Sie sind auf Seite 1von 927

INFECTIONS OF THE

CENTRAL NERVOUS
SYSTEM
FOURTH EDITION

Editors
W. Michael Scheld, MD Christina M. Marra, MD
Bayer-Gerald L. Mandell Professor of Infectious Professor of Neurology
Diseases Adjunct Professor of Medicine (Infectious Diseases)
Professor, Myles H. Thaler Center for AIDS and University of Washington School of Medicine
Human Retrovirus Research Seattle, Washington
Professor of Medicine
Clinical Professor of Neurosurgery
Director, Pfizer Initiative in
International Health
University of Virginia Health System
Charlottesville, Virginia

Richard J. Whitley, MD
Distinguished University Professor
Loeb Scholar in Pediatrics
Professor of Pediatrics, Microbiology, Medicine,
and Neurosurgery
University of Alabama at Birmingham
Birmingham, Alabama

Scheld_FM.indd i 2/22/14 5:41 AM


Acquisitions Editor: Julie Goolsby
Senior Product Development Editor: Kristina Oberle
Production Project Manager: David Orzechowski
Senior Manufacturing Coordinator: Beth Welsh
Marketing Manager: Stephanie Manzo
Design Manager: Teresa Mallon
Production Service: Absolute Service, Inc.

Copyright 2014 Wolters Kluwer Health


Two Commerce Square
2001 Market Street
Philadelphia, PA 19103 USA
LWW.com

2004 by Lippincott Williams & Wilkins

All rights reserved. This book is protected by copyright. No part of this book may be repro-
duced in any form by any means, including photocopying, or utilized by any information
storage and retrieval system without written permission from the copyright owner, except
for brief quotations embodied in critical articles and reviews. Materials appearing in this
book prepared by individuals as part of their official duties as U.S. government employees
are not covered by the above-mentioned copyright.

Printed in China

Library of Congress Cataloging-in-Publication Data


Infections of the central nervous system (Scheld)
Infections of the central nervous system / editors, W. Michael Scheld, Richard J. Whitley,
Christina M. Marra. Fourth edition.
p. ; cm.
Includes bibliographical references and index.
ISBN-13: 978-1-4511-7372-7 (hardback : alk. paper)
ISBN-10: 1-4511-7372-5 (hardback : alk. paper)
I. Scheld, W. Michael, editor of compilation. II. Whitley, Richard J., editor of compilation.
III. Marra, Christina M., editor of compilation. IV. Title.
[DNLM: 1. Central Nervous System Infections. WL 301]
RC359.5
616.8dc23
2014004822

Care has been taken to confirm the accuracy of the information presented and to
describe generally accepted practices. However, the authors, editors, and publisher are not
responsible for errors or omissions or for any consequences from application of the informa-
tion in this book and make no warranty, expressed or implied, with respect to the currency,
completeness, or accuracy of the contents of the publication. Application of the information
in a particular situation remains the professional responsibility of the practitioner.
The authors, editors, and publisher have exerted every effort to ensure that drug
selection and dosage set forth in this text are in accordance with current recommendations
and practice at the time of publication. However, in view of ongoing research, changes in
government regulations, and the constant flow of information relating to drug therapy and
drug reactions, the reader is urged to check the package insert for each drug for any change
in indications and dosage and for added warnings and precautions. This is particularly
important when the recommended agent is a new or infrequently employed drug.
Some drugs and medical devices presented in the publication have Food and Drug
Administration (FDA) clearance for limited use in restricted research settings. It is the
responsibility of the health care provider to ascertain the FDA status of each drug or device
planned for use in their clinical practice.
10 9 8 7 6 5 4 3 2 1

Scheld_FM.indd ii 2/22/14 5:41 AM


WITH GRATITUDE TO ALL OF OUR MENTORS
AND
TO OUR TRAINEES WHO UNIFORMLY GAVE MORE
THAN THEY RECEIVED

Scheld_FM.indd iii 2/22/14 5:41 AM


Scheld_FM.indd iv 2/22/14 5:41 AM
CONTRIBUTORS

Philipp Agyeman, MD Itzhak Brook, MD, MSc


Research Fellow Professor
Neuroinfection Laboratory, Institute for Infectious Diseases Department of Pediatrics
University of Bern Georgetown University School of Medicine
Attending Physician Attending Physician in Infectious Diseases, Pediatrics
Department of Pediatrics Georgetown University Hospital
University of Bern Washington DC, Washington
Inselspital
Bern, Switzerland Mary T. Caserta, MD
Professor
Kelly J. Baldwin, MD Department of Pediatrics
Clerkship Director University of Rochester Medical Center School of Medicine
Department of Neurology and Dentistry
Temple University, School of Medicine Attending Physician
Philadelphia, Pennsylvania Department of Pediatrics
Associate Golisano Childrens Hospital
Department of Neurology Rochester, New York
Geisinger Medical Center
Danville, Pennsylvania Kevin A. Cassady, MD
Associate Professor
Kyra J. Becker, MD Department of Pediatric Infectious Diseases
Professor University of Alabama at Birmingham
Department of Neurology and Neurological Surgery Birmingham, Alabama
University of Washington School of Medicine
Seattle, Washington Matthias Cavassini
Private-Decent and Senior Lecturer
J. David Beckham, MD Chief of Service, Department Chair
Assistant Professor of Medicine and Neurology Service of Infectious Diseases, Department of Medicine
Departments of Medicine (Infectious Diseases) and University Hospital of Lausanne
Neurology Lausanne, Switzerland
University of Colorado School of Medicine
Aurora, Colorado Maxine Caws, PhD, MSc, BSc
Research Lecturer
Jeana L. Benwill, MD Department of Clinical Sciences
Assistant Professor of Medicine Liverpool School of Tropical Medicine
The University of Texas Health Science Center at Tyler Liverpool, United Kingdom
Tyler, Texas Head of TB Research Programme
Oxford University Clinical Research Unit
Sven Bergstrm, PhD Hospital for Tropical Diseases
Professor Ho Chi Minh City, Vietnam
Department of Molecular Biology
Ume University Won K. Chung, MD
Ume, Sweden Post-doctoral Fellow
Department of Internal Medicine, Division of Infectious
Ari Bitnun, MD, MSc, FRCPC Disease
Associate Professor University of Texas Medical Branch
Department of Pediatrics Galveston, Texas
University of Toronto
Staff Physician David J. Coffey, MD
Department of Pediatrics Associate Professor
The Hospital for Sick Children Department of Neurology
Toronto, Ontario, Canada Geisel School of Medicine at Dartmouth
Lebanon, New Hampshire

Scheld_FM.indd v 2/22/14 5:41 AM


vi Contributors

Jeffrey I. Cohen, MD Carol Glaser, MD


Chief Chief
Laboratory of Infectious Diseases Encephalitis and Special Investigations Section
National Institute of Allergy and Infectious Diseases, Division of Communicable Disease Control
National Institutes of Health California Department of Public Health
Bethesda, Maryland Richmond, California
Associate Clinical Professor
Amanda C. Cohn, MD Department of Pediatrics, Division of Pediatric Infectious
Medical Epidemiologist Diseases
Division of Bacterial Diseases University of California, San Franciso
National Center for Immunizations and Respiratory Diseases, San Francisco, California
Centers for Disease Control and Prevention
Atlanta, Georgia John W. Gnann, Jr., MD
Professor
Moshe Ephros, MD Department of Medicine, Division of Infectious Diseases
Associate Clinical Professor Medical University of South Carolina
Department of Pediatrics Charleston, South Carolina
Faculty of Medicine
Technion-Israel Institute of Technology Denis Grandgirard, PhD
Director Senior Postdoc
Pediatric Infectious Disease Unit Neuroinfection Laboratory
Department of Pediatrics Institute for Infectious Diseases
Carmel Medical Center University of Bern
Haifa, Israel Bern, Switzerland

Jeremy Farrar, FRCP, FRCP(Ed), FMedSci, PhD, OBE Diane E. Griffin, MD, PhD
Director Professor and Alfred and Jill Sommer Chair
Oxford University Clinical Research Unit W. Harry Feinstone Department of Molecular Microbiology
Wellcome Trust Major Overseas Programme and Immunology
Ho Chi Minh City, Vietnam Johns Hopkins Bloomberg School of Public Health
Baltimore, Maryland
Kathleen R. Fink, MD
Assistant Professor Paul D. Griffiths, MD, DSc
Department of Radiology Professor of Virology
University of Washington Centre for Virology
Harborview Medical Center University College London Medical School
Seattle, Washington Royal Free London NHS Foundation Trust
London, United Kingdom
Sven Forner, BA
CJD Clinical Research Team John J. Halperin, MD
University of California, San Francisco Professor
UCSF Memory and Aging Center Departments of Neurology and Medicine
San Francisco, California Icahn School of Medicine at Mount Sinai
New York, New York
Michael D. Geschwind, MD, PhD Chair
Associate Professor Department of Neurosciences
Michael J. Homer Chair in Neurology Overlook Medical Center
University of California, San Francisco Summit, New Jersey
UCSF Memory and Aging Center
San Francisco, California Barry J. Hartman, MD
Clinical Professor of Medicine
Michael Giladi, MD, MSc Department of Medicine, Division of Infectious Diseases
Associate Professor of Medicine Weill Cornell Medical Center
Sackler Faculty of Medicine Attending Physician
Tel Aviv University Department of Medicine
The Infectious Disease Unit and the Bernard Pridan New York Presbyterian Hospital
Laboratory for Molecular Biology of Infectious Diseases New York, New York
Tel Aviv Medical Center
Tel Aviv, Israel Rodrigo Hasbun, MD, MPH
Associate Professor
Stefano Giulieri, MD Department of Medicine, Infectious Diseases
Chief Resident University of Texas Health Science Center
Service of Infectious Diseases, Department of Medicine Attending Physician
University Hospital of Lausanne Department of Medicine
Lausanne, Switzerland Memorial Hermann Hospital
Houston, Texas

Scheld_FM.indd vi 2/22/14 5:41 AM


Contributors vii

Dorothee Heemskerk, MSc, MD Matthias Klein, MD


Clinical Research Fellow Attending Physician
Oxford University Clinical Research Unit VN Department of Neurology
University of Oxford University of Munich
Ho Chi Minh City, Vietnam Klinikum Grosshadern
Munich, Germany
David C. Helfgott, MD
Assistant Professor of Medicine Serggio C. Lanata, MD
Internal Medicine Clinical Fellow
Weill Cornell Medical College Department of Neurology
Assistant Attending Physician University of California, San Francisco
Department of Medicine, Division of Infectious Diseases UCSF Memory and Aging Center
New York Presbyterian Hospital San Francisco, California
New York, New York
Stephen L. Leib, MD
Jerzy Hildebrand, MD, PhD Associate Professor
Professor of Neurology Neuroinfection Laboratory, Institute for Infectious Diseases
Department of Medicine University of Bern
Institut Jules Bordet Bern, Switzerland
Universit Libre de Bruxelles Head
Brussels, Belgium Biology Division, Spiez Laboratory
Swiss Federal Office for Civil Protection
Marc Hildebrand, MD, PhD Austrasse
Department of Medicine, Division of Infectious Diseases Spiez, Switzerland
Hpitaux Iris Sud
Brussels, Belgium Matthias Maiwald, MD, PhD
Adjunct Associate Professor
Susan E. Hoover, MD, PhD Department of Microbiology
Associate Professor National University of Singapore
Internal Medicine Consultant in Microbiology
University of South Dakota Sanford School of Medicine Department of Pathology and Laboratory Medicine
Sanford Health KK Womens and Childrens Hospital
Sioux Falls, South Dakota Singapore, Singapore

Jennifer L. Horan, MD, PharmD Carrie P. Marder, MD, PhD


Medical Instructor Acting Instructor
Department of Medicine Department of Radiology
Duke University Medical Center University of Washington
Durham, North Carolina Department of Radiology
University of Washington Medical Center
Alan C. Jackson, MD, FRCPC Seattle, Washington
Professor
Departments of Internal Medicine (Neurology) and Medical James D. Marks, MD, PhD
Microbiology Professor and Vice Chairman
University of Manitoba Department of Anesthesia and Perioperative Care
Head University of California, San Francisco
Section of Neurology, Internal Medicine Chief of Anesthesia
Health Sciences Centre Department of Anesthesia and Perioperative Care
Winnipeg, Canada San Francisco General Hospital
San Francisco, California
David W. Kimberlin, MD
Professor of Pediatrics Christina M. Marra, MD
Department of Pediatrics Professor of Neurology
University of Alabama at Birmingham Adjunct Professor of Medicine (Infectious Diseases)
Birmingham, Alabama University of Washington School of Medicine
Seattle, Washington
Louis V. Kirchhoff, MD, MPH
Professor Matthew McCarthy, MD
Departments of Internal Medicine (Infectious Diseases) and Fellow
Epidemiology Department of Medicine
University of Iowa Health Care Weill Cornell Medical Center
Staff Physician New York, New York
Medical Service
Department of Veterans Affairs Medical Center
Iowa City, Iowa


Deceased

Scheld_FM.indd vii 2/22/14 5:41 AM


viii Contributors

Tony M. McGrath, MD Adjanie Patabendige, PhD


Associate Professor NC3Rs David Sainsbury Fellow
Department of Pediatrics, Division of Child Neurology Department of Clinical Infection, Microbiology and
University of Alabama at Birmingham Immunology
Attending Physician Institute of Infection and Global Health
Division of Child Neurology University of Liverpool
Childrens of Alabama Liverpool, United Kingdom
Birmingham, Alabama
John R. Perfect, MD
Thomas O. McPharlin, RPh Professor
Clinical Associate Professor Department of Medicine
University of Washington School of Pharmacy Duke University Medical Center
Clinical Pharmacist Durham, North Carolina
Department of Neurology and Rehabilitation Medicine
Pharmacy William A. Petri, Jr., MD, PhD
Harborview Medical Center Professor and Chief
Seattle, Washington Division of Infectious Diseases
University of Virginia School of Medicine
Nancy E. Messonnier, MD Charlottesville, Virginia
Medical Epidemiologist
Division of Bacterial Diseases Hans-Walter Pfister, MD
National Center for Immunizations and Respiratory Diseases, Senior Consultant
Centers for Disease Control and Prevention Department of Neurology
Atlanta, Georgia University of Munich
Senior Consultant
Reto Antoine Meuli, MD, PhD Department of Neurology
Full Professor Klinikum Grosshadern
Department of Radiology Munich, Germany
University of Lausanne, Faculty of Biology and Medicine
Chief of Service, Department Chair Douglas G. Postels, MD
Department of Radiology Associate Professor
CHUV, University Hospital of Lausanne Department of Neurology
Lausanne, Switzerland Michigan State University
East Lansing, Michigan
Augusto Miravalle, MD
Assistant Professor of Neurology Didier Raoult, MD, PhD
Director, Neurology Residency Training Program Director
University of Colorado Denver School of Medicine URMITE UMR 7278, Facult de Mdecine
Aurora, Colorado Aix-Marseille Universit
Chief
John F. Modlin, MD Fdration de Microbiologie Clinique
Professor of Pediatrics and Medicine Hpital de la Timone
Department of Pediatrics and Medicine Marseille, France
Geisel School of Medicine at Dartmouth
Hanover, New Hampshire David A. Relman, MD
Deputy Director for Research, Polio Thomas C. and Joan M. Merigan Professor
Global Development Departments of Medicine and Microbiology and Immunology
Bill & Melinda Gates Foundation Stanford University School of Medicine
Seattle, Washington Stanford, California
Chief
Jose G. Montoya, MD, FACP, FIDSA Infectious Diseases Section
Professor Veterans Affairs Palo Alto Health Care System
Department of Medicine Palo Alto, California
Stanford University
Attending Physician Susan Richardson, MD, CM
Department of Medicine Professor
Stanford Hospital and Clinics Department of Laboratory Medicine and Pathobiology
Stanford, California University of Toronto
Head
Shannon Moonah, MD, ScM Department of Paediatric Laboratory Medicine, Division of
Clinical and Research Fellow Microbiology
Division of Infectious Diseases The Hospital for Sick Children
University of Virginia School of Medicine Toronto, Ontario, Canada
Charlottesville, Virginia

Scheld_FM.indd viii 2/22/14 5:41 AM


Contributors ix

Jos R. Romero, MD Khoi Duc Than, MD


Professor Chief Resident
Department of Pediatrics Department of Neurosurgery
University of Arkansas for Medical Sciences University of Michigan
Director Ann Arbor, Michigan
Section of Infectious Diseases
Arkansas Childrens Hospital Allan R. Tunkel, MD, PhD
Little Rock, Arkansas Professor of Medicine
Associate Dean for Medical Education
Karen L. Roos, MD Warren Alpert Medical School of Brown University
John and Nancy Nelson Professor of Neurology Providence, Rhode Island
Professor of Neurological Surgery
Indiana University Health Neuroscience Center Kenneth L. Tyler, MD
Indianapolis, Indiana Reuler-Lewin Family Professor and Chair of Neurology
Professor of Medicine & Microbiology
Jeffrey P. Ross, MD Departments of Neurology, Medicine, and Microbiology
Assistant Clinical Professor University of Colorado Denver School of Medicine
Medicine Chair
University of New Mexico School of Medicine Department of Neurology
Albuquerque, New Mexico University of Colorado Hospital
Aurora, Colorado
Oren Sagher, MD
William F. Chandler Collegiate Professor Diederik van de Beek, MD, PhD
Department of Neurosurgery Professor
University of Michigan Department of Neurology
Neurosurgery Faculty Academic Medical Center
Department of Neurosurgery University of Amsterdam
University of Michigan Health System Amsterdam, The Netherlands
Ann Arbor, Michigan
Arun Venkatesan, MD, PhD
W. Michael Scheld, MD Assistant Professor
Bayer-Gerald L. Mandell Professor of Infectious Diseases Department of Neurology
Professor, Myles H. Thaler Center for AIDS and Human Johns Hopkins University School of Medicine
Retrovirus Research Director
Professor of Medicine Encephalitis Center
Clinical Professor of Neurosurgery Johns Hopkins Hospital
Director, Pfizer Initiative in International Health Baltimore, Maryland
University of Virginia Health System
Charlottesville, Virginia Richard J. Wallace, Jr.
Chairman
Jose A. Serpa, MD, MS Department of Microbiology
Assistant Professor Chief
Department of Medicine Infectious Disease Section
Baylor College of Medicine The University of Texas Health Science Center at Tyler
Attending Physician Tyler, Texas
Medicine
Ben Taub Hospital Thomas J. Walsh, MD, PhD (hon), FCCP, FAAM, FIDSA
Houston, Texas Director
Transplantation-Oncology Infectious Diseases Program
Tom Solomon, MD Chief
Director Infectious Diseases Translational Research Laboratory
Institute of Infection and Global Health Professor of Medicine, Pediatrics, and Microbiology &
University of Liverpool Immunology
Honorary Consultant Neurologist Henry Schueler Foundation Scholar
Department of Neurology Weill Cornell Medical Center and New York Presbyterian
Walton Centre NHS Foundation Trust Hospital
Liverpool, United Kingdom New York, New York

Terrie E. Taylor, DO Anthony C. Wang, MD


University Distinguished Professor Department of Neurosurgery
Osteopathic Medical Specialties University of Michigan
Michigan State University College of Osteopathic Medicine House Officer
East Lansing, Michigan Department of Neurosurgery
Scientific Director University of Michigan Health System
Blantyre Malaria Project Ann Arbor, Michigan
University of Malawi College of Medicine
Blantyre, Malawi

Scheld_FM.indd ix 2/22/14 5:41 AM


x Contributors

David F. Welch, PhD, D(ABMM) Joseph R. Zunt, MD, MPH


Medical Microbiologist Professor
Department of Pathology Departments of Neurology, Global Health, Medicine
Medical City (Infectious Diseases), Epidemiology
Dallas, Texas University of Washington
Attending Neurologist
A. Clinton White, Jr., MD Department of Neurology
Paul R. Stalnaker Distinguished Professor and Director Harborview Medical Center
Infectious Disease Division, Department of Internal Medicine Seattle, Washington
University of Texas Medical Branch
Galveston, Texas John Zurasky, MD
Neurocritical Care Medical Director
Richard J. Whitley, MD Department of Neurology
Distinguished University Professor Chair of Neurology
Loeb Scholar in Pediatrics Providence Health and Services Oregon
Professor of Pediatrics, Microbiology, Medicine, and Portland, Oregon
Neurosurgery
University of Alabama at Birmingham
Birmingham, Alabama

Gary P. Wormser, MD
Professor of Medicine
Departments of Microbiology and Immunology and
Pharmacology
New York Medical College
Chief
Division of Infectious Diseases
Westchester Medical Center
Valhalla, New York

Scheld_FM.indd x 2/22/14 5:41 AM


PREFACE

The first edition of Infections of the Central Nervous System completely with essentially the same team of authors (e.g.,
was published in 1991, a comprehensive treatise addressing Chapter 23 on the Pathogenesis and Pathophysiology of
all aspects of central nervous system (CNS) infections for Bacterial Infections). We have added one new chapter, on
advanced readers. In its preface, we clearly stated our goal: to acute encephalitis, by Glaser and Venkatesan. A discussion of
develop the gold standard reference text using the best infor- the diagnostic approach to the acute encephalitis syndrome
mation from the best authors with the best format. Judging was lacking in the last edition as well as any mention of some
from the reviews that followed publication, and from feed- noninfectious entities (e.g., antiNMDA receptor encephalitis)
back that we received from colleagues, we believe that the first literally unknown at the time. Dr. Glaser headed the California
edition succeeded in meeting our objectives. Encephalitis Project for much of the last decade, an effort
The second edition of Infections of the Central Nervous which has contributed substantially to our current knowledge
System was published in 1997 with identical goals and objec- of encephalitis.
tives. The same format was followed, but the 37 chapters of As with prior editions, we chose contributors with clinical
the first edition were expanded to 51 chapters. Reviews and experience as well as basic and/or clinical investigative inter-
comments were again positive. The third edition, of 50 chap- ests in their topic. Although many of the primary authors of
ters, was published in 2004, with a similar positive reception the chapters of the third edition have been retained, fully 21
from readers. A great deal of new information has accumu- of the 51 chapters of this edition employ a new author team
lated in the past decade, and we wanted to further improve (11 chapters have completely new author teams). In choosing
the book in other ways: hence the fourth edition. With this these new authors, we attempted to maintain the excellence of
explosion of new knowledge, the text has changed dramati- the prior editions while emphasizing cutting edge science and
cally, but we have retained the same editorial team. In ad- a more international perspective. This is an outstanding group
dition, we have kept the same basic format: approximately overall drawn from the disciplines of medicine, pediatrics,
50 chapters divided into 10 sections. infectious diseases, neurology, neurosurgery, neurointensive
As for previous editions, the first three chapters of the book care, neuroradiology, virology, epidemiology, parasitology,
cover the approach to diagnosis of CNS infections, including vaccines and prevention, and the basic neurosciences. Tables,
detailed discussion of diagnostic tests. The chapters that fol- illustrations, and photographs have again been used liberally.
low provide an in-depth discussion of individual infectious In many chapters, more than 50% of the references have been
agents and the CNS diseases that they produce in humans, published since 2012.
including differential diagnosis, clinical symptoms and find- We plan to further develop and refine the book through
ings, abnormalities on laboratory and imaging studies, treat- future editions. We will continue to provide a comprehensive
ment, and prevention. readable resource for all physicians who deal with infections
Although the number of chapters is approximately the of the CNS. We welcome your comments.
same as the third edition (51 versus 50), several other changes
are noteworthy. Every chapter has been extensively revised
and updated appropriately, with cited references through W. Michael Scheld, MD
early 2014. Some, reflecting a huge amount of new infor- Richard J. Whitley, MD
mation accumulated in the past decade, have been rewritten Christina M. Marra, MD

xi

Scheld_FM.indd xi 2/22/14 5:41 AM


Scheld_FM.indd xii 2/22/14 5:41 AM
ACKNOWLEDGMENTS

We thank everyone who has helped us in the preparation of We are particularly grateful to our assistants, Lisa Cook and
this large book. Most importantly, we thank all of the authors Dunia Ritchey. The editorial staff at Lippincott Williams &
for their outstanding contributions, especially those who have WilkinsJulie Goolsby, acquisitions editor, and Kristina
replaced prior author teams. As editors, we were privileged Oberle, development editordeserve our gratitude for ensur-
to see their work first; as students of CNS infections, we ing completion of the project. Finally, we thank our families
admire their special insights and expertise. Numerous other for their tolerance and support during interminable hours
colleagues provided helpful discussion, advice, and criticism. required to bring this undertaking to closure.

xiii

Scheld_FM.indd xiii 2/22/14 5:41 AM


Scheld_FM.indd xiv 2/22/14 5:41 AM
PREFACE TO THE FIRST EDITION

From the brain, and from the brain only, arise our pleasures, joys, sequelae, elegantly described by Hippocrates above, may de-
laughter and jests, as well as sorrows, pains, griefs and tears.... It is prive survivors of hearing, intellect, or function, demeaning
the same thing which makes us mad or delirious, inspires us with the quality of human life and burdening health resources and
dread or fear, whether by night or by day, brings sleeplessness, social services.
inopportune mistakes, aimless anxieties, absent-mindedness, and
acts that are contrary to habit. These things that we suffer all come
The distinctive nature and natural history of CNS infections
from the brain, when it is not healthy, but becomes abnormally set them somewhat apart from the mainstream of infectious
hot, cold, moist or dry. diseases. The scope of todays knowledge of these infections
can no longer be presented adequately within the confines of
Hippocrates, The Sacred Disease, Section XVII a subsection in a general textbook. Indeed, the understanding
and management of CNS infections is evolving toward a sub-
Every physician, almost, hath his favourite disease, to which he specialty in its own right. For these reasons, a new major text
ascribes all the victories obtained over human nature. The gout, seems justifiedhence this book, devoted to a comprehensive
the rheumatism, the stone, the gravel, and the consumption have
coverage of human CNS infections.
all their several patrons in the faculty; and none more than the
nervous fever, or the fever on the spirits. The work is a comprehensive treatise for the advanced
reader on all aspects of CNS infections. The book occupies a
Henry Fielding, Tom Jones, Book II, Chapter 9 central niche between large general texts on pediatrics, medi-
cine, neurology, neurosurgery, and infectious diseases on the
I hasten to give you a sketch of the spotted fever in this place. It one hand, and specialized single-subject treatises on the other.
made its first appearance about the beginning of January last: but We have assembled an outstanding group of contributors,
the instances were few and distant from each other, until last week. drawn from the ranks of internal medicine, pediatrics, neurol-
Although it had proved fatal in most instances, seven only had died
ogy, neurosurgery, infectious diseases, epidemiology, virology,
belonging to this town, previous to the 25th of February. Since that
time the disorder has come upon us like a flood of mighty waters. neuroradiology, and the basic neurosciences.
We have buried eight persons within the last eight days. About After a brief introduction that emphasizes the syndrome-
twelve or fifteen new cases appeared on Thursday last; many of oriented clinical approach to the patient with a CNS syndrome
them very sudden and violent. This was the most melancholy and and fever, the book is divided into parts based on microorgan-
alarming day ever witnessed in this place. Seven or eight physicians isms. The major CNS pathogens (viruses, bacteria, fungi, and
were continually engaged in the neighborhood north of the meet- protozoa) receive the most attention, although rarer patho-
ing house, and I believe not one half hour passed in the forenoon gens such as mycoplasmas, slow viruses, and helminths are
without presenting a new case. Pale fear and extreme anxiety were also covered in depth. In keeping with our philosophy that
visible in every countenance....
advances in diagnosis, therapy, prognosis, and prevention
Reverend Festus Foster of Petersham, Massachusetts require better understanding of the pathogenesis and patho-
in a letter to the editor of The Worchester Spy, 6 March 1810 physiology of these disorders, an introductory chapter on
these subjects is included in each of the major sections of the
These vignettes concerning central nervous system (CNS) book. Within each section a syndromic approach has been
infections come down to us over a span of 25 centuries. maintained whenever possible, but in many instances we felt
The Reverend Fosters graphic description of an outbreak of that specific diseases required separate coveragefor example,
meningococcemia and meningococcal meningitis in the late tetanus, neurosyphilis, and Lyme disease. In some sections we
winter of 1810 makes it easy to understand why these infec- have separated processes that primarily present as meningitis
tions engendered fear among physicians and lay persons alike. or meningoencephalitis from those that usually present as
Today, even with the comforts of vastly better knowledge and focal CNS lesions. The book concludes with discussions on
treatments, CNS infections continue to pose serious problems two major diagnostic modalities: (i) evaluation of the cere-
in health care. Some CNS infections are common, occurring brospinal fluid and (ii) neurodiagnostic imaging by computed
either as sporadic cases or in epidemics. For example, major tomography and magnetic resonance imaging.
outbreaks of meningococcal disease have occurred in Africa In choosing the contributors, we have sought individuals
and Asia during the past few years. Furthermore, despite with clinical experience as well as with active basic and/or
the introduction of new antimicrobial agents and diagnostic clinical investigative interests in their topic. We asked them to
techniques, the mortality associated with some infections of take a comprehensive approach, ranging from recent advances
the central nervous system remains high, particularly in tu- in molecular pathogenesis to the clinical manifestations,
berculosis, pneumococcal, and gram-negative aerobic bacil- therapy, and prevention of CNS infections. We also estab-
lary meningitis; rabies; tetanus; cryptococcal meningitis in lished certain other ground rules. To gain a measure of unity
patients with acquired immunodeficiency syndrome (AIDS); among the chapters, each contributor was asked to write
and Jakob-Creutzfeldt disease. The morbidity associated under common subheadings: history of the syndrome, epide-
with CNS infections may be even more important than the miology, etiology, pathogenesis and pathology, clinical mani-
death rate, especially in developing countries. Neurologic festations, approach to diagnosis, therapy, and prevention.

xv

Scheld_FM.indd xv 2/22/14 5:41 AM


xvi Preface to the First Edition

We asked authors to provide an extensive but not exhaus- format. Inevitably, the size and complexity of the field means
tive bibliography, emphasizing classical papers and recent that we will fall short in some areas. Recognizing this, we hope
(19851991) references while limiting each chapter total to to develop and improve the book through future editions. Our
350 citations or fewer. We strongly encouraged the liberal use ambition will remain the same: to present the best available
of tables, drawings, and photographs. Although a degree of comprehensive resource and reference text for all who deal
overlap between chapters is inevitable (and sometimes even with infections of the central nervous system.
desirable) in a multiauthored volume, we have attempted to
minimize redundancies as much as possible.
From its inception 3 years ago, we intended that this should W. Michael Scheld
be a gold standard reference text. We set out to bring Richard J. Whitley
together the best information from the best authors in the best David T. Durack

Scheld_FM.indd xvi 2/22/14 5:41 AM


CONTENTS

Contributors v
Preface xi
Acknowledgments xiii
Preface to the First Edition xv

PART I APPROACH TO THE PATIENT AND DIAGNOSTIC EVALUATION

Chapter 1 Introduction: Approach to the Patient with


Central Nervous System Infection 1
Christina M. Marra, Richard J. Whitley, and W. Michael Scheld

Chapter 2 Cerebrospinal Fluid in Central Nervous System Infections 4


Rodrigo Hasbun

Chapter 3 Imaging of Intracranial Infections 24


Carrie P. Marder and Kathleen R. Fink

PART II VIRAL INFECTIONS AND RELATED DISORDERS

Chapter 4 Pathogenesis and Pathophysiology of


Viral Infections of the Central Nervous System 49
Kevin A. Cassady and Richard J. Whitley

Chapter 5 Viral Meningitis and Aseptic Meningitis Syndrome 65


Jos R. Romero

Chapter 6 Encephalitis 84
Carol Glaser and Arun Venkatesan

Chapter 7 Poliomyelitis, Polio Vaccines, and the Postpoliomyelitis Syndrome 112


John F. Modlin and David J. Coffey

Chapter 8 Measles and Rubella 125


Diane E. Griffin

Chapter 9 Herpes Simplex Virus 137


Richard J. Whitley

Chapter 10 Neurologic Manifestations of Varicella and Herpes Zoster 157


John W. Gnann, Jr. and Richard J. Whitley

Chapter 11 Cytomegalovirus 168


Paul D. Griffiths

xvii

Scheld_FM.indd xvii 2/22/14 5:41 AM


xviii Contents

Chapter 12 Epstein-Barr Virus 183


Susan E. Hoover, Jeffrey P. Ross, and Jeffrey I. Cohen

Chapter 13 Human Herpesvirus-6 191


Mary T. Caserta

Chapter 14 B Virus 204


Richard J. Whitley

Chapter 15 Arthropod-Borne Viral Encephalitides 210


Tom Solomon, Adjanie Patabendige, and Richard J. Whitley

Chapter 16 Meningitis and Encephalitis Caused by Mumps Virus 239


John W. Gnann, Jr.

Chapter 17 Rabies 251


Alan C. Jackson

Chapter 18 Human Prion Diseases 261


Serggio C. Lanata, Sven Forner, and Michael D. Geschwind

Chapter 19 Human Immunodeficiency Virus 286


Christina M. Marra

Chapter 20 Guillain-Barr Syndrome 299


Tony M. McGrath

Chapter 21 Acute Viral Myelitis 315


J. David Beckham and Kenneth L. Tyler

Chapter 22 Postinfectious Encephalomyelitis 331


Karen L. Roos and Augusto Miravalle

PART III BACTERIAL AND MYCOPLASMAL INFECTIONS

Chapter 23 Pathogenesis and Pathophysiology of Bacterial Infections 341


Philipp Agyeman, Denis Grandgirard, and Stephen L. Leib

Chapter 24 Acute Bacterial Meningitis 365


Karen L. Roos, Allan R. Tunkel, Diederik van de Beek, and W. Michael Scheld

Chapter 25 Mycoplasmal and Ureaplasmal Infections 420


Ari Bitnun and Susan Richardson

Chapter 26 Bartonella Infections, Including Cat-Scratch Disease 434


Michael Giladi, Moshe Ephros, and David F. Welch

Chapter 27 Rickettsioses, Anaplasmoses, and Q Fever 444


Didier Raoult

Chapter 28 Whipples Disease 461


Matthias Maiwald and David A. Relman

Chapter 29 Tuberculous Meningitis 474


Dorothee Heemskerk, Jeremy Farrar, and Maxine Caws

Chapter 30 Infections Due to Nontuberculous Mycobacteria 501


Jeana L. Benwill and Richard J. Wallace, Jr.

Chapter 31 Brain Abscess 522


Matthias Klein, Hans-Walter Pfister, Allan R. Tunkel, and W. Michael Scheld

Chapter 32 Epidural Abscess 550


Hans-Walter Pfister, Matthias Klein, Allan R. Tunkel, and W. Michael Scheld

Scheld_FM.indd xviii 2/22/14 5:41 AM


Contents xix

Chapter 33 Subdural Empyema and Suppurative Intracranial Phlebitis 566


Barry J. Hartman and David C. Helfgott

Chapter 34 Complications of Infective Endocarditis 579


Stefano Giulieri, Reto Antoine Meuli, and Matthias Cavassini

Chapter 35 Iatrogenic Infections of the Central Nervous System 608


Kelly J. Baldwin and Joseph R. Zunt

PART IV CENTRAL NERVOUS SYSTEM SYNDROMES MEDIATED BY BACTERIAL TOXINS

Chapter 36 Botulism 621


James D. Marks

Chapter 37 Tetanus 634


Itzhak Brook

PART V SPIROCHETAL INFECTIONS

Chapter 38 Neurosyphilis 659


Christina M. Marra

Chapter 39 Neuroborreliosis: Nervous System Involvement with Borrelia Species 674


John J. Halperin, Sven Bergstrm, and Gary P. Wormser

PART VI FUNGAL INFECTIONS

Chapter 40 Fungal Meningitis 687


Jennifer L. Horan and John R. Perfect

Chapter 41 Space-Occupying Fungal Lesions 711


Matthew McCarthy and Thomas J. Walsh

PART VII PROTOZOAL AND HELMINTHIC INFECTIONS

Chapter 42 Cerebral Malaria 729


Douglas G. Postels and Terrie E. Taylor

Chapter 43 Toxoplasma gondii and Toxoplasmosis 745


Jose G. Montoya

Chapter 44 Trypanosomiasis 756


Louis V. Kirchhoff

Chapter 45 Free-Living and Parasitic Amebic Infections 770


Shannon Moonah and William A. Petri, Jr.

Chapter 46 Helminthic Infections 776


Jose A. Serpa, Won K. Chung, and A. Clinton White, Jr.

Scheld_FM.indd xix 2/22/14 5:41 AM


xx Contents

PART VIII CHRONIC MENINGITIS AND MENINGITIS OF NONINFECTIVE OR


UNCERTAIN ETIOLOGY

Chapter 47 Chronic Meningitis Syndrome and Meningitis of 805


Noninfective or Uncertain Etiology
Jerzy Hildebrand and Marc Hildebrand

PART IX NEUROSURGICAL MANAGEMENT

Chapter 48 Surgical Management of Central Nervous System Infections 819


Anthony C. Wang, Khoi Duc Than, and Oren Sagher

Chapter 49 Critical Care of Central Nervous System Infections 849


John Zurasky, Thomas O. McPharlin, and Kyra J. Becker

PART X PREVENTION

Chapter 50 Vaccines for Viral Diseases with Significant


Central Nervous System Manifestations 859
David W. Kimberlin

Chapter 51 Vaccines Against Bacterial Meningitis 876


Amanda C. Cohn and Nancy E. Messonnier

Index 891

Scheld_FM.indd xx 2/22/14 5:41 AM


PART I APPROACH TO THE PATIENT AND
DIAGNOSTIC EVALUATION
CHAPTER 1 INTRODUCTION: APPROACH TO
THE PATIENT WITH CENTRAL NERVOUS SYSTEM
INFECTION
CHRISTINA M. MARRA, RICHARD J. WHITLEY, AND W. MICHAEL SCHELD

Infections of the central nervous system (CNS) are notable of brain or spinal cord herniation after lumbar puncture.
for their diversity. They range from common to rare, acute to Such findings mandate neuroimaging before lumbar puncture.
chronic, and benign to fatal. Although some are self-limited Identification of concomitant pneumonia, diarrhea, and skin
or are easily cured with modern treatment, others are relent- or bone lesions may offer clues to the etiology of infection.
lessly progressive despite treatment or have no known treat- Most importantly, findings on neurologic examination allow
ment. For the many CNS infections that are treatable, prompt for identification of the most likely site or sites of infection
diagnosis and aggressive management afford the best chance among cerebrospinal fluid (CSF) space, brain, or spinal cord
of recovery without sequelae. and allow for a syndrome recognition approach to diagno-
The clinical hallmarks of CNS infection are fever, head- sis, as described below.
ache, and alteration of mental status. Focal neurologic signs
may also be evident. Nonetheless, these four symptoms and
signs are nonspecific and can also be seen in noninfectious Acute Meningitis Syndrome
CNS syndromes. To narrow the differential diagnosis, other
characteristics must be evaluated. Among these, risk factors The dominant features of the acute meningitis syndrome are
for CNS infections are particularly helpful. Physical exami- acute onset over a few hours to a few days of fever, headache,
nation may also yield information that provides clues to the photophobia, stiff neck, and altered mental status. The latter
etiology of a given infection. An approach to the diagnosis of may range from simple irritability to confusion, obtundation,
CNS infections is shown in Figure 1.1. or coma. Vomiting may occur, especially in young children. In
many cases, there is no warning, but an acute upper respira-
tory tract infection may precede the onset of meningitis by
RISK FACTORS FOR CENTRAL a few days. The two leading causes of acute meningitis are
bacteria and viruses. The differential diagnosis includes nonin-
NERVOUS SYSTEM INFECTIONS fectious conditions, such as systemic lupus erythematosus and
Behet syndrome, or rare chemical meningitis caused by non-
Many infections of the CNS are geographically distributed or
steroidal antiinflammatory drugs.
occur seasonally. Therefore, a thorough travel history and con-
sideration of the date of onset of illness can provide clues to the
etiology. For example, Lyme disease is endemic in the northeast-
ern United States but uncommon in the southwestern states. Subacute or Chronic Meningitis Syndrome
Similarly, transmission of arborviral encephalitides requires
In contrast to acute meningitis, subacute and chronic men-
the presence of an insect vector, and thus these illnesses most
ingitis syndromes run their course over weeks, months, or
commonly occur in summer and fall. Certain environments
years. Because symptoms and signs may fluctuate, subacute or
facilitate acquisition and transmission of CNS infections, as ex-
chronic meningitides may be confused with the syndrome of
emplified by outbreaks of meningococcal infection in military
recurrent acute meningitis. Although in subacute and chronic
recruits and college students. Concomitant illnesses such as
meningitis the clinical findings of fever, headache, stiff neck,
HIV infection or diabetes, alcoholism, receipt of immunosup-
and altered mental status may resemble those of acute menin-
pressant medications, or cancer chemotherapy all predispose to
gitis, the time course is quite different. Onset is usually grad-
specific CNS infections. Similarly, receipt of prophylactic thera-
ual, often without any evident predisposing condition. Fever,
pies protects against individual CNS infections. For example,
though often present, tends to be lower and less hectic than in
primary prophylaxis against Pneumocystis jiroveci pneumonia
acute meningitis. The patient with chronic meningitis is likely
with trimethoprim-sulfamethoxazole in HIV-infected individu-
to be lethargic and generally debilitated, in addition to having
als decreases the risk of CNS toxoplasmosis.
symptoms referable to the CNS. Focal neurologic findings are
more common than in acute meningitis, although less common
than in the space-occupying syndromes.
CLUES ON PHYSICAL The differential diagnosis for subacute and chronic men-
EXAMINATION ingitis is extensive. The most likely infectious causes are
tuberculosis; fungal infections including cryptococcosis, coc-
Physical examination in the setting of suspected CNS infec- cidioidomycosis, and histoplasmosis; and spirochetal infections
tion has three purposes: (a) to identify contraindications to including syphilis and Lyme disease. Important noninfectious
lumbar puncture, (b) to identify concomitant sites of infec- conditions include sarcoidosis, systemic lupus erythematosus,
tion or pathology that provide clues to the infectious etiology, systemic or primary CNS vasculitides, and neoplastic menin-
and (c) to define the site of CNS infection. Depressed level gitis. Establishing a specific diagnosis is challenging. Of the
of consciousness, focal neurologic abnormalities, or seizures important treatable conditions, cryptococcosis and syphilis
may indicate a structural CNS abnormality that poses a risk usually can be diagnosed or excluded quickly on the basis of

Scheld_Ch01.indd 1 2/21/14 5:27 PM


2 Part I: Approach to the Patient and Diagnostic Evaluation

Assess Risks for Infection


Exposures
Travel
Environment (daycare, military)
Season
Concomitant illnesses
CMI dysfunction (HIV, organ transplant)
Neutropenia (cancer chemotherapy)
Diabetes
Alcoholism
Prophylactic agents

Perform Physical Examination


Assess safety of lumbar puncture
Identity concomitant illness or pathology
Pneumonia
Diarrhea
Skin or bone lesions

Define Probable Site of Infection Neuroimaging


CSF
Acute meningitis
Subacute or chronic meningitis
Recurrent meningitis
Brain
Acute encephalitis
Chronic encephalitis
Space-occupying lesions
Toxin-mediated syndromes
Encephalopathy with systemic infections
Postinfectious syndromes
Spinal cord
Acute encephalomyelitis
Chronic encephalomyelitis
Space-occupying lesions
Toxin-mediated syndromes
Postinfectious syndromes

Laboratory Evaluation
Pathogen-specific CSF Evaluation
Pathogen nonspecific

FIGURE 1.1 Approach to the diagnosis of CNS infections. CMI, cell mediated immunity.

Scheld_Ch01.indd 2 2/21/14 5:27 PM


Chapter 1: Introduction: Approach to the Patient with Central Nervous System Infection 3

serology or antigen detection. However, tuberculous meningi- as shooting sensations or electric shocks; third, motor
tis is more difficult to diagnose or exclude, and patients with weakness followed by sensory changes with bowel or bladder
suspected tuberculous meningitis should be treated empirically dysfunction; and fourth, paralysis, often accompanied by less-
while evaluation continues. The decision whether and when to ening in pain. The rate of progression from one stage to the
obtain a meningeal biopsy is complex and depends on many next is unpredictable. Because the incidence and severity of
factors, including the unfortunate fact that in practice even neurologic sequelae depend on the stage of the disease and
this invasive gold standard test often does not yield a defini- the degree of neural damage sustained before intervention,
tive diagnosis. this syndrome presents an emergency that requires immediate
diagnosis and treatment.

Acute Encephalitis Syndrome


Toxin-Mediated Syndromes
The acute encephalitis syndrome is characterized by inflam-
mation of the cerebral cortex and is most commonly caused Microbial toxins mediate several distinctive neurologic syn-
by viruses. It shares many features with the acute meningitis dromes. The leading examples are tetanus and botulism.
syndrome. Indeed, the two conditions often coexist as me- Toxin-mediated conditions are the least likely to show the
ningoencephalitis. Acute encephalitis may be either diffuse usual manifestations of CNS infection, including fever, head-
or focal. Focal encephalitis reflects tropism of some viruses ache, disturbance of consciousness, and focal neurologic signs.
for specific locations in the CNS, such as temporal lobe infec- For example, botulism is characterized by absence of fever and
tion by herpes simplex virus type 1 (HSV-1), or the anterior normal consciousness in most patients.
horn cells in flavivirus infections, such as West Nile. Nonviral
organisms that may produce the acute encephalitis syndrome
include Rickettsia, Mycoplasma, and Bartonella species Encephalopathy with Systemic Infection
that may cause encephalitis as one component of systemic
infection. Diverse conditions, including infective endocardi- Many systemic infections involve the CNS, for example, rick-
tis, Whipple disease, and recrudescent toxoplasmosis in the ettsial diseases, infective endocarditis, typhoid fever, malaria,
immunocompromised host may cause diffuse or focal acute and Whipple disease. Usually, the systemic manifestations of
encephalitis. the disease dominate the clinical picture, but sometimes the
CNS findings are prominent. In a few cases, CNS symptoms
are the only features. Because this is such a large and varied
Chronic Encephalitis Syndrome group of diseases, the syndromic approach to diagnosis is less
effective. In the setting of an undiagnosed CNS infectious syn-
The chronic encephalitis syndrome shares many features drome, an important principle is to consider systemic infection
with the acute encephalitis syndrome. However, the onset is as a possible underlying cause.
more gradual and the course is less hectic. The clinical find-
ings may be less dramatic or less severe. The patient with
chronic encephalitis is likely to be generally debilitated rather Postinfectious Syndromes
than acutely ill. Chronic encephalitis evolves over weeks to
months or years and relapses or recrudescences may occur. Several important CNS syndromes can develop following mi-
Complications such as pressure sores, contractures, or demen- crobial infections. The usual sequence begins with a common,
tia may ensue in the course of disease. often rather trivial, viral infection that may go unnoticed.
A postinfectious neurologic syndrome develops. Rarely, these
syndromes follow routine vaccinations. Examples include
Space-Occupying Lesion Syndrome postinfectious encephalitis, postinfectious encephalomyelitis,
and transverse myelitis. These reactions are presumably medi-
Patients with space-occupying brain lesions have focal neuro- ated by an immunologic response to the etiologic microbe or to
logic abnormalities referable to the location of the lesion or antigens revealed as a result of the initial infection. Although
lesions. These include cognitive abnormalities, weakness, sen- rare, these syndromes can be severe or fatal.
sory changes, and visual loss. Clinical manifestations, such as
headache, nausea, or vomiting, often begin intermittently, but
they progress steadily to a crisis at about the time the patient is CONCLUSION
admitted to the hospital. This crisis may consist of (a) a focal
or generalized seizure or (b) onset of obtundation progressing A systematic approach to the patient with a suspected CNS
to coma. infection can be undertaken. This includes assessment of risks
When a space-occupying lesion occurs in the extramedul- and a careful physical examination to assess safety of lumbar
lary space in the spinal canal, a distinctive set of manifestations puncture, identify non-CNS sites of infection, and define the
may develop in a typical sequence: first, localized back pain, site of CNS infection. A consideration of the different CNS
often severe; second, nerve root pain with associated altera- infection syndromes as outlined above can then be used to
tion in reflexes and sometimes paresthesias, often described promptly establish a diagnosis and implement therapy.

Scheld_Ch01.indd 3 2/21/14 5:27 PM


CHAPTER 2 CEREBROSPINAL FLUID IN
CENTRAL NERVOUS SYSTEM INFECTIONS
RODRIGO HASBUN

Infections within the central nervous system (CNS) frequently, The fourth ventricle drains into the subarachnoid space
but not always, produce changes in cerebrospinal fluid (CSF). through three small openings, the foramina of Luschka and the
The changes produced may provide invaluable information foramen of Magendie. The foramina of Luschka are located in
about the nature of the infectious process and, in many cases, the lateral recesses of the fourth ventricle and are absent in up
may permit specific identification of the offending organism. to 20% of the population. The foramen of Magendie is located
Despite the great diagnostic value of CSF analysis, however, in the midline and, in most persons, represents the major com-
injudicious attempts to obtain CSF (as in the setting of in- munication between the fourth ventricle and the subarachnoid
creased intracranial pressure) can sometimes cause brain her- space. As is discussed later, these narrow openings are impor-
niation or death, and casual handling of the CSF obtained may tant in CNS infections because they represent the sites at which
render the analysis useless. obstruction of CSF flow may most easily occur.
This chapter is divided into three parts. The first part re-
views the anatomy of the CSF spaces, the physiology of CSF
production and reabsorption, and the effect of infection on
CSF physiology and composition. The second part discusses
The Meninges and Subarachnoid Space
methods of CSF analysis in CNS infections, and the third part The brain and spinal cord are surrounded by three layers of
summarizes the CSF analysis in specific CNS infections. meninges (2). The outermost layer of the meninges is a tough
fibrous membrane, the dura mater. Within the skull, the dura
forms the inner layer of the cranial periosteum and is tightly
ANATOMY AND PHYSIOLOGY adherent to bone. Below the foramen magnum, the dura and
OF THE CEREBROSPINAL FLUID periosteum diverge and are separated by a fat-filled epidural
space. The middle layer of meninges, the arachnoid, is joined
COMPARTMENTS to the dura by a specialized layer of fibroblasts, the dural bor-
der cell layer. The cells of this inner dural border are devoid of
The CSF is contained within two connecting compartments, the collagen and have few cellular junctions, providing a cleavage
cerebral ventricles and the subarachnoid space (1). Infectious plane in which infection may develop and rapidly spread. The
organisms may affect both compartments, and analysis of CSF arachnoid covers the brain and spinal cord loosely and extends
from both may reflect changes produced by infectious or para- outward along the course of cranial and spinal nerves.
infectious processes within meninges, brain, or spinal cord. The third layer of meninges, the pia mater, is continuous
with the surface of the brain and spinal cord. The pia mater
also follows vessels into brain and spinal cord parenchyma
The Ventricular System and projects into the ventricles to form the choroid plexuses.
The pia mater and the ventricular ependyma merge at the
The cerebral ventricular system represents, in greatly elabo- foramina of Luschka and Magendie. The CSF is contained
rated form, the remnants of the embryologic neural tube. A in the subarachnoid space, enclosed between the arachnoid
single layer of neuroglial-derived cells, the ventricular epen- and the pia. The subarachnoid space surrounds the brain and
dyma, lines the ventricles; a dense network of astrocytic foot extends within the spinal canal to the level of the second sacral
processes backs these. The ventricular system consists of two vertebra. Within the skull, the subarachnoid space widens into
lateral ventricles, the third ventricle, and the fourth ventricle cisterns where pia and arachnoid are more widely separated by
(Fig. 2.1). The lateral ventricles are located within the cere- irregularities in the contour of the brain. The largest of these,
brum and consist of frontal, temporal, and occipital horns; the cisterna magna, surrounds the brainstem and the cerebel-
these join at the ventricular trigone within the parietal lobe. lum at the base of the skull and is occasionally used as a source
The third ventricle is an elongated, slitlike cavity that lies of CSF for analysis and culture. The subarachnoid space is
within the midbrain and is bounded inferiorly by the hypo- crossed by trabecular extensions of the arachnoid itself, by
thalamus. The fourth ventricle overlies the brainstem from the cranial nerves, by a network of small arteries, the rete mirabile,
level of the midpons to the extreme rostral end of the spinal and by numerous bridging veins, which connect the meningeal
cord. The roof of the fourth ventricle is the cerebellum poste- veins with the deeper intracranial venous system (2).
riorly and the superior and inferior medullary veli anteriorly. The subarachnoid space is normally a closed system.
The fourth ventricle is roughly diamond shaped and is wid- Occasionally, however, congenital or posttraumatic communi-
est at the lateral recesses, which lie between the superior and cations may exist between the subarachnoid space and super-
middle cerebral peduncles. ficial tissues and may provide a route for single or recurrent
The cerebral ventricles are connected to each other and with episodes of meningitis. Congenital defects arise from incom-
the subarachnoid space through a series of small openings. plete closure of the neural tube. These defects may extend for
Each lateral ventricle drains into the third ventricle through variable distances into subcutaneous tissues or to the cutaneous
the foramen of Monro, located in the inferomedial wall of the surface and are most common in the upper cervical regions
frontal horn. The third and fourth ventricles are connected by and over the sacrum. Their presence may be suggested by a
the aqueduct of Sylvius, which extends through the midbrain. cutaneous dimple or a patch of hair. Traumatic communications

Scheld_Ch02.indd 4 2/21/14 5:27 PM


Chapter 2: Cerebrospinal Fluid in Central Nervous System Infections 5

FIGURE 2.1 The cerebral ventricles. Inset:


Shown are the structure of the fourth ventricle
and the locations of the foramina of Luschka
and Magendie. (From Greenlee JE. Anatomical
considerations in central nervous system infec-
tions. In: Mandell GL, Bennett JE, Dolin R, eds.
Principles and Practice of Infectious Diseases.
4th ed. New York: Churchill Livingstone; 1994:
821831, with permission.)

into the subarachnoid space are most often associated with Formation of CSF involves both filtration and active
basilar skull fractures. The most common sites of involvement transport (1,3). Filtration of CSF varies inversely with
are (a) the thin layers of bone that separate the cranial cavity serum osmolality. In experimental animals, and possibly in
from the paranasal sinuses and (b) the petrous bone, which humans, CSF production changes 7% for each 1% change in
separates the auditory canals and mastoid from the cranial serum osmolality (4). Active secretion of CSF involves Na,
cavity. In rare instances, traumatic defects may occur over the K-adenosine triphosphatase (ATPase)mediated transport of
cranial convexities or along the spinal column. sodium across choroidal epithelium into the ventricular lumen,
with water, chloride, and bicarbonate ions following through
facilitated transport. In experimental animals, the carbonic
PHYSIOLOGY OF CEREBROSPINAL anhydrase inhibitor acetazolamide reduces CSF secretion by
approximately 50%, whereas furosemide and ethacrynic acid
FLUID PRODUCTION AND reduce CSF production by 25% to 35% (5). Simultaneous use
REABSORPTION of both agents reduces CSF formation by 75%.
Reabsorption of CSF occurs through arachnoid villi. Most
CSF is produced by the choroid plexuses of the lateral, of these are located along the superior sagittal sinus. Smaller
third, and fourth ventricles and, to a lesser extent, by extra- numbers of arachnoid villi are found along other intracranial
choroidal sites (1,3). In adults, the choroid plexus produces venous sinuses and around spinal nerve roots (1). During
approximately 500 mL of CSF per day, with 150 mL present health, the arachnoid villi along the superior sagittal sinus
in the ventricular system at any time. The choroid plexuses are provide the major site of CSF uptake. The arachnoid villi
specialized projections of vessels and pia mater into the ven- along other sinuses and surrounding spinal nerve roots may
tricular cavities. Each choroid plexus branches into frondlike provide alternative sites of CSF absorption following superior
villi, each of which contains a capillary surrounded by loose sagittal sinus thrombosis.
connective tissue and a layer of specialized ependymal cells Each arachnoid villus represents an extension of the arach-
termed choroid epithelium. Choroidal epithelial cells, in con- noid membrane through the dura mater into the lumen of the
trast to ependymal cells elsewhere in the ventricular system, venous sinus and functions as a one-way valve, permitting
are columnar in shape and are covered on their ventricular unidirectional flow from CSF into blood. Early work by
surfaces by a brush border of microvilli. The villous structure Welch (6) demonstrated that the arachnoid villi have a critical
of the choroid plexus and the presence of microvilli greatly in vitro opening pressure of 2 to 5 cm H2O; this study also
increase the surface area available for secretion of CSF (1). demonstrated that particles up to the size of erythrocytes

Scheld_Ch02.indd 5 2/21/14 5:27 PM


6 Part I: Approach to the Patient and Diagnostic Evaluation

that provide host defense elsewhere in the body. Normally,


T cells and B cells are present in very small numbers in CSF
and only rarely in brain; immunoglobulins and complement
are largely excluded from both CSF and brain; and opsonic
activity of CSF, even in the presence of meningitis, is far less
than that of serum (1114). Therefore, both the brain and the
CSF are poorly equipped to deal with infectious agents.
The barrier systems that isolate CSF, brain, and spinal
cord from blood are not static systems but, instead, are highly
dynamic in their ability to interact with and transport a
wide variety of substances (15). In addition, it is increasingly
recognized that the endothelial cells and astrocytes of the BBB
and the bloodCSF barrier are important sources of cytokines
(including tumor necrosis factor [TNF] and interleukins), and
that astrocytes, in addition to their abilities to regulate solute
entry into brain, have the ability to act as antigen-presenting
cells (16). The release of cytokines by endothelial cells and
astrocytes in response to bacterial endotoxins and other bacte-
rial products is fundamental in the production of inflammation
and injury during CNS infections and provides an extremely
important area for early therapy (9,1719).

MAINTENANCE OF
CEREBROSPINAL FLUID
HOMEOSTASIS
FIGURE 2.2 Uptake of CSF by an arachnoid villus. (From Fishman
RA. Cerebrospinal Fluid in Diseases of the Nervous System. 2nd ed. The BBB and the bloodCSF barrier maintain the cellular and
Philadelphia: WB Saunders; 1992, with permission.) chemical elements of the CSF within narrow ranges (1,3,20).
Lipid-soluble substances within blood readily diffuse across
choroidal epithelium or vascular endothelium into CSF or brain
(3). Passage of fluid and ionically polar substances, however,
readily pass from CSF into blood, whereas particles larger requires mechanisms for transport and facilitated diffusion.
than 7.5 m are excluded. Although these early data suggested Sodium enters CSF both by Na, K-ATPasemediated trans-
that the arachnoid villi might provide a direct communication port during secretion of CSF and by passive diffusion (20).
between CSF and blood, studies using electron microscopy Potassium is secreted into CSF by active transport mechanisms
have demonstrated that arachnoid villi and venous sinuses and is actively removed from CSF into brain by transport
are separated by a layer of endothelial cells connected by mechanisms that are believed to be located in astrocyte foot
tight junctions, and that movement of CSF and particulate processes. Movement of calcium, magnesium, and phospho-
matter across the arachnoid villi occurs by transport within rus into CSF and brain also occurs predominantly by active
giant vesicles (7,8) (Fig. 2.2). These giant vesicles, although transport, and the concentrations of these substances are rela-
they provide efficient transfer of CSF into blood under normal tively independent of their concentrations in serum. Chloride
circumstances, can become obstructed by bacteria and inflam- and bicarbonate, like potassium, are actively secreted into and
matory cells during meningitis or by red blood cells (RBCs) actively removed from CSF. Glucose, amino acids, amines,
during subarachnoid hemorrhage (9,10). and thyroid hormone enter the brain by carrier-mediated
transport mechanisms (1,15). Insulin and transferrin require
receptor-mediated transport (15). Although lipids complexed
BRAIN AND CEREBROSPINAL to proteins were once thought to be excluded from the CNS,
FLUID BARRIER SYSTEMS it is now known that complexed lipids undergo dissociation
from their carrier proteins at the bloodbrain interface and
The brain and CSF are contained within a series of barrier sys- may enter the CNS without significant exodus of protein from
tems (1). These prevent entry of fluids, electrolytes, and other brain capillaries (15).
substances from blood into CSF or brain by simple diffusion Chloride represents the major anion in CSF. Normal CSF
and isolate the CNS from systemic immune responses. The chloride concentration is 15 to 20 mEq/L higher than that in
bloodbrain barrier (BBB) is formed by tight junctions between serum. Early workers observed that CSF chloride concentrations
endothelial cells of CNS capillaries and is further reinforced were lowered in tuberculous meningitis; for many years, levels
by a surrounding layer of astrocytes, whose processes termi- of CSF chloride were used to diagnose and follow the course
nate in overlapping fashion on the capillary walls. In contrast, of this infection (1). It is now recognized, however, that the
the bloodCSF barrier is formed by the endothelial cells of the lowered CSF chloride concentration observed in tuberculous
choroid plexus and the tight junctions that link them. The cells meningitis is nothing more than a reflection of lowered serum
of the pia mater, like those of choroid plexus and arachnoid chloride values and has no diagnostic or prognostic value.
capillaries, are separated by gap junctions; entry of substances The acidbase balance of the CSF, like its electrolyte con-
from CSF into brain is modulated by a basement membrane centration, tends to remain fairly constant despite fluctuations
subjacent to the pia and by a continuous layer of astrocytes in systemic acidbase balance. In CSF, as opposed to plasma,
beneath the basement membrane, forming a CSFbrain barrier. however, movement of CO2 occurs readily by diffusion, whereas
The barrier systems that surround spinal cord and brain movement of bicarbonate occurs more slowly by carrier-
exclude from the CNS most of the immunologic mechanisms mediated transport. The discrepancy in the rate of movement

Scheld_Ch02.indd 6 2/21/14 5:27 PM


Chapter 2: Cerebrospinal Fluid in Central Nervous System Infections 7

of these two substances may produce delayed (and, at times, Occlusion of the aqueduct of Sylvius by granulomatous epen-
paradoxical) responses in CSF pH as compared to systemic pH dymitis may occur as a complication of tuberculosis, fungal
during rapid changes in bicarbonate concentration (1). The CSF infections, or sarcoidosis. Mumps virus, which replicates in ven-
acidbase balance is also maintained by the choroid plexuses, tricular ependymal cells, has been shown to produce congenital
which possess transport mechanisms capable of removing weak aqueductal stenosis in experimental animals (23). Rare cases of
organic acidsincluding antibiotics such as the penicillins, hydrocephalus have also been reported following mumps and
cephalosporins, and aminoglycosidesfrom CSF (21,22). with Toscana meningoencephalitis in humans (24,25). Extrinsic
Choroid plexus transport of antibiotics and other weak organic compression of the aqueduct of Sylvius may be produced by
acids can be blocked by probenecid. abscesses or other localized infections within the pons or mid-
brain. Involvement of the foramen of Monro is almost always
unilateral and is the consequence of severe brain shifts caused
ALTERATIONS OF by abscess, focal encephalitis, or hemorrhage. Hydrocephalus
CEREBROSPINAL FLUID caused by the occlusion of one foramen of Monro is particularly
dangerous because the CSF trapped within the involved lateral
DYNAMICS AND PRESSURE IN ventricle acts as a unilateral space-occupying lesion, greatly in-
CENTRAL NERVOUS SYSTEM creasing the risk of transtentorial brain herniation.
Computerized tomography (CT) and magnetic resonance
INFECTIONS: HYDROCEPHALUS, imaging (MRI) are invaluable in demonstrating the presence of
INTRACRANIAL HYPERTENSION, hydrocephalus and in determining its cause. Ventricular dilation
AND BRAIN HERNIATION is common in the elderly and is characterized by symmetric ven-
tricular dilation accompanied by evidence of cerebral cortical
Acute or chronic CNS infections may produce profound alter- atrophy. In contrast, hydrocephalus is defined as a frontal horn
ations in intracranial pressure (ICP) by obstructing CSF flow ratio (Evans index) of 0.3 or greater in the absence of cerebral
or reabsorption, by behaving as space-occupying lesions, or by atrophy (26). Hydrocephalus that occurs from impaired CSF
producing hemorrhage or cerebral edema. These pathologic circulation is accompanied by loss of cortical markings visible
consequences of infection, acting individually or together, may on CT or MRI as the brain is forced outward against the skull
cause brain herniation and death. and by periventricular areas of increased lucency, represent-
ing transependymal leakage of CSF. Communicating hydro-
cephalus and hydrocephalus from obstruction of the foramina
Alteration of Cerebrospinal Fluid Circulation of Luschka and Magendie are characterized by symmetric
enlargement of all four ventricles. Hydrocephalus from occlu-
in Central Nervous System Infections sion of the fourth ventricle or aqueduct of Sylvius results in
loss of that structure on CT or MRI, with dilation of the third
Impairment of normal CSF circulation may result in ventricu-
and lateral ventricles. Hydrocephalus following compression
lar enlargement and hydrocephalus. Interruption of CSF reab-
of the foramen of Monro is almost invariably associated with
sorption produces communicating hydrocephalus with normal
an identifiable space-occupying lesion and a prominent midline
circulation of CSF through the ventricular system and into the
shift. Thrombosis of the superior sagittal sinus may be difficult
subarachnoid space. Communicating hydrocephalus is a com-
to detect as a cause of communicating hydrocephalus and can
mon complication of bacterial meningitis and, in most cases,
be missed with the use of routine CT scanning. MRI and CT
results from obstruction of the arachnoid villi by bacteria and
venogram are more sensitive and are used to diagnose superior
white blood cells (WBCs) (9). Communicating hydrocephalus
sagittal sinus (SSS) thrombosis (27).
may also result from functional occlusion of arachnoid villi
during severe meningitis or by RBCs in the course of subarach-
noid hemorrhage during bland or septic subarachnoid hemor-
rhage (10). Thrombosis of the superior sagittal sinus may also Intracranial Hypertension and
block CSF reabsorption and thereby produce communicating Brain Herniation
hydrocephalus. Occlusion of a large portion of the superior
sagittal sinus usually produces catastrophic, often hemor- The normal mechanisms of CSF secretion and drainage main-
rhagic, cerebral infarction. Involvement of the anterior third tain CSF pressure at a level less than 150 mm of CSF in most
of the sinus, however, may be clinically silent except for the patients. Infection, however, greatly alters these homeostatic
development of hydrocephalus. mechanisms; moreover, death during the acute stages of intra-
Obstructive hydrocephalus results from interruption of CSF cranial CNS infections often results from extreme elevation in
flow within the ventricular system or at its point of exit into ICP followed by brain herniation and respiratory arrest.
the subarachnoid space (2). This may be the consequence of For a period of time, the intracranial contents are able to
infection of the ventricular ependyma or basilar meninges or compensate in response to space-occupying lesions before a rise
may result from extrinsic compression of the ventricular sys- in ICP occurs. This compensatory ability is termed compliance
tem by infection within brain parenchyma. Lesions producing (dV/dP) and represents the ratio of changes in volume (dV) to
obstructive hydrocephalus most commonly involve the ventric- changes in pressure (dP). Compliance in response to space-
ular system at its narrowest points: the foramina of Luschka and occupying intracranial lesions consists of several factors.
Magendie, the fourth ventricle, the aqueduct of Sylvius, and the These include increased rate of reabsorption of CSF (this may
foramina of Monro. Obstruction of the foramina of Luschka be prevented in meningitis by obstruction of the arachnoid
and Magendie is characteristic of exudative basilar menin- villi by cells and exudate); displacement of CSF; reduction
gitides such as those caused by Mycobacterium tuberculosis, in the total volume of intracranial blood, predominantly by
Coccidioides immitis, and Cryptococcus neoformans but may compression of veins and venous sinuses; and plasticity of the
also be seen in bacterial meningitis. Hydrocephalus as a result brain itself. Compliance is extremely limited when infection is
of obliteration of the fourth ventricle is almost always extrinsic accompanied by a rapid increase in ICP, such as during acute
and is the result of ventricular compression by large cerebel- bacterial meningitis or subdural empyema. In contrast, the
lar mass lesions such as cerebellar abscess or hemorrhage. ability of CNS compliance to compensate for increased ICP

Scheld_Ch02.indd 7 2/21/14 5:27 PM


8 Part I: Approach to the Patient and Diagnostic Evaluation

may be extensive where space-occupying lesions develop over patients with a negative head CT scan. The procedure is of
time (28). Once compliance is exceeded, however, the increase little specific diagnostic value in the diagnosis of brain abscess
in pressure in chronic lesions may occur rapidly. or parameningeal infections. Lumbar punctures (LPs) should
The elevation in CSF pressure seen in infections and other not be done in patients with impending herniation or with
pathologic conditions is not constant but fluctuates consider- intracranial mass lesions with severe mass effect. Furthermore,
ably. This fluctuation is usually not observed during the brief inappropriate LP can cause patient death or serious neurologic
period of measurement provided by LP but becomes an impor- injury, and the procedure should never be initiated without
tant parameter to observe during monitoring of ICP. Minor consideration of its potential danger to the patient.
variation in pressure occurs during Cheyne-Stokes respiration Clinicians have relied on the meningeal signs (nuchal rigid-
and during variations in blood pressure produced by Hering- ity, Kernig sign, Brudzinski sign) for over 100 years to evaluate
Breuer reflexes, the inflation and deflation reflexes that help patients with suspected meningitis to help them decide who
regulate the rhythmic ventilation of the lungs. More major should undergo a LP. A prospective study of 297 adults with
variations in ICP occur during plateau waves. These are suspected meningitis documented a very low sensitivity of the
abrupt elevations in ICP (usually lasting 5 to 20 minutes) in Kernig sign (sensitivity, 5%), Brudzinski sign (sensitivity, 5%),
which ICP may reach 600 to 1,300 mm of CSF (50 to 100 and nuchal rigidity (sensitivity, 30%) (35). The absence of the
mm Hg) (29,30). Plateau waves are believed to represent a meningeal signs should not defer the performance of the LP.
consequence of disturbed cerebrovascular autoregulation be- The decision to perform a LP on those suspected of having
cause of either abnormal sympathetic tone or cyclic changes meningitis is largely based on a combination of clinical signs
in perfusion in which mild hypotension is followed by cerebral and symptoms at presentations. The classic triad of fever, stiff
vasodilation and increased cerebral blood flow (30). Although neck, and altered mental status was present in only 44% of
plateau waves may be without any detectable clinical effect, patients in a prospective study involving 696 patients with
they may also be associated with signs of brainstem compres- confirmed bacterial meningitis (36). However, at least two
sion and impending herniation. of the four symptoms of headache, fever, neck stiffness, and
Increased pressure that exceeds intracranial compliance altered mental status were found in 95% of patients.
causes downward and backward shifting of the cerebrum
and brainstem (31). Minimal degrees of shift are well toler-
ated, but a more extensive shift may cause herniation of the Major Complications of Lumbar Puncture
cingulate gyrus beneath the falx cerebri, herniation of the
uncus of the temporal lobe over the tentorium cerebelli, and Role of Head Computerized Tomography Scan Before
ultimately, herniation of the lower brainstem and cerebellar
Lumbar Puncture and Risk of Brain Herniation
tonsils into the foramen magnum. Herniation of the cingu-
late gyrus is usually asymptomatic. Uncal herniation, however, It has become a routine practice to obtain a CT scan of the
initially produces compression of the third cranial nerve as it head prior to performing a LP in patients with suspected
passes beneath the tentorium; it subsequently causes compres- meningitis. This is done to rule out the possibility of an
sion of the midbrain, with resultant coma. The aqueduct of intracranial mass, hydrocephalus, edema, or any other signs of
Sylvius is often occluded during uncal herniation, and the re- increased ICP that could theoretically place the patient at risk
sultant hydrocephalus increases the mass effect already pres- for cerebral herniation after CSF removal during the LP (37).
ent. Herniation of the cerebellar tonsils through the foramen Herniation of the brain as the consequence of severe cerebral
magnum, with compression of medullary respiratory centers edema or acute hydrocephalus can sometimes occur in acute
and respiratory arrest, is often the terminal event in CNS in- bacterial meningitis and other CNS infections. Clinically, this
fections. Occasionally, space-occupying lesions within the cer- is manifested by altered state of consciousness, abnormalities
ebellum cause upward herniation of posterior fossa contents in pupil reflexes, and decerebrate or decorticate posturing. The
through the tentorial notch (32). Extreme elevation of CSF incidence of herniation after LP even in patients with papill-
pressure may elevate ICP above systemic arterial perfusion edema is approximately 1% (37).
pressure, producing global cerebral and brainstem infarction. In order to clarify the role of a screening CT scan, a pro-
Elevation in CSF pressure, as monitored by ICP monitor- spective study involving 301 adults with suspected meningitis
ing devices, may provide an indication of prognosis in bacterial was done (38). Baseline characteristics that were associated
meningitis and possibly in other CNS infections. Rebaud et al. with an abnormal finding on head CT were age 60 years and
(33) found that CSF pressures were significantly higher and older, immunocompromised host (i.e., HIV/AIDS, immuno-
cerebral perfusion pressure were significantly lower (mean sys- suppressive therapy, or transplantation), a history of CNS
temic arterial pressure minus ICP) in patients who died due to disease, a history of seizure within 1 week before presenta-
meningitis or encephalitis than in those who survived. Goitein tion, and any abnormality on neurologic examination. These
and Tamir (34) found that all pediatric patients with meningitis factors have now been included in the Infectious Diseases
or encephalitis who had a cerebral perfusion pressure more than Society of America guidelines to decide who should undergo
30 mm Hg survived, whereas those with lower pressures died. CT prior to the LP (39). The decision to obtain a brain CT
scan before LP should not result in delay in instituting anti-
biotic therapy because delay can increase mortality (40). It
CEREBROSPINAL FLUID ANALYSIS should be also noted that herniation can occur in patients
with bacterial meningitis who have a normal brain CT scan.
IN CENTRAL NERVOUS SYSTEM The most reliable clinical signs of impending herniation
INFECTIONS include deteriorating level of consciousness, brainstem signs,
and a very recent seizure (41).
Indications for Lumbar Puncture Spinal Hematoma with Cord Compression
LP is essential in the diagnosis of bacterial, viral, or fungal Case reports of LP in patients with severe disorders of blood
meningitis and may provide valuable information in encepha- coagulation, thrombocytopenia, or in patients anticoagulated
litis. LP is also used to diagnose subarachnoid hemorrhage in with heparin or Coumadin have described complications

Scheld_Ch02.indd 8 2/21/14 5:27 PM


Chapter 2: Cerebrospinal Fluid in Central Nervous System Infections 9

with either continued bleeding at the site of puncture or with decreased CSF volume, which would cause cerebral vasodila-
epidural or subdural hematomas that may compress the cauda tation and stretching of pain-sensitive cerebral structures (46).
equina, thereby producing permanent neurologic injury (42). The incidence of the postLP headache is not associated
These complications appear to be rare. In a study of 5,223 LPs with the volume of CSF removed, hydration, the position of
performed, no complications were seen in 941 children with the patient (lying on their side or sitting up), or the opening
leukemia who had severe thrombocytopenia (platelet count pressure (47). Factors that can be associated with a decrease
50) (43). in the incidence of headache are the type and size of needle,
the direction of the bevel during needle placement, the replace-
Introduction of Infection into the Subarachnoid Space ment of the stylet, and possibly the number of LP attempts
(47). Atraumatic needles with a blunt end are recommended by
Inadvertent LP through an area of infection overlying the spi-
the American Academy of Neurology to reduce the incidence
nal canal may result in seeding of the subarachnoid space and
of postLP headaches (48). The blunt end produces a more
meningitis. This is a particular risk in spinal epidural abscess
traumatic opening with tearing and disruption of the collagen
or subdural empyema but may occasionally occur in the set-
fibers that is closed faster by an immunologic reaction and
ting of superficial or deep paraspinal infections. The problem
thus associated with a decrease incidence of headache (49).
can be avoided by entering the subarachnoid space at a level
Additionally, smaller needles have been shown to decrease
well removed from the site of presumed infection. Thus, in
the incidence of the postLP headache (46). The direction
patients with known or suspected focal lumbar infection, spi-
of the bevel should be parallel to the long axis of the spine
nal fluid should be obtained under fluoroscopic guidance by
to decrease the incidence of headache. If the patient is lying
high cervical (C2) or cisternal puncture, whereas the lumbar
on his or her side, the bevel should face up. This way, the
route should be used in patients with suspected cervical or
needle will separate the dural collagen fibers, which also run
upper thoracic infections. Introduction of infection into the
along the long axis of the spine, rather than cutting them (47).
subarachnoid space during LP in uninfected individuals has
Several techniques to treat the postLP headache exist
been reported in 1 out of 50,000 LPs (44). The most com-
including the instillation of a blood patch, dextran, or saline
monly implicated organism is Streptococcus salivarius, and
into the epidural space. A blood patch refers to the injection
this could be potentially prevented by using a mask during the
of 20 to 30 mL of the patients fresh blood into the epidural
procedure (45).
space. It is thought to work by closing the CSF leak by forming
a clot, and it works in about 70% to 98% of patients (47). If a
PostLumbar Puncture Headache blood patch does not work, 20 mL of dextran or saline can be
The most frequent complication of LP is the postLP head- injected into the epidural space to raise the epidural pressure
ache, which can occur in 10% to 60% of patients, more com- and reduce the CSF leak. Oral or intravenous caffeine can be
monly in young women with a lower body mass index (BMI) used because they act as a cerebral vasoconstrictor and blocks
and in pregnancy (46). The diagnosis is a clinical one, and it is adenosine receptors. Surgical closure of the dural gap is the
usually defined as a bilateral headache that worsens while sit- last resort (46).
ting up and improves lying down, develops within 7 days after
a LP, and disappears within 14 days (Fig. 2.3). It is thought Less Common Complications of Lumbar Puncture
that the headache is caused by a CSF leak that decreases ICP.
Cortical Blindness. Downward displacement of the brainstem
This causes headache either by gravitational traction on sensi-
in states of increased ICP may compress the posterior cerebral
tive meningeal vascular coverings as a result of CSF volume
arteries against the edge of the tentorium cerebelli, causing isch-
depletion or by activation of adenosine receptors as a result of
emic infarction of the occipital lobes and cortical blindness (31).
Although this complication of intracranial hypertension is often
accompanied by signs of uncal or tonsillar herniation, compres-
sion of the posterior cerebral arteries may also occur before other
signs of herniation appear. Prognosis for return of vision is poor.

Cervical Spinal Cord Infarction. Rarely, LP in the setting of


bacterial meningitis may be followed within a few hours by
respiratory arrest accompanied by flaccid tetraplegia (50).
A variety of mechanisms, including hypotension and vasculi-
tis, have been postulated as the cause of cervical cord ischemia
in these patients. In some patients, however, it is likely that
displacement of the cerebellar tonsils through the foramen
magnum as the result of greatly elevated ICP compresses the
anterior spinal artery or its penetrating branches, with resul-
tant ischemic infarction of the upper cord (50).

Technique of Lumbar Puncture


The LP was first performed by Quincke in 1891 on children
suffering from headaches in hopes to relieve their symptoms.
Soon after, using CSF as a diagnostic tool became the standard
way for evaluating patients with meningitis (47). The LP is
generally performed with the patient in the lateral recumbent
FIGURE 2.3 Gadolinium-enhanced MRI scan of a patient with intra- position in a fetal position with the knees flexed toward the
cranial hypotension. There is diffuse, symmetric meningeal enhance- chest, and the neck slightly flexed. Only this position allows
ment (arrows). the opening pressure to be measured. The other positions

Scheld_Ch02.indd 9 2/21/14 5:27 PM


10 Part I: Approach to the Patient and Diagnostic Evaluation

include sitting the patient upright on the edge of the bed and TA B L E 2 . 1
bending forward over a bed stand or sitting with the feet sup-
ported and chest resting on the knees. MINIMAL VOLUMES OF CEREBROSPINAL FLUID
The spinal cord typically ends as the conus medullaris at REQUIRED FOR COMMON DIAGNOSTIC TESTSa
the L1 to L2 level in adults, and in children at the L3 to L4
level. The landmarks used are the anterior superior iliac crests, Test Volume of CSF Required
which correlate with the L4 to L5 interspace. The needle may
be inserted between the L3 and L4, L4 and L5, or L5 and Cell count and differential 0.55.0 mLb
S1 interspace (51). Insertion above the L3 level may puncture Glucose and protein 0.5 mLc
the conus medullaris and should not be attempted. Also, the Bacterial culture 35 mLd
needle should not be inserted over a skin infection or abscess Mycobacterial culture; fungal 20 mLe
because this has the potential of inserting bacteria into the culture (includes acid-fast smear
CSF. The performer of the LP should follow a sterile technique and India ink preparation)
including hand washing, gloves, gown, and mask. After the
anterior superior iliac spine is identified, the spinous process Viral culture and/or PCR 12 mL
superior to the interspace is palpated. Prior to inserting the Cryptococcal antigen 0.5 mL
spinal needle, local anesthetic should be utilized, usually 2 to 3 VDRL 0.5 mL
mL of lidocaine without epinephrine deposited subcutaneously Oligoclonal bands 2 mL serumf
and then deeper, allowing 1 to 2 minutes for it to take effect.
The needle should be inserted 1 cm below this and directed in
VDRL, Venereal Disease Research Laboratory.
a horizontal position toward the umbilicus to an approximate a
Volumes required represent minimal quantities of CSF required
depth of 2 cm (51). During the LP, if bone is encountered, the by most hospital laboratories. The clinician should determine the
needle should be withdrawn to the subcutaneous layer and amounts of CSF required by his or her hospital laboratory by each
reinserted at a slightly different angle. The needle is inserted intended test before performing the LP.
b
until a pop is felt indicating penetration of the ligamentum Approximately 0.5 mL will be needed for cell count. Amount of
flavum and presence of the needle in the subarachnoid space. CSF required for differential will vary, depending on whether cyto-
centrifugation is used or material from centrifuged CSF sediment is
The stylet is then removed and CSF obtained. A manometer studied.
to measure the CSF pressure should be attached in all cases c
Blood drawn before initiating the LP should also be submitted with
if possible. If CSF is not obtained, rotate the needle as part of spinal fluid for determination of simultaneous blood glucose level.
d
the dura may be blocking the hole of the needle. If this does As little as 0.5 mL may be submitted for culture if there is great
not work, reinsert the stylet and advance the needle, stopping difficulty obtaining fluid. However, the use of centrifuged sediment
from larger volumes of CSF will improve yield on culture in acute
frequently to withdraw the stylet (51). bacterial meningitis. The use of large volumes of CSF is essential in
more chronic infections.
e
Yield on culture for acid-fast bacilli and fungi is, in general,
Alternative Routes of Obtaining extremely poor unless large volumes of CSF (20 mL or more in
adults) are cultured.
Cerebrospinal Fluid f
Serum (25 mL) drawn before or after the LP should be submitted
for electrophoresis along with CSF.
Cisternal, high cervical (C2), and ventricular approaches may
be used to obtain CSF if a lumbar approach is contraindicated
by infection or is technically impossible (1). Cisternal puncture
was initially described in 1923, but it can cause vascular injuries
(52,53). Spinal puncture at the level of the second cervical ver- Cerebrospinal Fluid Pressure
tebra under fluoroscopic guidance has been suggested as a less
hazardous approach than cisternal puncture, but its actual CSF pressure must be measured in the lateral decubitus
value remains unproven. Ventricular CSF may be of great position with the head of the bed being flat. Opening CSF
diagnostic value if there is a predominantly intraventricular pressure in healthy adults lies between 50 and 195 mm CSF
infection with obstructive hydrocephalus or in the presence of (1). Values higher than 200 mm are abnormal. Normal lumbar
a ventriculoperitoneal shunt (54). CSF pressures in neonates and premature infants are signifi-
cantly lower, with mean values of 100 mm H2O and 95 mm
H2O, respectively (55). CSF pressure is not affected during
pregnancy (56). A CSF baseline pressure of greater than
Routine Studies of Cerebrospinal Fluid 250 mm H2O was associated with higher incidence of neuro-
logic complaints including papilledema, hearing loss, and with
Studies routinely obtained at the time of LP include measure-
mortality in AIDS patients with cryptococcal meningitis (57).
ment of CSF pressure, gross examination of the fluid for tur-
Extreme elevation of CSF pressure may also herald impending
bidity or changes in color, measurement of CSF protein and
brain herniation. Occasionally, CSF pressure may be normal
glucose concentrations, RBC and WBC counts, Gram and/or
or even low in the setting of ongoing tonsillar herniation. The
acid-fast stains of CSF sediment, and Gram stain and bacte-
falsely low readings obtained in this setting are believed to
rial culture of the fluid. Differentiation of bacterial meningitis
reflect occlusion of the CSF space at the foramen magnum by
from viral, mycobacterial, or fungal meningitis on the basis of
the herniated tonsils wedged against the lower brainstem. The
CSF abnormalities is presumptive unless an organism is cul-
possibility of complete spinal block should be kept in mind if
tured or detected by antigen tests or PCR. Amounts of CSF
CSF pressure falls to zero during the procedure.
required by most laboratories for commonly obtained deter-
minations are listed in Table 2.1. Because clinical laborato-
ries differ in the amounts of CSF required for individual tests,
Gross Appearance of the Spinal Fluid
however, the clinician must determine the amounts of CSF re- Once CSF is obtained, it is centrifuged down to give a super-
quired by the hospital laboratory for each intended test before natant. Normal CSF is colorless and clear. Under pathologic
performing the LP. conditions, CSF may become turbid, discolored, or both.

Scheld_Ch02.indd 10 2/21/14 5:27 PM


Chapter 2: Cerebrospinal Fluid in Central Nervous System Infections 11

The CSF may become turbid as a result of entry of cells, bacte- valuable in following the course of illness and response to
ria, or fat; it can be made turbid by as few as 200 WBCs/mm3 treatment (Table 2.2). Improperly handled or counted CSF,
or 400 RBCs/mm3 (1,58). CSF containing RBCs will be grossly however, can be a dangerous source of error. The cell count in
bloody if 6,000 or more RBCs are present per cubic millimeter, CSF tends to decrease over time and may be falsely low if mea-
and it will be cloudy and xanthochromic or pinkish if 400 to sured after 30 to 60 minutes. This decrease in cell count occurs
6,000 cells are present (1). partly because leukocytes and RBCs settle out over time if the
The yellow discoloration of the supernatant is termed xan- tube of CSF is allowed to stand. In addition, however, lysis of
thochromia and is often used to distinguish between a so-called RBCs, polymorphonuclear (PMN) leukocytes, and to a lesser
bloody tap and subarachnoid hemorrhage. Xanthochromia can extent, lymphocytes begins in vitro within 1 to 2 hours of the
be assessed visually or by spectrophotometric methodology by LP and may occasionally occur even more rapidly. WBCs also
scanning the CSF over a range of wavelengths. The discolor- adsorb to the glass or plastic walls of the tube and are not
ation is from degradation products of hemoglobin from lysis of easily dislodged by agitation. Because of these factors, the re-
RBCs. This usually forms 2 to 4 hours after RBCs have entered duction in cell count that occurs over time is only partially
the subarachnoid space (1), which is why some experts suggest reversible if the tube is vigorously agitated before counting.
waiting at least 6 hours after the onset of headache when a Any CSF destined for cell counts should, thus, be handled
subarachnoid bleed is suspected because you may get a false- carefully and expeditiously. Similarly, where serial tubes must
negative result (58). A traumatic tap should clear as the CSF be counted to exclude a traumatic tap, the samples must be
is collected in serial vials but not in all cases of subarachnoid handled in the same manner and counted at the same time by
hemorrhage. Xanthochromia resulting from lysis of RBCs is the same person.
initially a result of oxyhemoglobin. After 12 hours, the pig-
ment represents predominantly bilirubin (1). Visual assessment White Blood Cell Count
of xanthochromia can be deceitful because it may also be seen
Quantification of numbers of cells in CSF can be carried
in the presence of increased amounts of protein, in metastatic
out manually, using a Neubauer counting chamber, but this
melanoma, or as a consequence of systemic hyperbilirubinemia
methodology is labor-intensive, time-consuming, technique-
with a bilirubin level higher than 10 to 15 mg/dL. The most
dependent, and prone to variability. Although electronic cell
appropriate and sensitive way to assess xanthochromia is by
counters are available, they can have poor reproducibility
spectrophotometry of the CSF to detect the hemoglobin break-
especially if the CSF samples have low WBC counts. Novel
down products, oxyhemoglobin and bilirubin.
instruments using flow cell digital imaging have excellent cor-
relation with manual hemacytometer method and should be
Cell Count and Differential the method of choice (59). The accuracy of the cell count is
Enumeration and characterization of cells within spinal fluid open to question unless the specimen is examined immediately
is of crucial value in the diagnosis of CNS infections and is after the LP has been completed. Normally, CSF contains fewer

TA B L E 2 . 2
NORMAL CEREBROSPINAL FLUID VALUES OF IMPORTANCE IN INFECTIOUS
DISEASES OF THE NERVOUS SYSTEM: VALUES IN ADULTS, TERM INFANTS,
AND PREMATURE INFANTS

Parameter Adults Term Infants Premature Infants

Cell count (per cubic millimeter) 5 9a 9a


Percent polymorphonuclear leukocytes 0b 61b 57b
Protein (mg/dl) (lumbar)
Mean 30 90 115
Range 958 20170 65150
Glucose (mg/dl)
Meanc 62 52 50
Rangec 4580 34119 2463
CSF:blood glucose ratio
Mean 0.60 0.81 0.74
Range 0.50.8c 0.442.48 0.551.55

a
Cell counts in term and premature infants represent mean values. The range of cell counts found in nor-
mal neonates is 032 cells/mm3 and in premature infants is 029, with 2 standard deviations encompassing
a range of 022.4 cells/mm3 in term and 024.4 cells/mm3 in premature infants. By 1 month of age, normal
CSF contains 20 cells/mm3 (2).
b
Rare polymorphonuclear leukocytes may be seen in cytocentrifuged samples of CSF from normal adults.
This is not necessarily abnormal if the CSF leukocyte count is 4 cells/mm3 or less and if protein and glucose
levels are normal.
c
Assumes a blood glucose level of 70120 mg/dl. At high blood glucose levels (700 mg/dl), normal lower
limit of CSF: blood glucose ratios may approach 0.4 (see text).
Adapted from Fishman RA. Cerebrospinal Fluid in Diseases of the Nervous System. 2nd ed. Philadelphia:
WB Saunders; 1992, with permission.

Scheld_Ch02.indd 11 2/21/14 5:27 PM


12 Part I: Approach to the Patient and Diagnostic Evaluation

than five cells per cubic millimeter (Table 2.2). Most of these TA B L E 2 . 3
cells are small lymphocytes (nuclear diameter about 6 to 7 m)
with scant cytoplasm. Larger numbers of PMN leukocytes are CONDITIONS ASSOCIATED WITH CEREBROSPINAL
abnormal in uncentrifuged CSF. C. neoformans is similar in FLUID EOSINOPHILIA
size to a small CSF lymphocyte, and nonbudding forms may
be mistaken for these cells in the counting chamber, though not Parasitic infestations
in stained cytocentrifuged or otherwise concentrated samples. Taenia solium (cysticercosis)
Neonatal CSF usually contains 8 to 9 WBCs/mm3, and up to Angiostrongylus cantonensis
32 WBCs/mm3 has been reported in the absence of disease (60)
Gnathostoma spinigerum
(Table 2.2).
The WBC count is usually between 1,000 and 5,000/mm3 Trichinella spiralis
in untreated bacterial meningitis (60), and more than 90% Ascaris lumbricoides
of patients with bacterial meningitis will have a WBC count Toxoplasma gondii
greater than 100 cells/mm3 (61). The most common three types
of viral meningitis in the United States (enterovirus, West Nile Toxocara cati
virus, and herpes simplex virus) have a median WBC count Toxocara canis
between approximately 100 and 250 cells/mm3 (25). Other infectious agents or conditions
Mycobacterium tuberculosis
Differential White Blood Cell Count Treponema pallidum
A differential count of CSF leukocytes may be obtained fol- Mycoplasma pneumoniae
lowing concentration of CSF through a Millipore filter, cen- Rocky Mountain spotted fever
trifugation of a volume (usually 5 mL) of CSF, concentration Subacute sclerosing panencephalitis
by sedimentation, or cytocentrifugation. The number of neu- Lymphocytic choriomeningitis virus
trophils is increased in various conditions. In adults with bac-
terial meningitis, neutrophils make up an average of 86.4% Fungal meningitides
of cells counted, with neutrophils making up an average of Central nervous system disorders of noninfectious or
34.2% of cells counted in aseptic meningitis (1,62). In the unknown origin
early stages of meningitis, this distinction between bacterial Idiopathic eosinophilic meningitis
and viral etiologies may not be clear because a neutrophilic Granulomatous meningitis
pleocytosis (50% neutrophils) may accompany early viral
meningitis or encephalitis (63). Up to two thirds of enterovi- Malignant lymphoma
ral meningitis cases initially have a neutrophilic predominance Hodgkin disease
(64). Within 12 to 24 hours, there is usually a shift from a Leukemia
neutrophilic predominance to a lymphocytic predominance,
Multiple sclerosis
which is why some may suggest a repeat LP if the first LP
was nonspecific (47). A lymphocytic pleocytosis is typically Subarachnoid hemorrhage
observed in patients with viral meningitis, M. tuberculosis, Obstructive hydrocephalus with shunt
Borrelia burgdorferi, Treponema pallidum, or C. neoformans, Reaction to intrathecal antibiotics
as well as in neoplastic and drug-induced meningitis (63). In
AIDS-associated cryptococcal meningitis, CSF pleocytosis Data from references 1, 6165, with permission.
may be absent, a finding that is associated with a worse
prognosis. Only up to 30% of patients with AIDS-associated
cryptococcal meningitis have a CSF WBC greater than 20 with
a lymphocytic predominance (57). In tuberculous meningitis,
the range of CSF pleocytosis is more commonly between 50 of RBCs are present. In such cases, one should compare num-
and 300 cells/mm3 with a lymphocytic predominance. Plasma bers of RBCs present in CSF obtained at the beginning of
cells and eosinophils should not be present in normal CSF (60). the LP with numbers present in CSF obtained at the end of
Increased numbers of B cells can be seen in neurosyphilis and the procedure (e.g., one should count cells from tubes 1 or
could represent another diagnostic option (65). Eosinophilic 2 and then from tube 4). The presence of xanthochromia in
meningitis can be caused by several parasitic infections, the samples centrifuged immediately after obtaining CSF argues
most common being angiostrongyliasis, gnathostomiasis, against a traumatic tap, although it must be kept in mind that
toxocariasis, cysticercosis, schistosomiasis, baylisascariasis, lysis of RBCs in vitro in CSF obtained during a traumatic tap
and paragonimiasis (66) (Table 2.3). In addition, however, will produce xanthochromia if the specimen is allowed to sit.
CSF eosinophilia has been reported in a wide variety of other Crenation of RBCs may occur in vitro and has no diagnos-
infectious and noninfectious conditions (Table 2.3), so detec- tic significance (67). Blood entering CSF during spontaneous
tion of eosinophils within the CSF is not pathognomonic of subarachnoid hemorrhage or as the result of a traumatic tap
parasitic infestation (66). contains WBCs and RBCs, and thus, the CSF leukocyte count
will increase. Numbers of WBCs relative to those of RBCs in
CSF after a traumatic tap should be consistent with the leuko-
Red Blood Cells
cyte count of the peripheral blood, and the differential count
The presence of RBCs in CSF may result from a traumatic LP of CSF will be the same. In contrast, actual subarachnoid
or may indicate subarachnoid or parenchymal hemorrhage. hemorrhage often produces pleocytosis and alteration in the
Grossly bloody fluid that clears visibly as CSF is collected sug- differential count. A traumatic tap in the setting of CNS infec-
gests a traumatic tap. Differentiation between a traumatic LP tion will increase the numbers of WBCs already present by an
and subarachnoid blood as the result of intracranial or intra- amount that can be calculated by comparing the ratio of RBCs
spinal pathology becomes more difficult if only small numbers to WBCs in CSF with that seen in peripheral blood.

Scheld_Ch02.indd 12 2/21/14 5:27 PM


Chapter 2: Cerebrospinal Fluid in Central Nervous System Infections 13

Cerebrospinal Fluid Glucose greater than 0.3 was felt to exclude most (but not all) cases
of bacterial meningitis. Additionally, Spanos, Harrell, and
Most glucose present in CSF (Table 2.2) moves across the Durack (81) performed a retrospective study of 422 patients
choroid plexus and across ventricular and subarachnoid with acute bacterial or viral meningitis and found that CSF
capillaries by facilitated transport. A smaller amount of glu- glucose levels less than 18 mg/dL (1.9 mmol/L) and a CSF/
cose enters the CSF by simple diffusion. Glucose is removed blood glucose ratio less than 0.23 were predictors of bacterial
from CSF through utilization by cells lining the ventricles and meningitis. Furthermore, hypoglycorrhachia is associated with
subarachnoid space and by transport across capillaries and an adverse clinical outcome in patients with meningitis and a
arachnoid villi. Entry of glucose occurs over time, and more negative Gram stain (82).
than 2 to 4 hours is required before serum and CSF glucose
levels reach equilibrium (1). In the absence of infection or Cerebrospinal Fluid Protein
other pathologic conditions, CSF glucose levels are a predict-
able reflection of blood glucose, and the ratio of CSF to blood Protein is largely excluded from CSF by the bloodCSF bar-
glucose concentrations is approximately 0.6. The CSF glucose rier and, under normal conditions, reaches CSF by pinocytotic
level, equilibrated with a normal blood glucose level of 70 to transport across capillary endothelia (83). Total CSF protein
120 mg/dL, thus ranges between 45 and 80 mg/dL (Table 2.2). concentration in lumbar CSF of a healthy adult (Table 2.2) is
Levels of glucose in ventricular fluid are 6 to 18 mg/dL higher less than 45 mg, and the CSF/serum ratio of albumin is 1:200
than those in lumbar fluid (1,68). (1,13). Mean values of lumbar CSF protein in healthy children
CNS infections may alter glucose transport across the and adults have ranged from 23 to 38 mg/dL, and the extreme
bloodCSF barrier, resulting in a low CSF glucose level, upper and lower concentrations have been 58 and 9 mg,
termed hypoglycorrhachia (1). Further reduction in CSF glu- respectively (1). The CSF protein level in premature and full-
cose levels may result from glucose consumption by WBCs term neonates may range between 20 and 170 mg/dL, with
and organisms (1). Reduction of CSF glucose relative to blood a mean of 90 mg/dL (58) (Table 2.2). Protein concentrations
glucose is characteristic of meningitis caused by bacteria, in cisternal and lumbar CSF are lower, ranging from 13 to
mycobacteria, or fungi (69,70). The CSF glucose level is usu- 30 mg/dL (1). Elevation of protein concentration in the set-
ally normal during viral infections, but low CSF glucose levels ting of CNS infections results from disruption of tight junc-
are occasionally observed in meningoencephalitis caused by tions between endothelial cells of venules and, to a lesser
mumps, enteroviruses, lymphocytic choriomeningitis, herpes extent, other small meningeal or parenchymal vessels (83).
simplex, and herpes zoster viruses (25,71). Low CSF glucose Elevation of CSF protein level to more than 150 mg/dL may
values have also been described in CNS complications of cause the CSF to be xanthochromic. Extreme elevation of pro-
Mycoplasma pneumoniae infection, carcinomatous meningi- tein (to 1.5 g/dL) may cause formation of a weblike surface
tis, CNS sarcoidosis, and subarachnoid hemorrhage (7275). pellicle or an actual clot, as may high levels of fibrinogen (1).
During recovery from meningitis, CSF glucose levels tend Levels of CSF protein may be falsely elevated by deteriorating
to return toward normal more rapidly than cell counts and RBCs following subarachnoid hemorrhage or traumatic LP.
protein levels, making CSF glucose levels an important param- The amount of increase is roughly 1 mg/dL per 1,000 RBCs.
eter to follow in assessing response to therapy (76,77). Accurate assessment of the contribution to total CSF protein
Both reduction in CSF glucose values and altered ratios made by RBCs requires that the cell count and protein deter-
of CSF to blood glucose levels are used as indicators of mination be carried out on the same tube of CSF.
infection. However, the literature contains a variety of recom- Changes in the concentration of protein in CSF are the most
mendations about the point at which CSF glucose should be common and least specific of CSF alterations in disease and
considered abnormally low (78); this is partly because of the are seen in a wide variety of infectious and noninfectious neu-
prolonged interval over which CSF glucose equilibrates with rologic conditions. Thus, an elevated CSF protein level, taken
serum glucose. In general, a CSF/blood glucose ratio less than alone, has little specific value in the diagnosis of CNS infections.
0.5 should be considered abnormal. In premature and full- Elevation of CSF protein to levels more than 100 mg/dL, par-
term infants, however, the normal CSF/blood glucose ratio is ticularly if obtained on serial LPs, argues against viral infec-
0.74 to 0.96, and a ratio of 0.6 is usually considered abnor- tion, however, and Spanos, Harrell, and Durack (81) have
mal (79). In severe hyperglycemia, transport of glucose into demonstrated that elevation of protein to a level of 220 mg/dL
CSF may lag, and at a blood sugar level of 700 mg/dL, the (2.2 g/L) suggests bacterial meningitis. The CSF protein levels
CSF/blood glucose ratio may approach 0.4. For this reason, return to normal more slowly than glucose levels and cell count
a ratio of 0.3 has been suggested as abnormal in diabetics during recovery from meningitis and may remain abnormal for
(80). Silver and Todd (78) addressed the problem of diag- months after parenchymal infections. Although elevation of
nostically significant hypoglycorrhachia in a study of 181 CSF protein is common in CNS infections, normal protein val-
pediatric patients with CSF glucose levels less than 50 mg/dL ues are occasionally seen in all types of CNS infections, includ-
or a CSF/blood glucose ratio less than 50%. Patients ranged ing bacterial meningitis. In children with bacterial meningitis,
in age from younger than 1 week to 14 years, with an average antibiotic administration more than 12 hours before the LP
age of 1 years. Their series included patients with bacterial is associated with lower CSF protein and higher CSF glucose
meningitis, aseptic meningitis, subarachnoid hemorrhage, and concentrations (84).
CNS carcinomatosis but did not include patients with tuber-
culous or fungal meningitis. Blood for glucose analysis was
Cerebrospinal Fluid Immunoglobulins
obtained 1 to 114 minutes before the LP (average interval, Immunoglobulins are almost totally excluded from normal
30 minutes). Of 35 patients with bacterial meningitis in this CSF. The blood/CSF ratio of immunoglobulin G (IgG) in
series, 27 (77%) had CSF glucose levels of 20 mg/dL or less, normal CSF is usually in the range of 500:1. Immunoglobulin
whereas CSF glucose levels of 20 mg/dL or less were found in M (IgM) is essentially absent from CSF. Studies with radio-
only 10 (7%) of 146 patients with other conditions. A CSF iodinated IgG have demonstrated that CSF IgG in healthy
glucose level less than 20 mg/dL or a CSF/blood glucose ratio individuals is derived entirely from serum, requiring 3 to
less than 0.30 was highly correlated with bacterial meningi- 6 days to reach equilibrium (1). Immunoglobulins enter CSF
tis, whereas an absolute CSF glucose value between 20 and less readily than albumin; and in health, immunoglobulin/
50 mg/dL was nonspecific; also, a CSF/serum glucose ratio albumin ratios in CSF are reduced relative to those in serum.

Scheld_Ch02.indd 13 2/21/14 5:27 PM


14 Part I: Approach to the Patient and Diagnostic Evaluation

Elevation in CSF immunoglobulins may follow disruption glucose and protein levels if administered within 12 hours of
of the BBB, allowing passage of immunoglobulins across the LP (84). The sensitivity of the Gram stain in children and
capillary endothelium, or may result from local antibody adults with pneumococcal meningitis ranges between 69% and
synthesis within the brain. Increased levels of CSF IgG per se 95% and in meningococcal meningitis between 30% and 89%.
have little diagnostic value in CNS infections. Detection of Blood cultures will be positive in 50% to 80% of patients, and
oligoclonal IgG bands unique to CSF and not seen in serum the CSF cultures will be positive between 80% and 90% of
on gel electrophoresis provides strong evidence for an ongoing cases (86). In general, the sensitivity of the Gram stain ranges
immune response within the brain and is part of the diagnostic from 50% to 90%; however, the specificity approaches 100%
criteria for multiple sclerosis (85). (61,86). In one study, only 22 (4%) out of 567 patients with
community-acquired meningitis and a negative Gram stain had
culture-proven bacterial meningitis (82).
Microscopic Methods for Detecting
Infectious Organisms
Partially Treated Bacterial Meningitis
Gram Stain The diagnosis of patients that present with possible bacterial
Gram stain is of crucial value in providing rapid identification meningitis who have received antibiotics remain a challenge
of the offending organism in bacterial meningitis, and it is fast, to clinicians. The acridine orange stain is a fluorochrome
inexpensive, and fairly reliable (Fig. 2.4). It is usually the single stain that has been shown to improve detection of bacteria
most important piece of information the clinician uses to guide in CSF specimens, especially in patients who have partially
initial antibiotic therapy and should be an invariable part of treated bacterial meningitis (87). A more recent approach is
the CSF evaluation. Diagnostic accuracy of a properly prepared detection of Streptococcus pneumoniae C-polysaccharide,
Gram stain is a function of the number of organisms present, which is found in the cell wall and is common to all serotypes,
the type of meningeal pathogen, and by the receipt of prior in CSF by using rapid immunochromatographic membrane
antibiotic therapy (61). In one large study of bacterial men- assays (88,89). Two large, multicenter studies have shown
ingitis in children, prior antibiotic exposure did not alter the a sensitivity and specificity of 99% detecting S. pneumoniae
sensitivity of the Gram stain but decreased the sensitivity of even in patients who have been pretreated with antibiotics that
the blood and CSF cultures by 18% and also altered the CSF have negative CSF cultures (90,91).

FIGURE 2.4 Gram stains of CSF from patients with bacterial men-
ingitis. A: Streptococcus pneumoniae. B: Neisseria meningitides.
C: Gram-negative meningitis.

Scheld_Ch02.indd 14 2/21/14 5:27 PM


Chapter 2: Cerebrospinal Fluid in Central Nervous System Infections 15

Cerebrospinal Fluid Bacterial Culture


Choice of culture media, methods of handling, and lengths of
time over which cultures are to be maintained are thoroughly
discussed in standard reviews and texts (92,93). The CSF
should be submitted to the laboratory immediately after the LP
and should be placed in culture promptly to avoid loss of fas-
tidious organisms such as Haemophilus influenzae, Neisseria
meningitidis, or anaerobes. CSF cultured for bacteria should,
at a minimum, be plated on a 5% sheep blood agar, chocolate
agar, and inoculated into an enrichment broth (93). A mini-
mum of 2 mL (ideally 5 mL or more) should be submitted for
Gram stain and bacterial culture.

Cerebrospinal Fluid Acid-Fast Bacilli Stains


and Cultures
A positive acid-fast stain for detection of M. tuberculosis is FIGURE 2.5 India ink preparation of CSF from a patient with
highly suggestive of tuberculous meningitis, but positive results cryptococcal meningitis. The capsule of a cryptococcal organism is
occur in only 10% (94). The sensitivity of the acid-fast stain clearly outlined by ink particles.
depends greatly on the skill and persistence of the examiner and
the amount of fluid concentrated. In general, collecting four
serial samples and spinning of large volumes (20 mL) of CSF for
30 minutes enhances the rate of detection by smear microscopy,
but it is impractical (95). Isolating mycobacteria in culture is diffi- Viral Culture
cult with detection rates for M. tuberculosis between 10.2% and
55.8% for conventional Lowenstein-Jensen medium and from Isolation of viral agents by tissue culture methods has been the
4.3% to 48.9% for the automated commercial system BACTEC traditional means of diagnosis in cases of suspected viral men-
Mycobacteria Growth Indicator Tube (MGIT) 960 (96). ingitis or encephalitis. Newer methods of virus isolation have
improved diagnostic yield; these include the incorporation of
multiple tissue culture cell lines and a combination of culture
Microscopic Detection of Anaplasma, Fungi, and staining procedures (shell vial assay for early antigen
and Protozoa in Cerebrospinal Fluid detection, enzyme immunoassays, and immunofluorescence
staining). Enteroviruses can be isolated in 43% to 77% of
In a few cases, intracellular morulae have been detected in CSF of patients, depending on the predominant viral serotype in a
patients with meningitis due to Anaplasma infection (97). Fungi, particular community. In approximately half of these cases,
including C. neoformans, Blastomyces dermatitidis, C. immitis, virus will be isolated by day 3 and in more than 80% by day 7
and Candida albicans, may occasionally be detected on Gram or (104). Mumps virus and lymphocytic choriomeningitis virus,
silver stains of concentrated CSF (98). In many cases of fungal the agents of western and eastern equine encephalitides, may
meningitis, however, organisms are too few to be readily detect- also be recovered from CSF. Herpes simplex virus (HSV) types
able, and negative Gram or silver stains of CSF sediment in no 1 and 2 can be isolated from cases of meningitis but are rarely
way excludes the possibility of fungal infection. India ink prepa- recovered from CSF in cases of encephalitis. Varicella-zoster
rations, in which CSF sediment from 3 to 5 mL of CSF is mixed virus; cytomegalovirus; and California, St. Louis, and Japanese
with a drop of India ink, provide a useful means of outlining the encephalitis viruses are rarely recovered (105).
capsule of C. neoformans (Fig. 2.5). Sensitivity of the India ink
preparation is about 60% in patients who are not infected with
acquired immunodeficiency syndrome (AIDS) and more than
75% in patients with AIDS (98). Cryptococcal antigen detection
ADJUNCTIVE AND
has replaced India ink preparations in most laboratories because MOLECULAR STUDIES OF
of its high sensitivity and specificity (99).
Wet mount preparations may be used to identify motile
CEREBROSPINAL FLUID IN
trophozoites in the CSF of patients with primary amebic THE DIAGNOSIS OF CENTRAL
meningoencephalitis (100). Search for motile organisms in NERVOUS SYSTEM INFECTION
wet mounts may be made more reliable by the use of phase-
contrast microscopy. The need for rapidly available accurate diagnostic informa-
C. neoformans is cultured from the CSF in approximately tion in CNS infections, the poor sensitivity of microscopic
72% of patients on the first LP and in more than 90% on examination of CSF sediment, and the delays inherent in
multiple attempts (101). Frequency of recovery of C. albicans obtaining results of CSF culture have led to the development
from CSF is also high (102). Isolation of other organisms such of a wide variety of rapid diagnostic tests for CNS infections.
as Histoplasma capsulatum or Brucella species often proves At present, PCR methods have largely replaced tissue culture
difficult (103). methods for enteroviruses, Herpetoviridae (herpes simplex,
In most bacterial and fungal infections, extraneural sites herpes zoster, cytomegalovirus, Epstein-Barr virus), JC virus,
of possible infection should also be cultured. Depending on and West Nile virus. West Nile virus meningoencephalitis is
the organism being sought, these sites may include blood, diagnosed largely by serology because the virus is only rarely
urine, paranasal sinuses, ears, skin, oropharynx, sputum, bone isolated by tissue culture methods at the time patients present
marrow, prostate, or abscess material. with neurologic symptoms (106).

Scheld_Ch02.indd 15 2/21/14 5:27 PM


16 Part I: Approach to the Patient and Diagnostic Evaluation

S. pneumoniae, E. coli K1, and Streptococcus agalactiae. In


Bacterial Infections general, these tests find little application; antigen-detection tests
are less sensitive than bacterial culture, offer no advantages over
Lactic Acid the Gram stain, and their diagnostic reliability and usefulness
Elevation of lactic acid levels in CSF occurs more frequently may vary from institution to institution (114,115). Situations
in bacterial than in viral meningitis. A CSF lactate cutoff value in which antigen-detection tests are not likely to be helpful
of more than 3.5 to 4.2 mmol/L provides supportive evidence include cases in which the Gram stain is positive, in which the
for a bacterial infection in untreated patients. Two large CSF WBC count and chemistries are within normal limits, and
metaanalyses have concluded that the determination of CSF in which the CNS infection is hospital acquired (115).
lactate level is better than the CSF WBC, glucose, or protein
in differentiating bacterial meningitis from aseptic meningitis Polymerase Chain Reaction
(sensitivity of 93% and 97% and specificity of 96% and 94%, Molecular techniques such as polymerase chain reaction (PCR)
respectively) (107,108). are well suited for the diagnosis of CNS infections because the
presence of microorganisms is highly suggestive of infection.
C-Reactive Protein PCR has emerged as a novel technique for identifying viral,
C-reactive protein (CRP) is an acute phase reactant released bacterial, and mycobacterial causes of meningitis (116119).
from the liver in response to an inflammatory reaction, such as Although CSF makes an ideal source to examine by PCR because
meningitis. CRP is released within 6 hours of insult and peaks it is sterile and without contaminants, the presence of inhibitors,
after 36 hours. One of the functions of CRP is to bind to phos- assay contamination, and experimental conditions may some-
pholipid components of damaged cells or bacteria resulting in times alter its diagnostic value (118). There are a variety of PCR
activation of the classical complement pathway (109). Both methods used today including multiplex and nested PCR.
serum and CSF CRP have been studied as potentially useful Multiplex PCR has the advantage of potentially detecting more
tools for discriminating bacterial meningitis from aseptic than one organism in the same PCR reaction. This is done by
or viral meningitis. A large retrospective study in children using two or more primer pairs, each specific for a single agent.
showed a sensitivity of 93% and a specificity of 100% with In nested PCR, products from the first amplification are reampli-
a CRP greater than 40 mg/L to detect bacterial meningitis fied from a second set of primers that is nested between the first
(110). Furthermore, a metaanalysis showed that a CRP level set. This essentially overcomes nonspecific amplification (i.e.,
greater than 20mg/L was conveyed significantly higher odds of assay contaminations) and increases the sensitivity of detection.
bacterial meningitis (odds ratio [OR] 9.9 [4.8 to 20.8]) (111).
Current literature supports the assertion that CRP may be Bacterial Polymerase Chain Reaction. The use of PCR in
useful, but it should be used with caution as the sole criterion bacterial meningitis may serve a role in patients previously
in the differentiation of bacterial versus viral meningitis (61). treated with antibiotics or in the detection of difficult to
culture organisms such as Mycoplasma or Brucella. However,
Procalcitonin molecular tests are not routinely available, and Gram stain
Procalcitonin (PCT) is a calcitonin propeptide synthesized and culture of the CSF is still the gold standard for diagnosis.
by C cells of the thyroid gland and released from leukocytes Broad-range PCR amplifies the 16S ribosomal RNA (rRNA)
of the peripheral blood (112). It has been used as a marker gene that is present in all bacterial species. Broad-range PCR
of severe inflammation such as those caused by bacterial and organism-specific PCR have been used in detecting menin-
infections, pancreatitis, burns, or trauma (113). In a prospec- geal pathogens with sensitivities between 89% and 100% and
tive multicenter trial of 151 patients with a negative Gram specificities of 95% to 100% (61). In a study of 409 patients
stain, 18 had confirmed bacterial meningitis; a serum PCT with bacterial meningitis in Burkina Faso, PCR was able to
greater than 0.5 ng/mL (sensitivity 87%, specificity 100%, make the diagnosis in a third of patients who had negative
positive predictive value 1.0, negative predictive value 0.99) bacterial cultures (120). The availability, expense, and time to
had better diagnostic accuracy than serum CRP, CSF leukocyte run these test may prove difficult, and positive results ideally
count, CSF/blood glucose ratio, CSF protein, and the physi- should be confirmed by species-specific PCR. Furthermore,
cians assessment (109). A metaanalysis showed that a procal- contamination of PCR specimens may cause false-positive
citonin level greater than 0.5 g/L was predictive of bacterial results, which can be encountered by improper handling or
meningitis with an OR 434 (95% confidence interval [CI] contaminated work equipment from previous PCR reactions.
57 to 1,000) (111). Procalcitonin in the CSF may also be
elevated in patients with probable Alzheimer disease, vascular Viral Polymerase Chain Reaction. The use of PCR is the
dementia, dementia with Lewy bodies, frontotemporal demen- preferred method of diagnosing patients with suspected
tia, and encephalitis (113). viral encephalitis such as those caused by HSV, enterovirus,
varicella-zoster virus, cytomegalovirus, or Epstein-Barr virus
Additional Biomarkers (117). This is very important in cases of suspected HSV
encephalitis that can cause significant morbidity and mortality
Bacterial meningitis results in systemic and intrathecal inflamma- if untreated. There are over 100 known viruses that may infect
tory reactions that may lead to significant morbidity and mortal- the CNS although only a limited number of tests are available
ity. Several inflammatory markers (interleukin-1 [IL-1], IL-6 for confirmation (117). The decision to perform molecular
and -12, TNF-, soluble triggering receptor expressed on myeloid tests is physician-driven and is often based on the clinical
cells-1 (sTREM1), cortisol, heparin-binding protein, and comple- presentation. In a study of 760 adult patients presenting with
ment factor 3 and complement B) have all been evaluated in small meningitis and a negative Gram stain, only 44% had PCR in
studies with adequate diagnostic accuracy (61). the CSF performed for a viral pathogen (116).
Detection of Bacterial Antigens Herpes simplex virus. Herpes simplex encephalitis (HSE)
Antigen-detection tests using latex agglutination assays are is the most common cause of sporadic encephalitis in the
available to rapidly detect meningitis caused by H. influenzae United States, and it is the most common cause of severe viral
type b, N. meningitidis serogroups A, B, C, Y, and W135, encephalitis (121). In immunocompetent adults, 90% of HSE

Scheld_Ch02.indd 16 2/21/14 5:27 PM


Chapter 2: Cerebrospinal Fluid in Central Nervous System Infections 17

is caused by HSV-1, with the remaining 10% caused by HSV- the Centers for Disease Control and Prevention (CDC) recom-
2. In AIDS patients, the rate of HSV-2 may be higher (122). mends using a two-step process for testing serum from patients
Empirical antiviral therapy and prompt identification are of suspected of having Lyme disease (130). Step one involves
crucial importance because of the high morbidity and mor- screening with a sensitive assay such as enzyme-linked immu-
tality associated with HSE. Early initiation of intravenous nosorbent assay (ELISA) or immunofluorescence assay. Those
acyclovir has been shown to reduce mortality in patients with samples that are negative by such an assay are not tested further.
HSE (123). The availability of PCR has now become the diag- All equivocal or positive results are subsequently confirmed by
nostic modality of choice for diagnosing HSE (117,121,123). immunoblotting (Western blot) (131). Standardized criteria
Numerous studies have shown sensitivities greater than 90%, for interpretation of both IgM and IgG are outlined elsewhere
with specificities near 100% (117). Viral cultures are not rou- (131). Antibody to B. burgdorferi may be absent early in the
tinely recommended because they are positive in less than 5% course of infection, and seronegative Lyme disease, diagnosed
of adults. Prior to PCR, the gold standard for diagnosing HSE by T-cell proliferation to Lyme disease, has been reported
was brain biopsy (121). A negative PCR for HSV does not rule (132). False-positive test results for Lyme disease may be seen
out HSV encephalitis but rather makes it less likely (117). in patients with infectious mononucleosis, positive serology for
syphilis, and autoimmune conditions.
Enterovirus. Enteroviruses are one of the most common causes In the acute form of neuroborreliosis, there is usually
of aseptic meningitis in children and in adults (71). Enterovirus pronounced synthesis of IgM antibody production (133). IgG
PCR in the CSF has improved the detection rates compared to and IgA synthesis is seen in the chronic forms of disease (133).
viral cultures, and results can be available within 2 hours. The Detection of intrathecal antibody production is considered
Gene Xpert Enterovirus PCR had a sensitivity of 97.1% (95% the most specific test for neuroborreliosis (134). However,
CI, 84.7% to 99.9%) and a specificity of 100% (95% CI, 94.6% not all patients develop CSF antibodies. The CSF antibody
to 100%) for the diagnosis of enteroviral meningitis (124). production in subtle CNS disease is inconsistent and may be
A rapid diagnosis of enteroviral meningitis could impact care by lacking in patients with only peripheral nerve involvement
avoiding hospitalization or empirical antibiotic therapy (71). (135). Detection of CSF antibody is not essential for diagnosis
(134,135). Accuracy and reliability of tests vary considerably
between laboratories; therefore, positive values reported by
Mycobacterial Polymerase Chain Reaction laboratories unfamiliar to the physician must be approached
with caution and, if necessary, confirmed.
The diagnosis of tuberculous meningitis (TM) can be difficult There is currently not a validated PCR method commercially
because culture sensitivities are low, and the organism may available (136). There are several parameters that can have sig-
take up to 6 weeks to grow (96). PCR has received interest in nificant effects on the performance of the PCR such as sample
hopes it may serve as a rapid, sensitive, and specific test for type and volume, extraction method (nested PCR, PCR followed
TM. One of the more frequently used M. tuberculosis PCR by hybridization, real-time PCR), target, primers, template DNA,
targets is IS61100 (125). Investigators prospectively studied and PCR chemistry (136). A metaanalysis derived from published
677 CSF samples in patients with clinically suspected TM PCR results irrespective of methods or targets from patients with
(125). All culture-positive samples (n 136) were positive all stages of Lyme neuroborreliosis demonstrated an overall sensi-
(100%) by the PCR assay. In those patients with clinically sus- tivity of 19% and a specificity of 100% (137). Other reports have
pected (culture negative) TM, the assay was positive in 70% indicated that PCR is no more sensitive as a diagnostic tool than
(n 541). Not all studies have shown as good of results, with the measurement of intrathecal antibody production and overall
some sensitivities being reported as low as 33% (126) and as is less useful (138139). PCR should not be considered a stand
high as 87% (127). Specificities also have a wide range from alone test, and a negative result does not rule out neuroborrelio-
88% to 100% (94). This discrepancy could be due to the sis. At present, molecular assays may, at most, have a limited role
different types of measuring methods with the use of different as adjunctive tests in patients who are seronegative and who have
targets used in the laboratories (127). a high likelihood of infection (e.g., as in the case of the immuno-
deficient patient). Elevated levels of CSF CXCL13 (C-X-C motif
Measurement of Adenosine Deaminase Levels chemokine 13) have been described in Lyme neuroborreliosis and
in Cerebrospinal Fluid could serve as a diagnostic marker. Furthermore, CSF CXCL13
decreased with intravenous ceftriaxone and oral doxycycline
Adenosine deaminase is an enzyme that is widely distributed
(140). As research tools, molecular assays may provide insight
in human tissues and is present in high concentrations in
into pathogenesis and clinical course when used prospectively
lymphocytes. Elevation of CSF adenosine deaminase levels
during the course of illness.
may occur in a variety of neurologic disorders, including
bacterial meningitis, brain abscess, neurobrucellosis, cryp-
tococcal meningitis, and CNS lymphoma. Elevated levels of Fungal and Other Infections
lymphocyte adenosine deaminase are frequently present in the
CSF of patients with TM, and measurement of this enzyme has
Fungal Infections
been used to provide presumptive evidence of M. tuberculosis
infection and to evaluate response to treatment (128,129). The Detection of cryptococcal antigen in spinal fluid is the most
test, though both sensitive and useful, is not specific because practical diagnostic test for cryptococcal meningitis. The
it detects a component of host response and does not detect a test has a high degree of specificity and is positive in 83% to
structural component of the organism itself. 98% of patients (141). Detection of cryptococcal antigen has
replaced India ink stains. Despite its sensitivity and specificity,
however, assays for cryptococcal antigen may occasionally
Lyme Disease give false-negative results in both immunocompetent and
immunocompromised patients (142). Screening for cryptococ-
The overall sensitivity and specificity of tests for diagnosis of cal meningitis as a point of care test by using the cryptococcal
Lyme disease are still being determined, as is the accuracy of antigen in urine or plasma could help identify patients at high
tests used to diagnose CNS involvement. In the United States, risk sooner and possibly decrease mortality (143,144).

Scheld_Ch02.indd 17 2/21/14 5:27 PM


18 Part I: Approach to the Patient and Diagnostic Evaluation

Complement-fixing antibodies have been reported in CSF is less sensitive (117). Comparison of serum and CSF titers of
in up to 95% of cases of meningitis caused by C. immitis IgG and IgM antiviral antibodies has been proposed as a diag-
(145), although yield of detection has not been that high in nostic test in encephalitis caused by HSV and other agents, but
all series (146). Diagnosis of Histoplasma meningitis can be the test, which is dependent on intrathecal antibody synthesis,
extremely difficult, especially the chronic form of the disease, is of limited value at the time of presentation, and intrathecal
which may occur in the absence of other manifestations of antibody may become detectable only as virus is cleared from
disseminated infection (147,148). Detection of CSF antibodies the CSF compartment (161). Most patients presenting with
may be useful, but false-positive results have been reported West Nile virus infection already have CSF IgM antibodies to
in patients with fungal meningitis caused by other organisms the virus, making this the diagnostic method of choice (162).
and by diffusion of serum antibodies to H. capsulatum into Compared to viral culture (overall sensitivity, 14% to 24%),
the CSF during other inflammatory conditions of the meninges the sensitivity of PCR ranges from 75% to 100% depending
(149). Histoplasma polysaccharide antigen detection can be on the virus (163175). Molecular detection of viral nucleic
detected in CSF, serum, or in urine. Small studies of less than acid sequences in CSF has not only improved diagnosis but
20 patients have documented sensitivities between 38% and also has largely replaced invasive methods such as brain
71% (149,150). biopsy, has shortened time to specific diagnosis, and particu-
Development of molecular techniques for diagnosis of fun- larly in enteroviral CNS infections, has proven cost-effective
gal infections has lagged behind those assays for detection of through decreased use of empirical antibacterial therapy and
other pathogens. There are many reasons for this, but the most reduction in hospital stay (163165). Finally, molecular assays
compelling is the ubiquitous nature of fungi in the environment have added greatly to our understanding of the epidemiology
and the difficulties with contamination control. Initial assays and pathogenesis of these infections (125,166,167).
used species-specific, single-copy genes (150). More recently,
investigators have evaluated the use of highly conserved,
multicopy genes that are universal to all or most fungal species Other Adjunctive Tests in the Diagnosis
(150). Such targets have included 18S rRNA subunit genes, of Central Nervous System Infections
28S rRNA genes, mitochondrial genes, and the intergenic
transcribed spacer (ITS) region of the rRNA gene (149,150). Detection of Cytokines in Cerebrospinal Fluid
Toxoplasmosis TNF, IL-1, and other cytokines have received increasing
Encephalitis is the most common presentation of toxoplasmo- attention as mediators of the inflammatory response during
sis in the immunocompromised patient and most commonly bacterial meningitis and can help distinguish between bacterial
results from reactivation of latent infection (151). CSF antibody and viral meningitis (176180). Lopez-Cortez et al. (178) have
titers have been used to diagnose and follow CNS infections recently demonstrated that a TNF- level of more than 150 pg/
caused by Toxoplasma gondii in both patients with AIDS and mL and IL-1 level more than 90 pg/mL showed sensitivities of
patients without AIDS (152,153). PCR may be useful in the 74% and 90%, respectively, in discriminating viral from aseptic
absence of typical serologic or radiologic studies and could po- meningitis. Pinto Junior et al. (179) found that an elevated CSF
tentially decrease the need for a brain biopsy (154,155). IL-8 level was higher in patients with acute bacterial meningitis
compared to aseptic meningitis and controls (100% of sen-
Whipple Disease sitivity and 94% of specificity). Tang et al. (180) determined
the concentrations of IL-1 and TNF- in the CSF of 171
Whipple disease is a systemic illness caused by Tropheryma specimens of 144 patients whose cases were classified as fol-
whippelii. Illness is characterized by a predominance of intesti- lows: bacterial meningitis (n 23), aseptic meningitis (n 26),
nal manifestations, but extraintestinal manifestations including and nonmeningitis (n 95). Significantly higher serum IL-1
endocarditis, myocarditis, pericarditis, and CNS disease occur and TNF- concentrations were detected in those with bacterial
with relative frequency (156). Although cultivation of the meningitis than those with aseptic meningitis or among those
organism has been reported (157), diagnosis is generally made patients without meningitis (p 0.001). These findings, though
by a combination of cytologic analysis of tissue and fluids using requiring both confirmation and amplification, suggest that
periodic acidSchiff (PAS) staining to demonstrate the presence analysis of TNF and other cytokines, in particular IL-1, may
of macrophages laden with intracellular organisms and electron prove valuable in differentiating acute bacterial meningitis from
microscopy (158). PCR has also been used to determine the viral meningitis and possibly in detecting patients at particular
stage of disease and monitor response to therapy (158,159). risk of adverse outcome. Their role in guiding adjunctive
False-positive PCR results have been reported in asymptomatic therapy, such as corticosteroids and nonsteroidal treatment of
individuals, and for this reason, PCR cannot be recommended BBB injury, is also under investigation.
in place of standard diagnostic techniques (159).

CHARACTERISTIC
Viral Infections CEREBROSPINAL FLUID FINDINGS
PCR methods have had their greatest impact in the diagnosis IN MAJOR CENTRAL NERVOUS
of viral meningitis and encephalitis and have replaced tissue
culture methods (117, 123). Additionally, before the advent
SYSTEM INFECTIONS
of PCR, CSF antibody titers and determination of CSF/serum
antibody ratios were routinely used as methods of acute viral Bacterial Meningitis
diagnosis. Determination of CSF antibody titers per se has
been found valuable in the diagnosis of chronic CNS infec- Bacterial meningitis characteristically produces a neutrophilic
tions such as tropical spastic paraparesis or subacute sclerosing pleocytosis, hypoglycorrhachia (CSF glucose 45 mg/dL), and
panencephalitis (160), but CSF antibody titers alone are of an elevated protein level. Numbers of PMN leukocytes may
limited value in most cases of acute viral encephalitis with the vary from a few to many thousand and usually range between
exception of arboviruses such as West Nile virus where the PCR 1,000 and 10,000 cells. A predominantly (50% of cells)

Scheld_Ch02.indd 18 2/21/14 5:27 PM


Chapter 2: Cerebrospinal Fluid in Central Nervous System Infections 19

lymphocytic pleocytosis has been reported in up to 14% of H. capsulatum, Brucella, or other organisms as indicated by
patients (181), and atypical CSF profiles can especially be seen history and occupational exposure. Serum and CSF should be
in Listeria monocytogenes infection (182). A predominance frozen and held for future serologic studies. CSF findings in
of lymphocytes may also be seen in neonatal gram-negative fungal infections are similar to those described for tuberculous
meningitis (183). Leukocytes may be absent from CSF very meningitis, except that PMN leukocytes may be found less
early in the course of infection, in neonatal meningitis, or in often. The number of cells present may vary widely, and as
severely immunocompromised patients (184). in tuberculous meningitis, CSF may be acellular in severely
immunocompromised patients, including those with AIDS
(190). An exception to this rule is seen in infections with
Brain Abscess and Parameningeal Infection Mucorales, in which the extremely destructive nature of the
infection may result in large numbers of neutrophils (191).
Subdural empyema complicates community-acquired bacterial As in tuberculous meningitis, CSF glucose level may return
meningitis in 2.7% of cases and is usually caused by S. pneu- toward normal before changes are seen in cell count and
moniae in association with sinusitis or otitis (185). CSF protein.
cultures are positive in 93% of these patients. In contrast, LP
in brain abscess is not helpful and has been complicated with
brain herniation in 4 out of 296 (1.3%) of patients (186). The Neurosyphilis
CSF findings are nonspecific and may include (a) a mixed, pre-
dominantly lymphocytic pleocytosis, (b) normal glucose level, Suspicion of neurosyphilis is predicated on the presence of
and (c) elevated protein level. Organisms are not present un- reactive serum nontreponemal tests such as the rapid plasma
less there is accompanying meningitis, in which case CSF find- reagin (RPR) or the Venereal Disease Research Laboratory
ings will be those of bacterial meningitis (187188). (VDRL) and reactive serum treponemal tests such as the
fluorescent treponemal antibody-absorption (FTA-ABS),
Treponema pallidum particle agglutination (TPPA), or various
Tuberculous Meningitis enzyme immunoassays. A serum RPR titer 1:32 or greater is
associated with a higher probability of neurosyphilis in both
Typical findings in tuberculous meningitis are (a) a pleocytosis HIV and nonHIV-infected individuals; a CD4 less than 350
with lymphocytic predominance, (b) lowered glucose level, cells/L is another predictor in HIV-infected patients (192).
and (c) elevated protein level (94,95). In approximately 70% A reactive CSF VDRL test confirms the diagnosis but it may
of patients, the cell count is between 100 and 400 cells (95). be insensitive, and a nonreactive test does not rule out neu-
However, as many as 1,000 to 1,200 cells may be present, rosyphilis (193,194). The CSF may contain variable numbers
and in few patients, the CSF is acellular despite the presence of lymphocytes and an elevated protein level in asymptomatic
of organisms, elevation in protein, and hypoglycorrhachia. or symptomatic neurosyphilis (1). The findings are extremely
Although most cells in the CSF are lymphocytes, relative variable, however, and normal CSF cell count, protein, and
numbers of lymphocytes and PMN leukocytes may vary from glucose values do not exclude active disease (193,194). Rarely,
LP to LP. Protein levels are 100 to 500 mg/dL in 65% of syphilis may present as an acute meningitis, with CSF findings
patients and may reach levels of 1,000 mg or more if treat- similar to those of bacterial meningitis (1). The T. pallidum
ment is delayed (94,95). In 25% of patients, protein levels PCR detection has been of value in primary syphilis, but the
are normal (94). Glucose levels are 30 to 45 mg/dL in 50% utility in neurosyphilis is still under investigation (195).
of patients and may occasionally be less than 10 mg/dL. In
17% of patients, CSF glucose levels are normal (94). More
recently, clinical models that include a duration of symptoms Lyme Borreliosis
for more than 5 days, abnormal neurologic status, a CSF to
serum glucose ratio less than 0.5, a low CSF neutrophilic per- The CSF changes in Lyme neuroborreliosis are typically a
centage (50%), and a CSF protein greater than 100 mg/dL mild lymphocytic pleocytosis, modest elevation of protein
among others can aid clinicians distinguish between TM from level, normal glucose level, and may mimic viral meningitis.
bacterial meningitis but need to be validated in other patient A clinical model named the rule of 7s can help distinguish
populations (189). patients with Lyme meningitis from aseptic meningitis with a
M. tuberculosis may be extremely difficult to detect on sensitivity of 96%. If all the variables (7 days of headache,
smear or to recover by culture. When tuberculous meningi- 70% CSF mononuclear cells, and absence of seventh or
tis is strongly suspected, obtaining more than 6 mL of CSF other cranial nerve palsy) are absent, the patient has a low risk
and repeating LPs can be associated with a higher degree of of having Lyme meningitis (196).
positive acid-fast bacilli smears and cultures (189). PCR may
provide a rapid means of diagnosis superior to acid-fast stain
(125127). Infections Caused by Mycoplasma, Rickettsia,
Ehrlichia, Anaplasma
Fungal and Other Chronic Meningitides CSF in meningoencephalitis associated with M. pneumoniae
infections may be normal but has also been characterized by
Initial requirements for CSF analysis in suspected fungal infec- a usually lymphocytic pleocytosis, elevated protein level, and
tions are similar to those described for tuberculous meningi- mildly depressed glucose level (72). CSF in Rocky Mountain
tis, and the same material may be sent for both mycobacterial spotted fever is usually acellular but may contain increased
and fungal culture. CSF should be submitted for cryptococ- protein concentration (197); typhus may be accompanied by
cal antigen and, if the patient has a history of residence in lymphocytic pleocytosis and elevation of protein concentra-
an endemic area such as the southwestern United States, tion (198). Meningoencephalitis has been reported in human
for complement-fixing antibodies to C. immitis. Additional granulocytic anaplasmosis (Anaplasma phagocytophilum)
samples of CSF should be submitted for serologic studies for in approximately 1% of cases and in human monocytic

Scheld_Ch02.indd 19 2/21/14 5:27 PM


20 Part I: Approach to the Patient and Diagnostic Evaluation

ehrlichiosis (Ehrlichia chaffensis and E. ewingii) in about 20% other conditions. As in viral meningitis, CSF should be sent for
of cases, and it usually has a mild lymphocytic pleocytosis PCR and/or viral culture as appropriate, and both serum and
(199). One study has detected Ehrlichia morulae in CSF (97). CSF should be held for future serologic studies. Serum or CSF
should be sent for IgM and IgG antibody determination in cases
of suspected West Nile or other flavivirus encephalitis (123).
Viral and Other Acute Meningoencephalitis
Viral meningitis produces a lymphocytic pleocytosis, usually AIDS
in the range of 10 to 1,000 cells/mm3 with mildly elevated
protein and normal CSF glucose (25). PMN leukocytes may Abnormalities of CSF in HIV infection are protean and may
at times constitute more than 50% of the cells during the first reflect either (a) a response to CNS invasion by the agent
24 to 36 hours of the infection, and this can change in a re- itself, as in HIV-related meningitis, meningoencephalitis, and
peat LP (200,201) and in one series were shown to be present encephalopathy; (b) meningitis or parenchymal infection by
for several days (202). In some patients with coxsackievirus other agents; or (c) meningeal reaction to neoplastic or ischemic
infections of the CNS, PMN leukocytes may constitute 90% events within brain or spinal cord. The response to any of these
of cells at the onset of infection, and the predominance of conditions is often modified by the immunosuppressive effect
PMN leukocytes may persist for longer than 24 hours. There of the virus (206). In HIV-infected individuals, normal find-
are also reports of CSF samples with few or no cells yield- ings on routine CSF studies do not exclude infectious disease
ing enteroviruses on culture or by PCR (203). Protein is el- of the nervous system. The neurologic complications of HIV
evated in the range of 50 to 100 mg/dL but may sometimes infection and the approach to the patient with suspected neu-
be higher. Glucose is usually normal, but depression of glu- rologic involvement are discussed in detail elsewhere.
cose to levels approaching those of bacterial meningitis has
been reported in infections with HSV-2, herpes zoster virus,
mumps, and lymphocytic choriomeningitis virus (204,205). Prion Diseases
CSF should be routinely sent for PCR analysis for entero-
viruses, including parechoviruses and for HSV. In patients Prion diseases do not elicit a cellular reaction in CSF, so the
who have vesicular rashes, a varicella-zoster virus PCR presence of a CSF pleocytosis essentially excludes this group
should also be sent. Both CSF and serum should be frozen for of diseases. Mild elevation of protein may occasionally be
future serologic testing. seen (207). In recent years, 14-3-3 protein, S100 protein, tau
Requirements for CSF analysis in cases of suspected viral protein, and neuron-specific enolase in CSF have been studied
encephalitis are similar to those for viral meningitis, and CSF as markers for Creutzfeldt-Jakob disease; of these, CSF tau
findings are often similar. PMN leukocytes may be present and 14-3-3 protein has proven most valuable when used in
in large numbers in severe encephalitides accompanied by appropriate clinical context. CSF may contain 14-3-3 protein
extensive destruction of brain tissue. HSV classically produces in other neurologic conditions, however, and its detection
a hemorrhagic encephalitis. However, HSV is not unique is thus not specific for prion diseases (208). CSF from cases
in its ability to produce hemorrhagic necrosis of brain, and of known or suspected Creutzfeldt-Jakob disease should be
RBCs are often not detected; thus, the presence or absence of regarded as infectious and handled according to current guide-
RBCs cannot be used to differentiate HSV encephalitis from lines (208).

References
1. Fishman RA. Cerebrospinal Fluid in Diseases of the Nervous System. 11. Scheld WM. Bacterial meningitis in the patient at risk. Am J Med.
2nd ed. Philadelphia: WB Saunders; 1992. 1984;76(5A):193207.
2. Bleck TP, Greenlee JE. Approach to the patient with central nervous sys- 12. Simmerkoff MS, Moldover NH, Rahal JJ Jr. Absence of detectable bac-
tem infection. In: Mandell GL, Bennett JE, Dolin R, eds. Principles and tericidal and opsonic activities in normal and infected human cerebrospi-
Practice of Infectious Diseases. Philadelphia: Churchill Livingstone; 2000: nal fluids: a regional host defense deficiency. J Lab Clin Med. 1980;95:
950959. 362372.
3. Strazielle N, Ghersi-Egea JF. Choroid plexus in the central nervous system: 13. Leibowitz S, Hughes RAC. Immunology of the Nervous System. London:
biology and physiopathology. J Neuropathol Exp Neurol. 2000;59(7):561574. Arnold; 1983.
4. Dimattio J, Hochwald GM, Malhan C, et al. Effects of changes in serum 14. Zwahlen A, Nydegger UE, Vaudaux P, et al. Complement-mediated opsonic
osmolality on bulk flow of CSF into cerebral ventricles and on brain water activity in normal and infected human cerebrospinal fluid: early response
content. Pflugers Arch. 1975;359:253264. during bacterial meningitis. J Infect Dis. 1982;145(5):635646.
5. Johanson CE, Palm DE, Dyas ML, et al. Microdialysis analysis of effects of 15. Partridge WM. Recent advances in blood-brain barrier transport. Annu
loop diuretics and acetazolamide on chloride transport from blood to CSF. Rev Pharmacol Toxicol. 1988;28:2539.
Brain Res. 1994;641:121126. 16. Aschner M. Astrocytes as mediators of immune and inflammatory
6. Welch K. The principles of physiology of the cerebrospinal fluid in relation responses in the CNS. Neurotoxicology. 1998;19(2):269281.
to hydrocephalus including normal pressure hydrocephalus. In: Friedlander 17. Neal JW, Gasque P. How does the brain limit the severity of inflammation
WJ, ed. Advances in Neurology. Current Reviews. New York: Raven Press; and tissue injury during bacterial meningitis? J Neuropathol Exp Neurol.
1975:345375. 2013;72(5):370385.
7. Yamashima T. Functional ultrastructure of cerebrospinal fluid drainage 18. Titmarsh CJ, Moscovis SM, Hall S, et al. Comparison of cytokine gene
channels in human arachnoid villi. Neurosurgery. 1988;22:633641. polymorphisms among Greek patients with invasive meningococcal disease
8. Krisch B. Ultrastructure of the meninges at the site of penetration of veins or viral meningitis. J Med Microbiol. 2013;62(5):694700.
through the dura mater, with particular reference to Pacchionian granula- 19. Mook-Kanamori BB, Geldhoff M, van der Poll T, et al. Pathogenesis
tions: investigations in the rat and two species of New World monkeys and pathophysiology of pneumococcal meningitis. Clin Microbiol Rev.
(Cebus apella, Callitrix jacchus). Cell Tissue Res. 1988;251:621631. 2011;24(3):557591.
9. Woehrl B, Klein M, Grandgirard D, et al. Bacterial meningitis: current 20. Gloor SM, Wachtel M, Bolliger MF, et al. Molecular and cellular
therapy and possible future treatment options. Expert Rev Anti Infect permeability control at the blood-brain barrier. Brain Res Brain Res Rev.
Ther. 2011;9(11):10531065. 2001;36(23):258264.
10. Ellington E, Margolis G. Block of arachnoid villus by subarachnoid 21. Fishman RA. Blood-brain and CSF barriers to penicillin and related organic
hemorrhage. J Neurosurg. 2003;30:651657. acids. Arch Neurol Psychiatry. 1966;15:113124.

Scheld_Ch02.indd 20 2/21/14 5:27 PM


Chapter 2: Cerebrospinal Fluid in Central Nervous System Infections 21

22. Spector R. The transport of gentamicin in the choroid plexus and cerebro- 56. Davis LE. Normal laboratory values of CSF during pregnancy. Arch
spinal fluid. J Pharmacol Exp Ther. 1975;194(1):8288. Neurol. 1979;36:443.
23. Johnson RT, Johnson KP. Hydrocephalus following viral infection: the 57. Graybill JR, Sobel J, Saag M, et al. Diagnosis and management of increased
pathogenesis of aqueductal stenosis developing after experimental mumps intracranial pressure in patients with AIDS and cryptococcal meningitis.
virus infection. J Neuropathol Exp Neurol. 1968;27:591606. Clin Infect Dis. 2000;30(1):4754.
24. Timmons GD, Johnson KP. Aqueductal stenosis and hydrocephalus after 58. Edlow JA, Malek AM, Ogilvy CS. Aneurysmal subarachnoid hemorrhage:
mumps encephalitis. N Engl J Med. 1970;283:15051507. update for emergency physicians. J Emerg Med. 2008;34(3):237251.
25. Jaijakul S, Arias CA, Hossain M, et al. Toscana meningoencephalitis: a 59. Walker TJ, Nelson LD, Dunphy BW, et al. Comparative evaluation of the
comparison to other viral central nervous system infections. J Clin Virol. Iris iQ200 body fluid module with manual hemacytometer count. Am J
2012;55(3):204208. Clin Pathol. 2009;131:333338.
26. Evans WA. An encephalographic ratio for estimating ventricular and cere- 60. Tunkel AR. Bacterial Meningitis. Philadelphia: Lippincott Williams &
bral atrophy. Arch Neurol Psychiatry. 1942;47:931937. Wilkins; 2001.
27. Filippidis A, Kapsalaki E, Patramani G, et al. Cerebral venous sinus throm- 61. Brouwer M, Thwaites G, Tunkel AR, et al. Dilemmas in the diagnosis
bosis: review of the demographics, pathophysiology, current diagnosis, and of acute community-acquired bacterial meningitis. Lancet. 2012;380:
treatment. Neurosurg Focus. 2009;27(5):E3. 16841692.
28. Bergsneider M. Evolving concepts of cerebrospinal fluid physiology. 62. Mengel M. The use of the cytocentrifuge in the diagnosis of meningitis.
Neurosurg Clin N Am. 2001;12(4):631638, vii. Am J Clin Pathol. 1985;84:212216.
29. Mann JD, Butler AB, Rosenthal JE. Regulation of intracranial pressure in 63. Cinque P, Linde A. CSF Analysis in the diagnosis of viral encephalitis and
rat, dog, and man. Ann Neurol. 1978;3:156165. meningitis. In: Nath A, Berger JR, eds. Clinical Neurovirology. New York:
30. Lundberg N, Cronquist S, Kjallquist A. Clinical investigations on the inter- Marcel Dekker; 2003.
relationships between intracranial pressure and intracranial hemodynam- 64. Rice SK, Heinl RE, Thornton LL, et al. Clinical characteristics, manage-
ics. Prog Brain Res. 1968;30:6981. ment strategies, and cost implication of a statewide outbreak of enterovirus
31. Plum F, Posner JB. The Diagnosis of Stupor and Coma. 3rd ed. Philadelphia: meningitis. Clin Infect Dis. 1995;20:931937.
FA Davis; 1980. 65. Marra CM, Tantalo LC, Maxwell CL, et al. Alternative cerebrospinal fluid
32. Cuneo RA, Caronna JJ, Pitts L, et al. Upward transtentorial herniation: tests to diagnose neurosyphilis in HIV-infected individuals. Neurology.
seven cases and a literature review. Arch Neurol. 1979;36(10):618623. 2004;63(1):8588.
33. Rebaud P, Berthier JC, Hartemann E, et al. Intracranial pressure in child- 66. Graeff-Teixeira C, Armburu da Silva AC, Yoshimura K. Update on
hood central nervous system infections. Intensive Care Med. 1988;14: eosinophilic meningoencephalitis and its clinical relevance. Clin. Microbiol
522525. Rev. 2009;22(2):322348.
34. Goitein KJ, Tamir I. Cerebral perfusion pressure in central nervous system 67. Matthews WF, Frohmeyer WB Jr. The in vitro behavior of erythrocytes in
infections of infancy and childhood. J Pediatr. 1983;103:4043. human cerebrospinal fluid. J Lab Clin Med. 1955;45:508515.
35. Thomas K, Hasbun R, Jekel J, et al. The diagnostic accuracy of Kernigs 68. Meyer GC, Netsky MG. Relation of blood and cerebrospinal fluid glucose.
sign, Brudzinskis sign, and nuchal rigidity in patients with suspected men- Arch Neurol Psychiatry. 1962;6:1820.
ingitis. Clin Infect Dis. 2002;35:4652. 69. Swartz MN, Dodge PR. Bacterial meningitisa review of selected aspects 1.
36. van de Beek D, de Gans J, Spanjaard L, et al. Clinical features and General clinical features, special problems, and unusual meningeal reactions
prognostic factors in adults with bacterial meningitis. N Engl J Med. mimicking bacterial meningitis. N Engl J Med. 1965;272: 779787.
2004;351:18491859. 70. Ellner JJ, Bennett JE. Chronic meningitis. Medicine. 1976;55:341369.
37. Korein J, Cravioto H, Leicach M. Reevaluation of lumbar puncture: a study 71. Hasbun R. The acute aseptic meningitis. Cur Infect Dis Rep. 2000;2(4):
of 129 patients with papilledema or intracranial hypertension. Neurology. 345351.
1959;9:290297. 72. Tsiodras S, Kelesidis I, Kelesidis T. et al. Central Nervous system mani-
38. Hasbun R, Abrahams J, Jekel J, et al. Computed tomography of the head festations of Mycoplasma pneumoniae infections. J Infect. 2005;51(5):
before lumbar puncture in adults with suspected meningitis. N Engl J Med. 343354.
2001;345:17271733. 73. Sharma OP. Neurosarcoidosis: a personal perspective based on the study of
39. Tunkel AR, Hartman BJ, Kaplan SL, et al. Practice guidelines for the man- 37 patients. Chest. 1997;112(1):220228.
agement of bacterial meningitis. Clin Infect Dis. 2004;39(9):12671284. 74. Zeller JA, Zunker P, Witt K, et al. Unusual presentation of carcinomatous
40. Proulx N, Frechette D, Toye D, et al. Delays in the administration of anti- meningitis: case report and review of typical CSF findings. Neurol Res.
biotics are associated with mortality from adult acute bacterial meningitis. 2002;24(7):652654.
QJM. 2005;98:291298. 75. Pasquier F, Leys D, Vermersch P, et al. Severe hypoglycorrhachia after
41. Joffe AR. Lumbar puncture and brain herniation in acute bacterial menin- subarachnoid hemorrhage: two cases with spontaneous recovery in adults.
gitis: a review. J Intensive Care Med. 2007;22:194207. Acta Neurol Belg. 1987;87(2):7679.
42. Domenicucci M, Ramieri A, Ciappetta P, et al. Nontraumatic acute spinal 76. Valmari P, Peltola H, Kataja M. Cerebrospinal fluid white cell, glucose and
subdural hematoma: report of five cases and review of the literature. protein changes during the treatment of Haemophilus influenzae meningi-
J Neurosurg. 1999;91(1)(suppl):6573. tis. Scand J Infect Dis. 1986;18(1):3943.
43. Howard SC, Gajjar A, Ribeiro RC, et al. Safety of lumbar puncture for 77. Roine I, Foncea LM, Ledermann W, et al. Slow recovery of cerebrospinal
children with acute lymphoblastic leukemia and thrombocytopenia. JAMA. fluid glucose and protein concentrations distinguish pneumococcal from
2000;284(17):22222224. Haemophilus influenzae and meningococcal meningitis in children. Pediatr
44. Baer ET. Post-dural puncture bacterial meningitis. Anesthesiology. 2006; Infect Dis J. 1995;14(10):905907.
105:381393. 78. Silver TS, Todd JK. Hypoglycorrhachia in pediatric patients. Pediatrics.
45. Srinivasan V, Gertz RE Jr, Shewmaker PL, et al. Using PCR-based detection 1976;58:6771.
and genotyping to trace Streptococcus salivarius meningitis outbreak strain 79. Omene JA, Okolo AA, Longe AC, et al. The specificity and sensitivity of
to oral flora of radiology physician assistant. PLoS One. 2012;7(2):e32169. CSF and blood glucose concentration in the diagnosis of neonatal meningi-
46. Ahmed SV, Jayawarna C, Jude E. Post lumbar puncture headache: diagnosis tis. Ann Trop Paediatr. 1985;5(1):3739.
and management. Postgrad Med J. 2006;82:713716. 80. Powers WJ. Cerebrospinal fluid to serum glucose ratios in diabetes mellitus
47. Welch H, Hasbun R. Lumbar puncture and cerebrospinal fluid analysis. and bacterial meningitis. Am J Med. 1981;71(2):217220.
Handb Clin Neurol. 2010;96:3149. 81. Spanos A, Harrell FE, Durack DT. Differential diagnosis of acute
48. Armon C, Evans RW. Addendum to assessment: prevention of post-lumbar meningitis, an analysis of the predictive value of initial observations.
puncture headaches. Neurology. 2005;65:510512. JAMA. 1989;262:27002707.
49. Reina MA, de Leon-Casasola OA, Lopez A, et al. An in vitro study of dural 82. Khoury N, Hossain M, Wootton S, et al. Meningitis with a negative
lesions produced by 25-gauge Quincke and Whitacre needles evaluated by cerebrospinal fluid Gram stain in adults: risk classification for an adverse
scanning electron microscopy. Reg Anesth Pain Med. 2000;25(4):393402. clinical outcome. Mayo Clin Proc. 2012;87(12):11811188.
50. Norman MG. Respiratory arrest and cervical spinal cord infarction follow- 83. Quagliarello VJ, Ma A, Stukenbrok H, et al. Ultrastructural localization of
ing lumbar puncture in meningitis. Can J Neurol Sci. 1982;9:443447. albumin transport across the cerebral microvasculature during experimen-
51. Strauss SE, Thorpe KE, Holroyd-Ledu J. How do I perform a lumbar tal meningitis in the rat. J Exp Med. 1991;174(3):657672.
puncture and analyze the result to diagnose bacterial meningitis? JAMA. 84. Nigrovic LE, Malley R, Macias CG, et al. Effect of antibiotic pretreatment
2006;296:20122022. on cerebrospinal fluid profiles of children with bacterial meningitis.
52. Ayer JB. Puncture of the cisterna magna. Arch Neurol Psychiatry. Pediatrics. 2008;122(4):726730.
1923;4:529541. 85. Kelly SB, Kinsella K, Duggan M, et al. A proposed modification to the
53. Portela, L, Souza V, Pahl F et al. Laceration of the posterior inferior McDonald 2010 criteria for the diagnosis of primary progressive multiple
cerebellar artery by suboccipital puncture of the cisterna magna: case sclerosis. Mult Scler. 2013;19(8):10951100.
report. Arq Neuropsiquiatr. 2004;62(3b):882884. 86. Flitch MF, van de Beek D. Emergency diagnosis and treatment of adult
54. van de Beek D, Drake J, Tunkel AR. Nosocomial bacterial meningitis. meningitis. Lancet Infect Dis. 2007;7:191200.
N Engl J Med. 2010;362(2):146154. 87. Kleiman MB, Reynolds JK, Watts NH, et al. Superiority of acridine orange
55. Vidyasagar D, Raju TNK. A simple noninvasive technique of measuring stain versus Gram stain in partially treated bacterial meningitis. J Pediatr.
intracranial pressure in the newborn. Pediatrics. 1977;59:957961. 1984;104(3):401404.

Scheld_Ch02.indd 21 2/21/14 5:27 PM


22 Part I: Approach to the Patient and Diagnostic Evaluation

88. Genne D, Siegrist HH, Lienhard R. Enhancing the etiologic diagnosis of 119. Desai D, Nataraj G, Kulkarni S, et al. Utility of the polymerase chain
community-acquired pneumonia in adults using the urinary antigen assay reaction in the diagnosis of tuberculous meningitis. Res Microbiol.
(Binax NOW). Int J Infect Dis. 2006;10:124128. 2006;157:967970.
89. Marcos MA, Martinez E, Almela M, et al. New rapid antigen test for 120. Parent du Chatelet I, Traoere Y, Gessner BD, et al. Bacterial meningitis
diagnosis of pneumococcal meningitis. Lancet. 2001;357:14991500. in Burkina Faso: surveillance using field-based polymerase chain reaction
90. Samra Z, Schmuely H, Nahum E, et al. Use of the NOW Streptococcus testing. Clin Infect Dis. 2005;40:1725.
pneumoniae urinary antigen test in cerebrospinal fluid for rapid diagno- 121. Whitley RJ. Herpes simplex encephalitis: adolescents and adults. Antiviral
sis of pneumococcal meningitis. Diagn Microbiol Infect Dis. 2003;45: Res. 2006;71:141148.
237240. 122. Roos KL. Encephalitis. Neurol Clin. 1999;17:813833.
91. Mosi JC, Saha SK, Adegoke G., et al. Enhanced diagnosis of pneu- 123. Tunkel AR, Glaser CA, Bloch KC, et al. The management of encephalitis:
mococcal meningitis using the Binax NOW S. pneumoniae immuno- clinical practice guidelines by the Infectious Diseases Society of America.
chromatographic test: a multi-site study. Clin Infect Dis. 2009;48(suppl 2): Clin Infect Dis. 2008;47(3):303327.
S49S56. 124. Kost C, Rogers B, Oberste S, et al. Multicenter beta trial of the GeneXpert
92. Gray F, Alonso JM. Bacterial infections of the central nervous system. enterovirus assay. J Clin Microbiol. 2007;45(4);10811086.
In: Graham DI, Lantos PL, eds. Greenfields Neuropathology. 7th ed. 125. Rafi W, Venkataswamy MM, Nagarathna S, et al. Role of IS6110 uniplex
London: Arnold; 2002;151193. PCR in the diagnosis of tuberculous meningitis: experience at a tertiary
93. Karcher D, Mc Pherson R. Cerebrospinal fluid, synovial, serous body neurocentre. Int J Tuberc Lung Dis. 2007;11:209213.
fluids and alternative specimens. In: Henry JB, ed. Clinical Diagnosis 126. Nguyen LA, Kox LFF, Pham LD, et al. The potential contribution of the
and Management by Laboratory Methods. Philadelphia: WB Saunders; polycerase chain reaction to the diagnosis of tuberculous meningitis. Arch
2011:480508. Neurol. 1996;53:771776.
94. Thomson RB Jr, Bertram H. Laboratory diagnosis of central nervous 127. Takahashi T, Tamura M, Takasu T. The PCR-based diagnosis of central ner-
system infections. Infect Dis Clin North Am. 2001;15:10471071. vous system tuberculosis: up to date. Tuberc Res Treat. 2012;2012:831292.
95. Katti MK. Pathogenesis, diagnosis, treatment, and outcome aspects of 128. Choi SH, Kim YS, Bae IG, et al. The possible role of cerebrospinal fluid
cerebral tuberculosis. Med Sci Monit. 2004;10:RA215RA229. adenosine deaminase activity in the diagnosis of tuberculous meningitis in
96. Thakur R, Goyal R, Sarma S. Laboratory diagnosis of tuberculous adults. Clin Neurol Neurosurg. 2002;104(1):1015.
meningitisis there a scope for further improvement? J Lab Physicians. 129. Lopez-Cortes LF, Cruz-Ruiz M, Gomez-Mateos J, et al. Adenosine deami-
2010;2(1):2124. nase activity in the CSF of patients with aseptic meningitis: utility in the
97. Rikihisa Y. Anaplasma phagocytophilum and Ehrlichia chaffeensis: sub- diagnosis of tuberculous meningitis or neurobrucellosis. Clin Infect Dis.
versive manipulators of host cells. Nat Rev Microbiol. 2010;8(5):328339. 1995;20(3):525530.
98. Tunkel A. Bacterial Meningitis. Philadelphia: Lippincot Williams & 130. Centers for Disease Control and Prevention. Recommendations for test
Wilkins; 2001:142148. performance and interpretation from the second national conference on
99. McMullan BJ, Halliday C, Sorrell TC, et al. Clinical utility of crypto- serological diagnosis of Lyme disease. MMWR Morb Mortal Wkly Rep.
coccal antigen lateral flow assay in a diagnostic mycology laboratory. 1995;44:590.
PLoS One. 2012;7(11):e49541. 131. Dattwyler RJ, Volkman DJ, Luft BJ, et al. Seronegative Lyme disease: dis-
100. Singh U, Petri WA Jr. Free-living amebas. In: Mandell GL, Bennett JE, sociation of specific T- and B-lymphocyte responses to Borrelia burgdor-
Dolin R, eds. Principles and Practice of Infectious Diseases. Philadelphia: feri. N Engl J Med. 1988;319(22):14411446.
Churchill Livingstone; 2000:28112817. 132. Kaiser R. Neuroborreliosis. J Neurol. 1998;245(5):247255.
101. White MH. Cryptococcosis. Inf Dis Clin North Am. 1994;8:383398. 133. Steere AC, Berardi VP, Weeks KE, et al. Evaluation of the intrathecal
102. Fernandez M, Moylett EH, Noyola DE, et al. Candidal meningitis in antibody response to Borrelia burgdorferi as a diagnostic test for Lyme
neonates: a 10-year review. Clin Infect Dis. 2000;31(2):458463. neuroborreliosis. J Infect Dis. 1990;161:12031209.
103. Andrew R, Craig D, Woods M. Fungal infections of the central nervous sys- 134. Halperin JJ, Luft BJ, Anand AK, et al. Lyme neuroborreliosis: central ner-
tem: a review of fungal pathogens and treatments. Neur Ind. 2007;55(3): vous system manifestations. Neurology. 1989;39(6):753759.
251259. 135. Tylewska-Wierzbanowska S, Chmielewski T. Limitation of serologi-
104. Chonmaitree T, Baldwin CD, Lucia HL. Role of the virology laboratory cal testing for Lyme borreliosis: evaluation of ELISA and western blot
in diagnosis and management of patients with central nervous system in comparison with PCR and culture methods. Wien Klin Wochenschr.
disease. Clin Microbial Rev. 1989;2:114. 2002;114(1314):601605.
105. Storch GA. The diagnosis of viral infections. Infect Dis Clin Pract. 136. Nolte O. Nucleic acid amplification based diagnostic of Lyme (neuro-)
1993;2:120. borreliosislost in the jungle of methods, targets, and assays? Open
106. Campbell GL, Marfin AA, Lanciotti RS, et al. West Nile virus. Lancet Neurol J. 2012;6:129139.
Infect Dis. 2002;2:116. 137. Reed KD. Laboratory testing for Lyme disease: possibilities and practicali-
107. Huy NT, Thao NT, Diep DT, et al. Cerebrospinal fluid lactate concentra- ties. J Clin Microbiol. 2002;40(2):319324.
tion to distinguish bacterial from aseptic meningitis: a systemic review and 138. Zbinden R, Goldenberger D, Lucchini GM, et al. Comparison of two
meta-analysis. Crit Care. 2010;14:R240. methods for detecting intrathecal synthesis of Borrelia burgdorferi
108. Sakushima K, Hayashino Y, Kawaguchi T, et al. Diagnostic accuracy of specific antibodies and PCR for diagnosis of Lyme neuroborreliosis. J Clin
cerebrospinal fluid lactate for differentiating bacterial meningitis from Microbiol. 1994;32(7):17951798.
aseptic meningitis: a meta-analysis. J Infect. 2011;62:255262. 139. Christen HJ, Eiffert H, Ohlenbusch A, et al. Evaluation of the polymerase
109. Ray P, Badarou-Acossi G, Viallon A, et al. Accuracy of the cerebrospi- chain reaction for the detection of Borrelia burgdorferi in cerebrospi-
nal fluid results to differentiate bacterial from non bacterial meningi- nal fluid of children with acute peripheral facial palsy. Eur J Pediatr.
tis, in case of negative gram-stained smear. Am J Emerg Med. 2007;25: 1995;154(5):374377.
179184. 140. Bremell D, Mattsson N, Edsbagge M, et al. Cerebrospinal fluid CXCL13
110. Sormunen P, Kallio MJ, Kilpi T, et al. C-reactive protein is useful in in Lyme neuroborreliosis and asymptomatic HIV infection. BMC Neurol.
distinguishing Gram-stain negative bacterial meningitis from viral 2013;13:14712377.
meningitis in children. J Pediatr. 1999;134:725729. 141. Currie BP, Freundlich LF, Soto MA, et al. False-negative cerebrospinal fluid
111. Dubos F, Korczowski B, Aygun DA, et al. Serum procalcitonin level and cryptococcal latex agglutination tests for patients with culture-positive
other biomarkers to distinguish between bacterial meningitis and aseptic cryptococcal meningitis. J Clin Microbiol. 1993;31(9): 25192522.
meningitis: a meta-analysis. J Infect. 2011;62:255262. 142. Berlin L, Pincus JH. Cryptococcal meningitis: false-negative antigen test
112. Taskin E, Turgot M, Kilic M, et al. Serum procalcitonin and cerebrospi- results and cultures in nonimmunosuppressed patients. Arch Neurol.
nal fluid cytokines level in children with meningitis. Mediators Inflamm. 1989;46(12):13121316.
2004;13:269273. 143. Jarvis JN, Percival A, Bauman S, et al. Evaluation of a novel point-of-
113. Ernst A, Morgenthaler NG, Buerger K, et al. Procalcitonin is elevated in care cryptococcal antigen test on serum, plasma, and urine from patients
the cerebrospinal fluid of patients with dementia and acute neuroinflam- with HIV-associated cryptococcal meningitis. Clin Infect Dis. 2011;53:
mation. J Neuroimmunol. 2007;189:169174. 10191023.
114. Mein J, Lum G. CSF bacterial antigen detection tests offer no advantage 144. Roy M, Chiller T. Preventing deaths in cryptococcal meningitis: from
over Grams stain in the diagnosis of bacterial meningitis. Pathology. bench to bedside. Expert Rev Anti Infect Ther. 2011;9(9):715717.
1999;31(1):6769. 145. Bouza E, Dreyer JS, Hewitt WL, et al. Coccidioidal meningitis: an analy-
115. Perkins MD, Mirrett S, Reller LB. Rapid bacterial antigen detection is not sis of thirty-one cases and review of the literature. Medicine (Baltimore).
clinically useful. J Clin Microbiol. 1995;33(6):14861491. 1981;60(3):139172.
116. Hasbun R, Bijlsma M, Brouwer MC, et al. Risk score for identifying 146. Williams PL, Johnson R, Pappagianis D, et al. Vasculitic and encephalitic
adults with CSF pleocytosis and negative CSF Gram stain at low risk for complications associated with Coccidioides immitis infection of the cen-
an urgent treatable cause. J Infect. 2013;67(2):102110. tral nervous system in humans: report of 10 cases and review. Clin Infect
117. DeBiasi RL, Tyler KL. Molecular methods for diagnosis of viral encepha- Dis. 1992;14(3):673682.
litis. Clin Microbiol Rev. 2004;17:903925. 147. Wheat LJ, Kohler RB, Tewari RP, et al. Significance of Histoplasma
118. Yamamoto Y. PCR in diagnosis of infection: detection of bacteria in cere- antigen in the cerebrospinal fluid of patients with meningitis. Arch Intern
brospinal fluids. Clin Diagn Lab Immunol. 2002;9:508514. Med. 1989;149(2):302304.

Scheld_Ch02.indd 22 2/21/14 5:27 PM


Chapter 2: Cerebrospinal Fluid in Central Nervous System Infections 23

148. Wheat LJ, Batteiger BE, Sathapatayavongs B. Histoplasma capsulatum 175. Lanciotti RS, Kerst AJ. Nucleic acid sequence-based amplification assays
infections of the central nervous system: a clinical review. Medicine for rapid detection of West Nile and St. Louis encephalitis viruses. J Clin
(Baltimore). 1990;69(4):244260. Microbiol. 2001;39(12):45064513.
149. Wheat LJ, Musial CE, Jenny-Avital E. Diagnosis and management 176. Bociaga-Jasik M, Garlicki A, Ciesla A, et al. The diagnostic value of
of central nervous system histoplasmosis. Clin Infect Dis. 2005;40: cytokine and nitric oxide concentrations in cerebrospinal fluid for the dif-
844852. ferential diagnosis of meningitis. Adv Med Sci. 2012;57(1):142147.
150. Lindsley MD, Hurst SF, Iqbal NJ, et al. Rapid identification of dimor- 177. Mukai AO, Krebs VL, Bertoli CJ, et al. TNF-alpha and IL-6 in the diagnosis
phic and yeast-like fungal pathogens using specific DNA probes. J Clin of bacterial and aseptic meningitis in children. Pediatr Neurol. 2006;
Microbiol. 2001;39(10):35053511. 34(1):2529.
151. Montoya JG. Laboratory diagnosis of Toxoplasma gondii infection and 178. Lopez-Cortes LF, Marquez-Arbizu R, Jimenez-Jimenez LM, et al.
toxoplasmosis. J Infect Dis. 2002;185(suppl 1):S73S82. Cerebrospinal fluid tumor necrosis factor-alpha, interleukin-1beta, inter-
152. Luft BJ, Hafner R, Korzun AH, et al. Toxoplasmic encephalitis in leukin-6, and interleukin-8 as diagnostic markers of cerebrospinal fluid
patients with the acquired immunodeficiency syndrome. N Engl J Med. infection in neurosurgical patients. Crit Care Med. 2000;28(1):215219.
1993;329:9951000. 179. Pinto Junior VL, Rebelo MC, Gomes RN, et al. IL-6 and IL-8 in cerebro-
153. Potasman I, Resnick L, Luft BJ, et al. Intrathecal production of antibodies spinal fluid from patients with aseptic meningitis and bacterial meningitis:
against Toxoplasma gondii in patients with toxoplasmic encephalitis their potential role as a marker for differential diagnosis. Braz J Infect
and the acquired immunodeficiency syndrome (AIDS). Ann Intern Med. Dis. 2011;15(2):156158.
1988;108(1):4951. 180. Tang RB, Lee BH, Chung RL, et al. Interleukin-1 beta and tumor necrosis-
154. Alfonso Y, Fraga J. Cox R, et al. Conventional polymerase chain reac- alpha in cerebrospinal fluid of children with bacterial meninigitis. Childs
tion for the diagnosis of neurotoxoplasmosis: comparison of three sets of Nerv Syst. 2001;17(8):453486.
primers for the B1 gene using CSF samples. Diag Microb and Infect Dis. 181. Powers WJ. Cerebrospinal fluid lymphocytosis in acute bacterial meningi-
2013;75(2):150154. tis. Am J Med. 1985;79:216220.
155. Sukthana Y, Mahittikorn A, Wickert H, et al. A promising diagnostic 182. Brouwer MC, van de Beek D, Sebastiaan G. B., et al. Community-
tool for toxoplasmic encephalitis: tachyzoite/bradyzoite stage-specific acquired Listeria monocytogenes meningitis in adults. Clin Infect Dis.
RT-PCR. Int J Infect Dis. 2012;16:e279e284. 2006;43(10):12331238.
156. Gerard A, Sarrot-Reynauld F, Liozon E, et al. Neurologic presentation of 183. Bonadio WA. Acute bacterial meningitis: cerebrospinal fluid differential
Whipple disease: report of 12 cases and review of the literature. Medicine count. Clin Pediatr (Phila). 1988;27(9):445447.
(Baltimore). 2002;81(6):443457. 184. Fishbein DB, Palmer DL, Porter KM, et al. Bacterial meningitis in the
157. Raoult D, Birg ML, La Scola B, et al. Cultivation of the bacillus of absence of CSF pleocytosis. Arch Intern Med. 1981;141(10):13691372.
Whipples disease. N Engl J Med. 2000;342(9):620625. 185. Jim KK, Brouwer MC, van der Ende, et al. Subdural empyema in bacterial
158. von Herbay A, Ditton HJ, Schuhmacher F, et al. Whipples disease: meningitis. Neurology. 2012;79(21):21332139.
staging and monitoring by cytology and polymerase chain reac- 186. Tattevin P, Bruneel F, Rgnier B. Cranial CT before lumbar puncture in
tion analysis of cerebrospinal fluid. Gastroenterology. 1997;113(2): suspected meningitis. N Engl J Med. 2002;346(16):12481251.
434441. 187. King N. Brain abscess. In: Roos KL, Tunkel AR, eds. Handbook of
159. Fenollar F, Fournier PE, Raoult D, et al. Quantitative detection of Clinical Neurology: Bacterial Infections of the Central Nervous System.
Tropheryma whipplei DNA by real-time PCR. J Clin Microbiol. 2002; Amsterdam: Elsevier; 2010:6574.
40(3):11191120. 188. Helweg-Larsen J, Astradsson A, Richhall H, et al. Pyogenic brain abscess,
160. Puccioni-Sohler M, Rios M, Carvalho SM, et al. Diagnosis of HAM/ a 15 year survey. BMC Infect Dis. 2012;12:332.
TSP based on CSF proviral HTLV-I DNA and HTLV-I antibody index. 189. Thwaites G, Fisher M, Hemingway C, et al. British Infection Society
Neurology. 2001;57(4):725727. guidelines for the diagnosis and treatment of tuberculosis of the central
161. Sauerbrei A, Eichhorn U, Hottenrott G, et al. Virological diagnosis of nervous system in adults and children. J of Infect. 2009;59:167187.
herpes simplex encephalitis. J Clin Virol. 2000;17(1):3136. 190. Panther LA, Sande MA. Cryptococcal meningitis in the acquired immuno-
162. Murray K, Mertens E, Despress P. West Nile virus and its emergence in the deficiency syndrome. Semin Respir Infect. 1990;5(2):138145.
United States. Vet Res. 2010;41(6):67. 191. Lehrer RI, Hoard DH, Sypherd PS, et al. Mucormycosis. Ann Intern Med.
163. Ramers C, Billman G, Hartin M, et al. Impact of a diagnostic cerebrospi- 1980;93:93108.
nal fluid enterovirus polymerase chain reaction test on patient manage- 192. Marra CM, Maxwell CL, Smith SL, et al. Cerebrospinal fluid abnormali-
ment. JAMA. 2000;283(20):26802685. ties in patients with syphilis: association with clinical and laboratory fea-
164. Robinson CC, Willis M, Meagher A, et al. Impact of rapid polymerase tures. J Infect Dis. 2004;189(3):369374.
chain reaction results on management of pediatric patients with enterovi- 193. Ho EL, Marra CM. Treponemal tests for neurosyphilis-less accurate than
ral meningitis. Pediatr Infect Dis J. 2002;21(4):283286. what we thought? Sex Trasnm Dis. 2012;39(4):298299.
165. Rotbart HA, Sawyer MH, Fast S, et al. Diagnosis of enteroviral menin- 194. Marra CM, Tantalo LC, Maxwell CL, et al. The rapid regain test cannot
gitis by using PCR with a colorimetric microwell detection assay. J Clin replace the venereal disease research laboratory test for neurosyphilis. Sex
Microbiol. 1994;32(10):25902592. Transm Dis. 2012;39(6):453457.
166. Guffond T, Dewilde A, Lobert PE, et al. Significance and clinical relevance 195. Peng R-R, Wang AL, Li J, et al. Molecular typing of Treponema pallidum: a
of the detection of herpes simplex virus DNA by the polymerase chain systematic review and meta-analysis. PLoS Negl Trop Dis. 2011;5(11):e1273.
reaction in cerebrospinal fluid from patients with presumed encephalitis. 196. Cohn K, Thompson AD, Shah S, et al. Validation of a clinical prediction
Clin Infect Dis. 1994;18(5):744749. rule to distinguish Lyme meningitis from aseptic meningitis. Pediatrics.
167. Cinque P, Cleator GM, Weber T, et al. The role of laboratory investi- 2012;129(1):e46e53.
gation in the diagnosis and management of patients with suspected her- 197. Miller JQ, Price TR. The nervous system in Rocky Mountain spotted
pes simplex encephalitis: a consensus report. The EU Concerted Action fever. Neurology. 1972;22:561566.
on Virus Meningitis and Encephalitis. J Neurol Neurosurg Psychiatry. 198. Massung RF, Davis LE, Slater K, et al. Epidemic typhus meningitis in the
1996;61(4):339345. southwestern United States. Clin Infect Dis. 2001;32:979982.
168. Fodor PA, Levin MJ, Weinberg A, et al. Atypical herpes simplex virus 199. St Clair K, Decker CF. Ehrlichioses: anaplasmosis and human ehrlichiosis.
encephalitis diagnosed by PCR amplification of viral DNA from CSF. Dis Mon. 2012;58:346354.
Neurology. 1998;51(2):554559. 200. Feigin RD, Shackleford PG. Value of repeat lumbar puncture in the dif-
169. Read SJ, Kurtz JB. Laboratory diagnosis of common viral infections of the ferential diagnosis of meningitis. N Engl J Med. 1973;289:571574.
central nervous system by using a single multiplex PCR screening assay. 201. Varki AP, Puthuran P. Value of second lumbar puncture in confirming a
J Clin Microbiol. 1999;37(5):13521355. diagnosis of aseptic meningitis. Arch Neurol. 1979;36:581582.
170. Kleinschmidt-DeMasters BK, Gilden DH. Varicella-Zoster virus infections 202. Negrini B, Kelleher KJ, Wald ER. Cerebrospinal fluid findings in aseptic
of the nervous system: clinical and pathologic correlates. Arch Pathol Lab versus bacterial meningitis. Pediatrics. 2000;105(2):316319.
Med. 2001;125(6):770780. 203. Henquell C, Chambon M, Bailly JL, et al. Prospective analysis of 61 cases
171. Bestetti A, Pierotti C, Terreni M, et al. Comparison of three nucleic acid of enteroviral meningitis: interest of systematic genome detection in cere-
amplification assays of cerebrospinal fluid for diagnosis of cytomegalovi- brospinal fluid irrespective of cytologic examination results. J Clin Virol.
rus encephalitis. J Clin Microbiol. 2001;39(3):11481151. 2001;21(1):2935.
172. Weinberg A, Li S, Palmer M, et al. Quantitative CSF PCR in Epstein- 204. Brenton DW. Hypoglycorrhachia in herpes simplex type 2 meningitis.
Barr virus infections of the central nervous system. Ann Neurol. 2002;53: Arch Neurol. 1980;37:317.
543548. 205. Reimer LG, Beller LB. CSF in herpes zoster meningoencephalitis. Arch
173. Garcia de Viedma D, Alonso R, Miralles P, et al. Dual qualitative-quan- Neurol. 1981;38(10):668.
titative nested PCR for detection of JC virus in cerebrospinal fluid: high 206. Churchill M, Nath A. Where does HIV hide? A focus on the central ner-
potential for evaluation and monitoring of progressive multifocal leuko- vous system. Curr Opn HIV AIDS. 2013;8:165169.
encephalopathy in AIDS patients receiving highly active antiretroviral 207. Hamlin C, Puoti G, Berri S, et al. A comparison of tau and 1433 protein in the
therapy. J Clin Microbiol. 1999;37(3):724728. diagnosis of Creuktzfeldt-Jakob disease. Neurology. 2012;79(6):547552.
174. Briese T, Glass WG, Lipkin WI. Detection of West Nile virus sequences in 208. Koehler AP, Athan E, Collins SJ. Updated Creutzfeldt-Jakob disease infection
cerebrospinal fluid. Lancet. 2000;355(9215):16141615. control guidelines: sifting facts from fiction. Med J Aust. 2013;70(2):245246.

Scheld_Ch02.indd 23 2/21/14 5:27 PM


CHAPTER 3 IMAGING OF INTRACRANIAL
INFECTIONS
CARRIE P. MARDER AND KATHLEEN R. FINK

Intracranial infections are usually diagnosed by clinical assess- According to the American College of Radiology Appro-
ment and laboratory investigations, particularly cerebrospi- priateness Criteria for headache (3), both NCCT and contrast-
nal fluid (CSF) analysis, combined with radiologic findings. enhanced MRI of the head are usually appropriate for patients
Imaging plays an important role by providing or narrowing presenting with new headache and suspected meningitis or
the differential diagnosis and occasionally identifying a par- encephalitis, with the choice of test depending on local prefer-
ticular entity that has a characteristic appearance, such as ence and availability. When MRI is unavailable or contrain-
herpes simplex virus encephalitis, pyogenic abscess, or empy- dicated, contrast-enhanced CT may be a suitable alternative.
ema. Imaging is also crucial for identifying complications of Cerebrovascular complications of infection are relatively fre-
disease and assessing response to treatment. Finally, imaging quent, and therefore magnetic resonance angiography (MRA)
contributes to the evaluation of opportunistic infections in and magnetic resonance venography (MRV), either with or with-
immunocompromised patients and other patients at high risk out contrast, may also be appropriate tests. Computed tomogra-
for infection. phy angiography (CTA) may be performed when there is strong
Here, we emphasize the overall strategy for imaging intra- suspicion for vascular disease or to further evaluate abnormalities
cranial infections and highlight specific entities in which imag- detected by MRA. Other advanced imaging techniques such as
ing findings contribute to diagnosis or management. We stress computed tomography perfusion (CTP) or magnetic resonance
the major complications of intracranial infections and address perfusion (MRP), magnetic resonance spectroscopy (MRS), and
special considerations for immunocompromised patients. nuclear medicine studies such as single-photon emission com-
puted tomography (SPECT) and 18F-fluorodeoxyglucose posi-
tron emission tomography (FDG-PET) or PET/CT are usually
IMAGING STRATEGY only performed for problem solving after the standard imaging
evaluation has been performed.
Patients suspected of harboring intracranial infection who For evaluation of a new headache in an HIV-positive or
present with altered mental status, seizures, or focal neurologic immunocompromised individual, MRI with or without con-
deficits should emergently undergo noncontrast computed trast is usually appropriate as the initial imaging test (ACR
tomography (NCCT) to exclude life-threatening conditions. Appropriateness Criteria Headache [3]). For an indeterminate
In the acute setting, NCCT is the test of choice to assess for cerebral mass in an immunocompromised patient, additional
hydrocephalus, cerebral edema, mass lesions, or hemorrhage problem-solving techniques such as FDG-PET/CT, SPECT, MRS,
and is often performed prior to lumbar puncture (LP) to ex- and MRP may be helpful to narrow the differential diagnosis.
clude impending brain herniation. NCCT is widely available, In neonates in whom the anterior fontanelle is patent, cra-
and the images are rapidly acquired, making the examination nial ultrasound may be used to evaluate for hydrocephalus,
well tolerated even by critically ill patients. subdural and epidural collections, and parenchymal masses (4).
Clinically stable patients in whom immediately life- Uncomplicated rhinosinusitis is usually managed clinically
threatening conditions have been excluded by NCCT often without the need for imaging, but for suspected intracranial
require further evaluation with contrast-enhanced magnetic or orbital complications of sinonasal disease, both NCCT and
resonance imaging (MRI), which has a greater sensitiv- contrast-enhanced MRI of the head, orbits, and paranasal
ity for leptomeningitis, ventriculitis, cerebral abscess, and sinuses are usually appropriate, with CT and MRI serving as
empyema as well as downstream complications of infec- complementary examinations (ACR Appropriateness Criteria
tion such as infarctions. MRI lacks ionizing radiation, so it Sinonasal Disease [5]). Brain imaging is critical if there is a
is relatively safe to perform, but specific contraindications concern for intracranial extension and is best accomplished
include pacemakers and other implanted metallic devices or with contrast-enhanced MRI. If the patient is unable to tol-
metallic foreign bodies. The risks and benefits of MRI should erate gadolinium contrast, noncontrast MRI augmented with
be weighed carefully in pregnant patients. Studies have not contrast-enhanced CT of the head and sinuses is recommended.
proven any negative effects of MRI to the fetus, but the Immunocompromised patients with acute or subacute rhino-
American College of Radiology recommends deferring MRI sinusitis are at high risk for developing intracranial or orbital
until after pregnancy if possible (1). Because MRI acquisi- complications, and therefore the threshold for obtaining imag-
tion time is much longer than CT, MRI may not be feasible ing should be lower than for immunocompetent patients.
in critically ill patients who require intensive monitoring.
Additionally, if a patient is unable to lie still, motion artifact
may significantly degrade the images obtained.
Gadolinium contrast agents improve the sensitivity of
PYOGENIC INFECTIONS
MRI but are generally avoided in patients with severe renal
dysfunction and a glomerular filtration rate of less than Meningitis
30 mL/min/1.73 m2 due to the risk for nephrogenic systemic
fibrosis (2). Administration of gadolinium-based contrast Meningitis refers to inflammation of the pia and arachnoid
should be avoided in pregnancy due to the unknown effects of membranes. LP with CSF analysis is the test of choice for diag-
exposure to free gadolinium ions on the developing fetus (1). nosis (6), and imaging plays an ancillary role.

24

Scheld_Ch03.indd 24 2/21/14 6:24 PM


Chapter 3: Imaging of Intracranial Infections 25

FIGURE 3.1 Pyogenic (Streptococcus pneumoniae) meningitis. Contrast-enhanced CT (A) shows lepto-
meningeal enhancement (white arrows). MRI FLAIR sequence (B) shows subarachnoid hyperintensity
(black arrows).

Meningitis may be classified based on the pattern of involve- Imaging is also helpful to assess for complications of men-
ment. Pyogenic and viral meningitis typically involve the ce- ingitis, including extensive cerebral edema resulting in brain
rebral cortices. Granulomatous or chronic meningitis typically herniation, infarcts, hydrocephalus, extraaxial pus collections,
involves the basal surfaces of the brain (basilar meningitis) and ventriculitis, cerebritis, and cerebral abscess.
may be due to infectious or noninfectious causes. Pyogenic in- Sterile subdural effusions occur with meningitis but, unlike
fections may also produce a basilar pattern of meningitis. infected extraaxial collections, generally resolve spontaneously.
The primary goals of imaging are to evaluate for contrain- Sterile subdural effusions may develop internal membranes or
dications to LP and to exclude unexpected clinical mimics or septations and occasionally become infected, resulting in sub-
complications. NCCT can satisfy these goals, but not all patients dural empyema (SDE) (13). Subdural effusions occur more
with suspected meningitis require CT. Reported risk factors for often in children with bacterial meningitis and typically de-
an abnormal CT in patients with suspected bacterial meningitis velop over the frontal and temporal lobes (13). Sterile effusions
include age 60 years or older, immunocompromise, recent sei- appear similar to CSF in density or signal intensity or may be
zure, focal neurologic deficits, and impaired consciousness (7). In mildly proteinaceous, resulting in slight signal hyperintensity
the absence of these clinical indicators, CT may not be necessary. on FLAIR compared to CSF. They may be mistaken for promi-
Five percent of patients with acute bacterial meningitis suffer nent subarachnoid spaces, a normal finding in infants. One
brain herniation, and herniation accounts for 32% of deaths (8). distinguishing feature is the finding of bridging vessels crossing
A causal relationship between LP and brain herniation has not the collections, which are present in prominent subarachnoid
been proven, but generally accepted imaging contraindications spaces but not subdural effusions (14).
to LP include midline shift, effacement of the basilar cisterns, and Hydrocephalus is a potentially life-threatening complica-
posterior fossa mass effect (8, 9). Clinical signs of increased intra- tion of meningitis resulting from impaired resorption of CSF
cranial pressure are also a contraindication to LP. by the arachnoid granulations or by diminished CSF outflow
Imaging is usually normal in cases of bacterial meningi- due to viscous material in the ventricles or basilar cisterns.
tis (10). Imaging findings supporting the diagnosis include Hydrocephalus may be the only imaging finding in patients
cerebral edema, inflammatory material in the subarachnoid with meningitis, particularly those with basilar meningitis
spaces, and leptomeningeal enhancement. Cerebral edema (Fig. 3.2). In some cases, proteinaceous or enhancing material
manifests as narrowed or compressed sulci, ventricles, and may also be evident in the basilar cisterns on CT (Fig. 3.2A)
basilar cisterns. Inflammatory material in the subarachnoid or MRI (Fig. 3.2B).
spaces manifests as hyperdense or enhancing material on CT
(Fig. 3.1A) and abnormal fluid-attenuated inversion recovery
(FLAIR) signal hyperintensity (Fig. 3.1B), enhancement, or Cerebritis
restricted diffusion (11) on MRI. Leptomeningeal enhance-
ment manifests as thin, linear enhancement extending along Cerebritis refers to focal brain inflammation due to any cause,
the sulci and basilar cisterns in a gyriform pattern (12). including pyogenic infection (15). Unlike meningitis, which is
Leptomeningeal enhancement should not be confused with localized to the pia and arachnoid, cerebritis involves the brain
pachymeningeal enhancement, which refers to enhancement parenchyma and may occur adjacent to infected subdural or
of the dura mater. epidural collections.

Scheld_Ch03.indd 25 2/21/14 6:24 PM


26 Part I: Approach to the Patient and Diagnostic Evaluation

FIGURE 3.2 Pyogenic basilar meningitis. NCCT (A) demonstrates hydrocephalus, with enlarged temporal
horns of the lateral ventricles (white arrow). Additionally, there is subtle effacement of basilar cisterns,
including the interpeduncular fossa (arrowhead). Contrast-enhanced MRI (B) shows corresponding en-
hancement in the interpeduncular fossa (arrowhead).

Cerebritis has a nonspecific imaging appearance. CT find- Regardless of etiology, abscesses share common imaging
ings include focal low attenuation (16) (Fig. 3.3A) without features. On NCCT, abscesses appear as hypodense masses
enhancement or with nodular or peripheral enhancement, with surrounding vasogenic edema, sometimes with a hyper-
which may resemble infarct or mass lesion. MRI findings in- dense rim corresponding to the abscess capsule (16) (Fig. 3.4A).
clude hyperintensity on T2 and FLAIR (Fig. 3.3B) with vari- MRI also shows a localized mass with a T2-hyperintense ne-
able enhancement (Fig. 3.3C). There may be hemorrhage or crotic core and a markedly T2-hypointense rim surrounding
restricted diffusion. MRI appearance may be similar to that the collection that corresponds to the capsule (22) (Fig. 3.4B).
seen in status epilepticus, ischemia, or neoplasm. The central core demonstrates markedly restricted diffusion
If inadequately treated, cerebritis may develop into a cere- (hyperintense on diffusion-weighted image [DWI] sequence
bral abscess, following a well-described progression through [Fig. 3.4C] and dark on apparent diffusion coefficient [ADC]
the stages of early cerebritis, late cerebritis, early abscess for- maps). Quantitative ADC values of the necrotic core are signifi-
mation, and late abscess formation (16,17) (Fig. 3.3C and D). cantly lower for abscesses than necrotic neoplasms (23).
Features suggesting the formation of an abscess within an area On both CT and MRI, cerebral abscesses demonstrate thick
of cerebritis include development of a ring-enhancing mass smooth rim enhancement, sometimes with thinning of the me-
with restricted diffusion of the central cystic or necrotic core. dial wall. Focal wall rupture results in formation of a daugh-
This is in contrast to the restricted diffusion that may be seen ter abscess (22) (Fig. 3.4D). In immunocompromised patients,
with cerebritis, which affects the brain parenchyma itself. ring enhancement may be absent and vasogenic edema may be
mild (15), requiring a high index of suspicion for diagnosis.
Complications of cerebral abscess include mass effect and
Abscess brain herniation. It is important to evaluate for intraventricu-
lar rupture of the abscess with resulting ventriculitis because
Cerebral abscess refers to a focal pus collection within brain this is a marker of poor prognosis that requires aggressive
parenchyma with a surrounding capsule (15). Abscesses may treatment (21). Imaging findings indicating ventricular rupture
result from direct extension of local infection or from hema- include layering debris in the lateral ventricles and enhance-
togenous spread. Local infections associated with cerebral ment of the ependymal lining.
abscesses include otomastoiditis, sinusitis, and odontogenic The differential diagnosis of a ring-enhancing mass includes
infections (18). Bloodborne infections may be associated high-grade glial neoplasm, metastasis, and less commonly,
with intravenous drug use, bacterial endocarditis, pulmonary tumefactive demyelination or subacute infarction. Usually, the
infections, pulmonary arteriovenous malformations, congeni- clinical scenario helps distinguish these entities, but in difficult
tal heart disease, and other causes (1821). Cerebral abscesses cases, MRP and MRS may be helpful.
may also occur after trauma or neurosurgical intervention. MRP allows comparison of the cerebral blood volume of
Abscesses from bloodborne infections tend to be multiple the lesion with that of contralateral normal white matter, re-
and located at the graywhite junction, most commonly in sulting in a measure of relative cerebral blood volume (rCBV).
the frontal lobes (1821). Abscesses arising from local spread The enhancing component of high-grade tumors demonstrates
are often spatially related to the primary infection. For ex- elevated rCBV (increased perfusion) compared to normal
ample, a frontal abscess may develop adjacent to frontal si- white matter, whereas pyogenic abscesses demonstrate signifi-
nusitis. In these cases, the primary infection is usually visible cantly reduced rCBV, which is less than that of contralateral
on imaging. normal white matter (23,24). It is important to evaluate the

Scheld_Ch03.indd 26 2/21/14 6:24 PM


Chapter 3: Imaging of Intracranial Infections 27

FIGURE 3.3 Cerebritis progressing to abscess. NCCT (A) and MRI (B, C) on presentation shows focal
parenchymal abnormality with decreased density on CT and increased FLAIR signal involving gray and
white matter (white arrows), with ill-defined central enhancement (C) (white arrowhead). Despite treat-
ment, contrast-enhanced MRI 2 weeks later demonstrated rim-enhancing abscess (D) (black arrowhead).
Notice also development of leptomeningeal enhancement (black arrow).

Scheld_Ch03.indd 27 2/21/14 6:24 PM


28 Part I: Approach to the Patient and Diagnostic Evaluation

FIGURE 3.4 Pyogenic (Streptococcus milleri) abscess. NCCT (A) shows right frontal mass with slightly
hyperdense rim (white arrow) and surrounding vasogenic edema. MRI T2 sequence (B) shows hypointense
rim corresponding to abscess capsule (black arrow) and extensive surrounding edema. C: Central necrotic
core demonstrates restricted diffusion (DWI). Postcontrast T1 sequence (D) shows thick rim enhancement
with daughter abscess (white arrowhead).

Scheld_Ch03.indd 28 2/21/14 6:24 PM


Chapter 3: Imaging of Intracranial Infections 29

enhancing rim of these lesions rather than the central necrotic Cranial epidural abscesses appear on NCCT as hypo- or
regions, which do not statistically differ. isodense extraaxial collections in the epidural space (33) that
MRS evaluates the presence and relative ratios of metab- may contain gas (Fig. 3.6A). On MRI, epidural abscesses are
olites present in a region of interest. Metabolites commonly T2 hyperintense and T1 iso- or hypointense relative to brain,
evaluated include choline, a marker of cell membrane turn- depending on the viscosity of the infectious material. There is
over; N-acetylaspartate (NAA), a marker of neuronal integ- usually intense enhancement of the dura (Fig. 3.6B).
rity; lactate, a marker of anaerobic metabolism; and lipid, a Coexistence of subdural and epidural collections is com-
byproduct of necrosis. Creatine, a marker for energy metabo- mon, and differentiating them is sometimes difficult, particu-
lism, is often used as an internal control against which other larly by CT (31,34). MRI may help by allowing delineation
metabolite peaks are compared. In the central necrotic core of of the thickened or enhancing dura with respect to the col-
both tumors and abscesses, the metabolite peaks of NAA, cho- lection (34). Additionally, like epidural hematomas, epidural
line, and creatine are all depressed or absent (25). In brain ab- abscesses tend to be lentiform and can freely cross dural reflec-
scesses, additional metabolite peaks may be present, including tions such as the falx but are bounded by the cranial sutures.
amino acids, alanine, acetate, succinate, and lactate/lipid (26).
Although lactate and lipid may be present in necrotic tumors,
the other metabolites are more specific to abscesses (23,25,27). SPECIFIC ENTITIES
Treatment alters the metabolite profile of the central abscess
cavity and must be considered in the evaluation.
Tuberculosis
Tuberculous meningitis typically involves the basilar cisterns,
Ventriculitis which become filled with a thick inflammatory exudate (35).
NCCT findings may be subtle and hydrocephalus may be the
Ventriculitis may result from intraventricular rupture of an only finding, although isodense material in the basilar cisterns
abscess, severe pyogenic meningitis (13), or as a complica- may be evident. Contrast-enhanced CT may demonstrate
tion after ventricular drainage procedures. Imaging findings enhancing material in the basilar subarachnoid spaces (36),
include layering debris within the ventricles, which appears which may involve the pachymeninges. However, the absence
hyperdense on CT, hyperintense on FLAIR, and hypointense of basilar meningeal enhancement should not preclude the
on T2 compared to CSF (28). Infected ventricular debris may diagnosis (37). Additional findings that support the diagno-
also show markedly restricted diffusion, similar to the central sis include infarcts and tuberculomas (36,37). HIV patients
core of an abscess. Hydrocephalus is usually present, and there with tuberculous meningitis are more likely to have tuber-
may be enhancement of the ependymal lining. Ventricular sep- culomas and infarcts rather than basilar enhancement or
tations may develop as a late sequela of ventriculitis (28). hydrocephalus (35).
MRI is useful in the assessment for tuberculous meningitis
and allows better visualization of basilar meningeal enhance-
Subdural Empyema ment (Fig. 3.7A), infarcts, pachymeningeal involvement, and
tuberculomas than CT (38). MRI may also show enhancement
SDE refers to an infected subdural collection, occurring of cranial nerves.
between the dura and arachnoid membranes. SDE is often as- Tuberculomas are punctate or large granulomatous lesions
sociated with direct spread of infection from sinusitis or otitis that have a variable appearance depending on the extent of cen-
media but also occurs as a complication of meningitis, trauma, tral caseation (39). On CT, tuberculomas may appear target-
or neurosurgical procedures. Seeding of subdural effusions in like with central calcification surrounded by an enhancing rim.
infants with meningitis, or seeding of subdural hematomas, On MRI, tuberculomas may be uniformly T2 hypointense or
also leads to SDE (29,30). Early recognition of SDE is vital be- may appear target-like, with a T2-hyperintense core surrounded
cause urgent surgical decompression is usually required (31). by a low T2 rim (39). Enhancement may be solid, nodular, or
On CT, SDE appears as a hypo- or isodense crescentic ringlike.
subdural collection (Fig. 3.5A) with rim enhancement (31), Tuberculous abscesses are encapsulated masses contain-
which may be subtle. On MRI, SDE appears as a protein- ing pus and viable mycobacteria that occur more commonly
aceous subdural collection, hyperintense on T1 and FLAIR in immunocompromised patients (40). Unlike tuberculo-
relative to CSF (Fig. 3.5B). This is in contrast to a subdural mas, tuberculous abscesses are not primarily granulomatous
effusion, which follows CSF signal intensity on all sequences. (35). By imaging, differentiating a tuberculoma with central
SDEs are usually hyperintense on DWI (11) (Fig. 3.5C) and caseation from a tuberculous abscess is difficult. Tuberculous
demonstrate rim enhancement (Fig. 3.5D), similar to other pus abscesses also resemble pyogenic abscesses on conventional
collections. Sterile subdural effusions may demonstrate mild imaging, with rim enhancement and a central necrotic core
rim enhancement but do not typically demonstrate restricted demonstrating restricted diffusion. MRS may help differenti-
diffusion (32). Complications of SDE include dural venous ate a tuberculous abscess from pyogenic abscess or necrotic
sinus thrombosis (Fig. 3.5D), cerebral edema, cerebritis, and tumor. Metabolites specifically associated with tuberculous
cerebral abscess (Fig. 3.5B and C). abscesses include high lipid and lactate peaks (Fig. 3.7B).
Unlike pyogenic abscesses, amino acid, succinate, acetate, and
alanine peaks are absent (25,26).
Epidural Abscess
Epidural abscesses are usually the result of direct extension of Lyme Disease
adjacent infections, particularly sinusitis or otomastoiditis (33),
but may also occur after trauma (30) or neurosurgical proce- Lyme neuroborreliosis refers to central nervous system (CNS)
dures. Infected material collects between the dura and calvarium. involvement of Lyme disease. Although no specific imaging
Epidural abscesses may extend to involve the subdural space findings exist, neuroborreliosis is often included in the dif-
and may be associated with cerebritis or cerebral abscesses. ferential diagnosis for nonspecific white matter lesions and

Scheld_Ch03.indd 29 2/21/14 6:24 PM


30 Part I: Approach to the Patient and Diagnostic Evaluation

FIGURE 3.5 Subdural empyema. NCCT (A) shows isodense left hemispheric subdural collection (white
arrows). MRI FLAIR sequence (B) shows collection is hyperintense to CSF (white arrows) and is associated
with cerebritis (white arrowhead), and these areas demonstrate restricted diffusion (C). Contrast-enhanced
MRI (D) confirms rim-enhancing collection (arrows) with collection deep to the enhancing dura (black
arrowhead), confirming subdural location. Note also the left sigmoid sinus thrombosis (black arrowhead)
complicating the subdural empyema.

Scheld_Ch03.indd 30 2/21/14 6:24 PM


Chapter 3: Imaging of Intracranial Infections 31

FIGURE 3.6 Epidural abscess complicating maxillary sinusitis. Contrast-enhanced CT (A) shows left
maxillary sinus mucosal thickening and opacification (asterisk) with orbital subperiosteal rim-enhancing
collection (arrow) and intracranial extraaxial gas-containing collection (arrowhead). Contrast-enhanced
MRI (B) redemonstrates sinus disease (asterisk), subperiosteal abscess (black arrow), and enhancing
extraaxial collection (arrowhead), which can be localized to the epidural space by presence of the
overlying enhancing dura.

may share overlapping features with multiple sclerosis. Most Syphilis


patients with neuroborreliosis appear normal on MRI. When
imaging abnormalities exist, leptomeningeal enhancement and Neurosyphilis presents clinically in several discrete phases,
cranial nerve root enhancement may be equally as common as with variable imaging findings in each stage. A high index
white matter lesions (41). The seventh cranial nerve is most of clinical suspicion is needed for diagnosis. Imaging may
commonly involved, followed by the third and fifth cranial be normal or may show cerebral atrophy, nonspecific white
nerves (42). matter lesions, parenchymal masses, or vascular complications

FIGURE 3.7 Tuberculosis: Postcontrast T1-weighted MRI (A) shows thick basilar enhancement (white
arrowhead) and enlarged temporal horns of the lateral ventricles, indicating hydrocephalus. MRS (B) of
the central necrotic core of a tuberculous abscess (white arrow) (yellow box) shows a marked lipid peak
(black arrowhead) and relative paucity of other metabolites.

Scheld_Ch03.indd 31 2/21/14 6:24 PM


32 Part I: Approach to the Patient and Diagnostic Evaluation

TA B L E 3 . 1
PARENCHYMAL NEUROCYSTICERCOSIS:
IMAGING FINDINGS BY STAGE

Stage Description Imaging

Vesicular Cyst or cluster of Thin-walled cyst


cysts with scolex containing simple
fluid; no edema;
scolex may be
visible.
Colloidal Cyst degenerates; Cyst may become
vesicular incites dense on CT, T1
inflammatory hyperintense on
response MRI, with ring
enhancement;
vasogenic edema
Granular Cyst retracts; edema Cyst becomes
nodular begins to ebb. more solid and
smaller; decreasing
enhancement and
edema
Calcified Chronic Calcified nodules
nodular without edema
or enhancement;
FIGURE 3.8 Neurosyphilis. Postcontrast coronal T1-weighted MRI nodules dark on T2
shows left posterior parietal edema (dark area) (arrowhead), which is or gradient-echo
associated with parenchymal, leptomeningeal, and dural enhancement sequences.
(white arrow). (Case provided courtesy of James Fink, MD.)

including infarcts (43,44). T2/FLAIR-hyperintense lesions colloidal vesicular stage, the cyst degenerates, becomes pro-
measuring less than 1 cm occur in the deep periventricular and teinaceous, and the scolex may disappear (Fig. 3.9AC). The
subcortical white matter (43,44). granular nodular stage occurs as the cyst retracts. Vasogenic
Syphilitic gummas are granulomatous lesions of the menin- edema lessens, although nodular or ring enhancement per-
ges that subsequently involve brain parenchyma or dura or sists. Differentiating the colloidal vesicular stage from the
both and that may contain Treponema pallidum. Gummas are granular nodular stage may be difficult (47). Finally, dur-
hypodense on CT and T1 hypointense and T2 hyperintense ing the nodular calcified stage, edema and enhancement
on MRI, with mass-effect, enhancement, and surrounding va- subside, leaving a small calcified nodule (Fig. 3.9D). MRI
sogenic edema (44,45) (Fig. 3.8). Dural thickening indicates is best for identifying lesions in the vesicular, colloidal ve-
dural involvement. Gummas typically occur in the cerebral sicular, and granular nodular stages. CT is excellent at de-
hemispheres but may also appear in unexpected locations, tecting lesions in the nodular calcified stage (47), as is MR
such as the pituitary gland (45). gradient-echo sequence.
Meningeal syphilis may manifest as meningeal enhancement Intraventricular neurocysticercosis may occur alone or in
on CT or MRI. Cranial nerve enhancement may also occur, conjunction with parenchymal neurocysticercosis. Imaging
often involving cranial nerves VII and VIII (46). Syphilitic vas- findings include cystic lesions within the ventricles, commonly
culitis (meningovascular syphilis) may affect medium and large in the fourth ventricle (47). There may be associated noncom-
vessels (Heubner arteritis) or small vessels (Nissl-Alzheimer municating hydrocephalus. Cysts usually are thin walled and
type) (46). Infarctions can complicate either type. Angiographic contain CSF-like fluid, making them difficult to see on stan-
findings of infectious vasculitis are discussed further later in dard MRI sequences. High-resolution heavily T2-weighted
this chapter. MRI sequences (MR cisternography) may help to delineate the
cyst walls.
Neurocysticercosis may also involve the subarachnoid spaces,
Neurocysticercosis particularly the basilar cisterns, and appears as multilobular cys-
tic lesions (racemose neurocysticercosis) (Fig. 3.9E). The scolex
Neurocysticercosis results from CNS invasion by the parasitic is often not visible.
organism Taenia solium and may involve brain parenchyma,
ventricles, or subarachnoid spaces. The imaging appearance
varies with location and stage of infection. Creutzfeldt-Jakob Disease
Parenchymal neurocysticercosis can be classified into
four stages from acute to chronic (47), as summarized in Creutzfeldt-Jakob disease (CJD) is a prion disease that may
Table 3.1. In the vesicular stage, a thin-walled cyst forms be sporadic, hereditary, or acquired from exposure to infected
containing the invaginated scolex, which may be visible on CNS tissue. Variant CJD (vCJD) most commonly occurs
FLAIR and contrast-enhanced sequences (48). During the after consumption of meat from cows infected with bovine

Scheld_Ch03.indd 32 2/21/14 6:24 PM


Chapter 3: Imaging of Intracranial Infections 33

FIGURE 3.9 Neurocysticercosis. Colloidal vesicular phase on CT (A), FLAIR (B), and postcontrast T1
MRI (C) demonstrating central scolex (black arrows), thick rim enhancement (black arrowhead), and
surrounding vasogenic edema. D: Calcified nodular phase by CT (arrowheads). Subarachnoid neurocys-
ticercosis on T1 MRI (E), with multiple cysts (arrowheads) causing distortion of adjacent parenchyma.
(Cases provided courtesy of James Fink, MD.)

spongiform encephalopathy. Kuru occurs with cannibalism. (Fig. 3.10B). The precentral gyrus is usually spared (51).
Iatrogenic CJD occurs with surgical exposures such as corneal Globus pallidus, thalamus, and periaqueductal gray matter
transplantation. Most cases of CJD are sporadic (sCJD) (49). may be involved (49). Cortical atrophy occurs with disease
The imaging appearances of sCJD and vCJD have been best progression.
described. vCJD is associated with the pulvinar signsymmetric
MRI is the preferred imaging modality (49) to sup- FLAIR signal hyperintensity in the pulvinar nuclei of the
port the diagnosis and exclude other etiologies. Criteria thalamus (52). The dorsal medial nucleus of the thalamus may
to support the diagnosis of sCJD include DWI or FLAIR also be hyperintense (hockey stick sign) (49,52), as may be
signal hyperintensity in the caudate nucleus and putamen the tectal plate, periaqueductal gray matter, or cerebral cortex.
(Fig. 3.10A), with involvement of at least one cortical Cortical atrophy may occur but is usually less severe than with
gyrus or involvement of more than three cortical gyri (50) the sCJD.

Scheld_Ch03.indd 33 2/21/14 6:24 PM


34 Part I: Approach to the Patient and Diagnostic Evaluation

FIGURE 3.10 Sporadic Creutzfeldt-Jakob disease in two patients. DWI MRI (A) demonstrates restricted
diffusion in the caudate and anterior putamen (arrowhead). Cortex was also involved (not shown).
Predominantly cortical pattern of involvement in a second patient (B) showing restricted diffusion affect-
ing more than three gyri. (Case provided courtesy of James Fink, MD.)

VIRAL INFECTIONS and treatment are critical to limit morbidity and mortality.
Although all patients suspected of having HSV encephalitis
Viral infections of the CNS manifest variably as meningitis, en- should receive prompt treatment regardless of imaging find-
cephalitis, myelitis, radiculitis, postinfectious encephalomyeli- ings, it is important to recognize the characteristic imaging
tis, or various combinations thereof. Unlike most bacterial and features because these may precede clinical suspicion of the
fungal infections, the intracranial imaging findings in viral en- disease.
cephalitis may predict a specific causative organism (Table 3.2). Infection usually begins in the anterior and medial aspects
of the temporal lobe(s) but may extend to the lateral temporal
lobes, inferior frontal lobes, insular cortex, and frontal and
Viral Meningitis parietal cingulate gyri. Findings may be unilateral or bilateral.
Extension to the pons may occur through retrograde viral
Uncomplicated viral meningitis is usually diagnosed by clini- spread along the trigeminal nerve (55). CT may be normal
cal presentation combined with CSF evaluation. Enteroviruses initially or may show low attenuation in the affected regions,
are the most frequent cause (53). Imaging may be normal or sometimes with associated mass effect, gyral enhancement,
may show cortical leptomeningeal enhancement on contrast- or petechial hemorrhage (56,57) (Fig. 3.11A). MRI is more
enhanced CT or MRI (12). Viral meningitis is less likely than sensitive for early disease and better demonstrates the edema-
bacterial, fungal, or tuberculous meningitis to produce FLAIR tous changes as T2/FLAIR-hyperintense areas (Fig. 3.11B)
signal hyperintensity in the subarachnoid space (54), but this with concomitant decreased T1 signal intensity (55,58). DWI
finding is not reliable for distinguishing the entities. may show restricted diffusion (Fig. 3.11C), which may pre-
cede findings on other sequences (5961). Hemorrhages occur
with disease progression and are demonstrated with high
Herpes Simplex Virus sensitivity on MRI as petechial areas of intrinsic T1 signal
hyperintensity or as susceptibility artifact on gradient-echo se-
Herpes simplex virus (HSV) type 1 is the most common quences. Variable enhancement may also develop at this stage.
cause of sporadic acute viral encephalitis, and early diagnosis Progressive encephalomalacia occurs over several weeks, often

Scheld_Ch03.indd 34 2/21/14 6:24 PM


Chapter 3: Imaging of Intracranial Infections 35

TA B L E 3 . 2
CHARACTERISTICS OF VIRAL ENCEPHALITIS BY ENTITY

Viral Infection Pearls

Enteroviruses Normal or thin linear leptomeningeal enhancement

HSV Bilateral medial temporal and inferior frontal lobes, sparing basal ganglia
/ Abnormal DWI, enhancement, hemorrhages
Mass effect initially, followed by atrophy over several weeks

VZV Lesions at graywhite interface, white matter, gray matter


Vascular imaging may show vasculitis

Arboviruses Bilateral basal ganglia and thalami lesions


Can involve any brain area, including medial temporal lobes

ADEM Large, asymmetric lesions in supratentorial white matter


/ Deep gray nuclei, brainstem, spinal cord, optic nerves
Open ring sign of enhancement
Monophasic, with resolution on follow-up imaging

HIV Generalized cerebral atrophy


Symmetric periventricular white matter FLAIR hyperintensity with T1
isointensity

PML Asymmetric white matter signal abnormality (hyperintense T2,


hypointense T1) involving subcortical U-fibers
Cerebellar crescent-shaped lesions
Minimal mass effect and enhancement, except with IRIS
May see leading edge of abnormal DWI and enhancement
Progresses on follow-up; may lead to atrophy long-term

CMV Periventricular enhancement or calcification

Noteworthy features highlighted in bold text.

leading to marked temporal lobe atrophy (Fig. 3.11D) with aneurysm formation, and arterial dissection (66). Parenchymal
associated seizure disorder. lesions in VZV infection characteristically occur at the graywhite
The differential diagnosis includes infarct, glioma, limbic interface but may also occur in the cortical gray matter and deep
encephalitis (paraneoplastic syndrome), Rasmussen encephali- white matter. Multiple lesions at the graywhite interface should
tis (chronic viral encephalitis), and other viral infections, such specifically suggest VZV vasculopathy in the right clinical setting,
as arboviral encephalitis. Bilateral abnormalities, sparing of along with the differential diagnosis of emboli and metastases.
the basal ganglia, and involvement of both medial and lateral VZV vasculopathy may coexist with meningitis, radiculitis, and
portions of the temporal lobes (posterior cerebral artery [PCA] myelitis and may occur with or without rash (66).
and middle cerebral artery [MCA] vascular territories) are all
features that increase the specificity for HSV.
Advanced techniques are not typically required for diagnosis Arboviruses
but may be helpful for problem solving. Both CTP (62) and SPECT
(63) show hyperperfusion of the involved areas acutely. MRS may Arthropod-borne viruses, also known as arboviruses, consti-
show decreased NAA and increased choline levels acutely, which tute an important cause of viral meningoencephalitis world-
can mimic neoplasm (64). Follow-up conventional MRI usually wide. Examples from this diverse group include the viruses
distinguishes these entities however, because HSV leads to atro- causing Eastern equine, Western equine, Venezuelan equine,
phy, whereas infiltrating glioma persists or progresses (64,65). West Nile, Japanese, St. Louis, California, Murray Valley, and
tick-borne encephalitides. Affected patients may have normal
MRI findings or may have signal abnormalities on T2, FLAIR,
Varicella-Zoster Virus or DWI. Classically, lesions are located in the basal ganglia and
thalami bilaterally (Fig. 3.12), and this imaging pattern should
Neurologic manifestations of varicella-zoster virus (VZV) may strongly suggest arboviral encephalitis in a potentially exposed
occur in the setting of primary infection (chickenpox) or reacti- patient. The differential diagnosis includes anoxic or hypoxic
vation (shingles). Reactivation is usually associated with immu- encephalopathy, toxic exposures such as carbon monoxide
nosuppression or normal age-related declining immunity. VZV poisoning, metabolic disorders such as Wilson disease and
infection is distinct among viral infections in that it causes a vas- mitochondrial abnormalities, and other entities such as CJD.
culopathy, which may involve either small or large vessels. Small The basal ganglia and thalami are classically involved
artery involvement may lead to monocular visual loss. Large artery in arboviral infections, but additional nonspecific areas of
involvement classically leads to ischemic infarctions. Other mani- involvement include the meninges, the brainstem and spinal
festations of vasculopathy include subarachnoid hemorrhage, cord, the cortical gray matter, and the cerebral and cerebellar

Scheld_Ch03.indd 35 2/21/14 6:24 PM


36 Part I: Approach to the Patient and Diagnostic Evaluation

FIGURE 3.11 HSV encephalitis. A: NCCT shows left temporal low density and volume expansion with
petechial areas of hemorrhage (arrowhead). B: Coronal FLAIR MRI in a different patient shows bilateral
involvement of the anterior and medial temporal lobes and insula. C: DWI shows restricted diffusion in
the corresponding areas, confirmed on ADC map (not shown). D: Axial NCCT in a third patient with
prior HSV encephalitis shows left temporal lobe atrophy.

white matter (65,67,68). Leptomeningeal or parenchymal abruptly 2 to 3 weeks following a viral illness or vaccination.
enhancement is variable. Isolated substantia nigra lesions have In contrast to multiple sclerosis, ADEM follows a monophasic
been reported with St. Louis encephalitis (69). Arboviral en- course lasting several weeks.
cephalitis commonly involves the mesial temporal lobes, but CT may be normal or may show nonspecific areas of low
involvement of the basal ganglia and thalami with relative attenuation (Fig. 3.13A). Lesions may show peripheral en-
sparing of the anterior portions of the temporal lobes help to hancement (7072). MRI is more sensitive, but lesions may not
distinguish arboviral infections from HSV (65). be visible until several days after the onset of symptoms (73).
Like other demyelinating lesions, ADEM lesions have high T2
and FLAIR and low T1 signal intensity. They are located most
Acute Disseminated Encephalomyelitis commonly in the supratentorial white matter (Fig. 3.13B)
and are often multiple, bilateral, and asymmetric, although
Acute disseminated encephalomyelitis (ADEM), also known as involvement of the deep gray structures may be symmetric (74).
postinfectious or postvaccination encephalitis, is an inflamma- Lesions may be small and round or large and irregular, some-
tory autoimmune demyelinating condition that typically begins times with a central T2-hyperintense portion creating a fried

Scheld_Ch03.indd 36 2/21/14 6:24 PM


Chapter 3: Imaging of Intracranial Infections 37

egg appearance (75). Mass effect and surrounding edema


are typically absent. Tumefactive demyelinating lesions and
rare hemorrhagic forms of ADEM may show surrounding
edema (74,75). A peripheral incomplete ring of enhancement,
the open ring sign, may occur in lesions that partially abut
white matter and cortex (Fig. 3.13C). In this situation, only the
white matter edge enhances, a sign that is highly specific for
demyelinating lesions and only rarely occurs with neoplasms
or abscesses (76,77). Other patterns of enhancement observed
in ADEM include closed ring, solid, nodular, or gyral enhance-
ment (74,75). Patterns of signal abnormality on DWI are vari-
able (78,79), and single lesions may have heterogeneous signal
intensity on DWI and ADC map (80).
MRS shows selective reduction of NAA initially, with nor-
mal levels of the other metabolites. NAA returns to normal
levels on follow-up imaging (81). Elevated choline and/or lac-
tate may also be observed (79,82) (Fig. 3.13D), resembling
other demyelinating conditions and brain tumors. MRP usu-
ally reveals hypoperfusion (Fig. 3.13E) (80), which may help
to distinguish ADEM from neoplastic lesions.
ADEM lesions usually either resolve or improve on follow-
up imaging, although sometimes there is residual gliosis.
Clinical resolution may precede radiographic resolution (73).
FIGURE 3.12 Eastern equine encephalitis. Symmetric hyperintense FLAIR
A subset of patients may have a relapsing form of the disease
signal in the bilateral basal ganglia, including caudate, putamen, and glo-
bus pallidus. Bilateral thalamic involvement is also typical but not present resembling multiple sclerosis. The terms multiphasic or recur-
in this case. (Case provided courtesy of Mahmud Mossa-Basha, MD.) rent ADEM, however, should be reserved for cases in which

FIGURE 3.13 ADEM. NCCT (A) shows multiple large ill-defined areas of low attenuation bilaterally, which
are hyperintense on FLAIR (B). C: Postcontrast coronal T1-weighted image shows open ring signs (arrow-
heads) with incomplete peripheral enhancement abutting the white matter edge. Single voxel short echo time
MRS (D) of a left parietal lesion shows elevated choline:NAA ratio and a lactate peak (black arrows) mimick-
ing neoplasm. MRP (E) showing decreased perfusion in the lesions. Another patient with ADEM isolated to
the posterior fossa (F) demonstrating FLAIR hyperintense lesions in the pons and middle cerebellar peduncle.

Scheld_Ch03.indd 37 2/21/14 6:24 PM


38 Part I: Approach to the Patient and Diagnostic Evaluation

FIGURE 3.14 HIV. NCCT at presentation (A) and 5 years later (B) (obtained after trauma) demonstrate
progressive atrophy and patchy periventricular white matter hypoattenuation. B: Incidental right frontal
contusion and small intraventricular hemorrhage evident related to trauma. FLAIR (C) better shows
confluent periventricular white matter disease.

new MRI lesions develop or recur at least 3 months after the such as HIV, cancers such as leukemia, and medications such
initial demyelinating event and longer than 4 weeks after com- as chemotherapy and immunosuppressive agents, which are
pleting steroid therapy (74). These criteria help to distinguish used following bone marrow or solid organ transplantation to
relapse from monophasic disease with a protracted course or prevent rejection. Immunocompromised patients are prone to
an incomplete response to treatment (83). frequent, severe, and long-lasting CNS infections that may be
Supratentorial white matter lesions are most common in caused by opportunistic pathogens. Neutropenic patients are
ADEM, but lesions may also occur in the deep gray nuclei, in- particularly susceptible to bacterial and fungal infections. The
fratentorial white matter, spinal cord, and optic nerves (71,83). most common CNS opportunistic infections to affect immuno-
Lesions may sometimes be isolated to the infratentorial white compromised patients are discussed below.
matter (Fig. 3.13F).

Progressive Multifocal Leukoencephalopathy


HIV
PML is a progressive and frequently fatal demyelinating oppor-
Patients infected with HIV may have imaging findings related tunistic infection caused by the JC virus, a ubiquitous patho-
directly to HIV infection in the CNS or related to opportunis- gen that causes disease primarily in patients with impaired
tic infections. Associated opportunistic infections include viral T-cell immunity. The JC virus causes demyelination by directly
diseases such as progressive multifocal leukoencephalopathy infecting the myelin-producing oligodendrocytes. Most cases
(PML) and cytomegalovirus (CMV) infections and fungal occur in the setting of HIV infection, hematologic disorders,
infections such as Cryptococcus infection and coccidioidomy- organ transplantation, and treatment with the monoclonal
cosis. Tuberculosis and syphilis are also increasingly prevalent antibody natalizumab for multiple sclerosis or Crohn disease.
in HIV-infected patients. The diagnosis is established by demonstrating JC viral DNA
Direct effects of HIV infection include generalized cerebral in the CSF by polymerase chain reaction or by immunohisto-
volume loss and symmetric patchy or confluent periventricular chemical analysis of brain tissue, but characteristic findings on
white matter low attenuation on CT and signal hyperintensity MRI (Fig. 3.15) may first suggest the diagnosis.
on T2 and FLAIR (Fig 3.14), with corresponding isointensity PML appears as confluent areas of hypoattenuation on CT
on T1-weighted images. There is no contrast enhancement or and signal abnormality on MRI within the subcortical white
mass effect (40). HIV infection may be difficult to distinguish matter involving arcuate or U-fibers, sparing the cortex. MRI
from chronic microvascular ischemia and age-related volume shows confluent areas of T2 and FLAIR hyperintensity with
loss if comparison studies are not available. Symmetry, peri- progressive hypointensity on T1-weighted images and little to
ventricular location, and T1 isointensity help to distinguish no mass effect or contrast enhancement. Scant peripheral en-
imaging abnormalities due to HIV from those due to PML. hancement as well as restricted diffusion is sometimes observed
at the leading edge of demyelination (84,85). More marked
enhancement and mass effect may be observed when PML is
OPPORTUNISTIC INFECTIONS associated with the immune reconstitution inflammatory syn-
drome (IRIS), further discussed below (8688).
Immune system compromise may affect humoral immunity Parietooccipital lobes and corpus callosum are typically
(B cells) or cell-mediated immunity (T cells) and may be due to affected. Unlike multiple sclerosis, periventricular white
a primary immune deficiency or may be secondary to infections matter is relatively spared. Lesions are frequently multiple

Scheld_Ch03.indd 38 2/21/14 6:24 PM


Chapter 3: Imaging of Intracranial Infections 39

FIGURE 3.15 PML in a patient after autologous stem cell transplant. NCCT (A) shows a right frontal
hypodense lesion. MRI redemonstrates lesion (black arrowheads) as an area of subcortical T2 hyperin-
tensity (B) sparing the cortical ribbon. Restricted diffusion (C) and slight contrast enhancement (D) at
the posterior white matter margin corresponds to the leading edge of demyelination. There is hypo-
metabolism evident on FDG-PET (E). JC virus was confirmed by lumbar puncture. Posterior fossa PML
in a separate patient with advanced AIDS (F) shows a crescentic T1-hypointense lesion with minimal
enhancement (white arrow).

and asymmetric bilaterally and become increasingly conflu- The imaging features of PML can resemble ADEM, but
ent with progression to new areas on follow-up imaging. the two entities can usually be distinguished clinically. PML
Infratentorial lesions may occur, sometimes in isolation. occurs in immunocompromised patients and has a subacute
Cerebellar lesions often have a characteristic crescent-shaped onset and progressive course of worsening neurologic im-
morphology (Fig. 3.15) (85,89,90). pairment and lesion enlargement on MRI. ADEM occurs in
Although PML lesions have a fairly characteristic ap- immune competent individuals, usually children, following
pearance on conventional MRI, advanced techniques are a viral infection or vaccination and has an abrupt onset of
sometimes performed. MRS may show elevated choline and neurologic impairment followed by gradual improvement
depressed NAA peaks as well as lipid and lactate peaks (91). clinically and radiologically during the course of steroid
This spectrum of metabolites reflects the underlying demyelin- treatment.
ating process and resembles other demyelinating conditions
such as ADEM but also resembles high-grade neoplasms.
MRP typically shows hypoperfusion in the affected areas, and Cytomegalovirus Infection
SPECT and FDG-PET or PET/CT studies show reduced meta-
bolic activity (Fig. 3.15), helping to differentiate PML from CMV produces two distinct types of disease, one in newborns
lymphoma or a high-grade glioma. related to in utero transmission of maternal infection and one in

Scheld_Ch03.indd 39 2/21/14 6:24 PM


40 Part I: Approach to the Patient and Diagnostic Evaluation

immunocompromised patients. Intracranial imaging findings pyogenic abscesses, there is no central restricted diffusion
of congenital infection include periventricular calcifications, (97). Ring enhancement is common. The eccentric target
migrational abnormalities, atrophy, and leukoencephalopathy sign (Fig. 3.16) refers to an enhancing nodule along the
(55,65,92). In acquired CMV infections that occur in the set- ring-enhancing wall of the lesion, which is highly specific,
ting of immune deficiency, the intracranial imaging findings are but insensitive, for toxoplasmosis (98). Typical abscess
nonspecific but may include findings of meningoencephalitis, locations include the basal ganglia, thalamus, and cerebral
ventriculitis, and/or leukoencephalopathy (40,55,65,92,93). hemispheres at the graywhite junction (40). MRS typically
Rarely, acquired CMV infection manifests as a space-occupying shows a large lipid and lactate peak, and perfusion studies
or peripherally enhancing mass (40,90). Extracranial imaging demonstrate decreased rCBV (98).
findings of acquired CMV infection include chorioretinitis and Differentiating toxoplasmosis from lymphoma in a patient
polyradiculitis. with AIDS remains a clinical conundrum (46). FDG-PET and
The major differential diagnosis of CMV ventriculitis in thallium-201 SPECT have both been used for differentiating
an immunocompromised patient is CNS lymphoma. Tuber- the two entities, but the utility of thallium-201 SPECT is vari-
culosis, toxoplasmosis, and bacterial ventriculitis are addi- able (99101). DWI MRI sequence may sometimes be helpful,
tional considerations. The pattern of contrast enhancement but there is significant overlap of ADC values between toxo-
may help distinguish these entities because smooth linear plasmosis and lymphoma (102). Serial imaging confirming
enhancement favors viral or bacterial ventriculitis, whereas expected response to antibiotic treatment, namely, resolution
nodular or mass-like enhancement favors lymphoma (94). of ring-enhancing lesions, may be the most helpful in diagnos-
Involvement of the corpus callosum also suggests lymphoma ing toxoplasmosis (40).
but can rarely occur with toxoplasmosis (95). Definitive diag-
nosis of CMV ventriculitis requires polymerase chain reaction
analysis for detection of viral DNA in the CSF. Lymphoma is Cryptococcus Infection
usually confirmed by detection of abnormal cells in the CSF
or by biopsy. Cryptococcus CNS infection may affect immunocompe-
tent patients but is more common in immunocompromised
patients. Neuroimaging studies may be normal or may
Toxoplasmosis demonstrate findings of meningitis, meningoencephalitis,
or vasculitis, better depicted with MRI than CT (103).
Cerebral toxoplasmosis usually presents clinically due to Meningitis findings include leptomeningeal enhancement
reactivation of latent infection in immunocompromised pa- and subarachnoid space DWI hyperintensity (11). CNS
tients (46). Toxoplasmosis appears as one or more ring- or cryptococcal infection may also manifest as parenchymal
nodular-enhancing masses, corresponding to abscesses. T2 mass lesion(s) or hydrocephalus.
signal is heterogeneous and may vary according to the stage Cryptococcus infection often results in accumulation of
of abscess formation and treatment effects (96). Unlike gelatinous exudate, which causes dilation of the perivascular
spaces and formation of pseudocysts (Fig. 3.17). These gelatinous

FIGURE 3.16 CNS toxoplasmosis. Contrast-enhanced MR shows FIGURE 3.17 Cryptococcus. T2-weighted image demonstrates nodu-
ring-enhancing lesions, one of which demonstrates eccentric target-like lar lesions in the basal ganglia (black arrowhead) that are hyperintense
enhancement (arrowhead), a relatively specific sign for toxoplasmosis. to brain but hypointense compared to CSF, consistent with gelatinous
Note the second ring-enhancing lesion in the left thalamus (arrow). pseudocysts.

Scheld_Ch03.indd 40 2/21/14 6:24 PM


Chapter 3: Imaging of Intracranial Infections 41

FIGURE 3.18 PML-IRIS. Initial FLAIR MRI (A) shows subtle right frontal hyperintense lesion
(white arrow). Follow-up scan 2 1/2 months later (B) shows interval enlargement of the FLAIR hyper-
intense lesion (white arrow) with mild mass effect. Postcontrast T1 images (C) shows new enhancement
with an open-ring configuration (black arrow), consistent with demyelination.

pseudocysts favor the basal ganglia and may have enhanc- worsening opportunistic infection. The diagnosis is usually
ing walls, particularly in immunocompetent patients (104). suspected when there is paradoxical clinical deterioration
Pseudocysts are hyperintense to CSF on FLAIR due to the pro- with imaging findings that are atypical for a given oppor-
teinaceous contents. Prominent perivascular spaces without tunistic infection. For example, in IRIS-PML (Fig. 3.18),
gelatinous exudate also occur, in concert with cerebral volume there may be greater than expected enhancement or mass
loss, but should demonstrate the same signal characteristics as effect (40,8688,108).
CSF on all sequences.
Cryptococcomas may form, which are either granuloma-
tous lesions with few organisms or inflammatory lesions with CEREBROVASCULAR
many organisms (40). Cryptococcomas appear as a cluster
of nodules that are non- or minimally enhancing in immuno-
COMPLICATIONS OF INFECTION
compromised patients and enhancing in immunocompetent Intracranial vascular complications of infections may be arte-
patients (104). There is no central restricted diffusion, unlike rial or venous and may occur in the setting of meningitis, head
pyogenic abscesses. Most occur in the basal ganglia, thalamus, and neck infections, or systemic infections. Examples include
and cerebellum (40). infectious vasculitis, venous septic thrombophlebitis, septic
emboli, septic (mycotic) aneurysms, and disseminated intra-
vascular coagulation.
Coccidioidomycosis
Coccidioidal meningitis may occur in both immunocompetent
and immunocompromised patients and presents as chronic
Infectious Vasculitis
(basilar) meningitis. Associated hydrocephalus is common Cerebral vasculitis may be primary and idiopathic, known
(105,106). Vasculitis and infarctions may complicate the in- as primary angiitis of the central nervous system (PACNS),
fection (107). Focal-enhancing parenchymal brain lesions may or may be secondary to a variety of systemic vasculitides,
occur in severe disease as a result of direct extension of basilar drugs, or infections. Infectious causes of cerebral vasculitis
meningeal disease (105). The presence of either hydrocepha- are potentially treatable; therefore, patients with known or
lus alone or hydrocephalus with infarction is associated with suspected infections and new neurologic deficits warrant
higher mortality (106). evaluation with brain and cerebrovascular imaging. Vascular
complications of pyogenic bacterial meningitis are common
and outcomes are poor, with a high risk for stroke and asso-
Immune Reconstitution Inflammatory ciated morbidity and mortality (109,110). Diagnosis of infec-
Syndrome tious vasculitis is usually established by characteristic clinical
and radiologic signs combined with CSF analysis using cul-
IRIS is a complication of highly active antiretroviral therapy ture, PCR, and serologic tests directed toward the most com-
(HAART) that occurs in the setting of severe AIDS-related mon pathogens.
immunodeficiency shortly after the initiation of therapy. Pathogens known to cause infectious cerebral vasculitis in-
The syndrome is characterized by an exaggerated inflam- clude bacteria such as Streptococcus pneumoniae (Fig. 3.19A
matory response to dead, latent, or viable organisms or and B), Mycobacterium tuberculosis, and T. pallidum; vi-
self-antigens and may coexist with a variety of opportunis- ruses such as VZV (Fig. 3.19C and D); and a variety of fungi
tic infections, most commonly JC virus and Cryptococcus (Fig. 3.19E and F) and parasites (66,107,111116). Any
(108). The imaging findings might be confused with new or cause of infectious basilar meningitis can potentially lead to

Scheld_Ch03.indd 41 2/21/14 6:24 PM


42 Part I: Approach to the Patient and Diagnostic Evaluation

FIGURE 3.19 Infectious vasculitis. A, B: Pyogenic (Streptococcus pneumoniae). T2-weighted MRI at


presentation (A) and 2 weeks later (B) demonstrate interval decrease in caliber of the basilar artery flow
void (open arrowhead) and new pontine infarcts (white arrow). C, D: Varicella-zoster vasculitis. Cerebral
angiography of the left internal carotid artery in the lateral (C) and PA (D) projection demonstrate
irregular narrowing of the posterior cerebral artery (C) (arrow) and middle cerebral artery (D, arrow).
E, F: Fungal (coccidioidal). Postcontrast T1 image demonstrates extensive basilar and subarachnoid en-
hancement (E, arrowhead) with evidence of subacute infarct in the posterior limb of the left internal
capsule (F, arrow) due to vasculitis affecting the lenticulostriate arteries.

Scheld_Ch03.indd 42 2/21/14 6:24 PM


Chapter 3: Imaging of Intracranial Infections 43

vasculopathy involving the cerebral vessels at the base of the complicate meningitis (13). Septic thrombophlebitis can in-
brain either by inducing vasospasm or by inciting an inflam- volve dural venous sinuses, cavernous sinuses, or cortical
matory reaction within the vessel walls. In VZV vasculitis, veins. High-risk patients include those with diabetes or
there is productive viral infection within the media of cerebral immunosuppression and those presenting with coalescent
vessel walls (66,117). It is controversial whether other viruses mastoiditis or acute bacterial or fungal sinusitis involving
such as HIV also cause cerebral vasculitis, because opportunis- the frontal or sphenoid sinuses (119). Specific patterns of
tic infections frequently coexist (66,115). septic thrombophlebitis include sigmoid dural venous sinus
The vascular imaging findings of infectious vasculitis thrombosis secondary to coalescent mastoiditis, superior
are similar to other vasculitides and include segmental va- sagittal sinus thrombosis secondary to frontal sinusitis, and
soconstriction creating a beads-on-a-string appearance, cavernous sinus thrombophlebitis secondary to sphenoid
irregularities of the vessel wall, smooth vessel narrowing, dis- sinusitis or osteomyelitis (Fig. 3.20), orbital cellulitis, or
sections, occlusions, or aneurysm formation. VZV vasculitis other facial infections.
may show contrast enhancement of the vessel walls, possi- Imaging signs of cavernous sinus thrombosis include fill-
bly reflecting the underlying productive viral infection (114). ing defects, diminished enhancement, or an expanded con-
Leptomeningeal contrast enhancement on CT or MRI is vari- tour of the cavernous sinus (120,121). Indirect findings may
able in basilar meningitis but, if present, may suggest infection include proptosis; enlargement of the extraocular muscles;
as the cause of vasculitis. Complications of vasculitis visible and enlargement, nonenhancement, or filling defects within
on CT or MRI include infarcts and hemorrhages, including the superior ophthalmic vein(s). Occasionally, gas bubbles are
subarachnoid hemorrhage. present in the cavernous sinus due to dehiscence of the sphe-
noid sinus walls related to osteomyelitis. Secondary arterial
complications involving the cavernous internal carotid artery
Septic Thrombophlebitis may occur, including arteritis, thrombosis (Fig. 3.20A), and
aneurysm formation (Fig. 3.20B and C).
Cerebral venous septic thrombophlebitis represents an im- Imaging signs of dural venous sinus thrombosis include a
portant pathway for intracranial spread of extracranial hyperdense venous sinus on NCCT, abnormal flow voids on
infections from the paranasal sinuses, mastoid air cells, noncontrast MRI, absence of the normal flowrelated signal
orbits, and other facial structures (118,119) and may also on noncontrast MRV, and filling defects on contrast-enhanced

FIGURE 3.20 Septic thrombophlebitis and complications in two patients with sphenoid sinusitis.
A: Contrast-enhanced CT demonstrates mild expansion and nonopacification of the right cavernous
sinus (arrowhead) with thrombosis of the right cavernous carotid artery (white arrow). B: Contrast-
enhanced T1 MRI demonstrates patchy nonopacification of the cavernous sinuses bilaterally, with
abnormal left carotid artery flow void (black arrowhead). Cerebral angiogram (C) confirms mycotic
left cavernous carotid aneurysm (black arrowhead).

Scheld_Ch03.indd 43 2/21/14 6:24 PM


44 Part I: Approach to the Patient and Diagnostic Evaluation

computed tomography venography (CTV) or MRV. Findings emboli, septic (mycotic) aneurysms, or complex clotting
on noncontrast MRI or MRV may be subtle in cases of in- disorders such as disseminated intravascular coagulation
complete thrombosis or following partial recanalization. (DIC). Septic emboli may lead to multiple cerebral infarctions,
Obstructive dural venous sinus thrombosis may lead to microhemorrhages, and microabscesses. The imaging find-
secondary complications of venous hypertension and venous ings on NCCT include loss of graywhite differentiation or
infarction, visible on CT or MRI as vasogenic edema, paren- hypoattenuation corresponding to acute infarcts (Fig. 3.21A).
chymal hemorrhages, ischemic or hemorrhagic infarcts, or MRI may show multifocal areas of FLAIR signal hyperinten-
sometimes as isolated convexal subarachnoid hemorrhage. sity or restricted diffusion at the graywhite interface, often
Multiple infarcts or hemorrhages in a nonarterial distribu- associated with small areas of abnormal susceptibility on
tion may suggest the diagnosis. Unlike arterial infarcts, which gradient-echo images (Fig. 3.21B). Contrast-enhanced im-
show restricted diffusion, findings on DWI are variable in ages characteristically reveal multiple peripherally enhanc-
venous infarctions. ing lesions at the graywhite interface, but this finding is
Cortical vein thrombosis may be difficult to detect by imag- sometimes absent, especially early in disease. Multifocal sub-
ing. The characteristic finding on NCCT is the cord signa arachnoid hemorrhage isolated to the cerebral convexities is
hyperdense, serpiginous cortical structure corresponding to the another presentation of septic emboli, possibly secondary to
thrombosed vein, which does not opacify on CTV. MRI may focal arteritis or rupture of small vessels at the sites of em-
show abnormal susceptibility on gradient echo sequences, but bolic occlusion. Convexal subarachnoid hemorrhage may be
this finding is often obscured by susceptibility artifact from visible on NCCT as peripheral areas of hyperdensity within
the adjacent calvarium. Secondary complications related to the cerebral sulci or as subarachnoid space FLAIR signal hy-
venous hypertension are similar to those of dural venous sinus perintensity on MRI.
thrombosis. Septic aneurysms may form in association with septic em-
boli or as a consequence of systemic sepsis or spread from
local head and neck infections. Rupture of a septic aneurysm
Septic Emboli, Septic Aneurysms, and may result in diffuse subarachnoid hemorrhage indistin-
Disseminated Intravascular Coagulation guishable from saccular aneurysm rupture or a more local-
ized presentation of subarachnoid hemorrhage confined to
Infectious endocarditis, as well as systemic sepsis from any a sylvian fissure or a cerebral convexity. On angiography,
cause, can lead to intracranial complications from septic septic aneurysms often appear irregular in shape and arise in

FIGURE 3.21 Septic emboli. NCCT (A) demonstrates loss of graywhite differentiation consistent with
acute cortical infarct (arrowhead). Gradient-echo MRI sequence (B) demonstrates multiple small micro-
hemorrhages (arrow).

Scheld_Ch03.indd 44 2/21/14 6:24 PM


Chapter 3: Imaging of Intracranial Infections 45

atypical locations, usually more distally in the vascular tree


compared to saccular aneurysms.
DIC usually occurs in the setting of systemic sepsis and
may also result in multiple cerebral infarcts and hemorrhages.
Multifocal convexal subarachnoid hemorrhage is another pre-
sentation of DIC (Fig. 3.22). Infarcts and hemorrhages result
from clot formation, consumption of platelets and clotting
factors, and bleeding complications in multiple organ systems.
The imaging findings are nonspecific and may share overlap-
ping features with septic emboli.

CONCLUSION
The spectrum of imaging findings in CNS infections includes
leptomeningeal enhancement, extraaxial collections, cerebri-
tis, encephalitis, white matter abnormalities, and enhancing
lesions. The pattern of these findings, however, may aid in
diagnosis of the underlying infectious agent and sometimes
predicts a specific organism. Additionally, imaging is key in
evaluating complications of CNS infections, including hydro-
cephalus, brain herniation, cerebral edema, infarcts, and other
vascular abnormalities.

ACKNOWLEDGMENTS
We gratefully acknowledge the contribution of some illustra-
FIGURE 3.22 DIC. NCCT demonstrates multiple scattered areas of tive cases from Dr. Mahmoud Mossa-Basha and Dr. James
convexal subarachnoid hemorrhage (SAH) (arrows). Fink from the University of Washington.

References
1. Kanal E, Barkovich AJ, Bell C, et al. ACR guidance document on MR safe 16. Britt RH, Enzmann DR. Clinical stages of human brain abscesses on serial
practices: 2013. J Magn Reson Imaging. 2013;37:501530. CT scans after contrast infusion. Computerized tomographic, neuropatho-
2. American College of Radiology Committee on Drugs and Contrast Media. logical, and clinical correlations. J Neurosurg. 1983;59:972989.
ACR manual on contrast media. American College of Radiology Web site. 17. Hatta S, Mochizuki H, Kuru Y, et al. Serial neuroradiological studies in
http://www.acr.org/~/media/ACR/Documents/PDF/QualitySafety/Resources focal cerebritis. Neuroradiology. 1994;36:285288.
/Contrast%20Manual/2013_Contrast_Media.pdf. Accessed April 17, 2013. 18. Carpenter J, Stapleton S, Holliman R. Retrospective analysis of 49 cases of
3. Jordan JE, Wippold FJ II, Cornelius RS, et al. ACR Appropriateness brain abscess and review of the literature. Eur J Clin Microbiol Infect Dis.
Criteria: headache. American College of Radiology Web site. http://www 2007;26:111.
.acr.org/~/media/ACR/Documents/AppCriteria/Diagnostic/Headache.pdf. 19. Seydoux C, Francioli P. Bacterial brain abscesses: factors influencing mor-
Accessed December 19, 2012. tality and sequelae. Clin Infect Dis. 1992;15:394401.
4. Nickerson JP, Richner B, Santy K, et al. Neuroimaging of pediatric intracra- 20. Tseng JH, Tseng MY. Brain abscess in 142 patients: factors influencing out-
nial infectionpart 1: techniques and bacterial infections. J Neuroimaging. come and mortality. Surg Neurol. 2006;65:557562; discussion 562.
2012;22:e42e51. 21. Nathoo N, Nadvi SS, Narotam PK, et al. Brain abscess: management and
5. Cornelius RS, Martin J, Wippold FJ II, et al. ACR Appropriateness outcome analysis of a computed tomography era experience with 973 pa-
Criteria: sinonasal disease. American College of Radiology Web site. tients. World Neurosurg. 2011;75:716726; discussion 612617.
http://www.acr.org/~/media/8172B4DE503149248E64856857674BB5. 22. Haimes AB, Zimmerman RD, Morgello S, et al. MR imaging of brain
pdf. Accessed December 19, 2012. abscesses. AJR Am J Roentgenol. 1989;152:10731085.
6. van de Beek D, de Gans J, Tunkel AR, et al. Community-acquired bacterial 23. Chiang IC, Hsieh TJ, Chiu ML, et al. Distinction between pyogenic brain
meningitis in adults. N Engl J Med. 2006;354:4453. abscess and necrotic brain tumour using 3-tesla MR spectroscopy, diffusion
7. Hasbun R, Abrahams J, Jekel J, et al. Computed tomography of the head and perfusion imaging. Br J Radiol. 2009;82:813820.
before lumbar puncture in adults with suspected meningitis. N Engl J Med. 24. Hakyemez B, Erdogan C, Bolca N, et al. Evaluation of different cerebral
2001;345:17271733. mass lesions by perfusion-weighted MR imaging. J Magn Reson Imaging.
8. Joffe AR. Lumbar puncture and brain herniation in acute bacterial menin- 2006;24:817824.
gitis: a review. J Intensive Care Med. 2007;22:194207. 25. Al-Okaili RN, Krejza J, Wang S, et al. Advanced MR imaging techniques
9. Gower DJ, Baker AL, Bell WO, et al. Contraindications to lumbar punc- in the diagnosis of intraaxial brain tumors in adults. Radiographics.
ture as defined by computed cranial tomography. J Neurol Neurosurg 2006;26(suppl 1):S173S189.
Psychiatry. 1987;50:10711074. 26. Gupta RK, Vatsal DK, Husain N, et al. Differentiation of tuberculous
10. Cabral DA, Flodmark O, Farrell K, et al. Prospective study of computed from pyogenic brain abscesses with in vivo proton MR spectroscopy and
tomography in acute bacterial meningitis. J Pediatr. 1987;111:201205. magnetization transfer MR imaging. AJNR Am J Neuroradiol. 2001;22:
11. Kawaguchi T, Sakurai K, Hara M, et al. Clinico-radiological features of 15031509.
subarachnoid hyperintensity on diffusion-weighted images in patients with 27. Pal D, Bhattacharyya A, Husain M, et al. In vivo proton MR spectroscopy
meningitis. Clin Radiol. 2012;67:306312. evaluation of pyogenic brain abscesses: a report of 194 cases. AJNR Am J
12. Smirniotopoulos JG, Murphy FM, Rushing EJ, et al. Patterns of contrast Neuroradiol. 2010;31:360366.
enhancement in the brain and meninges. Radiographics. 2007;27:525551. 28. Fukui MB, Williams RL, Mudigonda S. CT and MR imaging features of
13. Castillo M. Imaging of meningitis. Semin Roentgenol. 2004;39:458464. pyogenic ventriculitis. AJNR Am J Neuroradiol. 2001;22:15101516.
14. Mohan S, Jain KK, Arabi M, et al. Imaging of meningitis and ventriculitis. 29. Osborn MK, Steinberg JP. Subdural empyema and other suppurative com-
Neuroimaging Clin N Am. 2012;22:557583. plications of paranasal sinusitis. Lancet Infect Dis. 2007;7:6267.
15. Rath TJ, Hughes M, Arabi M, et al. Imaging of cerebritis, encephalitis, and 30. Nathoo N, Nadvi SS, Van Dellen JR. Traumatic cranial empyemas: a
brain abscess. Neuroimaging Clin N Am. 2012;22:585607. review of 55 patients. Br J Neurosurg. 2000;14:326330.

Scheld_Ch03.indd 45 2/21/14 6:24 PM


46 Part I: Approach to the Patient and Diagnostic Evaluation

31. Nathoo N, Nadvi SS, van Dellen JR, et al. Intracranial subdural empyemas 63. Launes J, Nikkinen P, Lindroth L, et al. Diagnosis of acute herpes simplex
in the era of computed tomography: a review of 699 cases. Neurosurgery. encephalitis by brain perfusion single photon emission computed tomogra-
1999;44:529535; discussion 535536. phy. Lancet. 1988;1:11881191.
32. Wong AM, Zimmerman RA, Simon EM, et al. Diffusion-weighted MR 64. Calli C, Ozel AA, Savas R, et al. Proton MR spectroscopy in the diag-
imaging of subdural empyemas in children. AJNR Am J Neuroradiol. nosis and differentiation of encephalitis from other mimicking lesions.
2004;25:10161021. J Neuroradiol. 2002;29:2328.
33. Nathoo N, Nadvi SS, van Dellen JR. Cranial extradural empyema in 65. Gupta RK, Soni N, Kumar S, et al. Imaging of central nervous system viral
the era of computed tomography: a review of 82 cases. Neurosurgery. diseases. J Magn Reson Imaging. 2012;35:477491.
1999;44:748753; discussion 753754. 66. Gilden D, Cohrs RJ, Mahalingam R, et al. Varicella zoster virus vasculopa-
34. Weingarten K, Zimmerman RD, Becker RD, et al. Subdural and epidural thies: diverse clinical manifestations, laboratory features, pathogenesis, and
empyemas: MR imaging. AJR Am J Roentgenol. 1989;152:615621. treatment. Lancet Neurol. 2009;8:731740.
35. Patkar D, Narang J, Yanamandala R, et al. Central nervous system tuber- 67. Ali M, Safriel Y, Sohi J, et al. West Nile virus infection: MR imaging find-
culosis: pathophysiology and imaging findings. Neuroimaging Clin N Am. ings in the nervous system. AJNR Am J Neuroradiol. 2005;26:289297.
2012;22:677705. 68. Petropoulou KA, Gordon SM, Prayson RA, et al. West Nile virus menin-
36. Marais S, Thwaites G, Schoeman JF, et al. Tuberculous meningitis: a uniform goencephalitis: MR imaging findings. AJNR Am J Neuroradiol. 2005;26:
case definition for use in clinical research. Lancet Infect Dis. 2010;10:803812. 19861995.
37. Botha H, Ackerman C, Candy S, et al. Reliability and diagnostic perfor- 69. Cerna F, Mehrad B, Luby JP, et al. St. Louis encephalitis and the substantia
mance of CT imaging criteria in the diagnosis of tuberculous meningitis. nigra: MR imaging evaluation. AJNR Am J Neuroradiol. 1999;20:12811283.
PLoS One. 2012;7:e38982. 70. Lukes SA, Norman D. Computed tomography in acute disseminated
38. Abdelmalek R, Kanoun F, Kilani B, et al. Tuberculous meningitis in encephalomyelitis. Ann Neurol. 1983;13:567572.
adults: MRI contribution to the diagnosis in 29 patients. Int J Infect Dis. 71. Caldemeyer KS, Smith RR, Harris TM, et al. MRI in acute disseminated
2006;10:372377. encephalomyelitis. Neuroradiology. 1994;36:216220.
39. Wasay M, Kheleani BA, Moolani MK, et al. Brain CT and MRI findings in 72. Mader I, Stock KW, Ettlin T, et al. Acute disseminated encephalomyelitis:
100 consecutive patients with intracranial tuberculoma. J Neuroimaging. MR and CT features. AJNR Am J Neuroradiol. 1996;17:104109.
2003;13:240247. 73. Honkaniemi J, Dastidar P, Khr V, et al. Delayed MR imaging changes in
40. Smith AB, Smirniotopoulos JG, Rushing EJ. From the archives of the AFIP: acute disseminated encephalomyelitis. AJNR Am J Neuroradiol. 2001;22:
central nervous system infections associated with human immunodefi- 11171124.
ciency virus infection: radiologic-pathologic correlation. Radiographics. 74. Tenembaum S, Chitnis T, Ness J, et al. Acute disseminated encephalomyeli-
2008;28:20332058. tis. Neurology. 2007;68:S23S36.
41. Agarwal R, Sze G. Neuro-lyme disease: MR imaging findings. Radiology. 75. Singh S, Alexander M, Korah IP. Acute disseminated encephalomyelitis:
2009;253:167173. MR imaging features. AJR Am J Roentgenol. 1999;173:11011107.
42. Hildenbrand P, Craven DE, Jones R, et al. Lyme neuroborreliosis: mani- 76. Masdeu JC, Moreira J, Trasi S, et al. The open ring. A new imaging sign in
festations of a rapidly emerging zoonosis. AJNR Am J Neuroradiol. demyelinating disease. J Neuroimaging. 1996;6:104107.
2009;30:10791087. 77. Masdeu JC, Quinto C, Olivera C, et al. Open-ring imaging sign: highly
43. Peng F, Hu X, Zhong X, et al. CT and MR findings in HIV-negative neuro- specific for atypical brain demyelination. Neurology. 2000;54:14271433.
syphilis. Eur J Radiol. 2008;66:16. 78. Axer H, Ragoschke-Schumm A, Bttcher J, et al. Initial DWI and ADC im-
44. Brightbill TC, Ihmeidan IH, Post MJ, et al. Neurosyphilis in HIV-positive aging may predict outcome in acute disseminated encephalomyelitis: report
and HIV-negative patients: neuroimaging findings. AJNR Am J Neuroradiol. of two cases of brain stem encephalitis. J Neurol Neurosurg Psychiatry.
1995;16:703711. 2005;76:996998.
45. Fargen KM, Alvernia JE, Lin CS, et al. Cerebral syphilitic gummata: 79. Harada M, Hisaoka S, Mori K, et al. Differences in water diffusion and
a case presentation and analysis of 156 reported cases. Neurosurgery. lactate production in two different types of postinfectious encephalopathy.
2009;64:568575; discussion 575576. J Magn Reson Imaging. 2000;11:559563.
46. Akgoz A, Mukundan S, Lee TC. Imaging of rickettsial, spirochetal, and 80. Bernarding J, Braun J, Koennecke HC. Diffusion- and perfusion-weighted
parasitic infections. Neuroimaging Clin N Am. 2012;22:633657. MR imaging in a patient with acute demyelinating encephalomyelitis
47. Zee CS, Segall HD, Boswell W, et al. MR imaging of neurocysticercosis. (ADEM). J Magn Reson Imaging. 2002;15:96100.
J Comput Assist Tomogr. 1988;12:927934. 81. Bizzi A, Ulug AM, Crawford TO, et al. Quantitative proton MR spec-
48. Lucato LT, Guedes MS, Sato JR, et al. The role of conventional MR imaging troscopic imaging in acute disseminated encephalomyelitis. AJNR Am J
sequences in the evaluation of neurocysticercosis: impact on characterization of Neuroradiol. 2001;22:11251130.
the scolex and lesion burden. AJNR Am J Neuroradiol. 2007;28:15011504. 82. Balasubramanya KS, Kovoor JME, Jayakumar PN, et al. Diffusion-
49. Collie DA, Sellar RJ, Zeidler M, et al. MRI of Creutzfeldt-Jakob dis- weighted imaging and proton MR spectroscopy in the characterization
ease: imaging features and recommended MRI protocol. Clin Radiol. of acute disseminated encephalomyelitis. Neuroradiology. 2007;49:
2001;56:726739. 177183.
50. Young GS, Geschwind MD, Fischbein NJ, et al. Diffusion-weighted and 83. Hynson JL, Kornberg AJ, Coleman LT, et al. Clinical and neuroradiologic
fluid-attenuated inversion recovery imaging in Creutzfeldt-Jakob disease: features of acute disseminated encephalomyelitis in children. Neurology.
high sensitivity and specificity for diagnosis. AJNR Am J Neuroradiol. 2001;56:13081312.
2005;26:15511562. 84. Mader I, Herrlinger U, Klose U, et al. Progressive multifocal leukoencepha-
51. Vitali P, Maccagnano E, Caverzasi E, et al. Diffusion-weighted MRI hyper- lopathy: analysis of lesion development with diffusion-weighted MRI.
intensity patterns differentiate CJD from other rapid dementias. Neurology. Neuroradiology. 2003;45:717721.
2011;76:17111719. 85. Ohta K, Obara K, Sakauchi M, et al. Lesion extension detected by diffu-
52. Collie DA, Summers DM, Sellar RJ, et al. Diagnosing variant Creutzfeldt- sion-weighted magnetic resonance imaging in progressive multifocal leuko-
Jakob disease with the pulvinar sign: MR imaging findings in 86 neuropatho- encephalopathy. J Neurol. 2001;248:809811.
logically confirmed cases. AJNR Am J Neuroradiol. 2003;24:15601569. 86. Hoffmann C, Horst HA, Albrecht H, et al. Progressive multifocal leuco-
53. Chadwick DR. Viral meningitis. Br Med Bull. 2005;7576:114. encephalopathy with unusual inflammatory response during antiretroviral
54. Tha KK, Terae S, Kudo K, et al. Differential diagnosis of hyperintense cere- treatment. J Neurol Neurosurg Psychiatry. 2003;74:11421144.
brospinal fluid on fluid-attenuated inversion recovery images of the brain. 87. Martinez JV, Mazziotti JV, Efron ED, et al. Immune reconstitution
Part I: pathological conditions. Br J Radiol. 2009;82:426434. inflammatory syndrome associated with PML in AIDS: a treatable disorder.
55. Tien RD, Felsberg GJ, Osumi AK. Herpesvirus infections of the CNS: MR Neurology. 2006;67:16921694.
findings. AJR Am J Roentgenol. 1993;161:167176. 88. Vendrely A, Bienvenu B, Gasnault J, et al. Fulminant inflammatory leu-
56. Davis JM, Davis KR, Kleinman GM, et al. Computed tomography of her- koencephalopathy associated with HAART-induced immune restora-
pes simplex encephalitis, with clinicopathological correlation. Radiology. tion in AIDS-related progressive multifocal leukoencephalopathy. Acta
1978;129:409417. Neuropathol. 2005;109:449455.
57. Enzmann DR, Ranson B, Norman D, et al. Computed tomography of her- 89. Svensson PA, Larsson EM. Infratentorial progressive multifocal leuco-
pes simplex encephalitis. Radiology. 1978;129:419425. encephalopathy (PML) in a patient with SLE (2008: 4b). Eur Radiol.
58. Schroth G, Gawehn J, Thron A, et al. Early diagnosis of herpes simplex 2008;18:15261528.
encephalitis by MRI. Neurology. 1987;37:179183. 90. Boster A, Hreha S, Berger JR, et al. Progressive multifocal leukoencepha-
59. Tsuchiya K, Katase S, Yoshino A, et al. Diffusion-weighted MR imaging of lopathy and relapsing-remitting multiple sclerosis: a comparative study.
encephalitis. AJR Am J Roentgenol. 1999;173:10971099. Arch Neurol. 2009;66:593599.
60. Heiner L, Demaerel P. Diffusion-weighted MR imaging findings in a patient 91. Chang L, Ernst T, Tornatore C, et al. Metabolite abnormalities in progres-
with herpes simplex encephalitis. Eur J Radiol. 2003;45:195198. sive multifocal leukoencephalopathy by proton magnetic resonance spec-
61. Kker W, Ngele T, Schmidt F, et al. Diffusion-weighted MRI in herpes sim- troscopy. Neurology. 1997;48:836845.
plex encephalitis: a report of three cases. Neuroradiology. 2004;46:122125. 92. Fink KR, Thapa MM, Ishak GE, et al. Neuroimaging of pediatric central ner-
62. Marco de Lucas E, Gonzlez Mandly A, Gutirrez A, et al. Computed vous system cytomegalovirus infection. Radiographics. 2010;30:17791796.
tomography perfusion usefulness in early imaging diagnosis of herpes sim- 93. Griffiths P. Cytomegalovirus infection of the central nervous system.
plex virus encephalitis. Acta Radiol. 2006;47:878881. Herpes. 2004;11(suppl 2):95A104A.

Scheld_Ch03.indd 46 2/21/14 6:24 PM


Chapter 3: Imaging of Intracranial Infections 47

94. Guerini H, Helie O, Leveque C, et al. Diagnosis of periventricular epen- 108. Post MJD, Thurnher MM, Clifford DB, et al. CNS-immune reconstitution
dymal enhancement in MRI in adults [in French]. J Neuroradiol. 2003; inflammatory syndrome in the setting of HIV infection, part 1: overview
30:4656. and discussion of progressive multifocal leukoencephalopathy-immune
95. de Silva T, Raychaudhuri M, Poulton M, et al. Ventriculitis and hydro- reconstitution inflammatory syndrome and cryptococcal-immune recon-
cephalus: an unusual presentation of toxoplasmosis in an adult with human stitution inflammatory syndrome. AJNR Am J Neuroradiol. 2013;34:
immunodeficiency virus. J Neurol Neurosurg Psychiatry. 2005;76:1074. 12971307.
96. Brightbill TC, Post MJ, Hensley GT, et al. MR of Toxoplasma encephalitis: 109. Pfister HW, Borasio GD, Dirnagl U, et al. Cerebrovascular complications
signal characteristics on T2-weighted images and pathologic correlation. of bacterial meningitis in adults. Neurology. 1992;42:14971504.
J Comput Assist Tomogr. 1996;20:417422. 110. Kastenbauer S, Pfister HW. Pneumococcal meningitis in adults: spectrum
97. Chong-Han CH, Cortez SC, Tung GA. Diffusion-weighted MRI of cere- of complications and prognostic factors in a series of 87 cases. Brain.
bral Toxoplasma abscess. AJR Am J Roentgenol. 2003;181:17111714. 2003;126:10151025.
98. Batra A, Tripathi RP, Gorthi SP. Magnetic resonance evaluation of cere- 111. Somer T, Finegold SM. Vasculitides associated with infections, immuniza-
bral toxoplasmosis in patients with the acquired immunodeficiency syn- tion, and antimicrobial drugs. Clin Infect Dis. 1995;20:10101036.
drome. Acta Radiol. 2004;45:212221. 112. Gaa J, Weidauer S, Sitzer M, et al. Cerebral vasculitis due to Treponema
99. Licho R, Litofsky NS, Senitko M, et al. Inaccuracy of Tl-201 brain SPECT pallidum infection: MRI and MRA findings. Eur Radiol. 2004;14:
in distinguishing cerebral infections from lymphoma in patients with 746747.
AIDS. Clin Nucl Med. 2002;27:8186. 113. Jaurguiberry S, Ansart S, Perez L, et al. Acute neuroschistosomiasis: two
100. Skiest DJ, Erdman W, Chang WE, et al. SPECT thallium-201 combined with cases associated with cerebral vasculitis. Am J Trop Med Hyg. 2007;76:
Toxoplasma serology for the presumptive diagnosis of focal central nervous 964966.
system mass lesions in patients with AIDS. J Infect. 2000;40:274281. 114. Katchanov J, Siebert E, Klingebiel R, et al. Infectious vasculopathy of
101. OMalley JP, Ziessman HA, Kumar PN, et al. Diagnosis of intracranial intracranial large- and medium-sized vessels in neurological intensive care
lymphoma in patients with AIDS: value of 201TI single-photon emission unit: a clinico-radiological study. Neurocrit Care. 2010;12:369374.
computed tomography. AJR Am J Roentgenol. 1994;163:417421. 115. Chow FC, Marra CM, Cho TA. Cerebrovascular disease in central ner-
102. Schroeder PC, Post MJD, Oschatz E, et al. Analysis of the utility of vous system infections. Semin Neurol. 2011;31:286306.
diffusion-weighted MRI and apparent diffusion coefficient values in 116. Javaud N, Certal RDS, Stirnemann J, et al. Tuberculous cerebral vas-
distinguishing central nervous system toxoplasmosis from lymphoma. culitis: retrospective study of 10 cases. Eur J Intern Med. 2011;22:
Neuroradiology. 2006;48:715720. e99e104.
103. Charlier C, Dromer F, Lvque C, et al. Cryptococcal neuroradiological 117. Gilden DH, Kleinschmidt-DeMasters BK, Wellish M, et al. Varicella zoster
lesions correlate with severity during cryptococcal meningoencephalitis in virus, a cause of waxing and waning vasculitis: the New England Journal
HIV-positive patients in the HAART era. PLoS One. 2008;3:e1950. of Medicine case 51995 revisited. Neurology. 1996;47:14411446.
104. Chen S, Chen X, Zhang Z, et al. MRI findings of cerebral cryptococcosis in 118. Vazquez E, Castellote A, Piqueras J, et al. Imaging of complications of
immunocompetent patients. J Med Imaging Radiat Oncol. 2011;55:5257. acute mastoiditis in children. Radiographics. 2003;23:359372.
105. Drake KW, Adam RD. Coccidioidal meningitis and brain abscesses: anal- 119. Bayonne E, Kania R, Tran P, et al. Intracranial complications of rhino-
ysis of 71 cases at a referral center. Neurology. 2009;73:17801786. sinusitis. A review, typical imaging data and algorithm of management.
106. Arsura EL, Johnson R, Penrose J, et al. Neuroimaging as a guide to pre- Rhinology. 2009;47:5965.
dict outcomes for patients with coccidioidal meningitis. Clin Infect Dis. 120. Schuknecht B, Simmen D, Yksel C, et al. Tributary venosinus occlusion
2005;40:624627. and septic cavernous sinus thrombosis: CT and MR findings. AJNR Am J
107. Williams PL, Johnson R, Pappagianis D, et al. Vasculitic and encepha- Neuroradiol. 1998;19:617626.
litic complications associated with Coccidioides immitis infection of the 121. Lee JH, Lee HK, Park JK, et al. Cavernous sinus syndrome: clinical fea-
central nervous system in humans: report of 10 cases and review. Clin tures and differential diagnosis with MR imaging. AJR Am J Roentgenol.
Infect Dis. 1992;14:673682. 2003;181:583590.

Scheld_Ch03.indd 47 2/21/14 6:24 PM


Scheld_Ch03.indd 48 2/21/14 6:24 PM
PART II VIRAL INFECTIONS AND
RELATED DISORDERS
CHAPTER 4 PATHOGENESIS AND
PATHOPHYSIOLOGY OF VIRAL INFECTIONS
OF THE CENTRAL NERVOUS SYSTEM
KEVIN A. CASSADY AND RICHARD J. WHITLEY

Viral infections of the central nervous system (CNS) occur direct viral entry into the CNS that produces clinically evident
infrequently and most often result in relatively benign, self- cortical or brainstem dysfunction. Subsequent damage occurs
limited disease. Nevertheless, CNS infections have tremendous as a consequence of the host immune response, but invasion
importance because of the potential for death and neurologic by the pathogen initiates CNS damage (6). The parenchyma
damage. The highly specialized brain tissue is exquisitely sen- exhibits neuronophagia, and viral antigen or nucleic acids can
sitive to metabolic derangements. Injured brain tissue recovers be detected (6). Postinfectious or parainfectious encephalitis is
slowly and often incompletely. Even in patients who recover an acute demyelinating process temporally associated with a
fully from viral encephalitis, months may be required for re- systemic viral infection but without evidence of direct viral in-
turn to normal function (1). The brain and spinal cord pro- vasion in the CNS and is included as one of the causes of acute
vide diagnostic and therapeutic obstacles. On an anatomic disseminated encephalomyelitis (ADEM) (6,7). Pathologic
level, the brain is housed in a closed skull with the spinal cord specimens demonstrate demyelination and perivascular aggre-
suspended within a bony columnar cage. A unique immuno- gation of immune cells but no evidence of virus or viral antigen,
logic surveillance system and the bloodbrain barrier further suggesting an immune-mediated etiology (1). The presence of
distinguish infections of the CNS from those involving other immune cells distinguishes primary and postinfectious enceph-
organ systems. Pathologic processes in the CNS have limited alitis from an encephalopathy.
clinical expressions and frequently share pathogenic mecha- Inflammation occurs at multiple sites within the CNS and
nisms. Tumors, infections, and autoimmune processes in the accounts for the myriad of clinical descriptors of viral neuro-
CNS often produce similar signs and symptoms (2). Clinical logic disease. Inflammation of the spinal cord, leptomeninges,
presentation and patient history, though frequently suggestive dorsal nerve roots, or nerves results in myelitis, meningitis,
of a diagnosis, remain unreliable methods for determining the radiculitis, and neuritis, respectively. Aseptic meningitis is
specific etiology of CNS disease (2,3). Understanding the path- frequently used to refer to a benign, self-limited, viral infec-
ogenic mechanism of a disease provides a rational basis for tion causing inflammation of the leptomeninges (1). The term
the development of antiviral medications and strategies for the aseptic meningitis is used instead of viral meningitis because
prevention of viral CNS infections. a pathogen fails to grow in conventional culture media and
The pathogenesis of viral infections is multifactorial: age, reflects the historic ability to diagnose and treat only bacterial
immune status, cultural practices, and genetic makeup can and fungal CNS infections (6,8). This misnomer hinders epide-
influence the clinical manifestations of viral infection as read- miologic studies, because the definition fails to differentiate be-
ily as viral load, gene polymorphisms, receptor preference, and tween infectious (fungal, tuberculous, viral, or other infectious
cell tropism. Although asymptomatic enteroviral infection pre- etiologies) and noninfectious causes of meningitis. Meningitis
dominates, some patients progress to viral meningitis or, rarely, and encephalitis can represent separate clinical entities; how-
fulminant encephalitis (1,4). A detailed description of the ever, a continuum exists between these distinct forms of CNS
pathogenesis of the individual viral encephalitides is beyond the disease (1). A change in a patients clinical condition can reflect
scope of this chapter. Instead, general concepts of viral infec- disease progression, with involvement of different regions of
tion and the pathogenic mechanisms of viral CNS infection are the CNS making it difficult to predict the extent of CNS infec-
reviewed and specific examples developed where applicable. tion early in the clinical course. A patient may present with
meningismus and be diagnosed as having viral meningitis and
then progress to meningoencephalitis with altered conscious-
DEFINITIONS ness and focal CNS changes (6). Epidemiologic data in many
cases provide clues to the viral etiology.
Viruses display tissue tropism and cause illness with a charac-
teristic temporal course. The definition of viral CNS disease
is often based on both viral tropism and disease duration. EPIDEMIOLOGY
Encephalitis refers to inflammation of parenchymal brain
tissue. Acute encephalitis occurs over a relatively short pe- Epidemiology studies of meningitis and encephalitis potentially
riod of time (days), whereas chronic encephalitis presents over underestimate the true incidence of viral CNS infections. Even
weeks to months. The temporal course of slow infections and when aseptic meningitis was a reportable disease, not all pa-
spongiform encephalopathies of the CNS (kuru, visna, variant tients having a cerebrospinal fluid (CSF) pleocytosis or symp-
Creutzfeldt-Jakob disease) overlaps with that of the chronic en- toms consistent with a viral meningitis had viral cultures or
cephalitides. These progressive CNS diseases are distinguished other diagnostic studies performed. An overview is difficult,
by a long incubation period, eventually resulting in death or because each pathogen fills a different ecologic niche with
extreme neurologic disability over months to years (1,5). unique seasonal, host, and/or vector properties (1) (Tables 4.1
Viral disease in the CNS can also be classified by pathogen- and 4.2). Instead, it is useful to analyze the individual agents
esis. Neurologic disease is frequently categorized as either pri- responsible for viral brain infections in an effort to find popu-
mary or postinfectious (1). A primary encephalitis results from lation patterns and trends.

49

Scheld_Ch04.indd 49 2/21/14 5:30 PM


50 Part II: Viral Infections and Related Disorders

TA B L E 4 . 1
TYPE OF DISEASE, EPIDEMIOLOGIC DATA, AND PATHOGENESIS OF VIRAL INFECTIONS
OF THE CNS: DNA VIRUS

Temporal Pathway Laboratory


Viral Agent CNS Disease Course Transmission to CNS Frequency Confirmation

Herpesviridae
Herpes simplex Encephalitis, meningitis, Acute Human Neuronal  Gold standardcell
virus type 1 meningoencephalitis Latent Blood culture brain biopsy
and type 2 reactivation sample; PCR has
replaced routine
brain biopsy.
Cytomegalovirus Encephalitis Acute Blood  Gold standardcell
(immunosuppressed culture, brain biopsy
and neonate) or CSF sample; PCR
may supplant this.
Epstein-Barr Encephalitis, meningitis, Acute  Serologic evidence.
virus myelitis, Guillain-Barr
Varicella-zoster Cerebellitis, Postinfectious Blood  Clinical findings,
virus encephalitis, Acute and latent Neuronal cell culture from a
meningitis, myelitis, reactivation lesion, brain biopsy
zoster ophthalmicus (zoster) or necroscopy.
Human herpes Encephalitis, febrile Acute ? ? ?PCR; high rate of
virus-6 seizures, latent form? Latent infection latent virus in
certain sites makes
interpretation
difficult
B virus Encephalitis Acute Animal bite Neuronal  Culture.
and human
Adenoviridae
Adenovirus Meningitis, encephalitis Acute Human Blood  Cell culture of CSF or
brain.
Poxviridae
Vaccinia Encephalomyelitis Postinfectious Vaccine Blood Extinct Recent

, frequent;  , infrequent; , rare; ?, unknown.

Historically, laboratory techniques for identifying neuro- sensitivity; however, the techniques sensitivity can lead to
logic infections were insensitive, invasive, and required brain erroneous diagnosis, because PCR may detect latent or in-
biopsy. Over the last two decades, molecular detection tech- tegrated viral DNA potentially unrelated to the pathogenic
niques have improved the detection of pathogens nucleic process (1). The introduction and testing of new antiviral
acids in the CSF (8,9). Despite the improved sensitivity of drugs will likely provide an impetus for accurate and timely
these techniques, the pathogen remains unidentified in the diagnosis.
majority of cases of encephalitis. Depending on the study Acute viral meningitis and meningoencephalitis represent
and diagnostic methods used, investigators fail to identify an most viral brain infections and frequently occur in epidemics
agent in the majority of presumed CNS infections (10,11). (1). Enteroviruses cause an estimated 60% to 90% of cases,
CSF viral culture rates differ based on etiology. They can whereas arboviruses constitute the majority of the remain-
often be diagnosed only presumptively by acute and conva- ing reported cases (1,8). The Centers for Disease Control
lescent serologic testing or isolation of virus from another lo- and Prevention (CDC) received notification of approximately
cation in the body (6,12). In a retrospective review of patients 7,200 to 14,500 cases of aseptic meningitis annually (1).
who had positive bacterial CSF cultures, 1 of 20 had a con- Most of these cases occurred from the late spring to autumn
comitant virus isolated from the CSF (13,14). Historically, months, reflecting the increased incidence of enteroviral and
the definitive method for virus detection in encephalitis was arboviral infections during these seasons (17,18). The inci-
brain biopsy and viral culture (1,2). Polymerase chain reac- dence and etiology of encephalitis varies based on geography,
tion (PCR) techniques and other molecular biologic methods environmental factors, and frequency of exposure to vectors
from CSF samples have replaced culture and brain biopsy responsible for viral transmission (19,20).
as the standard for diagnosing encephalitis for some viruses The CDC received 740 to 1,340 annual reports of per-
(herpes simplex virus [HSV], enterovirus, varicella-zoster sons with encephalitis from 1990 to 1993 (1). Herpes sim-
virus [VZV], and JC virus) (8,15,16). PCR has exquisite plex virus infection of the brain occurs year round without

Scheld_Ch04.indd 50 2/21/14 5:30 PM


Chapter 4: Pathogenesis and Pathophysiology of Viral Infections of the Central Nervous System 51

TA B L E 4 . 2
TYPE OF DISEASE, EPIDEMIOLOGIC DATA, AND PATHOGENESIS OF VIRAL INFECTIONS OF THE CNS: RNA VIRUSES

Fatality Geographic Disease Pathway Laboratory


Viral Taxonomy CNS Disease Rate Vector Distribution Pattern to CNS Frequency Confirmation

TogaviridaeAlphavirus (arborvirus)
Western Meningitis, 310% Mosquito, U.S.west of Epidemic Blood  Serologic titers
equine encephalitis bird Mississippi (HI, CF,
encephalitis River NA, IFA),
virus viral antigen
detection in
brain. Rarely
culture.
Eastern 30% U.S.Atlantic Sporadic  Viral culture
equine and Gulf or antigen
encephalitis Coast states detection in
virus brain, serologic
titers (HI, CF,
NA, IFA), CSF
IgM ELISA.
Venezuelan 1% Mosquito, Central and Sporadic,  Serologic titers
equine horse South epidemic (HI, CF, NA,
encephalitis America, IFA), CSF IgM
virus southwestern ELISA.
U.S., and
Florida
FlaviviridaeFlavivirus (arbovirus)
Japanese Meningitis, 25% Mosquito, Japan, China, Epidemic, Blood  Peripheral blood
encephalitis encephalitis swine, Korea, endemic ELISA, serologic
virus bird Taiwan, titers (HI, CF,
Southeast NA, IFA), CSF
Asia, India, antigen tests.
Nepal
St. Louis 7% U.S.  CSF IgM ELISA,
encephalitis serologic titers
virus (HI, CF, NA,
IFA). Rarely
culture.
West Nile Rarely Uganda, Egypt,  Culture (rare),
fever virus Israel serology (HI,
IFA).
Murray Valley virus Encephalitis 2060% Australia  Viral culture,
serologic titer
(HI, CF, NA).
Tickborne 20% Tick, Eastern Epidemic,  Serologic titer
encephalitis unpasteur- Russia and sporadic (HI, CF, NA),
virus (TBE ized milk Central IgM ELISA.
complex) Europe
Bunyaviridae (arbovirus)
California Meningitis, 1% Mosquito, Northern Endemic Blood  Viral culture, CSF
(La Crosse) encephalitis rodent Midwest (LCV)  IgM ELISA,
encephalitis and (CEV) serologic titers
virus northeastern (HI, CF, NA,
U.S. IFA), CIE.
ReoviridaeOrbivirus (orvivirus)
Colorado Meningitis, 1% Tick, rodent Rocky Endemic Blood  Antigen detection
tick fever encephalitis Mountains, on RBC
Pacific membrane, viral
Coast states culture, serologic
titers (HI, CF,
NA, IFA).

(continued)

51

Scheld_Ch04.indd 51 2/21/14 5:30 PM


52 Part II: Viral Infections and Related Disorders

TA B L E 4 . 2
TYPE OF DISEASE, EPIDEMIOLOGIC DATA, AND PATHOGENESIS OF VIRAL INFECTIONS OF THE CNS:
RNA VIRUSES (CONTINUED)

Fatality Geographic Disease Pathway Laboratory


Viral Taxonomy CNS Disease Rate Vector Distribution Pattern to CNS Frequency Confirmation

Picornaviridae (enterovirus)
Poliovirus Meningitis 4.550%a Fecal-oral Worldwide Endemic Blood and  Viral culture
myelitis neuronal CSF or brain,
viral culture
from other
side. Serologic
testing for some
serotypes. PCR
becoming gold
standard.
Coxsackievirus Meningitis, Rarelyb Blood 
Echovirus Meningo- 
encephalitis,
myelitis

Paramyxoviridae (xanthematous virus)


Measles virus Encephalitis, 15% Postinfectious, Worldwide Sporadic Blood  Serology, ELISA,
SSPE blood clinically.
Mumps virus Meningitis, 1% Blood  CSF, viral culture.
encephalitis,
myelitis
Orthomyxoviridae (upper respiratory virus)
Influenza viruses Encephalitis 1% Postinfectious Worldwide Sporadic Blood  Viral culture from
another site.
Rhabdoviridae
Rabies virus Encephalitis, 100% Mammal Worldwide Sporadic Neuronal  Antigen detection
encephalo- in brain,
myelitis serologic tests
(IFA, CF, HI,
CIE), viral
culture.
Retroviridae
Human Encephalopathy, Ultimately Human Worldwide ? Blood  PCR autopsy
immunodeficiency encephalitis, 100% samples, MRI
virus type 1 leukoence- findings.
(HIV-1) phalopathy
Arenaviridae
Lymphocytic Meningitis, 2.5% Rodent Worldwide Sporadic Blood  CSF, blood culture,
choriomeningitis encephalitis urine culture,
virus serology.

CF, complement fixation; CIE, counterimmunoelectrophoresis; ELISA, enzyme-linked immunosorbent assay; HI, hemagglutination inhibition; IFA, immu-
nofluorescence antibody; NA, neutralizing antibody titer; SSPE, subacute sclerosing panencephalitis.
Frequency:   , frequent;  , infrequent; , rare; ?, unknown.
a
Case fatality from poliomyelitis is increased in sporadic cases. With vaccination, the epidemic forms of polio have decreased, as has morbidity. In turn, the
calculated case-fatality rate in the U.S. has increased in sporadic and vaccine-associated disease relative to the number of cases of disease.
b
Rarely fatal except in neonate and agammaglobulinemic patient in whom fatality rates can approach 50% even with treatment.

seasonal variation, affects all ages, and constitutes most infections vastly outnumber those that are symptomatic.
fatal cases of endemic encephalitis in the United States (21). Patients with symptomatic infections may develop a mild,
Arboviruses, a group of more than 500 arthropod-transmit- systemic febrile illness or a viral meningitis. Encephalitis oc-
ted RNA viruses, are the leading cause of encephalitis world- curs in a minority of persons with arboviral infections, but
wide and in the United States (1). Arboviral infections occur the case-fatality rate varies extremely from 5% to 70%, de-
in epidemics and show a seasonal predilection, reflecting the pending on viral etiology, age of the patient, and unique host
prevalence of the transmitting vector (22). Asymptomatic differences (1,23).

52

Scheld_Ch04.indd 52 2/21/14 5:30 PM


Chapter 4: Pathogenesis and Pathophysiology of Viral Infections of the Central Nervous System 53

WNV 2011
Japanese B encephalitis and rabies constitute most cases of
encephalitis outside of North America. Japanese B encephalitis
virus, a member of the genus Flavivirus, occurs throughout
Asia and causes epidemics in China despite routine immuni-
zation for the virus (24,25). In warmer locations, the virus
occurs endemically (26). The disease typically affects children,
although adults with no history of exposure to the virus are also
susceptible (27). As with the other arboviral infections, asymp-
tomatic infections occur more frequently than symptomatic in-
fections. However, the disease has a high case-fatality rate and
leaves half of the survivors with a high degree of neurologic
morbidity (27). Of note, West Nile virus (WNV) encephalitis,
a member of the Flavivirus family, has increased in incidence
strikingly in the United States (28). In 2002 alone, the CDC
reported more than 3,989 cases and nearly 250 deaths. WNV
infection declined in the United States such that between 2008
and 2011, only 712 to 1,356 cases were reported. There was
an increase in cases (5,387) in the United States in 2012. Many
of the cases occurred in the Mississippi Valley and Southern WNV 2012
and Central United States, suggesting an evolving epidemi-
ology for this introduced pathogen (Fig. 4.1) (http://www
.cdc.gov/ncidod/dvbid/westnile/surv&controlCaseCount12_
detailed.htm). Rabies virus, a bullet-shaped RNA virus of the
family Rhabdoviridae, remains endemic around the world
(29). Human infections in the United States decreased over
the last decades to one to three cases per year because of the
immunization of domesticated animals. Bat exposure is in-
creasingly recognized as the source of infection. Fifteen per-
cent (685 of 4,470) of bats tested carried the rabies virus in
one study analyzing risk of bat exposure and rabies (30). In
most cases, (22 of 24) there was no evidence of bite; how-
ever, in half of the cases, direct contact (handling of the bats)
was documented (31). There is experimental evidence that
bat-associated rabies virus variants transmit across the dermis FIGURE 4.1 Cases of West Nile virus (WNV) in the United States
and potentially through hair follicles (29). Alternatively, bat 2011 (top panel) and 2012 as of December 11, 2012 (bottom panel).
bites may not have been recognized (1). In areas outside the Shaded area demonstrates counties where human WNV infection was
United States, annual cases of rabies encephalitis number in reported. (Courtesy of CDC Arboviral Branch.)
the thousands.
Postinfectious encephalitis, an acute demyelinating pro-
cess, accounts for approximately 100 additional cases of en-
cephalitis reported to the CDC annually in historical studies BSE cases, suggesting animal to human transmission (5). The
(32,33). The disease historically produced approximately report of atypical CJD (unique clinical and histopathologic
one third of the encephalitis cases in the United States and findings) affecting young adults (an age at which CJD rarely
was associated with measles, mumps, and other exanthema- has been diagnosed) led to the designation of a new disease,
tous viral infections (1). Postinfectious encephalitis is now variant Creutzfeldt-Jakob disease (vCJD). From 1996 to
associated with antecedent upper respiratory viral infection 2011, there have been 224 cases of vCJD reported, with 175
(noticeably with influenza virus) and varicella in the United of these occurring in Ireland and Great Britain (World Health
States (32). Measles continues to be a leading cause of postin- Organization [WHO] Web site: www.who.int). The numbers
fectious encephalitis worldwide. In addition to the postin- have declined since the ban in the use of bovine offal in fer-
fectious process, patients with paraneoplastic syndrome and tilizers, pet food, or other animal feed. Active monitoring is
autoantibodies to the N-methyl-d-aspartate (NMDA) auto- still important, and detection of BSE continues to be reported
antibodies have also been recently described (34). Recent in North America (Fig. 4.2) (CDC BSE Web site: http://www.
studies suggest that antigenic variation in the N-terminal do- cdc.gov/ncidod/dvrd/bse/).
main of the NMDA receptor may predispose these patients Environmental factors influence infections of the CNS.
to the autoimmune encephalitis (35). The slow infections Changes in behavior, cultural beliefs, and modification of
of the CNS and transmissible spongiform encephalopathies the environment result in changes in disease patterns and
(TSEs) occur sporadically worldwide (5). The prototypical exposure to new infectious agents. Arboviral infections will
TSE is Creutzfeldt-Jakob disease (CJD); it occurs at high likely increase as populations encroach on wilderness habi-
rates within families and has an estimated incidence of 0.5 tats and flood plains (1). Vaccination has further changed the
to 1.5 cases per million populations. In 1986, cases of a TSE character of viral CNS disease. In 1952, poliomyelitis affected
in cattle, bovine spongiform encephalopathy (BSE), were re- 57,879 Americans (1). Widespread vaccination has eradicated
ported in the United Kingdom. In addition to affecting other the disease currently from the Western Hemisphere. As so-
livestock throughout Europe that were fed supplements con- cial and environmental changes occur globally, the character
taining meat and bone meal, cross-species transmission of and prevalence of CNS viral infections will also change. CNS
BSE has been documented, leading to a ban in the use of infections must be examined in a geographic, cultural, and
bovine offal in fertilizers, pet food, or other animal feed (5). environmental context as well as at the cellular, molecular,
Increases in atypical CJD cases coincided with the peak of and genetic levels.

Scheld_Ch04.indd 53 2/21/14 5:30 PM


54 Part II: Viral Infections and Related Disorders

7
N = 23
6

5

4

3

2

1
* #
0
1993 2003 2004 2005 2006 2007 2008 2009 2010 2011 2012
as of 04/24/2012
FIGURE 4.2 Monitoring and overview
U.S. Canada Canadaborn after 1997 feed ban Canadaunknown birth date
of BSE in North America. Columns rep-
resent number of cases of BSE in North
* Imported UK to Canada # Imported Canada to US

Born after March 1, 1999 America by country of origin and year.
(Courtesy of CDC and WHO.)

PATHOGENESIS be permissive, providing an adequate environment for viral


replication. The initial steps involved in hematogenous spread
of virus to the CNS consist of replication at the local site of
Viral Spread entry and primary viremia (1). Infection of a secondary tissue
frequently ensues, permitting secondary replication and an ex-
Viruses use two basic pathways with fundamentally different tensive viremia that seeds the CNS. Not all viruses follow this
steps to gain access to the CNS: hematogenous and neuronal. sequence, and genetic factors of both virus and host influence
Viruses must survive and multiply at the cellular level effi- the route of viral spread.
ciently and in sufficient quantity to infect the CNS. The mech- The cornified layers of dead skin cells provide a structural
anism of spread to the CNS is in large part determined by viral defense for the greatest potential infective surface area of the
factors, site of entry, and successful replication in intermediate human body. Layers of keratin protect the underlying epi-
cells (1). The local immune response at the site of entry, the thelium from viral contact, thereby decreasing the incidence
systemic immune responses, and the limited vascular access of viral entry (1). Breaks in this defensive layer can result in
afforded by the bloodbrain barrier further reduce the oppor-
tunity for viral neurologic infections (1). Differences in host
physiology and mechanism of spread to the CNS further influ-
ence the clinical manifestations of neurologic disease (1). For
example, adults with herpes simplex encephalitis (HSE) have
different presenting signs and symptoms than newborn babies Mouth Conjunctiva
with HSV infection of the CNS. The route of viral spread and Respiratory
areas of neurologic involvement differ based on the age of the tract
patient and mechanism of exposure (36). The subsequent neu- Scratch,
rologic damage and poor outcome, however, are similar (1). injury
Subtle differences at the epidemiologic, host, tissue, cellular,
and genetic levels can alter this balance between viral exposure Alimentary
and symptomatic infection. tract

Hematogenous Spread
Enteroviruses and arboviruses are prototypes for viremic
spread to the CNS. Although the location of viral entry dif-
fers for each family, both cause primary and secondary viremia
prior to infecting the CNS. Reviewing the necessary steps and Arthropod
Urogenital
the numerous barriers to hematogenous neurologic infection tract Capillary
explains the low incidence of symptomatic viral infection and
the even lower frequency of viral neurologic infections. A virus Anus
must first bypass or attach to and enter host epithelial cells to Skin
produce infection (37) (Fig. 4.3). In addition, the cell must FIGURE 4.3 Body surfaces as sites of virus infection and shedding.

Scheld_Ch04.indd 54 2/21/14 5:30 PM


Chapter 4: Pathogenesis and Pathophysiology of Viral Infections of the Central Nervous System 55

higher frequency of infection as well as more severe disease. opportunity for host eradication of viremia (1). Viruses infect
Some vector-borne viruses bypass the cornified epithelial layer tissues other than the liver and spleen, such as muscle, endo-
by inoculation into the subepithelial layer or directly into the thelium, and blood cells. These sites provide an environment
blood (1). The nonkeratinized epithelial layer that constitutes for viral replication in highly vascular locations that facilitate
the conjunctival, respiratory, oral, and nasopharyngeal sur- extensive viremia. Secondary viremia produces high titers
faces provides an ideal entry point for aerosolized viruses or of virus in the bloodstream for prolonged periods of time,
pathogens transmitted by large droplets. Parainfluenza and facilitating the seeding of target organs. Viral genetics and host
adenovirus, although uncommon, can cause primary encepha- physiology determine the location and extent of infection at
litis (1). More frequently, however, the respiratory viruses are these secondary sites (1).
associated with postinfectious encephalitis (32). A mucous Virus must localize in the vessels of the CNS before cross-
layer composed of mucopolysaccharides, secretory immu- ing the bloodbrain or bloodCSF barrier, a network of tight
noglobulins, and inflammatory cells provides a mechanical, endothelial junctions sheathed by glial cells that regulate mo-
chemical, and cellular defense against pathogens (1). In the lecular access to the CNS (43). The pathophysiology of viral
gastrointestinal and urogenital systems, constant transit pro- transport from blood to brain and of viral endothelial cell tro-
tects the mucosa. As in the respiratory mucosa, leukocytes pism is poorly understood. Virus infects endothelial cells, leaks
and secretory factors augment this mechanical defense. The across damaged endothelium, passively channels through en-
enteroviruses tolerate stomach acid, bile salts, proteolytic en- dothelium (pinocytosis or colloidal transport), or bridges the
zymes, and alkaline infusions to infect the host. Certain viruses endothelium within migrating leukocytes (1). Cell-associated
(coxsackievirus A9) actually require exposure to proteolytic and cell-free viruses can cross the endothelium and enter the
enzymes in the gut before they can infect select cell types (1). parenchyma or CSF. This bridging of the endothelium occurs
Once virus breaches the epithelial barrier and finds a per- in choroid plexus vessels, meningeal blood vessels, or cerebral
missive cell, primary replication occurs. Virus then can spread blood vessels (1) (Fig. 4.4). Once in the CSF, virus may remain
and replicate in the lymph node, or it can bypass the node limited to the meninges or may enter the brain parenchyma
and enter the circulatory system, where it seeds other tissues across either ependymal cells or the pial linings.
(arbovirus, enterovirus, measles virus, or varicella virus) (1).
Local immune responses are crucial in limiting systemic viral
infection. The generation of a swift inflammatory response
Neuronal Spread
can limit viremia and symptoms of infection. Some viruses Rabies and HSV infection are prototypes of viral CNS infections
resist phagolysosomal degradation, allowing them to circulate that access brain by peripheral neuronal spread. Historically,
and replicate within the protective sheath of a macrophage the peripheral neural pathway was considered the only path-
(38,39). Antigenic changes and the sequestration of viral re- way of viral neurologic infection. Experiments with HSV and
ceptors provide additional mechanisms that enable viruses rabies virus performed in the nineteenth and early twentieth
to evade lymphocytes. For example, human rhinovirus 14, centuries, combined with the discovery of the bloodbrain bar-
influenza virus, and poliovirus have receptors embedded in a rier at the turn of the century, led most investigators to con-
recess or canyon in the viral membrane (1). The virus is clude that all viral neurologic infections occurred by neuronal
able to evade the immune response by altering the molecules spread (1). Contemporary data, however, show that the blood-
on the surface surrounding the highly conserved, immunologi- stream provides the principal pathway for CNS infections in
cally inaccessible receptor molecules lining the canyon. Other humans. Some viruses (poliovirus and reovirus), previously
viruses have hypervariable sequences surrounding a small, thought to infect the CNS by the hematogenous route, have
molecularly conserved binding sequence. The viral binding been detected in peripheral neurons in experimental models
site may be smaller than the antigenic sequence recognized by (44). Viremia and neuronal spread to the CNS can occur con-
the immunoglobulin. Changes in the hypervariable molecules currently and are not mutually exclusive (1).
surrounding the binding site allow the virus to evade immune Neuronal spread occurs along peripheral or cranial
responses without disrupting the fidelity of the receptor bind- nerves. The nerve shields the virus from immune regulation
ing site (40,41). and allows access to the CNS. Rabies virus classically infects
Primary viremia allows virus to seed distant locations of by the myoneural route; however, infection has been docu-
the body and frequently marks the onset of clinical illness. mented from corneal transplantations, and aerosolized entry
Virus circulates in the vascular system attached to or within has occurred following spelunking in caves contaminated
host cells such or as free virus within the plasma (1). Viruses with infectious bat guano (29,45). These sources of infection
have limited access to the CNS via cerebral vessels and re- are infrequent and employ the same axonal mechanism of
quire sufficient numbers of progeny to overcome the improb- spread within the nerve, albeit from a different location than
ability of contact and entry into a permissive cell. In rare the myoneural route.
circumstances, such as disseminated neonatal herpes infec- Rabies virus replicates locally in the soft tissue following a
tion, virus infects the CNS after primary viremia. However, rabid animal bite, although entry into sensory nerves prior to
most infect an intermediate tissue prior to reaching the CNS. soft tissue replication has also been documented (1). Protection
Viral genes may be as important as host physiology in de- by antibody-mediated immune mechanisms in the soft tissue
termining the route and degree of viral dissemination. For provides the only known method of preventing neurologic dis-
example, the reovirus S1 gene determines the mechanism ease and death (29). After primary replication, the virus en-
of viral spread in the host. The S1 gene codes for a capsid ters the peripheral nerve. Experimental evidence demonstrates
hemagglutinin, 1, that binds to neuron receptors. Serotypes acetylcholine receptor binding as the mechanism of myocyte
with an intact 1 gene spread to the CNS by neuronal path- entry (1). However, viruses have also been documented in cells
ways, whereas 1-deficient mutants gain access to the CNS lacking these receptors. Once in the muscle, the virus buds
via the hematogenous route (42). from the plasma membrane and may cross myoneural spin-
The liver and spleen provide ideal locations for secondary dles or enter the nerve by the motor endplate. The virus then
viral replication because of their highly vascular structure. travels by anterograde and retrograde intraaxonal transport
The high degree of parenchymal contact and large number of to infect neurons in the brainstem and limbic system. Viruses
fixed mononuclear macrophage cells also provide an excellent appear to cross the transsynaptic space between neurons by

Scheld_Ch04.indd 55 2/21/14 5:30 PM


56 Part II: Viral Infections and Related Disorders

Brain substance
CSF
Cerebral blood vessel
Blood vessel in
choroid plexus

Ventricle

Nerve
Pia

CSF

Ependyma

From peripheral
nerve ending or
nasal mucosa Meningeal blood FIGURE 4.4 Routes of viral invasion of
or dorsal CSF vessel the central nervous system. CSF, cerebro-
root ganglion spinal fluid.

passive transport rather than receptor-mediated transport. viral neuroinvasion and neurovirulence. For example, reovirus
Recent evidence suggests rabies virus enters projections in the types 1 and 3 produce different CNS diseases in mice based on
postsynaptic neuron that extend into invaginations on the pre- serotype differences in receptor affinities (1,42). Escape mutant
synaptic side. These projections pinch off and fuse with the B4 of tick-borne encephalitis (TBE) virus also demonstrates
presynaptic membrane, allowing the virus to spread along viral differences in mouse neuroinvasiveness. A single amino
motor or sensory neural pathways (1,45). acid substitution (Tyr to His) in domain 2 of viral surface
Paresthesias near the location of the animal bite and protein E eliminates viral neuroinvasiveness without affecting
change in behavior follow over the next weeks. These signs neurovirulence (1). Receptor difference is only one determi-
and symptoms correlate temporally with the axoplasmic nant of viral neurotropism. Other viral factors may influence
transport of virus and infection of the brainstem and hippo- neurotropism. For example, enteroviruses in the same recep-
campal region (1). The infection spares cortical regions during tor family produce very different diseases. Coxsackieviruses
this phase, allowing animals to vacillate between periods of B1 through B5 readily produce CNS infections, whereas type
calm, normal activity and short episodes of rage and disorien- B6 rarely produces neurologic infection. Viral genes influence
tation (45). Eventually, the virus spreads from the diencephalic neurovirulence of HSV-1. Mutant HSV-1 viruses with either
and hippocampal structure to the remainder of the brain, 134.5 gene deletions or stop codons inserted into the gene
killing the animal. Experimental rabies infections in animals have a decreased ability to cause encephalitis and death fol-
demonstrate that the mode of acquisition influences the neu- lowing intracerebral inoculation in mice as compared with
roanatomic location of initial infection (1). wild type virus (46,47). Upon entering mouse neuronal cells,
Viruses also infect the CNS through cranial nerves. The these 134.5 () mutants trigger the shutdown of protein syn-
olfactory system is unique among cranial nerves in that the thesis and elicit interferon signaling responses that limit ef-
neurons regenerate and have approximately a 1-month life ficient viral replication (48).
span. The olfactory neurons are not protected by the blood Host physiology is also important in determining the
brain barrier, theoretically providing direct neuronal access extent and location of viral CNS disease. Age, sex, and ge-
to the brain (1). Animal studies have shown that HSV can netic differences between hosts influence viral infections and
infect the brain through the olfactory system as well as the clinical course. With respect to HSV infection, host muta-
trigeminal nerve. Moreover, the inferomedial temporal lobe, tions in pattern recognition receptors important for type I
the initial location of early HSV encephalitis, contains direct interferon production predispose patients to HSE (49). Host
connections with the olfactory bulb. The association of viral age influences the clinical manifestations and sequelae of
latency in the trigeminal ganglia, the relative infrequency of a viral infection (50). Differences in outcome are twofold:
HSE, and the confusing data regarding encephalitis from HSV mature neurons resist virally induced apoptosis, and younger
reactivation suggest that the pathogenesis is more complex patients can have more immature immune response to infec-
than described earlier (1). tion (51,52). Differences in macrophage function can alter
infections and disease. Moreover, macrophage processing
capacity can change with age in humans (1,38). Enteroviral
Host and Viral Factors Influencing infections exemplify the difference that host physiology plays
Neurotropism in determining the extent of viral disease. Enterovirus infec-
tions in children younger than 2 weeks of age can produce
As illustrated in Tables 4.3 and 4.4, viruses exhibit differences a severe systemic infection, including meningitis or menin-
in neurotropism (1). Strain and serotype differences influence goencephalitis (53). Ten percent of neonates with systemic

Scheld_Ch04.indd 56 2/21/14 5:30 PM


Chapter 4: Pathogenesis and Pathophysiology of Viral Infections of the Central Nervous System 57

TA B L E 4 . 3
PROPOSED NEUROVIRULENCE FACTORS

Virus Viral Factor Effect

HSV-1 Viral 134.5 gene Decreased viral replication in neuronal tissue and
protein synthesis shutdown
Viral gE and gI glycoproteins Mutant viruses lacking these receptors do not
cause encephalitis
VZV Host cell-mediated immunosuppression Increased frequency of herpes zoster, visceral, and
disseminated cutaneous disease
CMV Viral Fc receptor protein Immune evasion: bind neutralizing antibodies
Virus binds 2-microglobulin Immune evasion
EBV Viral glycoproteins gp220 and gp350 Infect lymphocytes
Lassa fever virus Viral polymerase Changes the function of endothelium and platelets
Sindbis virus E2 envelope protein Neurovirulence receptor protein
Reovirus Viral proteins 1, 3, 1 Receptor proteins that influence CNS infection
Rabies virus Viral protein gG Major antigenic determinant, essential for
pathogenicity
Enterovirus Nucleotide 472 in the noncoding region Neurovirulence gene
Capsid proteins VP1 and VP3 Single amino acid changes in the capsid proteins
reduce neurovirulence
Measles virus M protein Deficiency of this protein inhibits release of virus
and is associated with SSPE
Tickborne encephalitis (TBE) virus Glycoprotein E (domain B) Neuroinvasiveness receptor domain
Murray Valley encephalitis virus Glycoprotein E (domain B) Neuroinvasiveness receptor domain
Louping ill virus Glycoprotein E (domain B) Neuroinvasiveness receptor domain

CMV, cytomegalovirus; SSPE, subacute sclerosing panencephalitis; VZV, varicella-zoster virus.

enteroviral infections die, and as many as 76% are left with poliomyelitis and may result in an increased incidence of
permanent sequelae (1). In older children, however, entero- enteroviral myocarditis and aseptic meningitis (1,54). The
viral infections produce less severe disease. In addition to frequency of infections in groups frequently reflects epidemi-
age, physical activity may be another important host fac- ologic differences in exposure. Increasingly, host differences
tor that determines the severity of infection. Exercise and are recognized as equally important determinants of disease
trauma have been associated with increased risk for paralytic at the cellular and molecular levels.

TA B L E 4 . 4
VIRAL NEUROTROPISM

Virus CNS Location

Herpes simplex virus (postnatal) Neurons in the inferomedial temporal lobe,


orbitofrontal areas
Varicella-zoster virus (postinfectious) White matter of the cerebellum
Cytomegalovirus Neurons, glial, ependymal, and subependymal cells
Epstein-Barr virus Cerebral blood vesselsvasculitis
Lassa fever virus Choroid plexus
Japanese encephalitis virus Brainstem and basal ganglia in humans, retinula cells
in mosquitoes
Rabies virus Neurons in the hippocampal, cerebellar,
mesencephalic areas
Mumps virus Ependymal and choroid plexus cells
Polyomavirus (immunosuppressed) Oligodendrocyte damage in subcortical white matter
Poliovirus Motor neurons in the anterior horn of the spinal cord
and brainstem

Scheld_Ch04.indd 57 2/21/14 5:30 PM


58 Part II: Viral Infections and Related Disorders

Central Nervous System Physiology Arachnoid

The bloodbrain barrier limits chemical and environmental Subarachnoid


exposure to the CNS by a series of tight endothelial junctions space
bound and maintained by glial cell foot processes. This barrier
Pia
provides a physiologic boundary between the metabolically
sensitive neuronal cells and the chemical changes outside the Subpial
CNS (43). In addition, the endothelial cells and tight junctions space
Blood vessel
provide a physical barrier to most pathogens, limiting viral
access to the CNS. As with most biologic systems, the blood Glia
limitans Perivascular
brain barrier is more complex and heterogeneous than previ- macrophage
ously imagined. The bloodbrain barrier was first described
during the late nineteenth and early twentieth century, when Virchow-Robin
scientists noted that various dyes administered intravenously space
failed to penetrate the CNS (1). In the 1960s, experiments
certified that the tight junctions between endothelial cells
lining the cerebral vessels blocked the passage of small protein FIGURE 4.5 The relationship of the Virchow-Robin space to the sub-
molecules. pial and subarachnoid space.
The tight junctions between endothelial cells provide
a relatively impermeable layer to most polar substances.
Unique transport systems and enzymes further distinguish
the CNS capillaries from blood vessels in other organs. The
asymmetric distribution of transport proteins in the endothe- The brain is an immunologically privileged site into
lial cell membrane creates a highly resistant, polarized cell which immune cells do not readily enter. Increasingly, scien-
layer that limits paracellular diffusion (55). Hydrophilic sub- tists are discovering that immune cells reside in and circulate
stances cross the endothelial layer through receptor-mediated through the Virchow-Robin space, a lymphatic channel lining
endocytosis or through highly specific, saturable transport the perivascular space in the brain (1) (Fig. 4.5). Moreover,
systems. Respiratory gases and lipophilic chemicals passively many of the fixed glial support cells and pericytes surrounding
penetrate the layer of tight junctions readily. The cerebral the vessels in the CNS can transform to monocyte/macrophage
vessel endothelial cells also possess second-messenger mole- antigen-presenting cells. The circulating lymphocytes act as
cules that may regulate transmembrane permeability through surveillance cells, detecting small amounts of antigen presented
receptor binding (55). Substances produced during infection by the macrophages in the perivascular space and initiating the
or chemicals secreted by cells, such as histamine and inter- immune response either within the Virchow-Robin space or
leukins, change the permeability of the bloodbrain barrier, peripherally at the lymph node. During periods of infection,
thus modulating entry of viruses and immune cells into the immune cells readily enter the CNS and fill the Virchow-Robin
CNS. Astrocytes are metabolically important support cells space (1) (Fig. 4.6). The perivascular space provides a staging
of mononuclear macrophage origin that surround cerebral area where lymphocytes interact chemically and differentiate
capillaries, induce tight junctions, and may regulate immune prior to entering the neuropil. Cells in the perivascular space
cell entry (1). as well as cerebral capillary endothelial cells are capable of

Virchow-Robin space Enlarged Virchow-Robin


around CNS blood vessel- space around
resting state CNS blood vessel-
state during reaction
to intra-CNS immune
Some macrophages stimulation
containing antigen
migrate to lymph node
T cells stimulated
to respond
T and B
lymphocytes
activated

Virchow-Robin space Macrophage

T cells Plasma cells


Perivascular
macrophage

Rare Microglia
lymphocyte
Plasma cells

FIGURE 4.6 Illustration of the Virchow-Robin space as an immunologic space that can become expanded
with immunocompetent cells interacting together under conditions of immunostimulation in the brain.

Scheld_Ch04.indd 58 2/21/14 5:30 PM


Chapter 4: Pathogenesis and Pathophysiology of Viral Infections of the Central Nervous System 59

regulating T-helper cell subsets in vitro and may influence the


expression of the immune response, dictating which cells enter Attachment and Entry
the CNS. Different viruses may activate characteristic lympho-
Attachment is an essential first step in viral infection. Multiple
cyte subsets for entry into the parenchyma. In some cases, the
copies of proteins line the surface of a virus. These capsid or
immune response is instrumental in the pathogenesis of CNS
envelope proteins create high-affinity bonds with host recep-
damage (1,51,56,57).
tors and initiate viral infection or a host cell response (58,59).
Classically, this response involves viral entry but may include
a change in cellular metabolism or generation of immune
VIRAL REPLICATION IN THE responses by the cell. Temperature, pH, receptor affinity, and
CENTRAL NERVOUS SYSTEM the concentration of viral and host receptors influence the
hostviral receptor interaction similar to a receptor-ligand
The fundamental principles of viral replication and cell-to-cell reaction. The cell receptor consists of proteins, lipids, and/
spread provide a framework for examining the pathogenesis or oligosaccharides. Receptor binding provides close contact
and clinical repercussions of neurologic infections. The clini- between virus and cell, facilitating but not ensuring viral entry
cal manifestations and the severity of illness reflect the loca- into the cell. Some viruses require specific chemical or proteo-
tion and extent of viral replication in the CNS. Once virus lytic conditions before entering the cell (6062). For example,
accesses the CNS, it must introduce its genome and transport Semliki Forest virus requires the presence of cholesterol in the
proteins into the cytoplasm or the nucleus of the mammalian cell membrane as well as a pH change in the endosome for
cell. Once the viral genome has been uncoated, transcription entry (63). The presence of one type of receptor for cell entry
and translation proceed in a predictable and organized cas- does not preclude other mechanisms of entry into a host cell.
cade of gene expression, culminating in the replication of the Viral entry into the cell is essential. Although receptors have
viral genome. Translation of late viral genes produces struc- been identified, alternative entry mechanisms are being identi-
tural proteins essential for the construction of the next gen- fied for viruses (64). Studies determining the structure of viral
eration of viruses. Viral genomic material is packaged with glycoproteins and host receptor interactions as well as experi-
structural proteins and exits the cell (1) (Fig. 4.7). Viruses ments using viral recombinants and cell lines expressing cellular
exploit essential cell activities such as protein synthesis, in- receptors provide two methods used to characterize viral entry.
tracellular transport, and cellular communication to enter the Viruses can bind nonspecifically to the cell surface; however,
cell and replicate their genome. As in other biologic systems, these nonspecific interactions do not produce a biologic re-
both divergent and convergent evolution has resulted in an sponse. Viruses frequently target essential and/or tightly con-
array of mechanisms for successful viral reproduction. As a served host receptor domains. Some viruses appear to interact
result, numerous strategies exist for viral entry, gene expres- with neurotransmitter receptors in the CNS. Experimental data
sion, replication, assembly, and egress (1). The relative speed indicate that rabies virus binds to acetylcholine receptors on
and efficiency with which the virus replicates determine the mouse myocytes (1). Reovirus 3 binds to the -adrenergic recep-
progression of infection. tor. Viruses also bind to immunologic proteins on the surface of
cells. Poliovirus, HSV, and measles virus bind to receptors in the
immunoglobulin superfamily (65,66). Hormone and cytokine
receptors provide additional targets for viral cell entry. Viruses
can have more than one mechanism for entering a cell or differ-
ent receptors for different cell types (1). The number and distri-
1 Attachment bution of receptors help determine viral tissue tropism and the
extent of viral CNS disease. Receptor prevalence is not the only
determinant of viral tissue tropism (1). Transgenic mice, for
2 Penetration (endocytosis) example, develop poliovirus infection only in limited tissue sites
despite the widespread expression of the receptor. Some viruses
3 Uncoating require the presence of certain genes and transactivating factors
to infect a cell. While a cell may contain a certain receptor, a
4 Transcription of mRNA permissive environment for viral replication may not exist. The
tissue, in such a case, is resistant to infection (67).
5 Translation of early proteins Enveloped viruses have different mechanisms than nonen-
veloped viruses for cell entry. Once the virus binds to the host
6 Replication of viral DNA cell receptor domain, the virus can enter the cell by direct fu-
sion or receptor-mediated endocytosis. The receptor-bound
7 Transcription of mRNA virus frequently becomes encased in a clathrin-coated pit during
endocytosis. Other modes of endocytosis exist, and virus has
8 Translation of late proteins been found in uncoated vesicles (1,68). Fusion proteins contain
hydrophobic regions and initiate the union of viral and cell mem-
9 Assembly of virions branes in some enveloped viral infections. Nonenveloped viral
entry is more enigmatic. Conformational changes or proteolytic
cleavage may expose hydrophobic regions of capsid proteins,
enabling the protein capsid to fuse with or embed in the cell
membrane. The capsid then opens and releases the viral genome
10 Release by budding into the cytoplasm. Endocytosis may provide the pH change or
enzymes necessary for viruscell fusion and ensures that the cell
FIGURE 4.7 The viral multiplication cyclea stylized and greatly is metabolically viable. Furthermore, it has been suggested that
simplified diagram summarizing the key steps in the multiplication of endocytosis delivers the viral genome to the proper intracellular
a typical DNA virus. (From Fenner F, White DO. Medical Virology. location from which replication occurs. Viral fusion, in most
2nd ed. Orlando, FL: Academic Press; 1986, with permission.) cases, occurs before the endosome fuses with the lysosome.

Scheld_Ch04.indd 59 2/21/14 5:30 PM


60 Part II: Viral Infections and Related Disorders

glycosylation (72). Viruses contain regulatory proteins and


Replication and Egress promoter sequences that control the differential expression of
transcripts. Proteolytic modifications are made to the struc-
Viral replication begins after uncoating and delivery of the tural protein following attachment of fatty acids and oligo-
genome to a satisfactory intracellular location (68). Viruses saccharides in the Golgi apparatus (1). In some cases, these
replicating in the nucleus often contain nuclear targeting sig- proteolytic changes are necessary for producing infectious
nals and can use existing cellular mechanisms to enter the progeny. TBE virus, for example, requires cleavage of a mem-
nucleus (1). Alternatively, viruses that replicate in the cyto- brane-bound precursor protein (prM). The proteolytic change
plasm uncoat and deliver the genome to the perinuclear area produces a small, membrane-bound protein (M protein) that
(68). The production of positive stranded viral messenger RNA protects another membrane-bound protein from conforma-
(mRNA) and the subsequent translation of gene products pro- tional changes in the acidic secretory pathway. Viruses that
vide a unifying pathway for viral infections (Fig. 4.8). Viruses contain the uncleaved prM moiety lack fusion capability and
use either host enzymes or specialized viral polymerases car- are noninfectious (1).
ried with or encoded by the viral genome. Replication of the viral genome involves the synthesis of
Host protein synthesis decreases at the start of viral protein full-length, complementary genomic transcripts that act as
synthesis in most viral infections. Viruses have unique mecha- templates for replication of the viral genome. The efficiency
nisms for inhibiting protein synthesis and can interfere with and fidelity of genomic replication influence the likelihood of
the translation, transport, ribosomal binding, or stability of disease. Defects in the viral genome cause abortive replication
host transcripts (1). With some viruses, premade proteins and or result in conditionally defective viruses that multiply only in
synthesized viral gene products decrease the transcription of the presence of cells or viruses carrying complementary genes.
host mRNA (6971). Cellular transport of mRNA out of the The newly synthesized progeny genomes are transported
nucleus is inhibited late in adenovirus infections. Viral mRNA to capsid structures, where they enter viral capsid shells.
copies can outnumber host mRNA or can be more efficiently Enveloped viruses bud from the cell membrane, whereas non-
transcribed, thus restricting access to ribosomes (1). For ex- enveloped viruses exit the cell by lysis (1).
ample, poliovirus inactivates the host cap-binding protein.
This alters the cells ability to modify transcripts and results
in less efficient translation of host proteins (1). Degradation
of host mRNA is another mechanism used by some HSV to Viral Spread in the Central Nervous System
inhibit host protein synthesis (71). Some viral gene factors act
as repressors and inhibit host mRNA export (70). Viral disease of the CNS requires cell-to-cell spread of the
Viral protein translation occurs in a stereotyped progres- virus. The densely packed neuropil provides a unique envi-
sion. Early gene expression regulates the transcription and ronment with limited extracellular space for viral dispersion.
translation of the remaining viral genome, inhibits host pro- Viruses can spread through the CNS in four prototypical ways:
tein and nucleic acid synthesis, and codes for enzymes neces- (a) sequential cellular infection, (b) movement in the extracel-
sary for viral nucleic acid replication (1). After viral nucleic lular space, (c) neuronal axoplasmic transport, or (d) transit
acid replication, late viral genes are selectively expressed and via migrating lymphocytes or glial cells. Viruses may spread
transcribe templates for capsid and structural proteins nec- within the neural tissue using more than one mechanism.
essary for virion assembly. Proteins synthesized from viral Few viruses infect the CNS by contiguous cell-to-cell spread.
transcripts can undergo posttranslational modification and Sindbis virus provides one example of a virus that spreads
from ependymal cells directly to glial and neuronal cells in
experimentally infected mice (1). Viruses exhibit cell tropism,
frequently infecting one cell type more readily than another.
 DNA For example, HSV-1 infects neurons early during encephali-
Parvovirus tis but is not present in glial cells until late in the infection.
Herpesvirus spreads in the nervous system via axoplasmic
transport in neurons (73,74). Electron microscopy has dem-
 RNA  DNA DNA onstrated togaviruses within extracellular space in the CNS.
Retrovirus Some viruses enter the CNS through a Trojan horse mecha-
Papovavirus nism via leukocytes (1,75).
Adenovirus
Herpesvirus
Poxvirus
Hepadnavirus Host Defense and Immunopathogenesis
Intrinsic and systemic antiviral defenses limit viral replica-
Picornavirus
RNA
tion and infection (44,51,76). Viral replication can activate
Togavirus  mRNA
Coronavirus Reovirus enzymatic pathways that degrade viral nucleic acid tran-
scripts. Other cells undergo apoptosis, creating a nonviable
Orthomyxovirus environment for the virus (1). Interferon-mediated intrinsic
Paramyxovirus antiviral pathways within cells can retard viral penetration,
Arenavirus uncoating, transcription, translation, and assembly, represent-
Bunyavirus ing an important factor of host resistance to viral infection
Rhabdovirus (1,7779). Interferonstype I (interferon-
and interferon-),
type II (interferon-), and type III (interferon- )are secreted
by distinct cells, bind to different receptors, and represent
 RNA evolutionarily distinct molecules that limit viral replication
FIGURE 4.8 Six basic strategies for transcribing messenger RNA from (44,8084). Interferons activate a cascade of enzymes and ki-
different types of viral genome. (From Baltimore D. Expression of ani- nases that inhibit protein synthesis at different steps in the syn-
mal virus genomes. Bacteriol Rev. 1971;35:235, with permission.) thetic pathway. Interferons also modify the binding properties,

Scheld_Ch04.indd 60 2/21/14 5:30 PM


Chapter 4: Pathogenesis and Pathophysiology of Viral Infections of the Central Nervous System 61

electrostatic charge, and receptor expression (major histocom- of vCJD and most patients die less than a year after onset of
patibility complex [MHC] antigen, 2-microglobulin, and Fc their neurologic manifestations.
receptor) of cellular membranes, further restricting viral access These encephalopathies differ in mode of transmission.
and replication (1). These cytokines can enhance or suppress Although most of the TSEs are experimentally transmissible
expression of immune cell subsets (82,85). Although interferon by direct inoculation in the CNS, this mode rarely occurs
can protect host cells from viral infection, some pathogens except for iatrogenic transmissions (1). The scrapie agent
have developed resistance. Furthermore, the inflammatory re- spreads by contact and lateral transmission. There is no evi-
sponse in some cases causes damage to tissue and constitutes a dence for lateral transmission in the case of BSE or vCJD,
pathogenic mechanism for viral disease (57). and all cases appear to have occurred following parenteral
The presence of viral envelope proteins in the host cell or ingestion of affected materials. The transmissible agents
membrane elicits an immunologic response. The host immune remain infectious after treatments that would normally in-
response targets and destroys the infected CNS cells, thus lim- activate viruses or nucleic acids (detergent formalin, ionizing
iting spread of the virus but potentially compounding disease. radiation, nucleases). Most of the experimental work on TSEs
For example, rabies virus causes metabolic derangement in the has involved analysis of the scrapie agent. The current work-
neuron, usurps the cellular metabolic machinery, and inhibits ing model is that posttranslational alteration of the normally
the synthesis of cellular proteins (1). The actual pathogenic
-helical form of the PrP protein results in a protease resis-
cause of neuronal cell death is not known but may involve tant -pleated sheet structure that accumulates in neurons,
the synthesis of toxic metabolites by rabies virus. The immune leading to progressive dysfunction, cell death, and subsequent
response changes the character of disease and the pathologic astrocytosis. In studies on the scrapie agent, gastrointestinal
findings. In paralytic or dumb rabies, patients have disease lim- tract involvement with infection of abdominal lymph nodes
ited to the brainstem and demonstrate reduced B-cell, interleu- occurs first, followed by brain involvement a year or more
kin, and cellular activity in response to rabies antigens (86,87). later. Experimental subcutaneous inoculation in mice and
Patients with furious or classic rabies generate brisk, late intra- goats also lead to local lymph node involvement followed
cranial immune responses to rabies antigens. An experimental by splenic spread and then CNS involvement. The mode of
model, involving immunosuppression, demonstrates that this transmission to the CNS (direct vs. hematogenous) or the in-
late immune response compounds CNS damage in infected fectivity of body fluids at different stages of infection is not
animals (1). known at this time.
In postinfectious encephalitis, the immune response is misdi- The TSEs are currently only diagnosed by histologic ex-
rected against the brain itself. There is no evidence of direct viral amination, characteristic electroencephalography (EEG),
damage or viral antigens in the CNS (1). Viral antigens can share magnetic resonance imaging (MRI) changes, and the clini-
homology with host proteins, and the ensuing immune reaction cal context. Most laboratory tests are of little value in the
can damage normal host tissue resembling virally infected cells diagnosis. CSF examination shows normal values or slightly
(7,88). Immune deregulation may cause immune-mediated elevated protein levels. The EEG in classic CJD reveals gen-
demyelination. For example, most patients with (post-Semple) eralized slowing early in the disease, punctuated by biphasic
rabies vaccine encephalitis have antibodies against myelin basic or triphasic peaks late in the disease with the onset of myoc-
protein. Forty-seven percent of people with postinfectious lonus. MRI changes late in the illness reveal global atrophy
measles encephalitis have lymphocytes directed against myelin with hyperintense signal from the basal ganglia (5). Fluid-
basic protein, as compared with a 15% rate in nonencephalitic attenuated inversion recovery (FLAIR) MRI provides greater
patients with measles (1). The pathogenic mechanism of postin- sensitivity and demonstrates signal intensity changes in the
fectious encephalitis is not fully understood. cortex that are undetectable by T2-weighted spin-echo MRI.
HIV infection is associated with a variety of CNS diseases. Histopathologic examination of the brain using a specific an-
Patients can develop a leukoencephalopathy with diffuse glio- tibody to the PrP-res protein confirms the disease. In addition,
sis and loss of the cerebral white matter in addition to the evidence of gliosis, neuronal loss, and spongiform changes
opportunistic infections and neoplasms associated with the support the diagnosis. In cases of vCJD, characteristic amy-
disease (51,56,89,90). Pathologic specimens show a multifocal loid plaques (so-called florid plaques) microscopically define
accumulation of giant cells with focal cerebral necrosis. PCR the disease. The florid plaques are not seen in other TSEs and
in tissue samples demonstrates large amounts of HIV nucleic consist of flower-like amyloid deposits surrounded by vacu-
acids in multinucleated giant cells. The viral structural and/ olar halos. The detection of PrP-res in the tonsillar tissue by
or regulatory proteins may be toxic to the CNS tissue (91). immunohistochemical staining is also strongly supportive of
Alternatively, macrophages and T lymphocytes may dam- vCJD diagnosis (5).
age the brain by aberrant secretion of interleukin and tumor
necrosis factor (1).
Clinical Correlates to Disease
Transmissible Spongiform Encephalopathies Patients with encephalitis have clinical and laboratory evidence
of parenchymal disease. Some viruses (rabies, B virus) produce
The TSEs produce clinical changes related to CNS dysfunc- encephalitis without significant meningeal involvement; how-
tion similar to the encephalitides (1). Unlike encephalitis, the ever, most patients with encephalitis have concomitant men-
TSEs are slowly progressing noninflammatory CNS diseases ingitis (1). Most patients also have a prodromal illness with
with long incubation periods involving the accumulation of myalgias, fever, and anorexia reflecting the systemic viremia.
an abnormal form of a normal glycoprotein, the prion protein Neurologic symptoms can range from fever, headache, and
(PrP) (92). Sporadic CJD occurs between the ages of 50 and subtle neurologic deficits or change in level of consciousness
70 years and is characterized by dementia, tremors, and more to severe disease with seizures, behavioral changes, focal neu-
rarely abnormal movements and ataxia. Unlike sporadic CJD, rologic deficits, and coma (93,94). Clinical manifestations
vCJD disease affects young adults and adolescents and pro- reflect the location and degree of parenchymal involvement
duces cerebellar ataxia and sensory involvement (dysesthesias) and differ based on viral etiology. For example, HSE infects
with florid amyloid plaques detected in the brain on autopsy the inferomedial frontal area of the cortex, resulting in focal
(5). Neurologic deterioration progresses relentlessly in the case seizures, personality changes, and aphasia. These symptoms

Scheld_Ch04.indd 61 2/21/14 5:30 PM


62 Part II: Viral Infections and Related Disorders

reflect the neuroanatomic location of infection with inflamma- The age, immune status, and viral etiology also influ-
tion near the internal capsule, limbic, and Broca regions (1). ence the clinical manifestations of viral meningitis (51,100).
Paresthesias near the location of the animal bite and change in Patients with enterovirus meningitis often present with non-
behavior correlate temporally with the axoplasmic transport specific symptoms such as fever (38 to 40C) of 3 to 5 days
of rabies and the viral infection of the brainstem and hippo- duration, malaise, and headache (8,101). Approximately
campal region (94). Rabies has a predilection for the limbic 50% of patients have nausea or vomiting. Although nuchal
system, producing personality changes. The damage spares rigidity and photophobia are the hallmark sign and symptom
cortical regions during this phase, allowing humans to vacillate for meningitis, 33% of patients with viral meningitis have
between periods of calm, normal activity and short episodes of no evidence of meningismus. Fewer than 10% of children
rage and disorientation (1). Alternatively, Japanese encephali- younger than 2 years develop signs of meningeal irritation.
tis virus initially produces a systemic illness with fever, malaise, Most of these children with meningitis present with fever and
and anorexia, followed by photophobia, vomiting, headache, irritability. Children may also present with seizures secondary
and changes in brainstem function. Brainstem encephalitis to fever, electrolyte disturbances, or the infection itself (1). The
leads to difficulty with autonomic functions with increased clinician must have a high index of suspicion for meningitis
risk for cardiac and respiratory instability, reflecting infection especially in younger patients. In the immunocompromised
of brainstem nuclei (1,9597). Other patients have evidence of host, enterovirus infection is both a diagnostic quandary and
multifocal CNS disease involving the basal ganglia, thalamus, a potentially life-threatening disease. Immunocompromised
and lower cortex and develop tremors, dystonia, and parkin- patients frequently do not mount a brisk immune cell response,
sonian symptoms. and therefore CSF analysis may underrepresent the extent of
Seizures are frequent during encephalitis. For example, CNS involvement.
approximately 40% of patients with HSE develop seizures Symptoms of meningitis (nuchal rigidity, headache, and
(1). EEG patterns include focal slowing, spiking, and par- photophobia) occur in approximately 11% of men and 36%
oxysmal lateralizing epileptiform discharges. The cellular of women with primary HSV-2 genital infection (1,102104).
mechanisms for seizures are incompletely understood. This Examples exist of recurrent HSV-2 meningitis (with or with-
may result from dysfunction of the smaller, inhibitory, - out genital lesions), although cases associated with primary
aminobutyric acid (GABA)secreting neurons. Although the infection are more common (105,106). HSV meningitis may
seizures encountered in patients with HSE could be directly spread to the CSF by neuronal spread along the sacral nerves.
attributed to cellular destruction, an alternative hypothesis Alternatively, the virus may reach the CSF by hematogenous
for epileptogenesis in HSE centers on the uptake of virus in spread, as the virus has been cultured from the blood buffy coat
the long projections of neurons. This uptake causes pertur- layer. VZV, cytomegalovirus, Epstein-Barr virus (EBV), and
bations in the cellular machinery necessary for the retention parainfluenza virus have all been cultured or detected by PCR
of acetylcholine within the nerve terminal. As a result, the from the CSF of patients with meningitis (1). The three herpes-
excitatory neurotransmitter could leak from the cell and ul- virus infections occur more frequently in immunocompromised
timately trigger a seizure focus. In addition to this mecha- patients and rarely produce isolated meningitis. Instead, these
nism, suboptimal uptake of acetylcholine by malfunctioning infections usually progress and involve the parenchyma.
presynaptic and postsynaptic terminals can result in a rela-
tive excess of the neurotransmitter and abnormal electric dis-
charges. An excess of acetylcholine could also result from the
decreased synthesis of degradatory enzymes (such as acetyl- CONCLUSION
cholinesterase) as viral replication proceeds. Finally, chronic
seizure foci are known to be hypermetabolic during interictal Clinical symptoms produced by a disease have a pathophysio-
periods. The first stage of viral cellular infection is the in- logic basis. An understanding of the pathogenesis of viral CNS
hibition of the cells homeostatic mechanisms. The crippled disease provides the physician with a framework for studying
cell, unable to maintain homeostasis, may be predisposed to related neurologic diseases. Moreover, the pathogenic mecha-
disordered electric discharges (1). nism of a viral disease provides clues toward the development
Encephalitis, unlike meningitis, has higher mortality and of antiviral medications and strategies for the prevention of
complication rates. Case-fatality rates differ based on the viral viral CNS infections. Improved diagnostic techniques are
etiology and host factors. For example, within the arthropod- essential for advancing both research and therapy of viral
borne viral encephalitides, St. Louis encephalitis virus has an neurologic infections. Application of viral PCR and other
overall case mortality rate of 10%. The mortality rate is only molecular diagnostic techniques have already changed some
2% in children but increases to 20% in the elderly (1). Similarly, of the fundamental concepts of viral infection. Basic research
WNV meningoencephalitis produces greater mortality rates in in neurosciences and infectious diseases will result in a better
the elderly than in younger adults (98,99). Other viruses like understanding of the hostvirus interaction in the CNS. These
western equine and eastern equine encephalitis produce higher advances have the potential for improving the care of patients
mortality and morbidity in children than in adults (1). with neurologic diseases.

References
1. Cassady KA, Whitley RJ. Pathogenesis and pathophysiology of viral central 3. Bale JF Jr. Human herpesviruses and neurological disorders of childhood.
nervous system infections. In: Scheld WM, Whitley R, Marra MC, eds. Semin Pediatr Neurol. 1999;6(4):278287.
Infections of the Central Nervous System. 3rd ed. Philadelphia: Lippincott- 4. Rotbart H. Enteroviral infections of the central nervous system. Clin Infect
Raven; 2004:5774. Dis. 1995;20:971981.
2. Whitley RJ, Cobbs CG, Alford CA Jr, et al. Diseases that mimic her- 5. Whitley RJ, Macdonald N, Ascher DM. Technical report: transmis-
pes simplex encephalitis. Diagnosis, presentation, and outcome. NIAD sible spongiform encephalopathies: a review for pediatricians. Pediatrics.
Collaborative Antiviral Study Group. JAMA. 1989;262(2):234239. 2000;106(5):11601165.

Scheld_Ch04.indd 62 2/21/14 5:30 PM


Chapter 4: Pathogenesis and Pathophysiology of Viral Infections of the Central Nervous System 63

6. Cassady KA, Whitley RJ. Viral central nervous system infections. In: 38. Rios M, Zhang MJ, Grinev A, et al. Monocytes-macrophages are a poten-
Richman DD, Whitley RJ, Hayden FG, eds. Clinical Virology. 3rd ed. tial target in human infection with West Nile virus through blood transfu-
Washington, DC: ASM Press; 2009:2944. sion. Transfusion. 2006;46(4):659667.
7. Stuve O, Zamvil SS. Pathogenesis, diagnosis, and treatment of acute dis- 39. Johnston LJ, Halliday GM, King NJ. Langerhans cells migrate to local
seminated encephalomyelitis. Curr Opin Neurol. 1999;12(4):395401. lymph nodes following cutaneous infection with an arbovirus. J Invest
8. Lee BE, Davies HD. Aseptic meningitis. Curr Opin Infect Dis. 2007;20(3): Dermatol. 2000;114(3):560568.
272277. 40. Colman PM. Influenza virus neuraminidase: structure, antibodies, and
9. Domingues RB, Lakeman FD, Mayo MS, et al. Application of competi- inhibitors. Protein Sci. 1994;3(10):16871696.
tive PCR to cerebrospinal fluid samples from patients with herpes simplex 41. Colman PM, Varghese JN, Laver WG. Structure of the catalytic and anti-
encephalitis. J Clin Microbiol. 1998;36(8):22292234. genic sites in influenza virus neuraminidase. Nature. 1983;303(5912):4144.
10. Glaser CA, Gilliam S, Schnurr D, et al. In search of encephalitis etiologies: 42. Sharpe AH, Fields BN. Pathogenesis of viral infections. Basic concepts
diagnostic challenges in the California Encephalitis Project, 19982000. derived from the reovirus model. N Engl J Med. 1985;312(8):486497.
Clin Infect Dis. 2003;36(6):731742. 43. Spindler KR, Hsu TH. Viral disruption of the blood-brain barrier. Trends
11. Antona D, Leveque N, Chomel JJ, et al. Surveillance of enteroviruses in Microbiol. 2012;20(6):282290.
France, 20002004. Eur J Clin Microbiol Infect Dis. 2007;26(6):403412. 44. Lancaster KZ, Pfeiffer JK. Limited trafficking of a neurotropic virus
12. Hammer SM, Connolly KJ. Viral aseptic meningitis in the United States: through inefficient retrograde axonal transport and the type I interferon
clinical features, viral etiologies, and differential diagnosis. Curr Clin Top response. PLoS Pathog. 2010;6(3):e1000791.
Infect Dis. 1992;12:125. 45. Mrak RE, Young L. Rabies encephalitis in humans: pathology, pathogen-
13. Sferra TJ, Pacini DL. Simultaneous recovery of bacterial and viral patho- esis and pathophysiology. J Neuropathol Exp Neurol. 1994;53(1):110.
gens from cerebrospinal fluid. Pediatr Infect Dis J. 1988;7(8):552556. 46. Chou J, Kern ER, Whitley RJ, et al. Mapping of herpes simplex virus-1
14. Basmaci R, Mariani P, Delacroix G, et al. Enteroviral meningitis does not neurovirulence to gamma 134.5, a gene nonessential for growth in culture.
exclude concurrent bacterial meningitis. J Clin Microbiol. 2011;49(9): Science. 1990;250(4985):12621266.
34423443. 47. Bolovan CA, Sawtell NM, Thompson RL. ICP34.5 mutants of herpes
15. Kharfan-Dabaja MA, Ayala E, Greene J, et al. Two cases of progres- simplex virus type 1 strain 17syn are attenuated for neurovirulence in
sive multifocal leukoencephalopathy after allogeneic hematopoietic cell mice and for replication in confluent primary mouse embryo cell cultures.
transplantation and a review of the literature. Bone Marrow Transplant. J Virol. 1994;68(1):4855.
2007;39(2):101107. 48. Shah AC, Parker JN, Shimamura M, et al. Spontaneous and engineered
16. Leveque N, Galambrun C, Najioullah F, et al. Two cases of varicella zoster compensatory HSV mutants that counteract the host antiviral PKR
virus meningitis found in pediatric patients after bone marrow transplanta- response. Viruses. 2009;1(3):510522.
tion despite valaciclovir prophylaxis and without skin lesions. J Med Virol. 49. Jouanguy E, Zhang SY, Chapgier A, et al. Human primary immunodefi-
2006;78(4):514516. ciencies of type I interferons. Biochimie. 2007;89(67):878883.
17. Sejvar JJ. The evolving epidemiology of viral encephalitis. Curr Opin 50. Kunze U, Asokliene L, Bektimirov T, et al. Tick-borne encephalitis in child-
Neurol. 2006;19(4):350357. hoodconsensus 2004. Wien Med Wochenschr. 2004;154(910):242245.
18. Centers for Disease Control and Prevention. West Nile virus update 51. Kaushik DK, Gupta M, Basu A. Microglial response to viral challenges:
United States, January 1August 14, 2007. MMWR Morb Mortal Wkly every silver lining comes with a cloud. Front Biosci. 2011;16:21872205.
Rep. 2007;56(32):821822. 52. Shrestha B, Diamond MS. Role of CD8 T cells in control of West Nile
19. LaBeaud AD, Kile JR, Kippes C, et al. Exposure to West Nile virus during virus infection. J Virol. 2004;78(15):83128321.
the 2002 epidemic in Cuyahoga County, Ohio: a comparison of pediatric 53. Abzug MJ, Levin MJ, Rotbart HA. Profile of enterovirus disease in the first
and adult behaviors. Public Health Rep. 2007;122(3):356361. two weeks of life. Pediatr Infect Dis J. 1993;12(10):820824.
20. Rios J, Hacker CS, Hailey CA, et al. Demographic and spatial analysis of 54. Wyatt HV. Poliomyelitis and infantile paralysis: changes in host and virus.
West Nile virus and St. Louis encephalitis in Houston, Texas. J Am Mosq His Philos Life Sci. 1993;15(3):357396.
Control Assoc. 2006;22(2):254263. 55. Abbott NJ. Inflammatory mediators and modulation of blood-brain barrier
21. Whitley RJ, Kimberlin DW. Herpes simplex encephalitis: children and ado- permeability. Cell Mol Neurobiol. 2000;20(2):131147.
lescents. Semin Pediatr Infect Dis. 2005;16(1):1723. 56. Williams DW, Eugenin EA, Calderon TM, et al. Monocyte maturation,
22. Wonham MJ, Lewis MA, Renclawowicz J, et al. Transmission assumptions HIV susceptibility, and transmigration across the bloodbrain barrier are
generate conflicting predictions in host-vector disease models: a case study critical in HIV neuropathogenesis. J Leukoc Biol. 2012;91(3):401415.
in West Nile virus. Ecol Lett. 2006;9(6):706725. 57. Irani DN, Prow NA. Neuroprotective interventions targeting detrimental
23. Solomon T, Winter PM. Neurovirulence and host factors in flavivirus host immune responses protect mice from fatal alphavirus encephalitis.
encephalitisevidence from clinical epidemiology. Arch Virol Suppl. J Neuropathol Exp Neurol. 2007;66(6):533544.
2004;(18):161170. 58. Daley JK, Gechman LA, Skipworth J, et al. Poliovirus replication and
24. Liu W, Clemens JD, Yang JY, et al. Immunization against Japanese enceph- spread in primary neuron cultures. Virology. 2005;340(1):1020.
alitis in China: a policy analysis. Vaccine. 2006;24(24):51785182. 59. Tucker PC, Lee SH, Bui N, et al. Amino acid changes in the Sindbis virus
25. Solomon T. Control of Japanese encephalitiswithin our grasp? N Engl J E2 glycoprotein that increase neurovirulence improve entry into neuroblas-
Med. 2006;355(9):869871. toma cells. J Virol. 1997;71(8):61066112.
26. Kabilan L, Rajendran R, Arunachalam N, et al. Japanese encephalitis in 60. Soulie C, Calvez V, Marcelin AG. Coreceptor usage in different reservoirs.
India: an overview. Indian J Pediatr. 2004;71(7):609615. Curr Opin HIV AIDS. 2012;7(5):450455.
27. Vaughn DW, Hoke CH Jr. The epidemiology of Japanese encephalitis: pros- 61. Chang A, Dutch RE. Paramyxovirus fusion and entry: multiple paths to a
pects for prevention. Epidemiol Rev. 1992;14:197221. common end. Viruses. 2012;4(4):613636.
28. Roehr B. Texas records worst outbreak of West Nile virus on record. BMJ. 62. Campadelli-Fiume G, Menotti L, Avitabile E, et al. Viral and cellular con-
2012;345:e6019. tributions to herpes simplex virus entry into the cell. Curr Opin Virol.
29. Manning SE, Rupprecht CE, Fishbein D, et al. Human rabies prevention 2012;2(1):2836.
United States, 2008: recommendations of the Advisory Committee on 63. Fazakerley JK. Semliki Forest virus infection of laboratory mice: a
Immunization Practices. MMWR Recomm Rep. 2008;57(RR3):128. model to study the pathogenesis of viral encephalitis. Arch Virol Suppl.
30. Pape WJ, Fitzsimmons TD, Hoffman RE. Risk for rabies transmission 2004;(18):179190.
from encounters with bats, Colorado, 19771996. Emerg Infect Dis. 64. Noyce RS, Bondre DG, Ha MN, et al. Tumor cell marker PVRL4 (nectin 4)
1999;5(3):433437. is an epithelial cell receptor for measles virus. PLoS Pathog. 2011;7(8):
31. Centers for Disease Control and Prevention. Human rabiesCalifornia, e1002240.
Georgia, Minnesota, New York, and Wisconsin, 2000. MMWR Morb 65. Navaratnarajah CK, Miest TS, Carfi A, et al. Targeted entry of enveloped vi-
Mortal Wkly Rep. 2000;49(49):11111115. ruses: measles and herpes simplex virus I. Curr Opin Virol. 2012;2(1):4349.
32. Parrish JB, Yeh EA. Acuted disseminated encephalomyelitis. Adv Exp Med 66. Gromeier M, Bossert B, Arita M, et al. Dual stem loops within the
Biol. 2012;724:114. poliovirus internal ribosomal entry site control neurovirulence. J Virol.
33. McNabb SJ, Jajosky RA, Hall-Baker PA, et al. Summary of notifiable diseases 1999;73(2):958964.
United States, 2005. MMWR Morb Mortal Wkly Rep. 2007;54(53):192. 67. Kauder S, Kan S, Racaniello VR. Age-dependent poliovirus replication in
34. Dalmau J, Gleichman AJ, Hughes EG, et al. Anti-NMDA-receptor enceph- the mouse central nervous system is determined by internal ribosome entry
alitis: case series and analysis of the effects of antibodies. Lancet Neurol. site-mediated translation. J Virol. 2006;80(6):25892595.
2008;7(12):10911098. 68. Marsh M, Helenius A. Virus entry: open sesame. Cell. 2006;124(4):729740.
35. Gleichman AJ, Spruce LA, Dalmau J, et al. Anti-NMDA receptor encephalitis 69. Pilipenko EV, Pestova TV, Kolupaeva VG, et al. A cell cycle-dependent pro-
antibody binding is dependent on amino acid identity of a small region within tein serves as a template-specific translation initiation factor. Genes Dev.
the GluN1 amino terminal domain. J Neurosci. 2012;32(32):11082-11094. 2000;14(16):20282045.
36. Kimberlin DW. Herpes simplex virus infections of the newborn. Semin 70. Sandri-Goldin RM. The many roles of the regulatory protein ICP27 during
Perinatol. 2007;31(1):1925. herpes simplex virus infection. Front Biosci. 2008;13:52415256.
37. Abzug MJ, Keyserling HL, Lee ML, et al. Neonatal enterovirus infection: 71. Dauber B, Pelletier J, Smiley JR. The herpes simplex virus 1 vhs protein
virology, serology, and effects of intravenous immune globulin. Clin Infect enhances translation of viral true late mRNAs and virus production in a
Dis. 1995;20(5):12011206. cell type-dependent manner. J Virol. 2011;85(11):53635373.

Scheld_Ch04.indd 63 2/21/14 5:30 PM


64 Part II: Viral Infections and Related Disorders

72. Hunt CL, Lennemann NJ, Maury W. Filovirus entry: a novelty in the viral 88. Bennetto L, Scolding N. Inflammatory/post-infectious encephalomyelitis.
fusion world. Viruses. 2012;4(2):258275. J Neurol Neurosurg Psychiatry. 2004;75(suppl 1):i22i28.
73. Mehta H, Muller J, Markovitz NS. Ultrastructural analysis of ICP34.5- 89. Stroup JS, Stephens JR, Bury J, et al. Cytomegalovirus encephalitis in an
herpes simplex virus 1 replication in mouse brain cells in vivo. J Virol. HIV-seropositive person. AIDS Read. 2007;17(3):133134, 136.
2010;84(21):1098210990. 90. Khetsuriani N, Holman RC, Lamonte-Fowlkes AC, et al. Trends in
74. Penfold ME, Armati PJ, Mikloska Z, et al. The interaction of human encephalitis-associated deaths in the United States. Epidemiol Infect.
fetal neurons and epidermal cells in vitro. In Vitro Cell Dev Biol Anim. 2007;135(4):583591.
1996;32(7):420426. 91. Wiestler OD, Leib SL, Brustle O, et al. Neuropathology and pathogenesis
75. Reuter JD, Gomez DL, Wilson JH, et al. Systemic immune deficiency nec- of HIV encephalopathies. Acta Histochem Suppl. 1992;42:107114.
essary for cytomegalovirus invasion of the mature brain. J Virol. 2004; 92. Aguzzi A, Heikenwalder M, Polymenidou M. Insights into prion strains
78(3):14731487. and neurotoxicity. Nat Rev Mol Cell Biol. 2007;8(7):552561.
76. Klein RS, Lin E, Zhang B, et al. Neuronal CXCL10 directs CD8 93. Sejvar JJ, Marfin AA. Manifestations of West Nile neuroinvasive disease.
T-cell recruitment and control of West Nile virus encephalitis. J Virol. Rev Med Virol. 2006;16(4):209224.
2005;79(17):1145714566. 94. Laothamatas J, Sungkarat W, Hemachudha T. Neuroimaging in rabies.
77. Johnson KE, Knipe DM. Herpes simplex virus-1 infection causes the secre- Adv Virus Res. 2011;79:309327.
tion of a type I interferon-antagonizing protein and inhibits signaling at or 95. Jereb M, Lainscak M, Marin J, et al. Herpes simplex virus infection lim-
before Jak-1 activation. Virology. 2010;396(1):2129. ited to the brainstem. Wien Klin Wochenschr. 2005;117(1314):495499.
78. Leib DA, Harrison TE, Laslo KM, et al. Interferons regulate the pheno- 96. Gelpi E, Preusser M, Garzuly F, et al. Visualization of Central European
type of wild-type and mutant herpes simplex viruses in vivo. J Exp Med. tick-borne encephalitis infection in fatal human cases. J Neuropathol Exp
1999;189(4):663672. Neurol. 2005;64(6):506512.
79. Cassady KA. Human cytomegalovirus TRS1 and IRS1 gene products 97. Ayukawa R, Fujimoto H, Ayabe M, et al. An unexpected outbreak of
block the double-stranded-RNA-activated host protein shutoff response Japanese encephalitis in the Chugoku district of Japan, 2002. Jpn J Infect
induced by herpes simplex virus type 1 infection. J Virol. 2005;79(14): Dis. 2004;57(2):6366.
87078715. 98. Murray K, Baraniuk S, Resnick M, et al. Risk factors for encephalitis
80. Okabayashi T, Kojima T, Masaki T, et al. Type-III interferon, not type-I, is and death from West Nile virus infection. Epidemiol Infect. 2006;134(6):
the predominant interferon induced by respiratory viruses in nasal epithe- 13251332.
lial cells. Virus Res. 2011;160(12):360366. 99. LaBeaud AD, Lisgaris MV, King CH, et al. Pediatric West Nile virus
81. Pott J, Mahlakoiv T, Mordstein M, et al. IFN-lambda determines the infection: neurologic disease presentations during the 2002 epidemic in
intestinal epithelial antiviral host defense. Proc Natl Acad Sci U S A. Cuyahoga County, Ohio. Pediatr Infect Dis J. 2006;25(8):751753.
2011;108(19):79447949. 100. Padate BP, Keidan J. Enteroviral meningoencephalitis in a patient with
82. Bhowmick S, Duseja R, Das S, et al. Induction of IP-10 (CXCL10) in astro- non-Hodgkins lymphoma treated previously with rituximab. Clin Lab
cytes following Japanese encephalitis. Neurosci Lett. 2007;414(1):4550. Haematol. 2006;28(1):6971.
83. Wacher C, Muller M, Hofer MJ, et al. Coordinated regulation and wide- 101. Lee BE, Chawla R, Langley JM, et al. Paediatric Investigators Collaborative
spread cellular expression of interferon-stimulated genes (ISG) ISG-49, Network on Infections in Canada (PICNIC) study of aseptic meningitis.
ISG-54, and ISG-56 in the central nervous system after infection with dis- BMC Infect Dis. 2006;6:68.
tinct viruses. J Virol. 2007;81(2):860871. 102. Gorander S, Andersen O, Leiram B, et al. Multiphasic encephalomyelitis
84. Holub M, Beran O, Lacinova Z, et al. Interferon-gamma and cortisol levels in a patient with recurrent herpes simplex type 2 meningitis. Scand J Infect
in cerebrospinal fluid and its relationship to the etiology of aseptic menin- Dis. 2006;38(10):942925.
goencephalitis. Prague Med Rep. 2006;107(3):343353. 103. Kupila L, Vuorinen T, Vainionpaa R, et al. Etiology of aseptic meningitis
85. Christensen JE, de Lemos C, Moos T, et al. CXCL10 is the key ligand and encephalitis in an adult population. Neurology. 2006;66(1):7580.
for CXCR3 on CD8 effector T cells involved in immune surveillance of 104. Tyler KL. Herpes simplex virus infections of the central nervous system: en-
the lymphocytic choriomeningitis virus-infected central nervous system. cephalitis and meningitis, including Mollarets. Herpes. 2004;11(suppl 2):
J Immunol. 2006;176(7):42354243. 57A64A.
86. Niu X, Wang H, Fu ZF. Role of chemokines in rabies pathogenesis and 105. Kumar S, Kumar S, Kohlhoff SA. Recurrent HSV-2 meningitis in a 9-year-
protection. Adv Virus Res. 2011;79:7389. old girl. Scand J Infect Dis. 2006;38(67):570572.
87. Watson NF, Woo D, Doherty MJ, et al. Humoral immune responses after 106. Bergstrom T, Vahlne A, Alestig K, et al. Primary and recurrent herpes sim-
rabies infection. Arch Neurol. 2007;64(9):13551356. plex virus type 2-induced meningitis. J Infect Dis. 1990;162(2):322330.

Scheld_Ch04.indd 64 2/21/14 5:30 PM


CHAPTER 5 VIRAL MENINGITIS AND
ASEPTIC MENINGITIS SYNDROME
JOS R. ROMERO

Viral meningitis can be characterized as a central nervous sys- revealed a mononuclear pleocytosis and the absence of bacte-
tem (CNS) viral infection with signs of meningeal irritation ria on direct examination and by culture. In addition, no para-
(neck stiffness, Kernig and/or Brudzinski signs) and cerebro- meningeal process, acute/chronic systemic infectious disease,
spinal fluid (CSF) pleocytosis but without neurologic dysfunc- or community infectious disease could be identified that could
tion due to brain parenchymal involvement (1). It differs from produce the syndrome. With advances in diagnostic method-
viral encephalitis where evidence of brain parenchymal dys- ologies, it became evident that multiple infectious agents (e.g.,
function is manifested by an altered state of consciousness, Lyme disease), inflammatory conditions, drugs, environmental
change in personality, or other objective signs of neurologic agents, and so forth could cause the syndrome.
dysfunction (e.g., seizures, cranial nerve palsies, abnormal It is estimated that in the United States, the annual num-
reflexes, paralysis, etc.). Although it is common to discuss the ber of aseptic meningitis cases is at least 75,000. Viruses
two as wholly separate entities, it is important to note that account for the overwhelming majority of cases (Table 5.1).
overlap between them (i.e., meningoencephalitis) does occur Early reports indicated that mumps virus, lymphocytic cho-
following infection with many viral agents. riomeningitis virus (LCMV), and poliovirus (PV) were the
Almost 100 years ago, Wallgren (2) introduced the term major identifiable causes of aseptic meningitis (3). As diag-
acute aseptic meningitis to describe a short-lived, self-limited, nostic techniques improved as a result of the development of
benign CNS syndrome characterized by the acute onset of the cell culture, the enteroviruses were shown to have a major
signs of meningeal irritation in which examination of the CSF role in causation of syndrome (Table 5.2) (4,5). Nucleic acid

TA B L E 5 . 1
CAUSES OF THE ASEPTIC MENINGITIS SYNDROME AND CURRENT RELATIVE INCIDENCES

Common Rare
Viruses Viruses
Enteroviruses Herpes simplex virus type 1
Parechoviruses Varicella-zoster virus
Tick-borne encephalitis virusa Cytomegalovirus
Arbovirusesa Epstein-Barr virus
Herpes simplex virus type 2 Influenza A and B viruses
Bacteria Parainfluenza viruses
Borrelia burgdorferi (Lyme disease)a Human herpesvirus type 6
Partially treated bacterial meningitis (common pathogens) Measles virus
Parameningeal bacterial infection Rotavirus
Other Coronavirus
Kawasaki disease Encephalomyocarditis virus
Uncommon Parvovirus B19
Viruses Other
Mumps Brucella species
Lymphocytic choriomeningitis virus Mycoplasma hominis
Human immunodeficiency virus Mycoplasma pneumoniae
Bacteria Toxoplasma gondii
Mycobacterium tuberculosis Fungi (many)
Leptospira speciesa Autoimmune disorders
Other Behet syndrome
Fungi,a including Cryptococcus neoformans, Coccidioides Drugs (including immunomodulators, antibiotics)
immitis, Histoplasma capsulatum, Candida species, Malignancy
Blastomyces dermatitidis

a
Incidence varies greatly with geographic region.

65

Scheld_Ch05.indd 65 2/21/14 5:30 PM


66 Part II: Viral Infections and Related Disorders

TA B L E 5 . 2
VIRAL CAUSES OF ASEPTIC MENINGITIS IN SELECTED LARGE SERIES

Viruses Identified, %

Investigators No. Consistent


(Reference No.) Years Cases PV NPEV Arboviruses Mumps Herpes LCMV Other None Methodologya

Adair, Gauld, 194752 480 13.3 5.3 9.7 no 74.8 yes


and Smadel (3)
Meyer et al. (11) 195358 430 8.8 29.8 0.7 15.8 1.4 8.8 no 29.0 yes
Lennette, 1958 368 2.0 57.0 9.0 1.0 yesb 31.0 yes
Magoffin, and
Knouf (5)
Buescher, 195863 374 4.8 38.5 0.8 7.5 0.5 1.9 yesc 43.5 yes
Artenstein, and
Olson (12)
Berlin et al. (96) 198690 274 0.007 61.3 yesd 38.4 yes

LCMV, lymphocytic choriomeningitis viruses; NPEV, nonpolio enteroviruses; PV, polioviruses.


a
Virologic and/or serologic studies performed by a single laboratory with most or all specimens subjected to all tests.
b
1% adenovirus.
c
1% each measles, Epstein-Barr, influenza A.
d
1% adenovirus (1 case).

amplification tests (NAATs) have bolstered this finding and led viridae, viruses) family of viruses known to cause disease in
to the identification of novel causes (6). humans. The original classification of the EVs, based on evidence
The incidence of aseptic meningitis is influenced by many of human origin, pathology in animal models, patterns of replica-
factors, including effective vaccine programs, sanitation, pov- tion in cell culture, and physicochemical characteristics, identified
erty, and regional endemic viruses (710). Previous significant 72 serotypes (18). This schema, although initially useful, resulted
causes of viral meningitis such as PVs, mump virus, and LCMV to the misclassification of several EV and inclusion of several non-
are now rare or infrequent as a result of effective vaccines, san- EV into the genus (19). The development of experimental and
itation, or improved housing (3,5,8,1113). Although many of computational methodologies for the study of the molecular biol-
the infectious causes of aseptic meningitis are reportable (14), ogy and genomic analysis of the EV allowed for refinement in the
the true incidence of the syndrome is unknown due to incom- classification of and identification of the EVs.
plete reporting, failure to test for specific agents, and because Currently, identification and classification of the EVs
aseptic meningitis is not a reportable condition. In Finland, is based on the nucleotide sequence of VP1, the largest and
a 14-year birth cohort study found the annual incidence of most surface exposed of the viral capsid proteins containing
viral meningitis in children younger than 14 years of age to important neutralizing epitopes (2023). Using this approach,
be 27.8 per 100,000 (15). A more recent study from Greece the EVs have been speciated into four groups (enterovirus A
documented the annual incidence of aseptic meningitis to be to D) containing more than 100 serotypes (Table 5.3) (24).
17 per 100,000 in children younger than 14 years of age (16). In addition, this approach has revealed that several of the
Two studies from the United States give widely discrepant es- traditional EV serotypes are actually strains of the same se-
timates of the incidence of aseptic meningitis. A 32-year (1950 rotype or are not genetically related to the EVs (echoviruses 22
to 1981) study from Olmsted County, Minnesota found that and 23) (20,21,2527).
the adjusted incidence rate of aseptic meningitis was 10.9 per The EVs are nonenveloped viruses 30 nm in diameter with
100,000 person-years (range 7.9 to 17.8 per 100,000) (8,13). a buoyant density of 1.30 to 1.34 g/cm3 in caesium chloride
The Centers for Disease Control and Prevention reported that (CsCl) (19). The lack of an envelope confers to them relative
the national incidence for aseptic meningitis ranged from 1.5 environmental stability where they can survive for days at
to 4.0 per 100,000 for the period spanning 1971 to 1981 (17). room temperature. Infectivity can be preserved for weeks at
The lower incidence in the latter report is most likely the re- 20C or with little or no loss of infectivity for years when
sult of passive surveillance and, therefore, underreporting. The stored at 70C. Similarly, the lack of a lipid envelope renders
incidence of aseptic meningitis is greater in males and in chil- them insusceptible to ether, chloroform, and alcohol. The EVs
dren, particularly those younger than 1 year of age (8,15,16). are inactivated by heating to greater than or equal to 50C,
chlorine, and formaldehyde.
The capsid of all EVs is composed of 60 units each of four
structural or capsid proteins: VP1 to VP4, alternatively known
ENTEROVIRUSES as 1A to 1D, arranged so as to give the virion icosahedral sym-
metry (2833). Each of proteins VP1 to VP3 is wedge-shaped
Virology and Pathogenesis and composed of an eight-stranded antiparallel -barrel core.
Each of the stands is connected to the next by intervening loops
The enteroviruses (EVs) are one of six genera (Enterovirus, Car- that determine antigenicity, receptor specificity, and confer cap-
diovirus, Cosavirus, Hepatovirus, Parechovirus, and Kobuvirus) sid stability (34). The basic structural element of the viral capsid,
in the Picornaviridae (pico, small; rna, ribonucleic acid; the protomer, is initially composed of the proteins VP0, VP1,

Scheld_Ch05.indd 66 2/21/14 5:30 PM


Chapter 5: Viral Meningitis and Aseptic Meningitis Syndrome 67

TA B L E 5 . 3
HUMAN ENTEROVIRUS AND PARECHOVIRUS SEROTYPES

Genus: Enterovirus

Species Serotype

Enterovirus A CV-A2, CV-A3, CV-A4, CV-A5, CV-A6, CV-A7, CV-A8,


(18 serotypes) CV-A10, CV-A12, CV-A14, CV-A16,
EV-A71, EV-A76, EV-A89, EV-A90, EV-A91, EV-A92, EV-A114
Enterovirus B CV-A9,
(59 serotypes) CV-B1, CV-B2, CV-B3, CV-B4, CV-B5, CV-B6,
E-1, E-2, E-3, E-4, E-5, E-6, E-7, E-9, E-11, E-12, E-13, E-14, E-15, E-16, E-17, E-18, E-19, E-20,
E-21, E-24, E-25, E-26, E-27, E-29, E-30, E-31, E-32, E-33,
EV-B69, EV-B73, EV-B74, EV-B75, EV-B77, EV-B78, EV-B79, EV-B80, EV-B81, EV-B82, EV-B83,
EV-B84, EV-B85, EVB-86, EV-B87, EV-B88, EV-B93, EV-B97, EV-B98, EV-B100, EV-B101,
EV-B106, EV-B107, EV-B110
Enterovirus C PV-1, PV-2, PV-3,
(21 serotypes) CV-A1, CV-A11, CV-A13, CV-A17, CV-A19, CV-A20, CV-A21, CV-A22, CV-A24,
EV-C95, EV-C96, EV-C99, EV-C102, EV-C104, EV-C105,
EV-C109, EV-C113, EV-C116
Enterovirus D EV-D68, EV-D70, EV-D94, EV-D111
(4 serotypes)

Genus: Parechovirus

Human parechovirus HPeV- 1, HPeV-2, HPeV-3, HPeV-4, HPeV-5, HPeV-6, HPeV-7, HPeV-8, HPeV-9, HPeV-10, HPeV-11,
(16 serotypes) HPeV-12 HPeV-13, HPeV-14, HPeV-15, HPeV-16

CV, coxsackievirus; E, echovirus; EV, enterovirus; PV, poliovirus; HPeV, human parechovirus (24).

and VP3 (35). Five protomers self-assemble to form a pentamer. The 5 UTR contains multiple regions of predicted higher
Twelve pentamers, in turn, assemble around a single strand of order structure and highly conserved nucleotide identity
viral RNA to produce the immature virion. The cleavage of VP0 among the EV. This region of the genome contains elements
to yield VP2 and VP4 completes the formation of the mature vi- essential for viral RNA replication, translation of ORF, and, in
rion. VP1 to VP3, and in particular VP1, have surface-exposed the PVs, determinants of neurovirulence. Because of the highly
amino acids which determine the antigenic diversity and the re- conserved nucleotide sequences within the 5 UTR found
ceptor specificity of the EV (20,33,36,37). VP4 is not surface ex- among all the EV, it serves as the target for NAATs for the
posed but shares close association with the viral RNA and plays detection of the EV now in common use (42,43).
a vital role in release of the genome after viral attachment (38). Translation of the ORF by host cell ribosomes is accom-
The surface topographies of the various EVs share a num- plished in a nonconical, cap-independent manner giving rise to
ber of similarities. These include a plateau or mesa located at a single polyprotein that is posttranslationally cleaved by viral
the fivefold axis of symmetry formed by the union of five pro- and host proteinases to yield 11 viral proteins (four structural
tomers. Surrounding this plateau is a deep cleft or canyon into and seven nonstructural) as well as several functional protein
which a viral receptor inserts when the EV encounters a sus- intermediates. The ORF can be subdivided into three regions:
ceptible host cell (36). Additionally, the host immune response P1 to P3. The P1 region encodes for the four structural pro-
to EV infection generates serotype-specific antibodies directed teins (VP1 to VP4 or 1A to 1D) that comprise the viral capsid.
to antigenic sites around the fivefold axis and canyon walls, These are organized 5 to 3 as VP4 (1A), VP2 (1B), VP3 (1C),
thus blocking viral-host receptor interaction and infection. and VP4 (1D). The P2 and P3 regions code for seven non-
Lastly, beneath the canyon floor exists a hydrophobic pocket structural proteins (2A, 2B, 2C, 3A, 3B, 3C, and 3D) that are
containing a lipophilic factor. This pocket has been the target essential for the viral life cycle. The intermediate proteins play
for the development of anti-EV drugs that result in altered re- roles in viral replication.
ceptor binding and viral uncoating (39,40). Immediately downstream of the ORF is a short 3 UTR fol-
The EV genome consists of single-stranded, positive lowed by a terminal poly(A) tract. Similar to the 5 UTR, the
(messenger)-sense RNA of approximately 7,400 nucleotides 3 UTR is predicted to have higher order structures and play a
(nts) in length. The genome layout may be summarized as role in genome replication (44).
follows: VPg5UTR[1A-1B-1C-1D/2A-2B-2C/3A-3B-3C-3D] Following binding of the EV to a host cell receptor, confor-
3UTR-poly(A) (24,41). The 5 end of the genome is covalently mational changes in the virion result in release of the viral ge-
linked to a small protein, VPg, essential for viral RNA repli- nome into the cytoplasm of the host cell. The RNA genome is
cation. The genome is organized into a long 5 untranslated replicated through a double-stranded RNA intermediate that
region (5 UTR) of approximately 740 nts that immediately is formed by the EV RNA-dependent RNA polymerase (3D).
precedes a single open reading frame (ORF). The ORF mea- Despite more than 100 years of study of the pathogenesis of
sures approximately 6,630 nts and is followed by a short (ap- EV infections, much still needs to be learned. Most of what is
proximately 70 nts) 3 UTR and a terminal polyadenylated tail. known stems for the study of PV types 1 to 3 using information

Scheld_Ch05.indd 67 2/21/14 5:30 PM


68 Part II: Viral Infections and Related Disorders

derived from human disease and nonhuman primate, murine, the bloodbrain barrier (BBB) or via retrograde axonal trans-
and, most recently, transgenic (Tg) mouse models expressing port. For the development of paralytic disease in chimpanzees,
the PV receptor, PVR or CD155 (4550). It is likely that the viremia has been shown to be essential (76). This finding pro-
non-PV EVs share similar mechanisms of pathogenesis. vided initial support that the BBB may be a route to the CNS.
The majority of the EVs are transmitted via a fecal-oral Endothelial cells may express EV receptors that may influence
route. In addition to direct person-to-person or fecal-oral tissue susceptibility to the EV (77) and facilitate virus entry
transmission, experimental or clinical evidence exists for the into the CNS and other organs. Further buttressing of this hy-
transmission of the EV via houseflies, housefly-contaminated pothesis came from the finding that cultured human brain mi-
food, water, and sewage (5154). Bivalves have been found crovascular cells can support PV replication (78,79). However,
to accumulate EV (55,56). However, their role in transmis- in other studies, pharmacokinetic analysis of PV injected into
sion has not been established. Notable exceptions to fecal- Tg and non-Tg mice indicated that PV was delivered to the
oral transmission include coxsackievirus (CV)-A21, EV-D68, brain in significantly greater amounts than would be expected
and EV-D70, which may spread via contaminated fomites or from the vascular concentration (80). This suggests that PV
ocular and respiratory secretions. Evidence for transplacental may enter the CNS via the BBB but without need of PVR.
transmission exists, leading to congenital infection (5761). Evidence in humans supports EV access to the CNS via
Following ingestion of the EVs, infection of the cells of a neural route. Individuals inadvertently inoculated with an
the nasopharynx and, more significantly, the lower gastroin- incompletely inactivated PV vaccine developed initial paraly-
testinal (GI) tract occurs. The inherent acid-resistance of the sis in the limb receiving the vaccine (81). Trauma to a limb
EV favors the latter. Replication in the lymphatic tissues of preceding PV infection has been associated with the develop-
these sites (i.e., tonsils and Peyer patches) leads to shedding ment of paralysis of that limb. Provocation poliomyelitis
of the EV from the nasopharynx for 1 to 2 weeks and from following intramuscular injections into an extremity of a per-
the GI tract in feces for several weeks to months (7). Seeding son incubating wild type PV or those receiving live attenuated
of EV to the deep cervical and mesenteric lymph nodes ensues PV vaccines has been well documented (82,83).
and results in their spread to the systemic circulation via the Substantial evidence from nonhuman primate and murine
lymphatics. This primary or minor viremia leads to seeding models exists in support of EV access to the CNS via a neural
of various organs, including the CNS, liver, lungs, skin, and route. In monkeys, inoculation of the sciatic nerve with PV re-
heart. Further replication in the tissues of those organs results sults in viral spread along the inoculated nerve and the spinal
in a major (secondary) viremia. If the CNS was not seeded cord (84). The initial limb to develop paralysis following in-
during the initial viremic episode, spread there may occur with tramuscular inoculation of PV in monkeys and Tg mice is the
the major viremia. Viremia or presence of virus in the CNS one injected (8587). If the sciatic nerve of the intramuscularly
continues until the host develops type-specific neutralizing an- inoculated lower extremity is frozen or transected, paralysis of
tibodies directed to the capsid proteins, usually by day 7 to the limb is prevented (84,87). In Tg mice, it has been shown
10 postinfection. Immunoglobulin A (IgA) antibodies appear that in provocation PV, there appears to be induction of retro-
in the respiratory and GI tracts 2 to 4 weeks after infection. grade axonal transport (88). PV may gain access to neurons at
Unlike other viruses, which are largely contained by cellular the level of the neuromuscular junction.
immune mechanisms, the EVs are cleared from the host pri- The search for viral genomic determinants of neurotropism
marily by antibody-mediated mechanisms. and neurovirulence has focused on the PVs (89). After immu-
Great strides have been made in understanding the patho- nization with live, neuroattenuated vaccine (Sabin) PV strains,
genesis of EV infections at a molecular level. The presence of shedding of PV that has recovered the ability to cause pa-
an EV receptor is the primary, but not the sole, determinant ralysis (i.e., neurovirulent revertant strains) occurs routinely.
for cellular infection (62). As stated previously, PVR has been Comparison of wild type, vaccine, and revertant PV strains
shown to be the receptor for the PVs (62) and maps to chro- has identified a 10-nucleotide region within the 5 UTR (nts
mosome 19. PVR is a member of the immunoglobin superfam- 472 to 484 relative to PV type 3 Sabin strain) where neuroat-
ily and functions as an adhesion molecule. It helps to form tenuating mutations are found to cluster. Additional minor de-
adherens junctions and is a recognition molecule for natural terminants of virulence are localized to amino acids encoded
killer cells. In addition to PVR, at least 11 other cell proteins in the P1 and P3 regions. The search for determinants of neu-
have been identified as receptors or coreceptors for other rovirulence in other EV has been unsuccessful.
EV serotypes (Table 5.2) (6366).
The exact identity of the cells in the upper and lower in-
testinal tract infected by the PVs is unknown. However, they Histopathology
have been identified within the ileal wall and mesenteric nodes
in human infection (45,49,50). It is believed that the PV infect The benign nature of EV meningitis has made human patho-
either lower GI cells expressing the PVR or use transcytosis logic data for this disease sparse. A report of a child who died
through microfold (M) cells in the lower GI tract to gain access of CV-B5 myocarditis with concomitant meningitis describes
to lymphoid tissue. Supporting this is the finding of PVR on the inflammation of the choroid plexus of the lateral and fourth
surface of intestinal epithelium, M cells, and in the germinal ventricles, fibrosis of the vascular walls with focal destruction
centers of Peyer patches (67). Support for the latter comes from of the ependymal lining, and fibrotic basal leptomeninges (90).
the finding that M cells can bind and endocytose PVs (68,69). Parenchymal findings were limited to moderate symmetric dila-
Why the majority of EV infections do not result in clinically tion of the ventricles and an increase in the number and size of
apparent infection (7073) comes for a number of studies that subependymal astrocytes. The inflammatory reaction at the cho-
suggest that replication of PV in extraneural tissues is inhibited by roid plexus supports the concept of viremic spread to the CNS.
the host interferon (IFN) response. PVR Tg mice that are IFN / An adolescent presenting with a similar constellation of findings
receptor deficient are highly susceptible to PV infection, and PV died of systemic CV-B3 infection (91). The dura was grossly
replication in the small intestine is enhanced (74,75). Thus, IFN distended with swelling also of the pia, arachnoid, and brain
responses may be crucial in limiting the spread of EV infection. parenchyma. Microscopically, round cell infiltrates were noted
Lastly, the mechanism of EV entry into the CNS remains un- in the meninges overlying the cerebellum; the brain parenchyma
clear. Evidence for two pathways exist for PV: transit through was congested with increased numbers of oligodendrocytes.

Scheld_Ch05.indd 68 2/21/14 5:30 PM


Chapter 5: Viral Meningitis and Aseptic Meningitis Syndrome 69

Lymphocytic infiltration was most prominent around blood CV-B3, E-7, CV-B4, E-18, CV-B1, E-3, and -5 (93). Some se-
vessels in the cerebral white matter and in the basal ganglia, rotypes cycle with varying periodicity within a community
again suggesting viremic CNS access; focal areas of necrosis and (58,93,109,110), a reflection of the availability of new sus-
hemorrhage were also seen (91). ceptible host populations (especially children). Other sero-
types appear de novo as novel epidemic-associated viruses
(111). Around the world, the serotypes most commonly iso-
Epidemiology lated from the CSF and, therefore, from cases of meningitis
or meningoencephalitis, belong to the Enterovirus B species
The EVs have a ubiquitous worldwide distribution; humans (93,108,112,93,113,114). In the United States, the serotypes
are their only natural reservoir (58,92). It is estimated that most frequently isolated from CSF specimens over a 36-year
they result in more than 1 billion annual infections worldwide period, in descending order of frequency, are E-9, -11, -30,
(22). In the United States alone, the non-PV EVs are estimated CV-B5, E-6, CV-B2, CV-A9, E-4, CV-B4, E-7, -18, and -5 (93).
to cause 30 to 50 million infections each year. Due to under- Outbreaks of EV meningitis are common. Large nation-
reporting of EV infections (93), a well-grounded estimate of and community-wide outbreaks involving thousands of in-
the number of cases of EV meningitis that occurs each year is dividuals have been well documented (115,116). Outbreaks
not possible. However, conservative estimates place the annual involving more localized venues such as neonatal units, nurs-
number to be between 30,0000 and 75,000 cases (22,94). The eries, daycare centers, orphanages, schools, pools, camps, and
EVs are responsible for 80% to 90% of identifiable causes of sports teams occur (53,117122). Sequential episodes of EV
viral meningitis (9497). Wild type PV were eradicated for the meningitis involving different serotypes have been reported to
West Hemisphere in 1991 (98) and are currently endemic only occur within a month of each other (123125). Mixed infec-
in Afghanistan, Nigeria, and Pakistan (99). As such, they do tions involving EVs, other viruses, or bacteria have been well
not contribute to the burden to EV in most of the world. described (102130).
In regions with temperate climates, the non-PV EV exhibit Children represent the overwhelming majority of cases of
marked seasonality with the majority of infections occurring in EV meningitis. An incidence peak among young infants and
the summer and fall (Fig. 5.1) (92,100,101). This being said, EV school-aged children ages 5 to 10 years has been reported in
infections do occur during the winter, warranting their inclusion multiple studies (58,131133). Occasional outbreaks of EV
in the diagnostic evaluation of aseptic meningitis during that time CNS infections occur predominantly among adults (134136).
of year (102106). In tropical and subtropical areas, EVs occur A possible explanation for these findings may lie in the his-
year-round, but with higher incidence during the rainy season. tory of the particular serotypes in the geographic area studied.
Despite the existence of more than 100 serotypes of EV, Serotypes with endemic patterns, those occurring with sig-
only a limited number are responsible for the majority of dis- nificant incidence annually, are most likely to affect only the
ease observed annually in each geographic region (58,93,107 youngest children because of their absence of previous expo-
110). The rank of each serotype within the most frequently sure and immunity. Older children and adults are more likely
isolated EVs varies annually and geographically. In the United to predominate in an outbreak of a serotype that has not been
States, 15 serotypes accounted for approximately 80% of present in a community for several years, thereby creating a
all EVs reported from 1970 to 2005 (in descending order): reservoir of susceptible individuals among children born since
echoviruses (E)-9, -11, -30, CV-B5, E-6, CV-B2, CV-A9, E-4, the last appearance of that serotype.

1.2

1.1

1.0
Reported Cases per 100,000 Population

0.9

0.8

0.7

0.6

0.5

0.4
FIGURE 5.1 The seasonal occurrence
of aseptic meningitis in the United
0.3
States from 1986 to 1994, as reported
to the Centers for Disease Control and
0.2 Prevention. The striking predominance
of cases during the summer months
0.1 reflects the predominance of entero-
viruses as etiologic agents in aseptic
0.0 meningitis. (From Centers for Disease
1986 1987 1988 1989 1990 1991 1992 1993 1994 Control and Prevention. MMWR Morb
Mortal Wkly Rep. 1993;42:69, with
Year (Month) permission.)

Scheld_Ch05.indd 69 2/21/14 5:30 PM


70 Part II: Viral Infections and Related Disorders

Host factors that predispose to EV meningitis, other than and upper respiratory tract findings (60,139,140,142). On
young age and immunodeficiency, have been difficult to iden- physical examination, the fontanelle may be full or bulg-
tify. A slight male-to-female predominance in the incidence ing. Signs of meningeal irritation such as nuchal rigidity,
ratio for EV infections has been noted in large series. However, Brudzinski, and Kernig signs are generally absent. An exan-
in a recent report, male predominance was present only among them may be present. If encephalitis in addition to meningitis
persons younger than 20 years of age (male/female ratio (meningoencephalitis) is present, the neonate may present
1.4:0.9) (101). This most likely represents the larger number with profound lethargy, seizures, and focal neurologic abnor-
of females exposed to young children who are principal source malities that suggest herpes simplex virus (HSV) infection.
of household exposure. Infection rates are higher among per- In some newborns, encephalohepatomyocarditis syndrome
sons of lower socioeconomic status, in areas of crowding, and may develop in which signs and symptoms of severe hepa-
larger families (7). titis and myocarditis are superimposed on those of menin-
goencephalitis (143). Disseminated intravascular coagulation
and other findings of sepsis result in an illness that may
Clinical Manifestations be indistinguishable from that caused by overwhelming
bacterial infection.
The clinical manifestations of aseptic meningitis do not sig- In infants and children, following an incubation period
nificantly differ among the non-PV EVs causing the syndrome of 5 to 10 days, the onset of EV meningitis is usually abrupt
(9,137). However, clinical manifestations do vary with the age with fever (38 to 40C), the most common presenting sign
and immune status of the patient. Meningitis or meningoen- (137,143147). The natural history of EV meningitis is
cephalitis, singularly or in combination with other syndromes, depicted in Figure 5.2. The fever pattern may be biphasic, ini-
are common manifestations of symptomatic EV infection in tially appearing in association with nonspecific constitutional
neonates and young infants. Two large retrospective reviews signs and symptoms followed by resolution and subsequently
have documented that 62% of infants younger than 3 months reappearing with the onset of meningeal signs (137,148).
of age with group B coxsackievirus infections and 27% of neo- Headache is nearly always present in those individuals old
nates younger than 2 weeks of age with infections due to the enough to report it. Interestingly, it may be ameliorated fol-
echoviruses had associated meningitis or meningoencephalitis lowing the performance of a lumbar puncture, indicating
(60,138). In two prospective studies, clinical or laboratory that it may be the result of increased intracranial pressure
evidence of meningitis was found in 42% to 75% of neonates (149,150). Photophobia is commonly reported. Infants and
with EV infection (139,140). young children may be irritable or, less commonly, lethargic
Early presentation of EV infection following birth suggests (142). Nonspecific findings, singly or in combinations, include
transplacental, intrapartum, or immediate postpartum acqui- anorexia, exanthems, malaise, sore throat, abdominal pain,
sition of virus (60,138,139). Maternal illness (fever, symp- nausea, emesis, and myalgias (144,147). In infants, the fonta-
toms of upper respiratory tract infection, abdominal pain) nelle may be full or bulging. Less than 5% of infants younger
has been reported to occur in 14% to 68% of infected neo- than 3 months of age have signs of meningeal irritation (142).
nates (60,138141). In neonates, fever (38.0C) is almost However, these become more common in older patients
universal and accompanied by any or all of the following (144,147,151,152). Seizures are noted in less than 5% of
nonspecific signs: irritability, lethargy, poor feeding, emesis, children (142,146). Other uncommon complications include

DAYS 2 4 6 8 10 12 14 16 18 20 22 24 26 28
TEMPERATIURE

104

102

100

98

HEADACHE
NAUSEA & VOMITING
SYMPTOMS STIFF NECK
AND SIGNS (MYALGIA)
(RASH)
(MUSCLE WEAKNESS)

CSF
CELLS 50 1000
PROTEIN

VIRUS ISOLATION
BLOOD
THROAT FIGURE 5.2 The clinical course of
FECES enterovirus aseptic meningitis. CSF,
CSF cerebrospinal fluid. (From Horstmann
ANTIBODIES DM, Yamada N. Enterovirus infec-
NEUTRALIZING tions of the CNS. Res Public Assoc
Nerv Ment Dis. 1968;44:236253,
COMP. FIX.
with permission.)

Scheld_Ch05.indd 70 2/21/14 5:30 PM


Chapter 5: Viral Meningitis and Aseptic Meningitis Syndrome 71

coma, increased intracranial pressure, and inappropriate has resulted in stabilization of some of these patients; how-
secretion of antidiuretic hormone (142,153). The duration of ever, viral persistence has been documented during therapy
EV meningitis in infants and children is generally less than (159,162,163). With the availability of intravenous prepara-
1 week. tions of immune globulin and the early recognition of this
Fewer clinical reports exist documenting the presentation of illness, fewer patients appear to be progressing to the classic
EV meningitis in adolescents and adults (134,136,154157). description of this disease, and atypical neurologic presen-
Headache is the most frequently reported symptom and is tations have appeared. The mortality rate in patients with
nearly uniformly present. The severity of the headache may humoral immunodeficiencies may be as high as 50%.
be such as warrant the use of narcotic analgesics in order to The infected neonate is at greatest risk of severe morbidity
control the pain (157). Photophobia, fever, signs of meningeal and mortality when signs and symptoms develop in the first
irritation, nausea, emesis, and neck stiffness occur in more days of life (60,138,139,164). Neonates infected with CV-B4
than 60% of cases. Other less frequently encountered signs were found to be higher risk of death than those infected with
and symptoms include myalgia, exanthems, and abdominal other EV. The short-term prognosis of young children with EV
pain. Full recovery takes longer in adolescents and adults and meningitis early in life appears to be good; however, there has
may require up to 2.5 weeks (157). been some controversy over possible later sequelae. Neurologic,
In individuals with humoral immunodeficiencies (X-linked cognitive, developmental, and language abnormalities have
agammaglobulinemia, X-linked hyper IgM syndrome, com- been reported in controlled studies of long-term outcome in
mon variable immunodeficiency), EV infection may result in children with EV meningitis during infancy (165168). In the
chronic meningitis or meningoencephalitis that may last for largest and most meticulously controlled study, however, no
years and often have a fatal outcome (158161). In addition differences between patients and controls could be demon-
to the common signs and symptoms of EV meningitis men- strated in any of the neurodevelopmental parameters studied
tioned previously, neurologic manifestations include paraly- (142). Less well studied are the ultimate outcomes of aseptic
sis/paresis, seizures, cognitive impairment, developmental meningitis cases in older children and adolescents; preliminary
regression, sensorineural hearing loss, coma, dysarthria, data suggest possible school and learning difficulties, but con-
hydrocephalus, and aphasia. Extra CNS manifestations occur trol patients were not studied (169).
singly or in combination in a significant number of cases and
include dermatomyositis, chronic hepatitis, arthritis, myocar-
ditis, and subacute lumbosacral polyradiculopathy. In patients Laboratory Findings and Diagnosis
who have undergone repetitive, sequential lumbar punctures,
the cell culture detection of EV in CSF has been intermittent. Salient among the laboratory analyses performed for the evalu-
However, using NAATs, evidence of their persistence in CSF ation of EV meningitis is the evaluation of the CSF. CSF analysis
has been documented (162). Treatment with antibody prepa- can provide initial clues as to the etiology of the clinical syn-
rations intravenously and intrathecally or intraventricularly drome (Table 5.4). Cytochemical analysis of the CSF typically

TA B L E 5 . 4
TYPICAL CEREBROSPINAL FLUID PATTERNS DURING MENINGITIS

Pattern

Pathogen Number of WBCs Predominant Cell Glucose Protein

Bacteria (common) 100s1,000s Neutrophils


Viruses
Leptospira
Lyme disease
Mycoplasma
Mollarets 10s100s Mononuclears nl/sl nl/sl
Kawasaki disease
Parameningeal focusa
Partially treated meningitisa
Mycobacteria
Brucella 10s100s Mononuclears
Fungal
Toxoplasmab
Connective tissue disease
Parameningeala 10s100s Neutrophils nl/sl nl/sl
Partially treateda

nl, normal; sl, slightly; WBCs, white blood cells; , decrease; , increase.
a
Either pattern may be seen.
b
Glucose usually normal.

Scheld_Ch05.indd 71 2/21/14 5:30 PM


72 Part II: Viral Infections and Related Disorders

shows a mild to moderate lymphocytic pleocytosis ranging etiology for aseptic meningitis syndrome should rely on detec-
from 10 to 1,000 cells/mm3 (136,139,141,142,145,149,160). tion of the virus from the CSF. As previously discussed, follow-
White blood cell (WBC) counts exceeding 1,000 cells/mm3 are ing EV infection viral shedding may occur from the throat and
seen occasionally (60,103,147,170). Although pleocytosis is GI tract for up to several weeks (7,92). Therefore, detection
almost always present, EVs have been isolated by cell culture of the EV from the stool or throat of an individual with asep-
or detected by NAAT from the CSF of patients with clinical tic meningitis may represent an infection that occurred weeks
evidence of meningitis without pleocytosis (171). This is par- previously and is unrelated to the present syndrome. Shedding
ticularly true in the young infant. If the CSF is examined early from a past infection cannot be ruled out unless the virus is
in the course of the illness, a predominantly polymorphonu- detected in nonpermissive sites (i.e., CSF, blood, tissue) (189).
clear pleocytosis may be observed (136,139, 142,146,149). Lastly, rare reports of co-infections of the CSF by bacteria
Reexamination of the CSF several hours later will document and EVs exist (102,126,129,130). In these patients, the clini-
a typical lymphocytic pleocytosis (172174). The progres- cal and laboratory picture of bacterial meningitis dominated
sion of an initially polymorphonuclear pleocytosis to one of a and the virus was isolated incidentally. The patients were sick
more viral meningitis mononuclear pleocytosis has also been enough that identification of a virus before identification of
observed with St. Louis encephalitis virus (175). The CSF pro- the bacterium would have been unlikely to dissuade the cli-
tein concentration is mildly to moderately increased, whereas nician from continued use of antibiotics. Thus, the detection
the glucose concentration is generally normal. However, hypo- of an EV either by culture or NAAT must always be placed
glycorrhachia may occur, serving to confound the assessment in the context of the patients clinical picture and laboratory
by suggesting a bacterial etiology (146,147,170). findings. In the clinical presentation typical of viral meningitis,
Traditionally, the diagnosis of EV meningitis has relied on co-infection with a clinically silent bacterium would be ex-
isolation of the virus from CSF using cell culture or inocula- traordinarily unlikely.
tion of suckling mice (176). Although initially very useful, these
techniques have significant limitations. The sensitivity of tissue
culture for EVs is only 65% to 75% (177). The titer of EVs in Treatment and Prevention
the CSF of patients with aseptic meningitis may be as low as
101 to 103 TCID50 (median tissue culture infectious dose) per No specific treatment exists for EV meningitis. Supportive
milliliter of CSF. This results in slower growth than is observed measures include bed rest, antipyretics, and analgesics, as
with specimens of throat or rectal origin. The time to isolation indicated. Administration of parenteral fluids for individuals
of EV from CSF ranges from 4 to 8 days (178) using traditional unable to take adequate fluids orally, especially infants, is
cell culturetoo long to be clinically useful in patient man- indicated. Seizures should be controlled with appropriate
agement. Using shell vial culture, the time can be shortened anticonvulsant drugs.
to 2 to 3 days, but sensitivity may be lost (179,180). Lastly, Immune globulin preparations have been used for the treat-
optimum recovery of EVs from clinical specimens requires the ment of newborn infants with severe disease and immuno-
use of multiple cell lines, either individually or as mixtures, in compromised individuals, but their efficacy is not established
order to increase culture yield (181). Even using multiple cell (61,159,190). Intravenous, as well as intrathecal, administra-
lines, some EVs, in particular the group A CVs and some of the tion may be necessary to ameliorate or prevent CNS infection
newer EVs, fail to grow in cell culture (182). The added cost in immunocompromised patients.
and technical expertise required for suckling mouse inoculation The promising results from clinical trials of pleconaril
makes it impractical for use in the modern diagnostic labora- (39), an antiviral that inhibits EV binding and viral uncoat-
tory. Serologic confirmation of EV infection is also generally ing, were overshadowed by its adverse effects (40). In clini-
impractical and not useful in acute management of the patient. cal trials, pleconaril was found to induce cytochrome P450
As mentioned previously, the 5 UTR contains regions of 3A, resulting in menstrual irregularities in women taking
conserved nucleotide identity among the EVs. These regions hormonal contraceptives. This finding raised concerns that
have been exploited for the creation of primers and probes it might increase the metabolism of some hormonal contra-
that can be used in NAATs capable of detecting all EVs (43). ceptives and anti-HIV drugs, thereby reducing their efficacy,
Compared to cell culture, NAAT detection of EVs in the prompting the U.S. Food and Drug Administration not to
CSF has been shown to exhibit sensitivities that range from grant a license for its use.
86% to 100% and specificities ranging from 92% to 100%. The EVs are spread primarily through a lack of good
Furthermore, NAATs are capable of detecting EV genome in hygiene. Hand washing prevents the spread of the EVs and
CSF samples from individuals with syndromes clinically com- should be encouraged in families and institutions (191).
patible with aseptic meningitis previously deemed negative by In patients hospitalized with EV, meningitis infection con-
cell culture or without pleocytosis (163,162,183). These assays trol measures using standard precautions are sufficient.
are also able to detect EV that cannot be grown in cell culture. Community measures for the prevention of EV infections rely
Lastly, NAAT can be performed rapidly, generally in a mat- on the development and maintenance of sewage and potable
ter of hours. The results can be made available with sufficient water systems. No vaccines exist for the non-PV EV. However,
speed as to have an impact on patient management, resulting recent early studies suggest that it may be possible to develop
in a reduction in hospital stay, antibiotic use, and ordering an inactivated EV-A71 vaccine that can induce neutralizing
of ancillary tests (184188). A confirmatory polymerase chain antibodies and is well tolerated in humans (192).
reaction (PCR) test result obtained on a patient with clinical
aseptic meningitis can reassure the clinician that no further
diagnostic investigation is required. For these reasons, NAAT PARECHOVIRUSES
detection has become the method of choice for the diagnosis
of CNS EV infection. Because of the lack of sensitivity of viral The first two members of the genus human parechoviruses
culture for detection of the EV in CSF, it should be reserved for (HPeVs) were found in 1956 (193). However, it was not until
instances when NAAT is not available. the turn of the century that they were accorded their own genus.
Two caveats should be borne in mind when establishing Originally classified as EVs and designated as echoviruses 22
the diagnosis of EV meningitis. Confirmation of EV as the and 23 (18) (currently designated HPeV 1 and 2, respectively),

Scheld_Ch05.indd 72 2/21/14 5:30 PM


Chapter 5: Viral Meningitis and Aseptic Meningitis Syndrome 73

it became evident early on that they exhibited characteristics


that differed from the other members of the genus (193,194). ARBOVIRUSES
The development of methodologies to probe the molecular
aspects of viral replication and viral genetics confirmed that The arboviruses are a group of more than 500 viruses from
they differed substantively from the EV (2527,195,196) and various viral families that are transmitted by the bite of an
led to their reclassification in a separate genus (24). insect or tick (i.e., an arthropod vector) (213); hence, the
Morphologically, HPeVs exhibit similar characteristics as derivation of their name ararthropod, boborne, viruses.
the EV: small size, lack of an envelope, a positive sense, and Transmission to humans (epizootic transmission) occurs by
single-stranded RNA genome of the length and organization chance and is secondary to the natural enzootic cycle involv-
consistent with that of the EVs (6). Important differences ing an arthropod vector and an avian or mammalian host.
are the lack of maturational cleavage of VP0 to yield capsid Mosquitoes serve as the vectors for the majority of clinically
proteins VP2 and VP4. As a result, the HPeV capsid is significant arbovirus endemic to North America. However,
composed of three, rather than four, proteins. Differences Colorado tick fever virus, Powassan (POW) virus in North
also exist in the function of two nonstructural proteins. America, and tick-borne encephalitis (TBE) virus in Eurasia
A detailed discussion of these and other differences is beyond are transmitted by ticks (214,215).
the scope of this chapter, and the reader is directed to a recent
review (6).
The genus is currently composed of 16 serotypes (Table 5.3) Flaviviridae
(24). HPeVs have been reported worldwide. The epidemiology
of the HPeV continues to evolve as new serotypes are identified
and detection is improved using NAATs. Current data indicates
West Nile Virus
that the HPeVs account for approximately 2% of the EV iso- West Nile virus (WNV) was first isolated in Uganda in 1937
lated using traditional cell culture in clinical laboratories (6). (216). Its incursion into the United States in 1999 (217) pre-
HPeV1 followed by HPeV3 are the types most frequently iso- ceded a rapid spread throughout the contiguous continental
lated. Infection with HPeV appears to occur early in life. In the United States as well as North and South America (218,219).
United States, 73% of HPeV1 and 67.6% of HPeV2 isolated In the United States, WNV has displaced all autochthonous
come from infants younger than 1 year of age. A longitudinal arboviruses as the single major cause of CNS disease (220).
study of Norwegian infants documented that the cumulative WNV is a member of the Flaviviridae family of RNA viruses,
incidence of HPeV infection by 24 and 36 months of age was within the genus Flavivirus (221). All members of the family
86% and 94%, respectively (197). possess a host cellderived lipid envelope that is modified by
HPeV infections exhibit a strong seasonal epidemiology. the insertion of viral proteins and a positive (message)-sense,
Worldwide, the peak incidence of infections occurs during the single-stranded RNA genome.
summer and fall months (197204). A unique biennial pat- WNV is maintained in an enzootic cycle that involves pri-
tern of circulation has been reported for HPeV3 (205208). marily avian hosts and mosquitoes (222). Culex mosquitoes are
As with the EVs, multiple HPeV serotypes circulate within a important vectors in the United States (223). Mosquitoes are im-
community at the same time. The majority of cases of HPeV portant in transgenerational and transseasonal maintenance of
meningitis occur in male infants younger than 3 months of the WNV enzootic cycle (224,225). WNV infections occur dur-
age. HPeV3 is the overwhelmingly dominant cause of HPeV ing the summer months, generally from July through October in
meningitis. Considerable variation in the annual prevalence of the United States, coinciding with periods of increased activity
HPeV meningitis is observed. of its vectors. However, as WNV has spread southward in the
Transmission of the HPeV occurs via the fecal-oral and United States, reported transmissions to humans have occurred
respiratory routes. They may be shed from these sites for as early as April and as late as December (223).
weeks to months (197,201,209). The finding of HPeV in the Transmission to humans (i.e., epizootic transmission) is
stool of healthy, asymptomatic infants indicates that many, if incidental and, with rare exception, results in a dead-end
not the majority, of infections are subclinical. infection without subsequent human transmission. The lat-
Irritability is present in nearly all cases of HPeV meningitis ter has occurred through transfusion of blood products and
(202,207,210). An exanthem is frequently present. Emesis, organ transplantation (226229). In addition, confirmed or
diarrhea, and distention are reported in approximately one- suspected maternalfetal and maternalinfant vertical trans-
quarter to half of cases. Rhinorrhea, cough, tachypnea, mission of WNV has been reported (230,231).
apnea, and wheezing may be present. Notably, findings of The majority of WNV infections in adults and children
increased intracranial pressure (bulging fontanelle) or men- result in subclinical disease. Infection of children results in
ingeal irritation (nuchal stiffness, Kernig or Brudzinski signs) asymptomatic infection or milder disease more frequently
are absent. The CSF cytochemical evaluation reveals no or than in adults (232236). During one outbreak, children
minimal abnormalities in WBC count or protein and glu- were 4.5 times more likely to become infected with WNV
cose concentrations in the overwhelming majority of patients but 110 times less likely to develop West Nile neuroinvasive
(202,207,210). disease (WNND) (237). In adults, age is the single most sig-
Currently, NAAT is the methodology of choice for HPeV nificant risk factor for development of WNND (235,236,238).
detection because of its sensitivity and ability to detect all The incidence of severe neurologic disease is 10 times higher
known HPeV types in a clinically meaningful time frame (6). in persons aged 50 to 59 years and 43 times higher in those
Optimum diagnostic assays target the HPeV 5 UTR and aged 80 years or older compared with individuals 20 years
are not type-specific. They are designed for increased sen- of age or younger. Other risk factors include hypertension,
sitivity and to broadly detect all HPeV types from clinical diabetes mellitus, cardiovascular disease, alcohol abuse, and
specimens. immunosuppression. Approximately 80% of those infected
The HPeV can produce cytopathic effect on appropriate with WNV remain asymptomatic. The majority of the remain-
cell lines (211). However, cell culture detection is limited by der develops an acute, self-limited febrile illness known as West
those factors discussed for the EV (see previous discussion) Nile fever. It is characterized by a sudden onset of fever (38 to
(211,212). 40C), accompanied by fatigue, malaise, anorexia, headache,

Scheld_Ch05.indd 73 2/21/14 5:30 PM


74 Part II: Viral Infections and Related Disorders

myalgias, and weakness. Ocular pain on eye movement has each with a specific geographic distribution, are the principal
been reported. A diffuse nonpruritic, macular, papular, macu- vectors (257). Younger patients tend to have milder forms of
lopapular, or morbilliform exanthem and diffuse lymphade- SLEV-associated CNS disease (1,258). Approximately 15% of
nopathy may be seen (222,235,239241). all symptomatic cases of SLEV infection present as meningitis.
Less than 1% (1:140 to 320) of adults develop WNND: asep- The frequency of SLEV meningitis in children is much higher:
tic meningitis (WNV meningitis), encephalitis, or poliomyelitis- approximately 40%. In contrast, patients older than 60 years
like syndrome (241245). WNND may be even rarer in of age rarely (5% or less) present with aseptic meningitis.
children: approximately 1:4,200 infected children according to No specific therapy is available.
one study (237,246). The median age of patients with WNV
meningitis is less than those with encephalitis (247). The per- Tick-Borne Encephalitis Virus
centage of individuals with WNV meningitis that comprise cases
of WNND varies by report. In an outbreak in Israel, 15.9% Tick-borne encephalitis virus (TBEV) is an important cause of
of hospitalized patients with WNND had meningitis (233). In meningitis in Europe. Cases in returning travelers from Europe
the United States, WNV surveillance data from 1999 to 2008 and China have been seen in the United States (259). The disease
found that 33% of WNND cases reported to the Centers for was first described in Austria in 1931 (260). TBEV is endemic
Disease Control and Prevention (CDC) were meningitis (247). from Europe through far-eastern Russia, northern China, and
Individuals 19 years of age or younger accounted for 8% of all Japan. Three subtypes of the virus exist: European, Siberian, and
cases of meningitis. However, when cases of WNND in children Far Eastern (10,215). It is maintained in enzootic cycles involving
were analyzed for nearly the same time frame, 47% of all pe- Ixodid ticks and wild rodents (215). Humans may be infected
diatric cases were of meningitis as compared to encephalitis or through the bite of an infected tick or, less commonly, the con-
meningoencephalitis, which accounted for 37% of cases (236). sumption of virus-infected milk. The majority of cases occur from
In adults and children, West Nile meningitis is clinically March to November (10). The annual incidence of disease var-
indistinguishable from other causes of viral meningitis. The ill- ies widely by country: Latvia, Estonia, Lithuania, and southern
ness begins abruptly with fever, headache, nuchal rigidity, and Germany30, 16.5, 11.2, and 2 cases per 100,000 inhabitants,
meningeal signs (222,235,244). Photo- and phonophobia may respectively (10). Aseptic meningitis is seen with infection due
be present. The headache may be so severe as to need the use to the European and Siberian viral subtypes. Meningitis is more
of narcotic analgesics to control. Weakness and dyskinesias in frequently seen in younger age-groups (10). Sixty-six percent of
the form of tremors, myoclonus, or parkinsonism may occur. cases in infants and children 15 years of age or younger are of
The outcome of West Nile meningitis is favorable, although it meningitis. This decreases progressively with advancing age such
may take 2 to 3 weeks to fully recover. that for individuals 60 years of age or older, 32% or less develop
Analysis of the CSF shows a lymphocytic pleocytosis of meningitis. Diagnosis of TBEV infection is established by the
generally less than 500 cells/mm3 (235). If the CSF is exam- detection of virus-specific IgM and IgG in serum. Vaccines for the
ined early in the course of the illness, a polymorphonuclear prevention of TBEV-related disease are available in Europe and
pleocytosis may be seen (248). The presence of plasma cells Canada and are recommended for certain regions of the world
may be indicative of WNV as the etiology (249). (10,215,261). No specific therapy is available.
The diagnosis of WNV infection relies on the detection Other flaviviruses such as POW virus (214,262) and
of WNV-specific antibodies in CSF or paired serum samples Japanese encephalitis virus (263,264) are far less commonly
(235,250). The most sensitive and commonly used diagnostic identified as causes of meningitis.
assay is the IgM antibody-capture enzyme-linked immunosor-
bent assay (MAC-ELISA). It is capable of detecting CSF IgM Colorado Tick Fever Virus
antibodies 3 to 5 days into the clinical illness and 3 or more
Colorado tick fever (CTF) virus is an Orbivirus, a member of
days earlier than detectable serum antibody (251). However,
the family Reoviridae. Orbiviruses are double-stranded, seg-
a positive test should always be interpreted in the context of
mented RNA viruses. The virion is nonenveloped, with an
clinical syndrome because the presence of WNV-specific IgM
outer diameter of 80 nm and an inner core of 50 nm diameter.
has been detected in the CSF for up to 199 days after onset
CTF virus is found mostly in western and mountainous regions
of illness (252). The finding of WNV-specific IgM in the CSF
of the United States. Its tick vector is Dermacentor andersoni,
of an individual with a clinically compatible CNS syndrome
also known as the Rocky Mountain wood tick (265). After a
generally is considered diagnostic of WNND. If only serum is
bite by an infected tick, a 3- to 6-day incubation period follows.
used for establishing a diagnosis, a second convalescent serum
Hematopoietic cells, principally erythrocytes, are the major tar-
sample, obtained 2 or more weeks later, should be collected to
gets, wherein viral replication and dissemination occurs (265).
document a fourfold increase in specific antibody titers using
A biphasic illness is characteristic but is actually observed in
a functional assay such as neutralization or hemagglutination
only half of patients (265). It consists of initial sudden onset of
inhibition (235). Because serologic cross reactions among
high fever and headache with flulike constitutional symptoms.
St. Louis encephalitis virus, WNV, and POW virus can occur,
Hepatosplenomegaly may occur, as well as GI symptoms. Stiff
serologic testing ideally should include a battery of region-
neck and other meningeal signs occur in as many as 18% of
specific arboviral antigens (253).
confirmed CTF cases (266). Meningoencephalitis and encepha-
litis can occur but less commonly than meningitis. The period
St. Louis Encephalitis Virus of illness is usually brief (2 to 3 days). A peripheral leukope-
St. Louis encephalitis virus (SLEV) was first identified in 1933 nia with relative lymphocytosis is common. A lymphocyte-
following an epidemic of encephalitis in St. Louis, Missouri predominant CSF pleocytosis and elevated protein level are
(254). Like others of the genus, it is an enveloped, positive typically found in patients with neurologic manifestations.
(message)-sense, single-stranded RNA virus (221). It is recog- Certain patients transiently improve (1 to 2 days), and then
nized as the cause of sporadic and epidemic encephalitis and a second phase of illness of equal or greater severity follows
meningitis throughout the Americas. In the United States, most (lasting an additional 2 to 3 days). Severe sequelae and death,
cases are seen from July through September (255). In Florida, though rare, have been reported. Typically, recovery is rapid
epidemics have continued into December (256). Birds are and complete within 2 weeks. Laboratory diagnosis can be
the reservoir, and four different species of Culex mosquitoes, made using PCR (within the first 5 days of illness) or IgM-based

Scheld_Ch05.indd 74 2/21/14 5:30 PM


Chapter 5: Viral Meningitis and Aseptic Meningitis Syndrome 75

serology (214). Virus may also be detected in peripheral blood mononuclear cells (277). Half of patients have 500 or fewer
smears by indirect immunofluorescence. Paired acute and con- cells/mm3, 75% of cases have 1,000 or fewer cells/mm3, and
valescent serology is useful for retrospective diagnosis. the remainder have fewer than 5,000 cells/mm3. Exceptional
cases with more than 5,000 cells/mm3 have been reported.
Pleocytosis may persist for weeks. CSF protein level has been
Bunyaviridae reported as normal in more than half of patients with mumps
meningitis (280,282).
The California encephalitis group of viruses include five viruses Several approaches exist for the diagnosis of mumps
in the family Bunyaviridae. ThreeLa Crosse, Jamestown virus meningitis: viral isolation, documentation of increased
Canyon, and snowshoe hareviruses have been associated antibody titers between serum acute and convalescent serum
with aseptic meningitis (128,267). Numerically, La Crosse is samples obtained 2 to 3 weeks apart, documentation of
the most clinically relevant. From 1999 to 2007, La Crosse the presence of mumps-specific IgM antibodies, or NAAT
virus was reported from 25 states. However, 87% of cases detection of mumps virus genome (283). Mumps virus can
came from 7 states: West Virginia, Ohio, North Carolina, be detected in saliva, blood, urine, and CSF. It is present in
Tennessee, Wisconsin, Minnesota, and Illinois (255). saliva 9 days prior to and 8 days after the onset of parotitis.
In urine, it is detectable for up to 2 weeks after the onset of
symptoms. Mumps-specific IgM is present in the blood within
PARAMYXOVIRUSES 3 to 4 days of the start of symptoms and may persist for up
to 3 months. A sole serum sample demonstrating the presence
of mumps-specific IgM obtained within 10 days of the onset
Mumps Virus of illness is sufficient to establish the diagnosis. IgG antibod-
ies are detectable 7 to 10 days after the start of symptoms
Mumps virus is a member of the genus Rubulavirus within the
and persist for life. Antibodies to other paramyxoviruses may
subfamily Paramyxoviridae (268). It is an enveloped, pleomor-
cross-react with in mumps virus in serologic assays, leading to
phic virus possessing a single-stranded, negative-sense RNA
false-positive results.
genome. Only a single serotype exists, although 13 genotypes
have been identified (269,270). In the United States, prior to
1967 and the introduction of an effective vaccine, mumps
infections are observed during the winter and spring, with epi- Other Paramyxoviruses
demics occurring approximately every 3 to 5 years (271,272).
It was responsible for 2.5% to 15% of all cases of aseptic Measles infection may be associated with pleocytosis in as
meningitis and between 17.5% and 22% of known causes of many as 30% of uncomplicated cases in normal patients,
meningitis (5,8,9,11). As a result of mumps vaccine and effec- usually without signs or symptoms of meningitis (284). The
tive vaccination programs, mumps cases have been reduced by parainfluenza viruses have been associated with CNS infection
99% (273). However, mumps outbreaks continue to occur in (285). The dominant serotype reported has been parainfluenza
the United States (274,275). virus type 3.
Mumps is transmitted via respiratory droplets. Once infec-
tion occurs, viremia is the likely means of spread to distant
target organs, including the CNS (276). Meningitis is the most
common neurologic manifestation of mumps infection (277).
ARENAVIRUSES
Mumps once was the leading identifiable cause of aseptic men-
ingitis. The widespread use of the attenuated live-virus vac- Lymphocytic Choriomeningitis Virus
cine in the United States has resulted in a dramatic drop in
incidence of mumps as well as its major role as a cause of LCMV, a member of the family Arenaviridae, is an enveloped
meningitis (278). Neurologic involvement is three times more virus with a genome consisting of two single-stranded, ambi-
common in males. More than 50% of patients with mumps sense RNA molecules (286). LCMV was one of the earliest
parotitis have CSF pleocytosis (277); however, most are not and significant viruses to be associated with aseptic meningitis
symptomatic of meningitis. in humans (3) (Table 5.2). It is now rarely identified as a cause
Clinically symptomatic meningitis occurs in up to 10% of CNS infection in humans. The virus is endemic in wild
of patients with parotitis (279). Symptoms of meningitis are mice, which serve as its reservoir (287). In the United States,
reported in cases of mumps parotitis by 4 to 10 days of illness the prevalence in wild mice is estimated to be 3.9% to 13.4%
but may precede parotitis by as much as 7 days; half or more (287). Seroprevalence studies in the United States suggest that
cases of mumps meningitis may not be associated with par- the 0.4% to 5% of patients sampled in three large cities had
otitis at all (280,281). The clinical manifestations of mumps evidence of LCMV infection (287289). The virus is trans-
meningitis are nonspecific and differ little from those of EV mitted by rodents (hamsters, guinea pigs, rats, mice) via their
cases. Fever is universal, usually lasting 3 days but occasion- saliva, urine, feces, and nasal secretions (290). Individuals who
ally persisting for a week (280). Bradycardia, drowsiness, leth- work with or own rodents as well as those living under impov-
argy, and anemia are all reported. More significant neurologic erished and nonhygienic circumstances have traditionally been
involvement can occur. Encephalitis is described concomi- at greatest risk (291294). Human-to-human transmission can
tantly with meningitis in as many as 35% of cases (280) or as occur through transplantation (295,296). Approximately one
few as 4% (166). third of LCMV infections are asymptomatic. Clinically mani-
Mumps virus meningitis and meningoencephalitis are usu- fest infections are usually mild and of brief duration. The infec-
ally benign and self-limited diseases (277,280). The prognosis tion frequently results in a biphasic illness characterized by an
for rapid and full recovery from mumps meningitis is excellent initial flulike illness with fever, headache, malaise, myalgia,
(277,280). The occasional fatalities demonstrate pathologic anorexia, nausea, and emesis. A temporary, brief period of
findings of demyelination near blood vessels (277). improvement follows and precedes the onset of symptoms of
Most but not all cases of symptomatic mumps menin- meningitis or encephalitis (291,292,297). Occasional severe
gitis have a primarily monocytic CSF pleocytosis, primarily neurologic disease (meningoencephalitis, encephalitis) has

Scheld_Ch05.indd 75 2/21/14 5:30 PM


76 Part II: Viral Infections and Related Disorders

been reported (298). The course of meningitis and recovery are lethargy (321). In that study, all cases of meningitis occurred in
often prolonged (299), but permanent neurologic impairment children. When meningitis complicates zoster, typical vesicu-
is rare. CSF findings are indistinguishable from those of other lar lesions restricted to dermatomal distribution are seen. The
viral causes of aseptic meningitis: lymphocytic pleocytosis (up use of NAAT has identified individuals with meningitis due to
to several thousand), mildly elevated protein, and usually nor- reactivation of VZV but without skin lesions (301,321). The
mal or low glucose level. Other abnormalities include leukope- benign course is typical of other aseptic meningitides.
nia, thrombocytopenia, elevations of transaminase levels, and Reports of meningitis with EBV and CMV infection are
pulmonary infiltrates (297). Cell culture of CSF usually detects far less common that those of the agents discussed previously.
the presence of LCMV. In severe disseminated infection, virus Although HHV-6 and HHV-7 have been reported as causes
may be found in the blood, urine, and nasopharyngeal secre- of CNS infection, including meningitis (305,307,308), these
tions. NAAT can be used to detect viral genome in CSF. Acute reports must be interpreted with caution. HHV-6 has been
and convalescent serum specimens can be tested for raising shown to establish persistence in the CNS and has been found
antibody titers by enzyme immunoassays. in the CSF of asymptomatic individuals (322).

HUMAN HERPESVIRUSES OTHER VIRAL PATHOGENS


The majority of the human herpesvirusesHSV types 1 and HIV
2, varicella-zoster virus (VZV) (300,301), Epstein-Barr virus
(EBV) (302,303), cytomegalovirus (CMV) (304), human her- Aseptic meningitis is known to occur as part of the clinical
pesvirus (HHV)-6 (305308), and HHV-7 (308)have been constellation of syndromes associated with primary HIV infec-
associated with reports of aseptic meningitis. Numerically, tion (323325). The symptoms and signs are typical of aseptic
HSV-1 and HSV-2, in particular the latter, are the major causes meningitis and resolve rapidly. If the CSF is examined, a lym-
of aseptic meningitis among this family for viruses. phocytic pleocytosis is present along with mildly elevated pro-
HSVs appear to account for approximately 1% to 3% tein and normal glucose concentrations. HIV can be detected
of all cases of aseptic meningitis (Table 5.2). HSV-2 and, in the CSF. Pleocytosis may also be seen in asymptomatic HIV-
much less commonly, HSV-1 have been associated with asep- infected individuals.
tic meningitis in patients with primary genital herpes infec- Occasional cases of aseptic meningitis or meningoencephali-
tion (309311). HSV-2 meningitis following primary genital tis have been associated with adenoviruses in normal and immu-
infection is more frequently seen in women. Genital lesions nocompromised patients (326330), influenza A and B viruses
may not be present at the time of symptoms of aseptic men- (331333), parvovirus (334), and rotavirus (335337).
ingitis (310). Examination of the CSF demonstrates a lym-
phocytic pleocytosis, elevated protein, and normal glucose
concentrations. NONVIRAL PATHOGENS
Recurrent benign lymphocytic meningitis (RBLM), also
referred to as Mollaret meningitis, has been shown to be Multiple pro- and eukaryotic pathogens (bacteria, spirochete,
associated primarily with HSV-2 and, much less so, HSV-1 mycobacteria, fungi) can present with the classic features of
(312315). The prevalence of RBLM is difficult to assess aseptic meningitis. The majority of these agents would not be
because of its intermittent presentation, and it is not a report- readily detectable by Gram stain of the CSF and may require
able condition. However, two reports place it between 1 and special culture techniques to identify. As originally noted by
2.2 cases per 100,000 population (316,317). The syndrome Wallgren (2), parameningeal foci (sinusitis, otitis, mastoiditis,
is more frequently seen in young women and consists of trauma) can present similarly to viral meningitis and need to
recurrent episodes of aseptic meningitis, lasting 2 to 5 days, be always considered in the differential diagnosis of aseptic
which resolve spontaneously and without sequelae (318). The meningitis.
clinical presentation of RBLM is typical of viral meningitis.
However, approximately 50% of patients have transient neu-
rologic manifestations (seizures, hallucinations, diplopia, cra- Spirochetes
nial nerve palsies, altered consciousness). Lesions of genital
herpes are absent. Cytochemical analysis of the CSF reveals
a lymphocytic pleocytosis, mildly elevated protein, and nor-
Lyme Disease
mal glucose concentrations (318). Detection of HSV genome In areas of the United States where Lyme disease is endemic
is made by NAAT. The role of antiviral therapy in the manage- (338), CNS infection due to Borrelia burgdorferi may be
ment of RBLM is debated. Administration of acyclovir has encountered in 10% to 15% of individuals infected by this
been reported to result in rapid resolution of symptoms, and spirochete (339,340). B. burgdorferi is transmitted by Ixodes
suppressive therapy using valacyclovir or famciclovir may pre- species (scapularisEastern United States, or pacificus
vent recurrences. In individuals with HSV-2 detected from the Western United States) of ticks. Because the vector is so dimin-
CSF, counseling regarding prevention of transmission of geni- utive, a significant number of infected individuals do not recall
tal HSV should be undertaken (318). a tick bite. The illness is commonly seen in the spring and
VZV-related meningitis is a recognized complication of summer months and overlaps with EV and arboviral causes
primary infection (chickenpox) and reactivation (zoster) of meningitis. Meningitis usually occurs in the early dissemi-
(300,301,319,320). Meningitis has been associated with nated stage of infection. The clinical manifestations are similar
infection due to multiple different genotypes of the wild type to viral meningitis and may occur in association with cranial
strain as well the vaccine strain (Oka strain) of VZV (321) neuritis and radiculoneuritis (339343). The majority of chil-
and may be seen in immunocompetent and immunocompro- dren have associated findings such as facial or sixth nerve
mised individuals. The clinical and CSF presentation is typical palsies, papilledema, and increased intracranial pressure of
of aseptic meningitis. However, in recent report, only half of erythema migrans. Analysis of the CSF reveals a lymphocytic/
patients had fever, and nearly 25% had altered mental status or monocytic pleocytosis (500 cells/mm3) and elevated protein

Scheld_Ch05.indd 76 2/21/14 5:30 PM


Chapter 5: Viral Meningitis and Aseptic Meningitis Syndrome 77

concentration. The CSF glucose concentration is usually nor- hypoglycorrhachia. In addition to fungal culture of the CSF,
mal but may be slightly decreased. evidence of infection should sought through detection of fungal
The diagnosis of Lyme meningitis requires documenting antigen, fungal wall constituents, or specific antibodies in CSF
B. burgdorferi infection using a two-step diagnostic approach and blood. When attempting to detect fungi through culture, it
consisting of an initial enzyme-linked immunosorbent assay is important to use 10 to 15 mL of CSF. Evidence of fungal infec-
(ELISA) followed by a specific Western blot assay. In addi- tion in sites outside of the CNS should also be sought.
tion, the CSF should be examined for the presence of intra- A variety of neurologic manifestations have been reported to
thecal production of antibody to B. burgdorferi (339341). be associated with Mycoplasma pneumoniae infections (356).
Treatment with ceftriaxone, cefotaxime, or penicillin G for Of these, aseptic meningitis and encephalitis are the most com-
2 to 4 weeks is recommended (340,342,343). mon. Clinically, mycoplasmal meningitis is impossible to dis-
tinguish from viral meningitis, and as in typical viral infection,
sequelae are not observed. Diagnosis is by serology or PCR
Other Spirochetes (356,357). Mycoplasma hominis has been associated with cases
of neonatal meningitis, usually in preterm infants (358).
Leptospirosis is an acute systemic vasculitic disease caused by
a number of spirochetes in the Leptospira genus. Acquisition
is via contact with infected animal body fluids. Although more
Systemic Diseases
typically noted in the anicteric variety, meningitis is com- Kawasaki disease (KD) is an acute, self-limited vasculitis of
mon in icteric leptospirosis (Weil disease) as well (344). The unknown etiology affecting children. In the United States, the
CSF profile is indistinguishable from that caused by common estimated overall annual incidence among children younger than
viruses, except that overall, more patients develop elevated 5 years of age is 20 cases per 100,000 population (359). The
CSF protein than patients with common viral meningitis. incidence is higher among Asians and Pacific Islanders. Several
Aseptic meningitis is a relatively uncommon manifestation of reports indicate that between 40% and 60% of children with
secondary and tertiary syphilis (345). Meningitis due to tick- KD have a mild (usually 100 cells/mm3) mononuclear pleocy-
borne relapsing fever may be seen as frequently as with Lyme tosis on CSF examination (360,361). The incidence appears to be
disease (346). highest in reports from Japan. The CSF protein and glucose con-
centration are normal in the overwhelming majority of children.
Many other systemic vasculitides (e.g., polyarteritis nodosa,
Bacterial and Fungal Causes temporal arteritis, Takayasu arteritis, Wegener granulomatosis)
are associated with CSF pleocytosis (362). Meningitis has been
The majority of tuberculous CNS infections are caused by described as the initial manifestation of systemic lupus erythe-
Mycobacterium tuberculosis. A small percentage is due to matosus in several patients (363). Two percent to 4% of patients
bovine tuberculosis, Mycobacterium bovis. Children are par- with lupus may develop aseptic meningitis during the course of
ticularly prone to develop tuberculous meningitis (347). The their disease (363).
presentation is often subacute, occurring over 1 to 3 weeks
(348). The clinical course consists of three stages. Personality
change, irritability, anorexia, listlessness, and occasional fever Medication-Induced Aseptic Meningitis
characterize the first stage. Second stage signs and symptoms
reflect increased intracranial pressure and cerebral ischemia: The clinical features of medication-induced aseptic meningitis
drowsiness, nuchal rigidity, cranial nerve palsies, anisocoria, are not such that they set it apart from that caused by infectious
emesis, and seizures. In older children, adolescents, and adults, etiologies nor do they permit differentiation of the multiple
headache and emesis may be the dominant features. The third medication causes of the syndrome. A wide variety of medica-
stage is characterized by coma, autonomic instability, fever, and tions, vaccines, or dyes, administered systemically or within the
progressive cerebral dysfunction. Chest radiograph; tuberculin CNS, have been associated with aseptic meningitis (364372).
skin testing; and, in older children, adolescents, and adults, The most common class of medications associated with asep-
IFN gamma release assays should be performed, seeking evi- tic meningitis is nonsteroidal antiinflammatory drugs (NSAIDs)
dence of tuberculosis. The lumbar puncture reveals elevated (364). Of the nonselective and selective inhibitors of cyclooxygen-
opening pressure. The CSF reveals a markedly elevated protein ase 1 and 2, ibuprofen is the most frequently associated NSAID
and decreased glucose concentrations. A lymphocytic pleocy- with aseptic meningitis. Ibuprofen-associated aseptic meningitis
tosis of usually less than 500 cells/mm3 is seen. is frequently seen in association with systemic lupus erythemato-
CNS infections with Brucella species occur in less than 5% sus. The majority of cases are reported in women. The associated
of systemic brucella infections (349). Meningoencephalitis is pleocytosis is frequently polymorphonuclear in nature.
the usual presentation. Both acute and chronic forms have Antimicrobial agents have also been shown to cause
been reported. Antibiotic therapy is usually curative; however, aseptic meningitis (364). Leading this group of medications
some residual neurologic defects are the rule. are trimethoprim and penicillin (penicillin, amoxicillin, or
Fungal meningitis is more commonly seen in immunocom- amoxicillin-clavulanic acid). Intravenous immunoglobulin
promised individuals; however, occasional cases have been is a well-recognized cause of aseptic meningitis (364). In
reported in immunologically normal individuals (350355). pediatric clinical trials using intravenous immunoglobulin,
Cryptococcus is the most commonly recognized cause of fungal the incidence of aseptic meningitis ranged from 6% to 32%.
meningitis (352). Other causes include Candida, Histoplasma, Immunomodulators such as monoclonal antibodies against
Coccidioides, Blastomyces, and Aspergillus (350,351,353355). CD3 or tumor necrosis factor- are important causes of
The clinical presentation is that of a subacute or chronic meningi- aseptic meningitis (364,368). Vaccine strains of mumps have
tis with fever, headache, and altered consciousness. Meningismus been the most frequently associated (364,373). Greater than
and focal neurologic findings are common findings. Evaluation 10 mumps vaccine strains are used around the world (373).
of the CSF reveals a lymphocytic pleocytosis in most cases Although the Jeryl Lynn and related vaccine strains rarely, if
(a polymorphonuclear pleocytosis may be seen in some cases) ever, cause aseptic meningitis, other vaccine strains have been
in association with an increased protein concentration and associated with varying incidences of this side effect (373).

Scheld_Ch05.indd 77 2/21/14 5:30 PM


78 Part II: Viral Infections and Related Disorders

References
1. Brinker KR, Monath TP. The acute disease. In: Monath TP, ed. St. Louis 29. Hogle JM, Chow M, Filman DJ. Three-dimensional structure of poliovirus
Encephalitis. Washington, DC: American Public Health Association; at 2.9 A resolution. Science. 1985;229:13581365.
1980:503584. 30. Muckelbauer JK, Kremer M, Minor I, et al. Structure determination of
2. Wallgren A. Une nouvelle maladie infectieuse du systeme nerveux central coxsackievirus B3 to 3.5 A resolution. Acta Crystallogr D Biol Crystallogr.
(Mningite aseptica acuta). Acta Paediatr. 1925;4:158182. 1995;51:871887.
3. Adair CV, Gauld RL, Smadel JE. Aseptic meningitis, a disease of diverse 31. Hendry E, Hatanaka H, Fry E, et al. The crystal structure of coxsacki-
etiology: clinical and etiologic studies on 854 cases. Ann Intern Med. evirus A9: new insights into the uncoating mechanisms of enteroviruses.
1953;39:675704. Structure. 1999;7:15271538.
4. Meyer HM Jr, Rogers NG, Miesse ML, et al. Aseptic meningitis caused by 32. Xiao C, Bator-Kelly CM, Rieder E, et al. The crystal structure of coxsackie-
orphan viruses and other agents. Ann N Y Acad Sci. 1957:19;67:332337. virus A21 and its interaction with ICAM-1. Structure. 2005;13:10191033.
5. Lennette EH, Magoffin RL, Knouf EG. Viral central nervous system dis- 33. Plevka P, Perera R, Cardosa J, et al. Crystal structure of human enterovirus
ease. An etiologic study conducted at the Los Angeles County General 71. Science. 2012;336:1274.
Hospital. JAMA. 1962;179:687695. 34. Fry EE, Stuart DI. Virion structure. In: Ehrenfeld E, Domingo E, Ross RR,
6. Romero JR, Selvarangan R. The human parechoviruses: an overview. Adv eds. The Picornaviruses. Washington, DC: ASM Press; 2010:5971.
Pediatr. 2011;58:6585. 35. Hellen CUT, Wimmer E. Enterovirus structure and assembly. In: Rotbart
7. Hall CE, Cooney MK, Fox JP. The Seattle virus watch program. I. Infection HA, ed. Human Enterovirus Infections. Washington, DC: ASM Press;
and illness experience of virus watch families during a communitywide epi- 1998:155170.
demic of echovirus type 30 aseptic meningitis. Am J Public Health Nations 36. Rossmann MG. The canyon hypothesis. Hiding the host cell receptor
Health. 1970;60:14561465. attachment site on a viral surface from immune surveillance. J Biol Chem.
8. Beghi E, Nicolosi A, Kurland LT, et al. Encephalitis and aseptic meningitis, 1989;264:1458714590.
Olmsted County, Minnesota, 19501981: I. Epidemiology. Ann Neurol. 37. Harber J, Bernhardt G, Lu HH, et al. Canyon rim residues, including
1984;16:283294. antigenic determinants, modulate serotype-specific binding of polioviruses
9. Lepow ML, Carver DH, Wright HT, et al. A clinical, epidemiologic to mutants of the poliovirus receptor. Virology. 1995;214:559570.
and laboratory investigation of meningitis during the four year period: 38. Tuthill TJ, Bubeck D, Rowlands DJ, et al. Characterization of early steps in
19551958. I. Observations concerning etiology and epidemiology. N Engl the poliovirus infection process: receptor-decorated liposomes induce con-
J Med. 1962;23:11811187. version of the virus to membrane-anchored entry-intermediate particles.
10. Kaiser R. Tick-borne encephalitis. Infect Dis Clin North Am. 2008;22: J Virol. 2006;80:172180.
561575. 39. Romero JR. Pleconaril: a novel antipicornaviral drug. Expert Opin Investig
11. Meyer HM Jr, Johnson RT, Crawford IP, et al. Central nervous system syn- Drugs. 2001;10:369379.
dromes of viral etiology. Am J Med. 1960;29:334347. 40. Webster AD. Pleconarilan advance in the treatment of enteroviral infec-
12. Buescher EL, Artenstein MS, Olson LC. Central nervous system infections tion in immune-compromised patients. J Clin Virol. 2005;32:16.
of viral etiology: the changing pattern. Res Public Assoc Nerv Ment Dis. 41. Palmenberg A, Neubauer D, Skern T. Genome organization and encoded
1968;44:147216. proteins. In: Ehrenfeld E, Domingo E, Ross RR, eds. The Picornaviruses.
13. Nicolosi A, Hauser WA, Beghi E, et al. Epidemiology of central nervous Washington, DC: ASM Press; 2010:371.
system infections in Olmsted County, Minnesota, 19501981. J Infect Dis. 42. Rotbart HA, Romero JR. Laboratory diagnosis of enterovirus infections.
1986;154:399408. In: Rotbart HA, ed. Human Enterovirus Infections. Washington, DC:
14. Centers for Disease Control and Prevention Office of Surveillance, ASM; 1995:401418.
Epidemiology, and Laboratory Services. 2012 nationally notifiable diseases 43. Romero JR. Reverse transcription-polymerase chain reaction detection of
and conditions and current case definitions. Centers for Disease Control the enteroviruses: overview and clinical utility in pediatric enteroviral infec-
and Prevention Web site. http://wwwn.cdc.gov/nndss/document/2012 tions. Arch Pathol Lab Med. 1999;123:11611169.
_Case%20Definitions.pdf. Accessed January 10, 2013. 44. Rozovics JM, Semler BL. Genome replication I: the players. In: Ehrenfeld
15. Rantakallio PM, Leskinen M, von Wendt L. Incidence and prognosis of E, Domingo E, Ross RR, eds. The Picornaviruses. Washington, DC: ASM
central nervous system infections in a birth cohort of 12,000 children. Press; 2010:107125.
Scand Infect Dis. 1986:18:287294. 45. Sabin AB, Ward R. The natural history of human poliomyelitis: I.
16. Michos AG, Syriopoulou VP, Hadjichristodoulou C, et al. Aseptic meningi- Distribution of virus in nervous and non-nervous tissues. J Exp Med.
tis in children: analysis of 506 cases. PLoS One. 2007;2:e674. 1941;73:771793.
17. Centers for Disease Control and Prevention. Annual summary 1982: 46. Bodian D. Histopathologic basis of clinical findings in poliomyelitis. Am J
reported morbidity and mortality in the United States. MMWR Morb Med. 1949;6:563578.
Mortal Wkly Rep. 1983;31:142146. 47. Bodian D. Emerging concept of poliomyelitis infection. Science. 1955;122:
18. Committee on Enteroviruses. Classification of human enteroviruses. 105108.
Virology. 1962;16:501504. 48. Sabin AB. Pathogenesis of poliomyelitis: reappraisal in the light of new
19. Gradien M, Forsgren M, Ehrnst A. Enteroviruses and reoviruses. In: data. Science. 1956;123:11511157.
Schmidt NJ, Emmons RW, eds. Diagnostic Procedures for Viral, Rickettsial, 49. Racaniello VR. One hundred years of poliovirus pathogenesis. Virology.
and Chlamydial Infections. 6th ed. Washington, DC: American Public 2006;344:916.
Health Association; 1989:513578. 50. Koike S, Nomoto A. Poliomyelitis. In: Ehrenfeld E, Domingo E, Ross RR,
20. Oberste MS, Maher K, Kilpatrick DR, et al. Molecular evolution of the eds. The Picornaviruses. Washington, DC: ASM Press; 2010:339351.
human enteroviruses: correlation of serotype with VP1 sequence and 51. Ward R, Melnick JL, Horstmann DM. Poliomyelitis virus in fly-contami-
application to picornavirus classification. J Virol. 1999;73:19411948. nated food collected at an epidemic. Science. 1945;101:491493.
21. Oberste MS, Maher K, Kilpatrick DR, et al. Typing of human enteroviruses 52. Kelly SW, Winkelstein W Jr, Winsser J. Poliomyelitis and other enteric
by partial sequencing of VP1. J Clin Microbiol. 1999;37:12881293. viruses in sewage. Am J Public Health Nations Health. 1957;47:7277.
22. Oberste MS, Maher K, Flemister MR, et al. Comparison of classic and 53. Centers for Disease Control and Prevention. Aseptic meningitis out-
molecular approaches for the identification of untypeable enteroviruses. break associated with echovirus 9 among recreational vehicle campers
J Clin Microbiol. 2000;38:11701174. Connecticut, 2003. MMWR Morb Mortal Wkly Rep. 2004;53:710713.
23. Oberste M, Schnurr D, Maher K, et al. Molecular identification of new 54. Begier EM, Oberste MS, Landry ML, et al. An outbreak of concurrent echo-
picornaviruses and characterization of a proposed enterovirus 73 serotype. virus 30 and coxsackievirus A1 infections associated with sea swimming
J Gen Virol. 2001;82:409416. among a group of travelers to Mexico. Clin Infect Dis. 2008;47:616623.
24. Knowles NJ, Hovi T, Hyypi T, et al. Picornaviridae. In: King AMQ, 55. Mitchell JR, Presnell WM, Akin EW, et al. Accumulation and elimination
Adams MJ, Carstens EB, et al, eds. Virus Taxonomy. Classification and of poliovirus by the eastern oyster. Am J Epidemiol. 1966;84:4050.
Nomenclature of Viruses: Ninth Report of the International Committee on 56. Bendinelli M, Ruschi A. Isolation of human enterovirus from mussels. Appl
the Taxonomy of Viruses. London: Elsevier Academic Press; 2012:855880. Microbiol. 1969;18:531532.
25. Hyypi T, Horsnell C, Maaronen M, et al. A distinct picornavirus group iden- 57. Bates HR Jr. Coxsackie virus B3 calcific pancarditis and hydrops fetalis.
tified by sequence analysis. Proc Natl Acad Sci U S A. 1992;89:88478851. Am J Obstet Gynecol. 1970;106:629630.
26. Oberste MS, Maher K, Pallansch MA. Complete sequence of echovirus 23 58. Grist NR, Bell EJ, Assaad F. Enteroviruses in human disease. Prog Med
and its relationship to echovirus 22 and other human enteroviruses. Virus Virol. 1978;24:114157.
Res. 1998;56:217223. 59. Bendig JW, Franklin OM, Hebden AK, et al. Coxsackievirus B3 sequences
27. Ghazi F, Hughes PJ, Hyypia T, et al. Molecular analysis of human parecho- in the blood of a neonate with congenital myocarditis, plus serological evi-
virus type 2 (formerly echovirus 23). J Gen Virol. 1998;79:26412650. dence of maternal infection. J Med Virol. 2003;70:606609.
28. Rossmann MG, Arnold E, Erickson JW, et al. Structure of a human com- 60. Kaplan MH, Klein SW, McPhee J, et al. Group B coxsackievirus infections
mon cold virus and functional relationship to other picornaviruses. Nature. in infants younger than three months of age: a serious childhood illness.
1985;317:145153. Rev Infect Dis. 1983;5:10191032.

Scheld_Ch05.indd 78 2/21/14 5:30 PM


Chapter 5: Viral Meningitis and Aseptic Meningitis Syndrome 79

61. Abzug MJ. Presentation, diagnosis, and management of enterovirus infec- 91. Sutinen S, Kalliomaki JL, Pohjonen R, et al. Fatal generalized coxsackie
tions in neonates. Paediatr Drugs. 2004;6:110. B3 virus infection in an adolescent with successful isolation of the virus
62. Mendelsohn CL, Wimmer E, Racaniello VR. Cellular receptor for poliovi- from pericardial fluid. Ann Clin Res. 1971;3:241246.
rus: molecular cloning, nucleotide sequence and expression of a new mem- 92. Melnick JL. Enteroviruses In: AS Evans, ed. Viral Infections of Humans,
ber of the immunoglobulin superfamily. Cell. 1989;56:855865. Epidemiology and Control. 3rd ed. New York: Plenum, 1989:191263.
63. Bergelson J. Receptors. In: Ehrenfeld E, Domingo E, Ross RR, eds. The 93. Centers for Disease Control and Prevention. Enterovirus surveillance
Picornaviruses. Washington, DC: ASM Press; 2010:7386. United States, 19971999. MMWR. 2000:13:913916.
64. Israelsson S, Gullberg M, Jonsson N, et al. Studies of Echovirus 5 interac- 94. Rotbart HA. Meningitis and encephalitis. In: Rotbart HA, ed. Human
tions with the cell surface: heparan sulfate mediates attachment to the host enterovirus infections. Washington, DC: American Society for
cell. Virus Res. 2010;151:170176. Microbiology Press; 1995:271289.
65. Mistry N, Inoue H, Jamshidi F, et al. Coxsackievirus A24 variant uses sialic 95. Lepow ML, Coyne N, Thompson LB, et al. A clinical, epidemiologic and
acid-containing O-linked glycoconjugates as cellular receptors on human laboratory investigation of aseptic meningitis during the four-year period,
ocular cells. J Virol. 2011;85:1128311290. 19551958. II. N Engl J Med. 1962;266:11881193.
66. Yamayoshi S, Iizuka S, Yamashita T, et al. Human SCARB2-dependent 96. Berlin LE, Rorabaugh ML, Heldrich F, et al. Aseptic meningitis in infants
infection by coxsackievirus A7, A14, and A16 and enterovirus 71. J Virol. 2 years of age: diagnosis and etiology. J Infect Dis. 1993;168:888892.
2012;86:56865696. 97. de Ory F, Avelln A, Echevarra JE, et al. Viral infections of the central ner-
67. Iwasaki A, Welker R, Mueller S, et al. Immunofluorescence analysis of vous system in Spain: a prospective study. J Med Virol. 2013;85:554562.
poliovirus receptor expression in Peyers patches of humans, primates, and 98. de Quadros CA, Andrus JK, Olive JM, et al. Polio eradication from the
CD155 transgenic mice: implications for poliovirus infection. J Infect Dis. Western Hemisphere. Annu Rev Public Health. 1992;13:239252.
2002;186:585592. 99. Centers for Disease Control and Prevention. Progress toward poliomyeli-
68. Sicinski P, Rowinski J, Warchok JB, et al. Poliovirus type 1 enters the tis eradicationNigeria, January 2011September 2012. MMWR Morb
human host through intestinal M cells. Gastroenterology. 1990;98:5658. Mortal Wkly Rep. 2012;61:899904.
69. Ouzilou L, Caliot E, Pelletier I, et al. Poliovirus transcytosis through M-like 100. Assaad F, Cockburn C. Four-year study of WHO virus reports on enterovi-
cells. J Gen Virol. 2002;83:21772182. ruses other than poliovirus. Bull World Health Organ. 1972:46;329336.
70. Gelfand HM, Holguin AH, Enterovirous infections in healthy children. 101. Centers for Disease Control and Prevention. Enterovirus surveillance
Study during 1960. Arch Environ Health. 1962;5:404411. United States, 19702005. Surveillance Summaries. MMWR Surveill
71. Kogon A, Spigland I, Frothingham TE, et al. The virus watch program: Summ. 2006;55(8):120.
a continuing surveillance of viral infections in metropolitan New York 102. Nishmi M, Morr J, Abrahamov A, et al. A winter outbreak of ECHO
families. VII. Observations on viral excretion, seroimmunity, intrafamilial virus type 9 meningitis in Jerusalem, with cases of a simultaneous mixed
spread and illness association in coxsackie and echovirus infections. Am J pneumococcal-viral infection. Isr J Med Sci. 1971;7:12401247.
Epidemiol. 1969;89:5161. 103. Miller SA, Wald ER, Bergman I, et al. Enteroviral meningitis in January with
72. Whitehead JE. Silent infections and the epidemiology of viral carditis. Am marked cerebrospinal fluid pleocytosis. Pediatr Infect Dis. 1986;5:706707.
Heart J. 1973;85:711713. 104. Mori I, Matsumoto K, Hatano M, et al. An unseasonable winter outbreak
73. Melnick JL. Enteroviruses. In: AS Evans, ed. Viral Infections of Humans: of echovirus type 30 meningitis. J Infect. 1995;31:219223.
Epidemiology and Control. New York: Plenum Medical Book Co; 105. Chambon M, Archimbaud C, Bailly JL, et al. Circulation of enteroviruses
1976:163207. and persistence of meningitis cases in the winter of 19992000. J Med
74. Mller U, Steinhoff U, Reis LF, et al. Functional role of type I and type II Virol. 2001;65:340347.
interferons in antiviral defense. Science. 1994;264:19181921. 106. Huang QS, Carr JM, Nix WA, et al. An echovirus type 33 winter outbreak
75. Ohka S, Igarashi H, Nagata N, et al. Establishment of a poliovirus oral in New Zealand. Clin Infect Dis. 2003;37:650657.
infection system in human poliovirus receptor-expressing transgenic mice that 107. Antona D, Lvque N, Chomel JJ, et al. Surveillance of enteroviruses in
are deficient in alpha/beta interferon receptor. J Virol. 2007;81:79027912. France, 20002004. Eur J Clin Microbiol Infect Dis. 2007;26:403412.
76. Bodian D. Pathogenesis of poliomyelitis. Am J Public Health Nations 108. Druyts-Voets E. Epidemiological features of entero non-poliovirus isola-
Health. 1952;42:13881402. tions in Belgium 198094. Epidemiol Infect. 1997;119:7177.
77. Huber, SA, Haisch C, Lodge PA. Functional diversity in vascular endothe- 109. Cherry JD. Enteroviruses and parechoviruses. In: Feigin RD, Cherry JD,
lial cellsrole in coxsackievirus tropism. J Virol. 1990;64:45164522. Demmler GJ, et al, eds. Textbook of Pediatric Infectious Diseases. 5th ed.
78. Coyne CB, Kim KS, Bergelson JM. Poliovirus entry into human brain mi- Philadephia: WB Saunders; 2004:19842041.
crovascular cells requires receptor-induced activation of SHP-2. EMBO J. 110. Roth B, Enders M, Arents A, et al. Epidemiologic aspects and laboratory
2007;26:40164028. features of enterovirus infections in Western Germany, 20002005. J Med
79. Coyne CB, Bozym R, Morosky SA, et al. Comparative RNAi screening Virol. 2007;79:956962.
reveals host factors involved in enterovirus infection of polarized endothe- 111. Schmidt NJ, Lennette EH, Ho HH. An apparently new enterovirus iso-
lial monolayers. Cell Host Microbe. 2011;9:7082. lated from patients with disease of the central nervous system. J Infect
80. Yang WX, Terasaki T, Shiroki K, et al. Efficient delivery of circulating po- Dis. 1974;129:304309.
liovirus to the central nervous system independently of poliovirus receptor. 112. Trallero G, Casas I, Tenorio A, et al. Enteroviruses in Spain: virological
Virology. 1997;229:421428. and epidemiological studies over 10 years (198897). Epidemiol Infect.
81. Nathanson, N, Langmuir A. The Cutter incident: poliomyelitis following 2000;124:497506.
formaldehyde-inactivated poliovirus vaccination in the United States dur- 113. Gharbi J, Jadane H, Ben Mhadheb M, et al. Epidemiological study of
ing the spring of 1955: III. Comparison of the clinical character of vac- non-polio enterovirus neurological infections in children in the region of
cinated and contact cases occurring after use of high rate lots of Cutter Monastir, Tunisia. Diagn Microbiol Infect Dis. 2006;54:3136.
vaccine. Am J Hyg. 1963;78:6181. 114. Tan CY, Ninove L, Gaudart J, et al. A retrospective overview of enterovi-
82. Sutter RW, Patriarca PA, Suleiman AJ, et al. Attributable risk of DTP rus infection diagnosis and molecular epidemiology in the public hospitals
(diphtheria and tetanus toxoids and pertussis vaccine) injection in provok- of Marseille, France (19852005). PLoS One. 2011;6:e18022.
ing paralytic poliomyelitis during a large outbreak in Oman. J Infect Dis. 115. Yamashita K, Miyamura K, Yamadera S, et al. Epidemics of asep-
1992;165:444449. tic meningitis due to echovirus 30 in Japan. A report of the National
83. Strebel PM, Ion-Nedelcu N, Baughman AL, et al. Intramuscular injec- Epidemiological Surveillance of Infectious Agents in Japan. Jpn J Med Sci
tions within 30 days of immunization with oral poliovirus vaccinea Biol. 1994;47:221239.
risk factor for vaccine-associated paralytic poliomyelitis. N Engl J Med. 116. Centers for Disease Control and Prevention. Outbreak of aseptic men-
1995;332:500506. ingitis associated with multiple enterovirus serotypesRomania, 1999.
84. Hurst EW. The newer knowledge of virus diseases of the nervous system: a MMWR Morb Mortal Wkly Rep. 2000;49:669671.
review and an interpretation. Brain. 1936;59:134. 117. Baron RC, Hatch MH, Kleeman K, et al. Aseptic meningitis among mem-
85. Nathanson N, Bodian D. Experimental poliomyelitis following intramus- bers of a high school football team. An outbreak associated with echovi-
cular virus injection: 1. The effect of neural block on a neurotropic and a rus 16 infection. JAMA. 1982;248:17241727.
pantropic strain. Bull Johns Hopkins Hospital. 1961;108:308319. 118. Modlin JF, Kinney JS. Perinatal enterovirus infections. Adv Pediatr Infect
86. Ohka S, Yang WX, Terada E, et al. Retrograde transport of intact polio- Dis. 1987;2:5778.
virus through the axon via the fast transport system. Virology. 1998;250: 119. Lenaway DD, Brockmann R, Dolan GJ, et al. An outbreak of an enterovi-
6775. rus-like illness at a community wading pool: implications for public health
87. Ren R, Racaniello V. Human poliovirus receptor gene expression and inspection programs. Am J Public Health. 1989;79:889890.
poliovirus tissue tropism in transgenic mice. J Virol. 1992;66:296304. 120. Alexander JP Jr, Chapman LE, Pallansch MA, et al. Coxsackievirus B2
88. Gromeier M, Wimmer E. Mechanism of injury-provoked poliomyelitis. infection and aseptic meningitis: a focal outbreak among members of a
J Virol. 1998;72:50565060. high school football team. J Infect Dis. 1993;167:12011205.
89. Minor PD. The molecular biology of poliovaccines. J Gen Virol. 121. Somekh E, Shohat T, Handsher R, et al. An outbreak of echovirus 11 in a
1992;73:30653077. childrens home Epidemiol Infect. 2001;126:441444.
90. Price RA, Garcia JH, Rightsel WA. Choriomeningitis and myocarditis in 122. Faustini A, Fano V, Muscillo M, et al. An outbreak of aseptic meningitis
an adolescent with isolation of coxsackie B-5 virus. Am J Clin Pathol. due to echovirus 30 associated with attending school and swimming in
1970;53:825831. pools. Int J Infect Dis. 2006;10:291297.

Scheld_Ch05.indd 79 2/21/14 5:30 PM


80 Part II: Viral Infections and Related Disorders

123. Nakao T, Miura R. Recurrent virus meningitis. Pediatrics. 1971;47: 155. Helfand RF, Khan AS, Pallansch MA, et al. Echovirus 30 infection and
773776. aseptic meningitis in parents of children attending a child care center.
124. Chang TW. Recurrent viral infection (reinfection). N Engl J Med. J Infect Dis. 1994;169:11331137.
1971;284:765773. 156. Rice SK, Heinl RE, Thornton LL, et al. Clinical characteristics, manage-
125. Aintablian N, Pratt RD, Sawyer MH. Rapidly recurrent enteroviral ment strategies, and cost implications of a statewide outbreak of enterovi-
meningitis in non-immunocompromised infants caused by different viral rus meningitis. Clin Infect Dis. 1995;20:931937.
strains. J Med Virol. 1995;47:126129. 157. Rotbart HA, Brennan PJ, Fife KH, et al. Enterovirus meningitis in adults.
126. Wright HT Jr, McAllister RM, Ward R. Mixed meningitis. Report of Clin Infect Dis. 1998;27:896898.
a case with isolation of Haemophilus influenzae type B and ECHO virus 158. Wilfert CM, Buckley RH, Mohanakumar T, et al. Persistent and fatal
type 9 from the cerebrospinal fluid. N Engl J Med. 1962;267:142144. central-nervous-system ECHOvirus infections in patients with agamma-
127. Phillips CA, Melnick JL, Barrett FF, et al. Dual virus infections. globulinemia. N Engl J Med. 1977;296:14851489.
Simultaneous enteroviral disease and St. Louis encephalitis. JAMA. 159. McKinney RE Jr, Katz SL, Wilfert CM. Chronic enteroviral meningo-
1966;197:169172. encephalitis in agammaglobulinemic patients. Rev Infect Dis. 1987;9:
128. Balfour HH Jr, Seifert GL, Seifert MH Jr, et al. Meningoencephalitis and 334356.
laboratory evidence of triple infection with California encephalitis virus, 160. Cunningham CK, Bonville CA, Ochs HD, et al. Enteroviral meningoen-
echovirus 11 and mumps. Pediatrics. 1973;51:680684. cephalitis as a complication of X-linked hyper IgM syndrome. J Pediatr.
129. Eglin RP, Swann RA, Isaacs D, et al. Simultaneous bacterial and viral 1999;134(5):584588.
meningitis. Lancet. 1984;2:984. 161. Halliday E, Winkelstein J, Webster AD. Enteroviral infections in primary
130. Pelkonen T, Roine I, Anjos E, et al. Picornaviruses in cerebrospinal fluid immunodeficiency (PID): a survey of morbidity and mortality. J Infect.
of children with meningitis in Luanda, Angola. J Med Virol. 2012;84: 2003;46:18.
10801083. 162. Webster ADB, Rotbart HA, Warner T, et al. Diagnosis of enterovirus
131. Yamashita K, Miyamura K, Yamadera S, et al. Enteroviral aseptic menin- brain disease in hypogammaglobulinemic patients by polymerase chain
gitis in Japan, 19811991. Jpn J Med Sci Biol. 1992;45:151161. reaction. Clin Infect Dis. 1993;17:657661.
132. Dai-ming W, Guo-chang Z, Shi-mei Z, et al. An epidemic of encephali- 163. Rotbart HA, Kinsella JP, Wasserman RL. Persistent enterovirus infection
tis and meningoencephalitis in children caused by echovirus type 30 in in culture-negative meningoencephalitis-demonstration by enzymatic
Shanghai. Chin Med J. 1993;106:767769. RNA amplification. J Infect Dis. 1990;161:787791.
133. Ray CG, McCollough RH, Doto IL, et al. Echo 4 illness: epidemiological, 164. Khetsuriani N, Lamonte A, Oberste MS, et al. Neonatal enterovirus
clinical and laboratory studies of an outbreak in a rural community. Am J infections reported to the national enterovirus surveillance system in the
Epidemiol. 1966;84:253267. United States, 19832003. Pediatr Infect Dis J. 2006;25:889893.
134. Kinnunen E, Hovi T, Stenvik, et al. Localized outbreak of enteroviral men- 165. Farmer K, MacArthur GA, Clay MM. A follow-up study of 15 cases
ingitis in adults. Acta Neurol Scand. 1987;74:346351. of neonatal meningoencephalitis due to coxsackie virus B5. J Pediatr.
135. Kao CH, Lee SS, Liu YC, et al. Outbreak of aseptic meningitis among 1975;87:568571.
adults in southern Taiwan. J Microbiol Immunol Infect. 2003;36:192196. 166. Sells CJ, Carpenter RL, Ray CG. Sequelae of central-nervous-system
136. Carrol ED, Beadsworth MB, Jenkins N, et al. Clinical and diagnostic find- enterovirus infections. N Engl J Med. 1975;293:14.
ings of an echovirus meningitis outbreak in the north west of England. 167. Wilfert CM, Thompson RJ, Sunder TR, et al. Longitudinal assessment of
Postgrad Med J. 2006;82:6064. children with enteroviral meningitis during the first three months of life.
137. Horstmann DM, Yamada N. Enterovirus infections of the central nervous Pediatrics. 1981;67:811815.
system. Res Publ Assoc Res Nerv Ment Dis. 1968;44:236253. 168. Bergman I, Painter MJ, Wald ER, et al. Outcome in children with entero-
138. Modlin JF. Perinatal echovirus infection: insights from a literature review viral meningitis during the first year of life. J Pediatr. 1987;110:705709.
of 61 cases of serious infection and 16 outbreaks in nurseries. Rev Infect 169. Etter CG, Wedgwood J, Schaad UB. Aseptische memingitiden in der
Dis. 1986;8:918926. padiatrie. Schweiz Med Wochenschr. 1991;121:11201126.
139. Abzug MJ, Levin MJ, Rotbart HA. Profile of enterovirus disease in the 170. Severien C, Jacobs KH, Schoenemann W. Marked pleocytosis and hypogly-
first two weeks of life. Pediatr Infect Dis J. 1993;12:820824. corrhachia in coxsackie meningitis. Pediatr Infect Dis J. 1994;13:322323.
140. Lin TY, Kao HT, Hsieh SH, et al. Neonatal enterovirus infections: em- 171. Yeager AS, Bruhn FW, Clark J. Cerebrospinal fluid: presence of virus
phasis on risk factors of severe and fatal infections. Pediatr Infect Dis J. unaccompanied by pleocytosis. J Pediatr. 1974;85:578579.
2003;22:889894. 172. Miller DG, Gabrielson MO, Bart KJ, et al. An epidemic of aseptic menin-
141. Lake AM, Lauer BA, Clark JC, et al. Enterovirus infections in neonates. gitis, primarily among infants, caused by echovirus 11-prime. Pediatrics.
J Pediatr. 1976;89:787791. 1968;41:7790.
142. Rorabaugh ML, Berlin LE, Heldrich F, et al. Aseptic meningitis in infants 173. Feigin RD, Shackelford PG. Value of repeat lumbar puncture in the dif-
younger than 2 years of age: acute illness and neurologic complications. ferential diagnosis of meningitis. N Engl J Med. 1973;289:571574.
Pediatrics. 1993;92:206211. 174. Amir J, Harel L, Frydman M, et al. Shift of cerebrospinal polymorpho-
143. Kibrick S, Benirschke K. Severe generalized disease (encephalohepato- nuclear cell percentage in the early stage of aseptic meningitis. J Pediatr.
myocarditis) occurring in the newborn period and due to infection with 1991;119:938940.
coxsackie virus, group B: evidence of intrauterine infection with this 175. Bond JO, Quick DT, Lewis AL, et al. The 1962 epidemic of St. Louis
agent. Pediatrics. 1958;22:857874. encephalitis in Florida. II. Follow-up serologic surveys for prevalence of
144. Wilfert CM, Lehrman SN, Katz SL. Enteroviruses and meningitis. Pediatr Group B arbovirus antibodies. Am J Epidemiol. 1966;83:564570.
Infect Dis J. 1983;2:333341. 176. Melnick JL, Wenner HA, Phillip CA. Enteroviruses. In: Lennette EH,
145. Dagan R, Jenista JA, Menegus MA. Association of clinical presentation, lab- Schmidt NJ, eds. Diagnostic Procedures for Viral, Rickettsial and
oratory findings, and virus serotypes with the presence of meningitis in hos- Chlamydial Infections. 5th ed. Washington, DC: American Public Health
pitalized infants with enterovirus infection. J Pediatr. 1988;113:975978. Association; 1979:471534.
146. Jarvis WR, Tucker G. Echovirus type 7 meningitis in young children. Am 177. Chonmaitree T, Baldwin CD, Lucia HL. Role of the virology laboratory
J Dis Child. 1981;135:10091012. in diagnosis and management of patients with central nervous system dis-
147. Singer JI, Maur PR, Riley JP, et al. Management of central nervous system ease. Clin Microbiol Rev. 1989;2:114.
infections during an epidemic of enteroviral aseptic meningitis. J Pediatr. 178. Rotbart HA. Enteroviral infections of the central nervous system. Clin
1980;96:559563. Infect Dis. 1995;20:971981.
148. Moore M, Kaplan MH, McPhee J, et al. Epidemiologic, clinical, and 179. Van Doornum GJ, De Jong JC. Rapid shell vial culture technique for
laboratory features of Coxsackie B1-B5 infections in the United States, detection of enteroviruses and adenoviruses in fecal specimens: com-
197079. Public Health Rep. 1984;99:515522. parison with conventional virus isolation method. J Clin Microbiol.
149. Haynes RE, Cramblett HG, Kronfol HJ. Echovirus 9 meningoencephalitis 1998;36:28652868.
in infants and children. JAMA. 1969;208:16571660. 180. Buck GE, Wiesemann M, Stewart L. Comparison of mixed cell culture
150. Jaffe M, Srugo I, Tirosh E, et al. The ameliorating effect of lumbar punc- containing genetically engineered BGMK and CaCo-2 cells (Super EMix)
ture in viral meningitis. Am J Dis Child. 1989;143:682685. with RT-PCR and conventional cell culture for the diagnosis of enterovi-
151. Desmond RA, Accortt NA, Talley L, et al. Enteroviral meningitis: nat- rus meningitis. J Clin Virol. 2002;25:S13S18.
ural history and outcome of pleconaril therapy. Antimicrob Agents 181. Dagan R, Menegus MA. A combination of four cell types for rapid detec-
Chemother. 2006;50:24092414. tion of enteroviruses in clinical specimens. J Med Virol. 1986;19:219228.
152. Peigue-Lafeuille H, Croquez N, Laurichesse H, et al. Enterovirus meningi- 182. Melnick JL. Diagnostic Procedures for Viral, Rickettsial and Chlamydial
tis in adults in 19992000 and evaluation of clinical management. J Med Infections. 5th ed. Washington, DC: American Public Health Association;
Virol. 2002;67:4753. 1979.
153. Chemtob S, Reece ER, Mills EL. Syndrome of inappropriate secre- 183. Sawyer MH, Holland D, Aintablian N, et al. Diagnosis of enteroviral cen-
tion of antidiuretic hormone in enteroviral meningitis. Am J Dis Child. tral nervous system infection by polymerase chain reaction during a large
1985;139:292294. community outbreak. Pediatr Infect Dis J. 1994;13:177182.
154. Moses EB, Dean AG, Hatch MH, et al. An outbreak of aseptic meningitis 184. Ramers C, Billman G, Hartin M, et al. Impact of a diagnostic cerebrospi-
in the area of Fort Smith, Arkansas, 1975, due to echovirus type 4. J Ark nal fluid enterovirus polymerase chain reaction test on patient manage-
Med Soc. 1977;74:121125. ment. JAMA. 2000;283:26802685.

Scheld_Ch05.indd 80 2/21/14 5:30 PM


Chapter 5: Viral Meningitis and Aseptic Meningitis Syndrome 81

185. Robinson CC, Willis M, Meagher A, et al. Impact of rapid polymerase 214. Romero JR, Simonsen KA. Powassan encephalitis and Colorado tick
chain reaction results on management of pediatric patients with enterovi- fever. Infect Dis Clin North Am. 2008;22:545559.
ral meningitis. Pediatr Infect Dis J. 2002;21:283286. 215. Mansfield KL, Johnson N, Phipps LP, et al. Tick-borne encephalitis virusa
186. Archimbaud C, Mirand A, Chambon M, et al. Improved diagnosis on a review of an emerging zoonosis. J Gen Virol. 2009;90:17811794.
daily basis of enterovirus meningitis using a one-step real-time RT-PCR 216. Smithburn KC, Hughes TP, Burke AW, et al. A neurotropic virus isolated
assay. J Med Virol. 2004;74:604611. from the blood of a native of Uganda. Am J Trop Med Hyg. 1940;20:
187. Tattevin P, Minjolle S, Arvieux C, et al. Benefits of management strategy 471492.
adjustments during an outbreak of enterovirus meningitis in adults. Scand 217. Centers for Disease Control and Prevention. Outbreak of West Nile-like
J Infect Dis. 2002;34:359361. viral encephalitisNew York. MMWR Morb Mortal Wkly Rep.1999;
188. King RL, Lorch SA, Cohen DM, et al. Routine cerebrospinal fluid 48:845849.
enterovirus polymerase chain reaction testing reduces hospitalization 218. Centers for Disease Control and Prevention. Statistics, surveillance, and
and antibiotic use for infants 90 days of age or younger. Pediatrics. control archive. http://www.cdc.gov/ncidod/dvbid/westnile/surv&control_
2007;120:489496. archive.htm. Accessed January 10, 2013.
189. Johnson GM, McAbee GA, Seaton ED, et al. Suspect value of non-CSF 219. Petersen LR, Hayes EB. West Nile virus in the Americas. Med Clin North
viral cultures in the diagnosis of enteroviral CNS infection in young Am. 2008;92:13071322.
infants. Dev Med Child Neurol. 1992;34:876884. 220. Centers for Disease Control and Prevention. Summary of notifiable dis-
190. Abzug MJ, Keyserling HL, Lee ML, et al. Neonatal enterovirus infection: easesUnited States, 2010. MMWR Morb Mortal Wkly Rep. 2010;59:
virology, serology, and effects of intravenous immune globulin. Clin Infect 1111.
Dis. 1995;20:12011206. 221. Simmonds P, Becher P, Collet MD, et al. Flaviviridae. In: King AMQ,
191. Ruan F, Yang T, Ma H, et al. Risk factors for hand, foot, and mouth Adams MJ, Carstens EB, et al, eds. Virus Taxonomy. Classification and
disease and herpangina and the preventive effect of hand-washing. Nomenclature of Viruses: Ninth Report of the International Committee on
Pediatrics. 2011;127:e898e904. the Taxonomy of Viruses. London: Elsevier Academic Press; 2012:855880.
192. Meng FY, Li JX, Li XL, et al. Tolerability and immunogenicity of an inacti- 222. Romero JR. West Nile virus infections. In: Feigin RD, Cherry JD,
vated enterovirus 71 vaccine in Chinese healthy adults and children: an open Demmler GJ, et al, eds. Textbook of Pediatric Infectious Diseases. 6th ed.
label, phase 1 clinical trial. Hum Vaccin Immunother. 2012;8:668674. Philadelphia: Saunders Elsevier; 2009:23312343.
193. Wigand R, Sabin AB. Properties of ECHO types 22, 23 and 24 viruses. 223. Hayes EB, Komar N, Nasci RS, et al. Epidemiology and transmission
Arch Gesamte Virusforsch. 1961;11:224247. dynamics of West Nile virus disease. Emerg Infect Dis. 2005;11:11671173.
194. Shaver DN, Barron AL, Karzon DT. Distinctive cytopathology of ECHO 224. Unlu I, Mackay AJ, Roy A, et al. Evidence of vertical transmission of West
viruses type 22 and 23. Proc Soc Exp Biol Med. 1961;106:648652. Nile virus in field-collected mosquitoes. J Vector Ecol. 2010;35:9599.
195. Coller BA, Chapman NM, Beck MA, et al. Echovirus 22 is an atypical 225. Murray K, Walker C, Herrington E, et al. Persistent infection with West
enterovirus. J Virol. 1990;64:26922701. Nile virus years after initial infection. J Infect Dis. 2011;201:24.
196. Coller BG, Tracy SM, Etchison D. Cap-binding complex protein p220 226. Meny GM, Santos-Zabala L, Szallasi A, et al. West Nile virus infection
is not cleaved during echovirus 22 replication in HeLa cells. J Virol. transmitted by granulocyte transfusion. Blood. 2011;117:57785779.
1991;65:39033905. 227. Montgomery SP, Brown JA, Kuehnert M, et al. Transfusion-associated
197. Tapia G, Cinek O, Wits E, et al. Longitudinal observation of parecho- transmission of West Nile virus, United States 2003 through 2005.
virus in stool samples from Norwegian infants. J Med Virol. 2008;80: Transfusion. 2006;46:20382046.
18351842. 228. Iwamoto M, Jernigan DB, Guasch A, et al. Transmission of West Nile
198. Abed Y, Boivin G. Human parechovirus infections in Canada. Emerg virus from an organ donor to four transplant recipients. N Engl J Med.
Infect Dis. 2006;12:969975. 2003;348:21962203.
199. Pham NTK, Trinh QD, Khamrin P, et al. Diversity of human parechovi- 229. Pealer LN, Marfin AA, Petersen LR, et al. Transmission of West Nile virus
ruses isolated from stool samples collected from Thai children with acute through blood transfusion in the United States in 2002. N Engl J Med.
gastroenteritis. J Clin Microbiol. 2010;48:115119. 2003;349:12361245.
200. Tauriainen S, Martiskainen M, Oikarinen S, et al. Human parechovirus 1 230. Centers for Disease Control and Prevention. Possible West Nile virus
infections in young childrenno association with type 1 diabetes. J Med transmission to an infant through breast-feedingMichigan, 2002.
Virol. 2007;79:457462. MMWR Morb Mortal Wkly Rep. 2002;51:877878.
201. Harvala H, Robertson I, McWilliam, et al. Epidemiology and clini- 231. Centers for Disease Control and Prevention. Intrauterine West Nile
cal associations of human parechovirus respiratory infections. J Clin virus infectionNew York, 2002. MMWR Morb Mortal Wkly Rep.
Microbiol. 2008;46:34463453. 2002;51:11351136.
202. Selvarangan R, Nzabi M, Selvaraju SB, et al. Human parechovirus 3 caus- 232. Spigland I, Jasinska-Klingberg W, Hofsbi E, et al. Clinical and labora-
ing sepsis-like illness in children from Midwestern United States. Pediatr tory observations in an outbreak of West Nile fever in Israel. Harefuah.
Infect Dis. 2011;30:238242. 1958;54:275281.
203. Walters B, Pearanda S, Nix WA, et al. Detection of human parechovirus 233. Chowers MY, Lang R, Nassar F, et al. Clinical characteristics of the West
(HPeV)-3 in spinal fluid specimens from pediatric patients in the Chicago Nile fever outbreak, Israel, 2000. Emerg Infect Dis. 2001;7:675678.
area. J Clin Virol. 2011;52:187191. 234. Hayes EB, OLeary DR. West Nile virus infection: a pediatric perspective.
204. Renaud C, Kuypers J, Ficken E, et al. Introduction of a novel parecho- Pediatrics. 2004;113:13751381.
virus RT-PCR clinical test in a regional medical center. J Clin Virol. 235. Sejvar JJ, Marfin AA. Manifestations of West Nile neuroinvasive disease.
2011;51:5053. Rev Med Virol. 2006;16:209224.
205. Harvala H, Robertson I, Chieochansin T, et al. Specific association of 236. Lindsey NP, Hayes EB, Staples E, et al. West Nile disease in children,
human parechovirus type 3 with sepsis and fever in young infants, as United States, 19992007. Pediatrics. 2009;123:e10841089.
identified by direct typing of cerebrospinal fluid samples. J Infect Dis. 237. Mandalakas AM, Kippes C, Sedransk J, et al. West Nile virus epidemic,
2009;199:17531760. Northeast Ohio, 2002. Emerg Infect Dis. 2005;11:17741777.
206. Verboon-Maciolek MA, Krediet TG, Gerards LJ, et al. Severe neonatal 238. Murray K, Baraniuk S, Resnick M, et al. Risk factors for encephalitis
parechovirus infection and similarity with enterovirus infection. Pediatr and death from West Nile virus infection. Epidemiol Infect. 2006;134:
Infect Dis J. 2008;7:241245. 13251332.
207. Wolthers KC, Benschop KS, Schinkel J, et al. Human parechoviruses as 239. Goldblum N, Sterk VV, Paderski B. West Nile fever. The clinical features
an important viral cause of sepsis like illness and meningitis in young of the disease and the isolation of West Nile virus from the blood of nine
children. Clin Infect Dis. 2008;47:358363. human cases. Am J Hyg. 1954;59:89103.
208. van der Sanden S, de Bruin E, Vennema H, et al. Prevalence of human 240. Marberg K, Goldblum N, Sterk VV, et al. The natural history of West
parechovirus in the Netherlands in 2000 to 2007. J Clin Virol. 2008; Nile fever. Clinical observations during an epidemic in Israel. Am J Hyg.
46:28842889. 1956;64:259269.
209. Pajkrt D, Benschop KS, Westerhuis B, et al. Clinical characteristics of 241. Campbell GL, Marfin AA, Lanciotti RS, et al. West Nile virus. Lancet
human parechoviruses 4-6 infections in young children. Pediatr Infect Dis Infect Dis. 2002;2:519529.
J. 2009;28:10081010. 242. Tsai TF, Popovici F, Cernescu C, et al. West Nile encephalitis epidemic in
210. Pieiro L, Vicente D, Montes M, et al. Human parechovirus in infants southeastern Romania. Lancet. 1998;352:767771.
with systemic infection. J Med Virol. 2010;82:17901796. 243. Mostashari F, Bunning ML, Kitsutani PT, et al. Epidemic West Nile
211. Benschop K, Minnaar R, Koen G, et al. Detection of human enterovirus encephalitis, New York, 1999: results of a household-based seroepide-
and human parechovirus (HPeV) genotypes from clinical stool samples: miological survey. Lancet. 2001;358:261264.
polymerase chain reaction and direct molecular typing, culture character- 244. Sejvar JJ, Haddad MB, Tierney BC, et al. Neurologic manifestations and
istics, and serotyping. Diagn Microbiol Infect Dis. 2010;68:166173. outcome of West Nile virus infection. JAMA. 2003;290:511515.
212. Joki-Korpela P, Hyypi T. Diagnosis and epidemiology of echovirus 22 245. Emig M, Apple DJ. Severe West Nile virus disease in healthy adults. Clin
infections. Clin Infect Dis. 1998;27:129136. Infect Dis. 2004;38:289292.
213. Lanciotti RS, Tsai TF. Arboviruses. In: Murray PR, Baron EJ, Jorgensen 246. LaBeaud AD, Lisgaris MV, King CH, et al. Pediatric West Nile virus
JH, et al, eds. Manual of Clinical Microbiology. 9th ed. Washington, DC: infection: neurologic disease presentations during the 2002 epidemic in
American Society for Microbiology; 2007:14861500. Cuyahoga County, Ohio. Pediatr Infect Dis J. 2006;25:751753.

Scheld_Ch05.indd 81 2/21/14 5:30 PM


82 Part II: Viral Infections and Related Disorders

247. Centers for Disease Control and Prevention. Surveillance for human 277. Bang HO, Bang J. Involvement of the central nervous system in mumps.
West Nile DiseaseUnited States, 19992008. MMWR Surveill Summ. Acta Med Scand. 1943;113:487505.
2010;59(SS-2):118. 278. Centers for Disease Control and Prevention. Measles, mumps, and
248. Crichlow R, Bailey J, Gardner C. Cerebrospinal fluid pleocytosis in rubellavaccine use and strategies for elimination of measles, rubella,
hospitalized West Nile virus patients. J Am Board Fam Pract. 2004;17: and congenital rubella syndrome and control of mumps. MMWR
470472. Recomm Rep. 1998;47:167.
249. Carson PJ, Steidler T, Patron R, et al. Plasma cell pleocytosis in cerebro- 279. McLean DM, Bach RD, Larke RPB, et al. Mumps meningoencephalitis,
spinal fluid in patients with West Nile virus encephalitis. Clin Infect Dis. Toronto, 1963. Can Med Assoc J. 1964;90:458462.
2003;37:e12e15. 280. Azimi PH, Cramblett HG, Haynes RE. Mumps meningoencephalitis in
250. Hayes EB, Sejvar JJ, Zaki SR, et al. Virology, pathology, and clinical children. JAMA. 1969;207:509512.
manifestations of West Nile virus disease. Emerg Infect Dis. 2005;11: 281. Johnstone JA, Ross CAC, Dunn M. Meningitis and encephalitis associ-
11741179. ated and mumps infection: a 10-year survey. Arch Dis Child. 1972;47:
251. Tardei G, Ruta S, Chitu V, et al. Evaluation of immunoglobulin M (IgM) 647651.
and IgG enzyme immunoassays in serologic diagnosis of West Nile Virus 282. Strussberg S, Winter S, Friedman A, et al. Notes on mumps menin-
infection. J Clin Microbiol. 2000;38:22322239. goencephalitis: some features of 199 cases in children. Clin Pediatr.
252. Kapoor H, Signs K, Somsel P, et al. Persistence of West Nile virus (WNV) 1969;8:373377.
IgM antibodies in cerebrospinal fluid from patients with CNS disease. 283. Leland DS. Parainfluenza and mumps viruses. In: Murray PR, Baron
J Clin Virol. 2004;31:289291. EJ, Jorgensen JH, et al, eds. Manual of Clinical Microbiology. 9th ed.
253. Tsai TF, Chandler LJ. Arboviruses. In: Murray PR, Baron EJ, Jorgensen Washington, DC: American Society for Microbiology Press; 2007;
JH, eds. Manual of Clinical Microbiology. 8th ed. Washington, DC: 13521360.
American Society for Microbiology Press; 2003:1553. 284. Hnninen P, Arstila P, Lang H, et al. Involvement of the central nervous
254. Webster LT, Fite GL. A virus encountered in the study of material from system in acute, uncomplicated measles virus infection. J Clin Microbiol.
cases of encephalitis in the St. Louis and Kansas City epidemics of 1933. 1980;11:610613.
Science. 1933;78:463465. 285. Newland JG, Romero JR. Myxoviruses. In: Barton LL, Friedman NR,
255. Reimann CA, Hayes EB, DiGuiseppi C, et al. Epidemiology of neuroin- eds. The Neurological Manifestations of Pediatric Infectious Disease and
vasive arboviral disease in the United States, 19992007. Am J Trop Med Immunodeficiency Syndromes. Totowa, NJ: Humana Press; 2008:4168.
Hyg. 2008;79:974979. 286. Salvato MS, Clegg JCS, Buchmeier MJ, et al. Arenaviridae. In: King AMQ,
256. Meehan PJ, Wells DL, Paul W, et al. Epidemiological features of and Adams MJ, Carstens EB, et al, eds. Virus Taxonomy. Classification and
public health response to a St. Louis encephalitis epidemic in Florida, Nomenclature of Viruses: Ninth Report of the International Committee
19901991. Epidemiol Infect. 2000;125:181188. on the Taxonomy of Viruses. London: Elsevier Academic Press; 2012:
257. Centers for Disease Control and Prevention. St. Louis encephalitis 715723.
epidemiology & geographic distribution. http://www.cdc.gov/sle/technical 287. Childs JE, Glass GE, Korch GW, et al. Lymphocytic choriomeningitis
/epi.html. Accessed December 29, 2012. virus infection and house mouse (Mus musculus) distribution in urban
258. Zweighaft RM, Rasmussen C, Brolnitsky O, et al. St. Louis encephalitis: Baltimore. Am J Trop Med Hyg. 1992:47;2734.
the Chicago experience. Am J Trop Med Hyg. 1979;28:114118. 288. Park JY, Peters CJ, Rollin PE, et al. Age distribution of lymphocytic cho-
259. Centers for Disease Control and Prevention. Tick-borne encephalitis riomeningitis virus serum antibody in Birmingham, Alabama: evidence of
among US travelers to Europe and Asia20002009. MMWR Morb a decreased risk of infection. Am J Trop Med Hyg. 1997;57:3741.
Mortal Wkly Rep. 2010;59:335338. 289. Knust B, Macneil A, Wong SJ, et al. Exposure to lymphocytic choriomen-
260. Schneider H. ber epidemische akute Meningitis serosa. Wien Klin ingitis virus, New York, USA. Emerg Infect Dis. 2011;17:13241325.
Wochenschr. 1931;44:350352. 290. Lehmann-Grube F. Portraits of viruses: arenaviruses. Intervirology.
261. Barrett PN, Plotkin SA, Ehrlich HJ. Tick-borne encephalitis virus vac- 1984;22:121145.
cines. In: Plotkin SA, Orenstein WA, Offit PA, eds. Vaccines. 5th ed. 291. Deibel R, Woodall JP, Decher WJ, et al. Lymphocytic choriomeningitis
Philadelphia: Elsevier; 2008:841856. virus in man. Serologic evidence of association with pet hamsters. JAMA.
262. Centers for Disease Control and Prevention. West Nile virus disease and 1975;232:501504.
other arboviral diseasesUnited States, 2011. MMWR Morb Mortal 292. Vanzee BE, Doublas RG Jr, Betts RF, et al. Lymphocytic choriomen-
Wkly Rep. 2012;61:510514. ingitis in university hospital personnel. Clinical features. Am J Med.
263. Jeurissen A, Strauven T. A case of aseptic meningitis due to Japanese en- 1975;58:803809.
cephalitis virus in a traveller returning from the Philippines. Acta Neurol 293. Maetz HM, Sellers CA, Bailey WC, et al. Lymphocytic choriomeningitis
Belg. 2011;111:143145. from pet hamster exposure: a local public health experience. Am J Public
264. Kuwayama M, Ito M, Takao S, et al. Japanese encephalitis virus in men- Health. 1976;66:10821085.
ingitis patients, Japan. Emerg Infect Dis. 2005;11:471473. 294. Centers for Disease Control and Prevention. Notes from the field: lym-
265. Emmons RW. Ecology of Colorado tick fever. Annu Rev Microbiol. phocytic choriomeningitis virus infections in employees of a rodent breed-
1988;42:4964. ing facilityIndiana, MayJune 2012. MMWR Morb Mortal Wkly Rep.
266. Goodpasture HC, Poland JD, Francy DB, et al. Colorado tick fever: clini- 2012;61:622623.
cal, epidemiologic, and laboratory aspects of 228 cases in Colorado in 295. Fischer SA, Graham MB, Kuehnert MJ, et al. Human-to-human trans-
19731974. Ann Intern Med. 1978;88:303310. mission can occur through transmission of lymphocytic choriomeningitis
267. Srihongse S, Grayson MA, Deibel R. California serogroup viruses in New virus by organ transplantation. N Engl J Med. 2006;354:22352249.
York State: the role of subtypes in human infections. Am J Trop Med Hyg. 296. Macneil A, Strher U, Farnon E, et al. Solid organ transplant-associated
1984;33:12181227. lymphocytic choriomeningitis, United States, 2011. Emerg Infect Dis.
268. Wang LF, Collins PL, Fouchier RAM, et al. Paramyxoviridae. In: King 2012;18:12561262.
AMQ, Adams MJ, Carstens EB, et al, eds. Virus Taxonomy. Classification 297. Bonthius DJ. Lymphocytic choriomeningitis virus: an underrecognized
and Nomenclature of Viruses: Ninth Report of the International cause of neurologic disease in the fetus, child, and adult. Semin Pediatr
Committee on the Taxonomy of Viruses. London: Elsevier Academic Neurol. 2012;19:8995.
Press; 2012:672685. 298. Hirsch MS, Moellering RC Jr, Pope HG, et al. Lymphocytic-
269. Jin L, Rima B, Brown D, et al. Proposal for genetic characterisation of choriomeningitis-virus infection traced to a pet hamster. N Engl J Med.
wild-type mumps strains: preliminary standardisation of the nomencla- 1974;291:610612.
ture. Arch Virol. 2005;150:19031909. 299. Jahrling PB, Peters CJ. Lymphocytic choriomeningitis virus. A neglected
270. Santos CL, Ishida MA, Foster PG, et al. Detection of a new mumps virus pathogen of man. Arch Pathol Lab Med. 1992;116:486488.
genotype during parotitis epidemic of 20062007 in the state of So 300. Johnson R, Milbourn PE. Central nervous system manifestations of chick-
Paulo, Brazil. J Med Virol. 2008;80:323329. enpox. Can Med Assoc J. 1970;102:831834.
271. Centers for Disease Control and Prevention. Mumps surveillance 1973. 301. Leahy TR, Webb DWM, et al. Varicella zoster virus associated acute asep-
MMWR Morb Mortal Wkly Rep. 1974;23:431. tic meningitis without exanthem in an immunocompetent 14-year-old
272. Centers for Disease Control and Prevention. Summary of notifiable dis- boy. Pediatr Infect Dis J. 2008;27:362363.
eases, United States, 1991. MMWR Morb Mortal Wkly Rep. 1991;40:3. 302. Silverstein A, Steinberg G, Nathanson M. Nervous system involvement in
273. Centers for Disease Control and Prevention. Mumps surveillance, United infectious mononucleosis. Arch Neurol. 1972;26:353358.
States, 19881993. MMWR Morb Mortal Wkly Rep. 1995;44(SS-3):121. 303. Connelly KP, DeWitt LD. Neurologic complications of infectious mono-
274. Centers for Disease Control and Prevention. Update: multistate outbreak nucleosis. Pediatr Neurol. 1994;10:181184.
of mumpsUnited States, January 1May 2, 2006. MMWR Morb Mortal 304. Causey JQ. Spontaneous cytomegalovirus mononucleosis-like syndrome
Wkly Rep. 2006;55:559563. and aseptic meningitis. South Med J. 1976;69:13841387.
275. Barskey AE, Schulte C, Rosen JB, et al. Mumps outbreak in Orthodox 305. Ishiguro N, Yamada S, Takahashi T, et al. Meningo-encephalitis
Jewish communities in the United States. N Engl J Med. 2012;367: associated with HHV-6 related exanthem subitum. Acta Paediatr Scand.
17041713. 1990;79:987989.
276. Kilham L. Isolation of mumps virus from the blood of a patient. Proc Soc 306. Huang LM, Lee CY, Lee PI, et al. Meningitis caused by human herpesvi-
Exp Biol Med. 1948;69:99100. rus-6. Arch Dis Child. 1991;66:14431444.

Scheld_Ch05.indd 82 2/21/14 5:30 PM


Chapter 5: Viral Meningitis and Aseptic Meningitis Syndrome 83

307. Yoshikawa T, Nakashima T, Suga S, et al. Human herpesvirus-6 DNA 340. Wormser R, Dattwyler R, Shapiro E, et al. The clinical assessment, treat-
in cerebrospinal fluid of a child with exanthem subitum and meningoen- ment, and prevention of Lyme disease, human granulocytic anaplasmo-
cephalitis. Pediatrics. 1992;89:888890. sis, and babesiosis: clinical practice guidelines by the Infectious Diseases
308. Yoshikawa T, Ihira M, Suzuki K, et al. Invasion by human herpesvirus 6 Society of America. Clin Infect Dis. 2006;43:10891134.
and human herpesvirus 7 of the central nervous system in patients with 341. Halperin JJ. Nervous system Lyme disease. Infect Dis Clin North Am.
neurological signs and symptoms. Arch Dis Child. 2000;83:170171. 2008;22:261274.
309. Corey L, Adams HG, Brown ZA, et al. Genital herpes simplex virus infec- 342. Pachner AR, Steere AC. The triad of neurologic manifestations of Lyme
tions: clinical manifestations, course, and complications. Ann Intern Med. disease: meningitis, cranial neuritis, and radiculoneuritis. Neurology.
1983;98:958972. 1985;35:4753.
310. Bergstrm T, Vahlne A, Alestig K, et al. Primary and recurrent herpes 343. Feder HM. Lyme disease in children. Infect Dis Clin North Am.
simplex virus type 2-induced meningitis. J Infect Dis. 1990;162:322330. 2008;22:315326.
311. OSullivan CE, Aksamit AJ, Harrington JR, et al. Clinical spectrum 344. Lecour H, Miranda M, Magro C, et al. Human leptospirosisa review of
and laboratory characteristics associated with detection of herpes sim- 50 cases. Infection. 1989;17:1014.
plex virus DNA in cerebrospinal fluid. Mayo Clin Proc. 2003;78: 345. Merrit HH, Moore M. Acute neurosyphilitic meningitis. Medicine.
13471352. 1935;14:119.
312. Yamamoto LJ, Tedder DG, Ashley R, et al. Herpes simplex virus type 346. Cadavid D, Barbour AG. Neuroborreliosis during relapsing fever: review
1 DNA in cerebrospinal fluid of a patient with Mollarets meningitis. of the clinical manifestations, pathology, and treatment of infections in
N Engl J Med. 1991;325:10821085. humans and experimental animals. Clin Infect Dis. 1998;26:151164.
313. Tedder DG, Ashley R, Tyler KL, et al. Herpes simplex virus infection as 347. Ussery XT, Valway SE, McKenna M, et al. Epidemiology of tuberculosis
a cause of benign recurrent lymphocytic meningitis. Ann Intern Med. among children in the United States: 1985 to 1994. Pediatr Infect Dis J.
1994;121:334338. 1996;15:697704.
314. Kupila L, Vainionp R, Vuorinen T, et al. Recurrent lymphocytic menin- 348. Starke JR. Tuberculosis of the central nervous system in children. Semin
gitis: the role of herpesviruses. Arch Neurol. 2004;61:15531557. Pediatr Neurol. 1999;6:318331.
315. Kumar S, Kumar S, Kohlhoff SA. Recurrent HSV-2 meningitis in a 9-year- 349. Bouza E, Garcia de la Torre M, Parras F, et al. Brucellar meningitis. Rev
old-girl. Scand J Infect Dis. 2006;38:570572. Infect Dis. 1987;9:810822.
316. Jensenius M, Myrvang B, Strvold G, et al. Herpes simplex virus type 2 350. Wheat LJ, Batteiger BE, Sathapatayavongs B. Histoplasma capsulatum
DNA detected in cerebrospinal fluid of 9 patients with Mollarets menin- infections of the central nervous system. A clinical review. Medicine.
gitis. Acta Neurol Scand. 1998;98:209212. 1990;69:244260.
317. Kallio-Laine K, Seppnen M, Kautiainen H, et al. Recurrent lymphocytic 351. Friedman JA, Wijdicks EF, Fulgham JR, et al. Meningoencephalitis due
meningitis positive for herpes simplex virus type 2. Emerg Infect Dis. to Blastomyces dermatitidis: case report and literature review. Mayo Clin
2009;15:11191122. Proc. 2000;75:403408.
318. Shalabi M, Whitley RJ. Recurrent benign lymphocytic meningitis. Clin 352. Bicanic T, Harrison TS. Cryptococcal meningitis. Br Med Bull.
Infect Dis. 2006;43:11941197. 2005;72:99118.
319. Mayo DR, Booss J. Varicella zosterassociated neurologic disease without 353. Wheat LJ, Musial CE, Jenny-Avital E. Diagnosis and management of cen-
skin disease. Arch Neurol. 1989;46:313315. tral nervous system histoplasmosis. Clin Infect Dis. 2005;40:844852.
320. Pahud BA, Glaser CA, Dekker CL, et al. Varicella zoster disease of the 354. Johnson RH, Einstein HE. Coccidioidal meningitis. Clin Infect Dis.
central nervous system: epidemiological, clinical, and laboratory fea- 2006;42:103107.
tures 10 years after the introduction of the varicella vaccine. J Infect Dis. 355. Antinori S, Corbellino M, Meroni L, et al. Aspergillus meningitis: a rare
2011;203:316323. clinical manifestation of central nervous system aspergillosis. Case report
321. Jhaveri R, Sankar R, Yazdani S, et al. Varicella-zoster virus: an overlooked and review of 92 cases. J Infect. 2013;66:218238.
cause of aseptic meningitis. Pediatr Infect Dis J. 2003;23:9397. 356. Lind K, Zoffmann H, Larsen SO, et al. Mycoplasma pneumoniae infec-
322. Caserta MT, Hall CB, Schnabel K, et al. Neuroinvasion and persistence of tion associated with infection of the central nervous system. Acta Med
human herpesvirus 6 in children. J Infect Dis. 1994;170:15861589. Scand. 1979;205:325332.
323. Ho DD, Rota TR, Schooley RT, et al. Isolation of HTLV-III from cere- 357. Narita M, Itakura O, Matsuzono Y, et al. Analysis of mycoplasmal central
brospinal fluid and neural tissues of patients with neurologic syndromes nervous system involvement by polymerase chain reaction. Pediatr Infect
related to the acquired immunodeficiency syndrome. N Engl J Med. Dis J. 1995;14:236237.
1985;313:14931497. 358. Roe O, Jorgen D, Matre R. Isolation of Mycoplasma hominis from cere-
324. Ho DD, Sarngadharan MG, Resnick L, et al. Primary human brospinal fluid. Scand J Infect Dis. 1973;5:285288.
T-lymphotropic virus type III infection. Ann Intern Med. 1985;103: 359. Holman RC, Belay ED, Christensen KY, et al. Hospitalizations for
880883. Kawasaki syndrome among children in the United States, 19972007.
325. Schacker T, Collier AC, Hughes J, et al. Clinical and epidemiologic fea- Pediatr Infect Dis J. 2010;29:483488.
tures of primary HIV infection. Ann Intern Med. 1996;125:257264. 360. Shimura T, Arima H. Clinical features of aseptic meningitis in Kawasaki
326. Kelsey DS. Adenovirus meningoencephalitis. Pediatrics. 1978;61:291293. disease. J Jpn Pediatr. 1978;31:789792.
327. Davis D, Henslee PJ, Markesbery WR. Fatal adenovirus meningoen- 361. Watanabe J, Kawasaki T, Takemura T. Cytologic observation of spinal
cephalitis in a bone marrow transplant patient. Ann Neurol. 1988;23: fluid in Kawasaki disease. J Jpn Pediatr. 1980;84:12591263.
385389. 362. Fishman RA. CSF findings in diseases of the nervous system. In: Fishman
328. Landry ML, Hsiung GD. Adenovirus-associated meningoencephalitis in a RA, ed. Cerebrospinal Fluid in Diseases of the Nervous System. 2nd ed.
healthy adult. Ann Neurol. 1988;23:627628. Philadelphia: Saunders; 1992:277287.
329. Fianchi L, Scardocci A, Cattani P, et al. Adenovirus meningoencepha- 363. Johnson RT, Richardson EP. The neurological manifestations of systemic
litis in a patient with large B-cell lymphoma. Ann Hematol. 2003;82: lupus erythematosus. Medicine. 1968;47:337369.
313315. 364. Hopkins S, Jolles S. Drug-induced aseptic meningitis. Expert Opin Drug
330. Dubberke ER, Tu B, Rivet DJ, et al. Acute meningoencephalitis caused by Saf. 2005;4:285297.
adenovirus serotype 26. J Neurovirol. 2006;12:235240. 365. Kluger N, Girard C, Gonzalez V, et al. Efalizumab-induced aseptic menin-
331. Paisley JW, Bruhn FW, Lauer BA, et al. Type A2 influenza viral infections gitis. Br J Dermatol. 2007;156:189191.
in children. Am J Dis Child. 1978;132:3436. 366. Callen EC, Church CO, Patel M, et al. Aseptic meningitis associated with
332. Drago L, Attia MW, DePiero A, et al. Influenza A-associated meningoen- chronic sulindac use for osteoarthritis: a case report. Rheumatol Int.
cephalitis. Pediatr Emerg Care. 2005;21:437439. 2008;28:391393.
333. Kwon S, Kim S, Cho MH, et al. Neurologic complications and out- 367. Nagovskiy N, Agarwal M, Allerton J. Cetuximab-induced aseptic menin-
comes of pandemic (H1N1) 2009 in Korean children. J Korean Med Sci. gitis. J Thorac Oncol. 2010;5:751.
2012;27:402407. 368. Jazeron A, Lallier JC, Rihn B, et al. Aseptic meningitis possibly induced by
334. Douvoyiannis M, Litman N, Goldman DL. Neurologic manifestations adalimumab. Joint Bone Spine. 2010;77:618619.
associated with parvovirus B19 infection. Clin Infect Dis. 2009;48: 369. Imataka G, Nakagawa E, Yamanouchi H, et al. Drug-indiced aseptic
17131723. meningitis: development of subacute sclerosing panencephalitis following
335. Wong CJ, Price Z, Bruckner DA. Aseptic meningitis in an infant with repeated intraventricular infusion therapy with interferon alpha/beta. Cell
rotavirus gastroenteritis. Pediatr Infect Dis J. 1984;3:244246. Biochem Biophys. 2011;61:699701.
336. Okumura A, Ichikawa T. Aseptic meningitis caused by human parvovirus 370. Shah BK, OKeefe S. Pemetrexed induced aseptic meningitis. Acta Oncol.
B19. Arch Dis Child. 1993;68:784785. 2012;51:399400.
337. Furuya Y, Katayama T, Miyahara K, et al. Detection of the rotavirus A 371. Simms KM, Kortepeter C, Avigan M. Lamotrigine and aseptic meningitis.
genome from the cerebrospinal fluid of a gastroenteritis patient: a case Neurology. 2012;78:921927.
report. Jpn J Infect Dis. 2007;60:148149. 372. Galindo Bonilla PA, Snchez Rodrguez N, Castro Jimnez A, et al.
338. Centers for Disease Control and Prevention. 2010 Maplyme disease. Aseptic meningitis induced by vitamin B complex. J Investig Allergol Clin
http://www.cdc.gov/lyme/stats/maps/map2010.html. Accessed January Immunol. 2012;22:225226.
15, 2013. 373. Bonnet MC, Dutta A, Weinberger C, et al. Mumps vaccine strains and
339. Steere AC. Lyme disease. N Engl J Med. 2001;345:115125. aseptic meningitis. Vaccine. 2006;24:70377045.

Scheld_Ch05.indd 83 2/21/14 5:30 PM


CHAPTER 6 ENCEPHALITIS
CAROL GLASER AND ARUN VENKATESAN

Encephalitis is one of the most challenging syndromes for cli- and surveillance activities. In tropical regions of the world,
nicians to manage. Patients are often critically ill, and there the minimum estimated incidence of encephalitis is 6.3 per
are many potential etiologies. Despite exhaustive testing, an 100,000 (8). In the Western world, the incidence ranges
etiology is only identified in 40% to 70% of cases. Even when between 0.7 and 13.8 per 100,000 (810). Most reports find
a cause is identified, there may be no effective treatment (13). the incidence of encephalitis higher in the pediatric age-group
Mortality rates vary substantially across studies and range than in adults. For example, a study in England of hospitalized
from 3% to 15% (4,5). The frequency of sequelae, including patients with encephalitis over a 10-year period demonstrated
cognitive and motor impairment as well as seizures, is also an overall incidence of 1.5 per 100,000 population, a rate of
variable; some case series report severe disability in 20% to 2.8 per 100,000 in children, and a rate of 8.7 per 100,000 in
40% of patients (4,6,7). Not surprisingly, given the severity of infants (11). Somewhat higher rates in children were reported
the disease as well as the complexity of diagnosis and clinical from Finland (8.8 per 100,000 from 1973 to 1987) (12) and in
management, substantial health care costs are associated with Slovenia (6.7 per 100,000 from 1979 to 1991) (13).
encephalitis. The epidemiology of encephalitis is a dynamic process. In
The term encephalitis generally refers to inflammation of countries where vaccines are widely used for measles, mumps,
the brain parenchyma. However, without the identification rubella, and varicella infections, the incidence of encephali-
of a neurotropic agent or confirmation of inflammation in tis due to these viruses has decreased (14,15). However, there
brain tissue, the diagnosis of encephalitis is presumptive and is a growing list of emerging and reemerging pathogens
based on clinical features. Clinically, patients with encephalitis such as Nipah virus, enterovirus 71, Balamuthia mandril-
often present with fever, headache, and altered mental status. laris, European tick-borne encephalitis virus, Hendra virus,
Seizures or focal neurologic deficits may also be present. In Baylisascaris procyonis, and Chandipura virus that can cause
principle, alteration in mental status distinguishes encepha- encephalitis. Moreover, some agents are now identified in pre-
litis from uncomplicated meningitis as meningitis symptoms viously nonendemic regions of the world. Notable is West Nile
typically include fever, headache, and nuchal rigidity but lack virus, which has expanded its geographic region from Africa to
global or focal neurologic dysfunction. In practice, however, North and South America, Europe, the Middle East, Western
the distinction between these two entities is not always readily Asia, and Australia (16). Chikungunya virus is yet another
apparent, and in descriptions of central nervous system (CNS) striking example of a virus that has spread from its origin in
infections with mental status changes due to agents such as en- Africa to nearly 40 countries including a number of countries
teroviruses, rabies, West Nile virus or herpesviruses, the terms adjacent to the Indian Ocean: La Reunion Island, Madagascar,
encephalitis or meningoencephalitis are often broadly applied. the Maldives, the Seychelles, and India. (17).
In contrast to encephalitis, encephalopathy refers to The increasing recognition of specific autoimmune causes,
any diffuse disease of the brain that results in changes in as discussed later in this chapter, has also had a tremendous im-
function; the clinical hallmark of encephalopathy is an al- pact on our understanding of the epidemiology of encephalitis.
tered mental state. Many entities including metabolic or
mitochondrial dysfunction, toxins, trauma, poor nutrition,
or lack of oxygen or blood flow to the brain can lead to
encephalopathy.
INFECTIOUS CAUSES: SPECIFIC
This chapter focuses predominantly on the immunocom- EPIDEMIOLOGIC AND CLINICAL
petent host and pathogens in North America that either cause FEATURES
encephalitis or an encephalitis-like syndrome. Moreover, many
patients with encephalitis also have meningeal inflammation,
thus demonstrating the overlap of encephalitis and menin- Viruses
goencephalitis. For the purposes of this chapter, the terms
encephalitis and meningoencephalitis are used interchangeably. Many cases of viral encephalitis are either an uncommon
Other regions of the CNS may be variably affected, including complication of a common infection, such as a herpesvirus or
the spinal cord (myelitis), nerve roots (radiculitis), and nerves enterovirus infection, or a predictable presentation of a rare
(neuritis). pathogen such as rabies or lymphocytic choriomeningitis virus.
The clinical manifestations of encephalitis are variable and
reflect the degree of brain involvement, host factors, and the
inherent pathogenicity of the offending agent. Most patients
GENERAL: ETIOLOGIC AGENTS with encephalitis have headache and fever, followed by altered
AND EPIDEMIOLOGY mental status. Seizures, behavioral changes, impaired cogni-
tion, aphasia, hemiparesis, and other focal neurologic signs
Although the term encephalitis is often used in conjunc- may be seen. Arboviruses are often associated with global CNS
tion with a viral etiology, many other infections and nonin- dysfunction, whereas agents such as herpes simplex encepha-
fectious entities can cause encephalitis or encephalitis-like litis typically result in focal manifestations. Although there is
symptomatology. The incidence of encephalitis varies through- significant overlap in the clinical presentation of various agents
out the world and is contingent upon the population under and diagnosis can rarely be made on clinical grounds alone,
study, the geographic region, the availability of vaccines for the most typical and/or characteristic features of some of the
some causes of encephalitis, and differences in case definitions causes are highlighted in the following sections.

84

Scheld_Ch06.indd 84 2/21/14 5:31 PM


Chapter 6: Encephalitis 85

Herpes Simplex Virus Epstein-Barr Virus


Herpesviruses are enveloped DNA viruses that are among the Epstein-Barr virus (EBV), another herpesvirus, is most often
most common causes of infections in humans. At least eight associated with mononucleosis but can also cause several
herpesvirus types are known to infect humans. Herpes simplex distinct neurologic syndromes including aseptic meningi-
virus type 1 (HSV-1) is one the most common causes of spo- tis, Guillain-Barr syndrome, Bell palsy, transverse myelitis,
radic encephalitis in the world (see Chapters 9 and 10). The cerebellitis, and encephalitis (37,38) (see Chapter 12). Most
epidemiology and clinical features of neonatal herpes CNS neurologic complications due to EBV occur during primary
infections differ from children and adults and are not covered infection, typically in childhood. Importantly, many patients
in this chapter. The incidence of herpes simplex encephalitis with EBV-associated encephalitis do not have classic mononu-
(HSE) caused by HSV-1 is estimated to be 4 per 1,000,000 cleosis symptoms (39,40). In a case series of 216 pediatric en-
(11,18). HSE is responsible for 10% to 20% of adult en- cephalitis patients in Canada, 21 (9.7%) were identified with
cephalitis cases (3,19). HSE is less common in children than EBV-associated encephalitis (40). Of these, only one patient
in adults; in a large cohort of over 300 pediatric encephalitis had classic mononucleosis symptoms; most others had a non-
patients over a 12-year period, only 5% were due to HSE. In specific prodrome of fever (81%) and headache (66%) (40).
the pediatric age-group, HSE is often a result of a primary in- Seizures occurred in almost half (48%) (40). Some individuals
fection, whereas most HSE infections in adults are the result of with EBV-associated encephalitis experience micropsia, mac-
HSV reactivation. Importantly, the presence of herpes labialis ropsia, and/or size distortion. This pattern of unusual images
has no diagnostic specificity for encephalitis causality but does of body and objects is referred to as the Alice in Wonderland
serve as a marker of HSV infection. syndrome (41). There are sporadic reports in the literature
The characteristic clinical presentation for HSE includes that reactivation of chronic EBV infection in adults may cause
altered mental status (97%), fever (90%), and headache neurologic manifestations, including encephalitis (42).
(81%) (20). Other common neurologic findings include per-
sonality change (85%), aphasia (40%), ataxia (40%), hemi- Human Herpesviruses 6 and 7
paresis (38%), cranial nerve deficits (32%), and seizures
Human herpesvirus-6 (HHV-6), the primary cause of ro-
(31%) (4,21). Children are more likely to have extratempo-
seola infantum in young children, has been identified as the
ral involvement as manifested by clinical symptoms as well as
causal agent of 10% to 20% of febrile seizures and is also
neuroimaging (22).
occasionally associated with encephalitis (43,44) (see Chapter
Unlike HSV-1, HSV-2 is more likely to cause disseminated
13). Several studies of encephalitis in children have identified
encephalitis and does not generally localize to the temporal
HHV-6 as a causative agent, with the incidence of HHV-6
and inferior frontal regions of the brain (23). Most neuro-
encephalitis ranging from 1% to 11% of cases (4549). In
logic CNS HSV-2 infections present with lymphocytic men-
one study of nine children with HHV-6 CNS infections,
ingitis. Relapsing meningitis, encephalitis, and myelitis can
characteristic clinical features included fever, gastroenteritis,
also occur.
rash, seizures, and ataxia (50). Another case series reported
HSV-1 and HSV-2 can also cause brainstem encephalitis.
three pediatric patients with HHV-6associated rhomben-
A recent comprehensive literature review of HSV brainstem
cephalitis; clinical manifestations included encephalopathy,
encephalitis identified 24 cases: 79% due to HSV-1 and 29%
seizures, ataxia, and myoclonus (51). HHV-7, a recently de-
due to HSV-2 (24). The most prominent features were neuro-
scribed herpesvirus, is occasionally associated with roseola
ophthalmologic manifestations; these were seen in over 80%
and is typically acquired in the first few years of life. Recent
of patients and included nystagmus, impaired ocular move-
studies from the United Kingdom suggest HHV-7 may be an
ments, anisocoria, ptosis, oscillopsia, or spasmodic move-
important cause of febrile seizures and encephalitis in young
ments (24). Other cranial deficits, altered mentation, ataxia,
children (52).
and corticospinal tract findings (e.g., hyperreflexia) were also
described. Although not common, quadriplegia was also pres- Enteroviruses and Parechoviruses
ent in some (19%) of the patients (24).
Enteroviruses (EVs) are small, nonenveloped, single-stranded
RNA picornaviruses. Similar to herpesvirus infections, EV
Varicella-Zoster Virus
infections are very common, and neurologic complications,
Both primary infections with varicella-zoster virus (VZV) and including encephalitis, represent a rare complication of EV
endogenous reactivation (herpes zoster) can lead to encepha- infection. Because EV infections occur frequently in children,
litis (25) (see Chapter 10). The most characteristic manifesta- they are a leading cause of encephalitis in children and are
tion of VZV encephalitis in children is acute cerebellar ataxia responsible for 10% to 15% of encephalitis cases for which an
(e.g., nystagmus, dysarthria, and ataxia), which usually occurs etiology is identified (53). In general, EVs cause a milder clini-
1 week after rash onset (26). VZV encephalitis, once a leading cal illness than many other etiologies of encephalitis. In the
cause of encephalitis in children, is much less common due to California Encephalitis Project, EV encephalitis patients had
the widespread use of VZV vaccine (27). less severe manifestations, including lower frequencies of coma
However, VZV encephalitis is relatively common in adults, and shorter hospitalization stays than those due to other agents
and its incidence rivals that of HSE (2831). The clinical pre- (54). CNS infections with EV-71 are an important exception
sentation in adults is different than in children and includes to the decreased severity of EVs (55,56). In addition to caus-
diffuse brain dysfunction, seizures, cranial nerve palsies, and ing acute flaccid paralysis (aka polio-like syndrome), EV-71
other focal neurologic signs (4,32). The presence of a diffuse has also been associated with a distinctive form of encephalitis
varicella rash or a vesicular rash in a dermatome distribution initially described in Taiwan and Malaysia (55,57). Most cases
can be an important clue to diagnosing VZV encephalitis. were in young children (younger than 5 years of age), with a
Notably, however, as many as 44% of patients lack cutaneous characteristic hand, foot, and mouth rash along with ataxia,
findings, a condition termed herpes sine zoster or preeruptive nystagmus, myoclonus, and oculomotor palsies (55). The
varicella (3236). Although the pathophysiology of VZV- predominant neurologic presentations included myoclonus
associated encephalitis remains unclear, some cases appear to (68%), vomiting (53%), and ataxia (35%) (57). Many of the
be due to a medium and large vessel vasculopathy (25). fatalities associated with EV-71 are due to pulmonary edema

Scheld_Ch06.indd 85 2/21/14 5:31 PM


86 Part II: Viral Infections and Related Disorders

and hemorrhage, which are thought to be a consequence of Similar to many other arboviruses, most JEV infections are
pronounced autonomic instability due to lesions in the me- asymptomatic, with less than 1% of infections leading to clini-
dulla and spinal cord (58,59). Sporadic outbreaks involving cal disease. When symptoms occur, encephalitis is the most
substantial numbers of EV-71 encephalitis cases have been ob- common presentation. After a characteristic febrile prodrome
served among young children in Europe and Asia over the past including headache and vomiting, mental status changes,
several decades (6062). seizures, focal neurologic deficits, and movement disorders
The reclassification of former EVs, echovirus 21 and echo- develop. Similar to patients with WNV, those affected by JEV
virus 22, resulted in the human parechovirus (HPeV) genus. can also develop acute flaccid paralysis (77).
Echoviruses 21 and 22 are currently classified as HPeV-1 and In Europe, tick-borne encephalitis virus (TBEV), another
HPeV-2, respectively. At least 12 HPeV serotypes have been flavivirus, is the most common cause of arboviral encephali-
described to date; nearly all have been associated with enceph- tis (78). It is also found in China and Japan. Analogous to
alitis, typically in children younger than 2 years of age (63,64). WNV, neuroinvasive disease is more common in older popu-
Clusters of HPeV-3 CNS infections have been reported (65). lations (79). Growing numbers of cases have been recognized
Young children and infants with HPeV encephalitis develop in recent years as a result of improvements in diagnostics and
fever, seizures, irritability, feeding problems, and rash (66). case reporting as well as increased recreational activities in
The relative frequency of HPeV encephalitis is unknown, tick-infested areas (80). In Europe and Russia, there was an
particularly because HPeV testing has only recently become average of 8,755 reported cases per year from 1990 to 2007
available. compared to an average of 2,755 cases per year from 1976 to
1989 (81). TBEV is characterized by three different subtypes:
Arboviruses European (TBEV-Eu), Siberian (TBEV-Sib), and Far Eastern
(TBEV-Fe). The TBEV-Eu subtype circulates predominantly
Arboviruses, viruses transmitted by an arthropod vector, are
in Western, Central, Northern, and Eastern Europe; the
well-recognized causes of encephalitis. The vast majority of
TBEV-Sib circulates predominantly in Asian parts of Russia;
neurologic illnesses seen in humans are caused by three arbo-
and TBEV-Fe circulates predominantly in China, Japan, and
virus families: Togaviridae, Flaviviridae, and Bunyaviridae (see
Eastern Russia. The clinical spectrum of disease ranges from
Chapter 15).
mild meningitis to severe meningoencephalitis with or with-
West Nile virus (WNV), a flavivirus, was first detected in
out paralysis (82). In individuals affected with the European
the Western Hemisphere in 1999 in New York City and rapidly
virus subtype, the illness is often biphasic, with the first stage
spread across North America from the Atlantic to the Pacific
characterized by fever, fatigue, general malaise, headache,
coasts and into Mexico and Canada. It is the now the most
and body pain. During the second phase of the illness, clini-
common cause of arboviral encephalitis in the United States
cal manifestations range from mild meningitis to severe en-
(67). Most individuals infected with WNV will experience
cephalitis, with or without myelitis and paralysis. Seizures
subclinical infection (70% to 80%) or febrile illness (20% to
are uncommon. The disease associated with TBEV-Fe sub-
30%). Less than 1% of infected individuals develop West Nile
type is the most severe, with a case fatality of 20% to 40%
neuroinvasive disease (WNND), which includes meningitis,
and higher rates of neurologic sequelae compared with other
encephalitis, and/or acute flaccid paralysis.
subtypes (82).
WNND is more common in older individuals, with an inci-
Although JEV is recognized to cause more cases of enceph-
dence of 1.35 per 100,000 in persons 70 years of age or older
alitis than any other mosquito-borne virus worldwide, dengue
compared with 0.05 per 100,000 in persons younger than
viruses are the most prevalent arboviruses that infect humans
10 years of age (67). Other risk factors for WNND include
and result in an estimated 390 million infections every year
male gender, hypertension, diabetes, renal disease, and immu-
(83). Infections with dengue typically result in dengue fever,
nosuppression (68,69). Characteristic presentations of WNND
dengue hemorrhagic fever, and dengue shock syndrome.
include altered mental status or lethargy with or without move-
Unlike many of the aforementioned arboviruses, neurologic
ment disorders (tremors, Parkinsonism, or myoclonus). Acute
manifestations of dengue have traditionally been considered
flaccid paralysis is also a feature of WNV infection and can be
to be the result of an encephalopathy rather than encephalitis.
seen along with encephalitis or may occur in isolation (70,71).
However, detection of dengue viral RNA in brain tissue, virus
Although the number of WNND cases has far surpassed the
isolation in CSF, and the presence of dengue-specific CSF an-
number of cases due to other arboviruses in recent years, other
tibody suggesting intrathecal synthesis have been described in
arboviruses in the United States cause seasonal outbreaks and
recent studies and strongly suggest the neuroinvasive potential
sporadic cases of neurologic disease. These include La Crosse
of dengue (8486).
virus (LACV), eastern equine encephalitis virus (EEEV),
Powassan virus (POWV), and St. Louis encephalitis virus
(SLEV). In 2012, over 2,500 WNND cases were recognized
Rabies
in the United States compared with 78 LAC cases (71 neuro- Rabies virus is one of the oldest known infectious diseases and
invasive), 15 EEE cases (all neuroinvasive), 7 POW cases (all is considered to be the most deadly of all infectious diseases
neuroinvasive), and 2 SLE cases (1 neuroinvasive) (72). (see Chapter 17). The number of rabies encephalitis cases in
LACV, primarily found in the upper Midwestern, mid-Atlantic, the United States has declined dramatically from an average
and Southeastern regions of the United States, is the second most of 100 or more cases per year before 1940 to only 2 to 3 cases
common cause of arbovirus-associated CNS infections in the per year (87). Although rabies is rare in the United States, an
United States. Unlike WNV, most neuroinvasive LACV cases estimated 50,000 rabies cases occur annually worldwide; most
occur in the pediatric population rather than in adults; in 2012, are acquired via rabid dog contact (88). In Asia, Africa, and
86% of cases were younger than 20 years of age (73). Latin America, animal rabies control programs and postexpo-
Outside the United States, other arboviruses predominate. sure prophylaxis are limited. A recent report of 49 rabies cases
Japanese encephalitis virus (JEV) is the most common cause of in the United States, from 1995 to 2011, identified 10 im-
mosquito-borne encephalitis worldwide. An estimated 50,000 ported and 39 cases acquired in the United States; of the cases
cases of JEV clinical disease occur annually, primarily in chil- acquired in the United States, one was associated with a rac-
dren younger than 10 years of age in Asia, South Asia (east of coon strain of rabies, and the rest were due to bat exposures.
Pakistan), and Southeast Asia (7476). The incubation period is generally between 20 and 60 days but

Scheld_Ch06.indd 86 2/21/14 5:31 PM


Chapter 6: Encephalitis 87

can range from a few days to several years (89). Paresthesia and often fatal form of encephalitis that typically occurs 7
at the site of the bite is unique to rabies and can be an im- to 12 years after the initial infection and usually affects chil-
portant clue to the diagnosis. Approximately 80% of human dren between 10 and 14 years of age (98,99). A history of
rabies cases develop the encephalitic (furious) form charac- prior measles vaccination does not preclude the diagnosis of
terized by unusual behavior, extreme agitation, hydrophobia, SSPE because an unrecognized measles infection may occur
delirium, and seizures. The remainder of cases develops the at an early age prior to immunization. This is supported
paralytic (dumb) form which is characterized by ascending by molecular studies that have identified wild type measles
paralysis followed by confusion and coma. Patients generally virus (rather than vaccine-type virus) from brain specimens
have a predominance of one form, but many affected individu- of SSPE cases (100). Early clinical features of SSPE include
als have components of both forms. personality and behavior changes, lethargy, decline in school
performance, and hyperactivity. More pronounced neuro-
Lymphocytic Choriomeningitis Virus logic manifestations such as aphasia, difficulty walking, and
involuntary movements (e.g., tremors, myoclonic jerks, and
Lymphocytic choriomeningitis (LCM) virus is an Old World choreoathetosis) later ensue. In the final stages, neurologic
arenavirus that can be acquired from infected house mice, deterioration resulting in a vegetative state occurs in most
hamsters, and guinea pigs. Humans become infected with affected patients (99).
LCM virus when aerosolized saliva, respiratory secretions, or
urine from rodents or virus-contaminated dust are inhaled or
possibly ingested. Infections occur more frequently in the win-
ter months when rodents migrate indoors (90). The incidence Bacteria
of LCM is unknown but appears to have decreased substan-
tially in the last several decades due to improvements in hous- While many infectious encephalitis cases have a viral etiol-
ing, which have resulted in less contact between house mice ogy, bacterial causes are important to consider in the diag-
and humans. However, it is likely that cases continue to occur nosis either as a mimicker of encephalitis or as an actual
but are not recognized (91). LCM often results in a bipha- cause of encephalitis. For example, Neisseria meningitidis and
sic illness with an initial phase of fever, anorexia, headache, Streptococcus pneumoniae do not cause encephalitis per se
muscle aches, nausea, and vomiting. Several days later, CNS but can cause clinical manifestations that are indistinguishable
symptoms can occur with either meningitis or encephalitis. from encephalitis. Mental confusion, drowsiness, convulsions,
Extra-CNS manifestations may also be present, including or- and coma are not uncommon manifestations of N. meningiti-
chitis, parotitis, arthritis, or alopecia (92,93). dis and S. pneumoniae bacterial meningitides (see Chapter 24).

Hendra Virus Mycobacterium tuberculosis


Hendra virus is a paramyxovirus first recognized in Hendra, Although meningitis is the most common form of neurotu-
Australia where it was associated with an outbreak of respira- berculosis, Mycobacterium tuberculosis was the third leading
tory and neurologic disease in horses and humans in 1994. cause of encephalitis in a French study where it was identi-
The natural reservoir of the virus is thought to be flying foxes fied in 15% of cases (101). In England, from 2005 to 2006,
(bats of the genus Pteropus). The virus is transmitted from M. tuberculosis was the causative agent in 12% of encephalitis
bats to horses and then transmitted to humans as a result of cases with an identified cause (29). Analogously, in a multi-
direct contact with infected horses. More than 60 equine and center study of encephalitis in Taiwan, M. tuberculosis was
4 human fatalities have been reported (94). The high fatality the third most common cause of encephalitis in both pediat-
rate of this infection in horses and people, as well as the large ric and adult patients (31). Of 20 patients with M. tubercu-
reservoir species, underscores the potential of this virus, and losis encephalitis referred to California Encephalitis Project
other similar viruses, to emerge and cause outbreaks of severe (CEP) between 1998 and 2005, many had features in common
illness. Human illness due to Hendra virus is characterized by with patients with viral encephalitis, including fever (75%),
influenza-like symptoms often followed by acute encephalitis. altered consciousness (65%), personality change (45%), and
A relapsing neurologic syndrome has also been described in a hallucinations (16%) (102). Because the base of the brain is
few individuals (94). often affected by M. tuberculosis, signs referable to cranial
nerves are often seen along with fever, headache, irritability,
and drowsiness. Diffuse meningeal irritation may also result
Nipah Virus in impairment of CSF resorption with accompanying hydro-
Nipah virus, another emerging paramyxovirus, was first rec- cephalus (see Chapter 29).
ognized in 1999 and associated with an encephalitis outbreak
among pig farmers in Malaysia. This virus has also caused Listeria monocytogenes
outbreaks in Singapore, Australia, Bangladesh, and India
(9597). Human infections can range from asymptomatic in- Unlike S. pneumoniae and N. meningitidis that rarely cause
fection to fatal encephalitis. When neurologic illness occurs, parenchymal brain infections, Listeria monocytogenes has tro-
individuals often experience influenza-like illness followed by pism for the brain parenchyma itself as well as the meninges
dizziness, excess drowsiness, and altered consciousness. (103). The most common CNS manifestation of listeriosis is
of an isolated meningitis, but approximately 10% of patients
present with brainstem encephalitis, encephalitis, diffuse cere-
Measles Virus britis, or abscess in cerebral cortex or spinal cord (104). In the
Measles virus infection causes acute encephalitis in approxi- French study cited earlier, L. monocytogenes was the fourth
mately 1 in 1,000 cases, often resulting in permanent brain most common etiology identified (101). Reported risk factors
injury (see Chapter 8). Although indigenous transmission of for CNS Listeria include male gender, immunosuppression,
measles was eliminated in the United States in 2000, it is chronic illness, and advanced age (35,105108). Conversely,
still a common infection in much of the world. In addition an in-depth review of Listeria rhombencephalitis found that
to acute encephalitis, measles is associated with subacute it was reported primarily in healthy, nonimmunosuppressed
sclerosing panencephalitis (SSPE), an indolent, progressive, middle-aged adults and affected both genders equally (109).

Scheld_Ch06.indd 87 2/21/14 5:31 PM


88 Part II: Viral Infections and Related Disorders

Clinically, a biphasic disease course may be seen; a nonspecific B. procyonis occurs in raccoons in North America, Europe,
prodrome lasting several days is typical of the first phase. The and parts of Asia (119). More than 90% of juvenile raccoons
prodrome itself is similar to many viral illnesses with fever, are infected in some areas of the United States (118). Humans
headache, nausea, and vomiting, but the duration of prodrome become infected by ingesting raccoon roundworm eggs in rac-
is longer in CNS Listeria illnesses compared with viruses. The coon feces, by soil or water contaminated with raccoon feces,
second phase is characterized by progressive asymmetric cra- or via contaminated hands. Small children are particularly vul-
nial nerve palsies, cerebellar signs (e.g., ataxia or dysmetria), nerable to infection because of their propensity to place dirt
hemiparesis, and altered level of consciousness (109). and other objects in their mouths.
Other important neurotropic helminthes associated with
Treponema pallidum eosinophilic encephalitis or meningitis includes G. spinigerum
and Angiostrongylus species. Gnathostomiasis, most com-
Treponema pallidum, the cause of syphilis, is yet another bac-
monly caused by the nematode G. spinigerum, is a cause of
terial infection that can potentially be confused with CNS viral
eosinophilic myeloencephalitis. G. spinigerum is endemic in
etiologies. Syphilis, also known as the great mimicker, can be
Southeast Asia and is increasingly being recognized in Central
difficult to recognize; this is particularly true for neurologic pre-
and South America (120,121). Most cases are associated with
sentations. Neurologic manifestations can occur during any stage
the ingestion of raw or undercooked fish, frogs, snakes, chick-
of the infection and include meningeal syphilis, meningovascular
ens, or ducks. The median time from ingestion of infected food
syphilis, paretic neurosyphilis, and tabes dorsalis (see Chapter 38).
to onset of symptoms may be several weeks to several months
(122). Common early symptoms may include sporadic epi-
Borrelia Species sodes of cutaneous larva migrans (creeping eruption) with
Borrelia burgdorferi, the causative agent of Lyme disease, is localized pain and pruritus. When CNS involvement occurs,
primarily endemic to the eastern United States, although reser- it may result in the sudden onset of radicular pain or head-
voirs are also present in the Pacific Northwest and Midwestern ache. Paralysis of the extremities and loss of bladder control
states. Notably, the geographic range is expanding. Lyme dis- may also occur (123). Cranial nerve abnormalities are also de-
ease can affect both the peripheral and central nervous system; scribed. Intermittent symptoms can occur for 10 to 15 years
CNS involvement is typically characterized by meningitis, al- after exposure because the larvae are long lived (124).
though encephalitis can rarely occur (see Chapter 39). A. cantonensis, the rat lungworm, is the principal cause
of human eosinophilic meningitis worldwide, and although
many cases are self-limiting, severe forms of the disease
Rickettsia occur (125,126). Angiostrongylus spp. have been reported
in Louisiana and Hawaii as well as the South Pacific, Asia,
Rickettsial infections can also cause encephalitis. Of the rick- Australia, and the Caribbean (127,128). Humans become in-
ettsial diseases, Rocky Mountain spotted fever (RMSF) and fected by ingestion of the third stage larvae in the molluscan
epidemic typhus are most commonly associated with neuro- intermediate host (e.g., snails, crabs, freshwater prawns) or
logic manifestations (110). In patients with RMSF, an intense contaminated vegetables. Following ingestion, the larvae pen-
headache along with restlessness, irritability, confusion, and etrate the intestinal wall and reach the CNS via the blood-
delirium often occur. General or focal neurologic impairment stream. Clinical illness often consists of severe headache,
including vertigo, seizures, hemiparesis, and ataxia may also photophobia, meningeal signs, hyperesthesia, and paresthesia.
be present (111). In a study of 92 children hospitalized with Coma, paralysis of extremities, and seizures are seen in the se-
RMSF in the southeastern and south central United States from vere forms of the disease (125,126). Conjunctivitis, periorbital
1990 through 2002, 33% had altered mental status, 18% pho- swelling, retinal hemorrhage, retinal detachment, or blindness
tophobia, 17% seizures, and 10% coma (112). Ophthalmic may occur if the eye is infected (129,130) (see Chapter 46).
features including photophobia, conjunctivitis, petechiae of Free-living ameba are ubiquitous in nature, and a few have
the bulbar conjunctiva, exudates and retinal venous engorge- been associated with human disease. Those causing encephali-
ment, papilledema, and ocular palsies are frequently described tis are generally divided into two clinical entities: (a) primary
(113,114). Acute temporary hearing impairment may also amebic meningoencephalitis due to Naegleria fowleri (also
occur (115). Similar neurologic complications have also been known as the brain-eating ameba), and (b) granulomatous
described for other rickettsial infections, although generally amebic encephalitis.
are not as severe (116) (see Chapter 27). Primary amebic meningoencephalitis (PAM) is a fulminant
disease occurring in children and young adults. The disease has
been reported in Australia, Europe, Asia, Africa, and North
Parasites and Free-Living Ameba America. In the United States, most of the cases have developed
in the southern tier of the country, where warm water condi-
A number of parasites can cause encephalitis via direct in- tions are more likely to be encountered. Typically, humans be-
vasion of the brain. Helminthes including various ascaris, come infected with N. fowleri while swimming or washing in
hookworms, Gnathostoma spinigerum, Angiostrongylus canto- warm, fresh water containing the ameba. Recent cases associ-
nensis, Spirometra spp., Alaria spp., and others can cause larva ated with the use of neti pots where N. fowlericontaminated
migrans, which refers to the prolonged migration and persis- tap water were implicated as the source of infection have also
tence of helminth larvae in the tissues of humans (117,118). been described (131). The onset of PAM is usually within 2 to
Larva migrans can result in visceral (VLM), ocular (OLM), 3 days after exposure, and symptoms include severe headache,
neural (NLM), and cutaneous larva migrans (CLM) based on fever, stiff neck, nausea, vomiting, diplopia, seizures, behav-
the organ systems involved (118). VLM and NLM are usually ioral changes, and coma (132135). Distortion of taste or smell
diseases of childhood, affecting children ages 1 to 8 years old. may also be a clinical feature (136,137). The case fatality rate
B. procyonis, a common ascarid roundworm in raccoons, is very high; of 111 reported cases in the United States between
causes an eosinophilic encephalitis in humans and other 1962 and 2008, only one individual survived (131).
animals. It is most commonly identified in children and, Two different closely related ameba cause granuloma-
although it is rare, often results in a severe and fatal illness. tous encephalitis: Acanthamoeba spp. and B. mandrillaris.

Scheld_Ch06.indd 88 2/21/14 5:31 PM


Chapter 6: Encephalitis 89

Acanthamoeba granulomatous encephalitis is an opportunistic, poorly understood. Influenza-associated encephalitis (IAE)


chronic disease that may have a prodromal period of weeks to and encephalopathy is typically characterized by a rapidly
months. Predisposing factors to acanthamebiasis include ste- progressive neurologic illness. Cases have been described
roid treatments, autoimmune conditions, organ transplants, sporadically and follow the seasonal influenza pattern, with
chemotherapy, radiation therapy, alcoholism, and pregnancy illnesses typically occurring during winter months in temper-
(134,138). A small number of Acanthamoeba granulomatous ate climates. IAE is more common in the pediatric population
encephalitis cases have been described in immunocompetent than in adults. Evidence of neuroinvasion by influenza is rarely
children. Clinical features of CNS Acanthamoeba infections seen; these cases are better characterized as an encephalopathy
are variable but typically have a subacute to chronic presenta- rather than encephalitis. Many cases of IAE, especially acute
tion with fever, headache, seizures, personality change, leth- necrotizing encephalopathy (ANE), have been reported from
argy, or confusion. Cranial nerve palsies, meningeal signs, or Japan, but cases of encephalitis and encephalopathy have been
hemiparesis may be seen on physical exam (138). Children reported throughout the world, including the United States
infected with Acanthamoeba have exhibited headache, stiff (156,157). Several case reports and case series describe neu-
neck, vomiting, abnormal behavior, fever, ataxia, and tonic- rologic illness associated with the pandemic H1N1 influenza
clonic seizures (139141). virus, including encephalitis/encephalopathy (158160).
When first described, B. mandrillaris was considered Similarly, the neurologic illness associated with Bartonella is
to be very rare, but recent reports suggest it may be more often an encephalopathy rather than encephalitis. Cat-scratch
common than previously recognized (142145). Symptoms disease (CSD), typically caused by Bartonella henselae, is usu-
of Balamuthia granulomatous encephalitis include fever, ally a self-limited infection associated with fever, regional
headache, vomiting, ataxia, hemiparesis, tonic-clonic sei- lymphadenopathy, and malaise. As the name implies, many
zures, cranial nerve palsies (third and sixth cranial nerves), affected individuals have contact with a cat, often a kitten.
and diplopia (146). Otitis media has preceded the onset of Atypical presentations can occur especially in children and
Balamuthia granulomatous encephalitis in several pediat- young adults and immunocompromised individuals. In a case
ric cases (146,147). Hydrocephalus develops in many cases series of 130 CSD cases in Japan, 19 (15%) had encepha-
(147,148). Interestingly, two independent case reports involv- lopathy (161). Although lymphadenopathy is a hallmark of
ing Balamuthia encephalitis patients describe associated CNS CSD, it is not always present in encephalopathy cases (161).
aneurysms (149,150) (see Chapter 45). Neuroretinitis can be an associated feature as well (162).

Fungi ACUTE DISSEMINATED


Although fungal CNS infections do not cause encephalitis per ENCEPHALOMYELITIS
se and often manifest as meningitis and abscesses, some pa-
tients present with encephalitis-like symptoms. These illnesses The incidence of acute disseminated encephalomyelitis
are more common in immunocompromised individuals, but (ADEM) is estimated to be 0.4 to 0.8 per 100,000 and it ac-
fungal neurologic infections can be seen in immunocompetent counts for 10% to 15% of encephalitis cases in the United
individuals. Important fungal causes of CNS infections in the States (163165). It is more common in children than adults,
United States include Cryptococcus neoformans, Coccidioides and many cases of ADEM have an identifiable trigger such
immitis, Histoplasma capsulatum, and Blastomyces dermatiti- as a recent illness or vaccination. Neurologic symptoms typi-
dis. Cryptococcus gattii is an emerging fungal infection in the cally develop 2 to 4 weeks after the trigger with rapid pro-
United States, particularly in the Pacific Northwest, and has gression of symptoms (165). Prior to the widespread use of
a greater tendency to affect normal hosts than Cryptococcus vaccine-preventable diseases in the United States, measles and
neoformans (151) (see Chapter 40). mumps were common triggers of ADEM. In regions of the
world where vaccines are widely used to prevent measles and
mumps, such as the United States and Canada, upper respira-
Other Agents tory infections are now the most commonly identified triggers
of ADEM.
Hundreds of other infectious agents have been associated with ADEM affects multiple regions of the brain and spinal cord
encephalitis, but the frequency and significance of many of and is characterized by the rapid onset of encephalopathy
them are unknown. This is especially true when a nonneu- along with multifocal neurologic deficits. Up to three quar-
rotropic agent is found in a patient with encephalitis, par- ters of patients with ADEM will have altered mental status,
ticularly when the agent is identified outside the CNS. For whereas seizures occur in 10% to 35% of patients (165).
example, Mycoplasma pneumoniae is one of the most com- Motor deficits (e.g., acute hemiparesis), ataxia, decreased ver-
monly diagnosed infections among children with encephalitis bal output or mutism, cranial neuropathies, and urinary disor-
(see Chapter 25). However, the significance of this association ders are common (166). Recovery can be complete, although
is unclear, particularly as most of the diagnoses are based on residual deficits can occur in up to 30% of patients.
a positive immunoglobulin M (IgM) antibody to M. pneu-
moniae. M. pneumoniae is a ubiquitous pathogen, and there is
a high background incidence of acute infection. Furthermore, NONINFECTIOUS ETIOLOGIES
there are many limitations of Mycoplasma serologic test-
ing (152). Other similar examples of nonneurotropic agents There is growing recognition that immune-mediated condi-
implicated as causative agents include parvovirus B19, rotavi- tions result in a substantial proportion of cases of encephalitis.
rus, and human metapneumovirus (153155). AntiN-methyl-d-aspartate receptor (NMDAR) encephalitis,
Although the association of influenza viruses and en- a relatively newly recognized neuronal antibody-associated
cephalitis is better documented and more accepted than encephalitis, deserves special mention because of the seemingly
parvovirus B19, rotavirus, or human metapneumovirus, the high frequency of the syndrome. In the California Encephalitis
mechanism by which influenza leads to neurologic illness is Project, the frequency of anti-NMDAR encephalitis surpasses

Scheld_Ch06.indd 89 2/21/14 5:31 PM


90 Part II: Viral Infections and Related Disorders

that of any single viral entity in the pediatric population and patients with bacterial meningitis and rickettsial infections where
also contributes to cases in adulthood (167). associated vasculitis and elaboration of toxins can lead to CNS
Clinically, anti-NMDAR encephalitis is characterized by ab- dysfunction. Intense inflammatory responses to fungi, free-living
normal behavior, seizures, and movement disorders followed by ameba, and parasites can also lead to CNS dysfunction.
decreased level of consciousness and autonomic instability and Organisms enter the CNS by different routes. Most enter via
may be associated with ovarian teratoma (168). Importantly, the the bloodstream, as is the case for EVs, HPeVs, and arboviruses as
development of prodromal symptoms of headache, low-grade well as several bacteria, rickettsia, and fungal agents (171). Once
fever, or a nonspecific viral-like illness prior to the onset of neuro- the agent reaches the CNS, the bloodbrain barrier is penetrated
logic symptoms in many patients may initially suggest the diagno- via the choroid plexus or through vascular endothelium (172).
sis of an infectious, rather than autoimmune, encephalitis (168). The proposed mechanism for entry into brain of the ame-
Another important cause of immune-mediated encephalitis bae varies by organism. For example, Balamuthia is thought to
is antivoltage-gated potassium channel (VGKC) encephalitis. enter the CNS via a hematogenous route, with ameba entering
Patients are typically older than 50 years of age, present with the bloodstream either from the lungs or from cutaneous lesions.
symptoms of limbic encephalitis (memory dysfunction, be- Naegleria ameba, on the other hand, enter the nasal passages
havioral changes, and seizures) and hyponatremia, and rarely and directly extend into the CNS by penetrating the olfactory
have an underlying neoplasm. Although antibodies were ini- mucosa, entering the submucosal nervous plexus, migrating
tially thought to recognize the VGKC receptor, subsequent along the olfactory nerves, and traversing the cribriform plate. A
studies have demonstrated that the target of autoimmunity distinct mechanism of entry via axon transport resulting in intra-
is usually a different antigen (i.e., LGI1 or CASPR2) that is neuronal route is used by some viruses such as rabies and HSV-1.
tightly associated with the VGKC complex (168,169). Several Within the CNS, the pathogen often targets specific cells
other antibodies are also associated with limbic encephalitis, and, depending on the brain region affected, variable clinical
including those that recognize glutamic acid decarboxylase manifestations ensue (173). Agents with specific predilection to
(GAD), AMPA receptor, and the -aminobutyric acid (GABAb) areas such as the brainstem (e.g., EV-71 and Listeria) can cause
receptor. Intracellular onconeural antibodies associated with rapid decompensation with coma or respiratory failure. Herpes
paraneoplastic conditions (e.g., anti-Hu, Yo, Ma2, CV2, simplex encephalitis characteristically affect the temporal lobes
amphiphysin, CRMP5, etc.) also need to be considered (170). and cause hemorrhage and necrotizing lesions (174) (Fig. 6.2).
WNND has a predilection for the gray matter of the brain-
stem and spinal cord, but cerebellum, temporal lobe, basal gan-
PATHOLOGY glia, and thalamus may also be affected (175). Indeed, many of
the neuroinvasive flaviviruses such as JEV, TBEV, and WNV
Given the many different etiologies of encephalitis, the pathol- have a predilection for specific regions of the brain, including
ogy is highly variable and dependent not only on the under- those regions important for motor control (thalamus, basal
lying etiology but also the relative severity of the infection. ganglia, brainstem, and anterior horn cells of spinal cord) (176).
The characteristic histology of patients with viral encephalitis In other instances, the infectious agents do not necessarily in-
includes perivascular mononuclear cell inflammation, phago- fect neurons. Nonneuronal cells, such as oligodendroglia, may
cytosis of neurons, and microglial nodules. Distinctive char- be infected, with resultant demyelination (177). Alternatively, an
acteristic histopathologic features are seen with some viral infection may cause immune changes that result in damage. EBV-
infections; intranuclear inclusion bodies are sometimes seen in associated encephalitis, for example, may be a result of an im-
herpes simplex and varicella zoster, whereas Negri bodies (eo- munologic phenomena rather than acute neuroinvasion. There is
sinophilic cytoplasmic inclusions) are found within Purkinje typically a 1- to 3-week delay in the onset of neurologic symptoms
cells and are pathognomonic for rabies (Fig. 6.1). after acute EBV infection (40). Further, the virus itself is often not
EVs, parechoviruses, herpesviruses, arboviruses, and rabies found in the CSF (40). In one cohort of five EBV encephalitis pa-
have well-established neurotropic potential where the virus di- tients with CNS demyelination, four had prodromal symptoms
rectly invades the CNS and primarily affects the gray matter of the for 2 weeks or more and did not have EBV detected by polymerase
brain. Other viruses, such as measles and rubella viruses, primarily chain reaction (PCR) in CSF (40). Conversely, the development of
affect the white matter of the brain by triggering an autoimmune neurologic symptoms either in the absence of, or within a few days
reaction and result in a postinfectious encephalitis (e.g., ADEM). of prodrome onset in the presence of, EBV in the CSF is suggestive
Symptoms indistinguishable from viral encephalitis can be seen in of direct invasion. In the pediatric series cited previously, one such

FIGURE 6.1 Rabies encephalitis. Purkinje cell in cerebellum


with eosinophilic, intra-cytoplasmic inclusion (Negri body).
(Courtesy of Dr. Andrew Bollen, University of California, San
Francisco.)

Scheld_Ch06.indd 90 2/21/14 5:31 PM


Chapter 6: Encephalitis 91

FIGURE 6.2 Herpes simplex encephalitis. Herpes simplex type


1 encephalitis with hemorrhage and necrosis of the right tempo-
ral lobe. (Courtesy of Dr. Andrew Bollen, University of California,
San Francisco.)

patient had EBV detected by PCR in brain tissue, suggesting that necrotic lesions. Multinucleated giant cells, focal necrosis,
direct invasion of the brain may occur in some cases. and hemorrhage are seen in brain histopathology. In some
Viruses such as influenza are well known to be associated instances, large sheets of ameba can be found in the perivascu-
with CNS manifestations, but the mechanisms by which they lar areas of brain tissue (Fig. 6.3).
cause neurologic signs and symptoms are not well under- B. procyonis and other nematodes that invade the CNS
stood. The lack of viral detection in most IAE cases in the cause damage by CNS larval migration, with histology demon-
CNS strongly points to a different pathogenesis; a number of strating inflammation and necrosis with track-like spaces.
potential mechanisms have been invoked, including excessive Pathologic findings of fatal cases demonstrate necrosis and
production of proinflammatory cytokines, vascular endothe- inflammation with eosinophils, macrophages, lymphocytes,
lial dysfunction, and mitochondrial dysregulation (178). and plasma cells concentrated in periventricular white matter
As discussed earlier, Listeria may be associated either with and leptomeninges in brain tissue. A characteristic of NLM/
a pure meningitis or encephalitis. Pathologically, a suppurative Baylisascaris infection is a large number of eosinophils and eo-
reaction is seen in the meningitic form, whereas a granuloma- sinophil granules surrounding the nematode migration track
tous response is seen in the meningoencephalitis form (104). and blood vessels (183).
Rickettsial agents invade and multiply in vascular endothe-
lial cells, leading to vasculitis both within and outside the CNS
(179,180). Vasculitis in the small vessels in the brain leads to DIAGNOSTIC APPROACH OF
meningeal irritation with perivascular mononuclear infiltrates. PATIENTS WITH ENCEPHALITIS
Characteristic pathologic lesions within the CNS include mul-
tifocal glial nodules and arteriolar microinfarctions (181). Identification of a specific etiology, even if there is no available
In partially treated bacterial meningitis and in tuberculous treatment, is important for counseling of patients and fami-
and fungal meningitis, a chronic basilar meningeal inflamma- lies, potential postexposure prophylaxis of contacts, and other
tion can cause a subarachnoid exudate, leading to obstruction public health interventions. Additionally, the identification of
of CSF reabsorption with resultant communicating hydro- a specific agent may lead to withdrawal of unnecessary an-
cephalus and cranial nerve palsies. CNS vasculitis can also timicrobial agents and reduce further testing. Knowledge of
lead to infarcts and focal neurologic deficits. limitations of testing, appropriate test selection, and timing of
N. fowleri, a free-living ameba, causes destruction of gray sample collection is crucial to optimal diagnosis.
matter and devastation of the olfactory bulbs with purulent A thorough assessment of exposures highlighting ill contacts,
meningitis and pronounced brain edema (182). In Balamuthia occupational exposures, vector and animal exposures, outdoor
and Acanthamoeba CNS infections, a granulomatous reaction activities, and ingestions should be ascertained. Both recent (e.g.,
occurs with affected areas including the cerebrum, cerebel- for arbovirus) and remote (e.g., rabies, fungal) travel history are
lum, and brainstem, where the amebae produce hemorrhagic important. Any recent or current respiratory, gastrointestinal,

FIGURE 6.3 Histopathology of B. mandril-


laris infections: (A) Brain tissue stained with
hematoxylin and eosin (100) showing B. man-
drillaris. Note that these B. mandrillaris organisms
may be mistaken for lymphocytes or macrophages.
(B) Higher magnification (1000) demonstrat-
ing trophozoites of B. mandrillaris in brain tis-
sue, some with single nucleolus (dotted arrow) or
double nucleoli (solid arrow). (Courtesy of Dr.
A B Govinda Visvesavara, Centers for Disease Control
and Prevention.)

Scheld_Ch06.indd 91 2/21/14 5:31 PM


92 Part II: Viral Infections and Related Disorders

TA B L E 6 . 1
DIAGNOSTIC ALGORITHM FOR INITIAL EVALUATION OF ENCEPHALITIS IN ADULTS

Routine Studies

CSF

Collect at least 20 mL fluid, if possible; freeze at least 510 mL fluid, if possible


Opening pressure, WBC count with differential, RBC count, protein, glucose
Gram stain and bacterial culture
HSV-1/HSV-2 PCR (if test available, consider HSV CSF IgG and IgM in addition)
VZV PCR (sensitivity may be low; if test available, consider VZV CSF IgG and IgM in addition)
Enterovirus PCR
Cryptococcal antigen and/or India ink staining
Oligoclonal bands and IgG index
VDRL

Serum

Routine blood cultures


HIV serology (consider RNA)
Treponema testing (RPR, specific treponemal test)
Hold acute serum and collect convalescent serum 1014 days later for paired antibody testing

Imaging

Neuroimaging (MRI preferred to CT, if available)


Chest imaging (chest x-ray and/or CT)

Neurophysiology

EEG

Other Tissues/Fluids

When clinical features of extra-CNS involvement are present, we recommend additional testing (e.g., biopsy of skin lesions,
bronchoalveolar lavage and/or endobronchial biopsy in those with pneumonia/pulmonary lesions, throat swab PCR/culture in
those with upper respiratory illness, stool culture in those with diarrhea); also see below.

Conditional Studies

Host Factors

ImmunocompromisedCMV PCR, HHV-6/HHV-7 PCR, HIV PCR (CSF); Toxoplasma gondii serology and/or PCR;
Mycobacterium tuberculosis testing; fungal testing; WNV testing

Geographic Factors

Africamalaria (blood smear), trypanosomiasis (blood/CSF smear, serology from serum and CSF), dengue testing
AsiaJapanese encephalitis virus testing, dengue testing, malaria (blood smear), Nipah virus testing (serology from serum and CSF;
PCR, immunohistochemistry, and virus isolation in a BSL4 lab can also be used to substantiate diagnosis)
AustraliaMurray Valley encephalitis virus testing, Kunjin virus testing, Australian bat Lyssavirus (ABLV) testing
Europetick-borne encephalitis virus (TBEV) (serology); if southern Europe, consider WNV testing, Toscana virus testing
Central and South Americadengue testing, malaria (blood smear), WNV testing, Venezuelan equine encephalitis testing
North Americageographically appropriate arboviral testing (e.g., WNV, Powassan, La Crosse, eastern equine encephalitis viruses,
Lyme [serum ELISA and Western blot])

(continued)

Scheld_Ch06.indd 92 2/21/14 5:31 PM


Chapter 6: Encephalitis 93

TA B L E 6 . 1
DIAGNOSTIC ALGORITHM FOR INITIAL EVALUATION OF ENCEPHALITIS IN ADULTS (CONTINUED)

Conditional Studies

Season and Exposure

Summer/fallarbovirus and tick-borne disease testinga


Cat (particularly if with seizures, paucicellular CSF)Bartonella antibody (serum), ophthalmologic evaluation
Tick exposuretick-borne disease test a
Animal bite/bat exposurerabies testing
Swimming or diving in warm freshwater or nasal/sinus irrigationNaegleria fowleri (CSF wet mount and PCR)

Specific Signs and Symptoms

Psychotic features or movement disorderanti-NMDAR antibody (serum, CSF); rabies testing; screen for malignancy, Creutzfeldt-
Jakob disease
Prominent limbic symptomsautoimmune limbic encephalitis testing; HHV-6/HHV-7 PCR (CSF); screen for malignancy
Rapid decompensation (particularly with animal bite history or prior travel to rabies-endemic areas)rabies testing
Respiratory symptoms Mycoplasma pneumoniae serology and throat PCR (if either positive, then do CSF PCR); respiratory virus testing
Acute flaccid paralysisarbovirus testing, rabies testing
Parkinsonismarbovirus testing, Toxoplasma serology
Nonhealing skin lesionsBalamuthia mandrillaris, Acanthamoeba testing

Laboratory Features

Elevated transaminasesrickettsia serology, tick-borne diseases testinga


CSF protein 100mg/dL, or CSF glucose 2/3 peripheral glucose, or lymphocytic pleocytosis with subacute symptom onset
M. tuberculosis testing, fungal testing
CSF protein 100 mg/dL or CSF glucose 2/3 peripheral glucose and neutrophilic predominance with acute symptom onset and
recent antibiotic useCSF PCR for Streptococcus pneumoniae and Neisseria meningitidis
CSF eosinophiliaM. tuberculosis testing, fungal testing, Baylisascaris procyonis antibody (serum), Angiostrongylus cantonensis and
Gnathostoma sp. testing
RBCs in CSFNaegleria fowleri testing
Hyponatremiaanti-VGKC antibody (serum); M. tuberculosis testing

RBC, red blood cell; VDRL, Venereal Disease Research Laboratory; RPR, rapid plasma reagin; EEG, electroencephalogram; CMV, cytomegalovirus.
a
Tick-borne disease testing should be tailored to specific geographic region and typically consists of serology (i.e., Borrelia, Ehrlichia, Rickettsia sp.,
Anaplasma phagocytophilum, TBEV) and blood PCR (Ehrlichia, Anaplasma).
Data from Venkatesan A, Tunkel AR, Bloch KC, et al. Case definitions, diagnostic algorithms, and priorities in encephalitis: consensus statement of the
International Encephalitis Consortium. Clin Infect Dis. 2013;57(8):11141128.

or rash illness should be investigated. Based on this information, neutrophils degrade within a few hours (186). Additionally, if
specific assays can be performed (Tables 6.1 and 6.2) (184). the CSF cell counts are performed by an automated cell counter,
A complete blood count, renal function tests, hepatic enzyme eosinophils can be mistaken for neutrophils. Accurate identifica-
levels, and coagulation studies should be included in the evalua- tion of eosinophils is best done by Giemsa or Wright stain of CSF
tion of a patient with suspected encephalitis. An initial chest radio- (128). The identification of CSF oligoclonal bands, representing
graph should also be performed as focal infiltrates are suggestive intrathecal antibody synthesis, is a nonspecific finding but can be
of certain pathogens (e.g., fungal or mycobacterial infections) and helpful in corroborating an infectious or inflammatory etiology.
might lead to other diagnostic studies (e.g., bronchoscopy). A The CSF profile offers important diagnostic clues on
thorough eye exam by an ophthalmologist may identify a migra- whether a patient has an infectious or noninfectious etiology
tory nematode which would considerably narrow the differential. and, within the infectious sphere, whether the cause is likely
Unless there is a specific contraindication, a lumbar puncture viral, bacterial, fungal, or parasitic. The CSF profile in most
(LP) with cerebrospinal fluid (CSF) analysis (cell count with dif- viral encephalitis patients demonstrates CSF mononuclear
ferential analysis, glucose and protein concentrations) and a mea- cell pleocytosis, with cell counts ranging from 10 to 200 mg/
surement of the opening pressure should be performed (185). A dL. However, several studies have shown that pleocytosis can
simultaneous peripheral glucose should also be measured and is be absent or there may be an elevation in neutrophils early
particularly important in a diabetic patient because what appears in the course. Although many textbooks mention that poly-
to normal CSF glucose might actually be low if the ratio of morphonuclear leukocytes (PMNs) can predominate the first
CSF to serum glucose is not taken into consideration (102). The 24 hours of viral CNS infections and suggest that a shift to
CSF sample should be analyzed promptly, particularly because mononuclear cells occurs after that time period, there have

Scheld_Ch06.indd 93 2/21/14 5:31 PM


94 Part II: Viral Infections and Related Disorders

TA B L E 6 . 2
DIAGNOSTIC ALGORITHM FOR INITIAL EVALUATION OF ENCEPHALITIS IN CHILDREN

Routine Studies

CSF

Collect at least 5 mL fluid, if possible; freeze unused fluid for additional testing
Opening pressure, WBC count with differential, RBC count, protein, glucose
Gram stain and bacterial culture
HSV-1/HSV-2 PCR (if test available, consider HSV CSF IgG and IgM in addition)
Enterovirus PCR

Serum

Routine blood cultures


EBV serology (VCA IgG and IgM and EBNA IgG)
Mycoplasma pneumoniae IgM and IgG
Hold acute serum and collect convalescent serum 1014 days later for paired antibody testing

Imaging

Neuroimaging (MRI preferred to CT, if available)


Neurophysiology
EEG
Other Tissues/Fluids
M. pneumoniae PCR from throat sample
Enterovirus PCR and/or culture of throat and stool
When clinical features of extra-CNS involvement are present, we recommend additional testing (e.g., biopsy of skin lesions,
bronchoalveolar lavage and/or endobronchial biopsy in those with pneumonia/pulmonary lesions, throat swab PCR/culture in
those with upper respiratory illness, stool culture in those with diarrhea); also see below.

Conditional Studies

Host Factors

Age younger than 3 yearsparechovirus PCR (CSF)


ImmunocompromisedCMV PCR, HHV-6/HHV-7 PCR, HIV PCR (CSF); cryptococcal antigen; Toxoplasma gondii serology and/or
PCR; Mycobacterium tuberculosis testing; fungal testing; WNV testing

Geographic Factors

Africamalaria (blood smear), trypanosomiasis (blood/CSF smear, serology from serum and CSF), dengue testing
AsiaJapanese encephalitis virus testing, dengue testing, malaria (blood smear), Nipah virus testing (serology from serum and CSF;
PCR, immunohistochemistry, and virus isolation in a BSL4 lab can also be used to substantiate diagnosis)
AustraliaMurray Valley encephalitis virus testing, Kunjin virus testing, Australian bat Lyssavirus (ABLV) testing
Europetick-borne encephalitis virus (TBEV) (serology); if Southern Europe, consider WNV testing, Toscana virus testing
Central and South Americadengue testing, malaria (blood smear)
North Americageographically appropriate arboviral testing (e.g., WNV, Powassan, La Crosse, eastern equine encephalitis viruses,
Lyme [serum ELISA and Western blot])

Season and Exposure

Summer/fallarbovirus and tick-borne disease testinga


Cat (particularly if with seizures, paucicellular CSF)Bartonella antibody (serum), ophthalmologic evaluation
Tick exposuretick-borne disease testa
Animal bite/bat exposurerabies testing
Swimming or diving in warm freshwater or nasal/sinus irrigationNaegleria fowleri (CSF wet mount and PCR)

(continued)

Scheld_Ch06.indd 94 2/21/14 5:31 PM


Chapter 6: Encephalitis 95

TA B L E 6 . 2
DIAGNOSTIC ALGORITHM FOR INITIAL EVALUATION OF ENCEPHALITIS IN CHILDREN (CONTINUED)

Conditional Studies

Specific Signs and Symptoms

Abnormal behavior (e.g., new-onset temper tantrums, agitation, aggression), psychotic features, seizures, or movement disorder
anti-NMDAR antibody (serum, CSF), oligoclonal bands, IgG index, rabies testing
Behavior changes followed by myoclonic spasms/jerksmeasles IgG (CSF and serum)
Vesicular rashVZV PCR from CSF (sensitivity may be low; if test available, consider CSF IgG and IgM), VZV IgG and IgM from serum
Rapid decompensation (particularly with animal bite history or prior travel to rabies-endemic areas)rabies testing
Respiratory symptomschest imaging (chest x-ray and/or CT scan), respiratory virus testing, Mycoplasma pneumoniae PCR (CSF)
Acute flaccid paralysisarbovirus testing, rabies testing
Parkinsonismarbovirus testing, Toxoplasma serology
Nonhealing skin lesionsBalamuthia mandrillaris, Acanthamoeba testing
Prominent limbic symptomsautoimmune limbic encephalitis testing, HHV-6/HHV-7 PCR (CSF)

Laboratory Features

If EBV serology is suggestive of acute infection, perform EBV PCR (CSF)


Elevated transaminasesrickettsia serology, tick-borne diseases testinga
CSF protein 100 mg/dL, or CSF glucose 2/3 peripheral glucose, or lymphocytic pleocytosis with subacute symptom onset
M. tuberculosis testing, fungal testing, Balamuthia mandrillaris testing
CSF protein 100 mg/dL or CSF glucose 2/3 peripheral glucose and neutrophilic predominance with acute symptom onset and
recent antibiotic useCSF PCR for Streptococcus pneumoniae and Neisseria meningitidis
CSF eosinophiliaM. tuberculosis testing, fungal testing, Baylisascaris procyonis antibody (serum), Angiostrongylus cantonensis and
Gnathostoma sp. testing
HyponatremiaM. tuberculosis testing
M. pneumoniae serology or throat PCR positiveM. pneumoniae PCR (CSF)

RBC, red blood cell; VCA, viral capsid antigen; EBNA, Epstein-Barr nuclear antigen; EEG, electroencephalogram; CMV, cytomegalovirus.
a
Tick-borne disease testing should be tailored to specific geographic region and typically consists of serology (i.e., Borrelia, Ehrlichia, Rickettsia sp.,
Anaplasma phagocytophilum, TBEV) and blood PCR (Ehrlichia, Anaplasma).
Adapted from Venkatesan A, Tunkel AR, Bloch KC, et al. Case definitions, diagnostic algorithms, and priorities in encephalitis: consensus statement of
the International Encephalitis Consortium. Clin Infect Dis. 2013;57(8):11141128.

been studies showing that PMNs may still predominate well increased protein content (75 to 970 mg/dL). The amebae
beyond this 24-time period (187). Therefore, a repeat LP a few can sometimes be seen in wet-mount preparations of spinal
days following the first LP may be useful (188,189). In viral fluid (139,140,190). In patients with CNS Balamuthia infec-
encephalitis, the CSF protein generally is elevated but is typi- tions, elevated CSF WBC is often seen along with mild to
cally less than 100 mg/dL, whereas the glucose level is almost markedly elevated protein (1,000 mg/dL) and normal or
always normal with a few important exceptions (e.g., LCM slightly decreased glucose.
and mumps; Table 6.3). Although brain biopsy in the setting of suspected enceph-
As outlined earlier, viral infections can sometimes be associ- alitis has become less common, examination of brain tissue
ated with a neutrophilic predominance; however, when a neu- still has utility because of limitations in both molecular and
trophilic pleocytosis is observed, particularly in cases where serologic methods. The use of targeted biopsy was recently
CSF white blood cell (WBC) count is more than 1,000 cells/ demonstrated by a report of 16 patients with undiagnosed
mm3, protein more than 100 mg/dL, or CSF glucose level less CNS illness where brain biopsy detected bacterial abscess (6),
than 2/3 of serum levels, a nonviral entity should be strongly toxoplasmosis (3), HSV (1), Aspergillus infection (2), and M.
considered. For most bacterial pathogens, the CSF shows a tuberculosis infection (2). Biopsies can also be particularly
WBC of 1,000 to 5,000 cells/mL with predominance of neu- helpful for the diagnosis of noninfectious entities such as small
trophils (80% to 95%), glucose less than 40 mg/dL in over half vessel vasculitis and intravascular lymphoma (191). An au-
the cases, and a CSF glucose level less than 2/3 serum levels. topsy should be encouraged to determine the cause of death in
In fungal infections, moderate lymphocytic pleocytosis usually patients who die with unexplained encephalitis.
is found along with elevated protein and low CSF glucose. In addition to studies done acutely, an extra red top serum
Eosinophils in the CSF are suggestive of a helminth infec- tube should be drawn during the acute phase of illness and
tion (e.g., B. procyonis, Angiostrongylus spp., and G. spi- held for later serologic studies. A convalescent serum should
nigerum). Eosinophils can also be seen in patients with be collected 10 to 21 days later. Similarly, extra spinal fluid
neurococcidiomycosis or neuro-Mycobacterium tuberculosis. should also be frozen for future testing. Specific caveats of test-
In patients with PAM, the CSF typically contains a very high ing are outlined in Table 6.3 and selected agents are outlined in
CSF cell count with a predominance of neutrophils, a slight the following sections.
to pronounced decrease in glucose concentration, and an Text continues on page 100

Scheld_Ch06.indd 95 2/21/14 5:31 PM


96 Part II: Viral Infections and Related Disorders

TA B L E 6 . 3
DIAGNOSTIC TESTING FOR INFECTIOUS ENCEPHALITIS WITH FOCUS ON PATHOGENS FOUND IN
IMMUNOCOMPETENT INDIVIDUALS IN THE UNITED STATES

Agent Recommended Diagnostic Studies Laboratory Findings; Limitations and Caveats

Viral Many CNS viral infections result in a mild CSF pleocytosis


(typically with a monocytic cell predominance but PMNs may
predominate, especially early in course); CSF protein normal to
mildly elevated (100 mg/dL) and normal glucose

Adenovirus CSF, respiratory, brain tissue PCR Pathogen of unknown neurologic potential
or culture CSF PCR testing rarely positive
Arboviruses Serology is the best assay for most PCR often negative due to acute viremia stage often completed
arboviruses: CSF IgM, serum IgM by the time of clinical presentation
and IgG (paired sera if possible) PCR may be helpful in immunocompromised host or very early
for specific viruses as suggested in clinical course.
by geography: CSF IgG for specific arbovirus usually not helpful because
West Nile virus (WNV) bloodbrain barrier integrity may be compromised; CSF blood
California serogroup viruses (e.g., contamination can cause false-positive results for both IgM
La Crosse virus [LACV]) and IgG.
Eastern equine encephalitis virus IgM can persist for several months and sometimes 1 year;
(EEEV) more likely to persist in serum, but persistence has also been
Powassan virus (POWV) described in CSF.
St. Louis encephalitis virus Serologic cross-reactivity among arbovirus of same family
(SLEV) (WNV, SLEV, and POWV antibodies cross-react; e.g.,
Western equine encephalitis virus individual with prior dengue infection will test positive for
(WEEV) WNV IgG)
Antibody typically positive early in presentation but if
negative, repeat on later specimen
WNV: PMNs can persist in CSF.
WNV: Reactive lymphocytes in CSF (e.g., Mollaret-like cells)
have also been described.
EEEV: CSF WBC counts may be similar to bacterial meningitis
(e.g., counts up to 4,000/mm3 described).
POWV testing only available at CDC and a few state
laboratories
Cytomegalovirus (CMV)a CSF PCR Serology can be problematic; false-positive IgM not uncommon
PCR: false positive (consider if viral load is low); CMV in CSF
also seen presumably because of latent infection
Atypical lymphocytes in CSF
Enteroviruses (EV) CSF PCR Testing of EV PCR on CSF alone may miss infection because
Respiratory PCR EV present only transiently in CSF; test non-CNS site
Stool PCR or culture (respiratory sample PCR, viral stool culture) to increase yield.
Epstein-Barr virus (EBV) CSF PCR Both serology (serum) and PCR (CSF) for EBV is recommended.
Serum: anti-VCA IgM/IgG, anti- Heterophil low sensitivity in children younger than 5 years of age
EBNA PCR false positives can occur if EBV-infected (latent)
Heterophil mononuclear cells present
EBV CSF PCR can be negative in true cases because of
either timing of LP (late) or mechanism other than direct
neuroinvasion.
Atypical lymphocytes in CSF or peripheral blood is consistent
with EBV but not always present.
Hepatitis C virus CSF PCR Pathogen of unknown neurotropic potential
Herpes simplex virus 1 CSF PCR HSV-1 causes the majority of HSE (outside neonatal period).
(HSV-1) Intrathecal antibody if 1 week ~5%10% of HSE patients have a normal CSF formula in the
of symptoms first LP, particularly in children.
False-negative PCRs occur; if clinical suspicion for HSE,
consider repeat lumbar puncture early in the course of illness
to repeat CSF HSV PCR and intrathecal HSV antibody testing
Presence of either IgG or IgM may be indicative of CNS
infection; however, bloodbrain barrier integrity and CSF-
blood contamination needs to be considered when interpreting
results

(continued)

Scheld_Ch06.indd 96 2/21/14 5:31 PM


Chapter 6: Encephalitis 97

TA B L E 6 . 3
DIAGNOSTIC TESTING FOR INFECTIOUS ENCEPHALITIS WITH FOCUS ON PATHOGENS FOUND IN
IMMUNOCOMPETENT INDIVIDUALS IN THE UNITED STATES (CONTINUED)

Agent Recommended Diagnostic Studies Laboratory Findings; Limitations and Caveats

Herpes simplex virus 2 CSF PCR Presence of either IgG or IgM may be indicative of CNS
(HSV-2) Intrathecal antibody if 1 week infection; however, bloodbrain barrier integrity and CSF-
of symptoms blood contamination needs to be considered when interpreting
results
Human herpes virus 6 CSF PCR Pathogen of unknown neurologic potential
(HHV-6) (Quantitative blood PCR if Not all positive HHV-6 CSF PCR results correlate with disease;
CSF positive to determine when positive, important to consider chromosomal integration
chromosomal integration) or latent infection
Human herpes virus 7 CSF PCR Unknown frequency of CNS illness
(HHV-7) Pathogen of unknown neurologic potential; detection of virus
may represent acute disease but could also represent latent
virus.
HIV ELISA and Western blot (single
serum)
Plasma and CSF PCR
Human Respiratory tract (NP or throat) Pathogen of unknown neurologic potential
metapneumovirus PCR CSF PCR rarely positive
Influenza A/B virus Respiratory viral culture PCR of CSF rarely positive
Respiratory viral antigen test PCR on respiratory specimens more sensitive and specific than
Respiratory PCR (if above antigen assays
negative) Most cases are encephalopathy (vs. encephalitis).
JC virusa CSF PCR A positive result corroborates diagnosis; negative result does
not rule it out.
Lymphocytic CSF IgM/IgG CSF profile may resemble bacterial etiology (e.g., CSF WBC
choriomeningitis Serum IgM/IgG sometimes 3,000, PMNs predominance, low glucose and/or
(LCM) virus very elevated proteins).
One of the few viruses that can decrease glucose in CSF
Measles virusacute CSF antibodies CNS illness occurs ~1 week after fever/rash.
CSF PCR Measles PCR and antibody testing (serum and CSF)
Serum IgG/IgM (paired serumb
samples if possible)
Brain tissue PCR
Measles virusSSPEc CSF IgG antibodies Incubation period several years
Serum IgG antibodies Because SSPE is a result of long-standing infection, IgM will be
Brain tissue PCR negative; IgG levels in both CSF and serum very high
Measles CSF PCR rarely positive, PCR brain tissue positive
CSF formula generally unremarkable
Oligoclonal bands present in CSF
Mumps virus CSF culture or PCR One of few viruses that can cause low glucose in CSF
CSF IgM/IgM
Serum IgM/IgG (paired serumb
samples if possible)
Throat swab PCR
Parvovirus B19 Serum IgM Pathogen of unknown neurologic potential
CSF PCR
Rabies virus Antemortem: Coordinate testing with local and state health department and
Serum antibodies CDC.
Saliva PCR Antemortem testing is possible; multiple samples and different
Nuchal biopsy PCR and DFA assays are important to run. Negative tests antemortem do not
Brain tissue DFA rule out rabies.
Postmortem:
Brain tissue; viral isolation or
antigen detection
Respiratory syncytial RSV antigen Pathogen of unknown neurologic potential
virus (RSV) CSF PCR rarely positive
CSF profile often unremarkable

(continued)

Scheld_Ch06.indd 97 2/21/14 5:31 PM


98 Part II: Viral Infections and Related Disorders

TA B L E 6 . 3
DIAGNOSTIC TESTING FOR INFECTIOUS ENCEPHALITIS WITH FOCUS ON PATHOGENS FOUND IN
IMMUNOCOMPETENT INDIVIDUALS IN THE UNITED STATES (CONTINUED)

Agent Recommended Diagnostic Studies Laboratory Findings; Limitations and Caveats

Rotavirus Stool antigen Pathogen of unknown neurologic potential


CSF PCR (testing available Typically young child with history of diarrhea; mechanism
specialized laboratories, unclear
e.g., CDC) CSF profile often unremarkable
Rubella virus Serology One of viral causes of low CSF glucose
CSF antibodies
Varicella-zoster virus CSF PCR Because some CNS VZV infections are reactivation, IgM not
(VZV) Intrathecal antibody if 1 week always positive
of symptoms Positive VZV skin lesions do not prove CNS etiology but may
Serum IgM/IgG be suggestive of etiology.
Skin lesions: DFA or PCR
West Nile virus See arboviruses (above)

Fungal Fungal CNS infections are often associated with CSF pleocytosis
with lymphocytic predominance, low glucose, and protein
100 mg/dL. EDTA-heat treated antigen test reported to
increase sensitivity for CSF and serum samples. Isolated CNS
disease can be difficult to diagnose because of insensitive
assays.
Note geographic locations of different fungal infections; order
antigen for each specific fungi.

Coccidioides spp.c CSF culture (large volume) Alert laboratory that coccidiomycosis is being considered if
fungal culture sent
Cryptococcus spp.c CSF-specific antigen Eosinophils sometimes present in CSF in coccidiomycosis
patients
Histoplasma capsulatumc CSF-specific antibody
and Blastomyces sp.c CSF India ink (for Cryptococcus
only)
Serum-specific antigen
Serum-specific antibody
Urine antigen (Histoplasma and
Blastomyces)

Free-living amebas and


parasites

Naegleria fowleri Wet mount of warm CSF Coordinate testing with local and state health department and
Brain histopathology the CDC.
Demonstration of motile ameba on wet mount of CSF
LP often demonstrates very high WBC (often PMN
predominance), high protein (100 mg/dL), and low glucose
(50 mg/dL).
Balamuthia mandrillarisc Serology Coordinate testing with local and state health department and
CSF and/or brain PCR the CDC.
Brain histopathology (special Serology and PCR available at specialized laboratories (CDC)
stains) Brain tissue may show necrotic and hemorrhagic
meningoencephalitis.
LP often shows high WBC (L or PMN) and protein 100 mg/
dL, resembling tuberculous meningitis.
Acanthamoeba spp.a,c Serology (research laboratories) Coordinate testing with local and state health department and
CSF and/or brain PCR the CDC.
Brain histopathology (special Serology and PCR available at specialized laboratories
stains)
Baylisascaris procyonis CSF and serum antibodies Serology available in specialized laboratories (CDC)
Eosinophils almost always present in CSF and CBC
Toxoplasma gondiia Older child: IgG, IgM, and PCR Often reactivation of disease, so IgM may not be positive; IgG
titers persistently high

(continued)

Scheld_Ch06.indd 98 2/21/14 5:31 PM


Chapter 6: Encephalitis 99

TA B L E 6 . 3
DIAGNOSTIC TESTING FOR INFECTIOUS ENCEPHALITIS WITH FOCUS ON PATHOGENS FOUND IN
IMMUNOCOMPETENT INDIVIDUALS IN THE UNITED STATES (CONTINUED)

Agent Recommended Diagnostic Studies Laboratory Findings; Limitations and Caveats

Bacteria

Bartonella henselae Serology (IFA) CSF PCR (if available) may also be useful to perform
PCR of lymph node CSF often negative
Borrelia burgdorferi Serology (serial EIA and Western Test both serum and CSF for Borrelia antibody; may be
blot) delayed in CNS intrathecal synthesis
CSF antibody index CSF PCR rarely positive (in contrast to synovial fluid) but may
be useful in some cases
Brucella spp. CSF IgG and IgM Serologyperform on both CSF and serum; culture increases
CSF culture sensitivity.
Serum IgG and IgM PCR available in some research settings; unknown sensitivity
False-positive IgM not uncommon
Leptospira spp. Serum IgM and IgG If serology negative on acute serum, important to repeat on
Urine culture convalescent serum
Listeria monocytogenes Routine bacterial culture on both CSF culture for Listeria relatively insensitive
CSF and blood CSF profile does not necessarily look bacterial, that is, can be
Multiple CSF cultures may be normal or have only a few hundred WBCs with predominance
helpful of lymphocytes; glucose and protein may be normal.
It may also be helpful to test CSF Listeria antibody because
detection of CSF antibody may be indicative of CNS infection.
Mycobacterium CSF AFB smear, culture, PCR, CSF PCR is insensitive tool for detection of CNS M. tuberculosis;
tuberculosisc direct examination important to test multiple samples.
Respiratory culture highly CNS M. tuberculosis should be considered in patients with
suggestive lymphocytic pleocytosis (but neutrophilic predominance can still
occur), CSF protein 100 mg/dL, and CSF glucose 50 mg/dL.
Mycoplasma pneumoniae PCR of NP or other respiratory Pathogen of unknown neurologic potential
specimen Perform PCR on respiratory samples, serology on acute/
Serum IgM convalescent serum
Serum IgG paired CSF PCR rarely positive
Single IgG titer is not helpful.
Treponema pallidum CSF VDRL CSF VDRL specific but CSF FTA-ABS more sensitive
Serum PCR with confirmatory
FTA-ABS

Rickettsia In patients with rickettsial infections, low peripheral WBC, low


platelets, and increased liver function tests (LFTS) often present

Anaplasma Morulae in WBCs (granulocytes) If serology negative on acute serum, important to repeat on
phagocytophilum Whole blood PCR convalescent serum (seroconversion may occur a few weeks
Serology IgG/IgM (paired serab if after onset)
possible)
Coxiella burnetii Serology on paired serum samples Utility of Coxiella burnetii PCR in CSF unknown
Ehrlichia chaffeensis Morulae in WBCs (monocytes) If serology negative on acute serum, important to repeat on
Whole blood PCR convalescent serum (seroconversion may occur a few weeks
Serum IgG/IgM (paired serab if after onset)
possible)
Ehrlichia ewingii Morulae in WBCs (granulocytes) If serology negative on acute serum, important to repeat on
Whole blood PCR convalescent serum (seroconversion may occur a few weeks
Serum IgG/IgM after onset)
Rickettsia spp. Serology (IFA) If serology negative on acute serum, important to repeat on
Skin biopsy of rashPCR or convalescent serum (seroconversion may occur a few weeks
immunohistochemical staining after onset)

NP, nasopharyngeal; DFA, direct fluorescent antibody; EDTA, ethylenediaminetetraacetate; CBC, complete blood count; EIA, enzyme immunoassay; AFB,
acid-fast bacilli; VDRL, Venereal Disease Research Laboratory; FTA-ABS, fluorescent treponemal antibody-absorption; PMNs, polymorphonuclear cells.
a
Causes encephalitis primarily in neonate and/or immunocompromised host.
b
Testing should be done on acute serum and then later when acute and convalescent sera are available.
c
Typically presents as subacute chronic illness.
Data from Venkatesan A, Tunkel AR, Bloch KC, et al. Case definitions, diagnostic algorithms, and priorities in encephalitis: consensus statement of the
International Encephalitis Consortium. Clin Infect Dis. 2013;57(8):11141128.

Scheld_Ch06.indd 99 2/21/14 5:31 PM


100 Part II: Viral Infections and Related Disorders

CSF PCR testing is recommended for the diagnosis of HHV-6


TESTING OF SELECTED and HHV-7; however, the finding of a positive CSF PCR for
ETIOLOGIES HHV-6 or HHV-7 is not necessarily equivalent to a diagnosis
of HHV-6 or HHV-7 encephalitis. This is because HHV-6 DNA
can be present in normal brain tissue and HHV-6 may be iden-
Viral Testing tified in CSF as an incidental bystander rather than a cause
of infection (201). Additionally, chromosomal integration must
Specific studies for viral agents often employ molecular testing. be considered as a potential reason for a positive PCR in the
Molecular testing by nucleic acid amplification (e.g., PCR) test- CSF. If a positive HHV-6 CSF PCR is obtained, an evaluation
ing provides more timely and typically more sensitive results than of HHV-6 in the blood should be done to distinguish between
culture for viral CNS infections (116,192,193). However, the chromosomal integration and acute infection (48).
limitations of these tests are often not recognized by physicians,
particularly false-negative and false-positive results as discussed Enteroviruses and Parechoviruses
in the following sections for specific agents. Testing for intrathe-
cal antibody synthesis of specific pathogens such as arboviruses PCR testing of both CSF and a throat specimen should be per-
and herpesviruses can be useful adjunct for diagnosis, particu- formed; if only CSF is tested, the diagnosis may be missed. For
larly in the later stages of disease (see the following discussion). example, in an outbreak of EV-71 in the United States, EV PCR
In patients with either preceding or concurrent respiratory of CSF was positive in only 5 (31%) of 16 cases, whereas PCR
symptoms, a viral culture from the respiratory tract should be testing of throat specimens were uniformly positive (202,203).
performed early in the hospital course to optimize the virus Because EVs may be shed in the stool for weeks after infection,
recovery. When a particular agent such as influenza or another culturing the stool for EV may increase the diagnostic yield; for
virus is suspected but the viral culture is negative, agent-specific the same reason, a positive stool is suggestive, but not confir-
PCR testing should also be performed. Similarly, if there is a matory, of the diagnosis. Testing for parechoviruses should be
history of preceding or concurrent diarrhea, a stool culture for considered in all children younger than 3 years of age with the
viral and, possibly, bacterial pathogens should be performed. same range of biologic samples tested as EVs. Parechoviruses
In children, a stool viral culture and/or EV PCR should be can only be detected by parechovirus-specific PCR assays.
routinely performed to enhance EV detection, regardless of the Arbovirus
presence of diarrhea.
For most arboviruses, serologic testing of CSF and serum is
preferred to molecular testing because peak viremia gener-
Herpesviruses ally occurs prior to symptom onset (204). The diagnosis of
The diagnostic test of choice for HSE is CSF HSV-1 DNA WNV infection is typically made via detection of IgM cap-
nucleic acid (PCR) testing. If testing from the first LP is nega- ture enzyme-linked immunosorbent assay (MAC-ELISA) or
tive and HSE is still suspected (e.g., temporal lobe involvement a fourfold or greater change in antibody titer on paired or
seen on neuroimaging), a second LP should be performed convalescent serum. Approximately 90% of patients with neu-
within 24 to 48 hours as a number of studies have shown that roinvasive disease will have detectable CSF IgM antibody by
HSV PCR can be falsely negative, particularly early in course days 8 to 10 after symptom onset (205). However, WNV IgM
of illness and in the pediatric age-group (22,189). For HSE, may persist for prolonged periods, with detectable IgM titers
intrathecal antibodies can be performed as a complement to persisting more than 500 days after presentation in some indi-
molecular testing, and this may be particularly helpful in es- viduals with neuroinvasive disease (206). Furthermore, there
tablishing the diagnosis later in the course. Intrathecal synthe- is serologic cross-reactivity between the flaviviruses (SLEV,
sis of HSV-specific IgG antibodies can often be detected 10 to dengue virus, yellow fever virus, or JEV), such that individuals
14 days after the onset of illness (194). infected with, or vaccinated against, one of these agents could
As mentioned in the clinical and epidemiology section, the test positive for WNV by IgM MAC-ELISA. Differentiation of
clinical spectrum of CNS infections associated with HSV-2 WNV from other flaviviruses is achieved by plaque reduction
often differs from HSV-1. Molecular testing of CSF for the neutralization testing (PRNT) (207).
presence of nucleic acid is the most reliable method for diag- For other arboviruses, the standard method for diagnosis
nosis for HSV-2; intrathecal antibody testing can be used as an is similar, namely through detection of IgM or a fourfold or
adjunct as described previously for HSV-1. greater change in antibody titer on paired acute and convales-
Because VZV encephalitis is one of the most common causes cent serum (208).
of encephalitis in adults, VZV testing should be performed in
all adult patients, with or without skin lesions. Testing should Other Viral Testing
include both CSF VZV PCR and CSF VZV antibody (195). The diagnosis of rabies should be considered in any rapidly
Although detection of VZV antibody in the CSF can be more progressing encephalitis. Antemortem testing for rabies re-
sensitive than PCR, intrathecal synthesis may be delayed by a quires highly specialized testing in a public health laboratory
week or more after onset of neurologic symptoms (196,197). using a combination of different assays and specimen types
In the event of a traumatic tap, distinguishing intrathecal anti- (209). Whenever the diagnosis of rabies is entertained, consul-
body synthesis versus blood contamination of CSF may be tation with public health authorities is strongly recommended.
accomplished by calculation of an antibody index (198). Testing for other viruses should be performed as suggested by
For the diagnosis of EBV encephalitis, both serology and mo- exposures or travel history.
lecular testing is recommended (40). Quantitative viral load may
be helpful, as a high viral load supports a significant CNS infec-
tion, whereas a low positive PCR in CSF may represent latent virus Bacterial Testing
and may only be an incidental finding (199,200). Conversely, the
absence of a positive PCR in the setting of acute EBV serology In most instances, detection of routine bacterial pathogens
does not rule out acute infection, and a negative PCR may be a via Gram stain and/or culture is fairly straightforward. The
result of the timing of LP (i.e., after virus has cleared from CSF) CSF bacterial culture for L. monocytogenes, however, is rela-
or may reflect a mechanism other than direct infection. tively insensitive compared with many other bacterial agents.

Scheld_Ch06.indd 100 2/21/14 5:31 PM


Chapter 6: Encephalitis 101

Additionally, in contrast to many bacterial pathogens, the tomography (CT) and magnetic resonance imaging (MRI).
CSF profile of CNS listeriosis patients, particularly those with CT scanning involves the application of x-rays to produce to-
rhombencephalitis, can appear normal or similar to viral ill- mographic images, resulting in excellent delineation of bony
nesses with only a mild pleocytosis, a lymphocytic predomi- structures; conventional MRI uses nuclear magnetic resonance
nance rather than polynuclear predominance, and a normal to (MR) to detect hydrogen nuclei and provides good contrast
minimal protein elevation (109) (see Table 6.3). between soft tissues. Of the two, MRI is far more sensitive
Neurotuberculosis can also be difficult to diagnose. The and specific for the evaluation of infectious encephalitis (214
sensitivity of CSF tuberculosis (TB) PCR is very poor; reported 216). Conventional MRI imaging entails acquisition of vari-
sensitivity has been less than 25% in some studies (102). This ous sequences that provide complementary information. For
may be because of low bacillary load in the CSF, relatively example, T1-weighted imaging (T1W) is well suited to define
small CSF volumes submitted for testing, or PCR inhibitors structural abnormalities; T2-weighted imaging (T2W) and
in samples (210). Testing from both CNS and extra-CNS sites fluid-attenuated inversion recovery (FLAIR) sequences may
should be done for the workup of neuro-TB (see Table 6.3). identify areas of edema, demyelination, or inflammation; and
susceptibility-weighted imaging (SWI) is particularly useful
in identifying areas of hemorrhage. T1W imaging following
Fungal Testing gadolinium administration can identify parenchymal or men-
ingeal regions where the bloodbrain barrier has been com-
Fungal meningoencephalitis is typically a subacute illness. promised, as occurs in active inflammation. Of particular note
Fungal culture of the CSF should be performed in individu- is diffusion-weighted imaging (DWI), which identifies brain
als with indolent symptoms, immunocompromised persons, or regions where diffusion of water is restricted and may be more
when the CSF findings are suggestive (e.g., CSF lymphocytic sensitive than other conventional sequences for detection of
pleocytosis, high protein, and low glucose). If a fungal etiology early abnormalities in encephalitis (217,218).
is suspected, several different tests including culture, antigen Several additional imaging modalities have emerged for
test, and serology on both CNS as well as non-CNS specimens evaluation of encephalitis. MR spectroscopy can identify a
should be done (see Table 6.3) (211). number of metabolites, including N-acetylaspartate (NAA),
choline, creatine, and lactate, based on unique proton chemi-
cal shifts. Quantification of the ratios of these metabolites
Rickettsial Testing can provide information regarding inflammation, neuronal
loss, and demyelination. Both single-photon emission com-
Diagnostic testing is similar for most rickettsial infections. puted tomography (SPECT) imaging and fluorodeoxyglucose
Serology is the most widely used method, and the indirect positron emission tomography (FDG-PET) involve detec-
fluorescent antibody (IFA) assay is considered the gold stan- tion of radioisotopes within the brain following injection
dard. Because serologic tests may be negative early in the ill- into the bloodstream and can provide information regarding
ness, detection of the rickettsial agent in blood or tissues by metabolism. Other MR-based techniques, including mag-
molecular assays (e.g., PCR) may be useful. Confirmation of netization transfer and diffusion tensor imaging, as well as
disease is best accomplished by evaluation of paired serum molecular imaging techniques that allow for the detection of
specimens collected during the acute and convalescent phases disease-specific molecules, are currently being optimized and
of the illness for a fourfold or greater change in antibody titer. may eventually show promise in evaluation of patients with
Immunohistochemistry of patients tissues (e.g., brain) is an- encephalitis (219).
other methodology that can be used to diagnose rickettsial
infection (212).
Imaging of Selected Etiologies
Parasitic and Free-Living Ameba Testing Herpesviruses
The diagnosis of Baylisascaris is often suggested by a history of The neuroimaging findings of encephalitis caused by HSV-1
exposure to raccoons (or their feces), clinical presentation, and reflect its predilection for the limbic system, with frequent in-
persistent eosinophilia in the blood and spinal fluid. Serologic volvement of the mesial temporal and inferior frontal lobes
assays are available at the Centers for Disease Control and (Fig. 6.4).
Prevention (CDC). In patients with a history of travel outside In the acute stage, HSE typically results in edema, inflam-
the United States with presence of CSF eosinophilia, both mation, or hemorrhage. Although initial CT scans are nor-
gnathostomiasis and Angiostrongylus should be considered. mal in up to 25% of affected individuals, greater than 90%
An ELISA test has been developed but is not widely available of patients with HSE documented by CSF PCR will have MRI
(213). Molecular tests for Angiostrongylus are available at abnormalities (220223). Abnormalities on conventional MRI
the CDC. appear within 48 hours and include isointense or hypointense
Molecular technology is also available for the identification lesions on T1W images and hyperintense lesions on T2W or
of Balamuthia and Acanthamoeba DNA in tissues and CSF FLAIR images. Changes in DWI, which may reflect the rapid
(CSF PCR is less sensitive than brain tissue PCR) but is largely accumulation of intracellular water in the setting of impaired
limited to research laboratories and the CDC. ion transport during acute infection, are seen even earlier
(218,224,225). MRI is superior to CT in the detection of sub-
acute (1 week) hemorrhage. Contrast enhancement on MRI
NEUROIMAGING MODALITIES is typically in a cortical gyral pattern and often lags behind
symptom onset (226). Extratemporal abnormalities occur in
Neuroimaging plays a crucial role in the evaluation of patients over half of cases and may involve the cerebral hemispheres,
with suspected encephalitis, as it may support the diagnosis of thalamus, and brainstem (227229). Notably, in up to 15% of
a specific etiology or identify alternate conditions that mimic patients, extratemporal involvement may be the sole-observed
encephalitis (Table 6.4). Routinely available studies to evalu- neuroimaging abnormality (229) and is more likely to occur in
ate patients with suspected encephalitis include computed pediatric and immunocompromised populations (230232).

Scheld_Ch06.indd 101 2/21/14 5:31 PM


102 Part II: Viral Infections and Related Disorders

TA B L E 6 . 4
NEUROIMAGING IN ACUTE ENCEPHALITIS

Location Possible Etiologies Imaging Features Possible Etiologies

Basal ganglia Arboviruses Arteritis/ infarction Aspergillus spp.a


Enteroviruses Chikungunya virus
Epstein-Barr virus Nipah virus
Influenza virus (ANE) Treponema pallidum
Measles virus (acute infection) Varicella-zoster virus
Mycobacterium tuberculosis Mycobacterium tuberculosis
Respiratory viruses Other fungal infections
Anti-NMDAR encephalitis Calcifications Cytomegalovirus (neonate)
Brainstem Arboviruses Toxoplasma gondii (neonate;
Brucellosis periventricular)a
Enteroviruses Hemorrhage Herpes simplex virus 1
Epstein-Barr virus Influenza (ANE)
Herpes simplex virus 1 Varicella-zoster virus
Influenza virus (ANE)
Hydrocephalus Balamuthia mandrillaris
Japanese encephalitis
Coccidioides immitis
Listeria monocytogenes
Histoplasma capsulatum
Lyme
Mycobacterium tuberculosis
Mycobacterium tuberculosis
Toxoplasma gondii (neonate)a
Rabies
West Nile virus Microcephaly Cytomegalovirusa
Anti-NMDAR encephalitis Toxoplasma gondii (neonate)a
Cerebellum Epstein-Barr virus Space-occupying lesions Acanthamoeba spp.
Influenza virus (ANE) Balamuthia mandrillaris
Mycoplasma pneumoniae Mycobacterium tuberculosis
Varicella-zoster virus Toxoplasma gondiia
West Nile virus Fungal infections
Anti-NMDAR encephalitis White matter (different Balamuthia mandrillaris
Corpus callosum West Nile virus than ADEM) Baylisascaris procyonis
Epstein-Barr virus Borrelia burgdorferi
HPeV-3 (especially neonate)
Frontal lobe Enteroviruses
HIV
Herpes simplex virus 1
JC virusa
Herpes simplex virus 2
Measles virus (SSPE)
Treponema pallidum
West Nile virus
Spinal cord Arboviruses Cytomegalovirusa
Enteroviruses
Gnathostoma spp.
Rabies virus
Temporal lobeunilateral Enteroviruses
Herpes simplex virus 1
La Crosse virus
Treponema pallidum
Temporal lobebilateral Enteroviruses
Herpes simplex virus 1
Human herpesviruses 6/7/8a
Treponema pallidum
Anti-NMDAR encephalitis
Anti-VGKC encephalitis
Thalamus Arboviruses
Epstein-Barr virus
Influenza virus (ANE)

a
Causes encephalitis in immunocompromised and/or neonate host.

Scheld_Ch06.indd 102 2/21/14 5:31 PM


Chapter 6: Encephalitis 103

are associated with mesial temporal abnormalities, a recent


study suggested that early extratemporal involvement was
more likely in HSE (244). Rarely, HHV-6 infection of the CNS
has been associated with a syndrome similar to ANE (51,245).
Enteroviruses
Although poliovirus has almost completely disappeared in de-
veloped countries, nonpolio EVs are an important cause of dif-
fuse, generalized encephalitis. In such cases, neuroimaging is
typically normal. Certain EVs, however, may result in similar
clinical and radiographic manifestations as poliovirus. EV-71,
for example, is characterized by symmetric bilateral T2W
A hyperintensities in the dorsal brainstem, cerebellar dentate
nuclei, and anterior horns of the cervical spinal cord on MRI
(246,247) (Fig. 6.5).
As in many cases of acute encephalitis, DWI appears to be
more sensitive than other conventional sequences for detection
of EV-71 lesions (248). Echovirus 7-associated encephalomy-
elitis is also associated with a similar topographical distribu-
tion on MRI (249). Isolated reports of unilateral and bilateral
hippocampal lesions in the setting of acute enteroviral infec-
tion have also been described (250,251).
Arboviruses
Neuroimaging abnormalities in WNV often involve the deep
gray matter, although findings are highly variable and up to
half of patients may have normal brain MRI (233,252,253). In
those who develop a flaccid paralysis, spinal cord imaging may
demonstrate T2W hyperintensities in the anterior cord and
gadolinium enhancement of the cauda equina, conus medul-
laris, and nerve roots (252,254). Limited studies of other
B arboviruses, including SLE, EEE, TBE, and Murray Valley
encephalitis, suggest involvement of the substantia nigra and
FIGURE 6.4 Typical MRI findings in HSE on (A) coronal and (B) axial other deep gray structures, and the latter three viruses may
FLAIR sequences. also resemble WNV acute flaccid paralysis both clinically and
radiographically (255,256). The considerable overlap in the
neuroimaging spectrum of these arboviruses prevents discrimi-
nation between these etiologies based on imaging alone.
MR spectroscopic findings have also been described, although JEV is most commonly associated with T2W hyperintensi-
the clinical use of this imaging modality in the evaluation of ties on MRI in the thalami, with variable involvement of the
HSE remains undefined. Abnormalities, which typically in- substantia nigra, basal ganglia, brainstem, cerebellum, and
clude decreased NAA and increased choline, myoinositol, and cortex (257). Although the presence of thalamic abnormalities
lactate, are thought to represent loss of neuronal integrity and is highly sensitive for JEV in the appropriate clinical setting,
necrosis with accompanying macrophage infiltration and glio- their absence does not rule out JEV (258). Mesial temporal lobe
sis (233235). involvement has also been reported, although unlike herpes
In several case series of VZV encephalitis, the most notable
finding was the absence of brain imaging abnormalities in the
majority of patients. In a minority of patients, affected regions
included the temporal lobe and brainstem. A notable caveat is
that many patients in these series underwent CT scanning only
(32,236). VZV vasculopathy is often associated with T2W hy-
perintensities in the white matter and at the graywhite matter
junctions on MRI, with evidence of either ischemia or hemor-
rhage (25). The most commonly reported imaging abnormali-
ties in EBV-associated encephalitis include multiple, diffuse
hyperintensities on T2W and FLAIR imaging, often transient
and with involvement of the splenium of the corpus callosum
(237239).
HHV-6 associated limbic encephalitis typically involves
the mesial temporal lobe (hippocampus, entorhinal cortex,
and amygdala). Additional cortical and subcortical areas can
be involved and may be associated with specific clinical sub-
types (240,241). Involved areas are characterized by T2W
hyperintensity and variable degrees of enhancement. In addi-
tion, increased hippocampal uptake of glucose, suggestive of FIGURE 6.5 Axial T2 MRI demonstrating symmetric hyperintensity
increased metabolic activity, has been detected by FDG-PET in the medulla in EV-71 encephalitis. (Courtesy of Philip Britton, The
(242,243). Although both HSE and HHV-6 limbic encephalitis Childrens Hospital, Westmead, NSW, Australia.)

Scheld_Ch06.indd 103 2/21/14 5:31 PM


104 Part II: Viral Infections and Related Disorders

encephalitis, the insula and anterior portion of the temporal Mumps


lobe are usually spared, while additional thalamic lesions are
typically present (259,260). In JEV cases with biphasic illness, Encephalomyelitis following mumps is associated with white
regression of prior lesions and the appearance of new lesions matter changes on brain MRI and a longitudinally extensive
in typical areas are described (261,262). In cases of dengue myelopathy on spine imaging that may be indistinguishable
encephalopathy, focal MRI abnormalities in a number of from ADEM; rarely, hemorrhagic white matter lesions have
brain areas have been found. In addition, generalized cerebral been reported (276278).
edema has been reported and may be related to the prominent Influenza
metabolic disturbances observed in affected individuals (263).
Bilateral frontoparietal white matter lesions with restricted Neuroimaging findings in influenza-associated encephalopathy/
diffusion, suggestive of subcortical strokes, have been reported encephalitis are diverse. MRI can be normal or may demonstrate
in two cases of Chikungunya encephalitis (264). However, in diffuse brain edema or symmetric involvement of the thalami
another case series of children with various CNS manifesta- (233,279,280). Severe cases of encephalitis are characterized by
tions including encephalitis, MRI abnormalities were only T2W hyperintensities, hemorrhage, and restricted diffusion in
rarely observed (265). the thalami, basal ganglia, and cerebellum, consistent with ANE
(281). Milder cases may be associated with reversible lesions of
Rabies the splenium of the corpus callosum (282). Notably, imaging
abnormalities caused by the 2009 H1N1 virus do not appear
The often rapid clinical course of rabies encephalitis presents to differ significantly from other influenza strains (283285).
a challenge in obtaining neuroimaging. The limited available
data demonstrate diffuse T2W hyperintensities affecting both Mycobacterium tuberculosis
the deep and cortical gray matter of the brain and spinal cord
in both the paralytic and encephalitic forms of rabies (266). Cranial TB can present as tuberculous meningitis (TBM), tuber-
Although abnormalities may also be seen in the white mat- culomas, or abscesseseach of which have differing neuroim-
ter, the predominant gray matter involvement distinguishes aging characteristics. TBM is associated with basal meningeal
rabies encephalitis from ADEM. Initially nonenhancing, the enhancement, often accompanied by hydrocephalus, strokes
gray matter lesions may demonstrate gadolinium enhancement in the basal ganglia and internal capsule, and a focal or diffuse
once the patient becomes comatose (267). pachymeningitis (286288). Intracranial tuberculomas consisting
of granulomatous tissue may have variable appearance on T2W
imaging depending on the pathologic stage of the lesion; nonca-
Nipah seating tuberculomas are T2W hyperintense, whereas those with
In the acute phase, Nipah encephalitis is associated with small, solid caseation are isointense or hypointense. When central lique-
punctate T2W hyperintensities in the subcortical white mat- faction ensues, tuberculomas may take on an appearance more
ter, often restricting diffusion (268). This imaging evidence of similar to that of an abscess. Such lesions exhibit T2W hyperin-
microinfarction is congruent with the main pathologic find- tensity surrounded by a peripheral rim of hypointensity and dem-
ings of vasculitis-associated thrombosis and presence of Nipah onstrate peripheral rim enhancement on contrast imaging (289).
virus antigen in endothelial and smooth muscle cells of blood
vessels (269). For the subset of patients who suffer from a Fungal Infections
relapse of neurologic symptoms, MRI shows patchy areas of Neuroimaging findings in the setting of CNS fungal infections
confluent cortical involvement on MRI and focal hypoperfu- are often nonspecific and may be mistaken for TBM, pyogenic
sion on SPECT imaging (270). abscess, or brain tumor (290,291). Fungal infections may result
in basilar meningitis, hydrocephalus, vasculitis, or abscess, and
Hendra any combination of these may be present on neuroimaging. In
In contrast to Nipah virus, the closely related Hendra virus immunocompetent individuals, fungal abscesses are hypoin-
is associated predominantly with gray matter abnormalities tense on T1W imaging and hyperintense on T2W imaging, with
in the setting of meningoencephalitis. In three reported cases, a well-defined rim enhancement on postcontrast images, simi-
T2W hyperintense lesions involved the cortical gray matter, lar to pyogenic or tubercular abscesses (290). Recent progress
with relative sparing of the deep white matter tracts and cer- has been made in distinguishing fungal abscesses from other
ebellum. Cortical lesions may be confluent or multifocal, and abscesses. In one study, all of the fungal abscesses showed intra-
restriction of diffusion can be seen on DWI (271). cavitary projections directed centrally from the wall, a finding
not seen in the other abscess types. Overall, a ring-enhancing
lesion with irregular walls and nonenhancing intracavitary pro-
Measles jections was likely to be a fungal abscess (292). Detection of the
Measles virus can cause differing CNS syndromes, including disaccharide trehalose on MR spectroscopy may also distin-
acute measles encephalitis and SSPE. Neuroimaging in acute guish fungal from nonfungal abscesses (292294).
measles encephalitis typically demonstrates bilateral, sym- MRI is frequently normal in the setting of cryptococcal menin-
metric T2W hyperintense lesions involving the white matter gitis, although evidence of a basilar meningitis and/or hydroceph-
and deep gray matter structures in a pattern that can resemble alus indistinguishable from TBM may be observed. The adjacent
ADEM (272). Cortical gyral swelling may also be present in brain parenchyma may also be affected, giving rise to cryptococ-
the acute setting, and hemorrhage may develop in gray matter comas, most commonly seen in the midbrain and basal ganglia.
areas over days to weeks (273). These lesions are of variable density on CT scan and, on MRI, are
The earliest stages of SSPE are often not characterized by hypointense on T1W imaging and hyperintense on T2W imag-
abnormalities on conventional MRI, although FDG-PET can ing. Contrast enhancement is variable and more commonly seen
demonstrate cortical hypometabolism and MR spectroscopy in immunocompetent individuals. Unlike pyogenic abscesses,
may show decreased NAA levels (274). As the disease pro- cryptococcomas typically do not demonstrate restricted diffusion
gresses, T2W hyperintensities in the subcortical and periven- on DWI. MR spectroscopy demonstrates elevated lactate and
tricular white matter can develop, as does brain atrophy. By decreased NAA, choline, and creatine (26,290,295).
the late stages of SSPE, widespread changes in the cortex, Imaging abnormalities in patients with CNS coccidioido-
brainstem, and cerebellum can be seen (275). mycosis may reflect strokes, granulomas, and white matter

Scheld_Ch06.indd 104 2/21/14 5:31 PM


Chapter 6: Encephalitis 105

matter infarcts, or T2W hyperintensity in the mesial temporal


lobe (301,302).

Acute Disseminated Encephalomyelitis


MRI findings in ADEM typically include multiple, asymmetri-
cally distributed areas of T2W hyperintensity in the subcorti-
cal and deep white matter (Fig. 6.7).
Lesions in the deep gray matter, which are characteristic
of ADEM but atypical for multiple sclerosis and other demy-
elinating disorders, often have a symmetric appearance. In
approximately one third of cases, the cerebral cortex is also
involved. Infratentorial involvement occurs in over half of
cases, with lesions seen in the brainstem, middle cerebellar
peduncles, and cerebellar white matter (303305). The size
and morphology of lesions is highly variable, although they
tend to have poorer margination as compared to lesions in
other demyelinating diseases. In most cases, lesions appear si-
multaneously, and all lesions will exhibit a similar degree of
FIGURE 6.6 Listeria rhombencephalitis. Sagittal FLAIR MRI demon- contrast enhancement. Restricted diffusion may be observed
strates hyperintensity in the medulla. on DWI sequences in the acute stage, potentially representing
swelling of myelin sheaths, myelin vacuolation, or infiltration
of inflammatory cells with concomitant free radical produc-
tion (306,307). Rare variants of ADEM include hemorrhagic,
disease, in addition to basilar meningitis and hydrocephalus.
necrotizing, and relapsing forms (308).
Ischemia has been reported in over half of cases and typically
results in deep cerebral infarcts seen as areas of hyperintensity AntiN-Methyl-D-Aspartate Receptor Encephalitis
on T2W imaging. Less commonly, focal enhancing lesions in
the white matter or deep grey matter representing granulomas One third to one half of patients may have abnormalities on
are observed (292,296,297). brain MRI. Affected areas, seen as T2W or FLAIR hyperin-
tensities, include the mesial temporal lobes, periventricular
Listeria monocytogenes white matter, cortex, cerebellum, brainstem, or basal gan-
glia. Notably, on follow-up, many of these abnormalities
Rhombencephalitis in the setting of L. monocytogenes infec- improve, although atrophy may develop. Less than 20% of
tion is characterized by the presence of multiple, small, medul- affected individuals will have evidence of contrast enhance-
lary T2W hyperintensities that demonstrate rim enhancement ment (168,309).
on T1W imaging, reflecting a combination of microabscesses
and associated edema in the lower brainstem (298) (Fig. 6.6). AntiVoltage-Gated Potassium Channel Receptor
Encephalitis
Borrelia Species
The spectrum of diseases associated with antibodies to the
Acute CNS Lyme disease is associated with small T2W hyper- VGKC complex has been steadily growing. In the setting of
intense and T1W isointense lesions in the subcortical white encephalitis, the most common finding is unilateral or bilat-
matter, which may mimic those seen in multiple sclerosis eral T2W hyperintensity in the mesial temporal lobe (169)
(299). Multifocal lesions may also be observed in the brain- (Fig. 6.8).
stem and spinal cord (300). Recent reports have suggested additional or exclusive in-
volvement of the basal ganglia (310,311).
Treponema Pallidum
The clinical and MRI features of neurosyphilis are highly vari-
able. The MRI of patients with clinical encephalitis is charac-
terized by diffuse brain atrophy, evidence of subcortical white

A B

FIGURE 6.7 MRI abnormalities in ADEM. (A) Axial FLAIR shows


multiple subcortical T2 hyperintensities in both hemispheres. (B) T1 FIGURE 6.8 Autoimmune limbic encephalitis. Coronal FLAIR MRI
post gadolinium demonstrates incomplete rings of enhancement sur- demonstrates T2 hyperintensities in the bilateral mesial temporal lobes
rounding the lesions. (solid arrow) and cingulate gyrus (dashed arrow).

Scheld_Ch06.indd 105 2/21/14 5:31 PM


106 Part II: Viral Infections and Related Disorders

MANAGEMENT have an almost 100% fatality rate. Two of the most well-
studied viral causes of encephalitis are HSV-1 and EV. HSV-1
Clinicians should focus first on treatable and common causes encephalitis has been reported to have a worse prognosis than
of encephalitis. Empirical treatment for bacterial meningitis EV, with greater than 35% of patients with HSE suffering se-
(vancomycin plus a third-generation cephalosporin) should be vere sequelae or death (4). Although EV encephalitis patients
started because the clinical presentation may overlap with en- often have a good outcome, EV-71 is associated with fatali-
cephalitis. Therapy with ampicillin should also be considered ties and neurologic sequelae (58). Other viral etiologies are
when the demographics (i.e., older individuals), clinical pre- less well studied, with information limited to case reports and
sentation (i.e., rhombencephalitis), or CSF profile is suggestive small series. Persistent neurologic deficits after EBV encepha-
of Listeria infection. Antiviral therapy is generally restricted litis are reported to be rare (316). Influenza has been asso-
to treatment of herpesviruses (especially HSV-1 and VZV) and ciated with a severe type of encephalitis with high mortality
the unusual instance of HIV infection (312). Therapy with particularly in Japan and Taiwan, including several case re-
acyclovir should be started and continued until HSV-1 has ports of ANE (317,318). In the United States, cases appear to
been reasonably excluded as a diagnosis, which may require be less severe with better outcomes, although further studies
testing serial CSF samples (see prior discussion). Therapy for are needed (319).
M. tuberculosis or fungal meningitis should be initiated when Deaths and complications caused by arboviruses are bet-
clinical and laboratory testing is compatible. If rickettsial or ter documented as a result of comprehensive reporting to
Ehrlichia infections are suspected, doxycycline should be initi- the CDC. WNV infections are less severe in children than in
ated empirically. adults; children accounted for only 4% of WNND reported
For IAE, oseltamivir may be beneficial (313). Corticosteroids to the CDC from 1999 to 2007, with 63% of cases older than
or intravenous immune globulin may also be helpful in some 10 years of age and only 15% younger than 4 years of age.
IAE cases to combat the proposed hyperintense cytokine re- There were only three pediatric fatalities over this time period
sponse. There is no evidence that treatment of presumed CNS (1% of all cases of WNND), a case fatality rate substantially
Mycoplasma infection alters outcome. lower than for older adults (14% for adults 50 years of age or
In addition to directed therapy, aggressive supportive care older) (320). In a study of 127 children with LACV, 12% had
is critical, and minimizing secondary brain injury should be neurologic deficits at discharge (321). EEE is also known to
made a high priority (314). Seizures, status epilepticus, and ce- have a higher mortality (almost 30%) and potentially severe
rebral edema are important complications of encephalitis and neurologic complications.
encephalopathy and should be monitored closely in patients Bacterial infections also have variable outcomes. Both L.
who are not improving. An elevated CSF opening pressure monocytogenes and M. tuberculosis may have relatively high
may serve as a harbinger for impending complications. Repeat morbidity and mortality (109,322). In a French encephalitis
neuroimaging to monitor for cerebral edema is particularly study, these two etiologies accounted for the majority of fa-
important in comatose patients. Typical indicators of elevated talities due to encephalitis (together, 12 of 26 fatalities) (101).
intracranial pressure, such as poorly reactive dilated pupils, On the other hand, infection with bacterial agents, such as
decorticate or decerebrate posture, or Cushing triad (systolic Bartonella spp., which typically causes encephalopathy rather
hypertension, bradycardia, and shallow respirations) are late than encephalitis, has an excellent outcome; over 90% of
findings. Patients should also be monitored for the syndrome patients recover completely without sequelae. There are lim-
of inappropriate secretion of antidiuretic hormone (SIADH). ited studies reporting outcomes specifically on encephalitis of
SIADH and extrapontine myelinolysis (EPM) are important unknown etiology. One study, however, reported significant
examples by which acquired metabolic derangements can both sequelae in up to 53% of survivors hospitalized with unknown
complicate, and mimic, encephalitis (315). causes of encephalitis (323).
Conditions that mimic infectious encephalitis should be
considered, particularly if no etiology is identified in the first
week of hospitalization. Metabolic and toxic disorders caus- SUMMARY
ing encephalopathy and seizures should be excluded. Anti-
NMDAR encephalitis is of particular importance given its It is unfortunate that in this era of modern medicine, so little
apparently high incidence; when identified, immunotherapy progress has been made in the field of encephalitis. An eti-
and removal of the tumor, if present, have been associated ology is only identified in about half of the cases and for
with improvement. those patients who receive a viral diagnosis; very few specific
antivirals are available for treatment. Given the significant
morbidity and mortality of this syndrome along with the fi-
OUTCOME nancial costs of hospitalization, rehabilitation, and sequelae,
research to address gaps in our understanding of this entity
In general, the prognosis of encephalitis is highly dependent are urgently needed as well as development of effective anti-
on the underlying cause. Rabies and N. fowleri, for instance, viral agents.

References
1. Glaser CA, Gilliam S, Schnurr D, et al. In search of encephalitis etiologies: 4. Raschilas F, Wolff M, Delatour F, et al. Outcome of and prognostic factors
diagnostic challenges in the California Encephalitis Project, 19982000. for herpes simplex encephalitis in adult patients: results of a multicenter
Clin Infect Dis. 2003;36(6):731742. study. Clin Infect Dis. 2002;35(3):254260.
2. Kolski H, Ford-Jones EL, Richardson S, et al. Etiology of acute childhood 5. Rautonen JM, Koskiniemi M, Vaheri A. Prognostic factors in childhood
encephalitis at the Hospital for Sick Children, Toronto, 19941995. Clin acute encephalitis. Pediatr Infect Dis. 1991;10(6):441446.
Infect Dis. 1998;26(2):398409. 6. Fowler A, Stdberg T, Eriksson M, et al. Childhood encephalitis in
3. Rantalaiho T, Frkkil M, Vaheri A, et al. Acute encephalitis from 1967 to Sweden: etiology, clinical presentation and outcome. Eur J Pediatr Neurol.
1991. J Neurol Sci. 2001;184(2):169177. 2008;12(6):484490.

Scheld_Ch06.indd 106 2/21/14 5:31 PM


Chapter 6: Encephalitis 107

7. Fowler A, Stdberg T, Eriksson M, et al. Long-term outcomes of acute 40. Doja A, Bitnun A, Jones EL, et al. Pediatric Epstein-Barr virus-associated
encephalitis in childhood. Pediatrics. 2010;126(4):e828835. encephalitis: 10-year review. J Child Neurol. 2006;21(5):385391.
8. Jmor F, Emsley HC, Fischer M, et al. The incidence of acute encephalitis syn- 41. Liaw SB, Shen EY. Alice in Wonderland syndrome as a presenting symptom
drome in Western industrialised and tropical countries. Virol J. 2008;5:134. of EBV infection. Pediatr Neurol.1991;7(6):464466.
9. Michael BD, Sidhu M, Stoeter D, et al. Acute central nervous system 42. Fujimoto H, Asaoka K, Imaizumi T, et al. Epstein-Barr virus infections of
infections in adultsa retrospective cohort study in the NHS North West the central nervous sytem. Intern Med. 2003;42(1):3340.
region. QJM. 2010;103(10):749758. 43. Hall CB, Long CE, Schnabel KC, et al. Human herpesvirus-6 infection in
10. Granerod H, Cunningham R, Zuckerman M, et al. Causality in acute children. A prospective study of complications and reactivation. N Engl
encephalitis: defining aetiologies. Epidemiol Infect. 2010;138(6):783800. Med. 1994;331(7):432438.
11. Davison KL, Crowcroft NS, Ramsay ME, et al. Viral encephalitis in 44. Zerr DM, Meier AS, Selke SS, et al. A population-based study of primary
England, 19891998: what did we miss? Emerg Infect Dis. 2003;9(2):234 human herpesvirus 6 infection. N Engl J Med. 2005;352(8):768776.
240. 45. de Ory F, Avelln A, Echevarra JE, et al. Viral infections of the central ner-
12. Rantala H, Uhari M, Tuokko H, et al. Poliovaccine virus in the cerebrospi- vous system in Spain: a prospective study. J Med Virol. 2013;85(3):554562.
nal fluid after oral polio vaccination. J Infect. 1989;19(2):173176. 46. Beaver H. The need is here. Detroit Dent Bull. 1969;38(2):16.
13. Cizman M, Jazbec J. Etiology of acute encephalitis in childhood in Slovenia. 47. Xu Y, Zhaori G, Vene S, et al. Viral etiology of acute childhood encephalitis
Pediatr Infect Dis J. 1993;12(11):903908. in Beijing diagnosed by analysis of single samples. Pediatr Infect Dis J.
14. Koskiniemi M, Korppi M, Mustonen K, et al. Epidemiology of en- 1996;15(11):10181024.
cephalitis in children: a prospective multicenter study. Eur J Pediatr. 48. Ward KN, Thiruchelvam AD, Couto-Parada X. Unexpected occasional per-
1997;156(7):541545. sistence of high levels of HHV-6 DNA in sera: detection of variants A and
15.Beghi E, Nicolosi A, Kurland LT, et al. Encephalitis and aseptic meningitis, B. J Med Virol. 2005;76(4):563570.
Olmsted County, Minnesota, 19501981: I. Epidemiology. Ann Neurol. 49. Virtanen JO, Frkkil M, Multanen J, et al. Evidence for human herpesvi-
1984;16(3):283294. rus 6 variant A antibodies in multiple sclerosis: diagnostic and therapeutic
16. Gubler DJ. The continuing spread of West Nile virus in the western hemi- implications. J Neurovirol. 2007;13(4):347352.
sphere. Clin Infect Dis. 2007;45(8):10391046. 50. Mannonen L, Herrgrd E, Valmari P, et al. Primary human herpesvirus-6
17. Thiboutot MM, Kannan S, Kawalekar OU, et al. Chikungunya: a poten- infection in the central nervous sytem can cause severe diasease. Pediatr
tially emerging epidemic? PLoS Negl Trop Dis. 2010;4(4):e623. Neurol. 2007;37(3):186191.
18. Tyler KL. Update on herpes simplex encephalitis. Rev Neurol Dis. 51. Crawford JR, Kadom N, Santi MR, et al. Human herpesvirus 6 rhomb-
2004;1(4):169178. encephalitis in immunocompetent children. J Child Neurol. 2007;22(11):
19. Corey L, Spear PG. Infections with herpes simplex viruses (2). N Engl Med. 12601268.
1986;314(12):749757. 52. Ward KN, Andrews NJ, Verity CM, et al. Human herpesviruses-6 and -7
20. Whitley RJ, Soong SJ, Linneman C Jr, et al. Herpes simplex encephalitis. each cause significant neurological morbidity in Britain and Ireland. Arch
Clinical assessment. JAMA. 1982;247(3):317320. Dis Child. 2005;90(6):619623.
21. Schleede L, Bueter W, Baumgartner-Sigl S, et al. Pediatric herpes simplex 53. Modlin JF, Dagan R, Berlin LE, et al. Focal encephalitis with enterovirus
virus encephalitis: a retrospective multicenter experience. J Child Neurol. infections. Pediatrics. 1991;88(4);841845.
2013;28(3):321331. 54. Fowlkes AL, Honarmand S, Glaser C, et al. Enterovirus-associated en-
22. Elbers JM, Bitnun A, Richardson SE, et al. A 12-year prospective study cephalitis in the California encephalitis project, 19982005. J Infect Dis.
of childhood herpes simplex encephalitis: is there a broader spectrum of 2008;198(11):16851691.
disease? Pediatrics. 2007;119(2):e399e407. 55. Ho M, Chen ER, Hsu KH, et al. An epidemic of enterovirus 71 infection
23. Mccabe K, Tyler K, Tanabe J. Diffusion-weighted MRI abnormali- in Taiwan. Taiwan Enterovirus Epidemic Working Group. N Engl J Med.
ties as a clue to the diagnosis of herpes simplex encephalitis. Neurology. 1999;341(13):929935.
2003;61(7):10151016. 56. Gilbert GL, Dickson KE, Waters MJ, et al. Outbreak of enterovirus 71
24. Livorsi D, Anderson E, Qureshi S, et al. Brainstem encephalitis: an unusual pre- infection in Victoria, Australia, with a high incidence of neurologic involve-
sentationof herpes simplex virus infection. J Neurol. 2010;257(9):14321437. ment. Pediatr Infect Dis. 1988;7(7):484488.
25. Nagel MA, Cohrs RJ, Mahalingam R, et al. The varicella zoster virus 57. Wang SM, Liu CC, Tseng HW, et al. Clinical spectrum of enterovirus 71
vasculopathies: clinical, CSF, imaging, and virologic features. Neurology. infection in children in southern Taiwan, with an emphasis on neurological
2008;70(11):853860. complications. Clin Infect Dis. 1999;29(1):184190.
26. Ho TL, Lee HJ, Lee KW, et al. Diffusion-weighted and conventional 58. Chang LY, Huan YC, Lin TY. Fulminant neurogenic pulmonary oedema
magnetic resonance imaging in cerebral cryptococcoma. Acta Radiol. with hand, foot, and mouth disease. Lancet. 1998;352(9125):367368.
2005;46(4):411414. 59. Lum LC, Wong KT, Lam SK, et al. Fatal enterovirus 71 encephalomyelitis.
27. Seward JF, Watson BM, Peterson CL, et al. Varicella disease after intro- J Pediatr. 1998;133(6):795798.
duction of varicella vaccine in the United States , 19952000. JAMA. 60. Chumakov M, Voroshilova M, Shindarov L, et al. Enterovirus 71 isolated
2002;287(5):606611. from cases of epidemic poliomyelitis-like disease in Bulgaria. Arch Virol.
28. Quist-Paulsen E, Kran AM, Dunlop O, et al. Infectious encephalitis: a de- 1979;60(34):329340.
scription of a Norwegian cohort. Scand J Infect Dis. 2013;45(3):179185. 61. Lin TY, Chang LY, Hsia SH, et al. The 1998 enterovirus 71 outbreak in Taiwan:
29. Granerod J, Ambrose HE, Davies NW, et al. Causes of encephalitis and dif- pathogenesis and management. Clin Infect Dis. 2002;34(suppl 2):S52S57.
ferences in their clinical presentations in England: a multicentre, population- 62. Kung CM, King CC, Lee CN, et al. Differences in replication capacity
based prospective study. Lancet Infect Dis. 2010;10(12):835844. between enterovirus 71 isolates obtained from patients with encephalitis
30. Kulkarni MA, Lecocq AC, Artsob H, et al. Epidemiology and aetiology of and those obtained from patients with herpangina in Taiwan. J Med Virol.
encephalitis in Canada, 19942008: a case of underdiagnosed arboviral 2007;79(1):6068.
agents? Epidemiol Infect. 2013;141(11):22432255. 63. Alam MM, Khurshid A, Shaukat S, et al. Identification of human parecho-
31. Lee TC, Tsai CP, Yuan CL, et al. Encephalitis in Taiwan: a prospective virus genotype, HPeV-12, in a paralytic child with diarrhea. J Clin Virol.
hospital-based study. Jpn J Infect Dis. 2003;56(56):193199. 2012;55(4):339342.
32. De Broucker T, Mailles A, Chabrier S, et al. Acute varicella zoster encephali- 64. Harvala H, Wolthers KC, Simmonds P. Parechoviruses in children: under-
tis without evidence of primary vasculopathy in a case-series of 20 patients. standing a new infection. Curr Opin Infect Dis. 2010;23(3):224230.
Clin Microbiol Infect. 2012;18(8):808819. 65. Selvarangan R, Nzabi M, Selvaraju SB, et al. Human parechovirus 3 caus-
33. Ibraheem M, Marin M, Leung J, et al. Fatal wild type varicella-zoster ing sepsis-like illness in children from midwestern United States. Pediatr
virus encephalitis without a rash in a vaccinated child. Pediatr Infect Dis J. Infect Dis. 2011;30(3):238242.
2013;32(2):183185. 66. Verboon-Maciolek MA, Krediet TG, Gerards LJ, et al. Severe neonatal
34. Koskiniemi M, Piiparinen H, Rantalaiho T, et al. Acute central nervous parechovirus infection and similarity with enterovirus infection. Pediatr
sytem complications in varicella zoster virus infections. J Clin Virol. Infect Dis J. 2008;27(3):241245.
2002;25(3):293301. 67. Centers for Disease Control and Prevention. Summary of notifiable diseases
35. Pollak L, Dovrat S, Book M, et al. Varicella zoster vs. herpes simplex United States, 2008. MMWR Morb Mortal Wkly Rep. 2010;57(54):194.
meningoencephalitis in the PCR era. A single center study. J Neurol Sci. 68. Jean CM, Honarmand S, Louie JK, et al. Risk factors for West Nile virus
2012;314(12):2936. neuroinvasive disease, California, 2005. Emerg Infect Dis. 2007;13(12):
36. Echevarra JM, Casas I, Tenorio A, et al. Detection of varicella-zoster virus- 19181920.
specific DNA sequences in cerebrospinal fluid from patients with acute asep- 69. Tu Y. The discovery of artemisinin (qinghaosu) and gifts from Chinese
tic meningitis and no cutaneous lesions. J Med Virol. 1994;43(4):331335. medicine. Nat Med. 2011;17(10):12171220.
37. Grose C, Henle W, Henle G, et al. Primary Epstein-Barr-virus infections in 70. Davis LE, DeBias R, Goade DE, et al. West Nile virus neuroinvasive disease.
acute neurologic diseases. N Engl J Med. 1975;292(8):392395. Ann Neurol. 2006;60(3):286300.
38. Connelly KP, DeWitt LD. Neurologic complications of infectious mono- 71. Sejvar JJ, Haddad MB, Tierney BC, et al. Neurologic manifestations and
nucleosis. Pediatr Neurol. 1994;10(3):181184. outcome of West Nile virus infection. JAMA. 2003;290(4):511515.
39. Husler M, Ramaekers VT, Doenges M, et al. Neurological complications 72. Centers for Disease Control and Prevention. West Nile virus and other
of acute and persistent Epstein-Barr virus infection in paediatric patients. J arboviral diseasesUnited States, 2012. MMWR Morb Mortal Wkly Rep.
Med Virol. 2002;68(2):253263. 2013;62(25):513517.

Scheld_Ch06.indd 107 2/21/14 5:31 PM


108 Part II: Viral Infections and Related Disorders

73. Reimann CA, Hayes EB, DiGuiseppi C, et al. Epidemiology of neuroin- 109. Armstrong RW, Fung PC. Brainstem encephalitis (rhombencephalitis)
vasive arboviral disease in the United States, 19992007. Am J Trop Med due to Listeria monocytogenes: case report and review. Clin Infect Dis.
Hyg. 2008;79(6):974979. 1993;16(5):689702.
74. Tyler KL. Emerging viral infections of the central nervous system: part 1. 110. Silpapojakul K, Ukkachoke C, Krisanapan S, et al. Rickettsial meningitis
Arch Neurol. 2009;66(8):939948. and encephalitis. Arch Intern Med. 1991;151(9):17531757.
75. Solomon T. Control of Japanese encephalitiswithin our grasp? N Engl J 111. Marrie TJ, Raoult D. Rickettsial infections of the central nervous sytem.
Med. 2006;355(9):869871. Semin Neurol. 1992;12(3):213224.
76. Parida M, Dash PK, Tripathi NK, et al. Japanese encephalitis outbreak, 112. Buckingham SC, Marshall GS, Schutze GE, et al. Clinical and laboratory
India, 2005. Emerg Infect Dis. 2006;12(9):14271430. features, hospital course, and outcome of Rocky Mountain spotted fever
77. Solomon T, Kneen R, Dung NM, et al. Poliomyelitis-like illness due to in children. J Pediatr. 2007;150(2):180184.
Japanese encephalitis virus. Lancet. 1998;351(9109):10941097. 113. Raab EL, Leopold IH, Hodes HL. Retinopathy in Rocky Mountain spot-
78. Mansfield KL, Johnson N, Phipps LP, et al. Tick-borne encephalitis virus ted fever. Am J Ophthalmol. 1969;68(1):4246.
a review of an emerging zoonosis. J Gen Virol. 2009;90(pt 8): 17811794. 114. Smith TW, Burton TC. The retinal manifestations of Rocky Mountain
79. Lindquist L, Vapalahti O. Tick-borne encephalitis. Lancet. 2008;371 spotted fever. Am J Ophthalmol. 1977;84(2):259262.
(9627):18611871. 115. Kelsey DS. Rocky Mountain spotted fever. Pediatr Clin North Am.
80. Amicizia D, Domnich A, Panatto D, et al. Epidemiology of tick-borne 1979;26(2):367376.
encephalitis (TBE) in Europe and its prevention by available vaccines. 116. Glaser C, Christie L, Bloch KC. Rickettsial and erhlichial infections.
Hum Vaccin Immunother. 2013;9(5):11631171. Handb Clin Neurol. 2010;96:143158.
81. Suss J. Tick-borne encephalitis in Europe and beyondthe epidemiological 117. Beaver PC. The nature of visceral larva migrans. J Parasitol. 1969;55(1):312.
situation as of 2007. Euro Surveill. 2008;13(26):pii=18916. 118. Kazacos KR, Boyce WM. Baylisascaris larva migrans. J Am Vet Med
82. Bogovic P, Lotric-Furlan S, Strle F. What tick-borne encephalitis may look Assoc. 1989;195(7):894903.
like: clinical signs and symptoms. Travel Med Infect Dis. 2010;8(4):246250. 119. Gavin PJ, Kazacos KR, Shulman ST. Baylisascariasis. Clin Microbiol Rev.
83. Bhatt S, Gething PW, Brady OJ, et al. The global distribution and burden 2005;18(4):703718.
of dengue. Nature. 2013;496(7446):504507. 120. Rusnak JM, Lucey DR. Clinical gnathostomiasis: case report and review
84. Arajo FM, Arajo MS, Nogueira RM, et al. Central nervous system in- of the English-language literature. Clin Infect Dis. 1993;16(1):3350.
volvement in dengue: a study in fatal cases from a dengue endemic area. 121. Moore DAJ, McCroddan J, Dekumyoy P, et al. Gnathostomiasis: an
Neurology. 2012;78(10):736742. emerging imported disease. Emerg Infect Dis. 2003;9(6):647650.
85. Chen WJ, Hwang KP, Fang AH. Detection of IgM antibodies from cere- 122. Ogata K, Nawa Y, Akahane H, et al. Short report: gnathostomiasis in
brospinal fluid and sera of dengue fever patients. Southeast Asian J Trop Mexico. Am J Trop Med Hyg. 1998;58(3):316318.
Med Public Health. 1991;22(4):659663. 123. Boongird P, Phuapradit P, Siridej N, et al. Neurological manifestations of
86. Miagostovich MP, Ramos RG, Nicol AF, et al. Retrospective study on gnathostomiasis. J Neurol Sci. 1977;31(2):279291.
dengue fatal cases. Clin Neuropathol. 1997;16(4):204208. 124. Herman JS, Chiodini PL. Gnathostomiasis, another emerging imported
87. Petersen BW, Repprecht CE. Human rabies epidemiology and diagnosis. In: disease. Clin Microbiol Rev. 2009;22(3);484492.
Tkachev S, ed. Non-Flavivirus Encephalitis. Winchester, United Kingdom: 125. Fuller AJ, Munckhof W, Ricrs L, et al. Eosinophilic meningitis due to
InTech; 2011. Angiostrongylus cantonensis. West J Med. 1993;159(1):7880.
88. Drr S, Nassengar S, Mindekem R, et al. Rabies diagnosis for developing 126. Yii CY, Chen CY, Chen ER, et al. Epidemiologic studies of eosinophilic
countries. PLoS Negl Trop Dis. 2008;2(3):e206. meningitis in southern Taiwan. Am J Trop Med Hyg. 1975;24(3):447454.
89. Johnson N, Fooks A, McColl K. Human rabies case with long incubation, 127. Slom TJ, Cortese MM, Gerber SI, et al. An outbreak of eosinophilic men-
Australia. Emerg Infect Dis. 2008;14(12):19501951. ingitis caused by Angiostrongylus cantonensis in travelers returning from
90. Peters C. Arenaviruses. In: Richman DD, Whitley RJ, Hayden FG, eds. the Carribean. N Engl J Med. 2002;346(9):668675.
Clinical Virology. Washington, DC: American Society of Microbiology 128. Kuberski T, Wallace GD. Clinical manifestations of eosinophilic meningitis
Press; 2009:10091029. due to Angiostrongylus cantonensis. Neurology. 1979;29(12):15661570.
91. Jahrling PB, Peters CJ. Lymphocytic choriomeningitis virus. A neglected 129. Punyagupta S, Juttijudata P, Bunnag T. Eosinophilic meningitis in Thailand.
pathogen of man. Arch Pathol Lab Med. 1992;116(5):486488. Clinical studies of 484 typical cases probably caused by Angiostrongylus
92. Baum SG, Lewis AM Jr, Rowe WP, et al. Epidemic nonmeningitic cantonensis. Am J Trop Med Hyg. 1975;24(6, pt 1):921931.
lymphocytic-choriomeningitis-virus infection. An outbreak in a popula- 130. Toma H, Matsumura S, Oshiro C, et al. Ocular angiostrongyliasis without
tion of laboratory personnel. N Engl J Med. 1966;274(17):934936. meningitis symptoms in Okinawa, Japan. J Parasitol. 2002;88(1):211213.
93. Lewis JM, Utz JP. Orchitis, parotitis and meningoencephalitis due to 131. Yoder JS, Straif-Bourgeois S, Roy SL, et al. Primary amebic meningoen-
lymphocytic choriomeningitis virus. N Engl J Med. 1961;265:776780. cephalitis deaths associated with sinus irrigation using contaminated tap
94. Mahalingam S, Herrero LJ, Playford EG, et al. Hendra virus: an emerging water. Clin Infect Dis. 2012;55(9):e79e85.
paramyxovirus in Australia. Lancet Infect Dis. 2012;12(10):799807. 132. Duma RJ, Helwig WB, Martinez AJ. Meningoencephalitis and brain ab-
95. Chew MH, Arquin PM, Shay DK, et al. Risk factors for Nipah virus scess due to a free-living amoeba. Ann Intern Med. 1978;88(4):468473.
infection among abattoir workers in Singapore. J Infect Dis. 2000; 133. Duma RJ, Rosenblum WI, McGehee RF, et al. Primary amoebic meningo-
181(5):17601763. encephalitis caused by Naegleria: two new cases, response to amphoteri-
96. Wong KT. Emerging epidemic viral encephalitides with a special focus on cin B, and a review. Ann Intern Med. 1971;74(6):923931.
henipaviruses. Acta Neuropathol. 2010;120(3):317325. 134. Martnez AJ. Is Acanthamoeba encephalitis an opportunistic infection?
97. Montgomery JM, Hossain MJ, Gurley E, et al. Risk factors for Nipah virus Neurology. 1980;30(6):567574.
encephalitis in Bangladesh. Emerg Infect Dis. 2008;14(10):15261532. 135. Martinez AJ, Janitschke K. Acanthamoeba, an opportunistic microorganism:
98. Garg RK. Subacute sclerosing panencephalitis. J Neurol. 2008;255 a review. Infection. 1985;13(6):251256.
(12):18611871. 136. Martinez AJ, Visvesvara GS. Free-living, amphizoic and opportunistic
99. Dyken PR. Subacute sclerosing panencephalitis. Current status. Neurol amebas. Brain Pathol. 1997;7(1):583598.
Clin. 1985;3(1):179196. 137. Martinez AJ. Free-living amebas: infection of the central nervous system.
100. Miki K, Komase K, Mgone CS, et al. Molecular analysis of measles virus Mt Sinai J Med. 1993;60(4):271278.
genome derived from SSPE and acute measles patients in Papua, New 138. Cha JH, Furie K, Walensky RP, et al. Case records of the Massachusetts
Guinea. J Med Virol. 2002;68(1):105112. General Hospital. Case 39-2006. A 24-year-old woman with systemic lupus
101. Mailles A, Stahl JP. Infectious encephalitis in France in 2007: a national erythematosus, seizures, and right arm weakness. N Engl J Med. 2006;
prospective study. Clin Infect Dis. 2009;49(12):18381847. 355(25):26782689.
102. Christie LJ, Loeffler AM, Honarmand S, et al. Diagnostic challenges of cen- 139. Karande SC, Lahiri KR, Sheth SS, et al. Acanthamoeba meningoen-
tral nervous sytem tuberculosis. Emerg Infect Dis. 2008;14(9):14731475. cephalitis complicating pyogenic meningitis. Indian Pediatr. 1991;28(7):
103. Painter J, Slutsker L. Listeriosis in humans. In: Ryser ET, Marth EH, eds. 794797.
Listeria, Listeriosis, and Food safety. 3rd ed. Boca Raton, FL: CRC Press 140. Sharma PP, Gupta P, Murali MV, et al. Primary amebic meningoencepha-
Taylor & Francis Group; 2007:85109. litis caused by Acanthamoeba: successfully treated with cotrimoxazole.
104. Pollock SS, Pollock TM, Harrison MJ. Infection of the central nervous Indian Pediatr. 1993;30(10):12191222.
system by Listeria monocytogenes: a review of 54 adult and juvenile cases. 141. Singhal T, Baipai A, Kalra V, et al. Successful treatment of Acanthamoeba
Q J Med. 1984;53(211):331340. meningitis with combination oral antimicrobials. Pediatr Infect Dis J.
105. Bouvet E, Suter F, Gibert C, et al. Severe meningitis due to Listeria monocyto- 2001;20(6):623627.
genes. A review of 40 cases in adults. Scand J Infect Dis. 1982;14(4):267270. 142. Schuster FL, Yagi S, Gavali S, et al. Under the radar: Balamuthia amebic
106. Peeters A, Waer M, Michielsen P, et al. Listeria monocytogenes meningitis. encephalitis. Clin Infect Dis. 2009;48(7):879887.
Clin Neurol Neurosurg. 1989;91(1):2936. 143. Centers for Disease Control and Prevention. Balamuthia amebic en-
107. Louria DB, Hensle T, Armstrong D, et al. Listeriosis complicating malig- cephalitisCalifornia, 19992007. MMWR Morb Mortal Wkly Rep.
nant disease. A new association. Ann Intern Med. 1967;67(2):260281. 2008;57(28):768771.
108. Cherubin CE, Marr JS, Sierra MF, et al. Listeria and gram-negative bacil- 144. Centers for Disease Control and Prevention. Notes from the field: trans-
lary meningitis in New York City, 19721979. Frequent causes of menin- plant-transmitted Balamuthia mandrillarisArizona, 2010. MMWR
gitis in adults. Am J Med. 1981;71(2):199209. Morb Mortal Wkly Rep. 2010;59(36):1182.

Scheld_Ch06.indd 108 2/21/14 5:31 PM


Chapter 6: Encephalitis 109

145. Centers for Disease Control and Prevention. Balamuthia mandrillaris is 175. Guarner J, Shieh WJ, Hunter S, et al. Clinicopathologic study and labora-
transmitted through organ transplantationMississippi, 2009. MMWR tory diagnosis of 23 cases with West Nile virus encephalomyelitis. Hum
Morb Mortal Wkly Rep. 2010;59(36):11651170. Pathol. 2004;35(8):983990.
146. Bakardijev A, Azimi PH, Ashouri N, et al. Amebic encephalitis caused 176. Sips GJ, Wilschut J, Smit JM. Neuroinvasive flavivirus infections. Rev
by Balamuthia mandrillaris: report of four cases. Pediatr Infect Dis J. Med Virol. 2012;22(2):6987.
2003;22(5):447453. 177. Johnson RT. Selective vulnerability of neural cells to viral infection. Brain.
147. Kodet R, Nohnkov E, Tich M, et al. Amebic encephalitis caused by 1980;103(3):447472.
Balamuthia mandrillaris in a Czech child: description of the first case from 178. Surtees R, DeSousa C. Influenza virus associated encephalopathy. Arch
Europe. Pathol Res Pract. 1998;194(6):423429. Dis Child. 2006;91(6):455456.
148. Duke BJ, Tyson RW, DeBiasi R, et al. Balamuthia mandrillaris menin- 179. Miller JQ, Price TR. The nervous system in Rocky Mountain spotted
goencephalitis presenting with acute hydrocephalus. Pediatr Neurosurg. fever. Neurology. 1972;22(6):561566.
1997;26(2):107111. 180. Miller JQ, Price TR. Involvement of the brain in Rocky Mountain spotted
149. Orozco LD, Khan MA, Fratkin JD, et al. Asymptomatic aneurysm of the cav- fever. South Med J. 1972;65(4):437439.
ernous and supraclinoid internal carotid artery in a patient with Balamuthia 181. Walker DH, Mattern WD. Rickettsial vasculitis. Am Heart J. 1980;100
mandrillaris encephalitis. J Clin Neurosci. 2011;18(8):11181120. (6, pt 1):896906.
150. Hill CP, Damodaran O, Walsh P, et al. Balamuthia amebic menin- 182. Martinez AJ, Nelson EC, Duma RJ. Animal model of human disease.
goencephalitis and mycotic aneurysms in an infant. Pediatr Neurol. Primary amebic meningoencephalitis, Naegleria meningoencephalitis,
2011;45(1):4548. CNS protozoal infection. Am J Pathol. 1973;73(2):545548.
151. Centers for Disease Control and Prevention. Emergence of Cryptococcus 183. Hamann KJ, Kephart GM, Kazacos KR, et al. Immunofluorescent local-
gattiiPacific Northwest, 20042010. MMWR Morb Mortal Wkly Rep. ization of eosinophil granule major basic protein in fatal human cases of
2010;59(28):865868. Baylisascaris procyonis infection. Am J Trop Med Hyg. 1989;40(3):291297.
152. Beersma MF, Dirven K, van Dam AP, et al. Evaluation of 12 commercial 184. Venkatesan A, Tunkel AR, Bloch KC, et al. Case definitions, diagnostic algo-
tests and the complement fixation test for Myccoplasma pneumoniae- rithms, and priorities in encephalitis: consensus statement of the International
specific immunoglobulin (IgG) and IgM antibodies, with PCR used as the Encephalitis Consortium. Clin Infect Dis. 2013;57(8):11141128.
gold standard. J Clin Microbiol. 2005;43(5):22772285. 185. Tunkel AR, Hartman BJ, Kaplan SL, et al. Practice guidelines for the man-
153. Arnold JC, Singh KK, Milder E, et al. Human metapneumovirus associ- agement of bacterial meningitis. Clin Infect Dis. 2004;39(9):12671284.
ated with central nervous system infection in children. Pediatr Infect Dis 186. Steele RW, Marmer DJ, OBrien MD, et al. Leukocyte survival in cerebro-
J. 2009;28(12):10571060. spinal fluid. J Clin Microbiol. 1986;23(5):965966.
154. Lynch M, Lee B, Azimi P, et al. Rotavirus and central nervous system 187. Negrini B, Kelleher KJ, Wald ER. Cerebrospinal fluid findings in aseptic
symptoms: cause or contaminant? Case reports and review. Clin Infect versus bacterial meningitis. Pediatrics. 2000;105(2):316319.
Dis. 2001;33(7):932938. 188. Feigin RD, Shackelford PG. Value of repeat lumbar puncture in the dif-
155. Biancheri R, Pessagno A, Baglietto MG, et al. Anti-N-methyl-D-aspartate- ferential diagnosis of meningitis. N Engl J Med. 1973;289(11):571574.
receptor encephalitis in a four-year-old girl. J Pediatr. 2010;156(2):332334. 189. Solomon T, Michael BD, Smith PE, et al. Management of suspected viral
156. Morishima T, Togashi T, Yokota S, et al. Encephalitis and encephalopa- encephalitis in adultsAssociation of British Neurologists and British
thy associated with an influenza epidemic in Japan. Clin Infect Dis. Infection Association National Guidelines. J Infect. 2012;64(4):347373.
2002;35(5):512517. 190. Callicott JH Jr, Nelson EC, Jones MM, et al. Meningoencephalitis
157. Maricich SM, Neul JL, Lotze TE, et al. Neurologic complications associ- due to pathogenic free-living amoebae. Report of two cases. JAMA.
ated with influenza A in children during the 20032004 influenza season 1968;206(3):579582.
in Houston, Texas. Pediatrics. 2004;114(5):e626e633. 191. Elbers J, Halliday W, Hawkins C, et al. Brain biopsy in children with
158. Khandaker G, Zurynski Y, Buttery J, et al. Neurologic complications of in- primary small-vessel central nervous system vasculitis. Ann Neurol.
fluenza A(H1N1)pdm09: surveillance in 6 pediatric hospitals. Neurology. 2010;68(5):602610.
2012;79(14):14741481. 192. Verstrepen WA, Bruynseels P, Mertens AH. Evaluation of a rapid real-time
159. Wada K, Nishiura H, Kawana A. An epidemiological analysis of severe RT-PCR assay for detection of enterovirus RNA in cerebrospinal fluid
cases of the influenza A (H1N1) 2009 virus infection in Japan. Influenza specimens. J Clin Virol. 2002;25(suppl 1):3943.
Other Respi Viruses. 2010;4(4):179186. 193. Gorgievski-Hrisoho M, Schumacher JD, Vilimonovic N, et al. Detection by
160. Glaser CA, Winter K, DuBray K, et al. A population-based study of neu- PCR of enteroviruses in cerebrospinal fluid during a summer outbreak of
rologic manifestations of severe influenza A(H1N1)pdm09 in California. aseptic meningitis in Switzerland. J Clin Microbiol. 1998;36(9):24082412.
Clin Infect Dis. 2012;55(4):514520. 194. Klapper PE, Cleator GM. European guidelines for diagnosis and man-
161. Murakami K, Tsukahara M, Tsuneoka H, et al. Cat scratch disease: agement of patients with suspected herpes simplex encephalitis. Clin
analysis of 130 seropositive cases. J Infect Chemother. 2002;8(4):349352. Microbiol Infect. 1998;4(4):178180.
162. McGrath N, Wallis W, Ellis-Pegler R, et al. Neuroretinitis and encephalop- 195. Gilden D. Varicella zoster virus and central nervous system syndromes.
athy due to Bartonella henselae infection. Aust N Z Med. 1997;27(4):454. Herpes. 2004;11(suppl 2):89A94A.
163. Banwell B, Kennedy J, Sadovnick D, et al. Incidence of acquired demyelin- 196. Gregoire SM, van Pesch V, Goffette S, et al. Polymerase chain reaction
ation of the CNS in Canadian children. Neurology. 2009;72(3):232239. analysis and oligoclonal antibody in the cerebrospinal fluid from 34 pa-
164. Leake JA, Albani S, Kao AS, et al. Acute disseminated encephalomyeli- tients with varicella-zoster virus infection of the nervous system. J Neurol
tis in childhood: epidemiologic, clinical and laboratory features. Pediatr Neurosurg Psychiatry. 2006;77(8):938942.
Infect Dis J. 2004;23(8):756764. 197. Nagel MA, Forghani B, Mahalingam R, et al. The value of detecting anti-
165. Tenembaum S, Chitnis T, Ness J, et al. Acute disseminated encephalomy- VZV IgG antibody in CSF to diagnose VZV vasculopathy. Neurology.
elitis. Neurology. 2007;68(16)(suppl 2):S23S36. 2007;68(13):10691073.
166. Lin CH, Jeng JS, Hsieh ST, et al. Acute disseminated encephalomyelitis: 198. Denne C, Kleines M, Dieckhfer A, et al. Intrathecal synthesis of anti-viral
a follow-up study in Taiwan. J Neurol Neurosurg Psychiatry. 2007; antibodies in pediatric patients. Eur J Paediatr Neurol. 2007;11(1):2934.
78(2):162167. 199. Weinberg A, Li S, Palmer M, et al. Quantitative CSF PCR in Epstein-Barr
167. Gable MS, Sheriff H, Dalmau J, et al. The frequency of autoimmune virus infections of the central nervous system. Ann Neurol. 2002;52(5):
N-methyl-D-aspartate receptor encephalitis surpasses that of individual 543548.
viral etiologies in young individuals enrolled in the California Encephalitis 200. Weinberg A, Bloch KC, Li S, et al. Dual infections of the central nervous
Project. Clin Infect Dis. 2012;54(7):899904. system with Epstein-Barr virus. J Infect Dis. 2005;191(2):234237.
168. Dalmau J, Gleichman AJ, Hughes EG, et al. Anti-NMDA-receptor 201. Luppi M, Barozzi P, Maiorana A, et al. Human herpesvirus-6: a survey
encephalitis: case series and analysis of the effects of antibodies. Lancet of presence and distribution of genomic sequences in normal brain and
Neurol. 2008;7(12):10911098. neuroglial tumors. J Med Virol. 1995;47(1):105111.
169. Vincent A, Buckley C, Schott JM, et al. Potassium channel antibody- 202. Prez-Vlez CM, Anderson MS, Robinson CC, et al. Outbreak of neuro-
associated encephalopathy: a potentially immunotherapy-responsive form logic enterovirus type 71 disease: a diagnostic challenge. Clin Infect Dis.
of limbic encephalitis. Brain. 2004;127(pt 3):701712. 2007;45(8):950957.
170. Graus F, Dalmau J. Paraneoplastic neurological syndromes. Curr Opin 203. Landry ML, Fonseca SN, Cohen S, et al. Fatal enterovirus type 71 in-
Neurol. 2012;25(6):795801. fection: rapid detection and diagnostic pitfalls. Pediatr Infect Dis J.
171. Liu TH, Liang LC, Wang CC, et al. The blood-brain barrier in the ce- 1995;14(12):10951100.
rebrum is the initial site for the Japanese encephalitis virus entering the 204. Lanciotti RS, Kerst AJ, Nasci RS, et al. Rapid detection of West Nile virus
central nervous system. J Neurovirol. 2008;14(6):514521. from human clinical specimens, field-collected mosquitoes, and avian
172. Johnson RT. Virus in the nervous system. Ann Neurol. 1988;23(suppl):S210. samples by a TaqMan reverse transcriptase-PCR assay. J Clin Microbiol.
173. Johnson RT, Burke DS, Elwell M, et al. Japanese encephalitis: immunocy- 2000;38(11):40664071.
tochemical studies of viral antigen and inflammatory cells in fatal cases. 205. Solomon T, Ooi MH, Beasley DW, et al. West Nile encephalitis. BMJ.
Ann Neurol. 1985;18(5):567573. 2003;326(7394):865869.
174. Esiri MM. Herpes simplex encephalitis. An immunohistological study 206. Roehrig JT, Nash D, Maldin B, et al. Persistence of virus-reactive serum
of the distribution of viral antigen within the brain. J Neurol Sci. 1982; immunoglobulin M antibody in confirmed West Nile virus encephalitis
54(2):209226. cases. Emerg Infect Dis. 2003;9(3):376379.

Scheld_Ch06.indd 109 2/21/14 5:31 PM


110 Part II: Viral Infections and Related Disorders

207. Martin DA, Noga A, Kosoy O, et al. Evaluation of a diagnostic algorithm 238. Shian WJ, Chi CS. Epstein-Barr virus encephalitis and encephalomyelitis:
using immunoglobulin M enzyme-linked immunosorbent assay to differ- MR findings. Pediatr Radiol. 1996;26(9):690693.
entiate human West Nile virus and St. Louis encephalitis virus infections 239. Di Carlo P, Trizzino M, Titone L, et al. Unusual MRI findings in an im-
during the 2002 West Nile virus epidemic in the United States. Clin Diagn munocompetent patient with EBV encephalitis: a case report. BMC Med
Lab Immunol. 2004;11(6):11301133. Imaging. 2011;11:6.
208. Calisher CH. Medically important arboviruses of the United States and 240. Provenzale JM, vanLandingham KE, Lewis DV, et al. Extrahippocampal
Canada. Clin Microbiol Rev. 1994;7(1):89116. involvement in human herpesvirus 6 encephalitis depicted at MR imaging.
209. Noah DL, Drenzek CL, Smith JS, et al. Epidemiology of human rabies in Radiology. 2008;249(3):955963.
the United States, 1980 to 1996. Ann Intern Med. 1998;128(11):922930. 241. Provenzale JM, vanLandingham K, White LE. Clinical and imaging find-
210. Pai M, Flores LL, Pai N, et al. Diagnostic accuracy of nucleic acid ampli- ings suggesting human herpesvirus 6 encephalitis. Pediatr Neurol. 2010;
fication tests for tuberculous meningitis: a systematic review and meta- 42(1):3239.
analysis. Lancet Infect Dis. 2003;3(10):633643. 242. Seeley WW, Marty FM, Holmes TM, et al. Post-transplant acute limbic
211. Wheat LJ, Musial CE, Jenny-Avital E. Diagnosis and management of cen- encephalitis: clinical features and relationship to HHV6. Neurology.
tral nervous system histoplasmosis. Clin Infect Dis. 2005;40(6):844852. 2007;69(2):156165.
212. Paddock CD, Greer PW, Ferebee TL, et al. Hidden mortality attributable 243. Wainwright MS, Martin PL, Morse RP, et al. Human herpesvirus 6 lim-
to Rocky Mountain spotted fever: immunohistochemical detection of fatal, bic encephalitis after stem cell transplantation. Ann Neurol. 2001;50(5):
serologically unconfirmed disease. J Infect Dis. 1999;179(6):14691476. 612619.
213. Intapan PM, Maleewong W, Sawanyawisuth K, et al. Evaluation of 244. Noguchi T, Yoshiura T, Hiwatashi A, et al. CT and MRI findings of
human IgG subclass antibodies in the serodiagnosis of angiostrongyliasis. herpesvirus 6-associated encephalopathy: a comparison with findings of
Parasitol Res. 2003;89(6):425429. herpes simplex virus encephalitis. AJR Am J Roentgenol. 2010;194(3):
214. Steiner I, Budka H, Chaudhuri A, et al. Viral encephalitis: a review of 754760.
diagnostic methods and guidelines for management. Eur J Neurol. 245. Matsumoto H, Hatanaka D, Ogura Y, et al. Severe human herpesvirus
2005;12(5):331343. 6-associated encephalopathy in three children: analysis of cytokine pro-
215. Schroth G, Kretzschmar K, Gawehn J, et al. Advantage of magnetic reso- files and the carnitine palmitoyltransferase 2 gene. Pediatr Infect Dis.
nance imaging in the diagnosis of cerebral infections. Neuroradiology. 2011;30(11):9991001.
1987;29(2):120126. 246. Shen WC, Chiu HH, Chow KC, et al. MR imaging findings of enterovi-
216. Marchbank ND, Howlett DC, Sallomi DF, et al. Magnetic resonance ral encephalomyelitis: an outbreak in Taiwan. AJNR Am J Neuroradiol.
imaging is preferred in diagnosing suspected cerebral infections. BMJ. 1999;20(10):18891895.
2000;320(7228):187188. 247. Nolan MA, Craig ME, Lahra MM, et al. Survival after pulmonary edema
217. Kiroglu Y, Cali C, Yunten N, et al. Diffusion-weighted MR imaging of due to enterovirus 71 encephalitis. Neurology. 2003;60(10):16511656.
viral encephalitis. Neuroradiology. 2006;48(12):875880. 248. Lian ZY, Huang B, He SR, et al. Diffusion-weighted imaging in the diag-
218. Tsuchiya K, Katase S, Yoshino A, et al. Diffusion-weighted MR imaging nosis of enterovirus 71 encephalitis. Acta Radiol. 2012;53(2):208213.
of encephalitis. AJR Am J Roentgenol. 1999;173(4):10971099. 249. Lum LC, Chua KB, McMinn PC, et al. Echovirus 7 associated encephalo-
219. Gupta RK, Soni N, Kumar S, et al. Imaging of central nervous system viral myelitis. J Clin Virol. 2002;23(3):153160.
diseases. J Magn Reson Imaging. 2012;35(3):477491. 250. Liow K, Spanaki MV, Boyer RS, et al. Bilateral hippocampal encephalitis
220. Domingues RB, Fink MC, Tsanaclis AM, et al. Diagnosis of herpes sim- caused by enteroviral infection. Pediatr Neurol. 1999;21(5):836838.
plex encephalitis by magnetic resonance imaging and polymerase chain 251. Jang S, Suh SI, Ha SM, et al. Enterovirus 71-related encephalomyelitis:
reaction assay of cerebrospinal fluid. J Neurol Sci. 1998;157(2):148153. usual and unusual magnetic resonance imaging findings. Neuroradiology.
221. Schroth G, Gawehn J, Thron A, et al. Early diagnosis of herpes simplex 2012;54(3):239245.
encephalitis by MRI. Neurology. 1987;37(2):179183. 252. Petropoulou KA, Gordon SM, Prayson RA, et al. West Nile virus me-
222. Hollinger P, Matter L, Sturzenegger M. Normal MRI findings in herpes ningoencephalitis: MR imaging findings. AJNR Am J Neuroradiol. 2005;
simplex virus encephalitis. J Neurol. 2000;247(10):799801. 26(8):19861985.
223. Davis JM, Davis KR, Kleinman GM, et al. Computed tomography of her- 253. Ali M, Safriel Y, Sohi J, et al. West Nile virus infection: MR imaging find-
pes simplex encephalitis, with clinicopathological correlation. Radiology. ings in the nervous system. AJNR Am J Neuroradiol. 2005;26(2):289297.
1978;129(2):409417. 254. Kraushaar G, Patel R, Stoneham GW. West Nile virus: a case report with
224. Sener RN. Herpes simplex encephalitis: diffusion MR imaging findings. flaccid paralysis and cervical spinal cord: MR imaging findings. AJNR Am
Comput Med Imaging Graph. 2001;25(5):391397. J Neuroradiol. 2005;26(1):2629.
225. Kker W, Ngele T, Schmidt F, et al. Diffusion-weighted MRI in herpes simplex 255. Einsiedel L, Kat E, Ravindran J, et al. MR findings in Murray Valley en-
encephalitis: a report of three cases. Neuroradiology. 2004;46(2):122125. cephalitis. AJNR Am J Neuroradiol. 2003;24(7):13791382.
226. Smirniotopoulos JG, Murphy FM, Rushing EJ, et al. Patterns of contrast en- 256. Cerna F, Mehrad B, Luby JP, et al. St. Louis encephalitis and the substan-
hancement in the brain and meninges. Radiographics. 2007;27(2):525551. tia nigra: MR imaging evaluation. AJNR Am J Neuroradiol. 1999;20(7):
227. Tien RD, Dillon WP. Herpes trigeminal neuritis and rhombencepha- 12811283.
litis on Gd-DTPA-enhanced MR imaging. AJNR Am J Neuroradiol. 257. Kalita J, Misra UK, Pandey S, et al. A comparison of clinical and radio-
1990;11(2):413414. logical findings in adults and children with Japanese encephalitis. Arch
228. Gms H, Kumandas S, Per H, et al. Unusual presentation of herpes sim- Neurol. 2003;60(12):17601764.
plex virus encephalitis: bilateral thalamic involvement and normal imag- 258. Dung NM, Turtle L, Chong WK, et al. An evaluation of the usefulness
ing of early stage of the disease. Am J Emerg Med. 2007;25(1):8789. of neuroimaging for the diagnosis of Japanese encephalitis. J Neurol.
229. Wasay M, Mekan SF, Khelaeni B, et al. Extra temporal involvement in 2009;256(12):20522060.
herpes simplex encephalitis. Eur J Neurol. 2005;12(6):475479. 259. Handique SK, Das RR, Barman K, et al. Temporal lobe involvement in
230. Tan IL, McArthur JC, Venkatesan A, et al. Atypical manifestations and Japanese encephalitis: problems in differential diagnosis. AJNR Am J
poor outcome of herpes simplex encephalitis in the immunocompromised. Neuroradiol. 2006;27(5):10271031.
Neurology. 2012;79(21):21252132. 260. Basumatary LJ, Raja D, Bhuyan D, et al. Clinical and radiological spec-
231. Leonard JR, Moran CJ, Cross DT III, et al. MR imaging of herpes simplex trum of Japanese encephalitis. J Neurol Sci. 2013;325(12):1521.
type 1 encephalitis in infants and young children: a separate pattern of 261. Handique SK, Barkataky N. MR imaging in biphasic Japanese encephalitis.
findings. AJR AM J Roentgenol. 2000;174(6):16511655. AJNR Am J Neurol. 2008;29(3):E3.
232. Beattie GC, Glaser CA, Sheriff H, et al. Encephalitis with thalamic and 262. Pradhan S, Gupta RK, Singh MB, et al. Biphasic illness pattern due to early
basal ganglia abnormalities: etiologies, neuroimaging, and potential role relapse in Japanese-B virus encephalitis. J Neurol Sci. 2001;183(1):1318.
of respiratory viruses. Clin Infect Dis. 2013;56(6):825832. 263. Wasay M, Channa R, Jumani M, et al. Encephalitis and myelitis asso-
233. Menon DK, Sargentoni J, Peden CJ, et al. Proton MR spectroscopy in ciated with dengue viral infection: clinical and neuroimaging features.
herpes simplex encephalitis: assessment of neuronal loss. J Comput Assist Clin Neurol Neurosurg. 2008;110(6):635640.
Tomogr. 1990;14(3):449452. 264. Ganesan K, Diwan A, Shankar SK, et al. Chikungunya encephalomyelo-
234. Calli C, Ozel AA, Savas R, et al. Proton MR spectroscopy in the diag- radiculitis: report of 2 cases with neuroimaging and 1 case with autopsy
nosis and differentiation of encephalitis from other mimicking lesions. findings. AJNR Am J Neuroradiol. 2008;29(9):16361637.
J Neuroradiol. 2002;29(1);2328. 265. Robin S, Ramful D, Le Seach F, et al. Neurologic manifestations of pedi-
235. Takanashi J, Sugita K, Ishii M, et al. Longitudinal MR imaging and atric chikungunya infection. J Child Neurol. 2008;23(9):10281035.
proton MR spectroscopy in herpes simplex encephalitis. J Neurol Sci. 266. Laothamatas J, Hemachudha T, Mitrabhakdi E, et al. MR imaging in
1997;149(1):99102. human rabies. AJNR Am J Neuroradiol. 2003;24(6):11021109.
236. Pahud BA, Glaser CA, Dekker CL, et al. Varicella zoster disease of the 267. Laothamatas J, Sungkarat W, Hemachudha T. Neuroimaging in rabies.
central nervous system: epidemiological, clinical, and laboratory fea- Adv Virus Res. 2011;79:309327.
tures 10 years after the introduction of the varicella vaccine. J Infect Dis. 268. Sarji SA, Abdullah BJ, Goh KJ, et al. MR imaging features of Nipah
2011;203(3):316323. encephalitis. AJR Am J Roentgenol. 2000;175(2):437442.
237. Hagemann G, Mentzel HJ, Weisser H, et al. Multiple reversible MR 269. Wong KT, Shieh WJ, Kumar S, et al. Nipah virus infection: pathology
signal changes caused by Epstein-Barr virus encephalitis. AJNR AM J and pathogenesis of an emerging paramyxoviral zoonosis. Am J Pathol.
Neuroradiol. 2006;27(7):14471449. 2002;161(6):21532167.

Scheld_Ch06.indd 110 2/21/14 5:31 PM


Chapter 6: Encephalitis 111

270. Tan CT, Goh KJ, Wong KT, et al. Relapsed and late-onset Nipah encepha- 298. Lever N, Haas L. Serial MRI in listeria mesenrhombencephalitis: a case
litis. Ann Neurol. 2002;51(6):703708. report. J Neurol Neurosurg Psychiatry. 1995;59(5):524527.
271. Nakka P, Amos GJ, Saad N, et al. MRI findings in acute Hendra virus 299. Halperin JJ, Luft BJ, Anand AK, et al. Lyme neuroborreliosis: central
meningoencephalitis. Clin Radiol. 2012;67(5):420428. nervous system manifestations. Neurology. 1989;39(6):753759.
272. Baba Y, Tsuboi Y, Inoue H, et al. Acute measles encephalitis in adults. 300. Chang BL, Shih CM, Ro LS, et al. Acute neuroborreliosis with involve-
J Neurol. 2006;253(1):121124. ment of the central nervous system. J Neurol Sci. 2010;295(12):1015.
273. Lee KY, Cho WH, Kim SH, et al. Acute encephalitis associated with mea- 301. Nagappa M, Sinha S, Taly AB, et al. Neurosyphilis: MRI features and
sles: MRI features. Neuroradiology. 2003;45(2):100106. their phenotypic correlation in a cohort of 35 patients from a tertiary care
274. Seo YS, Kim HS, Jung DE. 18F-FDG PET and MRS of the early stages of university hospital. Neuroradiology. 2013;55(4):379388.
subacute sclerosing panencephalitis in a child with a normal initial MRI. 302. Brightbill TC, Ihmeidan IH, Post MJ, et al. Neurosyphilis in HIV-
Pediatr Radiol. 2010;40(11):18221825. positive and HIV-negative patients: neuroimaging findings. AJNR Am J
275. Cece H, Tokay L, Yildiz S, et al. Epidemiological findings and clinical and Neuroradiol. 1995;16(4):703711.
magnetic resonance presentations in subacute sclerosing panencephalitis. 303. Hynson JL, Kornberg AJ, Coleman LT, et al. Clinical and neuroradiologic
J Int Med Res. 2011;39(2):594602. features of acute disseminated encephalomyelitis in children. Neurology.
276. Sonmez FM, Odemis E, Ahmetoglu A, et al. Brainstem encephalitis and 2001;56(10):13081312.
acute disseminated encephalomyelitis following mumps. Pediatr Neurol. 304. Dale RC, de Sousa C, Chong WK, et al. Acute disseminated encephalomy-
2004;30(2):132134. elitis, multiphasic disseminated encephalomyelitis and multiple sclerosis
277. Unal A, Emre U, Atasoy HT, et al. Encephalomyelitis following mumps. in children. Brain. 2000;123(pt 12):24072422.
Spinal Cord. 2005;43(7):441444. 305. Baum PA, Barkovich AJ, Koch TK, et al. Deep gray matter involve-
278. Kumar RS, Kuruvilla A. Teaching neuroimages: acute hemorrhagic leuko- ment in children with acute disseminated encephalomyelitis. AJNR Am J
encephalitis after mumps. Neurology. 2009;73(20):e98. Neuroradiol. 1994;15(7):12751283.
279. Kimura S, Ohtuki N, Nezu A, et al. Clinical and radiological variability 306. Balasubramanya KS, Kovoor JM, Jayakumar PN, et al. Diffusion-
of influenza-related encephalopathy or encephalitis. Acta Paediatr Jpn. weighted imaging and proton MR spectroscopy in the characterization
1998;40(3):264270. of acute disseminated encephalomyelitis. Neuroradiology. 2007;49(2):
280. Amin R, Ford-Jones E, Richardson SE, et al. Acute childhood encephalitis 177183.
and encephalopathy associated with influenza: a prospective 11-year re- 307. Abou Zeid N, Pirko I, Erickson B, et al. Diffusion-weighted imaging
view. Pediatr Infect Dis J. 2008;27(5):390395. characteristics of biopsy-proven demyelinating brain lesions. Neurology.
281. Mizuguchi M. Acute necrotizing encephalopathy of childhood: a novel 2012;78(21):16551662.
form of acute encephalopathy prevalent in Japan and Taiwan. Brain Dev. 308. Rossi A. Imaging of acute disseminated encephalomyelitis. Neuroimaging
1997;19(2):8192. Clin N Am. 2008;18(1):149161.
282. Bulakbasi N, Kocaoglu M, Tayfun C, et al. Transient splenial lesion of 309. Florance NR, Davis RL, Lam C, et al. Anti-N-methyl-D-aspartate recep-
the corpus callosum in clinically mild influenza-associated encephalitis/ tor (NMDAR) encephalitis in children and adolescents. Ann Neurol.
encephalopathy. AJNR Am J Neuroradiol. 2006;27(9):19831986. 2009;66(1):1118.
283. Kedia S, Stroud B, Parsons J, et al. Pediatric neurological complications of 310. Hacohen Y, Wright S, Siddiqui A, et al. A clinico-radiological phenotype
2009 pandemic influenza A (H1N1). Arch Neurol. 2011;68(4):455462. of voltage-gated potassium channel complex antibody-mediated disor-
284. Kawashima H, Morichi S, Okumara A, et al. National survey of pan- der presenting with seizures and basal ganglia changes. Dev Med Child
demic influenza A (H1N1) 2009-associated encephalopathy in Japanese Neurol. 2012;54(12):11571159.
children. J Med Virol. 2012;84(8):11511156. 311. Jaychandran R, Patel MJ, Netravathi M, et al. Unusual clinical and
285. Surana P, Tang S, McDougall M, et al. Neurological complications of pan- imaging characteristics in a patient with voltage-gated potassium channel
demic influenza A H1N1 2009 infection: European case series and review. antibody-associated encephalitis. Neurol India. 2012;60(5):533534.
Eur J Pediatr. 2011;170(8):10071015. 312. Tunkel AR, Glaser CA, Bloch KC, et al. The management of encephalitis:
286. Gupta RK, Gupta S, Singh D, et al. MR imaging and angiography in clinical practice guidelines by the Infectious Diseases Society of America.
tuberculous meningitis. Neuroradiology. 1994;36(2):8792. Clin Infect Dis. 2008;47(3):303327.
287. Shukla R, Abbas A, Kumar P, et al. Evaluation of cerebral infarction in 313. Akins PT, Belko J, Uyeki TM, et al. H1N1 encephalitis with malignant
tuberculous meningitis by diffusion weighted imaging. J Infect. 2008; edema and review of neurologic complications from influenza. Neurocrit
57(4):298306. Care. 2010;13(3):396406.
288. Goyal M, Sharma A, Mishra NK, et al. Imaging appearance of pachyme- 314. Thakur KT, Motta M, Asemota AO, et al. Predictors of outcome in acute
ningeal tuberculosis. AJR Am J Roentgenol. 1997;169(5):14211424. encephalitis. Neurology. 2013;81(9):793800.
289. Gupta RK, Kumar S. Central nervous system tuberculosis. Neuroimaging 315. Martin RJ. Central pontine and extrapontine myelinolysis: the osmotic de-
Clin N Am. 2011;21(4):795814. myelination syndromes. J Neurol Neurosurg Psychiatry. 2004;75(suppl 3):
290. Jain KK, Mittal SK, Kumar S, et al. Imaging features of central nervous 2228.
system fungal infections. Neurol India. 2007;55(3):241250. 316. Caruso JM, Tung GA, Gascon GG, et al. Persistent preceding focal neu-
291. Kastrup O, Wanke I, Maschke M. Neuroimaging of infections of the cen- rologic deficits in children with chronic Epstein-Barr virus encephalitis.
tral nervous system. Semin Neurol. 2008;28(4):511522. J Child Neurol. 2000;15(12):791796.
292. Luthra G, Parihar A, Nath K, et al. Comparative evaluation of fungal, 317. Ormitti F, Ventura E, Summa A, et al. Acute necrotizing encephalopathy
tubercular, and pyogenic brain abscesses with conventional and diffusion in a child during the 2009 influenza A(H1N1) pandemia: MR imaging in
MR imaging and proton MR spectroscopy. AJNR Am J Neuroradiol. diagnosis and follow-up. AJNR Am J Neuroradiol. 2010;31(3):396400.
2007;28(7):13321338. 318. Wang GF, Li W, Li K. Acute encephalopathy and encephalitis caused by
293. Siegal JA, Cacayorinb ED, Nassif AS, et al. Cerebral mucormyco- influenza virus infection. Curr Opin Neurol. 2010;23(3):305311.
sis: proton MR spectroscopy and MR imaging. Magn Reson Imaging. 319. Drago L, Attia MW, DePiero A, et al. Influenza A-associated meningoen-
2000;18(7):915920. cephalitis. Pediatr Emerg Care. 2005;21(7):437439.
294. Himmelreich U, Dzendrowskyj TE, Allen C, et al. Cryptococcomas distin- 320. Lindsey NP, Staples JE, Lehman JA, et al. Surveillance for human West
guished from gliomas with MR spectroscopy: an experimental rat and cell Nile virus diseaseUnited States, 19992008. MMWR Surveill Summ.
culture study. Radiology. 2001;220(1):122128. 2010;59(2):117.
295. Chang L, Miller BL, McBride D, et al. Brain lesions in patients with AIDS: 321. McJunkin JE, de los Reyes EC, Irazuta JE, et al. La Crosse encephalitis in
H-1 MR spectroscopy. Radiology. 1995;197(2):525531. children. N Engl J Med. 2001;344(11):801807.
296. Wrobel CJ, Meyer S, Johnson RH, et al. MR findings in acute and 322. Kayaaslan BU, Akinci E, Bilen S, et al. Listerial rhombencephalitis in an
chronic coccidiodomycosis meningitis. AJNR Am J Neuroradiol. 1992; immunocompetent young adult. Int J Infect Dis. 2009;13(2):e65e67.
13(4):12411245. 323. Schmidt A, Bhler R, Mhlemann K, et al. Long-term outcome of acute
297. Erly WK, Bellon RJ, Seeger JF, et al. MR imaging of acute coccidioidal encephalitis of unknown aetiology in adults. Clin Microbiol Infect. 2011;
meningitis. AJNR Am J Neuroradiol. 1999;20(3):509514. 17(4):621626.

Scheld_Ch06.indd 111 2/21/14 5:31 PM


CHAPTER 7 POLIOMYELITIS, POLIO VACCINES,
AND THE POSTPOLIOMYELITIS SYNDROME
JOHN F. MODLIN AND DAVID J. COFFEY

Poliomyelitis is a systemic viral infection caused by any of three series of experiments in which they induced paralysis and spinal
human poliovirus serotypes that is characterized by acute men- cord lesions in a monkey by intraperitoneal injection of neural
ingitis and lytic infection of motor neurons in the brainstem and tissue from a fatal human case; shortly thereafter, Flexner and
spinal cord resulting in cranial nerve dysfunction and transient Lewis (10) at the Rockefeller Institute serially passed poliovi-
or permanent paresis of one or more extremities. In the United ruses in monkeys, fulfilling one of Robert Kochs postulates.
States and other developed countries, poliomyelitis due to nat- Serologic surveys in the 1930s and 1940s helped define the
urally occurring polioviruses has been eliminated by routine basis for the observed differences in the behavior of epidemics in
childhood immunization. Global eradication is a major prior- different locations and social conditions. Evidence of immunity
ity of the World Health Organization (WHO) and other major was found in 80% to 100% of adults tested, with generally
partners in the Global Poliomyelitis Eradication Initiative. lower levels in children (11). In time, it was shown that the
risk of infection was universal, but children living in poor sani-
tary conditions acquired infection at a much earlier age than
children from higher socioeconomic backgrounds, an observa-
A BRIEF HISTORY tion that explained the enigmatic shift during the early twen-
OF POLIOMYELITIS tieth century from an endemic disease involving only young
infants to an epidemic disease that also affected older children
Poliomyelitis has afflicted humans since antiquity (1). The and young adults (12). Later studies conducted by Paul and
first acknowledged description in English is a biographical Riordan (13) in a remote Eskimo population 15 to 20 years
narrative of the acute paralysis suffered by Sir Walter Scott following outbreak of poliomyelitis showed that immunity per-
in the second year of his life, circa 1772, and the first medi- sisted for at least two decades and perhaps for life.
cal description is recorded in the second edition of A Treatise The existence of more than one type of poliovirus was first
on Diseases of Children, published in 1789 by Michael inferred by Burnet and Macnamara (14) in 1931 when they dem-
Underwood, a London pediatrician and obstetrician (2,3). onstrated that monkeys who had recovered from infection with
Major contributions to understanding the disease were made a strain recovered in Melbourne subsequently developed disease
by the German orthopedist Heine, whose 1840 monograph when given the virulent MV strain and showed that the two
describes the clinical features of acute poliomyelitis in young strains differed qualitatively in in vitro neutralization tests. These
children (4), by the French anatomist Charcot who showed results were confirmed by Paul and Trask (11) at Yale and the
that paralysis was accompanied by a loss of motor nerve cells two virus groups were respectively designated Lansing-like, or
within the anterior horns of the cord (5), and by Karl Oskar Brunhilde-like, the latter in reference to the name of a labora-
Medin, a Swedish pediatrician who, in 1887, recorded the tory chimpanzee (1). A third type was predicted by the studies of
natural history of poliomyelitis and developed a classification Bodian (15), at Johns Hopkins in 1949. The existence of three
that became widely used for decades afterwards. The eponym types, and not more, was verified by the testing of poliovirus
Heine-Medin disease was widely used throughout Europe strains collected worldwide by a collaborative group organized
and North America during the early twentieth century. by the National Foundation for Infantile Paralysis (16).
Up to this time, only sporadic cases of poliomyelitis were The era of effective poliomyelitis treatment began in the
recognized. The first recorded outbreaks of poliomyelitis 1920s with the invention of the Drinker negative pressure ven-
occurred in the mid- and late nineteenth century in northern tilator or iron lung at the Harvard School of Public Health
Europe and later in North America. Charles Caverly, a (17) and its development at several Boston hospitals (18). The
Dartmouth College trained physician, described 132 cases with earliest devices were powered by vacuum cleaners and were
18 deaths in the Otter Creek Valley near Rutland, Vermont, fitted with boat portholes for access to the body of the patient.
the largest outbreak known to have occurred up to that time Progress in the management of the persistently paralyzed
in North America (6). These early outbreaks heralded larger patient was also made during this period. The practice of rigid
epidemics, including a countrywide epidemic of more than immobilization of even mildly involved extremities for long
1,000 cases in Sweden in 1905 (7) and 1,200 cases in New York periods gave way to the influential practices of Sister Elizabeth
City in 1907 (1), and the spread of disease throughout Europe Kenny using warm, moist packs and prolonged physical
and North America. The shift from endemic to epidemic dis- therapy (19).
ease has been ascribed to the improved hygienic conditions that In 1936, Sabin and Olitsky (20) produced the first un-
accompanied the increasing standards of living in the industrial- equivocal evidence that polioviruses could be grown in vitro
ized world. In theory, extensive exposure to polioviruses during when they propagated the MV poliovirus strain in human
infancy in the presence of passively acquired maternal antibody embryonic central nervous system (CNS) tissue, and a decade
results in harmless but immunizing infections. Conversely, later, Enders, Weller, and Robbins (21), succeeded in growing
infections delayed beyond infancy as a result of higher sanitary Lansing strain poliovirus in human embryonic tissues. This
standards and reduced opportunities for exposure are associ- achievement is widely acknowledged as the landmark event
ated with an increased risk of neurotropic infection (8). that opened the door to further characterization of the virus,
In 1908, Landsteiner and Popper (9) demonstrated that the better understanding of the pathophysiology of poliomyelitis,
etiologic agent of poliomyelitis was a filterable virus in a and development of successful vaccines.

112

Scheld_Ch07.indd 112 2/21/14 5:32 PM


Chapter 7: Poliomyelitis, Polio Vaccines, and the Postpoliomyelitis Syndrome 113

FIGURE 7.1 Incidence of poliomyelitis in the


United States, 1920 to 1960. (Data courtesy Dr.
Linda Quick, Centers for Disease Control and
Prevention.)

Building on these advances, Jonas Salk (22) developed the by health departments and medical societies. Trivalent OPV
first successful inactivated poliovirus vaccine (IPV), which was vaccine was introduced in 1964 and remained the principal
first administered to paralyzed patients in a home for crippled poliovirus vaccine used in the United States until 1997.
children in Pittsburgh in 1952. In 1954 to 1955, Salk IPV was Figure 7.1 illustrates the reported incidence of paralytic
successfully tested in a monumental controlled trial involving poliomyelitis cases in the United States from 1920 to 1960.
more than 1.8 million U.S. schoolchildren. In the meantime, A peak of 55,000 cases was reported in 1952. The introduc-
several investigators were studying live, attenuated polioviruses tion of IPV vaccine for general use in 1955 and OPV vaccine in
that could be delivered orally and induce mucosal as well as the early 1960s led to rapid and dramatic control of epidemic
systemic immunity (23). Live attenuated poliovirus strains were poliomyelitis, and the incidence of paralytic poliomyelitis fell
tested in humans in several countries and ultimately, three mon- from 13.9 cases per 100,000 in 1954 to less than 0.5 cases
ovalent strains developed by Sabin were licensed for use in the per 100,000 in 1965. By the 1970s, sustained transmission
United States in 1961 and 1962. Over the next 2 years, more of naturally occurring polioviruses ceased and the last case
than 100 million oral poliovirus (OPV) vaccine doses were dis- of domestically acquired poliomyelitis was reported in 1979
tributed in the United States via community programs organized (Fig. 7.2). Rare cases acquired in other countries continued to

FIGURE 7.2 Incidence of poliomyelitis


and vaccine-associated paralytic polio-
myelitis (VAPP) in the United States,
1961 to 2002. (Data courtesy Dr. Linda
Quick, Centers for Disease Control and
Prevention.)

Scheld_Ch07.indd 113 2/21/14 5:32 PM


114 Part II: Viral Infections and Related Disorders

be imported into the United States in the 1980s, but control of attenuated vaccine viruses replaced the epidemiologic niche
poliomyelitis in the Western Hemisphere by 1991 resulted in inhabited by the naturally occurring viruses. The very low
virtual disappearance of imported cases. Approximately 8 to levels of person-to-person transmission of naturally occurring
10 vaccine-associated paralytic poliomyelitis (VAPP) cases fol- polioviruses during the winter and spring months probably
lowing administration or transmission of OPV were observed abetted the disappearance of naturally occurring viruses and
until OPV vaccine was discontinued in favor of IPV in 1997 their replacement by OPV strains (32).
(24). Since then, only very rare poliomyelitis cases attributed
to imported VAPP or to acquisition of OPV vaccine-derived
polioviruses (VDPV) by immunodeficient persons have been Pathophysiology
reported in the United States (25,26).
Ingested polioviruses implant in the oropharynx and small
bowel and penetrate the mucosa via specialized microfold cells
VIROLOGY, TRANSMISSION, (M cells) and other epithelial cells overlying submucosal lym-
PATHOPHYSIOLOGY, phoid tissues (33). In primate models, the most efficient site of
viral replication is detected in Peyer patches in the intestinal
AND IMMUNITY submucosa (34,35). Spread to regional lymph nodes gives
rise to a transient and clinically silent minor viremia dur-
Virology ing which virus spreads to systemic reticuloendothelial tissue
including lymph nodes, bone marrow, liver, and spleen (36).
The polioviruses are prototypical human enteroviruses (species: For most poliovirus infections, viral replication is contained
Enterovirus C; family Picornaviridae). Three poliovirus sero- at this stage, resulting in subclinical infection. In a minority
types are distinguished from one another by in vitro neutral- of infections, further replication of virus in reticuloendothelial
ization with type-specific antisera. The icosahedral virion of tissues leads to a major viremia, which coincides with the
approximately 27 nm in diameter is composed of a protein onset of fever and other clinical symptoms (37).
capsid and an enclosed single-stranded RNA genome of ap- The path by which polioviruses reach the CNS remains
proximately 7,500 bases in length. The genome functions as a unsettled. It is well established that viremia precedes paraly-
monocistronic message with a single open reading frame coding sis, but the precise mechanism by which the virus breaches the
a 250 kDa polyprotein, which is subsequently cleaved by both bloodbrain barrier is not known (38,39). A study using trans-
viral coded and cellular proteases into capsid proteins, an RNA genic mice bearing the PVR gene found that spinal cord involve-
polymerase, proteases, and other regulatory proteins. The four ment can be blocked by sciatic nerve section after virus is injected
capsid proteins (VP1 through VP4) combine to form the 60 intramuscularly (40), suggesting that virus replication in skeletal
structural subunits that are assembled into the viral capsid. muscle precedes transport of virus to the cord via the peripheral
Like all picornaviruses, polioviruses exhibit substantial nerve (41,42), a concept consistent with the myotropic nature
genomic variability with up to 15% variation within the cap- of enteroviruses and the clinical observation of intense myalgias
sid coding region for each serotype. The three Sabin OPV preceding the onset of paralysis in affected patients.
vaccine strains differ from their naturally occurring parents Polioviruses are recoverable from the spinal cord for only
at fewer than 0.2% of positions across the full genome (27). the first several days of paralysis. The necrotic lesions and
For all three serotypes, analogous nucleotide substitutions in inflammatory infiltrates (Fig. 7.3C), which are distributed
the 5 noncoding region appear to be associated with dimin- within the gray matter of the anterior horn of the spinal cord,
ished ability to replicate in the gastrointestinal tract and with the motor nuclei of the pons and medulla, and occasionally
diminished neurovirulence. Attenuating mutations also map other locations (Fig. 7.3A and B), may persist for months (43).
to capsid proteins for each of the individual serotypes. The severity of clinical paralysis depends more on the intensity
All three poliovirus serotypes share a common cell mem- of the lesions, rather than their distribution.
brane receptor (poliovirus receptor [PVR]), a 67-kDa mem-
ber of the immunoglobulin superfamily coded on human
chromosome 19 (28,29). PVR-transgenic mice express PVR Immunity
in CNS and muscle tissue, endowing susceptibility to these tis-
sues but not on intestinal mucosal cells (30). Immunity to poliovirus infection is type specific; cross pro-
tection between the three serotypes is low, if it exists at all.
Reinfection with the same serotype occurs upon exposure to
Transmission of Polioviruses live poliovirus, regardless of whether prior immunity is based
on infection with naturally occurring polioviruses, infection
Before the introduction of poliovirus vaccines, naturally with live, attenuated viruses, or inactivated poliovirus vac-
occurring polioviruses circulated in temperate climates with cine administration. After household exposure to wild type
a marked seasonal variation resulting in peak activity from virus, 20% to 50% of naturally immune (31), 30% to 50% of
July to October and low levels from December to May. All OPV immune, and 90% to 100% of IPV immune persons are
three serotypes circulated concomitantly, although one or reinfected, as defined by virus excretion from the gastrointes-
two types often predominated within a defined community tinal tract or rise in antibody titer. Reinfections are universally
during a single season. Horizontal transmission in the com- asymptomatic and are rarely associated with oropharyngeal
munity occurred mostly among children 2 to 5 years of age, virus excretion.
and data from virus watch studies indicated that poliovirus Passively acquired poliovirus antibody protects against dis-
transmission was strongly associated with low socioeconomic ease, but not against infection (44). Preexisting humoral anti-
status and household contact. Once virus was introduced into body prevents or reduces oropharyngeal shedding of poliovirus
a household, 90% to 100% of susceptible persons became upon reinfection but has only a minor effect on fecal shed-
infected, along with 20% to 50% of seropositive persons (31). ding (38,4547). Serum-neutralizing antibodies are made in
Adults were only slightly less likely to become infected than response to natural infection and to immunization with either
young children. After the introduction of OPV vaccine, the OPV or IPV vaccine. Antibodies made in response to natural

Scheld_Ch07.indd 114 2/21/14 5:32 PM


Chapter 7: Poliomyelitis, Polio Vaccines, and the Postpoliomyelitis Syndrome 115

A B

FIGURE 7.3 A: T2-weighted axial image demonstrates abnormal,


increased signal intensity in the cerebral peduncles (arrow).
B: Pathologic section shows areas of necrosis (arrows) in the region
of the substantia nigra, corresponding to the areas of abnormal sig-
nal intensity in (A). C: Photomicrograph shows cystic necrosis sur-
rounded by an area of macrophage infiltration. (From Wasserstrom R,
Mamourian AC, McGary CT, et al. Bulbar poliomyelitis: MR findings
with pathologic correlation. Am J Neuroradiol. 1992;13:371373,
C with permission.)

infection appear to persist for life (48), and it is presumed that incubation period of 3 to 7 days and the major illness with
vaccine-induced protection is long term, if not lifelong. onset of symptoms generally 9 to 12 days after exposure (54).
Poliovirus secretory immunoglobulin A (IgA) appears in The minor illness, coinciding with viremia, consists of nonspe-
nasopharyngeal and intestinal secretions 1 to 3 weeks after nat- cific symptoms such as fever, headache, sore throat, anorexia,
ural infection or administration of OPV vaccine (49). Secretory and listlessness. Overall, 4% to 8% of infected persons expe-
IgA antibody persists for at least 5 to 6 years at low levels and, rience symptoms of the minor illness, and most resolve their
unlike humoral antibody, is not boosted significantly upon illness within 1 to 2 days without further symptoms.
rechallenge with OPV vaccine (50). The degree of protection The major illness is associated with CNS infection, which
conferred by local IgA antibody is relative. Upon rechallenge, has been variously estimated to occur in 0.1% to 1.0% of all
high secretory IgA titers inhibit virus replication, whereas poliovirus infections (8,55). About one third of young children
lower titers permit replication; virus shedding is dependent on who develop the major illness experience a biphasic illness
the challenge dose (46,51). Studies in the field show that child- with symptoms of the minor illness preceding onset of CNS
hood recipients of OPV are often reinfected and shed OPV disease; adults usually develop CNS disease without the pre-
virus in feces despite multiple prior OPV doses (52,53). ceding minor illness (37,56). The major illness is heralded by
the abrupt onset of fever, headache, vomiting, and meningis-
mus (Table 7.1). CSF pleocytosis is present at this early stage.
CLINICAL FEATURES Approximately one third of cases of CNS disease are limited
to meningitis without detectable motor neuron impairment,
OF POLIOMYELITIS which resolves within 5 to 10 days (i.e., nonparalytic polio-
myelitis).
Acute Illness Muscle weakness is preceded by intense myalgias of
the involved limb(s) and the axial skeleton. The pain may
More than 90% of naturally occurring poliovirus infections be relieved by exercise; patients may pace nervously in an
are inapparent. Acute clinical poliomyelitis is traditionally attempt to work off the pain. The hallmark of poliomy-
separated into two distinct phases: the minor illness with an elitis is asymmetric motor paresis, which ranges from mild

Scheld_Ch07.indd 115 2/21/14 5:32 PM


116 Part II: Viral Infections and Related Disorders

TA B L E 7 . 1 Overall mortality for spinal poliomyelitis is about 4% to


6%. During the poliomyelitis epidemics in the 1940s and
CLINICAL FEATURES OF ACUTE POLIOMYELITIS 1950s, bulbar polio caused a mortality of 20% to 60% when
respiratory assistance was required. With modern intensive
Minor illness care, deaths from temporary respiratory paralysis should be
Fever less common.
Headache
Sore throat
Major illness Differential Diagnosis
Abrupt onset of headache, vomiting, meningismus Sporadic cases of paralytic disease due to the nonpolio entero-
Aseptic meningitis viruses and West Nile virus is clinically indistinguishable from
Prodromal myalgias of involved muscles poliomyelitis. Only two nonpolio serotypes have been known
to cause epidemic disease: coxsackievirus A7 virus, which
Asymmetric motor weakness
has caused small outbreaks of paralytic disease in the for-
Complications mer Soviet Union (58), South Africa, and Scotland (59), and
Bulbar paralysis enterovirus 71, which has caused large outbreaks in Eastern
Respiratory failure Europe in the late 1970s and more recently in several countries
in Southeast Asia (6062).
Aspiration pneumonia
Poliomyelitis must also be distinguished from other causes
Myocarditis of acute paralysis including the Guillain-Barr syndrome,
Paralytic ileus and gastric dilatation which is associated with paralysis that is classically ascend-
Bladder ileus ing in nature, symmetric, and is accompanied by sensory
abnormalities in approximately 80% of cases. The CSF pleo-
cytosis that occurs during the major illness of poliomyelitis
is not characteristic of the Guillain-Barr syndrome, which is
associated with a normal CSF leukocyte count and an elevated
weakness of a single extremity to complete quadriplegia. CSF protein concentration. Other conditions produce acute
Proximal limb muscles are more involved than distal, and paralysis, including transverse myelitis, botulism, tick paraly-
legs are more commonly involved than arms. The deep ten- sis, epidural abscess, cord tumors, and hysteria; however, each
don reflexes, which were initially brisk, become absent. The of these diseases has features that readily separate them from
pace of development of the paresis ranges from several hours acute poliomyelitis.
to several days; most commonly occurring over 2 to 3 days.
Cranial nerve involvement (i.e., bulbar poliomyelitis)
occurs in 5% to 35% of paralytic cases. Any of the motor
cranial nerves can be involved, with the ninth and tenth being Risk Factors
the most common.
Although it was once believed that adults are more susceptible
to paralytic complications, there is probably little correlation
of age with severity of disease after the decline of maternal
Complications antibody (8,55). Studies during outbreaks suggest that infected
pregnant women have an increased risk of developing para-
Bulbar paralysis results in dysphagia, nasal speech, dyspnea, lytic disease (63,64). There is no evidence that either naturally
difficulty managing secretions, anxiety, and respiratory com- occurring polioviruses (65) or attenuated vaccine polioviruses
promise. Involvement of respiratory and vasomotor nuclei is (66) cause congenital defects.
less common but may portend serious complications due to Persons with B-cell immunodeficiency, primarily young
hypoventilation, blood pressure lability, and cardiac arrhyth- children with X-linked immunodeficiency syndromes, have
mias. Respiratory failure from paralysis of the diaphragm an increased risk of CNS disease when infected with either
and intercostal muscles represents the most serious compli- naturally occurring or attenuated vaccine polioviruses. These
cation of paralytic poliomyelitis. Aspiration pneumonia, pul- patients may develop acute paralysis or may have an atypical
monary edema, myocarditis, paralytic ileus, gastric dilation, course with an incubation period of several months, prolonged
and ileus of the bladder may also complicate acute paralytic febrile illness, chronic meningitis, and progressive neurologic
disease. dysfunction that includes both upper and lower motor neuron
involvement (6770).
Strenuous exercise during the early stages of the major ill-
Prognosis ness substantially increases the risk and severity of poliomy-
elitis. This effect has been well documented clinically (71,72);
Paralysis most often progresses for 1 to 3 days after onset, and experimental infections with other enteroviruses provide
rarely more than a week, halting about the time the patient supportive evidence (73).
becomes afebrile (57). Most patients with limb paresis experi- Paralytic poliomyelitis tends to localize in a limb that has
ence some recovery of function in the weeks to months after been the site of a recent intramuscular injection or injury
acute disease. An estimate of the eventual outcome can be (7476). Observations in experimental poliomyelitis (39,40)
made by a month after onset when most reversible weakness and after the administration of OPV vaccine (77) confirm this
will have resolved. Very little additional recovery of strength association but do not fully explain the mechanism of the
can be expected after a period of 9 months. Residual motor provoking effect. Tonsillectomy during the incubation period
deficits remain in about two thirds of initially paralyzed of poliomyelitis markedly increases the risk of bulbar disease
patients, ranging from minor debility to permanent, flaccid (78). The mechanism may be similar to the provoking effect of
paralysis. injections on spinal poliomyelitis.

Scheld_Ch07.indd 116 2/21/14 5:32 PM


Chapter 7: Poliomyelitis, Polio Vaccines, and the Postpoliomyelitis Syndrome 117

POSTPOLIOMYELITIS SYNDROME Epidemiology and Risk Factors


As many as 20% to 30% of patients who recover from para- Epidemiologic studies indicate that postpolio syndrome affects
lytic poliomyelitis experience new onset of muscle weakness, a substantial proportion of the 250,000 to 640,000 polio survi-
pain, atrophy, and fatigue many years after the acute illness. vors estimated to live in the United States (83,86,93). A cohort
With the eradication of acute poliomyelitis, the postpolio syn- study of 300 persons living in Olmsted County, Minnesota, who
drome remains the sole polio-related clinical disease encoun- had contracted paralytic poliomyelitis between 1935 and 1955
tered by physicians in all but a few polio-endemic nations. It is found that 32 (64%) of 50 poliomyelitis survivors had symp-
expected that the postpolio syndrome will ultimately decline in toms consistent with postpolio syndrome, and in another ret-
incidence and disappear as poliomyelitis is controlled around rospective study of Allegheny County, Pennsylvania, residents
the globe. with a history of paralytic poliomyelitis noted a prevalence of
28% (94,95). In the latter study, female gender and permanent
impairment sustained at the time of acute polio infection seemed
to convey a higher risk of developing postpolio syndrome.
History and Background of the However, the strongest risk factor for developing postpolio syn-
Postpoliomyelitis Syndrome drome was the interval from the original paralytic disease with
the incidence of postpolio syndrome peaking 30 to 34 years after
In 1875, Charcot (79) described a relationship between the original illness. Older age at the time of presentation with
acute poliomyelitis and late development of motor neuron postpolio syndrome has also been identified as a risk factor (86).
degeneration. By 1969, approximately 83 cases had been
described in which motor neuron degeneration occurred
decades after acute poliomyelitis (80), although the dis- Etiology
tinction between what we now describe as postpolio syn-
drome and other possible causes of late denervation, such Although the precise cause of postpolio syndrome remains
as amyotrophic lateral sclerosis (ALS), was not possible unknown, there is a consensus among authors that the cause
in many of these cases. The hypothesis was advanced that is probably related to exhaustion of motor units previously
the incidence of late motor neuron disease might increase overstressed by recovery from the acute denervation during
as children affected during the epidemics of the 1950s acute polio virus infection following which terminal elements
reached middle age (81,82). Initial concern that paralytic of surviving alpha motor neurons sprout to reinnervate adja-
poliomyelitis might increase the risk of subsequent ALS cent myofibrils. After acute poliomyelitis, fewer anterior horn
was allayed by careful epidemiologic studies, suggesting cells innervate a relatively larger number of myofibrils, thereby
that prior infection with poliomyelitis might actually pro- exaggerating the effects of the additional, physiologic cell loss
tect against the subsequent development of ALS (83). At at a later date attributed to aging (92,96).
present, it seems clear that the etiology and natural his- Other possible mechanisms have been considered, includ-
tory of the postpolio syndrome and ALS are quite differ- ing immunologic events and other virushost interactions.
ent. Postpolio syndrome is an indolent condition that rarely Prime among these alternative theories is the conjecture that
leads to severe disability or death (84). polioviruses can persist for many years within the CNS of
patients with postpolio syndrome and cause recrudescent dis-
ease by direct viral infection or immunopathologic pathways
Nomenclature (97). Evidence in support of this theory includes the demon-
stration of oligoclonal immunoglobulin G bands (84,98) or
Although the term postpolio syndrome has sometimes been poliovirus-specific immunoglobulin M bands (99) in CSF, and
applied to all neurologic symptoms occurring as late mani- the demonstration of poliovirus-like RNA sequences in CSF
festations of poliomyelitis, most authorities reserve this des- of postpolio syndrome patients by the polymerase chain reac-
ignation for a disorder characterized by new-onset muscle tion (100). However, the presence of CSF oligoclonal bands
weakness, fatigue, and pain, associated with loss of function has been inconsistent across studies (101,102), and virologists
that occurs years after the original acute poliomyelitis epi- debate the likelihood of infectious virus, or even viral RNA,
sode (8587). Another term, postpolio progressive muscular persisting for long periods in immunocompetent hosts.
atrophy (PPMA) refers specifically to the weakness with or
without atrophy that results from late denervation of muscles
of patients (8891). Criteria for PPMA include a reasonable Pathophysiology
past history of poliomyelitis with partial recovery of func-
tion and at least a decade of stabilization after recovery from Physical fatigue may result from damage to motor units in
the initial illness. PPMA is then the development of progres- previously affected muscle (103106), perhaps associated with
sive muscular weakness following these antecedent criteria. a change in acetylcholine receptor sensitivity and/or number.
It is worth noting that atrophy probably occurs in only half (This has a practical ramification for anesthesiologists who
or fewer of the patients who develop new weakness (92). may find it prudent to reduce the dose of paralytic agents for
Although the terms postpolio syndrome and PPMA are used patients with postpolio syndrome [107].) During exercise,
almost interchangeably, it should be recognized that postpo- there is decreased maximal voluntary contraction of muscles
lio syndrome or postpoliomyelitis sequelae may include other affected by postpolio syndrome, decreased tetanic force, and
symptoms such as fatigue (undoubtedly the most common of delayed recovery. Based on these and other data, Sharma et al.
all symptoms), subjective alterations in attention or cogni- (106) hypothesize that muscle fatigue in postpolio syndrome
tion, sleep disturbance, and alterations in pain perception. may be due to impaired activation beyond the muscle mem-
Bulbar symptoms, including dysphagia and some pulmonary brane at the level of excitationcontraction coupling. Based on
disturbances, can occur and may be more common and severe neuropathologic studies of patients with poliomyelitis in the
in those who had bulbar symptoms during their childhood 1940s and 1950s, another hypothesis suggests that the fatigue
illness. experienced by patients with postpolio syndrome has a central

Scheld_Ch07.indd 117 2/21/14 5:32 PM


118 Part II: Viral Infections and Related Disorders

because chronic residual weakness leads to abnormal joint


stresses. Scoliosis, poor mechanics, and abnormal postures
may all contribute. The degree of actual osteophytic change
may be less than one might anticipate given the amount of
residual weakness. Muscle pain may result from chronic
denervation or, if residual weakness is present, merely from
overuse of muscles. Secondary myopathy with elevation of
creatine kinase levels, usually of a mild degree, may occur in
some patients (95).
Postpoliomyelitis dysphagia, whether simply residual or
progressive could be a consequence of bulbar motor nerve
injury. Attention to the problems of dysphagia in patients with
postpolio syndrome was first drawn by Sonies and Dalakas
(113), who suggested that it was analogous to the postpolio
syndrome seen in the limbs, based on their series in which all
but 1 of the 32 patients studied had some abnormality in swal-
lowing function. Cinefluorography can demonstrate whether
there are impaired movements of the tongue, aspiration, or
pooling of material in the pyriform sinuses or valleculae.
Aspiration seems to be found only very infrequently and prob-
ably does not pose a severe risk in most instances (93,113).
Other symptoms that might be attributed to late-onset bulbar
dysfunction include the sleep-disordered breathing and chronic
alveolar hypoventilation that sometimes develop in patients
FIGURE 7.4 T2-weighted axial MRI scan demonstrating one of three with postpolio syndrome (114). Sleep-disordered breathing
hyperintense lesions (arrow) in the circum semiovale of a 42-year-old
may include obstructive sleep apnea syndrome and may occur
patient with postpoliomyelitis syndrome. (From Bruno RL, Cohen
JM, Galaski T, et al. The neuroanatomy of postpolio fatigue. Arch more commonly in patients with postpolio syndrome who suf-
Phys Med Rehabil. 1994;75:498504, with permission.) fered insult to respiratory control centers in the brainstem at
the time of their primary infection. Pulmonary function may
be further compromised if weakness of respiratory muscles,
impaired swallowing or cough mechanisms, chest wall defor-
origin. These studies showed lesions in specific areas of the
mity, or poor thoracoabdominal interaction is present (115).
brain whether or not clinical symptoms of encephalitis had
occurred, including the reticular formation in the brainstem,
vestibular nuclei, cerebellar nuclei, periaqueductal gray, hypo-
thalamic and thalamic nuclei, substantia nigra, locus ceruleus, Electrophysiology
and median raphe nuclei (43,107,108). The reticular activat-
ing system, responsible for maintaining normal alertness and Conventional electromyography (EMG) demonstrates chronic
attention, is composed of the brainstem reticular formation, denervation and occasionally reveals new or ongoing denerva-
posterior hypothalamus, and thalamus. Specific injuries to tion in the form of fasciculations, fibrillations, and positive
these structures from poliovirus infection could cause the acute sharp waves (87,91,116). Both symptomatic and asymptom-
disorientation and depression of consciousness seen in acute atic patients with postpolio syndrome show similar EMG
poliomyelitis and hypothetically could set the stage for late- findings. Similarly, the chronic denervation and reinnerva-
onset decompensation of arousal, attention, and subjective tion signs (including both enlarged and polyphasic motor
cognition in postpolio syndrome (109,110). It has been sug- unit potentials) and decreased interference patterns are seen
gested that T2-weighted hyperintense areas observed on brain whether or not the muscle being tested was originally weak
magnetic resonance imaging (MRI) scans could represent these (91,92,117). Enlarged motor units consistent with highly
areas of polio virusinduced damage (111) (Fig. 7.4). Injury to increased fiber density can be demonstrated in 90% of patients
the periaqueductal gray of the brainstem and substantia gela- with postpolio syndrome with single-fiber EMG (90). Jitter and
tinosa of the posterior horn of the spinal cord may explain the neuromuscular blocking effects are also noted, which correlate
hyperpathia that is reported by many polio survivors (111). with the interval since acute poliomyelitis. Like conventional
EMG, single-fiber EMG is unable to distinguish symptomatic
and asymptomatic patients with postpoliomyelitis. In con-
Clinical Features trast, macro-EMG may distinguish muscles with new weak-
ness, because previously increased amplitudes decline with
Fatigue, which may be the most distressing and disabling new weakness and atrophy (90).
symptom, is also the most common complaint, occurring in Although electrophysiologic studies in postpolio syn-
91% of polio survivors in national surveys (89,96,103,112). drome and PPMA have provided important pathophysiologic
Of these individuals, 41% experienced work limitation and information and supported the etiologic concept of overuse
25% had interference with self-care (111). Patients also report or accelerated aging of overcompensated motor units, the pri-
mental fatigue affecting attention and concentration (103). mary clinical role for nerve conduction and EMG studies is to
The most common neurologic sign of postpolio syndrome exclude other causes for the patients complaints.
is new weakness, which may be accompanied by atrophy and
pain. Weakness occurs in those muscles previously weakened
during acute paralytic poliomyelitis or in muscles thought to Psychologic Issues
be previously unaffected and may increase with exertion. The
pain may be perceived in muscles or joints and can occur in Although the cardinal manifestations of postpolio syndrome
the presence or absence of weakness. Joint pain may occur are neither caused nor modified by psychopathologic or

Scheld_Ch07.indd 118 2/21/14 5:32 PM


Chapter 7: Poliomyelitis, Polio Vaccines, and the Postpoliomyelitis Syndrome 119

personality factors, patients with postpolio syndrome may be OPV-immunized children, whereas mean titers to type 2 are
subject to characteristic psychologic symptoms (110,116,118). equivalent. Detectable antibody persists at protective levels
Bruno et al. (112) argue that anxiety, depression, and com- for at least 5 years, although mean titers decline considerably
pulsive behavior occur frequently in polio survivors, resulting (128). Vaccine efficacy for IPV distributed in the 1960s was
from the experience of a disabling disease in childhood from estimated to be 91% and 95% for 3 and 4 doses, respectively.
which recovery was achieved only by extreme effort (118). There are few data regarding vaccine efficacy of the current
Success in obtaining independence and in overcoming a dis- IPV formulations. A casecontrol study in Senegal indicated
ability was often accomplished by adopting very compulsive protection rates of 36% and 89% for recipients of one and
behavior and demanding very high standards of themselves. two doses, respectively (129). The efficacy for three doses is
One of the goals that many individuals set was to overcome assumed to be higher.
the need for assistive devices such as braces, crutches, wheel- IPV-immunized children develop little or no measurable
chairs, and the like. In consequence, polio survivors achieved secretory antibody (49). When challenged with live poliovi-
higher than expected life goals. ruses, IPV-immunized children shed the challenge virus in their
feces at a higher rate, higher titer, and for a longer period than
OPV-immunized children (51), indicating a greater potential
Effects of Exercise for asymptomatic infection and transmission of circulat-
ing polioviruses to unimmunized contacts. Although this is
Unfortunately, the avoidance of assistive devices at the time widely considered to be a disadvantage of IPV, there is strong
of acute weakness may be maladaptive in subsequently deal- evidence that widespread use of IPV results in protection that
ing with postpolio syndrome (112). Overtaxing motor units extends to unvaccinated persons in the community in devel-
with exercise may contribute to irreversible damage to mus- oped nations with good sanitation (130,131). IPV vaccine
cle fibers and more severe weakness (88,119,120). Patients may reduce community transmission of polioviruses due to
with postpolio syndrome may benefit from rest periods, in- reduction of pharyngeal shedding of poliovirus in reinfected
creased sleep time, and other energy-conservation methods IPV vaccine recipients, which may be the important mode of
to overcome fatigue (121). Compliance with appropriate community spread (47), although there is little effect of IPV
intervention to correct the overuse pattern appears to result vaccine in preventing spread within families in which fecal-
in improvement or resolution of new weakness and fatigue oral spread may be a more important mode of transmission
(122). However, because disuse can also enhance weakness, (132,133).
some moderation must be sought. Muscle strength may be im-
proved by nonfatiguing exercise (123). Specific protocols for
high-resistance exercise to improve isokinetic and isometric
strength for patients with postpolio syndrome have been de- Live, Attenuated Poliovirus Vaccine
scribed (124,125).
Live, attenuated trivalent (tOPV) vaccine remains the principal
vaccine used throughout the developing world. There are dif-
ferent manufacturers worldwide, but most follow a common
POLIOVIRUS VACCINES production method in which Sabin seed strains representing
each of the three poliovirus serotypes are individually grown
Inactivated Poliovirus Vaccine in monkey kidney cells and are combined for oral administra-
tion in concentrations of approximately 106.0 TCID50, 105.1
IPV vaccine is prepared by inactivation of poliovirus seed TCID50, and 105.8 TCID50 for poliovirus types 1, 2, and 3,
strains by formalin treatment for 12 to 14 days at 37C, the respectively. The unequal contribution of each type to the
method originally developed by Jonas Salk. However, con- trivalent preparation represents a balanced formulation
temporary IPV vaccines contain higher concentrations of designed to account for the more efficient replication of type
all three antigens compared with IPV vaccines introduced 2 OPV vaccine in the gastrointestinal tract (134). Because
in the 1950s. Three IPV formulations are now distributed type 2 virus regularly interferes with replication of types 1 and
in the United States: trivalent IPV (IPOL, Aventis Pasteur) 3, a primary series of three doses are routinely administered
and trivalent IPV combined with other recommended child- to enhance seroconversion to all three serotypes. Under con-
hood vaccines (Pediatrix, GlaxoSmithKline and Pentacel, ditions of good hygiene, seroconversion rates of 50%, 85%,
Sanofi Pasteur). Most available IPV vaccines are produced and 30% to serotypes 1, 2, and 3, respectively, are achieved
from wild type polioviruses grown in monkey kidney cells following the first tOPV dose (135), and a second and third
and contain 40-, 8-, and 32-D antigen units, respectively, for tOPV dose induce neutralizing antibodies to all three types in
poliovirus serotypes 1, 2, and 3. IPV formulations produced 86% and 96%, respectively (127,136). Serum antibody to all
from Sabin OPV vaccine strains are under development in three types persists in 84% to 98% of vaccinees 5 years after
China and elsewhere. The primary vaccination series for IPV primary immunization (137), although reexposure to vaccine
vaccine in the United States consists of four doses admin- viruses probably aids the maintenance of antibody levels in the
istered at 2 months, 4 months, 6 to 18 months, and 4 to population. Secretory IgA poliovirus antibody appears in oro-
6 years of age (126). Since 2000, IPV vaccine has been used pharyngeal and duodenal secretions 1 to 3 weeks after OPV
exclusively in the United States for routine immunization immunization (49) and persists for at least 5 to 6 years (138).
of infants against poliomyelitis and for all other recipients Challenge studies suggest that the intestinal immunity induced
including unimmunized adults, immunodeficient persons, by OPV vaccine is similar to intestinal immunity following
and anyone requiring polio vaccine boosters to travel to natural infection (139).
poliovirus endemic regions (126). The WHO Expanded Program on Immunization (EPI)
Neutralizing antibodies are detectable to all three poliovi- recommends one dose of tOPV at birth, a practice that
rus types in 99% of IPV vaccine recipients after two doses provides an opportunity to administer at least one dose of
and 100% after the third dose (127,128). A large boost vaccine to a child who may not present for routine health
in antibody titer follows the third dose (127,128). After maintenance care later, and three tOPV doses at 6, 10, and
three doses, mean titers to types 1 and 3 are higher than in 14 weeks of age (140). Even though passively acquired

Scheld_Ch07.indd 119 2/21/14 5:32 PM


120 Part II: Viral Infections and Related Disorders

maternal antibody to polioviruses present in the infants


circulation and in breast milk reduces vaccine virus repli- GLOBAL CONTROL OF
cation in the gastrointestinal tract and therefore blunts the POLIOMYELITIS
immune response in some infants, infants who receive OPV
at birth are more likely to have antibody to all three polio- With some notable exceptions (158), paralytic poliomyeli-
virus types at 4 months of age (141). Seroconversion rates tis affects mostly children between the ages of 6 months and
to tOPV are lower in many tropical countries compared to 2 years in developing countries, and most cases are caused by
seroconversion rates in developed countries (142144). Poor type 1 poliovirus. Lameness surveys of school-aged children in
responses to tOPV have contributed to outbreaks of polio- more than 20 developing nations revealed lower limb paralysis
myelitis in Oman, Israel, and Brazil despite relatively high prevalence rates of 2 to 11 per 1,000 in the 1960s and 1970s,
immunization rates existing before the appearance of epi- prevalence rates that equal or exceed those of the peak epi-
demic disease (145147). Although the reasons for the lower demic years in the United States (159,160). Currently, OPV
potency of tOPV vaccine in tropical areas remain incom- vaccine is used almost exclusively in underdeveloped nations
pletely understood, concurrent diarrheal disease at the time because of lower cost, ease of administration, enhanced
of immunization is an important factor (148,149). mucosal immunity, and enhancement of population immunity
Monovalent type 1 (mOPV1), monovalent type 3 (mOPV3), through transmission of vaccine viruses from immunized chil-
and bivalent type 1 and type 3 (bOPV) were added to the dren to nonimmune contacts.
Global Polio Eradication Program in 2006 and 2009, respec- Following the successful eradication of smallpox, an
tively to enhance seroconversion rates to type 1 and type 3 international conference held in Bellagio, Italy in 1983
OPV in the absence of the type 2 vaccine virus (150). Studies articulated the possibility of poliomyelitis eradication based
in Egypt and South Africa have confirmed the superior immu- on the unique epidemiology of poliovirus infections and the
nogenicity of the monovalent vaccines compared to tOPV in widespread availability of an inexpensive oral vaccine (tOPV)
resource poor settings (151,152). (161,162). By 1991, the Pan American Health Organization
Nonimmune OPV recipients shed vaccine viruses in the succeeded in eradicating polio in the Americas, and in 1988,
feces for 1 to 6 weeks and from the oropharynx for 1 to the World Health Assembly resolved to eradicate polio glob-
3 weeks. The spread of OPV viruses to unimmunized children ally by the year 2000 (163165). The Global Poliomyelitis
is an advantage in areas in which immunization levels are low. Eradication Initiative (GPEI) is a consortium of international
For example, a seroprevalence study in Houston and Detroit partners led by the WHO (159,166). The principal strategies
found that 11% to 42% of 11- to 35-month-old children pos- employed by the GPEI include enhancement of routine infant
sessed poliovirus neutralization antibodies, despite receiving tOPV immunization, the conduct of large scale supplementary
no prior OPV vaccine (153). immunization activities (SIAs) targeting all children younger
than 5 years of age, surveillance for poliomyelitis cases
through identification of persons with acute flaccid paralysis
(AFP), and maintenance of a global network of laboratories
Vaccine-Associated Paralytic Poliomyelitis capable of identification and characterization of polioviruses
in fecal specimens obtained from AFP cases. Seroconversion
The only serious adverse reaction associated with OPV is
rates during SIAs are higher than for routine immunization
the rare occurrence of VAPP. The incidence of VAPP has
(167), possibly because of spread of OPV or because they are
been estimated to be 2 to 4 cases per million individuals per
conducted during the dry season when diarrheal disease is less
year in countries using OPV (154). In industrialized coun-
prevalent. In the Americas, twice yearly mass campaigns were
tries, the relative frequency of paralysis associated with the
credited with rapid cessation of poliovirus circulation and dis-
first dose in the OPV series is about 10-fold higher than
appearance of disease (168,169).
with subsequent doses, whereas in developing countries,
Although there has been a reduction of more than 99% in
this ratio is lower, probably due to lower vaccine effective-
the burden of paralytic poliomyelitis and circulation of type 2
ness (155). OPV virus types 3 and 2 were most common
polioviruses ceased in 1999, the goal of complete eradication
causes of VAPP in the United States prior to discontinuation
has proven to be more difficult to attain than originally an-
of OPV use in 2000 (155). Approximately half of VAPP
ticipated (170172). Progress stalled in the last decade due to
cases are recent OPV vaccinees, most of whom develop
inability to stop transmission in some highly endemic areas,
paralysis 7 to 21 days after the first feeding of OPV. A
exportation from these areas to previously polio-free nations,
similar number of cases occur among parents, other family
social and cultural opposition to OPV immunization, civil
members, or other household contacts that develop paraly-
unrest, funding gaps, and the emergence of VDPV in areas
sis several weeks after the administration of OPV to a close
with low OPV coverage (173,174). Eventually, many of the
contact.
social, cultural, and political barriers were addressed, and in
Persons who have transient or hereditary B-cell immuno-
the mid-2000s, the deployment of monovalent and bivalent
deficiency, severe combined immunodeficiency syndrome, or
OPVs during SIAs led to improved seroconversion rates in
common variable immunodeficiency have an elevated risk of
many countries affected by low tOPV potency. These new
VAPP (69,156). For immunodeficient VAPP patients, interval
formulations are now credited with the eventual elimination
between the last OPV dose and onset of neurologic disease
of poliomyelitis in India and the marked reduction in case
is unusually long, with a typical range of 1 to 8 months, and
load in some particularly challenging areas in sub-Saharan
has been documented to be as long as 7 years (70). The illness
Africa (166).
may be protracted with chronic meningitis, progressive neu-
rologic dysfunction suggesting involvement of both upper and
lower motor neurons, progression of paralysis over several
weeks, and high mortality (67,68). Although fewer than 20% Vaccine-Derived Polioviruses
of surviving VAPP patients excrete polioviruses for longer
than 6 months (70), fecal excretion of virus has been esti- One unanticipated challenge to global poliomyelitis eradica-
mated to occur for as long as 18 years in one immunodeficient tion is the emergence of virulent polioviruses derived from
patient (157). OPV vaccine strains among underimmunized children living

Scheld_Ch07.indd 120 2/21/14 5:32 PM


Chapter 7: Poliomyelitis, Polio Vaccines, and the Postpoliomyelitis Syndrome 121

in certain economically deprived regions (175,176). A retro- IPV production, delivery, and storage, including development
spective laboratory investigation demonstrated previously of dose reduction by intradermal injection, development of
unrecognized type 2 VDPV isolates that circulated in Egypt new adjuvants, and combination of IPV with other vaccines
from approximately 1982 to 1993, and global surveillance routinely administered to infants (174).
since 2000 has uncovered more than 15 subsequent VDPV
outbreaks representing all three poliovirus serotypes (177). All
VDPV strains have been isolated from regions with low OPV SUMMARY
coverage, permitting VDPV to circulate, and all have acquired
biologic properties that are indistinguishable from naturally Epidemic paralytic poliomyelitis is a disease that has both
occurring wild type polioviruses, including neurovirulence in appeared and disappeared during the past 120 years. Most
monkeys and transgenic mice (178,179). medical practitioners in industrialized countries have never
The discovery of the potential for reintroduction of viru- seen a case of acute poliomyelitis but may encounter patients
lent polioviruses into previously polio-free areas via genera- who are now developing the symptoms of postpolio syndrome.
tion of VDPV has reinforced the necessity of maintaining high The clinical, epidemiologic, and scientific foundations for the
immunization levels in all polio-free regions and has created control of poliomyelitis were laid in the first half of the twen-
the necessity of discontinuing the use of all live, attenuated tieth century. Now, eradication has been achieved through-
poliovirus vaccines once eradication of naturally occurring out the developed world by routine immunization programs
poliovirus disease is assured (180,181). using two very effective vaccines, each of which possesses
unique advantages and disadvantages. In recent years, IPV has
become the preferred vaccine for developed countries because
OUTLOOK of the risk of rare VAPP cases associated with OPV.
Attention is now focused on the few remaining locations
As of 2013, renewed commitment to eradication has reduced where polio remains endemic. Despite setbacks, new strate-
the number of polio-endemic nations to threeNigeria, gies are being deployed and real progress has been made in
Afghanistan, and Pakistanand the annual global case count recent years. Hopefully, the goal of global poliomyelitis eradi-
to less than 300, and there is hope that the mission to eradicate cation will be achieved before the next edition of this text is
will soon be met. The polio endgame now calls for replace- published. The posteradication phase will require continued
ment of tOPV by bOPV once the threat from type 2 circulat- surveillance, maintenance of vaccine stockpiles for use should
ing vaccine-derived poliovirus (cVDPV) is diminished and the virulent polioviruses reemerge, and containment of labora-
ultimate discontinuation of all live OPV and a phased intro- tory stocks of naturally occurring and attenuated polioviruses
duction of IPV. IPV is known to be highly immunogenic when (189). The discovery of VDPV in multiple locations has lim-
administered to infants and children in resource-poor settings ited the options available for eventual discontinuation of polio
(182185) and has shown promise as a supplement to OPV immunization and underscored the critical need to maintain
in similar settings (147,186188). Furthermore, strategies are active surveillance and high levels of immunity worldwide for
being developed to overcome the higher costs associated with many years (180,181).

References
1. Paul JR. History of Poliomyelitis. New Haven Yale University Press; 15. Bodian D. Second attacks of paralytic poliomyelitis in human beings in
1971. relation to immunity, virus types and virulence. Am J Hyg. 1951;54:174190.
2. Lockhart JG. Memoirs of the Life of Sir Walter Scott, Bart. Philadelphia: 16. Committee on Typing of the National Foundation for Infantile Paralysis.
Carey Lean and Blanchard; 1837. A cooperative program for the typing of one hundred strains. Am J Hyg.
3. Underwood M. A Treatise on the Diseases of Children, with General 1951;54:191274.
Directions for the Management of Infants from the Birth. 2nd ed. London: 17. Drinker P, McKhann CF. The use of a new apparatus for the prolonged
Mathews; 1789. administration of artificial respiration: a fatal case of poliomyelitis. JAMA.
4. Heine J. Beobachtungen ber Lhmungszustnde der unteren Extremitten 1929;92:1658.
und deren Behandlung. Stuttgart: Khler; 1840. 18. Drinker P, Shaw LA. Apparatus for prolonged administration of
5. Charcot JM. Groupe des myopathies de cause spinale: paralysie infantile. artificial respiration: design for adults and children. J Clin Invest.
Rev Phot Hop (Paris). 1872;4:136. 1929;7:229.
6. Vermont State Department of Public Health. Infantile Paralysis in Vermont. 19. Pohl JF, Kenny E. The Kenny Concept of Infantile Paralysis and Its
Brattleboro: Vermont Printing Co; 1924. Treatment. Minneapolis: Bruce; 1943.
7. Wickman I. Beitrge zur Kenntnis der Heine-Medinschen Krankheit 20. Sabin AB, Olitsky PK. Cultivation of poliovirus in vitro in human embry-
(Poliomyelitis Acuta und Verwandter Erkrankungen). Berlin: Karger; onic nervous tissue. Proc Soc Exp Biol Med. 1936;34:357359.
1907. 21. Enders JF, Weller TH, Robbins FC. Cultivation of the Lansing strain of
8. Nathanson N, Martin JR. The epidemiology of poliomyelitis: enig- poliomyelitis virus in cultures of various human embryonic tissue. Science.
mas surrounding its appearance and disappearance. Am J Epidemiol. 1949;109:8587.
1979;110:672692. 22. Salk JE. Studies in human subjects on active immunization against
9. Landsteiner K, Popper E. Mikroscopische prparate von einem menschli- poliomyelitis. I. A preliminary report of experiments in progress. JAMA.
chen und zwei affenrckemarken. Wien Klin Wochenschr. 1908;21: 1953;151:10811098.
1830. 23. Sabin AB. Oral polio vaccine: history of its development and use and
10. Flexner S, Lewis PA. The transmission of poliomyelitis to monkeys. JAMA. current challenge to eliminate poliomyelitis from the world. J Infect Dis.
1909;53:1639. 1985;151:420436.
11. Paul JR, Trask JD. The neutralization test in poliomyelitis: comparative 24. Nkowane BM, Wassilak SCF, Orenstein WA, et al. Vaccine-associated
results with four strains of the virus. J Exp Med. 1935;61:447464. paralytic poliomyelitis. United States: 1973 through 1984. JAMA.
12. Turner TB, Young LE, Maxwell ES. Mouse-adapted Lansing strain of 1987;257:13351340.
poliomyelitis virus: neutralizing antibodies in serum of healthy children. 25. DeVries AS, Harper J, Murray A, et al. Vaccine-derived poliomyeli-
Am J Hyg. 1945;42:119127. tis 12 years after infection in Minnesota. N Engl J Med. 2011;364(24):
13. Paul JR, Riordan JT. Observations on serological epidemiology. Am J Hyg. 23162323.
1950;52:202212. 26. Alexander JP, Ehresmann K, Seward J, et al. Transmission of imported vac-
14. Burnet FM, Macnamara J. Immunological differences between strains of cine-derived poliovirus in an undervaccinated community in Minnesota. J
poliomyelitic virus. Br J Exp Pathol. 1931;12:5761. Infect Dis. 2009;199(3):391397.

Scheld_Ch07.indd 121 2/21/14 5:32 PM


122 Part II: Viral Infections and Related Disorders

27. Almond JW. The attenuation of poliovirus neurovirulence. Ann Rev 58. Voroshilova MK, Chumakov MP. Poliomyelitis-like properties of AB-IV-
Microbiol. 1987;41:154180. coxsackie A7 group of viruses. Prog Med Virol. 1959;2:106170.
28. Mendelsohn CL, Johnson B, Lionetti KA, et al. Transformation of a human 59. Grist NR, Bell EJ. Enteroviral etiology of the paralytic poliomyelitis syn-
poliovirus receptor gene into mouse cells. Proc Natl Acad Sci U S A. 1986; drome. Arch Environ Health. 1970;21:382387.
83:78457849. 60. Ho M, Chen ER, Hsu KH, et al. An epidemic of enterovirus 71 infection in
29. Mendelsohn CL, Wimmer E, Racaniello VR. Cellular receptor for poliovi- Taiwan. N Engl J Med. 1999;341:929935.
rus: molecular cloning, nucleotide sequence, and expression of a new mem- 61. Huang CC, Liu CC, Chang YC, et al. Neurologic complications in children
ber of the immunoglobulin super family. Cell. 1989;56:855865. with enterovirus 71 infection. N Engl J Med. 1999;341:936942.
30. Ren R, Costantini F, Gorgacz EJ, et al. Transgenic mice expressing a 62. Lee TC, Guo HR, Su HJ, et al. Diseases caused by enterovirus 71 infection.
human poliovirus receptor: a new model for poliomyelitis. Cell. 1990;63: Ped Infect Dis J. 2009;28(10):904910.
353362. 63. Anderson GW, Anderson G, Skaar A, et al. Poliomyelitis in pregnancy. Am
31. Gelfand HM, LeBlanc DR, Fox JP, et al. Studies on the development of J Hyg. 1952;55:127139.
natural immunity to poliomyelitis in Louisiana. II. Description and analysis 64. Weinstein L, Aycock WL, Feemster RF. Relation of sex, pregnancy, and men-
of episodes of infection observed in study group households. Am J Hyg. struation to susceptibility in poliomyelitis. N Engl J Med. 1951;245:5458.
1957;65:367385. 65. Harjulehto T, Aro T, Hovi T, et al. Congenital malformations and oral
32. Yorke JA, Nathanson N, Pianigiani G, et al. Seasonality and the require- poliovirus vaccination during pregnancy. Lancet. 1989;1:771772.
ments for perpetration and eradication of viruses in populations. Am J 66. Siegel M, Greenberg M. Poliomyelitis in pregnancy: effect on fetus and
Epidemiol. 1979;109:103123. newborn infant. J Pediatr. 1956;49:280288.
33. Iwasaki A, Welker R, Mueller S, et al. Immunofluorescence analysis of 67. Davis LE, Bodian D, Price D, et al. Chronic progressive poliomyelitis
poliovirus receptor expression in Peyers patches of humans, primates, and secondary to vaccination of an immunodeficient child. N Engl J Med.
CD155 transgenic mice: implications for poliovirus infection. J Infect Dis. 1977;297:241245.
2002;186:585592. 68. Wyatt HV. Poliomyelitis in hypogammaglobulinemics. J Infect Dis.
34. Bodian D. Emerging concept of poliomyelitis infection. Science. 1955; 1973;128:802806.
122:105108. 69. Sutter RW, Prevots DR. Vaccine-associated paralytic poliomyelitis among
35. Sabin AB. Behavior of chimpanzee-avirulent poliomyelitis viruses in experi- immunodeficient persons. Infect Med. 1994;11:426,429430,435438.
mentally infected human volunteers. Am J Med Sci. 1955;230:18. 70. Khetsuriani N, Prevots DR, Quick L, et al. Persistence of vaccine-derived
36. Bodian D. Poliovirus in chimpanzee tissues after virus feeding. Am J Hyg. polioviruses among immunodeficient persons with vaccine-associated para-
1956;64:187. lytic poliomyelitis. J Infect Dis. 2003;188:18451852.
37. Horstmann DM. Clinical aspects of acute poliomyelitis. Am J Med. 71. Horstmann DM. Acute poliomyelitis. Relation of physical activity at the
1949;6:592. time of onset to the course of the disease. JAMA. 1950;142:236241.
38. Bodian D. Viremia in experimental poliomyelitis. I. General aspects of 72. Russell WR. Paralytic poliomyelitis: the early symptoms and the effect of
infection after intravascular inoculation with strains of high and low inva- physical activity on the course of disease. Br Med J. 1949;1:465471.
siveness. Am J Hyg. 1954;60:339357. 73. Gatmaitan BG, Chason JL, Lerner AM. Augmentation of the virulence
39. Bodian D. Viremia in experimental poliomyelitis. II. Viremia and the of murine coxsackievirus B-3 myocardiopathy by exercise. J Exp Med.
mechanism of the provoking effect of injections or trauma. Am J Hyg. 1970;131:11211136.
1954;60:358370. 74. Greenberg M, Abramson H, Cooper HM, et al. The relation between
40. Ren R, Racaniello VR. Poliovirus spreads from muscle to the central ner- recent injections and paralytic poliomyelitis in children. Am J Public
vous system by neural pathways. J Infect Dis. 1992;166:747752. Health. 1952;42:142152.
41. Bodian D, Howe HA. An experimental study of the role of neurones in 75. Hill AB, Knowelden J. Inoculation and poliomyelitis: a statistical investiga-
the dissemination of poliomyelitis virus in the nervous system. Brain. tion in England and Wales in 1949. Br Med J. 1950;2:16.
1940;63:135. 76. Sutter RW, Patriarca PA, Suleiman AM, et al. Attributable risk of DTP
42. Jubelt B, Gallez-Hawkins G, Narayan O, et al. Pathogenesis of human (diphtheria and tetanus toxoids and pertussis vaccine) injection in provok-
poliovirus infection in mice. I. Clinical and pathological studies. J Neuropathol ing paralytic poliomyelitis during a large outbreak in Oman. J Infect Dis.
Exp Neurol. 1980;39:138148. 1992;165:444449.
43. Bodian D. Histopathological basis of clinical findings in poliomyelitis. Am 77. Strebel PM, Nedelcu NI, Baughman AL, et al. Intramuscular injec-
J Med. 1949;6:563578. tions within 30 days of immunization with oral poliovirus vaccinea
44. Hammon WM, Coriell LL, Wehrle PF, et al. Evaluation of Red Cross risk factor for vaccine-associated paralytic poliomyelitis. N Engl J Med.
gamma globulin as a prophylactic agent for poliomyelitis. IV. Final report 1995;332:500506.
of results based on clinical diagnoses. JAMA. 1953;151:12721285. 78. Paffenbarger RS, Wilson VO. Previous tonsillectomy and current preg-
45. Gelfand HM, LeBlanc DR, Fox JP, et al. Studies on the development of nancy as they affect risk of poliomyelitis. Ann N Y Acad Sci. 1955;61:
natural immunity to poliomyelitis in Louisiana. III. The serologic response 856868.
to commercially produced Salk vaccine of children totally or partially 79. Charcot JM. Paralysie essentielle de lenfance: atrophie musculaire con-
susceptible to poliovirus infection. Am J Hyg. 1959;70:303311. secutive. Gaz Med (Paris). 1875;4:225226.
46. Henry JL, Jaikaran ES, Davies JR, et al. A study of polio vaccination in 80. Campbell AMG, Williams MB, Pearce J. Late motor neuron degeneration
infancy: excretion following challenge with live virus by children given following poliomyelitis. Neurology. 1969;19:11011106.
killed or living poliovaccine. J Hyg. 1966;64:105120. 81. Martyn CN, Barker DJP, Osmond C. Motoneuron disease and past polio-
47. Wehrle PF, Carbonaro O, Day PA, et al. Transmission of polioviruses. III. myelitis in England and Wales. Lancet. 1988;1:13191320.
Prevalence of polioviruses in pharyngeal secretions of infected household 82. Mulder DW, Rosenbaum RA, Layton DD. Late progression of polio-
contacts of patients with clinical disease. Pediatrics. 1961;27:762764. myelitis or forme fruste amyotrophic lateral sclerosis? Mayo Clin Proc.
48. Paul JR, Riordan JT, Melnick JL. Antibodies to three different antigenic 1972;47:756761.
types of poliomyelitis virus in sera from North Alaskan Eskimos. Am J 83. Armon C, Jasper RD, Windebank AJ, et al. How frequently does clas-
Hyg. 1951;54:275285. sic amyotrophic lateral sclerosis develop in survivors of poliomyelitis?
49. Ogra PL, Karzon DT, Righthand F. Immunoglobulin response in serum and Neurology. 1990;40:172174.
secretions after immunization with live and inactivated poliovaccine and 84. Dalakas MC, Elder G, Hallet M, et al. A long-term follow-up study of
natural infection. N Engl J Med. 1968;279:893900. patients with post-poliomyelitis neuromuscular symptoms. N Engl J Med.
50. Ogra PL, Karzon DT. Formation and function of poliovirus antibody in 1986;314:959963.
different tissues. Prog Med Virol. 1971;13:157193. 85. Currie DM, Gershkoff AM, Cifu DX. Geriatric rehabilitation 3: mid-
51. Onorato IM, Modlin JF, McBean AM, et al. Mucosal immunity induced by and late-life effects of early-life disabilities. Arch Phys Med Rehabil.
enhanced potency IPV and OPV. J Infect Dis. 1991;163:16. 1993;74(suppl):S413S416.
52. Grassly NC, Jafari H, Bahl S, et al. Mucosal immunity after vaccination 86. Trojan DA, Cashman NR, Shapiro S, et al. Predictive factors for post-
with monovalent and trivalent oral poliovirus vaccine in India. J Infect Dis. poliomyelitis syndrome. Arch Phys Med Rehabil. 1994;75:770777.
2009;200(5):794801. 87. Trojan DA, Gendron D, Cashman NR. Stimulation frequency-dependent
53. Nishio O, Ishihara Y, Sakae K, et al. The trend of acquired immunity with neuromuscular junction transmission defects in patients with prior polio-
live poliovirus vaccine and the effect of revaccination: follow-up of vac- myelitis. J Neurol Sci. 1993;118:150157.
cinees for ten years. J Biol Stand. 1984;12:110. 88. Birk TJ. Poliomyelitis and the post-polio syndrome: exercise capaci-
54. Horstmann DM, Paul JR. The incubation period in human poliomyelitis ties and adapationcurrent research, future directions, and widespread
and its implications. JAMA. 1947;135:11. applicability. Med Sci Sports Exerc. 1993;25:466472.
55. Melnick JL, Ledinko N. Social serology: antibody levels in a normal young 89. Dalakas MC, Sever JL, Madden DL, et al. Late postpoliomyelitis muscu-
population during an epidemic of poliomyelitis. Am J Hyg. 1951;54: lar atrophy: clinical, virologic, and immunologic studies. Rev Infect Dis.
354382. 1984;6(2):S562S567.
56. Weinstein L, Shelokov A, Seltser R, et al. A comparison of the clinical features 90. Lange DJ, Smith T, Lovelace RE. Postpolio muscular atrophy: diagnostic
of poliomyelitis in adults and children. N Engl J Med. 1952;246:297302. utility of macroelectromyography. Arch Neurol. 1989;46:502506.
57. Russell WR, Fischer-Williams M. Recovery of muscular strength after 91. Ryniewicz B, Rowinska-Marcinska K, Emeryk B, et al. Disintegration
poliomyelitis. Lancet. 1954;1:330333. of the motor unit in post-polio syndrome. Part I. Electrophysiological

Scheld_Ch07.indd 122 2/21/14 5:32 PM


Chapter 7: Poliomyelitis, Polio Vaccines, and the Postpoliomyelitis Syndrome 123

findings in patients after poliomyelitis. Electromyogr Clin Neurophysiol. 127. McBean AM, Thoms ML, Albrecht P, et al. The serologic response to
1990;30:423427. oral polio vaccine and enhanced potency inactivated polio vaccines. Am J
92. Jubelt B, Drucker J. Post-polio syndrome: an update. Sem Neurol. Epidemiol. 1988;128:615628.
1993;13:283290. 128. Vidor E, Meschievitz C, Plotkin S. Fifteen years of experience of Vero-
93. Munsat TL. Poliomyelitisnew problems with an old disease. N Engl J produced enhanced potency inactivated poliovirus vaccine. Pediatr Infect
Med. 1991;324:12061207. Dis J. 1997;16:312322.
94. Ramlow J, Alexander M, LaPorte R, et al. Epidemiology of the post-polio 129. Robertson SE, Traverso HP, Drucker JA, et al. Clinical efficacy of a
syndrome. Am J Epidemiol. 1992;136:769786. new, enhanced-potency, inactivated poliovirus vaccine. Lancet. 1988;1:
95. Windebank AJ, Litchy WJ, Daube JR, et al. Late effects of paralytic 897899.
poliomyelitis in Olmsted County, Minnesota. Neurology. 1991;41: 130. Bijkerk H. Poliomyelitis epidemic in the Netherlands. Dev Biol Stand.
501507. 1979;43:195206.
96. Jubelt B, Cashman NR. Neurological manifestations of post-polio syn- 131. Stickle G. Observed and expected poliomyelitis in the United States,
drome. Crit Rev Neurobiol. 1987;3:199220. 19581961. Am J Public Health. 1964;54:12221229.
97. Dawson RB. Immune responses in the post-polio syndrome [letter]. 132. Gelfand HM, Potash L, LeBlanc DR, et al. Intrafamilial and interfamil-
N Engl J Med. 1992;326:641642. ial spread of living vaccine strains of polioviruses. JAMA. 1959;170:
98. Dalakas MC, Sever JL, Fletcher M, et al. Neuromuscular symptoms in 20392048.
patients with old poliomyelitis: clinical, virological and immunologic 133. Wehrle PF, Reichert R, Carbonaro O, et al. Influence of prior active immu-
studies. In: Halstead LS, Weichers DO, eds. Late Effects of Poliomyelitis. nization on the presence of poliomyelitis virus in the pharynx and stools
Miami, FL: Symposia Foundation; 1985:7389. of family contacts of patients with paralytic poliomyelitis. Pediatrics.
99. Sharief MK, Hentges R, Ciardi M. Intrathecal immune response in 1958;21:353361.
patients with the post-polio syndrome. N Engl J Med. 1991;325:749755. 134. Robertson HE, Acker MS, Dillenberg HO, et al. Community-wide use
100. Stone R. Post-polio syndrome: remembrance of viruses past. Science. of a balanced trivalent oral poliovirus vaccine (Sabin). Can J Public
1994;264:909. Health. 1962;53:179191.
101. Rosenberg LT, Hamparian VV. Immune responses in the post-polio syn- 135. Cohen-Abbo A, Culley BS, Reed GW, et al. Seroresponse to trivalent
drome [letter]. N Engl J Med. 1992;326:640642. oral poliovirus vaccine as a function of dosage interval. Ped Infect Dis J.
102. Salazar-Grueso EF, Grimaldi LME, Roos RP, et al. Isoelectric focusing 1995;14:100106.
studies of serum and cerebrospinal fluid in patients with antecedent polio- 136. Hardy GE, Hopkins CC, Linneman CC, et al. Trivalent oral poliovirus
myelitis. Ann Neurol. 1989;26:709713. vaccine: a comparison of two infant immunization schedules. Pediatrics.
103. Agre JC, Rodriquez AA. Neuromuscular function in polio survivors. 1970;45:444448.
Orthopedics. 1991;14:13431347. 137. Krugman RD, Hardy GE, Sellers C. Antibody persistence after pri-
104. Berlly MH, Strauser WW, Hall KM. Fatigue in post-polio syndrome. Arch mary immunization with trivalent oral poliovirus vaccine. Pediatrics.
Phys Med Rehabil. 1991;72:115118. 1977;60:8082.
105. Packer TL, Martins I, Krefting L, et al. Activity and post-polio fatigue. 138. Ogra PL. Mucosal immune response to poliovirus vaccines in childhood.
Orthopedics. 1991;14:12231226. Rev Infect Dis. 1984;6(2):S361S368.
106. Sharma KR, Kent-Braun J, Mynhier MA, et al. Excessive muscular fatigue 139. Ghendon YUZ, Sanakoyeva II. Comparison of the resistance of the
in the postpoliomyelitis syndrome. Neurology. 1994;44:642664. intestinal tract to poliomyelitis virus (Sabins strains) in person after natu-
107. Gyermek L. Increased potency of nondepolarizing relaxants after polio- rally and experimentally acquired immunity. Acta Virol (Praha). 1961;5:
myelitis. J Clin Pharmacol. 1990;30:170173. 265273.
108. Louhan JA. Epidemic poliomyelitis: some pathological observations on 140. Expanded Programme on Immunization. Poliomyelitis surveillance and
human material. Arch Pathol. 1946;42:245260. vaccine efficacy. Wkly Epidemiol Rec. 1988;63:249251.
109. Bodian D. Poliomyelitis: neuropathologic observations in relation to 141. Dong DX, Hu XM, Liu WJ, et al. Immunization of neonates with tri-
motor symptoms. JAMA. 1947;134:11481154. valent oral poliomyelitis vaccine (Sabin). Bull World Health Organ.
110. Bruno RL, Cohen JM, Galaski T, et al. The neuroanatomy of post-polio 1986;64:853860.
fatigue. Arch Phys Med Rehabil. 1994;75:498504. 142. John TJ, Jayabal P. Oral polio vaccination of children in the tropics:
111. Bruno RL. Post-polio sequelae: research and treatment in the second I. The poor seroconversion rates and the absence of viral interference.
decade [editorial]. Orthopedics. 1991;14:11691170. Am J Epidemiol. 1972;96:263269.
112. Bruno RL, Frick NM, Cohen J. Polioencephalitis, stress, and the etiology 143. Patriarca PA, Wright PF, John TJ. Factors affecting the immunogenic-
of post-polio sequelae. Orthopedics. 1991;14:12691276. ity of oral poliovirus vaccine in developing countries. Rev Infect Dis.
113. Sonies BC, Dalakas MC. Dysphagia in patients with the post-polio syn- 1991;13:926939.
drome. N Engl J Med. 1991;324:11621167. 144. Jenkins HE, Aylward RB, Gasasira A, et al. Effectiveness of immu-
114. Bach JR, Alba AS. Pulmonary dysfunction and sleep disordered breath- nization against paralytic poliomyelitis in Nigeria. N Engl J Med.
ing as post-polio sequelae: evaluation and management. Orthopedics. 2008;359(16):16661674.
1991;14:13291337. 145. Sutter RW, Patriarca PA, Brogan S, et al. Outbreak of paralytic poliomy-
115. Dean E, Ross H, Road JD, et al. Pulmonary function in individuals with a elitis in Oman: evidence for widespread transmission among fully vac-
history of poliomyelitis. Chest. 1991;100:118123. cinated children. Lancet. 1991;338:715720.
116. Bruno RL, Galski T, DeLuca J. The neuropsychology of post-polio fatigue. 146. de Brito Bastos NC, de Carvalho ES, Schatzmayr H, et al. Antipoliomyelitis
Arch Phys Med Rehabil. 1993;74(suppl):S413S416. program in Brazil: a serologic study of immunity. Bull Pan Am Health
117. Emeryk B, Rowinska-Marcinska K, Ryniewicz B, et al. Disintegration Organ. 1974;8:5465.
of the motor unit in post-polio syndrome. Part II. Electrophysiological 147. Lasch EE, Abed Y, Abdulla K, et al. Successful results of a program com-
findings in patients with post-polio syndrome. Electromyogr Clin bining live and inactivated poliovirus vaccines to control poliomyelitis in
Neurophysiol. 1990;30:451458. Gaza. Rev Infect Dis. 1984;6(2):467470.
118. Clark K, Dinsmore DO, Grafman J, et al. A personality profile of patients 148. Myaux JA, Unicomb L, Besser RE, et al. Effect of diarrhea on the humoral
diagnosed with post-polio syndrome. Neurology. 1994;44:18091811. response to oral polio vaccination. Ped Infect Dis J. 1996;15:204209.
119. Cashman NR, Maselli R, Wollman RL. Late denervation in patients with 149. World Health Organization Collaborative Study Group on Oral Poliovirus
antecedent paralytic poliomyelitis. N Engl J Med. 1987;317:712. Vaccine. Effect of diarrhea on seroconversion to oral poliovirus vaccine.
120. Jones DR, Speier J, Canine K, et al. Cardiorespiratory responses to Paper presented at: 32nd Interscience Conference on Antimicrobial
aerobic training by patients with postpoliomyelitis sequelae. JAMA. Agents and Chemotherapy; October 1114, 1992; Anaheim, CA.
1989;261:32553258. 150. Caceres VM, Sutter RW. Sabin monovalent oral polio vaccines: review of
121. Young GR. Energy conservation, occupational therapy, and the treatment past experiences and their potential use after polio eradication. Clin Infect
of post-polio sequelae. Orthopedics. 1991;14:12331239. Dis. 2001;33:531541.
122. Peach PE, Olejnik S. Effect of treatment and noncompliance on post-polio 151. el-Sayed N, el-Gamal Y, Abbassy AA, et al. Monovalent type 1 oral polio-
sequelae. Orthopedics. 1991;14:11991203. virus vaccine in newborns. N Engl J Med. 2008;359:16551665.
123. Fillyaw MJ, Badger GJ, Goodwin GD, et al. The effects of long-term 152. Waggie Z, Geldenhuys H, Sutter RW, et al. Randomized trial of type 1 and
non-fatiguing exercise in subjects with postpolio syndrome. Orthopedics. type 3 oral monovalent poliovirus vaccines in newborns in Africa. J Infect
1991;14:12531256. Dis. 2012;205:228236.
124. Einarsson G. Muscle conditioning in late poliomyelitis. Arch Phys Med 153. Chen RT, Hausinger S, Dajani AS, et al. Seroprevalence of anti-
Rehabil. 1991;72:1114. body against poliovirus in inner-city preschool children. JAMA.
125. Milner-Brown HS. Muscle strengthening in a post-polio subject through 1996;275:16391645.
a high-resistance weight-training program. Arch Phys Med Rehabil. 154. World Health Organization. Polio vaccines and polio immunization
1993;74:11651167. in the pre-eradication era: WHO position paper. Wkly Epidemiol Rec.
126. Centers for Disease Control and Prevention. Updated recommendations 2010;23:213228.
of the Advisory Committee on Immunization Practices (ACIP) regarding 155. Strebel PM, Sutter RW, Cochi SL, et al. Epidemiology of poliomyelitis in
routine poliovirus vaccination. MMWR Morb Mortal Wkly Rep. the United States one decade after the last reported case of indigenous
2009;58:829830. wild virus-associated disease. Clin Infect Dis. 1992;14:568579.

Scheld_Ch07.indd 123 2/21/14 5:32 PM


124 Part II: Viral Infections and Related Disorders

156. Wyatt HV. Risk of live poliovirus in immunodeficient children. J Pediatr. 175. Kew O, Morris-Glasgow V, Landaverde M, et al. Outbreak of poliomyeli-
1975;87:152153. tis in Hispaniola associated with circulating type 1 vaccine-derived polio-
157. Kew OM, Sutter RW, Nottay BK, et al. Prolonged replication of a type virus. Science. 2002;296:356359.
1 vaccine-derived poliovirus in an immunodeficient patient. J Clin 176. Centers for Disease Control and Prevention. Outbreak of poliomyelitis
Microbiol. 1998;36:28932899. Dominican Republic and Haiti, 2000. MMWR Morb Mortal Wkly Rep.
158. Le Menach A, Llosa AE, Mouniaman-Nara I, et al. Poliomyelitis out- 2000;49:1094.
break, Pointe-Noire, Republic of the Congo, September 2010February 177. Centers for Disease Control and Prevention. Circulation of a type 2 vac-
2011. Emerg Infect Dis. 2011;17(8):15061509. cine-derived poliovirusEgypt, 19821993. MMWR Morb Mortal Wkly
159. Henderson RH. The Expanded Programme on Immunization of the Rep. 2001;50:4142,51.
World Health Organization. Rev Infect Dis. 1984;6(2):475479. 178. Kew OM, Sutter RW, de Gourville EM, et al. Vaccine-derived poliovi-
160. Ofusu-Amaah S. The challenge of poliomyelitis in tropical Africa. Rev ruses and the endgame strategy for global polio eradication. Ann Rev
Infect Dis. 1984;6(2):318320. Microbiol. 2005;59:587635.
161. Rockefeller Foundation. Protecting the worlds children: vaccines and 179. Centers for Disease Control and Prevention. Update on vaccine-derived
immunization. Paper presented at: Bellagio Conference; March 1315, poliovirusesworldwide, January 2008June 2009. MMWR Morb
1984; Bellagio, Italy. Mortal Wkly Rep. 2009;58:10021006.
162. Horstmann DM. Preface to the 1983 International Symposium on 180. Dowdle WR, de Gourville E, Kew OM, et al. Polio eradication: the OPV
Poliomyelitis Control. Rev Infect Dis. 1984;6(suppl 2):S301. paradox. Rev Med Virol. 2003;13:277291.
163. Hull HF, Ward NA, Hull BP, et al. Paralytic poliomyelitis: seasoned strate- 181. Nathanson N, Fine P. Poliomyelitis eradicationa dangerous endgame.
gies, disappearing disease. Lancet. 1994;343:13311337. Science. 2002;296:269270.
164. Wright PF, Kim-Farley RJ, de Quadros CA, et al. Strategies for the 182. Sutter RW, Suleiman AJ, Malankar P, et al. Trial of a supplemental
global eradication of poliomyelitis by the year 2000. N Engl J Med. dose of four poliovirus vaccines. Sequential use of inactivated poliovi-
1991;325:17741779. rus vaccine followed by oral poliovirus vaccine in Oman. N Engl J Med.
165. World Health Organization. Global eradication of poliomyelitis by the 2000;343(11):767773.
year 2000. Wkly Epidemiol Rec. 1988;63:161162. 183. World Health Organization. Introduction of inactivated poliovirus vac-
166. Aylward B, Tangermann R. The global polio eradication initiative: lessons cine into oral poliovirus vaccine-using countries. Wkly Epidemiol Rec.
learned and prospects for success. Vaccine. 2011;29(suppl 4):D80D85. 2003;78:241250.
167. Richardson G, Linkins R, Eames M, et al. Immunogenicity of oral 184. Asturias EJ, Dueger EL, Omer SB, et al. Randomized trial of inactivated
poliovirus vaccine administered in mass campaigns versus routine and live polio vaccine schedules in Guatemalan infants. J Infect Dis.
immunization programmes. Bull World Heath Organ. 1995;73:769777. 2007;196:692698.
168. Cruz RR. Cuba: mass polio vaccination program, 19621982. Rev Infect 185. Dayan GH, Thorley M, Yamamura Y, et al. Serologic response to inacti-
Dis. 1984;6(2):408412. vated poliovirus vaccine: a randomized clinical trial comparing 2 vaccina-
169. Risi JB. The control of poliomyelitis in Brazil. Rev Infect Dis. 1984;6(2): tion schedules in Puerto Rico. J Infect Dis. 2007;195:1220.
400403. 186. du Chatelet IP, Merchant AT, Fisher-Hoch S, et al. Serological response
170. Abraham T. Polio eradication: a complex end game. Br Med J. 2012;344: and poliovirus excretion following different combined oral and inacti-
e2398. vated poliovirus vaccines immunization schedules. Vaccine. 2003;21:
171. Centers for Disease Control and Prevention. Progress toward the global 17101718.
interruption of wild poliovirus type 2 transmission, 1999. MMWR Morb 187. Lasch EE, Abed Y, Marcus O, et al. Combined live and inactivated polio-
Mortal Wkly Rep. 1999;48:736747. virus vaccine to control poliomyelitis in a developing countryfive years
172. Centers for Disease Control and Prevention. Progress towards global erad- after. Develop Biol Stand. 1986;65:137143.
ication of poliomyelitis, 2002. MMWR Morb Mortal Wkly Rep. 2003; 188. Morinere BJ, van Loon FPL, Rhodes PH, et al. Immunogenicity of a
52:366369. supplemental dose of oral versus inactivated poliovirus vaccine. Lancet.
173. Modlin JF. The bumpy road to polio eradication. N Engl J Med. 2010; 1993;341:15451550.
362:23462349. 189. Technical Consultative Group to the World Health Organization on the
174. Patriarca PA. Research and development and the polio eradication initiative: Global Eradication of Poliomyelitis. Endgame issues for the global
too much, too soon . . . too little, too late? Clin Infect Dis. 2012;55:13071311. polio eradication initiative. Clin Infect Dis. 2002;34:7277.

Scheld_Ch07.indd 124 2/21/14 5:32 PM


CHAPTER 8 MEASLES AND RUBELLA
DIANE E. GRIFFIN

Measles and rubella are rash diseases of childhood that can range. Nonprimate morbilliviruses cause respiratory disease in
be complicated by neurologic disease. Measles virus (MeV) dogs, horses, cows, goats, sheep, and marine mammals and
induced neurologic disease is associated with community- neurologic complications are common. Rinderpest, a disease
acquired infection of children, whereas the most important of cattle, has recently been eradicated (4,5).
rubella virus (RV)induced neurologic disease is associated Morbilliviruses have six structural proteins (Fig. 8.1). The
with congenital infection. For both diseases, effective and safe hemagglutinin (H) and fusion (F) are transmembrane proteins
vaccines exist. Despite wide distribution of measles vaccine, present on the surface of the virus and infected cell. These pro-
measles remains a frequent cause of morbidity and mortality teins are important for viral attachment and penetration of
worldwide, and even a low incidence of measles-associated the target cell. The matrix (M) protein is found on the inner
neurologic disease has a significant impact on society because surface of the membrane and interacts with the cytoplasmic
of the long-term neurologic disabilities that ensue (1). Rubella tails of H and F and with the nucleocapsid for virion assem-
vaccine is less widely distributed, but there is an intensifying bly and budding. The nucleocapsid (N) protein surrounds and
effort to increase coverage in developing countries and reduce encapsidates the viral RNA to form the helical nucleocapsid
the worldwide incidence of congenital rubella syndrome (CRS) structures. The phosphoprotein (P) and large (L) polymerase
(2). The World Health Organization has targeted both viruses protein are also associated with the nucleocapsid and com-
for elimination and potential eradication (3). plete the viral elements necessary for RNA transcription. Two
Neurologic complications may result from direct virus inva- nonstructural proteins, C and V, are encoded within the P gene
sion of the central nervous system (CNS) or from induction of and regulate the host innate response to infection (612).
an autoimmune response to CNS antigens. For measles, there MeV transmission from person to person is by the respi-
are three distinct neurologic diseases that occur either at the ratory route. The virus spreads from the initial site of repli-
time of the acute disease or months to many years after appar- cation in the respiratory tract to local draining lymph nodes
ent recovery: acute disseminated encephalomyelitis (ADEM), (1315). Replication in lymphatic tissue produces virus that
measles inclusion body encephalitis (MIBE), and subacute then spreads through the blood to multiple organs including
sclerosing panencephalitis (SSPE). Rubella causes ADEM and skin, lung, liver, spleen, and lymph nodes. The viremia is cell
progressive rubella panencephalitis (PRP) less frequently than associated with infected B cells, T cells, and monocytes in cir-
measles but is also teratogenic and causes CRS. culation (1618). MeV-infected leukocytes increase expression
and activation of the integrins lymphocyte function-associated
antigen (LFA)-1 and very late antigen (VLA)-4 that promote
BACKGROUND ON MEASLES adherence to the surface of endothelial cells and this interaction
is likely to facilitate the spread of infection to many organs
MeV, the etiologic agent of measles, is a member of the and tissues (19,20). In tissue, endothelial and epithelial cells
Morbillivirus genus of the Paramyxoviridae family of nonseg- are also targets for infection (2123).
mented, negative-stranded, enveloped RNA viruses. There are Three cellular receptors have been identified: membrane
several morbilliviruses and each has a relatively restricted host cofactor protein or CD46 (24,25), signaling lymphocyte acti-

Nucleocapsid protein (N)


Phosphoprotein (P)

Matrix protein (M)

Fusion protein (F)

Attachment protein (H)

FIGURE 8.1 Schematic diagram of


Polymerase (L) measles virus. (Brindley and Plemper,
reproduced from Griffin DE. Measles
virus. In: Knipe DM, Howley PM, Griffin
DE, et al, eds. Fields Virology. 6th ed.
Philadelphia: Lippincott Williams &
Wilkins; 2013, with permission.)

125

Scheld_Ch08.indd 125 2/21/14 5:33 PM


126 Part II: Viral Infections and Related Disorders

infection. However, these changes are also observed after mea-


rash
sles immunization, so it is not clear that they indicate virus
infection of the CNS (44).
The characteristic morbilliform rash of measles is due to
measles ADEM immune cell infiltration into sites of MeV infection of skin epi-
virus MIBE thelial cells and marks the onset of the immune response and
infection SSPE the initiation of virus clearance (45). It is a time of intense
immune activation (4648) which is accompanied by suppres-
sion of skin test responses to recall antigens such as tuberculin
(49,50) and decreased in vitro lymphoproliferative responses
7 14 21 1 3 6 9 1 3 6 9 12 to mitogens (51). This immune suppression contributes to an
days months years increased susceptibility to secondary infections, the most com-
mon cause of death due to measles (52). The immune activa-
FIGURE 8.2 Time after infection of the occurrence of the three major tion accompanying measles may contribute to the neurologic
neurologic complications of measles: acute disseminated encephalo- complications as well.
myelitis (ADEM), measles inclusion body encephalitis (MIBE), and Although clearance of infectious MeV is generally complete
subacute sclerosing panencephalitis (SSPE). (Reproduced from Griffin after the rash has resolved, clearance of viral RNA requires
DE. Measles virus. In Knipe DM, Howley PM, Griffin DE, et al, eds. many months (53,54). This continued presence of MeV RNA
Fields Virology. 6th ed. Philadelphia: Lippincott Williams & Wilkins;
in lymphoid tissue and circulating mononuclear cells may con-
2013, with permission.)
tribute to immune suppression and to development of acute
and chronic neurologic disease.
vation molecule (SLAM) or CD150 (26), and nectin 4 (27,28).
CD46 is a widely distributed human complement regulatory
protein expressed on all nucleated cells including the api- OVERVIEW OF THE NEUROLOGIC
cal surface of polarized epithelial cells. In the CNS, CD46 COMPLICATIONS OF MEASLES
is expressed on choroid plexus epithelial cells, on cerebral
endothelium, and ependymal cells (29,30). SLAM/CD150 is a The neurologic complications of measles are uncommon and
membrane glycoprotein expressed on cells of the immune sys- occur at three distinct times in relation to the primary infection
tem including immature thymocytes, activated T and B lym- and acute disease (Fig. 8.2). ADEM usually presents within
phocytes, activated monocytes, and mature dendritic cells 1 to 2 weeks of the appearance of the rash (55,56), MIBE
(31,32) but is not expressed by brain parenchymal cells (29). within a few months (57,58), and SSPE several years after ini-
Nectin 4 is an adherens junction protein expressed by epithe- tial infection (59). The age and general immune status of the
lial cells in the lungs, tonsils, and placenta (33,34). Studies of individuals susceptible to these complications are also distinct
different strains of MeV have shown that both vaccine and (Table 8.1). ADEM occurs in individuals who have apparently
wild type viruses can use CD150 and nectin 4 as a receptor. normal immune systems and are older than 2 years at the time
However, although most vaccine strains use CD46 efficiently, of primary infection (52). The incidence is 1:1,000 cases of
wild type strains do not (3537). The receptors used for neural measles (52,60). MIBE occurs in immunosuppressed patients
cell infection by wild type MeV and chick cell infection by vac- of any age (6165). The incidence is approximately 1:10 cases
cine virus (3840) have not been identified. of measles in immunocompromised children (66,67). SSPE
Through expression of the H and F proteins on the cell occurs in immunologically normal individuals who have often
surface, infected cells may fuse with nearby cells to form syn- had measles at younger than 2 years (59,68,69). The incidence
cytia or giant cells both in vitro and in the lungs and lymphoid is approximately 1:10,000 cases of measles (70).
tissues of infected patients. However, MeV is not detectable MeV is easily demonstrable in the brains of patients with
by usual pathologic or immunocytochemical techniques in MIBE and SSPE, but not ADEM (21). The current understand-
the brains of patients dying acutely with measles (21). Studies ing of these three neurologic complications of measles is that
using in situ hybridization have identified MeV infecting cere- ADEM is an autoimmune demyelinating disease triggered by
bral capillary endothelial cells during acute fatal disease (22). measles, MIBE is a progressive MeV infection of the brain in
In addition, electroencephalographic (EEG) abnormalities and patients unable to mount an effective immune response, and
a cerebrospinal fluid (CSF) pleocytosis are common in acute SSPE is a slowly progressive MeV infection of the CNS that
uncomplicated measles (4143), suggesting the possibility that is poorly controlled by the immune system and presents clini-
MeV infection of the CNS is common during uncomplicated cally many years after infection.

TA B L E 8 . 1
SUMMARY OF THE NEUROLOGIC COMPLICATIONS OF MEASLES

Age of
Disease Host Measles Onset Incidence Pathology Time Course

Acute disseminated Normal 2 yr 1:1,000 Inflammation, demyelination Monophasic, weeks


encephalomyelitis (ADEM)
Measles inclusion body Immunocompromised Any 1:10 Inclusion bodies Progressive, months
encephalitis (MIBE)
Subacute sclerosing Normal 2 yr 1:10,000 Inclusion bodies, inflammation Progressive, years
panencephalitis (SSPE)

Scheld_Ch08.indd 126 2/21/14 5:33 PM


Chapter 8: Measles and Rubella 127

of viral RNA extracted from the brains of MIBE patients has


MEASLES INCLUSION-BODY shown that messenger RNAs (mRNAs) for the envelope pro-
ENCEPHALITIS teins are limited in amount and that the virus has accumulated
many mutations, particularly in the M-protein gene, similar
to those found in SSPE (see later discussion) (79,80). These
History mutations often preclude or severely limit productive replica-
tion (8183). These mutations may reflect positive selection
Progressive MeV infection was reported first by Hecht (71) and adaption of the virus to growth in neurons or frequent
in 1910 as a case of giant cell pneumonia. Hecht pneu- transcription errors, including biased hypermutation due to
monia was suspected to be a complication of measles and the action of dsRNAdependent adenosine deaminase (84),
this hypothesis was proven in 1959 when Enders et al. (72) combined with a lack of selection for replication-competent
isolated MeV from the lungs and respiratory tract secretions virus. Production of infectious virus may not be important
of three young children with compromised immune systems for virus replication and spread in the CNS, because neu-
and progressive pulmonary disease. Pathologic examination rons can allow transsynaptic spread of viral RNAcontaining
of the lungs of these children showed giant cell pneumo- nucleocapsids from cell to cell without production of infec-
nia. None had a history of rash or other classic signs or tious virions (8588).
symptoms of measles, but all had histories of exposure to
measles in the preceding months. Giant cells were found in
other organs (e.g., liver, lymph nodes), but neurologic symp-
toms were not reported and it is not clear that the brain was Clinical Manifestations
examined (72).
MIBE usually presents 1 to 6 months after exposure to mea-
Breitfeld and colleagues (73) first reported progressive
sles with progressive neurologic deterioration in the absence
nervous system disease in 1973 as cases of SSPE. Two young
of fever. Signs and symptoms include altered mental status,
children with leukemia developed progressive neurologic dis-
lethargy, slurred speech, focal motor seizures or epilepsia
ease and died approximately 6 months after exposure to mea-
partialis continua, weakness, and occasionally blindness or
sles. At autopsy, giant cells were not present in the CNS, but
hearing loss (57,62,65,77). Typically, the seizures are resis-
there were intranuclear and intracytoplasmic inclusions now
tant to control by anticonvulsant medications (62). Disease
recognized to be frequent pathologic features of persistent
progresses over days to weeks to coma and death and may
MeV infection of the CNS. One also had Hecht pneumonia.
be accompanied by inappropriate secretion of antidiuretic
Subsequently, this neurologic disease, MIBE, was distinguished
hormone (58,66,73).
from SSPE by its time of onset in relationship to measles, lack
of inflammatory response in the brain (74), and occurrence
in immunocompromised individuals. Recently, this disease has
also been described in adults who are immunocompromised Diagnosis
due to HIV infection (65) and after immunization with the live
attenuated MeV vaccine (75). The diagnosis is often difficult because there may be no history
of a rash. History of exposure to measles or immunization in
the preceding months should be sought in immunosuppressed
Pathogenesis and Pathology individuals with progressive neurologic deterioration. Brain
biopsy with pathologic examination and reverse transcrip-
The rash marks the onset of the immune response with the tase-polymerase chain reaction (RT-PCR) to detect MeV
appearance of MeV-specific antibody and cellular immu- RNA is often necessary for definitive diagnosis (62,65,89).
nity, and this immune response to MeV is normally effective At the time of presentation, there is often no detectable
at clearing infectious virus from blood and tissues (53). In antibody to MeV, consistent with the poor immune response
individuals with severe acquired or genetic defects in cellular to infection, although at later times antibody may be pres-
immunity, a rash may not appear (72,73) and MeV infection ent in serum or CSF (62,66,74). CSF examination is usually
may not be controlled. These patients often develop progres- within normal limits, but occasionally there is a moderate
sive pulmonary or neurologic disease due to unrestrained MeV elevation in the protein concentration (62). EEG findings are
replication (66,73,74). usually abnormal, but nonspecific and nondiagnostic, with
Virus infection of the CNS is presumed to begin with diffuse slowing or periodic lateralized spike and wave activ-
infection of cerebral capillary endothelial cells or with in- ity (62,65,66,90). Computed tomographic (CT) scan and
filtration of infected leukocytes into the brain (20,22,76). magnetic resonance imaging (MRI) scans are often normal
Neurons or glial cells subsequently become infected and at the time of presentation with later development of T2 sig-
virus spreads slowly within the CNS. Intranuclear and intra- nal abnormalities, edema, cortical atrophy, and ventricular
cytoplasmic eosinophilic inclusion bodies are seen primar- dilation (62,65,66).
ily in gray matter areas (73). There is focal necrosis, and
neurons often show signs of degeneration (74). The areas
of brain most frequently involved are the parietooccipital Treatment and Prevention
areas, basal ganglia, and brainstem. Typically, there is little
evidence of an inflammatory response to the infection, al- Adequate measles immunization prior to the onset of leuke-
though proliferation of astrocytes and microglia is often ap- mia or other immunosuppressive illness undoubtedly pre-
parent (61,74). vents many cases of MIBE (57). It is difficult to discern the
On electron microscopic examination, inclusion bodies usefulness of postexposure immunoglobulin prophylaxis for
contain the microtubular structures of MeV nucleocapsids immunocompromised individuals. Several cases of MIBE
(58,74). MeV antigens (particularly N) can be identified by have occurred despite administration of immunoglobulin, but
immunocytochemical staining (77). However, the H, F, and many more may have been prevented. There is no established
M proteins are undetectable (78) and often MeV cannot be antiviral treatment for MIBE and essentially all cases are fatal.
recovered in culture from brain tissue (61,64). Examination However, there are case reports of neurologic improvement

Scheld_Ch08.indd 127 2/21/14 5:33 PM


128 Part II: Viral Infections and Related Disorders

with prolonged intravenous ribavirin treatment (62) and enters the brain at the time of the original acute infection or
slowed disease progression with initiation of antiretroviral during the prolonged phase of circulating viral RNA and sub-
therapy in patients with AIDS (65). sequently spreads slowly through the CNS eventually infecting
a sufficient number of cells to produce dysfunction and clinical
evidence of infection.
SUBACUTE SCLEROSING At the time that neurologic symptoms are recognized, the
infection is extensive. Neurons and oligodendrocytes contain
PANENCEPHALITIS nuclear and cytoplasmic viral inclusion bodies (114), antibody
responses are vigorous and evident both in serum and CSF
History (115,116), and there is an extensive mononuclear inflamma-
tory reaction in the brain (92,93,117). Gray matter is most
SSPE was first described in 1933 by Dawson (91) in a 16-year- prominently affected, but pathologic changes are present in
old boy with progressive neurologic deterioration character- white matter as well (92,93). Retinitis is frequently present
ized by failing memory, slow and deliberate movements, and with MeV antigen demonstrable in the retinal neuroepithe-
myoclonus. The following year, he reported an additional case lium. At autopsy, MeV RNA or antigen can be detected in a
of this subacute inclusion body encephalitis in a 5-year-old girl wide variety of tissues (118).
(92). Histologic examination of the brains of these patients Pathologic examination of the brain shows intranuclear
showed inflammation with eosinophilic intranuclear and and intracytoplasmic inclusions, in situ hybridization shows
intracytoplasmic inclusions in neurons. The disease became MeV RNA, and immunocytochemical staining shows MeV
known as Dawson encephalitis, and Dawson postulated that antigens (110). However, no virus is seen budding from the
the disease was of viral etiology, but he could not transmit the surface of infected cells (119). Nuclear inclusions are filled
disease to experimental animals. In 1945, van Bogaert (93) with smooth nucleocapsids (114,119) consistent with the
described a similar condition, subacute sclerosing leukoen- absence of the associated L and P proteins necessary for tran-
cephalitis with prominent white matter involvement. It was scription and replication of viral RNA. The cytoplasm con-
appreciated subsequently that the same disease could involve tains replication-competent fuzzy nucleocapsids that extend
both gray and white matter (94). In 1966, paramyxovirus-like into neuronal processes further suggesting that virus can
particles suggestive of MeV were seen on electron microscopic spread within the CNS by cell-to-cell synaptic transmission
examination of the inclusions (95). Reports of these virus of the ribonucleoprotein complex (85,87,88,113,120). The
particles were followed rapidly by observations of elevated observation that strains of MeV isolated from SSPE patients
MeV antibody in serum and CSF, staining of the inclusions are more likely than standard strains of MeV to cause neuro-
with antibody to MeV antigens and culture of MeV from logic disease after intracerebral inoculation into small animals
brains (96100). and primates suggests that the virus has adapted to growth in
neural tissue.
Like MIBE, the virus present in the brain of SSPE patients
Epidemiology is replication defective and cannot usually be recovered
from SSPE brain in a cell-free form (99,100,121). Extensive
SSPE is a rare (approximately 1 in 10,000) late complication sequence analysis of viral RNAs has shown that SSPE viruses
of measles (68,70,101). The mean time to onset of SSPE after are of the same lineage as viruses that cause acute measles but
measles is 6 to 10 years (59,102,103) (Fig. 8.2). Children distinguishable in the genes encoding the M, F, and H proteins
with SSPE often have a history of acquiring measles at an (112,122129). Studies of brain-associated viral proteins and
early age (59,6870,104107) when the immune system is RNA in SSPE have revealed differences in the relative amounts
immature and maternal antibody may still be present. In most of the various viral mRNAs and proteins (127130), in the
parts of the world, the disease occurs preferentially in boys antigenicity of viral proteins (131), and in RNA sequences cod-
(68,104,108,109). Exposure to birds has been identified as a ing for viral proteins (125127) between SSPE viruses and wild
risk factor (69,105,108). How these factors increase the risk type or vaccine strains of MeV. Because the disease cannot be
of developing SSPE is unknown. There is no clustering of cases studied prospectively, it is not known for certain whether these
to suggest that the virus causing the initial infection leading to differences represent unique features of the original infecting
SSPE is different from the virus causing uncomplicated disease. virus, selection for growth in the CNS, or selection for growth
in the presence of a vigorous antibody response.
In general, expression of M protein is low (130,132,133)
Pathogenesis and Pathology and the mRNA encoding M extracted from SSPE brain is
mutated throughout the gene. Construction of recombinant
SSPE is the most extensively studied of the neurologic com- viruses has shown that functional M is dispensable for virus
plications of measles. Nevertheless, the pathogenesis of this growth and spread in the CNS and may foster the formation
rare complication remains obscure. It is not known whether of nuclear and cytoplasmic inclusion bodies (83). Mutations in
infection of the nervous system occurs at the time of primary the H and F envelope proteins that interfere with assembly and
infection and progresses slowly with clinical evidence of dis- budding of infectious virus are also associated with persistent
ease apparent only after years or whether the infection is latent infection and SSPE (123,126,127,134).
at a site outside the CNS and then spreads to the brain. The The possibility that the development of SSPE represents
route of virus entry into the CNS is unknown, but infection of a defect in the immune response has led to investigations of
cerebral capillary endothelial cells is a likely possibility (76,11). cellular and humoral immune responses to MeV and other
Extensive sequence analysis of viral RNA from various parts antigens. In contrast to patients with MIBE, there is often an
of the brain in SSPE suggests that the virus in the nervous sys- intense perivascular mononuclear inflammatory response in
tem is clonal (111), implying that virus entered the brain at brain and high levels of antibody to MeV in serum. There is
one time and then gradually spread throughout the nervous also significant production of MeV-specific antibody by plasma
system. This gradual spread has also been suggested by serial cells residing in the CNS. This locally synthesized antibody
MRI studies (112,113). Therefore, it is most likely that MeV appears in the CSF leading to characteristic elevations in the

Scheld_Ch08.indd 128 2/21/14 5:33 PM


Chapter 8: Measles and Rubella 129

level of CSF immunoglobulin much of which is MeV specific a burst-suppression pattern often most easily demonstrable
(96,135137). Antibody produced in the CNS derives from during sleep (94,156,160), but this may not be present in
clones of resident antibody-secreting B cells and is therefore of adult-onset disease (112). The CSF is often normal on routine
restricted heterogeneity leading to the appearance of oligoclo- analysis of pressure, protein, glucose, and cells. However, the
nal immunoglobulin bands on electrophoretic analysis of the immunoglobulin concentration is usually elevated, oligoclo-
CSF from SSPE patients (138142). Antibodies against the N nal bands are present on electrophoretic analysis, and MeV-
and P proteins are particularly abundant and antibody against specific antibody is elevated (155). MeV RNA can be detected
the M protein is particularly deficient (132,143). Antibody to in the brain and CSF by RT-PCR (112,161,162). CT and MRI
CD9, a tetraspanin protein widely expressed in the CNS, is scans are generally unhelpful. CT scans often show evidence
also elevated, raising the possibility of an autoimmune com- of loss of parenchyma with ventricular dilation and cortical,
ponent to this progressive disease (144). brainstem, and cerebellar atrophy accompanied by low paren-
Experiments in small mammals have shown that treat- chymal attenuation (163). MRI scans tend to show hyperin-
ment with antibody after intracerebral infection with neuro- tense T2-weighted lesions in gray and white matter, with white
adapted strains of MeV attenuates acute disease but increases matter lesions becoming more prominent as disease progresses
the incidence of persistent virus infection and subacute or (112,164166).
chronic encephalitis (81,145). Cases of SSPE have been associ-
ated with passive transfer of immunoglobulin and persistent
infection has been induced experimentally by passive transfer Treatment and Prevention
of antibody (146). Therefore, antibody may contribute to the
establishment and maintenance of persistent nervous system Numerous therapeutic agents including bromodeoxyuridine,
infection. azaguanine, amantadine, interferon, isoprinosine, ribavirin,
The mononuclear inflammatory response in the brain and cimetidine have been used for the treatment of SSPE
includes CD4 and CD8 T cells as well as monocytes and (162,167170). Evaluation of the efficacy of any of these
immunoglobulin-secreting B cells (103,136,147,148). Expression regimens is difficult because the disease is rare, most reports
of class I and class II major histocompatibility antigens is are anecdotal or uncontrolled, and the benefits at best are
increased in brain, and 2-microglobulin, interleukin (IL)-1, sol- short term. Therefore, there is no established treatment for
uble intercellular adhesion molecule (ICAM), soluble IL-2 recep- this disease.
tor, and soluble CD8 are increased in CSF (149,150). Thus, there Timely measles immunization is the most effective means
is no evidence for a global defect in immune responses, although of prevention. The incidence of SSPE has decreased dramati-
MeV induction of interferon- (IFN-) is reduced in some cally since introduction of the live attenuated measles vaccine
individuals (151). It is likely that SSPE and MIBE are similar in and there is no evidence that the vaccine virus can cause SSPE
their pathogenesis, but that MeV replication and the appearance (70,101,102,104,160,171173).
of neurologic symptoms is slowed in SSPE by the presence of a
vigorous immune response (64). However, this immune response
is not able to clear the virus once parenchymal CNS infection has ACUTE DISSEMINATED
been established and infection is progressive.
ENCEPHALOMYELITIS
Clinical Manifestations History
The age of initial MeV infection in individuals subsequently In 1790, James Lucas (174), an English surgeon, described the
developing SSPE is often younger than 2 years. The age of first case of postmeasles encephalomyelitis in a 23-year-old
onset of neurologic disease is typically between 2 and 20 years woman who developed paraparesis and urinary retention as
of age but has been reported up to the age of 35 years the measles rash was fading. ADEM can develop after a num-
(59,68,102,109). The onset is insidious and the diagnosis is ber of infections and is known under many names including
often not suspected early in disease (152,153). The first symp- postinfectious and parainfectious encephalomyelitis (175).
toms are likely to be deterioration in school or work perfor- Acute hemorrhagic leukoencephalitis or acute hemorrhagic
mance and changes in personality (stage I). In adults, visual necrotizing encephalopathy probably represents a more severe
impairment is often an early sign (112,154). Alteration in men- form of the disease. In 1960, Koprowski (176) hypothesized
tal status is followed by the onset of myoclonus, convulsions, that ADEM was an autoimmune (hyperergic) disease based
abnormal postures and movements, and autonomic dysfunc- on the similarity of the pathology and clinical picture to
tion (stage II). Progressive neurologic deterioration is marked experimental autoimmune encephalomyelitis (EAE) and the
by rigidity (stage III), optic atrophy, and akinetic mutism, end- demyelinating disease that complicates immunization with the
ing in coma (stage IV) and death months to years after onset brain-derived Semple rabies vaccine. This remains the lead-
(155157). The course of SSPE is usually 1 to 3 years. More ing hypothesis (55), but understanding of the pathogenesis of
rapid progression has been reported with perinatally acquired ADEM is incomplete.
infection (102,158,159). Occasionally, with good supportive
care, patients survive for longer periods of time and remis-
sions lasting weeks to years with total clinical courses of 10 to Epidemiology
16 years have been reported (119).
ADEM occurs worldwide and complicates many infections
but is most frequent after measles (105,177). The overall
Diagnosis incidence of ADEM associated with measles is approximately
1 per 1,000 infections (52) but varies with age. ADEM is more
The diagnosis of SSPE can be made by demonstration of frequent in children older than 5 years of age and rare in those
high levels of antibody to MeV in CSF in the setting of a younger than 2 years of age (52). It occurs equally in males
characteristic clinical picture. Typical EEG changes include and females.

Scheld_Ch08.indd 129 2/21/14 5:33 PM


130 Part II: Viral Infections and Related Disorders

The disease has a monophasic course over 10 to 20 days.


Pathogenesis and Pathology Improvement is usually evident within a few days after onset.
Mortality ranges from 10% to 40% with substantial neuro-
ADEM occurs in close temporal association with acute measles logic residua in the majority of survivors (55,60,179). Prior
(Fig. 8.2) at a time of an active immune response to the infec- to widespread measles immunization, ADEM was a common
tion (178). Brain pathology shows perivascular inflammation cause of chronic neurologic disability.
and perivenular demyelination. During the acute phase, there
is patchy swelling in the walls of small cerebral vessels and
mononuclear cell infiltration. Later, perivenous inflammation
is more marked and demyelination is evident (60,179,180). Diagnosis
There is little evidence for direct infection of the brain by
MeV. Virus has rarely been recovered from the brain (56,181), The diagnosis is apparent on clinical grounds when neurologic
viral antigen is not detectable by immunocytochemical disease follows shortly after the rash. Routine laboratory tests
staining (21,182), and viral RNA has not been detected by are not particularly helpful. There are no consistent abnor-
in situ hybridization (21). Furthermore, there is no evidence malities in blood or urine. CSF may be normal or contain a
for a local virus-specific intrathecal antibody response as is modest elevation in protein and a mononuclear pleocytosis.
expected in diseases caused by direct virus infection of the The EEG shows a nonspecific diffuse slowing. The CT scan
CNS (55). Therefore, there is no evidence that virus is pres- may be normal, but the MRI scan usually shows multiple foci
ent in the CNS at the time of neurologic disease, but more of demyelination most likely to be seen on T2-weighted and
sensitive techniques such as RT-PCR have not been applied fluid-attenuated inversion recovery (FLAIR) images in subcor-
and specimens from early in disease have not generally been tical and central white matter of the cerebral hemispheres, cer-
available for study. It is possible that virus in cerebral endothe- ebellum, brainstem, and spinal cord (166,177,195).
lial cells plays a role in triggering the autoimmune disease or
allowing access of autoreactive cells (22). Thus, virus may no
longer be detectable by the methods used at the time that tissue Treatment and Prevention
has been examined.
The pattern of loss of the myelin proteins, myelin-associated Measles vaccine is highly effective in prevention of ADEM and
glycoprotein, and myelin basic protein resembles that seen in neurologic complications after immunization are rare (196).
EAE, a disease induced in animals by inoculation of myelin Treatment is not well established. Corticosteroids are widely
or myelin proteins (182). Furthermore, immunologic studies used, but the benefit is not clear (177,197199). One random-
of patients with ADEM have shown the presence of cellular ized study showed no benefit (200) and a retrospective study
immune responses to myelin basic protein (55,181,183) and found higher mortality and rates of sequelae in steroid-treated
antibody to myelin proteins similar to that seen in animals patients (201). Intravenous immunoglobulin and plasma
with EAE and in humans with Semple rabies vaccine-induced exchange have also been used with some reported success
encephalomyelitis (55,184187). It is postulated that ADEM (177,202).
is analogous to these autoimmune diseases, but the mecha-
nism by which an autoimmune response to myelin proteins
is induced during a systemic infection is not clear. The tim-
ing of this complication suggests that immune activation BACKGROUND ON RUBELLA
associated with MeV infection may play a role in induction of
ADEM. Immune activation may increase presentation of self- RV, the etiologic agent of the mild rash disease rubella or
peptides and allow the proliferation of autoreactive clones of German measles, is the only member of the Rubivirus genus
cells (178,188). Patients with ADEM differ from patients with of the Togaviridae family. Togaviruses are single-stranded,
uncomplicated measles by having more marked and prolonged positive-sense, enveloped RNA viruses and all, except RV,
immunologic abnormalities. In particular, IgE is more persis- are in the Alphavirus genus and are transmitted by mosqui-
tently elevated and soluble IL-2 receptor is lower in patients toes. No related animal viruses have been recognized. RV
with ADEM (47,189). IL-2 is one of the cytokines elevated has three structural proteins. The capsid protein surrounds
in plasma (190), and a similar disease has been reported as a the RNA and the two envelope surface glycoproteins E1
complication of IL-2 infusion (191). and E2 are important for attachment and entry. E1 induces
The possibility of immunologic cross reactivity between neutralizing antibodies. Two clades and 10 genotypes are
some component of myelin and a component of MeV has been recognized (203).
explored. Limited sequence homologies have been identified Like MeV, RV is a human virus that is transmitted between
but none for which biologic relevance has been shown (192). individuals by respiratory droplets or aerosol and spreads
Studies of MeV-specific antibodies and T cells have shown no through the blood via a cell-associated viremia to skin and
cross reactivity with myelin basic protein or galactocerebro- other organs (204). RV can infect many types of cells, and
side (193,194). myelin oligodendrocyte glycoprotein has recently been iden-
tified as a cellular receptor (205). The immune response is
associated with the appearance of the rash. There is substan-
tial evidence of immune activation, leukopenia, and immune
Clinical Manifestations suppression, as indicated by loss of skin test responses to recall
antigens (206,207).
Typically, patients recovering from measles present with an The neurologic complications of postnatally acquired
abrupt onset of renewed fever and obtundation accompanied rubella include ADEM, which is in every way similar to
by neurologic signs and symptoms that can include menin- ADEM complicating measles but occurs less frequently
gismus, seizures, altered mental status, multifocal neurologic (approximately 1:5,000 to 8,000 cases) (208), and PRP, a
deficits, and coma (177,180). The onset is most often between disease similar to SSPE. However, the most important neu-
2 and 7 days after the appearance of the rash but occasion- rologic complications occur when RV infection is acquired
ally predates or appears up to 3 weeks after the rash (55). prenatally (Table 8.2).

Scheld_Ch08.indd 130 2/21/14 5:33 PM


Chapter 8: Measles and Rubella 131

TA B L E 8 . 2
SUMMARY OF THE NEUROLOGIC COMPLICATIONS OF RUBELLA

Disease Host Incidence Time of Presentation

Congenital rubella syndrome First trimester fetus 1:2 Birth


(CRS)
Acute disseminated Normal child or adult 1:7,000 12 weeks after rubella
encephalomyelitis (ADEM)
Progressive rubella Prior CRS or rubella Very rare Years after CRS or rubella
panencephalitis (PRP)

CONGENITAL RUBELLA Pathogenesis and Pathology


SYNDROME During the viremia in pregnant women, RV can infect the pla-
centa and spread to the fetus. Fetal infection is systemic, per-
History sistent, and teratogenic. The most significant damage occurs
when infection occurs in the first trimester of pregnancy (212)
The first recognition of the connection between rubella and (Fig. 8.3). Spontaneous abortion is common during this period
birth defects was in 1941 when Norman Gregg (209), an (212). In infants who survive to term, common congenital
Australian ophthalmologist, noted an increase in cases of defects include cardiac abnormalities, hearing loss, low birth
congenital cataracts, frequently accompanied by cardiac weight, and cataracts. Neurologic complications that include
abnormalities, in babies born after an epidemic of rubella. microcephaly, mental retardation, retinopathy, and meningo-
Subsequent studies documented the relationship between encephalitis are common (213215).
maternal rubella early in pregnancy and multiple congenital RV infects vessels and causes obstructive vasculopathy and
defects in the infants born to these mothers. The virus was iso- CNS lesions due to ischemia (216). Vascular lesions include
lated in 1962 and an attenuated live virus vaccine was licensed focal areas of destruction of the walls of arteries and veins,
in 1969 (210,211). pericapillary debris, thickening and proliferation of vessel
walls, and vascular constriction. Foci of parenchymal necrosis
follow the paths of damaged brain vessels and involve both
white and gray matter. In addition, RV infection of CNS cells
Rubella Outcome inhibits cell division leading to microcephaly (217).
Infection is persistent and RV can be isolated from tissues,
blood, CSF, and secretions for 6 to 30 months after birth, with
100 boys clearing virus more slowly than girls (218). Failure to clear
infection may be due to the immaturity of the immune system
at the time of infection (219) and is associated with a num-
% infected ber of RV-specific and nonspecific immune deficits. RV-specific
80 IgM with neutralizing activity is present, but levels of IgG are
low and cellular immune responses are impaired (219221).
Percent of Fetuses

Peripheral blood mononuclear cells (PBMCs) do not proliferate


60
in response to mitogens (222) and responses to routine immuni-
zations are decreased compared to infants without CRS (218).

40
Clinical Manifestations
% with CRS Infants at the time of birth typically show intrauterine growth
retardation and frequently have thrombocytopenia purpura,
hepatomegaly, and splenomegaly. Signs of neural involve-
20
ment include a bulging anterior fontanel, lethargy, irritabil-
ity, psychomotor retardation, and abnormal motor tone.
Other common manifestations of CRS are congenital heart
0 disease, typically patent ductus arteriosus, valvular stenosis
or pulmonary artery stenosis; eye disease, most often cata-
11 1112 1314 1516 1718 1922 2336 36
racts, glaucoma, or retinopathy; and sensorineural hearing
Weeks Gestation at Time of Maternal Rubella loss (213,214). As infants with CRS mature, psychomotor
retardation, behavioral disorders, psychiatric disturbances,
FIGURE 8.3 Diagram showing the likelihood of fetal infection and
of congenital rubella syndrome (CRS) when rubella occurs in the
and autism, as well as endocrinopathies, ocular damage,
mother at various stages of gestation. (Graphed from data in Miller E, and hypertension may become apparent (223225). Prenatal
Cradock-Watson JE, Pollock TM. Consequences of confirmed mater- exposure to RV is also associated with subsequent diagnosis
nal rubella at successive stages of pregnancy. Lancet. 1982;2:781784, of schizophrenia spectrum disorders, often accompanied by a
with permission.) progressive cognitive decline (226,227).

Scheld_Ch08.indd 131 2/21/14 5:33 PM


132 Part II: Viral Infections and Related Disorders

On pathologic examination, white matter is preferentially


Diagnosis affected with mononuclear cell infiltration, microglial nodules,
astrocytosis, and amorphous vascular and perivascular depos-
Subependymal cysts and hypoechogenicity indicative of calci- its (244,245). Cerebellar atrophy is common (244). Inclusion
fication can be detected by cranial ultrasonography (228,229), bodies are not present.
intraventricular and periventricular calcification is often evident Clinical features include new onset of progressive intellec-
on CT scans, and parenchymal lesions are seen on MRI scans tual deterioration, seizures, ataxia, spasticity, and myoclonus
(230,231). CSF protein is typically elevated and RV can often be (246). The diagnosis is usually made by the presence of high
isolated from CSF as well as many other tissues, nasopharyngeal levels of antibody to RV in serum and CSF and the presence of
secretions, and urine. RV RNA can also be detected by RT-PCR. oligoclonal bands containing RV antibody in CSF (247,248).
RV-specific IgM is usually present in serum. The disease is progressive over several years and eventually
fatal. No effective treatment has been identified. Prevention
requires widespread use of the live attenuated rubella vaccine.
Prevention and Treatment
Routine childhood, premarital, and postnatal RV immunization
programs prevent maternal infection and thus CRS (232,233). ACUTE DISSEMINATED
Widespread vaccination has been successful in eliminating ENCEPHALOMYELITIS
rubella and CRS from the Americas (234,235). However,
introduction of rubella vaccine must be carefully planned to Rubella is one of the rash diseases associated with ADEM
avoid increasing the age of infection and thus the risk of CRS (175,208). Symptoms typically occur abruptly within the first
(236238). When rubella is diagnosed in early pregnancy, ther- week after the onset of the rash and often include headache,
apeutic abortion can be performed. Inadvertent immunization vomiting, meningismus, and seizures. Neurologic abnormali-
of pregnant women can result in placental or fetal infection but ties may include evidence of meningitis, transverse myelitis,
rarely results in congenital abnormalities (239,240) and is not radiculitis, and optic neuritis, in addition to encephalitis
considered an indication for therapeutic abortion. (249,250). In addition to ADEM, RV-induced arteritis may
lead to cerebral infarction (249). CSF shows a moderate
increase in mononuclear cells. The EEG is abnormal. RV IgM
PROGRESSIVE RUBELLA is positive, but virus is rarely isolated from CSF or CNS sam-
PANENCEPHALITIS ples (249,250). As with the other neurologic complications of
rubella, immunization will prevent ADEM.
CRS is not usually progressive and virus is gradually cleared.
However, in a few individuals, progressive neurologic dete-
rioration begins many years later. This disease, PRP, was first ACKNOWLEDGMENTS
recognized in 1975 and is characterized by motor and mental
deterioration accompanied by high levels of RV-specific anti- Studies from the authors laboratory were supported by
body in serum and CSF (241,242). The disease has also been research grants from the National Multiple Sclerosis Society,
recognized as an occasional late complication of postnatally the National Institutes of Health (AI23047), and the Bill &
acquired rubella (243). Melinda Gates Foundation.

References
1. Perry RT, Halsey NA. The clinical significance of measles: a review. J Infect 12. Devaux P, Cattaneo R. Measles virus phosphoprotein gene products:
Dis. 2004;189(suppl 1):S4S16. conformational flexibility of the P/V protein amino-terminal domain and
2. Reef SE, Strebel P, Dabbagh A, et al. Progress toward control of rubella and C protein infectivity factor function. J Virol. 2004;78:1163211640.
prevention of congenital rubella syndromeworldwide, 2009. J Infect Dis. 13. Shingai M, Inoue N, Okuno T, et al. Wild-type measles virus infection
2011;204(suppl 1):S24S27. in human CD46/CD150-transgenic mice: CD11c-positive dendritic cells
3. Andrus JK, de Quadros CA, Solorzano CC, et al. Measles and rubella erad- establish systemic viral infection. J Immunol. 2005;175:32523261.
ication in the Americas. Vaccine. 2011;29(suppl 4):D91D96. 14. Kamahora J, Nii S. Pathological and immunological studies of monkeys
4. Morens DM, Holmes EC, Davis AS, et al. Global rinderpest eradica- infected with measles virus. Arch Gesamte Virusforsch.1965;16:161167.
tion: lessons learned and why humans should celebrate too. J Infect Dis. 15. Sergiev PG, Ryazantseva NE, Shroit, IG. The dynamics of pathological pro-
2011;204:502205. cesses in experimental measles in monkeys. Acta Virol. 1960;4:265273.
5. Horzinek MC. Rinderpest: the second viral disease eradicated. Vet 16. Esolen LM, Ward BJ, Moench TR, et al. Infection of monocytes during
Microbiol. 2011;149:295297. measles. J Infect Dis.1993;168:4752.
6. Palosaari H, Parisien JP, Rodriguez JJ, et al. STAT protein interference and 17. Grivel JC, Garcia M, Moss WJ, et al. Inhibition of HIV-1 replica-
suppression of cytokine signal transduction by measles virus V protein. tion in human lymphoid tissues ex vivo by measles virus. J Infect Dis.
J Virol. 2003;77:76357644. 2005;192:7178.
7. Pfaller CK, Conzelmann KK. Measles virus V protein is a decoy substrate 18. McChesney MB, Fujinami RS, Lerche NW, et al. Virus-induced immuno-
for IkappaB kinase alpha and prevents Toll-like receptor 7/9-mediated suppression: infection of peripheral blood mononuclear cells and suppres-
interferon induction. J Virol. 2003;82:1236512373. sion of immunoglobulin synthesis during natural measles virus infection of
8. Obojes K, Andres O, Kim KS, et al. Indoleamine 2,3-dioxygenase mediates rhesus monkeys. J Infect Dis. 1989;159:757760.
cell type-specific anti-measles virus activity of gamma interferon. J Virol. 19. Ludlow M, Allen I, Schneider-Schaulies J. Systemic spread of measles virus:
2005;79:77687776. overcoming the epithelial and endothelial barriers. Thromb Haemost.
9. Caignard G, Bourai M, Jacob Y, et al. Inhibition of IFN-alpha/beta signal- 2009;102:10501056.
ing by two discrete peptides within measles virus V protein that specifically 20. Dittmar S, Harms H, Runkler N, et al. Measles virus-induced block of
bind STAT1 and STAT2. Virology. 2009;383:112120. transendothelial migration of T lymphocytes and infection-mediated virus
10. Devaux P, Hodge G, McChesney MB, et al. Attenuation of V- or C-defective spread across endothelial cell barriers. J Virol. 2008;82:1127311282.
measles viruses: infection control by the inflammatory and interferon 21. Moench TR, Griffin DE, Obriecht CR, et al. Acute measles in patients with
responses of rhesus monkeys. J Virol. 2008;82:53595367. and without neurological involvement: distribution of measles virus anti-
11. Nakatsu Y, Takeda M, Ohno S, et al. Measles virus circumvents the host gen and RNA. J Infect Dis. 1988;158:433442.
interferon response by different actions of the C and V proteins. J Virol. 22. Esolen LM, Takahashi K, Johnson RT, et al. Brain endothelial cell infection
2008;82:82968306. in children with acute fatal measles. J Clin Invest. 1995;96:24782481.

Scheld_Ch08.indd 132 2/21/14 5:33 PM


Chapter 8: Measles and Rubella 133

23. Takahashi H, Umino Y, Sato TA, et al. Detection and comparison of viral 57. Aicardi J, Goutieres F, Arsenio-Nunes ML, et al. Acute measles encephalitis
antigens in measles and rubella rashes. Clin Infect Dis. 1996;22:3639. in children with immunosuppression. Pediatrics. 1977;59:232239.
24. Drig RE, Marcil A, Chopra A, et al. The human CD46 molecule is a recep- 58. Murphy JV, Yunis EJ. Encephalopathy following measles infection in chil-
tor for measles virus (Edmonston strain). Cell. 1993;75:295305. dren with chronic illness. J Pediatr. 1976;88:937942.
25. Naniche D, Varior-Krishnan G, Cervoni F, et al. Human membrane cofactor 59. Modlin JF, Jabbour JT, Witte JJ, et al. Epidemiologic studies of measles,
protein (CD46) acts as a cellular receptor for measles virus. J Virol.1993; measles vaccine, and subacute sclerosing encephalitis. Pediatrics. 1997;59:
67:60256032. 505512.
26. Tatsuo H, Ono N, Tanaka K, et al. SLAM (CDw150) is a cellular receptor 60. Litvak AM, Sands IJ, Gibel H. Encephalitis complicating measles: report
for measles virus. Nature. 2000;406:893897. of fifty-six cases with followup studies in thirty-two. Am J Dis Child.
27. Mhlebach MD, Mateo M, Sinn PL, et al. Adherens junction protein nectin-4 1943;65:265295.
is the epithelial receptor for measles virus. Nature. 2011;480:530533. 61. Wolinsky JS, Swoveland P, Johnson KP, et al. Subacute measles encephalitis
28. Noyce RS, Bondre DG, Ha MN, et al. Tumor cell marker PVRL4 complicating Hodgkins disease in an adult. Ann Neurol. 1977;1:452457.
(Nectin 4) is an epithelial cell receptor for measles virus. PLoS Pathog. 62. Mustafa MM, Weitman SD, Winick NJ, et al. Subacute measles encephali-
2011;7:e1002240. tis in the young immunocompromised host: report of two cases diagnosed
29. McQuaid S, Cosby SL. An immunohistochemical study of the distribution by polymerase chain reaction and treated with ribavirin and review of the
of the measles virus receptors, CD46 and SLAM, in normal human tissues literature. Clin Infect Dis. 1993;16:654660.
and subacute sclerosing panencephalitis. Lab Invest. 2002;82:403409. 63. Graham DG, Gordon A, Ashworth B, et al. Immunodeficiency measles
30. Blau DM, Compans RW. Entry and release of measles virus are polarized in encephalitis. J Clin Lab Immunol. 1983;10:117120.
epithelial cells. Virology. 1995;210:9199. 64. Roos RP, Graves MC, Wollmann RL, et al. Immunologic and virologic
31. Cocks BG, Chang CC, Carballido JM, et al. A novel receptor involved in studies of measles inclusion body encephalitis in an immunosuppressed
T-cell activation. Nature.1995; 376:260263. host: the relationship to subacute sclerosing panencephalitis. Neurology.
32. Okamoto Y, Vricella LA, Moss WJ, et al. Immature CD4 CD8 thy- 1981;31:12631270.
mocytes are preferentially infected by measles virus in human thymic organ 65. Albertyn C, van der Plas H, Hardie D, et al. Silent casualties from the
cultures. PLoS One. 2012;7:e45999. measles outbreak in South Africa. S Afri Med J. 2011;101:313317.
33. Noyce RS, Richardson CD. Nectin 4 is the epithelial cell receptor for mea- 66. Hughes I, Jenney MEM, Newton RW, et al. Measles encephalitis during
sles virus. Trends Microbiol. 2012;20:429439. immunosuppressive treatment for acute lymphoblastic leukemia. Arch Dis
34. Reymond N, Fabre S, Lecocq E, et al. Nectin4/PRR4, a new afadin- Child. 1993;68:775778.
associated member of the nectin family that trans-interacts with nectin1/ 67. Kernahan J, McQuillin J, Craft AW. Measles in children who have malig-
PRR1 through V domain interaction. J Biol Chem. 2001;276:4320543215. nant disease. Br Med J. 1987;295:1518.
35. Yanagi Y, Takeda M, Ohno S. Measles virus: cellular receptors, tropism and 68. Jabbour JT, Duenas DA, Sever JL, et al. Epidemiology of subacute scle-
pathogenesis. J Gen Virol. 2006;87:27672779. rosing panencephalitis: a report of the SSPE registry. J Am Med Assoc.
36. Erlenhfer C, Duprex WP, Rima BK, et al. Analysis of receptor (CD46, 1972;220:959962.
CD150) usage by measles virus. J Gen Virol. 2002; 83:14311436. 69. Detels R, Brody JA, McNew J, et al. Further epidemiological studies of
37. Tahara M, Takeda M, Seki F, et al. Multiple amino acid substitutions in subacute sclerosing panencephalitis. Lancet. 1973;2:1114.
hemagglutinin are necessary for wild-type measles virus to acquire the abil- 70. Bellini WJ, Rota JS, Lowe LE, et al. Subacute sclerosing panencephalitis:
ity to use receptor CD46 efficiently. J Virol. 2007; 81:25642572. more cases of this fatal disease are prevented by measles immunization than
38. Escoffier C, Gerlier D. Infection of chicken embryonic fibroblasts by mea- was previously recognized. J Infect Dis. 2005;192:16861693.
sles virus: adaptation at the virus entry level. J Virol. 1999; 73:52205224. 71. Hecht V. Die Riesenzellenpneumonie im Kindesalter. Beitr Pathol Anat.
39. Kato S, Ohgimoto S, Sharma LB, et al. Reduced ability of hemagglutinin 1910;48:264310.
of the CAM-70 measles virus vaccine strain to use receptors CD46 and 72. Enders JF, McCarthy K, Mitus A, et al. Isolation of measles virus at
SLAM. Vaccine. 2009;27:38383848. autopsy in cases of giant cell pneumonia without rash. N Engl J Med.
40. Sharma LB, Ohgimoto S, Kato S, et al. Contribution of matrix, fusion, 1959;261:875881.
hemagglutinin, and large protein genes of the CAM-70 measles virus vac- 73. Breitfeld V, Hashida Y, Sherman FE, et al. Fatal measles infection in chil-
cine strain to efficient growth in chicken embryonic fibroblasts. J Virol. dren with leukemia. Lab Invest. 1973;28:279291.
2009;83:1164511654. 74. Pullan CR, Noble TC, Scott DJ, et al. Atypical measles infections in leu-
41. Gibbs FA, Gibbs EL, Carpenter PR, et al. Electroencephalographic kaemic children on immunosuppressive treatment. Br Med J. 1976;1:
abnormality in uncomplicated childhood diseases. J Am Med Assoc. 15621565.
1959;171:10501055. 75. Bitnun A, Shannon P, Durward A, et al. Measles inclusion-body encephali-
42. Hnninen P, Arstila P, Lang H, et al. Involvement of the central nervous tis caused by the vaccine strain of measles virus. Clin Infect Dis. 1999;29:
system in acute, uncomplicated measles virus infection. J Clin Microbiol. 855861.
1980;11:610613. 76. Kirk JL, Zhou AL, McQuaid S, et al. Cerebral endothelial cell infection
43. Pampiglione G. Prodromal phase of measles: some neurophysiological by measles virus in subacute sclerosing panencephalitis: ultrastructural
studies. Br Med J. 1964;2:12961300. and in situ hybridization evidence. Neuropathol Appl Neurobiol. 1991;17:
44. Pampiglione G, Griffith AH, Bramwell EC. Transient cerebral changes after 289297.
vaccination against measles. Lancet. 1971;2:58. 77. Haltia M, Paetau A, Vaheri A, et al. Fatal measles encephalopathy with reti-
45. Griffin DE. Measles virus. In Knipe DM, Howley PM, Griffin DE, et al, nopathy during cytotoxic chemotherapy. J Neurol Sci. 1977;32:323330.
eds. Fields Virology. 6th ed. Philadelphia: Lippincott Williams & Wilkins; 78. Baczko K, Liebert UG, Cattaneo R, et al. Restriction of measles virus
2013. gene expression in measles inclusion body encephalitis. J Infect Dis.
46. Griffin DE, Ward BJ, Jauregui E, et al. Immune activation during measles: 1988;158:144150.
interferon-gamma and neopterin in plasma and cerebrospinal fluid in com- 79. Ohuchi M, Ohuchi R, Homma M. Mode of subacute sclerosing panen-
plicated and uncomplicated disease. J Infect Dis. 1990;161:449453. cephalitis (SSPE) virus infection in tissue culture cells. Microbiol Immunol.
47. Griffin DE, Ward BJ, Jauregui E, et al. Immune activation during measles. 1981;225:887893.
N Engl J Med. 1989;320:16671672. 80. Cattaneo R, Rebmann G, Baczko K, et al. Altered ratios of measles virus
48. Griffin DE, Ward BJ, Jauregui E, et al. Immune activation during measles: transcripts in diseased human brains. Virology. 1987;160:523526.
beta-2-microglobulin in plasma and cerebrospinal fluid in complicated and 81. Liebert UG, Flanagan SG, Loffler S, et al. Antigenic determinants of
uncomplicated disease. J Infect Dis. 1992;166:11701173. measles virus haemagglutinin associated with neurovirulence. J Virol.
49. Tamashiro VG, Perez HH, Griffin DE. Prospective study of the magnitude 1994;68:14861493.
and duration of changes in tuberculin reactivity during complicated and 82. Cattaneo R, Schmid A, Billeter MA, et al. Multiple viral mutations rather
uncomplicated measles. Pediatr Infect Dis J. 1987;6:451454. than host factors cause defective measles virus gene expression in a sub-
50. von Pirquet C. Verhalten der kutanen tuberkulin-reaktion wahrend der acute sclerosing panencephalitis cell line. J Virol. 1988;62:13881397.
Masern. Deutsch Med Wochenschr. 1908;34:12971300. 83. Patterson JB, Cornu TI, Redwine J, et al. Evidence that the hypermutated
51. Mokhtarian F, Brooks BR, Griffin DE. Inhibition of chemotaxis by the CSF M protein of a subacute sclerosing panencephalitis measles virus actively
of prednisone-treated multiple sclerosis patients. Neurology. 33:121. contributes to the chronic progressive CNS disease. Virology. 2001;291:
52. Miller DL. Frequency of complications of measles, 1963. Br Med J. 215225.
1964;2:7578. 84. Cattaneo R, Billeter MA. Mutations and A/I hypermutations in measles
53. Lin WH, Kouyos RD, Adams RJ, et al. Prolonged persistence of measles virus persistent infections. Curr Top Microbiol Immunol. 1992;176:6374.
virus RNA is characteristic of primary infection dynamics. Proc Natl Acad 85. Ehrengruber MU, Ehler E, Billeter M, et al. Measles virus spreads in rat
Sci U S A. 2012;109:1498914994. hippocampal neurons by cell-to-cell contact and in a polarized fashion.
54. Griffin DE, Lin WH, Pan CH. Measles virus, immune control and persis- J Virol. 2002;76:57205728.
tence. FEMS Microbiol Rev. 2012;36(3):649662. 86. Makhortova NR, Askovich P, Patterson CE, et al. Neurokinin-1 enables
55. Johnson RT, Griffin DE, Hirsch RL, et al. Measles encephalomyelitis measles virus trans-synaptic spread in neurons. Virology. 2007;362:
clinical and immunologic studies. N Engl J Med. 1984;310:137141. 235244.
56. McLean DM, Best JM, Smith PA, et al. Viral infections of Toronto children 87. Lawrence DM, Patterson CE, Gales TL, et al. Measles virus spread between
during 1965: II. Measles encephalitis and other complications. Can Med neurons requires cell contact but not CD46 expression, syncytium forma-
Assoc J. 1966;94:905910. tion, or extracellular virus production. J Virol. 2000;74:19081918.

Scheld_Ch08.indd 133 2/21/14 5:33 PM


134 Part II: Viral Infections and Related Disorders

88. Iwasaki Y, Koprowski H. Cell to cell transmission of virus in the cen- 118. Brown HR, Goller NL, Rudelli RD, et al. Postmortem detection of mea-
tral nervous system. I. Subacute sclerosing panencephalitis. Lab Invest. sles virus in non-neural tissues in subacute sclerosing panencephalitis. Ann
1974;31:187196. Neurol. 1989;26:263268.
89. Poon TP, Tchertkoff V, Win H. Subacute measles encephalitis with AIDS 119. Dubois-Dalcq M, Coblentz JM, Pleet AB. Subacute sclerosing panenceph-
diagnosed by fine needle aspiration biopsy. A case report. Acta Cytol. alitis: unusual nuclear inclusions and lengthy clinical course. Arch Neurol.
1998;42:729733. 1974;31:355363.
90. Chien LT, Price RA, Murti KG, et al. Fatal subacute immunosuppres- 120. Duprex WP, McQuaid S, Roscic-Mrkic B, et al. In vitro and in vivo infec-
sive measles encephalitis (SIME) in children with acute lymphocytic leu- tion of neural cells by a recombinant measles virus expressing enhanced
kemiaclinical, electroencephalographic, and computerized tomographic green fluorescent protein. J Virol. 2000;74:79727979.
scan features. Clin Electroencephalogr. 1983;14:214220. 121. Katz M, Koprowski H. The significance of failure to isolate infec-
91. Dawson JR. Cellular inclusions in cerebral lesions of lethargic encephali- tious viruses in cases of subacute sclerosing panencephalitis. Arch ges
tis. Am J Pathol. 1993;9:715. Virusforsch. 1973;41:390393.
92. Dawson JR. Cellular inclusions in cerebral lesions of epidemic encephali- 122. Wong TC, Ayata M, Hirano A, et al. Generalized and localized biased
tis. Arch Neurol Psych. 1934;31:685700. hypermutation affecting the matrix gene of a measles virus strain that
93. Bogaert LV. Une leuco-encephalite sclerosante subaigue. J Neurol causes subacute sclerosing panencephalitis. J Virol. 1989;63:54645468.
Neurosurg Psych. 1945;8:101120. 123. Schmid A, Spielhofer P, Cattaneo R, et al. Subacute sclerosing panencepha-
94. Foley J, Williams D. Inclusion encephalitis and its relation to subacute litis is typically characterized by alterations in the fusion protein cytoplas-
sclerosing leucoencephalitis: a report of five cases. Q J Med. 1953;22: mic domain of the persisting measles virus. Virology. 1992;188:910915.
157198. 124. Ayata M, Hirano A, Wong TC. Structural defect linked to nonrandom
95. Tellez-Nagel I, Harter DH. Subacute sclerosing leukoencephalitis: ultra- mutations in the matrix gene of Biken strain of subacute sclerosing pan-
structure of intranuclear and intracytoplasmic inclusions. Science. 1966; encephalitis virus defined by cDNA cloning and expression of chimeric
154:899901. genes. J Virol. 1989;63:11621173.
96. Connolly JH, Allen IV, Hurwitz LJ, et al. Measles virus antibody and 125. Cattaneo R, Schmid A, Eschle D, et al. Biased hypermutation and other
antigen in subacute sclerosing panencephalitis. Lancet. 1967;1:542544. genetic changes in defective measles viruses in human brain infections.
97. Dayan AD, Gostling JVT, Greaves JL, et al. Evidence of a pseudomyxo- Cell. 1988;55:255265.
virus in the brain in subacute sclerosing leucoencephalitis. Lancet. 126. Cattaneo R, Schmid A, Spielhofer P, et al. Mutated and hypermutated
1967;1:980981. genes of persistent measles viruses which caused lethal human brain dis-
98. Freeman JM, Magoffin RL, Lennette EH, et al. Additional evidence of the eases. Virology. 1989;173:415425.
relation between subacute inclusion-body encephalitis and measles virus. 127. Baczko K, Liebert UG, Billeter M, et al. Expression of defective measles
Lancet. 1967;2:129131. virus genes in brain tissues of patients with subacute sclerosing panen-
99. Chen TT, Watanabe I, Zeman W, et al. Subacute sclerosing panenceph- cephalitis. J Virol. 1986;59:472478.
alitis: propagation of measles virus from brain biopsy in tissue culture. 128. Hall WW, Choppin PW. Measles-virus proteins in the brain tissue
Science. 1969;163:11931194. of patients with subacute sclerosing panencephalitis. N Engl J Med.
100. Payne FE, Baublis JV, Itabashi HH. Isolation of measles virus from cell 1981;304:11521155.
cultures of brain from a patient with subacute sclerosing panencephalitis. 129. Sidhu MS, Crowley J, Lowenthal A, et al. Defective measles virus in human
N Engl J Med. 1969;281:585. subacute sclerosing panencephalitis brain. Virology. 1994;202:631641.
101. Takasu T, Mgone JM, Mgone CS, et al. A continuing high incidence of 130. Liebert UG, Baczko K, Budka H, et al. Restricted expression of measles
subacute sclerosing panencephalitis (SSPE) in the Eastern Highlands of virus proteins in brains from cases of subacute sclerosing panencephalitis.
Papua New Guinea. Epidemiol Infect. 2003;131:887898. J Gen Virol. 1986;67:24352444.
102. Campbell H, Andrews N, Brown KE, et al. Review of the effect of mea- 131. Payne FE, Baublis JV. Decreased reactivity of SSPE strains of measles virus
sles vaccination on the epidemiology of SSPE. Int J Epidemiol. 2007;36: with antibody. J Infect Dis. 1973;127:505511.
13341348. 132. Dhib-Jalbut S, McFarland HF, Mingioli ES, et al. Humoral and cellular
103. Anlar B, Soylemezoglu F, Aysun S, et al. Tissue inflammatory response immune responses to matrix protein of measles virus in subacute scleros-
in subacute sclerosing panencephalitis (SSPE). J Child Neurol. 2001;16: ing panencephalitis. J Virol. 1988;62:24832489.
895900. 133. Hall WW, Choppin PW. Evidence for lack of synthesis of the M polypep-
104. Miller C, Farrington CP, Harbert K. The epidemiology of subacute scle- tide of measles virus in brain cells in subacute sclerosing panencephalitis.
rosing panencephalitis in England and Wales 19701989. Int J Epidemiol. Virology. 1979;99:443447.
1992;21:9981006. 134. Cattaneo R, Rose JK. Cell fusion by the envelope glycoproteins of per-
105. Halsey NA, Modlin JF, Jabbour JT, et al. Risk factors in subacute scle- sistent measles viruses which caused lethal human brain disease. J Virol.
rosing panencephalitis: a case-control study. Am J Epidemiol. 1980;111: 1993;67:14931502.
415424. 135. Appelbaum E, Greenberg M, Nelson J. Neurological complications fol-
106. Modlin JF, Halsey NA, Eddins DL, et al. Epidemiology of subacute scle- lowing antirabies vaccination. J Am Med Assoc. 1953;151:188191.
rosing panencephalitis. J Ped. 1970;94:231236. 136. Esiri MM, Oppenheimer DR, Brownell B, et al. Distribution of measles
107. Zilber N, Rannon L, Alter M, et al. Measles, measles vaccination and antigen and immunoglobulin containing cells in the CNS in subacute scle-
risk of subacute sclerosing panencephalitis (SSPE). Neurology. 1983;33: rosing panencephalitis (SSPE) and atypical measles encephalitis. J Neurol
15581564. Sci. 1982;53:2943.
108. Halsey NA, Modlin JF, Jabbour JT. Subacute sclerosing panencephalitis 137. Tourtellotte WW, Ma BI, Brandes DR, et al. Quantification of de novo
(SSPE): an epidemiologic review. In: Stevens JG, Todaro GJ, Fox CF, eds. central nervous system IgG measles antibody synthesis in SSPE. Ann
Persistent Viruses. New York: Academic Press; 1978:101114. Neurol. 1981;9:551556.
109. Khare S, Kumari S, Verghese T. Subacute sclerosing panencephalitis in 138. Lin FH, Mehta PD, Thormar H. Precipitation of measles virus proteins
Delhi. J Trop Ped. 1994;40:326328. by immunoglobulin G fractions containing groups of oligoclonal bands
110. Isaacson SH, Asher DM, Godec MS, et al. Widespread, restricted low- isolated from sera of patients with subacute sclerosing panencephalitis.
level measles virus infection of brain in a case of subacute sclerosing pan- Infect Immun. 1982;37:393396.
encephalitis. Acta Neuropathol (Berl). 1996;91:135139. 139. Ebers GC, Zabriskie JB, Kunkel HG. Oligoclonal immunoglobulins in
111. Baczko K, Lampe J, Liebert UG, et al. Clonal expansion of hypermutated subacute sclerosing panencephalitis and multiple sclerosis: a study of idio-
measles virus in a SSPE brain. Virology. 1993;197:188195. typic determinants. Clin Exp Immunol. 1979;35:6775.
112. Mawrin C, Lins H, Koenig B, et al. Spatial and temporal disease pro- 140. Vandvik B, Norrby E, Nordal HJ, et al. Oligoclonal measles virus specific
gression of adult-onset subacute sclerosing panencephalitis. Neurol. IgG antibodies isolated from cerebrospinal fluids, brain extracts and sera
2002;58:15681571. from patients with subacute sclerosing panencephalitis and multiple scle-
113. Sawaishi Y, Yano T, Watanabe Y, et al. Migratory basal ganglia lesions in rosis. Scand J Immunol. 1976;5:979992.
subacute sclerosing panencephalitis (SSPE): clinical implications of axonal 141. Strosberg AD, Karcher D, Lowenthal A. Structural homogeneity of
spread. J Neurol Sci. 1999;168:137140. human subacute sclerosing panencephalitis antibodies. J Immunol.
114. Herndon RM, Rubinstein LJ. Light and electron microscopy observations on 1975;115:157160.
the development of viral particles in the inclusions of Dawsons encephalitis 142. Burgoon MP, Owens GP, Smith-Jensen T, et al. Cloning the antibody
(subacute sclerosing panencephalitis). Neurology. 1968;18:820. response in humans with inflammatory central nervous system disease:
115. Brody JA, Detels R, Sever JL. Measles-antibody titres in sibships of analysis of the expressed IgG repertoire in subacute sclerosing panen-
patients with subacute sclerosing panencephalitis and controls. Lancet. cephalitis brain reveals disease-relevant antibodies that recognize specific
1972;1:177178. measles virus antigens. J Immunol. 1999;163:34963502.
116. Tourtellotte WW, Parker JA, Herndon RM, et al. Subacute sclerosing pan- 143. Wechsler SL, Weiner HL, Fields BN. Immune response in subacute scle-
encephalitis: brain immunoglobulin-G, measles antibody and albumin. rosing panencephalitis: reduced antibody response to the matrix protein
Neurology. 1968;18:117121. of measles virus. J Immunol. 1979;123:884889.
117. Oyanagi S, Rorke LB, Katz M, et al. Histopathology and electron micros- 144. Shimizu T, Matsuishi T, Iwamoto R, et al. Elevated levels of anti-CD9
copy of three cases of subacute sclerosing panencephalitis (SSPE). Acta antibodies in the cerebrospinal fluid of patients with subacute sclerosing
Neuropathol. 1971;18:5873. panencephalitis. J Infect Dis. 2002;185:13461350.

Scheld_Ch08.indd 134 2/21/14 5:33 PM


Chapter 8: Measles and Rubella 135

145. Rammohan KW, McFarland HF, McFarlin DE. Induction of subacute 174. Lucas J. An account of uncommon symptoms succeeding the measles;
murine measles encephalitis by monoclonal antibody to virus haemag- with additional remarks on the infection of measles and smallpox.
glutinin. Nature. 1981;290:588589. London Med J. 1790;11:325331.
146. Rammohan KW, McFarland HF, McFarlin DE. Subacute sclerosing panen- 175. Hung KL, Liao HT, Tsai ML. Postinfectious encephalomyelitis: etiologic
cephalitis after passive immunization and natural measles infection: role of and diagnostic trends. J Child Neurol. 2000;15:666670.
antibody in persistence of measles virus. Neurology. 1982;32:390394. 176. Koprowski H. The role of hyperergy in measles encephalitis. Amer. J Dis
147. Hofman FM, Hinton DR, Baemayr J, et al. Lymphokines and immunoreg- Child. 1962;103:273278.
ulatory molecules in subacute sclerosing panencephalitis. Clin Immunol 177. Noorbakhsh F, Johnson RT, Emery D, et al. Acute disseminated encepha-
Immunopathol. 1991;58:331342. lomyelitis: clinical and pathogenesis features. Neurol Clin. 2008;26:
148. Nagano I, Nakamura S, Yoshioka M, et al. Immunocytochemical analysis 759780.
of the cellular infiltrate in brain lesions in subacute sclerosing panencepha- 178. Griffin DE. Immune responses during measles virus infection. Curr Top
litis. Neurology. 1991;41:16391642. Microbiol Immunol. 1995;191:117134.
149. Mehta PD, Kulczycki J, Mehta SP, et al. Increased levels of beta- 179. Miller HG, Stanton JB. Neurological sequelae of prophylactic inocula-
2-microglobulin, soluble interleukin-2 receptor, and soluble CD8 tion. Q J Med. 1954;89:127.
in patients with subacute sclerosing panencephalitis. Clin Immunol 180. Young NP, Weinshenker BG, Parisi JE, et al. Perivenous demyelination:
Immunopathol. 1992;65:5359. association with clinically defined acute disseminated encephalomyelitis
150. Mehta PD, Kulczycki J, Mehta SP, et al. Increased levels of interleukin-1 and and comparison with pathologically confirmed multiple sclerosis. Brain.
Soluble intercellular adhesion molecule-1 in cerebrospinal fluid of patients 2010;133:333348.
with subacute sclerosing panencephalitis. J Infect Dis. 1997;175:689692. 181. Lisak RP, Behan PO, Zweiman B, et al. Cell-mediated immunity to
151. Hara T, Yamashita S, Aiba H, et al. Measles virus-specific T helper 1/T myelin basic protein in acute disseminated encephalomyelitis. Neurology.
helper 2-cytokine production in subacute sclerosing panencephalitis. 1974;24:560564.
J Neurovirol. 2000;6:121126. 182. Gendelman HE, Wolinsky JS, Johnson RT, et al. Measles encephalitis: lack
152. Prashanth LK, Taly AB, Sinha S, et al. Subacute sclerosing panencephalitis of evidence of viral invasion of the central nervous system and quantita-
(SSPE): an insight into the diagnostic errors from a tertiary care university tive study of the nature of demyelination. Ann Neurol. 1984;15:353360.
hospital. J Child Neurol. 2007;22:683688. 183. Lisak RP, Zweiman B. 1977. In vitro cell-mediated immunity of cerebro-
153. Honarmand S, Glaser CA, Chow E, et al. Subacute sclerosing pan- spinal fluid lymphocytes to myelin basic protein in primary demyelinating
encephalitis in the differential diagnosis of encephalitis. Neurology. diseases. N Engl J Med. 1977;297:850853.
2004;63:14891493. 184. Brilot F, Dale RC, Selter RC, et al. Antibodies to native myelin oligoden-
154. Singer C, Lang AE, Suchowersky O. Adult-onset subacute sclerosing drocyte glycoprotein in children with inflammatory demyelinating central
panencephalitis: case reports and review of the literature. Mov Disord. nervous system disease. Ann Neurol. 2009;66:833842.
1997;12:342353. 185. Lalive PH, Husler MG, Maurey H, et al. Highly reactive anti-myelin oli-
155. Jabbour JT, Garcia JH, Lemmi H, et al. Subacute sclerosing panen- godendrocyte glycoprotein antibodies differentiate demyelinating diseases
cephalitis: a multidisciplinary study of eight cases. J Am Med Assoc. from viral encephalitis in children. Mult Scler. 2011;17:297302.
1969;207:22482254. 186. Di Pauli F, Mader S, Rostasy K, et al. Temporal dynamics of anti-MOG
156. Freeman JM. The clinical spectrum and early diagnosis of Dawsons antibodies in CNS demyelinating diseases. Clin Immunol. 2001;138:
encephalitis. J Ped. 1969;75:590603. 247254.
157. Metz H, Gregoriou M, Sandifer P. Subacute sclerosing panencephalitis: 187. Hemachudha T, Phanthumchinda K, Phanuphak P, et al. Myoedema as a
a review of 17 cases with special reference to clinical diagnostic criteria. clinical sign in paralytic rabies. Lancet. 1987;1:1210.
Arch Dis Child. 1964;39:554557. 188. Herberts CA, van Gaans-van den Brink J, van der Heeft E, et al.
158. Zwiauer K, Forstenpointner E, Popow-Kraupp T, et al. Rapid progres- Autoreactivity against induced or upregulated abundant self-peptides
sive subacute sclerosing panencephalitis after perinatally acquired measles in HLA-A*0201 following measles virus infection. Hum Immunol.
virus infection. Lancet. 1995;345:1124. 2003;64:4455.
159. Simsek E, Ozturk A, Yavuz C, et al. Subacute sclerosing panencephali- 189. Griffin DE, Cooper SJ, Hirsch RL, et al. Changes in plasma IgE levels
tis (SSPE) associated with congenital measles infection. Turk J Pediatr. during complicated and uncomplicated measles virus infections. J Allergy
2005;47:5862. Clin Immunol. 1985;76:206213.
160. Garg RK. Subacute sclerosing panencephalitis. Postgrad Med J. 2002; 190. Griffin DE, Ward BJ. Differential CD4 T cell activation in measles. J Infect
78:6370. Dis. 1993;168:275281.
161. Godec MS, Asher DM, Swoveland PT, et al. Detection of measles virus 191. Goretsky M. Acute fatal leukoencephalopathy after interleukin-2 therapy.
genomic sequences in SSPE brain tissue by the polymerase chain reaction. N Engl J Med.1990; 323:11461147.
J Med Virol. 1990;30:237244. 192. Jahnke U, Fischer EH, Alvord EC Jr. Sequence homology between cer-
162. Tomoda A, Shiraishi S, Hosoya M, et al. Combined treatment with inter- tain viral proteins and proteins related to encephalomyelitis and neuritis.
feron-alpha and ribavirin for subacute sclerosing panencephalitis. Pediatr Science. 1985;229:282284.
Neurol. 2001;24:5459. 193. Richert JR, Robinson ED, Reuben-Burnside CA, et al. Measles virus
163. Krawiecki NS, Dyken PR, El Gammal T, et al. Computed tomogra- specific human T cell clones: studies of alloreactivity and antigenic cross
phy of the brain in subacute sclerosing panencephalitis. Ann Neurol. reactivity. J Neuroimmunol. 1988;19:5968.
1984;15:489493. 194. Rubio N, Cuesta A. Lack of cross-reaction between myelin basic pro-
164. Anlar B, Saati I, Kse G, et al. MRI findings in subacute sclerosing pan- teins and putative demyelinating virus envelope proteins. Mol Immunol.
encephalitis. Neurology. 1996; 47:12781283. 1988;26:663668.
165. Oztrk A, Grses C, Baykan B, et al. Subacute sclerosing panencephalitis: 195. Atlas SW, Grossman RI, Goldberg HI, et al.. MR diagnosis of acute dis-
clinical and magnetic resonance imaging evaluation of 36 patients. J Child seminated encephalomyelitis. J Comput Assist Tomogr. 1986;10:798801.
Neurol. 2002;17:2529. 196. Landrigan PJ, Witte JJ. Neurologic disorders following live measles-virus
166. Yilmaz K, Yilmaz M, Mete A, et al. A correlative study of FDG PET, MRI/ vaccination. JAMA. 1973;223:14591462.
CT, electroencephalography, and clinical features in subacute sclerosing 197. Tenembaum S, Chitnis T, Ness J, et al. Acute disseminated encephalomy-
panencephalitis. Clin Nucl Med. 2010;35:675681. elitis. Neurology. 2007;68:S23S36.
167. Anlar B, Gcyener K, Imir T, et al. Cimetidine as an immunomodulator 198. McDaneld LM, Fields JD, Bourdette DN, et al. Immunomodulatory ther-
in subacute sclerosing panencephalitis: a double blind, placebo-controlled apies in neurologic critical care. Neurocrit Care. 2010;12:132143.
study. Pediatr Infect Dis J. 1993;12:578581. 199. Tenembaum S, Chamoles N, Fejerman N. Acute disseminated encephalo-
168. Yalaz K, Anlar B, Oktem F, et al. Intraventricular interferon and oral ino- myelitis: a long-term follow-up study of 84 pediatric patients. Neurology.
siplex in the treatment of subacute sclerosing panencephalitis. Neurology. 2002;59:12241231.
1992;42:488491. 200. Ziegra SR. Corticosteroid treatment for measles encephalitis. J Pediatr.
169. Gutierrez J, Issacson RS, Koppel BS. Subacute sclerosing panencephalitis: 1961;59:322323.
an update. Dev Med Child Neurol. 2010;52:901907. 201. Boe J, Solberg CO, Saeter T. Corticosteroid treatment of acute meningoen-
170. Anlar B, Aydin OF, Guven A, et al. Retrospective evaluation of inter- cephalitis: a retrospective study of 346 cases. Br Med J. 1965;1:10941095.
feron-beta treatment in subacute sclerosing panencephalitis. Clin Ther. 202. Ravaglia S, Piccolo G, Ceroni M, et al. Severe steroid-resistant post-
2004;26:18901894. infectious encephalomyelitis: general features and effects of IVIg.
171. Suga S, Miyahara M, Obata M, et al. Detection by polymerase chain J Neurol. 2007;254:15181523.
reaction of wild-type measles virus genome in the cerebrospinal fluid 203. Zhou Y, Ushijima H, Frey TK. Genomic analysis of diverse rubella virus
of a patient with SSPE who had received measles vaccine. Clin Diagn genotypes. J Gen Virol. 2007;88:932941.
Virol.1996;5:3742. 204. Heggie AD. Pathogenesis of the rubella-exanthem: distribution of rubella
172. Bale JF Jr. Subacute sclerosing panencephalitis: a preventable disease that virus in the skin during rubella with and without rash. J Infect Dis.
needs to be prevented. Neurology. 2004;63:13521353. 1978;137:7477.
173. Dyken PR, Cunningham SC, Ward LC. Changing character of sub- 205. Cong H, Jiang Y, Tien P. Identification of the myelin oligodendrocyte
acute sclerosing panencephalitis in the United States. Pediatr Neurol. glycoprotein as a cellular receptor for rubella virus. J Virol. 2011;85:
1989;5:339341. 1103811047.

Scheld_Ch08.indd 135 2/21/14 5:33 PM


136 Part II: Viral Infections and Related Disorders

206. Hyypi T, Eskola J, Laine M, et al. Polyclonal activation of B cells during 230. Lane B, Sullivan EV, Lim KO, et al. White matter MR hyperintensi-
rubella infection. Scand J Immunol. 1985;21:615617. ties in adult patients with congenital rubella. AJNR Am J Neuroradiol.
207. Niwa Y, Kanoh T. Immunological behaviour following rubella infection. 1996;17:99103.
Clin Exp Immunol. 1979;37:470476. 231. Sugita K, Ando M, Makino M, et al. Magnetic resonance imaging of
208. Ueda K, Sasaki K, Tokugawa K, et al. The 19761977 rubella epidemic the brain in congenital rubella virus and cytomegalovirus infections.
in Fukuoka city in southern Japan: epidemiology and incidences of com- Neuroradiology. 1991;33:239242.
plications among 80,000 persons who were school children at 28 primary 232. Schluter WW, Reef SE, Redd SC, et al. Changing epidemiology of con-
schools and their family members. Biken J. 1984;27:161168. genital rubella syndrome in the United States. J Infect Dis. 1998;178:
209. Gregg NM. Congenital cataract following German measles in mother. 636641.
Tr Ophth Soc Australia. 1941;3:3546. 233. Berger BE, Navar-Boggan AM, Omer SB. Congenital rubella syndrome
210. Parkman PD, Buescher EL, Artenstein MS. Recovery of rubella virus from and autism spectrum disorder prevented by rubella vaccinationUnited
army recruits. Proc Soc Exp Biol Med. 1962;111:225230. States, 20012010. BMC Public Health. 2011;11:340.
211. Weller TH, Neva FA. Propagation in tissue culture of cytopathic 234. Castillo-Solrzano C, Marsigli C, Bravo-Alcntara P, et al. Elimination
agents from patients with rubella-like illness. Proc Soc Exp Biol Med. of rubella and congenital rubella syndrome in the Americas. J Infect Dis.
1962;111:215225. 2011;204(suppl 2):S571S578.
212. Miller E, Cradock-Watson JE, Pollock TM. Consequences of confirmed 235. Reef SE, Redd SB, Abernathy E, et al. Evidence used to support the
maternal rubella at successive stages of pregnancy. Lancet. 1982;2: achievement and maintenance of elimination of rubella and congeni-
781784. tal rubella syndrome in the United States. J Infect Dis. 204(suppl 2):
213. Cooper LZ, Krugman S. Clinical manifestations of postnatal and congeni- S593S597.
tal rubella. Arch Ophthalmol. 1967;77:434439. 236. Morice A, Ulloa-Gutierrez R, Avila-Agero ML. Congenital rubella syn-
214. Lindquist JM, Plotkin SA, Shaw L, et al. Congenital rubella syndrome as a drome: progress and future challenges. Expert Rev Vaccines. 2009;8:
systemic infection. Studies of affected infants born in Philadelphia, U.S.A. 323331.
Br Med J. 1965;2:14011406. 237. Strebel PM, Gacic-Dobo M, Reef S, et al. Global use of rubella vaccines,
215. Munro ND, Sheppard S, Smithells RW, et al. Temporal relations between 19802009. J Infect Dis. 2011;204(suppl 2):S579S584.
maternal rubella and congenital defects. Lancet. 1987;2:201204. 238. Cutts FT, Metcalf CJ, Lessler J, et al. Rubella vaccination: must not be
216. Rorke LB. Nervous system lesions in the congenital rubella syndrome. business as usual. Lancet. 2012;380:217218.
Arch Otolaryngol. 1973;98:249251. 239. Modlin JF, Herrmann K, Brandling-Bennett AD, et al. Risk of congenital
217. Kemper TL, Lecours AR, Gates MJ, et al. Retardation of the myelo- and abnormality after inadvertent rubella vaccination of pregnant women.
cytoarchitectonic maturation of the brain in the congenital rubella syn- N Engl J Med. 1976;294:972974.
drome. Res Pub Assoc Res Nerv Ment Dis. 1973;51:2362. 240. Soares RC, Siqueira MM, Toscano CM, et al. Follow-up study of
218. Michaels RH. Immunologic aspects of congenital rubella. Pediatrics. unknowingly pregnant women vaccinated against rubella in Brazil,
1969;43:339350. 20012002. J Infect Dis. 2011;204(suppl 2):S729S736.
219. Meitsch K, Enders G, Wolinsky JS, et al. The role of rubella-immunoblot 241. Townsend JJ, Baringer JR, Wolinsky JS, et al. Progressive rubella
and rubella-peptide-EIA for the diagnosis of the congenital rubella syn- panencephalitis: late onset after congenital rubella. N Engl J Med.
drome during the prenatal and newborn periods. J Med Virol. 1997;51: 1975;292:990993.
280283. 242. Weil ML, Itabashi HH, Cremer NE, et al. Chronic progressive panenceph-
220. Fuccillo DA, Steele RW, Hensen SA, et al. Impaired cellular immunity to alitis due to rubella virus simulating subacute sclerosing panencephalitis.
rubella virus in congenital rubella. Infect Immun. 1974;9:8184. N Engl J Med. 1975;292:994998.
221. de Mazancourt A, Waxham MN, Nicolas JC, et al. Antibody response to 243. Wolinsky JS, Berg BO, Maitalnd,CH. Progressive rubella panencephalitis.
the rubella virus structural proteins in infants with the congenital rubella Arch Neurol. 1976;33:722723.
syndrome. J Med Virol. 1986;19:111122. 244. Townsend JJ, Wolinsky JS, Baringer JR. The neuropathology of progres-
222. Olson GB, South MA, and Good RA. Phytohaemagglutinin unrespon- sive rubella panencephalitis of late onset. Brain. 1976;99:8190.
siveness of lymphocytes from babies with congenital rubella. Nature. 245. Townsend JJ, Stroop WG, Baringer JR, et al. Neuropathology of pro-
1967;214:696. gressive rubella panencephalitis after childhood rubella. Neurology.
223. Sever JL, South MA, Shaver KA. Delayed manifestations of congenital 1982;32:185190.
rubella. Rev Infect Dis. 1985;7(suppl 1):S164S169. 246. Guizzaro A, Volpe E, Lus G, et al. Progressive rubella panencephalitis.
224. Chess S, Fernandez P, Korn S. Behavioral consequences of congenital Follow-up EEG study of a case. Acta Neurol (Napoli). 1992;14:485492.
rubella. J Pediatr. 1978;93:699703. 247. Vandvik B, Weil ML, Grandien M, et al. Progressive rubella panencepha-
225. Chess S. Autism in children with congenital rubella. J Autism Child litis: synthesis of oligoclonal virus-specific IgG antibodies and homoge-
Schizophr. 1971;1:3347. neous free light chains in the central nervous system. Acta Neurol Scand.
226. Brown AS, Cohen P, Greenwald S, et al. Nonaffective psychosis after pre- 1978;57:5364.
natal exposure to rubella. Am J Psychiatry. 2000;157:438443. 248. Wolinksy JS, Waxham MN, Hess JL, et al. Immunochemical features
227. Brown AS, Cohen P, Harkavy-Friedman J, et al. A.E. Bennett Research of a case of progressive rubella panencephalitis. Clin Exp Immunol.
Award. Prenatal rubella, premorbid abnormalities, and adult schizophre- 1982;48:359366.
nia. Biol Psychiatry. 2001;49:473486. 249. Connolly JH, Hutchinson WM, Allen IV, et al. Carotid artery thrombosis,
228. Beltinger C, Saule H. Sonography of subependymal cysts in congenital encephalitis, myelitis and optic neuritis associated with rubella virus infec-
rubella syndrome. Eur J Pediatr. 1988;148:206207. tions. Brain. 1975;98:583594.
229. Chang YC, Huang CC, Liu CC. Frequency of linear hyperechogenicity 250. Bitzan M. Rubella myelitis and encephalitis in childhood. A report of
over the basal ganglia in young infants with congenital rubella syndrome. two cases with magnetic resonance imaging. Neuropediatrics. 1987;18:
Clin Infect Dis. 1996;22:569571. 8487.

Scheld_Ch08.indd 136 2/21/14 5:33 PM


CHAPTER 9 HERPES SIMPLEX VIRUS
RICHARD J. WHITLEY

Eight herpesviruses routinely cause human disease. The cur- even fever blisters. The Roman scholar Herodotus associated
rent classification of herpesviruses into subfamilies serves the mouth ulcers and lip vesicles with fever (2). He called this event
purposes of identifying evolutionary relatedness and summa- herpes febrilis. Genital herpetic infections were described first
rizing unique properties of each member. The three subfam- by a physician to the French royalty, Astruc (3).
ilies are alpha that include herpes simplex virus 1 (HSV-1), The transmissibility of these viruses was established un-
HSV-2, and varicella-zoster virus (VZV); beta consisting of equivocally by passage of virus from human lip and genital le-
cytomegalovirus (CMV), human herpesvirus-6 (HHV-6) and sions to either the cornea or the scarified skin of the rabbit (4).
HHV-7; and the gamma herpesviruses, Epstein-Barr virus Goodpasture (5) further demonstrated that material derived
(EBV), and Kaposi sarcoma herpesvirus (i.e., HHV-8). One from the lesions of herpes labialis consistently produced en-
simian herpesvirus, B virus (Cryptotetia crypta), also an alpha cephalitis when inoculated onto the scarified cornea of rabbits.
herpesvirus, can inadvertently infect humans, resulting in dev- Since the first suggestions of herpes simplex encephalitis
astating central nervous system (CNS) disease. These viruses (HSE) by the Mathewson Commission in 1926 (6) and subse-
share similar molecular and biologic characteristics, including quent description of the histopathologic changes (7), HSV is
the unique ability to establish latency and reactivate. These reported as the most common cause of sporadic fatal encepha-
agents, among the most common encountered by humans, are litis in the United States. Intranuclear inclusion bodies consis-
common causes of CNS infections (1). Members of the alpha tent with HSV infection were first demonstrated in the brain
herpesvirus subfamily are characterized by a very short repro- of a neonate with encephalitis (7) in 1941, as is described later
ductive cycle, prompt destruction of the host cell, and ability in this chapter. Virus was subsequently isolated from this brain
to establish latency, usually in sensory ganglia. Both HSV-1 tissue (7). The first adult case of HSE providing similar proof
and HSV-2 are routine causes of CNS disease and the subject of viral disease (i.e., intranuclear inclusions in brain tissue and
of this chapter. virus isolation) was described in 1944 (8). The most striking
pathologic findings in this patients brain were apparent in the
left temporal lobe where perivascular cuffs of lymphocytes
GENERAL CHARACTERISTICS OF and numerous small hemorrhages were found. This temporal
lobe localization subsequently has been determined to be char-
HERPESVIRUSES acteristic of adult HSE, and it differs from the patchy diffuse
All members of the family Herpesviridae have a similar mo- encephalitis of neonates with HSV brain infection.
lecular structure. These viruses contain double-stranded DNA, In the mid-1960s, Nahmias and Dowdle (9) demonstrated
which is located at the central core. The DNA is surrounded two antigenic types of HSV. Viral typing allowed the demon-
by a capsid, consisting of 262 capsomers and providing icos- stration that HSV-1 was historically primarily responsible for
apentahedral symmetry to the virus. Tightly adherent to the infections above the belt (including brain disease in adults),
capsid is an amorphous tegument. Loosely surrounding the whereas HSV-2 was primarily responsible for infections
capsid is a bilayered lipid envelope. The overall size of herpes below the belt (brain disease in neonates). However, more
virions varies from 120 to approximately 300 nm, depending recent studies (10,11) indicate that either virus can infect the
on the virus. The envelope consists of polyamines, lipids, and mouth, genital tract, or brain.
glycoproteins. The glycoproteins confer distinctive properties
to each virus, providing unique antigens to which the host is
capable of responding. INFECTIOUS AGENT
Herpesvirus DNA varies in molecular weight from approx-
imately 80 to 150 million and consists of 120,000 to 230,000 Recent detailed reviews highlight the importance of these or-
base pairs. Base composition of herpesvirus DNA varies be- ganisms as models of viral replication and as pathogens for
tween 31% and 75% of guanine plus cytosine. Of all the human infection (11). Our current understanding of the struc-
herpesviruses, HSV-1 and HSV-2 are the most closely related, ture of HSV indicates that the genome has a molecular weight
with approximately 50% genomic homology. With the excep- of approximately 100 million. The DNA encodes about 80
tion of HSV-1 and HSV-2, the structural and nonstructural polypeptides. The DNA strands of HSV-1 and HSV-2 are co-
proteins coded by the DNA of the HHVs are not immuno- linear with reasonable but not identical matching of base pairs.
logically related. However, HSV-1 and HSV-2 share common Of note, there is considerable overlap in the cross reactivity be-
types of proteins; therefore, cross-antigenic reactions do occur. tween the HSV-1 and HSV-2 glycoproteins, although unique-
ness can be demonstrated, as discussed later in this chapter.
Distinction between the two viral types can be demonstrated
HISTORY by restriction enzyme analysis of viral DNA patterns. This
technique has been applied to epidemiologic investigations of
Infections caused by HSV have been recognized since the time human HSV infections, as well.
of ancient Greece. Greek physicians used the word herpes to Replication of HSV is characterized by the expression
mean creeping or crawling in reference to observable skin of three gene classes: immediate early (alpha), early (beta),
lesions. Likely, this word was used to describe various skin and late (gamma), which are expressed temporally and in
conditions ranging from cancer to shingles and probably a rolling-circle sequence (11). A few relevant events will be

137

Scheld_Ch09.indd 137 2/21/14 5:33 PM


138 Part II: Viral Infections and Related Disorders

noted. There are five immediate early genes, one of which


is necessary for initiating viral replication. The early gene PATHOLOGY AND PATHOGENESIS
products include those enzymes necessary for viral replica-
tion (such as HSV thymidine kinase), as well as the regulatory General Observations of Pathology
proteins. Current antiviral drugs with selective mechanisms
of action are activated at the level of early gene expression. The pathologic changes induced by replicating HSV are similar
Acyclovir is an example of such a drug, being converted to its for both primary and recurrent infection and for both adults
active monophosphate derivative by HSV thymidine kinase. and newborns but vary in the quantitative extent of cytopa-
Early gene expression coincides with an irreversible shutoff of thology. The histopathologic characteristics of a skin lesion
host cellular macromolecular protein synthesis, which results induced by HSV represent a combination of virus-mediated
in cell death. Structural proteins are usually of the late gene cellular death and associated inflammatory responses. Changes
class. Assembly of the virus begins in the nucleus, and the induced by viral infection include ballooning of infected cells
envelope is acquired as the capsid buds through the inner la- and the appearance of chromatin within the nuclei of cells;
mella of the nuclear membrane, as shown in Figure 9.1. Virus this is followed by degeneration of the cellular nuclei, gener-
is transported through the cytoskeleton to the plasma mem- ally within parabasal and intermediate cells of the epithelium.
brane, where lysis of the cell and release of progeny virions Cells lose intact plasma membranes and form multinucleated
occur. The replicative efficiency of HSV is poor, as indicated giant cells. As host defenses are mounted, an influx of mono-
by the ratio of infectious to noninfectious virions. With the nuclear cells can be detected in infected tissue.
completion of replication, 11 glycoproteins are expressed on
the plasma membrane (12).
Pathology of Central Nervous System Disease
HSE results in acute inflammation, congestion, and/or hem-
orrhage, most prominently in the temporal lobes and usually
asymmetrically in adults (13) and more diffusely in the new-
born. Adjacent limbic areas show involvement as well. The
meninges overlying the temporal lobes may appear clouded or
congested. After approximately 2 weeks, these changes pro-
ceed to frank necrosis and liquefaction, as shown in Figure 9.2.
Microscopically, involvement extends beyond areas that
appear grossly abnormal. At the earliest stage, the histologic
changes are not dramatic and may be nonspecific. Congestion
of capillaries and other small vessels in the cortex and sub-
cortical white matter is evident; other changes are also evi-
dent, including petechiae. Vascular changes that have been
reported in the area of infection include areas of hemorrhagic
necrosis and perivascular cuffing (Fig. 9.3). The perivascular
cuffing becomes prominent in the second and third weeks of
infection. Glial nodules are common after the second week
(14,15). The microscopic appearance becomes dominated by
evidence of necrosis and eventually inflammation; the latter is
characterized by a diffuse perivascular subarachnoid mono-
nuclear cell infiltrate, gliosis, and satellitosis neuronophagia
(13,16). In such cases, widespread areas of hemorrhagic ne-
crosis, mirroring the area of infection, become most promi-
nent. Oligodendrocytic involvement and gliosis (as well as
astrocytosis) are common, but these changes develop very late
in the disease. Although found in only approximately 50% of
patients, the presence of intranuclear inclusions supports the
diagnosis of viral infection, and these inclusions are most often
visible in the first week of infection. Intranuclear inclusions
(Cowdry type A inclusions) are characterized by an eosino-
philic homogeneous appearance and are often surrounded by
a clear, unstained zone beyond which lies a rim of marginated
chromatin, as shown in Figure 9.4.

General Observations on the


Pathogenesis of Human Disease
The pathogenesis of human disease depends on intimate, per-
sonal contact of a susceptible individual (namely, one who is
FIGURE 9.1 Maturation of herpes simplex virus (HSV). A: Electron
micrograph showing early vesiculation at the inner nuclear membrane
seronegative) with someone excreting HSV. Virus must come
(INM). B: Electron micrograph showing development of an encapsi- in contact with mucosal surfaces or abraded skin for infection
dated HSV at the INM. (Courtesy S. Chatterjee, PhD, Departments to occur. With viral replication at the site of infection, the cap-
of Pediatrics and Clinical Virology, University of Alabama at sid is transported by neurons to the dorsal root ganglia, where
Birmingham.) after another round of viral replication, latency is established.

Scheld_Ch09.indd 138 2/21/14 5:33 PM


Chapter 9: Herpes Simplex Virus 139

FIGURE 9.2 Gross pathologic


findings in herpes simplex encepha-
litis, illustrating hemorrhagic ne-
crosis of the inferior medial portion
of the temporal lobe.

FIGURE 9.3 A: Hemorrhagic necrosis on microscopic


examination. B: Perivascular cuffing on histopatho-
logic examination of a patient with herpes simplex
encephalitis.
A B

FIGURE 9.4 Intranuclear inclusions.

Scheld_Ch09.indd 139 2/21/14 5:33 PM


140 Part II: Viral Infections and Related Disorders

These events have been demonstrated in a variety of animal 15%, have active herpes labialis and HSE, allowing compari-
models (17). Transport of the virion is by retrograde axonal son of the DNA by restriction enzyme analyses. The isolates
flow (18). In some instances, replication can lead to severe are usually identical; however, this is not always the case. The
CNS infection; however, more often, the hostvirus interac- virus isolated from the peripheral site can be different from that
tion results in latency. After latency is established, reactivation retrieved from the CNS (41). Thus, the issues of reactivation
can occur, with virus shedding at mucocutaneous sites appear- of virus directly within the CNS, the potential for enhanced
ing as skin vesicles or mucosal ulcers or being totally asymp- neurotropism of certain viruses, and the selective reactivation
tomatic. Occasionally, primary infection can become systemic, and access of one virus by the trigeminal route or other routes
affecting other organ systems besides the CNS and the periph- to the CNS remain for further elucidation.
eral nervous system. Such circumstances include disseminated The route of access of virus to the CNS in primary infection
neonatal HSV infection with multiorgan involvement, multi- is a subject of debate, especially in humans. Studies performed
organ disease of pregnancy, and infrequently dissemination in more than five decades ago defined pathways for HSV access
patients undergoing immunosuppressive therapy. Multiorgan to the brain in animals, including both the olfactory and tri-
disease is likely the consequence of viremia in a host not geminal nerves among others (42). However, which of these
capable of limiting replication to mucosal surfaces. nerve tracts uniformly leads to HSV infection in the CNS of
Infection with HSV-1 is transmitted either by respiratory humans is not clear. The anatomic distribution of nerves from
droplets or through direct contact (to a susceptible individual) the olfactory tract into the limbic system, along with the recov-
with infectious secretions (such as virus contained in orolabial ery of virus from the temporal lobe (the site of apparent onset
vesicular fluid). Acquisition of HSV-2 infection is usually the of HSE in the human brain), suggests that viral access to the
consequence of transmission via genital routes. Under these CNS via this route is a tenable hypothesis. Reports in the lit-
circumstances, virus replicates in the vaginal tract or on penile erature have found electron microscopic evidence that suggests
skin sites, with seeding of the sacral ganglia. this has been the case in some individuals with HSE (4345).
Animal model data support the contention that the olfactory
tract provides one neurologic avenue for viral access to the
Pathogenesis of Latency CNS and causes localization of the infection in brain regions
analogous to medial temporal structures in humans (46,47).
All of the herpesviruses have the ability to become latent, persist Definitive proof of such progression in humans is lacking.
in an apparent inactive state for varying durations, and be reac- Reactivation of HSV, leading to focal HSE, is a similarly
tivated by a provocative stimulus, as yet unidentified (11,19), confusing problem from the standpoint of pathogenesis.
as recently reviewed. As a biologic phenomenon, latency has Evidence of latent virus within infected brain tissue exists
been recognized since the beginning of the twentieth century (48); however, virus reactivation at that site remains purely
(2026). In 1905, Cushing (27) noted that patients treated for hypothetical. Reactivation of virus peripherally (namely, in
trigeminal neuralgia (by sectioning a branch of the trigeminal the olfactory bulb or the trigeminal ganglion) with subse-
nerve) developed HSV lesions along the innervated areas of quent neuronal transmission to the CNS has been suggested
the sectioned branch, as suggested previously by Goodpasture (42,49,50). Nevertheless, a relevant observation is that with
(28,48). Several investigators have demonstrated that micro- recurrent herpes labialis, whereby reactivation of virus from
vascular surgery of the trigeminal nerve tract for tic doulou- the trigeminal ganglia occurs, HSE is a very uncommon event.
reux resulted in recurrent herpetic lesions in more than 90% of Furthermore, HSE does not occur more frequently in immuno-
seropositive individuals (2932). Axonal injury and attempts at compromised patients.
excision of lesions have been associated with recurrences (33). Host immunity plays an important, but undefined, role
Reactivation of latent virus appears to depend on an intact an- in the pathogenesis of HSE. Possibly, the CNS is particularly
terior nerve route and peripheral nerve pathways. prone to HSV infection because intraneuronal spread may
Recurrences occur despite both cell-mediated and humoral im- shelter virus from host defense mechanisms. HSE is no more
mune responses and can be either symptomatic or asymptomatic. common in the immunosuppressed host than in the normal
Recurrences are spontaneous, but there have been associations host; however, when it does occur, the presentation is atypical,
with physical or emotional stress, fever, exposure to ultraviolet with a subacute but progressively deteriorating course.
light, tissue damage, and immune suppression (25,34,35). Viral More recently, a host genetic deficiency has been found to
DNA can be detected in neuronal tissue in the absence of cutane- play a role in recurrent HSE but certainly does not exist in all
ous lesions (17,3639). Latent virus has been retrieved from the patients (51).
trigeminal, sacral, and vagal ganglia of humans (2022,36,39).
EPIDEMIOLOGY
Pathogenesis of Encephalitis
Herpes Simplex Virus, Type 1
The pathogenesis of HSE in children (older than 3 months) and
adults is only partly understood. Both primary and recurrent The epidemiology of HSV infections is multifaceted. Because
HSV infections can cause disease of the CNS. From studies the focus of this book is CNS infections, only a brief review of
performed by the National Institute of Allergy and Infectious non-CNS HSV infections follows. The reader is referred to more
Diseases (NIAID) Collaborative Antiviral Study Group (CASG), complete reviews (11). HSV infections are distributed world-
approximately one third of the cases of HSE are the conse- wide and have been reported in both developed and developing
quence of primary infection. For the most part, the patients countries, including remote Brazilian tribes (52). Animal vec-
with primary infection are younger than 18 years. The remain- tors for human HSV infections have not been described; there-
ing two thirds of cases occur in the presence of preexisting anti- fore, humans remain the sole reservoir for transmission of these
bodies, but only approximately 10% of patients have a history viruses to other humans during close personal contact. There
of recurrent herpes labialis. Patients with preexisting antibod- is no seasonal variation in the incidence of infection. Because
ies are considered to have HSE as a consequence of reactiva- infection is rarely fatal, and because these viruses become
tion of HSV (40). A limited number of patients, approximately latent, more than two thirds of the worlds population can have

Scheld_Ch09.indd 140 2/21/14 5:33 PM


Chapter 9: Herpes Simplex Virus 141

recurrent HSV infections and, therefore, can transmit infection Women have the highest rates of infection, particularly
during episodes of reactivation. HSV disease ranges from mild prostitutes and others with multiple sex partners, including
(even indiscernible) in most patients to sporadic, severe, and those with HIV infection. The incidence of genital HSV infec-
life-threatening disease in a few infants, children, and adults. tions in both indigent women and those of middle and upper
Children, particularly those younger than 5 years, are most socioeconomic classes is significantly lower than the incidence
often infected; however, primary infections can also occur in found among women attending sexually transmitted disease
older individuals. With clinical illness, oropharyngeal disease, clinics (82). As with HSV-1 infections of the mouth, HSV-2
namely gingivostomatitis, usually is the manifestation. Primary can be excreted in the absence of symptoms at the time of
infection in young adults has been associated with pharyngitis primary, initial, or recurrent infection (83,84). The actual
and often a mononucleosis-like syndrome (53). Seroprevalence frequency of asymptomatic excretion of HSV-2 in women by
studies have demonstrated that acquisition of HSV-1 infection culture is approximately 3% to 5% of all days, and by poly-
is related to socioeconomic factors, namely lower socioeco- merase chain reaction (PCR) 15% to 20%. Furthermore, some
nomic populations acquire infection earlier in life than more individuals can start and stop shedding multiple times during
affluent individuals. The identification of primary gingivosto- the same day (85). Its occurrence creates a silent reservoir for
matitis that was proven to be caused by HSV infection (54,55) transmission of infection (86,87). The appearance of HSV-2
led to the definition of the natural history of infection, includ- antibodies reflects the time of exposure or more simply the
ing the appearance of neutralizing antibodies (56), absence of acquisition of infection and is positively correlated with the
virus shedding in children younger than 6 months (57), and a onset of sexual activity (70,71,88). However, crowded living
higher rate of occurrence among individuals of lower socioeco- conditions may indirectly contribute to antibody prevalence
nomic status. Contemporary surveys document the viral shed- (89). If HSV-2 type-specific antibodies are sought in healthy
ding data, ranging from 2% to 5% (5863). women, there is a wide discrepancy in prevalence, ranging
Antibody surveys have helped clarify the epidemiology from averages of 10% in England and Italy to 25% in the
of HSV infection. Geographic location, socioeconomic sta- United States and 77% in Uganda (90,91). Up to 50% to 60%
tus, and age all influence the acquisition of HSV infection of lower socioeconomic populations in the United States and
(54,6466). In developing countries, seroconversion occurs elsewhere develop antibodies to HSV-2 by adulthood. The
early in life. In Brazilian Indians, HSV antibodies are detect- reader is referred to a review for worldwide seroprevalence
able in more than 95% of children by the age of 15 years of HSV-2 (90). Seroprevalence is a function of age, number of
(67). Similarly, serologic studies performed in New Orleans sexual partners, race, and marital status (9294).
demonstrated acquisition of antibodies in more than 90% of
children by the age of 15 years (68). In developing countries,
such as Uruguay, or in lower socioeconomic populations in the Latent Genital Herpes
central United States, the appearance of antibodies occurred Simplex Virus Infections
at similar but lower frequencies (6871). By 5 years of age,
approximately one third of patients had seroconverted; this Latent genital infection with subsequent reactivation is the
frequency increased to 70% to 80% by early adolescence. largest reservoir for transmission of HSV-2. As with HSV-1
Middle-class individuals of industrialized societies acquired infection, recurrent HSV-2 infection can be either symptom-
infection later in life. Seroconversion occurred during the first atic or asymptomatic; however, recurrence is usually associ-
5 years of life in 20% of children; there was no significant ated with a shorter duration of viral shedding and fewer
increase until the second and third decades of life, at which time lesions. Several studies have implicated a frequency of recur-
the prevalence of antibodies increased to 40% and 60%, respec- rence as high as 60% (88,95). The type of genital infection,
tively (72,73). One study of university students demonstrated HSV-1 versus HSV-2, is predictive of the frequency of recur-
that seroconversion of susceptible individuals occurred at an rence (9597), with HSV-1 infection recurring less frequently
annual frequency of approximately 5% to 10% (53,74,75). than HSV-2 (98,99).
In summary, primary infection occurs very early in children of
underdeveloped countries and in those of lower socioeconomic
classes; however, in developed countries and more affluent HERPES SIMPLEX ENCEPHALITIS
classes, primary infection is delayed until adolescence or, per-
haps, even adulthood. The frequency of direct person-to-person
contact is the major mediator of acquisition of infection. Background
Over the past decade, HSV-1 has been increasingly associ-
HSV infections of the CNS are among the most severe of all
ated with primary genital herpes, reflecting a change in the
viral infections of the human brain. Currently, HSE is estimated
epidemiology of infection (11).
to occur in approximately 1 per 250,000 to 500,000 individu-
als per year. At the University of Alabama at Birmingham, the
diagnosis of HSE was proven by brain biopsy in an average of
Herpes Simplex Virus, Type 2 ten patients per year, for an incidence of approximately 1 in
300,000 individuals, an incidence similar to those in Sweden
Because HSV-2 infections are usually acquired through sexual and England (100,101). With the advent of PCR, brain biopsy
contact, antibodies to this virus are rarely found before the is no longer used. In the United States, HSE accounts for 10%
age at onset of sexual activity. Although most genital HSV to 20% of viral infections of the CNS (102).
infections are caused by HSV-2, an ever-increasing propor- The economic cost of HSE is considerable, as estimated
tion is attributable to HSV-1, as noted earlier, now as high as in 1983 for hospitalization alone of adults to be more than
50% of all new primary infections (10,7678). Approximately $25 million (103). The total medical cost is considerably
1.5 million new cases of HSV-2 occur annually in the United higher because of the long-term care and support services
States (79). Genital HSV infections are not reportable in the required for many of the survivors.
United States (80). Current estimates of infected individuals HSE occurs throughout the year and in patients of all
with genital herpes in the United States range from 40 to ages, with approximately one third of cases occurring in
60 million (7678,81). patients younger than 20 years but older than 6 months and

Scheld_Ch09.indd 141 2/21/14 5:33 PM


142 Part II: Viral Infections and Related Disorders

approximately one half in patients older than 50 years (102). TA B L E 9 . 1


Caucasians account for 95% of patients with biopsy-proven
disease. Both sexes are affected equally. COMPARISON OF FINDINGS IN BRAIN-POSITIVE
The severity of disease is best determined by the outcome AND BRAIN-NEGATIVE PATIENTS WITH HERPES
of patients who have received either no therapy or an ineffec- SIMPLEX ENCEPHALITIS
tive antiviral medication, such as idoxuridine or cytosine arabi-
noside. In such situations, mortality is in excess of 70%; only No. (%) of Patients
approximately 2.5% of all patients with confirmed disease (9.1%
Brain Positivea Brain Negativea
of survivors) returned to normal function after recovery from
their illness (104108). Because brain biopsy with isolation of Historic findings
HSV from brain tissue was the method of diagnosis in these early
studies, a far broader spectrum of HSV infections of the CNS Alteration of 109/112 (97) 82/84 (96)
actually was thought to exist. However, with the more recent use consciousness
of PCR for diagnosis of HSE, virtually all patients have a focal Cerebrospinal fluid 107/110 (97) 71/82 (87)
neurologic disease, suggesting a limited spectrum of disease (109). pleocytosis
Fever 101/112 (90) 68/85 (78)
Headache 89/110 (81) 56/73 (77)
Diagnosis Personality change 62/87 (71) 44/65 (68)
Seizures 73/109 (67) 48/81 (59)
Several aspects relating to the diagnosis of HSE merit discus-
sion: (a) the clinical presentation in regard to the sensitivity and Vomiting 51/111 (46) 38/82 (46)
specificity of various clinical characteristics, (b) the historical Hemiparesis 33/100 (33) 19/71 (26)
use of brain biopsy to establish the diagnosis, (c) conditions Memory loss 14/59 (24) 9/47 (19)
that mimic HSE, and (d) noninvasive means of diagnosis. Data
Clinical findings at presentation
from the NIAID CASG compare presentation and outcome
for brain biopsypositive and brain biopsynegative patients Fever 101/110 (92) 84/79 (81)
(137). Of 202 patients who were evaluated for HSE because of Personality change 69/81 (85) 43/58 (74)
focal neurologic findings, HSV was isolated from brain tissue Dysphasia 58/76 (76) 36/54 (67)
of only 113. Only three of the remaining patients had combi-
Autonomic dysfunction 53/88 (80) 40/71 (58)
nations of serologic and clinical findings suggestive of HSE.
These patients were subsequently shown to have HSV DNA in Ataxia 22/55 (40) 18/45 (40)
their cerebrospinal fluid (CSF) by PCR. Hemiparesis 41/107 (38) 24/81 (30)
As shown in Table 9.1, most patients with biopsy-proven Seizures 43/112 (38) 40/85 (47)
HSE presented with a focal encephalopathic process, includ-
Focal 28 13
ing (a) altered mentation and decreasing levels of conscious-
ness with focal neurologic findings; (b) CSF pleocytosis and Generalized 10 14
proteinosis; (c) the absence of bacterial and fungal pathogens Both 5 13
in the CSF; and (d) focal electroencephalographic (EEG), com- Cranial nerve defects 34/105 (32) 27/81 (33)
puted tomographic (CT), and/or magnetic resonance image
(MRI) findings (102,110120). The frequency of headache Visual field loss 8/58 (14) 4/33 (12)
and CSF pleocytosis is higher in patients with proven HSE Papilledemia 16/111 (14) 9/84 (11)
than in patients with diseases that mimic HSE. Nearly uni-
a
formly, patients with HSE present with fever and personality Positive or negative findings from brain tissue culture.
change. Seizures, whether focal or generalized, occur in only Data from Whitley RJ, Soong S-J, Linneman C Jr, et al. Herpes sim-
approximately two thirds of all patients with proven disease. plex encephalitis: clinical assessment. JAMA. 1982;247:317320, with
permission.
Thus, the clinical findings of HSE are nonspecific and do not
allow for empirical diagnosis of disease predicated solely on
clinical presentation. Although clinical evidence of a localized
temporal lobe lesion is often thought to indicate HSE, various discharges, which arise from the temporal lobe (114118). Early
other diseases can mimic this condition. in the disease, the abnormal electric activity usually involves one
Examination of the CSF is indicated in patients with temporal lobe and then spreads to the contralateral temporal
altered mentation, provided it is not contraindicated because lobe as the disease evolves, usually over 7 to 10 days. The sen-
of increased intracranial pressure. In patients with HSE, CSF sitivity of the EEG is approximately 84%, but the specificity
findings are nondiagnostic, being similar in patients with con- is only 32.5%. CT scans initially show low-density areas with
firmed disease or diseases that mimic HSE. Both the CSF white mass effect localized to the temporal lobe, which can progress to
blood cell (WBC) count (lymphocytes predominance) and the radiolucent and/or hemorrhagic lesions (119,120). Bitemporal
CSF protein level are elevated. The average CSF WBC count is disease is common in the absence of therapy, particularly late in
100 cells/L; the protein averages approximately 100 mg/dL. the disease course (Fig. 9.5). When these neurodiagnostic tests
Sequential evaluation of CSF specimens from patients with are used in combination, the sensitivity is enhanced; however,
HSE indicates increasing cell counts and levels of protein. the specificity remains inadequate. None of these neurodiag-
The presence of CSF red blood cells is not diagnostic for HSE. nostic tests is uniformly satisfactory for diagnosing HSE. MRI
Approximately 5% to 10% of patients have a normal CSF detects evidence of HSE earlier than CT scan (111).
formula on first evaluation. A sensitive and specific means of diagnosis is the isolation of
Noninvasive neurodiagnostic studies support a presump- HSV from tissue obtained at brain biopsy (121). However, PCR
tive diagnosis of HSE. These studies have included EEG, CT, detection of HSV DNA in the CSF has become the diagnostic
and MRI. Focal changes of the EEG are characterized by spike procedure of choice. Brain biopsy is of value in confusing
and slow-wave activity and periodic lateralized epileptiform clinical presentations. Complications, either acute or chronic,

Scheld_Ch09.indd 142 2/21/14 5:33 PM


Chapter 9: Herpes Simplex Virus 143

FIGURE 9.5 Sequential computed tomographic examination of a patient with confirmed herpes simplex
encephalitis.

occur in approximately 3% of patients. Fears of potentiating of therapy, including brain abscess, tuberculosis, cryptococ-
acute illness (by incising the brain in a diseased area) or of cal infection, and brain tumor. An additional 19 patients had
causing chronic seizure disorders have not been substantiated diseases that were indirectly treatable, and another 38 patients
by follow-up studies of patients in the NIAID CASG. had an alternative diagnosis established for which there was
no current therapy, usually other viral infections. Thus, those
diseases that mimic HSV infection of the CNS and that require
Serologic Evaluation immediate medical intervention should be considered if the
PCR is negative for HSV DNA.
Several strategies using antibody production as a means of diag-
nosing HSE have been utilized (58). Because most encephalitic
patients are HSV seropositive at presentation, seroconversion TA B L E 9 . 2
per se is usually not helpful because fever alone can reactivate DISEASES THAT MIMIC HERPES SIMPLEX
labial herpes, resulting in antibody elevations. A fourfold rise ENCEPHALITIS
in serum antibody was neither sensitive nor specific enough to
be useful. A fourfold or greater rise in CSF antibody occurred Nontreatable Diseases
significantly more often within a month after onset of dis- Treatable Diseases (N 46)a (N 49)a
ease in patients with biopsy-proven HSE: 85% versus 29%.
By 10 days after clinical presentation, however, only 50% of Abscess/subdural Vascular disease 11
brain biopsypositive patients had a fourfold rise in CSF anti- empyema Toxic encephalopathy 5
body. This test is useful only for retrospective diagnosis. The Bacterial 5 Reye syndrome 1
use of a ratio of serum to CSF antibody of 20 or less did not Listeria 1 Viral (N 40)
improve sensitivity during the first 10 days of disease. Fungal 2 Arbovirus infection
Mycoplasma 2 St. Louis encephalitis 7
Tuberculosis 6 Western equine 3
Polymerase Chain Reaction Detection of Cryptococcal 3 encephalitis
Viral DNA Rickettsial 2 California 4
Toxoplasmosis 1 encephalitis
PCR detection of HSV DNA in the CSF is the diagnostic Mucormycosis 1 Eastern equine 2
method of choice (121127). Data from the NIAID CASG Meningococcal 1 encephalitis
defined the sensitivity and specificity as 94% and 98%, meningitis Other herpesviruses
respectively. These CSF specimens were obtained from patients Other viruses Epstein-Barr virus 8
with biopsy-proven or biopsy-negative disease. Notably, the Cytomegalovirus 1 Others viruses
specificity would have been higher except that some tissue Influenza A 4 Mumps virus 3
specimens were fixed in formalin, which killed infectious Echovirus infectionb 3 Adenovirus 1
virus, prior to an attempt to isolate virus. HSV DNA persisted Tumor 5 Progressive multifocal 1
in 80% of tested CSF specimens for 1 week or more. Subdural hematoma 2 leukoencephalopathy
Systemic lupus 1 (JC virus)
erythematosus Lymphocytic 1
Adrenal 6 choriomeningitis
Diseases That Mimic Herpes Simplex leukodystrophy Subacute sclerosing 2
Encephalitis panencephalitis
In a compilation of the NIAID CASG data, 193 (45%) of a
Of 432 patients assessed.
432 patients undergoing brain biopsy for a focal encepha- b
Drug therapy under investigation.
lopathic process had HSE (128). As shown in Table 9.2, the From Whitley RJ, Gnann JW. Viral encephalitis: familiar infections
remaining patients were evaluated for diseases that mimic and emerging pathogens. Lancet. 2002;359:507513, with permission.
HSE (128). Thirty-eight had disease amenable to other forms

Scheld_Ch09.indd 143 2/21/14 5:33 PM


144 Part II: Viral Infections and Related Disorders

Level of Consciousness
Associated Neurologic Syndromes
Lethargic Semicoma Coma
HSV obviously involves areas of the nervous system other N = 23 N=7 N=7
than the brain. Primary and recurrent genital herpes have been 100
associated with neuritis localized to one extremity or even
transverse myelitis. Neuritis evident in such patients can be 80
associated with altered sensation of the lower extremities, as
well as dysesthesias, shooting pain, and motor impairment.
60
Urinary and fecal incontinence have been reported in a few
patients. An aseptic meningitis syndrome is also common, fre-
quently being associated with a Mollaret syndrome, and not 40
without complications.
Guillain-Barr syndrome and localized dermatomal rashes 20

Percent of Patients
associated with acute neuritis have also been attributed to
HSV infections. 0
Acute retinal necrosis has been reported as a long-term
complication of HSE (129). N = 26 N = 13 N = 14
100

Therapy 80

The first antiviral drug reported as efficacious therapy of HSE 60


was idoxuridine; however, it was soon proven both ineffec-
tive and toxic (104). Subsequent therapeutic trials defined
vidarabine as a useful medication for the management of 40
biopsy-proven HSE (107,108); however, it has been replaced
by acyclovir in the physicians armamentarium. During these 20
studies, the variables of age, disease duration, and level of con-
sciousness at the onset of therapy were proven major deter- 0
minants of clinical outcome. Patients younger than 30 years
and with a more normal level of consciousness (lethargic as Returned to normal function Moderate debility
opposed to comatose) were more likely to return to normal Severe impairment Death
function than older patients, especially those who were semico-
FIGURE 9.6 Influence of level of consciousness and age on mortality
matose or comatose (Fig. 9.6). From these data, older patients
and morbidity.
(older than 30 years), whether comatose or semicomatose, had
mortality rates that approached 70%, a figure very similar to
that encountered in the placebo recipients of the previously Of acyclovir recipients, 10% experienced an increased
cited studies. If therapy is to be effective, it must be instituted BUN level, and 6% developed a creatinine level in excess
before the onset of hemorrhagic necrosis of a dominant tem- of 2 mg/dL. No clinical evidence of toxicity was detected.
poral lobe and significant deterioration of consciousness. The current therapy of choice for the management of HSE is
Acyclovir is superior to vidarabine for the treatment of HSE acyclovir. This drug is administered at a dosage of 10 mg/kg
(166). The NIAID CASG study defined a mortality of 55% at every 8 hours (30 mg/kg per day) for 14 to 21 days.
6 and 18 months after the initiation of treatment for vida-
rabine recipients versus 19% and 28%, respectively, for the
acyclovir group (Fig. 9.7). Late deaths were not a consequence 100
of either persistent or reactivated HSV infection but occurred
in patients who were severely impaired as a consequence of
their disease. Acyclovir decreases mortality to 19% 6 months 80 ACV (N = 32)
after therapy. Importantly, 38% of patients, irrespective of
age, return to normal function.
Percent Surviving

Previous studies indicated that age and level of conscious-


ness influenced long-term outcome. A more objective reflection 60
of level of consciousness is the Glasgow Coma Scale (GCS). ARAA (N = 37)
Scores that approached normal predicted enhanced survival.
When GCS score and age were assessed simultaneously 40
(Fig. 9.8), a GCS score of 6 or less predicted a poor therapeutic
outcome, irrespective of the agent administered or of the age
of the patient (130).
20
Regarding morbidity for acyclovir recipients, 38% of
p = 0.008
patients were normal or with minor impairment, 9% of patients
had moderate sequelae, and 53% of patients were left with
severe impairment or died. No patient entered into the NIAID 0
trials suffered a relapse after completion of therapy. Relapse of 0 3 6 9 12 15 18
HSE has been reported, though not well documented, in a few
Months
patients following the administration of vidarabine (131133)
and acyclovir (133,134). Many patients were not afebrile at FIGURE 9.7 Comparison of survival in patients with biopsy-proven
the conclusion of treatment, suggesting that a longer duration herpes simplex encephalitis treated with vidarabine (AraA) or
of therapy to a minimum of 14 to 21 days may be desirable. acyclovir (ACV); p .008.

Scheld_Ch09.indd 144 2/21/14 5:33 PM


Chapter 9: Herpes Simplex Virus 145

N=4 N=5 N=7 N = 13 the newborn. Gross examination of the brain often reveals
100 100 encephalomalacia and hydranencephaly, which are likely the
1 consequence of extensive hemorrhagic necrosis. Porencephaly,
4 2 8 hydranencephaly, and multicystic lesions are often sequelae in
75 75 neonates who survive for several weeks or months following
neonatal HSV infection of the brain. The microscopic appear-
 30 ance of the brain is characterized by a mononuclear inflamma-
50 50 3 tion, necrosis, and hemorrhage.
3
3 Pathogenesis
25 25
2 In utero disease is likely a consequence of transplacental infec-
1 2 tion and usually involves skin, brain, eye, liver, and adrenals.
0 0 More commonly, the fetus comes in contact with infected
ARAA ACV ARAA ACV maternal genital secretions at the time of delivery. Viral rep-
6 6 lication in the newborn either remains limited to the portal
of entrynamely, the skin, eye, or mouthor progresses to
N=8 N=3 N = 18 N = 11 involve various other organs, including the brain (resulting
100 100 in encephalitis), causing life-threatening disease. Host mecha-
1 2 nisms responsible for control of viral replication at the site of
4 entry are unknown. For babies with encephalitis, intraneuro-
1 3
75 75 nal transmission of virus provides a privileged site that may be
1
2 impervious to circulating humoral and cell-mediated defense
4
mechanisms. Thus, transplacental maternal antibodies may be
 30 50 50 3 of less value in the prevention of encephalitic forms of neona-
tal HSV infections. Disseminated infection is a consequence of
5 viremia or secondary to extensive cell-to-cell spread, as occurs
25 25 9 with pneumonitis after aspiration of infected secretions.
1 4
Neonatal HSE illustrates the two major pathogenic routes
for virus access to the brain, namely, hematogenous and intra-
0 0 neuronal. For example, hematogenous spread of virus usually
ARAA ACV ARAA ACV
occurs with disseminated disease, and diffuse involvement of
6 6 the brain ensues in 60% to 80% of patients. In contrast, neu-
Normal and mild Moderate Severe ronal transmission probably results in the focal CNS disease
encountered in babies with encephalitis only when no distal
Dead Number of patients
organ involvement is documented (136).
FIGURE 9.8 Morbidity after vidarabine (AraA) or acyclovir (ACV)
treatment of biopsy-proven herpes simplex encephalitis, according to
age (younger than 30 years vs. older than 30 years) and the Glasgow Times of Transmission of Infection
Coma Scale score (6 vs. 6). The scale at the left side of each column
indicates percentages (0% to 100%). Neonatal HSV infection is acquired at one of three times: in
utero, intrapartum, or postnatally. Regardless of the time or
route of acquisition, the newborn is at risk of CNS disease.
Certainly, the mother is the most common source of infection
for the first two of these routes of transmission of infection to
NEONATAL HERPES SIMPLEX the newborn.
VIRUS INFECTIONS
Intrauterine Infection
History In utero acquisition of HSV infection is becoming increasingly
documented (137139). Manifestations of disease acquired
In 1941, Smith, Lennette, and Reames (7) reported the first in utero include chorioretinitis, cutaneous aplasia, hydran-
case of HSV infection of the CNS, as noted earlier. This case encephaly, and encephalomalacia (138) (Fig. 9.9). Risk fac-
occurred in a newborn with neonatal HSE. In 1952, Zuelzer, tors associated with intrauterine transmission of infection
Wolf, and Stulbery (135) reviewed eight cases of disseminated are unknown; however, both primary and recurrent maternal
HSV infection in neonates with involvement of most organs, infection can result in infection of the fetus in utero. In utero
including the brain in many instances. This report was fol- infection is the consequence of either transplacental or ascend-
lowed shortly by others indicating the association between ing infection.
HSV infection of the newborn and necrotizing encephalitis,
including the isolation of HSV in cell cultures from brain Intrapartum Infection
tissue.
The most common time of transmission of infection from
mother to the fetus is intrapartum. Transmission occurs when
the infant comes in contact with infected maternal genital
Pathology and Pathogenesis secretions at delivery, accounting for 85% of cases (140).
Prospective assessment of HSV excretion in the genital tract
at delivery indicates that shedding can occur in 0.5% to 1.3%
Pathology of women (141). Maternal primary versus recurrent infection
Although the pathology of HSE is discussed earlier in this influences the probability of neonatal infection, as discussed
chapter, a few characteristics appear more commonly in later in this chapter. With the increasing prevalence of HSV-2

Scheld_Ch09.indd 145 2/21/14 5:33 PM


146 Part II: Viral Infections and Related Disorders

infection in the population at large, the probability of excret- personnel or other babies) (155157), and as a consequence
ing HSV at delivery should increase (142,143). of the Jewish tradition of circumcision, known as metzitzah
Factors that influence intrapartum acquisition of infection bpeh (158).
by the fetus include the following: (a) type of maternal infec-
tion (primary vs. recurrent) (144), (b) maternal antibody sta-
tus (145,146), (c) duration of ruptured membranes (147), and Epidemiology of Maternal Infection
(d) placement of a fetal scalp monitor in a woman excreting
HSV (148,149). Disseminated Maternal Infection
Primary infection is associated with (a) larger quantities of
HSV replicating in the genital tract (more than 106 viral par- HSV infections of pregnant women may extend beyond the
ticles/0.2 mL of tissue culture inoculum) and (b) a period of usual sites of disease, the oropharynx and the genital tract.
viral excretion that on average persists for 3 weeks. In con- As first reported in 1969, dissemination can occur, leading
trast, in women with recurrent genital infection, HSV is shed to cutaneous or visceral disease; fortunately, this is a rare
for an average of only 2 to 5 days and at lower concentrations occurrence. In a limited number of cases (159162), dissemi-
(approximately 103/0.2 mL of tissue culture inoculum). Because nation following primary oropharyngeal or genital infection
of the larger quantity of virus and the longer period of viral has led to life-threatening diseases such as (a) hepatitis with
excretion, primary maternal infection is associated with a higher or without thrombocytopenia, (b) leukopenia, (c) consump-
rate of transmission to the fetusestimated between 30% and tive coagulopathy, and (d) encephalitis. Although only a small
50% (147). Reflecting the type of maternal infection, the deliv- number of patients have been studied, the mortality among
ery of transplacental maternal antibody to the fetus influences pregnant women with disseminated infection has been greater
both the severity of disease in the newborn and the likelihood of than 50%. Fetal deaths also occurred in more than 50% of
fetal infection (145,146,150). Lastly, placement of a fetal scalp cases, though not necessarily associated with the death of the
monitor in women excreting virus has been shown to lead to mother.
fetal infection. Monitor placement should be discouraged in
women with a history of genital herpes or visualized lesions. Localized Maternal Infection
The duration of ruptured membranes is reported to be
During the first 20 weeks of gestation, primary maternal geni-
an important indicator of risk for acquisition of neonatal
tal HSV infection is associated with an increased frequency of
infection. Cesarean section decreases the incidence of infection
spontaneous abortion (approximately 25%), stillbirth, and as
in women with lesions present at delivery (147).
noted earlier, congenital disease, particularly hydranencephaly
and chorioretinitis (138,163). Several studies have prospec-
Postnatal Infection tively evaluated the frequency and nature of viral shedding in
The third route of transmission is postnatal acquisition pregnant women with a known history of genital herpes. In
(151158). Documented sources include the mother (includ- a predominantly white middle-class population, documented
ing the breast as a source of virus [151,152]), the father recurrent infection occurred in 84% of pregnant women
(labial lesions) (153,154), nosocomial transmission (nursery (164). Moreover, asymptomatic viral shedding occurred in

FIGURE 9.9 Encephalomalacia resulting from in utero herpes simplex virus infection.

Scheld_Ch09.indd 146 2/21/14 5:33 PM


Chapter 9: Herpes Simplex Virus 147

at least 12% of the recurrent episodes. Viral shedding from tality. Children with disseminated infection usually present to
the cervix occurred in only 0.56% of symptomatic infections tertiary medical centers for therapy between 9 and 11 days of
and in 0.66% of asymptomatic infections (84,165,166). For life; however, signs of infection are, on average, usually pres-
asymptomatic pregnant women, an incidence of cervical shed- ent 4 to 5 days earlier.
ding as high as 3% has been reported (167). More than 60% The principal organs involved following disseminated
of women in various groups who give birth to infants with infection are the liver, brain, and adrenals; however, infection
neonatal HSV infection report no symptoms (168170). can involve various other organs, including the larynx, tra-
chea, lungs, esophagus, stomach, lower gastrointestinal tract,
spleen, kidneys, pancreas, and heart. Constitutional signs and
Incidence and Presentation of symptoms include irritability, seizures, respiratory distress,
jaundice, bleeding diatheses, and shock, in addition to a char-
Neonatal Infection acteristic vesicular exanthem that is often considered pathog-
The incidence of neonatal HSV infection is about 1 in 3,000 nomonic for neonatal HSV infection.
(0.03%) deliveries (136). Overall, two thirds of children with The vesicular rash, as described later in this chapter, is par-
neonatal HSV infection develop disease of the CNS, and the ticularly important in the diagnosis of HSV infection. Notably,
disease may remain localized to the brain or become dissemi- about 20% of children with disseminated neonatal HSV infec-
nated to involve various other organs. If untreated, newborns tion will not develop skin vesicles during the course of their
with disseminated disease have a mortality of 80%, and new- illness (140,171). In the absence of skin vesicles, the diag-
borns with disease limited to the CNS have a mortality of nosis becomes exceedingly difficult because the clinical signs
approximately 50%. are often vague and nonspecific, mimicking those of neonatal
Classification of newborns with HSV infection is manda- sepsis. Mortality in the absence of therapy exceeds 80%; if
tory for prognostic and therapeutic considerations (163). therapy is instituted before CNS disease ensues, outcome is
Babies with congenital infection, by definition, must be identi- usually good. The most common cause of death in babies with
fied within 48 hours of birth. Those babies who are infected disseminated disease is either HSV pneumonitis or dissemi-
(either during delivery or postnatally) are divided into three nated intravascular coagulopathy.
categories: (a) those with disease localized to the skin, eye, Evaluation of the extent of disease is imperative, as with
or mouth; (b) those having encephalitis with or without skin, all cases of neonatal HSV infection. The clinical laboratory
eye, and/or mouth involvement; and (c) those having dissemi- should be used to define hepatic enzyme elevation (serum ala-
nated disease involving multiple organs, such as CNS, lung, nine aminotransferase and AST), direct hyperbilirubinemia,
liver, adrenals, skin, eye, and/or mouth. This chapter focuses neutropenia, thrombocytopenia, and bleeding diatheses.
on CNS disease and considers prospectively acquired data Unless contraindicated, examination of the CSF is imperative.
obtained through the NIAID CASG. All babies, irrespective In addition, chest roentgenograms, abdominal x-rays, EEG,
of disease classification, should be considered at risk for CNS and CT or MRI of the head can be judiciously and serially
complications of infection. The presentation and outcome of employed to determine the extent of disease. The radiographic
infection (particularly prognosis after therapy) according to picture of HSV lung disease is characterized by a diffuse inter-
category vary significantly with regard to both mortality and stitial pattern that progresses to a hemorrhagic pneumonitis.
morbidity. Pneumatosis intestinalis can be detected when gastrointesti-
nal disease is present. Encephalitis is a common component
of disseminated infection, occurring in about 75% of these
newborns. Serial evaluation of the CSF and noninvasive neu-
Intrauterine Infection rodiagnostic tests, as defined later in this chapter, will help
Intrauterine infection is usually apparent at birth and is char- assess the extent of brain disease.
acterized by a triad of findings: (a) skin vesicles and/or scar-
ring (cuteus aplasia), (b) eye disease (chorioretinitis, optic
atrophy), and (c) brain disease (microcephaly, encephaloma- Encephalitis
lacia, or hydranencephaly). Serial ultrasound examination of
the mothers of those babies infected in utero has occasionally Infection of the CNS alone or in combination with dissemi-
demonstrated hydranencephaly. Retinitis alone can be a pre- nated disease presents with findings indicative of encephalitis.
senting sign and should alert the pediatrician to the possibility Overall, nearly 90% of babies with brain infection caused by
of intrauterine HSV infection, although HSV infection is a less HSV have evidence of an acute neurologic syndrome. Brain
common cause of chorioretinitis relative to other congenital infection can occur in one of two fashions: either as a com-
infections. The frequency of occurrence of intrauterine HSV ponent of multiorgan disseminated infection or as encepha-
infection has been estimated to range between 1 in 100,000 litis only, with or without skin, eye, and mouth involvement.
(0.001%) and 1 in 200,000 (0.0005%) deliveries (138). Nearly one third of all babies with neonatal HSV infection
A small group of children will have skin vesicles or eye have only the encephalitic component of disease.
lesions, which are present at the time of delivery. These Clinical manifestations of these two types of encephali-
neonates are often born to women who have had prolonged tis include seizures (both focal and generalized), lethargy,
rupture of membranes. The babies have no other findings of irritability, tremors, poor feeding, temperature instability,
invasive multiorgan involvement; specifically, there is no cho- bulging fontanel, and pyramidal tract signs. Whereas babies
rioretinitis, encephalitis, or evidence of other diseased organs. with disseminated infection often have skin vesicles in asso-
ciation with brain infection, the same is not true for babies
with encephalitis alone. In this latter group, only approxi-
Disseminated Infection mately 60% have skin vesicles at any time during the disease
course (140,169171). Cultures of CSF yield virus in 25% to
Table 9.3 summarizes the classification of 297 babies with 40% of all patients. Anticipated findings on CSF examina-
neonatal HSV infection from the NIAID CASG. Disseminated tion include pleocytosis and proteinosis (as high as 500 to
HSV infection has the worst prognosis with regard to mor- 1,000 mg/dL). Although a few babies with CNS infection,

Scheld_Ch09.indd 147 2/21/14 5:33 PM


148 Part II: Viral Infections and Related Disorders

TA B L E 9 . 3
DEMOGRAPHIC AND CLINICAL CHARACTERISTICS OF INFANTS
ENROLLED IN NIAID COLLABORATIVE ANTIVIRAL STUDY

Disease Classification

Disseminated Central Nervous Skin, Eye, and


(%) System (%) Mouth (%)

No. of babies 94 (32) 101 (34) 102 (34)


No. of boys/number of girls 55/39 51/50 53/49
Race
No. of whites/number of others 60/34 76/25 76/26
No. of premature infants (36 weeks) 39 (41) 24 (24) 30 (29)
Gestational age (mean weeks SEM) 36.5 0.41 37.9 0.36 37.7 0.33
Enrollment age (mean days SEM) 11.7 0.68 17.0 0.79 12.0 1.1
Maternal age (mean years SEM) 21.7 0.50 23.1 0.46 22.7 0.52
Clinical findings (number)
Skin lesions 67 (71) 68 (67) 87 (85)
Brain involvement 50/83a (60) 101 (100) 0
Pneumonia 35/82 (43)a 0 0
Mortality at 1 yearb 58 (62) 15 (15) 0
Neurologic impairment of survivorsc
(number affected/total number)
Total 10/27b (37) 37/75 (49) 7/82b (9)
Adenine arabinoside 7/19 (37) 21/46 (46) 3/32 (9)
Acyclovir 2/6 (33) 14/26 (54) 1/43 (2)
Placebo 1/2 (50) 2/3 (67) 3/7 (43)

NIAID, National Institute of Allergy and Infectious Diseases.


a
Denominators vary according to number of patients who were evaluated.
b
Regardless of therapy.
c
Denominators vary according to number of patients with follow-up available.
From Whitley RJ. Herpes simplex virus infections. In: Remington J, Klein J, eds. Infectious Diseases of the
Fetus and Newborn Infants. 3rd ed. Philadelphia: WB Saunders; 1990:282305, with permission.

demonstrated by brain biopsy, have been reported to have Several points warrant reiteration. Clinical manifestations
no abnormalities of their CSF, certainly this is very uncom- of disease in children with encephalitis alone are virtually
mon. Serial CSF examinations provide a useful diagnostic identical to those findings that occur with brain infection in
approach because the infected newborn with brain disease disseminated cases, in spite of the presumed differences in
demonstrates progressive increases in its protein content. The pathogenesis. For babies with encephalitis only, approximately
importance of CSF examinations in all infants is underscored
by the finding that even subtle changes have been associated
with significant developmental abnormalities (172). An EEG,
CT, or MRI can be very useful in defining the presence of
CNS abnormalities. A characteristic MRI scan is shown in
Figure 9.10. Death occurs in 50% of babies with localized
CNS disease who are not treated, and it is usually related
to involvement of the brainstem. In the absence of antiviral
therapy, with rare exceptions, survivors are left with neuro-
logic impairment, and the long-term prognosis after either
disseminated infection or encephalitis alone is particularly
poor. Up to 50% of surviving children have some degree of
psychomotor retardation, often in association with micro-
cephaly, hydranencephaly, porencephalic cysts, spasticity,
blindness, chorioretinitis, or learning disabilities. Whether
visceral or CNS damage can be progressive after initial clear-
ance of the viral infection is unclear, but it is a possibility
suggested by long-term assessment of children with skin, eye,
or mouth disease (140,173) and more recently by a study of FIGURE 9.10 MRI scans of CNS neonatal herpes simplex virus infec-
a group of babies with more severe disease (174). tion (bitemporal disease).

Scheld_Ch09.indd 148 2/21/14 5:33 PM


Chapter 9: Herpes Simplex Virus 149

60% develop evidence of a vesicular rash characteristic of HSV from these babies was subjected to PCR analysis, evidence of
infection. Thus, a newborn with pleocytosis and proteinosis HSV DNA was detected in virtually all of these children, indi-
of the CSF but without a rash can easily be misdiagnosed as cating an asymptomatic infection of the CNS (179).
having bacterial or other viral infection unless HSV infection
is carefully considered. In such circumstances, a history of
genital lesions in the mother or her sexual partner may be very Diagnosis
important in the incrimination of HSV as a cause of illness.
The appropriate use of laboratory tools is essential if a diag-
nosis of HSV infection is to be achieved (180). Virus isola-
Skin, Eye, and/or Mouth Infection tion remains one of two definitive diagnostic methods. If skin
lesions are present, a scraping of skin vesicles should be made
Infection localized to the skin, eye, and/or mouth is associated and transferred (in appropriate virus transport media) to a
with virtually no mortality. When infection is localized to the diagnostic virology laboratory. Clinical specimens should
skin, the presence of discrete vesicles remains the hallmark of be shipped on ice for prompt inoculation into cell culture
disease. Clusters of vesicles (Fig. 9.11) often appear initially systems that are susceptible for the demonstration of the cy-
upon the presenting part of the body that was in direct contact topathic effects characteristic of HSV replication. Specimen
with the virus during birth. With time, the rash can progress to shipping and processing should be expedited. In addition to
involve other areas of the body as well. Vesicles occur in 80% sampling vesicle fluid, other sites from which virus may be
of children with skin, eye, or mouth infection. Children with isolated include the CSF, stool, urine, throat, nasopharynx,
disease localized to the skin, eye, or mouth generally present and conjunctivae. In infants with evidence of hepatitis or
at about 10 to 11 days of life. Those babies with skin lesions other gastrointestinal abnormalities, duodenal aspirates are
invariably suffer from recurrences whether therapy is admin- useful for HSV isolation. The virologic results of cultures
istered or not. Although death is not associated with disease from these anatomic sites should be used in conjunction with
localized to the skin, eye, and/or mouth, approximately 30% clinical findings to define the extent of disease in the new-
of these children eventually develop evidence of neurologic im- born. Typing of an HSV isolate must be done for prognostic
pairment in the absence of antiviral therapy, which can result purposes.
in significant neurologic morbidity (140,173). Cytologic examination of cells from the maternal cer-
Infections involving the eye may manifest as keratocon- vix or from the infants skin, mouth, conjunctivae, or cor-
junctivitis or later chorioretinitis. The eye can be the only neal lesions has a sensitivity of only approximately 60%
site of HSV involvement in the newborn. Findings include to 70% and, therefore, should not be the sole diagnostic
keratoconjunctivitis, microphthalmia, or retinal dysplasia. determinant for infection in the newborn (11). Cellular mate-
In the presence of persistent disease and no therapy, chorio- rial obtained by scraping the periphery of the base of lesions
retinitis can result. Chorioretinitis can be caused by either should be smeared on a glass slide and promptly fixed in cold
HSV-1 or HSV-2 (175177). Keratoconjunctivitis, even in the ethanol. The slide can be stained according to the methods
presence of therapy, can progress to chorioretinitis, cataracts, of Papanicolaou, Giemsa, or Wright before examination by
and retinal detachment. Cataracts have been detected on a trained cytologist. Deployment of Giemsa or, alternatively,
long-term follow-up of proven perinatally acquired HSV in- Tzanck smears likely will not demonstrate the presence of
fections (178). intranuclear inclusions. Intranuclear inclusions and multinu-
Long-term neurologic impairment has been encountered cleated giant cells are indicative, but not diagnostic, of HSV
in children whose disease appeared localized to the skin, eye, infection. The use of HSV monoclonal antibodies for rapid
and/or mouth. The significant findings include spastic quad- diagnosis has gained widespread acceptance. These fluores-
riplegia, microcephaly, and blindness. Despite normal clinical cence studies should be performed by laboratories experi-
and CSF examinations at the time these children completed enced in the procedure.
antiviral therapy, neurologic impairment became apparent Serologic diagnosis of HSV infection is not of great clini-
between 6 months and 1 year of life. In retrospect, when CSF cal value. Therapeutic decisions cannot await the results of

FIGURE 9.11 Characteristic vesicular rash.

Scheld_Ch09.indd 149 2/21/14 5:33 PM


150 Part II: Viral Infections and Related Disorders

serologic studies. The inability to differentiate transplacentally Skin, eyes, or mouth, vidarabine (n = 31) or
acquired maternal immunoglobulin G from endogenously acyclovir (n = 54)
1.0
produced antibodies makes the assessment of the neonates
antibody status both difficult and of little value during acute 0.9 CNS, vidarabine (n = 36)
infection. Commercially available serologic tests are now
capable of distinguishing HSV-1 from HSV-2 antibodies. 0.8 CNS, acyclovir (n = 35)

Proportion Surviving
These assays are based on differences in glycoprotein gG1 and 0.7
gG2 (181). These are the only antibody assays that should be
used. Serial antibody assessments may be useful if a mother 0.6
without a history of HSV infection has a primary infection Disseminated, vidarabine (n = 28)
late in gestation and, therefore, transfers little or no antibody 0.5
to the fetus. 0.4
The use of CT and MRI scans to define CNS disease is Disseminated; acyclovir (n = 18)
essential, even in the child who appears normal. 0.3

0.2

Polymerase Chain Reaction to 0.1


Detect Viral DNA 0
0 60 120 180 240 300 360
The other definitive diagnostic method is PCR detection of
viral DNA, as discussed earlier in this chapter (121,179,182). Survival (days)
PCR has been applied to blood as an additional site for
FIGURE 9.12 Mortality in patients with disseminated disease and
diagnosis that is especially useful in babies with disseminated CNS disease.
disease.

Prevention possibility of disease progression should encourage the early


institution of therapy. Of children presenting with disease
Cesarean Section localized to the skin, eye, and/or mouth, approximately 70%
will progress to involve the CNS or result in disseminated
Mothers with active herpetic lesions should deliver their infection (169). When such events occur, the likelihood of an
child by cesarean section if delivery can be achieved within adequate outcome, even with efficacious drugs, is not optimal
4 hours of membrane rupture (147). Cesarean section is of because many of these children will either die or be left with
unproven benefit if membranes have been ruptured for more significant neurologic impairment. The following paragraphs
than 4 hours. Recent data support this approach (147). summarize our knowledge of therapy.
Furthermore, infection of the newborn has occurred in spite First, the overall mortality rate for babies with encephali-
of delivery by cesarean section (183), which was performed tis or disseminated infection 1 year after treatment with high
specifically to prevent neonatal infection. doses of acyclovir (20 mg/kg every 8 hours for 21 days) is
lower than that of prior studies of neonatal HSV infection
Vaccination that used lower doses, as compared in Figure 9.12 (184,185).
Though various strategies for prevention of neonatal infection There are no differences in either adverse effects or laboratory
must be instituted, eventual control of HSV infection is most toxicity.
likely to be achieved through vaccination, and there is consid- Second, irrespective of the therapeutic modality employed,
erable research under way to design and test HSV vaccines, there has been a significant increase in the number of babies
in spite of a recent failure to prevent HSV-2 infections (85). who returned to normal function. This can be accounted for
Several principles should be understood. First, the efficacy of largely by the introduction of therapy before the develop-
the vaccine must be sufficient to prevent transmission of infec- ment of encephalitis or disseminated disease, as reflected by
tion. It is highly unlikely that any vaccine will totally prevent Table 9.4 (185). Of the babies entered in the trial referenced in
infection. Second, high titers of antibody against HSV do not Table 9.4, more than 48% have disease localized to the skin,
protect humans from reactivation of latent infection. Third, eye, and mouth (185). This represents a threefold increase in
live virus vaccines tend to induce more potent and durable the number of babies with skin, eye, and mouth involvement,
humoral and cellular immune responses than subunit or puri- when compared with that of previous studies and historic data
fied glycoprotein vaccines. (p .001). The change in spectrum of disease presentation is
most likely related to earlier diagnosis. The number of babies
with encephalitis has remained fairly constant at approximately
Treatment 30%, whereas the number of babies with disseminated disease
has decreased to 22%. Nevertheless, improved morbidity by
disease classification is unchanged for encephalitis (Fig. 9.13).
Background Third, available data indicate that therapy has not been
Of all the perinatally acquired infections, the one most likely initiated any earlier in the most recent neonatal HSV stud-
to be amenable to successful therapy is that caused by HSV. ies (186) as compared to earlier studies (185). However, the
Because most babies acquire infection at the time of delivery or mean duration of disease for all children (irrespective of dis-
shortly thereafter, successful antiviral therapy should decrease ease classification) entered into these studies was 4 to 5 days;
mortality and improve long-term outcome. Inherent in these therapy can, therefore, be instituted even earlier in the disease
presumptions is the recognition that diagnosis early after the course. This window for earlier administration of therapy
onset of clinical illness is essential, as is the case with peri- is significant if further advances in therapeutic outcome are to
natally acquired bacterial infections. Equally importantly, the be achieved.

Scheld_Ch09.indd 150 2/21/14 5:33 PM


Chapter 9: Herpes Simplex Virus 151

TA B L E 9 . 4
ASSESSMENT OF MORBIDITY AFTER 12 MONTHS IN INFANTS WITH NEONATAL HSV INFECTION TREATED
WITH VIDARABINE OR ACYCLOVIR

Morbidity After 12 Months (Number of Infants)


Alive After 12 Months; Dead within
Extent of Disease Normal Mild Moderate Severe Subtotal Morbidity Unknown 12 Months Total
Skin, eye, or mouth infection
Vidarabine 22 1 1 1 25 6 0 31
Acyclovir 45 0 1 0 46 8 0 54
Central nervous system
infection
Vidarabine 13 1 5 11 30 1 5 36
Acyclovir 8 5 6 9 28 2 5 35
Disseminated disease
Vidarabine 7 1 0 4 12 2 14 28
Acyclovir 3 1 0 1 5 2 11 18
Total 98 9 13 26 146 21 35 202

From Whitley RJ, Arvin A, Prober C, et al. A controlled trial comparing vidarabine with acyclovir in neonatal herpes simplex virus infection. N Engl J
Med. 1991;324:444449, with permission.

The existing database from the NIAID CASG has provided Long-Term Therapy with Oral Acyclovir
insight into those factors that influence outcome, as summarized
in Table 9.5 (187). Those factors that have a major impact on The use of oral acyclovir therapy for prolonged periods for
outcome include disease classification, level of consciousness, 6 months has recently been shown to improve neurologic
time of initiation of therapy, virus type (Fig. 9.14), and the outcome in children with encephalitis such that over 60%
virus type and frequency of skin recurrences for babies whose of children with CNS disease returned to normal function
disease is localized to the skin, eye, and mouth. Our under- (188). This finding implies the chronic replication of HSV in
standing of these data implies that limitation of disease before the brain.
there has been extensive multiorgan involvement or disease of
the CNS is associated with the best prognosis. This informa-
Long-Term Follow-up
tion will be useful in developing therapeutic strategies and in Children with neonatal HSV infection require frequent and
counseling parents of children with neonatal HSV infection. detailed long-term follow-up. Children with CNS or dis-
From a laboratory perspective, a CSF that remains PCR seminated disease are at risk for neurologic impairment.
positive at the conclusion of therapy predicts a poor prognosis Management of resultant seizure disorders is standard. Even
and has resulted in prolonged therapy until the viral DNA can children with skin, eye, and/or mouth disease are at risk for
no longer be detected at that site. neurologic impairment and must be followed carefully.

n = 46 n=2 n = 28 n = 13 n=5 n = 18
100

80
Percentage

60

40

20

0
30 60 30 60 30 60
SEM Disease CNS Disease Disseminated
Disease FIGURE 9.13 Neonatal morbidity among
patients with known outcomes after
Severe Moderate Mild Normal 12 months of life.

Scheld_Ch09.indd 151 2/21/14 5:33 PM


152 Part II: Viral Infections and Related Disorders

TA B L E 9 . 5
PROGNOSTIC FACTORS IDENTIFIED BY MULTIVARIATE ANALYSES FOR
NEONATES WITH HSV INFECTION

Relative Risk

Dominant Factors Mortality Morbidity

Total group (n 202)


Extent of disease
Skin, eyes, or mouth 1 1
CNS 5.8b 4.4b
Disseminated 33b 2.1b
Level of consciousness
Alert or lethargic 1 NSa
Semicomatose or comatose 5.2b NSa
Disseminated intravascular coagulopathy 3.8b NSa
Prematurity 3.7b NSa
Virus type
1 2.3c 1
2 1 4.9b
Seizures NS 3.0c
Infants with disseminated disease (n 46)
Disseminated intravascular coagulopathy 3.5b NSa
Level of consciousness
Alert or lethargic 1 1
Semicomatose or comatose 3.9b 4.0b
Pneumonia 3.6b NSa
Infants with CNS involvement (n 71)
Level of consciousness
Alert or lethargic 1 NSa
Semicomatose or comatose 6.1b NSa
Prematurity 5.2b NSa
Seizures NS 3.4b
Infants with infection of the skin, eyes, or mouth (n 85)
No. of skin-vesicle recurrences
3 NA 1
3 NA 21b
Virus type
1 NA 1
2 NA 14c,d

a
NS, not statistically significant (p .05); NA, not applicable (no baby with disease confined to the skin,
eyes, or mouth died).
b
p .01.
c
p .05.
d
Because of the correlation between virus type and skin-vesicle recurrence, virus type was not significant in
the multivariate model; however, it was significant as a single factor.
From Whitley RJ, Arvin A, Prober C, et al. Predictors of morbidity and mortality in neonates with herpes
simplex virus infections. N Engl J Med. 1991;324:450454, with permission.

Scheld_Ch09.indd 152 2/21/14 5:33 PM


Chapter 9: Herpes Simplex Virus 153

100
90
80
70
Percentage

60
50
40
30
20
10
0
HSV-1 HSV-2 HSV-1 HSV-2 HSV-1 HSV-2
SEM Disease CNS Disease Disseminated
Disease FIGURE 9.14 Morbidity and mortality
among patients after 12 months of age
Dead Severe Moderate Mild Normal by viral type, from 1981 to 1997.

References
1. Whitley RJ, Gnann JW. Viral encephalitis: familiar infections and emerging 23. Nesburn AB, Cook ML, Stevens JG. Latent herpes simplex virus isolation
pathogens. Lancet. 2002;359:507513. from rabbit trigeminal ganglia between episodes of recurrent ocular infec-
2. Mettler C. History of Medicine. New York: McGraw-Hill (Blakiston); 1947. tion. Arch Ophthalmol. 1972;88:412417.
3. Astruc J. De Morbis Venereis Libri Sex. Paris: G. Cavelier; 1736. 24. Stevens JG, Cook ML. Latent herpes simplex virus in spinal ganglia.
4. Gruter W. Das herpesvirus, seine atiologische und klinische bedeutung. Science. 1971;173:843845.
Munch Med Wschr. 1924;71:1058. 25. Stevens JG, Cook ML. Latent herpes simplex virus in sensory ganglia.
5. Goodpasture E. The axis-cylinders of peripheral nerves as portals of entry Perspect Virol. 1974;8:171.
to the central nervous system for the virus of herpes simplex encephalitis in 26. Warren KG, Brown SM, Wrobelwska A, et al. Isolation of latent herpes
man. Am J Pathol. 1925;1:1128. simplex virus from the superior cervical and vagus ganglions of human
6. Mathewson Commission. Epidemic Encephalitis: Etiology, Epidemiology, beings. N Engl J Med. 1978;298:10681069.
Treatment. Report of a Survey by the Mathewson Commission. New York: 27. Cushing H. Surgical aspects of major neuralgia of trigeminal nerve: report
Columbia University Press; 1929. of 20 cases of operation upon the Gasserian ganglion with anatomic and
7. Smith MG, Lennette EH, Reames HR. Isolation of the virus of herpes sim- physiologic notes on the consequences of its removal. JAMA. 1905;44:
plex and the demonstration of intranuclear inclusions in a case of acute 7733379, 860865, 920929, 10021008.
encephalitis. Am J Pathol. 1941;17:5568. 28. Goodpasture EW. Herpetic infections with special reference to involvement
8. Zarafonetis CJD, Smodel MC, Adams JW, et al. Fatal herpes simplex of the nervous system. Medicine. 1929;8:223243.
encephalitis in man. Am J Pathol. 1944;20(3):429445. 29. Carlton CA, Kilbourne ED. Activation of latent herpes simplex by trigemi-
9. Nahmias AJ, Dowdle WR. Antigenic and biologic differences in herpesvi- nal sensory-root section. N Engl J Med. 1952;246:172.
rus hominis. Prog Med Virol. 1968;10:110159. 30. Pazin GJ, Ho M, Jannetta PJ. Reactivation of herpes simplex virus after
10. Bernstein DI, Bellamy AR, Hook EW III, et al. Epidemiology, clinical presen- decompression of the trigeminal nerve root. J Infect Dis. 1978;138:
tation, and antibody response to primary infection with herpes simplex virus 405409.
type 1 and type 2 in young women. Clin Infect Dis. 2013;56(3):344351. 31. Pazin GJ, Armstrong JA, Lam MT, et al. Prevention of reactivation of
11. Roizman B, Knipe D, Whitley R. Herpes simplex virus. In: Knipe D, herpes simplex virus infection by human leukocyte interferon after opera-
Howley D, Griffin DE, et al, eds. Fields Virology. 6th ed. Philadelphia: tion on the trigeminal route. N Engl J Med. 1979;301:225230.
Lippincott Williams & Wilkins; 2013. 32. Ellison SA, Carlton CA, Rose HM. Studies of recurrent herpes simplex
12. Whitley RJ, Roizman B. Herpes simplex viruses. In: Whitley RJ, Richman infections following section of the trigeminal nerve. J Infect Dis.
D, Hayden F, eds. Clinical Virology. 3rd ed. Washington, DC: ASM 1959;105:161167.
Publishing; 2009. 33. Walz MA, Price RW, Nortkins AL. Latent ganglionic infections with herpes
13. Boos J, Esiri MM. Sporadic encephalitis. In: Viral Encephalitis: Pathology, simplex virus types 1 and 2: viral reactivation in vivo after neurectomy.
Diagnosis and Management. Boston: Blackwell Scientific Publishers.1986; Science. 1974;184:11851187.
5593. 34. Rector JT, Lausch RN, Oakes JE. Use of monoclonal antibodies for analy-
14. Boos J, Kim JH. Biopsy histopathology in herpes simplex encephalitis and sis of antibody-dependent immunity to ocular herpes simplex virus type 1
in encephalitis of undefined etiology. Yale J Bio Med. 1984;57:751755. infection. Infect Immun. 1982;38:168174.
15. Kapur N, Barker S, Burrows EH, et al. Herpes simplex encephalitis: long 35. Segal AL, Katcher AH, Brightman VJ, et al. Recurrent herpes labialis,
term magnetic resonance imaging and neuropsychological profile. J Neurol recurrent aphthous ulcers and the menstrual cycles. J Dent Res. 1974;53:
Neurosurg Psychiatry.. 1994;57:13341342. 797803.
16. Garcia JH, Colon LE, Whitley RJ, et al. Diagnosis of viral encephalitis by 36. Baringer JR. Recovery of herpes simplex virus from human sacral ganglions.
brain biopsy. Semin Diagn Pathol. 1984;1:7181. N Engl J Med. 1974;291:828830.
17. Hill TJ. Herpes simplex virus latency. In: Roizman B, ed. The Herpesviruses. 37. Cabrera CV, Wholenberg C, Openshaw H, et al. Herpes simplex virus DNA
New York: Plenum Publishing; 1985:175240. sequences in the CNS of latently infected mice. Nature. 1980;288:288290.
18. Cook ML, Stevens JG. Pathogenesis of herpetic neuritis and ganglionitis 38. Fraser NW, Lawrence WC, Wroblewska A, et al. Herpes simplex type 1
in mice: evidence of intra-axonal transport of infection. Infect Immun. DNA in human brain tissue. Proc Natl Acad Sci. 1981;78:64616465.
1973;7:272288. 39. Warren KG, Gilden DH, Brown SM. Isolation of herpes simplex virus from
19. Roizman B, Whitley RJ. An inquiry into the molecular basis of HSV latency human trigeminal ganglia, including ganglia from one patient with multiple
and reactivation. Annu Rev Microbiol. 2013;67:355374. sclerosis. Lancet. 1977;2:637639.
20. Baringer JR. The biology of herpes simplex virus infection in humans. Surv 40. Nahmias AJ, Whitley RJ, Visintine AN, et al. Herpes simplex encephali-
Ophthalmol. 1976;21:171. tis: laboratory evaluations and their diagnostic significance. J Infect Dis.
21. Baringer JR, Swoveland P. Recovery of herpes simplex virus from human 1982;145:829836.
trigeminal ganglions. N Engl J Med. 1973;288:648650. 41. Whitley RJ, Lakeman AD, Nahmias AJ, et al. DNA restriction-enzyme
22. Bastian FO, Rabson AS, Yee CL. Herpesvirus hominis: isolation from analysis of herpes simplex virus isolates obtained from patients with
human trigeminal ganglion. Science. 1972;178:306. encephalitis. N Engl J Med. 1982;307:10601062.

Scheld_Ch09.indd 153 2/21/14 5:33 PM


154 Part II: Viral Infections and Related Disorders

42. Johnson RT, Olson LC, Buescher EL. Herpes simplex virus infections 75. Evans AS, Dick EC. Acute pharyngitis and tonsillitis in University of
of the nervous system. Problems in laboratory diagnosis. Arch Neurol. Wisconsin students. JAMA. 1964;190:699708.
1968;18(3):260264. 76. Crespi CM, Cumberland WG, Wald A, et al. Longitudinal study of herpes
43. Dinn JJ. Transolfactory spread of virus in herpes simplex encephalitis. simplex virus type 2 infection using viral dynamic modelling. Sex Transm
Br Med J. 1980;281:1392. Infect. 2007;83(5):359364.
44. Ojeda VJ, Archer M, Robertson TA, et al. Necropsy study of olfactory 77. Lee AJ, Ashkar AA. Herpes simplex virus-2 in the genital mucosa: insights
portal of entry in herpes simplex encephalitis. Med J Aust. 1983;1:7981. into the mucosal host response and vaccine development. Curr Opin
45. Twomey JA, Barker CM, Robinson G, et al. Olfactory mucosa in herpes Infect Dis. 2012;25(1):9299.
simplex encephalitis. J Neurol Neurosurg Psych. 1979;42:983987. 78. Sacks SL, Griffiths PD, Corey L, et al. HSV-2 transmission. Antiviral Res.
46. Schlitt M, Lakeman FD, Wilson ER, et al. A rabbit model of focal herpes 2004;63(suppl 1):S27S35.
simplex encephalitis. J Infect Dis. 1986;153:732735. 79. Armstrong GL, Schillinger J, Markowitz L, et al. Incidence of herpes sim-
47. Stroop WG, Schaefer DC. Production of encephalitis restricted to the tem- plex virus type 2 infection in the United States. Am J Epidemiol. 2001;
poral lobes by experimental reactivation of herpes simplex virus. J Infect 153:912920.
Dis. 1986;153:721731. 80. Institute of Medicine, Committee on Issues and Priorities for New Vaccine
48. Rock DL, Frasher NW. Detection of HSV-1 genome in central nervous sys- Development. Appendix I: Prospects for immunizing against herpes sim-
tem of latently infected mice. Nature. 1983;302:523531. plex virus 1 and 2. In: New Vaccine Development Establishing Priorities:
49. Davis LE, Johnson RT. An explanation for the localization of herpes sim- Disease of Importance in the United States. Washington, DC: National
plex encephalitis. Ann Neurol. 1979;5:25. Academy Press; 1985:280312.
50. Griffith JR, Kibrick S, Dodge PR, et al. Experimental herpes simplex 81. Bernstein D, Stokes-Riner A, Hook E, et al. Epidemiology, clinical pre-
encephalitis: electroencephalographic, clinical, virologic, and pathologic sentation and antibody response to primary infection with herpes simplex
observations in the rabbit. Electroencephalogr Clin Neurophysiol. 1967;23: virus type 1 and type 2 in young women. Clin Infect Dis. 2013;56(3):
263267. 344351.
51. Guo Y, Audry M, Ciancanelli M, et al. Herpes simplex virus encephalitis in 82. Ng ABP, Reagin JW, Yen SS. Herpes genitalisclinical and cytopathologic
a patient with complete TLR3 deficiency: TLR3 is otherwise redundant in experience with 256 patients. Obstet Gynecol. 1970;36:645651.
protective immunity. J Exp Med. 2011;208(10):20832098. 83. Ekwo E, Wong YW, Myers M. Asymptomatic cervicovaginal shedding of
52. Black FL. Infectious diseases in primitive societies. Science. 1975;187: herpes simplex virus. Am J Obstet Gynecol. 1979;134:102103.
515518. 84. Rattray MC, Corey L, Reeves WC, et al. Recurrent genital herpes among
53. Glezen WP, Fernald GW, Lohr JA. Acute respiratory disease of university women: symptomatic versus asymptomatic viral shedding. Br J Vener Dis.
students with special references to the etiologic role of herpesvirus hominis. 1978;54:262265.
Am J Epidemiol. 1975;101:111121. 85. Belshe RB, Leone PA, Bernstein DI, et al. Efficacy results of a trial of a
54. Burnet FM, Williams SW. Herpes simplex: new point of view. Med J Aust. herpes simplex vaccine. N Engl J Med. 2012;366(1):3443.
1939;1:637640. 86. Wald A, Zeh J, Selke S, et al. Reactivation of genital herpes simplex virus
55. Dodd K, Johnston LM, Buddingh GJ. Herpetic stomatitis. J Pediatr. type 2 infection in asymptomatic seropositive persons. N Engl J Med.
1938;12:95. 2000;342(12):844850.
56. Buddingh GH, Schrum DI, Lanier JC, et al. Studies of the natural history of 87. Wald A, Link K. Risk of human immunodeficiency virus infection in her-
herpes simplex infections. Pediatrics. 1953;11:595610. pes simplex virus type 2-seropositive persons: a meta-analysis. J Infect
57. Juretic M. Natural history of herpetic infection. Helv Pediatr Acta. Dis. 2002;185:4552.
1966;21:356. 88. Adam E, Kaufman RH, Mirkovic RR, et al. Persistence of virus shed-
58. Cesario TC, Poland JD, Wulff H, et al. Six years experiences with herpes ding in asymptomatic women after recovery from herpes genitalis. Obstet
simplex virus in a childrens home. Am J Epidemiol. 1969;90:416422. Gynecol. 1979;54:171173.
59. Douglas RG Jr, Couch RB. A prospective study of chronic herpes sim- 89. Becker WB. The epidemiology of herpesvirus infection in three racial com-
plex virus infection and recurrent herpes labialis in humans. J Immunol. munities in Cape Town. S Afr Med J. 1966;40:109111.
1970;104:289295. 90. International Herpes Management Forum. Strategies for interrupting
60. Kloene W, Bang FB, Chakroborty SM, et al. A two-year respiratory virus the transmission of genital and neonatal HSV infection. Herpes. 2004;
survey in four villages in West Bengal, India. Am J Epidemiol. 1970;92: 11(suppl 3):129A186A.
307320. 91. Rawls WE, Adam E, Melnick JL. Geographical variation in the associa-
61. Lindgren KM, Douglas RG Jr, Couch RB. Significance of herpesvirus homi- tion of antibodies to herpesvirus type 2 and carcinoma of the cervix. In:
nis in respiratory secretions of man. N Engl J Med. 1968;276:517523. Biggs PM, de The G, Payne LN, eds. Oncogenesis and Herpesviruses.
62. Stern H, Elek SD, Miller DM, et al. Herpetic whitlow, a form of cross- Lyon, France: Scientific Publication II, International Agency for Research
infection in hospitals. Lancet. 1959;2:871. on Cancer; 1972:424427.
63. Young SK, Rowe NH, Buchanan RA. A clinical study for the control of 92. Rawls WE, Gardner HL. Herpes genitalis: venereal aspects. Clin Obstet
facial mucocutaneous herpes virus infections. I. Characterization of natu- Gynecol. 1972;15:912918.
ral history in a professional school population. Oral Surg Oral Med Oral 93. Rawls WE, Garfield CH, Seth P, et al. Serological and epidemiological
Pathol. 1976;41:498507. considerations of the role of herpes simplex virus type 2 in cervical cancer.
64. Scott TF, Steigman AJ, Convey JH. Acute infectious gingivostomatitis: Cancer Res. 1976;36:829835.
etiology, epidemiology, and clinical pictures of a common disorder caused 94. Rawls WE, Gardner HL, Flanders RW, et al. Genital herpes in two social
by the virus of herpes simplex. JAMA. 1941;117:999. groups. Am J Obstet Gynecol. 1971;110:682689.
65. Templeton AC. Generalized herpes simplex in malnourished children. 95. Chang TW, Fiumara NJ, Weinstein L. Genital herpes: some clinical and
J Clin Pathol. 1970;23:2430. laboratory observations. JAMA. 1974;229:544545.
66. Rawls WE, Campione-Piccardo J. Epidemiology of herpes simplex virus 96. Reeves WC, Corey L, Adams HG, et al. Risk of recurrence after first
type 1 and 2. In: Nahmias A, Dowdle W, Schinazi R, eds. The Human episodes of genital herpes: relation to HSV type and antibody response.
Herpesviruses: An Interdiscipinary Perspective. Amsterdam: Elsevier/ N Engl J Med. 1981;305:315319.
North-Holland; 1981:137152. 97. Choi NW, Shettigara PT, Abu-Zeid HA, et al. Herpesvirus infection and cer-
67. Bader C, Crumpacker CS, Schnipper LE, et al. The natural history of vical anaplasia: a seroepidemiological study. Int J Cancer. 1977;19:167171.
recurrent facial-oral infection with herpes simplex virus. J Infect Dis. 98. Lafferty WE, Coombs RW, Benedetti J, et al. Recurrences after oral and
1978;138:897905. genital herpes simplex virus infection. Influence of site of infection and
68. Black FL, Hierholzer WJ, Pinheiro F, et al. Evidence for persistence viral type. N Engl J Med. 1987;316:14441449.
of infectious agents in isolated human populations. Am J Epidemiol. 99. Corey L, Ashley R, Benedetti J, et al. The effect of prior HSV-1 infection
1974;100:230250. on the subsequent natural history of genital HSV-2. Presented at: 28th
69. Nahmias AJ, Josey WE, Naib ZM, et al. Antibodies to Herpesvirus hominis Interscience Conference on Antimicrobial Agents and Chemotherapy;
type 1 and 1 in humans. I. Patients with genital herpetic infections. Am J October 23-26, 1988; Los Angeles, California.
Epidemiol. 1970;91:539546. 100. Longson M. The general nature of viral encephalitis in the United
70. Nahmias AJ, Josey WE, Naib ZM, et al. Antibodies to Herpesvirus hominis Kingdom. In: Ellis LS, ed. Viral Diseases of the Central Nervous System.
types 1 and 2 in humans. II. Women with cervical cancer. Am J Epidemiol. London: Bailliere Tindall; 1984:1931.
1970;91:547552. 101. Skoldenberg B, Forsgren M, Alestig K, et al. Acyclovir versus vidarabine
71. Rawls WE, Tompkins WA, Melnick JL. The association of herpesvirus type in herpes simplex encephalitis: a randomized multicentre study in con-
2 and carcinoma of the uterine cervix. Am J Epidemiol. 1969;89:547554. secutive Swedish patients. Lancet. 1984;2:707711.
72. Sawanabori S. Acquisition of herpes simplex virus infection in Japan. Acta 102. Whitley RJ, Soong SJ, Linneman C Jr, et al. Herpes simplex encephalitis.
Paediatr Jpn Overseas Ed. 1973;15:16. Clinical Assessment. JAMA. 1982;247(3):317320.
73. Wentworth BB, Alexander ER. Seroepidemiology of infections due to mem- 103. Khetsuriani N, Holman RC, Anderson LJ. Burden of encephalitis-
bers of herpesvirus group. Am J Epidemiol. 1971;94:496507. associated hospitalizations in the United States, 19881997. Clin Infect
74. Gibson JJ, Hornung CA, Alexander GR, et al. A cross-sectional study of Dis. 2002;35:175282.
herpes simplex virus types 1 and 2 in college students: occurrence and 104. Boston Interhospital Virus Study Group and the NIAID Sponsored
determinants of infection. J Infect Dis. 1990;162:306312. Cooperative Antiviral Clinical Study. Failure of high dose 5-deoxyuridine

Scheld_Ch09.indd 154 2/21/14 5:33 PM


Chapter 9: Herpes Simplex Virus 155

in the therapy of herpes simplex virus encephalitis: evidence of unaccept- 136. Whitley RJ. Herpes simplex virus infections of the central nervous system
able toxicity. N Engl J Med. 1975;292:600603. in children. Semin Neurol. 1982;2:8797.
105. Longson M. Le defi des encephalitis herpetiques. Ann Microbiol (Paris). 137. Florman AL, Gershon AA, Blackett RP, et al. Intrauterine infection
1979;130:5. with herpes simplex virus: resultant congenital malformation. JAMA.
106. Longson MM, Bailey AS, Klapper P. Herpes encephalitis. In: Waterson 1973;225:129132.
AP, ed. Recent Advances in Clinical Virology. New York: Churchill 138. Hutto C, Arvin A, Jacobs R, et al. Intrauterine herpes simplex virus infec-
Livingstone; 1980:147157. tions. J Pediatr. 1987;110:97101.
107. Whitley RJ, Soong S-J, Dolin R, et al. Adenine arabinoside therapy of 139. South MA, Tompkins WA, Morris CR, et al. Congenital malformation of
biopsy-proved herpes simplex encephalitis: National Institute of Allergy the central nervous system associated with genital type (type 2) herpesvi-
and Infectious Diseases collaborative antiviral study. N Engl J Med. rus. J Pediatr. 1969;75:1318.
1977;297:289294. 140. Gutierrez K, Whitley RJ, Arvin A. Herpes simplex virus infections. In:
108. Whitley RJ, Soong S-J, Hirsch MS, et al. Herpes simplex encephalitis: Remington JS, Klein JO, Wilson CB, eds. Infectious Diseases of the Fetus
vidarabine therapy and diagnostic problems. N Engl J Med. 1981;304: and Newborn Infant. Philadelphia: Elsevier; 2010.
313318. 141. Stagno S, Whitley RJ. Herpesvirus infections of pregnancy. I.
109. International Herpes Management Forum. Herpesvirus infections of the Cytomegalovirus and Epstein-Barr virus infections. N Engl J Med. 1985;
central nervous system. Herpes. 2004;11(suppl 2):47A120A. 313:12701274.
110. Demaerel P, Wilms G, Robberecht W, et al. MRI of herpes simplex 142. Johnson RE, Nahmias AJ, Magder LS, et al. A seroepidemiologic survey
encephalitis. Neuroradiology. 1992;34:490493. of the prevalence of herpes simplex virus type 2 infection in the United
111. Schlesinger Y, Buller RS, Brunstrom JE, et al. Expanded spectrum of States. N Engl J Med. 1989;321:712.
herpes simplex encephalitis in childhood. J Pediatr. 1995;126:234241. 143. Fleming DT, McQuillan GM, Johnson RE, et al. Herpes simplex virus
112. Sener RN. Diffusion MRI in Rasmussens encephalitis, herpes simplex type 2 in the United States, 1976 to 1994. N Engl J Med. 1997;337:
encephalitis, and bacterial meningoencephalitis. Comput Med Imaging 11051111.
Graph. 2002;26(5):327332. 144. Corey L, Adams HG, Brown ZA, et al. Genital herpes simplex virus infec-
113. Sener RN. Herpes simplex encephalitis: diffusion MR imaging findings. tions: clinical manifestations, course and complications. Ann Intern Med.
Comput Med Imaging Graph. 2001;25:391397. 1983;98:958972.
114. Chien LT, Boehm RM, Robinson H, et al. Characteristic early electro- 145. Prober CG, Sullender WM, Yasukawa LL, et al. Low risk of herpes sim-
encephalographic changes in herpes simplex encephalitis. Arch Neurol. plex virus infections in neonates exposed to the virus at the time of vaginal
1977;34:361364. delivery to mothers with recurrent genital herpes simplex virus infections.
115. Miller JHD, Coey A. The EEG in necrotizing encephalitis. Electroencephalogr N Engl J Med. 1987;316:240244.
Clin Neurophysiol. 1959;2:582585. 146. Yeager AS, Arvin AM, Urbani LJ, et al. Relationship of antibody to outcome
116. Radermecker J. Systematique its electrocencephalographic des encephali- in neonatal herpes simplex infections. Infect Immun. 1980;29:532538.
tis it encephalopathies. Electroencephalography. 1956;8(suppl 5):1243. 147. Brown ZA, Wald A, Morrow RM, et al. Effect of serologic status and
117. Smith JB, Westmoreland BF, Reagan TJ, et al. A distinctive clinical EEG cesarean delivery on transmission rates of herpes simplex virus from
profile in herpes simplex encephalitis. Mayo Clin Proc. 1975;50:469474. mother to infant. JAMA. 2003;289:203209.
118. Upton A, Grumpert J. Electroencephalography in diagnosis of herpes sim- 148. Kaye EM, Dooling EC. Neonatal herpes simplex meningoencephalitis asso-
plex encephalitis. Lancet. 1970;1:650652. ciated with fetal monitor scalp electrodes. Neurology. 1981;31:10451047.
119. Enzmann DR, Ransom B, Norman D. Computed tomography of herpes 149. Parvey LS, Chien LT. Neonatal herpes simplex virus infection introduced
simplex encephalitis. Radiology. 1978;129:419425. by fetal monitor scalp electrode. Pediatrics. 1980;65:11501153.
120. Zimmerman RD, Russell EJ, Leeds NE, et al. CT in the early diagnosis of 150. Kohl S, West MS, Prober CG, et al. Neonatal antibody-dependent cel-
herpes simplex encephalitis. Am J Roentgenol. 1980;134:6166. lular cytoxic antibody levels are associated with the clinical presenta-
121. Rowley A, Lakeman F, Whitley R, et al. Rapid detection of herpes simplex tion of neonatal herpes simplex virus infection. J Infect Dis. 1989;160:
virus DNA in cerebrospinal fluid of patients with herpes simplex encepha- 770776.
litis. Lancet. 1990;335:440441. 151. Dunkle LM, Schmidt RR, OConnor DM. Neonatal herpes simplex in-
122. Aurelius E, Johansson B, Skoldenberg B, et al. Rapid diagnosis of herpes fection possibly acquired via maternal breast milk. Pediatrics. 1979;63:
simplex encephalitis by nested polymerase chain reaction assay of cere- 250251.
brospinal fluid. Lancet. 1991;337(8735):189192. 152. Sullivan-Bolyai JZ, Fife KH, Jacobs RF, et al. Disseminated neonatal
123. Aurelius E, Johansson B, Skoldenberg B, et al. Encephalitis in immuno- herpes simplex virus type 1 from a maternal breast lesion. Pediatrics.
competent patients due to herpes simplex virus type 1 or 2 as determined 1983;71:455457.
by type-specific polymerase chain reaction and antibody assays of cere- 153. Yeager AS, Ashley RL, Corey L. Transmission of herpes simplex virus
brospinal fluid. J Med Virol. 1993;39:179186. from father to neonate. J Pediatr. 1983;103:905907.
124. Puchhammer-Stockl E, Heinz FX, Kundi M, et al. Evaluation of the poly- 154. Douglas J, Schmidt O, Corey L. Acquisition of neonatal HSV-1 infection
merase chain reaction for diagnosis of herpes simplex virus encephalitis. from a paternal source contact. J Pediatr. 1983;103:908910.
J Clin Microbiol. 1993;31:146148. 155. Linnemann CC Jr, Buchman TG, Light IJ, et al. Transmission of herpes
125. Shoji H, Koga M, Kusuhara T, et al. Differentiation of herpes simplex simplex virus type-1 in a nursery for the newborn: identification of viral
virus 1 and 2 in cerebrospinal fluid of patients with HSV encephalitis and isolates by D.N.A. fingerprinting. Lancet. 1978;1:964966.
meningitis by stringet hybridization of PCR-amplified DNAs. J Neurol. 156. Light IJ. Postnatal acquisition of herpes simplex virus by the newborn
1994;241:526530. infant: a review of the literature. Pediatrics. 1979;63:480482.
126. Sakrauski A, Weber B, Kessler HH, et al. Comparison of two hybrid- 157. Hammerberg O, Watts J, Chernesky M, et al. An outbreak of herpes
ization assays for the rapid detection of PCR amplified HSV genome simplex virus type 1 in an intensive care nursery. Pediatr Infect Dis.
sequences from cerebrospinal fluid. J Virol Methods. 1994;50:175184. 1983;2:290294.
127. Lakeman FD, Whitley RJ; National Institute of Allergy and Infectious 158. Centers for Disease Control and Prevention. Neonatal herpes simplex
Diseases Collaborative Antiviral Study Group. Diagnosis of herpes sim- virus infection following Jewish ritual circumcisions that included direct
plex encephalitis: application of polymerase chain reaction to cerebrospi- orogenital suctionNew York City, 20002011. MMWR Morb Mortal
nal fluid from brain biopsied patients and correlation with disease. J Infect Wkly Rep. 2012;61(22):405409.
Dis. 1995;172:857863. 159. Flewett TH, Parker RGF, Philip WM. Acute hepatitis due to herpes sim-
128. Whitley RJ, Cobbs CG, Alford CA Jr, et al. Diseases that mimic herpes plex virus in an adult. J Clin Pathol. 1969;22:6066.
simplex encephalitis: diagnosis, presentation and outcome. JAMA. 1989; 160. Anderson JM, Nicholls MWN. Herpes encephalitis in pregnancy. Br
262:234239. Med J. 1972;1:632.
129. Kim C, Yoon YH. Unilateral acute retinal necrosis occurring 2 years after her- 161. Goyette RE, Donowho EM, Hieger LR, et al. Fulminant herpesvirus hom-
pes simplex type 1 encephalitis. Ophthalmic Surg Lasers. 2002;33:250252. inis hepatitis during pregnancy. Obstet Gynecol. 1974;43:191196.
130. Whitley RJ, Alford CA Jr, Hirsch MS, et al. Vidarabine versus acyclovir 162. Young EJ, Killam AP, Greene JF. Disseminated herpesvirus infection as-
therapy of herpes simplex encephalitis. N Engl J Med. 1986;314:144149. sociated with primary genital herpes in pregnancy. JAMA. 1976;235:
131. Davis LE, McLaren LE. Relapsing herpes simplex encephalitis following 27312733.
antiviral therapy. Ann Neurol. 1983;13:192195. 163. Nahmias AJ, Josey WE, Naib ZM, et al. Perinatal risk associated with
132. Dix RD, Baringer JR, Panitch HS, et al. Recurrent herpes simplex encephali- maternal genital herpes simplex virus infection. Am J Obstet Gynecol.
tis: recovery of virus after Ara-A treatment. Ann Neurol. 1983;13:196200. 1971;110:825836.
133. Wang HS, Kuo MF, Huang SC, et al. Choreoathetosis as an initial sign of 164. Vontver LA, Hickok DE, Brown Z, et al. Recurrent genital herpes simplex
relapsing of herpes simplex encephalitis. Pediatr Neurol. 1994;11:341345. virus infection in pregnancy: infant outcome and frequency of asymptom-
134. VanLandingham KE, Marsteller HB, Ross GW, et al. Relapse of her- atic recurrences. Am J Obstet Gynecol. 1982;143:7584.
pes simplex encephalitis after conventional acyclovir therapy. JAMA. 165. Tejani N, Klein SW, Kaplan M. Subclinical herpes simplex genitalis infec-
1988;259:10511053. tions in the perinatal period. Am J Obstet Gynecol. 1979;135:547.
135. Zuelzer WW, Wolf W, Stulbery CS. Herpes simplex virus as the cause of 166. Bolognese RJ, Corson SL, Fuccillo DA, et al. Herpesvirus hominis
fulminating visceral disease and hepatitis in infancy. Am J Dis Child. 1952; type-2 infections in asymptomatic pregnant women. Obstet Gynecol.
83:421439. 1976;48:507510.

Scheld_Ch09.indd 155 2/21/14 5:33 PM


156 Part II: Viral Infections and Related Disorders

167. Harger JH, Pazin GJ, Armstrong JA, et al. Characteristics and manage- 179. Kimberlin DW, Lakeman FD, Arvin AM, et al. Application of the poly-
ment of pregnancy in women with genital herpes simplex virus infection. merase chain reaction to the diagnosis and management of neonatal herpes
Am J Obstet Gynecol. 1983;145:784791. simplex virus disease. National Institute of Allergy and Infectious Diseases
168. Jeansson S, Molin L. Genital herpesvirus hominis infection. A veneral Collaborative Antiviral Study Group. J Infect Dis. 1996;174(6):11621167.
disease. Lancet. 1970;1:10641065. 180. Ware JR Jr, Maruish ME, eds. The Use of Psychological Testing for
169. Whitley RJ, Nahmias AJ, Visitine AM, et al. The natural history of her- Treatment Planning and Outcomes Assessment. 2nd ed. Mahwah, NJ:
pes simplex virus infection of mother and newborn. Pediatrics. 1980;66: Lawrence Erlbaum Associates; 1999.
489494. 181. International Herpes Management Forum. Herpes. 2002;9:3155.
170. Yeager AS, Arvin AM. Reasons for the absence of a history of recurrent 182. Hardy DA, Arvin AM, Yasukawa LL, et al. Use of polymerase chain for suc-
genital infections in mothers of neonates infected with herpes simplex cessful identification of asymptomatic genital infection with herpes simplex
virus. Pediatrics. 1984;73:188193. virus in pregnant women in delivery. J Infect Dis. 1990;162:10311035.
171. Arvin AM, Yeager AS, Bruhn FW, et al. Neonatal herpes simplex infection 183. Stone KM, Brooks CA, Guinan ME, et al. National surveillance for neo-
in the absence of mucocutaneous lesions. J Pediatr. 1982;100:715721. natal herpes simplex virus infection. Sex Trans Dis. 1989;16:152156.
172. Mizrahi EM, Tharp BR. A unique electroencephalogram pattern in neo- 184. Kimberlin DW, Lin CY, Jacobs RF, et al. Safety and efficacy of high-dose
natal herpes simplex virus encephalitis. Neurology. 1981;31:164. intravenous acyclovir in the management of neonatal herpes simplex virus
173. Whitley RJ, Nahmias AJ, Soong S-J, et al. Vidarabine therapy of neonatal infections. Pediatrics. 2001;108(2):230238.
herpes simplex virus infection. Pediatrics. 1980;66:495501. 185. Whitley RJ, Arvin A, Prober C, et al. A controlled trial comparing vida-
174. Guttman LT, Wilfert CM, Eppes S. Herpes simplex virus encephalitis in rabine with acyclovir in neonatal herpes simplex virus infection. N Engl J
children: analysis of cerebrospinal fluid and progressive neurodevelop- Med. 1991;324:444449.
mental deterioration. J Infect Dis. 1986;154:415421. 186. Kimberlin DW, Lin CY, Jacobs RF, et al. Natural history of neonatal her-
175. Nahmias AJ, Visitine A, Caldwell A, et al. Eye infections with herpes sim- pes simplex virus infections in the acyclovir era. Pediatrics. 2001;108(2):
plex viruses in neonates. Surv Ophthalmol. 1976;21:100105. 223229.
176. Nahmias A, Hagler W. Ocular manifestations of herpes simplex in the 187. Whitley RJ, Arvin A, Prober C, et al. Predictors of morbidity and mor-
newborn. Int Ophthalmol Clin. 1972;12:191213. tality in neonates with herpes simplex virus infections. N Engl J Med.
177. Reested P, Hansen B. Chorioretinitis of the newborn with herpes simplex 1991;324:450454.
type 1: Report of a case. Acta Ophthalmol. 1979;57:10962100. 188. Kimberlin DW, Whitley RJ, Wan W, et al. Oral acyclovir suppression
178. Cibis A, Burde RM. Herpes simplex virus induced congenital cataracts. and neurodevelopment after neonatal herpes. N Engl J Med. 2011;
Arch Ophthalmol. 1971;85:220223. 365(14):12841292.

Scheld_Ch09.indd 156 2/21/14 5:33 PM


CHAPTER 10 NEUROLOGIC MANIFESTATIONS
OF VARICELLA AND HERPES ZOSTER
JOHN W. GNANN, JR. AND RICHARD J. WHITLEY

Varicella-zoster virus (VZV) causes two clinically distinct diseases. figure of 2.0 cases per 1,000 persons would predict about
Varicella (chickenpox), which results from primary infection of 500,000 cases of zoster annually in the United States.
a susceptible individual, is a common, extremely contagious, Expressed another way, an individual has a 10% to 20%
and usually benign acute illness that occurs in epidemics and is risk of developing herpes zoster at some point during
characterized by a generalized vesicular rash. Like all other her- his or her lifetime. Increasing age is the most important
pesviruses, VZV establishes latency following primary infection. risk factor for the development of herpes zoster. There is
Reactivation of latent VZV results in a localized cutaneous erup- an increase in the age-specific incidence of herpes zoster
tion termed herpes zoster or shingles, a common disorder among beginning at around age 55 years; individuals older than
the elderly. Both varicella and herpes zoster can be complicated 75 years have a zoster incidence of more than 10 cases per
by central nervous system (CNS) involvement (1). 1,000 person-years (7). Zoster occurs with equal frequency
in men and women, with no seasonal association. The
other well-defined risk factor for herpes zoster is altered
EPIDEMIOLOGY cell-mediated immunity. Patients with neoplastic diseases,
especially lymphoproliferative malignancies and organ
transplant recipients, are at very high risk for development
Varicella of herpes zoster. Approximately 15% to 30% of patients
with Hodgkin disease experience herpes zoster, compared
Humans are the only known reservoir for VZV. Primary
with 2% of patients with various solid tumors (8). The dis-
infection occurs when a susceptible individual is exposed to
ease occurs in 3% to 10% of renal transplant recipients,
airborne virus by the respiratory route. Patients with chicken-
20% to 25% of cardiac transplant recipients, and 20% to
pox are contagious for about 48 hours before and 4 to 5 days
50% of allogeneic hematopoietic stem cell transplant re-
after rash onset. Infection is usually acquired after exposure
cipients (9). Herpes zoster is also seen with increased fre-
to another individual with varicella but can also result from
quency in persons infected with human immunodeficiency
exposure to a patient with herpes zoster. Varicella is highly
virus (HIV) and often serves as an initial marker of disease
infectious; attack rates of 61% to 87% have been observed
in that population (10). A longitudinal study of a cohort
following household exposure.
of HIV-seropositive men in San Francisco demonstrated
In the United States, varicella epidemics occur annually in
an incidence of zoster of 29.4 cases per 1,000 person-
the late winter and early spring, with peak numbers of cases
years, compared with 2.0 cases per 1,000 person-years in
reported in March. Before the availability of the varicella vac-
a matched population of HIV-seronegative controls (11).
cine, about 3.8 million cases of chickenpox occurred each year
in the United States (15 cases per 1,000 population), which
approximately equaled the annual birth cohort (2). Varicella
was predominantly a disease of school-aged children. About INFECTIOUS AGENT
50% to 60% of varicella cases occurred in children between
5 and 9 years of age, and 90% of cases occurred in patients VZV shares structural characteristics with other members
younger than 15 years. Previous surveys indicated that more of the family Herpesviridae. The complete virion is approxi-
than 90% of the U.S. population was VZV seropositive by mately 180 to 200 nm in diameter. It is composed of an ico-
age 20 years (3). Mortality estimates for varicella in children sahedral nucleocapsid measuring 90 to 95 nm in diameter, an
(ages 1 to 14 years) are 1.4 per 100,000 cases and in adults amorphous tegument, and a lipid-containing envelope with
31 per 100,000 cases (4). glycoprotein spikes (12). The VZV genome consists of a lin-
Introduction of the varicella vaccine in the United States in ear, double-stranded DNA molecule containing 125,000 base
1995 resulted in striking changes in the epidemiology of chick- pairs with guanosine-plus-cytosine content of 46% (13).
enpox. By monitoring vaccine and disease activity at three The genome is organized in a manner similar to that of
sentinel sites, the Centers for Disease Control and Prevention other herpesviruses, having unique long (UL; 104.5 kb) and
(CDC) showed that vaccine coverage among preschool- unique short (US; 5.2 kb) regions flanked by inverted repeats.
aged children increased from 40% in 1997 to 70% in 1999. VZV encodes approximately 69 polypeptides, including seven
Between 1995 and 1999, varicella incidence declined 80% in glycoproteins.
the surveillance areas, with the greatest decline seen in chil- VZV can be propagated in vitro in a limited number of
dren aged 1 to 4 years (5). continuous and discontinuous cell culture monolayers, mostly
of human or simian origin. In human embryonic lung fibro-
blasts, cytopathic effects begin as a focal process with subse-
Herpes Zoster quent cell-to-cell spread. Approximately 8 to 10 hours after
infection, virus-specific immunofluorescence can be detected
Herpes zoster occurs as a result of reactivation of latent VZV. in cells adjacent to the initial focus of infection. VZV is highly
The annualized incidence of herpes zoster is 1.5 to 3.0 cases cell associated, with very limited release of infectious virions
per 1,000 persons in the population (6,7). An incidence into the media.

157

Scheld_Ch10.indd 157 2/21/14 5:34 PM


158 Part II: Viral Infections and Related Disorders

PATHOGENESIS AND PATHOLOGY Pathology of Herpes Zoster


Replication of VZV in the sensory ganglion results in intense
Pathogenesis of Varicella inflammation, neuronal destruction, and focal hemorrhage.
Less severe inflammatory changes often occur in the adjacent
Varicella is transmitted via the respiratory route. Virus in air- ganglia. Occasionally, inflammation and necrosis also extend
borne droplets enters the susceptible host via mucosal surfaces to the anterior nerve root, resulting in localized motor neu-
of the conjunctiva, oropharynx, or upper respiratory tract. ropathy. These changes are accompanied by lymphocytic
VZV undergoes an initial round of replication, presumably in pleocytosis. Movement of virus from the ganglion down the
cervical lymph nodes (14). When local immune responses are sensory nerve to the skin produces acute inflammation of the
overcome, a primary viremia occurs, with widespread dissemi- nerve (19). Virus reaching the skin replicates in epithelial cells
nation of VZV to the reticuloendothelial system and possibly of the epidermis, producing pathologic changes identical to
to epithelial cells of other organs. VZV then undergoes mul- those described for varicella. Inflammatory changes in the sen-
tiple cycles of replication, resulting in a second viremic phase sory nerve persist for months and may result in demyelination,
(about 1 week after the initial viremia) that is accompanied wallerian degeneration, and sclerosis.
by the onset of clinical symptoms. VZV localizes to endothe-
lial cells of cutaneous capillaries and then extends to epithelial
cells of the epidermis, where replication results in formation of
the characteristic vesicles. Viremia and new vesicle formation CLINICAL MANIFESTATIONS
continue for 3 to 5 days and then terminate when humoral and
cellular immune responses appear. Natural History of Varicella
Varicella is generally a benign disease in healthy children,
Pathology of Varicella although symptoms are often more severe in adolescents and
adults. Fewer than 5% of primary VZV infections are subclin-
The cutaneous manifestations of varicella begin with hema- ical. Symptoms develop after an incubation period of about
togenous infection of the endothelial cells of cutaneous blood 15 days (range, 10 to 20 days). A prodrome of fever, malaise,
vessels. VZV then begins to replicate in the skin, leading to headache, and anorexia is variably present, occurring more
ballooning degeneration of epithelial cells in the prickle cell commonly among older children and adults and lasting 1 to
(malpighian) layer of the epidermis. Local collections of extra- 2 days. A transient scarlatina-like rash is occasionally noted
cellular edema fluid result in acantholysis, with elevation of the just before or coincident with the appearance of the varicella
stratum corneum to form a clear vesicle. Multinucleated giant lesions. The lesions first appear on the head, then the trunk, and
cells are found at the base of the lesion. Infected cells contain finally the extremities. The greatest concentration of lesions is
eosinophilic intranuclear inclusion bodies (Cowdry type A on the trunk and proximal extremities. The rash of varicella is
inclusions) surrounded by a clear zone. A perivascular infil- characterized by rapid evolution of lesions over 8 to 12 hours
tration of mononuclear cells is seen around cutaneous vessels. and by successive crops of new lesions. Consequently, lesions of
The vesicular fluid becomes cloudy as it accumulates inflam- all stages are present simultaneously on involved skin surfaces.
matory cells and desquamated epidermal cells. The vesicular Lesions begin as pink macules that quickly become papular
fluid is resorbed, resulting in drying and crusting of the lesion. and evolve into fragile vesicles 1 to 4 mm in diameter sur-
Healing occurs with regeneration of the epithelial cell layers. rounded by a zone of erythema. As inflammatory cells migrate
into the vesicular fluid, the lesions become pustules; these are
often centrally umbilicated. The pustules become crusted and
Pathogenesis of Herpes Zoster the crusts detach after 1 to 3 weeks, usually healing without
scarring. Vesicles also appear on mucosal surfaces and rapidly
As VZV replicates in the skin during acute varicella, some evolve into shallow ulcerations. New lesion formation contin-
virions are transported via sensory nerves to the correspond- ues for 2 to 4 days, accompanied by pruritus, fever (100 to
ing dorsal root ganglia (15). The virus establishes a latent 102F), headache, malaise, and anorexia. The rash peaks on
infection within the ganglion, preferentially infecting satellite about the fifth day, with an average lesion count of 250 to 500;
cells rather than neurons (16). An alternative explanation is fewer lesions are seen in children younger than 5 years (20).
that VZV may reach the ganglion by viremic spread. VZV Older children and secondary cases within a household tend to
may periodically reactivate and undergo limited replication, have higher lesion counts and higher fever.
but replication is suppressed by the immune response before The most common complication of varicella in otherwise
any clinical symptoms result (17,18). The specific immune normal children is bacterial superinfection (usually staphylo-
responses that limit reactivation of VZV from the sensory gan- coccal or streptococcal infection) that can occasionally prog-
glia are poorly understood. The most important factor that ress to serious necrotizing cellulitis (1). Varicella pneumonia is
predisposes to the development of herpes zoster appears to be rare in children but occurs more often in adults (21). Pregnant
decline or suppression of VZV-specific cellular immunity. This women with chickenpox may be at especially high risk for
may occur naturally with aging or be induced by immunosup- severe varicella pneumonia. Neonatal varicella can occur
pressive illness or therapy. Following reactivation and replica- when the mother develops chickenpox within a period of
tion in the ganglion, virus travels along the sensory nerve to 5 days before to 2 days after delivery. The infected neonate can
the skin, where it again replicates in epithelial cells, produc- develop severe disseminated varicella with a mortality of 20%
ing the characteristic dermatomal vesicular rash of shingles. to 30%. Maternal varicella occurring during the first trimester
Unlike the lesions of varicella, in which different stages are of pregnancy has been associated with congenital abnormali-
seen simultaneously, most zoster lesions are in the same stage ties, but the risk appears low (about 1% to 2%) (1).
of development. A limited viremia occurs during many epi- In immunocompromised children, varicella is a serious and
sodes of herpes zoster, reflected by the appearance of a few potentially fatal infection. Children at highest risk are those
extradermatomal vesicles. with acute leukemia, although children with other malignan-

Scheld_Ch10.indd 158 2/21/14 5:34 PM


Chapter 10: Neurologic Manifestations of Varicella and Herpes Zoster 159

cies and those on cytotoxic or immunosuppressive medications The pathogenesis of this syndrome is unknown, partly
(including high-dose corticosteroids) can also develop compli- because of the lack of necropsy studies in this nonfatal illness.
cations. Immunocompromised children may develop severe The two pathogenic mechanisms that have been proposed are
hemorrhagic or necrotic skin lesions (purpura fulminans or direct viral involvement of the cerebellum or a parainfectious,
hemorrhagic varicella) or severe bacterial superinfection. immunologically mediated demyelinating process analogous to
Persistent viremia can result in dissemination of VZV, produc- that seen with other viral infections. Recovery of VZV from
ing pneumonitis, hepatitis, or encephalitis. A chronic form of brain or CSF by culture has not been reported in varicella-
varicella has been reported in HIV-infected children. associated cerebellar ataxia. VZV antibodies have been detected
in the CSF of patients with CNS abnormalities in association
with varicella but were absent in children who had varicella
Neurologic Complications of Varicella but no neurologic symptoms, perhaps reflecting intrathecal
antibody production (25). Analysis of CSF using polymerase
The incidence of CNS complications with varicella is reported chain reaction (PCR) revealed VZV-specific DNA in three of
to be 1 to 3 per 10,000 cases (3,21). Because many uncompli- five children with varicella cerebellitis (26). These observations
cated cases of varicella do not come to medical attention, any suggest that VZV replication within the CNS does occur, but
calculation of the frequency of complications is likely to be an detailed studies are lacking and the evidence for viral invasion
overestimation. The CNS manifestations most frequently asso- of the cerebellum is circumstantial.
ciated with chickenpox are cerebellar ataxia and encephalitis
(22,23). Uncommon neurologic complications include trans- Encephalitis
verse myelitis, aseptic meningitis, strokes, and Guillain-Barr
A less common but more severe CNS complication of chicken-
syndrome (23). Optic neuritis has been reported as a rare
pox is encephalitis or cerebritis. The incidence of encephalitis
complication of varicella in both pediatric and adult patients,
is estimated to be 1 to 2 cases per 10,000 cases of chicken-
with good visual recovery expected in most cases (24). Reye
pox (3). Most cases of encephalitis occur in children, but the
syndrome, a triad of acute hepatic failure, encephalopathy,
incidence is highest in adults (older than 20 years) and infants
and hypoglycemia, was previously associated with varicella
(younger than 1 year). Neurologic symptoms may occur from
(and with other viral infections) but is now known to be more
2 weeks before to several weeks after the varicella rash (most
specifically related to salicylate therapy in febrile children.
often about 1 week after), and the onset may be abrupt or
However, many cases of Reye syndrome are included in older
gradual (22,27). Headache, fever, vomiting, and an altered
reviews of varicella encephalitis, resulting in misleading esti-
sensorium are the usual presenting symptoms (23,28). Seizures
mates of incidence and mortality.
occur in 29% to 52% of patients (23). Focal neurologic abnor-
malities can include ataxia, hypertonia or hypotonia, hyperre-
Cerebellar Ataxia flexia or hyporeflexia, positive plantar reflexes, hemiparesis,
Cerebellar ataxia, the most common neurologic abnormal- and sensory changes (23,28).
ity associated with varicella, is diagnosed in approximately The CSF from patients with varicella-associated cerebri-
1 per 4,000 cases of chickenpox (21). Children can develop tis is frequently abnormal, with elevated opening pressure,
ataxia from several days before to 2 weeks after the onset a mild to moderate lymphocytic pleocytosis (usually 100
of the rash, although neurologic symptoms most often cells/L), elevated protein (50 to 100 mg/dL), and normal
occur simultaneously with the rash (23). Symptoms include glucose (22). The EEG is often abnormal, showing slow-
vomiting, headache, and lethargy accompanied by ataxia. wave activity consistent with a diffuse encephalitis. Focal
Fever, nuchal rigidity, and nystagmus occur in about 25% EEG abnormalities suggestive of epileptiform activity may
of patients. Seizures are rare and suggest a more diffuse occur even without clinical seizures. In patients who do have
encephalitis. In cases of ataxia presenting before the devel- seizures, these EEG abnormalities tend to persist and are
opment of rash, the correct diagnosis may not be clinically present in 43% of follow-up studies at 1 year. Abnormalities
apparent unless an association is made with recent varicella observed by CT in patients with varicella encephalitis have
exposure. included cerebral or cerebellar edema and areas of low at-
The extent of the diagnostic evaluation in patients with tenuation consistent with demyelination. MRI abnormalities
varicella-associated cerebellar dysfunction should be gov- in children with postvaricella encephalitis have included dif-
erned by the severity of the illness and the degree of certainty fuse gray and white matter lesions or bilateral basal ganglia
of the diagnosis. In uncomplicated cases, the clinical presenta- lesions (29).
tion alone is sufficient to establish the diagnosis, and no fur- The reported mortality for varicella encephalitis has var-
ther evaluation is necessary. In more complicated situations, ied from 5% to 35%, although many of these series included
a cerebrospinal fluid (CSF) examination, an electroencephalo- cases of Reye syndrome (22,23,28). The actual mortality
gram (EEG), and magnetic resonance imaging (MRI) scan of rate for varicella cerebritis is probably less than 10%, with
the brain usually are warranted. The CSF is frequently nor- complete or nearly complete recovery expected in most cases.
mal, but a moderate lymphocytic pleocytosis (100 cells/L) Long-term sequelae may be present in 10% to 20% of sur-
with mildly elevated protein and normal glucose levels may vivors (23). In a series of 59 cases of varicella with CNS
occur in 20% to 30% of patients (23). The EEG demonstrates involvement, the mortality rate was 5% (22). Two of the three
diffuse slow-wave activity in approximately 20% of patients deaths were associated with pneumonia, and 80% of the sur-
and normalizes as the symptoms resolve (23). Few data are vivors were discharged from the hospital without detectable
available regarding the utility of computed tomographic (CT) sequelae.
or MRI scan in diagnosing varicella-associated cerebellar The role of active viral replication in the CNS in varicella
ataxia. encephalitis remains uncertain (30). Postmortem studies of
Cerebellar ataxia associated with chickenpox is self- the brain have shown a wide range of histopathologic find-
limited, and most abnormalities completely resolve within 1 to ings (22,23,31). Diffuse cerebral edema is generally present.
3 weeks. Mortality is essentially zero, and deaths that occur Perivascular infiltration of mononuclear cells and demyelin-
are usually attributed to the development of nonneurologic ation have been seen in some cases, the latter suggesting a
complications such as pneumonia (22,23). postinfectious demyelinating process. Other cases have shown

Scheld_Ch10.indd 159 2/21/14 5:34 PM


160 Part II: Viral Infections and Related Disorders

focal hemorrhagic lesions. Intranuclear viral inclusions have MRI typically demonstrates unilateral infarctions in the MCA
been observed only rarely in the brain following varicella, usu- distribution (38,39). In one fatal case, histopathology revealed
ally in immunocompromised patients (31). active granulomatous arteritis of the MCA with lymphocytic
inflammatory infiltrate and VZV antigens in the smooth muscle
Transverse Myelitis layer (37). Patients have been treated with intravenous acyclo-
vir and corticosteroids, but no data from controlled studies are
On rare occasions, varicella has been associated with an isolated
available to assess the efficacy of these treatments. Most chil-
weakness of the lower extremities, sphincter dysfunction, abnor-
dren with hemiplegia following varicella infection have good
mal deep tendon reflexes, and extensor-plantar reflexes (32). The
neurologic recovery (which is a much better prognosis than
CSF is characterized by a lymphocytic pleocytosis and elevated
that associated with strokes in adults following HZO).
protein level with a normal glucose level. The completeness of
recovery is variable. The pathogenesis of varicella myelitis is
not known, although cases have been published in which VZV Natural History of Herpes Zoster
DNA was detected in CSF by PCR, suggesting that active viral
invasion of the spinal cord may be involved (33). The inflammatory changes that occur in the sensory gan-
glion and nerve as VZV reactivates are manifested by pain
Aseptic Meningitis in the corresponding dermatome. The patient may report
Aseptic meningitis has been reported as a complication of vari- sensations ranging from mild itching or tingling to severe
cella (23). Meningismus without evidence of cerebral or cere- pain that precedes the appearance of the skin lesions by 1 to
bellar dysfunction is suggestive of the diagnosis, and complete 5 days (or occasionally longer). Constitutional symptoms are
recovery is expected. CSF findings are typical of viral meningitis, reported by fewer than 5% of patients during the prodromal
with mild lymphocytic pleocytosis, slight elevation in the protein phase. The cutaneous eruption of herpes zoster is unilateral
content, and normal glucose level. VZV has not been cultured and does not cross the midline. The rash appears in the skin
from the CSF in this setting; PCR would be an appropriate diag- segment innervated by a single sensory ganglion (Fig. 10.1).
nostic test. It is probably appropriate to consider CNS involve- Overlap of lesions into adjacent dermatomes occurs in 20%
ment with varicella as a spectrum, with aseptic meningitis the of patients. Scattered cutaneous vesicles beyond the primary
most benign manifestation and encephalitis the most serious. or adjacent dermatomes appear in about one third of patients
with herpes zoster infection and have no prognostic signifi-
Stroke Syndromes cance. Disseminated VZV infection with visceral involvement
is extremely rare in immunocompetent patients with herpes
Arterial ischemic strokes are a well-recognized complication zoster infection, although occasional cases of encephalitis
of herpes zoster ophthalmicus (HZO) but can also occur after and myelitis have been reported. Herpes zoster appears with
varicella (34,35). By some estimations, young children with ar- increased frequency in certain dermatomes, and this is thought
terial ischemic strokes are threefold more likely than controls to reflect the distribution of skin lesions during varicella (18).
to have recently had varicella. In a prospective cohort study, 22 The most common sites for herpes zoster are the thoracic der-
(31%) of 70 children with arterial ischemic strokes had vari- matomes (50% of cases), followed by cervical (15%), lumbar
cella within the preceding 12 months, compared with 9% in (15%), and sacral (5%) dermatomes (18). About 15% of zos-
the healthy population (36). Children with strokes and recent ter cases occur in cranial nerve dermatomes, with most cases
varicella infection had higher rates of basal ganglia infarction, involving the first division of the trigeminal nerve (HZO).
abnormal cerebral vascular imaging, and recurrent ischemic Ocular involvement can be a serious complication of HZO.
attacks (p .05 for all) (36). The syndrome typically occurs Conjunctivitis is common and benign, but more serious ocu-
in otherwise healthy, immunocompetent children (median age, lar manifestations, such as keratitis, scleritis, and iridocyclitis,
5 years). The median interval between varicella infection and may develop in 50% to 70% of patients with HZO (40).
the onset of neurologic deficits is 2 months (37). The children Skin changes begin with an erythematous maculopapular
usually present with hemiplegia, and angiography reveals vas- rash, followed by the appearance of clear vesicles. New vesicle
culopathy of the branches of the middle cerebral artery (MCA). formation typically continues for 3 to 5 days, followed by lesion

A B

FIGURE 10.1 Anterior (A, left) and posterior (B, right) views of a patient with herpes zoster in the right
T3 dermatome. The vesicular eruption terminates at the midline.

Scheld_Ch10.indd 160 2/21/14 5:34 PM


Chapter 10: Neurologic Manifestations of Varicella and Herpes Zoster 161

pustulation and scabbing. The extent of involvement can range cation of herpes zoster is chronic pain, or PHN, which occurs
from a few vesicles to a confluent eruption filling the entire with sufficient frequency that it can be considered a part of the
dermatome. Most lesions are crusted by day 10, but complete natural history of the disease (48).
skin healing may require 2 to 4 weeks. Unlike varicella, herpes
zoster is associated with a significant risk of skin scarring and Encephalitis
permanent pigmentation changes. Skin necrosis and gangrene
Herpes zoster can be complicated by acute encephalitis that usu-
in the involved dermatome can occur but are more commonly
ally occurs a few days after the onset of rash but has been re-
encountered in immunocompromised patients.
ported from days to weeks before or after the skin eruption (49).
During the acute phase of herpes zoster, most patients expe-
Encephalitis has occasionally been documented in the absence of
rience dermatomal pruritus and pain, which can be severe. The
apparent cutaneous zoster and in patients who received appro-
acute neuritis is variably described as an aching, burning, or
priate antiviral therapy during the acute episode of herpes zoster
stabbing pain. Many patients complain of headache, photopho-
(50,51). Immunocompromised patients are clearly at increased
bia, and malaise, but significant fever is rare. CSF examination
risk for the development of encephalitis (49,52). Other markers
(which is not routinely necessary) reveals a lymphocytic pleocy-
of increased risk for CNS involvement include zoster in a cranial
tosis and increased protein concentration; VZV can occasion-
nerve dermatome or the presence of cutaneous dissemination.
ally be cultured from the CSF. Although the skin usually heals
The mortality of herpes zosterassociated encephalitis has been
in 2 to 4 weeks, pain (postherpetic neuralgia [PHN]) persists for
reported to be from 0% to 25%, and the true incidence is prob-
longer than 1 month in 20% to 70% of patients with herpes
ably about 10% (49). The clinical presentation is most often an
zoster, with elderly patients experiencing the highest frequency
acute or subacute delirium accompanied by few focal neurologic
of chronic pain. Although herpes zoster is most common
signs (6,49,53). Other findings can include headache, meningis-
among older adults, the disease can occur in patients of any age.
mus, fever, ataxia, and seizures.
Immunocompetent children and young adults with herpes zos-
CSF examination reveals increased opening pressure,
ter tend to have less extensive cutaneous eruptions, less severe
increased protein, and a lymphocytic pleocytosis. Caution must
pain, and a much lower risk for chronic pain.
be used in interpretation of the spinal fluid findings, because
Patients with deficiencies in cell-mediated immunity have
pleocytosis is also present in about half of patients with
an increased incidence of herpes zoster and a higher risk for
uncomplicated herpes zoster (54). Positive VZV cultures have
complications. In immunocompromised patients, zoster causes
been obtained from CSF in patients with zoster encephalitis,
more severe skin involvement within the dermatome and may
and cells containing VZV-specific antigens have been identified
be accompanied by viremia with cutaneous or visceral dissemi-
in CSF. Serologic assays have revealed increased levels of VZV-
nation. Without antiviral chemotherapy, cessation of new vesi-
specific intrathecal antibodies (25). EEGs show diffuse abnor-
cle formation does not occur until about day 8, pustulation on
malities with an excess of slow-wave activity (53). Images
day 9, and scabbing on day 18 (41). Patients with herpes zoster
obtained by CT are generally nondiagnostic; published experi-
infection at highest risk for dissemination are those with lym-
ence with MRI in this syndrome is limited. PCR is the most
phoproliferative malignancies or those who recently received
sensitive diagnostic tool for zoster encephalitis. In one study
induction chemotherapy (8,42). Without antiviral treatment,
of seven patients with herpes zoster with neurologic symp-
the reported incidence of cutaneous dissemination in immuno-
toms (meningitis or encephalitis), PCR performed on CSF was
compromised populations is 6% to 26% (43). In most patients,
positive for VZV DNA in all cases (26). The frequency of PCR
dissemination is limited to the skin and does not substantially
positivity in CSF from patients with uncomplicated cutaneous
alter the prognosis. However, 10% to 50% of patients who
herpes zoster remains to be established.
develop cutaneous dissemination also have evidence of visceral
As with virtually all neurologic syndromes caused by VZV,
dissemination (such as pneumonitis, meningoencephalitis, or
there is controversy regarding the relative contributions of
hepatitis), which carries a much more serious prognosis. Even
direct viral replication and postinfectious immunopathologic
with the availability of antiviral chemotherapy, the mortality
changes in the pathogenesis of herpes zosterassociated
rate for zoster with visceral dissemination is 5% to 15%, with
encephalitis. Histologic studies have shown demyelination
most deaths attributable to pneumonitis (44). Visceral VZV
with mononuclear cell infiltrates, especially in white matter
infection occurring without any evidence of skin lesions is
(19,55). In brain tissue from patients who died with herpes
uncommon. However, several patients with acquired immu-
zosterassociated encephalitis, intranuclear inclusion bodies
nodeficiency syndrome (AIDS) and VZV myelitis or encepha-
were often evident, and viral particles have been visualized
litis but without clinically apparent zoster have been reported.
by electron microscopy (49). Supporting the hypothesis that
Another syndrome unique to the immunocompromised host is
active viral invasion and replication play a role, there are
chronic cutaneous VZV infection, sometimes associated with
many anecdotal reports of rapid clinical improvement after
acyclovir-resistant virus.
initiation of acyclovir therapy (52).
Chronic VZV encephalitis is seen almost exclusively in
immunocompromised patients, especially patients with AIDS
Neurologic Complications of Herpes Zoster with marked depletion of CD4 lymphocytes (56). The
onset of the encephalitis may occur months after an episode
Neurologic complications of herpes zoster can occur during of herpes zoster and 30% to 40% of these patients have no
the acute eruption (e.g., segmental motor paresis) or may ap- recognized history of cutaneous VZV infection, making the
pear weeks to months after the herpes zoster rash has resolved diagnosis more difficult. Pathologic studies reveal multifocal
(e.g., delayed contralateral hemiparesis) (45). Neurologic com- leukoencephalopathy, with lesions in the white matter near the
plications appear more often in immunocompromised patients, graywhite junction, small vessel vasculitis, and demyelination
including patients with HIV infection. Several pathologically (46). The clinical presentation is usually subacute with head-
distinct syndromes have been defined, including large and ache, fever, mental status changes, and seizures. Patients may
small vessel vasculopathies and ventriculitis (46). Investigators have focal neurologic defects including aphasia, hemiplegia,
have also described myelitis and polyradiculitis, as well as a and visual-field cuts (5759). MRI demonstrates plaquelike
variety of cranial and peripheral nerve palsies in association lesions in deep white matter, changes consistent with demy-
with herpes zoster (47). The most common neurologic compli- elination, and late development of ischemic or hemorrhagic

Scheld_Ch10.indd 161 2/21/14 5:34 PM


162 Part II: Viral Infections and Related Disorders

infarcts of cortical and subcortical gray and white matter retention), often accompanied by weakness of the lower ex-
(60,61). CSF examination reveals mild mononuclear pleocy- tremities, asymmetric reflexes, and sensory disturbances (80).
tosis. VZV DNA has been amplified from the CSF of patients Prognosis for recovery of neurologic function has been vari-
with chronic encephalitis using PCR (62,63). Patients often able. In severe cases, the myelopathy can progress to a partial
follow a clinical course of progressive deterioration and death, Brown-Squard syndrome or total cord transection. MRI has
although anecdotal reports have suggested some benefit from been useful in diagnosing myelitis, with abnormal signal evi-
high-dose intravenous acyclovir therapy (46,64). Other VZV- dent in the cord at the level of inflammation (33,81,8486).
induced neurologic disorders described in patients with AIDS Pathologic studies have demonstrated necrosis of the spinal
include multifocal leukoencephalitis (57), ventriculitis (65), cord, with posterior column involvement, focal demyelination,
myelitis and myeloradiculitis (65,66), and focal brainstem and perivascular inflammation. The presence of intranuclear
lesions (67). inclusion bodies and positive immunofluorescence stains
for VZV antigens in spinal cord tissue are evidence for the
Herpes Zoster Ophthalmicus with Delayed direct role of the virus in this disease (80). VZV DNA has
Contralateral Hemiparesis been amplified from the spinal fluid of patients with myeli-
tis by PCR techniques (33,81). Although there are anecdotal
Hemiparesis is a rare but serious complication of herpes zos- reports of significant neurologic improvement with antiviral
ter that has been reported in immunocompetent and immu- treatment, this therapeutic approach has not been studied in a
nocompromised patients (68), including both children and prospective fashion (33,87).
adults (69). The mean interval from rash to onset of neuro-
logic symptoms in adults is about 7 weeks, although intervals
Cranial and Peripheral Nerve Palsies
of up to 6 months have been reported. The typical presenta-
tion is headache and hemiplegia occurring in a patient with Segmental motor paresis complicates about 2% to 3% of cases
a history of recent HZO (70). The mortality rate is 20% to of dermatomal herpes zoster. Weakness, which may be abrupt
25%. There is a high probability of permanent neurologic se- in onset, usually begins within a few days to 2 weeks after the
quelae among survivors (71). onset of the rash. The involved muscle groups are those whose
CSF examination reveals mononuclear cell pleocytosis (71). innervation corresponds with that of the affected dermatome.
Imaging studies (CT or MRI) show changes consistent with Thus, thoracic zoster may be associated with paralysis of limb or
brain infarction (72,73). Arteriography is usually diagnostic trunk muscles, while sacral zoster may result in bladder or anal
and demonstrates inflammation, narrowing, and thrombosis dysfunction. The peripheral motor neuropathy is caused by viral
of the proximal branches of the anterior cerebral artery or invasion and inflammatory changes affecting motor neurons in
the MCA (74). Rare cases of posterior circulation strokes fol- the anterior horn. Weakness is usually transient, and 75% to
lowing herpes zoster infection involving a cervical dermatome 85% of patients can expect total resolution, although a few pa-
have been reported (75). The pathogenesis of this unusual dis- tients experience prolonged paralysis with muscle atrophy.
order is thought to be direct VZV invasion of cerebral arteries Twelve percent to thirty percent of patients with herpes
by extension along intracranial branches of the trigeminal zoster involving a cephalic dermatome develop oculomotor
nerve. This results in inflammation of the internal carotid or facial palsies. HZO can be associated with partial or com-
artery or one of its branches on the side ipsilateral to the rash. plete palsies of the third, fourth, or sixth nerve. Facial palsy
The local inflammatory response and thrombosis produce vas- (sometimes including loss of taste on the anterior two thirds
cular occlusion or distal embolization, resulting in infarction of the tongue) may accompany otic zoster (Ramsay Hunt
and contralateral hemiparesis. Pathologic studies have demon- syndrome) or zoster involving the second or third cervical
strated a necrotizing arteritis of large and small cerebral vessels dermatomes (88,89).
with thrombosis in the proximal anterior cerebral artery or
the MCA (76). Although VZV has not been cultured from the Acute Retinal Necrosis
areas of inflammation, VZV-specific antigens and DNA have
been demonstrated in the smooth muscle cells of the media of VZV-associated acute retinal necrosis (ARN) has been described
affected vessels (77). in both immunocompetent and immunocompromised persons.
Both acyclovir (78) and corticosteroids (79) have been used Since the advent of the AIDS epidemic, a more aggressive variant
in an effort to treat this syndrome, although no therapy has of this disease (sometimes termed rapidly progressive herpetic
been evaluated in a controlled or prospective fashion. Antiviral retinal necrosis [RPHRN]) has been identified (90,91). Visual
therapy is warranted because of the demonstrated presence of changes are usually noted weeks to months after the antecedent
VZV in the inflamed vessels, but benefit of therapy is difficult herpes zoster. ARN can follow either HZO or herpes zoster in
to assess, because irreversible cerebral infarction has usually a remote dermatome. Furthermore, retinal involvement is bilat-
occurred by the time the diagnosis is made. eral in more than half of cases, suggesting that VZV reaches the
CNS via hematogenous spread, possibly with extension along
nerve pathways within the anterior visual system (91). VZV ret-
Myelitis initis presents with multifocal necrotizing lesions, often initially
Herpes zosterrelated myelitis is thought to result from direct involving the peripheral retina. The granular, nonhemorrhagic
invasion of the spinal cord by VZV, with unilateral motor and lesions rapidly extend and coalesce, accompanied by relatively
posterior column dysfunction evolving into paraplegia (33). little intraocular inflammation (92).
The disorder most often follows thoracic herpes zoster, with In patients with AIDS, VZV retinitis rapidly progresses to
weakness developing in the same spinal cord segment as the full-thickness retinal necrosis, usually with retinal detachment,
rash. Neurologic symptoms begin to develop an average resulting in blindness in 75% to 85% of involved eyes (91).
of 12 days after the onset of the rash (80). However, VZV Because the involved eye is rarely salvageable in HIV-infected
myelitis in patients with no history of antecedent zoster has patients with RPHRN, the goal is to try to prevent disease pro-
also been reported (81,82). Immunocompromised patients are gression in the uninvolved eye. Intravenous acyclovir alone is
at increased risk for postzoster myelitis, and the syndrome is ineffective (92). Some experts recommend intravenous therapy
well described in patients with AIDS (58,83). The most com- with ganciclovir or foscarnet (or a combination of the two) (93).
mon initial manifestation is bladder dysfunction (e.g., urinary Anecdotal success with cidofovir or with intravitreal injections

Scheld_Ch10.indd 162 2/21/14 5:34 PM


Chapter 10: Neurologic Manifestations of Varicella and Herpes Zoster 163

of ganciclovir has also been reported (94). Results of antiviral Topical treatments should be used only on intact, healed skin.
therapy for VZV retinitis in HIV-infected patients, regardless of In a controlled clinical trial of 277 patients with intractable
regimen, have been disappointing. PHN, intrathecal injection of 60 mg of methylprednisolone
ARN in immunocompetent patients is a less virulent disease acetate once weekly for 4 weeks resulted in significant pain re-
and responds better to antiviral therapy. In this setting, acyclo- duction (109); these promising initial results await validation.
vir is clearly beneficial for preserving useful vision (95). A sug-
gested antiviral regimen for ARN in the otherwise healthy host Zoster Sine Herpete
is intravenous acyclovir 10 to 15 mg/kg every 8 hours for 10 to
Clinicians occasionally encounter patients who present with
14 days, followed by oral valacyclovir 1 g three times daily for
zoster-like neuropathic pain but never develop the character-
4 to 6 weeks, although this treatment approach has not been
istic dermatomal rash (110,111). Recent detailed studies of
studied in a controlled fashion.
a few patients presenting with dermatomal pain have estab-
lished that some of these cases are due to VZV reactivation.
Postherpetic Neuralgia Patients with this syndrome, termed zoster sine herpete, have
PHN is the term traditionally used to describe the chronic rising titers of VZV-specific antibody in both serum and CSF
dermatomal pain that persists after the cutaneous eruption of and have VZV DNA in CSF and peripheral blood mononu-
herpes zoster has healed. PHN has been estimated to occur fol- clear cells detectable by PCR (112,113). Because there is no
lowing 10% to 40% of zoster cases; some of the variability in easy way to make the diagnosis, the incidence of zoster sine
incidence calculations can be attributed to differing definitions herpete is not known. In anecdotal reports, responses to anti-
of PHN (6,18,96). Ragozzino et al. (6) reported that 9.3% viral therapy have been inconsistent.
of the patients with herpes zoster in their study had PHN;
of these patients, 22% (or 2% of the total study population)
had pain that persisted for longer than 12 months. The inci- DIAGNOSIS
dence (and possibly the duration) of PHN correlates directly
with patient age. Pain that persists for longer than 1 year The appearance of varicella is quite distinctive, and in most
has been noted in 4%, 22%, and 47% of patients younger cases, a clinical diagnosis is accurate and reliable. The presenta-
than 20 years, older than 55 years, and older than 70 years tion of a child with mild constitutional symptoms, a diffuse ve-
of age, respectively (6,97,98). Within the affected dermatome, sicular rash, and no prior history of chickenpox (or vaccination)
patients with PHN experience a variety of sensory abnormali- is strongly suggestive of the diagnosis, especially during an epi-
ties (paresthesias, dysesthesias, allodynia) and neuralgic pain demic. Other infections that can occasionally mimic chickenpox
of varying quality and severity. The pathogenesis of PHN include vesicular exanthems caused by coxsackievirus, dissemi-
is not completely understood but apparently involves both nated herpes simplex virus (HSV) infection, diffuse impetigo,
peripheral and central mechanisms. Experimental evidence or rickettsialpox. Noninfectious cutaneous diseases that may
suggests that the damaged sensory nerve develops a lowered resemble varicella include contact dermatitis and dermatitis
activation threshold and superphysiologic responses to distal herpetiformis. Herpes zoster, with its characteristic dermatomal
stimuli. This excessive input from the peripheral sensory nerve vesicular rash, is also readily diagnosed on the basis of clini-
results in hyperexcitability of the dorsal horn, producing exag- cal appearance. The diagnosis may be less apparent in patients
gerated central responses that are perceived as pain (99). who present with dermatomal neuralgia before the emergence
Successful treatment of PHN often requires a multifaceted of skin lesions. The skin disease that is most commonly con-
approach (48,100,101). Opioid analgesics are the mainstay fused with herpes zoster is zosteriform HSV infection (usually
of therapy during the early phases of neuralgic pain. A clini- in the sacral area), which can closely mimic the appearance of
cal trial with oxycodone for patients with PHN demonstrated shingles. Contact dermatitis can also occasionally resemble her-
a significant level of pain reduction (67% of those receiv- pes zoster, but it tends to cross dermatomal boundaries.
ing oxycodone vs. 11% receiving placebo), as measured by There is no asymptomatic shedding of VZV, as there is with
a visual analog scale (102). Long-acting opioid preparations HSV and cytomegalovirus. Therefore, identification of VZV
(oral or transdermal) are preferable to short-acting analge- virions, antigens, or nucleic acids from cutaneous lesions or
sics for management of chronic PHN. Several randomized, nonneuronal tissues is diagnostic of active infection. VZV can
controlled clinical trials have shown tricyclic antidepressants be cultured by inoculation of vesicular fluid into monolay-
(including amitriptyline, nortriptyline, and desipramine) to ers of human fetal diploid kidney or lung cells (114). VZV is
be effective in reducing the pain of PHN, either as a single very labile, and every effort should be made to minimize time
agent or in combination with other drugs (103,104). Because spent in transport and storage. Ideally, fluid should be aspi-
tricyclic antidepressants are often associated with sedation rated from a clear vesicle using a tuberculin syringe and inocu-
and anticholinergic side effects, treatment should begin with a lated directly into tissue culture at the bedside. Characteristic
relatively low dose at bedtime, with a gradual increase in dos- cytopathic effects are usually seen in tissue culture within 3
age as required and tolerated. Clinical trials have shown the to 7 days. The culture process can be accelerated by use of
anticonvulsant gabapentin to significantly reduce established centrifugation in shell vials. Identification of the isolate can be
PHN when used alone or in combination with other modali- confirmed by direct immunofluorescent staining using VZV-
ties (105). For treatment of PHN, physicians should initiate specific monoclonal antibodies. In general, viral culture for
gabapentin at a dose of 300 mg three times daily and escalate VZV is highly specific but slow, insensitive, and expensive.
as required, watching for adverse effects such as somnolence, VZV infection can be demonstrated in infected tissue by his-
dizziness, and ataxia. The adverse effects of these medica- topathology or electron microscopy. However, visualization of
tions can be additive (such as sedation due to opioids, tricyclic multinucleated giant cells with inclusion bodies or herpesvirus
antidepressants, and gabapentin), especially in elderly patients. virions does not distinguish between VZV and HSV. Direct
Topical application of capsaicin provides relief of PHN for immunofluorescence staining using fluorescein-conjugated
some patients, but others find the local stinging and burning monoclonal antibodies to detect VZV glycoproteins in infected
associated with application of capsaicin cream to be intolera- epithelial cells is highly sensitive and specific (115). Specimens
ble (106). Transdermal administration of lidocaine via patches can also be stained with HSV-specific monoclonal antibodies
has been shown to reduce PHN in controlled trials (107,108). to distinguish VZV from HSV infection. This direct fluores-

Scheld_Ch10.indd 163 2/21/14 5:34 PM


164 Part II: Viral Infections and Related Disorders

cent antigen-detection method is rapid, simple, and more sen- morbidity, and the availability of some specific therapy would
sitive than virus isolation (especially in late stages of infection, be useful. As discussed earlier in this chapter, there is consid-
when virus isolation becomes more difficult) and is especially erable controversy regarding what role, if any, active VZV
helpful for making a rapid diagnosis when the clinical presen- replication plays in the pathogenesis of varicella encephalitis.
tation is atypical. However, because no other mode of therapy is available, and
The use of PCR to detect VZV nucleic acids in clinical speci- because acyclovir is extremely safe and well tolerated, therapy
mens holds great promise, but it is used primarily as a research with acyclovir in patients with varicella encephalitis is war-
tool (116,117). PCR overcomes the difficulties inherent in cul- ranted. Corticosteroids have been proposed for use in varicella
turing labile VZV and has been useful in detecting VZV DNA in encephalitis, but there are no reliable data to support their use.
CSF from patients with CNS infections (26,54,118120). In ad- In immunocompromised children, varicella is a serious
dition, the use of PCR makes it possible to distinguish between and potentially lethal infection that requires antiviral ther-
disease caused by wild type VZV or by the VZV vaccine virus, apy (126). Intravenous acyclovir was compared with placebo
which is not routinely possible by culture (121). in a small population of immunocompromised children with
Serologic techniques are used to determine susceptibility chickenpox (127). In this trial, the administration of acyclo-
of an individual to VZV infection and to document rising vir reduced the frequency of pneumonitis from 45% to 0%.
antibody titers in patients with acute varicella or herpes zoster. Despite the lack of data from large-scale controlled trials,
A serologic response is usually considered diagnostic if there is the safety and efficacy of intravenous acyclovir have led to
a fourfold rise in antibody titer. Serum antibodies appear sev- its acceptance as the drug of choice for varicella infection in
eral days after the onset of varicella and peak at 2 to 3 weeks, immunocompromised patients. For treatment of serious VZV
meaning that serologic determinations usually deliver a retro- infections, intravenous acyclovir is given at a dose of 10 mg/kg
spective rather than a real-time diagnosis. Acute infection can (or 500 mg/m2) every 8 hours.
be established by demonstration of VZV-specific serum immu-
noglobulin M titers. However, antigen-detection techniques
are usually faster and more reliable. Patients with herpes zos-
ter are VZV seropositive at the time of disease onset, but most Herpes Zoster
show a significant rise in titer during the convalescent phase.
Appropriate supportive care can help make patients with her-
Elevated antibody titers in CSF can support the diagnosis of
pes zoster more comfortable. The skin lesions should be kept
VZV encephalitis (25,54,122). Most laboratories have now
clean and dry to reduce the risk of bacterial superinfection.
adopted an enzyme-linked immunosorbent assay (ELISA) or
Astringent soaks (e.g., Domeboro solution) may be soothing.
a latex agglutination (LA) assay for VZV serodiagnosis (114).
Most patients with acute herpes zoster infection have signifi-
cant pain and require therapy with opioid analgesics.
Three oral antiviral drugs are approved in the United States
TREATMENT for treatment of herpes zoster infection in the normal host. Oral
acyclovir, initiated within 72 hours of the onset of lesions at a
Varicella dose of 800 mg five times daily, reduces the duration of viral
shedding, accelerates the cessation of new lesion formation,
For most immunocompetent children, chickenpox is a disease and accelerates the events of cutaneous healing (128130).
associated with very low morbidity and mortality and support- These clinical trials showed variable benefit from acyclovir
ive care alone is sufficient. Astringent soaks and nonaspirin anti- for reducing the duration of PHN. However, data from these
pyretics improve comfort. Trimming the fingernails closely helps studies have been reexamined in a metaanalysis, which con-
prevent bacterial superinfections caused by scratching. If bacte- clusively demonstrated that acyclovir was superior to placebo
rial cellulitis develops, antibiotics may be required. Oral acyclovir for reducing the duration of zoster-associated pain, defined
has been evaluated for treatment of uncomplicated varicella in as pain measured from the initial onset until final resolution
immunocompetent children (20,123). Acyclovir therapy, initiated (131). Oral administration of valacyclovir (a prodrug of acy-
within 24 hours of the onset of rash, resulted in shorter dura- clovir) as a dose of 1 g three times daily produces plasma acy-
tion of fever, fewer skin lesions, and accelerated lesion healing. clovir levels that are approximately threefold to fivefold higher
Overall, oral acyclovir reduced the duration of symptomatic ill- than those achievable with acyclovir. Valacyclovir and acyclo-
ness by about 24 hours. The populations studied in these con- vir have been compared in a controlled trial for treatment of
trolled trials were not sufficiently large to assess the impact of herpes zoster infection in the normal host (132). The two drugs
acyclovir therapy on the incidence of varicella complications. were equivalent in terms of accelerating events of cutaneous
Oral acyclovir has also been evaluated in immunocompetent healing; however, valacyclovir was superior in accelerating the
adolescents and adults (124,125). As seen in the pediatric studies, resolution of zoster-associated pain. When evaluated for treat-
initiation of acyclovir therapy within 24 hours of onset of rash ment of herpes zoster in the normal host, famciclovir (500 mg
resulted in reduction in time to cessation of new lesion formation, orally three times daily) was significantly superior to placebo in
reduced the number of lesions, and reduced constitutional symp- reducing the duration of viral shedding, limiting the duration
toms, including fever. The dose of oral acyclovir for chickenpox of new lesion formation, and accelerating the events of cutane-
is 20 mg/kg (up to a maximum of 800 mg) five times daily for 5 ous healing (133). Most importantly, famciclovir was signifi-
to 7 days. Famciclovir and valacyclovir are likely to be at least cantly superior to placebo in reducing the duration of PHN.
as effective as acyclovir for chickenpox but have not been exten- A study comparing valacyclovir and famciclovir for herpes zos-
sively studied. Oral antiviral therapy is optional in healthy chil- ter showed these drugs to be therapeutically equivalent (134).
dren with varicella, but it should definitely be used in adolescents All these compounds are safe and well tolerated for short-term
and adults because of their increased risk for more severe illness. administration. Patients who are most likely to benefit from an-
Few data exist to help address the question of antiviral ther- tiviral therapy of herpes zoster are those who present for medi-
apy for neurologic complications of varicella. In general, the cal attention within 72 hours after onset of lesions, patients
cerebellar ataxia syndrome is benign and self-limited, and there with severe pain or large lesion surface area involvement at
is no evidence that antiviral therapy alters the natural history. the time of presentation, and elderly patients who are at high
Varicella encephalitis is associated with a substantial degree of risk for long-term complications, especially chronic pain (135).

Scheld_Ch10.indd 164 2/21/14 5:34 PM


Chapter 10: Neurologic Manifestations of Varicella and Herpes Zoster 165

HZO is a special situation in which antiviral therapy is clearly varicelliform rash (median lesion count of two to five), occurring
beneficial and significantly reduces the risk for ocular compli- in about 7% of recipients (141). The VZV vaccine is also safe
cations (136). Consultation with an experienced ophthalmolo- and effective in seronegative adults, although the seroconversion
gist is also recommended for patients with HZO and ocular rate after a single dose is lower (79% to 82%) than that seen
involvement. Because of their convenience of administration, in pediatric populations (143). The varicella vaccine has been
famciclovir and valacyclovir are the preferred drugs for uncom- endorsed for routine use in children by the American Academy
plicated herpes zoster infection in the normal host. of Pediatrics (144).
Two large clinical studies have helped clarify the role of Widespread use of the vaccine should result in a reduction
corticosteroids in herpes zoster infection (137,138). Both in varicella and varicella-related neurologic complications.
studies demonstrated benefit from corticosteroids in reducing Herpes zoster does occasionally develop in patients who have
the duration of acute neuritis, but neither study showed any received the varicella vaccine, and in some instances, it has been
reduction in the incidence or duration of PHN among steroid documented to be caused by the VZVOKA vaccine virus (145).
recipients. Therefore, although corticosteroids (in combination Among immunocompromised children, however, the risk of
with antiviral therapy) can provide some symptomatic benefit herpes zoster is lower in children who have received the vaccine
during the early phases of herpes zoster, corticosteroids will than in those who have experienced wild type varicella.
not have an impact on the development of PHN. For immunocompetent children aged 12 months to 12 years,
Immunocompromised patients who develop herpes zos- the vaccine is administered as a single subcutaneous dose (0.5 mL
ter are at significant risk for morbidity and mortality related containing no fewer than 1,500 plaque-forming units of VZV).
to disseminated infection. Controlled trials have shown that Adolescents and adults should receive two doses of vaccine 4 to
intravenous acyclovir substantially reduces the risk for cutane- 8 weeks apart. VZV vaccine is not approved in the United States
ous and visceral dissemination (44,139). In patients who are for use in immunocompromised patients, but the vaccine has been
less severely immunocompromised, it may be feasible to use an shown to be effective in these populations, and further studies are
oral antiviral drug for the treatment of zoster (140). ongoing to clarify these indications (146). An inactivated virus
The role of antiviral drugs in the management of neuro- vaccine is also being evaluated for use in immunocompromised
logic complications of herpes zoster has not been evaluated in patients (147). Routine administration of a booster vaccine is not
a controlled fashion. For those diseases in which viral replica- recommended, but additional long-term surveillance studies are
tion likely plays an important role in pathogenesis (e.g., zoster in progress to better define the need for a booster.
myelitis), therapy with intravenous acyclovir is recommended; Another potential application of the VZV vaccine is to
this approach is supported by benefits noted in anecdotal stimulate waning cell-mediated immune responses in elderly
experience. For diseases such as delayed contralateral hemipa- individuals to prevent VZV reactivation and herpes zoster.
resis, in which the role of active viral replication is much less An enhanced VZV-specific cytotoxic lymphocyte response can
clear, the value of antiviral therapy is uncertain, but in general be demonstrated in elderly seropositive individuals receiving
the potential benefits of antiviral therapy probably outweigh the VZVOKA vaccine (148). Whether this enhanced immune
any potential risks. response will be clinically effective in the prevention or ame-
lioration of subsequent herpes zoster infection will be deter-
mined by a large placebo-controlled clinical trial.
PREVENTION
A live, attenuated varicella vaccine containing VZVOKA strain ACKNOWLEDGMENTS
was developed in the 1970s in Japan and approved for use in the
United States in 1995 (141). At the standard dose, the protection This project has been funded in whole or in part with Federal
provided by the vaccine against VZV disease following house- funds from the National Institute of Allergy and Infectious
hold exposure was approximately 70%. Any case of chickenpox Diseases, National Institutes of Health, Department of
occurring among vaccinated individuals was almost invariably Health and Human Services, under Contract (N01-AI-65306,
mild (median lesion count of 15 to 32) (142). The most com- NO1-AI-15113, NO1-AI-62554, NO1-AI-30025), the General
mon adverse event associated with the vaccine has been a mild Clinical Research Unit (RR-032), and the State of Alabama.

References
1. Gnann JW Jr. Varicella-zoster virus: atypical presentations and unusual 10. Glesby MJ, Moore RD, Chaisson RE. Clinical spectrum of herpes zoster
complications. J Infect Dis. 2002;186(suppl 1):S91S98. in adults infected with human immunodeficiency virus. Clin Infect Dis.
2. Weller TH. Varicella and herpes zoster: a perspective and overview. J Infect 1995;21:370375.
Dis. 1992;166(suppl 1):S1S6. 11. Buchbinder SP, Katz MH, Hessol NA, et al. Herpes zoster and human
3. Choo PW, Donahue JG, Manson JE, et al. The epidemiology of varicella immunodeficiency virus infection. J Infect Dis. 1992;166:11531156.
and its complications. J Infect Dis. 1995;172:706712. 12. Davison AJ. Varicella-zoster virus. The Fourteenth Fleming lecture. J Gen
4. Meyer PA, Seward JF, Jumaan AO, et al. Varicella mortality: trends before vac- Virol. 1991;72(pt 3):475486.
cine licensure in the United States, 19701994. J Infect Dis. 2000;182:383390. 13. Ostrove JM. Molecular biology of varicella zoster virus. Adv Virus Res.
5. Seward JF, Watson BM, Peterson CL, et al. Varicella disease after introduction of 1990;38:4598.
varicella vaccine in the United States, 19952000. JAMA. 2002;287:606611. 14. Grose C, Ye M, Padilla J. Pathogenesis of primary infection. In: Arvin
6. Ragozzino MW, Melton LJ III, Kurland LT, et al. Population-based study of AM, Gershon AA, eds. Varicella-Zoster Virus: Virology and Clinical
herpes zoster and its sequelae. Medicine (Baltimore). 1982;61:310316. Management. Cambridge: Cambridge University Press; 2000:105122.
7. Donahue JG, Choo PW, Manson JE, et al. The incidence of herpes zoster. 15. Gilden DH, Mahalingam R, Dueland AN, et al. Herpes zoster: pathogen-
Arch Intern Med. 1995;155:16051609. esis and latency. Prog Med Virol. 1992;39:1975.
8. Rusthoven JJ, Ahlgren P, Elhakim T, et al. Varicella-zoster infection in adult 16. Meier JL, Straus SE. Comparative biology of latent varicella-zoster virus
cancer patients: a population study. Arch Intern Med. 1988;148:15611566. and herpes simplex virus infections. J Infect Dis. 1992;166(suppl 1):
9. Gnann JW. Other herpesviruses: herpes simplex virus, varicella-zoster virus, S13S23.
human herpesviruses types 6, 7, and 8. In: Bowden RA, Ljungman P, Paya CV, 17. Croen KD, Straus SE. Varicella-Zoster virus latency. Annu Rev Microbiol.
eds. Transplant Infections. Philadelphia: LippincottRaven; 1998:265286. 1991;45:265282.

Scheld_Ch10.indd 165 2/21/14 5:34 PM


166 Part II: Viral Infections and Related Disorders

18. Hope-Simpson RE. The nature of herpes zoster; a long-term study and a 54. Haanpaa M, Dastidar P, Weinberg A, et al. CSF and MRI findings in
new hypothesis. Proc R Soc Med. 1965;58:920. patients with acute herpes zoster. Neurology. 1998;51:14051411.
19. Schmidbauer M, Budka H, Pilz P, et al. Presence, distribution and spread 55. Edgar JD, Crosbie JJ, Hawkins SA. Cerebral vasculitis associated with
of productive varicella zoster virus infection in nervous tissues. Brain. shingles. Ulster Med J. 1990;59:7781.
1992;115(pt 2):383398. 56. De La Blanchardiere A, Rozenberg F, Caumes E, et al. Neurological com-
20. Balfour HH Jr, Kelly JM, Suarez CS, et al. Acyclovir treatment of varicella plications of varicella-zoster virus infection in adults with human immuno-
in otherwise healthy children. J Pediatr. 1990;116:633639. deficiency virus infection. Scand J Infect Dis. 2000;32:263269.
21. Guess HA, Broughton DD, Melton LJ III, et al. Population-based studies of 57. Gray F, Mohr M, Rozenberg F, et al. Varicella-zoster virus encephalitis in
varicella complications. Pediatrics. 1986;78:723727. acquired immunodeficiency syndrome: report of four cases. Neuropathol
22. Applebaum E, Rachelson MH, Dolgopol VB. Varicella encephalitis. Am J Appl Neurobiol. 1992;18:502514.
Med. 1953;15:223230. 58. Gray F, Belec L, Lescs MC, et al. Varicella-zoster virus infection of the
23. Johnson R, Milbourn PE. Central nervous system manifestations of central nervous system in the acquired immune deficiency syndrome. Brain.
chickenpox. Can Med Assoc J. 1970;102:831834. 1994;117(pt 5):987999.
24. Lee CC, Venketasubramanian N, Lam MS. Optic neuritis: a rare complica- 59. Amlie-Lefond C, Kleinschmidt-DeMasters BK, Mahalingam R, et al. The vas-
tion of primary varicella infection. Clin Infect Dis. 1997;24:515516. culopathy of varicella-zoster virus encephalitis. Ann Neurol. 1995;37:784790.
25. Echevarria JM, Tellez A, Martinez-Martin P. Subclass distribution of the 60. Aygun N, Finelli DA, Rodgers MS, et al. Multifocal varicella-zoster virus
serum and intrathecal IgG antibody response in varicella-zoster virus infec- leukoencephalitis in a patient with AIDS: MR findings. AJNR Am J
tions. J Infect Dis. 1990;162:621626. Neuroradiol. 1998;19:18971899.
26. Puchhammer-Stockl E, Popow-Kraupp T, Heinz FX, et al. Detection of 61. Lentz D, Jordan JE, Pike GB, et al. MRI in varicella-zoster virus leuko-
varicella-zoster virus DNA by polymerase chain reaction in the cerebrospi- encephalitis in the immunocompromised host. J Comput Assist Tomogr.
nal fluid of patients suffering from neurological complications associated 1993;17:313316.
with chicken pox or herpes zoster. J Clin Microbiol. 1991;29:15131516. 62. Gilden DH, Kleinschmidt-DeMasters BK, LaGuardia JJ, et al. Neurologic
27. Liu GT, Urion DK. Pre-eruptive varicella encephalitis and cerebellar ataxia. complications of the reactivation of varicella-zoster virus. N Engl J Med.
Pediatr Neurol. 1992;8:6970. 2000;342:635645.
28. Shope TC. Chickenpox encephalitis and encephalopathy: evidence for dif- 63. Iten A, Chatelard P, Vuadens P, et al. Impact of cerebrospinal fluid PCR on
fering pathogenesis. Yale J Biol Med. 1982;55:321327. the management of HIV-infected patients with varicella-zoster virus infec-
29. Darling CF, Larsen MB, Byrd SE, et al. MR and CT imaging patterns in tion of the central nervous system. J Neurovirol. 1999;5:172180.
post-varicella encephalitis. Pediatr Radiol. 1995;25:241244. 64. Poscher ME. Successful treatment of varicella zoster virus meningoen-
30. Kennedy PG, Barrass JD, Graham DI, et al. Studies on the pathogenesis of cephalitis in patients with AIDS: report of four cases and review. AIDS.
neurological diseases associated with varicella zoster virus. Neuropathol 1994;8:11151117.
Appl Neurobiol. 1990;16:305316. 65. Chretien F, Gray F, Lescs MC, et al. Acute varicella-zoster virus ventriculi-
31. Carmack MA, Twiss J, Enzmann DR, et al. Multifocal leukoencephalitis tis and meningo-myelo-radiculitis in acquired immunodeficiency syndrome.
caused by varicella-zoster virus in a child with leukemia: successful treat- Acta Neuropathol (Berl). 1993;86:659665.
ment with acyclovir. Pediatr Infect Dis J. 1993;12:402406. 66. Snoeck R, Gerard M, Sadzot-Delvaux C, et al. Meningoradiculoneuritis
32. Rosenfeld J, Taylor CL, Atlas SW. Myelitis following chickenpox: a case due to acyclovir-resistant varicella zoster virus in an acquired immune defi-
report. Neurology. 1993;43:18341836. ciency syndrome patient. J Med Virol. 1994;42:338347.
33. Gilden DH, Beinlich BR, Rubinstien EM, et al. Varicella-zoster virus 67. Moulignier A, Pialoux G, Dega H, et al. Brain stem encephalitis due to varicella-
myelitis: an expanding spectrum. Neurology. 1994;44:18181823. zoster virus in a patient with AIDS. Clin Infect Dis. 1995;20:13781380.
34. Takeoka M, Takahashi T. Infectious and inflammatory disorders of the circu- 68. Verghese A, Sugar AM. Herpes zoster ophthalmicus and granulomatous angi-
latory system and stroke in childhood. Curr Opin Neurol. 2002;15:159164. itis: an ill-appreciated cause of stroke. J Am Geriatr Soc. 1986;34:309312.
35. deVeber G, Roach ES, Riela AR, et al. Stroke in children: recognition, treat- 69. Moriuchi H, Rodriguez W. Role of varicella-zoster virus in stroke syn-
ment, and future directions. Semin Pediatr Neurol. 2000;7:309317. dromes. Pediatr Infect Dis J. 2000;19:648653.
36. Askalan R, Laughlin S, Mayank S, et al. Chickenpox and stroke in child- 70. Joy JL, Carlo JR, Velez-Borras JR. Cerebral infarction following herpes
hood: a study of frequency and causation. Stroke. 2001;32:12571262. zoster: the enlarging clinical spectrum. Neurology. 1989;39:1640.
37. Berger TM, Caduff JH, Gebbers JO. Fatal varicella-zoster virus antigen- 71. Reshef E, Greenberg SB, Jankovic J. Herpes zoster ophthalmicus followed
positive giant cell arteritis of the central nervous system. Pediatr Infect by contralateral hemiparesis: report of two cases and review of literature.
Dis J. 2000;19:653656. J Neurol Neurosurg Psychiatry. 1985;48:122127.
38. Singhal AB, Singhal BS, Ursekar MA, et al. Serial MR angiography and 72. Bhat G, Mathur DS, Saxena GN, et al. Granulomatous angiitis of the cen-
contrast-enhanced MRI in chickenpox-associated stroke. Neurology. 2001; tral nervous system associated with herpes zoster. J Assoc Physicians India.
56:815817. 2002;50:977978.
39. Gilbert GJ. Serial MR angiography and contrast-enhanced MRI in 73. Nogueira RG, Sheen VL. Images in clinical medicine: herpes zoster oph-
chickenpox-associated stroke. Neurology. 2001;57:1742. thalmicus followed by contralateral hemiparesis. N Engl J Med. 2002;346:
40. Cobo LM. Corneal complications of herpes zoster ophthalmicus: preven- 1127.
tion and treatment. Cornea. 1988;7:5056. 74. Eidelberg D, Sotrel A, Horoupian DS, et al. Thrombotic cerebral vascu-
41. Whitley RJ, Soong SJ, Dolin R, et al. Early vidarabine therapy to control lopathy associated with herpes zoster. Ann Neurol. 1986;19:714.
the complications of herpes zoster in immunosuppressed patients. N Engl J 75. Patrick JT, Russell E, Meyer J, et al. Cervical (C2) herpes zoster infection
Med. 1982;307:971975. followed by pontine infarction. J Neuroimaging. 1995;5:192193.
42. Rusthoven JJ, Ahlgren P, Elhakim T, et al. Risk factors for varicella zoster 76. Blue MC, Rosenblum WI. Granulomatous angiitis of the brain with herpes
disseminated infection among adult cancer patients with localized zoster. zoster and varicella encephalitis. Arch Pathol Lab Med. 1983;107:126128.
Cancer. 1988;62:16411646. 77. Melanson M, Chalk C, Georgevich L, et al. Varicella-zoster virus DNA
43. Gnann JW, Whitley RJ. Natural history and treatment of varicella-zoster in in CSF and arteries in delayed contralateral hemiplegia: evidence for viral
high-risk populations. J Hosp Infect. 1991;18(suppl A):317329. invasion of cerebral arteries. Neurology. 1996;47:569570.
44. Balfour HH Jr, Bean B, Laskin OL, et al. Acyclovir halts progression of herpes 78. Booss J, Haak BB, Leroy RF. Delayed contralateral hemiplegia following
zoster in immunocompromised patients. N Engl J Med. 1983;308:14481453. herpes zoster ophthalmicus: should antiviral therapy be used? Eur Neurol.
45. Kleinschmidt-DeMasters BK, Gilden DH. Varicella-Zoster virus infections 1985;24:225228.
of the nervous system: clinical and pathologic correlates. Arch Pathol Lab 79. Freedman MS, Macdonald RD. Herpes zoster ophthalmicus with delayed cere-
Med. 2001;125:770780. bral infarction and meningoencephalitis. Can J Neurol Sci. 1987;14:312314.
46. Kleinschmidt-DeMasters BK, Amlie-Lefond C, Gilden DH. The patterns of 80. Devinsky O, Cho ES, Petito CK, et al. Herpes zoster myelitis. Brain.
varicella zoster virus encephalitis. Hum Pathol. 1996;27:927938. 1991;114(pt 3):11811196.
47. Elliott KJ. Other neurological complications of herpes zoster and their 81. Grant AD, Fox JD, Brink NS, et al. Detection of varicella-zoster virus DNA
management. Ann Neurol. 1994;35(suppl):S57S61. using the polymerase chain reaction in an immunocompromised patient
48. Dworkin RH, Schmader KE. Treatment and prevention of postherpetic with transverse myelitis secondary to herpes zoster. Genitourin Med.
neuralgia. Clin Infect Dis. 2003;36:877882. 1993;69:273275.
49. Jemsek J, Greenberg SB, Taber L, et al. Herpes zoster-associated encepha- 82. Heller HM, Carnevale NT, Steigbigel RT. Varicella zoster virus transverse
litis: clinicopathologic report of 12 cases and review of the literature. myelitis without cutaneous rash. Am J Med. 1990;88:550551.
Medicine (Baltimore). 1983;62:8197. 83. Manian FA, Kindred M, Fulling KH. Chronic varicella-zoster virus myelitis
50. Cheung WC, Yuen KY, Chang CM, et al. Herpes zoster associated encepha- without cutaneous eruption in a patient with AIDS: report of a fatal case.
litis in dialysis patients. J Infect. 1991;23:169174. Clin Infect Dis. 1995;21:986988.
51. Dueland AN, Devlin M, Martin JR, et al. Fatal varicella-zoster virus menin- 84. Hanakawa T, Hashimoto S, Kawamura J, et al. Magnetic resonance imaging
goradiculitis without skin involvement. Ann Neurol. 1991;29:569572. in a patient with segmental zoster paresis. Neurology. 1997;49:631632.
52. Hughes BA, Kimmel DW, Aksamit AJ. Herpes zoster-associated meningoen- 85. Friedman DP. Herpes zoster myelitis: MR appearance. AJNR Am J
cephalitis in patients with systemic cancer. Mayo Clin Proc. 1993;68:652655. Neuroradiol. 1992;13:14041406.
53. McKenna KF, Warneke LB. Encephalitis associated with herpes zoster: a 86. Hwang YM, Lee BI, Chung JW, et al. A case of herpes zoster myelitis: positive
case report and review. Can J Psychiatry. 1992;37:271273. magnetic resonance imaging finding. Eur Neurol. 1991;31:164167.

Scheld_Ch10.indd 166 2/21/14 5:34 PM


Chapter 10: Neurologic Manifestations of Varicella and Herpes Zoster 167

87. de Silva SM, Mark AS, Gilden DH, et al. Zoster myelitis: improvement 120. Cinque P, Bossolasco S, Vago L, et al. Varicella-zoster virus (VZV) DNA
with antiviral therapy in two cases. Neurology. 1996;47:929931. in cerebrospinal fluid of patients infected with human immunodeficiency
88. Adour KK. Otological complications of herpes zoster. Ann Neurol. virus: VZV disease of the central nervous system or subclinical reactiva-
1994;35(suppl):S62S64. tion of VZV infection? Clin Infect Dis. 1997;25:634639.
89. Mizock BA, Bartt R, Agbemazdo B. Herpes zoster oticus with pontine lesion: 121. LaRussa P, Lungu O, Hardy I, et al. Restriction fragment length polymor-
segmental brain-stem encephalitis. Clin Infect Dis. 2000;30:229230. phism of polymerase chain reaction products from vaccine and wild-type
90. Hellinger WC, Bolling JP, Smith TF, et al. Varicella-zoster virus retinitis in varicella-zoster virus isolates. J Virol. 1992;66:10161020.
a patient with AIDS-related complex: case report and brief review of the 122. Kronenberg A, Schupbach R, Schuknecht B, et al. Multifocal vasculopa-
acute retinal necrosis syndrome. Clin Infect Dis. 1993;16:208212. thy due to Varicella-Zoster virus (VZV): serial analysis of VZV DNA and
91. Ormerod LD, Larkin JA, Margo CA, et al. Rapidly progressive herpetic intrathecal synthesis of VZV antibody in cerebrospinal fluid. Clin Infect
retinal necrosis: a blinding disease characteristic of advanced AIDS. Clin Dis. 2002;35:330333.
Infect Dis. 1998;26:3447. 123. Dunkle LM, Arvin AM, Whitley RJ, et al. A controlled trial of acyclovir
92. Engstrom RE Jr, Holland GN, Margolis TP, et al. The progressive outer for chickenpox in normal children. N Engl J Med. 1991;325:15391544.
retinal necrosis syndrome: a variant of necrotizing herpetic retinopathy in 124. Balfour HH Jr, Rotbart HA, Feldman S, et al. Acyclovir treatment of
patients with AIDS. Ophthalmology. 1994;101:14881502. varicella in otherwise healthy adolescents. The Collaborative Acyclovir
93. Galindez OA, Sabates NR, Whitacre MM, et al. Rapidly progressive outer Varicella Study Group. J Pediatr. 1992;120:627633.
retinal necrosis caused by varicella zoster virus in a patient infected with 125. Wallace MR, Bowler WA, Murray NB, et al. Treatment of adult varicella
human immunodeficiency virus. Clin Infect Dis. 1996;22:149151. with oral acyclovir: a randomized, placebo-controlled trial. Ann Intern
94. Perez-Blazquez E, Traspas R, Mendez Marin I, et al. Intravitreal ganci- Med. 1992;117:358363.
clovir treatment in progressive outer retinal necrosis. Am J Ophthalmol. 126. Nyerges G, Meszner Z, Gyarmati E, et al. Acyclovir prevents dissemina-
1997;124:418421. tion of varicella in immunocompromised children. J Infect Dis. 1988;157:
95. Palay DA, Sternberg P Jr, Davis J, et al. Decrease in the risk of bilateral acute 309313.
retinal necrosis by acyclovir therapy. Am J Ophthalmol. 1991;112:250255. 127. Prober CG, Kirk LE, Keeney RE. Acyclovir therapy of chickenpox in immu-
96. Watson CP, Evans RJ, Watt VR, et al. Post-herpetic neuralgia: 208 cases. nosuppressed childrena collaborative study. J Pediatr. 1982;101:622625.
Pain. 1988;35:289297. 128. Huff JC, Bean B, Balfour HH Jr, et al. Therapy of herpes zoster with oral
97. de Morgas JM, Kierland RR. The outcome of patients with herpes zoster. acyclovir. Am J Med. 1988;85:8489.
Arch Dermatol. 1957;75:193196. 129. McKendrick MW, McGill JI, White JE, et al. Oral acyclovir in acute
98. Rogers RS III, Tindall JP. Geriatric herpes zoster. J Am Geriatr Soc. 1971; herpes zoster. Br Med J (Clin Res Ed). 1986;293:15291532.
19:495504. 130. Morton P, Thomson AN. Oral acyclovir in the treatment of herpes zoster
99. Bennett GJ. Hypotheses on the pathogenesis of herpes zoster-associated in general practice. N Z Med J. 1989;102:9395.
pain. Ann Neurol. 1994;35(suppl):S38S41. 131. Wood MJ, Kay R, Dworkin RH, et al. Oral acyclovir therapy accelerates
100. Kost RG, Straus SE. Postherpetic neuralgiapathogenesis, treatment, and pain resolution in patients with herpes zoster: a meta-analysis of placebo-
prevention. N Engl J Med. 1996;335:3242. controlled trials. Clin Infect Dis. 1996;22:341347.
101. Kanazi GE, Johnson RW, Dworkin RH. Treatment of postherpetic neural- 132. Beutner KR, Friedman DJ, Forszpaniak C, et al. Valacyclovir compared
gia: an update. Drugs. 2000;59:11131126. with acyclovir for improved therapy for herpes zoster in immunocompe-
102. Watson CP, Babul N. Efficacy of oxycodone in neuropathic pain: a ran- tent adults. Antimicrob Agents Chemother. 1995;39:15461553.
domized trial in postherpetic neuralgia. Neurology. 1998;50:18371841. 133. Tyring S, Barbarash RA, Nahlik JE, et al. Famciclovir for the treatment of
103. Kishore-Kumar R, Max MB, Schafer SC, et al. Desipramine relieves acute herpes zoster: effects on acute disease and postherpetic neuralgia: a ran-
postherpetic neuralgia. Clin Pharmacol Ther. 1990;47:305312. domized, double-blind, placebo-controlled trial. Collaborative Famciclovir
104. Watson CP, Vernich L, Chipman M, et al. Nortriptyline versus ami- Herpes Zoster Study Group. Ann Intern Med. 1995;123:8996.
triptyline in postherpetic neuralgia: a randomized trial. Neurology. 134. Tyring SK, Beutner KR, Tucker BA, et al. Antiviral therapy for herpes
1998;51:11661171. zoster: randomized, controlled clinical trial of valacyclovir and famciclovir
105. Rowbotham M, Harden N, Stacey B, et al. Gabapentin for the treat- therapy in immunocompetent patients 50 years and older. Arch Fam Med.
ment of postherpetic neuralgia: a randomized controlled trial. JAMA. 2000;9:863869.
1998;280:18371842. 135. Gnann JW Jr, Whitley RJ. Clinical practice: herpes zoster. N Engl J Med.
106. Bernstein JE, Korman NJ, Bickers DR, et al. Topical capsaicin treatment of 2002;347:340346.
chronic postherpetic neuralgia. J Am Acad Dermatol. 1989;21:265270. 136. Cobo LM, Foulks GN, Liesegang TJ, et al. Oral acyclovir in the treatment
107. Rowbotham MC, Davies PS, Verkempinck C, et al. Lidocaine patch: dou- of acute herpes zoster ophthalmicus. Ophthalmology. 1986;93:763770.
ble-blind controlled study of a new treatment method for post-herpetic 137. Whitley RJ, Weiss H, Gnann JW Jr, et al. Acyclovir with and without
neuralgia. Pain. 1996;65:3944. prednisone for the treatment of herpes zoster: a randomized, placebo-
108. Galer BS, Rowbotham MC, Perander J, et al. Topical lidocaine patch re- controlled trial. The National Institute of Allergy and Infectious Diseases
lieves postherpetic neuralgia more effectively than a vehicle topical patch: Collaborative Antiviral Study Group. Ann Intern Med. 1996;125:376383.
results of an enriched enrollment study. Pain. 1999;80:533538. 138. Wood MJ, Johnson RW, McKendrick MW, et al. A randomized trial of
109. Kotani N, Kushikata T, Hashimoto H, et al. Intrathecal methylprednisolone acyclovir for 7 days or 21 days with and without prednisolone for treat-
for intractable postherpetic neuralgia. N Engl J Med. 2000;343:15141519. ment of acute herpes zoster. N Engl J Med. 1994;330:896900.
110. McKendrick MW, Care CC, Kudesia G, et al. Is VZV reactivation a com- 139. Whitley RJ, Gnann JW Jr, Hinthorn D, et al. Disseminated herpes zoster
mon cause of unexplained unilateral pain? Results of a prospective study in the immunocompromised host: a comparative trial of acyclovir and
of 57 patients. J Infect. 1999;39:209212. vidarabine. The NIAID Collaborative Antiviral Study Group. J Infect Dis.
111. Gilden DH, Dueland AN, Devlin ME, et al. Varicella-zoster virus reactiva- 1992;165:450455.
tion without rash. J Infect Dis. 1992;166(suppl 1):S30S34. 140. Ljungman P, Lonnqvist B, Ringden O, et al. A randomized trial of oral
112. Gilden DH, Wright RR, Schneck SA, et al. Zoster sine herpete, a clinical versus intravenous acyclovir for treatment of herpes zoster in bone
variant. Ann Neurol. 1994;35:530533. marrow transplant recipients. Nordic Bone Marrow Transplant Group.
113. Amlie-Lefond C, Mackin GA, Ferguson M, et al. Another case of virologi- Bone Marrow Transplant. 1989;4:613615.
cally confirmed zoster sine herpete, with electrophysiologic correlation. 141. White CJ, Kuter BJ, Hildebrand CS, et al. Varicella vaccine (VARIVAX)
J Neurovirol. 1996;2:136138. in healthy children and adolescents: results from clinical trials, 1987 to
114. Gershon AA, LaRussa P, Steinberg SP. Varicella-zoster virus. In: Murray 1989. Pediatrics. 1991;87:604610.
PR, Barron EJ, Pfaller MA, et al, eds. Manual of Clinical Microbiology. 142. Watson BM, Piercy SA, Plotkin SA, et al. Modified chickenpox in children
6th ed. Washington, DC: ASM Press; 1995:896904. immunized with the Oka/Merck varicella vaccine. Pediatrics. 1993;91:1722.
115. Nahass GT, Goldstein BA, Zhu WY, et al. Comparison of Tzanck smear, 143. Gershon AA, Steinberg SP, LaRussa P, et al. Immunization of healthy adults
viral culture, and DNA diagnostic methods in detection of herpes simplex with live attenuated varicella vaccine. J Infect Dis. 1988;158:132137.
and varicella-zoster infection. JAMA. 1992;268:25412544. 144. American Academy of Pediatrics Committee on Infectious Diseases.
116. Dlugosch D, Eis-Hubinger AM, Kleim JP, et al. Diagnosis of acute and la- Recommendations for the use of live attenuated varicella vaccine.
tent varicella-zoster virus infections using the polymerase chain reaction. Pediatrics. 1995;95:791796.
J Med Virol. 1991;35:136141. 145. Hardy I, Gershon AA, Steinberg SP, et al. The incidence of zoster after
117. Kido S, Ozaki T, Asada H, et al. Detection of varicella-zoster virus (VZV) immunization with live attenuated varicella vaccine: a study in children
DNA in clinical samples from patients with VZV by the polymerase chain with leukemia. Varicella Vaccine Collaborative Study Group. N Engl J
reaction. J Clin Microbiol. 1991;29:7679. Med. 1991;325:15451550.
118. Shoji H, Honda Y, Murai I, et al. Detection of varicella-zoster virus DNA 146. Gershon AA, Steinberg SP, Gelb L. Live attenuated varicella vaccine use in
by polymerase chain reaction in cerebrospinal fluid of patients with herpes immunocompromised children and adults. Pediatrics. 1986;78:757762.
zoster meningitis. J Neurol. 1992;239:6970. 147. Hata A, Asanuma H, Rinki M, et al. Use of an inactivated varicella vaccine in
119. Burke DG, Kalayjian RC, Vann VR, et al. Polymerase chain reaction recipients of hematopoietic-cell transplants. N Engl J Med. 2002;347:2634.
detection and clinical significance of varicella-zoster virus in cerebrospinal 148. Levin MJ, Murray M, Zerbe GO, et al. Immune responses of elderly
fluid from human immunodeficiency virus-infected patients. J Infect Dis. persons 4 years after receiving a live attenuated varicella vaccine. J Infect
1997;176:10801084. Dis. 1994;170:522526.

Scheld_Ch10.indd 167 2/21/14 5:34 PM


CHAPTER 11 CYTOMEGALOVIRUS
PAUL D. GRIFFITHS

wild type strains (16). Propagation in fibroblasts selects for this


HISTORY and other genetic changes so viruses are now maintained in the
In the early years of the twentieth century, histopathologists laboratory as bacterial artificial chromosomes with approxi-
studying stillborn infants identified the characteristic intranu- mately 165 protein-coding genes in wild type strains (17). In
clear inclusions in fatal cases of what is now termed cytomegalic addition, the genome encodes approximately 10 micro-RNAs.
inclusion disease (1). At first, the inclusion bodies were attributed Recent results with ribosome profiling suggest that the genet-
to a new protozoal infection (2). However, in 1921 Goodpasture ics of this virus may be even more complex than described so
and Talbot (3) remarked on the similarity between these strange far (18). Salient features of the molecular biology of the virus
bodies and the intranuclear inclusions produced in the skin are summarized later in this chapter, with a focus on genes
lesions of varicella (4). The viral etiology of the inclusions was important for immune control or which are the targets of anti-
first proposed by von Glahn and Pappenheimer (5) in 1925. The viral chemotherapy; for extensive details, readers are referred
following year, Cole and Kuttner (6) were able to transmit the elsewhere (19). By international agreement, proteins encoded
guinea pig form of cytomegalic inclusion disease from salivary by the virus are described according to their map position (20)
gland material passed through a Berkefeld N filter. Later, three (Fig. 11.1). For example, gpUL55, the glycoprotein known
laboratories simultaneously reported the isolation of the caus- as glycoprotein B, originates from the fifty-fifth open reading
ative human virus in cell cultures: Smith (7) in 1956 from sali- frame of the unique long region, while ppUL82 is a tegument
vary glands, Rowe et al. (8) in 1956 from adenoid tissue, and phosphoprotein transactivator that maps to the eighty-second
Weller et al. (9) in 1957 from a liver biopsy specimen. In 1960, open reading frame of the unique long region.
Weller et al. (10) named the virus cytomegalovirus (CMV). CMV encodes several transactivators. The major immediate-
Since a characteristic feature of cytomegalic inclusion disease early region maps to UL122/123. Alternative splicing produces
is global mental retardation, scientists have known for more four major proteins, one of which interacts directly with TATA-
than 50 years the basic nature of CMV and its tropism for cells binding protein and downregulates its own promoter. This re-
of the central nervous system (CNS). However, whether this gion contains upstream enhancers, is a strong regulatory region,
tropism resulted from the immature phenotype of cells dividing and is a major target of cell-mediated immunity. pUL69 is a
to form the fetal CNS or whether CMV could also infect transactivator found in the virion. It synergizes with ppUL82 to
terminally differentiated neural cells of adult origin was not activate the major immediate-early promoter. These two proteins
known. Although individual cases of CMV encephalitis have together can thus be thought of as being functionally equivalent
been described in patients who were immunocompromised but to the -transinducing factor of herpes simplex virus (HSV).
not infected with human immunodeficiency virus (HIV), such A total of 11 genes are required for CMV replication (21).
cases are still the exception rather than the rule. For example, These proteins together provide in trans the functions required
CMV encephalitis was described in a renal transplant recipient to allow replication of the lytic origin of replication (22). Some
treated with augmented immunosuppression for graft rejec- of the functions are directly analogous to those required by
tion (11) and in a patient with Hodgkin disease who became HSV, for example, DNA polymerase (UL54) and its accessory
demented and died of pneumonia (12). The acquired immu- protein (UL44), although an equivalent to the origin-binding
nodeficiency syndrome (AIDS) epidemic changed this perspec- protein has not been identified.
tive; CMV infection was found so often at autopsy before the Structural proteins of the virus include the major capsid
advent of highly active antiretroviral therapy (HAART) that protein, UL86, and the minor capsid protein, UL46. The as-
it was the most common opportunistic infection of the CNS semblin gene complex (UL80a) includes a protein important
in patients with AIDS (13). Both CMV infection and disease, for packaging DNA as well as a protease. Several phosphopro-
including of the CNS, have decreased dramatically in coun- teins are found in the tegument (UL32, UL83, UL82, UL99).
tries where HAART is readily available. This clinical benefit is ppUL83, known as the lower matrix protein, is the major
consistent with the possibility that CMV infection of the CNS antigen detected in circulating leukocytes (23) and is the sec-
was a major and often unrecognized contributor to CNS dis- ond major target of the cell-mediated immune response (24).
ease. However, the clinical course of patients with AIDS is so Major envelope proteins exist on the plasma membrane
complex that it is not certain how much CNS disease could be as multimolecular complexes and are major targets for hu-
attributed to the presence of CMV as opposed to HIV, which moral immune responses. Glycoprotein complex I consists of
is also neurotropic and controlled by HAART. Interestingly, a homodimer of glycoprotein B (gB; gp UL55). Glycoprotein
CMV encephalopathy can predominate as the cause of death in complex II consists of gM (gp UL100) and gN (gp UL73).
a child who acquires both CMV and HIV from its mother (14). Glycoprotein complex III consists of gH (gp UL75), gL (gp
The aim of this chapter is to review what is known and how UL115), and gO (gp UL74) in fibroblasts but gH, gL, and
much remains to be defined about CMV infection of the CNS. three proteins (pUL128, pUL130, pUL131A) encoded within
the region of the genome which is deleted upon passage in
fibroblasts. This five-member complex of proteins mediates
INFECTIOUS AGENT viral entry into epithelial and endothelial cell lines (25).
CMV encodes a series of genes that interfere with immune
CMV has the largest genome (229 kb) of the viruses known to recognition of virus-infected cells. The product of US6 blocks
infect humans. More than 200 potential open reading frames the transporter associated with antigen presentation, which
were identified in strain Ad169 (15), with an additional 22 in normally takes peptides from the cytosol into the lumen of the

168

Scheld_Ch11.indd 168 2/21/14 5:34 PM


Chapter 11: Cytomegalovirus 169

0 5 10 15 20 25 30

RL1 RNA2.7 RL5A RNA1.2 RL10 RL13 UL4 UL7 UL10 UL13 UL16 UL19 UL22A UL24
RL6 RL8A RL11 UL1 UL5 UL8 UL11 UL14 UL17 UL20 UL23
RL9A RL12 UL2 UL6 UL9 UL15A UL18 UL21A

30 35 40 45 50 55 60

UL25 UL27 UL29 UL30 UL31 UL32 UL33 UL34 UL35 UL36 UL37 UL40 UL43 UL45
UL26 UL30A UL38 UL41A UL44
UL42

60 65 70 75 80 85 90

UL46 UL47 UL48 UL49 UL50 UL52 UL53 UL54 UL55 UL56 UL57
UL48A UL51

90 95 100 105 110 115 120

ori RNA4.9 UL69 UL70 UL71 UL74 UL75 UL76 UL78 UL80 UL82
UL72 UL74A UL77 UL79 UL80.5
UL73

120 125 130 135 140 145 150

UL83 UL84 UL85 UL86 UL87 UL89 UL93 UL95 UL97 UL98 UL100 UL102
UL88 UL91 UL94 UL96 UL99
UL92

150 155 160 165 170 175 180

UL104 UL105 RNA5.0 UL112 UL115 UL119 UL122 UL124 UL128 UL132
UL103 UL111A UL114 UL116 UL120 UL123 UL130 UL148
UL117 UL121 UL131A

180 185 190 195 200 205 210

UL147A UL144 UL141 UL138 UL133 UL148D IRS1 US1 US3 US7 US9 US12 US14
UL147 UL142 UL140 UL136 UL148A UL150 US2 US6 US8 US10 US13 US15
UL146 UL139 UL135 UL148B UL150A US11
UL145 UL148C

210 215 220 225 230 235 kbp

US16 US18 US20 US22 US23 US24 US26 US27 US29 US32 US34A TRS1
US17 US19 US21 US28 US30 US33A
US31 US34

FIGURE 11.1 Consensus genetic map of Merlin strain human cytomegalovirus (HCMV), updated from
the publication by Dolan et al. 2004. The unique long (UL), unique short (US), terminal repeat long (RL),
terminal repeat short (TRS), and inverted repeat short (IRS) regions are shown, with a thicker format for the
genome termini. Introns are shown as narrow white bars. Colored arrows indicate protein-coding regions
as well as the direction of transcription. The colors indicate degrees of conservation of genes between
the Alpha-, Beta-, and Gammaherpesvirinae (core genes) or between the Beta- and Gammaherpesvirinae
(subcore genes). Various subsets of the remaining noncore genes are grouped into gene families. Note that
the micro-RNAs encoded within the genome are not shown. (Figure kindly provided by Dr. A. Davison.)

Scheld_Ch11.indd 169 2/21/14 5:34 PM


170 Part II: Viral Infections and Related Disorders

endoplasmic reticulum. US3 binds to human leukocyte anti- of fetuses (53). Moreover, only 12.7% of infected infants have
gen (HLA) heavy chains and retains them in the endoplasmic clinical manifestations at birth (from very mild to severe), and
reticulum. Genes US2 and US11 take HLA complexes from the risk of subsequent sequelae averages 13.5% (54). Although
the endoplasmic reticulum and export them back into the the rate of transmission of virus in utero appears lower, the risk
cytosol for degradation in the proteasome. All of these effects of neonatal disease with attendant sequelae appears to be higher
combine to decrease HLA display at the plasma membrane when the mother has acquired CMV infection during the first
and thus allow the virus to escape from cytotoxic T cells. trimester of pregnancy (55).
However, because HLA molecules also provide nonantigen- Maternal immunity to CMV neither prevents virus reac-
specific signals to natural killer (NK) cells and macrophages, tivation nor controls the systemic spread of virus that can
their absence could trigger destruction of the virus-infected produce congenital infection (44,48,5660). Studies that use
cell via these innate immune effectors. To avoid this, CMV restriction endonuclease analysis of viral DNA to examine the
has additional genes: UL18 to provide a negative signal to genetic relatedness between CMV strains isolated repeatedly
macrophages and UL40 to provide a negative signal to NK from mother and offspring indicate reactivation of an identi-
cells. In addition, the proteins encoded by UL16, UL141, cal latent virus rather than reinfection with an exogenous one
and UL142 block the recognition of a positive signal for NK (61). In other cases, there is evidence for exogenous reinfection
cells. In addition, a viral micro-RNA interferes with recogni- with a different strain (62,63). In a highly seropositive (82% of
tion of another of these host defense signals. In combination, persons) urban population of low socioeconomic background
the function of these genes allows CMV to persist in the host in Alabama, recurrent CMV infection produced a 1.6%
despite strong humoral and cell-mediated immune responses. (13/835 infants) rate of congenital infection, thus indicating
CMV encodes four sets of proteins that are predicted that most intrauterine infections in this population result from
G proteincoupled receptors (US27 and US28; UL33; UL78). this phenomenon (49). In contrast, the infants of immune
UL97 is a protein kinase homolog responsible for phosphory- middle-class women with a seropositivity rate of 55% had a
lating ganciclovir and acyclovir (2628). lower rate of congenital infection (0.19%, i.e., 10/5,242). This
rate is not significantly different from that observed in Great
Britain and Sweden (44,48,57). Overall, the mothers of only
EPIDEMIOLOGY 25% of babies born with congenital CMV infection in the
United States had primary infection during pregnancy (64).
Humans are the only reservoir for CMV (29). Infection is Although maternal immunity is imperfect, congenital infec-
endemic worldwide and has no seasonal variation. The prev- tions that result from recurrent infection are less likely to pro-
alence of CMV infection increases directly with age, with duce fetal damage than those resulting from primary infections
significant variations according to geographic, ethnic, and (49). However, the larger number of seropositive women in a
socioeconomic backgrounds. Antibodies of immunoglobulin G community may provide a number of infected neonates equal
(IgG) class against CMV can be found in approximately 60% to those born to seroconverting mothers (65).
of adults in developed countries, and virtually 100% of adults During pregnancy and in the immediate postpartum period,
in developing countries (30). Within developed countries, CMV can be shed at variable rates from one or more sites (49).
acquisition of CMV infection is increased in poor socioeco- Rates of excretion are higher in younger, nonwhite women
nomic environments and by sexual contact. The presence of from lower socioeconomic backgrounds. Most women who
IgG antibodies indicates that infection has occurred some- excrete virus during pregnancy do so as a result of recurrent
time in the past and that the virus should be presumed to be infections (reactivation or reinfection) (61). There is little cor-
latent. The sites of latency of CMV are not known, although relation between CMV excretion from the cervix or in urine
myeloid dendritic cells are clearly one of them, with reactiva- during pregnancy and the subsequent birth of a congenitally
tion of virus linked to cellular differentiation (3133). CMV infected infant (47,50,66), but the presence of CMV in the
can be transmitted iatrogenically by all organ allografts and maternal genital tract at delivery and in the breast milk after
at autopsy can be found in most tissues of the body. Thus, delivery is strongly associated with intrapartum/postpartum
whether CMV has a single site of latency or whether silent in- transmission of infection to the infant (34,36). In full-term and
fection can become established in multiple tissues is debatable. otherwise healthy infants, perinatal infection has little clinical
Transmission of infection from one individual to another importance except for a small number of cases of interstitial
requires direct contact. Sources of virus include saliva, blood, pneumonia (36,67,68). However, such children are a major
vaginal secretions, semen, and breast milk (3436). Salivary source of infection for other children in the day care environ-
excretion of virus, particularly in association with mouthing ment (69,70) and represent an occupational risk to child care
of toys, can lead to baby-to-baby transmission in the child care personnel, particularly women of childbearing age (39,7173).
setting (3739). Susceptible children can thus acquire CMV in If a patient who is CMV IgG-seropositive becomes immuno-
the day care environment and transmit infection to susceptible suppressed, CMV reactivation is common, being found typically
family members in the home. Transmission of CMV from an in 50% of transplant recipients at some time after transplanta-
infant to his or her seronegative motheras well as from an tion. The donor organ can also transmit CMV to seronegative
infant to pregnant caretakers, with subsequent transmission to individuals (74) or to seropositive individuals (75), showing
the fetushas been documented (40,41). Overall, in the United that preexisting immunity to CMV does not provide protection
States, contact with young children is a much more important against infection. However, preexisting immunity does provide
source of CMV for mothers than is sexual exposure (42). moderate protection against CMV replication to high levels and
Prospective studies in the United States and Europe have disease (76). Primary CMV infection can also be acquired from
established that pregnancy itself does not increase the risk of blood transfusion (77).
acquiring CMV infection (4350). Primary CMV infections dur- In patients who are HIV positive, the same principles apply:
ing pregnancy are generally asymptomatic (43,4547,4951). CMV infection becomes increasingly common as the immu-
Among susceptible women, the risk of seroconversion during nocompromised state progresses, although formal studies of
pregnancy averages 1.7% (52). In primary CMV infection, an natural history have not been performed. However, CMV
innate barrier prevents in utero transmission; thus, primary disease, typically retinitis, clearly is uncommon before the
maternal CMV infection leads to transmission in only about 32% CD4 count declines to 100 106/L. It becomes increasingly

Scheld_Ch11.indd 170 2/21/14 5:34 PM


Chapter 11: Cytomegalovirus 171

common as such patients are followed, with typically 10% CMV infection include productive CMV replication leading
of patients developing CMV disease within 2 years of follow- to destruction of individual cells (lytic infection) and indirect
up (78). Retinitis is associated with CMV encephalitis (79). damage mediated by action of components of the immune sys-
By itself, it represents approximately 85% of CMV disease in tem (immunopathology).
patients with AIDS, with gastrointestinal tract infection rep-
resenting approximately 10% and CMV CNS disorders only
1% of total disease. Nevertheless, presentations in the CNS Lytic Infections
(and peripheral nervous system) are being increasingly recog-
nized, as described later in this chapter. Continuous viral replication in affected organs could explain
why some infants are severely affected and others remain free
of symptoms. Longitudinal studies have demonstrated that
ETIOLOGY excretion of CMV into urine and saliva persists for years.
Chronic viral replication probably also occurs at other ana-
In cases of congenital CMV, one can be certain that the CNS tomic sites that are less accessible to virologic examinations
disease is attributable to CMV, as no other infectious agent is (e.g., middle ear, brain).
present and the syndrome is characteristic. The same is prob- The histopathologic hallmark of CMV CNS infection is the
ably true in the case of a female preterm infant born to an characteristic intranuclear inclusion body (Fig. 11.2). This indi-
HIV-positive mother who acquired congenital CMV and HIV cates productive infection of the cell and represents a major site
infection vertically. At 5 months, the child developed AIDS of virus formation. Such cells are likely committed to die, so CNS
and died at age 11 months of a rapidly progressive diffuse dysfunction could result from loss of their function. Inclusion-
encephalopathy. At autopsy, CMV encephalitis was seen, with bearing cells were seen in six fetuses with polymerase chain reac-
occasional inclusion bodies, yet there was no evidence for HIV tion (PCR)positive amniotic fluids that were terminated at 19 to
infection of the brain (14). 35 weeks because of CNS lesions seen on ultrasound examina-
However, the same clear-cut picture is not seen in adult pa- tions (91). Meningoencephalomyelitis and micronodular enceph-
tients with AIDS, who typically have evidence for HIV involve- alitis were seen in all, consistent with bloodborne dissemination
ment of the CNS, with or without CMV infection (80). A case of virus. Diffuse periventricular microglial nodules were present.
definition for CNS conditions attributable to HIV has been Inclusions were seen in the stria vascularis, Reissner membrane,
provided (81). However, it is not possible to exclude an HIV and the organ of Corti as well as in epithelial cells of the saccule
contribution to CNS symptomatology in patients with CMV. and utricule. The authors suggested that CMV may gain access
Indeed, in vitro there are multiple ways in which CMV and to the inner ear via the highly vascularized stria vascularis and
HIV could interact so that each could enhance the replication damage the ability of this structure to maintain potassium ho-
of the other; this has been reviewed by Griffiths (82). CMV meostasis, dissipate electric potentials, and support recirculation
infection can also downregulate HIV (83). Other opportunistic of potassium within the perilymph.
infections of the CNS are usually spatially distinct from CMV,
with the exception of HSV type 1 (HSV-1); superimposed CMV/
HSV-1 infection has been described (84,85). Interestingly, su- Immunopathology
perimposed CMV/HSV-1 infection of the brain has also been
described in a patient who does not have AIDS (86). Studies of CMV polypeptides immunoprecipitated by IgG anti-
Cases have been described in which CMV appears to be the bodies indicate that symptomatically infected infants had a
cause of CNS dysfunction (8789). Until more is known about delay (until 12 months of age) in the appearance of precipitating
the pathogenesis of CMV infection of the CNS, however, it is
not possible to guarantee that CMV infection is the sole cause
of the CNS dysfunction in an individual patient. Nevertheless,
populations of patients with AIDS can be examined to discern
patterns of clinical syndromes, with the strong presumption
that CMV is a candidate etiologic agent.
Animal experiments are not particularly helpful, because
human CMV is strongly species specific and replicates only in
humans. Animal CMVs, such as murine or guinea pig CMV,
can be studied; although they are different viruses, they do
have the advantage of occupying the same ecologic niche in
their species as human CMV does for humans. For example,
studies of the direct intracerebral inoculation of murine CMV
into mice of different ages show that the virus has a predilec-
tion to infect developing brain cells (90).

PATHOGENESIS
Results of preliminary studies indicate that the virulence
of congenital CMV infection is not strain dependent (61).
Although only a few infants have been studied, viruses iso-
lated from congenitally or perinatally infected siblings were
genetically identical. In two of the pairs of siblings assessed, FIGURE 11.2 Cytomegalovirus-infected cell showing the enlarge-
the first-born baby was severely affected, whereas the second- ment of the cell with two inclusion bodies, each surrounded by a
born infant was only subclinically infected (60). Mechanisms clear zone, within a pleomorphic nucleus. Hematoxylin-eosin stain;
that possibly explain some of the CNS damage associated with magnification 252.

Scheld_Ch11.indd 171 2/21/14 5:34 PM


172 Part II: Viral Infections and Related Disorders

antibodies (92). However, when a humoral immune response


develops, antibodies to viral polypeptides are precipitated in
greater numbers and are maintained for longer periods. This
sustained humoral immune response to congenital CMV infec-
tion occurs in the presence of persistent viral replication, thereby
creating the potential for the formation of immune complexes
(93). During the first year of life, immune complexes circulate
in a large proportion of infants with congenital infection. The
molecular weight of these immune complexes is higher in symp-
tomatic infants than in asymptomatic infants. In a few fatal
symptomatic cases, deposition of immune complexes in renal
glomeruli has been demonstrated. Another factor in disease
pathogenesis is vasculitis, which may occur in utero or after
birth. Infants with congenital CMV infection who die soon after
birth usually have disseminated intravascular coagulopathy.
CMV pneumonitis in transplant recipients may be immu-
nopathologically mediated (94). Thus, the presence of the
virus is required for the syndrome, but the clinical disease may FIGURE 11.3 Cytomegalovirus-infected epithelial cell in the choroid
be caused by an abnormal, enhanced, cell-mediated immune plexus. Hematoxylin-eosin stain; magnification 252.
response. Immunopathology presumably also explains the im-
mune recovery vitreitis, which follows successful treatment
of CMV retinitis in patients with AIDS. Similarly, it has been cells and some astrocytes. In some cases, the microglial re-
suggested that the demyelinating component of the disease in sponse surrounds an inclusion-bearing cell. Other inclusion
the peripheral nervous system could be immunopathologi- body cells may be missed in the plane of section. In the series
cally mediated (95). It is unlikely that an identical scenario by Vinters et al. (85), CMV could not be found in every case
pertains in the CNS of patients with AIDS, because neurologic of microglial nodular encephalitis identified. Infected cells can
symptoms usually occur at profoundly low CD4 cell counts. likely be targets for microglial attack before reaching the stage
However, there are distinct possibilities that host cytokines, of bearing an inclusion body, because staining with monoclo-
released as a result of CMV infection, stimulated by CMV, or nal antibodies or the use of in situ hybridization reveals many
even encoded by CMV, could mediate extensive neural dam- more cells to be CMV infected (101,102).
age. For example, CMV activates tumor necrosis factor- and The second route of viremic spread is through the cere-
interleukin-6 (IL-6), both of which may induce HIV expres- brospinal fluid (CSF) following viral replication in infected
sion in macrophages (96). CMV replication is also enhanced epithelial cells of the choroid plexus (Fig. 11.3). Secondary
by IL-8. CMV infection induces production of messenger RNA seeding of the ependymal surfaces and internal spread pro-
(mRNA) of the IL-8 receptor, while IL-8 increases CMV rep- duce necroses of periventricular white matter (102). This con-
lication approximately fourfold (97). pUS28 has 33% amino dition, necrotizing ventricular encephalitis, may be extensive.
acid homology with the IL-8 receptor (97). Because IL-8 stim- Ependymal surfaces are replaced with CMV-infected cells,
ulates production of polymorphonuclear leukocytes, this is a infiltrating macrophages, inflammatory exudate, and necrotic
plausible explanation for the increased level of polymorpho- cells with or without hemorrhage (85,101).
nuclear leukocytes seen in the CNS in association with CMV, The histologic changes may be classified into one of five
particularly polyradiculopathy. The CMV genome encodes major groups: (a) nodular encephalitis, in which widely dis-
an -chemokine (UL146) and an IL-10 homolog (UL111A) seminated microglial nodules, only a minority of which may
whose function remains undefined in vivo. contain inclusion-bearing cells (Fig. 11.4), are seen dissemi-
nated over a wide area (103); (b) isolated inclusion-bearing

PATHOLOGY
CMV reaches the brain through viremic spread. In patients
with AIDS, viremia has been reported to have both a cel-
lular and a plasma component (98). In healthy individuals,
CMV can be found by PCR in monocytes (31). In transplant
recipients, ppUL83 (pp65) antigen has been demonstrated in
monocytes, polymorphonuclear cells, or endothelial cells (23).
Which, if any, of these cells are important for disseminating
CMV to the brain of patients with AIDS is not clear, but there
is evidence that microglia are derived from the peripheral
blood monocyte pool (99,100). Regardless, there is evidence
that CMV can spread through the brain by two routes.
The first route involves infection of the endothelial cells
of the brain, with spread to the contiguous astrocytes in foot
processes and their cognate neurons. This route of inoculation
is mirrored by the following histologic findings: (a) infection
of endothelial cells in the brain, which may sometimes be
associated with vascular thrombosis. Isolated endothelial cells
containing inclusion bodies may also be seen in the spinal cord
in association with a myelitis; (b) multiple foci of glial nodular FIGURE 11.4 A microglial nodule with an inclusion-bearing cell.
encephalitis. The glial nodules consist of multiple microglial Hematoxylin-eosin stain; magnification 160.

Scheld_Ch11.indd 172 2/21/14 5:34 PM


Chapter 11: Cytomegalovirus 173

FIGURE 11.5 Isolated inclusion-bearing astrocyte in the subependyma.


Hematoxylin-eosin stain; magnification 252.

cells, which may be astrocytes (Fig. 11.5), neurons, or macro-


phages (104); (c) focal parenchymal necroses in an otherwise
normal neuropil, whose lesions are characterized by accumula-
tions of macrophages with axonal destruction and occasional
inclusion-bearing cells (105); (d) ventricular encephalitis,
characterized by inflammation and necrosis involving the ep-
endyma and subependymal glia (Fig. 11.6), often over a wide FIGURE 11.7 Radiculomyelitis involving the lumbar cord. Many
area of the ventricular lining (106); and (e) radiculomyelitis, darkly staining infected cells are seen. Immunoperoxidase for cyto-
megalovirus (Dako); 40.
in which inflammatory changes, often with a marked infiltrate
of polymorphonuclear neutrophils, are seen involving the cord
(Fig. 11.7), spinal roots, and dorsal root ganglia. To some
extent, the histologic changes mirror the portal of entry of the
virus into the CNS and its dissemination throughout the brain CLINICAL MANIFESTATIONS
and spinal cord, as discussed earlier.
The incidence of such findings in seven separate AIDS Clinical Presentation of Congenital Infection
autopsy series is shown in Table 11.1. The proportion of
patients in whom CMV infection of the CNS was documented
varied from 7.9% to 28%. In total, 161 (15.7%) of 1,026
Symptomatic Infection
patients in these studies had CMV involvement of the CNS. Disease in the symptomatic newborn is characterized by hepato-
An increasing number of viruses are being described that megaly, splenomegaly, microcephaly, direct hyperbilirubinemia,
produce similar clinical and pathologic changes in patients petechiae, and thrombocytopenia (108). Intracranial calcifica-
with AIDS (107). Indeed, the clinical and pathologic features tions and retinitis are demonstrable with regularity. Intrauterine
may reflect more the route of infection and the state of the growth retardation is often present. Diabetes insipidus has been
immune system than the nature of the virus. reported in a few infants with congenital infection (109).

FIGURE 11.6 An area of ventricular encepha-


litis involving the roof of the fourth ventricle.
Hematoxylin-eosin stain; magnification 40.

Scheld_Ch11.indd 173 2/21/14 5:34 PM


174 Part II: Viral Infections and Related Disorders

TA B L E 1 1 . 1
COMPARISON OF INCIDENCE AND TYPE OF CYTOMEGALOVIRUS-INDUCED CENTRAL NERVOUS SYSTEM
LESIONS IN DIFFERENT AUTOPSY SERIES

Morgello and Klatt and Vinters McKenzie Artigas Chimelli


Findings Simpson (95) Shibata (103) et al. (85) et al. (104) et al. (105) et al. (106) RFH

Total no. 107 164 160 75 180 252 88


CMV (all organs) 81 31 61 91 53
CMV-induced CNS lesions 30 16 26 10/49 39 20 20
Microglial nodules 30 (100%)a 6 (38%) 29 (112%) 32 (82%) 17 (85%) 11 (55%)
Isolated inclusions 15 (50%) 6 (30%)
Necrosis 4 (13%) 2 (8%) 9 (23%) 4 (20%)
Necrotizing
leukoencephalopathy
Necrosis/infarcts
Necrotizing 3 (10%) 2 (13%) 7 (35%)
ventriculoencephalitis
Encephalitis 6 (38%)
Necrotizing 16 (41%) 2 (10%) 4 (20%)
radiculomyelitis
Myelopathy 1 (4%)
Leptomeningitis/ 5 (19%)
meningoradiculitis
Vasculitis 5 (31%) 4 (15%) 7 (18%)
Neuronal degeneration 4 (25%)
Meningitis 2 (13%)
Choroid plexusitis 2 (8%)

RFH, Royal Free Hospital; CMV, cytomegalovirus.


a
Percentage figures relate to total number of CMV-induced CNS lesions.

The mortality among congenitally infected infants may TA B L E 1 1 . 2


be attributed to hepatic dysfunction, bleeding, or secondary
bacterial infection (Table 11.2). Death after the first month of PUBLIC HEALTH CONSEQUENCES OF CONGENITAL
life is usually the consequence of hepatic dysfunction; how- CYTOMEGALOVIRUS INFECTION
ever, beyond the first year of life, death is usually the result of
complications in the neurologically handicapped child second- Variable Estimated Value
ary to malnutrition, aspiration pneumonia, or overwhelming
No. of livebirths per year 4,000,000
infection.
The CNS defects in these children of microcephaly and Average rate of newborns with 0.7%
intracranial calcifications are particularly striking. The CNS CMV infection
lesions of two children with intracranial calcifications evalu- Number of newborns with 28,000
ated by computed tomographic (CT) and magnetic resonance CMV infection
imaging (MRI) scans are shown in Figure 11.8. If calcifica- Number with symptomatic disease 3,556
tions are present on neurodiagnostic evaluation, long-term (rate, 12.7%)
neurologic impairment is unequivocal. Children with head
Number with fatal disease (rate, 0.5%) 18
circumferences less than the fifth percentile at birth may subse-
quently establish a normal brain growth pattern (110). Ocular Number of survivors with sequelae 1,769
and hearing defects are common in children with symptomatic (rate, 50%)
congenital CMV infection. The principal abnormality of the Number of asymptomatic infections 24,444
eye associated with congenital CMV infection is retinitis, with (rate, 87.3%)
optic atrophy being relatively uncommon (110). Retinitis in Number with sequelae (rate, 13.5%) 3,300
the newborn with CMV infection will usually resolve sponta-
Total number with sequelae or death 5,087
neously over the first several months of life, leaving residual,
characteristic pigmented scars. From a systematic review of the
literature (54), 50% of the survivors develop late-appearing CMV, cytomegalovirus.
Adapted from Dollard SC, Grosse SD, Ross DS. New estimates of
complications, such as hearing loss, mental retardation, delay the prevalence of neurological and sensory sequelae and mortality
in psychomotor development, chorioretinitis, optic atrophy, associated with congenital cytomegalovirus infection. Rev Med Virol.
seizures, expressive language delays, learning disabilities, and 2007;17(5):355363.
defects of dentition (48,56,108,110115).

Scheld_Ch11.indd 174 2/21/14 5:34 PM


Chapter 11: Cytomegalovirus 175

Factors That Influence Disease


The type of maternal infectionthat is, primary or recurrent
has a direct impact on neurologic sequelae (Table 11.3). Any
neurologic impairment occurred in 25% of children of mothers
with primary infection, as compared to 8% among offspring
of women with recurrent disease. Importantly, the time to
documentation of neurologic impairment was also influenced
by type of infection (120), as shown in Figure 11.9. Among
women with primary infections, those occurring in the first tri-
mester had a greater association with both the incidence and
severity of sensorineural hearing loss and CNS sequelae (55).
However, damage was observed at all gestational ages, so the
timing of maternal infection cannot explain all of the variabil-
ity seen in clinical practice.
The quantity of CSF protein is a predictor of neurologic
outcome (121). Protein concentrations more than 120 mg/dL
were more commonly associated with microcephaly, abnormal
hearing, and multiple neurologic sequelae (121). Microcephaly
and an abnormality detected in the neonate by CT predict
future mental retardation and motor disability (122).

Asymptomatic Infection
Most newborns with congenital CMV infection will have no
symptomatology but are still at risk for neurologic and senso-
rineural impairment, which usually become apparent within
the first 2 years of life (118,123127). These observations
underscore the necessity for early diagnosis and intervention
in CMV infection.

FIGURE 11.8 Congenital cytomegalovirus infection. CT scans show


TA B L E 1 1 . 3
cortical atrophy and intracranial calcification. SEQUELAE IN CHILDREN WITH CONGENITAL
CYTOMEGALOVIRUS INFECTION ACCORDING
Of the 87.3% of infants with no clinical manifestations at TO TYPE OF MATERNAL INFECTION
birth, an average of 13.5% is at risk for the development of
Sequela Primarya Recurrenta p
similar abnormalities (54). The single most important late-
appearing sequela is sensorineural hearing loss (5% to 10% Sensorineural 15 (18/120) 5 (3/56) .05
of cases), which is bilateral in nearly half of the cases and hearing loss
substantial enough to interfere with learning and verbal com-
munication. The hearing impairment may become progressive Bilateral 8 (10/120) 0 (0/56) .02
only after the first year of life. hearing loss
Sensorineural deafness is probably the most common handicap Speech threshold 8 (9/120) 0 (0/56) .03
caused by congenital CMV infection, as first identified by Medearis 60 dBb
(114). CMV can replicate in the inner ear, as evidenced either by IQ 70 13 (9/68) 0 (0/32) .03
the cytopathology of Reissner membrane, stria vascularis, or semi- Chorioretinitisc 6 (7/112) 2 (1/54) .20
circular canals or by the presence of disease in the organ of Corti.
Hearing impairment in the child with symptomatic congenital Other neurologic 6 (8/125) 2 (1/64) .13
CMV infection can be progressive; it often becomes significant in sequelaed
infancy and early childhood but has been documented to occur in Microcephaly 5 (6/125) 2 (1/64) .25
some patients between 4 and 14 years of age (116). Congenital Seizures 5 (6/125) 0 (0/64) .08
CMV is second only to genetic defects in connexin proteins as a
Paresis or paralysis 1 (1/125) 0 (0/64) .66
known cause of sensorineural hearing loss (117).
Late-onset appearance of CMV retinitis has also been doc- Deathe 2 (3/125) 0 (0/64) .29
umented, although previously, CMV retinitis in the congeni- Any sequela 25 (31/125) 8 (5/64) .003
tally infected infant was not thought to relapse (118).
As techniques for neuroimaging improve, (119) isolated a
Percentages are given; numbers in parenthesis represent those with
lesions may be identified by cranial ultrasound or MRI which the sequela and the number evaluated.
b
were not part of the original case definitions of symptomatic For the ear with better hearing.
c
CMV infection given earlier. A current example is lenticulostri- Three of the seven children with retinitis (43%) in the primary
infection group had visual impairment.
ated vasculopathy, which some investigators consider a high d
Four of the eight children (50%) had more than one abnormality.
risk of future sensorineural hearing loss. There is a need for e
After the newborn period.
an internationally agreed case definition which takes account of From ref. 120.
changes in diagnostic practice.

Scheld_Ch11.indd 175 2/21/14 5:34 PM


176 Part II: Viral Infections and Related Disorders

100
Recurrent infection (n = 64)
Polyradiculopathy
90
Patients present with a subacute onset of leg weakness and
Children Free of Sequelae (%)

80 Primary infection (n = 125) numbness, leading to flaccid paraparesis. There may be pre-
ceding pain and paresthesias in the legs and perineum, and
70
areflexia with or without bladder dysfunction (131133).
60 CMV retinitis is also frequently found (131). A distinctive CSF
P = 0.02 feature is the florid polymorphonuclear pleocytosis and low
50 glucose concentration.
40

30 Stem Cell Transplant Patients


20 Encephalitis caused by CMV is emerging as a late complication
in patients with severe and protracted T-cell immunocompro-
10 mise due to receipt of cord blood donation, T-cell depletion,
and/or antithymocyte globulin (134). These patients have an
0 extensive history of treatment of recurrent episodes of CMV
0 6 12 18 24 30 36 42 48 54 60 66 72 78 viremia and often have strains of virus resistant to ganciclovir
Months Since Birth and/or foscarnet before encephalitis develops. The mortality
is high.
FIGURE 11.9 Percentage of children with congenital cytomegalovi-
rus infection who remained free of sequelae, according to the type
of maternal infection. The p value was obtained by the log rank test.
(From ref. 120.) Rasmussen Syndrome
There is some evidence that Rasmussen syndrome, with onset
Long-Term Follow-Up and Economic Impact in childhood following an infectious illness and characterized
by seizures and a progressive, unilateral cerebral atrophy and
Long-term follow-up is mandatory in children with congenital neurologic deficits, may in some cases be associated with CMV
CMV infection because of late-onset hearing and ocular dis- infection (135,136). However, detection of latent viral infec-
ease. The economic significance of this disease cannot be un- tion remains a possibility, and other investigators have not
derestimated, as reflected by a recent report from the Institute been able to confirm a pathogenic role for CMV (137,138).
of Medicine (128), which estimates that the present value of An uncontrolled case series of four cases reported benefit after
the annualized health care costs of caring for individuals dam- treatment with ganciclovir (139).
aged by CMV is $4 billion (mostly driven by congenital CMV).
Peripheral Nervous System Syndromes
Patients with Acquired For completeness, distinctive clinical syndromes of the periph-
Immunodeficiency Syndrome eral nervous system that have been associated with CMV in-
fection are described in the following sections.
With the exception of polyradiculopathy, CMV infection
of the CNS produces no unique clinical features. Diagnosis Distal Symmetric Peripheral Neuropathy
is often made at postmortem. Cell culture techniques were
used to test for CMV in brain tissue from 47 patients with Often termed painful peripheral neuropathy, distal symmetric
AIDS (13). A clinicopathologic association was seen between peripheral neuropathy has a subacute presentation, with pain
the detection of CMV in the CNS and an antemortem diagno- in the feet and clinical evidence of axonal atrophy (140). Some
sis of undiagnosed encephalopathy, consistent with, but not of the early syndromes of the peripheral nervous system may
proof of, an etiologic role for CMV. be the result of abnormal immune reaction.

Mononeuritis Multiplex
Encephalitis Patients with mononeuritis multiplex may present with sen-
sory and motor involvement of peripheral or cranial nerves.
CMV may cause encephalitis that is clinically indistinguishable Histologically, multiple peripheral nerves are involved in an
from HIV dementia and is often not diagnosed until autopsy inflammatory process in which infiltrating polymorphonuclear
(129). When 14 autopsy-confirmed cases were compared with leukocytes and necrotizing vasculitis are seen (141).
17 controls with HIV dementia, the median survival of the
cases was shorter and they were more likely to have CMV dis-
ease outside the CNS (129). Typically, patients have a subacute DIAGNOSIS
or chronic course, with cortical dysfunction leading to confu-
sion, lethargy, disorientation, and perhaps seizures. Brainstem The differential diagnosis of the clinical syndromes caused
lesions, if present, may produce focal signs. by CMV is complex and discussed elsewhere (140,142).
In six patients with necrotizing ventricular encephalitis, Electromyography, nerve conduction studies, sural nerve biop-
chart review revealed distinctive clinical associations with sies, and MRI are frequently employed.
cranial nerve defects, nystagmus, and progressive ventriculo- Evaluation of urine or saliva specimens from a child with
megaly detected by serial CT scans (130). This form of CMV suspected congenital infection should lead to prompt detec-
encephalitis is rapidly fatal. tion of CMV. Classically, this was done by cell culture but is

Scheld_Ch11.indd 176 2/21/14 5:34 PM


Chapter 11: Cytomegalovirus 177

usually performed nowadays by PCR, which has equivalent of CSF levels, ganciclovir and foscarnet do not penetrate the
sensitivity and specificity (143). Repeat samples of urine and CNS well, with estimates from animal experiments and small
saliva should then be tested to confirm the diagnosis, because studies in humans indicating that the CSF level is about one
a positive saliva test can occasionally represent infected breast third that found in plasma (155,156). Rather more patients
milk imbibed by the baby rather than true congenital infection. have been tested for foscarnet, whose CSF penetration appears
A blood sample should also be tested by PCR to detect viremia to be between one third and one half of plasma levels in most
which has a moderate association with the risk of developing patients (157,158). In addition to this pharmacologic disad-
subsequent sensorineural hearing loss (144). vantage, all drugs have toxicities, discussed in the following
Diagnosis of CMV CNS infection is best made by PCR. paragraphs. There are no reported double-blinded, placebo-
Several investigators have reported that PCR is a method of controlled trials of anti-CMV therapy in patients with AIDS
high sensitivity and specificity for detecting CNS involvement with CMV involvement of the CNS.
(145147). One exception to this is CSF samples collected at A case series examined the survival of two groups of AIDS
autopsy, of which 70% were found to be PCR positive for patients presenting with first-episode retinitis who had all been
CMV with no clear correlation with histologic findings (148). treated with an intraocular device that released ganciclovir.
This indicates that CSF samples must be collected during life. Some of the patients were given systemic ganciclovir treatment
Cytospin preparations of CSF from 25 patients with AIDS in addition and had reduced mortality (159).
have been studied and stained with monoclonal antibodies Five patients with CMV retinitis treated with maintenance
against pUL83 (pp65). Ten CSF samples were positive for levels of ganciclovir have been described in whom abrupt pre-
CMV with this technique, and it was reported that the poly- terminal changes in mental status occurred. At autopsy, fulmi-
morphonuclear leukocytes were hypersegmented, whether nant CMV encephalitis was found in all five, with prominent
or not they stained with the monoclonal antibody (149). ependymal and periventricular necrosis (160). These cases
The same technique has also been shown to detect CMV in demonstrate that prophylaxis with ganciclovir, in the doses
13 cases of painful peripheral neuropathy, although these cells used for maintenance of CMV retinitis, cannot guarantee pro-
are also found in cases of CMV retinitis (150). tection against CMV encephalitis. Furthermore, one patient
Conventional cell culture for the propagation of CMV is far has been described who developed polyradiculopathy attrib-
too insensitive to detect the small amounts of CMV that are uted to a ganciclovir-resistant strain of CMV after receiving
found in the CSF, so this test is not recommended. Serology, maintenance therapy for 8 months following a diagnosis of
either of IgG or IgM antibodies, has no diagnostic value in the CMV retinitis (161). Mathematical models of the replication
investigation of these patients. dynamics of CMV in vivo predicted that resistance should be
more common than was recognized clinically or by the use
of cell cultures for detecting CMV (162). This prediction has
TREATMENT been borne out by clinical cohort studies reporting that 15%
to 22% of transplant recipients or patients with AIDS given
Individual neonates with CMV disease have been given gan- long-term oral ganciclovir therapy develop CMV disease due
ciclovir, and inhibition of CMV has been reported. However, to resistant strains (163165).
ganciclovir has important potential toxicities in the neonate, Most patients given ganciclovir for polyradiculopathy do
including neutropenia. The drug is also carcinogenic in animals not have clinical improvement (135,166,167). Some, however,
and has a cytostatic effect on the testes. It requires intravenous appear to respond (132,135,168). Some authorities use a com-
administration, which may also produce significant morbidity. bination of ganciclovir plus foscarnet (each at full dose) but
To begin to study the potential safety and toxicity of ganciclovir again with disappointing results (169). There is no evidence for
in neonates, Whitley et al. (151) conducted a randomized trial synergy between ganciclovir and foscarnet in vivo, although
comparing two doses of ganciclovir (4 mg/kg or 6 mg/kg given these drugs can be used in combination (each at half the nor-
intravenously twice daily for 6 weeks). The higher dose was tol- mal dosage) to reduce the incidence of side effects (170).
erated as well as the lower dose, and so was taken forward into The complications of ganciclovir include neutropenia and
a randomized trial of efficacy (152). The primary end point was thrombocytopenia. The drug is poorly bioavailable and is usu-
the proportion of children at 6 months with improved brain- ally given by the intravenous route. Trials of high-dose oral
stem evoked potentials (or normal results if the baseline test ganciclovir showed that maintenance doses can be provided
result was normal). This primary end point was met in 69% orally (171). This had the advantage of freeing the patient
of children given ganciclovir compared with 39% allocated to from daily intravenous administrations, with the associated
no treatment. In addition, the proportion of children in whom risk of intravenous lines, but it did not overcome the problem
hearing worsened was 0% in the ganciclovir arm and 42% in of subtherapeutic doses at the target organ, which facilitates
the no-treatment arm. Follow-up shows that these significant the emergence of resistant strains of CMV. More recently, the
changes are still present at 12 months of age. The neurologic valine ester prodrug, valganciclovir, has been licensed, which
development of these children was worse in those not given provides the same area under the timeconcentration ganci-
ganciclovir (153). This benefit is so important clinically that clovir curve as does intravenous ganciclovir (172). It should
a 6-week course of ganciclovir should now be considered to provide a more convenient and safer means of treating CMV,
be the standard of care for any child born with symptomatic especially because intravenous lines may predispose to bacte-
congenital CMV infection who would have met the entry cri- remias with a drug that causes neutropenia, and should reduce
teria for the trial described by Kimberlin et al. (152). The same the incidence of resistance.
investigators showed that the prodrug valganciclovir could be Foscarnet is nephrotoxic and alters plasma levels of elec-
given to newborns (154). They are conducting a randomized trolytes, particularly ionized calcium, which may precipitate
controlled trial comparing 6 weeks versus 6 months of valgan- seizures. The drug also causes a fixed drug eruption on genital
ciclovir given to symptomatic neonates with congenital CMV skin. Although the incidence of nephrotoxicity can be reduced
infection, and the results are awaited with interest (http://www significantly by adequate hydration, this drug remains second-
.clinical trials.gov/show/NCT00466817). line therapy for the treatment of CMV disease. This is despite
Three drugs are used for the systemic treatment of CMV dis- the fact that a randomized comparison of ganciclovir and fos-
ease: ganciclovir, foscarnet, and cidofovir. Based on assessment carnet for the treatment and maintenance of CMV retinitis in

Scheld_Ch11.indd 177 2/21/14 5:34 PM


178 Part II: Viral Infections and Related Disorders

HIV-infected individuals showed the drugs to be equipotent been attempted. Oral ganciclovir has been reported to reduce
for treating retinitis but that foscarnet had a significant sur- the rate of CMV retinitis, whereas oral acyclovir in high doses
vival benefit over ganciclovir (173). There are several possible had no effect on CMV retinitis (186,187) despite its signifi-
explanations for this finding including the potential efficacy of cant effect in preventing CMV pneumonitis in bone marrow
foscarnet in treating HIV itself (174). transplant recipients (188,189). Oral valacyclovir significantly
Cidofovir is nephrotoxic and must not be administered if reduced CMV disease in patients with AIDS, although the dose
renal function is abnormal or if proteinuria is present. The chosen was poorly tolerated (190).
nephrotoxicity can be reduced by prehydration and adminis- Because CMV disease of the CNS often occurs in patients
tration of probenecid. with CMV retinitis, routine ophthalmoscopy to detect early
A lipid prodrug of cidofovir (CMX001) is currently under- cases of retinitis may be helpful (although this remains to be
going clinical trial (175). proved), whereas treatment modalities for CMV disease out-
Maribavir is a drug that directly inhibits CMV UL97. There side the CNS should include consideration of controlling CMV
was evidence of antiviral activity in a phase 2 study of bone CNS disease. For example, intraocular ganciclovir implants
marrow transplant patients where the need for preemptive suppress CMV retinitis, but these would not be expected to
therapy was the primary end point (176). However, there was have any effect on systemic CMV infection, including dissemi-
no significant benefit when the lowest dose was compared to nation to the brain. Indeed, a randomized trial showed that
placebo in a phase 3 study of bone marrow transplant patients addition of oral ganciclovir to an intraocular implant signifi-
managed by preemptive therapy but where the end point was cantly reduced the incidence of CMV retinitis in the contralat-
CMV disease (177). eral eye (191).
AiCuris 246 (Letermovir) inhibits the terminase enzyme Experimental work to produce CMV vaccines is under way.
complex of CMV (responsible for packaging unit length Glycoprotein B absorbs much of the neutralizing antibody from
DNA into virions) and is currently in clinical trial in transplant sera and is a promising target for a prototype CMV vaccine
patients (178). (192194). A phase 2 randomized, placebo-controlled study
of recombinant soluble glycoprotein B plus MF59 adjuvant
in seronegative women reported significant protection against
PREVENTION acquiring primary CMV infection (195). The same vaccine and
adjuvant reduced the viral load parameters after transplant of
Improved control of CMV disease of the CNS would be a kidney or a liver when patients were immunized while on the
expected if infection could be detected at an early stage and waiting list for transplantation (196). This finding is consistent
treatment instituted promptly. However, in the absence of with that described previously for the live attenuated Towne
natural history studies, it is not possible to design such trials strain of CMV, which reduced the severity of CMV disease
rationally. In my opinion, such trials should be organized so in those originally seronegative (197). In addition, seroposi-
that intervention studies, preferably prophylaxis of infection, tive transplant candidates were randomized to receive the same
can be designed. Such studies should also include quantitative vaccine and adjuvant or placebo with evidence of boosted an-
measures of CMV load to determine whether a threshold value tibody levels and improved control of viral load parameters
exists above which CMV disease becomes more likely, as has (196). Likewise, glycoprotein B plus MF59 adjuvant given to
been reported for renal, liver, and marrow transplant recipi- seropositive women boosted their neutralizing antibody levels
ents (179181). In transplant recipients, preemptive therapy and gB specific CD4 T cells (198).
is given when CMV viremia is detected by PCR or antigen- Because virtually all patients with AIDS are CMV seroposi-
emia (182,183), although its relative merits compared with tive, such an immunotherapeutic use of vaccine could be consid-
prophylaxis are debated (184). A trial of preemptive therapy ered for placebo-controlled trials in individuals with relatively
in patients with AIDS (ACTG protocol 5030) failed to recruit high CD4 cell counts, with the objective of delaying the onset of
sufficient cases because HAART became available (185). CMV disease. Finally, work is under way to devise techniques
Controlled trials of antiviral chemotherapy for the prophy- for adoptive transfer of immunocommitted T cells, as has been
laxis of CMV disease in individuals infected with HIV have described in bone marrow transplant recipients (199).

References
1. Ribbert H. ber protozoenartige Zellen in der Niere eines syphilitischen 9. Weller TH, Macauley JC, Craig JM, et al. Isolation of intranuclear inclu-
Neugeborenen und in der Parotis von Kindern. Zentralbl Allg Pathol. sion producing agents from infants with illnesses resembling cytomegalic
1904;15:945948. inclusion disease. Proc Soc Exp Biol Med. 1957;94(1):412.
2. Smith AJ, Weidman F. Infection of a still-born infant by an amebi- 10. Weller T, Hanshaw J, Scott DE. Serologic differentiation of viruses respon-
form protozon (Entamoeba mortinatalium, N.S.). Univ Pa Med Bull. sible for cytomegalic inclusion disease. Virology. 1960;12:130132.
1910;23:285298. 11. Bamborschke S, Wullen T, Huber M, et al. Early diagnosis and successful
3. Goodpasture E, Talbot F. Concerning the nature of protozoan-like cells treatment of acute cytomegalovirus encephalitis in a renal transplant recipi-
in certain lesions of infancy. Am J Dis Child. 1921;21:415425. ent. J Neurol. 1992;239(4):205208.
4. Tyzzer EE. The histology of the skin lesions in varicella. J Med Res. 12. Suzumiya J, Marutsuka K, Ueda S, et al. An autopsy case of necrotizing
1906;14(2):361392. ventriculo-encephalitis caused by cytomegalovirus in Hodgkins disease.
5. Von Glahn WC, Pappenheimer AM. Intranuclear inclusions in visceral dis- Acta Pathol Jpn. 1991;41(4):291298.
ease. Am J Pathol. 1925;1(5):445466. 13. Pillay D, Lipman MC, Lee CA, et al. A clinico-pathological audit of
6. Cole R, Kuttner AG. A filterable virus present in the submaxillary glands of opportunistic viral infections in HIV-infected patients. AIDS. 1993;7:
guinea pigs. J Exp Med. 1926;44(6):855873. 969974.
7. Smith MG. Propagation in tissue cultures of a cytopathogenic virus 14. Belec L, Tayot J, Tron P, et al. Cytomegalovirus encephalopathy in an infant
from human salivary gland virus (SGV) disease. Proc Soc Exp Biol Med. with congenital acquired immuno-deficiency syndrome. Neuropediatrics.
1956;92(2):424430. 1990;21(3):124129.
8. Rowe WP, Hartley JW, Waterman S, et al. Cytopathogenic agent resem- 15. Chee MS, Bankier AT, Beck S, et al. Analysis of the protein-coding con-
bling human salivary gland virus recovered from tissue cultures of human tent of the sequence of human cytomegalovirus strain AD169. Curr Top
adenoids. Proc Soc Exp Biol Med. 1956;92(2):418424. Microbiol Immunol. 1990;154:125169.

Scheld_Ch11.indd 178 2/21/14 5:34 PM


Chapter 11: Cytomegalovirus 179

16. Prichard MN, Penfold ME, Duke GM, et al. A review of genetic differences 46. Monif GR, Egan EA, Held B, et al. The correlation of maternal cytomega-
between limited and extensively passaged human cytomegalovirus strains. lovirus infection during varying stages in gestation with neonatal involve-
Rev Med Virol. 2001;11(3):191200. ment. J Pediatr. 1972;80(1):1720.
17. Dolan A, Cunningham C, Hector RD, et al. Genetic content of wild-type 47. Nankervis GA, Kumar ML, Cox FE, et al. A prospective study of maternal
human cytomegalovirus. J Gen Virol. 2004;85(pt 5):13011312. cytomegalovirus infection and its effect on the fetus. Am J Obstet Gynecol.
18. Stern-Ginossar N, Weisburd B, Michalski A, et al. Decoding human cyto- 1984;149(4):435440.
megalovirus. Science. 2012;6110:10881093. 48. Preece PM, Pearl KN, Peckham CS. Congenital cytomegalovirus infection.
19. Mocarski ES Jr. Cytomegalovirus Biology and Replication. Philadelphia: Arch Dis Child. 1984;59(12):11201126.
Lipincott Williams & Wilkins; 2001. 49. Stagno S, Pass RF, Dworsky ME, et al. Congenital cytomegalovirus infec-
20. Landini MP, Sapete R. Human cytomegalovirus structural proteins: a tion: the relative importance of primary and recurrent maternal infection.
report of the first nomenclature workshop. In: Michelson S, Plotkin S, eds. N Engl J Med. 1982;306(16):945949.
Multidisciplinary Approach to Understanding Cytomegalovirus Diseases: 50. Stern H, Tucker SM. Prospective study of cytomegalovirus infection in
Proceedings of the Fourth International Cytomegalovirus Workshop. Paris: pregnancy. Br Med J. 1973;2(5861):268270.
Elsevier; 1993:6574. 51. Griffiths PD, Baboonian C. A prospective study of primary cytomegalovi-
21. Pari GS, Anders DG. Eleven loci encoding trans-acting factors are required rus infection during pregnancy: final report. Br J Obstet Gynaecol. 1984;
for transient complementation of human cytomegalovirus oriLyt-dependent 91(4):307315.
DNA replication. J Virol. 1993;67(12):69796988. 52. Hyde TB, Schmid DS, Cannon MJ. Cytomegalovirus seroconversion
22. Anders DG, Kacica MA, Pari G, et al. Boundaries and structure of human rates and risk factors: implications for congenital CMV. Rev Med Virol.
cytomegalovirus oriLyt, a complex origin for lytic-phase DNA replication. 2010;20(5):311326.
J Virol. 1992;66(6):33733384. 53. Kenneson A, Cannon MJ. Review and meta-analysis of the epidemiology of
23. Grefte A, Blom N, van der Giessen M, et al. Ultrastructural analysis of congenital cytomegalovirus (CMV) infection. Rev Med Virol. 2007;17(4):
circulating cytomegalic cells in patients with active cytomegalovirus infec- 253276.
tion: evidence for virus production and endothelial origin. J Infect Dis. 54. Dollard SC, Grosse SD, Ross DS. New estimates of the prevalence of neu-
1993;168(5):11101118. rological and sensory sequelae and mortality associated with congenital
24. Sylwester AW, Mitchell BL, Edgar JB, et al. Broadly targeted human cyto- cytomegalovirus infection. Rev Med Virol. 2007;17(5):355363.
megalovirus-specific CD4 and CD8 T cells dominate the memory com- 55. Pass RF, Fowler KB, Boppana SB, et al. Congenital cytomegalovirus infec-
partments of exposed subjects. J Exp Med. 2005;202(5):673685. tion following first trimester maternal infection: symptoms at birth and
25. Revello MG, Gerna G. Human cytomegalovirus tropism for endothelial/ outcome. J Clin Virol. 2006;35(2):216220.
epithelial cells: scientific background and clinical implications. Rev Med 56. Stagno S, Pass RF, Dworsky ME, et al. Congenital and perinatal cytomega-
Virol. 2010;20(3):136155. lovirus infections. Semin Perinatol. 1983;7(1):3142.
26. Littler E, Stuart AD, Chee MS. Human cytomegalovirus UL97 open read- 57. Griffiths PD, Baboonian C. Intra-uterine transmission of cytomegalovirus
ing frame encodes a protein that phosphorylates the antiviral nucleoside in women known to be immune before conception. J Hyg (Lond). 1984;
analogue ganciclovir. Nature. 1992;358(6382):160162. 92(1):8995.
27. Sullivan V, Talarico CL, Stanat SC, et al. A protein kinase homologue con- 58. Kamada M, Komori A, Chiba S, et al. A prospective study of congeni-
trols phosphorylation of ganciclovir in human cytomegalovirus-infected tal cytomegalovirus infection in Japan. Scand J Infect Dis. 1983;15(3):
cells. Nature. 1992;358(6382):162164. 227232.
28. Talarico CL, Burnette TC, Miller WH, et al. Acyclovir is phosphorylated by 59. Schopfer K, Lauber E, Krech U. Congenital cytomegalovirus infection in
the human cytomegalovirus UL97 protein. Antimicrob Agents Chemother. newborn infants of mothers infected before pregnancy. Arch Dis Child.
1999;43(8):19411946. 1978;53(7):536539.
29. Weller TH. The cytomegaloviruses: ubiquitous agents with protean clinical 60. Stagno S, Reynolds DW, Huang ES, et al. Congenital cytomegalovirus
manifestations. II. N Engl J Med. 1971;285(5):267274. infection. N Engl J Med. 1977;296(22):12541258.
30. Cannon MJ, Schmid DS, Hyde TB. Review of cytomegalovirus seropreva- 61. Huang ES, Alford CA, Reynolds DW, et al. Molecular epidemiology of
lence and demographic characteristics associated with infection. Rev Med cytomegalovirus infections in women and their infants. N Engl J Med.
Virol. 2010;20(4):202213. 1980;303(17):958962.
31. Taylor-Wiedeman J, Sissons JG, Borysiewicz LK, et al. Monocytes are a 62. Boppana SB, Rivera LB, Fowler KB, et al. Intrauterine transmission of
major site of persistence of human cytomegalovirus in peripheral blood cytomegalovirus to infants of women with preconceptional immunity.
mononuclear cells. J Gen Virol. 1991;72(pt 9):20592064. N Engl J Med. 2001;344(18):13661371.
32. Goodrum FD, Jordan CT, High K, et al. Human cytomegalovirus gene 63. Yamamoto AY, Mussi-Pinhata MM, Boppana SB, et al. Human cytomega-
expression during infection of primary hematopoietic progenitor cells: a lovirus reinfection is associated with intrauterine transmission in a highly
model for latency. Proc Natl Acad Sci U S A. 2002;99(25):1625516260. cytomegalovirus-immune maternal population. Am J Obstet Gynecol.
33. Reeves MB, MacAry PA, Lehner PJ, et al. Latency, chromatin remodeling, 2010;202(3):297298.
and reactivation of human cytomegalovirus in the dendritic cells of healthy 64. Wang C, Zhang X, Bialek S, et al. Attribution of congenital cytomegalovi-
carriers. Proc Natl Acad Sci U S A. 2005;102(11):41404145. rus infection to primary versus non-primary maternal infection. Clin Infect
34. Hayes K, Danks DM, Gibas H, et al. Cytomegalovirus in human milk. Dis. 2011;52(2):e11e13.
N Engl J Med. 1972;287(4):177178. 65. Boppana SB, Fowler KB, Britt WJ, et al. Symptomatic congenital cytomega-
35. Lang DJ, Kummer JF. Cytomegalovirus in semen: observations in selected lovirus infection in infants born to mothers with preexisting immunity to
populations. J Infect Dis. 1975;132(4):472473. cytomegalovirus. Pediatrics. 1999;104(1, pt 1):5560.
36. Stagno S, Reynolds DW, Pass RF, et al. Breast milk and the risk of cyto- 66. Reynolds DW, Stagno S, Hosty TS, et al. Maternal cytomegalovirus excre-
megalovirus infection. N Engl J Med. 1980;302(19):10731076. tion and perinatal infection. N Engl J Med. 1973;289(1):15.
37. Adler SP, Wilson M, Lawrence LT. Cytomegalovirus transmission among 67. Stagno S, Brasfield DM, Brown MB, et al. Infant pneumonitis associated
children attending a day care center. Pediatr Res. 1985;19:285A. with cytomegalovirus, Chlamydia, Pneumocystis, and Ureaplasma: a pro-
38. Murph JR, Bale JF, Perlman S. The prevalence of cytomegalovirus infection spective study. Pediatrics. 1981;68(3):322329.
in a midwest day care center. Pediatr Res. 1985;19:205A. 68. Whitley RJ, Brasfield D, Reynolds DW, et al. Protracted pneumonitis in
39. Pass RF, Hutto C, Ricks R, Cloud GA. Increased rate of cytomegalovirus young infants associated with perinatally acquired cytomegaloviral infec-
infection among parents of children attending day-care centers. N Engl J tion. J Pediatr. 1976;89(1):1622.
Med. 1986;314(22):14141418. 69. Faix RG. Survival of cytomegalovirus on environmental surfaces. J Pediatr.
40. Dworsky M, Lakeman A, Stagno S. Cytomegalovirus transmission within a 1985;106(4):649652.
family. Pediatr Infect Dis. 1984;3:236238. 70. Hutto C, Little EA, Ricks R, et al. Isolation of cytomegalovirus from toys
41. Spector SA, Spector DH. Molecular epidemiology of cytomegalovirus and hands in a day care center. J Infect Dis. 1986;154(3):527530.
infections in premature twin infants and their mother. Pediatr Infect Dis. 71. Adler SP. Molecular epidemiology of cytomegalovirus: evidence for viral
1982;1(6):405409. transmission to parents from children infected at a day care center. Pediatr
42. Staras SA, Flanders WD, Dollard SC, et al. Influence of sexual activity Infect Dis. 1986;5(3):315318.
on cytomegalovirus seroprevalence in the United States, 19881994. Sex 72. Adler SP. Molecular epidemiology of cytomegalovirus: viral transmission
Transm Dis. 2008;35(5):472479. among children attending a day care center, their parents, and caretakers.
43. Ahlfors K, Forsgren M, Ivarsson SA, et al. Congenital cytomegalovirus J Pediatr. 1988;112(3):366372.
infection: on the relation between type and time of maternal infection and 73. Centers for Disease Control and Prevention. Prevalence of cytomegalovirus
infants symptoms. Scand J Infect Dis. 1983;15(2):129138. excretion from children in five day-care centersAlabama. MMWR Morb
44. Ahlfors K, Ivarsson SA, Harris S, et al. Congenital cytomegalovirus infec- Mortal Wkly Rep. 1985;34(4):4951.
tion and disease in Sweden and the relative importance of primary and sec- 74. Wertheim P, Buurman C, Geelen J, et al. Transmission of cytomegalovi-
ondary maternal infections. Preliminary findings from a prospective study. rus by renal allograft demonstrated by restriction enzyme analysis. Lancet.
Scand J Infect Dis. 1984;16(2):129137. 1983;1(8331):980981.
45. Grant S, Edmond E, Syme J. A prospective study of cytomegalovirus infec- 75. Grundy JE, Lui SF, Super M, et al. Symptomatic cytomegalovirus infection
tion in pregnancy. I. Laboratory evidence of congenital infection following in seropositive kidney recipients: reinfection with donor virus rather than
maternal primary and reactivated infection. J Infect. 1981;3(1):2431. reactivation of recipient virus. Lancet. 1988;2(8603):132135.

Scheld_Ch11.indd 179 2/21/14 5:34 PM


180 Part II: Viral Infections and Related Disorders

76. Emery VC, Hassan-Walker AF, Burroughs AK, et al. Human cytomega- 105. Artigas J, Gross G, Niedobitek F. Neuropathology of AIDS. In: Artigas
lovirus (HCMV) replication dynamics in HCMV-naive and -experienced J, Gross G, Niedobitek F, eds. The Central Nervous System in AIDS:
immunocompromised hosts. J Infect Dis. 2002;185(12):17231728. Neurology Radiology Pathology Ophthalmology. Berlin: Springer-Verlag;
77. Roback JD. CMV and blood transfusions. Rev Med Virol. 2002;12(4): 1993:101110.
211219. 106. Chimelli L, Rosemberg S, Hahn MD, et al. Pathology of the central ner-
78. Gallant JE, Moore RD, Richman DD, et al. Incidence and natural his- vous system in patients infected with the human immunodeficiency virus
tory of cytomegalovirus disease in patients with advanced human im- (HIV): a report of 252 autopsy cases from Brazil. Neuropathol Appl
munodeficiency virus disease treated with zidovudine. The Zidovudine Neurobiol. 1992;18(5):478488.
Epidemiology Study Group. J Infect Dis. 1992;166(6):12231227. 107. Gray F, Belec L, Lescs MC, et al. Varicella-zoster virus infection of the cen-
79. Bylsma SS, Achim CL, Wiley CA, et al. The predictive value of cytomega- tral nervous system in the acquired immune deficiency syndrome. Brain.
lovirus retinitis for cytomegalovirus encephalitis in acquired immunodefi- 1994;117(pt 5):987999.
ciency syndrome. Arch Ophthalmol. 1995;113(1):8995. 108. Weller T, Hanshaw J. Virologic and clinical observations on cytomegalic
80. Shaw GM, Harper ME, Hahn BH, et al. HTLV-III infection in brains of inclusion disease. N Engl J Med. 1962;266:12331244.
children and adults with AIDS encephalopathy. Science. 1985;227(4683): 109. Mena W, Royal S, Pass RF, et al. Diabetes insipidus associated with symptom-
177182. atic congenital cytomegalovirus infection. J Pediatr. 1993;122(6):911913.
81. Budka H. The definition of HIV-specific neuropathology. Acta Pathol Jpn. 110. Hanshaw JB. Congenital cytomegalovirus infection: a fifteen-year per-
1991;41(3):182191. spective. J Infect Dis. 1971;123(5):555561.
82. Griffiths PD. Studies to define viral cofactors for human immunodefi- 111. McCracken GH Jr, Shinefield HM, Cobb K, et al. Congenital cytomegalic
ciency virus. Infect Agents Dis. 1992;1(5):237244. inclusion disease. A longitudinal study of 20 patients. Am J Dis Child.
83. Koval V, Clark C, Vaishnav M, et al. Human cytomegalovirus inhibits 1969;117(5):522539.
human immunodeficiency virus replication in cells productively infected 112. Pass RF, Stagno S, Myers GJ, et al. Outcome of symptomatic congenital
by both viruses. J Virol. 1991;65(12):69696978. cytomegalovirus infection: results of long-term longitudinal follow-up.
84. Pepose JS, Hilborne LH, Cancilla PA, et al. Concurrent herpes simplex Pediatrics. 1980;66(5):758762.
and cytomegalovirus retinitis and encephalitis in the acquired immune 113. Berenberg W, Nankervis G. Long-term follow-up of cytomegalic inclusion
deficiency syndrome (AIDS). Ophthalmology. 1984;91(12):16691677. disease of infancy. Pediatrics. 1970;46(3):403410.
85. Vinters HV, Kwok MK, Ho HW, et al. Cytomegalovirus in the nervous 114. Medearis DJ. Observations concerning human cytomegalovirus infection
system of patients with the acquired immune deficiency syndrome. Brain. and disease. Bull Johns Hopkins Hosp. 1964;114:181211.
1989;112(pt 1):245268. 115. Williamson WD, Desmond MM, LaFevers N, et al. Symptomatic congeni-
86. Yanagisawa N, Toyokura Y, Shiraki H. Double encephalitis with herpes tal cytomegalovirus. Disorders of language, learning, and hearing. Am J
simplex virus and cytomegalovirus in an adult. Acta Neuropathol. Dis Child. 1982;136(10):902905.
1975;33(2):153164. 116. Stagno S, Whitley RJ. Herpesvirus infection in the neonate and children.
87. Rousseau F, Perronne C, Deny P, et al. Prolonged and fatal disseminated In: Stamms W, Holmes W, Mardh P, eds. Sexually Transmitted Diseases.
cytomegalovirus infection in an immunocompetent woman. Eur J Clin New York: McGraw-Hill; 1990:863.
Microbiol Infect Dis. 1993;12:938940. 117. Morton CC, Nance WE. Newborn hearing screeninga silent revolution.
88. Studahl M, Ricksten A, Sandberg T, et al. Cytomegalovirus infection of N Engl J Med. 2006;354(20):21512164.
the CNS in non-compromised patients. Acta Neurol Scand. 1994;89(6): 118. Boppana S, Amos C, Britt W, et al. Late onset and reactivation of cho-
451457. rioretinitis in children with congenital cytomegalovirus infection. Pediatr
89. Tobita M, Komiyama A, Naito M, et al. Cytomegalovirus ascending Infect Dis J. 1994;13(12):11391142.
myelitis in an immunocompetent adult [in Japanese]. Rinsho Shinkeigaku. 119. de Vries LS, Gunardi H, Barth PG, et al. The spectrum of cranial ultra-
1993;33(8):915917. sound and magnetic resonance imaging abnormalities in congenital cyto-
90. van den Pol AN, Reuter JD, Santarelli JG. Enhanced cytomegalovirus megalovirus infection. Neuropediatrics. 2004;35(2):113119.
infection of developing brain independent of the adaptive immune system. 120. Fowler KB, Stagno S, Pass RF, et al. The outcome of congenital cytomega-
J Virol. 2002;76(17):88428854. lovirus infection in relation to maternal antibody status. N Engl J Med.
91. Teissier N, Delezoide AL, Mas AE, et al. Inner ear lesions in congenital 1992;326(10):663667.
cytomegalovirus infection of human fetuses. Acta Neuropathol. 2011; 121. Boppana SB, Pass RF, Britt WJ, et al. Symptomatic congenital cytomega-
122(6):763774. lovirus infection: neonatal morbidity and mortality. Pediatr Infect Dis J.
92. Pereira L, Stagno S, Hoffman M, et al. Cytomegalovirus-infected cell 1992;11(2):9399.
polypeptides immune-precipitated by sera from children with congenital 122. Noyola DE, Demmler GJ, Nelson CT, et al. Early predictors of neurode-
and perinatal infections. Infect Immun. 1983;39(1):100108. velopmental outcome in symptomatic congenital cytomegalovirus infec-
93. Stagno S, Volanakis JE, Reynolds DW, et al. Immune complexes in congen- tion. J Pediatr. 2001;138(3):325331.
ital and natal cytomegalovirus infections of man. J Clin Invest. 1977;60 123. Melish ME, Hanshaw J. Congenital cytomegalovirus infection.
(4):838845. Developmental progress of infants detected by routine screening. Am J
94. Grundy JE, Shanley JD, Griffiths PD. Is cytomegalovirus interstitial Dis Child. 1973;126(2):190194.
pneumonitis in transplant recipients an immunopathological condition? 124. Reynolds DW, Stagno S, Stubbs KG, et al. Inapparent congenital cyto-
Lancet. 1987;2(8566):996999. megalovirus infection with elevated cord IgM levels. Casual relation with
95. Morgello S, Simpson DM. Multifocal cytomegalovirus demyelinative poly- auditory and mental deficiency. N Engl J Med. 1974;290(6):291296.
neuropathy associated with AIDS. Muscle Nerve. 1994;17(2):176182. 125. Saigal S, Lunyk O, Larke RP, et al. The outcome in children with congeni-
96. Vitkovic L, Kalebic T, de Cunha A, et al. Astrocyte-conditioned medium tal cytomegalovirus infection. A longitudinal follow-up study. Am J Dis
stimulates HIV-1 expression in a chronically infected promonocyte clone. Child. 1982;136(10):896901.
J Neuroimmunol. 1990;30(23):153160. 126. Stagno S, Reynolds DW, Amos CS, et al. Auditory and visual defects
97. Murayama T, Kuno K, Jisaki F, et al. Enhancement human cytomega- resulting from symptomatic and subclinical congenital cytomegaloviral
lovirus replication in a human lung fibroblast cell line by interleukin-8. and toxoplasma infections. Pediatrics. 1977;59(5):669678.
J Virol. 1994;68(11):75827585. 127. Kumar ML, Nankervis GA, Gold E. Inapparent congenital cytomegalovirus
98. Spector SA, Merrill R, Wolf D, et al. Detection of human cytomegalovirus infection. A follow-up study. N Engl J Med. 1973;288(26):13701372.
in plasma of AIDS patients during acute visceral disease by DNA amplifi- 128. Stratton KR, Durch JS, Lawrence RS. Vaccines for the 21st century
cation. J Clin Microbiol. 1992;30(9):23592365. (2001). Washington, DC: National Academy Press; 2000.
99. Graeber MB, Streit WJ, Kreutzberg GW. Identity of ED2-positive perivas- 129. Holland NR, Power C, Mathews VP, et al. Cytomegalovirus encephali-
cular cells in rat brain. J Neurosci Res. 1989;22(1):103106. tis in acquired immunodeficiency syndrome (AIDS). Neurology. 1994;44
100. Hickey WF, Kimura H. Perivascular microglial cells of the CNS are (3, pt 1):507514.
bone marrow-derived and present antigen in vivo. Science. 1988;239 130. Kalayjian RC, Cohen ML, Bonomo RA, et al. Cytomegalovirus ventricu-
(4837):290292. loencephalitis in AIDS: a syndrome with distinct clinical and pathologic
101. Morgello S, Cho ES, Nielsen S, et al. Cytomegalovirus encephalitis in features. Medicine (Baltimore). 1993;72(2):6777.
patients with acquired immunodeficiency syndrome: an autopsy study of 131. Cohen BA, McArthur JC, Grohman S, et al. Neurologic prognosis of
30 cases and a review of the literature. Hum Pathol. 1987;18(3):289297. cytomegalovirus polyradiculomyelopathy in AIDS. Neurology. 1993;43
102. Wiley CA, Schrier RD, Denaro FJ, et al. Localization of cytomegalovirus (3, pt 1):493499.
proteins and genome during fulminant central nervous system infection in 132. de Gans J, Portegies P, Tiessens G, et al. Therapy for cytomegalovirus
an AIDS patient. J Neuropathol Exp Neurol. 1986;45(2):127139. polyradiculomyelitis in patients with AIDS: treatment with ganciclovir.
103. Klatt EC, Shibata D. Cytomegalovirus infection in the acquired immu- AIDS. 1990;4(5):421425.
nodeficiency syndrome. Clinical and autopsy findings. Arch Pathol Lab 133. Miller RG, Storey JR, Greco CM. Ganciclovir in the treatment of progres-
Med. 1988;112(5):540544. sive AIDS-related polyradiculopathy. Neurology. 1990;40(4):569574.
104. McKenzie R, Travis WD, Dolan SA, et al. The causes of death in patients 134. Reddy KS, Devarapalli Y, Mazur M, et al. Alemtuzumab with rapid ste-
with human immunodeficiency virus infection: a clinical and patho- roid taper in simultaneous kidney and pancreas transplantation: com-
logic study with emphasis on the role of pulmonary diseases. Medicine parison to induction with antithymocyte globulin. Transplant Proc.
(Baltimore). 1991;70(5):326343. 2010;42(6):20062008.

Scheld_Ch11.indd 180 2/21/14 5:34 PM


Chapter 11: Cytomegalovirus 181

135. Jay V, Becker LE. Surgical pathology of epilepsy: a review. Pediatr Pathol. 164. Hu H, Jabs DA, Forman MS, et al. Comparison of cytomegalovirus
1994;14(4):731750. (CMV) UL97 gene sequences in the blood and vitreous of patients with
136. Power C, Poland SD, Blume WT, et al. Cytomegalovirus and Rasmussens acquired immunodeficiency syndrome and CMV retinitis. J Infect Dis.
encephalitis. Lancet. 1990;336(8726):12821284. 2002;185(7):861867.
137. Farrell MA, Cheng L, Cornford ME, et al. Cytomegalovirus and 165. Limaye AP, Corey L, Koelle DM, et al. Emergence of ganciclovir-resistant
Rasmussens encephalitis. Lancet. 1991;337(8756):15511552. cytomegalovirus disease among recipients of solid-organ transplants.
138. Vinters HV, Wang R, Wiley CA. Herpesviruses in chronic encephalitis Lancet. 2000;356(9230):645649.
associated with intractable childhood epilepsy. Hum Pathol. 1993;24(8): 166. Jacobson MA, Mills J, Rush J, et al. Failure of antiviral therapy for
871879. acquired immunodeficiency syndrome-related cytomegalovirus myelitis.
139. McLachlan RS, Levin S, Blume WT. Treatment of Rasmussens syndrome Arch Neurol. 1988;45(10):10901092.
with ganciclovir. Neurology. 1996;47(4):925928. 167. Jeantils V, Lemaitre MO, Robert J, et al. Subacute polyneuropathy with
140. Fuller GN. Cytomegalovirus and the peripheral nervous system in AIDS. encephalopathy in AIDS with human cytomegalovirus pathogenicity?
J Acquir Immune Defic Syndr. 1992;5(suppl 1):S33S36. Lancet. 1986;2(8514):1039.
141. Said G, Lacroix C, Chemouilli P, et al. Cytomegalovirus neuropathy in 168. Graveleau P, Perol R, Chapman A. Regression of cauda equina syndrome in
acquired immunodeficiency syndrome: a clinical and pathological study. AIDS patient being treated with ganciclovir. Lancet. 1989;2(8661):511512.
Ann Neurol. 1991;29(2):139146. 169. Anduze-Faris BM, Fillet AM, Gozlan J, et al. Induction and maintenance
142. Simpson DM, Olney RK. Peripheral neuropathies associated with human therapy of cytomegalovirus central nervous system infection in HIV-
immunodeficiency virus infection. Neurol Clin. 1992;10(3):685711. infected patients. AIDS. 2000;14(5):517524.
143. Boppana SB, Ross SA, Shimamura M, et al. Saliva polymerase-chain- 170. Mattes FM, Hainsworth EG, Geretti AM, et al. A randomized, controlled
reaction assay for cytomegalovirus screening in newborns. N Engl J Med. trial comparing ganciclovir to ganciclovir plus foscarnet (each at half
2011;364(22):21112118. dose) for preemptive therapy of cytomegalovirus infection in transplant
144. Bradford RD, Cloud G, Lakeman AD, et al. Detection of cytomegalovirus recipients. J Infect Dis. 2004;189(8):13551361.
(CMV) DNA by polymerase chain reaction is associated with hearing loss 171. Spector SA, McKinley GF, Lalezari JP, et al. Oral ganciclovir for the preven-
in newborns with symptomatic congenital CMV infection involving the tion of cytomegalovirus disease in persons with AIDS. Roche Cooperative
central nervous system. J Infect Dis. 2005;191(2):227233. Oral Ganciclovir Study Group. N Engl J Med. 1996;334(23):14911497.
145. Cinque P, Vago L, Brytting M, et al. Cytomegalovirus infection of the 172. Martin DF, Sierra-Madero J, Walmsley S, et al. A controlled trial of val-
central nervous system in patients with AIDS: diagnosis by DNA amplifi- ganciclovir as induction therapy for cytomegalovirus retinitis. N Engl J
cation from cerebrospinal fluid. J Infect Dis. 1992;166(6):14081411. Med. 2002;346(15):11191126.
146. Clifford DB, Buller RS, Mohammed S, et al. Use of polymerase chain reac- 173. Mortality in patients with the acquired immunodeficiency syndrome treated
tion to demonstrate cytomegalovirus DNA in CSF of patients with human with either foscarnet or ganciclovir for cytomegalovirus retinitis. Studies of
immunodeficiency virus infection. Neurology. 1993;43(1):7579. Ocular Complications of AIDS Research Group, in collaboration with the
147. Wolf DG, Spector SA. Diagnosis of human cytomegalovirus central ner- AIDS Clinical Trials Group. N Engl J Med. 1992;326(4):213220.
vous system disease in AIDS patients by DNA amplification from cerebro- 174. Seeing the way forward for treatment of CMV retinitis. Lancet. 1991;
spinal fluid. J Infect Dis. 1992;166(6):14121415. 338(8781):14941495.
148. Achim CL, Nagra RM, Wang R, et al. Detection of cytomegalovirus in 175. Painter W, Robertson A, Trost LC, et al. First pharmacokinetic and
cerebrospinal fluid autopsy specimens from AIDS patients. J Infect Dis. safety study in humans of the novel lipid antiviral conjugate CMX001,
1994;169(3):623627. a broad-spectrum oral drug active against double-stranded DNA viruses.
149. Revello MG, Percivalle E, Sarasini A, et al. Diagnosis of human cytomega- Antimicrob Agents Chemother. 2012;56(5):27262734.
lovirus infection of the nervous system by pp65 detection in polymorpho- 176. Winston DJ, Young JA, Pullarkat V, et al. Maribavir prophylaxis for pre-
nuclear leukocytes of cerebrospinal fluid from AIDS patients. J Infect Dis. vention of cytomegalovirus infection in allogeneic stem-cell transplant
1994;170(5):12751279. recipients: a multicenter, randomized, double-blind, placebo-controlled,
150. Mastroianni CM, Sebastiani G, Folgori F, et al. Detection of cytomega- dose-ranging study. Blood. 2008;111(11):54035410.
lovirus-matrix protein (pp65) in leukocytes of HIV-infected patients with 177. Marty FM, Ljungman P, Papanicolaou GA, et al. Maribavir prophylaxis
painful peripheral neuropathy. J Med Virol. 1994;44(2):172175. for prevention of cytomegalovirus disease in recipients of allogeneic stem-
151. Whitley RJ, Cloud G, Gruber W, et al. Ganciclovir treatment of symp- cell transplants: a phase 3, double-blind, placebo-controlled, randomised
tomatic congenital cytomegalovirus infection: results of a phase II study. trial. Lancet Infect Dis. 2011;11(4):284292.
National Institute of Allergy and Infectious Diseases Collaborative 178. Marschall M, Stamminger T, Urban A, et al. In vitro evaluation of the ac-
Antiviral Study Group. J Infect Dis. 1997;175(5):10801086. tivities of the novel anticytomegalovirus compound AIC246 (letermovir)
152. Kimberlin DW, Lin CY, Sanchez PJ, et al. Effect of ganciclovir therapy against herpesviruses and other human pathogenic viruses. Antimicrob
on hearing in symptomatic congenital cytomegalovirus disease involv- Agents Chemother. 2012;56(2):11351137.
ing the central nervous system: a randomized, controlled trial. J Pediatr. 179. Cope AV, Sabin C, Burroughs A, et al. Interrelationships among quantity of
2003;143(1):1625. human cytomegalovirus (HCMV) DNA in blood, donor-recipient serostatus,
153. Oliver SE, Cloud GA, Sanchez PJ, et al. Neurodevelopmental outcomes and administration of methylprednisolone as risk factors for HCMV disease
following ganciclovir therapy in symptomatic congenital cytomega- following liver transplantation. J Infect Dis. 1997;176(6):14841490.
lovirus infections involving the central nervous system. J Clin Virol. 180. Cope AV, Sweny P, Sabin C, et al. Quantity of cytomegalovirus viruria is a
2009;46(suppl 4):S22S26. major risk factor for cytomegalovirus disease after renal transplantation.
154. Kimberlin DW, Acosta EP, Sanchez PJ, et al. Pharmacokinetic and pharma- J Med Virol. 1997;52(2):200205.
codynamic assessment of oral valganciclovir in the treatment of symptom- 181. Gor D, Sabin C, Prentice HG, et al. Longitudinal fluctuations in cytomeg-
atic congenital cytomegalovirus disease. J Infect Dis. 2008;197(6):836845. alovirus load in bone marrow transplant patients: relationship between
155. Brewster ME, Raghavan K, Pop E, et al. Enhanced delivery of ganciclo- peak virus load, donor/recipient serostatus, acute GVHD and CMV dis-
vir to the brain through the use of redox targeting. Antimicrob Agents ease. Bone Marrow Transplant. 1998;21(6):597605.
Chemother. 1994;38(4):817823. 182. Einsele H, Ehninger G, Hebart H, et al. Polymerase chain reaction moni-
156. Fletcher C, Sawchuk R, Chinnock B, et al. Human pharmacokinetics of toring reduces the incidence of cytomegalovirus disease and the duration
the antiviral drug DHPG. Clin Pharmacol Ther. 1986;40(3):281286. and side effects of antiviral therapy after bone marrow transplantation.
157. Raffi F, Taburet AM, Ghaleh B, et al. Penetration of foscarnet into cere- Blood. 1995;86(7):28152820.
brospinal fluid of AIDS patients. Antimicrob Agents Chemother. 1993;37 183. Paya CV, Wilson JA, Espy MJ, et al. Preemptive use of oral ganciclovir to
(9):17771780. prevent cytomegalovirus infection in liver transplant patients: a random-
158. Sjovall J, Bergdahl S, Movin G, et al. Pharmacokinetics of foscarnet and ized, placebo-controlled trial. J Infect Dis. 2002;185(7):854860.
distribution to cerebrospinal fluid after intravenous infusion in patients 184. Emery VC. Prophylaxis for CMV should not now replace pre-emptive
with human immunodeficiency virus infection. Antimicrob Agents therapy in solid organ transplantation. Rev Med Virol. 2001;11(2):8386.
Chemother. 1989;33(7):10231031. 185. Wohl D, Kendall MA, Andersen J, et al. Low rate of CMV end-organ disease
159. Kempen JH, Jabs DA, Wilson LA, et al. Mortality risk for patients with in HIV-infected patients despite low CD4 cell counts and CMV viremia:
cytomegalovirus retinitis and acquired immune deficiency syndrome. Clin results of ACTG protocol A5030. HIV Clin Trials. 2009;10(3):143152.
Infect Dis. 2003;37(10):13651373. 186. Cooper DA, Pehrson PO, Pedersen C, et al. The efficacy and safety of zid-
160. Berman SM, Kim RC. The development of cytomegalovirus encephalitis ovudine alone or as cotherapy with acyclovir for the treatment of patients
in AIDS patients receiving ganciclovir. Am J Med. 1994;96(5):415419. with AIDS and AIDS-related complex: a double-blind randomized trial.
161. Tokumoto JI, Hollander H. Cytomegalovirus polyradiculopathy caused European-Australian Collaborative Group. AIDS. 1993;7(2):197207.
by a ganciclovir-resistant strain. Clin Infect Dis. 1993;17(5):854856. 187. Youle MS, Gazzard BG, Johnson MA, et al. Effects of high-dose oral acy-
162. Emery VC, Griffiths PD. Prediction of cytomegalovirus load and resis- clovir on herpesvirus disease and survival in patients with advanced HIV
tance patterns after antiviral chemotherapy. Proc Natl Acad Sci U S A. disease: a double-blind, placebo-controlled study. European-Australian
2000;97(14):80398044. Acyclovir Study Group. AIDS. 1994;8(5):641649.
163. Bowen EF, Emery VC, Wilson P, et al. Cytomegalovirus polymerase chain 188. Meyers JD, Reed EC, Shepp DH, et al. Acyclovir for prevention of cyto-
reaction viraemia in patients receiving ganciclovir maintenance therapy megalovirus infection and disease after allogeneic marrow transplanta-
for retinitis. AIDS. 1998;12(6):605611. tion. N Engl J Med. 1988;318(2):7075.

Scheld_Ch11.indd 181 2/21/14 5:34 PM


182 Part II: Viral Infections and Related Disorders

189. Prentice HG, Gluckman E, Powles RL, et al. Impact of long-term acy- 194. Pass RF, Duliege AM, Boppana S, et al. A subunit cytomegalovirus vac-
clovir on cytomegalovirus infection and survival after allogeneic bone cine based on recombinant envelope glycoprotein B and a new adjuvant.
marrow transplantation. European Acyclovir for CMV Prophylaxis Study J Infect Dis. 1999;180(4):970975.
Group. Lancet. 1994;343(8900):749753. 195. Pass RF. Development and evidence for efficacy of CMV glycoprotein
190. Feinberg JE, Hurwitz S, Cooper D, et al. A randomized, double-blind B vaccine with MF59 adjuvant. J Clin Virol. 2009;46(suppl 4):S73S76.
trial of valaciclovir prophylaxis for cytomegalovirus disease in patients 196. Griffiths PD, Stanton A, McCarrell E, et al. Cytomegalovirus gly-
with advanced human immunodeficiency virus infection. AIDS Clinical coprotein-B vaccine with MF59 adjuvant in transplant recipients: a
Trials Group Protocol 204/Glaxo Wellcome 123-014 International CMV phase 2 randomised placebo-controlled trial. Lancet. 2011;377(9773):
Prophylaxis Study Group. J Infect Dis. 1998;177(1):4856. 12561263.
191. Martin DF, Kuppermann BD, Wolitz RA, et al. Oral ganciclovir for patients 197. Plotkin SA, Starr SE, Friedman HM, et al. Effect of Towne live virus vac-
with cytomegalovirus retinitis treated with a ganciclovir implant. Roche cine on cytomegalovirus disease after renal transplant: a controlled trial.
Ganciclovir Study Group. N Engl J Med. 1999;340(14):10631070. Ann Intern Med. 1991;114(7):525531.
192. Frey SE, Harrison C, Pass RF, et al. Effects of antigen dose and immuniza- 198. Sabbaj S, Pass RF, Goepfert PA, et al. Glycoprotein B vaccine is capable of
tion regimens on antibody responses to a cytomegalovirus glycoprotein boosting both antibody and CD4 T-cell responses to cytomegalovirus in
B subunit vaccine. J Infect Dis. 1999;180(5):17001703. chronically infected women. J Infect Dis. 2011;203(11):15341541.
193. Mitchell DK, Holmes SJ, Burke RL, et al. Immunogenicity of a recombi- 199. Riddell SR, Watanabe KS, Goodrich JM, et al. Restoration of viral immu-
nant human cytomegalovirus gB vaccine in seronegative toddlers. Pediatr nity in immunodeficient humans by the adoptive transfer of T cell clones.
Infect Dis J. 2002;21(2):133138. Science. 1992;257(5067):238241.

Scheld_Ch11.indd 182 2/21/14 5:34 PM


CHAPTER 12 EPSTEIN-BARR VIRUS
SUSAN E. HOOVER, JEFFREY P. ROSS, AND JEFFREY I. COHEN

result in encephalitis, myelitis, or neuropathy in nonimmu-


ETIOLOGY nocompromised persons. Although encephalitis or radiculo-
neuropathy can occur in patients with CMV infection, these
Epstein-Barr virus (EBV) is a member of the herpesvirus family patients are usually immunocompromised, and the disease is
(1) and, like other herpesviruses, contains a double-stranded progressive. Encephalitis due to herpes simplex virus (HSV) is
DNA core surrounded by a nucleocapsid. A tegument layer usually localized to the temporal lobes.
containing several proteins envelops the nucleocapsid, which
is surrounded by an outer lipid envelope studded with viral
glycoprotein spikes. CLINICAL MANIFESTATIONS
EBV infects B lymphocytes and epithelial cells in the oro-
pharynx (2). Infection of epithelial cells results in lytic infec-
tion with production of virions, lysis of the cell, and release of Infectious Mononucleosis
infectious virus. During virus replication, multiple EBV pro-
teins are expressed including the EBV early antigens (EAs), EBV-associated infectious mononucleosis is an acute illness
DNA polymerase (the target of acyclovir), viral capsid antigen characterized by fever, pharyngitis, lymphadenopathy, and
(VCA), and viral glycoproteins. Infection of B lymphocytes mononuclear leukocytosis with atypical lymphocytes (3).
usually results in latent infection and immortalization of the Symptoms of infectious mononucleosis commonly include sore
cells in vitro, without production of infectious virus. These throat, headache, and malaise in more than 50% of patients,
latently infected cells express only the EBV-encoded RNAs and myalgias, anorexia, nausea, and abdominal discomfort
(EBER-1 and EBER-2), nuclear proteins (EBNA-1, -2, -3A, in 10% to 20% of individuals. Physical examination shows
-3B, -3C, and LP), and latent membrane proteins (LMP-1, lymphadenopathy, pharyngitis, and splenomegaly in most pa-
-2A, and -2B). tients, whereas 10% have hepatomegaly, jaundice, palatal enan-
EBV has a worldwide distribution, and the vast majority them, or rash. Posterior cervical lymphadenopathy is most often
of adults show serologic evidence of past infection. Primary noted; however, any group of lymph nodes may be involved. A
EBV infection is usually asymptomatic or mildly symptomatic pruritic maculopapular eruption develops in nearly all patients
in early childhood; in contrast, symptomatic infectious mono- after the administration of ampicillin. Important complications
nucleosis is more common in individuals who become infected of infectious mononucleosis include splenic rupture, autoim-
with EBV after the first decade of life. Transmission of EBV mune hemolytic anemia, thrombocytopenia, hepatitis, myocar-
probably occurs most often by exposure to saliva from asymp- ditis, airway obstruction from enlarged tonsils, and neurologic
tomatic individuals who shed the virus. Serologic studies sug- abnormalities. Neurologic complications are cited as the lead-
gest that EBV is commonly spread among susceptible siblings ing cause of death resulting from infectious mononucleosis,
within a family. EBV-associated infectious mononucleosis has followed by secondary infection and splenic rupture. Fatalities
been transmitted by blood transfusion. occur in fewer than 0.03% of cases of infectious mononucleosis.
The incubation period for symptomatic infectious mono- Rare individuals infected with EBV develop a chronic ac-
nucleosis is 30 to 50 days. During the first few weeks of the ill- tive infection, persisting 6 months and characterized by per-
ness, peripheral lymphocytosis occurs, with a marked increase sistent adenopathy and hepatosplenomegaly and involvement
in T cells and natural killer cells; many of these cells appear of the lungs, eyes, or nervous system with EBV-infected lym-
as atypical lymphocytes on the peripheral blood smear. phocytes. EBV infection of patients with the X-linked lympho-
Most of the clinical features associated with infectious mono- proliferative disorder results in a fatal disease characterized
nucleosis are manifestations of the vigorous cellular immune by lymphoid infiltration of organs, lymphomas, and opportu-
responses to EBV infection rather than to direct cytotoxic ef- nistic infections. Immunocompromised patients may develop
fects of the virus. B cells are also increased in number, and up EBV-associated lymphomas.
to 20% may express EBV nuclear antigen during the first week
of illness.
Neurologic Complications of Acute
Epstein-Barr Virus Infection
DIFFERENTIAL DIAGNOSIS
Involvement of the nervous system in infectious mononu-
Other infectious or noninfectious diseases can produce the cleosis is more common than generally appreciated. About
syndrome of infectious mononucleosis. Acute infection with 50% of patients have headache on presentation. Neck stiff-
cytomegalovirus (CMV), Toxoplasma gondii, human immu- ness without meningitis is a common finding. In one study,
nodeficiency virus type 1 (HIV-1), human herpesvirus type 6, about 25% of patients (43 of 162) had more than five cells
or hepatitis viruses can result in fever, lymphadenopathy, sore per cubic millimeter in the cerebrospinal fluid (CSF) (4). Many
throat, and atypical lymphocytes. Acute leukemia, lymphoma, of these patients had no symptoms of meningitis. Abnormal
and reactions to certain drugs (e.g., phenytoin and sulfon-
amides) can also produce a syndrome resembling infectious
mononucleosis. Disclaimer: This chapter was prepared by Drs. Hoover, Ross, and Cohen
Other herpesviruses also produce neurologic symptoms in their personal capacities. The views expressed herein do not necessarily
similar to EBV. Infection with varicella-zoster virus also can represent the views of the NIH, DHHS, or the federal government.

183

Scheld_Ch12.indd 183 2/21/14 5:35 PM


184 Part II: Viral Infections and Related Disorders

electroencephalograms (EEGs), which most often reveal dif-


fuse slowing patterns, have also been reported in patients with
infectious mononucleosis, but without clinically significant
central nervous system (CNS) disease (4).
Significant neurologic complications of acute infectious
mononucleosis are rare, occurring in fewer than 0.5% of more
than 4,300 cases of infectious mononucleosis in one review
(5). Neurologic complications, however, are an important
cause of morbidity in infectious mononucleosis and have been
reported in 6% to 7% of patients hospitalized with infectious
mononucleosis (6,7,7a). A study of children admitted to the
hospital with new neurologic symptoms found acute EBV in-
fections in 4% of patients and reactivated infections in 17%
of patients (8).
CNS symptoms may occur shortly before, during, or after
infectious mononucleosis or following acute EBV infection in
the absence of symptomatic infectious mononucleosis. Cases
of neurologic symptoms associated with infectious mono-
nucleosis before the 1970s, when EBV-specific serologic tests
were developed, must be regarded with caution because a
number of other infectious agents can produce the clinical
syndrome of mononucleosis. Only 1 of 14 cases of neurologic
complications associated with primary EBV infection reported
by Grose et al. (9) presented with clinical infectious mononu-
cleosis. Similarly, only 1 of 21 children with EBV encephalitis
identified prospectively at the Hospital for Sick Children in
Toronto had infectious mononucleosis; the others had nonspe-
cific symptoms of fever and headache (9a).
Examination of biopsy or autopsy specimens suggests that
immunologic mechanisms are probably more important in
the pathogenesis of neurologic complications than active viral
replication. Perivascular lymphocytic infiltrates, which may
include atypical lymphocytes, are often found in the leptomen-
inges, CNS parenchyma, nerve roots, and peripheral nerves
(10) (Fig. 12.1). Additional findings include parenchymal
edema and microglial proliferation (11). Inflammatory demy-
elinating lesions similar to those found in multiple sclerosis
or postinfectious encephalomyelitis or neuritis have also
been described (10,12). A report of antineuronal antibodies FIGURE 12.1 Brain biopsy from a 17-year-old girl with Epstein-Barr
in the serum of a patient with EBV-associated acute cerebellar virus encephalomyelitis. Top: Abnormal white matter showing peri-
ataxia, but not in patients with other neurologic diseases, pro- vascular infiltrates of inflammatory cells and foam cells. Some myelin
has been replaced by lipid-filled macrophages and hyperplastic astro-
vides support for an immune-mediated mechanism for CNS cytes. Hematoxylin-eosin stain; magnification 200. Bottom: Higher
disease (13). power view showing perivascular infiltrates and microglial reaction.
These studies indicate that the pathogenesis of EBV- Hematoxylin-eosin stain; magnification 400. (From Ambler M, Stoll
associated neurologic disease is different from that of HSV J, Tzamaloukas A, et al. Focal encephalomyelitis in infectious mono-
CNS disease. Although viral proteins are present in the brain nucleosis: a report with pathological description. Ann Intern Med.
of patients with herpes simplex encephalitis, brain biopsies 1971;75:579585, with permission.)
from patients with EBV encephalitis do not show viral nucleic
acids or proteins (14). Thus, autoimmune mechanisms are
probably responsible for the pathogenesis of EBV-associated
CNS disease.
Encephalitis
Patients having encephalitis associated with acute EBV infec-
Aseptic Meningitis tion may present with seizures, coma, personality changes,
distortions of perception, cerebellar ataxia, or focal brainstem
Aseptic meningitis is the most common neurologic complica- or cerebral findings. These complications usually occur 1 to 3
tion of primary EBV infection. Patients present with headache, weeks after the onset of clinical infectious mononucleosis, but
malaise, stiff neck, and occasionally photophobia (15). The they have occurred shortly before, during, or after other symp-
CSF usually shows a mild to moderate lymphocytic pleocyto- toms of infectious mononucleosis (15,16,17a). Alternatively,
sis, occasionally with atypical lymphocytes. The CSF protein they may be the only symptom of acute infection with EBV
concentration is normal or mildly elevated, and the glucose (6,18,19). Patients usually present with fever and headache
concentration is usually normal (15,16). Although the CSF progressing to encephalitis over a few days. Most cases occur
opening pressure is usually normal or slightly elevated, in in older children and young adults.
some patients, it is markedly increased, and repeated lumbar Generalized seizures, as well as coma and other signs of
puncture provides symptomatic relief (17). Aseptic meningitis diffuse encephalopathy, may occur without focal neurologic
associated with EBV has a good prognosis and usually resolves findings (5,7,15,16). Patients have presented with new onset
without CNS sequelae. of seizures and only later have been found by serology to have

Scheld_Ch12.indd 184 2/21/14 5:35 PM


Chapter 12: Epstein-Barr Virus 185

recent EBV infection (20). EEGs in patients with diffuse en- occurred over 3 months. Magnetic resonance imaging (MRI)
cephalopathy show generalized slowing with occasional sharp showed rapidly changing CNS lesions that correlated with the
wave activity, but they may reveal periodic discharges in pa- fluctuating neurologic examination results (Fig. 12.2).
tients with seizures (20).
Focal neurologic findings in EBV-associated encephalitis in-
clude localized seizures, hemiparesis, and unilateral Babinski Myelitis
reflexes. In a large prospective study of encephalitis by the
Collaborative Antiviral Study Group, in which focal neuro- Spinal cord lesions can accompany encephalitis and present
logic findings were required for entry, 2% of patients (8 of as paraplegia, quadriplegia, or other motor or sensory losses
432) were found to have EBV encephalitis (21). (5,34). The CSF shows a mononuclear pleocytosis with ele-
Although EBV encephalitis may involve any area of the vated levels of protein, but the glucose level is often normal.
brain, the cerebellum is commonly involved. Numerous cases Isolated transverse myelitis with sensory and motor loss in the
of cerebellitis, usually manifested as acute cerebellar ataxia, absence of encephalopathy has also been reported (6,9), with
have been reported (13,21a). Most patients present with ab- viral DNA being detected in the CSF (35,36). Most cases of
normalities of gait. A complete recovery is usual, but severe myelitis occur in older children and young adults and eventu-
cases with tonsillar herniation and death have been reported ally resolve without residua, although several cases in older
(21b). Other commonly involved areas include the basal gan- adults have resulted in long-term sequelae such as muscle
glia and cerebral hemispheres, with many patients having weakness and sensory deficits (34).
involvement of multiple areas (21c21e). Several cases of in-
creased intracranial pressure associated with ischemic lesions
of the corpus callosum have been reported, two requiring Cranial and Peripheral Neuritis
decompressive craniectomy (17a,21f,22). Encephalitis result-
ing from EBV infection has been reported as a cause of acute Although any of the cranial nerves may be affected during
hemiplegia in children and young adults (23). Some patients EBV infection, palsies of cranial nerve VII are most com-
present with a typical history of infectious mononucleosis fol- mon. Numerous cases of unilateral or bilateral Bell palsy have
lowed by psychosis and personality changes. Children with been reported in the setting of acute infectious mononucleosis
EBV encephalitis may present with metamorphopsia, the so- (5,6,15). With the advent of specific EBV serologic tests, in-
called Alice in Wonderland syndrome, in which affected in- creased numbers of cases of Bell palsy have been attributed
dividuals have perceptual distortions of personal body image, to otherwise asymptomatic EBV infection (9). Other forms
size, and spatial relationships (8,18). Movement disorders in- of cranial nerve involvement include anosmia (37); Fisher
dicative of extrapyramidal involvement have been reported. syndrome, involving multiple cranial nerves (38); bilateral
These include cogwheel rigidity and athetosis (24), Parkinson- sensorineural hearing loss (39); unilateral hearing loss with
like syndrome (25,21d,21e), and Sydenham-type chorea (26). ipsilateral involvement of cranial nerves V and VII (40); ves-
Brainstem encephalitis with cranial nerve and long-tract signs tibular neuritis with oscillopsia; hypoglossal (XII) nerve palsy
(27) and the locked-in syndrome have been described. (41); vocal fold paralysis due to vagal (X) nerve palsy (41);
Acute psychosis (28) and transient global amnesia (29) may be extraocular muscle palsies involving cranial nerves III (42), IV,
the only neurologic manifestation of EBV infection. Unusual or VI (6); and acute optic neuritis (15,43). Other ocular com-
complications of EBV encephalitis include acute aqueductal plications include papilledema, retrobulbar neuritis, chiasmic
stenosis (30) and the syndrome of inappropriate antidiuretic neuritis, ptosis, uveitis, episcleritis, and opsoclonus (44,45).
hormone secretion (31). Reye syndrome, acute fatty degenera- Among peripheral nerves that are affected during EBV in-
tion of the liver accompanied by encephalopathy, has followed fection, the brachial plexus and its branches are most com-
primary EBV infection in a number of reported cases (32). monly involved. Brachial plexopathy in the setting of infectious
Rare cases of relapsing or chronic encephalitis associated mononucleosis usually presents with shoulder pain followed
with acute EBV infection have been reported. Tolly, Wells, and by paralysis of the serratus anterior for weeks to months
Sty (33) described a 10-year-old girl with waxing and wan- (7,46). Five patients with lumbosacral radicular plexopathy
ing neurologic symptoms associated with EBV infection that and one patient with femoral neuropathy were described who

FIGURE 12.2 MRI scan of a 10-year-old girl


with a prolonged neurologic syndrome associated
with Epstein-Barr virus infection. T2-weighted im-
ages reveal a right cerebellar lesion (A) and bilat-
eral frontal lesions (B). These were two of several
transient lesions present on MRI scans that cor-
related with neurologic findings over a 3-month
period and resolved completely over subsequent
weeks. (From Tolly TL, Wells RG, Sty JR. MR
features of fleeting CNS lesions associated with
A B Epstein-Barr virus infection. J Comput Assist
Tomogr. 1989;13:665668, with permission.)

Scheld_Ch12.indd 185 2/21/14 5:35 PM


186 Part II: Viral Infections and Related Disorders

had serologic evidence of recent EBV infection (47). Acute au-


tonomic neuropathy has also been reported with acute EBV
infection (48).

Guillain-Barr Syndrome
Guillain-Barr syndrome, or acute inflammatory demyelin-
ating polyneuropathy, is characterized by progressive motor
weakness of more than one limb and areflexia. Guillain-
Barr syndrome usually presents several days to weeks after
the onset of infectious mononucleosis, or it may present in
patients with otherwise asymptomatic EBV seroconversion.
Grose et al. (9) found evidence of acute EBV infection in 12
of 24 patients with Guillain-Barr syndrome. None of these
patients had symptomatic infectious mononucleosis before the
onset of Guillain-Barr syndrome. Dowling and Cook (49) re-
ported that 8 of 100 patients with Guillain-Barr syndrome A
had an immunoglobulin M (IgM)positive test result for EBV,
indicative of recent infection. A prospective study found evi-
dence of acute EBV infection in only 2 of 99 patients with
Guillain-Barr syndrome, with a similar frequency (2%) in
age-matched controls (50). However, the lack of association
of Guillain-Barr syndrome with EBV may have been a conse-
quence of the older age of the patients studied, because most
older individuals are already seropositive for EBV. A case
control study from the Netherlands found evidence of acute
EBV infection in 16 (10%) of 154 patients with Guillain-
Barr, compared with 1% of age-matched controls with other
neurologic diseases (51).

Epstein-Barr VirusAssociated Central B


Nervous System Lymphoma
FIGURE 12.3 Immunohistochemical staining of a central nervous
EBV has been associated with CNS lymphomas in patients system lymphoma from a patient with acquired immunodeficiency
without any apparent immunodeficiency or, more commonly, syndrome. A: Staining with a monoclonal antibody to EBNA-2
in patients with underlying immunodeficiencies. Although shows nuclear staining in most of the tumor cells (closed arrow) but
some studies of immunologically normal individuals have not in cells lining a blood vessel (open arrows) (magnification 250).
demonstrated EBV DNA in up to 46% (11/24 patients) of B: Staining with a monoclonal antibody to latent membrane protein
B-cell CNS lymphomas by in situ hybridization (52), most shows cytoplasmic and membrane staining in several of the tumor
primary CNS B-cell lymphomas in nonimmunocompromised cells (closed arrows) (magnification 400). (From Bashir R, Luka J,
persons do not contain EBV genomes. Cheloha K, et al. Expression of Epstein-Barr virus proteins in primary
CNS lymphoma in AIDS patients. Neurology. 1993;43:23582362,
EBV-associated CNS lymphomas have been reported in pa- with permission.)
tients receiving solid organ or bone marrow transplants and in
patients with leukemia, X-linked lymphoproliferative disease,
and acquired immunodeficiency syndrome (AIDS) (53). From
10% to 25% of lymphomas in patients with heart, heartlung, used EBV DNA PCR to test CSF of patients with HIV and CNS
liver, kidney, or bone marrow transplants involve the CNS and symptoms. They found that PCR for EBV had a sensitivity of
contain EBV genomes (54). 97% (35 patients PCR positive/36 with disease), specificity of
EBV has been detected in virtually 100% of CNS lym- 98% (180 patients PCR negative/183 without disease), and a
phomas in patients with AIDS (5456). Nearly all EBV- positive predictive value of 90% for the diagnosis of CNS lym-
positive lymphomas in human immunodeficiency virus phoma. Antinori et al. (60) showed a sensitivity of 85% and a
(HIV)positive patients express the EBV latency-associated specificity of 100% for PCR of CSF in their evaluation of pa-
proteins EBNA-2 and LMP-1 (57,58) (Fig. 12.3). Viral tients for CNS lymphoma. In some patients, EBV was detected
DNA is present in the lymphoma, not in neuronal cells (55). in CSF by PCR before CNS lesions were apparent on computed
EBV-associated CNS lymphoma in patients with AIDS oc- tomographic (CT) or MRI scanning (59). However, more recent
curs more often in patients with very low CD4 cell counts. studies in the era of HAART indicate that PCR alone is less
HIV patients who later developed CNS lymphomas lacked specific for a diagnosis of CNS lymphoma with a positive pre-
EBV-specific CD4 T cells (58a). CNS lymphomas are more dictive value of 30% or less (60a,60b).
often monoclonal and lack c-Myc translocations when com- The combination of CSF EBV PCR and single-photon
pared with systemic (not involving the CNS) lymphomas in emission computed tomography (SPECT) scanning has shown
patients with AIDS. promise for distinguishing CNS lymphoma from cerebral toxo-
Early studies, prior to the era of highly active antiretroviral plasmosis and other infections (61,62). A SPECT-to-thallium
therapy (HAART), showed that polymerase chain reaction (PCR) uptake ratio is calculated as the amount of thallium-201 up-
detection of EBV DNA in the CSF was highly predictive for pri- take in the lesion divided by uptake in uninvolved brain or
mary CNS lymphoma in patients with AIDS. Cinque et al. (59) scalp. Using SPECT scanning in HIV-infected patients with

Scheld_Ch12.indd 186 2/21/14 5:35 PM


Chapter 12: Epstein-Barr Virus 187

focal brain lesions on CT or MRI, the sensitivity of a positive antibodies to EBV in patients with chronic fatigue compared
scan for lymphoma ranged from 86% to 92%, and the speci- with healthy controls (75). Although some patients have per-
ficity ranged from 83% to 89% (61,62). When both SPECT sistent fatigue lasting several months after acute infectious
scanning and CSF EBV PCR were performed, the sensitivity mononucleosis, at present, there is no evidence for persistent
increased to 100% (61). EBV infection as a cause of the chronic fatigue syndrome.
Several patients have been reported with the new onset of
depressive illness following acute infectious mononucleosis
Other Neurologic Syndromes Possibly (76). Subsequent casecontrol studies have yielded conflict-
ing results (77,78). Review of these studies indicates that EBV
Associated with Epstein-Barr Virus does not play a significant role in the pathogenesis of depres-
sion. Three cases have been reported in which fatal subacute
Epidemiologic data suggest an association between EBV and
sclerosing panencephalitis (SSPE), which is associated with
multiple sclerosis (MS). Eight casecontrol studies found that
a measles-like agent, occurred concomitantly with infectious
97% to 100% of patients with MS were seropositive for EBV,
mononucleosis (79). Whether EBV infection or its associated
whereas only 65% to 95% of age- and sex-matched controls
immune alterations contributed to the development of SSPE in
were seropositive (63). Compilation of these studies, involving
these cases is not known.
about 1,000 patients with MS and a similar number of controls,
yielded an odds ratio for MS in EBV-seropositive persons ver-
sus EBV-seronegative persons of 13.5 (95% confidence interval,
6.331.4). A casecontrol study of 305 persons who developed LABORATORY AND
MS and 610 matched controls found that all 10 of the EBV- IMAGING STUDIES
negative persons who subsequently developed MS became EBV
positive before MS onset, whereas only 10 of 28 controls who The humoral immune response to EBV infection involves
never developed MS became EBV positive (64). Although a both viral-specific and nonspecific antibodies (Table 12.1).
metaanalysis showed a 2.2-fold relative risk of MS in persons Detection of heterophile antibodies at a titer of more than 40,
with infectious mononucleosis (65), the vast majority of patients along with symptoms of infectious mononucleosis and atypi-
with MS have no history of infectious mononucleosis (66). cal lymphocytes, is diagnostic of acute EBV infection. The
A nested casecontrol study of stored sera from more than 3 titer of heterophile antibody is defined as the maximum serum
million military personnel found that serum levels of IgG VCA dilution after absorption with guinea pig kidney cells, which
or EBNA complex were proportionate to the risk of MS with can agglutinate sheep, horse, or beef erythrocytes. The com-
an average time of 4 years between serum collection and di- mercially available monospot test is based on the heterophile
agnosis of MS (67). The relative risk of MS was 20 and 34 in test. False-positive results may occur in some patients with col-
those with the highest level of EBV VCA and EBNA complex lagen-vascular disease, lymphoma, or hepatitis; false-negative
antibodies, respectively, compared with those with the lowest results are more common in children younger than 5 years.
titers of these antibodies. A follow-up study showed a similar
increase in risk with antibodies to EBV EBNA complex and
EBNA-1 antibodies (68). Wandinger et al. (69) reported an as-
sociation between EBV reactivation and MS disease activity. In TA B L E 1 2 . 1
their study of 19 patients with MS followed monthly for 1 year, DIAGNOSTIC TESTS IN INFECTIOUS
patients with clinically documented relapses or progression of MONONUCLEOSIS AND EBV-ASSOCIATED
MS had more frequent anti-EBV EA immunoglobulin A (IgA) NEUROLOGIC SYNDROMES
seroconversion or detectable EBV DNA in the serum com-
pared with those with stable MS. Several patients have been I. Serology
reported who initially presented with acute EBV encephalitis
and developed new demyelinating CNS lesions over a period Antibody Onset Duration
of years, resulting in the diagnosis of MS (12,70). MS patients
have increased T-cell responses to EBV EBNA-1 which cross- VCA IgM Before or at time of symptoms 12 mo
react with myelin antigens and result in production of inter- VCA IgG Before or at time of symptoms Lifelong
feron (IFN)- (70a). Although one study reported finding EBV
RNA and protein in the brains from patients with MS (71), EBNA 34 wk after onset of symptoms Lifelong
EBV RNA has generally not been detected in MS plaques or in EA Peaks 34 wk after symptom onset; Months to
CSF from patients with MS, and intrathecal anti-EBV antibody present in 70% of patients years
synthesis did not differ in patients with MS and other controls Heterophile Before or at time of symptoms; Weeks to
(72). EBV might be a cofactor for MS, or EBV infection and present in about 90% of patients months
MS may share a common genetic predisposition or a common
immunologic abnormality. The true role of the virus in the II. Studies in EBV-associated neurologic syndromes
pathogenesis of MS is unclear. Additional studies are needed,
particularly to look at CSF for EBV DNA or RNA, oligoclonal Cerebrospinal fluid: protein, lymphocytes, atypical
bands specific for EBV, intrathecal synthesis of EBV antibody, lymphocytes, VCA IgG, PCR for EBV DNA
and EBV-specific T cells in patients with MS and controls. Computed tomography
In the mid-1980s, two small casecontrol studies reported
Magnetic resonance imaging
elevated antibody titers to EBV EA in patients with the chronic
fatigue syndrome (73,74). These patients presented with per- Single-photon emission computed tomography
sistent fatigue, myalgias, and mild cognitive impairment. Electroencephalography
Other studies have shown that antibody titers to EBV EA
remain elevated in a substantial number of otherwise healthy EA, early antigen; EBNA, EBV nuclear antigen; PCR, polymerase chain
individuals. A study of 548 patients attending a referral clinic reaction; VCA, viral capsid antigen.
found no differences in the seroprevalence or mean titers of

Scheld_Ch12.indd 187 2/21/14 5:35 PM


188 Part II: Viral Infections and Related Disorders

Specific antibody responses directed against EBV are progressive disease (60). Because EBV is present in circulating
more reliable than heterophile antibodies in making a diag- lymphocytes in asymptomatic seropositive individuals, culture or
nosis of EBV infection. Detection IgM antibodies to VCA, PCR of cells from the CSF of EBV-seropositive patients with other
which are usually present for 1 to 2 months after infection, causes of CNS inflammation is an important negative control be-
is particularly useful in the diagnosis of acute EBV infection. fore a causal association with EBV can be shown. Measurement
Immunoglobulin G (IgG) antibodies to VCA are less helpful of EBV DNA is frequently used for diagnosis, monitoring, and
for diagnosis, because they persist for life. EBV EA antibodies prevention of lymphoproliferative disease after transplant (81a).
develop later after infection and generally are not useful for Intrathecal production of EBV-specific antibody is consid-
diagnosis of acute infection. Patients with chronic active EBV ered present when the ratio of the CSF-to-serum EBV antibody
infection may have elevated VCA IgG or EA antibody titers. titer is greater than the ratio of the CSF-to-serum -globulin
Antibodies to EBNAs are usually detectable 3 to 4 weeks after concentration. Production of EBV antibody in the CSF has
onset of symptoms of infectious mononucleosis and persist been demonstrated in several studies and suggests that active
for life. Seroconversion to anti-EBNA positivity is evidence virus replication may occur in the CNS (31,34,81).
for recent infection with EBV. Anti-EBNA antibodies may be CT may show changes typical of viral encephalitis or
absent in immunocompromised patients. acute disseminated encephalomyelitis with low attenuation,
Establishing the diagnosis of an EBV-associated neurologic nonenhancing cerebral lesions that may be associated with
syndrome relies on an appropriate clinical neurologic diag- mass effect, or edema in progressive disease (33). MRI is con-
nosis in the setting of serologic evidence of acute or rarely siderably more sensitive and can reveal lesions, particularly
chronic active EBV infection (Table 12.1). Examination of the on T2-weighted images, when CT findings are normal (82)
CSF often reveals a lymphocytic pleocytosis with an elevated (Fig. 12.4). SPECT brain scans show lesions with reduced
protein level, particularly in EBV encephalitis; atypical lym- uptake in patients with EBV encephalitis (18) and increased
phocytes, if present, provide a clue to diagnosis. The CSF in uptake in patients with EBV lymphomas (61,62). EEGs fre-
Guillain-Barr syndrome typically contains increased levels of quently show generalized slowing with occasional bursts of
protein without a significant pleocytosis. activity in patients with encephalitis.
EBV has rarely been cultured from the mononuclear
cells obtained from the CSF of patients with neurologic
complications of EBV (80). EBV DNA has been detected by TREATMENT AND OUTCOME
PCR of CSF or neural tissue in patients with EBV-associated
neurologic conditions (14,26,81). Imai et al. (81) studied five The treatment of neurologic complications associated with
children with EBV-associated neurologic conditions diagnosed acute EBV infection is generally supportive and may include an-
by standard EBV serology. EBV DNA, but not other herpes- ticonvulsants, judicious use of fluids (to avoid cerebral edema),
virus DNA, was detected by PCR of the CSF from each case. and mechanical ventilation. Corticosteroids may be required
EBV DNA became undetectable by PCR as patients recovered in cases of increased intracranial pressure. Steroid treatment in
from their neurologic complications. Weinberg et al. (36) per- cases of uncomplicated infectious mononucleosis reduces the
formed quantitative PCR of the CSF and demonstrated that duration of fever and sore throat but does not reduce lymph-
patients with CNS lymphoma and encephalitis had high EBV adenopathy or splenomegaly (83). Corticosteroids have been
loads (104 EBV copies/mL); patients with encephalitis had reported to be helpful in several anecdotes describing neuro-
higher leukocyte counts than those with lymphoma. Two logic complications related to EBV infection (including enceph-
patients with myelitis had lower EBV loads (103 EBV copies/ alomyelitis, cerebellar ataxia, transverse myelitis, and cranial
mL), but high leukocyte counts. Reverse transcriptase PCR and peripheral neuropathies) without intracranial hypertension
(RT-PCR) detected EBV lytic cycle messenger RNAs (mRNAs) (24,38,47). Steroids, however, have also been associated with
in the CSF of several patients with lymphoma or encephalitis. the new onset of neurologic symptoms in patients with acute
EBV in the CNS declined in two patients with lymphomas infectious mononucleosis (84) and with a worse outcome in
that responded to treatment, and increased in four patients with Guillain-Barr syndrome (85). The limited experience with

FIGURE 12.4 Cerebral imaging from a 10-year-


old girl with Epstein-Barr virus encephalitis.
A: Findings of contrast-enhanced computed
tomographic scanning are normal. B: T2-weighted
MRI scan at the same level demonstrates bilat-
eral abnormalities in the posterior medial tempo-
ral areas (arrows). (From Bale JF, Andersen RD,
Grose C. Magnetic resonance imaging of the brain
in childhood herpesvirus infections. Pediatr Infect
Dis J. 1987;6:644647, with permission.)

Scheld_Ch12.indd 188 2/21/14 5:35 PM


Chapter 12: Epstein-Barr Virus 189

corticosteroids for EBV-associated CNS disease suggests that AIDS-related primary CNS lymphoma is decreasing in the era
they are useful in select cases; however, their potential benefits of HAART (92). A regimen including high-dose methotrexate,
must be balanced against their numerous risks. corticosteroids, and HAART is usually used for CNS lympho-
Acyclovir inhibits EBV replication in vitro; however, a meta- mas in patients with HIV. Although some patients with CNS
analysis of five clinical trials did not show any benefit of acyclovir lymphoma who received HAART and cranial irradiation had
in the therapy of acute infectious mononucleosis (86). Although prolonged survival compared with those who received only
acyclovir has been used in some cases of CNS disease associated cranial irradiation, insufficient data are available to verify the
with EBV, it probably provides little or no benefit (87). Acyclovir efficacy of HAART in the treatment of CNS lymphoma (93).
might be used in conjunction with corticosteroids for select cases Most patients with neurologic complications of acute EBV
of EBV-associated CNS disease in an attempt to reduce viral rep- infection recover completely within weeks to months; however,
lication in the setting of an immunosuppressing agent. some have sequelae. Of 35 patients with neurologic syndromes
EBV-associated lymphomas involving the CNS in posttrans- associated with heterophile antibodypositive infectious
plant recipients are usually treated with cytotoxic chemotherapy mononucleosis in whom outcomes were noted, 80% (28/35)
and/or radiation therapy. Immunosuppression is reduced when recovered completely, 14% (5/35) had neurologic residua, and
possible. Newer treatments include monoclonal antibodies to 6% (2/35) died (15). Fatalities are usually caused by respira-
B-cell surface antigens (88,89), interferon- therapy (90), or tory arrest related to encephalitis or Guillain-Barr syndrome
infusion of EBV-specific cytotoxic T cells (91). The incidence of or occasionally to secondary infections.

References
1. Longnecker R, Kieff E, Cohen JI. Epstein-Barr virus. In: Knipe DM, 21a. McCarthy CL, McColgan P, Martin P. Acute cerebellar ataxia due to
Howley PM, eds. Fields Virology. Philadelphia: Lippincott Williams & Epstein-Barr virus. Pract Neurol. 2012;12:238240.
Wilkins; 2013. 21b. Van Lierde A, Righini A, Tremolati E. Acute cerebellitis with tonsillar her-
2. Cohen JI. Epstein-Barr virus infection. N Engl J Med. 2000;343:481492. niation and hydrocephalus in Epstein-Barr virus infection. Eur J Pediatr.
3. Johannsen EC, Kaye KM. Epstein-Barr virus. In: Mandell GL, Bennett JE, 2004;163:689691.
Dolin R, eds. Principles and Practice of Infectious Diseases. 7th ed. New 21c. Abul-Kasim K, Palm L, Maly P, et al. The neuroanatomic localization
York: Churchill Livingstone; 2010:19892010. of Epstein-Barr virus encephalitis may be a predictive factor for its clini-
4. Pejme J. Infectious mononucleosis. A clinical and haematological study of cal outcome: a case report and review of 100 cases in 28 reports. J Child
patients and contacts, and a comparison with healthy subjects. Acta Med Neurol. 2009;24:720726.
Scand Suppl. 1964;413(suppl):183. 21d. Guan J, Lu Z, Zhou Q. Reversible parkinsonism due to involvement of sub-
5. Silversides JL, Richardson JC. Neurological complications of infectious stantia nigra in Epstein-Barr virus encephalitis. Mov Disord. 2011;27:156157.
mononucleosis. Can Med Assoc J. 1950;63:138143. 21e. Espay A, Henderson K. Postencephalitic parkinsonism and basal ganglia
6. Silverstein A, Steinberg G, Nathanson M. Nervous system involvement in necrosis due to Epstein-Barr virus infection. Neurology. 2011;76:15291530.
infectious mononucleosis. Arch Neurol. 1972;26:353358. 21f. Takeuchi S, Takasato Y, Masaoka H. Epstein-Barr virus encephalitis with a
7. Gautier-Smith PC. Neurological complications of glandular fever (infectious reversible splenial lesion. Intern Med. 2012;51:341342.
mononucleosis). Brain. 1965;88:323334. 22. Hagemann G, Mentzel HJ, Weisser H, et al. Multiple reversible MR
7a. Fujimoto H, Asaoka K, Imaizumi T, et al. Epstein-Barr virus infections of signal changes caused by Epstein-Barr virus encephalitis. AJNR Am J
the central nervous system. Intern Med. 2003;42:3340. Neuroradiol. 2006;27:14471449.
8. Hausler M, Ramaekers VT, Doenges M, et al. Neurological complications 23. Baker FJ, Kotchmar GS, Foshee WS, et al. Acute hemiplegia of childhood
of acute and persistent Epstein-Barr virus infection in pediatric patients. associated with Epstein-Barr virus infection. Pediatr Infect Dis. 1983;2:
J Med Virol. 2002;68:253263. 136138.
9. Grose C, Henle W, Henle G, et al. Primary Epstein-Barr virus infections in 24. Slade J. Involvement of the central nervous system in infectious mononucleosis.
acute neurologic diseases. N Engl J Med. 1975;292:392395. N Engl J Med. 1946;234:753757.
9a. Doja A, Bitnun A, Ford Jones E, et al. Pediatric Epstein-Barr virus-associ- 25. Hsieh JC, Lue KH, Lee YL. Parkinson-like syndrome as the major present-
ated encephalitis: 10-year review. J Child Neurol. 2006;21:384391. ing symptom of Epstein-Barr virus encephalitis [letter]. Arch Dis Child.
10. Ambler M, Stoll J, Tzamaloukas A, et al. Focal encephalomyelitis in infec- 2002;87:358359.
tious mononucleosis: a report with pathological description. Ann Intern 26. Tachi N, Nagata N, Chiba S, et al. Detection of Epstein-Barr virus genome
Med. 1971;75:579585. in cerebrospinal fluid from a patient with acquired chorea by the poly-
11. Custer RP, Smith EB. The pathology of infectious mononucleosis. Blood. merase chain reaction. J Child Neurol. 1993;8:424425.
1948;3:830857. 27. North K, de Silva L, Procopis P. Brain-stem encephalitis caused by Epstein-Barr
12. Bray PF, Culp KW, McFarlin DE, et al. Demyelinating disease after neu- virus. J Child Neurol. 1993;8:4042.
rologically complicated primary Epstein-Barr virus infection. Neurology. 28. Jarvis MR, Wasserman AL, Todd RD. Acute psychosis in a patient with
1992;42:278282. Epstein-Barr virus infection. J Am Acad Child Adolesc Psychiatry. 1990;29:
13. Ito H, Sayama S, Irie S, et al. Antineuronal antibodies in acute cerebel- 468469.
lar ataxia following Epstein-Barr virus infection. Neurology. 1994;44: 29. Pommer B, Pilz P, Harrer G. Transient global amnesia as a manifestation of
15061507. Epstein-Barr virus encephalitis. J Neurol. 1983;229:125127.
14. Pedneault L, Katz BZ, Miller G. Detection of Epstein-Barr virus in the brain 30. Cotton MF, Reiley T, Robinson CC, et al. Acute aqueductal stenosis in
by the polymerase chain reaction. Ann Neurol. 1992;32:184192. a patient with Epstein-Barr virus infectious mononucleosis. Pediatr Infect
15. Bernstein TC, Wolff HG. Involvement of the nervous system in infectious Dis J. 1994;13:224227.
mononucleosis. Ann Intern Med. 1950;33:11201138. 31. McKendall RR, Sakiq SA, Calverley JR. Unusual manifestations of Epstein-
16. Schnell RG, Dyck PJ, Bowie EJW, et al. Infectious mononucleosis: neuro- Barr virus encephalomyelitis. Infection. 1990;18:4143.
logic and EEG findings. Medicine. 1966;45:5163. 32. Fleisher G, Schwartz J, Lennette E. Primary Epstein-Barr virus infection in
17. Weschler HF, Rosenblum AH, Sills CT. Infectious mononucleosis: report of association with Reye syndrome. J Pediatr. 1980;97:935937.
an epidemic in an army post. Ann Intern Med. 1946;25:236265. 33. Tolly TL, Wells RG, Sty JR. MR features of fleeting CNS lesions associated
17a. Hayton E, Wakerley B, Bowler IC, et al. Successful outcome of Epstein-Barr with Epstein-Barr virus infection. J Comput Assist Tomogr. 1989;13:665668.
virus encephalitis managed with bilateral craniectomy, corticosteroids and 34. Majid A, Galetta SL, Sweeney CJ, et al. Epstein-Barr virus myeloradiculitis
aciclovir. Pract Neurol. 2012;12:234237. and encephalomyeloradiculitis. Brain. 2002;125(pt 1):159165.
18. Hung KL, Liao HT, Tsai ML. Epstein-Barr virus encephalitis in children. 35. Clevenbergh P, Jacobs F, Thys JP, et al. Infectious mononucleosis compli-
Acta Paediatr Taiwan. 2000;41:140146. cated by transverse myelitis: detection of the viral genome by polymerase
19. Domachowske JB, Cunningham CK, Cummings D, et al. Acute manifesta- chain reaction in the cerebrospinal fluid. J Neurol. 1997;244:592594.
tions and neurologic sequelae of Epstein-Barr virus encephalitis in children. 36. Weinberg A, Li S, Palmer M, et al. Quantitative CSF PCR in Epstein-Barr
Pediatr Infect Dis J. 1996;15:871875. virus infections of the central nervous system. Ann Neurol. 2002;52:543548.
20. Russell J, Fisher M, Zivin JA, et al. Status epilepticus and Epstein-Barr virus 37. Weinstein M. Epstein-Barr virus infection and neurologic disease. Neurol
encephalopathy. Arch Neurol. 1985;42:789792. Res. 1991;13:199200.
21. Whitley RJ, Cobbs CG, Alford CA, et al. Diseases that mimic herpes simplex 38. Slavick HE, Shapiro RA. Fishers syndrome associated with Epstein-Barr
encephalitis: diagnosis, presentation, and outcome. JAMA. 1989;262:234239. virus. Arch Neurol. 1981;38:134135.

Scheld_Ch12.indd 189 2/21/14 5:35 PM


190 Part II: Viral Infections and Related Disorders

39. Beg JA. Bilateral sensorineural hearing loss as a complication of infectious 66. Marrie RA, Wolfson C, Sturkenboom MC, et al. Multiple sclerosis and
mononucleosis. Arch Otolaryngol. 1981;107:620622. antecedent infections. Neurology. 2000;54:23072310.
40. Taylor L, Parsons-Smith G. Infectious mononucleosis, deafness and facial 67. Levin LI, Munger KL, Rubertone MV, et al. Multiple sclerosis and Epstein-
paralysis. J Laryngol Otol. 1969;83:613616. Barr virus. JAMA. 2003;289:15331536.
41. Johns MM, Hogikyan ND. Simultaneous vocal fold and tongue paresis 68. Munger KL, Levin LI, OReilly EJ, et al. Anti-Epstein-Barr virus antibod-
secondary to Epstein-Barr virus infection. Arch Otolaryngol Head Neck ies as serological markers of multiple sclerosis: a prospective study among
Surg. 2000;126:14911494. United States military personnel. Mult Scler. 2011;17:11851193.
42. Nellhaus G. Isolated oculomotor nerve palsy in infectious mononucleosis. 69. Wandinger KP, Jabs W, Siekhaus A, et al. Association between clinical
Neurology. 1966;16:221224. disease activity and Epstein-Barr virus reactivation in MS. Neurology.
43. Corssmit EP, Leverstein-van Hall MA, Portegies P, et al. Severe neuro- 2000;55:178184.
logical complications in association with Epstein-Barr virus infection. 70. Paskavitz JF, Anderson CA, Filley CM, et al. Acute arcuate fiber demyelin-
J Neurovirol. 1997;3:460464. ating encephalopathy following Epstein-Barr virus infection. Ann Neurol.
44. Tanner OR. Ocular manifestations of infectious mononucleosis. Arch 1995;38:127131.
Ophthalmol. 1954;51:229239. 70a. Lunemann JD, Jelcic I, Roberts S, et al. EBNA1-specific T cells from patients
45. Verma A, Brozman B. Opsoclonus-myoclonus syndrome following Epstein- with multiple sclerosis cross react with myelin antigens and co-produce
Barr virus infection. Neurology. 2002;58:11311132. IFN-gamma and IL-2. J Exp Med. 2008;205:17631773.
46. Watson P, Ashby P. Brachial plexus neuropathy associated with infectious 71. Serafini B, Rosicarelli B, Franciotta D, et al. Dysregulated Epstein-Barr virus
mononucleosis. Can Med Assoc J. 1976;51:758767. infection in the multiple sclerosis brain. J Exp Med. 2007;204:28992912.
47. Sharma KR, Sriram S, Fries T, et al. Lumbosacral radiculoplexopathy as a mani- 72. Sargsyan SA, Shearer AJ, Ritchie AM, et al. Absence of Epstein-Barr
festation of Epstein-Barr virus infection. Neurology. 1993;43:25502554. virus in the brain and CSF of patients with multiple sclerosis. Neurology.
48. Bennett JL, Mahalingam R, Wellish MC, et al. Epstein-Barr virus-associ- 2010;74:11271135.
ated acute autonomic neuropathy. Ann Neurol. 1996;40:453455. 73. Jones JF, Ray CG, Minnich LL, et al. Evidence for active Epstein-Barr virus
49. Dowling PC, Cook SD. Role of infection in Guillain-Barr syndrome: infection in patients with persistent, unexplained illnesses: elevated anti-
laboratory confirmation of herpesviruses in 41 cases. Ann Neurol. early antigen antibodies. Ann Intern Med. 1985;102:17.
1981;9(suppl):4455. 74. Straus SE, Tosato G, Armstrong G, et al. Persisting illness and fatigue
50. Winer JB, Hughes RAC, Anderson MJ, et al. A prospective study of acute in adults with evidence of Epstein-Barr virus infection. Ann Intern Med.
idiopathic neuropathy: antecedent events. J Neurol Neurosurg Psychiatry. 1985;102:716.
1988;51:613618. 75. Buchwald D, Ashley RL, Pearlman T, et al. Viral serologies in patients with
51. Jacobs BC, Rothbarth PH, van der Meche FGA, et al. The spectrum of chronic fatigue and chronic fatigue syndrome. J Med Virol. 1996;50:2530.
antecedent infections in Guillain-Barr syndrome: a casecontrol study. 76. Hendler N, Leahy W. Psychiatric and neurologic sequelae of infectious
Neurology. 1998;51:11101115. mononucleosis. Am J Psychiatry. 1978;135:842844.
52. Murphy JK, Young LS, Bevan IS, et al. Demonstration of Epstein-Barr 77. Miller AH, Silberstein C, Asnis GM, et al. Epstein-Barr virus infection and
virus in primary brain lymphoma by in situ DNA hybridization in paraffin depression. J Clin Psychiatry. 1986;46:529530.
wax-embedded tissue. J Clin Pathol. 1990;43:220223. 78. Amsterdam JD, Henle W, Winokur A, et al. Serum antibodies to Epstein-
53. Bashir RM, Hochberg FH, Harris NL, et al. Variable expression of Epstein- Barr virus in patients with major depressive disorder. Am J Psychiatry.
Barr virus genome as demonstrated by in situ hybridization in central ner- 1986;143:15931596.
vous system lymphomas in immunocompromised patients. Mod Pathol. 79. Feorino PM, Humphrey D, Hochberg F, et al. Mononucleosis-associated
1990;3:429434. subacute sclerosing panencephalitis. Lancet. 1975;2:530533.
54. Cohen JI. Epstein-Barr virus lymphoproliferative disease associated with 80. Schiff JA, Schaefer JA, Robinson JE. Epstein-Barr virus in cerebrospi-
acquired immunodeficiency. Medicine. 1991;70:137160. nal fluid during infectious mononucleosis encephalitis. Yale J Biol Med.
55. MacMahon EME, Glass JD, Hayward SD, et al. Epstein-Barr virus in AIDS- 1982;55:5963.
related primary central nervous system lymphoma. Lancet. 1991;338:969973. 81. Imai S, Usui N, Sugiura M, et al. Epstein-Barr virus genomic sequences and
56. Skiest DJ. Focal neurological disease in patients with acquired immunode- specific antibodies in cerebrospinal fluid in children with neurologic com-
ficiency syndrome. Clin Infect Dis. 2002;34:103115. plications of acute and reactivated EBV infections. J Med Virol. 1993;40:
57. Bashir R, Luka J, Cheloha K, et al. Expression of Epstein-Barr virus proteins in 278284.
primary CNS lymphoma in AIDS patients. Neurology. 1993;43:23582362. 81a. Gulley ML, Tang W. Using Epstein-Barr viral load assays to diagnose,
58. Hamilton-Dutoit SJ, Rea D, Raphael M, et al. Epstein-Barr virus-latent gene monitor, and prevent posttransplant lymphoproliferative disorder. Clin
expression and tumor cell phenotype in acquired immunodeficiency syndrome- Microbiol Rev. 2010;23(2):350366.
related non-Hodgkins lymphoma: correlation of lymphoma phenotype with 82. Bale JF, Andersen RD, Grose C. Magnetic resonance imaging of the brain
three distinct patterns of viral latency. Am J Pathol. 1993;143:10721085. in childhood herpesvirus infections. Pediatr Infect Dis J. 1987;6:644647.
58a. Gasser O, Bihl FK, Wolbers M, et al. HIV patients developing primary CNS 83. Andersson J, Ernberg I. Management of Epstein-Barr virus infections. Am
lymphoma lack EBV-specific CD4 T cell function irrespective of absolute J Med 1988;85(suppl 2A):107115.
CD4 T cell counts. PLoS Med. 2007;4(3):e96. 84. Waldo RT. Neurologic complications of infectious mononucleosis after steroid
59. Cinque P, Vago L, Dahl H, et al. Polymerase chain reaction on cerebro- therapy. South Med J. 1981;74:11591160.
spinal fluid for diagnosis of virus-associated opportunistic diseases of the 85. Hughes RA, Newsom-Davis JM, Perkin GD, et al. Controlled trial of pred-
central nervous system in HIV-infected patients. AIDS. 1996;10:951958. nisolone in acute polyneuropathy. Lancet. 1978;2:750753.
60. Antinori A, Cingolani A, De Luca A, et al. Epstein-Barr virus in monitor- 86. Torre D, Tambini R. Acyclovir for treatment of infectious mononucleosis:
ing the response to therapy of acquired immunodeficiency syndrome-related a meta-analysis. Scand J Infect Dis. 1999;31:543547.
primary central nervous system lymphoma. Ann Neurol. 1999;45:259261. 87. van der Horst C, Joncas J, Ahronheim G, et al. Lack of effect of peroral
60a. Corcoran C, Rebe K, van der Plas HJ, et al. The predictive value of cerebro- acyclovir for the treatment of acute infectious mononucleosis. J Infect Dis.
spinal fluid Epstein-Barr viral load as a marker of primary central nervous 1991;164:788792.
system lymphoma in HIV-infected persons. J Clin Virol. 2008;42:433436. 88. Faye A, Quartier P, Reguerre Y, et al. Chimeric anti-CD20 monoclonal
60b. Ivers LC, Kim AY, Sax PE. Predictive value of polymerase chain reaction antibody (rituximab) in posttransplant B-lymphoproliferative disorder follow-
of cerebrospinal fluid for detection of Epstein-Barr virus to establish the ing stem cell transplantation in children. Br J Haematol. 2001;115:112118.
diagnosis of HIV-related primary central nervous system lymphoma. Clin 89. Pels H, Schulz H, Manzke O, et al. Intraventricular and intravenous treat-
Infect Dis. 2004;38:16291632. ment of a patient with refractory primary CNS lymphoma using rituximab.
61. Antinori A, De Rossi G, Ammassari A, et al. Value of combined approach J Neurooncol. 2002;59:213216.
with thallium-201 single-photon emission computed tomography and 90. Davis CL, Wood BL, Sabath DE, et al. Interferon-alpha treatment of post-
Epstein-Barr virus DNA polymerase chain reaction in CSF for the diagnosis transplant lymphoproliferative disorder in recipients of solid organ trans-
of AIDS-related primary CNS lymphoma. J Clin Oncol. 1999;17:554560. plants. Transplantation. 1998;66:17701779.
62. Skiest DJ, Erdman W, Chang WE, et al. SPECT thallium-201 combined with 91. Sun Q, Burton R, Reddy V, et al. Safety of allogenic Epstein-Barr virus
Toxoplasma serology for the presumptive diagnosis of focal central nervous (EBV)-specific cytotoxic T lymphocytes for patients with refractory EBV-
system mass lesions in patients with AIDS. J Infect. 2000;40:274281. related lymphoma. Br J Haematol. 2002;118:799808.
63. Ascherio A, Munch M. Epstein-Barr virus and multiple sclerosis. 92. Ammassari A, Cingolani A, Pezzotti P, et al. AIDS-related focal brain
Epidemiology. 2000;11:220224. lesions in the era of highly active antiretroviral therapy. Neurology.
64. Levin LI, Munger KL, OReilly EJ, et al. Primary infection with the Epstein- 2000;55:11941200.
Barr virus and risk of multiple sclerosis. Ann Neurol. 2010;67(6):824830. 93. Hoffmann C, Tabrizian S, Wolf E, et al. Survival of AIDS patients with
65. Handel AE, Williamson AJ, Disanto G, et al. An updated meta-analysis of risk primary central nervous system lymphoma is dramatically improved by
of multiple sclerosis following infectious mononucleosis. PLoS One. 2010;5(9). HAART-induced immune recovery. AIDS. 2001;15:21192127.

Scheld_Ch12.indd 190 2/21/14 5:35 PM


CHAPTER 13 HUMAN HERPESVIRUS-6
MARY T. CASERTA

of transplacental antibody (17,18). Following this nadir, the


HISTORY geometric mean titers increase steadily to reach a peak at
1 year of age (7,19,20). By 3 years of age, 100% of children
Human herpesvirus-6 (HHV-6) was first isolated from six have acquired serologic evidence of infection with HHV-6. No
adults with acquired immunodeficiency syndrome (AIDS) or seasonal predilection for primary HHV-6 infection has been
lymphoproliferative disorders by Salahuddin et al. (1) in 1986. noted in prospective studies, but small outbreaks of roseola
Limited genomic analysis and morphologic studies suggested among infants documented by seroconversion to HHV-6 have
that the new virus was a member of the herpesvirus family been reported (21,22). Seroprevalence rates remain stable
and it was named human B lymphotropic virus (HBLV) based throughout childhood, adolescence, and adulthood until age
on an apparent B-cell tropism (2). Further experiments dem- 40 years, when a gradual fall in the prevalence and geometric
onstrated the propagation of HBLV in various human cell mean titer of antibody to HHV-6 is noted (23,24).
lines, including those of T-cell, B-cell, megakaryocytic, glial,
and endothelial origin, and the designation of HHV-6 was
proposed (3,4).
In 1988, Yamanishi et al. (5) were the first to report an etio- Transmission
logic role for HHV-6 in human disease. They isolated HHV-6
from the peripheral blood mononuclear cells (PBMCs) of four The pattern of widespread and rapid acquisition of HHV-6
Japanese children with exanthem subitum (roseola infantum) infection in infancy without contact with similarly ill individu-
and demonstrated seroconversion to HHV-6 in the patients als implies that the virus is spread readily from asymptomatic
convalescent sera. Since its discovery, numerous other diseases children and adults to susceptible infants. HHV-6 DNA has
have been linked with HHV-6 in both immunocompromised been frequently detected via polymerase chain reaction (PCR)
and immunocompetent hosts during primary or reactivated in saliva and throat swabs from healthy adults and children
infection, but a clear causal connection has not always been both in cross-sectional studies and over time (25,26). Salivary
proven. Diseases of the central nervous system (CNS) that gland tissue specimens screened by in situ hybridization,
have been associated with HHV-6 include encephalitis, pri- immunohistochemistry (IHC), and PCR have also been shown
marily in immunocompromised hosts, seizures, and multiple to contain HHV-6, suggesting that saliva may be a major mode
sclerosis (MS) (6). Chronic fatigue syndrome, Sjgren syn- of transmission and that the salivary glands are a site of persis-
drome, and several lymphoproliferative disorders including tent infection (27,28). In a prospective, longitudinal study of
malignancies were all initially associated with HHV-6 but infants, the risk of acquiring primary HHV-6 infection in the
have now been shown by further study to be unlikely due to first year of life was greater among subjects whose parents re-
HHV-6 infection (79). ported saliva-sharing practices, further supporting saliva as an
The syndrome of roseola has been well known among pedi- important mode of transmission (29). Recently, HHV-6 DNA
atricians for generations. Most authors credit Zahorsky (10) in has been identified in a majority of nasal mucous and olfactory
1913 with the first definite description of the illness as a com- tissue specimens suggesting that respiratory droplets may also
mon, self-limited, febrile exanthematous disease of infancy. In be a source of viral spread (30).
1941, Breese (11) described the epidemiologic, clinical, and Evidence of primary HHV-6 infection in infants as young
laboratory manifestations of roseola based on his experience as 14 to 21 days documented by viremia and rising antibody
with 100 patients. Eight years later, Berenberg, Wright, and titers demonstrates that the protection provided by maternal
Janeway (12) reported convulsions as the most common com- antibody is incomplete and suggests the possibility of perina-
plication of roseola, occurring in up to one third of children. tal transmission of HHV-6 (21,31). These data are consistent
Kempe et al. (13) were able to document the infectious nature with reports of HHV-6 DNA in both the blood and the cer-
of the illness in 1950 by directly transmitting disease via serum vical/vaginal samples from 12% to 25.8% of both pregnant
and throat washings from an infected child to an uninfected and nonpregnant women, implying possible sexual transmis-
child and to rhesus monkeys (Macaca mulatta). sion of HHV-6 as well (3235). HHV-6 DNA was not detected
in 120 specimens of breast milk, making breast-feeding an
unlikely mode of transmission of HHV-6 (36).
Congenital infection with HHV-6, defined as the presence
EPIDEMIOLOGY of viral DNA in cord blood specimens, has been documented
in 1.19% (exact CI 1.02, 1.38) of newborns (37). Eighty-
Seroprevalence six percent of congenital infections are due to chromosomal
integration. Transplacental HHV-6 infection accounts for the
Worldwide surveys have demonstrated ubiquitous infection remaining 14% of congenital infections (38). Chromosomal
with HHV-6. Seroprevalence rates of more than 80% have integration is unique to HHV-6 among the human herpesvi-
been reported consistently in various populations of adults by ruses and has been identified in 0.85% of unselected subjects
several methods (1416). Age-specific studies have revealed (39). In individuals with chromosomal integration of HHV-6
that antibody to HHV-6 is universally present in cord blood (ciHHV-6), the entire viral genome is present in every nucle-
and newborn sera at titers higher than in maternal sera, with ated cell of the body, including the germline, and transmitted
a gradual fall in geometric mean titers and seroprevalence via Mendelian genetics (40). Fluorescent in situ hybridization
noted in the first 4 months of life, corresponding to the loss has demonstrated that multiple chromosomes may harbor

191

Scheld_Ch13.indd 191 2/21/14 6:39 PM


192 Part II: Viral Infections and Related Disorder

FIGURE 13.1 Schematic comparing the genome structure of HHV-6 to inherited HHV-6 (iHHV-6).
A: The linear HHV-6 genome contains a unique long (UL) region of 143 kb with left and right direct
repeats (DR) of 8 kb at the termini. Perfect and imperfect telomere sequence (TTAGGG) positioned at
the end of DR play a role in HHV-6 chromosome integration into the telomere. B: Fluorescence in situ
hybridization (FISH) of PBMCs from a patient with iHHV-6A integrated into the telomere of chromo-
some 22q. Metaphase chromosomes counterstained with DAPI (blue), cy5-PNA telomere probe (red),
and FITC-conjugated HHV-6 cosmid probe (green). C: Genome structure of iHHV-6 in which HHV-6
DRR is fused with the telomere repeats near the chromosome subtelomere and the DRL fused with the
remaining telomere repeat array (TTAGGG)n. (From Arbuckle JH, Medveczky PG. The molecular biol-
ogy of human herpesvirus-6 latency and telomere integration. Microbes Infect. 2011;13:731741, with
permission. Copyright 2011 Elsevier Masson SAS. All rights reserved.)

integrated virus; however, the viral genome is always identi- analysis, and restriction fragment length polymorphisms
fied at the proximal end of the telomere with one site usually (7,47,48). In North America, Europe, and Japan, HHV-6B
identified per family (41,42). The HHV-6 U94 gene product was found in approximately 97% of both healthy and immu-
is thought to play a role in HHV-6 latency and in mediating nocompromised hosts and in infants during primary infection
HHV-6 chromosomal integration via homologous recombi- (49,50). Recent PCR data have suggested that HHV-6A is the
nation between the perfect telomere repeat sequences (TRS) predominant virus identified in children in sub-Saharan Africa
found in the direct repeat segments of the HHV-6 genome and (51). In infants identified with ciHHV-6 in the United States,
human telomeres (43) (Fig. 13.1). approximately one third had HHV-6A, a frequency substan-
Individuals with ciHHV-6 are notable for having persis- tially higher than in children with primary infection (21,38).
tently high HHV-6 DNA viral loads in whole blood, serum, Dual infection with HHV-6A and HHV-6B has also been docu-
plasma, and hair follicle specimens, with one or more genome mented in both adults and children. Because of the differences
copies per cell (44). Although replicating virus can be induced noted in epidemiology, tissue tropism, genome composition,
from cultured lymphocytes of individuals with ciHHV-6 via disease associations, and possible virulence between HHV-6A
treatment with histone deacetylase inhibitors, it is unknown and HHV-6B, the two were reclassified as separate species
whether the production of infectious viral particles occurs in in the genus Roseolovirus by the International Committee on
vivo (41). The observation that infants with transplacental Taxonomy of Viruses in 2012. Further clarification of the epi-
HHV-6 infection have mothers with ciHHV-6 suggests that demiology of HHV-6A and HHV-6B awaits the development
integrated virus may be able to replicate as free virus and be of reliable species-specific serologic assays (52,53).
transmitted (45). No clinical manifestations have been rec-
ognized in infants with ciHHV-6 and the long-term clinical
significance of chromosomally integrated HHV-6 is unknown. VIRAL CHARACTERISTICS
Preliminary data have demonstrated the rate of ciHHV-6
in groups of solid organ transplant patients and those with HHV-6 contains a linear, double-stranded DNA genome of
malignancies to be approximately double that found in the 160 to 170 kb that consists of a single unique sequence of
general population suggesting possible pathogenic potential; approximately 143 kb bounded by a single direct repeat (DR)
however, this has yet to be confirmed (39). at both termini. The complete sequences of representative
isolates of both HHV-6A and HHV-6B have been published
and shown to be approximately 90% identical at the nucleo-
Taxonomy tide level (5456). The seven conserved gene blocks shared by
all HHVs are organized in an identical fashion in HHV-6A,
Soon after the initial description of HHV-6, two groups of HHV-6B, HHV-7, and CMV, and the unique segment of both
viruses were identified: HHV-6 variant A and variant B (46). the HHV6A and HHV-6B genomes are colinear with the long
Both were included as members of the Roseolovirus genus unique segment of CMV (7). In addition, CMV, HHV-6A,
in the Betaherpesvirinae subfamily, along with HHV-7 and HHV-6B, and HHV-7 share a unique set of genes called beta
human cytomegalovirus (CMV) (7). The two HHV-6 vari- genes that are found only in CMV, HHV-6A, HHV-6B, and
ants were found to be highly genetically related but could HHV-7 (7).
be distinguished by the pattern of reactivity with monoclo- HHV-6 displays the typical ultrastructural characteristics of
nal antibodies, in vitro cellular tropism, nucleotide sequence the herpesvirus family. It is an enveloped virus, approximately

Scheld_Ch13.indd 192 2/21/14 6:39 PM


Chapter 13: Human Herpesvirus-6 193

Santoro et al. (66) described CD46 (i.e., membrane cofac-


tor protein) as an essential cellular receptor for HHV-6. CD46
is also the receptor for the Edmonston strain of measles virus
and other bacterial microbes and functions as a complement
regulatory protein present on the surface of all nucleated cells.
This finding is in keeping with the observation that HHV-6
can infect a broad range of cell types in vitro including primary
T cells, monocytes, natural killer (NK) cells, dendritic cells,
oligodendrocytes, microglia, and astrocytes, with infection of
T-cell, B-cell, megakaryocytic, endothelial, glial, and epithelial
cell lines described (4,7,60,6774). Variability has been noted
in infectivity between HHV-6A and HHV-6B (69). HHV-6A
viruses are most efficiently propagated in the HSB2 or J-Jhan
T-cell lines, whereas HHV-6B isolates can be adapted to grow
in the Molt-3 T-cell line. A viral glycoprotein complex of gH,
gL, gQ1, and gQ2 has been demonstrated to be the viral li-
gand for many, but not all, laboratory isolates of HHV-6A
and HHV-6B (75). Given these observations, the possibility of
a second cellular receptor that could explain the differential
tropism between HHV-6A and HHV-6B has been postulated.
Ablashi et al. (60) studied the kinetics of host cell destruc-
FIGURE 13.2 An extracellular human herpesvirus type 6 virion. tion in T-cell lines and reported that HHV-6A isolates demon-
c, capsid; d, DNA core; e, envelope; s, surface spikes; t, tegument. strated enhanced cytopathogenicity compared with HHV-6B
(From Kramarsky B, Sander C. Electron microscopy of human herpes- isolates suggesting greater virulence of HHV-6A. Despite the
virus-6 [HHV-6]. In: Ablashi DV, Krueger GRF, Salahuddin SZ, eds. broad tropism of HHV-6 noted in vitro, viral replication in
Human Herpesvirus-6: Epidemiology, Molecular Biology and Clinical nonlymphoid cell lines appears to be substantially restricted
Pathology. Amsterdam: Elsevier; 1992:5966, with permission.) with decreased production of infectious virus (58,76).
A previous study of PBMCs from children with roseola
suggested that HHV-6 acutely infects primarily mature CD4
T cells in vivo (77). However, higher viral loads and evidence
160 to 200 nm in diameter, and contains an electron-dense of viral gene transcription have been demonstrated in mono-
core. The core is surrounded by a nucleocapsid with icosahe- cytes and macrophages compared with CD4 T cells during
dral symmetry with a prominent tegument between the nu- the acute phase of primary HHV-6 infection (78). Following
cleocapsid and outer lipid membrane (57) (Fig. 13.2). HHV-6 primary infection, the HHV-6 genome persists in PBMCs with
viral replication occurs in the nucleus of the infected cell. Viral evidence suggesting that a true state of viral latency is induced
particles are initially released by exocytosis and then cell death. in cells of the myeloid lineage including CD34 stem cells,
Both HHV-6A and HHV-6B have the potential to encode which may function as a reservoir of latent virus (79,80).
for approximately 97 proteins including those involved in reg- The neuroinvasive potential of HHV-6 has been docu-
ulatory functions, DNA replication, and structural proteins as mented in several reports by the detection of HHV-6 DNA in
well as several unique genes not found in other herpesviruses. 32% to 85% of brain tissue specimens from immunocompe-
Many of these gene products have been identified and re- tent adults at autopsy (8183). Chan et al. (84) sampled areas
viewed previously and thus are not mentioned here (7,48,58). of the cerebellum, frontal, temporal, parietal, and occipital
One notable exception is HHV-6 U94, a protein that is homol- lobes bilaterally from 40 patients postmortem and found that
ogous to the parvovirus adeno-associated virus type 2 (AAV-2) 85% had one or more positive specimens. However, there was
Rep 68/78 gene that has been implicated in the maintenance no association between the presence of HHV-6 DNA and the
of latency and chromosomal integration as noted earlier (59). location of the sample, suggesting that HHV-6 may be widely
disseminated in the human brain (84). In addition, HHV-6
DNA has been found in gray matter and white matter samples
PATHOGENESIS and in primary brain tumor tissue (82,85). Primary infection
with HHV-6 identified by viral isolation from PBMCs is due
almost exclusively to HHV-6B. In contrast, HHV-6A DNA
Viral Growth Characteristics and was detected in 12% to 70% of human brain tissue specimens
Cellular Tropism in the studies described previously, with up to 21% of patients
harboring both HHV-6A and HHV-6B in different regions of
All HHV-6 isolates can be grown in the laboratory in primary the brain (81,82,84,86). In vitro studies have demonstrated
cord blood mononuclear cell cultures that have been pretreated HHV-6A and HHV-6B infection of primary astrocytes and
with phytohemagglutinin (PHA) (60). HHV-6 has also been oligodendroglia with the formation of multinucleated syncytia
propagated in adult PBMCs that have been stimulated with (70,87). Attempts to infect neuronal cells have led to conflict-
monoclonal antibody to CD3 but not PHA (61). HHV-6 rep- ing results but generally suggest these cells are not a signifi-
lication in vitro is augmented by T-cell activation and enhanced cant source of productive infection (67,68,87). Enhanced
in the presence of low concentrations of interleukin-2 (IL-2), neurovirulence of HHV-6A has been suggested by several in
with increasing concentrations producing inhibition (62,63). vitro studies examining the degree of cytopathic effect, DNA
Following infection, HHV-6 reproducibly induces a cytopathic replication, induction of apoptosis, and inhibition of cell pro-
effect characterized by large refractile mononucleated or binucle- liferation induced by HHV-6A compared to HHV-6B in both
ated cells with intracytoplasmic and/or intranuclear inclusions primary glial cells and established cell lines (67,8891).
(64). Infected cells exhibit a slightly prolonged life span in PHA- The clinical relevance of detecting HHV-6 DNA in cerebro-
stimulated cultures compared with uninfected controls; however, spinal fluid (CSF) samples has not been fully clarified. HHV-6
lytic infection of mononuclear cells has been the rule (65). DNA has been detected in the CSF of one quarter to one half of

Scheld_Ch13.indd 193 2/21/14 6:39 PM


194 Part II: Viral Infections and Related Disorder

children with primary infection, indicating that viral DNA may 1 week (105). The IgM response is short lived, persisting for
be present in the CSF of a substantial number of immunocom- only 2 to 3 weeks, and the IgG response lasts for extended
petent children during primary infection (9294). In one report periods. Low level neutralizing antibody develops at approxi-
of young febrile children with evidence of past HHV-6 infec- mately day 3 of illness, coincident with a falloff in the rate
tion, approximately 30% had detectable HHV-6 DNA present of viral isolation, and peaks at 2 to 4 weeks into the illness
in the CSF (92). The CSF was the only site of viral DNA detec- (106,107). Although the immune response to HHV-6 is gen-
tion in 29% of children, and these children were significantly erally sensitive and specific, IgM has also been detected in
younger than those with HHV-6 DNA in both the CSF and the approximately 5% of healthy adults and has been shown to
PBMCs, indicating that the CNS may be a preferred site of la- be cross reactive with HHV-7, limiting the usefulness of this
tency in younger children (92). Typing of CSF samples revealed test for the diagnosis of HHV-6 infection or reactivation (108).
a greater frequency of HHV-6A in CSF (14%) than in PBMCs HHV-6specific CD4 and CD8 T-cell responses have also
(5%), also implying a greater neurotropism of HHV-6A (50). been described, supporting a role for cellular factors in the
These data imply that finding HHV-6 DNA in the CSF of nor- immune response to HHV-6 (109,110).
mal hosts is not a rare event. However, in a single report of Following infection of PBMCs, HHV-6 interacts with the
bone marrow transplant (BMT) recipients, HHV-6 DNA was immune system in several ways including downregulation of
identified in 22% of CSF samples from patients with symptoms CD3 and CD46 cell surface molecule expression, upregula-
of CNS dysfunction compared with only 1% of control patients tion of CD4, induction and suppression of cytokines and type
with leukemia (95). Unfortunately, the cases and controls in this I interferons, and effects on NK cell activity (107,111114).
study were not well matched by degree of immunosuppression. In vitro HHV-6 infection of dendritic cells has been described
Nevertheless, these data suggest that detecting HHV-6 DNA in with conflicting reports on the functional outcome of this
CSF may be a useful marker of disease in the proper clinical set- infection (115,116). Apoptosis of T cells both in vitro and in
ting. Part of the discrepancy between the results from studies of vivo has been detected following HHV-6 infection, as well as
children with primary infection and the BMT patients may be the previously well-described cell lysis (117,118). In addition
due to the young age of the immunocompetent children studied to the immune response generated to HHV-6, the viral genome
and the significant percentage with primary HHV-6 infection itself encodes several factors that have immunologic regulatory
compared to the older group of BMT patients. Given the recent activity including two chemokine receptor homologs (U12 and
identification of ciHHV-6 and the fact that most subjects with U51) and two putative chemokines (U22 and U83) (111). It
ciHHV-6 have no symptoms referable to the integrated virus, has been postulated that through these proteins, HHV-6 may
caution should be used in interpreting the presence of HHV-6 either enhance viral replication in infected cells, enlarge the
DNA in CSF in order to avoid misdiagnosis of active HHV-6 pool of available virus susceptible cells, or circumvent the host
infection in an individual with ciHHV-6 (39,93). Obtaining immune response to infection.
HHV-6 viral load measurements in blood and CSF concur-
rently may be able to help distinguish between these different
clinical scenarios (93). Studies of healthy adults have demon-
strated the presence of HHV-6 DNA in saliva, PBMCs, urine,
CLINICAL MANIFESTATIONS
and vaginal secretions, with evidence of continued presence of
the viral genome at these sites over several months consistent Primary Infection
with the broad in vitro tropism of HHV-6 (32,9698). Despite
initial reports of isolation of HHV-6 from adult saliva, sub- Roseola is an acute self-limited disease of infancy and child-
sequent studies have shown that viral isolates obtained from hood. It is characterized by the abrupt onset of high fever
saliva are HHV-7 (99). Additionally, attempts at isolating infec- lasting approximately 72 hours. The fever resolves by crisis
tious HHV-6 from healthy subjects PBMCs have been gener- coincident with the appearance of a faint morbilliform rash
ally unsuccessful. In contrast, isolates can be readily obtained on the neck and trunk. Associated signs are notably sparse
from immunocompromised hosts, demonstrating reactivation but can include mild injection of the pharynx, palpebral con-
of latent HHV-6 (100). Reactivation documented either by junctivae, or tympanic membranes and enlarged suboccipital
HHV-6 viral isolation or by quantitative DNA PCR has also nodes (11).
been reported in immunocompetent children with measles or Prospective studies have extended our knowledge of the
dengue hemorrhagic fever and in adults with critical illness re- clinical manifestations of acute HHV-6 infection beyond that
quiring intensive care unit care or those with drug induced hy- of classic roseola. Hall et al. (21) identified 160 children with
persensitivity syndrome (DIHS) or drug rash with eosinophilia primary HHV-6 infection by viremia and seroconversion from
and systemic symptoms (DRESS) (101104). The cumulative a large number of children evaluated in a pediatric emergency
data support the concept of active HHV-6 viral replication in department. The mean age at primary HHV-6 infection was
mononuclear cells during primary infection, with the produc- 9.4 months, with few cases occurring before 2 months or after
tion of abundant infectious particles. The virus then becomes 25 months of age (Fig. 13.3). High fever was the most con-
latent or persistent in the host PBMCs, salivary gland tissue, sistent manifestation of infection. All children had an acute
the genitourinary (GU) tract, and the CNS, with the detection febrile illness, and 87% had fever of more than 39C. Fever
of the HHV-6 genome in the absence of significant amounts of persisted for 6 days or longer in 15%. Associated clinical
infectious virus. Immune dysfunction allows for HHV-6 reacti- symptoms and signs included irritability or a toxic appearance
vation, as evidenced by renewed viral isolation with subsequent in 72%, inflamed tympanic membranes in 46%, and upper
disease manifestations in a subset of patients. respiratory tract signs in 41%. Children with primary HHV-6
infection were significantly more likely to exhibit these signs
than matched controls. Cough and lower respiratory tract
Immune Response signs were seen significantly less often in children with pri-
mary HHV-6 infection than controls. Only 17% of children
Children with primary HHV-6 infection develop an immu- with primary HHV-6 infection developed a rash, making it
noglobulin M (IgM) response on day 5 of illness, with the difficult to identify correctly the cause of the illness. Most chil-
appearance of immunoglobulin G (IgG) at approximately dren were given a diagnosis of fever with otitis media, fever

Scheld_Ch13.indd 194 2/21/14 6:39 PM


Chapter 13: Human Herpesvirus-6 195

30% 50
Visits Due to HHV-6 (% of All Visits)

Number with HHV-6 Infection


40

20%
30 FIGURE 13.3 Number of children pre-
senting to the emergency department
with illness from primary HHV-6 infec-
20 tion (bars) and visits due to such infec-
10% tion as a percentage of all emergency
department visits for acute febrile ill-
10 nesses (curve) according to age. (From
Hall CB, Long CE, Schnabel KC, et al.
Human herpesvirus infection in chil-
dren. A prospective study of complica-
0% 0 tions and reactivation. N Engl J Med.
02 35 68 911 1214 1517 1820 2123 24 1994;331:432438, with permission.)

of undetermined origin, or presumed sepsis, and 13% were patients (125,126). However, in the study by Zerr et al. (119)
hospitalized. A more recent prospective study of an outpatient noted earlier, no seizures were recorded among 81 ambulatory
cohort of infants with primary HHV-6 infection identified by children with primary HHV-6 infection. Although the exact
HHV-6 DNA detection in saliva confirmed that over 90% of rate of seizures due to primary infection with HHV-6 has not
the infants had symptoms with fever, fussiness, and rhinorrhea been precisely defined, a review of the viral causes of febrile
noted most often (119). Rash was identified in a little less than seizures examined almost 1,000 published cases of primary
one third of the cohort (119). HHV-6 infection and found seizures to be a complication in
In a report from Japan of 176 children with roseola identi- 16.5% (127).
fied retrospectively, fever and rash were noted in 98% of the Clinically, no differences have been identified between
patients (120). This study also detected a higher percentage children with febrile seizures associated with primary HHV-6
of children with gastrointestinal tract disturbances (68% with infection and those with febrile seizures not associated with
diarrhea), in addition to edematous eyelids in 30%, erythema- primary HHV-6 infection, including the height or duration of
tous papules in the pharynx in 65%, cough in 50%, and mild fever or the recurrence rate of seizures in the first 24 hours
cervical lymphadenopathy in 31%. The differences between (125). Additionally, CSF analysis does not typically reveal
these studies may be attributed in part to study design and case pleocytosis or abnormal glucose or protein levels in patients
definition. Also, the severity of the clinical symptoms associ- with primary HHV-6 infection. Suga et al. (128) reported a
ated with acute HHV-6 infection has been shown to be related higher frequency of partial seizures, postictal paralysis, pro-
to the degree of viremia, which may contribute to the range longed seizures, and clustering of seizures in 19 children
of reported manifestations of primary HHV-6 infection (121). with primary HHV-6 infection compared with children with
The laboratory evaluation of children with primary HHV-6 seizures not associated with primary HHV-6 infection, sug-
infection is generally nonspecific and reveals a depressed total gesting more significant CNS pathology and implying an in-
white blood cell count, with lymphopenia noted early in the creased risk for the later development of epilepsy (92,129).
course of the infection. C-reactive protein levels have been Early reports did suggest an increased risk of recurrent febrile
reported to be less than 10 mg/L in most patients (21,122). seizures in children due to HHV-6 infection, but a casecontrol
Rarely reported complications of primary HHV-6 infection study of children with febrile seizures associated with primary
include intussusception, Kawasaki disease, hepatitis, hemo- HHV-6 infection compared with children with febrile seizures
phagocytic syndrome, and fatal multisystem disease (7,123). not associated with primary HHV-6 infection demonstrated
A heterophil-negative mononucleosis syndrome has been associ- a statistically significant lower rate of seizures over the fol-
ated with primary HHV-6 infection in immunocompetent adults lowing 3 years in the children with primary HHV-6, casting
(124). However, most reports have relied on seroconversion as doubt on this association (130). However, more recent data
evidence of HHV-6 infection, a method that is not optimal. have suggested a link between chronic smoldering or reacti-
vated HHV-6 infection of the CNS and the later development
of medial temporal lobe epilepsy (MTLE) (131). One line of
Central Nervous System Complications of evidence supporting this association comes from a prospec-
Primary Infection tive study of children with febrile status epilepticus (FSE), a
known risk factor for the development of MTLE that identi-
Seizures have been reported as a complication of primary fied active HHV-6 infection in peripheral blood samples of
HHV-6 infection. In the study from Japan noted earlier, 8% of almost one third of the patients (132). Similar to the data
the children with roseola had seizures during the febrile phase from children with simple febrile seizures, the children with
of the illness (120). Seizures were the most common complica- FSE associated with HHV-6 did not have CSF pleocytosis or
tion of primary HHV-6 infection, occurring in 13% of chil- differences in peripheral white blood cell counts from pa-
dren with acute infection described by Hall et al. (21) from a tients with FSE not associated with HHV-6. There were also
pediatric emergency department population. The highest inci- no differences in the proportion of children with EEG ab-
dence of seizures was 36%, noted in children 12 to 15 months normalities or acute hippocampal changes on MRI. Seizure
of age. This rate was significantly greater than that of age- characteristics between children with FSE associated with
matched controls with other febrile illnesses. In prospective HHV-6 and those not associated with HHV-6 were similar
studies of children with first time febrile seizures, primary in- (Table 13.1), suggesting a shared pathogenesis of FSE among
fection with HHV-6 has been observed in 18% to 26% of the different etiologies (132).

Scheld_Ch13.indd 195 2/21/14 6:39 PM


196 Part II: Viral Infections and Related Disorder

TA B L E 1 3 . 1
DESCRIPTIVE INFORMATION FOR CHILDREN WITH FSE WITH HHV6 OR HHV7 INFECTIONS (PRIMARY OR
REACTIVATED) AT BASELINE COMPARED TO CHILDREN WITH PRIOR OR NO HHV6 AND HHV7 INFECTIONS

HHV6 or HHV7 HHV6 and HHV7


Total or Reactivation or Prior Infections p
Feature (N 169) (n 58) (n 111) Value

Age (months)
Media (interquartile range) 15.8 (12.023.2) 14.8 (12.122.2) 17.4 (12.025.2) 0.67
Age 18 months 71 (42.0%) 20 (34.5%) 51 (46.0%) 0.15
Gender
Male 88 (52.1%) 30 (51.7%) 58 (52.2%) 0.95
Female 81 (47.9) 28 (48.3%) 53 (47.8%)
Development
Normal 148 (87.6%) 47 (81.0%) 101 (91.0%) 0.06
Suspect 10 (5.9%) 7 (12.1%) 3 (2.7%)
Abnormal 11 (6.5%) 4 (6.9%) 7 (6.3%)
Prior febrile seizures
Yes 35 (20.7%) 12 (20.7%) 23 (20.7%) 1.00
No 134 (79.3%) 46 (79.3%) 88 (79.3%)
Sick behavior before FSEa
Yes 78 (46.4%) 25 (43.1%) 53 (48.2%) 0.53
No 90 (53.6%) 33 (56.9%) 57 (51.8%)
Duration of recognized fever before FSE
1 h 74 (43.8%) 28 (48.3%) 46 (41.4%) 0.68
124 h 68 (40.2%) 21 (36.2%) 47 (42.3%)
24 h 27 (16.0%) 9 (15.5%) 18 (16.2%)
Peak temperature during illness
Mean peak temperature F (SD) 103.3 (1.3) 103.2 (1.0) 103.3 (1.5) 0.59
Mean ED peak temperature F (SD)a 102.2 (1.9) 102.4 (1.6) 102.0 (2.0) 0.22
Duration of FSE (min)
Median (interquartile range) 70.0 (47.0110.0) 67.5 (45.0105.0) 70.0 (50.0120.0) 0.30
Continuous versus intermittent FSE
Continuous 95 (56.2%) 29 (50.0%) 66 (59.5%) 0.50
Intermittent without full recovery 53 (31.4%) 21 (36.2%) 32 (28.8%)
in between
Intermittent with full recovery 21 (12.4%) 8 (13.8%) 13 (11.7%)
in between
Focal FSE
Yes (definitely or probably) 113 (66.9%) 44 (75.9%) 69 (62.2%) 0.07
No (maybe or no) 56 (33.1%) 14 (24.1%) 42 (37.8%)
Family history of FS or FSEa
Yes 52 (31.0%) 21 (36.2%) 31 (28.2%) 0.28
No 116 (69.0%) 37 (63.8%) 79 (71.8%)

FSE, febrile status epilepticus; FS, febrile seizure.


a
One with missing information.
From Epstein LG, Shinnar S, Hesdorffer DC, et al. Human herpesvirus 6 and 7 in febrile status epilepticus: the FEBSTAT study. Epilepsia.
2012;53:14811488.

Scheld_Ch13.indd 196 2/21/14 6:39 PM


Chapter 13: Human Herpesvirus-6 197

Further data supporting a link between HHV-6 and MTLE earlier, HHV-6 reactivation has been reported in children with
comes from a series of studies of adult and adolescent patients measles or dengue hemorrhagic fever and in seriously ill adults
undergoing epilepsy surgery. HHV-6B DNA was found in in an intensive care unit or with DIHS or DRESS. Evidence
28% to 63% of resected temporal lobe specimens in patients of reactivation as determined by quantitative PCR of HHV-6
with MTLE compared to zero specimens from patients with DNA in PBMCs in the critically ill adult patients was not asso-
other forms of epilepsy or 8% of specimens obtained from ciated with any specific symptoms and did not have an impact
trauma patients (133137). Active HHV-6 replication was on mortality. However, it is notable that all but one of these
identified via the detection of higher viral loads of HHV-6 in patients had HHV-6A detected, which may actually have been
the hippocampus than the surrounding tissue, HHV-6 protein due to ciHHV-6 and not true viral reactivation given the appar-
expression via IHC, and HHV-6 messenger RNA (mRNA) pro- ent low prevalence of HHV-6A in the general U.S. population
duction in the hippocampus and temporal lobe tissue as well (103). Reactivation of HHV-6 does occur in immunocompro-
as cultured astrocytes from the resected MTLE patient speci- mised hosts of all ages and has been associated with a range of
mens (133,134,136). Loss of glutamate transporter EAAT2 illnesses in this population. Following hematopoietic cell trans-
protein in infected astrocytes due to active HHV-6 infection plantation (HCT), HHV-6 reactivation documented by viral
with a resultant inability to process extracellular levels of the culture or quantitative PCR has been documented in 35% to
excitatory neurotransmitter glutamate is one proposed mecha- 65% of patients (150,151). Several diseases including fever and
nism of pathogenesis (131,134). Alternatively, increased lev- rash, encephalitis, interstitial pneumonitis, acute graft-versus-
els of expression of NFB have been detected in hippocampus host disease, delayed engraftment or myelosuppression, CMV
and temporal lobe specimens from MTLE patients suggesting disease, and increased mortality have been associated with
a role for inflammation in the development or progression of HHV-6 reactivation in the HCT population (48,100,152). Two
MTLE (136). of the best documented diseases linked to HHV-6 reactivation
Several older case reports have described encephalitis or in HCT recipients are fever with rash and encephalitis, occur-
encephalopathy in children with roseola (138). Fever, de- ring from 2 to 4 weeks after transplantation (48,100).
pressed level of consciousness, and seizures were the promi- HHV-6 reactivation has also been associated with both di-
nent symptoms noted in these patients, with normal CSF rect and indirect clinical effects in approximately 1% of solid
findings or mild pleocytosis with elevated protein levels. organ transplant recipients including fever, rash, encephalitis,
Computed tomography (CT) was often normal at onset but bone marrow suppression, interstitial pneumonitis, gastroin-
progressed to reveal hypodense lesions in multiple areas of testinal infections, CMV disease, fungal infections, and graft
the cerebral cortex, including the frontal, parietal, temporal, rejection or dysfunction (153162). Many of these reports
and occipital lobes, the putamen, and internal capsule (138 provide conflicting results and older studies often relied on se-
140). A single report including magnetic resonance imaging rologic markers of reactivation. In prospective studies of HCT
(MRI) results demonstrated multiple lesions of demyelination recipients that have included both serology and viral culture or
in the thalamus and parietooccipital deep white matter (141). quantitative PCR, the serologic results did not correlate with
Electroencephalographic (EEG) findings were either normal viral assays, suggesting caution in diagnosing reactivation of
or nonspecific, with most studies remarkable only for dif- HHV-6 based on serology alone (163165).
fuse slowing following seizures. Single photon emission CT Encephalitis associated with HHV-6 reactivation has been
(SPECT) performed in one patient showed diffuse hypoperfu- reported in both immunocompetent and immunocompromised
sion of the left hemisphere that persisted for at least 50 days hosts. Single case reports or small series have described disease in
(142). Although complete or near-complete recovery was previously healthy adults and children (166173). The quality of
noted in several cases, mortality was also reported. More re- these reports varies substantially and no clear group of symptoms
cent reports of young children with probable primary infection or unique clinical presentation has been identified. Clinical char-
with HHV-6 have described a syndrome of rhombencephalitis acteristics have included both acute and subacute presentations,
with progressive neurologic symptoms including ataxia and diffuse headache, altered mental status, and seizures as well as
cranial nerve deficits associated with imaging abnormalities of focal neurologic signs. Variable degrees of CSF pleocytosis and
the brainstem and cerebellum (143,144). Pleocytosis, HHV-6 protein elevations have been reported. Imaging studies have been
DNA in the CSF, and elevated CSF total protein values were normal or with a range of findings including focal abnormalities
found with identification of HHV-6 protein by IHC in a brain in the motor cortex, parietal lobe, temporal lobe, or white matter
biopsy sample from a single patient (143). Other reports from disease in the pons, internal capsule, and midbrain (166174).
single cases of encephalitis have not identified a unique patho- Outcomes have been mixed with both death and full recovery
logic description from brain tissue specimens and have re- reported. One point that appears consistent among these cases is
ported inconsistent results of DNA and IHC testing suggesting the identification of HHV-6 DNA in CSF as the main diagnostic
a possible immune-mediated pathogenesis (145,146). In chil- criteria. Given the relatively recent recognition of ciHHV-6 and
dren with primary HHV-6associated encephalitis or encepha- the persistent detection of HHV-6 DNA in all body fluids associ-
lopathy with or without HHV-6 DNA in the CSF, significantly ated with viral integration, it is possible that several of the earlier
greater concentrations of both interleukin (IL)-6 and IL-8 have reports were confounded by ciHHV-6. Alternatively, individuals
been identified in serum and CSF compared to values from with ciHHV-6 may develop encephalitis due to the integrated
patients with HHV-6associated febrile seizures or normal virus as has been suggested in a prior report (175). The ques-
controls supporting this hypothesis (147149). tion of whether HHV-6 reactivation can cause encephalitis in
immunocompetent individuals with ciHHV-6 or prior childhood
infection requires further careful study.
Reactivation Encephalitis associated with HHV-6 reactivation in HCT
recipients has been well defined with the majority of reports
The lifelong consequences of infection with HHV-6 are still not describing a syndrome of acute limbic encephalitis also re-
well known. Although HHV-6 DNA can be detected in several ferred to as posttransplantation acute limbic encephalitis
body sites in adults years after primary infection, the occurrence (PALE) (176181). In addition, HHV-6 reactivation has been
and frequency of reactivation, as well as the clinical relevance temporally associated with more common CNS symptoms
of reactivation, have not been completely defined. As noted including delirium during the early posttransplant period and

Scheld_Ch13.indd 197 2/21/14 6:39 PM


198 Part II: Viral Infections and Related Disorder

neurocognitive decline measured at 12 weeks after transplant, an association between high level viremia (25,000 genome
suggesting a significant impact on the clinical course of trans- copies/mL of whole blood) and CNS symptoms. Patients with
plantation (182). HHV-6 reactivation were more likely to have mental status
The incidence of PALE among HCT recipients is es- changes, and it is notable that the only patient who died with
timated at 1.5% overall but may be as high as 28% in pa- neurologic complications had the highest level of HHV-6 vire-
tients receiving two or more cord blood stem cell transplants mia among patients with CNS symptoms (185).
(176,177,179). PALE typically develops around the time of Wainwright et al. (180) were the first to describe five pa-
neutrophil engraftment, usually from 2 to 6 weeks after trans- tients with limbic encephalitis associated with HHV-6 reactiva-
plantation (176,179,181). Risk factors for the development of tion. Since then, multiple reports have confirmed the typical
PALE appear to be similar to the risk factors identified for clinical presentation of PALE as an acute onset of altered men-
HHV-6 reactivation in HCT and include umbilical cord blood tal status with confusion and prominent anterograde amnesia
as a donor source, human leukocyte antigen (HLA) mismatch, (176,179,181,183). Fever, insomnia, hallucinations, agitation,
time-dependent acute graft-versus-host disease, and the receipt or emotional lability have also been described (179,186).
of corticosteroids during the first 4 weeks after transplanta- Seizures are frequently identified either clinically or via EEG
tion (176,181,183). Hill et al. (176) have reported that all monitoring. Reported laboratory findings include the syndrome
patients who develop PALE have evidence of HHV-6 reactiva- of inappropriate antidiuretic hormone with minimally elevated
tion with positive DNA detection in plasma (176). In addi- levels of CSF total protein (176,179181). CSF pleocytosis is
tion, patients with PALE have been found to have significantly noted less commonly, probably due to the marked cytopenias
higher HHV-6 viral loads measured in plasma than patients found posttransplantation (176,179181). MRI studies may
with HHV-6 reactivation without neurologic symptoms with be normal early in the course of disease but typically show
the peak viral load correlating with the time of development focal nonenhancing abnormalities in the medial temporal lobe
of neurologic symptoms (176,183,184). Various plasma often including edema in the hippocampus, uncus, and amyg-
viral load cutoff values have been proposed as a threshold dala either unilaterally or bilaterally on T2 fluid attenuation
for predicting PALE with variable sensitivity and specificity inversion recovery (FLAIR) images with hypermetabolism in
(176,183). However, not all studies have consistently noted these regions on positron emission tomography (PET) scan-
an association between HHV-6 viral loads and the develop- ning (186,187) (Fig. 13.4). Extension of abnormalities beyond
ment of disease. Betts et al. (185) performed a prospective the medial temporal lobe into other limbic structures or ex-
surveillance study for HHV-6 reactivation following HCT tralimbic areas has also been described (176,186,188). A small
in 82 subjects and measured weekly whole blood viral loads. number of postmortem examinations have been performed
They did not find an association between HHV-6 reactivation with neuronal loss and gliosis limited to the hippocampus and
and total CNS symptoms, including headache. Nor was there amygdala regions (179,180). HHV-6B DNA has been identified

FIGURE 13.4 Matched level transverse slices showing acute (AD) and chronic (EH) phase morpho-
functional imaging in human herpesvirus 6 limbic encephalitis: postcontrast T1-weighted (A, E) and fluid-
attenuated inversion recovery (B, F) 3-T magnetic resonance imaging (MRI), fused MRI PET images
(C, G), and fluorodeoxyglucose positron emission tomography (FDG-PET) (D, H). On acute phase imag-
ing acquired 10 days after the beginning of neurologic signs and symptoms, FDG-PET shows bilateral in-
tense FDG uptake in both hippocampi and amygdalae (C, D). At the same time, MRI showed only a mild
increased signal on fluid-attenuated inversion recovery sequence (B) without contrast enhancement (A).
The follow-up study, done after 3 months, showed FDG hypometabolism (G, H) associated with a hippo-
campal sclerosis (E, F). (From Hubele F, Bilger K, Kremer S, et al. Sequential FDG PET and MRI findings
in a case of human herpes virus 6 limbic encephalitis. Clin Nucl Med. 2012;37:716717, with permission.)

Scheld_Ch13.indd 198 2/21/14 6:39 PM


Chapter 13: Human Herpesvirus-6 199

in the CSF in the majority of patients with PALE and in one (IFA), neutralization assay, immunoblot assay, and enzyme-linked
autopsy specimen HHV-6 protein was detected within astro- immunosorbent assay (ELISA) (199,200). Although antibody to
cytes and neurons in the hippocampal areas (180). A role for a HHV-7 limits specificity at low titer, higher titer responses have
direct viral effect on disease pathogenesis is also supported by been shown to be reliable (200). IFAs that include the measure-
data from a small study demonstrating that the viral load of ment of antibody avidity have also been described and have the
HHV-6 in CSF of HCT patients with encephalitis was signifi- added benefit of being able to provide an approximate timing of
cantly greater than from children with primary infection (149). primary infection (200). The IgM response to HHV-6 is short
However, similar to findings in children with primary HHV-6 lived, cross reactive with HHV-7, and can reappear after initial
infection and encephalitis or encephalopathy, elevated levels of infection diminishing the use of this test in the clinical diagnosis
CSF IL-6, IL-8, and IL-10 have been identified in patients with of primary infection or reactivation (108).
HHV-6 reactivation and associated limbic encephalitis com- Rapid diagnostic methods for the confirmation of HHV-6
pared to controls suggesting neuroinflammation as one alter- infection are not available. The detection of HHV-6 DNA in
native pathogenic mechanism (149). PBMC samples does not distinguish between latent infection
Mortality rates reported due to HHV-6associated PALE and active viral replication associated with primary infection
have varied from 0% to 50% but do not appear to be differ- or reactivation. Secchiero et al. (201) detected HHV-6 DNA in
ent than mortality in the general HCT population (176179). the serum of six of seven children with roseola, suggesting that
Differences may become more apparent when patients with this method was both sensitive and specific for the timely detec-
cord blood transplantation are evaluated separately (176). tion of active viral replication if performed on cell-free speci-
There does not appear to be an association between mortal- mens. In a separate report of 47 plasma samples obtained from
ity and viral load measurements in the CSF or plasma, time children with roseola, HHV-6 could be isolated in culture from
to antiviral treatment, or other clinical characteristics, sug- only 21% of the samples, implying that plasma contained infec-
gesting that although plasma viral loads may be useful as a tious virus in only a minority of patients with primary HHV-6
marker for disease development, they do not predict outcome infection (106). This was confirmed by Achour et al. (202) in a
(176178). Morbidity is common following PALE with ap- study demonstrating that HHV-6 DNA in cell-free specimens is
proximately 40% of patients reported to make a full recovery primarily due to the presence of free DNA, not viral particles,
(181). Most patients have persistent memory difficulties that and that detection in plasma or serum correlates well with the
limit their day-to-day function, and symptomatic generalized viral load in PBMC samples (202). However, the description
epilepsy has also been described (176179,181). of ciHHV-6 and the recognition that asymptomatic individuals
HHV-6 infection has also been etiologically linked to MS with ciHHV-6 have HHV-6 DNA detectable in all body fluids
and hypothesized to act as a trigger for disease development with high viral loads has limited the use of detecting HHV-6
(6,189). Studies have demonstrated a statistically significant DNA in plasma or serum as the sole test of active viral replica-
increased detection of HHV-6 proteins and both early and tion. Reverse transcriptase polymerase chain reaction (RT-PCR)
late gene mRNA in oligodendrocytes from brain specimens assays for HHV-6 have been developed to amplify mRNA cor-
of patients with MS compared to control brains and from MS responding to late structural proteins of the virus produced dur-
plaques compared to normal-appearing white matter (189,190). ing lytic infection in order to detect active replication (203,204).
An increased frequency of detection of HHV-6 DNA in serum The performance characteristics of each of these assays com-
and PBMC samples from patients with MS has also been de- pared to viral culture varies with the population being tested,
scribed, as well as an increased percentage of patients with MS but in general, the detection of U90 and U100 mRNA is 90%
who have positive IgM antibody assay results to protein p41/38 to 100% sensitive and 90% to 98% specific for primary and re-
(191,192). This last finding is tempered by a study that found activated HHV-6 infection (203205). Preliminary data suggest
no difference in antibody reactivity to recombinant HHV-6 p41 that approximately 5% to 15% of samples of PBMCs from in-
protein between patients with MS and controls and suggested dividuals with ciHHV-6 are positive in the U100 RT-PCR assay
that previous results may have been confounded by reactivity slightly diminishing the discriminatory power of this test but
to p38, a cellular protein (193). However, multiple reports have raising the possibility of viral protein production and or active
continued to describe an increased frequency of detection of replication of the integrated genome (205). The measurement
HHV-6 DNA in the serum of patients with MS and a correlation of HHV-6 viral loads in whole blood has been suggested as a
between clinical disease activity and HHV-6 DNA detection in means of differentiating between primary infection, reactiva-
serum as well as mRNA detection in whole blood was noted tion, and ciHHV-6 with viral loads in individuals with ciHHV-6
in a group of patients with relapsing and remitting MS over often greater than 5.5 log10/mL or at least one genome equiva-
time (194). Additionally, an increased prevalence of detection lent per cell with lower levels found in primary infection, reac-
of HHV-6A in serum has been reported in patients with MS, tivation, and during latency (39,206). Additionally, individuals
suggesting a potential association between HHV-6A and this with ciHHV-6 will have persistently high viral load measure-
disorder (194). These findings are in contrast to multiple studies ments in whole blood over time with HHV-6 DNA present in all
of CSF, serum, PBMCs, and brain tissue including demyelinated tissues including hair follicle specimens, which has been used in
plaques from patients with MS and controls demonstrating no research settings to identify subjects with integrated virus. From
difference in the detection of HHV-6 DNA by PCR (195198). a practical standpoint, it appears as though a combination of
The ability to compare results across studies is hampered by the DNA detection in plasma coupled with whole blood quanti-
methodologic differences between these reports, making it dif- tation may provide the best discriminatory power to identify
ficult to form a conclusion about the strength of the association different types of infection with HHV-6 because both of these
between HHV-6 and MS. assays are commercially available (39).

DIAGNOSIS TREATMENT
The diagnosis of primary HHV-6 infection is confirmed by isola- In vitro studies have evaluated the susceptibility of HHV-6 to
tion of the virus in culture coupled with seroconversion. Several available antiviral agents. Varying methods used in different
serologic methods have been developed for the detection of anti- experimental systems make absolute comparisons between
body to HHV-6 including an indirect immunofluorescence assay reports difficult. In general, however, the pattern of antiviral

Scheld_Ch13.indd 199 2/21/14 6:39 PM


200 Part II: Viral Infections and Related Disorder

inhibition of HHV-6 resembles that of HCMV, with ganciclo- HHV-6associated encephalitis in HCT recipients using gan-
vir, foscarnet, and cidofovir demonstrating effective in vitro ciclovir, foscarnet, or cidofovir, either alone or in combina-
activity (48,58). This pattern of antiviral susceptibility is in tion, with a suggestion of benefit in preventing CNS disease in
agreement with the results of in vitro assays and genomic anal- small trials of prophylaxis (208213). Unfortunately, in vitro
yses that have failed to identify a virally encoded thymidine resistance of HHV-6 to each of these drugs has been identified
kinase in HHV-6 and suggest that ganciclovir, foscarnet, and (58). Nevertheless, treatment of HHV-6associated PALE is
cidofovir may be of use in the clinical setting, whereas acyclo- recommended with ganciclovir or foscarnet suggested as first-
vir- and thymidine kinasedependent drugs should have little line agents for a minimum of 3 weeks (100).
effect (207).
Randomized controlled clinical trials aimed at evaluating
the effectiveness of antiviral agents in patients with primary ACKNOWLEDGMENTS
or reactivated HHV-6 infections are lacking, and no drug is
currently licensed for the treatment of HHV-6 infection or dis- The author is indebted to Andrew Schultz and Pam LaDuke
ease. Case reports have described the successful treatment of for their expert technical assistance.

References
1. Salahuddin SZ, Ablashi DV, Markham PD, et al. Isolation of a new 25. Cone RW, Huang ML, Ashley R, et al. Human herpesvirus 6 DNA in
virus, HBLV, in patients with lymphoproliferative disorders. Science. peripheral blood cells and saliva from immunocompetent individuals. J
1986;234:596601. Clin Microbiol. 1993;31:12621267.
2. Biberfeld P, Kramarsky B, Salahuddin SZ, et al. Ultrastructural character- 26. Di Luca D, Mirandola P, Ravaioli T, et al. Human herpesviruses 6 and 7 in
ization of a new human B lymphotropic DNA virus (human herpesvirus 6) salivary glands and shedding in saliva of healthy and human immunodefi-
isolated from patients with lymphoproliferative disease. J Natl Cancer Inst. ciency virus positive individuals. J Med Virol. 1995;45:462468.
1987;79:93341. 27. Fox JD, Briggs M, Ward PA, et al. Human herpesvirus 6 in salivary glands.
3. Ablashi DV, Salahuddin SZ, Josephs SF, et al. HBLV (or HHV-6) in human Lancet. 1990;336:590593.
cell lines. Nature. 1987;329:207. 28. Sada E, Yasukawa M, Ito C, et al. Detection of human herpesvirus 6 and
4. Caruso A, Rotola A, Comar M, et al. HHV-6 infects human aortic and human herpesvirus 7 in the submandibular gland, parotid gland, and lip
heart microvascular endothelial cells, increasing their ability to secrete pro- salivary gland by PCR. J Clin Microbiol. 1996;34:23202321.
inflammatory chemokines. J Med Virol. 2002;67:528533. 29. Rhoads MP, Magaret AS, Zerr DM. Family saliva sharing behaviors and
5. Yamanishi K, Okuno T, Shiraki K, et al. Identification of human herpesvi- age of human herpesvirus-6B infection. J Infect. 2007;54:623626.
rus-6 as a causal agent for exanthem subitum. Lancet. 1988;1:10651067. 30. Harberts E, Yao K, Wohler JE, et al. Human herpesvirus-6 entry into the
6. Yao K, Crawford JR, Komaroff AL, et al. Review part 2: human herpesvirus-6 central nervous system through the olfactory pathway. Proc Natl Acad Sci
in central nervous system diseases. J Med Virol. 2010;82:16691678. U S A. 2011;108:1373413739.
7. Braun DK, Dominguez G, Pellett PE. Human herpesvirus 6. Clin Microbiol 31. Kawaguchi S, Suga S, Kozawa T, et al. Primary human herpesvirus-6 infec-
Rev. 1997;10:521567. tion (exanthem subitum) in the newborn. Pediatrics. 1992;90:628630.
8. Ranger-Rogez S, Vidal E, Labrousse F, et al. Large-scale study suggests no 32. Caserta MT, Hall CB, Schnabel K, et al. Human herpesvirus (HHV)-6 and
direct link between human herpesvirus-6 and primary Sjgrens syndrome. HHV-7 infections in pregnant women. J Infect Dis. 2007;196:12961303.
J Med Virol. 1995;47:198203. 33. Dahl H, Fjaertoft G, Norsted T, et al. Reactivation of human herpesvirus 6
9. Reeves WC, Stamey FR, Black JB, et al. Human herpesviruses 6 and 7 in chronic during pregnancy. J Infect Dis. 1999;180:20352038.
fatigue syndrome: a case-control study. Clin Infect Dis. 2000;31:4852. 34. Leach CT, Newton ER, McParlin S, et al. Human herpesvirus 6 infection of
10. Zahorsky J. Roseola infantum. JAMA. 1913;61:446450. the female genital tract. J Infect Dis. 1994;169:12811283.
11. Breese B. Roseola infantum (exanthem subitum). NYSJ Med. 1941;41: 35. Okuno T, Oishi H, Hayashi K, et al. Human herpesviruses 6 and 7 in cer-
18541859. vixes of pregnant women. J Clin Microbiol. 1995;33:19681970.
12. Berenberg W, Wright S, Janeway C. Roseola infantum (exanthem subitum). 36. Dunne WM Jr, Jevon M. Examination of human breast milk for evi-
N Engl J Med. 1949;241:253259. dence of human herpesvirus 6 by polymerase chain reaction. J Infect Dis.
13. Kempe C, Shaw E, Jackson J, et al. Studies on the etiology of exanthema 1993;168:250.
subitum (roseola infantum). J Pediatr. 1950;37:561568. 37. Hall CB, Caserta MT, Schnabel KC, et al. Congenital infections with
14. Saxinger C, Polesky H, Eby N, et al. Antibody reactivity with HBLV (HHV- human herpesvirus 6 (HHV6) and human herpesvirus 7 (HHV7). J Pediatr.
6) in U.S. populations. J Virol Methods. 1988;21:199208. 2004;145:472477.
15. Sloots TP, Kapeleris JP, Mackay IM, et al. Evaluation of a commercial 38. Hall CB, Caserta MT, Schnabel K, et al. Chromosomal integration of
enzyme-linked immunosorbent assay for detection of serum immunoglobulin human herpesvirus 6 is the major mode of congenital human herpesvirus 6
G response to human herpesvirus 6. J Clin Microbiol. 1996;34:675679. infection. Pediatrics. 2008;122:513520.
16. Yoshikawa T, Suga S, Asano Y, et al. Neutralizing antibodies to human 39. Pellett PE, Ablashi DV, Ambros PF, et al. Chromosomally integrated human
herpesvirus-6 in healthy individuals. Pediatr Infect Dis J. 1990;9:589590. herpesvirus 6: questions and answers. Rev Med Virol. 2012;22:144155.
17. Ohashi M, Ihira M, Suzuki K, et al. Transfer of human herpesvirus 6 40. Tanaka-Taya K, Sashihara J, Kurahashi H, et al. Human herpesvirus 6
and 7 antibodies from mothers to their offspring. Pediatr Infect Dis J. (HHV-6) is transmitted from parent to child in an integrated form and
2001;20:449450. characterization of cases with chromosomally integrated HHV-6 DNA.
18. Yoshida M, Torigoe S, Ikeue K, et al. Neutralizing antibody responses J Med Virol. 2004;73:465473.
to human herpesviruses 6 and 7 do not cross-react with each other, and 41. Arbuckle JH, Medveczky MM, Luka J, et al. The latent human herpesvirus-
maternal neutralizing antibodies contribute to sequential infection with 6A genome specifically integrates in telomeres of human chromosomes in
these viruses in childhood. Clin Diagn Lab Immunol. 2002;9:388393. vivo and in vitro. Proc Natl Acad Sci U S A. 2010;107:55635568.
19. Cermelli C, Fabio G, Montorsi M, et al. Prevalence of antibodies to human 42. Nacheva EP, Ward KN, Brazma D, et al. Human herpesvirus 6 integrates
herpesviruses 6 and 7 in early infancy and age at primary infection. New within telomeric regions as evidenced by five different chromosomal sites.
Microbiol. 1996;19:18. J Med Virol. 2008;80:19521958.
20. Yoshikawa T, Suga S, Asano Y, et al. Distribution of antibodies to a caus- 43. Arbuckle JH, Medveczky PG. The molecular biology of human herpesvi-
ative agent of exanthem subitum (human herpesvirus-6) in healthy indi- rus-6 latency and telomere integration. Microbes Infect. 2011;13:731741.
viduals. Pediatrics. 1989;84:675677. 44. Ward KN, Leong HN, Nacheva EP, et al. Human herpesvirus 6 chromosomal
21. Hall CB, Long CE, Schnabel KC, et al. Human herpesvirus-6 infection in integration in immunocompetent patients results in high levels of viral DNA
children. A prospective study of complications and reactivation. N Engl J in blood, sera, and hair follicles. J Clin Microbiol. 2006;44:15711574.
Med. 1994;331:432438. 45. Hall CB, Caserta MT, Schnabel KC, et al. Transplacental congenital human
22. Okuno T, Mukai T, Baba K, et al. Outbreak of exanthem subitum in an herpesvirus 6 infection caused by maternal chromosomally integrated
orphanage. J Pediatr. 1991;119:759761. virus. J Infect Dis. 2010;201:505507.
23. Brown NA, Sumaya CV, Liu CR, et al. Fall in human herpesvirus 6 sero- 46. Schirmer EC, Wyatt LS, Yamanishi K, et al. Differentiation between two
positivity with age. Lancet. 1988;2:396. distinct classes of viruses now classified as human herpesvirus 6. Proc Natl
24. Yanagi K, Harada S, Ban F, et al. High prevalence of antibody to human Acad Sci U S A. 1991;88:59225966.
herpesvirus-6 and decrease in titer with increase in age in Japan. J Infect 47. Ablashi D, Agut H, Berneman Z, et al. Human herpesvirus-6 strain groups:
Dis. 1990;161:153154. a nomenclature. Arch Virol. 1993;129:363366.

Scheld_Ch13.indd 200 2/21/14 6:39 PM


Chapter 13: Human Herpesvirus-6 201

48. De Bolle L, Naesens L, De Clercq E. Update on human herpesvirus 6 biol- 79. Andre-Garnier E, Milpied N, Boutolleau D, et al. Reactivation of human
ogy, clinical features, and therapy. Clin Microbiol Rev. 2005;18:217245. herpesvirus 6 during ex vivo expansion of circulating CD34 haematopoi-
49. Dewhurst S, McIntyre K, Schnabel K, et al. Human herpesvirus 6 (HHV-6) etic stem cells. J Gen Virol. 2004;85:33333336.
variant B accounts for the majority of symptomatic primary HHV-6 infec- 80. Rotola A, Ravaioli T, Gonelli A, et al. U94 of human herpesvirus 6 is
tions in a population of U.S. infants. J Clin Microbiol. 1993;31:416418. expressed in latently infected peripheral blood mononuclear cells and
50. Hall CB, Caserta MT, Schnabel KC, et al. Persistence of human herpesvirus blocks viral gene expression in transformed lymphocytes in culture. Proc
6 according to site and variant: possible greater neurotropism of variant A. Natl Acad Sci U S A. 1998;95:1391113916.
Clin Infect Dis. 1998;26:132137. 81. Chan PK, Ng HK, Hui M, et al. Presence of human herpesviruses 6, 7, and
51. Bates M, Monze M, Bima H, et al. Predominant human herpesvirus 6 vari- 8 DNA sequences in normal brain tissue. J Med Virol. 1999;59:491495.
ant A infant infections in an HIV-1 endemic region of Sub-Saharan Africa. 82. Cuomo L, Trivedi P, Cardillo MR, et al. Human herpesvirus 6 infection in
J Med Virol. 2009;81:779789. neoplastic and normal brain tissue. J Med Virol. 2001;63:4551.
52. Burbelo PD, Bayat A, Wagner J, et al. No serological evidence for a 83. Luppi M, Barozzi P, Maiorana A, et al. Human herpesvirus 6 infection in
role of HHV-6 infection in chronic fatigue syndrome. Am J Transl Res. normal human brain tissue. J Infect Dis. 1994;169:943944.
2012;4:443451. 84. Chan PK, Ng HK, Hui M, et al. Prevalence and distribution of human
53. Higashimoto Y, Ohta A, Nishiyama Y, et al. Development of a human herpesvirus 6 variants A and B in adult human brain. J Med Virol.
herpesvirus 6 species-specific immunoblotting assay. J Clin Microbiol. 2001;64:4246.
2012;50:12451251. 85. Saddawi-Konefka R, Crawford JR. Chronic viral infection and primary
54. Dominguez G, Dambaugh TR, Stamey FR, et al. Human herpesvirus 6B central nervous system malignancy. J Neuroimmune Pharmacol. 2010;
genome sequence: coding content and comparison with human herpesvirus 5:387403.
6A. J Virol. 1999;73:80408052. 86. Luppi M, Barozzi P, Maiorana A, et al. Human herpesvirus-6: a survey
55. Gompels UA, Nicholas J, Lawrence G, et al. The DNA sequence of human of presence and distribution of genomic sequences in normal brain and
herpesvirus-6: structure, coding content, and genome evolution. Virology. neuroglial tumors. J Med Virol. 1995;47:105111.
1995;209:2951. 87. Albright AV, Lavi E, Black JB, et al. The effect of human herpesvirus-6
56. Isegawa Y, Mukai T, Nakano K, et al. Comparison of the complete DNA (HHV-6) on cultured human neural cells: oligodendrocytes and microglia.
sequences of human herpesvirus 6 variants A and B. J Virol. 1999;73: J Neurovirol. 1998;4:486494.
80538063. 88. Ahlqvist J, Fotheringham J, Akhyani N, et al. Differential tropism of
57. Kramarsky B, Sander C. Electron microscopy of human herpesvirus-6 human herpesvirus 6 (HHV-6) variants and induction of latency by
(HHV-6). In: Ablashi D, Krueger G, Salahuddin SZ, eds. Human HHV-6A in oligodendrocytes. J Neurovirol. 2005;11:384394.
Herpesvirus-6: Epidemiology, Molecular Biology and Clinical Pathology. 89. Donati D, Martinelli E, Cassiani-Ingoni R, et al. Variant-specific tropism
Amsterdam: Elsevier; 1992:5966. of human herpesvirus 6 in human astrocytes. J Virol. 2005;79:94399448.
58. Flamand L, Komaroff AL, Arbuckle JH, et al. Review, part 1: human her- 90. Gardell JL, Dazin P, Islar J, et al. Apoptotic effects of Human
pesvirus-6-basic biology, diagnostic testing, and antiviral efficacy. J Med Herpesvirus-6A on glia and neurons as potential triggers for central ner-
Virol. 2010;82:15601568. vous system autoimmunity. J Clin Virol. 2006;37(suppl 1):S11S16.
59. Morissette G, Flamand L. Herpesviruses and chromosomal integration. 91. Kong H, Baerbig Q, Duncan L, et al. Human herpesvirus type 6 indirectly
J Virol. 2010;84:1210012109. enhances oligodendrocyte cell death. J Neurovirol. 2003;9:539150.
60. Ablashi DV, Balachandran N, Josephs SF, et al. Genomic polymorphism, 92. Caserta MT, Hall CB, Schnabel K, et al. Neuroinvasion and persistence of
growth properties, and immunologic variations in human herpesvirus-6 human herpesvirus 6 in children. J Infect Dis. 1994;170:15861589.
isolates. Virology. 1991;184:545552. 93. Ward KN, Leong HN, Thiruchelvam AD, et al. Human herpesvirus 6
61. Kikuta H, Lu H, Tomizawa K, et al. Enhancement of human herpesvirus 6 DNA levels in cerebrospinal fluid due to primary infection differ from
replication in adult human lymphocytes by monoclonal antibody to CD3. J those due to chromosomal viral integration and have implications for
Infect Dis. 1990;161:10851087. diagnosis of encephalitis. J Clin Microbiol. 2007;45:12981304.
62. Frenkel N, Roffman E, Schirmer EC, et al. Cellular and growth-factor 94. Yoshikawa T, Ihira M, Suzuki K, et al. Invasion by human herpesvirus 6
requirements for the replication of human herpesvirus 6 in primary lym- and human herpesvirus 7 of the central nervous system in patients with
phocyte cultures. Adv Exp Med Biol. 1990;278:18. neurological signs and symptoms. Arch Dis Child. 2000;83:170171.
63. Frenkel N, Schirmer EC, Katsafanas G, et al. T-cell activation is re- 95. Wang FZ, Linde A, Hagglund H, et al. Human herpesvirus 6 DNA in
quired for efficient replication of human herpesvirus 6. J Virol. 1990;64: cerebrospinal fluid specimens from allogeneic bone marrow transplant pa-
45984602. tients: does it have clinical significance? Clin Infect Dis. 1999;28:562568.
64. Ablashi DV, Hung CL. In vitro propagation systems for HHV-6. In: 96. Cone RW, Huang ML, Ashley R, et al. Human herpesvirus 6 DNA in
Ablashi DV, Krueger GRF, Salahuddin SZ, eds. Human Herpesvirus-6: peripheral blood cells and saliva from immunocompetent individuals.
Epidemiology, Molecular Biology and Clinical Pathology. Amsterdam: J Clin Microbiol. 1994;32:2633.
Elsevier; 1992:3747. 97. Cuende JI, Ruiz J, Civeira MP, et al. High prevalence of HHV-6 DNA
65. Ablashi DV, Lusso P, Hung CL, et al. Utilization of human hematopoietic in peripheral blood mononuclear cells of healthy individuals detected by
cell lines for the propagation and characterization of HBLV (human herpes- nested-PCR. J Med Virol. 1994;43:115118.
virus 6). Int J Cancer. 1988;42:787791. 98. Pellett PE, Black JB, Yamamoto M. Human herpesvirus 6: the virus and
66. Santoro F, Kennedy PE, Locatelli G, et al. CD46 is a cellular receptor for the search for its role as a human pathogen. Adv Virus Res. 1992;41:152.
human herpesvirus 6. Cell. 1999;99:817827. 99. Wyatt LS, Frenkel N. Human herpesvirus 7 is a constitutive inhabitant of
67. De Bolle L, Van Loon J, De Clercq E, et al. Quantitative analysis of human adult human saliva. J Virol. 1992;66:32063209.
herpesvirus 6 cell tropism. J Med Virol. 2005;75:7685. 100. Zerr DM. Human herpesvirus 6 (HHV-6) disease in the setting of trans-
68. De Filippis L, Foglieni C, Silva S, et al. Differentiated human neural stem plantation. Curr Opin Infect Dis. 2012;25:438444.
cells: a new ex vivo model to study HHV-6 infection of the central nervous 101. Ablashi DV, Devin CL, Yoshikawa T, et al. Review part 3: human her-
system. J Clin Virol. 2006;37(suppl 1):S27S32. pesvirus-6 in multiple non-neurological diseases. J Med Virol. 2010;82:
69. Dewhurst S, Chandran B, McIntyre K, et al. Phenotypic and genetic poly- 19031910.
morphisms among human herpesvirus-6 isolates from North American 102. Balachandra K, Chimabutra K, Supromajakr P, et al. High rate of reacti-
infants. Virology. 1992;190:490493. vation of human herpesvirus 6 in children with dengue hemorrhagic fever.
70. He J, McCarthy M, Zhou Y, et al. Infection of primary human fetal astro- J Infect Dis. 1994;170:746748.
cytes by human herpesvirus 6. J Virol. 1996;70:12961300. 103. Razonable RR, Fanning C, Brown RA, et al. Selective reactivation of
71. Hirata Y, Kondo K, Yamanishi K. Human herpesvirus 6 downregulates human herpesvirus 6 variant a occurs in critically ill immunocompetent
major histocompatibility complex class I in dendritic cells. J Med Virol. hosts. J Infect Dis. 2002;185:110113.
2001;65:576583. 104. Suga S, Yoshikawa T, Asano Y, et al. Activation of human herpesvirus-6
72. Lusso P, Malnati MS, Garzino-Demo A, et al. Infection of natural killer in children with acute measles. J Med Virol. 1992;38:278282.
cells by human herpesvirus 6. Nature. 1993;362:458462. 105. Yamanishi K, Kondo T, Kondo K, et al. Exanthem subitum and human
73. Simmons A, Demmrich Y, La Vista A, et al. Replication of human herpes- herpesvirus 6 (HHV-6) infection. Adv Exp Med Biol. 1990;278:2937.
virus 6 in epithelial cells in vitro. J Infect Dis. 1992;166:202205. 106. Asano Y, Yoshikawa T, Suga S, et al. Viremia and neutralizing antibody
74. Yao K, Mandel M, Akyani N, et al. Differential HHV-6A gene expres- response in infants with exanthem subitum. J Pediatr. 1989;114:535539.
sion in T cells and primary human astrocytes based on multi-virus array 107. Kumagai T, Yoshikawa T, Yoshida M, et al. Time course characteristics of
analysis. Glia. 2006;53:789798. human herpesvirus 6 specific cellular immune response and natural killer cell
75. Mori Y. Recent topics related to human herpesvirus 6 cell tropism. Cell activity in patients with exanthema subitum. J Med Virol. 2006;78:792799.
Microbiol. 2009;11:10011006. 108. Yoshida M, Torigoe S, Yamada M. Elucidation of the cross-reactive
76. Campadelli-Fiume G, Mirandola P, Menotti L. Human herpesvirus 6: an immunoglobulin M response to human herpesviruses 6 and 7 on the basis
emerging pathogen. Emerg Infect Dis. 1999;5:353366. of neutralizing antibodies. Clin Diagn Lab Immunol. 2002;9:394402.
77. Takahashi K, Sonoda S, Higashi K, et al. Predominant CD4 T-lymphocyte 109. de Pagter AP, Boelens JJ, Scherrenburg J, et al. First analysis of human
tropism of human herpesvirus 6-related virus. J Virol. 1989;63:31613163. herpesvirus 6T-cell responses: specific boosting after HHV6 reactivation
78. Kondo K, Kondo T, Shimada K, et al. Strong interaction between human in stem cell transplantation recipients. Clin Immunol. 2012;144:179189.
herpesvirus 6 and peripheral blood monocytes/macrophages during acute 110. Nastke MD, Becerra A, Yin L, et al. Human CD4 T cell response to
infection. J Med Virol. 2002;67:364369. human herpesvirus 6. J Virol. 2012;86:47764792.

Scheld_Ch13.indd 201 2/21/14 6:39 PM


202 Part II: Viral Infections and Related Disorder

111. Lusso P. HHV-6 and the immune system: mechanisms of immunomodula- 142. Yanagihara K, Tanaka-Taya K, Itagaki Y, et al. Human herpesvirus 6
tion and viral escape. J Clin Virol. 2006;37(suppl 1):S4S10. meningoencephalitis with sequelae. Pediatr Infect Dis J. 1995;14:240242.
112. Sullivan BM, Coscoy L. Downregulation of the T-cell receptor complex 143. Crawford JR, Kadom N, Santi MR, et al. Human herpesvirus 6 rhomb-
and impairment of T-cell activation by human herpesvirus 6 u24 protein. encephalitis in immunocompetent children. J Child Neurol. 2007;22:
J Virol. 2008;82:602608. 12601268.
113. Jaworska J, Gravel A, Fink K, et al. Inhibition of transcription of the beta 144. Kato Z, Kozawa R, Teramoto T, et al. Acute cerebellitis in primary human
interferon gene by the human herpesvirus 6 immediate-early 1 protein. herpesvirus-6 infection. Eur J Pediatr. 2003;162:801803.
J Virol. 2007;81:57375748. 145. Ahtiluoto S, Mannonen L, Paetau A, et al. In situ hybridization detection
114. Jaworska J, Gravel A, Flamand L. Divergent susceptibilities of human of human herpesvirus 6 in brain tissue from fatal encephalitis. Pediatrics.
herpesvirus 6 variants to type I interferons. Proc Natl Acad Sci U S A. 2000;105:431433.
2010;107:83698374. 146. Asano Y, Yoshikawa T, Kajita Y, et al. Fatal encephalitis/encephalopa-
115. Kakimoto M, Hasegawa A, Fujita S, et al. Phenotypic and functional thy in primary human herpesvirus-6 infection. Arch Dis Child. 1992;67:
alterations of dendritic cells induced by human herpesvirus 6 infection. 14841485.
J Virol. 2002;76:1033810345. 147. Ichiyama T, Ito Y, Kubota M, et al. Serum and cerebrospinal fluid levels of
116. Smith AP, Paolucci C, Di Lullo G, et al. Viral replication-independent cytokines in acute encephalopathy associated with human herpesvirus-6
blockade of dendritic cell maturation and interleukin-12 production by infection. Brain Dev. 2009;31:731738.
human herpesvirus 6. J Virol. 2005;79:28072813. 148. Kawabe S, Ito Y, Ohta R, et al. Comparison of the levels of human her-
117. Inoue Y, Yasukawa M, Fujita S. Induction of T-cell apoptosis by human pesvirus 6 (HHV-6) DNA and cytokines in the cerebrospinal fluid and
herpesvirus 6. J Virol. 1997;71:37513759. serum of children with HHV-6 encephalopathy. J Med Virol. 2010;82:
118. Yasukawa M, Inoue Y, Ohminami H, et al. Apoptosis of CD4 T lympho- 14101415.
cytes in human herpesvirus-6 infection. J Gen Virol. 1998;79:143147. 149. Kawamura Y, Sugata K, Ihira M, et al. Different characteristics of human
119. Zerr DM, Meier AS, Selke SS, et al. A population-based study of primary herpesvirus 6 encephalitis between primary infection and viral reactiva-
human herpesvirus 6 infection. N Engl J Med. 2005;352:768776. tion. J Clin Virol. 2011;51:1219.
120. Asano Y, Yoshikawa T, Suga S, et al. Clinical features of infants with 150. Ogata M, Satou T, Kawano R, et al. Plasma HHV-6 viral load-guided
primary human herpesvirus 6 infection (exanthem subitum, roseola infan- preemptive therapy against HHV-6 encephalopathy after allogeneic stem
tum). Pediatrics. 1994;93:104108. cell transplantation: a prospective evaluation. Bone Marrow Transplant.
121. Asano Y, Nakashima T, Yoshikawa T, et al. Severity of human herpesvirus-6 2008;41:279285.
viremia and clinical findings in infants with exanthem subitum. J Pediatr. 151. Zerr DM, Boeckh M, Delaney C, et al. HHV-6 reactivation and associated
1991;118:891895. sequelae after hematopoietic cell transplantation. Biol Blood Marrow
122. Okada K, Ueda K, Kusuhara K, et al. Exanthema subitum and human Transplant. 2012;18:17001708.
herpesvirus 6 infection: clinical observations in fifty-seven cases. Pediatr 152. Yoshikawa T, Asano Y, Ihira M, et al. Human herpesvirus 6 viremia in
Infect Dis J. 1993;12:204208. bone marrow transplant recipients: clinical features and risk factors.
123. Portolani M, Cermelli C, Meacci M, et al. Primary infection by HHV-6 J Infect Dis. 2002;185:847853.
variant B associated with a fatal case of hemophagocytic syndrome. New 153. Chang FY, Singh N, Gayowski T, et al. Fever in liver transplant reci-
Microbiol. 1997;20:711. pients: changing spectrum of etiologic agents. Clin Infect Dis. 1998;26:
124. Akashi K, Eizuru Y, Sumiyoshi Y, et al. Brief report: severe infectious 5965.
mononucleosis-like syndrome and primary human herpesvirus 6 infection 154. DesJardin JA, Cho E, Supran S, et al. Association of human herpesvirus 6
in an adult. N Engl J Med. 1993;329:168171. reactivation with severe cytomegalovirus-associated disease in orthotopic
125. Barone SR, Kaplan MH, Krilov LR. Human herpesvirus-6 infection in liver transplant recipients. Clin Infect Dis. 2001;33:13581362.
children with first febrile seizures. J Pediatr. 1995;127:9597. 155. Dockrell DH, Mendez JC, Jones M, et al. Human herpesvirus 6 seronega-
126. Laina I, Syriopoulou VP, Daikos GL, et al. Febrile seizures and primary tivity before transplantation predicts the occurrence of fungal infection in
human herpesvirus 6 infection. Pediatr Neurol. 2010;42:2831. liver transplant recipients. Transplantation. 1999;67:399403.
127. Millichap JG, Millichap JJ. Role of viral infections in the etiology of 156. Dockrell DH, Prada J, Jones MF, et al. Seroconversion to human her-
febrile seizures. Pediatr Neurol. 2006;35:165172. pesvirus 6 following liver transplantation is a marker of cytomegalovirus
128. Suga S, Suzuki K, Ihira M, et al. Clinical characteristics of febrile convul- disease. J Infect Dis. 1997;176:11351140.
sions during primary HHV-6 infection. Arch Dis Child. 2000;82:6266. 157. Emery VC. Human herpesviruses 6 and 7 in solid organ transplant recipi-
129. Kondo K, Nagafuji H, Hata A, et al. Association of human herpesvirus 6 ents. Clin Infect Dis. 2001;32:13571360.
infection of the central nervous system with recurrence of febrile convul- 158. Kidd IM, Clark DA, Sabin CA, et al. Prospective study of human betaherpes-
sions. J Infect Dis. 1993;167:11971200. viruses after renal transplantation: association of human herpesvirus 7 and
130. Jee SH, Long CE, Schnabel KC, et al. Risk of recurrent seizures after a cytomegalovirus co-infection with cytomegalovirus disease and increased
primary human herpesvirus 6-induced febrile seizure. Pediatr Infect Dis J. rejection. Transplantation. 2000;69:24002404.
1998;17:4348. 159. Lautenschlager I, Hckerstedt K, Linnavuori K, et al. Human herpesvirus-6
131. Theodore WH, Epstein L, Gaillard WD, et al. Human herpes virus 6B: a infection after liver transplantation. Clin Infect Dis. 1998;26:702707.
possible role in epilepsy? Epilepsia. 2008;49:18281837. 160. Lautenschlager I, Razonable RR. Human herpesvirus-6 infections
132. Epstein LG, Shinnar S, Hesdorffer DC, et al. Human herpesvirus 6 and in kidney, liver, lung, and heart transplantation: review. Transpl Int.
7 in febrile status epilepticus: the FEBSTAT study. Epilepsia. 2012;53: 2012;25:493502.
14811488. 161. Ratnamohan VM, Chapman J, Howse H, et al. Cytomegalovirus and
133. Donati D, Akhyani N, Fogdell-Hahn A, et al. Detection of human her- human herpesvirus 6 both cause viral disease after renal transplantation.
pesvirus-6 in mesial temporal lobe epilepsy surgical brain resections. Transplantation. 1998;66:877882.
Neurology. 2003;61:14051411. 162. Rogers J, Rohal S, Carrigan DR, et al. Human herpesvirus-6 in liver trans-
134. Fotheringham J, Donati D, Akhyani N, et al. Association of human her- plant recipients: role in pathogenesis of fungal infections, neurologic com-
pesvirus-6B with mesial temporal lobe epilepsy. PLoS Med. 2007;4:e180. plications, and outcome. Transplantation. 2000;69:25662573.
135. Karatas H, Gurer G, Pinar A, et al. Investigation of HSV-1, HSV-2, CMV, 163. Cone RW, Huang ML, Corey L, et al. Human herpesvirus 6 infections
HHV-6 and HHV-8 DNA by real-time PCR in surgical resection materi- after bone marrow transplantation: clinical and virologic manifestations.
als of epilepsy patients with mesial temporal lobe sclerosis. J Neurol Sci. J Infect Dis. 1999;179:311318.
2008;264:151156. 164. Kadakia MP, Rybka WB, Stewart JA, et al. Human herpesvirus 6: infec-
136. Li JM, Lei D, Peng F, et al. Detection of human herpes virus 6B in patients tion and disease following autologous and allogeneic bone marrow trans-
with mesial temporal lobe epilepsy in West China and the possible asso- plantation. Blood. 1996;87:53415354.
ciation with elevated NF-B expression. Epilepsy Res. 2011;94:19. 165. Yoshikawa T, Suga S, Asano Y, et al. Human herpesvirus-6 infection in
137. Uesugi H, Shimizu H, Maehara T, et al. Presence of human herpesvirus 6 bone marrow transplantation. Blood. 1991;78:13811384.
and herpes simplex virus detected by polymerase chain reaction in surgi- 166. Beovic B, Pecaric-Meglic N, Marin J, et al. Fatal human herpesvirus
cal tissue from temporal lobe epileptic patients. Psychiatry Clin Neurosci. 6-associated multifocal meningoencephalitis in an adult female patient.
2000;54:589593. Scand J Infect Dis. 2001;33:942944.
138. Suga S, Yoshikawa T, Asano Y, et al. Clinical and virological analyses of 167. Birnbaum T, Padovan CS, Sporer B, et al. Severe meningoencephalitis
21 infants with exanthem subitum (roseola infantum) and central nervous caused by human herpesvirus 6 type B in an immunocompetent woman
system complications. Ann Neurol. 1993;33:597603. treated with ganciclovir. Clin Infect Dis. 2005;40:887889.
139. Ishiguro N, Yamada S, Takahashi T, et al. Meningo-encephalitis associ- 168. Isaacson E, Glaser CA, Forghani B, et al. Evidence of human herpesvirus 6
ated with HHV-6 related exanthem subitum. Acta Paediatr Scand. 1990; infection in 4 immunocompetent patients with encephalitis. Clin Infect Dis.
79:987999. 2005;40:890893.
140. Yoshikawa T, Nakashima T, Suga S, et al. Human herpesvirus-6 DNA 169. McCullers JA, Lakeman FD, Whitley RJ. Human herpesvirus 6 is associ-
in cerebrospinal fluid of a child with exanthem subitum and meningoen- ated with focal encephalitis. Clin Infect Dis. 1995;21:571576.
cephalitis. Pediatrics. 1992;89:888890. 170. Portolani M, Pecorari M, Gennari W, et al. Severe encephalopathy associ-
141. Kamei A, Ichinohe S, Onuma R, et al. Acute disseminated demyelination due ated with reactivated human herpesvirus 6 in a six year-old immunocom-
to primary human herpesvirus-6 infection. Eur J Pediatr. 1997;156:709712. petent child. Minerva Pediatr. 2002;54:459464.

Scheld_Ch13.indd 202 2/21/14 6:39 PM


Chapter 13: Human Herpesvirus-6 203

171. Portolani M, Pecorari M, Tamassia MG, et al. Case of fatal encephalitis 193. Xu Y, Linde A, Fredrikson S, et al. HHV-6 A- or B-specific P41 anti-
by HHV-6 variant A. J Med Virol. 2001;65:133137. gens do not reveal virus variant- specific IgG or IgM responses in human
172. Sloots TP, Mackay IM, Carroll P. Meningoencephalitis in an adult with serum. J Med Virol. 2002;66:394399.
human herpesvirus-6 infection. Med J Aust. 1993;159:838. 194. Alvarez-Lafuente R, de las Heras V, Garca-Montojo M, et al. Human
173. Torre D, Speranza F, Martegani R, et al. Meningoencephalitis caused by herpesvirus-6 and multiple sclerosis: relapsing-remitting versus secondary
human herpesvirus-6 in an immunocompetent adult patient: case report progressive. Mult Scler. 2007;13:578583.
and review of the literature. Infection. 1998;26:402404. 195. Enbom M, Linde A, Evengard B. No evidence of active infection with
174. Novoa LJ, Nagra RM, Nakawatase T, et al. Fulminant demyelinating human herpesvirus 6 (HHV-6) or HHV- 8 in chronic fatigue syndrome.
encephalomyelitis associated with productive HHV-6 infection in an J Clin Microbiol. 2000;38:2457.
immunocompetent adult. J Med Virol. 1997;52:301308. 196. Goldberg SH, Albright AV, Lisak RP, et al. Polymerase chain reaction
175. Troy SB, Blackburn BG, Yeom K, et al. Severe encephalomyelitis in an im- analysis of human herpesvirus-6 sequences in the sera and cerebro-
munocompetent adult with chromosomally integrated human herpesvirus spinal fluid of patients with multiple sclerosis. J Neurovirol. 1999;5:
6 and clinical response to treatment with foscarnet plus ganciclovir. Clin 134139.
Infect Dis. 2008;47:e93e96. 197. Merelli E, Bedin R, Sola P, et al. Human herpes virus 6 and human herpes
176. Hill JA, Koo S, Guzman Suarez BB, et al. Cord-blood hematopoietic virus 8 DNA sequences in brains of multiple sclerosis patients, normal
stem-cell transplantation confers an increased risk for human herpesvirus- adults and children. J Neurol. 1997;244:450454.
6-associated acute limbic encephalitis: a cohort analysis. Biol Blood 198. Taus C, Pucci E, Cartechini E, et al. Absence of HHV-6 and HHV-7 in
Marrow Transplant. 2012;18:16381648. cerebrospinal fluid in relapsing- remitting multiple sclerosis. Acta Neurol
177. Mori Y, Miyamoto T, Nagafuji K, et al. High incidence of human herpes Scand. 2000;101:224248.
virus 6-associated encephalitis/myelitis following a second unrelated cord 199. Chokephaibulkit K, Brunell PA, Vimal V, et al. Enzyme-linked immu-
blood transplantation. Biol Blood Marrow Transplant. 2010;16:15961602. nosorbent assay for detection of antibody to human herpesvirus 6. Clin
178. Sakai R, Kanamori H, Motohashi K, et al. Long-term outcome of human Diagn Lab Immunol. 1997;4:687691.
herpesvirus-6 encephalitis after allogeneic stem cell transplantation. Biol 200. Ward KN. The natural history and laboratory diagnosis of human her-
Blood Marrow Transplant. 2011;17:13891394. pesviruses-6 and -7 infections in the immunocompetent. J Clin Virol.
179. Seeley WW, Marty FM, Holmes TM, et al. Post-transplant acute limbic 2005;32:183193.
encephalitis: clinical features and relationship to HHV6. Neurology. 201. Secchiero P, Carrigan DR, Asano Y, et al. Detection of human herpes-
2007;69:156165. virus 6 in plasma of children with primary infection and immunosup-
180. Wainwright MS, Martin PL, Morse RP, et al. Human herpesvirus 6 limbic pressed patients by polymerase chain reaction. J Infect Dis. 1995;171:
encephalitis after stem cell transplantation. Ann Neurol. 2001;50:612619. 273280.
181. Zerr DM. Human herpesvirus 6 and central nervous system disease in he- 202. Achour A, Boutolleau D, Slim A, et al. Human herpesvirus-6 (HHV-6)
matopoietic cell transplantation. J Clin Virol. 2006;37(suppl 1):S52S56. DNA in plasma reflects the presence of infected blood cells rather than
182. Zerr DM, Fann JR, Breiger D, et al. HHV-6 reactivation and its effect on circulating viral particles. J Clin Virol. 2007;38:280285.
delirium and cognitive functioning in hematopoietic cell transplant recipi- 203. Norton RA, Caserta MT, Hall CB, et al. Detection of human herpesvirus
ents. Blood. 2011;117:52435249. 6 by reverse transcription-PCR. J Clin Microbiol. 1999;37:36723675.
183. Ogata M, Kikuchi H, Satou T, et al. Human herpesvirus 6 DNA in plasma 204. Ihira M, Enomoto Y, Kawamura Y, et al. Development of quantitative
after allogeneic stem cell transplantation: incidence and clinical signifi- RT-PCR assays for detection of three classes of HHV-6B gene transcripts.
cance. J Infect Dis. 2006;193:6879. J Med Virol. 2012;84:13881395.
184. Zerr DM, Corey L, Kim HW, et al. Clinical outcomes of human herpes- 205. Caserta MT, Hall CB, Schnabel K, et al. Diagnostic assays for active infec-
virus 6 reactivation after hematopoietic stem cell transplantation. Clin tion with human herpesvirus 6 (HHV-6). J Clin Virol. 2010;48:5557.
Infect Dis. 2005;40:932940. 206. Graudie B, Charrier M, Bonnafous P, et al. Quantitation of human
185. Betts BC, Young JA, Ustun C, et al. Human herpesvirus 6 infection after herpesvirus-6A, -6B and -7 DNAs in whole blood, mononuclear and poly-
hematopoietic cell transplantation: is routine surveillance necessary? Biol morphonuclear cell fractions from healthy blood donors. J Clin Virol.
Blood Marrow Transplant. 2011;17:15621568. 2012;53:151155.
186. Provenzale JM, van Landingham K, White LE. Clinical and imaging 207. Dewhurst S. Human herpesvirus type 6 and human herpesvirus type
findings suggesting human herpesvirus 6 encephalitis. Pediatr Neurol. 7 infections of the central nervous system. Herpes. 2004;11(suppl
2010;42:3239. 2):105A111A.
187. Hubele F, Bilger K, Kremer S, et al. Sequential FDG PET and MRI find- 208. Cheng FW, Lee V, Leung WK, et al. HHV-6 encephalitis in pediatric un-
ings in a case of human herpes virus 6 limbic encephalitis. Clin Nucl Med. related umbilical cord transplantation: a role for ganciclovir prophylaxis?
2012;37:716717. Pediatr Transplant. 2010;14:483487.
188. Provenzale JM, vanLandingham KE, Lewis DV, et al. Extrahippocampal 209. Ishiyama K, Katagiri T, Ohata K, et al. Safety of pre-engraftment prophy-
involvement in human herpesvirus 6 encephalitis depicted at MR imaging. lactic foscarnet administration after allogeneic stem cell transplantation.
Radiology. 2008;249:955963. Transpl Infect Dis. 2012;14:3339.
189. Opsahl ML, Kennedy PGE. Early and late HHV-6 gene transcripts in 210. Mookerjee BP, Vogelsang G. Human herpes virus-6 encephalitis after
multiple sclerosis lesions and normal appearing white matter. Brain. bone marrow transplantation: successful treatment with ganciclovir. Bone
2005;128:516527. Marrow Transplant. 1997;20:905906.
190. Challoner PB, Smith KT, Parker JD, et al. Plaque-associated expression 211. Phlmann C, Schetelig J, Reuner U, et al. Cidofovir and foscarnet for
of human herpesvirus 6 in multiple sclerosis. Proc Natl Acad Sci U S A. treatment of human herpesvirus 6 encephalitis in a neutropenic stem cell
1995;92:74407444. transplant recipient. Clin Infect Dis. 2007;44:e118e120.
191. Alvarez-Lafuente R, Martn-Estefana C, de Las Heras V, et al. Active 212. Rapaport D, Engelhard D, Tagger G, et al. Antiviral prophylaxis may
human herpesvirus 6 infection in patients with multiple sclerosis. Arch prevent human herpesvirus-6 reactivation in bone marrow transplant
Neurol. 2002;59:929933. recipients. Transpl Infect Dis. 2002;4:1016.
192. Soldan SS, Berti R, Salem N, et al. Association of human herpes virus 6 213. Tokimasa S, Hara J, Osugi Y, et al. Ganciclovir is effective for prophylaxis
(HHV-6) with multiple sclerosis: increased IgM response to HHV-6 early and treatment of human herpesvirus-6 in allogeneic stem cell transplantation.
antigen and detection of serum HHV-6 DNA. Nat Med. 1997;3:13941397. Bone Marrow Transplant. 2002;29:595598.

Scheld_Ch13.indd 203 2/21/14 6:39 PM


CHAPTER 14 B VIRUS
RICHARD J. WHITLEY

HISTORY INFECTIOUS AGENT


Of the nearly 35 herpesviruses that have been isolated B virus was propagated on the chorioallantoic membrane
from nonhuman primates, only oneB virus of Old World of embryonated eggs in 1939 (8). Tissue culture isolation of
monkeysis highly pathogenic for humans. In 1932, a young B virus did not occur until 1954, when it was recovered from
physician (W.B.) bitten by a monkey developed localized ery- rhesus kidney tissue used for preparation of poliomyelitis vac-
thema at the site of the animal bite. This apparently local- cines (9). During vaccine production, suspended cell culture
ized infection was followed by lymphangitis, lymphadenitis, systems from six kidneys elicited cytopathic effects similar to
and ultimately transverse myelitis. The demise of W.B. was those of HSV.
ascribed to respiratory failure. Tissue specimens from W.B. Notwithstanding numerous attempts at isolation of virus
were obtained by two independent research groups. In 1933, from macaques, the first report of virus isolation is that of
Gay and Holden (1) reported the identification of an ultra- Reissig and Melnick (10), who recovered B virus from tissue
filterable agent that recreated in rabbits a disease similar to cultures of purportedly normal rhesus monkeys. Further stud-
that observed in W.B. This virus was recovered from the neu- ies led to the realization that this virus was easily propagated
rologic tissues of W.B. The inoculation of this virus by either in monkey kidney and chick embryo cell lines (10, 11). B virus
intradermal or intracranial routes was lethal in the animal also multiplies well in rabbit kidney cells and other established
model. These investigators thought that the virus was simi- cell lines such as BS-C-1 (12) and LLC-RK (13). Virus is stable
lar to herpes simplex virus (HSV) and referred to the isolate in tissue culture media when stored at 4C and viral infectivity
as W. While attempts at transmission of this virus to rhesus is maintained if the culture is stored at 72C.
monkeys failed, infection of Cebus monkeys was successful. Replication characteristics of B virus have been reported by
In 1934, Sabin and Wright (2) reported the identification of a several laboratories (4,10,11,1416). The reproductive cycle
similar filterable agent, which they identified as B virus, named is relatively short. The virus inhibits host cell DNA and pro-
W.B. They isolated virus from several neurologic tissues as tein synthesis during the first 4 hours after infection. Infectious
well as from peripheral organs (spleen but not lymph nodes). virus is detectable approximately 6 hours after infection, and
As with the isolate named W, B-virus infection resulted in both extracellular and intracellular virus levels reach a pla-
lethal disease when inoculated by either intradermal or intra- teau approximately 24 to 36 hours after infection and decline
cerebral routes into rabbits. However, the virus was avirulent thereafter (10,16).
when inoculated by the same routes into a variety of other
species, including mice, dogs, and guinea pigs. Furthermore,
Sabin (3) recognized by immunologic characterization of the
isolate that it was related to both HSV and pseudorabies virus
PATHOLOGY AND PATHOGENESIS
(3,4). At the time when latinization of viral names was fash-
ionable, B virus acquired the name herpes simiae. The name Pathology
is a misnomer because nonhuman primates have yielded nu-
merous unrelated herpesviruses. According to the present no- The characteristic cytopathic effect is similar to that of HSV.
menclature, it is designated as the cercopithecine herpesvirus Cells balloon, fuse into polykaryocytes, and form clusters as
type 1. In this chapter, the virus is designated by its common focal areas of infection spread through the entire cell sheet. On
name, B virus. fixation and staining, Cowdry type A eosinophilic intranuclear
B virus is indigenous to Old World monkeys (5,6), inclusions are demonstrable.
being enzootic in rhesus (Macaca mulatta) and cynomolgus Pathologic findings in humans are indicative of the target
(Macaca fascicularis) monkeys, as well as in other Asiatic spe- organs involved. As described later in this chapter, there is lit-
cies of the genus Macaca. Most reported human cases result tle difference in the histopathologic findings encountered fol-
from bites by rhesus monkeys. Since the original reports of lowing either infection of humans or simians.
human herpes B-virus infections, advances in our knowledge
about human disease caused by this virus have proceeded
slowly. Although B-virus infection of humans is not a com- Pathogenesis of Human Infection
mon public health problem, the diagnosis of B virus in four
individuals living in Pensacola, Florida; two in Kalamazoo, Disease in humans usually results from an animal bite or
Michigan; and an animal handler at Yerkes Primate Center scratch, although disease that is the consequence of respiratory
has refocused medical and public health attention on the spread and reactivation has been reported (see later discus-
neuropathogenicity of infection (7). Approximately 50 cases sion). Following exposure, replication of virus at the local site
have occurred, although only 26 are well documented (7). of inoculation occurs. If the skin is the primary site of inocula-
Other cases are pending verification. Person-to-person trans- tion, virus replicates in the skin; this leads to erythema and
mission of infection from one of the fatal cases to a relative calor at the initial site of involvement. A vesicular rash may ap-
was documented for the first time. Additionally, a case has pear in association with tingling and numbness. Usually, there
been identified that led to infection, but without evidence of is evidence of local inflammation at the site of infection with
life-threatening clinical disease (8). Lastly, ocular inoculation mononuclear infiltrate followed by evidence of lymphangitis
of virus proved lethal (7). and subsequent lymphadenopathy. Although viremia has been

204

Scheld_Ch14.indd 204 2/21/14 5:38 PM


Chapter 14: B Virus 205

documented in both rabbits and monkeys, this route of patho- infection is transmitted. Virus cannot penetrate intact skin;
genesis has not been documented for humans. With lymphatic thus, a break in the skin is required for acquisition of infec-
involvement, it is possible for the virus to spread by lymphan- tion. Sites that have been documented as initially infected in
gial routes. Visceral organs that are involved, including the humans include wounds, the eyes, and perhaps the respiratory
heart, liver, spleen, lungs, kidneys, and adrenals, demonstrate tract. Because it is well established that infected animals can
evidence of congestion and focal hemorrhagic necrosis; how- excrete virus in saliva, from the eyes, in vesicular fluid, and
ever, the degree of involvement varies from one patient to the perhaps in the stool, these must all be considered potential
next. A striking characteristic of human B-virus infection is the sources for human infection. Furthermore, as has been well
propensity to involve the central nervous system (CNS) and documented in the history of the development of poliomyelitis
likely reflects spread via neuronal routes. Transverse myelitis is vaccine, infected cell culture tissues can also be a source of
a prominent neurologic finding, with ultimate progression of viral infections for humans.
infection to the brain. B-virus infection can involve all regions Person-to-person transmission of B virus has been docu-
of the brain without evidence of localization to any particular mented (17). In this instance, the wife of an animal worker,
region, in contrast to HSV infection of the CNS, which tends who subsequently died of B-virus infection of the CNS, ac-
to localize in the temporal lobe. Histopathologic findings of quired vesicular lesions of her ring finger. She provided direct
the brain include hemorrhagic foci, necrosis, and inflamma- care of her husbands vesicular lesions. Although she survived
tory changes, as evidenced by perivascular cuffing with mono- infection without evidence of neurologic disease, the extent of
nuclear infiltrates. Edema and degeneration of motor neurons disease may have been limited by early intervention with acy-
are prominent. Even with advanced disease, Cowdry type A clovir. Notably, individuals who developed B-virus infection
eosinophilic intranuclear inclusions can be found in only a had either skin-to-skin or face-to-face contact with at least 40
few cases. In addition, gliosis and astrocytosis are late histo- other individuals during a cluster of cases in Pensacola, Florida.
pathologic findings. Thus, there can be evidence of myelitis, Although these other individuals were under close surveillance,
encephalomyelitis, encephalitis, or combinations thereof (17). none developed evidence of infection. Thus, casual contact can
be excluded as a method of transmission of B-virus infection.
This is particularly important from a public health standpoint.
Pathogenesis of Latency in Macaques Nonfatal infection has not been well documented in reports
of human disease. One recent animal bite resulted in localized
Like other herpesviruses, B-virus infection in macaques be- viral replication without systemic involvement (20). Serologic
comes latent and can recur. As is discussed later in this chap- assessment of humans for evidence of B virus as a marker of
ter, prevalence of antibodies to B virus is widespread in Old subclinical infection is confounded by cross reactivity to HSV.
World rhesus monkeys. Rhesus monkeys captured in the wild
and shipped to primate centers develop vesicular lesions of the
oropharynx, suggesting a pattern of virus reactivation and re- MANIFESTATIONS OF CENTRAL
crudescence of lesions similar to that encountered with HSV.
Unequivocal evidence of latent B-virus infection in rhesus
NERVOUS SYSTEM DISEASE
monkeys evolved with the studies of the recovery of B virus in Human B-virus disease is characterized by an ascending myeli-
monkey kidney cell culture systems. Thus, Wood and Shimada tis or encephalomyelitis. To date, there are a limited number
(9) obtained six isolates from 650 pools of monkey kidneys, of cases of human disease caused by B virus in the literature,
suggesting that at least 1% of rhesus monkeys contain latent depending on the criteria for case definition (7). Several cases
virus that can be reactivated by culturing kidney cells. Virus have been cited in the literature but have not been docu-
was isolated from rhesus tissues also by Boulter (18). B virus mented. Regardless of the exact number of cases, the total
was recovered by cocultivation from a variety of neuronal tis- number of patients who have experienced disease is limited.
sues (including gasserian ganglia, trigeminal ganglia, and dor- Probably thousands of animal workers have been exposed to
sal root ganglia) and the spinal cord (19). Latent virus was animals excreting virus, but very few have developed disease.
also isolated by cocultivation of tissues from experimentally As noted earlier, casual transmission is unlikely. Nevertheless,
infected rabbits (9). the lessons learned regarding the clinical manifestations of dis-
As in the case of HSV infections in humans, the most prom- ease in individuals exposed to B virus, as well as the necessity
inent factor associated with reactivation of B virus in rhesus for methods of intervention and prevention, are reinforced by
monkeys is stress, particularly stress associated with the cap- the findings in these particular patients.
ture and shipping of animals from the wild to captivity. To Infection can occur in one of four fashions. First, infection
date, the state of viral DNA during latency or on the molecular may be asymptomatic. An animal worker exposed to a rhesus
or biochemical events associated with the establishment and monkey with conjunctivitis was stuck by a needle that had been
reactivation of latent virus are unknown. used for inoculation of this animal (20). Subsequent biopsy of
the needlestick site revealed evidence of multinucleated giant
cells. B-virus DNA was detected in the biopsy specimen, but
EPIDEMIOLOGY OF there was no evidence of clinical disease. This form of infec-
HUMAN INFECTION tion may be an underreported event for individuals exposed to
B virus. A second, and more common, route of infection is by
B-virus infections of humans must be considered in the con- animal bite. Following a bite by a monkey excreting B virus, a
text of the epidemiology of any infectious disease, including localized vesicular lesion associated with erythema and edema
the susceptibility of the host and quantity of virus to which develops at the site of viral inoculation. Subsequently, lym-
they are exposed. Importantly, the frequency of excretion of phangitis with spread to regional lymph nodes occurs, along
B virus in infected animals averages 2% to 3% at most; thus, with the development of secondary lymphadenopathy. These
if a human is bitten by an infected animal, the probability of early stages of disease are generally accompanied by fever,
infection is low, albeit existent. Direct contact with a source of myalgia, vomiting, abdominal cramping, meningeal irrita-
virusnamely, an actively or latently infected animal (or cells tion, and such cranial nerve signs as nystagmus and diplopia.
obtained from an infected animal)is the means by which an Neurologic symptoms develop very rapidly. Altered sensation,

Scheld_Ch14.indd 205 2/21/14 5:38 PM


206 Part II: Viral Infections and Related Disorders

hyperesthesia, or paresthesia of the limbs usually precede evi- contact with workers exposed to these animals should be
dence of weakness, areflexia, and flaccid paralysis. Transverse considered as having B-virus infection if compatible clinical
myelitis with urinary retention is likely to occur. With progres- findings occur. The suspicion of B-virus infection should be
sive involvement of the CNS, decreased levels of conscious- reinforced in the presence of historical evidence of direct con-
ness, altered mentation, respiratory depression, seizures, and tact with a rhesus monkey, either by bite, scratch, or labora-
ultimately neurologic death ensue. tory accident. In addition, some cases have been acquired by
Although the incubation period for B virus has been de- exposure to monkey tissues; consequently, this route of trans-
bated, most cases have a relatively short incubation period of mission should not be excluded as a possibility for human
approximately 3 to 5 days; however, some cases have been infection.
reported to occur as late as 24 days after an animal bite. The Clinical evidence of disease has been defined earlier. Of par-
onset of neurologic symptoms generally occurs 3 to 7 days ticular interest to the clinician is the presence of a vesicular
after the appearance of a vesicular rash. Time to death var- rash at a bite site with ipsilateral regional lymphadenopathy.
ies and can occur as early as within 10 to 14 days or much With a compatible incubation period, the progressive appear-
later. Improvements in intensive care for critically ill patients ance of neurologic symptomatologyparticularly altered sen-
probably have influenced these survival data. The progression sation in the extremities, weakness, hyporeflexia or areflexia,
of infection is likely related to a variety of factors, including and altered mentationin the presence of a wound should
host immunologic status quantity of neutralizing antibodies suggest the possibility of B-virus infection.
to HSV, age of the patient, site of the bite, and quantity of
virus inoculated. Prolonged incubation periods, as well as the
mechanism for these prolonged incubation periods, have not Virus Isolation
been well characterized (7).
Third, cases have been documented following exposure to Recovery of virus from humans suspected of having B-virus
infected secretions by an ocular or respiratory route. An ani- infection must be attempted. Sources for retrieval of virus are
mal care provider was exposed to direct inoculation of saliva those indicative of the pathogenesis of disease. These include
into the eye. In spite of rigorous rinsing, she became infected vesicles, conjunctivae, pharyngeal swabs, and tissue biopsy
and subsequently died (7). Two other cases illustrate the po- material. Furthermore, retrieval of virus from the cerebro-
tential for respiratory spread. Symptoms in these individuals spinal fluid (CSF) should be attempted, although the yield is
were localized to the respiratory tract, including coryza, cough, very low. Specimens for virus isolation should be processed
laryngitis, and pharyngitis. While associated with fever, these in cell lines that are susceptible to B virus. As noted earlier
symptoms progressed to respiratory distress, as evidenced by a in this chapter, these include primary vervet monkey kidney,
radiographic evidence of interstitial pneumonitis. One of these rabbit kidney cells, or established strains such as BS-C-1 or
two patients had virus isolated from a vesicle. Subsequently, LLC-RK-1. B virus replicates in all these cell lines, in contrast
neurologic symptoms developed, ultimately leading to death to other simian viruses, which have a more limited spectrum of
of these patients (5). susceptible cell lines (24).
The fourth manifestation of human disease is recurrent in- Once viral isolation has been achieved, identification of
fection. At least two patients illustrate that a recurrent vesicu- virus can be accomplished using either molecular methods
lar rash can occur as a consequence of B-virus infection. In one (25,26) or neutralization assays, although the latter assay is
case, infection was acquired as a consequence of person-to- cumbersome and tedious (5,27,28). Rapid identification of
person transmission. In another case, an individual developed isolates is essential for purposes of intervention with appropri-
lesions compatible with the diagnosis of herpes zoster; however, ate therapeutic agents (29,30).
B virus was isolated from these lesions. Of the well-described
cases of herpes B-virus infection, the majority developed en-
cephalitis (90%), with a documented mortality rate of 75%.
Survivors of B-virus infection of the CNS have a broad spec-
Serologic Evaluation
trum of neurologic impairment. Of those documented in the Serologic determination of B-virus infection is exceedingly
literature before the availability of antiviral therapy, two had difficult because of its cross reactivity with HSV. Attempts
mild or minimal impairment, one had moderate neurologic im- at antibody absorption are not satisfactory. The extensive
pairment, and the remainder had severe impairment. The lit- cross reactions of sera to simian viruses in the presence of
erature also documents three patients who were infected with HSV antibodies have made diagnosis difficult (5,31,32).
B virus and survived infection but did not develop encephalitis. Animal workers providing care for Macaca monkeys should
One had severe neurologic impairment (presentation of herpes be serially bled for antibody determinations. Sera should be
zoster), and two appeared normal on follow-up. With the use banked for future reference. The standard serologic assays
of antiherpetic drugs, survivors who were treated early, even that have been employed generally use either neutralization
if they had CNS disease, tended to survive without significant assays or complement fixation (33). In general, complement-
impairment (2123). Thus, taken together, these cases serve to fixing antibody titers to HSV are higher in humans than in
emphasize the severity of B-virus disease. monkeys, whereas titers against B virus are higher in Macaca
monkeys than in humans. As previously mentioned, human
sera contain higher levels of neutralizing antibodies to HSV
DIAGNOSIS than to B virus. Furthermore, sera derived from monkeys
with B-virus infection have been reported to neutralize
Clinical HSV better than they neutralize the endogenous virus. No
simple test exists to distinguish antibodies to HSV from
The total number of cases of B-virus infection is small. The antibodies to B virus. Nevertheless, a variety of assays for
possibility of B-virus infection must be considered in any indi- the rapid evaluation of human antibody response are under
vidual having contact with Old World monkeys, particularly development (29,34). The advent of these newer diagnostic
Asiatic Macaca species. Because of the recent identification of assays will be of value for prospectively evaluating patients
person-to-person transmission, individuals having intimate exposed to B virus.

Scheld_Ch14.indd 206 2/21/14 5:38 PM


Chapter 14: B Virus 207

TA B L E 1 4 . 1 animal workers (7,37). Although the risk of acquisition of in-


fection by B virus is extremely low, caution is indicated (37).
LABORATORIES THAT PERFORM TESTS FOR Guidelines for the management of macaques in captivity
B VIRUS (7) for research purposes include the following:
Physician, Laboratory, and 1. Macaque monkeys should be used only for research pur-
Contact Information Available Tests for B Virus poses and only when clearly indicated.
2. It is desirable to use macaques that are free of B-virus in-
Dr. Julia Hilliard Culture, serologic testing, fection. Thus, screening for B-virus infection before their
B Virus Research Resource and PCR analysis of use for research purposes is necessary. Furthermore, these
Laboratory specimens from humans animals should be maintained in a state that will prevent
Georgia State University or nonhuman primates acquisition of infection (5,6). Attempts are in progress to
P.O. Box 4118 establish B-virusfree colonies.
Atlanta, GA 30302-4118 3. All macaques should be considered infected unless proven
Tel.: (404) 651-0808 otherwise. This point warrants emphasis because it en-
E-mail: biojkh@panther.gsu.edu courages careful methods of handling these animals.
Dr. David Brown Culture, serologic testing, 4. Macaque handlers should be properly attired (arm-length
Central Public Health and PCR analysis of leather gloves, protective eyewear, and mask) and prepared
Laboratory specimens from humans to use proper restraints or anesthetic agents, such as ket-
Enteric, Respiratory, and or nonhuman primates amine, before manipulating animals.
Neurological 5. Cages and equipment for housing and transport of
Virus Laboratory macaques, as well as laboratory equipment with which
61 Colindale Avenue macaques have contact, should be thoroughly cleaned.
London NW9 5HT, England Sharp edges can lead to wounds in the macaques, which
Tel.: 44-208-200-4400 may ultimately be a source of infection.
E-mail: dbrown@phls.org.uk 6. Animal handlers, investigators, and support personnel
who handle macaques should be properly trained in the
Dr. Seymour S. Kalter Culture and serologic methods of animal restraint and manipulation. Every ef-
Esoterix testing of specimens fort must be made to educate personnel in bite prevention.
7540 Louis Pasteur Drive, from nonhuman Serial and prospective serologic evaluation of individuals
Suite 200 primates only who have continual contact with these animals should be
San Antonio, TX 78229 performed and sera specimens maintained at a repository
Tel.: (210) 614-7350 for future evaluation, if necessary. Knowledgeable medical
E-mail: sy.kalter@esoterix.com personnel should be readily available for consultation.
7. Any bite or scratch wound incurred from a macaque or as
From Cohen JI, Davenport DS, Stewart JA, et al. Recommendations a consequence of contact with a macaque cage should be
for prevention of and therapy for exposure to B virus (cercopithecine decontaminated as quickly as possible. Cleansing with soap
herpesvirus 1). Clin Infect Dis. 2002;35:11911203.
and water should be done for at least 15 minutes. If an eye
is involved, a sterile saline wash should be used. Cultures of
both the animal and the wound site should be obtained im-
mediately. Follow-up evaluation of these individuals should
include clinical and serologic evaluation. If there is evidence
Polymerase Chain Reaction Detection of suspected disease, assessments should proceed, with virus
isolation and further serologic evaluation. With evidence of
of B-Virus DNA disease, specific antiviral therapy should be instituted, as
noted in the next section, Prevention and Treatment.
Recently, a single-step polymerase chain reaction (PCR) assay
has been reported to be of value for diagnostic purposes (35). These recommendations have been provided as guidelines
Because of the rarity of disease, field performance character- for the management of macaques in captivity for research
istics need to be determined. Primate workers should carry a purposes (7,39). Variations from these guidelines can be dis-
card that indicates symptoms of B virus and contact informa- cussed with investigators at the CDC (Viral Exanthems and
tion for clinical and laboratory consultation regarding B virus Herpesvirus Branch, Division of Viral Diseases, Centers for
(Table 14.1). Disease Control and Prevention, 404-329-1338, www.cdc
.gov/ncidod/diseases/bvirus.htm).

CONTROL OF B-VIRUS
INFECTION PREVENTION AND TREATMENT
As with all other infectious diseases, the principal goal is to Treatment
prevent infection. Prevention has been stressed on numerous
occasions by the Centers for Disease Control and Prevention Although vaccines have been evaluated in animal models,
(CDC) and in reviews published over the last 15 years (5 none has proven efficacious in humans (17,4042). The use of
7,3638). Although a vaccine is not readily available for the hyperimmune serum or -globulin has not been proven effec-
prevention of B-virus infections, one is under development tive for the treatment of human infections caused by B virus
(37). Proper laboratory and animal breeding and handling (43). Other investigators have demonstrated protection with
procedures will significantly decrease the risk of infection. hyperimmune horse and rabbit sera for rabbit infection (44).
A CDC Advisory Committee has recommended procedures The use of other forms of hyperimmune sera has not been
for control and management of infected animals and exposed proven effective.

Scheld_Ch14.indd 207 2/21/14 5:38 PM


208 Part II: Viral Infections and Related Disorders

With the advent of antiviral therapy, nucleoside analogs a minimum of 14 days at dosages of 5 mg/kg every 12 hours
have been deployed for the management of B-virus infection IV (7). The duration of therapy is not known but should con-
of humans. Four drugs have been used experimentally: vida- tinue until all CNS symptoms have resolved. Subsequent oral
rabine, acyclovir, valacyclovir, and ganciclovir. Each of these administration of valacyclovir for suppression of latent infec-
drugs has been reported to have varying activity. Studies with tion is considered by many experts; however, there are no data
an acyclovir-based medication have attracted the most atten- regarding the value of therapy for prolonged periods under
tion. In cell culture systems, acyclovir is active against B-virus such circumstances. Furthermore, the value of valacyclovir for
infection. One milligram of acyclovir decreased the yield of life is recommended by some experts.
virus by approximately 90% (45). Subsequent experiments in
animal models provided varying evidence of efficacy, depending
on the quantity of inoculation, delay in therapy, and duration Prophylaxis
of treatment. Overall, a beneficial effect could be achieved in
animals (45). Smith et al. (46) have demonstrated that ganciclo- Valacyclovir should be used for prophylaxis in patients ex-
vir is extremely active against B virus at concentrations of 0.55 posed to B virus. The dosage is 1 g every 8 hours and should
mg/mL or less. These concentrations are achievable in humans. be continued until the results of laboratory tests are available.
More recently, animal model studies have shown the efficacy of
acyclovir and superiority of ganciclovir for disease therapy (15).
The use of acyclovir and its prodrug valacyclovir for ther- FUTURE DIRECTIONS
apy of human disease has been more limited. The literature
includes several cases in which therapy instituted early in the B-virus infection in humans should not occur. The proper
disease course may have slowed progression and led to a return deployment of control procedures among animal handlers
to normal function (20). Two of these cases were reported in should avoid all human infections. Regardless, the future will
the Pensacola outbreak. The value of acyclovir under such cir- allow for the development of more rapid and precise diagnos-
cumstances cannot be unequivocally determined because of the tic assays and the further definition of appropriate procedures
small number of patients who have this disease for evaluation for handling infected animals and humans at risk for infection.
in controlled studies. If acyclovir is used initially for cutaneous
disease, some experts recommend a dose of 12.5 to 15.0 mg/
kg every 8 hours intravenously (IV) (7). With the availability ACKNOWLEDGMENTS
of the prodrug of acyclovir, valacyclovir, higher plasma levels
can be achieved (45). Thus, a recommendation for valacyclovir Original studies performed by the investigators were sup-
therapy is made because of the severity of disease and its uni- ported by grants from the National Institute of Allergy and
formly near-fatal outcome (7). The dose is 1 g every 8 hours. Infectious Diseases (NO1-AI-62554, NO1-AI-30025), the
Some experts recommend ganciclovir therapy for symp- Division of Research Resources (RR0023) of the National
tomatic B-virus disease, particularly if the CNS is involved, for Institutes of Health, and the State of Alabama.

References
1. Gay FP, Holden M. The herpes encephalitis problem. J Infect Dis. 14. Benda R, Cinatl J. Isolation of two plaque variants from the prototype
1933;53:287303. strain of B virus (herpesvirus simiae). Acta Virol. 1966;10:178.
2. Sabin AB, Wright WM. Acute ascending myelitis following a monkey bite, 15. Falke D. Isolation of two variants with different cytopathic properties from
with the isolation of a virus capable of reproducing the disease. J Exp Med. a strain of herpes B virus. Virology. 1961;14:492.
1934;59:115136. 16. Hilliard JK, Eberle R, Lipper SL, et al. Herpesvirus simiae (B virus): repli-
3. Sabin AB. Studies on the B virus. I. The immunological identity of a virus cation of the virus and identification of viral polypeptides in infected cells.
isolated from a human case of ascending myelitis associated with visceral Arch Virol. 1987;93:185198.
necrosis. Br J Exp Pathol. 1934;15:248268. 17. Whitley R, Hilliard J. Cercopithecine herpes virus 1 (B virus). In: Knipe
4. Sabin AB. Studies on the B virus. II. Properties of the virus and pathogenesis D, Howley P, Griffin D, et al., eds. Fields Virology. 5th ed. Philadelphia:
of the experimental disease in rabbits. Br J Exp Pathol. 1934;15:269279. Lippincott Williams & Wilkins; 2007:2889903.
5. Hull RN. The simian herpesviruses. In: Kaplan AS, ed. The Herpesviruses. 18. Boulter EA. The isolation of monkey B virus (herpesvirus simiae) from the
New York: Academic Press; 1973:389425. trigeminal ganglia of a healthy seropositive rhesus monkey. J Biol Stand.
6. Palmer AE. B virus, herpesvirus simiae: historical perspective. J Med 1975;3:279280.
Primatol. 1987;16:99130. 19. Kalter SS, Weiss SA, Heberling RL, et al. The isolation of a herpesvirus
7. Cohen JI, Davenport DS, Stewart JA, et al. Recommendations for preven- from the trigeminal ganglia of normal baboons (Papio cynocephalus). Lab
tion of and therapy for exposure to B virus (cercopithecine herpesvirus Anim Sci. 1978;28:705709.
1). Clinical infectious diseases : an official publication of the Infectious 20. Benson PM, Malane SL, Banks R, et al. B virus (herpesvirus simiae) and
Diseases Society of America. Clin Infect Dis. 2002;35(10):1191203. human infection. Arch Dermatol. 1989;125(9):12471248.
8. Burnet FM, Lush D, Jackson AV. The propagation of herpes B and pseudora- 21. Holmes GP, Hilliard JK, Klontz KC. B virus (herpes virus simiae) infec-
bies viruses on the chorioallantois. Aust J Exp Biol Med Sci. 1939;17:3552. tion in humans; epidemiologic investigation of a cluster. Ann Intern Med.
9. Wood W, Shimada FT. Isolation of strains of virus B from tissue cultures of 1990;112:833839.
cynomolgus and rhesus kidney. Can J Public Health. 1954;45:509518. 22. Davenport DS, Johnson DR, Holmes GP, et al. Diagnosis and management
10. Reissig M, Melnick JL. The cellular changes produced in tissue cultures by of human B virus (herpesvirus simiae) infections in Michigan. Clin Infect
herpes B virus correlated with concurrent multiplication of the virus. J Exp Dis. 1994;19:3341.
Med. 1955;101:341351. 23. Centers for Disease Control and Prevention. B virus infections in humans
11. Krech U, Lewis LJ. Propagation of B virus in tissue culture. Proc Soc Exp Michigan. MMWR Morb Mortal Wkly Rep. 1989;38:453454.
Biol Med. 1954;87:174178. 24. Vizoso AD. Heterogeneity in herpes simiae (B virus) and some antigenic
12. Hopps HE, Bernheim BC, Nisalak A, et al. Biologic characteristics of a relationship in the herpes group. Br J Exp Pathol. 1974;55:471477.
continuous kidney cell line derived from the African Green Monkey. J 25. Benda R, Prochazka O, Cerva L, et al. Demonstration of B virus (her-
Immunol. 1963;91:416424. pesvirus simiae) by the direct fluorescent antibody technique. Acta Virol.
13. Hull RN, Dwyer AC, Cherry WR, et al. Development and character- 1966;10:149154.
istics of the rabbit kidney cell strain, LLC-RK. Proc Soc Exp Biol Med. 26. Hilliard JK, Kalter SS. Development of molecular probes for simian herpes-
1965;118:10541059. virus detection. Dev Biol Stand. 1985;59:7986.

Scheld_Ch14.indd 208 2/21/14 5:38 PM


Chapter 14: B Virus 209

27. Kalter SS. Virus studies on the normal baboon. In: Vagtborg H, ed. The 37. Hirano M, Nakamura S, Mitsunaga F, et al. Efficacy of a B virus gD
Baboon in Medical Research. Austin: Austin University Texas Press; DNA vaccine for induction of humoral and cellular immune responses in
1965:416417. Japanese macaques. Vaccine. 2002;20:25232532.
28. Kalter SS, Hutt R. Serodiagnosis of herpesvirus infection in primates. Dev 38. Liao G, Li R, Li C, et al. Safety and immunogenicity of inactivated polio-
Biol Stand. 1978;41:23540. virus vaccine made from Sabin strains: a phase II, randomized, positive-
29. Heberling RL, Kalter SS. A dot-immunoblotting assay on nitrocellulose controlled trial. J Infect Dis. 2012;205(2):237243.
with psoralen inactivated herpesvirus simiae (B virus). Lab Anim Sci. 39. Centers for Disease Control and Prevention. Updated CDC recommenda-
1987;37:304308. tions for the management of hepatitis B virus-infected health-care providers
30. Hilliard JK, Munoz RM, Lipper SL, et al. Rapid identification of herpes- and students. MMWR Recomm Rep. 2012;61(RR-3):112.
virus simiae (B virus) DNA from clinical isolates in nonhuman primate 40. Buthala DA. Studies on herpesvirus simiae (B virus) inactivation and at-
colonies. J Virol Methods. 1986;13:5562. tempts at vaccine production. J Infect Dis. 1962;111:95100.
31. Boulter EA, Kalter SS, Heberling RL, et al. A comparison of neutralization 41. Hull RN, Nash JC. Immunization against B virus infection. I. Preparation
tests for the detection of antibodies to herpesvirus simiae (monkey B virus). of an experimental vaccine. Am J Hyg. 1960;71:1528.
Lab Anim Sci. 1982;32:150152. 42. Hull RN, Peck FB Jr, Ward TG, et al. Immunization against B virus infec-
32. van Hoosier GL, Melnick JL. Neutralizing antibodies in human sera to tion. II. Further laboratory and clinical studies with an experimental vac-
herpesvirus simiae. Tex Rep Biol Med. 1961;19:376380. cine. Am J Hyg. 1962;76:239251.
33. Gary WG, Palmer EL. Comparative complement fixation and serum neu- 43. Boulter EA, Zwartouw HT, Thornton B. Postexposure immunoprophylaxis
tralization antibody titers to herpes simplex virus type 1 and herpesvirus against B virus infection. Br Med J (Clin Res Ed). 1982;284:746.
simiae in Macaca mulatta and humans. J Clin Microbiol. 1977;5:465470. 44. Buthala DA. Hyperimmunized horse anti-B virus globulin: preparation and
34. Katz D, Hilliard JK, Eberle R, et al. Elisa for detection of group-common effectiveness. J Infect Dis. 1962;111:101106.
and virus-specific antibodies in human and simian sera induced by herpes 45. Boulter EA, Thornton B, Bauer EJ, et al. Successful treatment of exper-
simplex and related simina viruses. J Virol Methods. 1986;14:99109. imental B virus (herpesvirus simiae) infection with acyclovir. Br Med J.
35. Hirano M, Nakamura S, Mitsunaga F, et al. One-step PCR to distinguish B 1980;280:681683.
virus from related primate alphaherpesviruses. Clin Diagn Lab Immunol. 46. Smith KO, Galloway KS, Hodges SL, et al. Sensitivity of equine herpes-
2002;9:716719. viruses 1 and 3 in vitro to a new nucleoside analogue, 9-[{2-hydroxy-
36. Estep RD, Messaoudi I, Wong SW. Simian herpesviruses and their risk to 1-(hydroxymethyl)ethoxy} methyl]guanine. Am J Vet Res. 1983;44:
humans. Vaccine. 2010;28(suppl 2):B78B84. 10321035.

Scheld_Ch14.indd 209 2/21/14 5:38 PM


CHAPTER 15 ARTHROPOD-BORNE VIRAL
ENCEPHALITIDES
TOM SOLOMON, ADJANIE PATABENDIGE, AND RICHARD J. WHITLEY

Conventionally, viruses are placed within families and genera includes Toscana virus (TOSV), an important emerging
according to the taxonomic system, based on morphologic, cause of CNS disease in southern Europe. The Reoviridae
biochemical, antigenic, and genetic similarities. However, family includes Colorado tick fever virus (CTFV) and similar
because the taxonomic system tells us nothing about how members of the Coltivirus genus that causes febrile illness
a virus is transmitted or about the patterns of disease it and CNS disease.
causes, it is of relatively little help to the practicing clini- Arboviruses classically cause three disease patterns in
cian. For this reason, alternative groupings that incorpo- humans (Fig. 15.1): fever-arthralgia-rash syndromes, viral
rate these clinical and epidemiologic aspects are often used. hemorrhagic fevers (often associated with hepatitis), and neu-
One such group is the arthropod-borne viral (or arboviral) rologic disease, which are the focus of this chapter. They con-
encephalitides that includes viruses transmitted by arthro- stitute some of the most important emerging and reemerging
pods (insect, ticks, sandflies, and biting midges) that are infectious diseases. General principles of arboviral ecology,
important causes of central nervous system (CNS) disease epidemiology, and pathogenesis, as well as diagnosis, manage-
in humans. Altogether, there are more than 500 arthro- ment, and prevention of arboviral encephalitis, are discussed
pod-borne viruses, or arboviruses, that come from four before the more important diseases are considered in more
viral families (Togaviridae, Flaviviridae, Bunyaviridae, and detail.
Reoviridae); only a relatively small number of these viruses
are responsible for human disease (Table 15.1). Within the
family Togaviridae, the Alphavirus genus contains eastern, ECOLOGY OF ARBOVIRUSES
western, and Venezuelan equine encephalitis (EEE, WEE,
and VEE) virusesimportant causes of disease in horses and A basic understanding of the ecology of arboviruses is help-
humans in the Americasand chikungunya virus (CHIKV), ful in understanding the epidemiology of human disease.
which caused major outbreaks associated with severe mor- For most of the arboviruses that cause neurologic disease
bidity in the Indian Ocean islands and in India during in humans, including many of the flaviviruses, alphaviruses,
2005 to 2007 (1). The flaviviruses (genus Flavivirus, fam- and bunyaviruses, humans are not part of the natural cycle,
ily Flaviviridae) include Japanese encephalitis virus (JEV), being incidental hosts (Fig. 15.2). In nature, these viruses
which occurs across much of southern and eastern Asia and are typically transmitted between birds or small rodents
is the most important cause of epidemic encephalitis world- in enzootic cycles that use mosquitoes or ticks as vectors.
wide; West Nile virus (WNV), which has spread to cause To be evolutionarily successful, arboviruses require immu-
large encephalitis outbreaks in the United States; dengue nologically naive hosts, which is why most have evolved
virus (DENV), which continue to be a global threat with to use rapidly reproducing animals. Humans are infected
over 100 million infected annually; and tick-borne encepha- when they encroach upon this natural enzootic cycle and
litis (TBE) virusesimportant in Europe and Russia. Within are bitten by the vector. This may be because they live in or
the family Bunyaviridae, the Bunyavirus genus includes La have entered areas where the virus circulates naturally or
Crosse virus (LACV) and other California serogroup viruses because changes in ecologic or environmental circumstances
that cause encephalitis, whereas the Phlebovirus genus have caused the virus to move closer to areas inhabited

TA B L E 1 5 . 1
VIRAL FAMILIES AND GENERA CONTAINING ARBOVIRUSES

Family Arboviral Genera Nonarboviral Genera

Togaviridae Alphavirus (eastern equine encephalitis) Rubivirus (rubella)


Flaviviridae Flavivirus (West Nile) Hepacivirus (hepatitis C)
Pestivirus (bovine viral diarrhea)
Bunyaviridae Bunyavirus (La Crosse) Hantavirus (sin nombre)
Phlebovirus (Toscana)
Nairovirus (Crimean-Congo
hemorrhagic fever)
Tospovirus (tomato spotted wilt)
Reoviridae Coltivirus (Colorado tick fever) Rotavirus (rotavirus)

An example from each genus is shown in parentheses.

210

Scheld_Ch15.indd 210 2/21/14 5:38 PM


Chapter 15: Arthropod-Borne Viral Encephalitides 211

FAR

CHK (a)
ONN (a)
RR (a)

VEE (a)
TOS (p)
RVF (p)
CTF (c)
EEE (a) WN (f) DEN (f) YF (f)
WEE (a) OHF (f)
KFD (f)
JE (f)
LAC (b) SLE (f)
MVE (f)
CNS TBE (f) CCHF (n) VHF

(a) Alphavirus genus (family Togaviridae) (f) Flavivirus genus (family Flaviviridae)
CHK = Chikungunya WN = West Nile
ONN = Onyong nyong JE = Japanese encephalitis
RR = Ross river SLE = St Louis encephalitis
VEE = Venezuelan equine encephalitis MVE = Murray valley encephalitis
EEE = Eastern equine encephalitis TBE = Tick-borne encephalitis
WEE = Western equine encephalitis DEN = Dengue
YF = Yellow Fever
(b) Bunyavirus genus (family Bunyaviridae) OHF = Omsk hemorrhagic fever
LAC = La Crosse KFD = Kyasanur Forest disease
FIGURE 15.1 Medically important
(c) Coltivirus genus (family Reoviridae) (p) Phlebovirus genus (family Bunyaviridae) arboviruses grouped according to
CTF = Colorado tick fever TOS = Toscana disease syndrome (top) and listed by
RVF = Rift Valley fever genus (bottom). FAR, fever-arthralgia-
(n) Nairovirus genus (family Bunyaviridae) rash; VHF, viral hemorrhagic fever;
CCHF = Crimean-Congo haemorrhagic fever CNS, central nervous system.

by humans. Sometimes a different mosquito carries the virus Arboviruses replicate in both the vertebrate host and
to humans (a bridging vector), and sometimes there is an the vector in an extrinsic cycle. Whereas vertebrate hosts
intermediate vertebrate cycle that brings the virus closer to typically mount an immune response and clear the virus,
humans (e.g., equines for some of the alphavirus encepha- the arthropod vectors are usually infected for life. Virus is
litides and swine for JEV). A large outbreak of disease in ingested by the vector with an infected blood meal, repli-
animals is an epizootic. Whereas natural hosts have pro- cates in the epithelium of the mesenteron (midgut), then
longed and high viremia, humans have transient and low disseminates to the salivary glands, where it replicates fur-
viremia, insufficient to transmit the virus further, and are ther and is excreted with each blood meal. The virus may
considered dead-end hosts. Most human infections are also be passed on from the adult mosquitoes and ticks to
asymptomatic or result in a mild nonspecific febrile illness, their offspring, via transovarial transmission (Fig. 15.2).
but in a small proportion, the virus enters the CNS to cause For ticks, virus can also be transmitted vertically from the
the syndrome for which the virus is known. The brief and larval to nymph then adult stages (transtadial transmission).
low viremia makes diagnosis of human infection by virus Vertical transmission is an important means of overwinter-
isolation or nucleic acid amplification difficult, and for the ing (i.e., surviving the cool winter months) for some viruses,
most part, serologic tests are used. Although humans are but for others, the means of overwintering is uncertain. An
dead-end hosts for most arboviruses, some arboviruses exception to a typical arboviral cycle includes mechanical
are better adapted to replicate in humans, resulting in per- transmission via the vectors mouth parts. More importantly,
sistent and high viremias, allowing for further transmission some viruses, as well as being arthropod borne, can also
to a biting insect. Examples include VEEV, TOSV, CTFV, be directly transmitted between vertebrate hosts, bypassing
and DENV. These high viremias typically cause a charac- the arthropod vector, for example, humans can become in-
teristic arboviral fever-arthralgia-rash syndrome in most fected with Rift Valley fever virus (RVFV) from the body
patients; CNS manifestations tend to be milder, for example, fluids of infected animals (2), TBE virus can be transmit-
aseptic meningitis. Because of the high viremias, virus isola- ted to humans via infected goats milk (3), and WNV can
tion and polymerase chain reaction (PCR) are more success- be transmitted in infected blood products or transplanted
ful for these viruses. organs (4).

Scheld_Ch15.indd 211 2/21/14 5:38 PM


212 Part II: Viral Infections and Related Disorders

Bridging vector
Vertical transmission
Sometimes, the vector carrying
In transovarial transmission, the virus the virus to humans differs from
is passed from the vector to its eggs. that in the enzootic cycle. These
For ticks, transtadial transmission are usually anthropophillic
from larvae to nymphs, then to adults vectors i.e., they prefer biting
also occurs. humans.

For some viruses, this provides


the means of overwintering
(surviving through the winter
months).

Dead end host

For most arbovirus, humans


do not transmit the virus
further. But there are few
Enzootic cycle
important exceptions.
Most arboviruses exist naturally in an
animal cycle (strictly the virus should
cause disease in animals to be
called enzootic). If these are wild
animals, the term Sylvatic cycle is Epizootic cycle
also used. Ornithophilic mosquitoes
prefer feeding on birds. A large outbreak of disease in animal is FIGURE 15.2 Overview of arboviral
an epizootic. Sometime, this also acts as ecology. A hypothetical arboviral cycle,
an amplifying/bridging cycle, bringing with an explanation of some commonly
the virus closer to humans. used terms.

TLR7, TLR8, and TLR9 are the main viral sensors in humans.
PATHOGENESIS OF ARBOVIRAL TLR7, TLR8, and TLR9 are localized in the endosomal com-
ENCEPHALITIS partments and signal through the MyD88 adapter, leading to
type 1 IFNs and inflammatory cytokine production. TLR3 is
Despite the wide range of arboviruses that cause encephalitis, located both on the cell surface and in endosomal compart-
there are many similarities in the pathogenesis and pathology ments and induces production of type 1 IFNs through toll/
(see Table 15.2 for an overview of the arboviral encephalitides). interleukin-1 (IL-1) receptor (TIR) domain-containing adapter-
Much of the data has come from rodent models of alphavirus inducing IFN- (TRIF) adapter. Protein kinase R (PKR) and
encephalitis. Following inoculation, usually during the bite of a 2, 5-oligoadenylate synthetase (OAS) are two other cellular
blood-sucking mosquito or tick, arboviruses typically replicate viral sensors. Viral sensors activate IFN regulatory factors and
in the skin and local lymph nodes, causing a viremia, before transcriptional induction of IFNs, which then signal through the
entering the CNS (5). Langerhans cells in the skin have been JAK/STAT pathway leading to transcription of many interferon-
implicated as a site of replication for alphaviruses and some fla- stimulated genes (ISGs). The expression of ISGs and induction
viviruses (68). Keratinocytes have also been shown to be one of their products (e.g., RIG-I, MDA5, TLRs, OAS, PKR) are
of the initial targets for WNV replication and could be contrib- critical for limiting replication of arboviruses. Many arbovi-
uting to the persistence of WNV in the skin (9). Replication in ruses encode nonstructural proteins to counter the host immune
local skeletal muscle may also be important for alphaviruses. response. The main function of these nonstructural proteins
Invasion of target organs appears to depend in part on the is to antagonize the initial IFN response and is an important
extent of viremia, but also on other invasive viral characteristics adaptation that has helped arboviruses to establish themselves
(10). The two peaks of the characteristic saddleback or bipha- in numerous vertebrate hosts (12).
sic fever curve seen in many arboviral infections correspond to The mechanisms of virus entering the CNS may include
production of interferon (IFN) and proinflammatory cytokines infection of or transport across brain microvascular endothelial
during the initial viremia, and then a mononuclear inflamma- cells that form the bloodbrain barrier (BBB) and infection of
tory response in infected target organs (11). IFN production is olfactory neurons or of choroid plexus epithelial cells (1316).
induced when viral components are detected by cytoplasmic However, the exact mechanisms of virus entry into the CNS
proteins such as retinoic acid-inducible gene I (RIG-I) and mela- across the BBB and interactions of the virus with the BBB remain
noma differentiation-associated gene 5 (MDA5) during the incompletely understood, despite the critical role of the BBB in
unwrapping of the viral RNA from the nucleocapsid to allow preventing pathogen entry to the CNS (1720). In animal mod-
replication. In addition, cells use toll-like receptors (TLRs) els, the term neuroinvasiveness is used to describe a virus abil-
to recognize pathogen-associated molecular patterns. TLR3, ity to enter the nervous system following peripheral inoculation.

Scheld_Ch15.indd 212 2/21/14 5:38 PM


Chapter 15: Arthropod-Borne Viral Encephalitides 213

TA B L E 1 5 . 2
OVERVIEW OF THE MAJOR ARBOVIRAL ENCEPHALITIDES

Main Vectors
Geographic (Mosquitoes Unless
Family/Genus Genus Virus Distribution Natural Hosts Otherwise Stated) Human Disease

Togaviridae Alphavirus Eastern equine North, Central, Freshwater swamp Culiseta melanura; High mortality; CSF
encephalitis and South birds in North Aedes species are may resemble
America America, rodents bridging vectors bacterial meningitis
and marsupials
in South America
Western Western Canada, Passerine birds Culex tarsalis; Aedes No human cases since
equine western and (sparrows, melanimon 1994, relatively low
encephalitis midwestern house finches); mortality
United States blacktail
jackrabbit
Venezuelan Central and Rodents, aquatic Culex FAR syndrome
equine South America, birds (Melanoconion) common; CNS disease
encephalitis occasionally species in enzootic rare. Humans have
extending cycle; Psorophora high viremias; virus
further north and Aedes species also isolated from
invoved in equine pharynx; Nonlicensed
epizootics, and live attenuated vaccine
epidemics (TC-83) available
Chikungunya Tropical and Non-human Aedes aegypti and FAR syndrome;
subtropical primates and Aedes albopictus polyarthralgia is
regions small mammals are main vectors. common; persistent
of Africa, (monkeys, bats) Aedes furcifer- arthralgia in some
Southeast Asia, taylori, Aedes cases; CNS disease
and Australia africanus, Aedes more common in
luteocephalus, and children; evidence of
Aedes neoafricanus vertical transmission
are enzootic vectors. from mother-to-child
Flaviviridae Flavivirus West Nile Africa, Middle Passeriform birds Culex pipiens and Nosocomial
East, Europe, (jays, blackbirds, others transmission also
Asia, America, finches, occurs; febrile
Australia warblers, syndrome common;
sparrows, crows) disease is more
severe in elderly and
immunosuppressed
Japanese Southeast Asia, Herons, egrets, Culex Most important
encephalitis Pacific Rim, migrating birds; tritaeniorhynchus, arboviral
Northern swine Culex gelidus and encephalitis; seizures
Australia, Asian others and parkinsonian
subcontinent features common;
inactivated vaccine
(Biken) is licensed;
live attenuated
vaccine in China
St. Louis North, Central, Passeriform and Culex tarsalis in Endemic disease
encephalitis and South columbiform the west; Culex in the western
America birds pipiens, Culex United States; large
quinquefasciatus, outbreaks in the
Culex nigripalpus eastern United States
in the east
Murray Valley New Guinea, Herons, egrets, Culex annulirostris, Mostly affects
Australia aquatic birds and others aboriginal children
Tick-borne Europe, Siberia, Small forest Ixodes ricinus, Poliomyelitis-like
encephalitis Far East rodents Ixodes persulcatus, paralysis of upper
and other ticks limbs is common.
Inactivated vaccines
are licensed

(continued)

Scheld_Ch15.indd 213 2/21/14 5:38 PM


214 Part II: Viral Infections and Related Disorders

TA B L E 1 5 . 2
OVERVIEW OF THE MAJOR ARBOVIRAL ENCEPHALITIDES (CONTINUED)

Main Vectors
Geographic (Mosquitoes Unless
Family/Genus Genus Virus Distribution Natural Hosts Otherwise Stated) Human Disease

Dengue Almost every Humans, and Aedes aegypti and FAR syndrome (dengue
country nonhuman Aedes albopictus fever); syndrome of
between tropics primates vascular leak and
of Capricorn hemorrhage (dengue
and Cancer hemorrhagic fever);
neurologic disease
Bunyaviridae Bunyavirus La Crosse Midwestern and Chipmunks, Aedes triseriatus, Major pediatric
Eastern United squirrels Aedes albopictus arboviral encephalitis
States in United States;
fatalities are rare
Jamestown North America White tailed deer Aedes species, Culex Encephalitis in the
Canyon inornata elderly
California Western United Rabbits, other Aedes melanimon, Rare cause of human
encephalitis States, Canada rodents Aedes dorsalis disease
Tahyna Europe, Russia Domestic animals, Aedes vexans, Culex Newly recognized
rabbits annulata cause of febrile
illness with CNS
disease, especially in
Russia
Phlebovirus Toscana Mediterranean Not known Phlebotomus Mostly aseptic
countries perniciosus meningitis,
sandflies encephalitis rare
Rift Valley Africa, Middle Livestock Aedes and other Febrile illness with
Fever East mosquitoes; also retinitis hemorrhagic
direct transmission fever, encephalitis
from livestock (02%)
body fluids
Reoviridae Coltivirus Colorado tick Western United Squirrels, Dermacentor Meningitis in patient
fever States and chipmunks, andersoni ticks with leukopenia and
Canada other small thrombocytopenia
mammals

FAR, fever arthralgia rash.

Neurovirulence describes its ability to cause damage once is associated with a fatal outcome in human disease (3032).
within the CNS (e.g., following invasion from the periphery Antibody appears to be protective by neutralization, nonneu-
or following direct intracranial inoculation). Once inside the tralizing binding, and complement-mediated lysis of infected
nervous system, the pathogenesis may involve a combination cells (33). In animal models of alphavirus encephalitis, anti-
of infection and dysfunction of neurons, caused by direct viral body assists with nonlytic clearance of virally infected cells by
or indirect inflammatory damage and other mechanisms such a mechanism that appears to be synergistic with the effects of
as programmed cell death. The immune response to alphavirus IFN- (34). This may be especially important for clearing virus
infection has been characterized in experimental animals and from the CNS, where limited expression of major histocom-
includes an early innate response with type I interferon (IFN- patibility class (MHC) antigens in mice (or human leukocyte
and IFN-) production, followed by specific humoral and cel- antigen [HLA] antigens in humans) may restrict the role of T
lular immune responses (21). Experiments with knockout mice lymphocytes. Infiltration into the CNS of antibody-producing
have shown type I IFN limits early viral replication, possibly B cells has been demonstrated in animal models (35). Cellular
by production of MxA protein, a large cytoplasmic guanosine immune responses to alphavirus infection include lymphopro-
triphosphatase that prevents accumulation of viral genomic and liferative, cytokine, and cytotoxic. In alphavirus encephalitis,
subgenomic RNA (22). Tumor necrosis factor- (TNF-) and there is infiltration of natural killer cells, B cells, CD4, CD8
other proinflammatory cytokines are produced, contributing to T cells, and macrophages. CD8 T cells may be important in
the pathogenesis (23). Production of IFN- and other cytokines virus clearance from infected macrophages (36). Studies on
and chemokines has been implicated in JEV in humans (2426). humoral responses to flavivirus infection have shown protec-
The humoral immune response in human alphavirus and tion of mice by passive transfer of antibodies against lethal
flavivirus encephalitis includes early production of immuno- challenge by several flaviviruses (3739). Furthermore, B cell
globulin M (IgM), followed by immunoglobulin G (IgG) that deficient mice were shown to be highly susceptible to flavivirus
persists for years (2730). The failure of antibody production infection (37,4042). In contrast, the role of T cells during

Scheld_Ch15.indd 214 2/21/14 5:38 PM


Chapter 15: Arthropod-Borne Viral Encephalitides 215

flavivirus infection is less well characterized. A few studies parenchyma causes encephalitis; and damage in the spinal
have shown a protective role for CD4 T cells by enhancing cord causes myelitis. The term meningoencephalomyelitis
antibody production and sustaining flavivirus-specific CD8 encompasses the concept that all three components may be
T-cell responses (43,44). CD8 T cells have been shown to be affected. Despite the wide range of etiologic agents, there
important for clearance of virus and survival of mouse models are many similarities in the clinical presentations of arbovi-
of WNV (4548), whereas cytotoxic T cells caused accelerated ral encephalitis. After a brief incubation period, which typi-
and more severe pathogenesis in mice infected with MVEV cally is 4 to 8 days but may range from 1 to 28 days, patients
(49). In mice infected with JEV, CD8 T cells did not affect develop a nonspecific flulike illness with fever, headache, nau-
the outcome of the infection. Therefore, a conflicting role for sea, and vomiting. Often, there are upper respiratory tract
CD8 T cells in flavivirus encephalitis is suggested (40). symptoms, such as cough and sore throat, or abdominal pain
and diarrhea. Sometimes, a rash, conjunctival or pharyngeal
injection, or lymphadenopathy may give a clue to the diagno-
PATHOLOGY OF ARBOVIRAL sis. This may be seen for the fever-arthralgia-rash viruses,
ENCEPHALITIS for example, WNV, DENV, CTF, TOSV, or VEEV. The febrile
phase usually lasts a few days and may immediately be fol-
Few data are available for CNS infections caused by col- lowed by continued fever with the development of neurologic
tiviruses and phleboviruses because they are rarely fatal. symptoms and signs, or there may be an intervening asymp-
Pathologic studies of fatal human encephalitis caused by alpha- tomatic period, thus giving a saddleback or biphasic fever
virus, flavivirus, and bunyaviruses show many common find- curve characteristic of some arboviral infections, such as TBE
ings (5056). The leptomeninges are normal or slightly hazy, or CTF. Neurologic disease may manifest as subtle changes
and histologic examination shows an inflammatory infiltrate. in behavior, or mutism, resulting in an initial misdiagnosis
The brain parenchyma is congested with focal petechiae or of psychiatric illness. More often though, there is a profound
hemorrhage. Lesions are distributed through the gray matter reduction in consciousness level, which in children may be
of the cerebral cortex, midbrain, basal ganglia, cerebellum, heralded by seizures. Seizures and status epilepticus are espe-
brainstem, and spinal cord, providing the anatomic correlates cially common in children with JEV and LACV encephalitis
for many of the clinical features seen (see later discussion). (67,68). As well as obvious generalized tonic-clonic seizures,
Microscopically, there is perivascular cuffing, with infiltration subtle motor status epilepticus is being increasingly recog-
of mononuclear and polymorphonuclear cells into the paren- nized. A great range of focal neurologic signs may be seen in
chyma, as well as neuronophagia of infected cells by glial cells. arboviral encephalitis, including those of upper motor neu-
Immunohistochemical analysis of the lymphocyte populations ron damage, such as a hemiparesis, and cranial nerve signs
typically shows CD4 and CD8 cells, which are presumed indicative of brainstem damage. Flaccid limb weakness due
to be cytotoxic T cells. Similar changes are seen in animal to destruction of the anterior horn cells in the spinal cord is
models. Initially, there is perivascular infiltration of mono- seen in flavivirus encephalitis and EEEV. Most flaviviruses can
nuclear cells, with a few polymorphonuclear cells, which is also cause a poliomyelitis-like flaccid paralysis that typically
accompanied by perivascular extravasation of red blood cells affects the lower limbs for mosquito-borne flaviviruses such
(RBCs), endothelial cell swelling, and hyperplasia (57,58). as JEV, WNV, and Murray Valley encephalitis (MVE), and
This is followed by migration of lymphocytes and monocytes the upper limbs for TBE (6971). Extrapyramidal signs are
toward virus-infected neurons. The inflammation is accompa- common in both alphavirus and flavivirus encephalitis and
nied by gliosis and by inflammatory and glial cell apoptosis are thought to be the clinical correlates of the basal ganglia
(59). Demyelination is seen in encephalitis caused by some damage evident pathologically and radiologically, including
alphaviruses but does not seem to be important in the flavivi- increased tone causing generalized axial and limb rigidity, or
ruses (60). Although the cellular immunity and the inflamma- rigidity/spasm such as opisthotonus, and tremors of the limbs,
tory response are important in most infections, some patients eyelids, and tongue.
die from virally induced neuronal cell death before there is
evidence of a cellular immune response and inflammation.
Various processes contribute to neuronal cell death includ- DIFFERENTIAL DIAGNOSIS OF
ing apoptosis, cytoplasmic swelling, vacuolation, and mem- ARBOVIRAL ENCEPHALITIS
brane breakdown (61). In some instances, alphaviruses are
inadequately cleared from the CNS, as illustrated by detection The differential diagnosis of arboviral encephalitis is broad
of viral RNA in the brains of mice long after recovery (62,63). and includes other causes of viral encephalitis (Table 15.3),
Chronic progressive human disease with continuing brain in- as well as diseases that mimic viral encephalitis (Table 15.4).
flammation long after the acute disease has been reported in Certain epidemiologic features may give a clue that a patient
some alphavirus encephalitides (64). For example, persistent has an arboviral infection. For example, in parts of Asia, epi-
symptoms such as joint and muscle pains can last for several demics of JEV occur with great predictability after the start
years post CHIKV infection (1). Chronic infection does not of the rainy season. In the Americas, the alphavirus equine
appear to be important for most of the mosquito-borne flavivi- encephalitides are typically associated with illness in horses.
ruses, but there is evidence of persistent infection and chronic More recently, birds falling from the sky have heralded the
inflammation for humans infected with TBEV and in animal arrival of WNV. The age and occupation of the patient can
models (65,66). also point to epidemiologic risk factors. JEV tends to affect
children in rural parts of Asia, recent WNV encephalitis out-
breaks have affected the elderly or patients on immunosup-
CLINICAL FEATURES pressive drugs, and LACV encephalitis tends to affect children.
OF ARBOVIRAL ENCEPHALITIS A recent tick bite may suggest TBE in Europe, Russia, and the
Far East, or CTF in parts of North America, though other tick-
The clinical syndrome following neurologic infection with borne diseases, such as borreliosis, ehrlichiosis, and rickettsial
an arbovirus depends on the site of attack. If the meninges disease must be considered. A biphasic illness is characteris-
are involved, meningitis results; inflammation of the brain tic of some arboviral encephalitides. On examination, certain

Scheld_Ch15.indd 215 2/21/14 5:38 PM


216 Part II: Viral Infections and Related Disorders

TA B L E 1 5 . 3
VIRAL CAUSES OF ENCEPHALITIS

Encephalitis due to arboviruses, by geographic regiona Australasia


The Americas Murray Valley encephalitis, Japanese encephalitis, Chikungunya
West Nile, La Crosse, St. Louis, Rocio, Powassan Encephalitis due to other viruses, most of which are not geographically
encephalitis, restricted
Venezuelan, eastern and western equine encephalitis, Herpes viruses
dengue, Colorado tick fever
Herpes simplex virus, herpes zoster virus, Epstein-Barr virus,
Europe/Middle East cytomegalovirus
Tick-borne encephalitis, West Nile, Toscana, Dengue, Enteroviruses
Louping ill, Chikungunya
Poliovirus, coxsackieviruses, echoviruses, enteroviruses-70 and -71
Africa
Paramyxoviruses
West Nile, Chikungunya, dengue, (Rift Valley fever, Congo-
Measles virus, mumps virus, (Nipah virus, found only in Asia)
Crimean hemorrhagic fever)
Others
Asia
Rabies, influenza viruses
Japanese encephalitis, West Nile, dengue, Murray Valley
encephalitis, Chikungunya

a
Rarer or suspected arboviral causes are shown in parentheses.

TA B L E 1 5 . 4
DISEASES MIMICKING VIRAL MENINGOENCEPHALITIS

Central nervous system infections Parasites Neoplastic


Bacteria Cerebral malaria Primary brain tumor
Bacterial meningitis Toxoplasmosis Metastases
Tuberculosis Cysticercosis Paraneoplastic limbic encephalitis
Brain abscess Trypanosomiasis Metabolic
Typhoid fever Echinococcus Hepatic encephalopathy
Parameningeal infection Trichinosis Renal encephalopathy
Lyme disease Amoebiasis Hypoglycemia
Syphilis Rickettsiae Reye syndrome
Relapsing fever Rocky Mountain spotted fever Other
Leptospirosis Typhus Antibody-associated encephalitis
Mycoplasma pneumoniae Q fever - Voltage-gated potassium channels (VGKC)
Listeriosis Ehrlichiosis - N-methyl-d-aspartate receptor (NMDA-R)
Brucellosis Cat-scratch fever Drug reactions
Subacute bacterial endocarditis Parainfectious/postinfectious causes Subarachnoid hemorrhage
Whipple disease Guillain-Barr syndromea Cerebrovascular accidents
Nocardia Acute disseminated encephalomyelitisa Epilepsy
Actinomycosis Viral illnesses with febrile convulsions Hysteria
Fungi Shigella
Cryptococcus Viral infections associated with swollen
fontanelle
Coccidiomycosis Noninfectious diseases
Histoplasmosis Vasculitic
North American blastomycosis Behet disease
Candidiasis Cerebral systemic lupus erythematosus

a
Guillain-Barr syndrome and acute disseminated encephalomyelitis may follow viral or bacterial infections or vaccinations.

Scheld_Ch15.indd 216 2/21/14 5:38 PM


Chapter 15: Arthropod-Borne Viral Encephalitides 217

neurologic signs may suggest a patient has arboviral encepha- For most arboviral infections of humans, viremias are so
litis rather than encephalitis caused by herpes or other viruses. low and brief that they are undetectable by the time a patient
For example, in an unconscious patient, the presence of flaccid comes to the hospital (75) (Fig. 15.3); exceptions are the viruses
limbs consistent with anterior horn cell damage, or the trem- for which humans are important natural or amplifying hosts,
ors, increased tone, and rigidity spasms suggestive of basal such as DENV, VEEV, and TOSV. Virus may also sometimes
ganglia damage may suggest an arboviral cause. be isolated from the CSF, particularly for infections that prove
fatal, or from brain tissue at either biopsy or autopsy. The tradi-
tional method for virus isolation was inoculation into suckling
INVESTIGATION AND DIAGNOSIS mice, but this has largely been replaced by continuous cell lines
OF ARBOVIRAL ENCEPHALITIS (e.g., C6/36 cells derived from Aedes albopictus mosquitoes,
and Vero cells derived from African green monkey kidney).
A mild leukocytosis is common in many arboviral encepha- Amplification by PCR of viral nucleic acid in the CSF or serum
litides, but leukopenia with thrombocytopenia is sometimes tends to be more sensitive than virus isolation, particularly if
seen in TBE and CTF. Hyponatremia due to a syndrome of real-time Taq-Man PCR is used and has helped with diagnosis
inappropriate antidiuretic hormone (SIADH) secretion is com- of WNV and TOSV infections, although false-positive results
mon, and in some infections, alanine transaminase may be do occur. Immunohistochemistry of brain tissue may demon-
mildly elevated. In Western settings, lumbar puncture is often strate viral antigen in fatal cases, and antigen-capture enzyme-
delayed in severely ill patients until imaging has ruled out a linked immunosorbent assays (ELISAs) have been developed,
space-occupying lesion, or incipient brainstem herniation. In but they have limited utility.
many tropical settings where imaging is not available, lumbar Serologic methods to detect antiviral antibody are the main-
punctures are performed on most encephalopathic patients stay of the diagnosis of arboviral infections. The traditional
who do not have signs of brainstem damage because of the techniques such as hemagglutination inhibition, complement
importance of excluding other treatable causes, particularly fixation, indirect immunofluorescence, and neutralization tests
bacterial infections (72,73). Lumbar puncture will often re- require the demonstration of a greater than fourfold rise in an-
veal mildly elevated cerebrospinal fluid (CSF) opening pres- tibody titer in samples taken several weeks apart and thus have
sures (up to 250 mm of CSF), a lymphocytic pleocytosis with practical limitations; for example, they do not give a diagnosis
a normal CSF-to-plasma glucose ratio, and slightly elevated acutely and are not helpful in patients who die or are lost to
protein level. However, an early lumbar puncture may reveal follow-up. However, they do have specific research uses. For
no cells, or a neutrophil predominance, rather than a lympho- example, hemagglutinating antibodies are often broadly cross
cytic CSF. A low glucose ratio has been reported for EEEV and reactive, so in a patient with an undiagnosed encephalitis, a
CTF, whereas an elevated ratio has been reported for VEEVs. panel of antigens can be used to indicate whether a patient
Radiologic investigations may be helpful in excluding other was infected with a flavivirus, an alphavirus, a bunyavirus, or
conditions and in highlighting changes such as the high sig- other virus; this can then be followed by more specific tests
nal intensity seen on magnetic resonance imaging (MRI) in the within that group. Neutralizing antibodies persist for life, so
thalamus and basal ganglia that characterizes both alphavirus they can be used for serologic surveys to indicate whether in-
and flavivirus encephalitis (74). Computed tomography (CT) dividuals have ever been infected with a virus. In recent years,
scans may show low signal in the same areas or just edema. IgM and IgG capture ELISAs have become the most useful and
Although they do not reveal diagnostic information, electro- widely deployed tests for diagnosing arboviral encephalitis.
encephalograms (EEGs) can help identify seizures (particularly Depending on the virus, typically 50% of patients have anti-
the subtle motor seizures described earlier) and distinguish body on admission to hospital, and more than 90% have anti-
focal seizures from tremors. body 1 week later, so in patients whose initial test results are

Percentage
100
of patients
positive
(Log scale)
10

CSF lgM antibody


1
Viraemia

Day of illness: 3 1 1 3 5 7 9 11 30 60 90

Infection FIGURE 15.3 Diagnosis of arboviral


infections. As an example, the clinical
Clinical features: Fever CNS Disease Recovery
course of West Nile encephalitis: viremia,
development of antibody, and the impli-
Diagnosis: cations for diagnosis are shown. The
limits of virus detection are expressed
Virus detection Antibody detection as plaque-forming units (pfu)/100 L;
(limits of detection) human viremia is thought to be less than
Real-time PCR 0.1 pfu lgM ELISA of CSF 10 pfu/100 L. The first day of fever is
Virus Isolation 1 pfu ~50% positive by hospital admission taken as the first day of illness; most pa-
RT-PCR 10 pfu ~95% positive by day 10 of illness tients are not hospitalized until the third
to fifth day of illness.

Scheld_Ch15.indd 217 2/21/14 5:38 PM


218 Part II: Viral Infections and Related Disorders

negative, the test should be repeated after 1 week (Fig. 15.3). of the greatest importance (86,87). In many encephalopa-
Although they are the most useful tests, ELISAs too have their thies, if the blood pressure is well maintained, but increased
limitations. For example, because closely related viruses have ICP is suspected, 20% mannitol (0.5 to 1.0 g/kg) is given,
many shared epitopes, there may be cross reactivity in IgM although the little evidence available suggests the benefits, if
and IgG capture ELISAs between the viruses. This problem any, are only short term (68,88). Steroids are sometimes given,
can usually be overcome by testing for the related viruses in although they were not shown to be beneficial in JEV (89).
parallel, with the strongest reaction indicating the infecting Hypoglycemia is common in many tropical pediatric condi-
agent. However, sometimes a neutralization test, which is tions (90) and must be looked for and corrected.
more specific, may be needed. Importantly, antibody will be
detected in the serum of individuals who have had recent as-
ymptomatic or febrile infection but no neurologic disease. Its PREVENTION OF ARBOVIRAL
detection in the CSF is usually taken as a better indicator of
viral replication in the CNS. Finally, some patients may make ENCEPHALITIS
antibody late or not at all. These patients are more likely to
A consideration of the typical arboviral ecologic cycle
die and to have virus detected by another means such as isola-
(Fig. 15.1) reveals points at which intervention might be effec-
tion or PCR.
tive. These can be divided into measures to interrupt the natu-
Newer approaches for detection of flaviviruses antibody
ral cycle, measures to stop humans being bitten by infected
include the lateral flow assay and the microsphere immunoas-
vectors, and measures to stop infection resulting in disease (i.e.,
say (MIA). The lateral flow device was recently approved by
vaccination). For most of the arboviruses that cause encepha-
the U.S Food and Drug Administration (FDA) for the diag-
litis, because the natural cycle involves small wild birds and
nosis of WNV infection in humans, which can give a result in
rodents, interrupting this cycle is not possible. However, when
15 minutes (76). In MIA, an antigen is attached to encoded
an intermediate or amplifying vertebrate host brings the virus
microbeads that will bind the target analytes during the assay
closer to humans, for example, equines for VEEV and swine
procedure. These can then be identified using a fluorescence-
for JEV, measures can be taken. For example, vaccines that
activated cell sorting system (FACS). MIA is used by many
protect horses against VEE probably also reduce the number
laboratories in the United States for WNV diagnosis as it is
of human cases. Immunization of swine against JEV has been
highly sensitive compared to a standard ELISA. Furthermore,
practiced in Asia, though with limited success. However, relo-
one serum sample can be analyzed for reactivity to several an-
cation of swine away from homes is probably a more effective
tigens in a single assay (77). Other approaches include high-
way of reducing transmission of JEV to humans. Surveillance
throughput rapid microneutralization assays, biosensor-based
of mosquitoes, dead birds, and sentinel chickens (i.e., chickens
assays, microfluidic systems based on virus-coated magnetic
deliberately exposed to mosquito bites) for viral infection is
beads, and multiplex real-time RT-PCR assays (7881).
used in many Western settings as an indicator of viral activ-
ity and possible impending human disease, and antimosquito
measures are then implemented. Interventions to reduce the
TREATMENT OF ARBOVIRAL number of circulating mosquitoes depend on the species and
ENCEPHALITIS the setting. Measures include removing stagnant water from
swampy areas, ditches, and drains favored by many Culex
There are no established antiviral drugs for any arboviral en- species and removing the freshwater containers (car tires, tree
cephalitides. IFN-, ribavirin, and intravenous immunoglob- holes, litter) favored by Aedes species. Larvicides can also be
ulins have been given to patients with flavivirus encephalitis, applied to potential breeding sites (either biologic larvicides
based on in vitro and animal data. However, the few clini- such as Bacillus thuringiensis variance israelensis or the chemi-
cal data available suggest that IFN- is not effective in JEV cal larvicide, metoprine). With continuing human cases, public
(82). In the West, most patients with encephalitis will be health authorities have practiced ground or aerosol spray-
given acyclovir until herpes simplex virus type 1 (HSV-1) en- ing with pyrethroid formulations that kill adult mosquitoes.
cephalitis has been ruled out. The management of arboviral Newer interventions are under development including the
encephalitis focuses on treating the complications of disease, sterile insect technique and the Wolbachia-based strategies to
in particular hyponatremia, seizures, and increased intracra- control Aedes species (9194).
nial pressure (ICP). In most Western settings, severely ill pa- For many of these measures, evidence of beneficial impact
tients will be electively ventilated and managed in intensive is limited, but they demonstrate that public health authorities
care settings, allowing airway protection, seizure control, are taking the situation seriously. Personal measures to reduce
optimal assessment of fluid balance, and hyperventilation to the chance of being bitten by infected vectors are important.
reduce increased ICP. However, in many of the settings in These include using repellents that contain 10% to 30% DEET
which arboviral encephalitis occurs, this is not possible. At a (N, N-diethyl-3-methyl benzamide) on skin and clothing,
minimum, patients at risk of raised ICP should be nursed at applying permethrin insecticides on clothes, and wearing long-
30C, with the neck held straight to ensure the jugular venous sleeved shirts and trouser. In areas where tick-borne viruses
outflow is not impaired and with urine output monitored and circulate, regular inspection of the limbs ensures no ticks are
oxygen given. attached. Many of the Culex vectors bite at night, and the use
Seizures are common, particularly in children with flavivi- of impregnated bed nets is recommended. Public education is
rus and bunyavirus infections. In addition to obvious tonic- an important part of any of these measures. For some flavi-
clonic seizures, subtle motor seizures may be indicative of viruses (e.g., JEV and TBEV), inactivated or live attenuated
subtle motor status epilepticus (68). An EEG is advisable, par- vaccines are available, although their use is limited by cost and
ticularly in patients with twitching of a finger, lip, or eyelid. availability. The dramatic reduction in TBE in Austria follow-
Simple or complicated seizures should be managed according ing widespread vaccination demonstrates feasibility. However,
to local standard of care (8385) (see Chapter 1 where the because most of these viruses are enzootic, eradication by vac-
management of the critically ill patient with CNS infections is cinating humans is not possible. For other arboviral diseases,
discussed). A few specific notes relating to arboviral encepha- the sporadic nature and low incidence mean that the vaccines
litis are indicated. Attention to the etiology of increased ICP is would not be economically viable, even if they existed.

Scheld_Ch15.indd 218 2/21/14 5:38 PM


Chapter 15: Arthropod-Borne Viral Encephalitides 219

5 years later. The geographical area affected by EEE extends


ALPHAVIRUSES from Ontario and Quebec provinces in Canada, down the
eastern seaboard of the United States, and into South America
The alphavirus genus is one of two important genera in the as far as Argentina. To the West, the area extends as far as
family Togaviridae (named after the cloak, or toga that envelops Wisconsin and eastern Texas. The virus is transmitted between
the viruses) (11). Whereas alphaviruses are arthropod-borne wading birds, passerines, and other swamp birds by Culiseta
members of the other genus, the Rubivirus genus organisms melanura and other mosquitoes. It has also been isolated from
(e.g., rubella virus) are not arthropod borne and so are not con- crows (98). These mosquitoes are strictly ornithophilic (bird
sidered further here (Table 15.1). Alphaviruses cause two clini- biting). However, other mosquitoes that feed on both birds
cal syndromes. In Africa, Old World alphaviruses CHIKV and and mammals serve as bridging vectors carrying the virus to
onyong-nyong virus cause large outbreaks of fever arthralgia horses, humans, and other mammals (Fig. 15.2, Table 15.2).
and rash; a similar syndrome in which arthritis is predominant In South America, rodents and marsupials may be more im-
is caused by Ross River virus in Australia. In the Americas, portant natural hosts than birds. Outbreaks of disease have
New World alphaviruses EEE, WEE, and VEE viruses cause also occurred in commercial flocks of pheasants, partridges,
outbreaks of encephalitis in horses and humans and are the turkeys, and emus (99). In these circumstances, direct trans-
focus of this section (Table 15.2). EEE is notable for having mission between birds because of preening and pecking and
a higher case-fatality rate than other viruses in the group, transmission via the fecal-oral route have also occurred. The
whereas WEE is remarkable because although it has caused means by which the virus persists during the winter is not
large outbreaks in the past, it has now virtually disappeared known, but persisting viremia in birds or vertical transmis-
as a disease of humans. Although VEEV causes encephalitis sion into mosquito eggs has been postulated. Cases occur year
in equines, in humans it causes a febrile disease, with only a round but are more likely in the summer, especially when asso-
small proportion developing usually mild CNS disease. Two ciated with increased rainfall, which by creating a high water
other alphaviruses, Sindbis virus and Semliki Forest virus, have table augments the breeding habitat of C. melanura.
animal models of the pathogenesis of alphavirus encephalitis. EEE usually occurs as single cases and rarely as clusters
Phylogenetic analyses suggest that the alphaviruses arose in the that may be preceded by disease in horses. The median annual
New World with subsequent introductions into the Old World. number of cases in the United States is three (range, 1 to 14),
The WEE lineage appears to have derived as a recombinant of and the reported incidence is less than 0.1 to 0.4 per 107.
an ancestral EEE-like and Sindbis-like viruses (95). Seroepidemiologic studies show that humans are only rarely
infected with the virus. Fewer than 7% of residents older than
45 years had antibody in one study (100). Most infected pa-
Infectious Agent tients are asymptomatic with a ratio of symptomatic to asymp-
tomatic infections of about 1 in 30. However, the elderly and
Alphaviruses consist of a single strand of positive-sense RNA young children are more likely to develop disease.
wrapped in a nucleocapsid and surrounded by a glycoprotein-
containing lipid membrane that is derived from host cell plasma
membrane (11). The virion is 60 to 70 nm in diameter. The 11- to Clinical Features
12-kb RNA has a 5 cap and a 3 polyadenylated tail. It consists After an incubation period, which is thought to range from 3 to
of message-sense RNA that is directly infectious. It codes from the 10 days, patients present with a few days of febrile prodrome
5 to the 3 end for four nonstructural proteins, nsP1, nsP2, nsP3, and then neurologic disease. Early symptoms include head-
and nsP4, and five structural proteinscapsid (C), envelope 3 ache, myalgia, photophobia, abnormal sensations, vomiting,
(E3), E2, 6K (a 6-kd protein), and E1. The nonstructural proteins dizziness, and lethargy, followed by neck stiffness, a reduced
are involved in replication of the viral RNA and production of level of consciousness, and seizures. In infants, progression to
subgenomic RNA. The C protein has a conserved end with many coma may be more rapid, occurring over 48 hours. On exami-
basic amino acids and is presumed to bind the viral genomic RNA nation, a high pyrexia (39C) is common and one third of
to form the nucleocapsid. This nucleocapsid is an icosahedron patients have signs of meningismus. Clinical signs suggestive of
with T 4 symmetry. The E1 and E2 glycoproteins project from brainstem involvement include gaze deviation, nystagmus, and
the lipid membrane, forming heterodimers that group as trimers papillary abnormalities. This may be because of inflammatory
to form 80 knobs on the virion surface (96). Alphaviruses gain lesions in the brainstem, but uncal and subtentorial hernia-
entry into cells by attaching to the cell surface and entering via the tion have been seen at autopsy. Hemiparesis and limb spastic-
endocytic pathway to form vesicles. As the pH level of the vesicles ity suggest upper motor neuron involvement, whereas flaccid
decreases, the E1/R2 heterodimer dissociates, revealing a highly limbs suggest involvement of lower motor neurons in the spi-
conserved hydrophobic sequence in E1. This is thought to act as nal cord. Seizures may be generalized or focal. In patients who
a fusion peptide, causing viral membrane and host cell membrane remain conscious, aphasia and emotional lability may occur.
to fuse, and thus releasing the viral nucleocapsid into the host Limb dysesthesia and flaccid paralysis have also been reported
cytoplasm. Here the nucleocapsid is uncoated of its capsid protein in a fully conscious patient, suggesting myelitis may occur
so that naked RNA is exposed to ribosomes for initiation of trans- without brain involvement (101). Approximately one third of
lation as the first step of viral replication. Virus is replicated in patients die, but for those older than 60 years, the propor-
association with the endoplasmic reticulum and Golgi apparatus, tion is 50%. One third of survivors have moderate or severe
and new virions are released by budding at the host cell surface. sequelae (102).

Diagnosis
Eastern Equine Encephalitis
Peripheral leukocytosis, with neutrophil predominance can
occur. The CSF resembles that of acute bacterial meningitis
Epidemiology
with a neutrophil pleocytosis (as high as 5,000 cells/mm3); the
EEE virus was first isolated from the brain of a horse that RBC count and protein level are often elevated, and the glu-
died during an epizootic of equine encephalitis in New Jersey cose ratio is less than 50% in half of the patients (102,103). CT
and Virginia in 1933 (97). The first human isolate was made scans show diffuse cerebral edema in most patients. On MRI,

Scheld_Ch15.indd 219 2/21/14 5:38 PM


220 Part II: Viral Infections and Related Disorders

lesions that do not enhance with gadolinium are seen in the nerve palsies, hemipareses, hyporeflexia, spasticity, generalized
basal ganglia, thalamus, and brainstem (102). EEGs show rigidity, and occasionally opisthotonus. The overall case-fa-
background and focal slowing, as well as burst suppression. tality rate is approximately 4% to 10% (112), being higher
High-voltage delta waves carry a poor prognosis (103). among the elderly. WEE virus is more severe in infants, with
Virus isolation from the blood is unusual, although it has rapid progression from nonspecific illness to convulsions
been reported for EEE early in the illness (101). In fatal cases, and coma. Transplacental infection has been reported (113).
virus may be isolated from brain tissue or detected by PCR or The overall case-fatality rate is 3%, increasing to 8% for those
immunohistochemical staining. In most cases, the diagnosis is older than 50 years (114). Neurologic sequelae are common
made serologically by detecting antibody in the blood. Older in young children. Parkinsonian features including cogwheel
serologic tests such as hemagglutination inhibition, comple- rigidity and tremors have also been reported (115).
ment fixation, and neutralization tests are being replaced by
IgM capture ELISAs, which detect antibody in a single blood Diagnosis
or CSF sample, thus providing an earlier diagnosis.
The peripheral white blood cell count is usually normal or
Treatment and Prevention mildly elevated. Hyponatremia due to SIADH has been re-
ported. CSF opening pressures of more than 200 mm occur
Because there are no antiviral drugs, treatment is focused in two thirds of patients. CSF white blood cell counts are usu-
on the complications of infection, particularly seizures and ally less than 100 cells/mm3 but may range from less than 10
raised ICP and pneumonia. No vaccine against EEE is com- to 500 cells/mm3 with lymphocyte predominance, a slightly
mercially available, although an inactivated vaccine is used by elevated protein level, and a normal glucose ratio. CT scans
laboratory workers and others at high risk (104). Recently, a have been reported as normal, but EEGs show diffuse slowing
chimeric Sindbis (SINV)/EEEV candidates have been shown with focal delta activity in the temporal region, which may
to be safe and effective against protecting mice against EEEV mimic HSV-1 encephalitis (116). WEE virus is occasionally
(105). A similar vaccine is used to protect horses. Personal isolated from the CSF or from diagnostic brain biopsies (117).
protective measures against mosquito bites are described Detection of IgM in the CSF or serum is the preferred diagnos-
earlier in this chapter. tic test (30).

Treatment and Prevention


Western Equine Encephalitis
There is no specific treatment, although equine immune serum
Epidemiology was used following laboratory exposure in one patient (118).
A nonlicensed inactivated vaccine is available for laboratory
The isolation of WEE virus from the brain of horses dur- workers and others at high risk (119). A recent study has shown
ing an epizootic episode of unknown etiology in 1930 in the the potential of an adenovirus-vectored WEEV vaccine, Ad5-
San Joaquin Valley of California marked the first time an WEEV, for development into an emergency vaccine during an
arbovirus was isolated in the territorial United States (106). outbreak of WEEV (120). Furthermore, a human adenoviral
Subsequently, in 1938, WEE virus was obtained from a child vector (Ad5-mIFN) expressing mouse IFN- was shown to
who succumbed to encephalitis (107). Virus activity has been offer 100% protection against mice challenged with various
reported from western Canada, the western and midwestern WEEV strains (121). Surveillance and vector control measures
United States, and as far south as Argentina. In the western are practiced in some areas where the virus circulates.
United States, WEE virus is transmitted between passerine
birds (sparrows and house finches) by Culex tarsalis mosqui-
toes. Epidemic and epizootic activity occurs mainly in the sum- Venezuelan Equine Encephalitis
mer months. Highlands J virus, which is found in Florida, is
one of several related viruses in the WEE complex that do not
appear to cause disease in humans (108). Epidemiology
VEEV was first isolated from the brains of dead horses in 1938
Clinical Epidemiology (122), and subsequently six antigenically related subtypes (I
Until recently, exposure to WEE virus was common, with up through VI) have been identified based on serologic cross reac-
to 20% of humans having antibodies in some areas. However, tivity (123). Tonate virus is a subtype IIIB VEE complex virus
a decline in the rural population and changes in land use have that circulates in French Guiuan and has caused encephalitis
meant that even in areas where the virus circulates, this fig- (124,125). Subtype I viruses were further subdivided serologi-
ure is now less than 1% (109). The largest outbreak was in cally into IAB, IC, ID, IE, and IF. More recent molecular ge-
1941, with more than 3,000 human cases and hundreds of netic studies suggest a revision of some of these groups. VEEV
thousands of equine cases (110) and an estimated incidence of circulates in an enzootic sylvatic (forest) cycle (Fig. 15.2) and
167 per 100,000 in North Dakota, but since the 1990s, there emerges to cause major epizootics in horses and human epi-
have been only a handful of reported cases each year. The ratio demics every 10 to 20 years. Sylvatic VEEV is found in Central
of apparent to inapparent infections is estimated to be fewer and South America, and although most epizootics have oc-
than 1 in 1,000 in adults and 1 in 60 in children. curred in northern South America, particularly Venezuela and
surrounding countries, they have extended as far north as
Mexico and Texas (21,85,126). In its sylvatic cycle, VEEV is
Clinical Features transmitted between small mammals and aquatic birds mainly
The clinical features of WEE tend to be milder than those by Culex melanoconion mosquitoes (Table 15.2). Sporadic
of EEE. After an incubation period, which ranges from 2 to human cases occur when people enter the swampy and forested
10 days, there is a nonspecific febrile prodrome before neu- locations where the viruses circulate or when bridging mosqui-
rologic disease. Meningismus is seen in half the patients, and toes such as Aedes taeniorhynchus carry the virus to nearby
although weakness and tremors are common, fewer than 10% human habitats. In contrast, human epidemics occur at the
of patients develop coma (111). Focal signs include cranial same time as equine epizootics when the virus emerges from

Scheld_Ch15.indd 220 2/21/14 5:38 PM


Chapter 15: Arthropod-Borne Viral Encephalitides 221

its enzootic cycle and is transmitted between susceptible horses pleocytosis that may reach several hundred cells/mm3 and is
by Aedes, Psorophora, Mansonia, and Deinocerites mosquito reported to be associated with an elevated CSF glucose level.
species. Some studies suggest that the E2 protein encodes mos- As expected, given the relatively high viremias, VEEV can be
quito infection determinants for VEE, suggesting selection for isolated from blood and pharynx up to the eighth day of ill-
efficient infection of epizootic mosquito vectors may mediate ness and can be detected by PCR (134). VEEV antibody can
VEE emergence (127). Major epizootics and epidemics of VEE be detected by IgM capture ELISAs (138). A VEEV-specific
have occurred every 10 to 20 years in cattle-ranching areas blocking ELISA that also identifies serotype-specific antibodies
of Venezuela, Peru, Colombia, and Ecuador. Although epizo- against VEEV in sera of humans, equids, or rodents has been
otic viruses (subtypes IAB and IC) were originally thought to reported (139).
be maintained in a separate transmission cycle from enzootic
viruses (subtypes 1D, 1E, or 1F), later studies have suggested Treatment and Prevention
that epizootic IC viruses arise by mutation from sylvatic ID
viruses (128131). The 1C strains remain in the sylvatic cycle There is no antiviral treatment for VEE. Supportive measures
until the ecologic factors support its emergence into the epi- include attention to seizures and increased ICP. Equine vaccines
zootic cycle. Such outbreaks occur when heavy rainfall and for VEE interrupt transmission during outbreaks and prevent
flooding expand the mosquito breeding habitats and support epizootics. An inactivated vaccine has largely been replaced by
expansion of the rodent population but also require a large a live vaccine (TC-83), attenuated by serial passage in guinea
susceptible equine population. Outbreaks of VEE IAB subtype pig heart cells (140). VEEV is considered a useful agent for
that spread to Central America and Texas in the late 1960s and biological warfare, because of its potential for droplet spread
early 1970s are thought to have originated from improperly and tendency to cause an incapacitating flulike illness, rather
activated equine vaccine strains, as indicated by the fact that than a fatal encephalitis, and for this reason, the vaccine was
the epizootic strains are genetically almost identical to the vac- produced by the U.S. military. TC-83 is given to laboratory
cine strains (132). workers and produces neutralizing antibodies in more than
Seroprevalence studies indicate that up to 50% of the popula- 80% of recipients; however, it is associated with a high inci-
tion have antibody to VEEV in some sylvatic areas, whereas the dence of adverse events including fever and aseptic meningitis
remainder of the population is immunologically naive until an (37%). Most of those who do not respond immunologically
outbreak occurs. During the 1995 outbreak of VEE in Venezuela to the vaccine seroconvert when subsequently given a killed
and Colombia, an estimated 85,000 human cases occurred, of vaccine (TC-84). An improved genetically engineered vaccine
which 3,000 had neurologic disease and 300 were fatal (128,133). V3526 is being developed (141,142). Another vaccine can-
One third of the human population seroconverted, and 8% of the didate in development is the recombinant, chimeric Sindbis/
equines died. Unlike most of the other arboviral encephalitides, VEEV (SIN-83) that is more highly attenuated than TC-83
human viremias are sufficiently high to infect mosquitoes, sug- (143,144). In addition, the use of antisense technology and
gesting humans may not always be dead-end hosts. Isolation of humanized monoclonal antibody has been shown to be effec-
virus from the pharynx of up to 40% of patients suggests direct tive against mice infected with VEEV (145,146).
spread between humans is possible (134). No epidemiologic evi-
dence of such spread has ever been demonstrated (133,134).
Chikungunya Virus
Clinical Features
Epidemiology
Compared with the other alphavirus encephalitides, only a
very small proportion of symptomatic VEE infections results A large outbreak of a disease locally known as Chikungunya
in severe neurologic disease. The incubation period is brief in the Newala district of Tanganyika (now Tanzania) occurred
(1 to 5 days), and most patients then develop a febrile ill- in 1952 to 1953 (147). However, CHIKV may have caused
ness with severe headache, made worse by eye movements, outbreaks as early as 1779 as the disease was frequently mis-
photophobia, facial flushing, conjunctival injection, myal- taken with dengue (148). In the local language, Chikungunya
gia, arthralgia, nausea, vomiting, and dizziness. Pharyngeal means the disease that bends up the bones. CHIKV was
inflammation and pain associated with cervical lymphade- first isolated in 1952 from the serum of a febrile human dur-
nopathy is common, and there may be subcostal tenderness ing this outbreak (147,149). Although initially assessed as a
(133,135,136). Somnolence and tremulousness occur often. dengue outbreak, further characterization of the isolates by
More severe neurologic features occur in 4% to 14% of pa- serologic and antigenic techniques indicated that CHIKV was
tients, particularly the young and elderly. Seizures, particu- an alphavirus (150). Since then, frequent outbreaks of CHIKV
larly focal seizures, and raised ICP are common (133,137). have been reported from many African and Asian countries.
In 5% to 10% of hospitalized patients, cranial nerve palsies, There are three CHIKV genotypes (West African, East/Central
motor weakness, paralysis, or cerebellar signs are reported. African, and Asian) but only one CHIKV serotype, and there-
Respiratory tract infections, including interstitial pneumonia fore, infection with a given genotype should result in lifelong
and tracheobronchitis, are common and may result in second- immunity against any genotype (151).
ary bacterial infection. Overall, 0.2% to 1.0% of symptomatic In Africa, CHIKV typically circulates in a sylvatic, enzo-
VEE infections are fatal, but this increases to up to 10.0% to otic cycle involving wild primates and forest-dwelling mosqui-
25.0% of those with encephalitis. Children are estimated to toes with sporadic infection of rural human populations. The
have ten times the risk of neurologic disease of adults, and principle enzootic mosquito vectors include Aedes furcifer-
younger children have a greater case-fatality rate. taylori (southern and western Africa), Aedes africanus (cen-
tral Africa), Aedes luteocephalus and Aedes dalzieli (Senegal),
Aedes cordellieri (South Africa) and Aedes neoafricanus.
Diagnosis Human migration has attributed to the introduction of CHIKV
Rather like the arboviral fever-arthralgia-rash syndromes, to the urban setting, where human-to-human transmission oc-
VEEV infection is typically associated with a leukopenia and curs through domestic and peridomestic mosquito vectors.
elevated serum aspartate amino transferase levels (135). When The main domestic vector, Aedes aegypti has been responsible
lumbar punctures are performed, the CSF reveals a lymphocyte for the major outbreaks of CHIKV in Comoros, India, Kenya,

Scheld_Ch15.indd 221 2/21/14 5:38 PM


222 Part II: Viral Infections and Related Disorders

Seychelles, and Singapore during the early phases of the probable if the patient is living or has visited an endemic area
global emergence of CHIKV during 2004 to 2011 (151,152). within 15 days before onset of symptoms. The case is con-
However, during the later phases (2006 onward), genetic ad- firmed when the patient test positive for one of the diagnos-
aptation of CHIKV to a new vector, A. albopictus led to major tic laboratory tests for CHIKV (163). Diagnosis of CHIKV is
outbreaks in several Indian Ocean islands (Reunion Island, made during the acute phase of infection by detection of viral
Sri Lanka), India, and Southeast Asian countries (Singapore, RNA in serum samples by RT-PCR and by virus isolation. Viral
Malaysia and Thailand) (153,154). The Indian outbreak re- RNA can be detected in samples obtained from 1 day before
sulted in an estimated 1.4 million cases during 2006 to 2009, symptom onset to day 7. Real-time RT-PCR assays capable of
whereas an estimated 266,000 cases (258 deaths) in the detecting a region of the nonstructural protein (nsp1) gene or
Reunion Island were reported, with an overall attack rate of the envelope (E) gene are also used for diagnosis (173,174).
34% during 2005 to 2006 (152,155). Later studies confirmed For later samples, indirect immunofluorescence and ELISA
that the epidemics in the Indian Ocean islands and in India can be used to rapidly and sensitively distinguish between IgG
were associated with an amino acid substitution in the E1 en- and IgM antibodies. CHIKV antigen can be detected in serum
velop glycoprotein (E1-A226V) in CHIKV, which allowed the and CSF samples obtained 2 days after onset of symptoms by
virus to adapt for dissemination by A. albopictus. A further antigen capture ELISA. Several commercial serologic assays as
mutation (E2-L210Q) has been described in the 2009 Indian well as in-house ELISA techniques have been used for CHIKV
outbreak, which again resulted in an increase in the dissemi- diagnosis and have been shown to accurately distinguish
nation of CHIKV in A. albopictus (156). Autochthonous CHIKV from other alphaviruses (except onyong-nyong virus)
transmission of CHIKV has been reported in Italy (2007) with and flaviviruses (175177).
over 250 cases and in France (2010), signaling the emergence
of CHIKV in Europe for the first time (157,158). Imported Treatment and Prevention
cases of CHIKV infection have now been reported in several
European countries, Australia, and the Americas (159161). Nonsteroidal antiinflammatory drugs are the only recom-
mended treatment for arthralgia caused by CHIKV infection.
Clinical Features Ribavirin, chloroquine, and passive transfer of antibodies
are other potential treatments being investigated (163,178).
The clinical features of CHIKV infection have some similari-
Vector control remains the main method of CHIKV prevention
ties with dengue infection (162). Asymptomatic CHIKV infec-
(91,179). Several vaccine candidates are under development.
tions are rare. After an incubation period that can range from
These include inactivated, whole-virus, DNA-based, viruslike
1 to 12 (average of 2 to 4) days, a sudden onset of high fever
particle, and adenovirus-vectored vaccine approaches as well
followed by severe arthralgia and a skin rash develops. Almost
as a chimeric alphavirus vaccine (105,180183).
all CHIKV patients have symmetrical polyarthralgia. The most
affected joints are the fingers, wrists, ankles, elbows, toes, and
knees. The fever can reach as high as 40C and may last up
to 10 days. A transient maculopapular and erythematous rash FLAVIVIRUSES
on the face, limbs, and torso is common and may last for 2 to
3 days. Other symptoms include myalgia, headaches, photo- Infectious Agent
phobia, lumbar back pain, chills, weakness, malaise, nausea,
and vomiting (151,163). The acute phase of CHIKV infection There are three important genera in the family Flaviviridae: the
usually resolves within 1 to 2 weeks, but the arthralgia may genus Hepacivirus, which includes hepatitis C virus, the genus
persist for many months or years in some patients (164,165). Pestivirus, which includes bovine viral diarrhea viruses, and
Age and underlying joint disorders have been shown to in- the genus Flavivirus (Table 15.1). This genus includes many
crease the likelihood of developing persistent arthralgia. The important causes of arboviral encephalitis, as well as arbo-
case-fatality rate for CHIKV is about 0.1%, with most deaths viral fever-arthralgia-rash and hemorrhagic fever syndromes,
occurring in the elderly, neonates, and adults with an underly- such as dengue viruses and yellow fever virus (Fig. 15.1). The
ing disorder. Heart failure, multiple organ failure, hepatitis, genus and family are named after the Latin word for yellow
and encephalitis are the most common causes of death (163). (flavus). Flaviviruses are thought to have evolved from a com-
CHIKV has not been generally considered to be a neuro- mon ancestor as recently as 10,000 years ago and are rapidly
tropic virus. However, reports from recent outbreaks have given evolving to fill new ecologic niches (184,185). They consist of
strong evidence of neurologic involvement of CHIKV. The a single strand of positive-sense RNA, wrapped in a nucleo-
main neurologic manifestations of adults infected with CHIKV capsid and surrounded by a glycoprotein-containing envelope.
are encephalopathy, acute flaccid paralysis, and Guillain-Barr The RNA comprises a short 58 untranslated region (UTR), a
syndrome (166169). A recent study has reported that about longer 38 UTR, and between them a single open reading frame
25% of adults admitted with atypical CHIKV infection during (186). This codes for a single polyprotein, which is cotransla-
the 2005 to 2006 Reunion Island had neurologic involvement tionally and posttranslationally cleaved by viral and host pro-
(mainly encephalitis, malaise, and meningoencephalitis) (170). teases into three structural proteins (core, or C, premembrane,
Neurologic manifestations were more common in children. or PrM, and envelope, or E), and seven nonstructural (NS)
The most common manifestations were encephalitis, febrile proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5).
seizures, and acute encephalopathies (171). For the first time, The C protein is highly basic and combines with the RNA
vertical transmission of CHIKV, from mother-to-child was to form the nucleocapsid. The PrM is closely associated with
reported during the Reunion Island outbreak. Encephalopathy the E protein, forming a heterodimer, and is thought to act as
was the most common neurologic manifestation among new- a chaperone to it, impairing its function until after virion
borns infected by vertical transmission (172). release. Immediately before virion release, the PrM protein is
cleaved by a furin-like protease to its mature M protein form.
Diagnosis
This allows the formation of E protein homodimers, which
A possible CHIKV case is suspected when a patient is pre- are thus activated (187). The E protein is the largest struc-
sented with an acute onset of high fever and severe arthralgia tural protein, consisting of nearly 500 amino acids with up to
unexplained by other medical conditions. The case becomes two potential glycosylation sites. It is the major target for the

Scheld_Ch15.indd 222 2/21/14 5:38 PM


Chapter 15: Arthropod-Borne Viral Encephalitides 223

humoral immune response and is thought to be important in the risk of febrile disease and of neurologic disease increased
viral entry into host cells. Studies with monoclonal antibodies with age, which may in part explain the differences compared
and more recently with x-ray crystallography have determined to parts of Africa. In Egypt, most of the population is infected
the composition of the E proteins three domains (188,189). during childhood, and neurologic disease is rare (205). In a
Domain III is the putative receptor-binding domain (by which South African outbreak of 18,000 people of all ages, only a
virions attach to the yet-to-be-identified host cell receptor), do- single encephalitis case was reported (192).
main II is the dimerization domain, and domain I has a central Recent molecular phylogenetic studies show that isolates of
beta barrel and is the hinge domain that links the other two WNV can be divided into seven lineages. Lineage 1 has been
domains. Following viral attachment to the cell surface, fla- subdivided into a further three clades: 1a, 1b, and 1c. Lineage
viviruses enter cells by endocytosis. Subsequent fusion of the 1b has spread to Australasia as Kunjin virus, whereas lineages
virus lipid membrane with the endosome membrane allows 1a and 5 are found in India, and lineage 6 in Malaysia (206).
viral RNA to penetrate into the cytoplasm of the infected cell Lineage 2 strains have mostly been found in Africa, whereas
(186). Recent cryoelectron microscopic studies have shown an lineage 1 strains are more widely distributed and are responsi-
arrangement of 90 E-protein dimers lying flat on the surface ble for all the large outbreaks during 1998 to 1999 (207,208).
of the virion, which rearrange to form E homodimers as the This has led to the suggestion that they may be more virulent,
pH level decreases, exposing an internal fusion peptide and a although neuroinvasive strains have been shown in both lin-
patch of viral membrane for fusion. Interestingly, recent stud- eages in animal models (209). However, lineage 2 was more
ies have shown the E1 protein of alphaviruses has a striking prevalent during the recent outbreaks in Europe (Greece,
similarity to the flavivirus E protein, in terms of structure and Romania, and Italy) and therefore may disperse wider in the
function (96). Together, they have been labeled class II fusion future (210,211).
peptides (190). In nature, WNV is transmitted in an enzootic cycle between
birds by mosquitoes. WNV infects over 300 different bird
species and 50 mosquito species (1). Members of the order
West Nile Virus Passeriformes (jays, blackbirds, finches, warblers, sparrows,
and crows) appear to be important in maintenance of the virus
Epidemiology in nature (because of viremias); the family Corvidae (crows,
blue jays) seems to be particularly susceptible Culex mosqui-
WNV was first isolated from the blood of a febrile woman
tos, particularly Culex pipiens, appear to be important in the
in the West Nile region of northern Uganda in 1937 (191).
enzootic cycle, although different species may act as bridging
It was soon shown to be transmitted between vertebrate hosts
vectors transmitting the virus to humans.
(especially birds) by mosquitoes. Sporadic cases and larger
How WNV is introduced to new areas is not completely
outbreaks of febrile disease (West Nile fever) were reported in
understood. Migratory birds are thought to be important for
Africa, the Middle East, and Asia (192). Although meningeal
the movement of WNV from Africa into southern Europe.
irritation was noted in some patients with West Nile fever, the
They may have been involved in the virus introduction into
first cases of encephalitis due to WNV occurred when the virus
the United States, although imported exotic birds, a viremic
was given as an experimental (and unsuccessful) treatment for
human, or inadvertently transported mosquitoes seem more
advanced cancer in 1951. The first naturally occurring West
likely (212). Molecular genetic evidence suggests there was a
Nile encephalitis cases were in the elderly residents of a nurs-
single introduction into the United States of a strain closely
ing home in Israel (193). Outbreaks of equine and human
related to one isolated from a goose in Israel (207). A com-
meningoencephalitis occurred in southern France during the
plex interplay of viral, avian, mosquito, human, and cli-
1960s, and a subtype of WNV (Kunjin virus) was isolated
matic factors may contribute to the large outbreaks that have
in Australasia. Since the 1990s, the clinical epidemiology of
characterized the disease recently (Fig. 15.4). During the 2002
WNV appears to have changed with increasing frequency and
outbreak in the United States, transmission also occurred via
severity of outbreaks, including urban disease. These included
transplanted organs, infected blood products, and possibly
an outbreak with nearly 400 confirmed cases in Romania
breast milk (213).
in 1996, nearly 200 cases in the Volgograd region of Russia in
1999 and more than 200 cases in Israel in 2000 (194197). In
Clinical Features
1999, the virus appeared in North America for the first time,
with 62 confirmed cases (198,199). It spread across the conti- After an incubation period, which is typically 2 to 6 days but
nent during 2000 and 2001, causing a largest encephalitis out- may extend to 14 days, patients with West Nile fever develop
break in 2002, with nearly 3,000 cases and 250 deaths (75). a sudden onset of an acute nonspecific flulike illness, char-
WNV is now the leading cause of domestically acquired arbo- acterized by high fever with chills, malaise, headache, back-
viral disease in the United States of America and has spread ache, arthralgia, myalgia, and retroorbital pain (86). Other
throughout Canada, the Caribbean, Colombia, and Mexico nonspecific features include anorexia, nausea, vomiting, diar-
(200,201). In 2010, WNV outbreaks occurred in Greece with rhea, cough, and sore throat. In epidemics of West Nile fever,
262 confirmed human cases with 197 cases of neuroinvasive a flushed face, conjunctival injection, and generalized lymph-
WNV and in Romania with 57 cases, out of which 54 was neu- adenopathy were common. A maculopapular or pale roseolar
roinvasive (202,203). Another outbreak occurred in Greece in rash was reported in about 50% of patients and was more
2011 with 31 cases of neuroinvasive WNV (204). common in children. In one outbreak, 20% of patients with
Most human infections with WNV are asymptomatic. In West Nile fever had hepatomegaly and 10% had splenomegaly
the 1998 to 1999 outbreaks, approximately 1 in 5 infections (214). Myocarditis, pancreatitis, and hepatitis occur occasion-
resulted in West Nile fever and only about 1 in 150 developed ally in WNV infection.
CNS disease (194,197). Over 75% of patients infected with Patients with neurologic disease due to WNV typically
WNV had neuroinvasive disease (encephalitis, meningitis, or have a febrile prodrome lasting 1 to 7 days that may be bi-
acute flaccid paralysis) during the recent outbreak in Greece phasic, before developing neurologic symptoms. Although in
and over 94% during the Romanian outbreak (202,203). In most cases, the prodrome is nonspecific, 15% to 20% of pa-
contrast, the proportions were much lower in earlier outbreaks tients may have features suggestive of West Nile fever, includ-
in Africa (192,205). In outbreaks across Europe and America, ing eye pain, facial congestion, or a rash, though less than 5%

Scheld_Ch15.indd 223 2/21/14 5:38 PM


224 Part II: Viral Infections and Related Disorders

Behavioral factors Human immune response

Exposure to biting Impaired (e.g., immunocompromised


mosquitoes patients) allows virus to replicate
Avian hosts ? genetic susceptibility
Compromised blood brain barrier
Large population of
immunologically nave ? Allows virus to enter brain
susceptible hosts
e.g., cerebrovascular disease

Age

? impaired immunity
? coexisting disease
Enzootic vectors

Feed naturally on birds Bridging vectors


Breed in water with high Strain virulence determinants
Feed on birds and humans
organic content (ditches,
swamps) May be different to enzootic ? affect rate of viral replication
vectors ? some strains neuroinvasive
e.g., Culex pipiens and
others e.g., Culex salinarius and others

Climate

Increased temperature enhances virus replication in mosquitoes


Lack of rainfall increases organic content of water collections, and
enhances mosquito replication

FIGURE 15.4 Factors known or postulated to be involved in the enzootic cycle of West Nile virus, and
epidemics of human disease.

had lymphadenopathy (215). In the 1998 to 1999 outbreaks, as hypertension and diabetes mellitus (199,218). Neurologic
approximately two thirds of hospitalized patients had enceph- sequelae are common among survivors. In one study, half of
alitis (with or without signs of meningeal irritation), and one patients hospitalized still had a deficit at discharge and only
third had meningitis (194,196,199). Severe generalized muscle 33% recovered fully at 1 year (219).
weakness was a common feature in the 1999 New York out-
break and subsequent outbreaks in the United States (199). Diagnosis
In some patients, this affects only the limbs, but in others,
respiratory and bulbar musculature are affected, requiring Approximately 50% of patients have a peripheral leukocy-
ventilation. Though initially ascribed to Guillain-Barr syn- tosis, and 15% have leukopenia (199,219). Hyponatremia
drome, the weakness was probably due to anterior horn cell sometimes occurs in encephalitic patients. Examination of
damage (myelitis), as is seen in other flavivirus infections (86). the CSF typically shows a moderate lymphocytic pleocytosis,
Conscious patients with a polio-like flaccid paralysis due to although sometimes no cells or neutrophils may predominate.
WNV were recognized in the 1970s and 2002 (70,216). In the The protein is moderately elevated and the glucose ratio is
recent outbreaks in Greece, 85% of patients had encephalitis/ typically normal. CT scans of the brain are usually normal.
meningoencephalitis and 12% had meningitis, followed by MRI may show nonspecific enhancement of the meninges or
3% with acute flaccid paralysis (202). periventricular areas (199,220) or high signal intensities on
Although seizures occurred in approximately 30% of pa- T2-weighted images in the thalamus and other basal ganglia
tients in the early descriptions of West Nile encephalitis, they (32). Nerve conduction studies typically show reduced motor
did not appear to be an important feature in the Romania or axonal amplitudes consistent with anterior horn cell damage,
the New York outbreak (199). Other neurologic features in- although there may also be some slowing of conduction velo-
clude cranial neuropathies, optic neuritis, and ataxia. Stiffness, cities and some changes to sensory nerves (86,216). The dif-
rigidity, spasms, and tremors associated with basal ganglia ferential diagnosis is broad and includes other causes of viral
damage similar to that seen in JE (217) have also recently been encephalitis (Table 15.3) and other causes of fever and reduced
recognized in West Nile encephalitis (32). consciousness (Table 15.4).
Overall case-fatality rates for patients hospitalized with Attempts at WNV isolation from serum or CSF are usu-
West Nile encephalitis outbreaks ranged from 4% to 14% but ally unsuccessful, because viremias are low, and the virus
were higher in older patients (196,199). However, the overall has cleared by the time most patients present (Fig. 15.3).
case-fatality rate during the recent Greece outbreak was much Techniques include detection of viral antigen by ELISA or of
higher at 17%, increasing to nearly 35% for those older than viral nucleic acid using reverse transcriptase PCR (RT-PCR)
80 years of age (202). Other risk factors for death include the or real-time PCR (Taq-Man). Real-time PCR, the most sensi-
presence of profound weakness, deep coma, failure to produce tive of these techniques, detects only 55% of patients (221).
IgM antibody, impaired immunity, and coexisting illness such Newer real-time RT-PCR assays, including multiplex RT-PCR

Scheld_Ch15.indd 224 2/21/14 5:38 PM


Chapter 15: Arthropod-Borne Viral Encephalitides 225

assays are more sensitive and offer rapid detection of WNV recently isolated strains from C. tritaeniorhynchus in China
(81,210,222). The accepted standard for diagnosing WNV and from Culex bitaeniorhynchus in Republic of Korea have
infection is the detection of IgM-specific antibodies in CSF been identified as genotype V, suggesting that this genotype
and/or serum using IgM capture ELISAs (29). Whereas an- may be reemerging in Asia (237,238).
tibody is detected in the serum of those with West Nile fever
or even asymptomatic infection, IgM in the CSF is specific for
CNS infection. Half of patients have antibody on admission, Clinical Features
and almost all have antibody by the seventh day of admission.
Those who do not are more likely to have virus isolated and After a 4- to 14-day incubation period, most patients develop
are more likely to die (32,223). a brief nonspecific febrile illness and then neurologic disease,
manifesting as a reduction in the level of consciousness and
Treatment and Prevention seizures. Seizures occur in approximately 85% of children
and 10% of adults with JEV (68,239). Multiple seizures and
There is no established antiviral treatment for West Nile en- status epilepticus are associated with a poor outcome. Subtle
cephalitis, although IFN-, ribavirin, and human immune motor status epilepticus, in which the only clinical manifesta-
globulin have been used in some patients (196). Treatment for tion might be the twitching of a finger or eyebrow, is important
patients with West Nile encephalitis is supportive. Mosquito in JEV (68). Multiple uncontrolled seizures may be associated
repellents are an identified prophylaxis being particularly with increased ICP and clinical signs of brainstem herniation
important for those in at-risk groups, such as the elderly syndromes (68). Movement disorders are common in JEV, both
and the immunocompromised. There is no human vaccine for in the acute stages of infection and as part of the spectrum of
WNV, although a crude formalin-inactivated vaccine and a neuropsychiatric sequelae. One quarter of patients showed acute
recombinant plasmidic DNA vaccine has been developed for manifestations in one series (68). A characteristic parkinsonian
horses (224,225). Several new vaccine candidates for human syndrome includes masklike facies, tremors, and cogwheel rigid-
are being developed and evaluated with some vaccines in ity. Other movement disorders include generalized rigidity, jaw
phase I/II human trials (210,226,227). dystonias, opisthotonus, choreoathetosis, orofacial dyskinesias
(involuntary tongue protrusions), myoclonic jerks, and opsoclo-
nus myoclonus (68,217). These disorders are thought to be the
Japanese Encephalitis clinical correlate of the inflammation often seen in the basal gan-
gliaparticularly the thalamus and substantia nigraon MRI
Epidemiology and at autopsy (217,240). In some patients, intention tremors
and ataxia reflect cerebellar involvement. Other focal neurologic
Outbreaks of encephalitis have been described in Japan since
signs include cranial nerve palsies, upper motor neuron weak-
the 1870s (228). The virus was first isolated in 1935 and has
ness (in 30% to 50% of patients), and flaccid limb weakness,
been recognized across much of eastern and southern Asia
with reduced or absent reflexes, which is often associated with
since then. All of Southeast Asia, the western Pacific region,
respiratory or bulbar paralysis (241). The combination of upper
most of the Asian subcontinent, and much of southern China
and lower motor neuron damage can lead to bizarre mixtures
is now affected, and the virus recently reached Australia for
of clinical signs that can change hourly during the acute stages
the first time (229). An estimated 35,000 to 50,000 cases and
of infection. In addition to causing flaccid weakness in coma-
10,000 deaths occur annually, making JEV numerically the
tose patients with encephalitis, JEV can cause a poliomyelitis-
most important cause of epidemic viral encephalitis worldwide
like acute flaccid paralysis in fully conscious patients (69). Acute
(230). Approximately half the survivors have severe neuropsy-
retention of urine, due to an atonic bladder, may be an early clue
chiatric sequelae, imposing a large socioeconomic burden in
that paralysis is due to a flavivirus (69). Overall, 20% to 30%
the areas affected. JEV is transmitted naturally in an enzootic
of hospitalized patients die, and half the survivors have neuro-
cycle between birds, pigs, and other vertebrate hosts by mos-
psychiatric sequelae. Poor prognostic signs include a depressed
quitoes, especially Culex tritaeniorhynchus, Culex gelidus,
level of consciousness, decerebrate posturing, multiple seizures,
and other species that breed in rice paddies. Humans become
increased ICP, isolation of virus from the CSF, and low levels of
infected when they encroach upon this enzootic cycle. Because
JEV-specific IgM and IgG in CSF and serum (31,68,239,242).
the virus is so ubiquitous, in rural areas almost all humans be-
Other nonspecific indicators include elevated admission temper-
come infected during childhood, but only a small proportion,
ature, absent abdominal reflexes, hyponatremia, low serum iron
estimated at around 1 in 300 develop symptoms (230), which
level, and elevated CSF white blood cell counts and protein level
may range from a nonspecific febrile illness through to a se-
(68,126,239,243,244).
vere meningoencephalomyelitis. In endemic areas, most cases
of JEV occur in children because by the time they reach adult-
hood, almost all individuals have been exposed to the virus
Diagnosis
and will have developed neutralizing antibodies. However, A peripheral polymorphonuclear leukocytosis is common.
when immunologically naive adults are exposed to the virus, CSF examination reveals an elevated opening pressure in 50%
for example, when the virus moves to new geographical areas of patients and usually a lymphocytic pleocytosis with a nor-
or when travelers or service personnel visit Asia, they too may mal glucose ratio and slightly elevated protein. MRI may show
develop disease (231). In northern temperate parts of Asia, high signal intensity on T2-weighted scans in the thalamus,
JEV causes large summer epidemics, but in southern tropi- other basal ganglia, midbrain, brainstem, and sometimes an-
cal areas, disease is year round, with a peak in the summer terior spinal cord. EEGs may reveal seizure activity, including
months (232). There are at least four genotypes of JEV across periodic lateralized epileptiform discharges (PLEDs) as occurs
Asia; molecular phylogenetic studies suggest that the distribu- with HSV encephalitis. This is usually on a background of
tion of genotypes is best explained by the virus origin in the slow-wave activity. Nerve conduction studies typically show
Indonesia-Malaysia region and spread from here (233235). reduced motor amplitudes, consistent with damage to the
A recent molecular phylogenetic study has identified the Muar lower motor neurons in the anterior horns of the spinal cord
strain of JEV as the fifth genotype of JEV with approximately (69,241), as is seen at autopsy (240). JEV is confirmed by IgM
20% divergence from other JEV strains (236). Furthermore, and IgG capture ELISAs, revealing antibody in the serum and

Scheld_Ch15.indd 225 2/21/14 5:38 PM


226 Part II: Viral Infections and Related Disorders

the CSF (27,245,246). A robust and user- friendly rapid IgM in 1931 (264) and is transmitted in nature between passeri-
capture ELISA (JEV Chex) has been developed with similar form and columbiform birds by Culex mosquitoes. The area
sensitivity as conventional ELISAs (247). Virus isolation and affected by SLE extends from Canada through the United
PCR from serum are usually negative; they may be positive in States to Central and South America. Strains of virus circu-
the CSF of fatal cases and in postmortem brain tissue (31,248). lating in the United States are genetically distinct from those
in Central and South America, forming two major clades,
Treatment and Prevention within which there are subgroups. Strains in the western
United States, which are transmitted by C. tarsalis mosqui-
A number of antiviral compounds against JEV have shown toes, differ from those found in the east that are transmitted
promise in vitro or animal models, including nitric oxide, by C. pipiens/Culex quinquefasciatus and Culex nigripalpus.
ribavirin, and IFN- (249251). IFN- showed promise Eastern strains are associated with greater epidemic potential,
in open clinical trials (252), but in a randomized placebo- being more readily transmitted by mosquitoes, causing higher
controlled trial in Vietnamese children, it failed to make any viremias in sparrows (one of the major vertebrate hosts)
difference to the outcome (82). Treatment of JEV focuses on and a higher case-fatality rate in humans (265). However,
controlling the complications of infection such as seizures between epidemics in the east, there may be long intervals
and ICP. High-dose dexamethasone failed to show any ben- during which no cases are seen. In contrast, in the western
efit (89). Recent studies on mice have shown protective ef- United States, transmission is endemic and sporadic cases are
fects of pentoxifylline and minocycline against JEV challenge seen every year, but the case-fatality rates are lower. The in-
(253255). cidence rates are highest in the Ohio and Mississippi River
Given that the vectors of JEV breed in rice paddies, vector basins and the Gulf Coast. The annual reported incidence has
control may seem an impossible task, but measures that may fluctuated from 0.003 to 0.752 per 100,000, with a median
have an impact include intermittent irrigation of the rice pad- of 35 cases reported each year (265). However, larger num-
dies and use of natural larvicides, such as larvivorous fish and bers have occurred in some years. In 1975, the largest out-
the plant neem (Azadirachta indica) (256). Personal protection break recorded caused 2,800 cases in 31 states, and in 1990,
with insect repellents and by using mosquito netting is also there were 222 cases during an epidemic in Florida. Serologic
recommended. However, the most effective prevention is with surveys suggest that on average approximately 1 in 300 in-
vaccines. A mouse-brainderived formalin-inactivated vac- fections is symptomatic. This rises with age, from 1 in 800
cine (JE-Vax) has been available for many years, produced by among children younger than 10 years to 1 in 85 for adults.
the Japanese under the Biken label and by the Korean Green Human immunodeficiency viruspositive individuals appear
Cross. Efficacy was demonstrated in a double-blind trial in to be at greater risk of acquiring SLE (266). More recently, an
Thailand (257), and it is used by travelers and service per- outbreak of SLE occurred in Argentina with 47 laboratory-
sonnel and in some of the wealthier Asian countries (231). confirmed cases and nine deaths (267,268).
Earlier crude preparations of the vaccine were associated with
adverse events, and in the early 1990s, there were reports of Clinical Features
hypersensitivity reactions, although the risk of serious neuro-
logic adverse events is low, at about 1 in 1 million (258). JE- After an incubation period, which is usually around 4 days
Vax is no longer manufactured and all existing stocks expired but may extend to 21 days, patients present with fever, head-
in May 2011 (259). A live attenuated vaccine (SA14-14-2), ache, malaise, myalgias, and other nonspecific features such as
developed by passage through primary hamster kidney cells, diarrhea, nausea, vomiting, cough, and sore throat. Dysuria,
has been used in China since the 1980s and was shown to urgency, and incontinence are early features in some patients.
be safe and efficacious (260262). It is also relatively cheap In patients younger than 20 years, about 55% have encephali-
and has been licensed to use in Korea, Nepal, and Sri Lanka. tis, and 40% have meningitis, but in those older than 60 years,
However, regulatory approval for its wider use has been de- more than 90% have encephalitis (269). The most common
layed because of concerns about its production, in particular manifestation of encephalitis is a reduced level of conscious-
whether it meets international good manufacturing practice. ness, which may range from mild confusion to deep coma.
A new inactivated Vero cell culturederived vaccine (IC51 or Tremulousness of the eyelids, tongue, lips, and extremities
IXIARO) using the attenuated SA14-14-2 strain for adults is is one of the most common neurologic signs. More marked
now approved for use in the United States, Europe, Canada, involuntary movements such as myoclonus, nystagmus, and
Australia, Hong Kong, and Switzerland. Clinical trials are on- opsoclonus are less common. About 25% of patients have
going to determine the safety and efficacy of this vaccine for cranial nerve palsies, usually unilateral. Cerebellar ataxias are
use in children (259). Newer vaccines in clinical trials include also common. Seizures occur in children and in more severe
a chimeric vaccine (JE-CV or IMOJEV) in which the prM and adult cases and are a poor prognostic sign (270). Subtle motor
E genes of JEVSA14-14-2 strain are inserted into the genome seizures have also been reported (271). About 17% of patients
of the yellow fever 17D backbone containing the yellow die (269), and neuropsychiatric sequelae including emotional
fever nonstructural gene, which has been used safely for many disturbances, forgetfulness, tremor, unsteadiness, and visual
years (226,263). disturbances occur in about 33% of patients.
Diagnosis
St. Louis Encephalitis The peripheral white blood cell count is normal or slightly
elevated. In some patients, microscopic hematuria, protein-
uria, and pyuria have been reported. Hyponatremia due to
Epidemiology
SIADH secretion occurs in more than one third of patients,
Before West Nile encephalitis in New York in 1999, St. Louis and alanine aminotransferase and creatinine phosphokinase
encephalitis (SLE) was the most important flavivirus in North levels may be slightly elevated. One third of patients have an
America. Outbreaks compatible with SLE were described elevated CSF opening pressure, and there is typically a moder-
from the 1920s, but the disease was first recognized in Paris, ate mononuclear pleocytosis, with an elevated protein level.
Illinois and then St. Louis and Kansas City, Missouri in the EEGs show diffuse slowing and occasional spike-and-wave
1930s. The virus was isolated from the brain of a fatal case activity, including PLEDs. The MRI may show high signal

Scheld_Ch15.indd 226 2/21/14 5:38 PM


Chapter 15: Arthropod-Borne Viral Encephalitides 227

intensity in the substantia nigra (269). The diagnosis is usu-


ally made by IgM capture ELISA or immunofluorescence, with Tick-Borne Encephalitis
nearly 100% of CSF samples being positive by day 7 of illness
(272). Antibody can also be measured in the serum, but there Epidemiology
may be problems with cross-reactive antibody in patients pre- Descriptions of a disease compatible with TBE appeared from
viously exposed to other flaviviruses, such as dengue. Virus the 1930s, and the virus was first isolated by Russian scientists
can sometimes be cultured from the brain tissue of fatal cases in the Far East in 1937 (280). Three closely related subtypes
or occasionally demonstrated by electron microscopy. Viral of TBE virus exist, whose names reflect the geographical areas
antigen detection and PCR results may be positive but are not they principally affect: European/Western, Siberian, and the
reliable for routine diagnosis. Far East. However, across this vast geographical area, the dis-
ease was given a range of different names (central European
Treatment and Prevention encephalitis, Russian spring/summer encephalitis, Far East
There is no antiviral treatment; thus, care is supportive. There Russian encephalitis, biphasic milk fever, Taiga encephalitis,
is no vaccine. Measures to reduce the risk of being bitten by an Kumlinge disease, Fruhsommer meningoenzephalitis) before it
infected mosquito are described earlier in this chapter. was realized that they are essentially the same disease. The TBE
group serocomplex (recently renamed the mammalian group
of tick-borne flaviviruses) also includes Powassan virus (a rare
cause of encephalitis in Canada); louping ill virus (a very rare
Murray Valley Encephalitis cause of CNS disease in the British Isles and Scandinavia);
and viruses that cause hemorrhagic disease, such as Kyasanur
Epidemiology Forest disease virus (in India) and Omsk hemorrhagic fever
Outbreaks of encephalitis in Australia in 1917 and 1922, la- virus (in Siberia) (Fig. 15.1). TBE virus is naturally transmit-
beled Australian X disease, or an aberrant form of polio- ted between small rodents by hard Ixodes ticks (particularly
myelitis (273), have subsequently been attributed to MVEV, Ixodes ricinus and Ixodes persulcatus). Because ticks may live
which was first isolated from the brain of a fatal case in 1951 for months or even years, and because they can pass the virus
(274). The virus has been found across parts of Australia and to their offspring transovarially and transtadially, they act as
New Guinea and is transmitted between herons, egrets, and important reservoirs. Following the bite of an infected tick,
other aquatic birds, by Culex annulirostris, and other mos- small mammals become viremic, and subsequent biting ticks
quitoes. The overwintering mechanism and factors responsible then become infected. In addition to this classic arboviral
for the intermittent epidemics are not known. MVEV is en- transmission cycle, virus may also be transmitted from tick
zootic in the Kimberley region of Western Australia and the to tick via infected reticuloendothelial and inflammatory cells
Northern Territory and is thought to only reach southeast- (Langerhans cells, neutrophils, monocytes, and macrophages)
ern Australia during extreme weather conditions (275). Only in the skin of the vertebrate host without the need for the ani-
about 1 in 1,000 to 1 in 2,000 infections result in disease, mal to become viremic (8). Adult ticks tend to feed on larger
and the number of cases is small, 25 between 1990 and 1998. mammals and thus infect humans walking through dense for-
Most cases are in aboriginal children living in areas where they est between spring and autumn. Across the large geographical
are exposed to the virus, but cases have also occurred in travel- area where TBE occurs, the virus tends to circulate in small
ers to these areas (276). More recently, the number of MVEV natural foci where the vegetation, temperature, moisture,
cases increased (16 cases and three deaths) in endemic areas and presence of appropriate mammalian hosts supports
of Australia and reemerged in southeastern Australia. This their existence. Humans can also become infected by drink-
outbreak in 2011 was followed after heavy rainfall and flood- ing the unpasteurized milk or cheese of infected goats, cows,
ing resulting in increased numbers of C. annulirostris mosqui- and sheep, causing biphasic milk fever. There has been a
toes and widespread seroconversion among sentinel chicken dramatic increase in the number of cases of TBE in humans
flocks (277). following drinking TBEV-infected milk in Russia and Europe
(281). The incidence of TBE varies according to location and
year (282). TBE is endemic in 27 European countries. The
Clinical Features, Diagnosis, and Treatment endemic area spans from central and Eastern Europe to Siberia
After a brief nonspecific febrile illness, milder cases develop and some parts of Asia, including China and Japan. Nearly
meningism and tremulousness, whereas in more severe cases, 170,000 clinical cases of TBE have been reported in Europe
there are seizures, flaccid paralysis requiring mechanical ven- and Russia between 1990 and 1999. In Europe, there have
tilation, and coma (276). Infants may deteriorate rapidly and been on average about 3,000 cases annually in the last 5 years
die within 24 hours. Approximately one fourth of hospital- (281). Following the collapse of the former Soviet Union and
ized cases die and one fourth have severe sequelae. Typically, reduced use of pesticides and vaccine against TBE, the annual
there is a CSF pleocytosis, with mildly elevated protein and incidence rose to more than 10,000 cases (3). Serologic sur-
normal glucose levels. MRI may show high signal intensities in veys indicate that in endemic regions, 70% to 95% of infec-
the basal ganglia (278). Virus has been isolated by inoculating tions are subclinical.
postmortem brain tissue into chick embryos, but the diagnosis
is usually made serologically with an ELISA, hemagglutina-
tion inhibition, complement fixation, or neutralization test.
Clinical Features
Treatment is supportive, because there is no antiviral drug. Three fourths of patients recall a tick bite, a median of 8 days
Patients with MVE have been treated with corticosteroids but (range, 4 to 28 days) before symptoms develop (283). A cou-
the effectiveness has not been assessed in controlled clinical ple days of headache, fatigue, and pain in the neck, shoulders,
studies (277). No vaccine is available, and the low incidence of and lower back precede the onset of high fever, nausea, and
disease makes it unlikely that one will be developed. However, vomiting. In three fourths of patients, the illness is biphasic,
recent studies in mice have shown that the live chimeric JEV with this febrile prodrome of several days, followed by an
vaccine offered cross protective immunity against lethal chal- afebrile period of about 8 days (range, 1 to 33 days) before
lenge with MVEV (279). neurologic disease develops (283,284). In the mildest form of

Scheld_Ch15.indd 227 2/21/14 5:38 PM


228 Part II: Viral Infections and Related Disorders

diseaselabeled the febrile form by Russian cliniciansthe FSME-Bulin label by Baxter in Austria, and the Encegam label
febrile phase lasts from 1 to 5 days, sometimes with severe by Chiron/Behringwerke in Germany. An intramuscular dose of
muscle pains and sometimes numbness and fasciculations, 0.2 mL/kg is recommended up to 4 days after the tick bite (288).
before resolving with no residual deficit (3). In the menin- Prophylaxis later than this is not recommended because of the
geal form of disease, there is neck stiffness and photopho- theoretical potential of antibody aggravating inflammation in
bia. The meningoencephalitic form is characterized by a patients with established neurologic disease. Postexposure pro-
reduced level of consciousness, which may range from drowsi- phylaxis is not recommended for children. The preparations
ness and hallucination to deep coma, focal neurologic signs, have also been used (at 0.4 mL/kg) as short-term preexposure
including hemiplegia, and seizures. There may also be cardiac prophylaxis for those about to visit areas at risk.
arrhythmias. A poliomyelitic form with poliomyelitis-like A formalin-inactivated vaccine based on an Austrian virus
flaccid paralysis is common in TBE virus infection. This usu- isolate (TBEV-Eu strain Neudoerfl) grown in chick embryo
ally affects the neck and upper limbs to cause pain, sometimes cells has been commercially available in Europe since 1976. It
with periodic muscle contractions and numbness, then upper had a relatively high rate of side effects (fever, headache, mal-
limb weakness with winging of the scapula, wristdrop, or a aise) but has been replaced by a highly purified form, prepared
hanged head due to neck extensor weakness. Muscle atro- by ultracentrifugation (289). This vaccine (FMSE-Immun,
phy begins after the second or third week and persists. In con- Baxter), which contains aluminum hydroxide as an adjuvant,
trast, some patients develop a polyradiculoneurotic form of has an efficacy of about 98%. Vaccination is recommended for
disease: neuropathy occurs 1 to 2 weeks after the initial febrile those living, traveling, or working in areas at risk, as well as
phase and is associated with a recurrence of fever, but there is laboratory workers. Mass vaccination since 1981 in Austria
usually complete recovery. Finally, in Russia, a chronic form resulted in a marked decline in the number of TBE cases (290).
of TBE has been described and is believed to be caused only Another formalin-inactivated vaccine, based on the TBEV-Eu
by the Siberian subtype of TBEV. In some patients, deteriora- strain K23 (Encepur, Novartis) manufactured in Germany
tion continues long after the acute disease, postmortem ex- is also used in Europe. This vaccine was initially withdrawn
amination suggests chronic inflammation, and viral RNA may from pediatric use because of allergic reactions related to poly-
be detected by nucleic acid hybridization. Other patients are gelin, which was removed in 2001 (291). Children are given
asymptomatic following the initial tick bite but present years half the adult dose to reduce the high fever that was observed
later with a progressive form of disease, with virus being iso- when given the full dose (292). Two other formalin-inactivated
lated at autopsy (65). Spontaneous regular contractions (myo- vaccines, TBE-Moscow vaccine (based on the Far-Eastern
clonic jerks) of the limbs are seen in about 25% of patients Sofjin strain) and EnceVir vaccine (based on the Far-Eastern
with all neurologic forms of TBE disease and may persist as 205 strain), are widely used in Russia and several Asian coun-
epilepsia partialis continua (Kozhevnikov epilepsy). tries. EnceVir is not currently recommended for pediatric use
Traditionally, disease caused by the Far East subtype of because of high rates of fever and allergic reactions observed
TBE virus is thought to be more severe than that caused by the in 2010 and 2011. These two vaccines are produced similar to
European subtype, with case-fatality rates of 20% to 60% com- the vaccines used in Europe except the TBE-Moscow vaccine,
pared with 1% to 3%, respectively. Differences in criteria for which is finally lyophilized. All four vaccines are produced
hospitalization may account for much of this apparent differ- according to the World Health Organization (WHO) manu-
ence, although differences in virulence have been found in animal facturing standards (293). Live attenuated vaccines against
models. The case-fatality rate of the Siberian subtype appears TBEV are currently under development (294). In 2011, the
to be similar to that of the European subtype, with neurologic WHO published its first position paper on TBE vaccines, giv-
sequelae reported for 30% to 60% of survivors. Interestingly, ing recommendation for TBE vaccination (295).
in biphasic milk fever following oral ingestion of infected milk,
TBE virus typically causes only the febrile form of disease.
Although there is sometimes mild meningism, encephalitis and Neurologic Dengue Disease
paralysis are not seen, and recovery is the rule (3).
Epidemiology
Diagnosis
Most patients affected by one of the four dengue viruses
During the initial febrile stage, leukopenia and thrombo- (dengue 1, 2, 3, and 4) present with a fever-arthralgia-rash
cytopenia are common (285), but by the time patients have syndrome or a viral hemorrhagic fever (296). The cause and
neurologic disease, many have a leukocytosis (283). CSF ex- even existence of neurologic manifestations of dengue has
amination usually reveals a mild pleocytosis with elevated pro- been a long-standing controversy (297302). However, more
tein level. EEGs of patients with encephalitis show slowing recent studies have given strong evidence to support neuro-
with or without focal activity (283). On MRI, abnormalities invasion of DENV (303306). Most authorities now accept
have been shown in about one fourth of patients with enceph- that dengue viruses can cause a nonspecific encephalopathy
alitis, usually in the thalamus but sometimes in the caudate, and occasionally encephalitis, even if the mechanism is not
cerebellum, and brainstem. The diagnosis is usually suspected yet clear (307,308). The four types of dengue virus were iso-
because of the history of tick bite, although other tick-borne lated from febrile patients in the 1940s, and dengue is unusual
febrile illnesses including borreliosis and ehrlichiosis should among arboviruses in that humans are the main natural host,
be considered. The diagnosis is confirmed by detection of although a sylvatic cycle exists in primates in Southeast Asia
IgM antibodies in CSF and/or serum using an ELISA (286), and West Africa (309). The virus is transmitted between hu-
although during the early viremic phase, virus may also be mans by A. aegypti and A. albopictus mosquitoes that breed
cultured from the blood. PCR assay have also been shown to in small collections of fresh water around the home (e.g.,
be useful in detecting TBEV in the first phase of illness (287). collected in rubbish, water containers, and tires). Dengue is
very widely distributed, being found in almost every country
Treatment and Prevention between the tropics of Capricorn and Cancer (310). According
There is no antiviral treatment, but an anti-TBE immune globu- to the WHO, over 50 million cases of dengue occur every year.
lin has been used for postexposure prophylaxis in those bitten by Three outbreaks of dengue occurred during 2001 and 2011
ticks in areas where the virus circulates. It is licensed under the in the United States, and the reemergence of autochthonous

Scheld_Ch15.indd 228 2/21/14 5:38 PM


Chapter 15: Arthropod-Borne Viral Encephalitides 229

dengue transmission within Europe has also been described (325). No vaccines are available yet, but several vaccine
(311,312). Neurologic disease has been reported from virtu- candidates are currently undergoing preclinical development
ally every country where dengue occurs (297). It is now rec- and some in clinical trials (326,327). Currently, the most ad-
ognized that in parts of Southeast Asia, dengue is responsible vanced dengue vaccine in phase III clinical trials is ChimeriVax,
for 1 in 20 patients admitted with a suspected CNS infection the chimeric tetravalent live vaccine (yellow fever 17D back-
(300) and as many as 1 in 5 of those with a clinical diagnosis bone) manufactured by Sanofi Pasteur (226). Several other
of encephalitis (299). chimeric and live attenuated vaccines are also in clinical trials
(326,328,329). There is no important enzootic cycle, and be-
Clinical Features cause the disease is spread by peridomestic mosquitoes, vector
control measures can have an impact. These include educating
Most patients with neurologic dengue present with a reduced
people to remove Aedes breeding sites, treating stored water
level of consciousness and other signs of severe dengue infec-
with larvicide (e.g., temephos), or the copepod Mesocyclops
tion, including the shock, vascular leakage, and hemorrhage
that feeds on A. aegypti larvae, covering water storage contain-
that characterize dengue hemorrhagic fever (DHF). They may
ers, and ultralow volume spraying of organophosphorus insec-
also have metabolic disturbances such as hyponatremia and
ticides during epidemics (330). In some settings, legislation and
acidosis, and in many patients, the encephalopathy is thought
fines for those who fail to remove Aedes breeding sites from
to be secondary to these complications. However, in other
the home have been effective. Personal protection with insect
patients with no rash, hemorrhage, or other signs of dengue
repellents is also recommended. The evidence for the efficacy of
fever or DHF, viral invasion across the BBB causing encephali-
these measures is variable. The only undoubtedly effective vec-
tis is thought to occur. In support of this is virus isolation, PCR
tor control measure was the near eradication of A. aegypti from
detection, and detection of anti-dengue IgM antibody in the
South America, using DDT, during the yellow fever campaign
CSF of some patients (300,313315). A recent study on 150
of the 1950s to 1970s. Since that campaign ended, Aedes has re-
CSF samples from fatal cases during a dengue epidemic pe-
infested South America (331). Worldwide, A. aegypti continues
riod in Brazil found evidence of DENV in 41 CSF samples out
to spread, and dengue is increasing as a global health problem
of 84 dengue-positive patients (306). Clinically, these patients
(332). In 2007, sequencing of the A. aegypti genome was com-
present with a brief febrile illness followed by a reduction in
pleted and has led to the development of transgenic mosquitoes
consciousness level, which may range from lethargy, drowsi-
that could be useful in preventing dengue transmission to hu-
ness, and irritability to deep coma. Seizures are also common,
mans (333). These include development of sterile mosquitoes,
particularly in young children (316,317). These may be sim-
interfering with mosquito feeding and reproduction and the use
ple febrile seizures or those associated with prolonged coma.
of Wolbachia to infect mosquitoes (9294).
Pyramidal or long tract signs also occur, but the extrapyra-
midal tremors and tone abnormalities that characterize other
arboviral encephalitides such as JEV and West Nile encepha-
litis (86) are less common in dengue, although they have been BUNYAVIRIDAE
described (318,319). Presentations consistent with acute dis-
seminated encephalomyelitis have also been described, some The Bunyaviridae family is the largest family of animal viruses,
time after a dengue infection (320). Meningismus occurs in with more than 350 members. It is named after Bunyamwera
up to 30% of patients, usually as part of an encephalopathic Village in Uganda where the prototype virus was isolated. The
illness. Uncomplicated viral meningitis due to dengue viruses family consists of five genera, the first two of which contain
is rare. Other neurologic manifestations include mononeu- important arboviral causes of CNS disease. Within each genus
ropathies, polyradiculopathies, Guillain-Barr syndrome, and of the family, viruses are placed into serogroups according to
acute neuromuscular weakness due to myositis and hypokale- their cross reactivity in serologic tests.
mia (303,307,321,322).
The Orthobunyavirus genus contains the mosquito-borne
LACV and other members of the California encephalitis
Diagnosis serogroup of viruses.
Patients with dengue often have leukopenia and thrombocy- The Phlebovirus genus includes the sandfly-borne TOSV,
topenia, as well as mildly elevated hepatitic transaminases. In which is emerging as an important cause of CNS disease
patients with DHF, an elevated hematocrit is found because of in southern Europe, and RVFV, which is transmitted by
fluid loss from blood vessels due to increased vascular perme- mosquitoes and causes febrile, hemorrhagic, and occasional
ability (301,323). Hyponatremia is common, particularly once CNS disease.
fluid resuscitation has begun. CSF examination reveals a mod- The Nairovirus genus includes Crimean-Congo hemor-
erate lymphocytic pleocytosis in up to 30% of patients with rhagic fever virus, which is transmitted by ticks.
neurologic disease. CT and MRI may show diffuse cerebral The Hantavirus genus, whose members are not arthropod
edema, although focal abnormalities have also been reported borne, includes viruses that cause hantavirus pulmonary
(300,313,319,324). Dengue can be confirmed by isolating the syndrome, and hemorrhagic fever with renal syndrome.
virus from serum or CSF, PCR detection, or demonstrating The Tospovirus genus contains only plant viruses transmitted
IgM antibodies. Because dengue is so common in many parts by thrips (woodworm), and no human pathogens.
of the world, and because IgM antibody may persist in the
blood for up to 3 months, confirmation of antibody or virus
in the CSF provides stronger evidence that dengue was the
cause of the neurologic symptoms (307). In addition, rigorous Infectious Agent
efforts must be made to rule out other causes.
All members of the family Bunyaviridae are 90 to 120 nm
in diameter and are spherical in shape, except hantaviruses,
Treatment and Prevention
which are either spherical or rod-shaped particles. These en-
No effective drugs against dengue are available. Because pa- veloped viruses have glycoprotein spikes assembled from the
tients with DHF are particularly likely to develop shock, fluid glycoproteins Gn and Gc, and enclose three circular nucleo-
management is critical, and detailed guidelines are available capsids, containing three linear segments of single-stranded

Scheld_Ch15.indd 229 2/21/14 5:38 PM


230 Part II: Viral Infections and Related Disorders

negative-sense RNA, designated L (large), M (medium), and thought to be an important amplifying host. In Europe and
S (small) (334,335). These range in size from 1 to 2.2 kb for Russia, the California encephalitis virus serogroup Tahyna
the S, 3.5 to 6 kb for the M, and 6.3 to 12.0 kb for the L. The virus is a cause of fever, respiratory symptoms, and occasion-
L segment codes for a single L protein, RNA-dependent RNA ally CNS disease (345,346).
polymerase (RdRp) protein. The M segment codes for surface Most encephalitis cases due to California serogroup viruses
glycoproteins G1 and G2, and for a nonstructural protein occur during the summer and early fall, particularly among
NSm. The S segment codes for nucleocapsid protein N (forms residents of the Mississippi and Ohio River basins (347); how-
the ribonucleoprotein [RNP] complex by encapsidating the ever, 31 states have now reported cases, with 130 cases (116
viral RNA replication products) and for a nonstructural pro- neuroinvasive cases) reported from 14 states in 2011. Nearly
tein NSs, although for hantaviruses, the nonstructural proteins 95% of patients were younger than 18 years of age (348).
NSm and NSs have not been detected (335,336). The recent spread of LACV to Tennessee, North Carolina,
Attachment of G1 and/or G2 glycoproteins to an as yet and West Virginia has been attributed to invasive mosqui-
unidentified cell receptor is thought to be the first step in viral toes including A. albopictus (349). The incidence of LACV
replication. However, some studies suggest that integrins are encephalitis is estimated at up to 30 per 100,000 in endemic
involved in hantavirus attachment, and phebovirus entry into areas (67). Most cases occur in children living close to wooded
dendritic cells involves DC-SIGN (337,338). Entry into the forests or visiting them for recreation; risk factors include the
cell by endocytosis is followed by uncoating and release of time spent outdoors and proximity of homes to tree holes
the three nucleocapsids into the cytoplasm, possibly mediated (343). Approximately 75 encephalitis cases are reported each
by pH-dependent fusion of the viral envelope and host endo- year. The ratio of asymptomatic to symptomatic infections is
somal membrane. A recent study suggests that after clathrin- about 1,000:1, and in endemic areas, 20% of the population
independent endocytosis, phleboviruses pass through early is seropositive by age 60 years. Jamestown Canyon virus is
and late endosomes, followed by late endosomal acidification distributed across the northern United States. Encephalitis
and release of RNPs and accumulation in the Golgi (339). due to Jamestown Canyon virus occurs mostly in the elderly.
Replication of viral RNA occurs via a positive-sense comple- Seroprevalence is 4% to 10% in some areas (350).
mentary RNA (cRNA) intermediate. Transcription of viral
RNA to messenger RNA (mRNA) is followed by translation Clinical Features
to produce viral proteins. For Phlebovirus and Tospovirus,
the SA segment is transcribed in both positive and negative Children with LACV encephalitis typically present with fever,
directions (ambisense). The G1 and G2 proteins are glycosyl- headache, malaise, nausea, and vomiting, which are often ac-
ated, and viral particles assemble by budding into the Golgi companied by meningeal signs (67,351353). Seizures, which
vesicles. These are transported to the cell surface, and fu- often include a focal component, occur in approximately 50%
sion of the cytoplasmic vesicles with the plasma membrane of children, one third of whom have status epilepticus. Half
results in virion release through exocytosis. Although this the patients have altered consciousness, and 10% are coma-
mechanism of viral release applies to most viruses in the fam- tose. Focal neurologic signs include hemiparesis, aphasia, dys-
ily Bunyaviridae, RVFV can also bud off directly at the cell arthria, chorea, and in about 10% clinical signs of raised ICP.
surface. Poor prognostic indicators include reduced plasma sodium
level, increased body temperature, and a Glasgow Coma Scale
score of less than 13 out of 15, and possibly increased number
of seizures. Although fatalities are rare, approximately 10%
La Crosse and Other California of patients have sequelae at hospital discharge. About 20% of
Serogroup Viruses those with seizures in the hospital develop epilepsy. Jamestown
Canyon virus causes a similar clinical pattern (354).
The California serogroup viruses are the only major cause of
CNS disease in the Bunyavirus.
Diagnosis
Epidemiology A peripheral leukocytosis is common, and although there is
usually a CSF lymphocytic pleocytosis up to 600 cells/mm3,
California encephalitis virus was isolated from mosquitoes an early lymphocytic pleocytosis may be normal. CSF pro-
by Reeves and Hammon in 1941 (340) and was shown to tein is elevated in about 30% of patients, but the glucose
be associated with three cases of encephalitis. Although the level is usually normal. Low sodium level due to SIADH se-
serogroup was named after this virus, human disease is rare cretion is reported in about 20% of patients. Diagnosis is
(341). LACV, which was isolated from the brain of a fatal en- usually serologic because attempts to isolate virus from CSF
cephalitis case in 1960 (342), is the most important member or blood are negative, although virus has been isolated from
of the group and was probably the most common cause of brain tissue.
arboviral encephalitis in the United States until the arrival of
WNV. Other members of the California serogroup include
Jamestown Canyon virus, which is also a relatively common
Treatment and Prevention
cause of CNS disease, and snowshoe hare virus, which is not There is no antiviral treatment for California serogroup vi-
(Table 15.2). California serogroup viruses are typically trans- ruses, or indeed any members of the Bunyavirus genus. A
mitted in nature between rodents by Aedes mosquitoes. LACV recent study investigated the safety and pharmacokinetics
is transmitted between chipmunks and squirrels by Aedes tri- of intravenous ribavirin treatment for children with LACV
seriatus, a forest-dwelling tree-hole breeding mosquito that encephalitis, but the results did not support the use of riba-
breeds in rainwater collected in artificial containers in urban virin for LACV encephalitis (355). Attention should focus on
settings. Evidence is accumulating that the recently introduced treating the complications of infection, particularly hypona-
mosquito species A. albopictus may be involved in the emer- tremia and seizures. There is no vaccine. Preventive measures
gence of LACV infection (343). During the winter months, include environmental measures to decrease breeding sites, for
the virus overwinters by vertical transmission into Aedes eggs example, removing tree holes, tires, and litter, which provide
(344). For Jamestown Canyon virus, the white-tailed deer is pools of water favored by Aedes mosquitoes for breeding.

Scheld_Ch15.indd 230 2/21/14 5:38 PM


Chapter 15: Arthropod-Borne Viral Encephalitides 231

Phleboviruses Causing Central Nervous Rift Valley Fever Virus


System Disease
RVF virus was first isolated in 1930, during an investigation of
Most members of this genus are transmitted by phlebotomine a large epizootic of disease causing abortion and death in sheep.
sandflies, although RVFV, which causes epidemics of febrile Originally found in the Rift Valley of Kenya, the virus is dis-
illness, and occasional hemorrhagic disease, retinitis, or neu- tributed across sub-Saharan Africa, Egypt, Saudi Arabia, and
rologic disease, across sub-Saharan Africa, is a notable excep- Yemen (332,372,373). Disease in animals follows the explosive
tion, being transmitted principally by Culex mosquitoes. increases in mosquito populations caused by heavy rains or new
irrigation projects. Humans become infected by mosquitoes or
contact with animal blood or other products. In humans, the
virus causes epidemics of febrile illness with hemorrhagic mani-
Sandfly Fever Viruses festations in about 1% to 2% (2). About 0.5% to 1.0% of pa-
tients develop retinal vasculitis or encephalitis as a late sequelae
A febrile illness associated with sandfly bites was recognized
1 to 4 weeks after the acute illness (374). Encephalitis mani-
since the early twentieth century. During outbreaks in World
fests as headache, meningismus, and clouding of consciousness,
War II, Albert Sabin isolated sandfly fever Sicilian virus and
sometimes with a recrudescence of fever. In fatal cases, focal
sandfly fever Naples virus from U.S. troops (356). Both vi-
necrosis is seen pathologically, which may be due to viral cell
ruses decreased or disappeared after the 1940s in countries
damage. However, the late presentation after viral clearance and
performing insecticide spraying for malaria eradication. More
development of antibodies suggest an immunopathologic pro-
recently, an outbreak occurred among Greek soldiers stationed
cess may be responsible, possibly due to a delayed hypersensi-
in Cyprus with 256 out of 581 soldiers infected with sandfly
tivity reaction (375). Because this virus is directly transmissible
fever Sicilian virus (357). In 1971, TOSV, a new virus antigeni-
to humans from the body fluids of livestock, abattoir workers
cally closely related to sandfly fever Naples virus, was isolated
in affected areas should wear protective clothing. There are no
in Italy from the sandfly Phlebotomus perniciosus (358,359).
human vaccines against phleboviruses, but vaccines are available
It has since emerged as the most important cause of pediatric
to protect livestock against RVFV and are the main countermea-
CNS infections in some parts of Italy, causing up to 80% of
sure against eliminating the source of human infection (376).
cases during the summer (360). Children from rural or semi-
urban areas are especially vulnerable. TOSV has also been rec-
ognized in other Mediterranean countries, including Cyprus,
France, Greece, Portugal, Spain, and Turkey (361,362). In af- COLTIVIRUSES
fected areas, seroprevalence levels of 10% to 25% have been
The genera of the family Reoviridae include the Rotavirus genus
found (361,363). The viruss natural cycle has not been fully
and three genera of arboviruses, Orbivirus, Coltivirus, and the re-
determined. In particular, although it has been shown that the
cently designated Seadornavirus. The orbiviruses include animal
virus replicates in sandflies and can be passed vertically to off-
pathogens such as bluetongue virus and African horse sickness
spring via transovarial and transtadial transmission, no natu-
virus, but no important human pathogens, and so are not consid-
ral vertebrate host has yet been identified. However, humans
ered further here. The most important coltivirus is CTFV, which
have sufficiently long and high viremias to transmit sandfly
because of similarities in its geographical distribution, vector and
viruses to biting sandflies, which contributed to some urban
symptoms were not distinguished from the rickettsial disease
outbreaks.
Rocky Mountain spotted fever until the 1930s. Its viral etiology
was demonstrated in the 1940s. Eyach virus is a coltivirus iso-
Clinical Features, Investigation, and Diagnosis lated from ticks in central Europe, where patients with CNS dis-
Aseptic meningitis is the most common presentation of TOSV, ease have been shown to have antibodies against the virus (377).
but encephalitis has also been described (363367). In one Banna virus was isolated from the CSF and blood of patients
series, all 14 patients had neck stiffness and 3 were encepha- with encephalitis in China (378,379). It had been classified as a
lopathic, including 1 with repeated seizures (360). Peripheral coltivirus, but following genome sequencing, it has been placed
white blood cell counts are usually normal, and the CSF shows with Kadipiro virus in the new genus, Seadornavirus (380,381).
a lymphocytic pleocytosis with normal glucose and mildly el-
evated protein levels. The outcome is generally good, with no
deaths or severe sequelae reported. Enterovirus infections are Infectious Agent
high on the differential for patients with aseptic meningitis in
the summer months, and a duplex PCR has been developed Like other Reoviridae, coltiviruses are nonenveloped particles
to distinguish between these infections (368). TOSV can be consisting of a double-capsid structure with icosahedral symmetry
isolated from the serum and CSF by inoculation into suckling containing double-stranded RNA. The 29-kb genomes are divided
mice or cell lines. Because humans have high viremias, virus into 12 segments of double-stranded RNA (ranging from 0.3 to 3.7
isolation and PCR from serum and CSF is often successful thousand base pairs), which can undergo resortment (382). Based
(369). IgM ELISAs have also been developed (368,370,371). on comparisons with other Reoviridae, each segment is thought to
encode the mRNA for a single gene product, which in the case of
VP1 is thought to be the RNA-dependent RNA polymerase.
Treatment and Prevention
There is no established antiviral treatment for TOSV infection
or other CNS Phlebovirus infections, although ribavirin may Epidemiology
be effective in RVF (250). Widespread use of DDT was effec-
tive in controlling sandfly fevers in the past, particularly re- CTFV is transmitted between ground squirrels, chipmunks, and
sidual spraying of internal walls in homes where sandflies rest. other small mammals by Dermacentor andersoni (wood ticks).
Although sandflies are small enough to penetrate mosquito net- The geographical distribution of cases is limited by the distri-
ting, impregnated bed nets are a deterrent, and the application bution of the vectors and host to mountainous areas between
of insecticides on the skin is recommended. 4,000 and 10,000 feet in the western United States and Canada

Scheld_Ch15.indd 231 2/21/14 5:38 PM


232 Part II: Viral Infections and Related Disorders

(383). CTFV or a closely related virus may be transmitted by


Dermacentor variabilis in parts of California. Larval, nymphal, Diagnosis
or adult ticks may be infected. The virus persists transtadially
between the maturing tick forms but is not passed transovari- Moderate leukopenia with a relative lymphocytosis and a
ally (Fig. 15.2). Ticks are infected for life, which may be up to left shift have been reported along with thrombocytope-
3 years. The vertebrate hosts usually have subclinical viremias nia, anemia, and elevated liver transaminase and creatinine
that can last for months or even longer in hibernating animals, phosphokinase levels (383). In the CSF, a moderate lym-
possibly providing one means for viral overwintering. Whereas phocytic pleocytosis (up to 500 cells/mm3) is described with
immature ticks feed on small mammals, adults feed on larger normal or slightly elevated protein and reduced glucose. The
mammals, deer, elk, and occasionally humans. Human cases of differential diagnosis includes other tick-borne conditions,
CTF occur in the summer months, when the number of ticks including Rocky Mountain spotted fever, ehrlichiosis, tulare-
and vertebrate hosts is increased, as is human exposure to the mia, and Lyme disease. The diagnosis of CTF is confirmed
ticks, because of recreational and occupational activities. by virus isolation from blood clots or red blood cells, direct
immunofluorescence of blood smears to detect antigen in
erythrocytes, or antibody detection using ELISAs or neutral-
ization assays.
Pathogenesis and Clinical Features
CTFV infects hematopoietic cells, resulting in persisting in-
fection of erythrocytes, leukopenia, and thrombocytopenia Treatment and Prevention
(384). The relative contributions of viral cytopathology and
the host immune response in the pancytopenia is not certain There is no established antiviral treatment, although ribavirin
(385). Because CTF is rarely fatal, there are few reports of the has activity in vitro and in the mouse model (388). Treatment
pathology, but hyaline membrane disease, endothelial swell- is symptomatic; aspirin and other nonsteroidal antiinflamma-
ing, and focal necrosis of the brain, liver, and spleen have been tory drugs should be avoided because of antiplatelet effects.
reported, similar to the changes seen in animal models (386). To avoid infection, standard methods to reduce the chances
IFN- levels in the plasma are high during the first 10 days of tick bites should be used, including wearing long-sleeved
of infection. After this neutralizing antibody, titers raise, but shirts and trousers, using repellents containing DEET, and fre-
despite this, viremias persist in nearly half the patients (383). quently inspecting for attached ticks.
Transplacental transmission has been recorded, but most
patients present with a history of exposure to ticks in an area
where the virus circulates, and many will recall a tick attach- ACKNOWLEDGMENTS
ment. Following 3 to 5 days of incubation (range, 1 to 14
days), there is fever, headache, malaise, and gastrointestinal Some of the work on flaviviruses described in this chapter was
disturbances. After 2 to 3 days, the fever may subside for a supported by the Wellcome Trust and the Medical Research
couple of days, before recurring, illustrating the biphasic or Council of Great Britain. T.S. is a professor of Neurology.
saddleback fever pattern (50% of patients with CTF). There A.P. is an NC3Rs David Sainsbury Fellow. This project has
is sometimes mild conjunctivitis, lymphadenopathy, and a faint been funded in whole or in part with Federal funds from the
maculopapular rash, or petechiae (383). Neurologic features National Institute of Allergy and Infectious Diseases, National
include neck stiffness, photophobia, and mildly reduced con- Institutes of Health, Department of Health and Human
sciousness. Rarely, there is encephalitis hepatitis, perimyocar- Services, under Contract (NO1-AI-65306, NO1-AI-15113,
ditis, and pneumonitis. Intravascular coagulopathy has been NO1-AI-62554, NO1-AI-30025), the General Research Unit
reported; only three deaths, all in children, are noted (387). (RR-032), and the State of Alabama.

References
1. Hollidge BS, Gonzalez-Scarano F, Soldan SS. Arboviral encephalitides: 12. Hollidge BS, Weiss SR, Soldan SS. The role of interferon antagonist, non-
transmission, emergence, and pathogenesis. J Neuroimmune Pharmacol. structural proteins in the pathogenesis and emergence of arboviruses.
2010;5:428442. Viruses. 2011;3:629658.
2. Solomon T. Viral haemorrhagic fevers. In: Cook G, Zumlar A, eds. 13. Johnson RT. Virus invasion of the nervous system: a study of sindbis
Mansons Tropical Diseases. London: Saunders; 2002:773793. virus infection in the mouse using fluorescent antibody. Am J Pathol.
3. Gritsun TS, Lashkevich VA, Gould EA. Tick-borne encephalitis. Antiviral 1965;46:929931.
Res. 2003;57:129146. 14. Johnson RT, Mimms CA. Pathogenesis of viral infections of the central
4. Centers for Disease Control and Prevention. West Nile virus infection nervous system. N Engl J Med. 1968;278:2325.
in organ donor and transplant recipientsGeorgia and Florida, 2002. 15. Liu C, Voth DW, Rodina P, et al. A comparative study of the pathogen-
MMWR Morb Mortal Wkly Rep. 2002;51:790. esis of western equine and eastern equine encephalomyelitis viral infec-
5. Griffin D. Arboviruses and the central nervous system. Semin tions in mice by intracerebral and subcutaneous inoculations. J Infect Dis.
Immunopathol. 1995;17:121132. 1970;122:5363.
6. Johnston LJ, Halliday GM, King NJ. Langerhans cells migrate to local 16. Monath TP, Cropp CP, Harrison AK. Mode of entry of a neurotropic virus
lymph nodes following cutaneous infection with an arbovirus. J Invest into the central nervous system. Reinvestigation of an old controversy. Lab
Dermatol. 2000;114:560568. Invest. 1983;48:399410.
7. Wu SJ, Grouard-Vogel G, Sun W, et al. Human skin Langerhans cells are 17. Diamond MS, Klein RS. West Nile virus: crossing the blood-brain barrier.
targets of dengue virus infection. Nat Med. 2000;6:816820. Nat Med. 2004;10:12941295.
8. Labuda M, Austyn JM, Zuffova E, et al. Importance of localized skin infection 18. Olsen AL, Morrey JD, Smee DF, et al. Correlation between breakdown of
in tick-borne encephalitis virus transmission. Virology. 1996;219:357366. the bloodbrain barrier and disease outcome of viral encephalitis in mice.
9. Lim PY, Behr MJ, Chadwick CM, et al. Keratinocytes are cell targets of Antiviral Res. 2007;75:104112.
West Nile virus in vivo. J Virol. 2011;85:51975201. 19. Liu TH, Liang LC, Wang CC, et al. The blood-brain barrier in the cerebrum
10. Lustig S, Halevy M, Ben-Nathan D, et al. The role of host immunocompe- is the initial site for the Japanese encephalitis virus entering the central
tence in neuroinvasion of Sindbis virus. Arch Virol. 1999;144:11591171. nervous system. J Neurovirol. 2008;14:514521.
11. Griffin DE. Alphaviruses. In: Knipe DM, Howley PM, eds. Fields Virology. 20. Roe K, Kumar M, Lum S, et al. West Nile virus-induced disruption of
Philadelphia: Lippincott Williams & Wilkins; 2001:917962. the bloodbrain barrier in mice is characterized by the degradation of the

Scheld_Ch15.indd 232 2/21/14 5:38 PM


Chapter 15: Arthropod-Borne Viral Encephalitides 233

junctional complex proteins and increase in multiple matrix metallopro- 50. Johnson RT, Burke DS, Elwell M, et al. Japanese encephalitis: immunocyto-
teinases. J Gen Virol. 2012;93:11931203. chemical studies of viral antigen and inflammatory cells in fatal cases. Ann
21. Griffin DE, Levine B, Tyor WR, et al. The immune response in viral en- Neurol. 1985;18:567573.
cephalitis. Semin Immunol. 1992;4:111119. 51. Miyake M. The pathology of Japanese encephalitis. Bull World Health
22. Landis H, Simon-Jodicke A, Kloti A, et al. Human MxA protein confers Organ. 1964;30:153160.
resistance to Semliki Forest virus and inhibits the amplification of a Semliki 52. Sampson BA, Ambrosi C, Charlot A, et al. The pathology of human West
Forest virus-based replicon in the absence of viral structural proteins. Nile Virus infection. Hum Pathol. 2000;31:527531.
J Virol. 1998;72:15161522. 53. Shieh WJ, Guarner J, Layton M, et al. The role of pathology in an investiga-
23. Klimstra WB, Ryman KD, Bernard KA, et al. Infection of neonatal mice tion of an outbreak of West Nile encephalitis in New York, 1999. Emerg
with sindbis virus results in a systemic inflammatory response syndrome. Infect Dis. 2000;6:370372.
J Virol. 1999;73:1038710398. 54. Kalfayan B. Pathology of La Crosse virus infection in humans. Prog Clin
24. Burke DS, Morill JC. Levels of interferon in the plasma and cerebrospinal fluid Biol Res. 1983;123:179186.
of patients with acute Japanese encephalitis. J Infect Dis. 1987;155:797799. 55. Bastian FO, Wende RD, Singer DB, et al. Eastern equine encephalomyelitis.
25. Ravi V, Parida S, Desai A, et al. Correlation of tumor necrosis factor levels Histopathologic and ultrastructural changes with isolation of the virus in a
in the serum and cerebrospinal fluid with clinical outcome in Japanese en- human case. Am J Clin Pathol. 1975;64:1013.
cephalitis patients. J Med Virol. 1997;51:132136. 56. Rozdilsky B, Robertson HE, Chorney J. Western encephalitis: report of
26. Winter PM, Dung NM, Loan HT, et al. Proinflammatory cytokines and eight fatal cases. Saskatchewan epidemic, 1965. Can Med Assoc J. 1968;98:
chemokines in humans with Japanese encephalitis. J Infect Dis. 2004;190: 7986.
16181626. 57. Moench TR, Griffin DE. Immunocytochemical identification and quanti-
27. Burke DS, Nisalak A, Ussery MA, et al. Kinetics of IgM and IgG responses tation of the mononuclear cells in the cerebrospinal fluid, meninges, and
to Japanese encephalitis virus in human serum and cerebrospinal fluid. brain during acute viral meningoencephalitis. J Exp Med. 1984;159:7788.
J Infect Dis. 1985;151:10931099. 58. Griffin DE, Levine B, Ubol S, et al. The effects of alphavirus infection on
28. Tardei G, Ruta S, Chitu V, et al. Evaluation of immunoglobulin M (IgM) neurons. Ann Neurol. 1994;35(suppl):S23S27.
and IgG enzyme immunoassays in serologic diagnosis of West Nile virus 59. Garen PD, Tsai TF, Powers JM. Human eastern equine encephalitis:
infection. J Clin Microbiol. 2000;38:22322239. immunohistochemistry and ultrastructure. Mod Pathol. 1999;12:646652.
29. Martin DA, Biggerstaff BJ, Allen B, et al. Use of immunoglobulin M cross- 60. Fazakerley JK. Pathogenesis of Semliki Forest virus encephalitis.
reactions in differential diagnosis of human flaviviral encephalitis infections J Neurovirol. 2002;8(suppl 2):6674.
in the United States. Clin Diagn Lab Immunol. 2002;9:544549. 61. Murphy FA, Whitfield SG. Eastern equine encephalitis virus infection: elec-
30. Calisher CH, Berardi VP, Muth DJ, et al. Specificity of immunoglobulin tron microscopic studies of mouse central nervous system. Exp Mol Pathol.
M and G antibody responses in humans infected with eastern and western 1970;13:131146.
equine encephalitis viruses: application to rapid serodiagnosis. J Clin 62. Tyor WR, Wesselingh S, Levine B, et al. Long term intraparenchymal Ig secre-
Microbiol. 1986;23:369372. tion after acute viral encephalitis in mice. J Immunol. 1992;149:40164020.
31. Burke DS, Lorsomrudee W, Leake CJ, et al. Fatal outcome in Japanese 63. Lewallen S, White VA, Whitten RO, et al. Clinical-histopathological corre-
encephalitis. Am J Trop Med Hyg. 1985;34:12031210. lation of the abnormal retinal vessels in cerebral malaria. Arch Ophthalmol.
32. Solomon T, Fisher AF, Beasley DWC, et al. Natural and nosocomial infec- 2000;118:924928.
tion in a patient with West Nile encephalitis and extrapyramidal movement 64. Noran HH. Chronic equine encephalitis. Am J Pathol. 1944;20:259267.
disorders. Clin Infect Dis. 2003;36:E140E145. 65. Gritsun TS, Frolova TV, Zhankov AI, et al. Characterization of a siberian
33. Griffin D, Levine B, Tyor W, et al. The role of antibody in recovery from virus isolated from a patient with progressive chronic tick-borne encephalitis.
alphavirus encephalitis. Immunol Rev. 1997;159:155161. J Virol. 2003;77:2536.
34. Desprs P, Griffin JW, Griffin DE. Effects of anti-E2 monoclonal anti- 66. Tesh RB, Siirin M, Guzman H, et al. Persistent West Nile Virus infection in
body on sindbis virus replication in AT3 cells expressing bcl-2. J Virol. the golden hamster: studies on its mechanism and possible implications for
1995;69:70067014. other flavivirus infections. J Infect Dis. 2005;192:287295.
35. Tyor WR, Moench TR, Griffin DE. Characterization of B cells and IA expres- 67. McJunkin JE, de los Reyes EC, Irazuzta JE, et al. La Crosse encephalitis in
sion in Sindbis virus encephalitis. Ann N Y Acad Sci. 1988;540: 491492. children. N Engl J Med. 2001;344:801807.
36. Linn ML, Mateo L, Gardner J, et al. Alphavirus-specific cytotoxic T 68. Solomon T, Dung NM, Kneen R, et al. Seizures and raised intracranial
lymphocytes recognize a cross-reactive epitope from the capsid protein pressure in Vietnamese patients with Japanese encephalitis. Brain. 2002;
and can eliminate virus from persistently infected macrophages. J Virol. 125:10841093.
1998;72:51465153. 69. Solomon T, Kneen R, Dung NM, et al. Poliomyelitis-like illness due to
37. Diamond MS, Shrestha B, Marri A, et al. B cells and antibody play criti- Japanese encephalitis virus. Lancet. 1998;351:10941097.
cal roles in the immediate defense of disseminated infection by West Nile 70. Gadoth N, Weitzman S, Lehmann EE. Acute anterior myelitis complicating
encephalitis virus. J Virol. 2003;77:25782586. West Nile fever. Arch Neurol. 1979;36:172173.
38. Heinz FX, Berger R, Tuma W, et al. A topological and functional model 71. Schellinger PD, Schmutzhard E, Fiebach JB, et al. Poliomyelitic-like illness
of epitopes on the structural glycoprotein of tick-borne encephalitis virus in central European encephalitis. Neurology. 2000;55:299302.
defined by monoclonal antibodies. Virology. 1983;126:525537. 72. Kneen R, Solomon T, Appleton R. The role of lumbar puncture in suspected
39. Kaufman BM, Summers PL, Dubois DR, et al. Monoclonal antibodies for CNS infectiona disappearing skill? Arch Dis Child. 2002;87:181183.
dengue virus prM glycoprotein protect mice against lethal dengue infection. 73. White NJ. Lumbar puncture in cerebral malaria (letter). Lancet. 1991;
Am J Trop Med Hyg. 1989;41:576580. 338:640641.
40. Larena M, Regner M, Lee E, et al. Pivotal role of antibody and subsidiary 74. Solomon T. Flavivirus encephalitis. N Engl J Med. 2004;351:370378.
contribution of CD8 T cells to recovery from infection in a murine model 75. Solomon T, Ooi MH, Beasley DWC, et al. West Nile encephalitis. BMJ.
of japanese encephalitis. J Virol. 2011;85:54465455. 2003;326:865869.
41. Charlier N, Leyssen P, Paeshuyse J, et al. Infection of SCID mice with 76. Sambol AR, Hinrichs SH, Hogrefe WR, et al. Performance of a commercial
Montana Myotis leukoencephalitis virus as a model for flavivirus encepha- immunoglobulin M antibody capture assay using analyte-specific reagents
litis. J Gen Virol. 2002;83:18871896. to screen for interfering factors during a West Nile virus epidemic season in
42. Halevy M, Akov Y, Ben-Nathan D, et al. Loss of active neuroinvasiveness Nebraska. Clin Vaccine Immunol. 2007;14:8789.
in attenuated strains of West Nile virus: pathogenicity in immunocompe- 77. Johnson AJ, Noga AJ, Kosoy O, et al. Duplex microsphere-based immu-
tent and SCID mice. Arch Virol. 1994;137:355370. noassay for detection of anti-west nile virus and anti-St. Louis encephalitis
43. Biswas SM, Ayachit VM, Sapkal GN, et al. Japanese encephalitis virus virus immunoglobulin M antibodies. Clin Diagn Lab Immunol. 2005;12:
produces a CD4 Th2 response and associated immunoprotection in an 566574.
adoptive-transfer murine model. J Gen Virol. 2009;90:818826. 78. Taketa-Graham M, Powell-Pereira JL, Baylis E, et al. High throughput
44. Sitati EM, Diamond MS. CD4 T-Cell responses are required for quantitative colorimetric microneutralization assay for the confirmation
clearance of West Nile Virus from the central nervous system. J Virol. and differentiation of West Nile Virus and St. Louis encephalitis virus. Am
2006;80:1206012069. J Trop Med Hyg. 2010:82;501504.
45. Shrestha B, Diamond MS. Role of CD8 T cells in control of West Nile 79. Teles FSRR. Biosensors and rapid diagnostic tests on the frontier between
virus infection. J Virol. 2004;78:83128321. analytical and clinical chemistry for biomolecular diagnosis of dengue
46. Shrestha B, Samuel MA, Diamond MS. CD8 T Cells Require Perforin To disease: a review. Analytica Chimica Acta. 2011;687:2842.
Clear West Nile Virus from Infected Neurons. J Virol. 2006;80:119129. 80. Lee Y-F, Lien K-Y, Lei H-Y, et al. An integrated microfluidic system for rapid
47. Wang Y, Lobigs M, Lee E, et al. CD8 T cells mediate recovery and immu- diagnosis of dengue virus infection. Biosens Bioelectron. 2009;25:745752.
nopathology in West Nile virus encephalitis. J Virol. 2003;77:1332313334. 81. Naze F, Le Roux K, Schuffenecker I, et al. Simultaneous detection and
48. Wang Y, Lobigs M, Lee E, et al. Exocytosis and Fas mediated cytolytic quantitation of Chikungunya, dengue and West Nile viruses by multiplex
mechanisms exert protection from West Nile virus induced encephalitis in RT-PCR assays and dengue virus typing using high resolution melting.
mice. Immunol Cell Biol. 2004;82:170173. J Virol Methods. 2009;162:17.
49. Licon Luna RM, Lee E, Mllbacher A, et al. Lack of both Fas ligand and 82. Solomon T, Dung NM, Wills B, et al. Interferon alfa-2a in Japanese enceph-
perforin protects from flavivirus-mediated encephalitis in mice. J Virol. alitis: a randomised double-blind placebo-controlled trial. Lancet. 2003;
2002;76:32023211. 361:821826.

Scheld_Ch15.indd 233 2/21/14 5:38 PM


234 Part II: Viral Infections and Related Disorders

83. Appleton R, Choonara I, Martland T, et al. The treatment of convulsive sta- 114. Longshore WA, Stevens IM, Hollister AC, et al. Epidemiologic observa-
tus epilepticus in children. The Status Epilepticus Working Party, Members of tions on acute infectious encephalitis in California with special reference
the Status Epilepticus Working Party. Arch Dis Child. 2000;83:415419. to the 1952 outbreak. Am J Hyg. 1956;63:6986.
84. Kirkham FJ. Non-traumatic coma in children. Arch Dis Child. 2001;85: 115. Schultz DR, Barthal JS, Garrett G. Western equine encephalitis with rapid
303312. onset of parkinsonism. Neurology. 1977;27:10951096.
85. Crawley J, Waruiru C, Mithwani S, et al. Effect of phenobarbital on sei- 116. Bia FJ, Thornton GF, Main AJ, et al. Western equine encephalitis mimick-
zure frequency and mortality in childhood cerebral malaria: a randomised, ing herpes simplex encephalitis. JAMA. 1980;244:367369.
controlled intervention study. Lancet. 2000;355:701706. 117. Sciple GW, Ray G, LaMotte LC, et al. Western encephalitis with recovery
86. Solomon T, Vaughn DW. Pathogenesis and clinical features of Japanese of virus from cerebrospinal fluid. Neurology. 1967;17:169171.
encephalitis and West Nile virus infections. In: Mackenzie JS, Barrett 118. Gold H, Hampil B. Equine encephalomyelitis in a laboratoy technician
AD, Deubel V. eds. Current Topics in Microbiology and Immunology: with recovery. Ann Intern Med. 1942;16:556569.
Japanese Encephalitis and West Nile Virus Infections. Berlin: Springer- 119. Randall R, Mills JW, Engel LL. The preparation and properties of a puri-
Verlag; 2002:171194. fied equine encephalomyelitis vaccine. J Immunol. 1947;55:4152.
87. Plum F, Posner JB. The pathologic physiology of signs and symptoms of 120. Barabe ND, Rayner GA, Christopher ME, et al. Single-dose, fast-acting
coma. In: Plum F, Posner JB, eds. The Diagnosis of Stupor and Coma. vaccine candidate against western equine encephalitis virus completely
Philadelphia: F. A. Davis Co.; 1982:173. protects mice from intranasal challenge with different strains of the virus.
88. Newton CRJC, Crawley J, Sowumni A, et al. Intracranial hypertension in Vaccine. 2007;25:62716276.
Africans with cerebral malaria. Arch Dis Child. 1997;76:219226. 121. Wu JQ, Barab ND, Chau D, et al. Complete protection of mice against a
89. Hoke CH, Vaughn DW, Nisalak A, et al. Effect of high dose dexametha- lethal dose challenge of western equine encephalitis virus after immuniza-
sone on the outcome of acute encephalitis due to Japanese encephalitis tion with an adenovirus-vectored vaccine. Vaccine. 2007;25:43684375.
virus. J Infect Dis. 1992;165:631637. 122. Beck CE, Wyckoff RWG. Venezuelan equine encephalomyelitis. Science.
90. Solomon T, Felix JM, Samuel M, et al. Hypoglycaemia in paediatric 1938;88:530.
admissions in Mozambique. Lancet. 1994;343:149150. 123. Calisher CH, Monath TP, Mitchell CJ, et al. Arbovirus investigations in
91. Oliva CF, Jacquet M, Gilles J, et al. The sterile insect technique for con- Argentina, 19771980. III. Identification and characterization of viruses
trolling populations of Aedes albopictus (diptera: culicidae) on Reunion isolated, including new subtypes of western and Venezuelan equine en-
island: mating vigour of sterilized males. PLoS ONE. 2012;7:e49414. cephalitis viruses and four new bunyaviruses (Las Maloyas, Resistencia,
92. Fu G, Lees RS, Nimmo D, et al. Female-specific flightless phenotype for Barranqueras, and Antequera). Am J Trop Med Hyg. 1985;34:956965.
mosquito control. Proc Natl Acad Sci U S A. 2010;107:45504554. 124. Talarmin A, Trochu J, Gardon J, et al. Tonate virus infection in French
93. Hoffmann AA, Montgomery BL, Popovici J, et al. Successful establish- Guiana: clinical aspects and seroepidemiologic study. Am J Trop Med
ment of Wolbachia in Aedes populations to suppress dengue transmission. Hyg. 2001;64:274279.
Nature. 2011;476:454457. 125. Hommel D, Heraud JM, Hulin A, et al. Association of Tonate virus (sub-
94. Caragata EP, Walker T. Using bacteria to treat diseases. Expert Opin Biol type IIIB of the Venezuelan equine encephalitis complex) with encephalitis
Ther. 2012;12:701712. in a human. Clin Infect Dis. 2000;30:188190.
95. Weaver SC, Kang W, Shirako Y, et al. Recombinational history and molec- 126. Libraty DH, Nisalak A, Endy TP, et al. Clinical and immunological risk
ular evolution of western equine encephalomyelitis complex alphaviruses. factors for severe disease in Japanese encephalitis. Trans R Soc Trop Med
J Virol. 1997;71:613623. Hyg. 2002;96:173178.
96. Lescar J, Roussel A, Wien MW, et al. The Fusion glycoprotein shell of 127. Brault AC, Powers AM, Weaver SC. Vector infection determinants of
Semliki Forest virus: an icosahedral assembly primed for fusogenic activa- Venezuelan equine encephalitis virus reside within the E2 envelope glyco-
tion at endosomal pH. Cell. 2001;105:137148. protein. J Virol. 2002;76:63876392.
97. TenBroeck C, Merrill MH. A serological difference between eastern 128. Weaver SC, Salas R, Rico-Hesse R, et al. Re-emergence of epidemic
and western equine encephalomyelitis virus. Proc Soc Exp Biol Med. Venezuelan equine encephalomyelitis in South America. VEE Study
1933;31:217220. Group. Lancet. 1996;348:436440.
98. Beckwith WH, Sirpenski S, French RA, et al. Isolation of eastern equine 129. Wang E, Barrera R, Boshell J, et al. Genetic and phenotypic changes
encephalitis virus and West Nile virus from crows during increased accompanying the emergence of epizootic subtype IC Venezuelan equine
arbovirus surveillance in Connecticut, 2000. Am J Trop Med Hyg. encephalitis viruses from an enzootic subtype ID progenitor. J Virol.
2002;66:422426. 1999;73:42664271.
99. Day JF, Stark LM. Eastern equine encephalitis transmission to emus 130. Powers AM, Oberste MS, Brault AC, et al. Repeated emergence of epi-
(Dromaius novaehollandiae) in Volusia County, Florida: 1992 through demic/epizootic Venezuelan equine encephalitis from a single genotype of
1994. J Am Mosq Control Assoc. 1996;12:429436. enzootic subtype ID virus. J Virol. 1997;71:66976705.
100. Goldfield M, Welsh JN, Taylor BF. The 1959 outbreak of Eastern 131. Rico-Hesse R, Weaver SC, de Siger J, et al. Emergence of a new epidemic/
encephalitis in New Jersey. 5. The inapparent infection:disease ratio. Am epizootic Venezuelan equine encephalitis virus in South America. Proc
J Epidemiol. 1968;87:3233. Natl Acad Sci U S A. 1995:92;52785281.
101. Clarke DH. Two nonfatal human infections with the virus of eastern 132. Weaver SC, Pfeffer M, Marriott K, et al. Genetic evidence for the ori-
equine encephalitis. Am J Trop Med Hyg. 1961;10:6770. gins of Venezuelan equine encephalitis virus subtype IAB outbreaks. Am J
102. Deresiewicz RL, Thaler SJ, Hsu L, et al. Clinical and neuroradiographic Trop Med Hyg. 1999;60:441448.
manifestations of eastern equine encephalitis. N Engl J Med. 1997; 133. Rivas F, Diaz LA, Cardenas VM, et al. Epidemic Venezuelan equine enceph-
336:18671874. alitis in La Guajira, Colombia, 1995. J Infect Dis. 1997;175:828832.
103. Przelomski MM, ORourke E, Grady GF, et al. Eastern equine encepha- 134. Bowen GS, Calisher CH. Virological and serological studies of Venezuelan
litis in Massachusetts: a report of 16 cases, 19701984. Neurology. equine encephalomyelitis in humans. J Clin Microbiol. 1976;4:2227.
1988;38:736739. 135. Bowen GS, Fashinell TR, Dean PB, et al. Clinical aspects of human
104. Cole FE Jr. Inactivated eastern equine encephalomyelitis vaccine propa- Venezuelan equine encephalitis in Texas. Bull Pan Am Health Organ.
gated in rolling-bottle cultures of chick embryo cells. Appl Microbiol. 1976;10:4657.
1971;22:842845. 136. Sanchez JL, Takafuji ET, Lednar WM, et al. Venezuelan equine encephalo-
105. Wang E, Volkova E, Adams AP, et al. Chimeric alphavirus vaccine candi- myelitis: report of an outbreak associated with jungle exposure. Mil Med.
dates for chikungunya. Vaccine. 2008;26:50305039. 1984;149:618621.
106. Meyer KF, Haring CM, Howitt B. The etiology of encephalomyelitis in 137. Molina OM, Morales MC, Soto ID, et al. Venezuelan equine encephalitis.
horses in the San Joaquin Valley. Science. 1931;74:227228. 1995 outbreak: clinical profile of the case with neurologic involvement.
107. Howitt BF. Recovery of the virus of equine encephalomyelitis from the Rev Neurol. 1999;29:296298.
brain of a child. Science. 1938;88:455456. 138. Watts DM, Callahan J, Rossi C, et al. Venezuelan equine encephalitis
108. Calisher CH, Karabatsos N, Lazuick JS, et al. Reevaluation of the western febrile cases among humans in the Peruvian Amazon River region. Am J
equine encephalitis antigenic complex of alphaviruses (family Togaviridae) as Trop Med Hyg. 1998;58:3540.
determined by neutralization tests. Am J Trop Med Hyg. 1988;38:447452. 139. Wang E, Paessler S, Aguilar PV, et al. A novel, rapid assay for detection
109. Reisen WK, Chiles RE, Lothrup HD, et al. Prevalence of antibodies to and differentiation of serotype-specific antibodies to Venezuelan equine en-
mosquito-borne encephalitis viruses in residents of the Coachella Valley, cephalitis complex alphaviruses. Am J Trop Med Hyg. 2005;72:805810.
California. Am J Trop Med Hyg. 1996;55:667671. 140. Berge TO, Banks IS, Tigertt WD. Attenuation of Venezuelan equine
110. Calisher CH. Medically important arboviruses of the United States and encephalomyelitis virus by in vitro cultivation in guinea-pig heart cells.
Canada. Clin Microbiol Rev. 1994;7:89116. Am J Hyg. 1961;73:209218.
111. Bigler WJ, Lassing EB, Buff EE, et al. Endemic eastern equine encephalo- 141. Hart MK, Caswell-Stephan K, Bakken R, et al. Improved mucosal pro-
myelitis in Florida: a twenty-year analysis, 19551974. Am J Trop Med tection against Venezuelan equine encephalitis virus is induced by the
Hyg. 1976;25:884890. molecularly defined, live-attenuated V3526 vaccine candidate. Vaccine.
112. Arboviral infections of the central nervous systemUnited States, 1987. 2000;18:30673075.
MMWR Morb Mortal Wkly Rep. 1988;37:506508, 513505. 142. Fine DL, Roberts BA, Terpening SJ, et al. Neurovirulence evaluation of
113. Copps SC, Giddings LE. Transplacental transmission of western equine Venezuelan equine encephalitis (VEE) vaccine candidate V3526 in non-
encephalitis. Pediatrics. 1959;24:3133. human primates. Vaccine. 2008;26:34973506.

Scheld_Ch15.indd 234 2/21/14 5:38 PM


Chapter 15: Arthropod-Borne Viral Encephalitides 235

143. Paessler S, Fayzulin RZ, Anishchenko M, et al. Recombinant sindbis/ 174. Hasebe F, Parquet MC, Pandey BD, et al. Combined detection and
Venezuelan equine encephalitis virus is highly attenuated and immuno- genotyping of Chikungunya virus by a specific reverse transcription-
genic. J Virol. 2003;77:92789286. polymerase chain reaction. J Med Virol. 2002;67:370374.
144. Paessler S, Ni H, Petrakova O, et al. Replication and clearance of 175. Litzba N, Schuffenecker I, Zeller H, et al. Evaluation of the first commer-
Venezuelan equine encephalitis virus from the brains of animals vacci- cial chikungunya virus indirect immunofluorescence test. J Virol Methods.
nated with chimeric SIN/VEE viruses. J Virol. 2006;80:27842796. 2008;149:175179.
145. Paessler S, Rijnbrand R, Stein DA, et al. Inhibition of alphavirus infection 176. Shukla J, Khan M, Tiwari M, et al. Development and evaluation of
in cell culture and in mice with antisense morpholino oligomers. Virology. antigen capture ELISA for early clinical diagnosis of chikungunya. Diagn
2008;376:357370. Microbiol Infect Dis. 2009;65:142149.
146. Goodchild SA, OBrien LM, Steven J, et al. A humanised murine mono- 177. Yap G, Pok KY, Lai YL, et al. Evaluation of chikungunya diagnostic assays:
clonal antibody with broad serogroup specificity protects mice from differences in sensitivity of serology assays in two independent outbreaks.
challenge with Venezuelan equine encephalitis virus. Antiviral Res. PLoS Negl Trop Dis. 2010;4:e753.
2011;90:18. 178. de Lamballerie X, Ninove L, Charrel RN. Antiviral treatment of chikun-
147. Lumsden WH. An epidemic of virus disease in Southern Province, gunya virus infection. Infect Disord Drug Targets. 2009;9:101104.
Tanganyika Territory, in 1952-53. II. General description and epidemiol- 179. Moulay D, Aziz-Alaoui MA, Kwon HD. Optimal control of chikungunya
ogy. Trans R Soc Trop Med Hyg. 1955;49:3357. disease: larvae reduction, treatment and prevention. Math Biosci Eng.
148. Carey DE. Chikungunya and dengue: a case of mistaken identity? J Hist 2012;9:369392.
Med Allied Sci. 1971;26:243262. 180. Tiwari M, Parida M, Santhosh SR, et al. Assessment of immunogenic
149. Ross RW. The Newala epidemic. III. The virus: isolation, pathogenic prop- potential of Vero adapted formalin inactivated vaccine derived from novel
erties and relationship to the epidemic. J Hyg (Lond). 1956;54:177191. ECSA genotype of Chikungunya virus. Vaccine. 2009;27:25132522.
150. Spence LP, Thomas L. Application of haemagglutination and comple- 181. Mallilankaraman K, Shedlock DJ, Bao H, et al. A DNA vaccine against
mentfixation techniques to the identification and serological classification chikungunya virus is protective in mice and induces neutralizing antibodies
of arthropod-borne viruses; studies on Chikungunya and Makonde vi- in mice and nonhuman primates. PLoS Negl Trop Dis. 2011;5:e928.
ruses. Trans R Soc Trop Med Hyg. 1959;53:248255. 182. Akahata W, Yang ZY, Andersen H, et al. A virus-like particle vaccine
151. Powers AM, Logue CH. Changing patterns of chikungunya virus: for epidemic Chikungunya virus protects nonhuman primates against
re-emergence of a zoonotic arbovirus. J Gen Virol. 2007;88:23632377. infection. Nat Med. 2010;16:334338.
152. Ng LC, Hapuarachchi HC. Tracing the path of Chikungunya virusevo- 183. Wang D, Suhrbier A, Penn-Nicholson A, et al. A complex adenovirus
lution and adaptation. Infect Genet Evol. 2010;10:876885. vaccine against chikungunya virus provides complete protection against
153. Tsetsarkin KA, Chen R, Sherman MB, et al. Chikungunya virus: evolution viraemia and arthritis. Vaccine. 2011;29:28032809.
and genetic determinants of emergence. Curr Opin Virol. 2011;1:310317. 184. Zanotto PM, Gould EA, Gao GF, et al. Population dynamics of flavi-
154. Pulmanausahakul R, Roytrakul S, Auewarakul P, et al. Chikungunya in viruses revealed by molecular phylogenies. Proc Natl Acad Sci U S A.
Southeast Asia: understanding the emergence and finding solutions. Int J 1996;93:548553.
Infect Dis. 2011;15:e671e676. 185. Gould EA, Zanotto PM, Holmes EC. The genetic evolution of flavivi-
155. Renault P, Balleydier E, DOrtenzio E, et al. Epidemiology of chikungunya ruses. In: Saluzzo JF, Dodet B, eds. Factors in the Emergence of Arbovirus
infection on Reunion Island, Mayotte, and neighboring countries. Med Diseases. Paris: Elselvier; 1997:5163.
Mal Infect. 2012;42:93101. 186. Chambers TJ, Hahn CS, Galler R, et al. Flavivirus genome organisation,
156. Tsetsarkin KA, Weaver SC. Sequential adaptive mutations enhance effi- expression, and replication. Annu Rev Microbiol. 1990;44:649688.
cient vector switching by Chikungunya virus and its epidemic emergence. 187. Stadler K, Allison SL, Schalich J, et al. Proteolytic activation of tick-borne
PLoS Pathog. 2011;7:e1002412. encephalitis virus by furin. J Virol. 1997;71:84758481.
157. Rezza G, Nicoletti L, Angelini R, et al. Infection with chikungunya virus 188. Roehrig JT, Hunt AR, Johnson AJ, et al. Synthetic peptides derived from
in Italy: an outbreak in a temperate region. Lancet. 2007;370:18401846. the deduced amino acid sequence of the E-glycoprotein of Murray Valley
158. Gould EA, Gallian P, De Lamballerie X, et al. First cases of autochthonous encephalitis virus elicit antiviral antibody. Virology. 1989;171:4960.
dengue fever and chikungunya fever in France: from bad dream to reality! 189. Rey FA, Heinz FX, Mandl C, et al. The envelope glycoprotein from tick-
Clin Microbiol Infect. 2010;16:17021704. borne encephalitis virus at 2 resolution. Nature. 1995;375:291298.
159. Lanciotti RS, Kosoy OL, Laven JJ, et al. Chikungunya virus in US travelers 190. Heinz FX, Allison SL. The machinery for flavivirus fusion with host cell
returning from India, 2006. Emerg Infect Dis. 2007;13:764767. membranes. Curr Opin Microbiol. 2001;4:450455.
160. Johnson DF, Druce JD, Chapman S, et al. Chikungunya virus infection in 191. Smithburn KC, Hughes TP, Burke AW, et al. A neurotropic virus isolated
travellers to Australia. Med J Aust. 2008;188:4143. from the bood of a native of Uganda. Am J Trop Med Hyg. 1940;20:
161. Panning M, Grywna K, van Esbroeck M, et al. Chikungunya fever in 471492.
travelers returning to Europe from the Indian Ocean region, 2006. Emerg 192. McIntosh BM, Jupp PG, Dos Santos I, et al. Epidemics of West Nile and
Infect Dis. 2008;14:416422. Sindbis viruses in South Africa with Culex (culex) univittatus Theobold as
162. Halstead SB, Nimmannitya S, Margiotta MR. Dengue and Chikungunya vector. S Afr J Sci. 1976;72:295300.
virus infection in man in Thailand, 19621964: II. Observations on dis- 193. Spigland I, Jasinska-Klingberg W, Hofsbi E, et al. Clinical and labora-
ease in outpatients. Am J Trop Med Hyg. 1969;18:972983. tory observations in an outbreak of West Nile fever in Israel. Harefua
163. Burt FJ, Rolph MS, Rulli NE, et al. Chikungunya: a re-emerging virus. (Jerusalem). 1958;54:275281.
Lancet. 2012;379:662671. 194. Tsai TF, Popovici F, Carnescu C, et al. West Nile encephalitis epidemic in
164. Sissoko D, Malvy D, Ezzedine K, et al. Post-epidemic chikungunya disease Southeastern Romania. Lancet. 1998;352:767771.
on Reunion island: course of rheumatic manifestations and associated fac- 195. Platonov AE, Shipulin GA, Shipulina OY, et al. Outbreak of West Nile
tors over a 15-month period. PLoS Negl Trop Dis. 2009;3:e389. virus infection, Volgograd Region, Russia, 1999. Emerg Infect Dis.
165. Manimunda SP, Vijayachari P, Uppoor R, et al. Clinical progression 2001;7:128132.
of chikungunya fever during acute and chronic arthritic stages and the 196. Chowers MY, Lang R, Nassar F, et al. Clinical characteristics of the West
changes in joint morphology as revealed by imaging. Trans R Soc Trop Nile fever outbreak, Israel, 2000. Emerg Infect Dis. 2001;7:675678.
Med Hyg. 2010;104:392399. 197. Mostashari F, Bunning ML, Kitsutani PT, et al. Epidemic West Nile en-
166. Singh SS, Manimunda SP, Sugunan AP, et al. Four cases of acute flaccid cephalitis, New York, 1999: results of a household-based seroepidemio-
paralysis associated with chikungunya virus infection. Epidemiol Infect. logical survey. Lancet. 2001;358:261264.
2008;136:12771280. 198. Briese T, Jia X-Y, Huang C, et al. Indentification of a Kunjin/West Nile-like
167. Rampal, Sharda M, Meena H. Neurological complications in chikungunya flavivirus in brains of patients with New York encephalitis. Lancet. 1999;
fever. J Assoc Physicians India. 2007;55:765769. 354:12611262.
168. Wielanek AC, Monredon JD, Amrani ME, et al. Guillain-Barr syndrome 199. Nash D, Mostashari F, Fine A, et al. The outbreak of West Nile virus infection
complicating a Chikungunya virus infection. Neurology. 2007;69:21052107. in the New York City area in 1999. N Engl J Med. 2001;344:18071814.
169. Arpino C, Curatolo P, Rezza G. Chikungunya and the nervous system: 200. Murray KO, Mertens E, Despres P. West Nile virus and its emergence in
what we do and do not know. Rev Med Virol. 2009;19:121129. the United States of America. Vet Res. 2010;41:67.
170. Economopoulou A, Dominguez M, Helynck B, et al. Atypical chikun- 201. Davis LE, DeBiasi R, Goade DE, et al. West Nile virus neuroinvasive dis-
gunya virus infections: clinical manifestations, mortality and risk factors ease. Ann Neurol. 2006;60:286300.
for severe disease during the 2005-2006 outbreak on Runion. Epidemiol 202. Danis K, Papa A, Theocharopoulos G, et al. Outbreak of West Nile virus
Infect. 2009;137:534541. infection in Greece, 2010. Emerg Infect Dis. 2011;17:18681872.
171. Robin S, Ramful D, Le Seach F, et al. Neurologic manifestations of pedi- 203. Sirbu A, Ceianu CS, Panculescu-Gatej RI, et al. Outbreak of West Nile
atric Chikungunya infection. J Child Neurol. 2008;23:10281035. virus infection in humans, Romania, July to October 2010. Euro Surveill.
172. Grardin P, Barau G, Michault A. Multidisciplinary prospective study 2011;16:pii:19762.
of mother-to-child chikungunya virus infections on the Island of La R 204. Danis K, Papa A, Papanikolaou E, et al. Ongoing outbreak of West Nile
union. PLoS Med. 2008;5 (3):e60. virus infection in humans, Greece, July to August 2011. Euro Surveill.
173. Panning M, Charrel RN, Mantke OD, et al. Coordinated implementa- 2011;16:pii:19951.
tion of chikungunya virus reverse transcription-PCR. Emerg Infect Dis. 205. Taylor R, Work TH, Harlbut HS, et al. A study of the ecology of West
2009;15:469471. Nile virus in Egypt. Am J Trop Med Hyg. 1956;5:579620.

Scheld_Ch15.indd 235 2/21/14 5:38 PM


236 Part II: Viral Infections and Related Disorders

206. Mackenzie JS, Williams DT. The zoonotic flaviviruses of Southern, South- 239. Kumar R, Mathur A, Kumar A, et al. Clinical features and prognostic
Eastern and Eastern Asia, and Australasia: the potential for emergent indicators of Japanese encephalitis in children in Lucknow (India). Indian
viruses. Zoonoses Public Health. 2009;56:338356. J Med Res. 1990;91:321327.
207. Lanciotti RS, Roehrig JT, Deubel V, et al. Origin of the West Nile virus 240. Zimmerman HM. The pathology of Japanese B encephalitis. Am J Pathol.
responsible for an outbreak of encephalitis in the northeastern United 1946;22:965991.
States. Science. 1999;286:23332337. 241. Misra UK, Kalita J. Anterior horn cells are also involved in Japanese
208. Lanciotti RS, Ebel GD, Deubel V, et al. Complete genome sequences and encephalitis. Acta Neurol Scand. 1997;96:114117.
phylogenetic analysis of West Nile virus strains isolated from the United 242. Desai A, Ravi V, Guru SC, et al. Detection of autoantibodies to neural
States, Europe, and the Middle East. Virology. 2002;298:96105. antigens in the CSF of Japanese encephalitis patients and correlation of
209. Beasley DW, Li L, Suderman MT, et al. Mouse neuroinvasive phenotype findings with the outcome. J Neurol Sci. 1994;122:109116.
of West Nile virus strains varies depending upon virus genotype. Virology. 243. Dapeng L, Jindou S, Huijun Y, et al. Prognostic factors of early sequelae
2002;296:1723. and fatal outcome of Japanese encephalitis. Southeast Asian J Trop Med
210. De Filette M, Ulbert S, Diamond M, et al. Recent progress in West Nile Public Health. 1995;26:694698.
virus diagnosis and vaccination. Vet Res. 2012;43:16. 244. Misra UK, Kalita J, Srivastava M. Prognosis of Japanese encephalitis: a
211. Papa A. West Nile virus infections in Greece: an update. Expert Rev Anti multivariate analysis. J Neurol Sci. 1998;161:143147.
Infect Ther. 2012;10:743750. 245. Solomon T, Thao LTT, Dung NM, et al. Rapid diagnosis of Japanese
212. Rappole JH, Derrickson SR, Hublek Z. Migratory birds and spread of West encephalitis by using an immunoglobulin M dot enzyme immunoassay.
Nile virus in the Western Hemisphere. Emerg Infect Dis. 2000;6:319328. J Clin Microbiol. 1998;36:20302034.
213. Centers for Disease Control and Prevention. West Nile virus activity 246. Burke DS, Nisalak A, Lorsomrudee W, et al. Virus-specific antibody-
United States, October 10-16, 2002, and update on West Nile virus infec- producing cells in blood and cerebrospinal fluid in acute Japanese enceph-
tions in recipients of blood transfusions. MMWR Morb Mortal Wkly Rep. alitis. J Med Virol. 1985;17:283292.
2002;51:929931. 247. Ravi V, Desai A, Balaji M, et al. Development and evaluation of a rapid
214. Goldblum N, Sterk VV, Paderski B. West Nile fever: the clinical features IgM capture ELISA (JEV-Chex) for the diagnosis of Japanese encephalitis.
of the disease and the isolation of West Nile virus from the blood of nine J Clin Virol. 2006;35:429434.
human cases. Am J Hyg. 1954;59:89103. 248. Igarashi A, Tanaka M, Morita K, et al. Detection of West Nile and Japanese
215. Asnis DS, Conetta R, Teixeira AA, et al. The West Nile virus outbreak encephalitis viral genome sequences in cerebrospinal fluid from acute enceph-
of 1999 in New York: the Flushing Hospital experience. Clin Infect Dis. alitis cases in Karachi, Pakistan. Microbiol Immunol. 1994;38:827830.
2000;30:413418. 249. Harinasuta C, Wasi C, Vithanomsat S. The effect of interferon on
216. Leis AA, Stokic DS, Polk JL, et al. A poliomyelitis-like syndrome from Japanese encephalitis virus in vitro. Southeast Asian J Trop Med Public
West Nile virus infection. N Engl J Med. 2002;347:12791280. Health. 1984;15:564568.
217. Misra UK, Kalita J. Movement disorders in Japanese encephalitis. 250. Huggins JW, Robins RK, Canonico P. Synergistic antiviral effects of
J Neurol. 1997;244:299303. ribavirin and the C-nucleoside analogs tiazofurin and selenazofurin
218. Cernesu C, Ruta SM, Tardei G, et al. A high number of severe neurologic against togaviruses, bunyaviruses, and arenaviruses. Antimicrob Agents
clinical forms during an epidemic of West Nile virus infection. Rom J Chemother. 1984;26:476480.
Virol. 1997;48:1325. 251. Saxena SK, Singh A, Mathur A. Antiviral effect of nitric oxide during
219. Weiss D, Carr D, Kellachan J, et al. Clinical findings of West Nile virus Japanese encephalitis virus infection. Int J Exp Pathol. 2000;81:165172.
infection in hospitalized patients, New York and New Jersey, 2000. 252. Harinasuta C, Nimmanitya S, Titsyakorn U. The effect of interferon alpha
Emerg Infect Dis. 2001;7:654658. A on two cases of Japanese encephalitis in Thailand. Southeast Asian J
220. Petersen LR, Marfin AA. West Nile virus: a primer for the clinician. Ann Trop Med Public Health. 1985;16:332336.
Intern Med. 2002;137:173179. 253. Das S, Dutta K, Kumawat KL, et al. Abrogated inflammatory response
221. Lanciotti RS, Kerst AJ, Nasci RS, et al. Rapid detection of West Nile virus promotes neurogenesis in a murine model of Japanese encephalitis. PLoS
from human clinical specimens, field-collected mosquitoes, and avian One. 2011;6:e17225.
samples by a TaqMan reverse transcriptase-PCR assay. J Clin Microbiol. 254. Mishra MK, Dutta K, Saheb SK, et al. Understanding the molecular
2000;38:40664071. mechanism of blood-brain barrier damage in an experimental model of
222. Dauphin G, Zientara S. West Nile virus: recent trends in diagnosis and Japanese encephalitis: correlation with minocycline administration as a
vaccine development. Vaccine. 2007;25:55635576. therapeutic agent. Neurochem Int. 2009;55:717723.
223. Huang C, Slater B, Rudd R, et al. First isolation of West Nile virus 255. Sebastian L, Desai A, Madhusudana SN, et al. Pentoxifylline inhibits rep-
from a patient with encephalitis in the United States. Emerg Infect Dis. lication of Japanese encephalitis virus: a comparative study with ribavirin.
2002;8:13671371. Int J Antimicrob Agents. 2009;33:168173.
224. Davis BS, Chang GJ, Cropp B, et al. West Nile virus recombinant DNA 256. Lacey LA, Lacey CM. The medical importance of riceland mosquitoes
vaccine protects mouse and horse from virus challenge and expresses in and their control using alternatives to chemical insecticides. J Am Mosq
vitro a noninfectious recombinant antigen that can be used in enzyme- Control Assoc Suppl. 1990;2:193.
linked immunosorbent assays. J Virol. 2001;75:40404047. 257. Hoke CH, Nisalak A, Sangawhipa N, et al. Protection against Japanese
225. Ng T, Hathaway D, Jennings N, et al. Equine vaccine for West Nile virus. encephalitis by inactivated vaccines. N Engl J Med. 1988;319:608614.
Dev Biol (Basel). 2003;114:221227. 258. Takahashi H, Pool V, Tsai TF, et al. Adverse events after Japanese en-
226. Guy B, Barrere B, Malinowski C, et al. From research to phase III: preclin- cephalitis vaccination: review of post-marketing surveillance data from
ical, industrial and clinical development of the Sanofi Pasteur tetravalent Japan and the United States. The VAERS Working Group. Vaccine.
dengue vaccine. Vaccine. 2011;29:72297241. 2000;18:29632969.
227. Beasley DW. Vaccines and immunotherapeutics for the prevention and treat- 259. McArthur MA, Holbrook MR. Japanese encephalitis vaccines. J Bioterror
ment of infections with West Nile virus. Immunotherapy. 2011;3:269285. Biodef. 2011;S1:2.
228. Kaneko R, Aoki Y. Uber die Encephalitis epidemica in Japan. Ergebn d 260. Xin YY, Ming ZG, Peng GY, et al. Safety of a live-attenuated Japanese
inn Med u Kinderh. 1928;34:342456. encephalitis virus vaccine (SA14-14-2) for children. Am J Trop Med Hyg.
229. Solomon T. Japanese encephalitis. In: Gilman S, Goldstein GW, Waxman 1988;39:214217.
SG, eds. Neurobase. San Diego: Medlink Publishing; 2000. 261. Ma X, Yu YX, Wang SG. Observations on safety and serological efficacy
230. Vaughn DW, Hoke CH. The epidemiology of Japanese encephalitis: pros- from a large scale field trial of Japanese encephalitis vaccine. Chin J Biol.
pects for prevention. Epidemiol Rev. 1992;14:197221. 1993;6:188191.
231. Shlim DR, Solomon T. Japanese encephalitis vaccine for travelers: explor- 262. Hennessy S, Zhengle L, Tsai TF, et al. Effectiveness of live-attenuated
ing the limits of risk. Clin Infect Dis. 2002;35:183188. Japanese encephalitis vaccine (SA14-14-2): a case control study. Lancet.
232. Solomon T, Dung NM, Kneen R, et al. Japanese encephalitis. J Neurol 1996;347:15831586.
Neurosurg Psychiatry. 2000;68:405415. 263. Monath TP, McCarthy K, Bedford P, et al. Clinical proof of principle
233. Chen WR, Tesh RB, Ricco-Hesse R. Genetic variation of Japanese for ChimeriVax: recombinant live, attenuated vaccines against flavivirus
encephalitis virus in nature. J Gen Virol. 1990;71:29152922. infections. Vaccine. 2002;20:10041018.
234. Chen WR, Ricco-Hesse R, Tesh RB. A new genotype of Japanese virus 264. Muckenfuss RW, Armstrong C, McCordock HA. Encephalitis: studies on
from Indonesia. Am J Trop Med Hyg. 1992;47:6169. experimental transmission. Public Health Rep. 1933;48:13411343.
235. Solomon T, Ni H, Beasley DW, et al. Origin and evolution of Japanese 265. Monath TP. Epidemiology. In: Monath TP, ed. St Louis Encephalitis.
encephalitis virus in Southeast Asia. J Virol. 2003;77:30913098. Washington, DC: American Public Health Associates; 1980:239312.
236. Mohammed MAF, Galbraith SE, Radford AD, et al. Molecular phylogenetic 266. Okhuysen PC, Crane JK, Pappas J. St. Louis encephalitis in patients
and evolutionary analyses of Muar strain of Japanese encephalitis virus with human immunodeficiency virus infection. Clin Infect Dis. 1993;17:
reveal it is the missing fifth genotype. Infect Genet Evol. 2011;11:855862. 140141.
237. Li M-H, Fu S-H, Chen W-X, et al. Genotype V Japanese encephalitis virus 267. Diaz LA, Re V, Almiron WR, et al. Genotype III Saint Louis encephalitis
is emerging. PLoS Negl Trop Dis. 2011;5:e1231. virus outbreak, Argentina, 2005. Emerg Infect Dis. 2006;12:17521754.
238. Takhampunya R, Kim H-C, Tippayachai B, et al. Emergence of 268. Spinsanti LI, Daz LA, Glatstein N, et al. Human outbreak of St.
Japanese encephalitis virus genotype V in the Republic of Korea. Virol J. Louis encephalitis detected in Argentina, 2005. J Clin Virol. 2008;42:
2011;8:449. 2733.

Scheld_Ch15.indd 236 2/21/14 5:38 PM


Chapter 15: Arthropod-Borne Viral Encephalitides 237

269. Brinker KR, Monath TP. The acute disease. In: Monath TP, ed. St. Louis 302. Nimmannitya S. Clinical spectrum and management of dengue haemor-
Encephalitis. Washington, DC: American Public Health Associates; 1980: rhagic fever. Trans R Soc Trop Med Hyg. 1987;18:292297.
503534. 303. Soares CN, Faria LC, Peralta JM, et al. Dengue infection: neurologi-
270. Southern PM Jr, Smith JW, Luby JP, et al. Clinical and laboratory features cal manifestations and cerebrospinal fluid (CSF) analysis. J Neurol Sci.
of epidemic St. Louis encephalitis. Ann Intern Med. 1969;71:681689. 2006;249:1924.
271. Wasay M, Diaz-Arrastia R, Suss RA, et al. St Louis encephalitis: a review of 304. Puccioni-Sohler M, Soares CN, Papaiz-Alvarenga R, et al. Neurologic den-
11 cases in a 1995 Dallas, Tex, epidemic. Arch Neurol. 2000;57:114118. gue manifestations associated with intrathecal specific immune response.
272. Monath TP, Nystrom RR, Bailey RE, et al. Immunoglobulin M antibody Neurology. 2009;73:14131417.
capture enzyme-linked immunosorbent assay for diagnosis of St. Louis 305. Arajo FMC, Brilhante RSN, Cavalcanti LPG, et al. Detection of the den-
encephalitis. J Clin Microbiol. 1984:20:784790. gue non-structural 1 antigen in cerebral spinal fluid samples using a com-
273. Breinl A. Clinical pathological and experimental observations on the mercially available enzyme-linked immunosorbent assay. J Virol Methods.
mysterious disease, a clinically abberant form of poliomyelitis. Med J 2011;177:128131.
Aust. 1918;1:209229. 306. Arajo FMC, Arajo MS, Nogueira RM, et al. Central nervous system
274. French EL. Murray Valley encephalitis: isolation and characterization of involvement in dengue: a study in fatal cases from a dengue endemic area.
the aetiological agent. Med J Aust. 1952;1:100103. Neurology. 2012;78:736742.
275. Mackenzie JS, Broom AK, Hall RA, et al. Arboviruses in the Australian 307. Solomon T, Barrett AD. Dengue. In: Nath A, Berger J, eds. Neurovirology.
region, 1990 to 1998. Commun Dis Intell. 1998;22:93100. New York: Marcel Decker; 2003.
276. Burrow JN, Whelan PI, Kilburn CJ, et al. Australian encephalitis in the 308. Varatharaj A. Encephalitis in the clinical spectrum of dengue infection.
Northern Territory: clinical and epidemiological features, 19871996. Neurol India. 2010;58:585591.
Aust N Z J Med. 1998;28:590596. 309. Wang E, Ni H, Xu R, et al. Evolutionary relationships of endemic/
277. Knox J, Cowan RU, Doyle JS, et al. Murray Valley encephalitis: a review of epidemic and sylvatic dengue viruses. J Virol. 2000;74:32273234.
clinical features, diagnosis and treatment. Med J Aust. 2012;196:322326. 310. Halstead SB. Global epidemiology of dengue hemorrhagic fever. Southeast
278. Kienzle N, Boyes L. Murray Valley encephalitis: case report and review of Asian J Trop Med Public Health. 1990;21:636641.
neuroradiological features. Australas Radiol. 2003;47:6163. 311. Medlock JM, Hansford KM, Schaffner F, et al. A review of the invasive
279. Lobigs M, Larena M, Alsharifi M, et al. Live chimeric and inactivated mosquitoes in Europe: ecology, public health risks, and control options.
Japanese encephalitis virus vaccines differ in their cross-protective values Vector Borne Zoonotic Dis. 2012;12:435447.
against Murray Valley encephalitis virus. J Virol. 2009;83:24362445. 312. Adalja AA, Sell TK, Bouri N, et al. Lessons learned during dengue out-
280. Smorodintsev AA. Tick-borne spring-summer encephalitis. Progr Med breaks in the United States, 20012011. Emerg Infect Dis. 2012;18:
Virol. 1958;1:210247. 608614.
281. Sss J. Tick-borne encephalitis 2010: epidemiology, risk areas, and virus 313. Lum LC, Lam SK, Choy YS, et al. Dengue encephalitis: a true entity? Am
strains in Europe and Asiaan overview. Ticks Tick Borne Dis. 2011;2:215. J Trop Med Hyg. 1996;54:256259.
282. Dobler G, Gniel D, Petermann R, et al. Epidemiology and distribution of 314. Hommel D, Talarmin A, Deubel V, et al. Dengue encephalitis in French
tick-borne encephalitis. Wien Med Wochenschr. 2012;162:230238. Guiana. Res Virol. 1998;149:235238.
283. Kaiser R. The clinical and epidemiological profile of tick-borne encepha- 315. Chokephaibulkit K, Kankirawatana P, Apintanapong S, et al. Viral eti-
litis in southern Germany 1994-98: a prospective study of 656 patients. ologies of encephalitis in Thai children. Pediatr Infect Dis J. 2001;20:
Brain. 1999;122:20672078. 216218.
284. Gunther G, Haglund M, Lindquist L, et al. Tick-bone encephalitis in Sweden 316. Pancharoen C, Thisyakorn U. Neurological manifestations in dengue
in relation to aseptic meningo-encephalitis of other etiology: a prospective patients. Southeast Asian J Trop Med Public Health. 2001;32:341345.
study of clinical course and outcome. J Neurol. 1997;244:230238. 317. Familusi JB, Moore DL, Fomufod AK, et al. Virus isolates from children
285. Haglund M, Gunther G. Tick-borne encephalitis-pathogenesis, clinical with febrile convulsions in Nigeria. A correlation study of clinical and
course and long-term follow-up. Vaccine. 2003;21(suppl 1):S11S18. laboratory observations. Clin Pediatr (Phila). 1972;11:272276.
286. Hofmann H, Frisch-Niggemeyer W, and Heinz F. Rapid diagnosis of tick- 318. George R, Liam CK, Chua CT, et al. Unusual clinical manifestations
borne encephalitis by means of enzyme linked immunosorbent assay. of dengue virus infection. Southeast Asian J Trop Med Public Health.
J Gen Virol. 1979;42:505511. 1988;19:585590.
287. Schultze D, Dollenmaier G, Rohner A, et al. Benefit of detecting tick- 319. Thakare J, Walhekar B, Banerjee K. Hemorrhagic manifestations and
borne encephalitis viremia in the first phase of illness. J Clin Virol. 2007; encephalopathy in cases of dengue in India. Southeast Asian J Trop Med
38:172175. Public Health. 1996; 27(3):471475.
288. Chiba N, Osada M, Komoro K, et al. Protection against tick-borne 320. Chimelli L, Hahn MD, Netto MB, et al. Dengue: neuropathological find-
encephalitis virus isolated in Japan by active and passive immunization. ings in 5 fatal cases from Brazil. Clin Neuropathol. 1990;9:157162.
Vaccine. 1999;17:15321539. 321. Misra UK, Kalita J, Syam UK, et al. Neurological manifestations of den-
289. Heinz FX, Kunz C, Fauma H. Preparation of a highly purified vaccine gue virus infection. J Neurol Sci. 2006;244:117122.
against tick-borne encephalitis by continuous flow zonal ultracentrifugation. 322. Hira HS, Kaur A, Shukla A. Acute neuromuscular weakness associated
J Med Virol. 1980;6:213221. with dengue infection. J Neurosci Rural Pract. 2012;3:3639.
290. Kunz C. Vaccination against TBE in Austria: the success story continues. 323. Bethell DB, Gamble J, Pham PL, et al. Noninvasive measurement of mi-
Int J Med Microbiol. 2002;291(suppl 33):5657. crovascular leakage in patients with dengue hemorrhagic fever. Clin Infect
291. Zent O, Hennig R. Post-marketing surveillance of immediate allergic Dis. 2001;32:243253.
reactions: polygeline-based versus polygeline-free pediatric TBE vaccine. 324. Cam BV, Fonsmark L, Hue NB, et al. Prospective case-control study of
Vaccine. 2004;23:579584. encephalopathy in children with dengue hemorrhagic fever. Am J Trop
292. Girgsdies OE, Rosenkranz G. Tick-borne encephalitis: development of Med Hyg. 2001;65:848851.
a paediatric vaccine. A controlled, randomized, double-blind and multi- 325. World Health Organization. Dengue Hemorrhagic Fever: Diagnosis,
centre study. Vaccine. 1996;14:14211428. Treatment and Control. 2nd ed. Geneva: World Health Organization; 1997.
293. Kollaritsch H, Paulke-Korinek M, Holzmann H, et al. Vaccines and vac- 326. Webster DP, Farrar J, Rowland-Jones S. Progress towards a dengue vaccine.
cination against tick-borne encephalitis. Expert Rev Vaccines. 2012;11: Lancet Infect Dis. 2009;9:678687.
11031119. 327. Schmitz J, Roehrig J, Barrett A, et al. Next generation dengue vaccines:
294. Rendi-Wagner P. Advances in vaccination against tick-borne encephalitis. a review of candidates in preclinical development. Vaccine. 2011;29:
Expert Rev Vaccines. 2008;7:589596. 72767284.
295. World Health Organization. Vaccines against tick-borne encephalitis: 328. Laughlin CA, Morens DM, Cassetti MC, et al. Dengue research opportu-
WHO position paperrecommendations. Vaccine. 2011;29:87698770. nities in the Americas. J Infect Dis. 2012;206:11211127.
296. Solomon T, Mallewa MJ. Dengue and other emerging flaviviruses. 329. Heinz FX, Stiasny K. Flaviviruses and flavivirus vaccines. Vaccine.
J Infect. 2001;42:104115. 2012;30:43014306.
297. Gubler DJ, Kuno G, Wareman SH. Neurological disorders associated with 330. Rigau-Perez JG, Clark GG, Gubler DJ, et al. Dengue and dengue haemor-
dengue fever infection. In: Proceedings of the International Conference on rhagic fever. Lancet. 1998;352:971977.
Dengue/DHF; 1983; Kuala Lumpur, Malaysia. 331. Monath TP. Epidemiology of yellow fever: current status and speculations
298. Thisyakorn U, Thisyakorn C, Limpitikul W, et al. Dengue infection with on future trends. In: Saluzzo J-F, Dodet B, eds. Factors in the Emergence
central nervous system manifestations. Southeast Asian J Trop Med Public of Arbovirus Diseases. Paris: Elsevier; 1997:143158.
Health. 1999;30:504506. 332. Gubler DJ. The global emergence/resurgence of arboviral diseases as pub-
299. Kankirawatana P, Chokephaibulkit K, Puthavathana P, et al. Dengue lic health problems. Arch Med Res. 2002;33:330342.
infection presenting with central nervous system manifestation. J Child 333. Nene V, Wortman JR, Lawson D, et al. Genome sequence of Aedes
Neurol. 2000;15:544547. aegypti, a major arbovirus vector. Science. 2007;316:17181723.
300. Solomon T, Dung NM, Vaughn DW, et al. Neurological manifestations of 334. Schmaljohn CS, Hooper JW. Bunyaviridae: the viruses and their repli-
dengue infection. Lancet. 2000;355:10531059. cation. In: Knipe DM, Howley PM, eds. Fields Virology. Philadelphia:
301. Nimmannitya S, Thisyakorn U, Hemsrichart V. Dengue haemorrhagic Lippincott Williams & Wilkins; 2001:15811602.
fever with unusual manifestations. Southeast Asian J Trop Med Pub 335. Guu TS, Zheng W, Tao YJ. Bunyavirus: structure and replication. Adv
Health. 1987;18:398406. Exp Med Biol. 2012;726:245266.

Scheld_Ch15.indd 237 2/21/14 5:38 PM


238 Part II: Viral Infections and Related Disorders

336. Walter CT, Barr JN. Recent advances in the molecular and cellular biology 364. Nicoletti L, Verani P, Caciolli S, et al. Central nervous system involve-
of bunyaviruses. J Gen Virol. 2011;92:24672484. ment during infection by Phlebovirus toscana of residents in natural
337. Gavrilovskaya IN, Shepley M, Shaw R, et al. 3 integrins mediate the cel- foci in central Italy (19771988). Am J Trop Med Hyg. 1991;45:
lular entry of hantaviruses that cause respiratory failure. Proc Natl Acad 429434.
Sci U S A. 1998;95:70747079. 365. Schwarz TF, Gilch S, Jager G. Aseptic meningitis caused by sandfly fever
338. Lozach PY, Kuhbacher A, Meier R, et al. DC-SIGN as a receptor for virus, serotype Toscana. Clin Infect Dis. 1995;21:669671.
phleboviruses. Cell Host Microbe. 2011;10:7588. 366. Braito A, Ciufolini MG, Pippi L, et al. Phlebotomus-transmitted toscana
339. Lozach P-Y, Mancini R, Bitto D, et al. Entry of bunyaviruses into mam- virus infections of the central nervous system: a seven-year experience in
malian cells. Cell Host Microbe. 2010;7:488499. Tuscany. Scand J Infect Dis. 1998;30:505508.
340. Hammon WM, Reeves WC. California encephalitis virus, a newly de- 367. Dionisio D, Valassina M, Ciufolini MG, et al. Encephalitis without
scribed agent. Calif Med. 1952;77:303309. meningitis due to sandfly fever virus serotype toscana. Clin Infect Dis.
341. Eldridge BF, Glaser C, Pedrin RE, et al. The first reported case of California 2001;32:12411243.
encephalitis in more than 50 years. Emerg Infect Dis. 2001;7:451452. 368. Valassina M, Valentini M, Valensin PE, et al. Fast duplex one-step RT-
342. Thompson W, Kalfayan B, Anslow R. Isolation of California encephalitis PCR for rapid differential diagnosis of entero- or toscana virus meningitis.
group virus from a fatal human illness. Am J Epidemiol. 1965;81:245253. Diagn Microbiol Infect Dis. 2002;43:201205.
343. Erwin PC, Jones TF, Gerhardt RR, et al. La Crosse encephalitis in Eastern 369. Cusi MG, Savellini GG. Diagnostic tools for Toscana virus infection.
Tennessee: clinical, environmental, and entomological characteristics Expert Rev Anti Infect Ther. 2011;9:799805.
from a blinded cohort study. Am J Epidemiol. 2002;155:10601065. 370. Valassina M, Cusi MG, Valensin PE. Rapid identification of Toscana
344. Watts DM, Thompson WH, Yuill TM, et al. Overwintering of La Crosse virus by nested PCR during an outbreak in the Siena area of Italy. J Clin
virus in Aedes triseriatus. Am J Trop Med Hyg. 1974;23:694700. Microbiol. 1996;34:25002502.
345. Demikhov VG, Chaitsev VG, Butenko AM, et al. California serogroup 371. Ciufolini MG, Fiorentini C, di Bonito P, et al. Detection of Toscana virus-
virus infections in the Ryazan region of the USSR. Am J Trop Med Hyg. specific immunoglobulins G and M by an enzyme-linked immunosor-
1991;45:371376. bent assay based on recombinant viral nucleoprotein. J Clin Microbiol.
346. Lundstrom JO. Mosquito-borne viruses in western Europe: a review. 1999;37:20102012.
J Vector Ecol. 1999;24:139. 372. Centers for Disease Control and Prevention. Outbreak of Rift Valley
347. Kappus KD, Monath TP, Kaminski RM, et al. Reported encephalitis feverYemen, August-October 2000. MMWR Morb Mortal Wkly Rep.
associated with California serogroup virus infections in the United States, 2000;49:10651066.
19631981. Prog Clin Biol Res. 1983;123:3141. 373. Laughlin LW, Meegan JM, Strausbaugh LJ, et al. Epidemic Rift Valley
348. Centers for Disease Control and Prevention. West nile virus disease and fever in Egypt: observations of the spectrum of human illness. Trans R Soc
other arboviral diseasesUnited States, 2011. MMWR Morb Mortal Trop Med Hyg. 1979;73:630633.
Wkly Rep. 2012;61:510514. 374. Maar SA, Swanepoel R, Gelfand M. Rift Valley fever encephalitis. A descrip-
349. Elliott RM. Bunyaviruses and climate change. Clin Microbiol Infect. tion of a case. Cent Afr J Med. 1979;25:811.
2009;15:510517. 375. Peters CJ, Anderson GW Jr. Pathogenesis of Rift Valley fever. Contrib
350. Mayo D, Karabatsos N, Scarano FJ, et al. Jamestown Canyon virus: sero- Epidemiol Biostat. 1981;3:2141.
prevalence in Connecticut. Emerg Infect Dis. 2001;7:911912. 376. Bird BH, Nichol ST. Breaking the chain: Rift Valley fever virus control via
351. Balkhy HH, Schreiber JR. Severe La Crosse encephalitis with significant livestock vaccination. Curr Opin Virol. 2012;2:315323.
neurologic sequelae. Pediatr Infect Dis J. 2000;19:7780. 377. Rehse-Kpper B, Casals J, Rehse E, et al. Eyachan arthropod-borne
352. Cramblett HG, Stegmiller H, Spencer C. California encephalitis virus related to Colorado tick fever virus in the Federal Republic of
virus infections in children. Clinical and laboratory studies. JAMA. Germany. Acta Virol. 1976;20:339342.
1966;198:108112. 378. Xu P, Wang Y, Zuo J, et al. New orbiviruses isolated from patients
353. Balfour HH Jr, Siem RA, Bauer H, et al. California arbovirus (La Crosse) with unknown fever and encephalitis in Yunnan province. Chin J Virol.
infections. I. Clinical and laboratory findings in 66 children with menin- 1990;6:2733.
goencephalitis. Pediatrics. 1973;52:680691. 379. Song L, Chen B, Zhao Z. Isolation and identification of new members
354. Grimstad PR, Shabino CL, Calisher CH, et al. A case of encephalitis in a of coltivirus from mosquitoes collected in China. Chin J Exp Clin Virol.
human associated with a serologic rise to Jamestown Canyon virus. Am J 1995;9:710.
Trop Med Hyg. 1982;31:12381244. 380. Attoui H, Billoir F, Biagini P, et al. Complete sequence determination and
355. McJunkin JE, Nahata MC, De Los Reyes EC, et al. Safety and pharma- genetic analysis of Banna virus and Kadipiro virus: proposal for assign-
cokinetics of ribavirin for the treatment of la crosse encephalitis. Pediatr ment to a new genus (Seadornavirus) within the family Reoviridae. J Gen
Infect Dis J. 2011;30:860865. Virol. 2000;81:15071515.
356. Sabin AB. Phlebotomus fever. In: Rivers TM, ed. Viral and Rickettsial 381. Attoui H, Mohd Jaafar F, Biagini P, et al. Genus Coltivirus (family
Diseases of Man. Philadelphia: JB Lippincott; 1948:454460. Reoviridae): genomic and morphologic characterization of Old World
357. Konstantinou GN, Papa A, Antoniadis A. Sandfly-fever outbreak in and New World viruses. Arch Virol. 2002;147:533561.
Cyprus: are phleboviruses still a health problem? Travel Med Infect Dis. 382. Knudson DL. Genome of Colorado tick fever virus. Virology. 1981;112:
2007;5:239242. 361364.
358. Verani P, Nicoletti L, Ciufolini MG. Antigenic and biological character- 383. Goodpasture HC, Poland JD, Francy DB, et al. Colorado tick fever: clini-
ization of Toscana virus, a new Phlebotomus fever group virus isolated in cal, epidemiologic, and laboratory aspects of 228 cases in Colorado in
Italy. Acta Virol. 1984;28:3947. 19731974. Ann Intern Med. 1978;88:303310.
359. Nicoletti L, Ciufolini MG, Verani P. Sandfly fever viruses in Italy. Arch 384. Bowen GS. Colorado tick fever. In: Monath TP, ed. The Arboviruses:
Virol Suppl. 1996;11:4147. Ecology and Epidemiology. Boca Raton, FL: CRC Press Inc; 1988:
360. Braito A, Corbisiero R, Corradini S, et al. Toscana virus infections of the central 159176.
nervous system in children: a report of 14 cases. J Pediatr. 1998;132:144148. 385. Philipp CS, Callaway C, Chu MC, et al. Replication of Colorado tick
361. Eitrem R, Stylianou M, Niklasson B. High prevalence rates of antibody fever virus within human hematopoietic progenitor cells. J Virol.
to three sandfly fever viruses (Sicilian, Naples, and Toscana) among 1993;67:23892395.
Cypriots. Epidemiol Infect. 1991;107:685691. 386. Hierholzer WJ, Barry DW. Colorado tick fever pericarditis. JAMA. 1971;
362. Depaquit J, Grandadam M, Fouque F, et al. Arthropod-borne viruses 217:825.
transmitted by Phlebotomine sandflies in Europe: a review. Euro Surveill. 387. Spruance SL, Bailey A. Colorado tick fever. A review of 115 laboratory
2010;15:19507. confirmed cases. Arch Intern Med. 1973;131:288293.
363. Mendoza-Montero J, Gmez-Rueda MI, Navarro-Mari JM, et al. 388. Smee DF, Sidwell RW, Clark SM, et al. Inhibition of bluetongue and
Infections due to sandfly fever virus serotype Toscana in Spain. Clin Infect Colorado tick fever orbiviruses by selected antiviral substances. Antimicrob
Dis. 1998;27:434436. Agents Chemother. 1981;20:533538.

Scheld_Ch15.indd 238 2/21/14 5:38 PM


CHAPTER 16 MENINGITIS AND ENCEPHALITIS
CAUSED BY MUMPS VIRUS
JOHN W. GNANN, JR.

Mumps is an acute systemic human infection caused by a the Centers for Disease Control and Preventions Advisory
paramyxovirus. Mumps virus is highly transmissible and, in Committee on Immunization Practices recommended that all
unimmunized populations, causes epidemics of mumps among children receive a second dose of the measles-mumps-rubella
school-aged children. Although salivary gland enlargement, (MMR) vaccine at the time of school entry, which reduced the
especially parotitis, is the most readily recognized clinical rate of primary vaccine failure (4). The incidence of mumps in
manifestation of mumps, the infection involves many other the United States declined steadily after 1987; only 387 cases
organs, including the central nervous system (CNS). In the were reported in 1999 (7). By testing sera from the 1999 to
past, nonsuppurative parotitis was considered the sine qua 2004 U.S. National Health and Nutrition Examination Survey
non of mumps, and involvement of any other organ system (NHANES), the overall age-adjusted seroprevalence of immu-
was viewed as a complication. However, studies conducted noglobulin G (IgG) to mumps virus was shown to be 90% (8).
over the last 60 years have made it clear that CNS involve- However, a series of outbreaks in the United States and
ment during mumps occurs with such high frequency that it Canada in 2005 to 2006 reemphasized the potential for
should be considered a part of the natural history of the infec- mumps virus to cause localized epidemics even in highly vac-
tion and not as an aberrant manifestation or complication. cinated populations (9,10). In 2006, a total of 6,584 cases of
Indeed, some virologists have classified mumps virus as pri- mumps were reported, with a national incidence of 2.2 cases
marily neurotropic. per 100,000 population (9). The U.S. outbreak was centered
The spectrum of CNS diseases associated with mumps in the Midwest and peaked in April 2006; 34% of the cases
ranges from mild aseptic meningitis, which is very common, to occurred in Iowa. The highest age-specific age rate was in per-
fulminant and potentially fatal encephalitis, which is very rare. sons aged 18 to 24 years (median age 22 years), many of whom
The literature describing CNS involvement with mumps can be were college students (11). Among the Iowa cases, 7% were
difficult to assess critically because some authors have grouped unvaccinated; 14% had received one dose of MMR; 49% had
all cases of mumps neurologic disease under the label mumps received two or more doses of MMR; and 30% had unknown
meningoencephalitis. Although this term is convenient, its vaccine status. Although some cases can be attributed to fail-
use obscures the fact that the clinical course and prognosis ure to vaccinate, most represented primary (insufficient initial
of mumps aseptic meningitis differs markedly from that of immune response) or secondary (waning immune response)
mumps encephalitis, although there is considerable overlap vaccine failure (12). These findings suggest the need for a
between the two syndromes. It is important for the clinician to mumps vaccine with a longer duration of protection or modifi-
establish whether an individual patient with mumps has clear cation in vaccination policy, perhaps focusing on young adults
evidence of encephalitis because that diagnosis has important (9,13). In selected outbreaks, a third dose of mumps vaccine
implications for management and prognosis. has been administered for outbreak control, which appears to
be a safe and effective measure (14,15).
The importance of immunization of adolescents and young
EPIDEMIOLOGY adults was emphasized by a large outbreak (over 56,000 re-
ported cases) of mumps in the United Kingdom in 2004 to
In unvaccinated urban populations, mumps (or epidemic par- 2005 (16,17). Many of the cases occurred in university students
otitis) is a disease of school-aged children with a worldwide who were too old to have been vaccinated in childhood but too
distribution. Mumps infrequently occurs in infants younger young to have been exposed to epidemic natural infection (18).
than 1 year of age, presumably because of transplacentally ac- Conversely, a large mumps outbreak (3,500 cases) occurred
quired antibody. Most mumps cases occur in children between in 2009 to 2010 in New York and New Jersey, causing disease
4 and 7 years of age (1). By age 15 years, over 90% of children primarily among adolescent boys attending Orthodox Jewish
have antibodies against mumps virus. schools (19). Mumps transmission in this highly vaccinated
Prior to the release of the live attenuated mumps vaccine in population was apparently facilitated by the close face-to-face
the United States in 1967, mumps was an endemic disease with interactions among the students. The propensity for mumps to
a seasonal peak of activity occurring between January and May spread among vaccinated adolescents in schools and commu-
(2). The largest number of cases reported in the United States nal living environments raises the question of whether a revised
occurred in 1941, when the incidence of mumps was 250 cases vaccination schedule should be considered (2022).
per 100,000 population (3). In 1968, when the live attenu- In unimmunized populations, mumps is one of the most
ated vaccine was first being put to clinical use, the incidence of common causes of aseptic meningitis and encephalitis (23). In a
mumps was 76 cases per 100,000 population. In 1985, a total survey conducted in Minnesota from 1950 to 1981, mumps
of only 2,982 cases of mumps were reported, an incidence of was identified as the cause of encephalitis in 6 of 189 cases and
1.1 cases per 100,000 population, representing a 98% decline the cause of aseptic meningitis in 7 of 283 cases (24). In that
from the 185,691 cases reported in 1967 (4). Sporadic out- series, mumps was the second most common cause of both
breaks of mumps in secondary schools occurring in the United encephalitis and aseptic meningitis (behind California virus
States were attributed to primary vaccine failure (5). Between and enteroviruses, respectively). Meyer et al. (25) reviewed
1988 and 1993, 36% of all mumps cases in the United States 713 cases of encephalitis and aseptic meningitis treated at U.S.
were in patients older than 15 years of age (6). In 1989, military hospitals between 1953 and 1958. In those patients in

239

Scheld_Ch16.indd 239 2/21/14 5:38 PM


240 Part II: Viral Infections and Related Disorders

whom a specific etiology was established, mumps was identi- shares some serologic cross reactivity with other paramyxovi-
fied in 91 cases, poliomyelitis in 156, coxsackievirus B in 80, ruses, especially parainfluenza viruses, which can complicate
lymphocytic choriomeningitis virus in 58, and echovirus in 53. interpretation of serodiagnostic assays (48).
In an analysis of 191 cases of acute encephalitis occurring in Mumps virions are pleomorphic, irregularly spherical, en-
adults in Finland between 1967 and 1978, mumps virus was veloped particles with an average diameter of about 200 nm.
found to be the second most common etiologic agent (following Glycoprotein spikes project from the outer surface of the lipid
herpes simplex virus), accounting for 6.8% of the cases (26). envelope, which encloses a helical nucleocapsid composed of
The frequency with which CNS manifestations occur during RNA and nucleoproteins. The mumps virus genome consists
acute mumps has varied tremendously (from 1% to 70%) of a linear molecule of single-stranded, negative-sense RNA
among published series (27,28). A more realistic estimate of approximately 15.3 kilobases in size that encodes seven major
the frequency of symptomatic CNS involvement during mumps proteins and several minor proteins. The three proteins con-
appears to be 10% to 30%. Potential causes for this variability tained in the ribonucleoprotein complex are the nucleocapsid
in reported incidence include the interest and skill of the ob- protein (NP), which is the major structural protein of mumps
server, the population studied (e.g., schoolchildren vs. military virus, plus the phosphoprotein (P) and large (L) protein, which
recruits, outpatients vs. hospitalized patients), the case defini- are thought to function as the RNA-dependent RNA poly-
tions used, and the frequency of use of lumbar puncture for merase (49). Full-length transcription of the V/P gene yields
diagnosis. Symptomatic encephalitis is observed much less fre- the V protein (also known as NS1), whereas RNA editing dur-
quently than aseptic meningitis, probably occurring in fewer ing the transcription of V/P produces the phosphoprotein as
than 0.1% of cases of acute mumps (2931). Cerebrospinal well as the nonstructural I protein (also known as NS2) (50).
fluid (CSF) pleocytosis occurs in 40% to 60% of patients with The V protein blocks host antiviral responses by inhibiting the
acute mumps, although only 10% to 30% of patients with interferon signal transduction pathway and interleukin (IL)-6
mumps have clinical evidence of meningeal irritation. That is, expression and may serve as a virulence factor (5153). The
about half of the patients with mumps with CSF pleocytosis function of the I protein is not known. The envelope contains
do not have CNS symptoms (28,32). Bang and Bang (27) per- the matrix (M) protein and the two surface glycoproteins that
formed lumbar punctures on 371 patients with mumps paroti- mediate hemagglutinin-neuraminidase (HN) and fusion (F)
tis and found that 235 (63%) had elevated CSF white blood activities. The HN glycoprotein is responsible for attachment
cell counts; of these 235 patients, 129 showed no clinical evi- of mumps virus to the receptors on the host cell, and the F
dence of meningitis or encephalitis. Similarly, Finkelstein (33) glycoprotein induces the fusion of lipid membranes neces-
found elevated CSF white blood cell counts in 16 of 40 patients sary for penetration of the virus nucleocapsid into the cell. In
(40%) with mumps parotitis; 6 of these had no clinical evi- traditional complement fixation (CF) assays, HN and NP are
dence of CNS involvement. Clearly, mumps CNS disease can known as V antigen and S antigen, respectively.
also occur in patients without evidence of parotitis; indeed, The function of the membrane-associated protein encoded
40% to 50% of patients with mumps meningitis have no evi- by the small hydrophobic (SH) gene has not been fully defined,
dence of salivary gland enlargement (34,35). If we accept that although it does not appear to be essential for viral replication
CSF pleocytosis occurring during acute mumps is indicative of (54). Characterization of the genetically divergent SH region
CNS infection, then we must conclude that CNS involvement is a useful tool for distinguishing wild type and vaccine strains
during mumps is quite common and frequently asymptomatic. of mumps virus (55). Furthermore, studies employing reverse
Although male and female patients have the same risk for transcriptase polymerase chain reaction (RT-PCR) methodol-
mumps parotitis, there is a distinct male predominance (70% ogy have demonstrated 13 distinct viral genotypes (designated
to 80%) with respect to development of CNS disease among A to M) based on sequence of the SH gene (5658). These
children with mumps. In virtually every published series, the molecular tools will allow more precise studies of mumps
ratio of boys to girls is between 3:1 and 4:1 (28,3032,3639). transmission and population genetics (5962). In the Western
This striking difference in the incidence of mumps CNS disease hemisphere, genotypes A, C, D, E, and H are more common,
between the sexes has not been satisfactorily explained. Among whereas genotypes B, F, and I predominate in Asia. Although
young adults with mumps and CNS involvement, the ratio of there is only a single mumps serotype, some investigators have
men to women is closer to 1:1 (39). The peak incidence of CNS found that cross-neutralization between mumps virus geno-
involvement in mumps occurs at about age 7 years in both types is reduced (63,64). Some studies have suggested that
sexes (31,39,40). Of all cases, 60% to 70% occur in children infection with certain genotypes (e.g., C, D, H, or J) may result
between 5 and 9 years of age (28,30,37,38,41). Several authors in enhanced neuropathogenicity (60,65), but this observation
have reported seasonal differences in the frequency of mumps was not confirmed by other investigators (58).
meningoencephalitis, with and without parotitis (34,35,4044). Humans are the only known natural hosts for mumps virus,
although infection can be experimentally induced in a variety
of mammalian species. In vitro, mumps virus can be cultured
INFECTIOUS AGENT in many mammalian cell lines, including primary Rhesus
monkey kidney, human embryonic kidney, BSC-1, Vero, and
Mumps virus is classified as a member of the family HeLa cells, as well as in embryonated hens eggs (66). In tis-
Paramyxoviridae and the genus Rubulavirus (45). In gen- sue culture, the cytopathic effects caused by mumps virus
eral, members of the family Paramyxoviridae possess nonseg- appear within 3 to 6 days and are highly variable. They in-
mented RNA genomes, bind to neuraminic acid receptors on clude development of rounded cells, formation of eosinophilic
host cells, replicate in the cytoplasm, are inactivated by ether, intracytoplasmic inclusions, and fusion of cells to form giant
and cause respiratory infections in humans (46). Serologic multinucleated syncytia. Guinea pig or chick erythrocytes will
testing with hemagglutination inhibition (HI) or neutraliza- adhere to mumps virusinfected cells, a phenomenon medi-
tion assays has identified only one serotype of mumps virus, ated by hemagglutinins present on the surface of infected cells.
although minor antigenic differences among mumps virus Traditionally, verification of the isolate as mumps virus has been
isolates can be detected using panels of glycoprotein-specific accomplished by a hemadsorption inhibition assay, in which
monoclonal antibodies (47). Infection can be prevented by mumps virusspecific antiserum is used to block the adherence
vaccination with a single strain of mumps virus. Mumps virus of erythrocytes to mumps-infected cells. Most laboratories

Scheld_Ch16.indd 240 2/21/14 5:38 PM


Chapter 16: Meningitis and Encephalitis Caused by Mumps Virus 241

now use direct or indirect immunofluorescence methods to cerebellum and in the white and gray matter of the brainstem
identify mumps virus in tissue culture (66). Molecular methods and spinal cord. Because of the pattern of perivascular demy-
based on PCR have also been described (67). elinization, Donohue (82) and Schwarz et al. (83) suggested
that these findings were more suggestive of a postinfectious or
parainfectious encephalopathy rather than of tissue destruction
PATHOGENESIS AND caused directly by mumps virus. Conversely, other authors have
PATHOPHYSIOLOGY interpreted the findings of cellular destruction as consistent
with primary mumps virus cytopathic effect (85). In most cases
Mumps is highly contagious, although some studies have sug- of fatal mumps encephalitis that have been carefully studied,
gested that it is less contagious than varicella or measles (68). there has been histologic evidence of both cellular destruction
This clinical observation may be skewed by the fact that up to (suggestive of direct virus effect) and demyelinization (sugges-
30% of all mumps infections are subclinical and asymptom- tive of an autoimmune process). The pathogenesis of mumps
atic (69). Over 90% of adults who give negative histories for encephalitis remains incompletely understood.
mumps are, in fact, seropositive when tested for mumps anti- Investigations using the hamster model have made major con-
bodies, indicating prior subclinical infection (70). tributions to our understanding of the pathogenesis of mumps
Mumps can be experimentally transmitted to humans by in- CNS disease. CNS mumps infections have been produced exper-
oculation of virus onto the nasal or buccal mucosa, suggesting imentally by the intracerebral or intraperitoneal inoculation of
that most natural infections result from droplet spread of upper newborn hamsters with a neuroadapted strain of mumps virus
respiratory secretions from infected to susceptible individuals. (77,86). About 9 to 12 days after intracerebral inoculation with
Virus can be isolated from saliva for 5 to 7 days before and 5 mumps virus, suckling hamsters developed wasting, ruffled hair,
to 8 days after the onset of clinical symptoms, meaning that an and arched backs; they usually died within 24 hours. Maximum
infected individual is potentially able to transmit mumps for a titers of mumps virus were detected in hamster brains at 5 days,
period of up to 2 weeks (71,72). The average incubation period and the virus titer declined as neutralizing antibodies appeared.
for mumps is about 18 days (range 14 to 28 days) (73,74). The clinical symptoms occurred 3 to 5 days after the decline
During this interval, primary viral replication is thought to take in virus titers and the concurrent development of neutralizing
place in epithelial cells of the upper respiratory tract, followed antibody (86). During the first week after virus inoculation, the
by spread of virus to regional lymph nodes and then viremic neuropathologic appearance of the brain was characterized by
dissemination to glandular and neural tissue (75,76). intense perivascular mononuclear cell infiltrates. During the sec-
Mumps virus is delivered to the CNS either via free plasma ond week, microglial cell proliferation and small areas of necro-
viremia or by infected host mononuclear cells (77,78). Virus is sis were observed. Notably, no foci of demyelinization (as has
thought to spread across the endothelium of the choroid plexus been observed in human brains) were apparent.
and to infect choroidal epithelial cells. Replication of mumps Immunofluorescence studies of suckling hamster brains fol-
virus then takes place in the choroidal epithelium, and progeny lowing intracerebral inoculation with mumps virus showed that
virus is shed into the CSF. In support of this model is the observa- mumps virus antigens were present in endothelial cells of the cho-
tion that mumps virus can be easily recovered from CSF during roid plexus, in ependymal cells, and in neurons of the brainstem,
the early phases of mumps meningitis in humans. Additionally, hippocampus, and cerebral cortex. Most of the infected neurons
choroidal and ependymal epithelial cells containing mumps an- were morphologically normal, and there was little correlation be-
tigens can be recovered from the CSF of patients with mumps tween the major sites of neuronal infection and the observed sites
meningoencephalitis (79). Replication of mumps virus in the of perivascular inflammation (86). The histologic appearance
choroid plexus and ependyma (the tissue that lines the cerebral of mumps encephalitis in suckling hamsters is more consistent
ventricles and covers the choroid plexus) has been demonstrated with direct virus-induced pathology than with immunologically
in rodent (77) and primate (80) models of mumps CNS infec- mediated pathologic changes. Studies using the suckling hamster
tion. When mumps encephalitis develops, it is presumed that model have also demonstrated an association of mumps CNS
virus replicating in ependymal cells spreads by direct extension infection with the development of stenosis of the aqueduct of
into neurons within the brain parenchyma, as has been observed Sylvius and with granular ependymitis (87). These findings sug-
in the hamster model of mumps encephalitis (81). gest a possible (but unproven) linkage between mumps CNS in-
fections and aqueductal stenosis in children (88,89).
The ability of mumps virus to establish chronic infections
PATHOLOGY OF MUMPS in tissue culture systems is well known (90,91). In the ham-
ster model, cell-associated mumps virus can be recovered
CENTRAL NERVOUS SYSTEM from brain explant cultures for up to 50 days after infection,
INFECTION well after the appearance of specific humoral immunity (77).
In humans with mumps meningoencephalitis, the persistence
Autopsy reports of patients who have died with acute encepha- of leukocytes and oligoclonal mumps-specific immunoglobu-
litis caused by mumps virus have been reviewed by Donohue lins in the CSF for months after the acute infection suggests
(82) and Schwarz et al. (83). Many of these cases occurred the possibility of ongoing antigenic stimulation from chronic
prior to the availability of modern virologic or serologic meth- mumps CNS infection (92,93). Sufficient data do not yet exist
ods for confirmation of the diagnosis, and some were likely to confirm or refute a potential for mumps virus as a cause of
caused by pathogens other than mumps virus. Among those chronic CNS infection in humans, but chronic infection, if it
cases in which mumps virus was the probable cause of the fatal occurs at all, is uncommon (92,94).
encephalitis, neuropathological findings were quite variable
(28,8284). The most commonly recorded features were dif-
fuse edema of the brain, limited mononuclear cell infiltration of IMMUNE RESPONSES
the meninges, perivascular infiltration with mononuclear cells,
glial cell proliferation, focal areas of neuronal cell destruc- Specific humoral and cell-mediated immune responses develop
tion, and localized demyelinization. These histologic changes during the course of acute mumps infection, but the relative
were seen in the white matter of the cerebral hemispheres and contributions of antibody and cellular immunity to viral

Scheld_Ch16.indd 241 2/21/14 5:38 PM


242 Part II: Viral Infections and Related Disorders

clearance have not been precisely determined. Interestingly,


mumps does not often cause unusually severe or prolonged CLINICAL COURSE AND
infections in immunocompromised patients, although severe NATURAL HISTORY
nephritis due to mumps virus has been reported in renal trans-
plant recipients (95). Mumps usually begins with a short prodromal phase charac-
Within 10 days of infection, mumps-specific immunoglobu- terized by low-grade fever, malaise, headache, and anorexia.
lin M (IgM) appears in serum and immunoglobulin A (IgA) Young children may initially complain of ear pain. The patient
can be detected in saliva (96,97). Serum IgM titers begin to then develops the characteristic salivary gland enlargement
wane shortly after the acute illness and are usually undetect- and tenderness (113). The parotid glands are most commonly
able after 6 months (96,98). A mumps-specific IgG response is involved, although other salivary glands (e.g., submandibu-
detectable during the first week of the acute infection, peaks lar and sublingual) may be enlarged in about 10% of cases.
about 3 to 4 weeks after the onset of the infection, and persists Parotitis may initially be unilateral, with swelling of the con-
for decades (99). Using the CF assay, a fourfold rise in mumps- tralateral parotid gland occurring 2 to 3 days later; bilateral
specific IgG can usually be demonstrated within 10 to 14 days parotitis eventually develops in over 80% of patients with
after the onset of the disease (100). Lifelong immunity follows symptomatic salivary gland involvement. Painful parotid gland
natural infection. Symptomatic mumps virus reinfections have enlargement progresses over the course of 2 to 3 days, lifting
been reported, but most patients who report more than one the earlobe outward and obscuring the angle of the mandible
episode of mumps probably had parotitis caused by infection (Fig. 16.1). The orifice of Stensen duct is often red and edema-
with a different pathogen (101). tous. Lymphatic obstruction resulting from bilateral glandular
Mumps-specific immunoglobulins are also detectable enlargement can occasionally result in presternal edema (114).
in CSF of patients with mumps meningoencephalitis (102). Fever and parotid gland swelling peak on about the third
Using a sensitive enzyme-linked immunoadsorbent assay day of the illness, followed by defervescence and resolution
(ELISA) method, mumps-specific IgG was detected in almost of parotid pain and swelling within about 7 days. Long-term
all CSF specimens from patients with mumps meningitis, and sequelae of parotitis are uncommon. Children with mumps
mumps virus IgM was detected in about 50% of patients
(103). By measuring the levels of immunoglobulin in serum
and CSF and then comparing this ratio with that of an index
antibody (such as measles antibody), intrathecal synthesis of
mumps-specific immunoglobulin was shown to occur during
mumps CNS infection. There was no apparent correlation
between the severity of clinical disease and the presence or
absence of mumps IgM in the CSF (104). Furthermore, no
correlation between the CSF leukocyte count and the CSF
mumps antibody titer was demonstrated (104). Infection
(or vaccination) with mumps virus also elicits a cell-mediated
immune response (105). Peripheral blood lymphocytes that
proliferate when stimulated with mumps S and V antigens can
be detected by an in vitro blastogenesis assay following natu-
ral infection or immunization (106). Lymphocytes recovered
from the CSF of patients with mumps meningitis also prolif-
erate when stimulated with mumps antigens (107). Human
leukocyte antigen (HLA)restricted cytotoxic T lymphocytes
(CTLs) are detectable in peripheral blood following mumps
infection or immunization and in CSF of patients with mumps
CNS infection (108). Kreth et al. (109) demonstrated that
T lymphocytes from CSF and from blood from 10 children
with mumps meningitis were cytotoxic to autologous mumps
virusinfected target cells. Lymphocyte-mediated toxicity was
present during the acute phase of mumps meningitis, declined
over 2 to 3 weeks after the onset of symptoms, and was no lon-
ger apparent after 50 days. Fleischer and Kreth (110) cloned
mononuclear cells directly from the CSF of a patient with
mumps meningitis and demonstrated that 90% of the cells
were T lymphocytes and that 60% were CD8 suppressor/
cytotoxic T cells. A high percentage of the T-cell clones
showed specificity for the autologous mumps virusinfected
target cells. In patients with self-limited mumps meningitis,
CSF interferon disappeared within a week, whereas interferon
levels remained elevated in the CSF from those patients who
had persistent CSF pleocytosis (111). These findings suggest
that the recruitment of CTLs into the CNS in mumps menin-
gitis is highly antigen specific and that mumps-specific CTLs
could play a role in the immunopathologic changes observed
in human brains after fatal mumps encephalitis. Recent stud- FIGURE 16.1 Child with mumps demonstrating neck swelling due
ies have also demonstrated increased levels of IL-8, IL-10, to salivary gland enlargement. (Provided by the Centes for Disease
IL-12, IL-13, and interferon (IFN)-gamma in CSF from chil- Control and Prevention [CDC] Public Health Image Library, Patricia
dren with mumps meningitis (112). Smith and Barbara Rice, image #1861.)

Scheld_Ch16.indd 242 2/21/14 5:38 PM


Chapter 16: Meningitis and Encephalitis Caused by Mumps Virus 243

are usually isolated for about 5 days after the appearance of


parotitis, although this practice is of dubious benefit to class- Central Nervous System Manifestation
mates, because the virus is excreted for several days prior to of Mumps
the onset of clinical symptoms (115).
Epididymoorchitis is rare in boys with mumps, but it CNS infection is the most common extrasalivary manifesta-
occurs in 25% of postpubertal men with mumps infection tion of mumps and may precede, accompany, or follow the
(19,116,117). Orchitis is caused by replication of mumps development of parotitis (35). There is no association between
virus in seminiferous tubules, with resulting lymphocytic the severity of the parotitis and the likelihood or severity of
infiltration and edema (118120). Orchitis is most often uni- meningitis (27). Most frequently, CNS symptoms follow the
lateral, but bilateral involvement occurs in 10% to 30% of onset of parotitis by about 5 days (140). In a study of 41 cases
cases (121). Orchitis typically develops within 1 week after of mumps encephalitis reported by Koskiniemi et al. (31),
the onset of parotitis, although orchitis can develop prior to parotitis appeared 3 to 14 days prior to CNS involvement in
or even in the absence of parotitis. Mumps orchitis is char- 15 patients, was coincident with encephalitis in 9 patients,
acterized by marked testicular swelling and severe pain, occurred 1 to 4 days after the onset of CNS symptoms in
accompanied by fever, nausea, and headache (121123). The 2 patients, and was absent in 15 patients. In another series
pain and swelling resolve within 5 to 7 days, although residual of 24 patients with mumps parotitis and meningoencephalitis,
testicular tenderness can persist for weeks. Testicular atrophy 11 patients had parotitis for 6 to 21 days before the onset of
may follow orchitis in about 35% to 50% of cases, but steril- the CNS symptoms, 9 patients had neurologic symptoms pre-
ity is an uncommon complication, even among patients with ceding the parotitis by 1 to 8 days, and 4 patients had parotitis
bilateral orchitis. Orchitis occurring in vaccinated males who and CNS symptoms that occurred simultaneously (42). Levitt
develop breakthrough mumps tends to be less clinically et al. (30) noted a mean interval of 2.7 days between the onset
severe (124). of parotitis and the development of CNS symptoms, but the
Mumps can cause inflammation of other glandular tissues, range was wide, with parotitis developing from 20 days before
including pancreatitis (125) and thyroiditis (126). Investigators to 7 days after CNS disease. These data clearly indicate that
in the 1970s proposed an epidemiologic association between the development of CNS disease in mumps does not depend
mumps and juvenile diabetes mellitus (127); however, the on prior development of parotitis. The physician examining a
dramatic decline in the incidence of mumps has not been patient with suspected meningoencephalitis may not exclude
reflected by a similar decline in the occurrence of juvenile- the possibility of mumps simply because the patient does not
onset diabetes. Oophoritis and mastitis have been reported have clinically apparent salivary gland involvement.
in postpubertal women with mumps (113,128). Renal func- The common presenting signs and symptoms seen in patients
tion abnormalities are common in mumps and virus can be with mumps CNS infection are summarized in Table 16.1. The
readily isolated from urine, but significant or permanent renal most frequently reported presentation is a triad of fever, vom-
damage is rare (129,130). Other infrequent manifestations of iting, and headache. Salivary gland enlargement is present in
mumps include arthritis (131,132), myocarditis (133,134), only 50% of patients with mumps CNS disease. The fever is
and thrombocytopenia (135). frequently high (39C to 40C) and lasts for 72 to 96 hours.
Maternal mumps infection during the first trimester of The headache and vomiting may also be quite severe and usu-
pregnancy results in an increased frequency of spontaneous ally persist for about 48 hours (32,141). Other frequently
abortions (136). However, no clear association between con- noted clinical findings include neck stiffness, lethargy or som-
genital malformations and maternal mumps has been demon- nolence, and abdominal pain. Most patients with mumps with
strated (137). An etiologic relationship between gestational CNS involvement have signs of meningitis (e.g., headache
mumps and endocardial fibroelastosis has been postulated and nuchal rigidity), but no evidence of cortical dysfunction.
(138). Mumps occurring during the perinatal period is usually Defervescence is usually accompanied by overall clinical recov-
benign, perhaps due the partial protective effect of transpla- ery, and the total duration of illness in uncomplicated cases is
cental antibody (139). 7 to 10 days (40,41).

TA B L E 1 6 . 1
PRESENTING SIGNS AND SYMPTOMS OF PATIENTS WITH MUMPS INFECTION OF THE CENTRAL NERVOUS SYSTEM

Kravis et al. (36) Murray et al. (37) McLean et al. (44) Azimi et al. (42) Levitt et al. (30)
(N 74) (N 50) (N 30) (N 51) (N 64)

Fever NR 88% 100% 94% 100%


Vomiting 68% 76% 79% 84% 78%
Headache 47% 72% 62% 47% 88%
Parotitisa 53% 54% 62% 47% 53%
Neck stiffness 43% 76% 77% 71% 93%
Lethargy 28% 34% 31% 69% NR
Abdominal pain 15% 16% 23% 14% NR
Seizures 14% NR 18% 18% NR

NR, not reported.


a
Or swelling of other glands.

Scheld_Ch16.indd 243 2/21/14 5:38 PM


244 Part II: Viral Infections and Related Disorders

The presence of seizures, pronounced changes in level of fourfold rise in mumps antibody titer, providing a retrospective
consciousness, or focal neurologic findings are indicative of diagnosis. Parotitis can occasionally be caused by other viruses
significant encephalitis (30). Koskiniemi et al. (31) reviewed (e.g., Epstein-Barr, adenovirus, influenza A, parainfluenza,
41 cases of mumps encephalitis occurring in Helsinki, Finland coxsackievirus, lymphocytic choriomeningitis) or by bacteria
between 1968 and 1980 and reported high fever (39) in (e.g., Staphylococcus aureus) (157,158). Noninfectious causes
83%, vomiting in 88%, headaches in 71%, difficulty walking of parotid gland enlargement include Sjgren syndrome, sar-
in 37%, nuchal rigidity in 27%, seizures in 24%, psychiatric coidosis, thiazide ingestion, iodine sensitivity, tumor, or salivary
disturbances in 22%, and significantly depressed levels of con- duct obstruction (159,160). Parotid gland enlargement has been
sciousness in 20% of patients. described in patients with AIDS, especially children (161,162).
Mumps meningitis is a benign disease with essentially no risk A careful physical examination should permit parotitis to be
of mortality or long-term morbidity. It is difficult to judge accu- distinguished from lymphadenitis or lymphadenopathy.
rately the true incidence of neurologic sequelae following mumps When aseptic meningitis occurs in the context of parotitis,
CNS disease from published reports because of the variability the diagnosis of mumps is usually obvious. The cause of the
of the populations studied. Patients who develop permanent se- meningitis may be obscured, however, if there is no accompa-
quelae following mumps with CNS involvement are presumed nying salivary gland enlargement. Mumps meningoencephalitis
to have had mumps encephalitis. However, the mortality rate for has been confused with nonparalytic poliomyelitis, especially
patients with mumps encephalitis is 1.5% or less, and perma- when it occurs during the summer (34). In general, the CSF leu-
nent sequelae are rare. As with many childhood viral infections, kocyte count is higher in mumps than in poliomyelitis, and the
the mortality rate for mumps appears to be substantially higher clinical findings of neck stiffness and fever resolve more quickly
among adults than among children. Between 1982 and 1991, in cases of mumps than in polio (28). Lennette et al. (163) re-
14 mumps-associated deaths were reported in the United States, ported 11 cases of encephalitis with mild local muscle weakness
and more than half occurred in patients older than 20 years of that were all clinically considered to be polio but that were sero-
age. Even among patients who are profoundly encephalopathic, logically demonstrated to be caused by mumps virus.
the probability for complete recovery is high; sustained seizures Confirmation of CNS involvement in patients with mumps
and focal neurologic deficits (both of which are uncommon) may is based on examination of the CSF. CSF pleocytosis (5
predict a less favorable outcome (31). In many large series of white blood cells/mm3) occurs in 40% to 60% of patients
patients with mumps CNS infection, no long-term neurologic with mumps parotitis (27,32). The spinal fluid opening pres-
sequelae were identified (28,42,44,141). Ataxia, behavioral sure is normal in virtually all cases (28). The CSF white blood
changes, and electroencephalographic abnormalities have been cell count is usually in the range of 200 to 600 cells/mm3
noted in children in the immediate postencephalitis period, but (Table 16.2), although cell counts of 1,000 to 2,000 cells/mm3
these usually resolve within a few weeks (42). are not uncommon. In the series of 45 patients with mumps
A wide variety of other neurologic complications have been meningoencephalitis reported by Wilfert (38), the CSF white
observed following mumps encephalitis. Among the reported blood cell count on the initial lumbar puncture was less than
sequelae are behavioral disturbances and personality changes 100 cells/mm3 in 13% of patients, 100 to 500 in 53%, 500 to
(31,34,36,142), seizure disorders (40,143), cranial nerve palsies 1,000 in 29%, and more than 1,000 cells/mm3 in 5% of pa-
(especially facial and ocular palsies) (36,144), muscle weakness tients. The differential count of CSF leukocytes demonstrates
including hemiparesis (34,37), cerebellitis and ataxia (31,145), more than 80% lymphocytes in 80% to 90% of patients
acute hydrocephalus (146), and chronic headaches (143,147). (28,30,36,38). A small number of neutrophils are commonly
Myelitis and polyneuritis have also been reported as sequelae seen, but neutrophil predominance in the initial CSF sample
of mumps (148,149). occurs in fewer than 5% of patients with mumps CNS infec-
Sensorineural hearing loss is an uncommon but well-rec- tion (28,38,42). The CSF protein is normal in about one half
ognized complication of mumps that occurs with an estimated the patients and moderately elevated (100 mg/dL) in the re-
frequency of 0.5 to 5.0/100,000 cases (143,147,150,151). In a mainder (31,38,40,41). The CSF glucose is normal in most
recent survey conducted in Japan, the incidence of hearing loss in patients, but moderate hypoglycorrhachia (20 to 40 mg/dL)
children with mumps was reported to be much higher (7 in 7,400 may be present in 10% to 20% of patients with mumps men-
cases, or approximately 1/1,000 cases) (152). Deafness may be ingitis (30,31,41,42). In the series reported by Wilfert (38), 14
either transient or permanent and probably results from direct of 45 patients had CSF glucose of less than 40 mg/dL. Marked
damage to the cochlea by the mumps virus (154). Rare ocular hypoglycorrhachia (10 mg/dL), as can be seen in pyogenic
complications of mumps include keratitis, iritis, and central reti- bacterial meningitis, is very uncommon in mumps meningitis.
nal vein occlusion (154,155). An abnormally low CSF glucose is an unusual finding in viral
meningitis and has been reported most often in meningitis
caused by mumps virus, lymphocytic choriomeningitis virus,
DIAGNOSIS AND LABORATORY or herpes simplex virus.
FINDINGS No clear association has been established between the mag-
nitude of the CSF pleocytosis and the clinical course. The CSF
The presentation of a febrile child with parotitis strongly sug- white blood cell count may be higher in patients with parotitis
gests the diagnosis of mumps, particularly if the individual is and signs of meningitis than in patients with parotitis alone
known to be susceptible and has been exposed to mumps dur- (27). However, there is no correlation between the level of CSF
ing the preceding 2 to 3 weeks. However, the reduced frequency pleocytosis and severity of illness (40). CSF findings do not
of mumps in countries where vaccination is routine may result differ significantly between those patients with meningitis only
in physician inexperience and reduced accuracy of clinical diag- and those with mumps encephalitis (30). The magnitude of the
nosis (156). An atypical clinical presentation (e.g., meningitis or spinal fluid abnormalities is not predictive of the risk for long-
orchitis without parotitis) will usually require laboratory con- term sequelae following mumps encephalitis (31).
firmation. Culturing for mumps virus is the definitive diagnostic Studies employing sequential lumbar punctures have dem-
test but is frequently not available and has largely been replaced onstrated that the CSF white blood cell count frequently
by polymerase chain reaction (PCR) assays. Testing of paired increases during the first 2 to 3 days after the onset of CNS
acute and convalescent sera should demonstrate a diagnostic symptoms and then begins to decline (34,38,164). Even 2 weeks

Scheld_Ch16.indd 244 2/21/14 5:38 PM


Chapter 16: Meningitis and Encephalitis Caused by Mumps Virus 245

TA B L E 1 6 . 2
INITIAL CEREBROSPINAL FLUID FINDINGS IN PATIENTS WITH
MUMPS INFECTION OF THE CENTRAL NERVOUS SYSTEM

Russell and
Kilham (226) Donald (164) Ritter (40)
(N 22) (N 19) (N 30)

WBC (per mm3)


Mean 503 416 357
Range 1002,920 41,260 151212
Differential (% lymphs)
Mean 96 69 89
Range 89100 2898 30100
Protein (mg/dL)
Mean 56 79 49
Range 17145 20240 17140
Glucose (mg/dL)
Mean NR 53 NR
Range NR 3469 NR

WBC, white blood cell; NR, not reported.

after the onset of CNS symptoms, when most patients with antibodies directed against the V (HN) and S (nucleocapsid)
mumps meningoencephalitis are asymptomatic or substantially antigens. Highly sensitive ELISAs that can be readily auto-
improved, the CSF white blood cell count may still be in the mated have now largely replaced CF as the preferred method
range of 100 to 500 cells/mm3. Complete normalization of the for serodiagnosis (174,175). All these assays are designed to
spinal fluid and disappearance of CSF pleocytosis may require measure a fourfold increase in mumps-specific IgG between the
several weeks (28,40,165). acute serum (collected at the time of clinical disease) and the
Laboratory confirmation of the clinical diagnosis depends on convalescent serum (collected 2 to 4 weeks later). Alternatively,
isolation of mumps virus, nucleic acid amplification, or dem- demonstration of mumps-specific IgM by ELISA is indicative of
onstration of an appropriate serologic response (166). Mumps recent infection and is widely used for serodiagnosis of acute
virus can be isolated from saliva from virtually all patients with disease (176). An IgM response is detectable during the first
acute mumps parotitis (34). Virus can also be recovered from week of illness and persists for at least 6 weeks (177179).
the urine for up to 2 weeks after the onset of illness. Virus can be In cases of mumps meningoencephalitis, involvement of the
isolated from 30% to 50% of CSF samples collected early during CNS can be confirmed by demonstration of IgM or elevated
the course of mumps CNS infection (28,39,44). Wolontis and ratios of mumps-specific IgG in CSF (103,180). Presence of IgG
Bjrvatn (167) attempted virus isolation from CSF specimens by a standard serologic test provides presumptive laboratory
of 655 patients with mumps and CSF pleocytosis and were suc- evidence of prior infection, but there is no universally accepted
cessful in 33% of cases. Interestingly, no significant association surrogate immunologic marker of protection (181). The mumps
between the magnitude of the CSF pleocytosis and the probabil- skin test is not a reliable indicator of immune status.
ity of viral isolation could be demonstrated (34,167). Other routine laboratory studies are not generally helpful.
Widespread availability of molecular diagnostic techniques The average peripheral white blood cell count in patients with
such as RT-PCR has replaced viral culture in many settings mumps is 10,000 to 12,000 cells/mm3, with a differential of
and may eventually replace serology (67,168,169). Detection 30% to 40% lymphocytes (36,40,42). Approximately 30%
of mumps virus RNA is diagnostic of infection; sequence of patients have an elevated serum amylase, reflecting inflam-
analysis of the amplified SH gene can be used for molecular mation of the salivary glands or pancreas (42). During acute
epidemiologic studies. Quantitation of viral RNA is readily mumps encephalitis, the electroencephalogram characteristi-
accomplished using real-time RT-PCR methods (170172). cally shows moderate to severe slowing without spikes or later-
RT-PCR appears to be more sensitive than culture or immuno- alizing signs (31,182). Little information is available regarding
histochemical staining for detection of mumps virus in CSF or the utility of modern imaging methods (computed tomography
oropharyngeal swabs (168,173). or magnetic resonance imaging) in the diagnosis and manage-
A variety of assays have been developed to measure the ment of mumps CNS infections (183). Scrotal ultrasonography
humoral immune responses to mumps virus infection (66). may help with the evaluation of orchitis (184).
To greater or lesser degrees, all these serologic tests are lim-
ited by cross reactions between mumps virus and other human
parainfluenza viruses (48). The neutralizing antibody assay THERAPY
has been considered the gold standard test, but it is techni-
cally demanding. The HI assay is simple and sensitive, but re- Clinical management of patients with mumps consists of con-
agents may not be commercially available. In the past, the most servative measures to provide symptomatic relief and ensure
widely used serologic test has been the CF assay, which detects adequate rest, hydration, and nutritional support. There is

Scheld_Ch16.indd 245 2/21/14 5:38 PM


246 Part II: Viral Infections and Related Disorders

currently no established role for antiviral chemotherapy or pas- not recommended for persons with a history of anaphylac-
sive immunotherapy in mumps. Treatment of orchitis includes tic reactions to those substances. Desensitization protocols to
bed rest, scrotal support, analgesics, and ice packs. In anec- permit safe administration of the vaccine to egg-allergic chil-
dotal reports, men with mumps orchitis were said to improve dren have been described (210).
and have a lower frequency of testicular atrophy after admin- Questions regarding prevention often arise when an indi-
istration of interferon--2B, but this therapy has not been vidual with no history of mumps (typically a male adult) is
adequately studied in a controlled fashion (121,185188). exposed to a patient with active mumps. The immune status of
Patients with clinical evidence of significant CNS involvement the exposed individual can be determined by ELISA, although
(altered mental status, seizures, or focal neurologic findings) this may involve some delay (98). Mumps vaccine can be
require hospitalization for observation. Supportive care for safely administered to an individual of unknown immune sta-
patients with mumps encephalitis includes hydration, fever tus (211). However, vaccine given to a susceptible individual
control, antiemetics, and anticonvulsants as required. Lumbar after exposure to mumps may not provide protection. The vast
puncture has been reported to relieve the headache associated majority of adults born in the United States before 1957 have
with mumps meningitis in some patients. There have been an- been naturally infected and are therefore immune, although
ecdotal reports of the use of corticosteroids in patients with susceptibility rates among younger adults are higher (212).
mumps encephalitis, but no benefits have been proven (40). Widespread use of the mumps vaccine has had a major im-
In the series of patients with mumps meningoencephalitis pact on the incidence of mumps and mumps meningoenceph-
reported by Ritter (40), the average duration of hospitaliza- alitis (190). Through the mid-1960s, mumps was a leading
tion was 9.1 days, with a range of 5 to 19 days. cause of viral encephalitis (213). By the mid-1980s, however,
mumps had been reduced to the seventh most common cause
of viral encephalitis in the United States, accounting for only
PREVENTION 0.5% of cases of viral encephalitis. This trend was documented
in Minnesota, where mumps was the second most common
The cornerstone of mumps prevention is active immuniza- cause of encephalitis between 1950 and 1972, but where no
tion with the live attenuated mumps vaccine (189,190). The cases of mumps CNS infection were noted in 1972 to 1981
U.S. Public Health Service recommends administration of the (24). Similarly, mumps and measles disappeared as leading
MMR vaccine by subcutaneous injection in two doses, with causes of encephalitis in children in Finland after institution of
the first dose given at 12 to 15 months of age and the second a nationwide MMR vaccination program in 1982 (214). Not
at ages 4 to 6 years, prior to school entry (191). In 2011 to surprisingly, the resurgence in mumps over the last decade has
2012, median coverage of children with two doses of MMR in been accompanied by increased numbers of cases of mumps-
the United States was 94.8% (192). Clinical trials conducted associated meningitis and encephalitis.
during vaccine development indicated a vaccine efficacy rate The Jeryl Lynn strain of attenuated mumps virus used in
of 97% (193). However, studies conducted during more re- the United States since 1967 is a very well-tolerated vaccine,
cent mumps outbreaks have suggested that the vaccine is 80% although rare instances of fever, parotitis, orchitis, or asep-
to 90% effective in preventing clinical mumps (26,194198). tic meningitis following immunization have been reported
Vaccine efficacy may be lower among household contacts (215). Other attenuated mumps viruses used for vaccina-
(65 to 75%) (198). In countries where universal vaccination tion in various countries include RIT 4385, Urambe Am9,
is practiced, mumps outbreaks due to failure to vaccinate con- Rubini, Leningrad-3, Leningrad-Zagreb, Hoshino, and Torii
tinue to occur among immigrants and in specific groups where strains (216). Beginning in 1988, an increased frequency of
vaccination is not accepted (199202). Furthermore, large- vaccine-related mumps meningitis cases (rates as high as 1 to
scale mumps outbreaks in highly vaccinated populations con- 3/1,000 vaccinations) was recognized in Japan (217), Canada
tinue to occur and can be attributed to primary (inadequate (218), Brazil (219), and the United Kingdom (220). These
immune response) or to secondary (waning immunity after cases followed administration of an MMR vaccine containing
successful immunization) vaccine failure (1922,203,204). the Urabe Am9 strain of mumps virus. In several instances,
Asymptomatic mumps virus infection has been demonstrated mumps virus was recovered from the CSF (221). A similar
in vaccinated children (with an attack rate of 7% to 10%) situation was reported from Yugoslavia following adminis-
and may contribute to epidemic spread (205). Because wild tration of the Leningrad-3 mumps vaccine (222). The clini-
type mumps virus is always genetically distinct from vaccine- cal presentation and CSF findings of the vaccine-related cases
strain virus, concerns have been raised that vaccine-induced are similar to those of naturally occurring mumps meningitis,
immunity might not provide protection against some wild- with onset of symptoms about 3 weeks after vaccination. In
type variants (206). However, in vitro studies have shown that general, the patients with vaccine-related meningitis were not
genetically diverse mumps virus isolates collected during out- seriously ill, and long-term sequelae have not been noted. This
breaks were all effectively neutralized by sera from vaccinated adverse effect of vaccination was presumed to result from in-
children, arguing against immune escape (207). adequate attenuation of the vaccine virus. However, nucleo-
Administration of the live mumps vaccine is contraindi- tide sequencing of the mumps virus HN gene from the Urabe
cated in pregnant women (208). Vaccination is also not rec- vaccine showed that the vaccine was actually a mixture of wild
ommended in persons who have received immunoglobulin type and variant mumps viruses with identifiable genetic dif-
therapy within the preceding 3 months (which might interfere ferences, which appear to confer differences in neurovirulence
with the immune response to the vaccine) or in persons with (223,224). For example, most isolates recovered from patients
severe systemic immunosuppression caused by disease or med- who developed postvaccination parotitis or meningitis were
ical therapy. Mumps immunization (using MMR) is recom- wild type (Lys335) rather than variant (Glu335) virus (223).
mended for children infected with human immunodeficiency In some countries, the highly immunogenic Urabe mumps
virus (HIV) who do not have evidence of severe immunosup- vaccine strain was replaced by the highly attenuated Rubini
pression (defined as CD4 count less than 200 lymphocytes/ strain, which appears to provide unacceptably low levels of
mm3 or CD4 less than 15% (209). Since mumps vaccine is clinical protection (190,225). These problems have not been
produced in cell cultures of chick embryos and may contain recognized in the United States, where the Jeryl Lynn mumps
trace amounts of egg protein and neomycin, immunization is vaccine is still used.

Scheld_Ch16.indd 246 2/21/14 5:38 PM


Chapter 16: Meningitis and Encephalitis Caused by Mumps Virus 247

Selected Readings
Barskey AE, Schulte C, Rosen JB, et al. Mumps outbreak in Orthodox Jewish methods of virus detection and nested PCR. J Clin Virol. 2006;37:
communities in the United States. N Engl J Med. 2012;367:17041713. 184189.
Carr MJ, Moss E, Waters A, et al. Molecular epidemiological evaluation of the Kutty PK, Kyaw MH, Dayan GH, et al. Guidance for isolation precautions
recent resurgence in mumps virus infections in Ireland. J Clin Microbiol. for mumps in the United States: a review of the scientific basis for policy
2010;48:32883294. change. Clin Infect Dis. 2010;50:16191628.
Centers for Disease Control and Prevention. Prevention of measles, rubella, con- Lievano F, Galea SA, Thornton M, et al. Measles, mumps, and rubella virus
genital rubella syndrome, and mumps, 2013: summary recommendations vaccine (M-M-RII): a review of 32 years of clinical and postmarketing
of the Advisory Committee on Immunization Practices (ACIP). MMWR experience. Vaccine. 2012;30:69186926.
Recomm Rep. 2013;62(RR-04):134. Muhlemann K. The molecular epidemiology of mumps virus. Infect Genet Evol.
Dayan GH, Quinlisk MP, Parker AA, et al. Recent resurgence of mumps in the 2004;4:215219.
United States. N Engl J Med. 2008;358:15801589. Ternavasio-de la Vega HG, Boronat M, Ojeda A, et al. Mumps orchitis
Hviid A, Rubin S, Muhlemann K. Mumps. Lancet. 2008;371:932944. in the post-vaccine era (19672009): a single-center series of 67 pa-
Krause CH, Eastick K, Ogilvie MM. Real time PCR for mumps diag- tients and review of clinical outcome and trends. Medicine. 2010;89:
nosis on clinical specimenscomparison with results of conventional 96116.

References
1. Anderson RM, Crombie JA, Grenfell BT. The epidemiology of mumps in 23. McIntyre JP, Keen GA. Labortory surveillance of viral meningitis by
the UK: a preliminary study of virus transmission, herd immunity and the examination of cerebirospinal fluid in Cape Town. Epidemiol Infect.
potential impact of immunization. Epidemiol Infect. 1987;99:6584. 1993;111(2):357371.
2. Bennett AD, Modlin J, Orenstein WA, et al. Current status of mumps in the 24. Beghi E, Nicolosi A, Kurland LT, et al. Encephalitis and aseptic meningitis,
United States. J Infect Dis. 1975;132:106109. Olmsted County, Minnesota, 19501981: I. Epidemiology. Ann Neurol.
3. Cochi SL, Preblud SR, Orenstein WA. Perspective on the realtive resurgence 1984;16:283294.
of mumps in the United States. Am J Dis Child. 1988;142:449507. 25. Meyer HM, Johnson RT, Crawford IP, et al. Central nervous system
4. Centers for Disease Control and Prevention. Mumps prevention. MMWR syndromes of viral etiology. Am J Med. 1960;29:334347.
Morb Mortal Wkly Rep. 1989;38:388392,397400. 26. Koskiniemi ML, Vaheri A. Acute encephalitis of viral origin. Scand J Infect
5. Cheek JE, Baron R, Atlas H, et al. Mumps outbreak in a highly vacci- Dis. 1982;14:181187.
nated school population. Evidence for large scale vaccination failure. Arch 27. Bang HO, Bang J. Involvement of the central nervous system in mumps.
Pediatr Adolesc Med. 1995;149:774778. Acta Med Scand. 1943;113:487505.
6. van Loon FP, Holmes SJ, Sirotkin BI, et al. Mumps surveillanceUnited 28. Bruyn HB, Sexton HM, Brainend HD. Mumps meningoencephalitis:
States, 1988-1993. MMWR CDC Surveill Summ. 1995;44:114. a clinical review of 119 cases with one death. Calif Med. 1957;86:
7. Centers for Disease Control and Prevention. Summary of notifiable 153160.
diseases, United States, 2000. MMWR Morb Mortal Wkly Rep. 2002;49: 29. Klemola E, Kaariainen L, Ollila O, et al. Studies on viral encephalitis. Acta
1100. Med Scand. 1965;177:707716.
8. Kutty PK, Kruszon-Moran DM, Dayan GH, et al. Seroprevalence of 30. Levitt LP, Rich TA, Kindle SW, et al. Central nervous system mumps.
antibody to mumps virus in the US population, 19992004. J Infect Dis. Neurology. 1970;20:829834.
2010;202:667674. 31. Koskiniemi M, Donner M, Pettay O. Clinical appearance and outcome in
9. Dayan GH, Quinlisk MP, Parker AA, et al. Recent resurgence of mumps in mumps encephalitis in children. Acta Pediatr Scand. 1983;72:603609.
the United States. N Engl J Med. 2008;358:15801589. 32. Bowers D, Weatherhead DSP. Mumps encephalitis. Can Med Assoc J.
10. Kancherla VS, Hanson IC. Mumps resurgence in the United States. 1953;69:4955.
J Allergy Clin Immunol. 2006;118:938941. 33. Finkelstein H. Meningoencephalitis in mumps. JAMA. 1938;117:1719.
11. Centers for Disease Control and Prevention. Brief report: update: mumps 34. Kilham L, Levens J, Enders J. Non-paralytic poliomyelitis and mumps
activityUnited States, January 1October 7, 2006. MMWR Morb Mortal meningoencephalitis. JAMA. 1949;140:934936.
Wkly Rep. 2006;55:11521153. 35. Eberlein WR, Lynxwiler CP. Clinical picture of mumps meningoencephalitis
12. Peltola H, Kulkarni PS, Kapre SV, et al. Mumps outbreaks in Canada and and report of case without parotitis. J Pediatr. 1947;31:513520.
the United States: time for new thinking on mumps vaccines. Clin Infect 36. Kravis LP, Sigel MM, Henle G. Mumps meningoencephalitis, with special
Dis. 2007;45:459466. reference to the use of the complement fixation test in diagnosis. Pediatrics.
13. Davidkin I, Jokinen S, Broman M, et al. Persistence of measles, mumps, 1951;8:204214.
and rubella antibodies in an MMR-vaccinated cohort: a 20-year follow-up. 37. Murray HGS, Field CMB, McLeod WJ. Mumps menigo-encephalitis.
J Infect Dis. 2008;197:950956. Br Med J. 1960;1:18501853.
14. Ogbuanu IU, Kutty PK, Hudson JM, et al. Impact of a third dose of 38. Wilfert CM. Mumps meningoencephalitis with low cerebrospinal fluid
measles-mumps-rubella vaccine on a mumps outbreak. Pediatrics. glucose, prolonged pleocytosis and elevation of protein. N Engl J Med.
2012;130:e1567e1574. 1969;280:855859.
15. Abedi GR, Mutuc JD, Lawler J, et al. Adverse events following a third dose 39. Bjrvatn N, Wolontis S. Mumps meningoencephalitis in Stockholm,
of measles, mumps, and rubella vaccine in a mumps outbreak. Vaccine. November 1964July 1971. I. Analysis of a hospitalized study group.
2012;30:70527058. Questions of selection and representativity. Scand J Infect Dis. 1973;5:
16. Centers for Disease Control and Prevention. Mumps epidemicUnited 253260.
Kingdom, 20042005. MMWR Morb Mortal Wkly Rep. 2006;55: 40. Ritter BS. Mumps meningoencephalitis in children. J Pediatr. 1958;52:
173175. 424433.
17. Gupta RK, Best J, MacMahon E. Mumps and the UK epidemic 2005. BMJ. 41. Johnstone JA, Ross CAC, Dunn M. Meningitis and encephalitis associated
2005;330:11321135. with mumps infection. Arch Dis Child. 1972;47:647651.
18. Savage E, Ramsay M, White J, et al. Mumps outbreaks across England and 42. Azimi PH, Cramblett HG, Hayers RE. Mumps meningoencephalitis in
Wales in 2004: observational study. BMJ. 2005;330:11191120. children. JAMA. 1969;207:509512.
19. Barskey AE, Schulte C, Rosen JB, et al. Mumps outbreak in Orthodox 43. McLean DM, Walker SJ, McNaughton GA. Mumps meningoencephalitis:
Jewish communities in the United States. N Engl J Med. 2012;367: a virological and clinical study. Can Med Assoc J. 1960;83:148151.
17041713. 44. McLean DM, Bach RD, Larke RP, et al. Mumps meningoencephalitis,
20. Greenland K, Whelan J, Fanoy E, et al. Mumps outbreak among vaccinated Toronto, 1963. Can Med Assoc J. 1964;90:458462.
university students associated with a large party, the Netherlands, 2010. 45. Galinski MS, Wechsler SL. The molecular biology of the paramyxovirus
Vaccine. 2012;30:46764680. genus. In: Kingbury DW, ed. The Paramyxoviruses. New York: Plenum
21. Whelan J, van Binnendijk R, Greenland K, et al. Ongoing mumps outbreak Press; 1991:4182.
in a student population with high vaccination coverage, Netherlands, 2010. 46. Chang A, Dutch RE. Paramyxovirus fusion and entry: multiple paths to a
Euro Surveill. 2010;15(17). common end. Viruses. 2012;4:613636.
22. Anis E, Grotto I, Moerman L, et al. Mumps outbreak in Israels highly 47. Server AC, Merz DC, Waxham MN, et al. Differentiation of mumps virus
vaccinated society: are two doses enough? Epidemiol Infect. 2012;140: strains with monoclonal antibody to the HN glycoprotein. Infect Immun.
439446. 1982;35:179186.

Scheld_Ch16.indd 247 2/21/14 5:38 PM


248 Part II: Viral Infections and Related Disorders

48. Ito Y, Tsurudome M, Hishiyama M, et al. Immunological interrelationships 78. Fleischer B, Kreth HW. Mumps virus replication in human lymphoid cell
among human and non-human paramyxoviruses revealed by immuno- lines and in peripheral blood lymphocytes: preference for T cells. Infect
precipitation. J Gen Virol. 1987;68(pt 5):12891297. Immun. 1982;35:2531.
49. Okazaki K, Tanabayashi K, Takeuchi K, et al. Molecular cloning and 79. Lindeman J, Muller WK, Versteeg J, et al. Rapid diagnosis of meningoen-
sequence analysis of the mumps virus gene encoding the L protein and the cephalitis: immunofluorescent examination of fresh and in vitro cultured
trailer sequence. Virology. 1992;188:926930. cerebrospinal fluid cells. Neurology. 1974;24:143148.
50. Elliott GD, Yeo RP, Afzal MA, et al. Strain-variable editing during transcrip- 80. Rozina EE, Hilgenfeldt M. Comparative study on the neurovirulence
tion of the P gene of mumps virus may lead to the generation of non-structural of different vaccine strains of parotitis virus in monkeys. Acta Virol.
proteins NS1 (V) and NS2. J Gen Virol. 1990;71(pt 7):15551560. 1985;29:225230.
51. Xu P, Luthra P, Li Z, et al. The V protein of mumps virus plays a critical 81. Wolinsky JS, Baringer JR, Margolis G, et al. Ultrastructure of mumps
role in pathogenesis. J Virol. 2012;86:17681776. virus replication in newborn hamster central nervous system. Lab Invest.
52. Andrejeva J, Childs KS, Young DF, et al. The V proteins of paramyxoviruses 1974;31:402412.
bind the IFN-inducible RNA helicase, mda-5, and inhibit its activation of 82. Donohue WL. The pathology of mumps encephalitis with report of a fatal
the IFN-beta promoter. Proc Natl Acad Sci U S A. 2004;101:1726417269. case. J Pediatr. 1941;19:4552.
53. Kubota T, Yokosawa N, Yokota S, et al. Mumps virus V protein antago- 83. Schwartz GA, Yang DC, Noone EL. Meningoencephalomyelitis with
nizes interferon without the complete degradation of STAT1. J Virol. epidemic parotitis. Arch Neurol. 1964;11:453462.
2005;79:44514459. 84. Bistrian B, Phillips CA, Kaye IS. Fatal mumps meningoencephalitis.
54. Takeuchi K, Tanabayashi K, Hishiyama M, et al. The mumps virus SH JAMA. 1972;222:478479.
protein is a membrane protein and not essential for virus growth. Virology. 85. Taylor FB, Toreson WE. Primary mumps meningo-encephalitis. Arch
1996;225:156162. Intern Med. 1963;112:216.
55. Afzal MA, Buchanan J, Dias JA, et al. RT-PCR based diagnosis and 86. Johnson RT. Mumps virus encephalitis in the hamster. J Neuropath Exp
molecular characterisation of mumps viruses derived from clinical speci- Neurol. 1968;27:8095.
mens collected during the 1996 mumps outbreak in Portugal. J Med Virol. 87. Uno M, Takano T, Yamano T, et al. Age-dependent susceptibility in
1997;52:349353. mumps-associated hydrocephalus: neuropathologic features and brain
56. Jin L, Rima B, Brown D, et al. Proposal for genetic characterisation of wild- barriers. Acta Neuropathol (Berl). 1997;94:207215.
type mumps strains: preliminary standardisation of the nomenclature. Arch 88. Spataro RF, Lin SR, Horner FA, et al. Aqueductal stenosis and hydro-
Virol. 2005;150:19031909. cephalus: a rare sequelae of mumps virus infection. Neuroradiology.
57. Johansson B, Tecle T, Orvell C. Proposed criteria for classification of new 1976;12:1113.
genotypes of mumps virus. Scand J Infect Dis. 2002;34:355357. 89. Viola L, Chiaretti A, Castorina M, et al. Acute hydrocephalus as a con-
58. Kidokoro M, Tuul R, Komase K, et al. Characterization of mumps viruses sequence of mumps meningoencephalitis. Pediatr Emerg Care. 1998;14:
circulating in Mongolia: identification of a novel cluster of genotype H. 212214.
J Clin Microbiol. 2011;49:19171925. 90. McCarthy M, Walinsky JS, Lazzarini RA. A persistent infection of Vero
59. Jin L, Brown DW, Litton PA, et al. Genetic diversity of mumps virus in oral cells by egg-adapted mumps virus. Virology. 1981;114:343356.
fluid specimens: application to mumps epidemiological study. J Infect Dis. 91. Lve A, Andersson T, Norrby E, et al. Mumps virus infection of disso-
2004;189:10011008. ciated rodent spinal ganglion in vitro. Expression and disappearance of
60. Muhlemann K. The molecular epidemiology of mumps virus. Infect Genet viral structural protein in neurons. J Gen Virol. 1987;68:17551759.
Evol. 2004;4:215219. 92. Vaheri A, Juklunen I, Koskiniemi ML. Chronic encephalomyelitis with spe-
61. Palacios G, Jabado O, Cisterna D, et al. Molecular identification of mumps cific increase in intrathecal mumps antibodies. Lancet. 1982;2:685688.
virus genotypes from clinical samples: standardized method of analysis. 93. Ito M, Go T, Okuno T, et al. Chronic mumps virus encephalitis. Pediatr
J Clin Microbiol. 2005;43:18691878. Neurol. 1991;7:467470.
62. Carr MJ, Moss E, Waters A, et al. Molecular epidemiological evalua- 94. Haginoya K, Ike K, Iinuma K, et al. Chronic progressive mumps virus
tion of the recent resurgence in mumps virus infections in Ireland. J Clin encephalitis in a child [letter]. Lancet. 1995;346:50.
Microbiol. 2010;48:32883294. 95. Baas MC, van Donselaar KA, Florquin S, et al. Mumps: not an innocent by-
63. Nojd J, Tecle T, Samuelsson A, et al. Mumps virus neutralizing antibodies stander in solid organ transplantation. Am J Transplant. 2009;9:21862189.
do not protect against reinfection with a heterologous mumps virus geno- 96. Nigro G, Nanni F, Midulla M. Determination of vaccine-induced and nat-
type. Vaccine. 2001;19:17271731. urally acquired class-specific antibodies by two indirect ELISAs. J Virol
64. Orvell C, Tecle T, Johansson B, et al. Antigenic relationships between six Methods. 1986;13:91106.
genotypes of the small hydrophobic protein gene of mumps virus. J Gen 97. Chiba Y, Horino K, Umetsu M, et al. Virus excretion and antibody responses
Virol. 2002;83:24892496. in saliva in natural mumps. Tohoku J Exp Med. 1973;111:229238.
65. Tecle T, Mickiene A, Johansson B, et al. Molecular characterisation of two 98. Linde GA, Granstrom M, rvell C. Immunoglobulin class and immu-
mumps virus genotypes circulating during an epidemic in Lithuania from noglobulin G subclass enzyme-linked immunosorbent assays compared
1998 to 2000. Arch Virol. 2002;147:243253. with microneutralization assay for serodiagnosis of mumps infection and
66. Swierkosz EM. Mumps virus. In: Murray P, Baron E, Pfaller M, et al, eds. determination of immunity. J Clin Microbiol. 1987;25:16531658.
Manual of Clinical Microbiology. 6th ed. Washington, DC: ASM Press; 99. Ukkonen P, Grandstrom ML, Penttinen K. Mumps-specific immuno-
1995:936967. globulin M and G antibodies in natural mumps infection as measured by
67. Boriskin Yu S, Booth JC, Yamada A. Rapid detection of mumps virus by the enzyme-linked immunosorbent assay. J Med Virol. 1981;8:131142.
polymerase chain reaction. J Virol Methods. 1993;42:2332. 100. Lve A, Rydbeck R, Kristensson K, et al. Hemagglutinin-neuraminidase
68. Reed D, Brown G, Merrick R, et al. A mumps epidemic on St. George glycoprotein as a determinant of pathogenicity in mumps virus hamster
Island, Alaska. JAMA. 1967;199:967971. encephalitis: analysis of mutants selected with monoclonal antibodies.
69. Levitt LP, Mahoney DH, Casey HL, et al. Mumps in a general population: J Virol. 1985;58:6774.
a sero-epidemiologic study. Am J Dis Child. 1970;120:134138. 101. Gut JP, Lablache C, Behr S, et al. Symptomatic mumps virus reinfections.
70. Meyer MD, Stifler WC, Joseph JM. Evaluation of mumps vaccine given J Med Virol. 1995;45:1723.
after exposure to mumps, with special reference to the exposed adult. 102. Glikmann G, Pedersen M, Mordhorst CH. Detection of specific immuno-
Pediatrics. 1966;37:304315. globulin M to mumps virus in serum and cerebrospinal fluid samples from
71. Henle G, Henle W, Wendell KK, et al. Isolation of mumps virus from patients with acute mumps infection, using an antibody capture enzyme
human beings with induced apparent or inapparent infections. J Exp Med. immunoassay. Acta Pathol Microbiol Immunol Scand. 1986;94:145156.
1946;88:223232. 103. Forsberg P, Fryden A, Link H, et al. Viral IgM and IgG antibody synthe-
72. Ennis FA, Jackson D. Isolation of virus during the incubation period of sis within the central nervous system in mumps meningitis. Acta Neurol
mumps infection. J Pediatr. 1968;72:536537. Scand. 1986;73:372380.
73. Meyer MB. An epidemiologic study of mumps: its spread in schools and 104. Ukkonen P, Granstrom ML, Rasanen J, et al. Local production of mumps
families. Am J Hyg. 1962;75:259281. IgG and IgM antibodies in the cerebrospinal fluid of meningitis patients.
74. Richardson M, Elliman D, Maguire H, et al. Evidence base of incubation J Med Virol. 1981;8:257265.
periods, periods of infectiousness and exclusion policies for the control 105. Vandermeulen C, Clement F, Roelants M, et al. Evaluation of cellular
of communicable diseases in schools and preschools. Pediatr Infect Dis J. immunity to mumps in vaccinated individuals with or without circulat-
2001;20:380391. ing antibodies up to 16 years after their last vaccination. J Infect Dis.
75. Overman JR. Viremia in human mumps infection. Arch Intern Med. 2009;199:14571460.
1958;102:354356. 106. Bruserud O, Thorsby E. HLA control of the proliferative T lymphocyte
76. Foy HM, Cooney MK, Hall CE, et al. Isolation of mumps virus from response to antigenic determinants on mumps virus. Scand J Immunol.
children with acute lower respiratory tract disease. Am J Epidemiol. 1985;22:509518.
1971;94:467472. 107. Reunanen M, Salonen R, Salmi A. Intrathecal immune responses in
77. Wolinsky JS, Klassen T, Baringer JR. Persistence of neuroadapted mumps mumps meningitis patients. Scand J Immunol. 1982;15:419426.
virus in brains of newborn hamsters after intraperitoneal inoculation. 108. Kress HG, Kreth HW. HLA-restriction of secondary mumps specific cyto-
J Infect Dis. 1976;133:260267. toxic T lymphocytes. J Immunol. 1982;129:844849.

Scheld_Ch16.indd 248 2/21/14 5:38 PM


Chapter 16: Meningitis and Encephalitis Caused by Mumps Virus 249

109. Kreth HW, Kress L, Kress HG, et al. Demonstration of primary cytotoxic 144. Beardwell A. Facial palsy due to the mumps virus. Br J Clin Pract.
T cells in venous blood and cerebrospinal fluid of children with mumps 1969;23:3738.
meningitis. J Immunol. 1982;128:24112415. 145. Majda-Stanislawska E. Mumps cerebellitis. Eur Neurol. 2000;43:117.
110. Fleischer B, Kreth HW. Clonal analysis of HLA-restricted virus-specific 146. Aydemir C, Eldes N, Kolsal E, et al. Acute tetraventricular hydrocepha-
cytotoxic T lymphocytes from cerebrospinal fluid in mumps meningitis. lus caused by mumps meningoencephalitis in a child. Pediatr Neurosurg.
J Immunol. 1983;130:21872190. 2009;45:419421.
111. Martin R, Vallbracht A, Kreth HW. Interferon-gamma secretion by in vivo 147. Laurence D, McGavin D. The complications of mumps. Brit Med J.
activated cytotoxic T lymphocytes from the blood and cerebrospinal fluid 1948;1:9497.
during mumps meningitis. J Neuroimmunol. 1991;33:191198. 148. Venketasubramanian N. Transverse myelitis following mumps in an
112. Asano T, Ichiki K, Koizumi S, et al. Enhanced expression of cytokines/ adulta case report with MRI correlation. Acta Neurol Scand. 1997;
chemokines in cerebrospinal fluids in mumps meningitis in children. 96:328331.
Pediatr Int. 2011;53:143146. 149. Bansal R, Kalita J, Misra UK, et al. Myelitis: a rare presentation of
113. Philip RN, Reinhard KR, Lackman DB. Observations on a mumps epi- mumps. Pediatr Neurosurg. 1998;28:204206.
demic in a virgin population. Am J Hyg. 1959;69:91111. 150. McKenna MJ. Measles, mumps, and sensorineural hearing loss. Ann N Y
114. Garty BZ, Danon YL, Nitzan M. Presternal swelling in mumps. Cutis. Acad Sci. 1997;830:291298.
1987;39:149150. 151. Kayan A. Bilateral sensorineural hearing loss due to mumps. Brit J Clin
115. Kutty PK, Kyaw MH, Dayan GH, et al. Guidance for isolation precau- Pathol. 1990;44:757759.
tions for mumps in the United States: a review of the scientific basis for 152. Hashimoto H, Fujioka M, Kinumaki H. An office-based prospective study
policy change. Clin Infect Dis. 2010;50:16191628. of deafness in mumps. Pediatr Infect Dis J. 2009;28:173175.
116. Beard CM, Benson RC, Kelalis PP, et al. The incidence and outcome of 153. Smith GA, Guessen R. Inner ear pathologic features following mumps
mumps orchitis in Rochester, Minnesota, 1935 to 1974. Mayo Clinic infection: report of a case in an adult. Arch Otolarynogol. 1976;102:
Proc. 1977;52:37. 108111.
117. Manson AL. Mumps orchitis. Urology. 1990;36:355358. 154. Riffenburgh RS. Ocular manifestations of mumps. Arch Ophthal.
118. Gall EA. The histopathology of acute mumps orchitis. Am J Pathol. 1961;66:739743.
1947;23:637651. 155. Foster RE, Lowder CY, Meisler DM, et al. Mumps neuroretinitis in an
119. Bjorvatn B. Mumps virus recovered from testicles by fine-needle aspira- adolescent. Am J Ophthalmol. 1990;110:9192.
tion biopsy in cases of mumps orchitis. Scand J Infect Dis. 1973;5:35. 156. Hatchette TF, Mahony JB, Chong S, et al. Difficulty with mumps
120. Jalal H, Bahadur G, Knowles W, et al. Mumps epididymo-orchitis with diagnosis: what is the contribution of mumps mimickers? J Clin Virol.
prolonged detection of virus in semen and the development of anti-sperm 2009;46:381383.
antibodies. J Med Virol. 2004;73:147150. 157. McQuone SJ. Acute viral and bacterial infections of the salivary glands.
121. Ternavasio-de la Vega HG, Boronat M, Ojeda A, et al. Mumps orchitis in Otolaryngol Clin North Am. 1999;32:793811.
the post-vaccine era (1967-2009): a single-center series of 67 patients and 158. Turner MD, Glickman R. Case presentations of salivary gland infections.
review of clinical outcome and trends. Medicine (Baltimore). 2010;89: Oral Maxillofac Surg Clin North Am. 2009;21:359362.
96116. 159. Travis LW, Hecht DW. Acute and chronic inflammatory diseases of the
122. Casella R, Leibundgut B, Lehmann K, et al. Mumps orchitis: report of a salivary glands: diagnosis and management. Otolaryngol Clin North Am.
mini-epidemic. J Urol. 1997;158:21582161. 1977;10:329338.
123. Wharton IP, Chaudhry AH, French ME. A case of mumps epididymitis. 160. Lei L, Velasco O, Nobay F. Iodide mumps: a case report of complicated
Lancet. 2006;367:702. radioactive iodine causing sialadenitis. Am J Emerg Med. 2012;30:512.
124. Tae BS, Ham BK, Kim JH, et al. Clinical features of mumps orchitis in vac- e515e516.
cinated postpubertal males: a single-center series of 62 patients. Korean J 161. Soberman N, Leonidas JC, Berdon WE, et al. Parotid enlargement in chil-
Urol. 2012;53:865869. dren seropositive for human immunodeficiency virus: imaging findings.
125. Suskovic T, Vukicevic-Baudoin D, Vucicevic Z, et al. Severe pancreatitis Am J Roentgenol. 1991;157:553556.
as first symptom of mumps complicated with pseudocyst and abscess of 162. Bem C, Barucha H, Patil PS. Parotid disease and human immunodefi-
pancreas. Infection. 1997;25:3940. ciency virus infection in Zambia. Br J Surg. 1992;79:768770.
126. Eyland E, Zmucky R, Sheba C. Mumps virus and subactue thyroiditis: 163. Lennette EH, Caplan GE, Magoffin RL. Mumps virus infection simulating
evidence of a causal association. Lancet. 1957;1:10621065. paralytic poliomyelitis: a report of 11 cases. Pediatrics. 1960;25:788797.
127. Sultz HA, Hart BA, Zielezny M, et al. Is mumps virus an etiologic factor 164. Russell RR, Donald JC. The neurological complications of mumps.
in juvenile diabetes mellitus? J Pediatr. 1975;86:654656. Br Med J. 1958;2:2730.
128. Morrison JC, Givens JR, Wiser WL. Mumps oophoritis: a cause of prema- 165. Azimi PH, Shaban S, Hilty MD, et al. Mumps meningoencephalitis: pro-
ture menopause. Fertil Steril. 1975;26:655660. longed abnormality of cerebrospinal fluid. JAMA. 1975;234:11611162.
129. Utz JP, Houk VN, Alling DW. Clinical and laboratory studies of mumps. 166. Person DA, Smith TF, Herrmann EC Jr. Experiences in laboratory diagno-
IV. Viruria and abnormal renal function. N Engl J Med. 1964;270: sis of mumps virus infections in routine medical practice. Mayo Clin Proc.
12831286. 1971;46:544548.
130. Kabakus N, Aydinoglu H, Bakkaloglu SA, et al. Mumps interstitial 167. Wolontis S, Bjorvatn B. Mumps meningoencephalitis in Stockholm,
nephritis: a case report. Pediatr Nephrol. 1999;13:930931. November 1964July 1971. Isolation attempts from the cerebrospinal
131. Gordon SC, Lauter CB. Mumps arthritis: a review of the literature. Rev fluid in a hospitalized group. Scand J Infect Dis. 1973;5:261271.
Infect Dis. 1984;6:338343. 168. Poggio GP, Rodriguez C, Cisterna D, et al. Nested PCR for rapid detec-
132. Harel L, Amir J, Reish O, et al. Mumps arthritis in children. Pediatr Infect tion of mumps virus in cerebrospinal fluid from patients with neurological
Dis J. 1990;9:928929. diseases. J Clin Microbiol. 2000;38:274278.
133. Chaudary S, Jaski BE. Fulminant mumps myocarditis. Ann Int Med. 169. Royuela E, Castellanos A, Sanchez-Herrero C, et al. Mumps virus
1989;110:569570. diagnosis and genotyping using a novel single RT-PCR. J Clin Virol.
134. Kussy JC. Fatal mumps myocarditis. Min Med. 1974;57:285286. 2011;52:359362.
135. Graham DY, Brown CH, Benrey J, et al. Thrombocytopenia: a complica- 170. Kubar A, Yapar M, Besirbellioglu B, et al. Rapid and quantitative
tion of mumps. JAMA. 1974;227:11621164. detection of mumps virus RNA by one-step real-time RT-PCR. Diagn
136. Siegal M, Fuerst HT, Peress NS. Comparative fetal mortality in maternal Microbiol Infect Dis. 2004;49:8388.
virus diseases: a prospective study on rubella measles, mumps, chicken- 171. Leblanc JJ, Pettipas J, Davidson RJ, et al. Detection of mumps virus RNA
pox, and hepatitis. New Engl J Med. 1966;274:768771. by real-time one-step reverse transcriptase PCR using the LightCycler
137. Katz M. Is there mumps embryopathy? An unanswered question. Clin platform. J Clin Microbiol. 2008;46:40494051.
Pediatr. 1967;6:321322. 172. Krause CH, Eastick K, Ogilvie MM. Real-time PCR for mumps diagnosis
138. St Geme JW Jr, Peralta H, Farias E, et al. Experimental gestational mumps on clinical specimenscomparison with results of conventional methods
virus infection and endocardial fibroelastosis. Pediatrics. 1971;48: of virus detection and nested PCR. J Clin Virol. 2006;37:184189.
821826. 173. Uchida K, Shinohara M, Shimada S, et al. Rapid and sensitive detection of
139. Jones JF, Ray CG, Fulginiti VA. Perinatal mumps infection. J Pediatr. mumps virus RNA directly from clinical samples by real-time PCR. J Med
1980;96:912914. Virol. 2005;75:470474.
140. Kaplan KM, Marder DC, Cochi SL, et al. Mumps in the workplace: 174. Mauldin J, Carbone K, Hsu H, et al. Mumps virus-specific antibody titers
further evidence of the changing epidemiology of a childhood vaccine- from pre-vaccine era sera: comparison of the plaque reduction neutralization
preventable disease. JAMA. 1988;260:14341438. assay and enzyme immunoassays. J Clin Microbiol. 2005;43:48474851.
141. Henderson W. Mumps meningo-encephalitis an outbreak in preparatory 175. Backhouse JL, Gidding HF, McIntyre PB, et al. Evaluation of two enzyme
school. Lancet. 1952;1:386388. immunoassays for detection of immunoglobulin G antibodies to mumps
142. Brown EH, Dunnett WH. A retrospective survey of the complications of virus. Clin Vaccine Immunol. 2006;13:764767.
mumps. J R Coll Gen Pract. 1974;24:552556. 176. Warrener L, Samuel D. Evaluation of a commercial assay for the detec-
143. Oldfelt V. Sequelae of mumps-meningoencephalitis. Acta Medica Scand. tion of mumps specific IgM antibodies in oral fluid and serum specimens.
1949;134:405414. J Clin Virol. 2006;35:130134.

Scheld_Ch16.indd 249 2/21/14 5:38 PM


250 Part II: Viral Infections and Related Disorders

177. Gut JP, Spiess C, Schmitt S, et al. Rapid diagnosis of acute mumps infec- 203. Hassan J, Dean J, Moss E, et al. Seroepidemiology of the recent mumps
tion by a direct immunoglobulin M antibody capture enzyme immunoas- virus outbreaks in Ireland. J Clin Virol. 2012;53:320324.
say with labeled antigen. J Clin Microbiol. 1985;21:346352. 204. Centers for Disease Control and Prevention. Mumps outbreak on a uni-
178. Cunningham C, Faherty C, Cormican M, et al. Importance of clinical versity campusCalifornia, 2011. MMWR Morb Mortal Wkly Rep.
features in diagnosis of mumps during a community outbreak. Ir Med J. 2012;61:986989.
2006;99:171173. 205. Dittrich S, Hahne S, van Lier A, et al. Assessment of serological evidence
179. Krause CH, Molyneaux PJ, Ho-Yen DO, et al. Comparison of mumps- for mumps virus infection in vaccinated children. Vaccine. 2011;29:
IgM ELISAs in acute infection. J Clin Virol. 2007;38:153156. 92719275.
180. Sharief MF, Thompson EJ. A sensitive ELISA system for the rapid detec- 206. Utz S, Richard JL, Capaul S, et al. Phylogenetic analysis of clinical mumps
tion of virus specific IgM antibodies in the cerebrospinal fluid. J Immunol virus isolates from vaccinated and non-vaccinated patients with mumps
Methods. 1990;130:1924. during an outbreak, Switzerland 19982000. J Med Virol. 2004;73:
181. Allwinn R, Zeidler B, Steinhagen K, et al. Assessment of mumps 9196.
virus-specific antibodies by different serological assays: which test cor- 207. Rubin SA, Link MA, Sauder CJ, et al. Recent mumps outbreaks in vac-
relates best with mumps immunity? Eur J Clin Microbiol. 2011;30: cinated populations: no evidence of immune escape. J Virol. 2012;86:
12231228. 615620.
182. Gibbs FA, Gibbs EL, Spies HW, et al. Common types of childhood 208. White SJ, Boldt KL, Holditch SJ, et al. Measles, mumps, and rubella. Clin
encephalitis: electroencephalographic and clinical relationships. Arch Obstet Gynecol. 2012;55:550559.
Neurol. 1964;10:111. 209. Atkinson WL, Pickering LK, Schwartz B, et al. General recommenda-
183. Sugita K, Ando M, Minamitani K, et al. Magnetic resonance imaging in a tions on immunization. Recommendations of the Advisory Committee
case of mumps postinfectious encephalitis with asymptomatic optic neuri- on Immunization Practices (ACIP) and the American Academy of Family
tis. Eur J Pediatr. 1991;150:773775. Physicians (AAFP). MMWR Recomm Rep. 2002;51:135.
184. Basekim CC, Kizilkaya E, Pekkafali Z, et al. Mumps epididymo-orchitis: 210. Lavi S, Zimmerman B, Koren G, et al. Administration of measles,
sonography and color Doppler sonographic findings. Abdom Imaging. mumps, and rubella virus vaccine (live) to egg allergic children. JAMA.
2000;25:322325. 1990;263:269271.
185. Erpenbach KH. Systemic treatment with interferon-alpha 2B: an effec- 211. Davidson WL, Buynak EB, Leagus MB, et al. Vaccination of adults with
tive method to prevent sterility after bilateral mumps orchitis. J Urol. live attenuated mumps virus vaccine. JAMA. 1967;201:995998.
1991;146:5456. 212. Kelley PW, Petruccelli BP, Stehr-Green P, et al. The susceptibility of young
186. Rther U, Stilz S, Rhl E, et al. Successful interferon-alpha 2a therapy for adult Americans to vaccine-preventable infections: a national serosurvey
a patient with acute mumps orchitis. Eur Urol. 1995;27:174176. of U.S. Army recruits. JAMA. 1991;266:27242729.
187. Ku JH, Kim YH, Jeon YS, et al. The preventive effect of systemic treat- 213. Rantakallio P, Leskinen M, VonWendt L. Incidence and prognosis of cen-
ment with interferon-alpha2B for infertility from mumps orchitis. BJU tral nervous system infections in a birth cohort of 12,000 children. Scand
Int. 1999;84:839842. J Infect Dis. 1986;18:287297.
188. Yapanoglu T, Kocaturk H, Aksoy Y, et al. Long-term efficacy and safety 214. Peltola H, Heinonen OP, Valle M, et al. The elimination of indigenous
of interferon-alpha-2B in patients with mumps orchitis. Int Urol Nephrol. measles, mumps, and rubella from Finland by a 12-year, two-dose vac-
2010;42:867871. cination program. N Engl J Med. 1994;331:13971402.
189. Wharton M, Cochi SL, Williams WW. Measles, mumps and rubella 215. Lievano F, Galea SA, Thornton M, et al. Measles, mumps, and rubella
vaccines. Infect Dis Clin North Am. 1990;4:4773. virus vaccine (M-M-RII): a review of 32 years of clinical and postmarket-
190. Galazka AM, Robertson SE, Kraigher A. Mumps and mumps vaccine: a ing experience. Vaccine. 2012;30:69186926.
global review. Bull World Health Organ. 1999;77:314. 216. Sawada A, Yamaji Y, Nakayama T. Mumps Hoshino and Torii vac-
191. Centers for Disease Control and Prevention. Prevention of measles, cine strains were distinguished from circulating wild strains. J Infect
rubella, congenital rubella syndrome, and mumps, 2013: summary rec- Chemother. 2013;19:480485.
ommendations of the Advisory Committee on Immunization Practices 217. Fujinaga T, Motegi Y, Tamura H, et al. A prefecture-wide survey of
(ACIP). MMWR Recomm Rep. 2013;62:134. mumps meningitis associated with measles, mumps, and rubella vaccine.
192. Centers for Disease Control and Prevention. Vaccination coverage among Pediatr Infect Dis J. 1991;10:204209.
children in kindergartenUnited States, 2011-12 school year. MMWR 218. McDonald JC, Moore DL, Quennec P. Clinical and epidemiologic fea-
Morb Mortal Wkly Rep. 2012;61:647652. tures of mumps meningoencephalitis and possible vaccine-related disease.
193. Hilleman MR, Buynak EB, Weibel RE, et al. Live, attenuated mumps- Pediatr Infect Dis J. 1989;8:751755.
virus vaccine. N Engl J Med. 1968;278:227232. 219. Dourado I, Cunha S, Teixeira MG, et al. Outbreak of aseptic meningi-
194. Sullivan KM, Halpin TJ, Marks JS, et al. Effectiveness of mumps vaccine tis associated with mass vaccination with a urabe-containing measles-
in a school outbreak. Am J Dis Child. 1985;139:909912. mumps-rubella vaccine: implications for immunization programs. Am J
195. Kim-Farley R, Bart S, Stetler H, et al. Clinical mumps vaccine efficacy. Epidemiol. 2000;151:524530.
Am J Epidemiol. 1985;121:593597. 220. Gray JA, Burns SM. Mumps meningitis following measles, mumps, and
196. Richard JL, Zwahlen M, Feuz M, et al. Comparison of the effectiveness rubella immunisation. Lancet. 1989;2:98.
of two mumps vaccines during an outbreak in Switzerland in 1999 and 221. Brown EG, Furesz J, Dimock K, et al. Nucleotide sequence analysis of
2000: a case-cohort study. Eur J Epidemiol. 2003;18:569577. Urabe mumps vaccine strain that caused meningitis in vaccine recipients.
197. Dominguez A, Torner N, Castilla J, et al. Mumps vaccine effectiveness in Vaccine. 1991;9:840842.
highly immunized populations. Vaccine. 2010;28:35673570. 222. Cizman M, Mozetic M, Radescek-Rakar R, et al. Aseptic meningitis after
198. Snijders BE, van Lier A, van de Kassteele J, et al. Mumps vaccine effective- vaccination against measles and mumps. Pediatr Infect Dis J. 1989;8:
ness in primary schools and households, the Netherlands, 2008. Vaccine. 302308.
2012;30:29993002. 223. Brown EG, Dimock K, Wright KE. The Urabe AM9 mumps vaccine is
199. Vivancos R, Keenan A, Farmer S, et al. An ongoing large outbreak of mea- a mixture of viruses differing at amino acid 335 of the hemagglutinin-
sles in Merseyside, England, January to June 2012. Euro Surveill. 2012;17. neuraminidase gene with one form associated with disease. J Infect Dis.
200. Tan KE, Anderson M, Krajden M, et al. Mumps virus detection during an 1996;174:619622.
outbreak in a highly unvaccinated population in British Columbia. Can J 224. Shah D, Vidal S, Link MA, et al. Identification of genetic mutations
Public Health. 2011;102:4750. associated with attenuation and changes in tropism of Urabe mumps
201. Wielders CC, van Binnendijk RS, Snijders BE, et al. Mumps epidemic in virus. J Med Virol. 2009;81:130138.
orthodox religious low-vaccination communities in the Netherlands and 225. Schlegel M, Osterwalder JJ, Galeazzi RL, et al. Comparative efficacy of
Canada, 2007 to 2009. Euro Surveill. 2011;16. three mumps vaccines during disease outbreak in Eastern Switzerland:
202. Ruijs WL, Hautvast JL, Akkermans RP, et al. The role of schools in the cohort study. BMJ. 1999;319:352.
spread of mumps among unvaccinated children: a retrospective cohort 226. Kilham L. Mumps meningoencephalitis with and without parotitis. Am J
study. BMC Infect Dis. 2011;11:227. Dis Child. 1949;78:324333.

Scheld_Ch16.indd 250 2/21/14 5:38 PM


CHAPTER 17 RABIES
ALAN C. JACKSON

Rabies is an acute viral infection of the central nervous sys- applied in order to demonstrate rabies virus antigens in tissues
tem (CNS) that also involves the peripheral nervous system (9), which became very useful for rabies diagnosis and also
both in its pathogenesis and clinical manifestations. Rabies is a for early rabies pathogenesis studies in animals performed by
zoonotic disease affecting mammals that is usually transmitted Richard Johnson (10) and Frederick Murphy (11,12) and their
to humans by bites from animal vectors. Rabies virus infec- colleagues.
tion causes most cases of rabies, although nonrabies virus
lyssaviruses have been recognized to very rarely cause disease
with identical clinical and pathologic features, but not in the RABIES VIRUS
Americas. Physicians in geographic regions where rabies is
rare must consider the diagnosis based on the clinical picture Rabies virus is in the family Rhabdoviridae and genus
alone, because there may not be a history of an animal expo- Lyssavirus and is a nonsegmented negative-strand (antisense)
sure. Rabies can nearly always be prevented after a recognized RNA virus consisting of 11,932 nucleotides that code for five
exposure with initiation of appropriate prophylactic therapy. viral proteins: nucleocapsid protein (N), matrix protein (M),
Once rabies develops, the disease is almost invariably fatal phosphoprotein (P), glycoprotein (G), and large polymerase
with rare exceptions, and survival has usually been associated protein (L) (13) (Fig. 17.1). Helical genomic RNA forms a ri-
with the administration of rabies vaccine prior to the onset of bonucleoprotein (RNP) core associated with the N, P, and L
the disease. proteins. The RNP serves as a functional template for viral
transcription and replication. The G and M proteins are as-
sociated with a lipid bilayer envelope that surrounds the RNP
HISTORY core. The G protein forms spikelike projections on the sur-
face of the viral envelope and serves as the major surface an-
Rabies has a long and colorful history going back to antiq- tigen of the virus and binds viral neutralizing antibodies and
uity. Perhaps the earliest reference to rabies was in the pre- is important for immunity. Rabies virus belongs to genotype
Mosaic Eshnunna Code of Mesopotamia in about 2300 bc 1 of lyssaviruses, which includes wild type rabies virus strains
(1). Rabid dogs were recognized in China centuries before (also called street rabies viruses) and also laboratory-adapted
the birth of Christ (2). The works of Democritus, Aristotle, strains, including vaccine strains. The nonrabies virus lyssavi-
Hippocrates, and Celsus made reference to rabies in both hu- ruses that have been recognized to rarely cause human disease,
mans and animals (3). In 100 ad, Celsus described human which is indistinguishable from rabies, include Mokola virus
rabies and used the term hydrophobia, which is derived from (genotype 3), Duvenhage virus (genotype 4), European bat lys-
the Greek words meaning fear of water. Celsus recognized savirus 1 (genotype 5), European bat lyssavirus 2 (genotype 6),
that the saliva of the biting animal contained the poisonous Australian bat lyssavirus (genotype 7), and Irkut virus (geno-
agent, and he recommended the practice of using caustics, type pending) (14).
burning, cupping, and also sucking the wounds of individu-
als bitten by rabid dogs in order to prevent the subsequent
development of rabies (3). PATHOGENESIS
In the early nineteenth century, Zinke (4) demonstrated
experimentally that the infectious agent causing rabies was Rabies virus is usually transmitted in the saliva to humans and
transmitted in the saliva by painting saliva from a rabid dog animals via a bite, although a scratch or abrasion with salivary
into incisions made in healthy animals. In 1879, Galtier, who contamination can also result in viral transmission. Aerosol
was working at a veterinary school in Lyon, France, used rab- transmission in a laboratory accident (15,16) and in a cave
bits in his rabies experiments and noted that it was technically containing millions of bats (17) has also been documented but
much less difficult and dangerous than experiments using dogs only occurs very rarely. Organ and tissue (corneal and vascu-
and cats (5). Subsequently, Louis Pasteur took up this experi- lar conduit) transplantation are also well-documented causes
mental rabbit model of rabies. He transmitted rabies virus by of transmission of rabies virus in humans, which account for
inoculating CNS tissues of rabid animals into the brains of a total of 16 well-documented cases (14,18,19). Much has
other animals and noted that sequential brain passages led to been learned about the steps involved in rabies pathogenesis
attenuation for peripheral inoculation (6). In 1885, Pasteur (7) (Fig. 17.2) from experimental studies in animals. Usually, the
successfully immunized a 9-year-old boy, Joseph Meister, who incubation period lasts about 20 to 90 days after the time
had been severely bitten by a rabid dog, with a series of inocu- of the exposure (e.g., bite), although it may vary from a few
lations of infected rabbit spinal cord tissues that had been par- days to over a year. Based on studies in animals, the virus is
tially inactivated after variable periods of desiccation. Joseph thought to remain close to the site of viral entry during most
Meister never developed rabies and, subsequently, many peo- of this incubation period. After an exposure involving muscle,
ple were immunized with nervous system vaccinations in Paris rabies virus is known to bind to nicotinic acetylcholine recep-
and other locations throughout the world. tors (20), which are located in the postsynaptic membrane
In 1903, Adelchi Negri (8) described eosinophilic cytoplas- of the neuromuscular junction. The virus spreads across the
mic inclusions in infected neurons, which are now called Negri synaptic cleft and then spreads centripetally toward the spi-
bodies. Negri bodies proved to be useful for a pathologic di- nal cord in motor nerve fibers of peripheral nerves by retro-
agnosis of rabies. In 1958, fluorescent antibody staining was grade fast axonal transport (21). Bats cause more superficial

251

Scheld_Ch17.indd 251 2/21/14 5:39 PM


252 Part II: Viral Infections and Related Disorders

Glycoprotein (G)
505 a.a. Lipid membrane

Phosphoprotein (P)
297 a.a. (-) viral RNA
12 kb

Matrix protein (M)


RNA-dependent
Nucleoprotein (N) 202 a.a
RNA-polymerase (L)
2130 a.a. 450 a.a.

FIGURE 17.1 Schematic representation of the rabies virus particle. Viral proteins: N for nucleoprotein,
P for phosphoprotein, M for matrix protein, G for glycoprotein, and L for large protein; their length in
amino acids are indicated. The viral membrane is covered by the glycoprotein G, whereas M is located
beneath the membrane. N is bound to the genomic RNA and together with P and L forms the ribonu-
cleoprotein, which constitutes the active viral replication unit. (Reproduced from Albertini AAV, Ruigrok
RW, Blondel D. Rabies virus transcription and replication. Adv Virus Res. 2011;79:122, with permis-
sion. Copyright Elsevier.)

exposures with their bites involving cutaneous and subcuta-


neous tissues, but experimental studies have not yet examined EPIDEMIOLOGY
the detailed pathways of viral spread in animal hosts. After
infecting spinal cord neurons (e.g., ventral horn neurons), ra- Worldwide, about 99% of human rabies cases are related to
bies virus spreads within axons of the CNS by fast axonal transmission from dogs related to the presence of endemic dog
transport along neuroanatomic connections. After CNS infec- rabies, particularly in Asia and Africa. Although the means of
tion is established, there is centrifugal spread of rabies virus controlling dog rabies are very well established, for a variety
to multiple organs along sensory and/or autonomic nerves. In of economic, cultural, and political reasons, dog rabies persists
rabies vectors, viral spread to the salivary glands is important, in many countries, and people in these regions are at a con-
and saliva is secreted containing infectious rabies virus, which tinued risk of transmission via dog bites. In other countries,
is important for transmission to new hosts via bite exposures. rabies is endemic in wildlife, and this poses the main risk for
Viral spread also occurs to multiple organs, including the skin transmission of rabies virus. Rabies virus variants can be iden-
(important for rabies diagnosis using a skin biopsy), heart tified with molecular techniques, including monoclonal anti-
(with myocarditis in some cases), adrenal medulla, and gas- body characterization and reverse transcriptase polymerase
trointestinal tract (22). chain reaction (RT-PCR) amplification with sequencing. Bats
are the most important wildlife rabies vectors, and in the
United States, bat rabies is present in every state except Hawaii
PATHOLOGY (Fig. 17.4). Because of their small size, bat bites may not be
recognized, leaving no opportunity for initiating effective pre-
Characteristic microscopic features of rabies encephalomyeli- ventive measures. Patients infected by bat rabies virus variants
tis include mild mononuclear inflammatory changes involving may not even be aware that they have had contact with bats
the leptomeninges, perivascular regions, and the parenchyma. (Table 17.1). A bat rabies virus variant associated with silver-
There are microglial nodules called Babes nodules in the pa- haired bats and tricolored bats are most frequently associated
renchyma, which were described by Babes (23), that consist with human rabies in the United States and Canada. A variant
of activated microglia and monocytes. Degenerative neuronal associated with Brazilian (Mexican) free-tailed bats is the sec-
changes are not usually prominent in rabies. However, neu- ond most common variant associated with human rabies cases
ronophagia can be observed with accumulations of activated in the United States. Although big brown and little brown bats
microglia/macrophages in the process of phagocytosing de- are often found in houses and they are commonly found to be
generating or dying neurons (24). Infected neurons may con- infected with rabies virus, they are not frequently responsible
tain characteristic eosinophilic inclusions called Negri bodies for human rabies cases.
(Fig. 17.3), which were described by and named after Adelchi Terrestrial animals that are vectors of rabies in North
Negri (8,25). Electron microscopy has demonstrated that America include raccoons, skunks, and foxes (30) (Fig. 17.5).
Negri bodies are composed of large aggregates of granulofila- Raccoon rabies is endemic along the entire eastern coast of
mentous matrix material and variable numbers of viral par- the United States. In the 1940s, raccoon rabies was present in
ticles (24). Florida, and over a period of decades, it gradually spread north

Scheld_Ch17.indd 252 2/21/14 5:39 PM


Chapter 17: Rabies 253

FIGURE 17.2 Schematic diagram


showing the sequential steps in the
pathogenesis of rabies after an animal
bite/peripheral inoculation of rabies
virus. (Reproduced from Jackson AC,
Fu ZF. Pathogenesis. In: Jackson AC,
ed. Rabies: Scientific Basis of the Disease
and Its Management. 3rd ed. Oxford:
Elsevier Academic Press; 2013:299349;
Copyright Elsevier.)

and the first incursion occurred into Ontario, Canada in 1999. with an incubation period as long as 6 years (32). Prodromal
There are only two documented cases of human rabies due to symptoms in rabies are nonspecific and include fever, chills,
a raccoon rabies virus variant (19,31). Skunk rabies is present malaise, fatigue, insomnia, anorexia, headache, anxiety, and
in midwestern states, the prairie provinces of Canada, and in irritability. They may last for up to 10 days prior to the onset
California. Fox rabies is now uncommon in North America of neurologic symptoms. The earliest neurologic features of
because it has been well controlled, particularly in Ontario rabies include paresthesias, pain, and pruritus at or close to
(red fox) and Texas (gray fox), and in Europe with oral vac- the site of exposure, which likely reflects infection and inflam-
cination programs. Companion animals, especially dogs and matory changes in local sensory ganglia (e.g., dorsal root gan-
cats, are also at risk of developing rabies transmitted from glia or cranial sensory ganglia). By this time, the wound may
wildlife vectors, and they may then pose a danger to humans. have completely healed. There are two clinical forms of disease
in rabies: encephalitic rabies (in 80% of cases) and paralytic
rabies (in 20% of cases). The main burden of the infection
CLINICAL FEATURES in encephalitic rabies involves the brain, whereas in paralytic
rabies, the main burden likely involves the spinal cord, nerve
Typically, the incubation period from the time of the expo- roots, and peripheral nerves. Fever occurs in both forms.
sure until the time of the onset of clinical disease is between In encephalitic rabies, there may be episodes of generalized
20 and 90 days but may be as short as only a few days or arousal or hyperexcitability, which are separated by lucid pe-
exceed a year. There is a well-documented report of rabies riods (33). Patients may have aggressive behavior, confusion,

Scheld_Ch17.indd 253 2/21/14 5:39 PM


254 Part II: Viral Infections and Related Disorders

FIGURE 17.3 Hematoxylin and eosinstained


sections showing Negri bodies in the perikarya of
(AC) cerebellar Purkinje cells and (D, E) pyrami-
dal neurons in the cerebral cortex of human rabies
cases. The arrow in (C) indicates a Negri body
in an apical dendrite. (Magnifications: A, 355;
B, 515; C, 615; D, 820; E, 970). (Adapted
from Rossiter JP, Jackson AC. Pathology. In:
Jackson AC, ed. Rabies: Scientific Basis of the
Disease and Its Management. 3rd ed. Oxford:
Elsevier Academic Press; 2013:351386; Copyright
Elsevier.)

and hallucinations. Features of autonomic dysfunction are difficulty swallowing. When they attempt to swallow, they ex-
common, and these may include hypersalivation, piloerection perience contractions of the diaphragm and other inspiratory
(gooseflesh), sweating, priapism, and cardiac arrhythmias. muscles, typically lasting for 5 to 15 seconds. Subsequently,
Hydrophobia is a characteristic clinical feature of encepha- this may become a conditioned reflex and the sight, sound,
litic rabies and occurs more frequently with infections due or even mentioning water (or other liquids) may trigger the
to rabies virus variants associated with dogs than with bats. spasms. Aerophobia is the occurrence of these same spasms
Patients may initially experience pain in the throat or have precipitated by a draft of air on the skin. There is progressive

FIGURE 17.4 Reported cases of ra-


bies involving bats, by county, in 2011.
Histogram represents the number of
counties in each category for total
number of bats submitted for testing
(excluding Oklahoma). (Reproduced
from Blanton JD, Dyer J, McBrayer
J, et al. Rabies surveillance in the
United States during 2011. J Am Vet
Med Assoc. 2012;241:712722, with
permission.)

Scheld_Ch17.indd 254 2/21/14 5:39 PM


Chapter 17: Rabies 255

TA B L E 1 7 . 1 become infected with rabies virus, and in some cases, there


is an associated myocarditis (3436). Respiratory complica-
INDIGENOUSLY ACQUIRED CASES OF HUMAN tions include hyperventilation, hypoxemia, respiratory depres-
RABIES FROM BATS IN THE UNITED STATES AND sion with apnea, atelectasis, and aspiration pneumonia (37).
CANADA, 19502011a Hyperthermia or hypothermia may occur, likely secondary
to hypothalamic infection. Endocrine complications include
Type of Case Number of Cases inappropriate secretion of antidiuretic hormone and diabetes
insipidus (37,38). Multiple organ failure commonly occurs in
Bite or scratch 23 (36.5%) patients treated aggressively in critical care units.
Direct contact with no 11 (17.5%)
recognized bite
House exposure but no 8 (12.7%) DIFFERENTIAL DIAGNOSIS
direct contact
No history of bat contact 21 (33.3%) The diagnosis of rabies may be difficult without a history of an
Total 63
animal exposure. Physicians may not ask about animal expo-
sures, and the patient may not recall an exposure or may not
a
be able to provide this information at the time of presentation.
Data from De Serres et al. (26), Pue et al. (27), and
Blanton et al. (2830).
In early phases, encephalitic rabies may be misdiagnosed as a
Adapted from Jackson AC. Update on rabies. Res Rep Trop Med. psychiatric disorder and paralytic rabies as Guillain-Barr syn-
2011;2:3143, with permission from Dove Medical Press. drome. Rabies hysteria is a conversion disorder (somatoform
disorder) that likely occurs as a psychologic response to the
fear of developing rabies (39). It is characterized by a shorter
incubation period than rabies, aggressive behavior (not com-
mon in humans), inability for the patient to communicate, and
neurologic deterioration with worsening in the level of con- a long clinical course with recovery.
sciousness to coma and the development of paralysis. Other viral encephalitides may show behavioral changes
In paralytic rabies, there is early prominent weakness that with fluctuations in the level of consciousness. Hydrophobic
usually initially involves the bitten extremity and progresses spasms are not observed, and the presence of brainstem signs
to involve the other extremities and facial muscles. Sphincter is unusual in conscious patients in most of the other viral
involvement, pain, and sensory disturbances also occur. encephalitides. Herpes simiae (B virus) encephalomyelitis,
Hydrophobia is unusual in paralytic rabies, although weak- which is transmitted by monkey bites, is usually associated
ness of bulbar and respiratory muscles also develops. Patients with a shorter incubation period and recovery may occur (40)
with paralytic rabies later develop neurologic deterioration (see Chapter 14). Tetanus has a shorter incubation period
with progression to coma, and they typically survive longer (3 to 21 days) than rabies and is characterized by sustained
than patients with encephalitic rabies. muscle rigidity involving paraspinal, abdominal, masseter
Medical complications are common in rabies patients (trismus), laryngeal, and respiratory muscles with superim-
treated aggressively in a critical care unit. Cardiac and respi- posed brief recurrent muscle spasms (41) (see Chapter 37).
ratory complications are common. Cardiac disorders include In tetanus, consciousness is preserved, there is no cerebrospi-
heart failure, hypotension, a variety of arrhythmias, and car- nal fluid (CSF) pleocytosis, and the prognosis is much better
diac arrest. Both cardiac ganglia and the myocardium may than in rabies. In Africa, rabies is commonly misdiagnosed
as cerebral malaria (42). Anti-N-methyl-d-aspartate recep-
tor (anti-NMDA) encephalitis occurs in young patients (es-
pecially females) and is characterized by behavioral changes,
autonomic instability, hypoventilation, and seizures, and it has
recently been recognized that this autoimmune disease rivals
viral etiologies as a cause of encephalitis (43). Postvaccinal
encephalomyelitis is an important differential diagnosis in pa-
tients immunized with a vaccine derived from neural tissues
(e.g., Semple vaccine), which is currently used in only a few
resource-poor countries. Patients with paralytic rabies may
resemble the Guillain-Barr syndrome, and the pathologic fea-
tures may also be similar (44). Local symptoms at the site of
the bite, piloerection, early or persistent bladder dysfunction,
and fever are all more suggestive of paralytic rabies.

INVESTIGATIONS
Routine blood tests and computed tomography (CT) head
scans are typically normal in rabies. Magnetic resonance
(MR) imaging may be normal or show signal abnormalities
in the brain, spinal cord, and nerve roots/plexuses, but these
FIGURE 17.5 Distribution of the major rabies virus variants among
findings are not specific for rabies and the main usefulness of
wild terrestrial reservoirs in the United States and Puerto Rico, 2008 MR imaging is to exclude other diagnostic possibilities (45).
to 2012. *Potential host shift event. (Reproduced from Dyer JL, CSF analysis usually shows a mononuclear pleocytosis with a
Wallace R, Orciari L, et al. Rabies surveillance in the United States cell count of less than 100 cells/L. Serum-neutralizing anti
during 2012. J Am Vet Med Assoc. 2013;243:805815.) rabies virus antibodies may develop in unvaccinated patients

Scheld_Ch17.indd 255 2/21/14 5:39 PM


256 Part II: Viral Infections and Related Disorders

but may not appear for a week or more during the clinical and, if available, a virucidal agent (e.g., povidone) should be
course of disease, and some patients never develop antibodies used to irrigate the wounds. In the United States, four doses
prior to death. Neutralizing antirabies virus antibodies may of rabies vaccine, which was recently reduced from five doses,
also develop in the CSF, whereas CSF antibodies are not pres- are recommended on days 0, 3, 7, and 14 (50), and for each
ent in vaccinated patients who do not have rabies encephali- dose, 1.0 mL of vaccine should be given intramuscularly in
tis. Specific laboratory tests for confirmation of a diagnosis the deltoid muscle. Two rabies vaccines are currently licensed
of rabies include a full-thickness skin biopsy taken from the in the United States and Canada: purified chick embryo cell
posterior region of the neck at the hairline. Rabies virus anti- vaccine (PCECV) (RabAvert) and human diploid cell vaccine
gen may be detected in nerve fibers around hair follicles with (Imovax). Pregnancy is not a contraindication for immuniza-
direct fluorescent antibody staining. A recent advance in the tion. Local and mild systemic adverse effects are common.
laboratory diagnosis of rabies is detection of rabies virus RNA Local reactions include pain, erythema, edema, and pruritus;
in fluids or tissues using RT-PCR amplification. Detection of systemic reactions include fever, myalgias, headache, and nau-
rabies virus RNA in saliva is the most useful. RT-PCR can sea. Antiinflammatory medications and antipyretics may be
also be used on skin biopsies (46) and CSF but is much less used, but immunization should not be discontinued. The dose
sensitive on CSF. A negative laboratory test for rabies never of HRIG is based on weight (20 IU/kg), and HRIG should
excludes rabies unless performed on brain tissues, and the tests be infiltrated into and around the wound and the remaining
may need to be repeated for diagnostic confirmation of a ra- portion of the dose can be given intramuscularly in a differ-
bies diagnosis. Brain tissues are only very rarely obtained by ent location (e.g., gluteal muscles) than the vaccine is given. If
biopsy antemortem but are routinely evaluated postmortem by there are multiple or extensive wounds and a large volume of
direct fluorescent antibody staining and by culture techniques. HRIG is needed for infiltration, then HRIG can be diluted as
required for satisfactory wound infiltration. HRIG should not
be given later than 7 days after the first dose of rabies vaccine.
Adverse effects of HRIG include local pain and low-grade
PREVENTION OF RABIES fever. If HRIG is not available, then purified equine rabies im-
Rabies can be very effectively prevented after recognized ex- mune globulin, which is much more readily available in some
posures. Detailed guidelines that are periodically updated are rabies-endemic countries (e.g., Thailand), may be used in the
available from the Centers for Disease Control and Prevention same manner at a dose of 40 IU/kg.
(47) and from the World Health Organization (48), and these In persons at risk of rabies exposures, including laboratory
documents are available on the Morbidity and Mortality workers, veterinarians, and travelers to places with endemic
Weekly Report (http://www.cdc.gov/mmwr/) and World dog rabies (e.g., Asia and Africa), preexposure rabies immuni-
Health Organization (http://www.who.int/en/) Web sites. zation should be considered. Three doses of vaccine are given
Algorithms can be very useful in making decisions concerning on days 0, 7, and 21. When prolonged protection is needed,
postexposure rabies prophylaxis (Fig. 17.6). The first step is to booster doses of rabies vaccine can be given periodically as re-
determine whether there is a real risk of rabies virus transmis- quired based on a serum-neutralizing antirabies antibody titer.
sion, which depends on obtaining the details of the exposure, After a rabies exposure in preimmunized individuals, in addi-
the species of animal involved, and also on knowledge about tion to wound cleansing, two doses of rabies vaccine should be
the local epidemiologic situation. Advice from local public given on days 0 and 3 and HRIG should not be given.
health officials can be very helpful in determining whether
postexposure rabies prophylaxis measures should be initiated.
Laboratory testing on brain tissues from an animal is needed THERAPY OF HUMAN RABIES
for a definitive diagnosis of rabies, which is usually performed
by an antigen detection method using the fluorescent antibody There is no effective therapy for human rabies, and the disease
technique. If a dog, cat, or ferret remains healthy for a 10-day is virtually always fatal. In all except a single case, survivors
period after an exposure, then a confident conclusion can be have received rabies vaccine prior to the onset of the clinical
made that rabies virus transmission did not occur during the disease. In 2003, a viewpoint article was published outlining
exposure because the brainstem infection associated with the the therapeutic options for consideration of an aggressive ap-
salivary excretion of infectious virus would have progressed proach for a patient with rabies (51). Young and previously
to overt clinical signs within the period. Of course, unwanted healthy patients with an early clinical diagnosis of rabies were
animals may be tested without an observation period. Other felt to be the best potential candidates for aggressive therapy
animals must not be observed after an exposure because there (51). Therapies suggested for consideration include rabies vac-
is uncertainty about the period of time for clinical disease to cine, HRIG, monoclonal antibodies (for the future), ribavirin,
develop, and this period may substantially exceed 10 days. If interferon-, and ketamine. It was felt that a combination of
an animal escapes after an exposure, then it should be consid- therapies might improve efficacy in situations in which specific
ered rabid unless information from public health officials indi- therapies used individually had failed in the past, similar to the
cates that this is unlikely. Current recommendations indicate situation for current therapies of a variety of infectious and
that the physical presence of a bat may warrant postexposure other noninfectious diseases.
prophylaxis when a person such as a small child or sleeping In 2004, a 15-year-old female who had been bitten on her
adult is unable to reliably report contact that could have re- finger by a bat and did not receive postexposure prophylaxis
sulted in a bite (47). However, in light of the low risks and therapy survived rabies (52). She developed typical clinical fea-
high costs, recommendations for bedroom exposures to a bat tures of rabies encephalitis about a month after the bite. On
while sleeping and without known physical contact have been arrival at a tertiary care hospital in Milwaukee, Wisconsin, neu-
questioned (49). tralizing antirabies virus antibodies were detected in sera and
Postexposure rabies prophylaxis in previously unvacci- CSF (initially at titers of 1:102 and 1:47, respectively). Nuchal
nated persons includes wound cleansing and active immu- skin biopsies were negative for rabies virus antigen, and rabies
nization with rabies vaccine and passive immunization with virus RNA was not detected in saliva or in the skin biopsies
human rabies immune globulin (HRIG). All animal bite using RT-PCR. She was intubated and put into a drug-induced
wounds should be thoroughly cleaned with soap and water coma, which included the noncompetitive NMDA antagonist

Scheld_Ch17.indd 256 2/21/14 5:39 PM


Chapter 17: Rabies 257

Did an exposure to rabies occur?


Did an animal bite the patient, or did
a potentially unrecognized exposure occur?
Was there direct contact of the patients open
bleeding wound, broken skin, or mucous
membranes with potentially infectious Yes
Was the animal a mammal?
material such as animal saliva or central
nervous system tissue?
No Yes
No

Yes
No PEP Was the animal a small rodent or rabbit?

No

Observe the animal for Yes Yes


Is the animal available
10 days. Does it exhibit Was the animal a dog, cat, or ferret?
for observation?
signs of rabies?

No
No

Yes
Was the animal domestic livestock?
No Yes Consult public health
officials for local
rabies epidemiology.
Was the animal No
a bat, or is
terrestrial rabies
present? No
Is the animal (brain) available for testing?

Yes

No Yes

Did the animal Negative for Positive for


exhibit any rabies rabies
signs of rabies?

No Yes

No PEP PEP No PEP PEP No PEP PEP

FIGURE 17.6 Algorithm for rabies postexposure prophylaxis (PEP) in the United States. (Reproduced
from Rupprecht CE, Gibbons RV. Prophylaxis against rabies. N Engl J Med. 2004;351:26262635, with
permission.)

ketamine at 48 mg/kg/day as a continuous infusion and intra- This patient is the first documented rabies survivor who
venous midazolam for 7 days. A burst-suppression pattern on did not receive any rabies vaccine prior to the onset of clini-
her electroencephalogram was maintained, and supplemental cal rabies (Table 17.2). It remains uncertain if therapy with
phenobarbital was given as needed. She also received antivi- one or more specific agents played a significant role in her fa-
ral therapy, including intravenous ribavirin and amantadine vorable outcome (60). Since that time, there have been over
200 mg per day administered enterally. She improved and was 25 cases in which the main components of this approach (the
discharged from hospital with neurologic deficits, and she sub- Milwaukee protocol) have been used, and the therapy failed
sequently demonstrated further neurologic improvement (53). with fatal outcomes (61). The induction of coma per se has no

Scheld_Ch17.indd 257 2/21/14 5:39 PM


258 Part II: Viral Infections and Related Disorders

TA B L E 1 7 . 2
CASES OF HUMAN RABIES WITH RECOVERY

Age of
Location Year Patient Transmission Immunization Prior to Onset Outcome Reference

United States 1970 6 Bat bite Duck embryo vaccine Complete recovery (54)
Argentina 1972 45 Dog bites Suckling mouse brain vaccine Mild sequelae (55)
United States 1977 32 Laboratory (vaccine strain) Preexposure vaccination Sequelae (16,56)
Mexico 1992 9 Dog bites Postexposure vaccination Severe sequelaea (57)
(combination)
India 2000 6 Dog bites Postexposure vaccination Severe sequelaeb (58)
(combination)
United States 2004 15 Bat bite None Mild sequelae (52,53)
Brazil 2008 15 Vampire bat bite Postexposure vaccination Severe sequelae (59)

a
Patient died less than 4 years after developing rabies with marked neurologic sequelae (Dr. L. Alvarez, personal communication, 1997).
b
Patient died about 2 years after developing rabies with marked neurologic sequelae (Dr. S. Mahusudana, personal communication, 2005).
(Adapted from Jackson AC. Therapy of human rabies. In: Jackson AC, ed. Rabies: Scientific Basis of the Disease and Its Management. 3rd ed. Oxford:
Elsevier Academic Press; 2013:573587; Copyright Elsevier.)

established benefit for the management of infectious diseases and tissues, and brain tissues have not been tested. This may
of the nervous system, and there is no evidence to date sup- reflect effective viral clearance in which centrifugal spread of
porting this approach in rabies or other viral encephalitides. the infection to peripheral organ sites is reduced or very rapid
Hence, therapeutic coma should not become a routine therapy clearance occurs through immune-mediated mechanisms.
for the management of rabies. Recent experimental evidence Pathologic data from a number of human rabies cases treated
does not support a mechanism of excitotoxicity in a mouse with the Milwaukee protocol indicate that the therapy is inef-
model of rabies and also in rabies virus infection of cultured fective in clearing rabies virus infection from the brain and from
neurons, and there was also a lack of efficacy of ketamine ther- preventing neuronal injury. A case from Edmonton (Canada)
apy in cultured neurons and in the mouse model (62). Even in was treated with the Milwaukee protocol and after termination
situations in which there is very strong experimental evidence of the therapeutic coma remained in a brain deathlike state for
of excitotoxicity in animal models, numerous clinical trials in about 4 weeks (68). At autopsy, there was complete loss of neu-
humans have failed to demonstrate efficacy of neuroprotec- rons in the cerebral cortex, and positive staining for rabies virus
tive agents in stroke (63). Hence, a neuroprotective effect of a antigen was observed in both brainstem and cerebellar neurons,
therapy given to a single patient without a credible scientific indicating a failure of clearance of the viral infection from the
rationale is highly doubtful. It is likely that this patient would brain and also failure of protection against neuronal injury and
also have recovered with only good supportive therapy. loss (68). In Germany, lung and kidney/pancreas recipients from
Neutralizing antirabies virus antibodies are an important a rabies virusinfected donor developed rabies and were treated
marker of an adaptive immune response that is essential for with major components of the Milwaukee protocol, including
clearance of rabies virus and recovery (64). The presence of intravenous midazolam, ketamine, and phenobarbital (in one)
serum-neutralizing antirabies virus antibodies early in a pa- (19). One patient died within 2 days, whereas the other survived
tients clinical course probably occurs in less than 20% of pa- 64 days after the onset of clinical rabies. At autopsy, the two
tients with rabies and is likely an important factor contributing patients had 1.2 to 2.3 109 RNA copies per mg of CNS tis-
to a favorable outcome in this patient. There have been six sur- sue, indicating ineffective viral clearance. The long surviving pa-
vivors of rabies who received rabies vaccine prior to the onset tient showed viral clearance from systemic organs and peripheral
of their disease and only one without vaccine. This suggests nerve. Hence, Milwaukee protocol therapy has proved ineffective
that an early immune response is associated with a positive in promoting viral clearance from the CNS in rabies. It remains
outcome. Recovery of cases with atypical clinical features of highly doubtful that the Milwaukee protocol will prove to be
rabies without the development of antirabies virus neutraliz- useful in the management of human rabies. Unfortunately, pro-
ing antibodies (65,66) were probably not actual cases of rabies motion and repetition of this flawed therapy has likely already
and should not be considered survivors. Bat rabies virus vari- impeded progress in the development of new effective therapies
ants are probably less neurovirulent than canine virus variants for rabies. A better understanding of basic mechanisms underly-
or other variants that are responsible for most human cases of ing rabies pathogenesis in humans and animals is needed, which
rabies (67), and human rabies due to canine rabies virus vari- may prove to be very helpful in the development of novel thera-
ants likely has a reduced chance of a favorable outcome than peutic approaches for the management of this dreaded disease.
cases caused by bat rabies virus variants. One previous sur-
vivor of rabies, who was also infected with a bat rabies virus
variant received rabies vaccine prior to the onset of disease OTHER LYSSAVIRUS INFECTIONS
and made an excellent neurologic recovery (54). It is unknown
if the causative bat rabies virus variant in the Milwaukee case Nonrabies virus lyssaviruses may cause fatal neurologic ill-
was attenuated and had different biologic properties than other nesses that are clinically and pathologically indistinguishable
isolated variants because there was no viral isolation in this from rabies. Mokola virus has been isolated from shrews,
case. However, in rabies survivors, diagnostic laboratory tests although the reservoir is unknown. In 1971, a 6-year-old girl
are usually negative for rabies virus antigen and RNA in fluids died with Mokola virus infection (69); another case with mild

Scheld_Ch17.indd 258 2/21/14 5:39 PM


Chapter 17: Rabies 259

illness was more likely due to cross-contamination of specimens (76,77). In 1996, 1998, and 2013, cases due to Australian bat
in the laboratory (70). The index case of Duvenhage virus infec- lyssavirus were likely transmitted by a fruit-eating bat (flying
tion was transmitted by a bat and occurred in South Africa (71), fox) and an insect-eating bat, respectively (7880). In 2007, a
and two additional cases were recently reported (72,73). There 20-year-old female died in the Primorye Territory, which is in
have been two cases reported due to European bat lyssavirus 1 the Russian Far East, due to Irkut virus, which had been previ-
(74,75) and another two cases due to European bat lyssavirus 2 ously isolated from a greater tube-nosed bat (81).

Suggested Readings
Baer GM. The Natural History of Rabies. 2nd ed. Boca Raton, FL: CRC Press; 1991. Jackson AC. Rabies: Scientific Basis of the Disease and Its Management. 3rd ed.
Baer GM, Neville J, Turner GS. Rabbis and Rabies: A Pictorial History of Oxford: Elsevier Academic Press; 2013.
Rabies through the Ages. Mexico City, Mexico: Laboratorios Baer; 1996. Meslin FX, Kaplan MM, Koprowski H. Laboratory Techniques in Rabies. 4th
Jackson AC. Update on rabies. Res Rep Trop Med. 2011;2:3143. ed. Geneva: World Health Organization; 1996.
Jackson AC. Research advances in rabies. In: Jackson AC, ed. Advances in Virus Schnell MJ, McGettigan JP, Wirblich C, et al. The cell biology of rabies virus:
Research. Vol 79. London: Elsevier Academic Press; 2011. using stealth to reach the brain. Nat Rev Microbiol. 2010;8:5161.

References
1. Rosner F. Rabies in the Talmud. Med Hist. 1974;18:198200. 26. De Serres G, Dallaire F, Cote M, et al. Bat rabies in the United States and
2. Wilkinson L. Understanding the nature of rabies: an historical perspective. Canada from 1950 through 2007: human cases with and without bat con-
In: Campbell JB, Charlton KM, eds. Rabies. Boston: Kluwer Academic tact. Clin Infect Dis. 2008;46:13291337.
Publishers; 1988:123. 27. Pue HL, Turabelidze G, Patrick S, et al. Human rabiesMissouri, 2008.
3. Fleming G. Rabies and Hydrophobia: Their History, Nature, Causes, MMWR Morb Mortal Wkly Rep. 2009;58:12071209.
Symptoms, and Prevention. London: Chapman & Hall; 1872. 28. Blanton JD, Palmer D, Rupprecht CE. Rabies surveillance in the United
4. Zinke GG. Neue Ansichten der Hundswuth; ihrer Ursachen und Folgen, States during 2009. J Am Vet Med Assoc. 2010;237:646657.
nebst einer sichern Behandlungsart der von tollen Thieren gebissenen 29. Blanton JD, Palmer D, Dyer J, et al. Rabies surveillance in the United States
Menschen. Gabler Jena. 1804;16:212234. during 2010. J Am Vet Med Assoc. 2011;239:773783.
5. Wilkinson L. The development of the virus concept as reflected in corpora 30. Blanton JD, Dyer J, McBrayer J, et al. Rabies surveillance in the United
of studies on individual pathogens: rabiestwo millennia of ideas and con- States during 2011. J Am Vet Med Assoc. 2012;241:712722.
jecture on the aetiology of a virus disease. Med Hist. 1977;21:1531. 31. Silverstein MA, Salgado CD, Bassin S, et al. First human death associated
6. Pasteur L, Chamberland CE, Roux E. Physiologie experimentaleNouvelle with raccoon rabiesVirginia, 2003. MMWR Morb Mortal Wkly Rep.
communication sur la rage. C R Acad Sci. 1884;98:457463. 2003;52:11021103.
7. Pasteur L. Mthode pour prvenir la rage aprs morsure. C R Acad Sci. 32. Smith JS, Fishbein DB, Rupprecht CE, et al. Unexplained rabies in three
1885;101:765774. immigrants in the United States: a virologic investigation. N Engl J Med.
8. Negri A. Beitrag zum studium der aetiologie der tollwuth. Z Hyg 1991;324:205211.
Infektionskr. 1903;43:507528. 33. Warrell DA. The clinical picture of rabies in man. Trans R Soc Trop Med
9. Goldwasser RA, Kissling RE. Fluorescent antibody staining of street and Hyg. 1976;70:188195.
fixed rabies virus antigens. Proc Soc Exp Biol Med. 1958;98:219223. 34. Cheetham HD, Hart J, Coghill NF, et al. Rabies with myocarditis: two
10. Johnson RT. Experimental rabies: studies of cellular vulnerability and cases in England. Lancet. 1970;1:921922.
pathogenesis using fluorescent antibody staining. J Neuropathol Exp 35. Raman GV, Prosser A, Spreadbury PL, et al. Rabies presenting with myo-
Neurol. 1965;24:662674. carditis and encephalitis. J Infect. 1988;17:155158.
11. Murphy FA, Bauer SP, Harrison AK, et al. Comparative pathogenesis of 36. Ross E, Armentrout SA. Myocarditis associated with rabies: report of a
rabies and rabies-like viruses: viral infection and transit from inoculation case. N Engl J Med. 1962;266:10871089.
site to the central nervous system. Lab Invest. 1973;28:361376. 37. Hattwick MAW. Human rabies. Public Health Rev. 1974;3:229274.
12. Murphy FA, Harrison AK, Winn WC, et al. Comparative pathogenesis of 38. Bhatt DR, Hattwick MAW, Gerdsen R, et al. Human rabies: diagnosis,
rabies and rabies-like viruses: infection of the central nervous system and complications, and management. Am J Dis Child. 1974;127:862869.
centrifugal spread of virus to peripheral tissues. Lab Invest. 1973;29:116. 39. Wilson JM, Hettiarachchi J, Wijesuriya LM. Presenting features and diag-
13. Wunner WH, Conzelmann KK. Rabies virus. In: Jackson AC, ed. Rabies: nosis of rabies. Lancet. 1975;2:11391140.
Scientific Basis of the Disease and Its Management. 3rd ed. Oxford, United 40. Whitley RJ. B virus. In: Scheld WM, Whitley RJ, Marra CM, eds. Infections
Kingdom: Elsevier Academic Press; 2013:1760. of the Central Nervous System. 4th ed. Philadelphia: Lippincott Williams
14. Jackson AC. Human disease. In: Jackson AC, ed. Rabies: Scientific Basis & Wilkins; 2014:204209.
of the Disease and Its Management. 3rd ed. Oxford, United Kingdom: 41. Brook I. Tetanus. In: Scheld WM, Whitley RJ, Marra CM, eds. Infections
Elsevier Academic Press; 2013:269298. of the Central Nervous System. 4th ed. Philadelphia: Lippincott Williams
15. Winkler WG, Fashinell TR, Leffingwell L, et al. Airborne rabies transmis- & Wilkins; 2014:634658.
sion in a laboratory worker. JAMA. 1973;226:12191221. 42. Mallewa M, Fooks AR, Banda D, et al. Rabies encephalitis in malaria-
16. Tillotson JR, Axelrod D, Lyman DO. Rabies in a laboratory workerNew endemic area, Malawi, Africa. Emerg Infect Dis. 2007;13:136139.
York. MMWR Morb Mortal Wkly Rep. 1977;26:183184. 43. Gable MS, Sheriff H, Dalmau J, et al. The frequency of autoimmune
17. Constantine DG. Rabies transmission by nonbite route. Pub Health Rep. N-methyl-D-aspartate receptor encephalitis surpasses that of individual
1962;77:287289. viral etiologies in young individuals enrolled in the California Encephalitis
18. Maier T, Schwarting A, Mauer D, et al. Management and outcomes after Project. Clin Infect Dis. 2012;54:899904.
multiple corneal and solid organ transplantations from a donor infected 44. Sheikh KA, Ramos-Alvarez M, Jackson AC, et al. Overlap of pathology
with rabies virus. Clin Infect Dis. 2010;50:11121119. in paralytic rabies and axonal Guillain-Barr syndrome. Ann Neurol.
19. Vora NM, Basavaraju SV, Feldman KA, et al. Raccoon rabies virus variant 2005;57:768767.
transmission through solid organ transplantation. JAMA. 2013;310:398407. 45. Laothamatas J, Sungkarat W, Hemachudha T. Neuroimaging in rabies. Adv
20. Lentz TL, Burrage TG, Smith AL, et al. Is the acetylcholine receptor a ra- Virus Res. 2011;79:309327.
bies virus receptor? Science. 1982;215:182184. 46. Dacheux L, Reynes JM, Buchy P, et al. A reliable diagnosis of human rabies
21. Tsiang H. Evidence for an intraaxonal transport of fixed and street rabies based on analysis of skin biopsy specimens. Clin Infect Dis. 2008;47:
virus. J Neuropathol Exp Neurol. 1979;38:286296. 14101417.
22. Jackson AC, Ye H, Phelan CC, et al. Extraneural organ involvement in 47. Manning SE, Rupprecht CE, Fishbein D, et al. Human rabies prevention
human rabies. Lab Invest. 1999;79:945951. United States, 2008: recommendations of the Advisory Committee on
23. Babes VM. Sur certains caracteres des lesions histologiques de la rage. Immunization Practices. MMWR Recomm Rep. 2008;57(RR3):128.
Annales de LInstitut Pasteur. 1892;6:209223. 48. World Health Organization. WHO Expert Consultation on Rabies: Second
24. Rossiter JP, Jackson AC. Pathology. In: Jackson AC, ed. Rabies: Scientific Report (WHO technical report series; no. 982). Geneva: World Health
Basis of the Disease and Its Management. 3rd ed. Oxford, United Kingdom: Organization; 2013.
Elsevier Academic Press; 2013:351386. 49. De Serres G, Skowronski DM, Mimault P, et al. Bats in the bedroom, bats in
25. Negri A. Zur aetiologie der tollwuth. Die diagnose der tollwuth auf grund the belfry: reanalysis of the rationale for rabies postexposure prophylaxis.
der neuen befunde. Z Hyg Infektionskr. 1903;44:519. Clin Infect Dis. 2009;48:14931499.

Scheld_Ch17.indd 259 2/21/14 5:39 PM


260 Part II: Viral Infections and Related Disorders

50. Rupprecht CE, Briggs D, Brown CM, et al. Use of a reduced (4-dose) 67. Lafon M. Bat rabiesthe Achilles heel of a viral killer? Lancet. 2005;366:
vaccine schedule for postexposure prophylaxis to prevent human rabies: 876877.
recommendations of the Advisory Committee on Immunization Practices. 68. McDermid RC, Saxinger L, Lee B, et al. Human rabies encephalitis follow-
MMWR Recomm Rep. 2010;59:19. ing bat exposure: failure of therapeutic coma. CMAJ. 2008;178:557561.
51. Jackson AC, Warrell MJ, Rupprecht CE, et al. Management of rabies in 69. Familusi JB, Osunkoya BO, Moore DL, et al. A fatal human infection with
humans. Clin Infect Dis. 2003;36:6063. Mokola virus. Am J Trop Med Hyg. 1972;21:959963.
52. Willoughby RE Jr, Tieves KS, Hoffman GM, et al. Survival after treatment 70. Familusi JB, Moore DL. Isolation of a rabies related virus from the cere-
of rabies with induction of coma. N Engl J Med. 2005;352:25082514. brospinal fluid of a child with aseptic meningitis. Afr J Med Sci. 1972;3:
53. Hu WT, Willoughby RE Jr, Dhonau H, et al. Long-term follow-up after 9396.
treatment of rabies by induction of coma [letter]. N Engl J Med. 2007;357: 71. Meredith CD, Prossouw AP, Koch Hv. An unusual case of human rabies
945946. thought to be of chiropteran origin. S Afr Med J. 1971;45:767769.
54. Hattwick MAW, Weis TT, Stechschulte CJ, et al. Recovery from rabies: a 72. Paweska JT, Blumberg LH, Liebenberg C, et al. Fatal human infection with
case report. Ann Intern Med. 1972;76:931942. rabies-related Duvenhage virus, South Africa. Emerg Infect Dis. 2006;12:
55. Porras C, Barboza JJ, Fuenzalida E, et al. Recovery from rabies in man. 19651967.
Ann Intern Med. 1976;85:4448. 73. van Thiel PP, de Bie RM, Eftimov F, et al. Fatal human rabies due to
56. Tillotson JR, Axelrod D, Lyman DO. Follow-up on rabiesNew York. Duvenhage virus from a bat in Kenya: failure of treatment with coma
MMWR Morb Mortal Wkly Rep. 1977;26:249250. -induction, ketamine, and antiviral drugs. PLoS Negl Trop Dis.
57. Alvarez L, Fajardo R, Lopez E, et al. Partial recovery from rabies in a nine- 2009;3:e428.
year-old boy. Pediatr Infect Dis J. 1994;13:11541155. 74. Selimov MA, Tatarov AG, Botvinkin AD, et al. Rabies-related Yuli virus;
58. Madhusudana SN, Nagaraj D, Uday M, et al. Partial recovery from rabies Identification with a panel of monoclonal antibodies. Acta Virol. 1989;33:
in a six-year-old girl [letter]. Int J Infect Dis. 2002;6:8586. 542546.
59. Ministerio da Saude in Brazil. Rabies, human survival, batBrazil: 75. Botvinkin AD, Selnikova OP, Antonova LA, et al. Human rabies case
(Pernambuco). ProMED-mail. 2008;20081114.3599. http://www.promed caused from a bat bite in Ukraine. Rabies Bull Europe. 2005;29:57.
mail.org. Accessed November 23, 2012. 76. Roine RO, Hillbom M, Valle M, et al. Fatal encephalitis caused by a bat-
60. Jackson AC. Recovery from rabies [editorial]. N Engl J Med. 2005;352: borne rabies-related virus: clinical findings. Brain. 1988;111:15051516.
25492550. 77. Nathwani D, McIntyre PG, White K, et al. Fatal human rabies caused by
61. Jackson AC. Current and future approaches to the therapy of human European bat lyssavirus type 2a infection in Scotland. Clin Infect Dis. 2003;37:
rabies. Antiviral Res. 2013;99:6167. 598601.
62. Weli SC, Scott CA, Ward CA, et al. Rabies virus infection of primary neuro- 78. Samaratunga H, Searle JW, Hudson N. Non-rabies lyssavirus human en-
nal cultures and adult mice: failure to demonstrate evidence of excitotoxic- cephalitis from fruit bats: Australian bat lyssavirus (pteropid lyssavirus)
ity. J Virol. 2006;80:1027010273. infection. Neuropathol Appl Neurobiol. 1998;24:331335.
63. Ginsberg MD. Current status of neuroprotection for cerebral ischemia: 79. Hanna JN, Carney IK, Smith GA, et al. Australian bat lyssavirus infection:
synoptic overview. Stroke. 2009;40(suppl 3):S111S114. a second human case, with a long incubation period. Med J Aust. 2000;172:
64. Lafon M. Immunology. In: Jackson AC, Wunner WH, eds. Rabies. 2nd ed. 597599.
London: Elsevier Academic Press; 2007:489504. 80. Anonymous. Lyssavirus story breaks. Whitsunday Times. June 6, 2013.
65. Holzmann-Pazgal G, Wanger A, Degaffe G, et al. Presumptive abortive http://www.whitsundaytimes.com.au/news/lyssavirus-story-breaks/
human rabiesTexas, 2009. MMWR Morb Mortal Wkly Rep. 2010;59: 1896182/. Accessed December 17, 2013
185190. 81. Leonova GN, Belikov SI, Kondratov IG, et al. A fatal case of bat lyssa-
66. Wiedeman J, Plant J, Glaser C, et al. Recovery of a patient from clinical virus infection in Primorye Territory of the Russian Far East. Rabies Bull
rabiesCalifornia, 2011. MMWR Morb Mortal Wkly Rep. 2012;61:6165. Europe. 2009;33:58.

Scheld_Ch17.indd 260 2/21/14 5:39 PM


CHAPTER 18 HUMAN PRION DISEASES
SERGGIO C. LANATA, SVEN FORNER, AND MICHAEL D. GESCHWIND

The transmissible spongiform encephalopathies (TSEs) are a


group of closely related fatal neurodegenerative diseases that SCRAPIE, KURU, AND THE
affect humans and several other mammals (Tables 18.1 and DISCOVERY OF PRIONS
18.2). TSEs have a few characteristics in common: (1) they are
transmissible in nature and/or experimentally; (2) they invari- The history of the discovery of prions is an inspiring journey
ably lead to brain dysfunction or encephalopathy, which of multidisciplinary scientific collaboration that profoundly
manifests clinically as cognitive, behavioral, sensory, and/or changed our understanding of infectious and neurodegenerative
motor dysfunction; (3) they are uniformly fatal; and (4) they diseases. This journey began with the careful study of the prion
currently can only be diagnosed definitively by neuropathol- disease called scrapie, which primarily affects sheep and goats.
ogy (or genetically in certain cases). Scrapie is probably the earliest described prion disease,
TSEs are caused by prions (for proteinaceous infectious with the first reports published in 1750, but the disease was
particles), misshapen forms of the endogenous prion protein, probably recognized as early as 1732 in Great Britain. The
in the central nervous system (1); hence, TSEs are now referred clinical signs of scrapie are variable; the most frequently ob-
to as prion diseases, which is the term that will be used served clinical signs are emaciation, repetitive head rubbing,
for this chapter. Prion (pree-ahn) diseases are unique in medi- hyperesthesia, and pruritus. Pruritus is often severe enough
cine in that they occur in three forms: sporadic, infectious/ to make the affected animal rub and scrape (hence the term
acquired, and genetic. All three mechanisms occur in humans. scrapie) its body against rough surfaces, leading to wool
Prions are considered infectious because they rarely can be loss and skin abrasions (2). Affected animals often become hy-
transmitted from person to person (iatrogenically) or even less persensitive to outside stimuli, including noises, sudden move-
commonly by ingestion. When they are transmitted into, or ments, or handling by humans, to the extent some stimuli will
develop in, the nervous system, they cause the conversion of cause the animal to startle violently or convulse. Other com-
normal-shaped prion proteins into an abnormal, pathogenic mon signs include tremor and ataxia leading to impaired gait
conformation. This conversion process becomes exponen- and falls. Behavioral changes are prominent and common. A
tial and leads to the spread of prions throughout the brain, nibbling response of the mouth is often evoked by scratching
causing neuronal dysfunction and cell death and, ultimately, the animals back (positive scratch response). Scrapie has a
neurodegeneration. Prions are intrinsically different from all long incubation period, ranging between 2 and 5 years, and
other naturally occurring infectious agents, however, in that affected animals live up to 6 months after symptom onset.
they do not rely on nucleic acid to replicate within the host Scrapie is transmissible both naturally and experimentally.
organism. Furthermore, the presence of sporadic and heritable The natural means of transmission is a matter of debate, with
forms of prion disease also differentiates prions from all other both environmental and genetic susceptibility factors influenc-
infectious agents. This chapter will focus primarily on human ing transmission (3). Experimental transmissibility of scrapie,
prion diseases, with the caveat that most of our knowledge of however, was firmly demonstrated in 1936 when Cuill and
the pathophysiologic mechanisms of prion disease has been Chelle (4) showed that healthy sheep inoculated with scrapie-
obtained from animal and cell culture studies. infected spinal cord tissue developed the disease. At the time,
however, the infectious agent of scrapie was unknown.
Puzzled by scrapies long incubation period and devastat-
ing course, Bjorn Sigurdsson, an Icelandic veterinarian, first
TA B L E 1 8 . 1 proposed the notion of slow viral infections in the 1950s

PRION DISEASES IN HUMANS


TA B L E 1 8 . 2
Class Disease Abbreviation
PRION DISEASES IN NONHUMAN MAMMALS
Sporadic Sporadic Jakob-Creutzfeldt sJCD
disease Disease Abbreviation Animal Hosts
Fatal insomnia sFI
Scrapie Sc Sheep, goat
Variably protease-sensitive VPSPr
Transmissible mink TME Mink
prionopathy
encephalopathy
Genetic Genetic Jakob-Creutzfeldt gJCD
Bovine spongiform BSE Cattle
Disease
encephalopathy
Gerstmann-Strussler-Scheinker GSS
Chronic wasting CWD Mule deer, elk,
Fatal familial insomnia FFI disease moose
Acquired Iatrogenic Jakob-Creutzfeldt iJCD Feline spongiform FSE Cats
disease encephalopathy
Variant Jakob-Creutzfeldt disease vJCD Exotic ungulate EUE Nyala, greater kudu
Kuru Kuru encephalopathy

261

Scheld_Ch18.indd 261 2/21/14 5:39 PM


262 Part II: Viral Infections and Related Disorders

regarding a paralytic encephalitis of sheep called rida (5). In (1). This observation further motivated scientists to take
his view, these neurologic diseases were caused by unknown on the challenge of identifying the causative agent of these
viruses with prolonged incubation periods (years to decades) diseases.
that led to death after a short and progressive clinical course. Toward this goal, a series of careful experiments done on
Sigurdssons ideas shed light on the etiology of many diseases scrapie-infected brains demonstrated that the scrapie agent
that were poorly understood at the time (6). For example, was resistant to conditions that normally inactivated other
maedi, a deadly form of pneumonitis in sheep, and visna, a infectious organisms like viruses and bacteria, such as expo-
slowly progressive encephalitis, were found to be caused by sure to formalin (17), very high temperatures (18), and other
the same virus, now known as the maedi-visna lentivirus (7). solvents (19). Scrapie, and presumably kuru, was beginning
The suspected viral cause of scrapie, on the other hand, still to show properties that distinguished it from other infectious
remained elusive. diseases.
Concurrently, in the 1950s, D. Carleton Gajdusek, an Siggurdsons and Hadlows hypotheses were eventually
American physician and virologist, initiated the scientific supported experimentally in 1966, when the transmission
study of a human neurologic disease known as kuru in Papua of kuru was demonstrated by Gajdusek and colleagues (20)
New Guinea. Papua New Guinea is one of the most culturally through the intracerebral inoculation of diseased brain tissue
diverse countries in the world, with more than 800 languages in the chimpanzee. The proven transmissibility of kuru helped
spoken among roughly 6.3 million people segregated in differ- solidify the natural transmissibility theory that had already
ent tribes. The kuru epidemic emerged in the early twentieth been gaining momentum based on anthropologic and epide-
century among the Fore tribe that inhabits one of the highland miologic observations: kuru was transmitted through endo-
provinces. Kuru means to shiver from fever or cold in the lan- cannibalism (21). Furthermore, neuropathologic similarities
guage of the Fore people (8). The disease is characterized clini- between kuru and Jakob-Creutzfeldt disease (JCD) had been
cally by the insidious onset of progressive cerebellar ataxia, clearly recognized in the late 1950s (22). And the same group
eventually leading to inability to walk. A typical early feature of investigators that demonstrated transmission of kuru to
of the disease is the presence of unusual laughter, giving rise to chimpanzees also demonstrated transmission of JCD to the
the phrase the laughing death to describe kuru (9). In later chimpanzee in 1968 (23). This was an important experimental
stages, brainstem findings typically manifesting as eye move- discovery, as JCD had previously been regarded as a human
ment abnormalities and severe dysarthria are also common. neurodegenerative disease of unknown etiology. Taken to-
Like scrapie, the incubation period for kuru is long, as long as gether, these observations led to the introduction of the term
50 years (10), and disease duration is relatively short, usually transmissible spongiform encephalopathies to highlight the
less than 12 months. many neuropathologic and clinical characteristics shared by
Early anthropologic studies of the Fore people described these three diseases.
a ritualistic form of cannibalism that prevailed across all age Still, although experimental transmissibility had been
groups. Fore people consumed the bodies of their deceased proven for scrapie, kuru, and JCD, and natural transmissi-
relatives (endocannibalism), including those who died of bility had been shown for scrapie and kuru, the infectious
kuru (11). This was done with the hope of maintaining the substrate of these diseases remained unknown. As mentioned
spirit of the deceased within the village community. Perhaps earlier, the scrapie agent did not share basic biochemical and
partly for this reason, Gajdusek and his team initially sus- biophysical properties with other known infectious organ-
pected an infectious, probably viral, etiology to kuru, bor- isms. This notion was strengthened in 1966, when Alper
rowing from Sigurdssons concept of slow viral infections and colleagues (24) showed that extracts of scrapie-infected
(12). It soon became clear, however, that kuru was not a typ- brains retained their infective nature even after exposure to
ical infectious disease, in that victims did not exhibit fever large doses of ultraviolet light that were known to irrevers-
or other clinical signs of infection, including no signs of ibly damage the DNA and RNA molecules, thus implying
meningoencephalitis, no obvious cerebrospinal fluid (CSF) that the scrapie agent lacked nucleic acid. Later, similar stud-
abnormalities, and patients did not respond to antibiotic ies were conducted on tissue containing the agent responsible
medications. Furthermore, autopsy studies of brains affected for kuru and JCD (25).
by kuru did not demonstrate the inflammatory changes The idea of an infectious agent lacking nucleic acid was
typically seen in many forms of nervous system infections. considered outlandish at the time. It implied that the agent
Additionally, it was not transmissible to small laboratory that transmitted these diseases was, at its core, unlike any
animals and no infectious agent had been identified (8). This other known infectious agent. This idea captured the curi-
led Gajdusek and others to conduct an exhaustive search of osity of neurologist Stanley Prusiner and other researchers.
environmental factors that could explain the transmissible Through a series of careful experiments using animal models,
and seemingly infectious nature of kuru (13,14), but none Prusiner confirmed that the scrapie agent indeed resists inacti-
were found. The absence of an environmental cause for kuru vation by procedures that hydrolyze, modify, or shear nucleic
coupled with the well-described tribal and familial distribu- acids; furthermore, Prusiner and colleagues demonstrated that
tion of the disease led some to believe that kuru was a genet- the scrapie agent was inactivated by treatments that denatured
ically inherited condition (15). Large epidemiologic studies protein (26). Based largely on this line of evidence, Prusiner
showed that the incidence of kuru dramatically decreased postulated that a protein, which he named prion (pree-ahn)
among members of tribes who abolished endocannibalism, for proteinaceous infectious particle, was the principal car-
thus pointing toward a transmissible disease mechanism rier of scrapie (27).
rather than a genetic one (8). The discovery of prions (PrP, for prion protein) shattered
A major breakthrough came when William J. Hadlow, a the concept that nucleic acids are the sole carriers of trans-
veterinarian pathologist, made note of the neuropathologic missible infectious disease. Alternative theories to explain
similarities between kuru and scrapie and postulated that the nature of the scrapie agent were proposed for years after
a similar infectious agent was the cause of both diseases Prusiners postulate (28,29), but none of these theories has
(16). This was an important conceptual contribution to the been proven experimentally and the prion model has become
study of prion diseases in general, as prior to the 1950s, widely accepted over the last two decades, supported by a vast
the efforts to understand scrapie and kuru were disjoined body of evidence.

Scheld_Ch18.indd 262 2/21/14 5:39 PM


Chapter 18: Human Prion Diseases 263

NORMAL AND PATHOGENIC The Prion Protein Gene (PRNP)


PRION PROTEINS: PrPC AND PrPSc The prion gene, PRNP, is located in the short arm of chromo-
some 20 in humans. It is composed of two exons and one in-
After PrP was found to be the disease-producing substrate in
tron, and the protein-coding region is located in exon 2. PRNP
concentrates of scrapie-infected brain tissue (27), the question encodes the 253 amino acid PrP. A schematic of PRNP and
arose whether PrP was acquired exclusively from the environ- the prion gene in other species is shown in Figure 18.1. There
ment via an infectious mechanism or if it was also produced are more than 30 autosomal dominant mutations of PRNP
endogenously by the diseased animal. that lead to inherited forms of prion disease in humans. These
In 1986, for the first time, researchers were able to isolate occur primarily in the form of point mutations as well as some
PrP-related genomic clones in normal hamsters using comple- insertion, and possibly deletion mutations. Point mutations
mentary DNA (cDNA) obtained from PrP messenger RNA normally occur in the central and C-terminal portions of PrP.
(mRNA), thus demonstrating that mammalian cells carry the Insertion mutations occur in the N-terminal end of the pro-
gene responsible for encoding PrP (30). Similar experiments tein, which is normally composed of a nonapeptide followed
were performed in mice (31) and other mammals including by four octapeptide repeats. Different mutations lead to varied
humans (32). The gene that encodes PrP is known as PRNP. familial forms of the disease (33,34).
PRNP encodes a normal form of PrP with incompletely under- Historically, clinically-based studies of the heritable forms
stood cellular functions. The misshapen, pathogenic form of of prion disease in humans led to the identification of three
PrP, on the other hand, has unique biochemical properties that classic phenotypes: Gerstmann-Strussler-Scheinker disease
distinguish it from its normal counterpart (as will be discussed (GSS), familial Jakob-Creutzfeldt disease (fJCD), and fatal fa-
in the following sections). This distinction led to the current milial insomnia (FFI). These three forms of familial JCD were
designation of cellular PrP (PrPC) to describe PrP in its nor- described based on clinicopathologic features, prior to the
mal form and scrapie PrP (PrPSc) to describe the misfolded, identification of PRNP. The classification of familial (genetic)
pathogenic form of PrP. The terms PrPC and PrPSc will be used prion diseases, however, has changed with the division of fa-
accordingly in this chapter. milial forms based on specific PRNP mutations. Genetic prion

FIGURE 18.1 Mutations and polymorphisms of the prion protein gene; PrP mutations of the prion pro-
tein gene; PrP mutations causing genetic human prion disease; and PrP polymorphisms found in humans,
mice, sheep, elk, and cattle. Above the line of the human sequence are mutations that cause gPrD. Below
the lines are polymorphisms, some, but not all, of which are known to influence both the onset and the
phenotype of disease. Residue numbers in parentheses correspond to the human codons. (Much of the
data were compiled by J.-L. Laplanche.) (From Knipe DM, Howley PM, eds. Fields Virology. 5th ed.
Philadelphia: Lippincott Williams & Wilkins; 2007:3072.)

Scheld_Ch18.indd 263 2/21/14 5:39 PM


264 Part II: Viral Infections and Related Disorders

diseases (gPrDs) are discussed in detail in the section, Human cellular proteins are able to interact with each other and
Genetic Prion Diseases. thereby dictate an organisms physiology.
Polymorphisms in the PRNP gene have been shown to in- The primary structure of PrPC across different mamma-
fluence prion disease susceptibility and the clinical phenotype. lian species is highly conserved (50) and is nearly identical to
The most important polymorphism in humans is at amino acid that of PrPSc (51). PrPSc and PrPC also share important post-
129 of PRNP, which can be methionine (M) or valine (V). As translational modifications, including the presence of two
every person has two copies of each gene, humans can be ei- N-glycosylation sites, a single disulfide bond between cysteine
ther MM, MV, or VV. Homozygosity (either MM or VV) at residues, and a glycosylphosphatidylinositol membrane an-
codon 129 increases susceptibility to sJCD (35,36), for exam- chor at the C-terminus (this is the site of attachment of PrPC to
ple. Likewise, in patients with gPrD, the cis codon 129 poly- the cell membrane) (52).
morphism (i.e., the codon 129 allele on the same DNA strand The biochemical and biophysical differences that exist be-
as the mutation) can greatly influence how a disease presents tween PrPC and PrPSc are the result of specific posttranslational
(37). The phenotypic influence of these polymorphisms will be modifications that affect the secondary, tertiary, and quaternary
discussed further under the gPrDs section. structures of these proteins. In this respect, the most important
difference between PrPC and PrPSc relates to their response upon
exposure to detergents and proteases. PrPC is soluble in nonde-
naturing detergents and degrades when exposed to proteases.
The Function of Cellular PrP PrPSc, on the other hand, forms amyloid structures (prion rods
that show the typical fluorescence birefringence of amyloids)
PrPC is expressed mostly in the brain, but it has also been de- when exposed to detergents and is highly resistant to cellular
tected in a wide variety of mammalian tissues, including leu- proteases, particularly proteinase K (PK) (52). It is important
kocytes, heart, skeletal muscle, lung, intestinal tract, spleen, to note that PrPSc is not fully resistant to PK, however, and this
and reproductive organs (38). The role of PrPC in the nervous will be discussed further in the following texts. The insolubility
system remains elusive and controversial. PrPC is a cell-sur- of PrPSc to detergents, on the other hand, is universal, and this
face glycoprotein that forms part of cell membrane structures property is used in the laboratory to accurately diagnose the
known as lipid rafts (39). In the nervous system, lipid rafts presence of prion disease.
are heavily involved in cellular signal transduction pathways, Our knowledge of the structural differences that exist be-
including neurotrophic factor signaling, cell adhesion and mi- tween PrPSc and PrPC at an atomic level has been obtained via
gration, axon guidance, myelin genesis, and synaptic trans- careful studies on different transgenic animal models using
mission (40). It is therefore not surprising that most of the nuclear magnetic resonance (NMR) and infrared (IR) spec-
cellular functions of PrPC that have been described over the troscopy and x-ray crystal diffraction. IR spectroscopy stud-
past two decades relate to a wide range of signal transduction ies have shown that PrPC has a high alpha-helix content and
pathways (41) involving molecular interactions with multiple virtually no beta-sheet content, whereas PrPSc is characterized
cellular proteins (42), some of which are vital for normal neu- by roughly equal amounts of alpha-helices and beta-sheets
ronal function. For example, in mouse neuroblast brain cells, (53). Alpha-helices and beta-sheets are secondary protein
PrPC overexpression enhances cell proliferation and cell cycle structures. NMR spectroscopy studies also reveal that PrPC
reentrance, whereas PrPC silencing slows down the cell cycle; forms a unique three-dimensional structure in its C-terminus,
these effects are mediated via PrPC interactions with epidermal consisting of three alpha-helices bundled with two short
growth factor receptor (EGFR) in the plasma membrane (43). antiparallel beta-sheets (52), whereas the N-terminus does
Similarly, PrPC expressed in pluripotent human embryonic not arrange into a specific conformation. These findings
stem cells have been shown to induce and modulate cell cycle have been largely replicated using x-ray crystal diffraction
dynamics and partly determine cell differentiation (44). PrPC methods (54).
has also been shown to be essential for myelin maintenance,
as depletion of PrPC in neurons triggers a form of chronic
demyelinating neuropathy in mice (45). PrPC has been impli-
cated in neuroprotective and neurotoxic signaling cascades Possible Mechanisms of PrPC Conversion and
(46). Furthermore, there is an expanding body of evidence
that supports clear interactions between PrPC and metal ions PrPSc Propagation
in different signaling pathways, particularly copper, zinc, and
iron (47). The infectious nature of prions in animals was perhaps
Despite the role of PrPC in a variety of important cellular most influentially demonstrated by Legname and colleagues
functions (48), however, some PrPC knockout mice have been (55) in 2004, when they inoculated fibrils of N-terminally
shown to develop normally and do not have signs of neuro- truncated recombinant mouse PrP made in Escherichia coli
logic disease (49), thereby suggesting that the absence of PrPC into the brains of transgenic PrP knockout mice expressing
is not a sufficient cause of neurodegeneration or neurodevel- N-terminally truncated PrP and showed that these mice devel-
opmental abnormalities. oped neurologic dysfunction between 380 and 660 days after
inoculation, with neuropathologic findings consistent with
prion disease. When brain extracts from these mice were then
inoculated into the brains of wild-type mice, these mice devel-
Structural Differences Between PrPC and PrPSc oped a prion disease. Many other similar experiments have
confirmed that prions have the ability to replicate and propa-
All mammalian proteins are composed of a specific structure of gate within the nervous system.
amino acids (primary structure) that are arranged into identifi- As stated previously, the mode of propagation of prions
able repeating structures held together by hydrogen bonding is unique among infectious agents, in that prions do not ap-
(secondary structure), which in turn cause protein molecules to pear to use nucleic acid to replicate within the host species.
adopt specific three-dimensional conformations (tertiary and Instead, according to the protein-only hypothesis of prion
quaternary structures). Via their three-dimensional structure, spread, the presence of PrPSc by itself induces endogenous

Scheld_Ch18.indd 264 2/21/14 5:39 PM


Chapter 18: Human Prion Diseases 265

FIGURE 18.2 Molecular pathogen-


esis of prion disorders. In sJCD, PrPC
spontaneously misfolds into PrPSc.
In gPrD, mutations in the prion gene
make it easier for PrPC to misfold into
PrPSc. In iatrogenic forms of prion
disease, PrPSc is accidentally transmit-
ted into the brain of a person, causing
their PrPC to misfold into PrPSc by a
templating mechanism. Conversion of
PrPC to PrPSc becomes exponential and
results in neurodegeneration or spongi-
form encephalopathy. (From Rubin R,
Strayer DS, Rubin E. Rubins Pathology.
6th ed. Baltimore: Lippincott Williams
& Wilkins; 2011:1334.)

PrPC to misfold into PrPSc via the modification of the second- receptors have been implicated in the initial steps of trans-
ary and tertiary structures of PrPC in a process referred to as formation of PrPC into PrPSc. Among these, the low-density
template-directed misfolding. In other words, PrPSc acts as a lipoprotein receptorrelated protein 1 (LRP1) might be an
template that guides PrPC misfolding: one molecule of PrPSc important player (61) as well as glycosaminoglycan (62) and
induces the conversion of one molecule of PrPC into PrPSc, laminin receptors (63). There are likely other cellular recep-
two molecules of PrPSc induce the transformation of two new tors involved in this process.
molecules of PrPC, and so PrPSc accumulates in an exponential Once incorporated to the host cell, PrPSc leads to PrPC mis-
fashion (see Figs. 18.2 and 18.3. The progressive accumula- folding within minutes. This was elegantly demonstrated in in
tion of pathogenic PrPSc in the nervous system leads to the vitro experiments using epitope-tagged PrPC in neuroblastoma
clinical manifestations and neurodegeneration seen in prion cell lines (64). Moreover, it appears that PrPC misfolding oc-
disease (1). curs largely within the cell surface, from where PrPSc is then
The exact mechanisms by which PrPSc leads to the mis- endocytosed and either recycled back to the plasma membrane
folding of PrPC are yet to be determined. It has been demon- or deposited within the Golgi system (65).
strated experimentally that the first step in prion infectivity Several biochemical and biophysical factors have been
involves the successful cellular uptake of PrPSc in susceptible shown to facilitate the misfolding of PrPC into PrPSc in vitro,
host cells (56,57), thereby allowing the physical interac- including the presence of acidic and/or salt solutions and inter-
tion between PrPC and PrPSc that is required for misfolding. actions between PrPC and metal ions (66). The cellular mecha-
Although it is widely accepted that the presence of cel- nisms of PrPC misfolding remain elusive, however, and there is
lular PrPC is required for the propagation of prion disease mounting evidence suggesting that there are several different
(58,59), the initial cellular uptake of PrPSc occurs without cellular structures and cofactors mediating the transformation
participation of PrPC (60). Instead, several cell membrane of PrPC to PrPSc (67,68).

Scheld_Ch18.indd 265 2/21/14 5:39 PM


266 Part II: Viral Infections and Related Disorders

SPORADIC HUMAN PRION


DISEASE: JAKOB-CREUTZFELDT
DISEASE, FATAL INSOMNIA, AND
VARIABLY PROTEASE-SENSITIVE
PRIONOPATHY
Sporadic Jakob-Creutzfeldt Disease
The disease that we now recognize as sporadic Jakob-
Creutzfeldt disease (sJCD) clinically and neuropathologically
was first described by Alfons Maria Jakob (69,70) in the
early 1920s. The single case that his mentor Hans Gerhard
Creutzfeldt (71) described in 1920, on the other hand, was nei-
ther clinically nor neuropathologically representative of sJCD
or what we now know as prion disease (72,73). Therefore,
some authors prefer the more correct eponym Jakobs or
Jakob-Creutzfeldt disease rather than Creutzfeldt-Jakob dis-
ease (CJD) when referring to prion disease. The authors of this
chapter prefer the name Jakob-Creutzfeldt over Creutzfeldt-
Jakob. Thus, the term should probably be Jakob-Creutzfeldt
disease (or even Jakobs disease [JD]). Of note, some physicians
have mistakenly thought prion disease was due to the JC virus
and incorrectly sent the JC virus PCR as a test for prion dis-
ease. In this chapter, we use the term Jakob-Creutzfeldt disease.
Most studies have shown that about 85% of human prion
diseases are sporadic, 10% to 15% are genetic, and less than 1%
is acquired. In a large prospective systematic study of human
prion disease in Europe, Australia, and Canada from 1993
to 2002, which included more than 4,400 pathology-proven
cases, the incidence of sporadic prion disease was approxi-
mately 85%, genetic was about 10%, and acquired were about
6% (74). The larger percentage of acquired cases than many
other studies is likely due to the emergence of vJCD primar-
ily in the United Kingdom and France during this time period
(75) as well as the emergence of human pituitary hormone
cases from prior exposure in the United Kingdom, France, and
Australia (76). The incidence of sporadic human prion diseases
in most populations is about 1 per million, of gPrD about 1 per
10 million, and even less for acquired prion disease (77).
Before recent advances in molecular biology were applied to
study human prion disease, sJCD was subdivided into separate
phenotypes based on clinical and histopathologic character-
istics. Classic subtypes of sJCD described in the early litera-
ture include the Heidenhain variant of sJCD, characterized
by prominent early visuospatial symptomatology, sometimes
leading to cortical blindness (78); the Jakob variant or spas-
tic pseudosclerosis, which presents with prominent frontal,
pyramidal, and extrapyramidal symptoms; and the Brownell-
Oppenheimer variant, characterized by early cerebellar dys-
function and late dementia. Other clinical variants exist in the
literature.
It is now known that the clinicopathologic variability of
sJCD is in part due to two factors: the polymorphisms at
codon 129 of PRNP (36) and at least two distinct species
of PrPSc (PrPSc type 1 and type 2). In the laboratory, the two
types of PrPSc are distinguished by their mobility on Western
blot transfers after exposure to PK (Fig. 18.4) (79). Taken
FIGURE 18.3 One proposed mechanism for reproduction of the
infectious prion protein (PrP). PrPC, noninfectious PrP; PrPSc, infec-
together, this line of evidence suggests that the different sJCD
tious PrP. (From Harvey RA, Cornelissen CN. Microbiology. 3rd ed. phenotypes are an expression of interactions of the host PRNP
Baltimore: Lippincott Williams & Wilkins; 2012:328.) genotype and PrPSc type, and this notion forms the basis of our
current classification of sJCD.
Large studies have been carried out comparing molecular,
neuropathologic, and clinical characteristics of sJCD subjects
(80). Results of these studies prompted a classification scheme

Scheld_Ch18.indd 266 2/21/14 5:39 PM


Chapter 18: Human Prion Diseases 267

sJCD include seizures (including status epilepticus) (88); these


have been reported in some studies in up to 10% of cases, but
usually later in the disease course. In our experience, seizures
are far less common than this (84,86,88). Clinically evident
peripheral neuropathy (including cranial neuropathy) is very
uncommon in sJCD (89,90) but is seen more commonly in
some gPrDs (9195).
The average age of onset of sJCD is 65 years, with a peak
incidence in the 7th decade. The average disease duration is 4
to 7 months. The great majority of patients (90%) die within
1 year of diagnosis, but up to 5% of patients may survive for
2 or more years (36,84,96). Certain factors such as younger
age at onset, female gender, and heterozygosity at codon 129
have been associated with longer survival in sJCD (36,77,79).
Invariably, however, sJCD leads to a relatively rapidly progres-
sive dementia (RPD) (compared to most other neurodegenera-
tive diseases), with varied clinical manifestations, depending
on which areas of the brain are affected by prion neurode-
generation. Regardless of pattern of prion spread, most cases
develop akinetic mutism prior to death (87). Death is usually
FIGURE 18.4 Western blot. Western blot analysis of brain homog- due to aspiration pneumonia.
enates from cerebral cortex and probed with antibody 3F4 (against Brain magnetic resonance imaging (MRI) is the most sen-
PrP) after digestion with PK. In each column there are three bands, sitive and specific diagnostic test for sJCD, with published
representing from top to bottom: diglycosylated, monoglycosylated, sensitivity of approximately 91% to 96% and specificity of
and unglycosylated PrPSc. Type 1 PrPSc has an unglycosylated band
approximately 92% to 94% (79,97,98). The high diagnos-
running at a molecular weight of 21 kDa. Type 2 PrPSc has an ungly-
cosylated band running at a molecular weight of 19 kDa. The electro- tic sensitivity of brain MRI is largely a reflection of its ability
phoretic migration of the unglycosylated PK-resistant PrPSc in lane 1 is to detect changes very early in the disease course (98). The
from an sJCD MM1 case and shows type 1 PrPSc, whereas lane 2 from most sensitive MRI sequences for the diagnosis of sJCD are
an MM2 sJCD case shows type 2 PrPSc, with a 19 kDa unglycosylated diffusion-weighted imaging (DWI) and apparent diffusion
PK-resistant PrPSc. (From Tartaglia M, Thai J, See T, et al. Pathologic coefficient (ADC) sequences; indeed, DWI has been shown
evidence that the T188R mutation in PRNP is associated with prion to be more sensitive than fluid-attenuated inversion recovery
disease. J Neuropathol Exp Neurol. 2010;69[12]:12201227.) (FLAIR)/T2 sequences abnormalities (98,99).
The radiographic hallmark of sJCD is the presence of DWI
MRI hyperintensities in the neocortex, basal ganglia, and/or
that included six molecular subtypes based on PRNP codon thalamus, often associated with corresponding ADC map hy-
129 polymorphisms (M/M, M/V, or V/V) and PrPSc type (type pointensities, suggesting restricted flow of water molecules in
1 or 2). There is considerable overlap between classic sJCD these regions (97), probably due to pathological changes oc-
clinical phenotypes and the clinical phenotypes associated curing in prion disease, particularly vacuolation (spongiform
with each molecular subtype to the extent that five combined change), prion deposition, and/or gliosis (astrocyte prolifera-
sJCD subtypes were initially recognized: MM1/MV1, VV1, tion) (100). These MRI findings have been included in various
VV2, MV2, and MM2 cortical and MM2 thalamic (the lat- criteria for sJCD (101), although some criteria allow T2 or
ter of which corresponds to sporadic fatal insomnia) (79,81). FLAIR and do not require DWI abnormalities (102), which
The discovery that many patients have both type 1 and type 2 might lead to false-positive diagnoses (97). An example of the
prions has complicated this earlier molecular classification and typical brain MRI findings seen in sJCD is shown in Figure
expanded the subtypes of sJCD (79,82). It is beyond the scope 18.5. Three major patterns of DWI MRI hyperintensities have
of this chapter to describe the clinicopathologic characteristics been identified in sJCD: neocortical and subcortical (approxi-
of each subtype in great detail. The main characteristics of mately 68% of cases), predominantly neocortical (24% to
each subtype are shown in Table 18.3 (79). 30%), and predominantly subcortical (mostly striatal hyperin-
The clinical presentation of sJCD in general is protean, how- tensities, with or without thalamic involvement) (2% to 5%)
ever. Studies show that 40% to 60% of affected individuals (97,103) (Fig. 18.6). Cortical hyperintensities (or cortical rib-
initially develop cognitive symptoms (8385), generally mani- boning) can be unilateral or bilateral but typically spares the
festing as memory, executive, and language dysfunction and/or precentral cortex (97,104). Striatal involvement often begins
confusion. Other early clinical manifestations include cerebel- unilaterally, but eventually becomes bilateral, and typically
lar dysfunction, constitutional symptoms, and behavioral/per- has a decreasing anterior-posterior gradient (i.e., the anterior
sonality changes (e.g., depression, irritability) in around 20% caudate appears more hyperintense than the posterior puta-
of pathology-proven cases of sJCD. Furthermore, impaired vi- men) (97). Moreover, up to 90% of sJCD cases demonstrate
sion (blurred vision, diplopia, and visual hallucinations) has involvement of limbic and paralimbic cortical structures (i.e.,
been reported as an initial complaint in 9% to 15% of cases insula, anterior cingulate, hippocampus), visualized as hyper-
(8486). Myoclonus, which is anecdotally considered to be a intensities and hypointensities of these regions on DWI and
sensitive sign of sJCD by many clinicians, is a late feature of ADC sequences, respectively. In our experience, isolated lim-
the disease in up to 90% of cases (84). Other movement dis- bic involvement is atypical for sJCD, however, and should
orders are often seen in patients with sJCD, either early or late alert the clinician toward alternative diagnoses (97).
in their disease course, including dystonia (fixed posturing of Cortical ribboning is not 100% sensitive and specific for
a body part), choreoathetosis (slow, writhing or quick dance- sJCD, however, as it is also seen in cases of viral and/or autoim-
like movements), parkinsonism (slowed movements, tremor mune encephalitis, hypoglycemia, seizures and status epilepticus,
at rest, etc.), and supranuclear gaze palsy (inability to move hyperammonemic encephalopathy, hypoxic injury, mitochondrial
eyes volitionally) (87). Other rare initial manifestations of encephalopathy, vasculitis, and other conditions (97,98,105).

Scheld_Ch18.indd 267 2/21/14 5:39 PM


268 Part II: Viral Infections and Related Disorders

TA B L E 1 8 . 3
MAIN CLINICOPATHOLOGIC CHARACTERISTICS OF sJCD SUBTYPES

Average Age of Average Disease Characteristic Clinical DWI MRI


sJCD Subtype Onset (years) Duration (months) Features at Onset Findings234,235 PrPSc Neuropathology

MM1/MV1 66 (4291) 4 (124) Cognitive decline is by Primarily cortical Predominantly non-


far the most common hyperintensities in amyloid synaptic
initial manifestation; frontal, parietal, PrPSc deposition
ataxia, visual temporal, or
impairment, aphasia, insular regions
and psychiatric
features can be initial
manifestations in
around 1/3 of patients.
MM2-cortical 66 (4982) 14 (324) Nearly all patients Widespread cortical Perivacuolar PrPSc
present with cognitive hyperintensities deposits
decline; aphasia
may be initial
manifestation in up
to 1/3 of patients.
Ataxia and psychiatric
manifestations are far
less frequent at onset.
MM2-thalamic 46 (2474) 16 (1073) Cognitive decline and Often normal Mild PrPSc deposition
(sporadic fatal ataxia occur in nearly in the thalamus
insomnia) half of patients at
onset; insomnia,
psychiatric symptoms,
and visual signs
occur in around
1/3 of patients at
onset. Dysautonomia
is a rare initial
manifestation.
MV2 62 (4081) 17 (443) Ataxia is most common Predominant signal Kuru-like plaques
initial manifestation, increase in the
followed closely basal ganglia and
by cognitive thalamus
decline. Psychiatric
manifestations
occur in up to 1/3
of patients at onset.
Aphasia is a rare
initial manifestation.
VV2 64 (4183) 6 (318) Nearly all patients Predominant signal Focal plaque-like
present with ataxia; increase in the PrPSc deposits
cognitive decline basal ganglia and
occurs in up to cingulate gyrus
1/3 of patients.
Psychiatric symptoms,
visual impairment,
and aphasia are
uncommon at onset.
VV1 43 (1971) 19 (472) Cognitive decline Cortical Mild synaptic
is most common hyperintensities PrPSc deposition
initial manifestation; in the cingulate,
psychiatric signs are insular cortex, and
initial manifestation in hippocampus
up to 1/3 of patients.

From refs 36, 81, and 233.

Scheld_Ch18.indd 268 2/21/14 5:39 PM


Chapter 18: Human Prion Diseases 269

FIGURE 18.5 Three common variations of sporadic Jakob-Creutzfeldt disease presentation on MRI.
A: Neocortical (solid arrow), limbic (dashed arrow), and subcortical gray matter (dotted arrow).
B: Neocortical and limbic cortex. C: Limbic and subcortical. Note that the DWI shows the hyperintensities
much more than the corresponding FLAIR sequences, and that DWI hyperintensities often have correspond-
ing ADC hypointensity. Pattern A was found in 54% of cases, pattern B in 27% of cases, and pattern C in
9% of cases. Note that the abnormalities are more readily seen on DWI than on FLAIR. ADC hypointensity,
indicating restricted diffusion, corresponds to most DWI hyperintensities. ADC abnormalities are most eas-
ily identified in the basal ganglia. (From Vitali P, Maccagnano E, Caverzasi E, et al. Diffusion-weighted MRI
hyperintensity patterns differentiate CJD from other rapid dementias. Neurology. 2011;76[20]:17111719.)

Hence, clinicians should consider these and other conditions in setting (77,108). This EEG finding is found only in about two
the presence of cortical ribboning on brain MRI. Likewise, stria- thirds of sJCD patients, however, and is most often present
tal and diencephalic DWI hyperintensities with ADC hypointen- late in the disease course and may require repeated EEG test-
sities should not be considered pathognomonic of sJCD, as these ing (108). In general, compared to brain MRI, EEG probably
changes have been observed in extrapontine myelinolysis, Wilson provides little value for diagnosis early in the disease course.
disease, Wernicke encephalopathy, and hyperglycemia with Furthermore, the presence of PSWCs rarely is seen in other
seizures (97,105). These DWI hyperintensities might decrease in neurologic conditions such as Alzheimer disease, dementia
late stages of the disease due to brain atrophy, particularly in pa- with Lewy bodies, toxic-metabolic and anoxic encephalopa-
tients with disease duration of over 1 year (97). Despite the high thies (e.g., hepatic), progressive multifocal leukoencephalopa-
sensitivity and specificity of MRI DWI and ADC sequences for thy, and Hashimoto encephalopathy (109,110).
the diagnosis of sJCD in the right clinical setting, there is evidence
to suggest that radiologists often miss the diagnosis of sJCD Cerebrospinal fluid
because they fail to recognize these changes (106,107).
Routine CSF analysis is typically normal in sJCD, although
sometimes a mild elevation in the total protein level can be seen
Electroencephalogram (typically less than 75 mg/dL). CSF pleocytosis (10 cells/L
The diagnostic value of electroencephalography (EEG) has less- white blood cells [WBCs]), an elevated immunoglobulin G (IgG)
ened to some extent due to the high sensitivity and specificity index, and/or the presence of oligoclonal bands are unusual in
of brain MRI for the diagnosis of sJCD (98). Still, the presence sJCD and should lead the clinician to consider other conditions,
of periodic sharp wave complexes (PSWCs), often biphasic or particularly infectious or autoimmune disorders (111,112).
triphasic, occurring every 0.5 to 2 seconds (Fig. 18.7) remains The clinical use of several CSF biomarkers is quite contro-
a useful finding that helps rule in sJCD in the right clinical versial, with evidence showing varying degrees of sensitivity and

Scheld_Ch18.indd 269 2/21/14 5:39 PM


270 Part II: Viral Infections and Related Disorders

FIGURE 18.6 Brain MRI of a patient with pathology-proven sJCD showing three key sequences for
prion disease diagnosis: FLAIR, DWI, and ADC. Note that the abnormal hyperintensities (bright areas)
in the deep nuclei (solid arrows) and cortex (dotted arrows; cortical ribboning) are more evident on the
DWI than the FLAIR MRI, which is typical of JCD. The hyperintense regions on DWI have correspond-
ing hypointense (dark) regions on the ADC sequence showing that the DWI and FLAIR hyperintensities
are due to restricted diffusion of water molecules.

specificity for the diagnosis of sJCD. The 14-3-3 protein, for neurodegeneration, seizures, and many others (118122). In this
example, which is one of the first CSF proteins reported to be respect, a large multicenter European study found that the speci-
elevated in sJCD, and initially reported to have 100% sensitivity ficity of 14-3-3 protein is lowered when used to differentiate sJCD
and 96% specificity for the diagnosis of sJCD (113), has been from acute neurologic disorders (such as vascular, inflammatory,
subsequently found to have more limited sensitivity and specific- or seizures) versus neurodegenerative dementias (82% to 87%
ity when studied in larger patient cohorts (114,115). A more re- vs. 95% to 97% respectively). This suggests taking the differen-
cent analysis in the United Kingdom showed CSF 14-3-3 protein tial diagnoses into consideration is important when interpreting
sensitivity of 86% and specificity of 74% in a pathologically con- the test result (123). In summary, a positive 14-3-3 protein does
firmed cohort (116). Similarly, an analysis of data obtained from not necessarily equate with prion disease. Regardless, recent rec-
the U.S. National Prion Disease Pathology Surveillance Center ommendations from the American Academy of Neurology sug-
(NPDPSC) showed the 14-3-3 protein Western blot only had gest ordering CSF 14-3-3 protein when there is a strong clinical
a receiver operating characteristic area under the curve (ROC suspicion of JCD (pretest probability of 20% to 90%) in order
AUC) value of 0.68 (117), which is poor considering that a test to reduce diagnostic uncertainty (124).
with perfect sensitivity and specificity would be 1.0. Some stud- Total-tau (t-tau), neuron-specific enolase (NSE), and the as-
ies on the clinical use of CSF 14-3-3 protein suggests it should trocytic protein S100 are also used as CSF biomarkers for sJCD
be considered a general marker of neuronal injury and death. diagnosis. The sensitivity and specificity of these biomarkers var-
Indeed, 14-3-3 protein is ubiquitously present in the intracellu- ies greatly among studies. In a large multicenter, retrospective
lar compartment of brain neurons and is released into the CSF European study (115), the combined sensitivity and specificity
compartment due to neuronal injury from varied causes. Thus, of the four main sJCD biomarkers (14-3-3 protein, t-tau, NSE,
14-3-3 protein can be elevated in many non-prion neurologic and S100) was found to be higher than the individual sensitiv-
conditions, such as multiple sclerosis, acute stroke, Alzheimer ity and specificity. Not all patients in this study underwent all
disease and other neurodegenerative dementias, HIV-related four tests nor were the tests performed in the same CSF samples,

Scheld_Ch18.indd 270 2/21/14 5:39 PM


Chapter 18: Human Prion Diseases 271

FIGURE 18.7 Periodic complexes associated with Jakob-Creutzfeld disease. A 65-year-old woman with
history of rapid cognitive decline, myoclonus, and gait ataxia. Her EEG showed bilaterally synchronous
and independently sharp wave discharges. Her CSF 14-3-3 prion protein was positive, further supportive
of Jakob-Creutzfeld disease. (From Greenfield LJ, Geyer JD, Carney PR, eds. Reading EEGs: A Practical
Approach. Philadelphia: Lippincott Williams & Wilkins; 2010:253.)

however; hence, this study did not permit a fair comparison be-
tween biomarkers. Nevertheless, the study found the sensitivity
Two Uncommon Forms of Sporadic Human
and specificity of the 14-3-3 protein to be 85% and 84%, t-tau Prion Disease: Fatal Insomnia and Variably
(cutoff 1,300 pg/mL) 86% and 88%, NSE 73% and 95%, and Protease-Sensitive Prionopathy
S100 82% and 76%, respectively (115). Other studies, how-
ever, have shown sensitivity and specificity values higher than The MM2 thalamic form of sJCD is sometimes referred to as
90% for t-tau in sJCD (125,126), although there is still no agree- sporadic fatal insomnia (sFI), the least common of the six forms
ment over its cutoff value (usually higher than 1,150 pg/mL). of sJCD based on molecular classification (36,81). It is similar
In reviewing all of the data, the opinion of the authors of clinically and pathologically to the rare gPrD FFI, as described
this chapter is that among 14-3-3, t-tau and NSE, the single under gPrDs section. It is the rarest form of sporadic human
best CSF biomarker is the t-tau, although NSE has somewhat prion disease, with approximately 31 cases reported in the lit-
higher specificity, whereas the 14-3-3 protein is overall the erature. On average, affected patients are 46 years old at dis-
least clinically useful. There probably is not sufficient data to ease onset, mean survival is 24 months (79). Clinicians should
make a determination of the clinical utility of S100B (beta). consider this form of sporadic prion disease in patients with
At our center, we send all three biomarkers when looking for rapidly progressing dementia associated with sleep abnormali-
evidence of rapid neuronal injury. In summary, these CSF bio- ties and dysautonomia. Unlike sJCD, EEG, CSF, and brain MRI
markers are likely markers of rapid neuronal injury and do not investigations have not been shown to be helpful for the diag-
equate with prion disease, but might be helpful in some cases, nosis of this disorder (79). Instead, polysomnography, which
yet have overall less diagnostic use than brain MRI (97,98). shows impaired sleep-related wave forms (such as K-complexes
New CSF diagnostic tests, such as real-time quaking-induced and spindles), and brain positron emission tomography (PET)
conversion reaction (RT-QuIC), that are relatively specific for examinations, which consistently show thalamic hypometabo-
prions (and not just biomarkers) are under development (127). lism, have proven to be more useful diagnostic tools (79).

Scheld_Ch18.indd 271 2/21/14 5:39 PM


272 Part II: Viral Infections and Related Disorders

The variably protease-sensitive prionopathy (VPSPr) is the lack parkinsonism and ataxia. As is the case in sJCD MM2-
latest addition to the list of sporadic human prion diseases thalamic form, MRI, CSF, and EEG investigations often do not
introduced into the literature in 2008 (128). The term vari- show the typical features diagnostic for sJCD. It is not clear if
ably protease-sensitive prionopathy is derived from the fact VPSPr is as transmissible as other prion diseases, as the prions
that prions extracted from brain tissue from such cases pro- in VPSPr are more sensitive to proteases and thus might be
duces a distinctive electrophoretic profile on Western blot due more easily degraded.
to sensitivity to protease digestion, which distinguishes it from
other forms of prion diseases. As most laboratory techniques
for diagnosis of prion disease (e.g., Western blot and immu-
Basic Neuropathology of Human Prion Diseases
nohistochemistry) rely on identification of protease-resistant At a macroscopic level, atrophy is the only gross neuroana-
PrPSc, as described previously, detecting this form of prion dis- tomic finding in sJCD brains. Atrophy can be mild to moderate,
ease can be quite difficult (129). VPSPr has certain distinctive localized to the cerebral cortex, basal ganglia, and cerebel-
clinical, molecular, and pathologic characteristics (79,129). lar regions. As in other neurodegenerative diseases, atrophy
Clinically, patients generally present with prominent psychi- is most pronounced in patients with a long clinical course.
atric manifestations and rapid cognitive decline (primarily Unlike what is seen in other neurodegenerative diseases, how-
aphasia), followed by motor manifestations (ataxia and par- ever, prominent hippocampal atrophy is usually not seen in
kinsonism). Clinical presentations, however, vary according human prion disease (130).
to codon 129 genotype (79). Based on the cases identified Microscopically, the histopathologic hallmarks of human
through 2010, cases associated with methionine homozygosity prion disease are the presence of neuronal loss, astrocytic glio-
(VPSPr-129MM) seem to not have prominent psychiatric fea- sis, vacuolation (or spongiform changes), and deposition of
tures, whereas VPSPr-129MV and VSPr-129VV cases usually prions (PrPSc) in the brain (Fig. 18.8) (131). These changes

FIGURE 18.8 Photomicrographs from right (side relatively spared radiologically and pathologically)
and left (side affected radiologically and pathologically) cortex of a patient with sJCD with very asym-
metric brain involvement. A and D show hematoxylin and eosin (H&E) stains, B and E show CD68
immunostaining for glia (astrocytic gliosis), and C and F show a synaptic pattern of staining for PrPSc
using the 3F4 antibody (against PrP) after hydrolytic autoclaving (which degrades PrP, but not PrPSc).
(From Geschwind M, Potter CA, Sattavat M, et al. Correlating DWI MRI with pathologic and other
features of Jakob-Creutzfeldt disease. Alzheimer Dis Assoc Disord. 2009;23[1]:8287.)

Scheld_Ch18.indd 272 2/21/14 5:39 PM


Chapter 18: Human Prion Diseases 273

occur without signs of inflammation in the brain, other than amyloid. A different type of plaque known as florid plaque
those seen typically with neuronal loss in neurodegenerative is commonly seen in vJCD (see the following texts); it con-
disease, such as microglia activation, astrocytosis, and re- sists of an amyloid core surrounded by a halo of spongiform
lease of cytokines (132134). The distribution of vacuola- change, giving it a flower (florid) appearance. This is a find-
tion changes seen in human prion disease vary between cases ing strongly suggestive for vJCD, as it is rarely seen in other
and disease subtypes (77), although the head of the caudate prion diseases. FFI (see the following texts), a gPrD, is charac-
nucleus is most often involved, whereas the brainstem is terized by severe neuronal loss and gliosis in thalamic nuclei,
relatively spared (130). More than 95% of sJCD cases show whereas vacuolation and amyloid deposition are minimal or
vacuolation and the remaining cases are diagnosed by immu- even absent (130). Small amounts of PrPSc are usually found
nohistochemistry to PrPSc. PrPSc deposition in sJCD is usually but often restricted to the thalamus, temporal lobe, or other
in a nonamyloid form, with frequent synaptic or punctate regions (77).
deposition in the cortex as well as perivacuolar PrPSc deposi-
tion in brain regions containing vacuolation. The Differential Diagnosis of
Amyloid plaques of PrPSc are very common in kuru and
the gPrD GSS (see the following texts) but are only found in
Sporadic Jakob-Creutzfeldt Disease
about 10% or less of sJCD cases. More specifically, GSS is Because of the protean manifestations of sJCD, it is often
usually characterized by multicentric plaques, composed of confused with many other conditions. Table 18.4 shows the
a dense core of amyloid surrounded by smaller globules of breakdown of non-prion diagnoses of patients referred to

TA B L E 1 8 . 4
DIAGNOSTIC BREAKDOWN OF NONJAKOB-CREUTZFELDT DISEASE RAPIDLY PROGRESSIVE DEMENTIA
REFERRALS TO TWO JAKOB-CREUTZFELDT DISEASE CENTERS

University of California, San Francisco German Cohort n 124


Cohort n 104 (21 Pathology-Proven)136 Total (37 Pathology-Proven)135 Total

Unclassified dementia 14 Alzheimer disease 34


Psychiatric 12 Unclassified dementia 20
Dementia with Lewy bodies 8 Cerebrovascular disease (vascular dementia, 11
cerebrovascular accident)
Encephalitis 8 Encephalitis (chronic), unknown 10
Frontotemporal degeneration (FTD) or FTD motor 7 Parkinson disease 6
neuron disease
Corticobasal degeneration 6 Psychiatric 6
Autoantibodya 4 Motor neuron disease 3
Metastatic encephalopathy 4 Multiple sclerosis 3
Primary CNS lymphomab 4 Paraneoplastic 3
Alzheimer disease 3 Intoxication 3
Encephalopathy 3 Alcohol-induced atrophy 2
Leukoencephalopathy 3 Brain tumord 2
Progressive supranuclear palsy 3 Chronic epilepsy 2
Vasculitis 3 Corticostriatonigral degeneration 2
Alzheimer disease/dementia with Lewy bodies 2 Familial spastic paraplegia 2
Cerebrovascular (vascular dementia, 2 Hashimoto encephalopathy 2
cerebrovascular accident)
Paraneoplastic 2 Hereditary ataxia 2
Otherc 8 Huntington disease 2
Metabolic disorder 2
Primary CNS lymphoma 2
Otherc 5

CNS, central nervous system.


a
One antiglutamic acid decarboxylase, 65 isoform (GAD65), voltage-gated potassium channel, and two novel antineuronal antibodies.
b
One lymphomatosis cerebri.
c
Other University of California, San Francisco Cohort: hydrocephalus, mesial temporal sclerosis, vertigo, germinoma, methylmalonic acidemia,
mulsclerosis, methotrexate toxicity, pathology-proven sarcoid.
d
Gliomatosis cerebri and astrocytoma.
Other German Cohort: leukodystrophy, congophilic angiopathy, fatal familial insomnia, hypoxia, and Niemann-Pick lipoid histiocytosis.
Modified from Geschwind MD. Rapidly progressive dementia: prion diseases and other rapid dementias. Continuum (Minneap Minn). 2010;
16(2 Dementia):3156.

Scheld_Ch18.indd 273 2/21/14 5:39 PM


274 Part II: Viral Infections and Related Disorders

two large prion referral centers. Many cases initially mis-


diagnosed as prion disease had other neurodegenerative ACQUIRED HUMAN PRION
diseases, encephalitis, or autoimmune (antibody-mediated) DISEASES: KURU, IATROGENIC
encephalopathies (135138). Among 1,106 brain autopsies
from patients referred with potential JCD to the NPDPSC,
JAKOB-CREUTZFELDT
32% (342 cases) were negative pathologically for prion dis- DISEASE, AND VARIANT JAKOB-
ease. Among the 304 cases with adequate tissue for diagno- CREUTZFELDT DISEASE
sis, 51% had Alzheimer disease, 12% had vascular disease,
but almost 25% had possibly treatable conditions, most By far, the most efficient route of transmission of prion dis-
commonly autoimmune, neoplastic, or infectious condi- eases is through direct intracerebral inoculation, yet acquired
tions (139). Unfortunately, the reverse is also true in that forms of prion disease would be virtually nonexistent in nature
prion diseases are often misdiagnosed as other conditions. if this were the only mode of transmission. Indeed, similarly to
In our own recent study of 163 pathology-proven sJCD sub- viruses, bacteria, and parasites, prions have been shown to in-
jects, patients were an average of two thirds of the way vade host mammalian organisms via parenteral and oral routes.
through their disease course before sJCD was diagnosed Oral transmission of prion disease has been demonstrated in
clinically. Infections (non-prion) was the third most com- humans as well as several animal prion diseases (142,143).
mon misdiagnosis category; the top two diagnostic misdi- More specifically, in humans, variant Jakob-Creutzfeldt disease
agnosis categories were other neurodegenerative diseases (vJCD) and kuru are acquired via oral transmission (although
and autoimmune encephalopathies (91). The authors of vJCD can also be transmitted by blood products; see the follow-
this chapter have written several papers on the workup and ing section on vJCD). Acquired prion diseases can also occur
evaluation of patients with RPD, including prion disease iatrogenically via neurosurgical manipulation with contami-
(112,140,141). Some suggested diagnostic tests for a pa- nated instruments, from contaminated corneal and dura mater
tient with suspected JCD or another form of RPD is shown grafts, through EEG electrodes, and from intramuscular injec-
in Figure 18.9. tion of cadaveric-derived human pituitary hormones (144,145).

Blood tests CSF Imaging Urine/Other

Basic panel of tests

- Complete blood count - Cell count and - Brain MRI - Urine analysis
- Basic metabolic panel (Ca,P,Mg) differential (including FLAIR, (and culture if
- Liver function tests (including ammonia) - Protein DWI and ADC indicated)
- Renal function tests - Glucose sequences), at - EEG
- Thyroid function tests - lgG index least one scan with
- Oligoclonal bands and without
- Anti-TG and Anti-TP antibodies
contrast
- Vitamin B12/MMA/homocysteine - VDRL
- Rheumatologic screen (ANA, ESR, CRP, - 14-3-3/NSE/total
RF, ANCAs, SSA, SSB) tau
- Rapid plasma reagin (RPR)
- HIV serology
- Paraneoplastic/autoimmune antibodies

Tests to consider in selected cases


- Lyme disease (in endemic areas) - Bacterial, fungal, - Cancer screen (CT - Heavy metal screen
- Cancer screen acid-fast bacilli chest, abdomen, (24h urine)
- Blood smear stains and cultures and pelvis with and - Copper (24h urine)
- Cytology without contrast;
- Coagulation profile - Porphobilinogen
- Flow cytometry mammogram;
- Hypercoagulability testing (PBG)/delta-
body PET scan)
- Whipple PCR aminolevulinic acid
- Copper and ceruloplasmin - MR angiography (ALA) in urine (24h)
- Additional rheumatologic tests - Cryptococcal or brain
antigen - EMG/nerve
(complement, dsDNA, anti-Sm, anti-RNP, angiogram
- Viral PCRs and conduction study
anticardiolipin, anti-SCL 70, Anti-Jo, anti- - MR spectroscopy
cultures - Brain biopsy
centromere antibodies) - Carotid ultrasound
- Echocardiogram

FIGURE 18.9 Suggested diagnostic tests for initial rapidly progressive dementia evaluation. anti-TG,
antithyroglobulin; anti-TP, antithyroperoxidase; MMA, methylmalonic acid; ANA, antinuclear antibody;
ESR, erythrocyte sedimentation rate; CRP, C-reactive protein; RF, rheumatoid factor; ANCA, antineutro-
phil cytoplasmic antibody; VDRL, Venereal Disease Research Laboratory; NSE, neuron-specific enolase;
PCR, polymerase chain reaction. (From Paterson RW, Takada LT, Geschwind MD. Diagnosis and treat-
ment of rapidly progressive dementias. Neurol Clin Pract. 2012;2[3]:187200.)

Scheld_Ch18.indd 274 2/21/14 5:39 PM


Chapter 18: Human Prion Diseases 275

of prion disease, as well as in vJCD, the spleen and lymph


Prions Invade the Host Organism nodes have been shown to be the first sites of PrPSc replication
Analogously to Other Infectious Agents after peripheral inoculation. This notion is supported by the
fact that splenectomy delays the onset of orally transmitted
A basic understanding of the mechanisms of spread of pri- prion disease in mice (164), whereas athymia does not (165),
ons after oral inoculation in animal models serves to high- thus indicating that the B cell immune system plays a major
light the infectious nature of prions. Owing to the unique role in prion disease spread, as has been shown experimentally
biochemical and biophysical properties of PrPSc, prions are (166168). Moreover, prions accumulate in the appendix of
able to resist the low pH and proteolytic environment of patients with vJCD before they become symptomatic (169),
the upper gastrointestinal tract. Indeed, even direct intra- and tonsil biopsy has been used to diagnose vJCD in the pre-
gastric inoculation of PrPSc, in which it is exposed to the clinical phases (170). As discussed previously, PrPSc requires
low pH of stomach acid, leads to the development of prion the presence of PrPC for infectivity (171,172). Therefore, in
disease (146). Once prions reach the small intestine, they order for prion disease to propagate within the lymphoid sys-
enter the gut-associated lymphoid tissue (GALT), accumu- tem, it must express PrPC; this has been shown experimentally
lating in Peyer patches (147,148). The exact mechanisms of (173). Taken together, this evidence suggests that lymphoid or-
absorption of PrPSc from the gut lumen into Peyer patches is gans influence the hosts susceptibility to peripheral infection
a matter of debate (149); there is strong evidence to suggest and serve as foci of propagation and spread to the CNS via the
that microcytic cells (M cells), which are specialized cells hemopoietic system.
within follicle-associated epithelia that overlie organized Aside from direct transmission of prions occurring from
mucosa-associated lymphoid tissues (MALTs), are key play- the autonomic nervous system into the central nervous system,
ers for the transportation of PrPSc across the gastrointes- there is also considerable evidence to suggest that prions are
tinal epithelium (150,151). Recent studies also suggest M carried to the brain through the blood by crossing the blood
cellindependent routes of PrPSc transportation (149,152). brain barrier (BBB). Prions can be detected in blood plasma
Once inside enterocytes, prions are transported into Peyer in animals (174), and vJCD can be transmitted via blood
patches mainly by dendritic cells, which are bone marrow transfusion (175), thus suggesting that blood has the ability
derived antigen-presenting cells (153), with macrophages to harbor and transmit disease. Furthermore, PrPC has been
also playing a somewhat unclear role in this process (154). shown to participate in the transport of molecules across the
Accumulation of PrPSc within Peyer patches is a crucial step BBB (176), and radioactively labeled PrPC has been shown to
in allowing efficient spread of prions from the gastrointesti- cross the BBB in both the brain-to-blood and blood-to-brain
nal system into the central nervous system. Indeed, the ab- directions (177).
sence of Peyer patches impairs neural invasion of PrPSc in
animals (155).
After PrPSc accumulates in GALT, and in Peyer patches
in particular, prions infiltrate the enteric nervous system. Kuru
Evidence for direct spread from GALT to the enteric nervous
system is most clearly supported by the fact that the regions As discussed in detail previously in the section on the histori-
of myenteric (Auerbach) plexuses with the most robust accu- cal background of prion diseases, kuru is an acquired form of
mulation of PrPSc are those closest to Peyer patches harboring prion disease due to endocannibalism, confined to the Fore
PrPSc (151). Clinicopathologic studies in animals show that ethnic group in Papua New Guinea. It has largely become ex-
after entering the enteric nervous system, prions reach the tinct after the cessation of endocannibalism by missionaries.
brain via the autonomic peripheral nervous system (primarily Due to an incubation period as long as 50 years or greater,
the sympathetic nervous system) and spinal cord or by direct very rare cases are still being identified (10).
spread into the medulla oblongata through vagal nerve efferent
fibers. In mice infected intraperitoneally with PrPSc, prion ac-
cumulation occurred first in the splanchnic autonomic (mostly
sympathetic) fibers, which served as conduits for prion spread Variant Jakob-Creutzfeldt Disease
to the midthoracic spinal cord, from where prions ascended
into the brain via the intermediolateral cell columns (156). A vJCD was first identified in the United Kingdom in 1995 (178)
similar pattern of spread has been described in a hamster ani- and quickly garnered global media and epidemiologic atten-
mal model (157). Furthermore, PrPSc inoculated into the sci- tion due to its association with bovine spongiform encepha-
atic (158) and optic (159) nerves of mice leads to prion-related lopathy (BSE) (mad cow disease) and that it was affecting
brain degeneration, further proof that the peripheral nervous young persons. BSE is unique in that it is the only form of
system can serve as a conduit for prion spread. Moreover, nonhuman prion disease known to be transmissible directly
sympathectomy delayed or prevented neural invasion of PrPSc to humans. It is hypothesized that BSE in bovine populations
from GALT. (160) occurred through contamination of cattle feed with scrapie-
Interestingly, other experiments that probed the temporo- infected sheep products (179181). Worldwide, more than
spatial spread of PrPSc using immunohistochemical methods 180,000 cattle are thought to have been infected with BSE, and
showed an alternative, direct route of spread from the enteric the vast majority of these were in the United Kingdom (182).
nervous system to the medullary dorsal motor nucleus in the Due to heightened awareness and more rigorous screening of
medulla oblongata, from where prions ascend into the brain livestock, however, the incidence of BSE has been dramatically
(161,162). Similar patterns of neural invasion of prion disease reduced since 1992 (182). Two new vJCD cases were identi-
have been described in other animals, including cattle (163). fied in the United Kingdom in 2011, and to our knowledge,
Together, these experiments suggest that the vagus nerve serves no new cases of vJCD were reported in the United Kingdom
as conduit of prion spread. in 2012 or 2013. As of November 2013, 228 cases have been
There is also experimental evidence suggesting that prions reported worldwide (183). The relationship between the num-
spread from GALT into more distant lymphoid organs before ber of BSE and vJCD cases over time in the United Kingdom is
reaching the central nervous system. In several animal forms shown in Figure 18.10.

Scheld_Ch18.indd 275 2/21/14 5:39 PM


276 Part II: Viral Infections and Related Disorders

FIGURE 18.10 Annual frequency of bovine spongiform encephalopathy (BSE) and variant JCD in Great
Britain, 1988 to 2010. (From Engleberg NC, Dermody T, DiRita V. Schaechters Mechanisms of Microbial
Disease. 5th ed. Baltimore: Lippincott Williams & Wilkins; 2013:557.)

Although vJCD shares many features with sJCD, the clinical has focused heavily on T2-weighted sequences (i.e., proton
presentation of vJCD differs in several salient ways. Patients density, T2 and FLAIR imaging), primarily because the pre-
infected with vJCD are often younger, with a median age of ponderance of data has been collected in the United Kingdom,
onset around 27 years (range 12 to 74 years) (184) and a lon- where the acquisition of DWI sequences is uncommon. Thus,
ger median disease duration of about 14.5 months (184) than the sensitivity and specificity of DWI for the pulvinar sign is
patients with sJCD. The PRNP codon 129 polymorphism has not yet known.
been identified as a critical susceptibility factor for the acquisi-
tion of vJCD, with nearly every reported case thus far having
been found to be homozygous for methionine at this location
(185,186). Psychiatric symptoms are often an early, prominent
feature, occurring as long as 6 months before obvious neuro-
logic impairments manifest. As a result, many patients with
vJCD are often initially thought to have psychiatric etiologies
for their illness (184). Sensory symptoms (dysesthesias), often
painful; cerebellar signs; and involuntary movements such as
dystonia, myoclonus, and chorea often become more evident
later on in the disease course (184,187).
Diagnostic test results are often also divergent with those
seen in sJCD. CSF tests are often less revealing, with lower
sensitivity and specificity than they are in the differential
diagnosis of sJCD (188). Patients with vJCD rarely exhibit
the classic PSWCs seen in sJCD on EEG, and if so, only in
the final stages of disease progression (189). The brain MRI
hallmark of vJCD is the pulvinar sign, in which bilateral
hyperintensity in the pulvinar nucleus in the posterior region
of the thalamus is brighter than in the anterior putamen
FIGURE 18.11 Magnetic resonance imaging of a patient with vari-
on T2-weighted, FLAIR, or possibly other MRI sequences ant Jakob-Creutzfeldt disease (vJCD). A: Increased signal intensity
(Fig. 18.11). In 85% of patients with vJCD, the pulvinar sign in the pulvinar (arrows; pulvinar sign). B: Increased signal intensity
is seen on their first exam (190). This is a finding rarely exhib- in the pulvinar and dorsomedial thalamus (arrow; double hockey
ited in other forms of human prion disease (191), although it stick sign). (From Engleberg NC, Dermody T, DiRita V. Schaechters
has been reported in sJCD (192). The literature investigating Mechanisms of Microbial Disease. 5th ed. Baltimore: Lippincott
the use of brain MRI in the differential diagnosis of vJCD Williams & Wilkins; 2013:558.)

Scheld_Ch18.indd 276 2/21/14 5:39 PM


Chapter 18: Human Prion Diseases 277

FIGURE 18.12 Human tissues and blood involved


in propagation and transport of prions in vJCD.
Orally ingested prions are intestinally absorbed and
transported to the blood and lymphoid fluids. After a
peripheral replication step in the spleen, appendix, ton-
sils, or other lymphoid tissues, prions are transported
to the brain primarily by peripheral nerves. Direct
penetration into the brain across the bloodbrain bar-
rier is conceivable. (From Aguzzi A, Heikenwalder M.
Pathogenesis of prion disease: current status and future
outlook. Nat Rev Microbiol. 2006;4[10]:765.)

As with other forms of non-gPrD, the definitive diagnosis medical/surgical procedures or blood products is not known.
of vJCD relies on neuropathologic evidence of the presence of Measures were taken in the United Kingdom to prevent trans-
PrPSc in the central nervous system. In vJCD, there is typically mission of vJCD through blood products with donor selection,
abundant PrPSc deposition and vacuolation in the neuropil as leukodepletion of blood (WBCs carry a majority of prions in
well as the presence of florid plaques composed of multiple blood), and efforts toward developing methods to detect PrP
fibrillary PrP plaques surrounded by spongiform vacuoles. in blood (182). It is important to note that although trans-
These are seen most commonly in the molecular layer of the mission through blood products has been reported in vJCD,
cerebellum (130). Because vJCD is acquired peripherally, PrPSc there are no known cases to date of transmission from sJCD
accumulation often is identified in tonsillar tissue and other patients through blood transfusion.
parts of the lymphoreticular system (see Fig. 18.12) (193).
Importantly from an infectious disease perspective, it was re-
cently discovered that vJCD could also be acquired from trans- Iatrogenic Jakob-Creutzfeldt Disease
fusion of vJCD-contaminated blood products (194,195). Three
cases of vJCD have been reported in patients who received To date, with the exception of kuru, human-to-human trans-
(contaminated) blood transfusions prior to 1999 and developed mission of prion disease has only occurred iatrogenically. The
symptoms from 6.5 to 7.8 years after the date of implicated first reported case of suspected iatrogenic prion disease was
transfusion; all cases clinically and neuropathologically were published in 1974 of a 55-year-old female who developed
similar to other cases of vJCD (196). Two additional patients, RPD beginning 18 months after receiving a corneal transplant
both heterozygous at codon 129 in PRPN (129MV), received from a donor who later was found to have autopsy-confirmed
contaminated blood products (nonleukodepleted red blood cells JCD. The patient died of JCD 8 months later (199).
and factor VIII, respectively) and died of nonneurologic causes Multiple other cases of suspected iatrogenic Jakob-
but had positive prion testing in their lymphoreticular system Creutzfeldt Disease (iJCD) have been reported in the litera-
but without clinical signs of vJCD and without vJCD prions in ture since the 1970s. These have been carefully reviewed by
their brains; it is not known if they would have ever developed Paul Brown and colleagues up to 2012 (76,144,200). Several
clinical vJCD (145,186). Of great concern is that these cases other vectors of transmission have been implicated in iJCD:
show that vJCD was transmissible by blood product donation dura mater grafts, EEG leads, neurosurgical instrumentation,
years before the donors became symptomatic. human pituitary hormone extracts, and packed red blood
In order to determine the number of persons in the United cells. These modes of transmission will be reviewed briefly.
Kingdom who might be harboring latent vJCD, and be po- Since the original report of iJCD via corneal transplanta-
tential transmissible carriers of vJCD prions, a few stud- tion, only two other similar cases have been published in the
ies have analyzed anonymized U.K. appendix and/or tonsil literature. The first occurred in Japan and the second case in-
samples. An initial large study found a prevalence of 1 per volved a German patient (201). Neither case is very conclusive
4,000 appendices positive for vJCD prions (197), whereas for a causal relationship, however, as the medical history of
a follow-up larger study found a higher prevalence of 1 per the donor of the Japanese case is unknown and the German
2,000 appendices positive, leading to assumptions that there patient developed JCD 30 years after receiving corneal trans-
are subclinically infected persons in the population who might plants from a donor who died of JCD. Nevertheless, potential
be transmissible carriers (198); their risk of either develop- donors of any tissue, including corneas, should be screened for
ing symptomatic vJCD and/or passing it on to others through dementia, particularly prion disease.

Scheld_Ch18.indd 277 2/21/14 5:39 PM


278 Part II: Viral Infections and Related Disorders

Three years after the first case of iJCD was published to produce the NHPP-distributed hGH (207). The first case in-
in 1974, Bernoulli and colleagues (202) reported on two volved a 39-year-old Austrian man who died of JCD approxi-
patientsa 23-year-old woman and a 17-year-old boywho mately 22 years after receiving commercial cadaveric hGH in
developed JCD within 2 years of having depth electrodes placed the mid-1980s. The second case was a 33-year-old man who
as part of an EEG study for epilepsy. It was later found that developed rapid dementia approximately 26 years after the
the same electrodes had been previously used in a 69-year-old midpoint of commercial hGH treatment and died 9 months
woman who died from JCD. Transmission occurred despite later from probable sJCD. His clinical syndrome and CSF and
standard sterilization procedures at the time (70% alcohol brain MRI findings were consistent with JCD, but autopsy
and formaldehyde vapor). More than 2 years later, and after was not performed. Both patients were treated with different
repeated sterilizations, the same electrodes were implanted in extracts of commercial cadaveric hGH, but it appears that they
the cortex of a chimpanzee: the animal became symptomatic received some extracts from the same brand in the same year
18 months later and subsequently died of JCD, confirming (Crescormon, by Kabi Pharmaceuticals, Sweden) (207). More
the cases were iatrogenic (203). To date, this represents the than 10,000 people, mostly outside the United States, received
only case of experimentally proven iJCD. A handful of other commercial cadaveric hGH produced by different companies.
cases of JCD have been associated with neurosurgical instru- Many details regarding sourcing and production methods of
mentation, but not proven, with a mean incubation period of these extracts are lacking.
17 months (range 12 to 28 months). Incubation periods for hGH-associated iJCD range from 5
By far, the most common sources of iJCD has come from to 42 years (mean 17 years) from the midpoint of treatment
cadaveric prion-contaminated growth hormone extracts and duration (206). Clinically, iJCD secondary to hGH therapy
dura mater grafts. The first published case associated with tends to produce a clinical syndrome that is reminiscent of
cadaveric human growth hormone (hGH) extracts involved a kuru, with prominent early cerebellar signs and late cognitive
20-year-old man with congenital idiopathic hypopituitarism dysfunction. Although there is not much data on MRI findings
who had been treated for approximately 14 years with growth in hGH-associated iJCD, they appear to range from isolated
hormone extracts obtained from pooled human cadaver pitu- cerebellar involvement (208) to appearing similar to sJCD,
itary glands (204). He developed ataxic gait and dysarthria, often with deep nuclei involvement (207).
followed by cerebellar dysfunction, myoclonus, and dementia Susceptibility to infection is influenced by the methio-
that progressed rapidly to death due to JCD. Concurrently, nine/valine polymorphism at codon 129 of the PRNP gene.
two similar cases were reported to the Centers for Disease Methionine homozygosity is present in 55% of affected U.S.
Control and Prevention (CDC): one involved a 22-year-old and French individuals (compared to 40% methionine homo-
man with long-standing growth hormone deficiency who had zygosity in the general population), whereas valine homozygos-
been treated with cadaveric hGH for approximately 8 years ity is far more common in the U.K. cohort. Moreover, patients
and the other case involved a 34-year-old man with congenital who were heterozygous at codon 129 tend to have longer incu-
hypopituitarism who had received cadaveric hGH for approx- bation periods (30 years) than normal (mean 17 years) (209).
imately 6 years; the former case died of autopsy-confirmed Dura materassociated iJCD is as common as hGH-associ-
CJD and the latter case died after a course of RPD highly sus- ated iJCD worldwide. As of 2012, there are 228 reported cases
picious for CJD (205) (http://www.cdc.gov/mmwr/preview/ of dura materassociated JCD, and 142 of these have occurred
mmwrhtml/00000563.htm). in Japan (200,210). The remaining cases have been reported
The hormone extracts used to treat these patients was pro- in Argentina, Australia, Canada, and Europe. Most iJCD cases
duced and distributed by the National Hormone and Pituitary due to dura mater graft contamination have been traced back
Program (NHPP). Approximately 10,000 U.S. patients re- to grafts manufactured by one German company, which was
ceived cadaveric hGH through the NHPP between 1963 and a global provider of dura mater grafts in the 1980s (manufac-
early 1985 from different hGH batches derived from a pool of turing ceased in 1987, but patients received grafts until 1993)
approximately 16,000 cadaver pituitary glands. The average (200). The main reason why so many cases have been reported
duration of therapy was 4 years. Each patient received extracts in Japan relates to the high frequency of use of these grafts in
from up to three different batches per year. Since the original Japan compared to other countries; it is estimated that 20,000
reports of hGH-associated iJCD, a total of 226 similar cases Japanese patients received contaminated grafts annually dur-
have been reported throughout the world, with most originat- ing the 1980s, often for nonlife-threatening conditions. Other
ing in France (119 cases), in the United Kingdom (65 cases), factors, however, may have contributed to the high incidence,
and in the United States (29 cases). In the United States, all re- such as longer survival, and hence longer incubation times,
ported cases have occurred in patients who received cadaveric among Japanese recipients (210).
hGH prior to 1977, before a column chromatography step was In most cases the clinical and neuropathologic features of
added to the manufacturing purification process of cadaveric dura materassociated iJCD appear similar to those of sJCD
hGH extracts (206). It is estimated that around 2,700 people (200). In Japan, however, there is evidence suggesting that
received NHPP extracts prior to 1977, and to our knowledge, dura materassociated iJCD may have distinctive clinical and
the last case in the United States was reported in 2009. In the molecular features. In Japanese patients with dura materas-
United Kingdom, on the other hand, cases continue to appear sociated iJCD, two distinct phenotypes were identified: a
at a rate of approximately 2 per year, the most recent case was dominant type, comprising nearly two thirds of cases, that is
reported in 2011. In France, all cases are thought to have arisen pathologically and clinically nearly identical to sJCD and an
within a 20-month period between December 1983 and July atypical type with prominent plaque formation in the brain
1985, when up to 50% of hormone extracts produced became and other atypical clinical features such as slower disease pro-
contaminated due to faulty manufacturing processes (206). gression. Both of these types were associated with methionine
It is important to note that the vast majority of pituitary homozygosity at codon 129 of the PRNP gene. These differ-
hormoneassociated iJCD cases have been associated with ences have led some investigators to postulate contamination
hormone extracts produced and distributed by the NHPP. of the dura mater grafts with different prion strains (211).
There are at least two case reports, however, of iJCD asso- Worldwide, the mean incubation period of dura materassoci-
ciated with commercially available cadaveric hGH extracts, ated iJCD is 12 years, with the shortest and longest reported
which were produced in different laboratories from those used incubation times being 16 months and 30 years (200).

Scheld_Ch18.indd 278 2/21/14 5:39 PM


Chapter 18: Human Prion Diseases 279

The emergence of dura materassociated iJCD occurred ing these three familial presentations of gPrD, it is important
almost concurrently to that of hGH-related iJCD, and the inci- to mention that all three forms were described prior to the
dence of both peaked in the mid-to-late 1990s and has steadily identification of the PRNP gene, hence the description of these
declined since the institution of recombinant growth hormone diseases is primarily based on clinicopathologic observations
extracts (growth hormone extracts and gonadotropins have (214). After the identification of PRNP and its mutations, a
been manufactured in vitro since 1987) and synthetic dura much wider spectrum of phenotypes associated with gPrD is
mater grafts (77). Nevertheless, given the long incubation emerging. In a recent study, for example, researchers identified
times reported for both modes of transmission, it is expected a consistent phenotype of chronic diarrhea with autonomic
that additional cases might still occur from past exposure. failure and a length-dependent axonal, predominantly sensory,
Likewise, the medical community across different specialties peripheral polyneuropathy associated with a specific PRNP
has adopted effective infection control measures that have de- mutation (PRNP Y163X truncation mutation) (218). These
creased the risk of iJCD significantly. Patients with suspected patients begin to develop peripheral neurologic symptoms in
JCD are excluded as blood, tissue, or organ donors. Clinicians early adulthood, whereas cognitive decline does not occur
evaluating patients with suspected JCD should always ask until their 40s or 60s, thereby suggesting that genetic forms
about any prior human pituitary hormone use, ophthalmo- of prion diseases can affect the peripheral nervous system be-
logic surgery, and neurosurgical procedures. fore leading to encephalopathy and dementia. Furthermore,
Measures to prevent iJCD vary by country and by health a recent case of atypical parkinsonism was linked to a spe-
care institution. Our institutions infection control guide- cific mutation in the PRNP gene, the D202N mutation, that
lines for managing suspected human prion cases are avail- had previously only been associated with GSS (219). Indeed,
able online at http://infectioncontrol.ucsfmedicalcenter the phenotypic heterogeneity associated with PRNP muta-
.org/ICMANUAL2007/Section4/Sec%204.2%20 tions is certainly not encompassed by the three classic forms
Human%20Prion%20Policy_5-31-2012.pdf. The World of gPrD mentioned previously. Clinical features of each gPrD
Health Organization and the CDC have published iJCD can be highly variable, not only within the same mutation but
prevention guidelines as well as information on the dis- also within a single family (215). In order to help categorize
tribution of tissue infectivity (212,213) (http://www.who gPrDs more easily, however, we will briefly discuss each one
.int/csr/resources/publications/bse/whocdscsraph2003.pdf, of these forms.
http://www.cdc.gov/ncidod/dvrd/cjd/qa_cjd_ infection_
control.htm).
Familial Jakob-Creutzfeldt Disease
HUMAN GENETIC PRION For most mutations causing fJCD, the clinical presentation usu-
ally is indistinguishable to that of sJCD, with RPD, ataxia, and
DISEASES: FAMILIAL JAKOB- other motor manifestations. More than 15 mutations have been
CREUTZFELDT DISEASE, associated with fJCD, most are point (missense) mutations but
GERSTMANN-STRUSSLER- some are insertion mutations and a deletion (34,214). The most
common fJCD mutation worldwide is E200K (214) found most
SCHEINKER DISEASE, AND FATAL commonly among Libyan Jews and Slovakians (220). Similarly
FAMILIAL INSOMNIA to sJCD, brain MRI is helpful for the diagnosis of fJCD, and
both fJCD and sJCD have overlapping findings on MRI. The
gPrDs account for approximately 10% to 15% of all cases of MRI appearance of fJCD seems to vary according to mutation
prion disease. They are associated with autosomal dominant type, however. For example, fJCD patients due to E200K muta-
mutations in the human PrP gene, PRNP, on the short arm of tion typically show symmetric, prominent striatal hyperinten-
chromosome 20. PRNP mutations are of different kinds, in- sities on MRI, with less prominent cortical ribboning (221),
cluding point mutations, insertions, truncations, and deletions whereas patients with V180I mutations often shows cortical
(34,214,215). There are presently more than 30 PRNP muta- ribboning with minimal or absent basal ganglia hyperintensities
tions associated with gPrD, all of which lead to disease via the (222). An MRI of a patient with fJCD due to E200K is shown in
production of a mutant PrP molecule (PrPSc) (34,214,215). Figure 18.13A. Also, depending on the mutation, the EEG often
Some gPrDs have been shown to be transmissible to laboratory shows PSWCs in fJCD, particularly in late stages. CSF biomark-
animals, although most have not been tested for transmission ers (14-3-3 protein, NSE, t-tau) in fJCD often are elevated, but
and a few have failed to transmit (216). Genetic forms of prion with typically lower sensitivity than in sJCD (115).
disease can be diagnosed through DNA testing of blood in
vivo or through autopsy tissue. Most known PRNP mutations
have very high penetrance (nearly 100%), yet more than 60% Gerstmann-Strussler-Scheinker Disease
of patients with gPrD do not report a positive family history
of prion disease. This is likely due to a combination of factors, Patients with GSS often become symptomatic earlier than pa-
including misdiagnosis. Some mutations, such as the E200K tients with sporadic prion disease (disease onset is typically in
mutation, however, have reduced penetrance (217). Patients the 50s or younger). Clinically, GSS often manifests with the
with gPrD are typically younger at onset (approximately 40s insidious onset of movement abnormalities, including progres-
to 60s) and have a slower and longer disease course compared sive ataxia or parkinsonism. Cognitive impairment is usually
to sJCD patients, although there is great clinical variability a late feature, although some mutations present with early
among different mutations, in part due to other genetic (codon dementia and/or behavioral abnormalities. Among all forms
129 polymorphisms) and epigenetic factors that influence phe- of human prion disease, patients with GSS are perhaps the
notypic expression (34,214). most likely to be misdiagnosed as having other neurodegen-
As stated previously in the basic prion genetics section of erative diseases or even non-neurodegenerative neurologic and
this chapter, there are three main forms of gPrD in humans: psychiatric illness. This occurs largely because GSS may have
Gerstmann-Strussler-Scheinker disease, familial Jakob- an unusually long survival compared to other forms of prion
Creutzfeldt disease, and fatal familial insomnia. Before discuss- disease (214,223,224).

Scheld_Ch18.indd 279 2/21/14 5:39 PM


280 Part II: Viral Infections and Related Disorders

FIGURE 18.13 Axial MRI from representative cases of familial Creutzfeldt-Jakob disease (fCJD)
(A), Gerstmann-Strussler-Scheinker (GSS) (B), and non-prion causes of rapidly progressive dementia
(npRPD) (C). A: An fCJD case (E200K mutation) showing neocortical (solid arrow) involvement more
evident in the right hemisphere, especially in the right frontal lobe; limbic involvement (dashed arrow)
more evident in the right anterior cingulate and right insula; and subcortical (dotted arrow) hyperintensi-
ties, greater in DWI than in FLAIR images. Note the subcortical ADC hypointensity in bilateral striatum.
The image was read as Creutzfeldt-Jakob disease (CJD). B: A GSS case (F198S mutation) with bilateral
limbic hyperintensity in the anterior cingulate, insula, and subtle involvement in the mesiotemporal cor-
tex, equally evident in DWI and FLAIR images. Image read as not CJD. C: An npRPD patient with limbic
encephalopathy due to anti-AMPAR with anti-Sox2 antibodies and small cell lung cancer. Note signifi-
cant bilateral hyperintensity in mesiotemporal cortex (including hippocampus and amygdala), insula, and
cingulate, greater on FLAIR than on DWI images. MRI read as not consistent with CJD. (From Vitali
P, Maccagnano E, Caverzasi E, et al. Diffusion-weighted MRI hyperintensity patterns differentiate CJD
from other rapid dementias. Neurology. 2011;76[20]:17111719.)

There is considerable phenotypic variability in GSS within and/or personality changes (irritability, short temper, aggres-
and between mutations and families (34,225,226). There are sion, antisocial behavior, and aggression) years before the onset
at least 10 PRNP mutations associated with GSS, these include of GSS (33). Neuropsychologic testing of these cases almost in-
several missense mutations, a stop mutation, and insertion variably shows frontal executive impairment as well as frequent
mutations (octapeptide repeat insertions [OPRIs]). In general, visuospatial, language, and memory impairments (224).
OPRI mutations with 5 or more additional octapeptide repeats EEGs of patients with GSS usually reveal slow waves, not
(repeats of 24 base pairs) are associated with a long duration the PSWCs seen in sJCD. Likewise, CSF biomarkers that offer
(several years), GSS phenotype, whereas OPRI mutations with some diagnostic use in sJCD (protein 14-3-3, NSE, and t-tau)
4 or fewer repeats may present as sJCD (34,214). An 8-octa- are typically not elevated in GSS (115). Given that these CSF
peptide repeat insertion mutation (OPRI-8) of PRNP results in a biomarkers are thought to be released into the CSF as a result
GSS phenotype with prominent early neuropsychiatric features, of rapid neuronal injury or neurodegeneration, it is possible
often in the late 20s, with a long disease course of several years. that the slow course of GSS is preventing the accumulation of
These patients have ended up hospitalized in psychiatric institu- these markers in the CSF of affected patients (121). Finally,
tions for management of manic manifestations (223). Likewise, brain MRI investigations in patients with GSS usually do not
patients with 6-OPRI mutations may demonstrate psychologic reveal the cortical ribboning or deep nuclei hyperintensities

Scheld_Ch18.indd 280 2/21/14 5:39 PM


Chapter 18: Human Prion Diseases 281

seen in sJCD and in other forms of prion disease, although not have a PRNP mutation, then all blood relatives are no more
patients with some PRNP mutations associated with GSS do at risk of developing JCD than anyone else in the population.
have brain MRI findings characteristic of prion disease (97).
An MRI of a GSS patient due to an F198S mutation is shown
in Figure 18.13B. TRANSMISSIBILITY OF HUMAN
PRION DISEASES: EVALUATING
Fatal Familial Insomnia AND CARING FOR PATIENTS
FFI is a very rare form of gPrD caused by a single PRNP point Although human prion diseases can be infectious, they are
mutation, D178N, with codon 129 having methionine (129M) very difficult to transmit, typically requiring direct contact
on the same chromosome (cis). Curiously, patients with the with the nervous system. Most cases of human prion disease
D178N mutation with the cis codon 129 being valine (129V) are not due to infection, however, but occur spontaneously. As
usually present with an sJCD clinicopathologic phenotype. discussed in the section, Acquired Human Prion Diseases,
Thus, codon 129 can have a profound effect on the expres- the risk of transmission through certain neuroinvasive medical
sion of human prion disease. The codon 129 polymorphism procedures is not insignificant (76), and new cases of poten-
on the trans or normal (without the mutation) allele also af- tial transmission through neurosurgical procedures, particu-
fects disease presentation; for example, FFI patients with larly brain biopsies without using JCD precautions (229), are
codon 129 methionine homozygosity have shorter disease still occurring. This is particularly important as all prion dis-
durations than those with a trans valine at codon 129 (227). eases are uniformly fatal, and to date, no treatments or cures
Patients with FFI usually become symptomatic in their late 40s have been identified (230). Therefore, certain precautions are
(48 to 49 years; range 20 to 72 years), usually with severe, advised when evaluating patients with prion disease (229).
progressive insomnia over several months, followed by other There is no evidence that human prion diseases are transmit-
manifestations such as dysautonomia (e.g., tachycardia, hy- ted through casual, or even intimate, contact. In recorded
perhidrosis, and hyperpyrexia) as well as motor and cognitive history, to our knowledge, there is only one case of spouses
manifestations appearing later in the illness. Mean survival is both developing sJCD (231) given the incidence and a persons
about 18 months (range 7 to 33 months), which is slightly lifetime risk of developing prion disease (estimated to be 1 in
longer than most sJCD patients (34,77). Similarly to patients 10,000s), one would have expected there to be more famil-
with GSS, EEG investigations in patients with FFI usually ial nongenetic cases to occur by chance alone. This suggests
show generalized slowing, not PSWCs. Brain MRI is usually that even intimate contact is not a risk factor for human prion
unremarkable, but fluorodeoxyglucose positron emission to- disease transmission. Although prion diseases are not trans-
mography (FDG-PET) imaging reveals thalamic and cingulate mitted easily, standard universal precautions, such as using
hypometabolism, often even before disease onset (228) CSF gloves when handling bodily fluids and waste, are advised
biomarkers in general have very low sensitivity in FFI (115). when caring for patients with suspected prion disease. More
The infectious nature of human gPrDs is not clear, but over- information regarding care, management, autopsy, and burial-
all they appear very difficult to transmit and the risk of trans- related issues can be found at the Creutzfeldt-Jakob Disease
missibility is presumed to be very low. There are no known Foundation (www.cjdfoundation.org) and UCSF Memory and
cases of transmission of gPrD from human to human. Because Aging Center (http://memory.ucsf.edu) Web sites.
of the theoretical risk of transmission through blood products,
persons with sJCD, gPrD, or at risk for gPrD (asymptomatic
either with a mutation or have not had PRNP testing) should CONCLUSIONS
not donate blood products. The American Red Cross does
not accept blood product donations from blood relatives of Prion diseases are unique in medicine because of their mul-
someone who has died from JCD (http://www.redcrossblood tiple modes of occurrence from a single protein or agent. New
.org/donating-blood/eligibility-requirements/eligibility-criteria- data suggest that the mechanisms for prion propagation in the
alphabetical-listing); their presumed reasoning is that there is a body might be relevant for most other neurodegenerative dis-
chance that the JCD patient had gPrD and thus blood relatives eases (232). This suggests that what we learn from this rare
are at risk for also carrying a mutation and thus being potential family of diseases might have far greater applicability in our
transmissible carriers. Realistically, if a patient with JCD does understanding of more common conditions.

References

1. Prusiner SB. Prions. Proc Natl Acad Sci U S A. 1998;95(23):1336313383. 9. Stocklin WH. Kuru, the laughing death. Medical and ethnological aspects
2. Cockcroft PD, Clark AM. The Shetland Islands scrapie monitoring and of a puzzling disease in the highland of New Guinea [in German]. Acta
control programme: analysis of the clinical data collected from 772 scrapie Trop. 1967;24(3):193224.
suspects 19851997. Res Vet Sci. 2006;80(1):3344. 10. Collinge J, Whitfield J, McKintosh E, et al. Kuru in the 21st centuryan
3. Hoinville LJ, Tongue SC, Wilesmith JW. Evidence for maternal transmis- acquired human prion disease with very long incubation periods. Lancet.
sion of scrapie in naturally affected flocks. Prev Vet Med. 2010;93(23): 2006;367(9528):20682074.
121128. 11. Berndt RM. In the steps of kuru. Lancet. 1981;1(8216):381.
4. Cuill J, Chelle PL. La maladie dite tremblante du mouton est-elle inocu- 12. Gajdusek DC. Unconventional viruses and the origin and disappearance of
lable? C R Acad Sci. 1936;203:15521554. kuru. Science. 1977;197(4307):943960.
5. Poser CM. Notes on the history of the prion diseases. Part II. Clin Neurol 13. Reid LH, Gajdusek DC. Nutrition in the Kuru region. II. A nutritional
Neurosurg. 2002;104(2):7786. evaluation of traditional Fore diet in Moke village in 1957. Acta Trop.
6. Palsson PA. Dr. Bjorn Sigurdsson (19131959). A memorial tribute. Ann N 1969;26(4):331345.
Y Acad Sci. 1994;724:15. 14. Sorenson ER, Gajdusek C. Nutrition in the Kuru region. I. Gardening, food
7. Thormar H. The origin of lentivirus research: maedi-visna virus. Curr HIV handling, and diet of the Fore people. Acta Trop. 1969;26(4):281330.
Res. 2013;11(1):29. 15. Bennett JH, Rhodes FA, Robson HN. A possible genetic basis for kuru. Am
8. Liberski PP. Kuru and D. Carleton Gajdusek: a close encounter. Folia J Hum Genet. 1959;11(2, pt 1):169187.
Neuropathol. 2009;47(2):114137. 16. Hadlow WJ. Scrapie and kuru. Lancet. 1959;2:289290.

Scheld_Ch18.indd 281 2/21/14 5:39 PM


282 Part II: Viral Infections and Related Disorders

17. Pattison IH. Resistance of the scrapie agent to formalin. J Comp Pathol. 51. Stahl N, Baldwin MA, Teplow DB, et al. Structural studies of the scra-
1965;75:159164. pie prion protein using mass spectrometry and amino acid sequencing.
18. Hunter GD, Millson GC. Studies on the heat stability and chromatographic Biochemistry. 1993;32(8):19912002.
behaviour of the scrapie agent. J Gen Microbiol. 1964;37:251258. 52. Colby DW, Prusiner SB. Prions. Cold Spring Harb Perspect Biol. 2011;
19. Mould DL, Dawson AM, Smith W. Scrapie in mice. The stability of the 3(1):a006833.
agent to various suspending media, pH and solvent extraction. Res Vet Sci. 53. Pan KM, Baldwin M, Nguyen J, et al. Conversion of alpha-helices into
1965;6:151154. beta-sheets features in the formation of the scrapie prion proteins. Proc
20. Gajdusek DC, Gibbs CJ, Alpers M. Experimental transmission of a kuru- Natl Acad Sci U S A. 1993;90(23):1096210966.
like syndrome to chimpanzees. Nature. 1966;209(25):794796. 54. Wille H, Bian W, McDonald M, et al. Natural and synthetic prion
21. Lindenbaum S. Cannibalism, kuru and anthropology. Folia Neuropathol. structure from X-ray fiber diffraction. Proc Natl Acad Sci U S A. 2009;
2009;47(2):138144. 106(40):1699016995.
22. Klatzo I, Gajdusek DC, Zigas V. Pathology of kuru. Lab Invest. 55. Legname G, Baskakov IV, Nguyen HO, et al. Synthetic mammalian prions.
1959;8(4):799847. Science. 2004;305(5684):673676.
23. Gibbs CJ Jr, Gajdusek DC, Asher DM, et al. Creutzfeldt-Jakob disease 56. Greil CS, Vorberg IM, Ward AE, et al. Acute cellular uptake of abnor-
(spongiform encephalopathy): transmission to the chimpanzee. Science. mal prion protein is cell type and scrapie-strain independent. Virology.
1968;161(839):388389. 2008;379(2):284293.
24. Alper T, Haig DA, Clarke MC. The exceptionally small size of the scrapie 57. Magalhaes AC, Baron GS, Lee KS, et al. Uptake and neuritic transport of
agent. Biochem Biophys Res Commun. 1966;22(3):278284. scrapie prion protein coincident with infection of neuronal cells. J Neurosci.
25. Gibbs CJ Jr, Gajdusek DC, Latarjet R. Unusual resistance to ionizing radia- 2005;25(21):52075216.
tion of the viruses of kuru, Creutzfeldt-Jakob disease, and scrapie. Proc 58. Manson JC, Clarke AR, Hooper ML, et al. 129/Ola mice carrying a null
Natl Acad Sci U S A. 1978;75(12):62686270. mutation in PrP that abolishes mRNA production are developmentally nor-
26. Diener TO, McKinley MP, Prusiner SB. Viroids and prions. Proc Natl Acad mal. Mol Neurobiol. 1994;8(23):121127.
Sci U S A. 1982;79(17):52205224. 59. Brandner S, Isenmann S, Raeber A, et al. Normal host prion protein
27. Prusiner SB. Novel proteinaceous infectious particles cause scrapie. Science. necessary for scrapie-induced neurotoxicity. Nature. 1996;379(6563):
1982;216(4542):136144. 339343.
28. Carp RI, Merz PA, Kascsak RJ, et al. Nature of the scrapie agent: current 60. Paquet S, Daude N, Courageot MP, et al. PrPc does not mediate internaliza-
status of facts and hypotheses. J Gen Virol. 1985;66(pt 7):13571368. tion of PrPSc but is required at an early stage for de novo prion infection of
29. Manuelidis EE. Creutzfeldt-Jakob disease. J Neuropathol Exp Neurol. Rov cells. J Virol. 2007;81(19):1078610791.
1985;44(1):117. 61. Jen A, Parkyn CJ, Mootoosamy RC, et al. Neuronal low-density lipopro-
30. Basler K, Oesch B, Scott M, et al. Scrapie and cellular PrP isoforms are tein receptor-related protein 1 binds and endocytoses prion fibrils via recep-
encoded by the same chromosomal gene. Cell. 1986;46(3):417428. tor cluster 4. J Cell Sci. 2010;123(pt 2):246255.
31. Locht C, Chesebro B, Race R, et al. Molecular cloning and complete 62. Hijazi N, Kariv-Inbal Z, Gasset M, et al. PrPSc incorporation to cells
sequence of prion protein cDNA from mouse brain infected with the scra- requires endogenous glycosaminoglycan expression. J Biol Chem. 2005;
pie agent. Proc Natl Acad Sci U S A. 1986;83(17):63726376. 280(17):1705717061.
32. Robakis NK, Devine-Gage EA, Jenkins EC, et al. Localization of a human 63. Rieger R, Edenhofer F, Lasmezas CI, et al. The human 37-kDa laminin
gene homologous to the PrP gene on the p arm of chromosome 20 and receptor precursor interacts with the prion protein in eukaryotic cells. Nat
detection of PrP-related antigens in normal human brain. Biochem Biophys Med. 1997;3(12):13831388.
Res Commun. 1986;140(2):758765. 64. Goold R, Rabbanian S, Sutton L, et al. Rapid cell-surface prion protein
33. Lloyd S, Mead S, Collinge J. Genetics of prion disease. In: Tatzelt J, ed. conversion revealed using a novel cell system. Nat Commun. 2011;2:281.
Prion Proteins. Berlin-Heidelberg: Springer; 2011:122. 65. Goold R, McKinnon C, Rabbanian S, et al. Alternative fates of newly
34. Kong Q, Surewicz WK, Petersen RB, et al. Inherited prion diseases. In: formed PrPSc upon prion conversion on the plasma membrane. J Cell Sci.
Prusiner SB, ed. Prion Biology and Diseases. 2nd ed. Cold Spring Harbor, 2013;126(pt 16):35523562.
NY: Cold Spring Harbor Laboratory Press; 2004:673775. 66. Guest WC, Plotkin SS, Cashman NR. Toward a mechanism of prion mis-
35. Palmer MS, Dryden AJ, Hughes JT, et al. Homozygous prion protein folding and structural models of PrP(Sc): current knowledge and future
genotype predisposes to sporadic CreutzfeldtJakob disease. Nature. directions. J Toxicol Environ Health A. 2011;74(24):154160.
1991;352(6333):340342. 67. Abid K, Morales R, Soto C. Cellular factors implicated in prion replication.
36. Parchi P, Giese A, Capellari S, et al. Classification of sporadic Creutzfeldt- FEBS Lett. 2010;584(11):24092414.
Jakob disease based on molecular and phenotypic analysis of 300 subjects. 68. Miller MB, Wang DW, Wang F, et al. Cofactor molecules induce struc-
Ann Neurol. 1999;46(2):224233. tural transformation during infectious prion formation. Structure.
37. Montagna P, Cortelli P, Avoni P, et al. Clinical features of fatal familial 2013;21(11):20612068.
insomnia: phenotypic variability in relation to a polymorphism at codon 69. Jakob A. Concerning a disorder of the central nervous system clini-
129 of the prion protein gene. Brain Pathol. 1998;8(3):515520. cally resembling multiple sclerosis with remarkable anatomic findings
38. Bendheim PE, Brown HR, Rudelli RD, et al. Nearly ubiquitous tissue dis- (spastic pseudosclerosis). Report of a fourth case. Med Klin. 1921;17:
tribution of the scrapie agent precursor protein. Neurology. 1992;42(1): 372376.
149156. 70. Jakob A. Concerning a disorder of the central nervous system clinically
39. Taraboulos A, Raeber AJ, Borchelt DR, et al. Synthesis and trafficking of resembling multiple sclerosis with remarkable anatomic findings (spastic
prion proteins in cultured cells. Mol Biol Cell. 1992;3(8):851863. pseudosclerosis). Report of a fourth case. Alzheimer Dis Assoc Disord.
40. Sonnino S, Prinetti A. Membrane domains and the lipid raft concept. 1989;3(12):2645.
Curr Med Chem. 2013;20(1):421. 71. Creutzfeldt HG. On a particular focal disease of the central nervous sys-
41. Didonna A. Prion protein and its role in signal transduction. Cell Mol Biol tem (preliminary communication), 1920. Alzheimer Dis Assoc Disord.
Lett. 2013;18(2):209230. 1989;3(12):325.
42. Gauczynski S, Hundt C, Leucht C, et al. Interaction of prion proteins with 72. Richardson EP Jr, Masters CL. The nosology of Creutzfeldt-Jakob disease
cell surface receptors, molecular chaperones, and other molecules. Adv and conditions related to the accumulation of PrPCJD in the nervous system.
Protein Chem. 2001;57:229272. Brain Pathol. 1995;5(1):3341.
43. Llorens F, Carulla P, Villa A, et al. PrP(C) regulates epidermal growth factor 73. Masters CL. Creutzfeldt-Jakob disease: its origins. Alzheimer Dis Assoc
receptor function and cell shape dynamics in Neuro2a cells. J Neurochem. Disord. 1989;3(12):4651.
2013;127(1):124138. 74. Ladogana A, Puopolo M, Croes EA, et al. Mortality from Creutzfeldt-
44. Lee YJ, Baskakov IV. The cellular form of the prion protein is involved in Jakob disease and related disorders in Europe, Australia, and Canada.
controlling cell cycle dynamics, self-renewal, and the fate of human embry- Neurology. 2005;64(9):15861591.
onic stem cell differentiation. J Neurochem. 2013;124(3):310322. 75. Heath CA, Cooper SA, Murray K, et al. Validation of diagnostic criteria for
45. Bremer J, Baumann F, Tiberi C, et al. Axonal prion protein is required for variant Creutzfeldt-Jakob disease. Ann Neurol. 2010;67(6):761770.
peripheral myelin maintenance. Nat Neurosci. 2010;13(3):310318. 76. Brown P, Brandel JP, Preece M, et al. Iatrogenic Creutzfeldt-Jakob disease:
46. Resenberger UK, Winklhofer KF, Tatzelt J. Neuroprotective and neurotoxic the waning of an era. Neurology. 2006;67(3):389393.
signaling by the prion protein. Top Curr Chem. 2011;305:101119. 77. Brown K, Mastrianni JA. The prion diseases. J Geriatr Psychiatry Neurol.
47. Singh N, Das D, Singh A, et al. Prion protein and metal interaction: physiolog- 2010;23(4):277298.
ical and pathological implications. Curr Issues Mol Biol. 2010;12(2):99107. 78. Heidenhain A. Klinische und anatomische Untersuchungen ber eine eigen-
48. Linden R, Martins VR, Prado MA, et al. Physiology of the prion protein. artige organische Erkrankung des Zentralnervensystems im Praesenium.
Physiol Rev. 2008;88(2):673728. Z Gesamte Neurol Psychiat. 1929;118:49114.
49. Behrens A, Aguzzi A. Small is not beautiful: antagonizing functions for 79. Puoti G, Bizzi A, Forloni G, et al. Sporadic human prion diseases: molecu-
the prion protein PrP(C) and its homologue Dpl. Trends Neurosci. lar insights and diagnosis. Lancet Neurol. 2012;11(7):618628.
2002;25(3):150154. 80. Cali I, Castellani R, Yuan J, et al. Classification of sporadic Creutzfeldt-
50. Wopfner F, Weidenhofer G, Schneider R, et al. Analysis of 27 mammalian Jakob disease revisited. Brain. 2006;129(pt 9):22662277.
and 9 avian PrPs reveals high conservation of flexible regions of the prion 81. Parchi P, Capellari S, Chin S, et al. A subtype of sporadic prion disease
protein. J Mol Biol. 1999;289(5):11631178. mimicking fatal familial insomnia. Neurology. 1999;52(9):17571763.

Scheld_Ch18.indd 282 2/21/14 5:39 PM


Chapter 18: Human Prion Diseases 283

82. Polymenidou M, Stoeck K, Glatzel M, et al. Coexistence of multiple 111. Green A, Sanchez-Juan P, Ladogana A, et al. CSF analysis in patients with
PrPSc types in individuals with Creutzfeldt-Jakob disease. Lancet Neurol. sporadic CJD and other transmissible spongiform encephalopathies. Eur J
2005;4(12):805814. Neurol. 2007;14(2):121124.
83. Brown P, Cathala F, Sadowsky D, et al. Creutzfeldt-Jakob disease in 112. Paterson RW, Takada LT, Geschwind MD. Diagnosis and treatment of
France: II. Clinical characteristics of 124 consecutive verified cases during rapidly progressive dementias. Neurol Clin Prac. 2012;2(3):187200.
the decade 19681977. Ann Neurol. 1979;6(5):430437. 113. Hsich G, Kenney K, Gibbs CJ, et al. The 14-3-3 brain protein in cerebro-
84. Brown P, Cathala F, Castaigne P, et al. Creutzfeldt-Jakob disease: clinical spinal fluid as a marker for transmissible spongiform encephalopathies.
analysis of a consecutive series of 230 neuropathologically verified cases. N Engl J Med. 1996;335(13):924930.
Ann Neurol. 1986;20(5):597602. 114. Chapman T, McKeel DW Jr, Morris JC. Misleading results with the
85. Rabinovici GD, Wang PN, Levin J, et al. First symptom in sporadic 14-3-3 assay for the diagnosis of Creutzfeldt-Jakob disease. Neurology.
Creutzfeldt-Jakob disease. Neurology. 2006;66(2):286287. 2000;55(9):13961397.
86. Will RG, Matthews WB. A retrospective study of Creutzfeldt-Jakob 115. Sanchez-Juan P, Green A, Ladogana A, et al. CSF tests in the differ-
disease in England and Wales 1970-79. I: Clinical features. J Neurol ential diagnosis of Creutzfeldt-Jakob disease. Neurology. 2006;67(4):
Neurosurg Psychiatry. 1984;47(2):134140. 637643.
87. Maltete D, Guyant-Marechal L, Mihout B, et al. Movement disorders 116. Chohan G, Llewelyn C, Mackenzie J, et al. Variant Creutzfeldt-Jakob
and Creutzfeldt-Jakob disease: a review. Parkinsonism Relat Disord. disease in a transfusion recipient: coincidence or cause? Transfusion.
2006;12(2):6571. 2010;50(5):10031006.
88. Espinosa PS, Bensalem-Owen MK, Fee DB. Sporadic Creutzfeldt-Jakob 117. Hamlin C, Puoti G, Berri S, et al. A comparison of tau and 14-3-3 protein
disease presenting as nonconvulsive status epilepticus case report and in the diagnosis of Creutzfeldt-Jakob disease. Neurology. 2012;79(6):
review of the literature. Clin Neurol Neurosurg. 2010;112(6):537540. 547552.
89. Kovacs T, Aranyi Z, Szirmai I, et al. Creutzfeldt-Jakob disease with 118. Deisenhammer F, Egg R, Giovannoni G, et al. EFNS guidelines on disease-
amyotrophy and demyelinating polyneuropathy. Arch Neurol. specific CSF investigations. Eur J Neurol. 2009;16(6):760770.
2002;59(11):18111184. 119. Satoh J, Kurohara K, Yukitake M, et al. The 14-3-3 protein detectable
90. Niewiadomska M, Kulczycki J, Wochnik-Dyjas D, et al. Impairment of in the cerebrospinal fluid of patients with prion-unrelated neurological
the peripheral nervous system in Creutzfeldt-Jakob disease. Arch Neurol. diseases is expressed constitutively in neurons and glial cells in culture.
2002;59(9):14301436. Eur Neurol. 1999;41(4):216225.
91. Paterson RW, Torres-Chae CC, Kuo AL, et al. Differential diagnosis of 120. Shimada T, Fournier AE, Yamagata K. Neuroprotective function of 14-
Jakob-Creutzfeldt disease. Arch Neurol. 2012;69(12):15781582. 3-3 proteins in neurodegeneration. Biomed Res Int. 2013;2013:564534.
92. Kovacs GG, Seguin J, Quadrio I, et al. Genetic Creutzfeldt-Jakob disease 121. Geschwind MD, Martindale J, Miller D, et al. Challenging the clinical
associated with the E200K mutation: characterization of a complex pro- utility of the 14-3-3 protein for the diagnosis of sporadic Creutzfeldt-
teinopathy. Acta Neuropathol. 2011;121(1):3957. Jakob disease. Arch Neurol. 2003;60(6):813816.
93. Esiri MM, Gordon WI, Collinge J, et al. Peripheral neuropathy in 122. Pruss H, Dalmau J, Harms L, et al. Retrospective analysis of NMDA
Creutzfeldt-Jakob disease. Neurology. 1997;48(3):784. receptor antibodies in encephalitis of unknown origin. Neurology.
94. Antoine JC, Laplanche JL, Mosnier JF, et al. Demyelinating peripheral 2010;75(19):17351739.
neuropathy with Creutzfeldt-Jakob disease and mutation at codon 200 of 123. Stoeck K, Sanchez-Juan P, Gawinecka J, et al. Cerebrospinal fluid bio-
the prion protein gene. Neurology. 1996;46(4):11231127. marker supported diagnosis of Creutzfeldt-Jakob disease and rapid de-
95. Chapman J, Brown P, Goldfarb LG, et al. Clinical heterogeneity and un- mentias: a longitudinal multicentre study over 10 years. Brain. 2012;
usual presentations of Creutzfeldt-Jakob disease in Jewish patients with 135(pt 10):30513061.
the PRNP codon 200 mutation. J Neurol Neurosurg Psychiatry. 1993; 124. Muayqil T, Gronseth G, Camicioli R. Evidence-based guideline: diag-
56(10):11091112. nostic accuracy of CSF 14-3-3 protein in sporadic Creutzfeldt-Jakob dis-
96. Johnson DY, Dunkelberger DL, Henry M, et al. Sporadic Jakob- ease: report of the guideline development subcommittee of the American
Creutzfeldt disease presenting as primary progressive aphasia. JAMA Academy of Neurology. Neurology. 2012;79(14):14991506.
Neurol. 2013;70(2):254257. 125. Coulthart MB, Jansen GH, Olsen E, et al. Diagnostic accuracy of cere-
97. Vitali P, Maccagnano E, Caverzasi E, et al. Diffusion-weighted MRI brospinal fluid protein markers for sporadic Creutzfeldt-Jakob disease in
hyperintensity patterns differentiate CJD from other rapid dementias. Canada: a 6-year prospective study. BMC Neurol. 2011;11:133.
Neurology. 2011;76(20):17111719. 126. van Harten AC, Kester MI, Visser PJ, et al. Tau and p-tau as CSF bio-
98. Shiga Y, Miyazawa K, Sato S, et al. Diffusion-weighted MRI abnormalities markers in dementia: a meta-analysis. Clin Chem Lab Med. 2011;49(3):
as an early diagnostic marker for Creutzfeldt-Jakob disease. Neurology. 353366.
2004;63:443449. 127. McGuire LI, Peden AH, Orru CD, et al. Real time quaking-induced con-
99. Tschampa HJ, Kallenberg K, Urbach H, et al. MRI in the diagnosis of version analysis of cerebrospinal fluid in sporadic Creutzfeldt-Jakob dis-
sporadic Creutzfeldt-Jakob disease: a study on inter-observer agreement. ease. Ann Neurol. 2012;72(2):278285.
Brain. 2005;128(pt 9):20262033. 128. Gambetti P, Dong Z, Yuan J, et al. A novel human disease with abnormal
100. Geschwind MD, Potter CA, Sattavat M, et al. Correlating DWI MRI with prion protein sensitive to protease. Ann Neurol. 2008;63(6):697708.
pathologic and other features of Jakob-Creutzfeldt disease. Alzheimer Dis 129. Zou WQ, Puoti G, Xiao X, et al. Variably protease-sensitive prionopathy:
Assoc Disord. 2009;23(1):8287. a new sporadic disease of the prion protein. Ann Neurol. 2010;68(2):
101. Geschwind MD, Josephs KA, Parisi JE, et al. A 54-year-old man with 162172.
slowness of movement and confusion. Neurology. 2007;69(19):1881 130. Budka H. Neuropathology of prion diseases. Br Med Bull. 2003;66:
1887. 121130.
102. Zerr I, Kallenberg K, Summers DM, et al. Updated clinical diagnostic 131. Budka H, Aguzzi A, Brown P, et al. Neuropathological diagnostic cri-
criteria for sporadic Creutzfeldt-Jakob disease. Brain. 2009;132(pt 10): teria for Creutzfeldt-Jakob disease (CJD) and other human spongiform
26592668. encephalopathies (prion diseases). Brain Pathol. 1995;5(4):459466.
103. Young GS, Geschwind MD, Fischbein NJ, et al. Diffusion-weighted and 132. Stoeck K, Zerr I. Cellular immune activation markers neopterin and
fluid-attenuated inversion recovery imaging in Creutzfeldt-Jakob disease: beta2-microglobulin are not elevated in the cerebrospinal fluid of pa-
high sensitivity and specificity for diagnosis. AJNR Am J Neuroradiol. tients with Creutzfeldt-Jakob disease. J Neuroimmunol. 2011;233(12):
2005;26(6):15511562. 228232.
104. Lin YR, Young GS, Chen NK, et al. Creutzfeldt-jakob disease involve- 133. Stoeck K, Bodemer M, Zerr I. Pro- and anti-inflammatory cytokines in
ment of rolandic cortex: a quantitative apparent diffusion coefficient the CSF of patients with Creutzfeldt-Jakob disease. J Neuroimmunol.
evaluation. AJNR Am J Neuroradiol. 2006;27(8):17551759. 2006;172(12):175181.
105. Letourneau-Guillon L, Wada R, Kucharczyk W. Imaging of prion diseases. 134. Van Everbroeck B, Dewulf E, Pals P, et al. The role of cytokines, astro-
J Magn Reson Imaging. 2012;35(5):9981012. cytes, microglia and apoptosis in Creutzfeldt-Jakob disease. Neurobiol
106. Geschwind MD, Kuryan C, Cattaruzza T, et al. Brain MRI in sporadic Aging. 2002;23(1):5964.
JakobCreutzfeldt disease is often misread. Neurology. 2010;78(suppl 135. Poser S, Mollenhauer B, Kraubeta A, et al. How to improve the clinical di-
1):P03.018. agnosis of Creutzfeldt-Jakob disease. Brain. 1999;122(pt 12):23452351.
107. Carswell C, Thompson A, Lukic A, et al. MRI findings are often missed 136. Geschwind MD, Shu H, Haman A, et al. Rapidly progressive dementia.
in the diagnosis of Creutzfeldt-Jakob disease. BMC Neurol. 2012; Ann Neurol. 2008;64(1):97108.
12(1):153. 137. Geschwind MD, Tan KM, Lennon VA, et al. Voltage-gated potassium
108. Steinhoff BJ, Zerr I, Glatting M, et al. Diagnostic value of periodic com- channel autoimmunity mimicking Creutzfeldt-Jakob disease. Arch
plexes in Creutzfeldt-Jakob disease. Ann Neurol. 2004;56(5):702708. Neurol. 2008;65(10):13411346.
109. Seipelt M, Zerr I, Nau R, et al. Hashimotos encephalitis as a differential 138. Van Everbroeck B, Dobbeleir I, De Waele M, et al. Differential diag-
diagnosis of Creutzfeldt-Jakob disease. J Neurol Neurosurg Psychiatry. nosis of 201 possible Creutzfeldt-Jakob disease patients. J Neurol.
1999;66(2):172176. 2004;251(3):298304.
110. Tschampa HJ, Neumann M, Zerr I, et al. Patients with Alzheimers dis- 139. Chitravas N, Jung RS, Kofskey DM, et al. Treatable neurological disor-
ease and dementia with Lewy bodies mistaken for Creutzfeldt-Jakob dis- ders misdiagnosed as Creutzfeldt-Jakob disease. Ann Neurol. 2011;70(3):
ease. J Neurol Neurosurg Psychiatry. 2001;71(1):3339. 437444.

Scheld_Ch18.indd 283 2/21/14 5:39 PM


284 Part II: Viral Infections and Related Disorders

140. Geschwind MD. Rapidly progressive dementia: prion diseases and other 171. Bueler H, Aguzzi A, Sailer A, et al. Mice devoid of PrP are resistant to
rapid dementias. Continuum (Minneap Minn). 2010;16(2 Dementia):3156. scrapie. Cell. 1993;73(7):13391347.
141. Takada L, Geschwind MD. Prion diseases. Sem Neurol. 2013;33: 172. Sailer A, Bueler H, Fischer M, et al. No propagation of prions in mice
338356. devoid of PrP. Cell. 1994;77(7):967968.
142. Bons N, Mestre-Frances N, Belli P, et al. Natural and experimental oral 173. Durig J, Giese A, Schulz-Schaeffer W, et al. Differential constitutive and
infection of nonhuman primates by bovine spongiform encephalopathy activation-dependent expression of prion protein in human peripheral
agents. Proc Natl Acad Sci U S A. 1999;96(7):40464051. blood leucocytes. Br J Haematol. 2000;108(3):488495.
143. Ridley RM, Baker HF. Oral transmission of BSE to primates. Lancet. 174. Bannach O, Birkmann E, Reinartz E, et al. Detection of prion pro-
1996;348(9035):1174. tein particles in blood plasma of scrapie infected sheep. PLoS One.
144. Will RG. Acquired prion disease: iatrogenic CJD, variant CJD, kuru. 2012;7(5):e36620.
Br Med Bull. 2003;66:255265. 175. Andreoletti O, Litaise C, Simmons H, et al. Highly efficient prion trans-
145. Peden A, McCardle L, Head MW, et al. Variant CJD infection in the mission by blood transfusion. PLoS Pathog. 2012;8(6):e1002782.
spleen of a neurologically asymptomatic UK adult patient with haemo- 176. Pflanzner T, Petsch B, Andre-Dohmen B, et al. Cellular prion protein
philia. Haemophilia. 2010;16(2):296304. participates in amyloid-beta transcytosis across the blood-brain barrier.
146. Kimberlin RH, Walker CA. Pathogenesis of scrapie in mice after intragas- J Cereb Blood Flow Metab. 2012;32(4):628632.
tric infection. Virus Res. 1989;12(3):213220. 177. Banks WA, Robinson SM, Diaz-Espinoza R, et al. Transport of prion pro-
147. Wells GA, Dawson M, Hawkins SA, et al. Infectivity in the ileum of cat- tein across the blood-brain barrier. Exp Neurol. 2009;218(1):162167.
tle challenged orally with bovine spongiform encephalopathy. Vet Rec. 178. Will RG, Ironside JW, Zeidler M, et al. A new variant of Creutzfeldt-
1994;135(2):4041. Jakob disease in the UK. Lancet. 1996;347(9006):921925.
148. Bons N, Mestre-Frances N, Guiraud I, et al. Prion immunoreactivity in 179. Will RG, Ironside JW, Hornlimann B, et al. Creutzfeldt-Jakob disease.
brain, tonsil, gastrointestinal epithelial cells, and blood and lymph vessels Lancet. 1996;347(8993):6566.
in lemurian zoo primates with spongiform encephalopathy. C R Acad Sci 180. Bruce ME, Will RG, Ironside JW, et al. Transmissions to mice in-
III. 1997;320(12):971979. dicate that new variant CJD is caused by the BSE agent. Nature.
149. Kujala P, Raymond CR, Romeijn M, et al. Prion uptake in the gut: 1997;389(6650):498501.
identification of the first uptake and replication sites. PLoS Pathog. 181. Scott MR, Will R, Ironside J, et al. Compelling transgenetic evidence for
2011;7(12):e1002449. transmission of bovine spongiform encephalopathy prions to humans.
150. Davies GA, Bryant AR, Reynolds JD, et al. Prion diseases and the gastro- Proc Natl Acad Sci U S A. 1999;96(26):1513715142.
intestinal tract. Can J Gastroenterol. 2006;20(1):1824. 182. Norrby E. Prions and protein-folding diseases. J Intern Med. 2011;270(1):
151. Miyazawa K, Kanaya T, Takakura I, et al. Transcytosis of murine-adapted 114.
bovine spongiform encephalopathy agents in an in vitro bovine M cell 183. UK National Creutzfeldt-Jakob Disease Surveillance Unit. Variant
model. J Virol. 2010;84(23):1228512291. Creutzfeldt-Jakob Disease Worldwide Current Data (November 2013).
152. Aguzzi A, Nuvolone M, Zhu C. The immunobiology of prion diseases. Edinburgh: Western General Hospital; 2013. http://www.cjd.ed.ac.uk
Nat Review Immunol. 2013;13(12):888902. /documents/worldfigs.pdf. Accessed January 30, 2014.
153. Huang FP, Farquhar CF, Mabbott NA, et al. Migrating intestinal dendritic 184. Heath CA, Cooper SA, Murray K, et al. Diagnosing variant Creutzfeldt-
cells transport PrP(Sc) from the gut. J Gen Virol. 2002;83(pt 1):267271. Jakob disease: a retrospective analysis of the first 150 cases in the UK.
154. Maignien T, Shakweh M, Calvo P, et al. Role of gut macrophages in J Neurol Neurosurg Psychiatry. 2011;82(6):646651.
mice orally contaminated with scrapie or BSE. Int J Pharm. 2005;298(2): 185. Kaski D, Mead S, Hyare H, et al. Variant CJD in an individual heterozy-
293304. gous for PRNP codon 129. Lancet. 2009;374(9707):2128.
155. Prinz M, Huber G, Macpherson AJ, et al. Oral prion infection requires 186. Peden AH, Head MW, Ritchie DL, et al. Preclinical vCJD after blood
normal numbers of Peyers patches but not of enteric lymphocytes. Am J transfusion in a PRNP codon 129 heterozygous patient. Lancet.
Pathol. 2003;162(4):11031111. 2004;364(9433):527529.
156. Kimberlin RH, Walker CA. Pathogenesis of mouse scrapie: patterns of 187. UK National Creutzfeldt-Jakob Disease Surveillance Unit. Diagnosis &
agent replication in different parts of the CNS following intraperitoneal Testing. Diagnostic criteria for human prion disease. (December 2012).
infection. J R Soc Med. 1982;75(8):618624. Edinburgh: Western General Hospital; 2012. http://www.cjd.ed.ac.uk
157. Beekes M, Baldauf E, Diringer H. Sequential appearance and accumula- /documents/criteria.pdf. Accessed January 30, 2014.
tion of pathognomonic markers in the central nervous system of hamsters 188. Will RG, Zeidler M, Stewart GE, et al. Diagnosis of new variant
orally infected with scrapie. J Gen Virol. 1996;77 (pt 8):19251934. Creutzfeldt-Jakob disease. Ann Neurol. 2000;47(5):575582.
158. Kimberlin RH, Hall SM, Walker CA. Pathogenesis of mouse scrapie. 189. Binelli S, Agazzi P, Giaccone G, et al. Periodic electroencephalogram com-
Evidence for direct neural spread of infection to the CNS after injection of plexes in a patient with variant Creutzfeldt-Jakob disease. Ann Neurol.
sciatic nerve. J Neurol Sci. 1983;61(3):315325. 2006;59(2):423427.
159. Kimberlin RH, Walker CA. Pathogenesis of scrapie (strain 263K) in ham- 190. Collie DA, Summers DM, Sellar RJ, et al. Diagnosing variant
sters infected intracerebrally, intraperitoneally or intraocularly. J Gen Creutzfeldt-Jakob disease with the pulvinar sign: MR imaging findings
Virol. 1986;67 (pt 2):255263. in 86 neuropathologically confirmed cases. AJNR Am J Neuroradiol.
160. Glatzel M, Heppner FL, Albers KM, et al. Sympathetic innervation of 2003;24(8):15601569.
lymphoreticular organs is rate limiting for prion neuroinvasion. Neuron. 191. Petzold GC, Westner I, Bohner G, et al. False-positive pulvinar sign on
2001;31(1):2534. MRI in sporadic Creutzfeldt-Jakob disease. Neurology. 2004;62(7):
161. Beekes M, McBride PA, Baldauf E. Cerebral targeting indicates vagal 12351236.
spread of infection in hamsters fed with scrapie. J Gen Virol. 1998;79 192. Haik S, Brandel JP, Oppenheim C, et al. Sporadic CJD clinically mimick-
(pt 3):601607. ing variant CJD with bilateral increased signal in the pulvinar. Neurology.
162. McBride PA, Schulz-Schaeffer WJ, Donaldson M, et al. Early spread 2002;58(1):148149.
of scrapie from the gastrointestinal tract to the central nervous system 193. Will R. Variant Creutzfeldt-Jakob disease. Folia Neuropathol. 2004;42
involves autonomic fibers of the splanchnic and vagus nerves. J Virol. (suppl A):7783.
2001;75(19):93209327. 194. Llewelyn CA, Hewitt PE, Knight RS, et al. Possible transmission of
163. Kaatz M, Fast C, Ziegler U, et al. Spread of classic BSE prions from variant Creutzfeldt-Jakob disease by blood transfusion. Lancet. 2004;
the gut via the peripheral nervous system to the brain. Am J Pathol. 363(9407):417421.
2012;181(2):515524. 195. Dorsey K, Zou S, Schonberger LB, et al. Lack of evidence of transfu-
164. Fraser H, Dickinson AG. Pathogenesis of scrapie in the mouse: the role of sion transmission of Creutzfeldt-Jakob disease in a US surveillance study.
the spleen. Nature. 1970;226(5244):462463. Transfusion. 2009;49(5):977984.
165. Fraser H, Dickinson AG. Studies of the lymphoreticular system in the 196. Ironside JW. Variant Creutzfeldt-Jakob disease: an update. Folia
pathogenesis of scrapie: the role of spleen and thymus. J Comp Pathol. Neuropathol. 2012;50(1):5056.
1978;88(4):563573. 197. Hilton DA, Ghani AC, Conyers L, et al. Prevalence of lymphore-
166. Klein MA, Frigg R, Flechsig E, et al. A crucial role for B cells in neuroin- ticular prion protein accumulation in UK tissue samples. J Pathol.
vasive scrapie. Nature. 1997;390(6661):687690. 2004;203(3):733739.
167. Kitamoto T, Muramoto T, Mohri S, et al. Abnormal isoform of prion 198. Gill ON, Spencer Y, Richard-Loendt A, et al. Prevalent abnormal prion
protein accumulates in follicular dendritic cells in mice with Creutzfeldt- protein in human appendixes after bovine spongiform encephalopathy
Jakob disease. J Virol. 1991;65(11):62926295. epizootic: large scale survey. BMJ. 2013;347:f5675.
168. Montrasio F, Frigg R, Glatzel M, et al. Impaired prion replication in 199. Duffy P, Wolf J, Collins G, et al. Letter: possible person-to-person trans-
spleens of mice lacking functional follicular dendritic cells. Science. mission of Creutzfeldt-Jakob disease. N Engl J Med. 1974;290(12):
2000;288(5469):12571259. 692693.
169. Hilton DA, Fathers E, Edwards P, et al. Prion immunoreactivity in ap- 200. Brown P, Brandel JP, Sato T, et al. Iatrogenic Creutzfeldt-Jakob disease,
pendix before clinical onset of variant Creutzfeldt-Jakob disease. Lancet. final assessment. Emerg Infect Dis. 2012;18(6):901907.
1998;352(9129):703704. 201. Heckmann JG, Lang CJ, Petruch F, et al. Transmission of Creutzfeldt-
170. Hill AF, Zeidler M, Ironside J, et al. Diagnosis of new variant Creutzfeldt- Jakob disease via a corneal transplant. J Neurol Neurosurg Psychiatry.
Jakob disease by tonsil biopsy. Lancet. 1997;349(9045):99100. 1997;63(3):388390.

Scheld_Ch18.indd 284 2/21/14 5:39 PM


Chapter 18: Human Prion Diseases 285

202. Bernoulli C, Siegfried J, Baumgartner G, et al. Danger of accidental 219. Plate A, Benninghoff J, Jansen GH, et al. Atypical parkinsonism due to a
person-to-person transmission of Creutzfeldt-Jakob disease by surgery. D202N Gerstmann-Straussler-Scheinker prion protein mutation: first in
Lancet. 1977;1(8009):478479. vivo diagnosed case. Mov Disord. 2013;28(2):241244.
203. Gibbs CJ Jr, Asher DM, Kobrine A, et al. Transmission of Creutzfeldt- 220. Kovacs GG, Horvath S, Strobel T, et al. Genetic Creutzfeldt-Jakob dis-
Jakob disease to a chimpanzee by electrodes contaminated during neuro- ease mimicking variant Creutzfeldt-Jakob disease. J Neurol Neurosurg
surgery. J Neurol Neurosurg Psychiatry. 1994;57(6):757758. Psychiatry. 2009;80(12):14101411.
204. Koch TK, Berg BO, De Armond SJ, et al. Creutzfeldt-Jakob disease in 221. Vitali P, Migliaccio R, Agosta F, et al. Neuroimaging in dementia. Semin
a young adult with idiopathic hypopituitarism. Possible relation to the Neurol. 2008;28(4):467483.
administration of cadaveric human growth hormone. N Engl J Med. 222. Yang TI, Jung DS, Ahn BY, et al. Familial Creutzfeldt-Jakob disease with
1985;313(12):731733. V180I mutation. J Korean Med Sci. 2010;25(7):10971100.
205. Centers for Disease Control and Prevention. Fatal degenerative neuro- 223. Laplanche JL, Hachimi KH, Durieux I, et al. Prominent psychiatric fea-
logic disease in patients who received pituitary-derived human growth tures and early onset in an inherited prion disease with a new insertional
hormone. MMWR Morb Mortal Wkly Rep. 1985;34:359360, 365366. mutation in the prion protein gene. Brain. 1999;122(pt 12):23752386.
206. Abrams JY, Schonberger LB, Belay ED, et al. Lower Risk of Creutzfeldt- 224. Alner K, Hyare H, Mead S, et al. Distinct neuropsychological profiles corre-
Jakob disease in pituitary growth hormone recipients initiating treatment spond to distribution of cortical thinning in inherited prion disease caused by
after 1977. J Clin Endocrinol Metab. 2011;96(10):E1666E1669. insertional mutation. J Neurol Neurosurg Psychiatry. 2012;83(1):109114.
207. Appleby BS, Lu M, Bizzi A, et al. Iatrogenic Creutzfeldt-Jakob disease 225. Giovagnoli AR, Di Fede G, Aresi A, et al. Atypical frontotemporal demen-
from commercial cadaveric human growth hormone. Emerg Infect Dis. tia as a new clinical phenotype of Gerstmann-Straussler-Scheinker disease
2013;19(4):682684. with the PrP-P102L mutation. Description of a previously unreported
208. Lewis AM, Yu M, DeArmond SJ, et al. Human growth hormone-related Italian family. Neurol Sci. 2008;29(6):405410.
iatrogenic Creutzfeldt-Jakob disease with abnormal imaging. Arch Neurol. 226. Webb TE, Poulter M, Beck J, et al. Phenotypic heterogeneity and genetic
2006;63(2):288290. modification of P102L inherited prion disease in an international series.
209. Brandel JP, Preece M, Brown P, et al. Distribution of codon 129 genotype Brain. 2008;131(pt 10):26322646.
in human growth hormone-treated CJD patients in France and the UK. 227. Gambetti P, Parchi P, Petersen RB, et al. Fatal familial insomnia and fa-
Lancet. 2003;362(9378):128130. milial Creutzfeldt-Jakob disease: clinical, pathological and molecular fea-
210. Hamaguchi T, Sakai K, Noguchi-Shinohara M, et al. Insight into the fre- tures. Brain Pathol. 1995;5(1):4351.
quent occurrence of dura mater graft-associated Creutzfeldt-Jakob disease 228. Cortelli P, Perani D, Montagna P, et al. Pre-symptomatic diagnosis in fatal
in Japan. J Neurol Neurosurg Psychiatry. 2013;84(10):11711175. familial insomnia: serial neurophysiological and 18FDG-PET studies.
211. Yamada M, Noguchi-Shinohara M, Hamaguchi T, et al. Dura mater graft- Brain. 2006;129(pt 3):668675.
associated Creutzfeldt-Jakob disease in Japan: clinicopathological and 229. University of California, San Francisco Medical Center. Policies and
molecular characterization of the two distinct subtypes. Neuropathology. Procedures for Patients with Suspected or Confirmed Human Prion
2009;29(5):609618. Disease (e.g., Creutzfeldt-Jakob disease [CJD]). San Francisco, CA:
212. World Health Organization. WHO Guidelines on Tissue Infectivity University of California, San Francisco; 2012. http://infectioncontrol
Distribution in Transmissible Spongiform Encephalopathies. Geneva: .ucsfmedicalcenter.org/ICMANUAL2007/Section4/Sec%204.2%20
World Health Organization; 2006. Human%20Prion%20Policy_5-31-2012.pdf.
213. Centers for Disease Control and Prevention Healthcare Infection Control 230. Geschwind MD, Kuo AL, Wong KS, et al. Quinacrine treatment trial for
Practices Advisory Committee . Guidelines for Environmental Infection sporadic Creutzfeldt-Jakob disease. Neurology. 2013;81(23):20152023.
Control in Health-Care Facilities. Atlanta: U.S. Department of Health and 231. Brown P, Cervenakova L, McShane L, et al. Creutzfeldt-Jakob disease in
Human Services, Centers for Disease Control and Prevention; 2003. a husband and wife. Neurology. 1998;50(3):684688.
214. Mead S. Prion disease genetics. Eur J Hum Genet. 2006;14(3):273281. 232. Prusiner SB. Cell biology. A unifying role for prions in neurodegenerative
215. Lloyd S, Mead S, Collinge J. Genetics of prion disease. Topics in current diseases. Science. 2012;336(6088):15111513.
chemistry 2011;305:122. 233. Puoti G, Rossi G, Giaccone G, et al. Polymorphism at codon 129 of PRNP
216. Collinge J. Prion diseases of humans and animals: their causes and mo- affects the phenotypic expression of Creutzfeldt-Jakob disease linked to
lecular basis. Annu Rev Neurosci. 2001;24:519550. E200K mutation. Ann Neurol. 2000;48(2):269270.
217. Kovacs GG, Puopolo M, Ladogana A, et al. Genetic prion disease: the 234. Meissner B, Kallenberg K, Sanchez-Juan P, et al. MRI lesion profiles in
EUROCJD experience. Hum Genet. 2005;118(2):166174. sporadic Creutzfeldt-Jakob disease. Neurology. 2009;72(23):19942001.
218. Mead S, Gandhi S, Beck J, et al. A novel prion disease associated with diar- 235. Hamaguchi T, Kitamoto T, Sato T, et al. Clinical diagnosis of MM2-type
rhea and autonomic neuropathy. N Engl J Med. 2013;369(20):19041914. sporadic Creutzfeldt-Jakob disease. Neurology. 2005;64(4):643648.

Scheld_Ch18.indd 285 2/21/14 5:39 PM


CHAPTER 19 HUMAN IMMUNODEFICIENCY
VIRUS
CHRISTINA M. MARRA

HIV can directly infect the cerebrospinal fluid (CSF) and the impairment is increasing (24). In addition, treatment of HIV
brain. Infection of the CSF is a notable feature of the acute itself may be associated with neurologic complications. The
retroviral syndrome, and CSF pleocytosis is common in HIV clinical findings and treatment of CNS opportunistic infec-
infection, particularly among individuals with high peripheral tions seen in HIV-infected individuals are covered in several
blood CD4 T-cell counts, detectable plasma HIV RNA, chapters of this book. In particular, a detailed discussion of
and in those not on antiretroviral therapy (1). Infection of cerebral toxoplasmosis is provided in Chapter 43, and neu-
the brain likely is the substrate for dementia in HIV infec- rosyphilis is covered in Chapter 38. This chapter addresses
tion. The immunodeficiency caused by HIV predisposes indi- two opportunistic infections not covered elsewhere: primary
viduals to secondary or opportunistic central nervous system CNS lymphoma, which is considered with infections because
(CNS) infections or neoplasms (Table 19.1). The incidence of its association with Epstein-Barr virus infection, and pro-
of dementia and most CNS opportunistic infections has de- gressive multifocal leukoencephalopathy (PML) caused by the
clined in the developed world with widespread use of com- JC virus. In addition, the most common CNS complications
bination antiretroviral therapy (CART). However, although associated with HIV itself are discussed, including newly rec-
the incidence may be decreasing, the prevalence of cognitive ognized syndromes.

TA B L E 1 9 . 1
CENTRAL NERVOUS SYSTEM INFECTIONS ASSOCIATED WITH HIV

Type of Infection Organisms Focal Lesion Encephalitis Meningitis

Protozoal Toxoplasma gondii X X (rare)


Trypanosoma cruzi X
Amoebae (rare) X X X
Viral Cytomegalovirus X X X
Herpes simplex type 1 X
Herpes simplex type 2 X X
Varicella zoster X X X
JC virus (causes PML) X X (rare) X (rare)
Epstein-Barr virus (associated X
with primary CNS lymphoma)
Bacterial Mycobacterium tuberculosis X X
Treponema pallidum X X
Mycobacterium avium complex X X
Listeria monocytogenes X X
Salmonella sp. X
Streptococcus pneumoniae X
Nocardia sp. X
Bartonella sp. (rare) X X
Mixed (pyogenic brain abscess) X
Fungal Cryptococcus neoformans X X
Candida sp. X X
Aspergillus sp. X X
Coccidioides immitis X
Histoplasma capsulatum X
Pneumocystis jirovecii (rare) X

286

Scheld_Ch19.indd 286 2/21/14 5:40 PM


Chapter 19: Human Immunodeficiency Virus 287

FOCAL BRAIN DISEASE Differential Diagnosis


Opportunistic infections, neoplasms, and cerebrovascular dis- As noted earlier, the most common causes of brain lesions as-
ease are the most common causes of focal findings referable sociated with contrast enhancement and mass effect are toxo-
to the brain in HIV-infected patients. In such individuals, the plasmosis, primary CNS lymphoma, and tuberculoma. Other
presence or absence of contrast enhancement and mass effect causes are listed in Table 19.1.
on cranial neuroimaging studies helps to narrow the differen-
tial diagnosis. Lesions associated with contrast enhancement
and mass effect are most commonly due to toxoplasmosis, pri- Clinical Symptoms and Findings
mary CNS lymphoma, or tuberculoma. Lesions with minimal
or no contrast enhancement and no mass effect are most com- The presenting clinical features of HIV-associated primary
monly due to PML. CNS lymphoma include confusion, lethargy, memory loss,
hemiparesis, speech and language disorders, seizures, and cra-
nial nerve palsies (7,8).
Primary Central Nervous
System Lymphoma
Laboratory and Imaging Studies
Etiology HIV-infected patients with primary CNS lymphoma typi-
Primary CNS lymphomas in HIV-infected patients are typically cally have peripheral blood CD4 T-cell counts less than 50/
of B-cell origin and are classified as diffuse large cell or immu- L (9). Brain computed tomography (CT) or magnetic reso-
noblastic (5). Epstein-Barr virus (EBV) is detectable in these nance imaging (MRI) show ring- or homogeneously enhanc-
tumors in virtually all HIV-infected patients (5). Prolonged im- ing lesion(s), often located periventricularly or in the frontal
mune suppression and EBV-induced B-cell stimulation likely lobes, although other locations may be seen (Fig. 19.1). These
contribute to tumor development (6). lesions may cross the midline in the corpus callosum and may

FIGURE 19.1 Paired contrast-enhanced T1-weighted (top) and fluid-attenuated inversion recovery
(FLAIR) (bottom) magnetic resonance images in HIV-associated primary CNS lymphoma showing a
single well-circumscribed lesion with contrast enhancement and surrounding edema. Biopsy showed dif-
fuse large B-cell lymphoma. (With permission from Claire Beiser, MD.)

Scheld_Ch19.indd 287 2/21/14 5:40 PM


288 Part II: Viral Infections and Related Disorders

be associated with patchy nodular ventricular enhancement


(10). Thallium-201 (201Tl) single-photon emission computed Progressive Multifocal
tomography (SPECT) may be helpful in distinguishing be- Leukoencephalopathy
tween CNS infections and primary CNS lymphoma in HIV-
infected individuals. Focal areas of increased 201Tl uptake are Etiology
seen in patients with lymphoma, whereas no brain uptake
is seen in patients with CNS infections such as Toxoplasma PML is caused by a polyoma virus called JC virus. This
encephalitis or tuberculoma (11,12). Delayed imaging and virus is usually acquired in childhood. It remains latent in
calculation of a retention index increase the specificity of kidney and perhaps in brain, reactivates in the setting of
the test (12). immunosuppression, and infects oligodendrocytes and less so
Although the diagnosis of primary CNS lymphoma can astrocytes. Death of oligodendrocytes leads to demyelination.
only be proven by histopathology, examination of CSF for
EBV DNA by polymerase chain reaction (PCR) may be sensi-
tive and specific for establishing the diagnosis (13) when it is
used in the appropriate clinical setting: an HIV-infected patient
Differential Diagnosis
with a focal brain lesion with mass effect and enhancement. The diagnosis of PML should be considered in patients with
The positive predictive value of the test is highest in patients dementia, particularly if there are also focal neurologic find-
with a low likelihood of CNS toxoplasmosis, such as those ings. Varicella-zoster encephalitis may cause demyelination
who are Toxoplasma seronegative or who have been taking and should also be considered (34). Cytomegalovirus (CMV)
trimethoprim-sulfamethoxazole for prophylaxis against toxo- encephalitis is sometimes accompanied by focal lesions that
plasmosis. When the test is used more broadly in clinical set- could mimic PML (35). Substance abuse, particularly a form
tings, the positive predictive value of detection of EBV DNA of heroin use called chasing the dragon, can cause clinical
in CSF is lower (14,15). and radiographic abnormalities similar to PML (36). A se-
vere form of HIV-associated leukoencephalopathy charac-
terized pathologically by extensive perivascular macrophage
infiltration and demyelination and high levels of brain and
Treatment and Prevention CSF HIV RNA has been described (37). Patchy or confluent
white matter high signal intensity is seen on brain MRI. The
HIV-associated primary CNS lymphomas are sensitive to
syndrome occurs in patients failing CART and is accompa-
radiation therapy. In a retrospective analysis conducted be-
nied by cognitive abnormalities. Leukoencephalopathy has
fore the availability of CART, median survival was about 3
been described in individuals whose HIV is successfully
to 4 months with radiation therapy and dexamethasone and
treated, and has been attributed both to CNS immune
untreated survival was 3 to 4 weeks (16). Death was usually
reconstitution (3840) and to CNS escape in individu-
due to opportunistic infection rather than lymphoma (7,8).
als with suppressed plasma HIV RNA but detectable CSF
Absence of opportunistic infection, younger age, higher
HIV RNA (41). The latter two diagnoses may be distin-
Karnofsky performance status, and delivery of higher biologi-
guished from PML because they may be associated with
cally effective doses of radiation were associated with longer
CSF pleocytosis.
survival (17).
More modern studies show that whole brain radiation
therapy (at least 30 Gy) (1820) and CART (18,20,21) are
associated with better survival in HIV-infected patients with Clinical Findings
primary CNS lymphoma, often with best survival seen in
those who receive both modalities. Prolonged survival has Patients with PML typically experience insidious onset of
also been reported in HIV-infected patients with primary progressive neurologic deficits, most commonly cognitive dys-
CNS lymphoma who only received CART (2224). Some function, limb weakness, gait disturbance, coordination dif-
have suggested that whole brain radiation be reserved for ficulties, and visual loss. Headache is a complaint in about
patients who do not respond to antiretrovirals to avoid the one fourth of patients. Neurologic examination shows focal
morbidity, particularly leukoencephalopathy, associated with deficits, particularly hemiparesis or visual field abnormali-
this treatment modality. Because of the association with EBV, ties (42,43). Less common manifestations of CNS JC infec-
some investigators have advocated combination therapy for tion include cerebellar granule cell neuronopathy (44) and JC
HIV-associated primary CNS lymphoma that targets this virus infection of cortical pyramidal neurons, causing an acute en-
with agents such as ganciclovir or foscarnet. Individual re- cephalopathy (45).
ports of success with these regimens can be difficult to eval-
uate because patients are often also treated with radiation,
CART, and immune modulators (2530). Overall, treatment
data for HIV-associated primary CNS lymphoma are mostly Laboratory and Imaging Studies
from retrospective case reports and series and are limited by
selection bias. The best treatment for HIV-associated primary HIV-infected patients with PML typically have peripheral
CNS lymphoma is not known because a randomized trial has blood CD4 T cells less than 200/L, although even before
not been conducted, and, given the low incidence of the disor- the advent of CART, about 10% of patients had CD4 counts
der, may never be feasible. above this threshold (43). In the current treatment era, indi-
Prior to the advent of CART, primary CNS lymphoma oc- viduals with PML and much higher or even near-normal pe-
curred in 1% to 4% of HIV-infected persons. Since CART has ripheral blood CD4 T cell concentrations have been reported
become widely available, the incidence of this disorder has (46,47).
declined (31,32) and survival has improved (18,33). Thus, Brain CT in patients with PML may sometimes be normal.
prevention lies in antiretroviral treatment to prevent pro- More often, it shows multiple, often confluent, white matter
longed immunosuppression. lesions that are most commonly located in the parietooccipital

Scheld_Ch19.indd 288 2/21/14 5:40 PM


Chapter 19: Human Immunodeficiency Virus 289

FIGURE 19.2 Paired T1-weighted (top) and fluid-attenuated inversion recovery (FLAIR) (bottom) mag-
netic resonance images in HIV-associated PML. Most notable is the large left frontal white matter lesion
that crosses the midline in the corpus callosum. This abnormality is seen on both the T1-weighted and
FLAIR images but is better seen on the FLAIR images. Another smaller lesion is evident in the right fron-
tal white matter. None of the lesions has associated mass effect.

regions. These lesions are low density and have little, if any,
mass effect (43). Enhancement may be seen in 10% of CT Treatment and Prevention
scans and is usually faint and peripheral (43). Brain MRI
shows more lesions than CT (48). In contrast to the white mat- Recent studies suggest that the current incidence of PML has
ter lesions seen in HIV-associated dementia, which are visible declined approximately three- to fourfold compared to before
only on T2-weighted images, PML lesions are low intensity 1996, with estimates among HIV-infected individuals ranging
on T1-weighted and high intensity on T2-weighted images from 0.6 to 1.3 per 1,000 person-years (51,52). Treatment
(Fig. 19.2); about 15% show faint contrast enhancement (43). with potent antiretrovirals has also resulted in significantly
Contrast enhancement is more likely to be seen in the setting improved survival in patients with PML (5153).
of immune reconstitution (see the following discussion). In ad- Several prognostic factors have been identified in HIV-
dition to the abnormalities seen on T1- and T2-weighted MRI, associated PML. Mass effect on MRI (54) and brainstem and
restricted diffusion may be seen; additional examples of MRI cerebellar involvement (55,56) are associated with poorer out-
abnormalities in PML are shown in Chapter 3. come. Higher peripheral blood CD4 T-cell count at diagnosis
Diagnostic criteria for PML have been recently published (48,51,5760), PML as the AIDS-defining illness (58), lower
(49). In patients with characteristic clinical and neuroimaging concentration and decline in CSF JC virus DNA concentration
findings, histologic examination or identification of JC virus during antiretroviral therapy (5964), and the presence of JC
DNA by PCR in CSF can confirm the diagnosis. However, virus-specific cytotoxic T cells in blood (6567) are associated
a negative PCR does not exclude the diagnosis. Individuals with improved survival.
who develop PML while receiving CART as well as those with All HIV-infected patients with PML should be treated with
higher peripheral blood CD4 T cell concentrations are more CART aimed at complete suppression of plasma HIV viremia.
likely to have a negative diagnostic PCR (50). Whether certain types of antiretroviral regimens are more

Scheld_Ch19.indd 289 2/21/14 5:40 PM


290 Part II: Viral Infections and Related Disorders

effective than others remains a matter of debate because few


prospective studies have been conducted. Two retrospective DIFFUSE CENTRAL NERVOUS
analyses suggest that a regimen that includes a protease inhibitor SYSTEM DISEASE
may be more effective than one that does not (68,69). Gasnault
and colleagues (60) conducted a multicenter, prospective, open-
label trial of an individualized regimen of five antiretrovirals, HIV-Associated Neurocognitive Disorders
including enfuvirtide for the first 6 months, in 28 HIV-infected
patients with PML. The 1-year survival was 75%, significantly Cognitive impairment due to HIV infection was first character-
higher than the historically reported survival of 45% at the ized by Navia et al. (85) in 1986 and termed AIDS dementia
time the trial was planned (63,7072). In this trial, all deaths complex or ADC to emphasize a triad of cognitive, motor,
occurred within the first 4 months of treatment (60). and behavioral abnormalities: (a) forgetfulness and loss of
As a consequence of an immune reconstitution inflamma- concentration; (b) apathy, social withdrawal, and irritability;
tory syndrome (IRIS), PML may develop (unmasking or simul- and (c) loss of balance and leg weakness. On neurologic ex-
taneous IRIS) or worsen (paradoxical or delayed IRIS) after amination, such patients typically demonstrated slowed ver-
beginning antiretroviral therapy. In an observational study bal and motor responses, reminiscent of neurologic findings in
in Spain of 61 patients with PML, 14 (23%) developed PML Parkinson disease. Not every patient with ADC had all three
IRIS, of which 8 were unmasking IRIS and 6 paradoxical IRIS components of the triad at onset of disease, and not all patients
(53). In a case series and retrospective review, PML IRIS devel- with mild disease progressed to more severe disease. As will be
oped 1 week to 26 months (median 7 months) after beginning discussed further in the following section, with the advent of
antiretroviral therapy, with a shorter latency, greater number CART, the incidence of HIV-associated dementia (HAD) has
of MRI-defined brain lesions, and poorer outcome in paradox- decreased, but the incidence and prevalence of less severe forms
ical compared to unmasking IRIS (73). A retrospective case se- of cognitive impairment have increased. In 2006, a working
ries showed that peripheral blood CD4 T cells 50/L at the group addressed the criteria for diagnosis of HIV-associated
time of initiating antiretroviral therapy significantly increases neurocognitive disorders (HAND) (86). These criteria are com-
the risk of PML IRIS and that the prognosis of patients with monly referred to as the Frascati criteria, because the work-
PML IRIS is no different than PML patients without IRIS (74). ing group met in Frascati, Italy. Three categories of impairment
Paradoxical IRIS is characterized by clinical worsening and were established: asymptomatic neurocognitive impairment
progression of previously defined MRI lesions or development (ANI), mild neurocognitive disorder (MND), and HAD. The
of new lesions, often, but not always, with evidence of contrast diagnosis of all three disorders is based on results of neuro-
enhancement. In the absence of new enhancement, paradoxi- psychologic (NP) tests covering at least the domains of verbal/
cal IRIS can be difficult to distinguish from progressive dis- language, attention/working memory, abstraction/executive,
ease, although the onset of clinical worsening in IRIS may be memory (learning, recall), speed of information processing,
more acute. Both fatal and benign courses have been described and sensoryperceptual or motor skills, in combination with
(73,7577) as well as individuals that do and do not respond an assessment of everyday functioning. In all categories, cogni-
to immunosuppression with steroids (73,78). A single case of tive or functional impairment cannot be explained by delirium,
rapid improvement in paradoxical PML IRIS after treatment opportunistic CNS disease, systemic illness, psychiatric illness,
with the CCR5 inhibitor, maraviroc, but not treatment with substance use, or medications with CNS effects (Table 19.2).
methylprednisolone, has been reported (79). The authors specu- Although these categories were intended for research purposes,
lated that blocking of CCR5 leukocyte recruitment to the CNS they have become increasingly applied to clinical settings.
might have been the underlying mechanism of improvement.
No specific therapy for HIV-associated (or nonassociated)
Epidemiology and Risks
PML has been identified. Cytosine arabinoside (Ara-C) given Estimates early in the HIV epidemic suggested that at least
intravenously (IV) or intrathecally did not confer a survival 60% of patients with AIDS developed overt or subclini-
benefit in a clinical trial conducted before the availability of cal HAD (85,87). The proportion decreased to about 7% in
CART (80). A metaanalysis of cidofovir showed no benefit the first years after antiretrovirals became available (88,89).
beyond that of CART (72). Mefloquine showed anti-JC activ- Several studies in the CART era show that the prevalence of
ity in an in vitro assay (81), but an open-label randomized HAD is low but the prevalence of milder impairment (MND
trial was stopped early because of lack of efficacy (64). Based and ANI) is surprisingly high. For example, in the multi-
on the observation that the cellular receptor for JC virus is the center U.S. CNS HIV Antiretroviral Therapy Effects Research
5-HT2A serotonin receptor (82), mirtazapine has been used for (CHARTER) study, among 1,316 participants without severe
PML treatment with anecdotal reports of success (83,84), but confounding disorders that could independently explain cog-
no clinical trial has been conducted. nitive impairment, 2% had dementia, 12% had MND, and
HIV-infected patients with PML who survive generally 33% had ANI (90). In a study of 100 Italian patients with
have persistent neurologic deficits, although some regain in- undetectable plasma HIV RNA, 2% met criteria for HAD,
dependence. In the trial by Gasnault and colleagues referenced 17% for MND, and 50% for ANI (91), and in a study of
earlier, the median modified Rankin scale at 12 months in the 400 French patients, 7% had HAD, 31% had MND, and
21 survivors was 3 (moderate disability). Eight patients had 21% had ANI (92). Although symptomatic HAND (MND
only slight disability and were independent (60). In contrast, and HAD) remains a risk factor for poorer survival (4), the
in the Swiss HIV Cohort Study, among 47 individuals who relevance of an ANI diagnosis has been questioned for sev-
survived more than 1 year, only 8 (17%) had clinical improve- eral reasons. These include concerns that (a) normative NP
ment (52). In a convenience sample of 23 HIV-infected pa- data are from nonconfounded normal individuals rather
tients with PML who survived at least 5 years, 9 remained than background-matched controls; (b) in a normally distrib-
neurologically stable, 10 had partial improvement, and 4 had uted population, 16% will score 1 SD below the norm on a
marked improvement; 8 had only slight disability and lived in- given test; and (c) no published study has demonstrated that
dependently (55). In a population-based study from Denmark, ANI confers a risk for subsequent dementia (93,94). In addi-
5 of 11 patients followed for 3 years (from an original group of tion, patients frequently transition from mild impairment to
47 patients) returned to their pre-PML level of function (51). normal cognition and vice versa despite stable CART regimens

Scheld_Ch19.indd 290 2/21/14 5:40 PM


Chapter 19: Human Immunodeficiency Virus 291

TA B L E 1 9 . 2
DIAGNOSTIC CRITERIA FOR HIV-ASSOCIATED NEUROCOGNITIVE DISORDERS

Disorder Neuropsychologic Testing Available Neuropsychologic Testing Not Available

Asymptomatic neurocognitive Impairment (1 SD below a Mental status exam (MSE) impairment


impairment (ANI) demographically appropriate normative (1 SD below a demographically
mean) in 2 cognitive domains appropriate normative mean) involving
2 cognitive domains
No reported or demonstrated functional No reported or demonstrated functional
decline decline
Mild neurocognitive disorder (MND) Mild NP impairment (1 SD below a Mild MSE impairment (1 SD below a
demographically appropriate normative demographically appropriate normative
mean), involving 2 cognitive domains mean), involving 2 cognitive domains
Reported or demonstrated mild functional Reported or demonstrated mild
decline functional decline
HIV-associated dementia (HAD) Moderate NP impairment (2 SD below a Moderate MSE impairment (2 SD
Note: Severity of NP impairment and demographically appropriate normative below a demographically appropriate
functional decline must both meet mean) on 2 cognitive domains. normative mean), involving 2
these standards in order to diagnose Alternatively, one domain could be more cognitive domains
the person as having HAD. If either severely impaired (2.5 SD below the
NP impairment or functional decline mean) and another less severely impaired
is mild, the condition should be (1 SD below the mean)
classified as MND.
Reported or demonstrated major functional Reported or demonstrated major
decline functional decline

Adapted from Antinori A, Arendt G, Becker JT, et al. Updated research nosology for HIV-associated neurocognitive disorders. Neurology.
2007;69(18):17891799.

(95,96). Whether this reflects diagnostic imprecision, practice tients with cognitive impairment than in those with normal
effects or normal variation remains undefined (97). cognition (112). Astrocytes may be infected but do not sup-
Low current, and in particular low nadir, CD4 is a risk fac- port viral replication (113). The prevailing view is that HIV
tor for prevalent neurocognitive impairment in the CART era causes brain injury and subsequent cognitive impairment via
(90,95,98,99). However, the association between HIV-related indirect mechanisms (114116). Proposed models suggest that
factors and incident cognitive impairment is less clear. Although HIV-infected mononuclear phagocytes release toxic viral gene
current but not nadir CD4 remained a significant predictor of products such as gp120 or tat. Alternatively, infected brain
incident dementia in the Concerted Action on Seroconversion macrophages or microglia may release cell-derived toxins such
to AIDS and Death in Europe (CASCADE) cohort (100), in the as quinolinic acid; cytokines, including tumor necrosis factor-
AIDS Clinical Trials Group Longitudinal Linked Randomized (TNF-); eicosanoids; platelet-activating factor; or nitric
Trials (ALLRT) study, neither current nor nadir CD4 or plasma oxide. This hypothesis is particularly compelling because the
HIV RNA predicted incident cognitive impairment (95). In a neuropathology of HAD is characterized by increased num-
population-based study of 1,320 HIV-infected patients in bers of activated macrophages, and the severity of dementia
Canada, incident symptomatic cognitive impairment was correlates better with the degree of macrophage staining in
more common with lower nadir CD4 and plasma HIV RNA brain than with the number of HIV-infected cells in brain
greater than 1 million copies/mL (101). In this study and in the (117). Toxic substances released by activated macrophages
CASCADE study, longer estimated duration of HIV infection may injure neurons directly, may injure astrocytes or oligoden-
conferred greater risk of incident impairment (100,101). drocytes and interfere with their supporting functions, or may
stimulate astrocytes or oligodendrocytes to release toxic prod-
Etiology ucts that may augment neurotoxicity. Nonproductive HIV in-
HIV can be recovered from CSF (102) or brain (103) early in fection of astrocytes may contribute to macrophage activation,
the course of infection, and acute meningitis or encephalitis a 180 augmenting neurotoxicity.
may be part of the acute retroviral syndrome (102,104,105). The relationship between brain HIV infection and clini-
Moreover, brain inflammation and injury is evident in primary cally evident cognitive impairment may be changing in the
HIV infection in some, but not all, patients (106108). Most current treatment era, particularly among patients with mild
experts agree that productive brain HIV infection is restricted impairment. A recent pathologic study of 589 individuals with
to perivascular macrophages and less so microglia (109). advanced disease showed no relationship between brain HIV
Infection of the CNS may be driven by activation of peripheral infection and HAND (118), and, as discussed in the follow-
monocytes as a consequence of microbial translocation from ing section, although detectable CSF HIV was associated with
the gut early in infection (110). Activated monocytes are bet- prevalent dementia and predicted it in the pre-CART era, these
ter able to support productive HIV infection (111), and HIV associations are less robust in the era of CART. Moreover, the re-
DNA concentration in activated monocytes is higher in pa- lationship between lower nadir CD4 and cognitive impairment

Scheld_Ch19.indd 291 2/21/14 5:40 PM


292 Part II: Viral Infections and Related Disorders

suggests that CNS injury may have occurred in the past, and
that although impairment may be persistent, it may not reflect
the effect of ongoing CNS infection, inflammation, or injury.
This legacy, however, might lessen cognitive reserve. As
HIV-infected patients live longer, aging (119), vascular disease
(120122), metabolic abnormalities (123), and CART toxicities
(124126) may increasingly contribute to cognitive impairment.

Differential Diagnosis
Before the advent of highly active antiretroviral therapy, HAD
was a disease of those with advanced immunosuppression,
and CMV encephalitis was an important alternative diagnosis
to consider. In the current treatment era, patients with HAD
may present with peripheral blood CD4 T cells more than
200 cells/L (2,3) and CMV encephalitis is rare. The differ-
ential diagnosis includes psychiatric disease, particularly de-
pression; adverse effects from prescription or illicit drugs; and
cerebral opportunistic infections including toxoplasmosis, tu-
berculosis, cryptococcal meningitis, and neurosyphilis. Rarely,
PML may present solely with cognitive changes, but focal neu-
rologic findings and neuroimaging almost always distinguish
patients with PML from those with HAD (127).

Clinical Symptoms and Findings


FIGURE 19.3 T2-weighted brain magnetic resonance images in HIV-
Patients with HAD generally present with subacute onset of
associated dementia show patchy and diffuse increased signal inten-
cognitive impairment, often with complaints of mental and sity in the white matter.
physical slowness. Neurologic examination typically is re-
markable for slowed verbal and motor responses. Ataxia and
hyperreflexia may also be seen. A staging system for HAD has
been used to characterize clinical severity (128,129). It is based the periventricular regions and in the centrum semiovale that
on functional disability and ranges from normal cognitive and are not seen on T1-weighted sequences (Fig. 19.3). However,
motor function (stage 0), to mild impairment (stage 1) with atrophy and focal white matter abnormalities may be seen in
preserved ability to perform all but the more demanding tasks HIV-infected individuals without cognitive changes (137).
of work or daily activities, to end-stage disease (stage 4) dis-
tinguished by rudimentary comprehension and responses. Not
all patients with mild disease progress to more severe stages. Treatment and Prevention
Early studies showed that even zidovudine monotherapy im-
Laboratory and Imaging Studies proved cognition and survival in patients with HAD (138,139).
Studies early in the CART era showed that decline in CSF HIV
The diagnosis of HAD is based on clinical findings and re- RNA and improvement in NP test performance could be seen
mains one of exclusion. There is no specific test that estab- in individuals treated with CART (140142), and CART is
lishes the diagnosis. However, CSF studies may be the most also associated with increased survival in patients with HAD
informative. Conventional CSF evaluation helps exclude other (143). Improvement in cognition after starting CART may lag
disorders. Data from the Multicenter AIDS Cohort Study sug- behind improvement in CD4 or decline in plasma HIV RNA;
gest that a CSF 2-microglobulin concentration more than in one study, improvement peaked at 24 to 36 weeks after
3.8 mg/dL in a CSF specimen with a normal white blood cell beginning CART (144). That CART may also play a role in
(WBC) count is specific, but not sensitive, for the diagnosis of preventing development of cognitive impairment is shown
HAD (130). In the pre-CART era, among patients with AIDS, by a recent study demonstrating that HIV-infected patients
CSF HIV RNA levels were elevated in those with cognitive who initiated CART early in the course of disease had a low
impairment (131); they correlated with severity of dementia prevalence of cognitive impairment that was comparable to
(132,133) and predicted incident impairment (134). In the matched HIV-uninfected individuals (145).
CART era, CSF HIV RNA does not distinguish between those In the last few years, much attention has been given to
with and without cognitive impairment (135,136). However, whether antiretroviral regimens with good CNS penetration
in the clinical setting, measurement of CSF HIV RNA can be are more effective in decreasing CSF HIV RNA concentra-
useful. A CSF concentration higher than plasma suggests, but tion and in improving cognitive function than regimens with
does not prove, that cognitive impairment is due to ongoing poorer penetration. Letendre and colleagues (146,147) have
CNS HIV infection and is one definition of CNS escape (see published two versions of the CNS Penetration Effectiveness
the following discussions). (CPE) rank, which estimates CNS drug penetration based on
Neuroimaging is useful in excluding other disorders but virologic and pharmacologic data. The most recent version
does not establish the diagnosis of HAD. Cranial CT may be uses four categories (14), with higher values assigned to drugs
normal or show atrophy or patchy white matter attenuation with better predicted penetration (147). The score for a given
(85). Cranial MRI is more sensitive than CT for demonstrating regimen is calculated as the sum of the scores of its component
white matter abnormalities and may show high T2 signal in drugs. Higher CPE regimens convey increased likelihood of

Scheld_Ch19.indd 292 2/21/14 5:40 PM


Chapter 19: Human Immunodeficiency Virus 293

suppressed CSF HIV RNA (146,147). However, the relation- HIV RNA, those with CNS escape had a longer duration of
ship between regimen CPE and improvement in cognition is CART and had experienced more treatment interruptions, rais-
less clear. Although several studies suggest that regimens with ing the question of whether CSF viral blips, similar to what
higher CPE improve cognitive function more than regimens has been described in plasma, may occur. A review of the lit-
with lower scores, this finding is not universal (148). A recent erature shows overlap between CNS IRIS and CNS escape. The
study suggested that the revised CPE rank is superior to the distinguishing feature should be detection of CSF HIV RNA,
original, which might explain some, but not all, discrepan- which is not seen in IRIS, but that is an integral part of the
cies between different studies (149). An observational study diagnosis of CNS escape. Reports describe successful treatment
showed that higher CPE conveyed an increased risk of cogni- of CNS IRIS with steroids, whereas reports of successful treat-
tive worsening in patients with advanced HIV (124), and a ment of CNS escape usually entail changes in CART based on
small randomized trial of three CART regimens showed that genotypic analysis of CSF viral sensitivity or by substitution or
although patients in the arm with the highest CPE had greater addition of agents thought to have good CNS drug penetration.
cognitive improvement, these patients also had greater evi- Of note, the pathogenesis of CNS IRIS and CNS escape may be
dence of brain inflammation by neuroimaging (150). similar. Lescure and colleagues (168) describe 14 HIV-infected
Shikuma and colleagues (151) have proposed a monocyte patients who were initially diagnosed with encephalitis of un-
efficacy (ME) score as a means of determining the effectiveness known etiology. Two patients had CNS IRIS, two had CNS
of an antiretroviral regimen in treating CNS HIV infection. escape, one had viral rebound after stopping CART, and six
The ME score is defined as the summed reciprocal ( 1,000) had a minor infection a few days before onset of their neuro-
of each agents median effective concentration (EC50) in a rest- logic illness. Brain MRI showed diffuse T2 and fluid-attenuated
ing macrophage acute infection model (151). The relationship inversion recovery (FLAIR) signal intensities in gray and white
between cognitive impairment and ME score was examined matter with punctate or linear gadolinium enhancement that
in a cohort of 139 individuals on stable CART for at least was best seen with T1-weighted spin-echo sequences combined
6 months; those who were taking atazanavir or lopinavir were with magnetization transfer. Histology showed intense perivas-
excluded because of lack of EC50 data. Patients whose regi- cular infiltration of polyclonal CD8 lymphocytes.
mens had higher ME scores had lower odds of symptomatic
cognitive impairment. Of note, the CPE rank and ME score
of individual regimens may not be congruent. For example, an SPINAL CORD DISEASE
acceptable regimen of abacavir, lamivudine, and nevirapine
yields a high CPE of 9 but a low ME of 73 (152). HIV-Associated Myelopathy
Several adjunctive agents have been investigated for the
treatment of HAD. Their rationale is based on data reviewed Several processes can affect the spinal cord in HIV-infected pa-
above suggesting that neuronal damage is a downstream event tients (Table 19.3). HIV-associated myelopathy (HAM) is likely
triggered by release of toxic viral gene products or inflamma- the most common cause of spinal cord dysfunction in this pop-
tory mediators from infected mononuclear cells. None have ulation in the U.S. Most of what we know about the disorder
proved to be beneficial, including nimodipine (153), CPI-1189 comes from studies conducted before the advent of CART, and
(an antioxidant that blocks TNF- in animal models) (154), the overall incidence of the disease is now low.
selegiline (155,156), memantine (157,158), minocycline
(159161), and rivastigmine (162). Etiology
The pathogenesis of HAM, pathologically termed vacuolar
myelopathy, is poorly understood. Detection of HIV in spinal cord
Encephalitis in the Setting of Combination is not associated with presence or severity of vacuolar changes
Antiretroviral Therapy: Central Nervous (169). Activated macrophages can be identified in the posterior
System Immune Reconstitution Inflammatory and lateral columns of spinal cords from HIV-infected individu-
als with and without vacuolar myelopathy (170), and degree of
Syndrome, Central Nervous System Escape, myelin damage is proportional to the number of infiltrating mac-
and CD8 T-Cell Encephalitis rophages (171). Thus, the pathogenesis of HAM may be simi-
lar to that proposed for HAD. Because of pathologic similarities
Just as there are reports of IRIS in patients with CNS opportu- between HAM and myelopathy associated with vitamin B12 or
nistic infections treated with CART, CNS IRIS due to HIV it- cobalamin deficiency, nutritional or vitamin deficiencies have
self has been reported after CART initiation (3840). Typically, also been suggested as a cause of HAM. In autopsy-based series,
patients develop diffuse or focal neurologic symptoms and vitamin B12 levels in patients with vacuolar myelopathy are not
signs usually accompanied by CSF pleocytosis in the absence of low (170,172). However, cobalamin is a cofactor for conversion
an identifiable infectious agent. Such patients need to be distin- of homocysteine to methionine, which is subsequently converted
guished from those with CNS escape in the setting of apparently to S-adenosylmethionine (SAM). SAM is needed for myelin for-
effective CART. These patients develop diffuse or focal neuro- mation and repair. In a small study, CSF SAM levels were sig-
logic symptoms while on CART, with detectable CSF HIV RNA nificantly lower in patients with HAM compared to HIV-infected
in the setting of suppressed peripheral viremia or with CSF HIV patients without myelopathy and normal controls (173). These
RNA concentrations higher than simultaneous plasma levels results support the hypothesis that HAM may be due to abnor-
(163165). Canestri and colleagues (166) described CNS escape malities of a B12-dependent metabolic pathway.
in 11 patients defined as CSF HIV RNA more than 200 copies/
mL with corresponding plasma HIV RNA less than 50 copies/
mL or CSF HIV RNA more than 1 log higher than plasma. Differential Diagnosis
Clinical syndromes included meningitis, encephalitis, and my-
elitis. Based on genotypic analysis, CSF virus was not sensitive Differential diagnosis of myelopathy in HIV-infected patients
to the CART regimen in five patients (166). Asymptomatic low- includes extrinsic cord compression from tumor such as lym-
level CNS escape with CSF HIV RNA concentrations ranging phoma, infection, such as bacterial or mycobacterial osteomy-
from 52 to 860 copies/mL has been described in a small series elitis, or hemorrhage (174). These entities usually have a rapid
of patients (167). Compared to patients with undetectable CSF course and associated back pain and sensory level, which

Scheld_Ch19.indd 293 2/21/14 5:40 PM


294 Part II: Viral Infections and Related Disorders

TA B L E 1 9 . 3 are uncharacteristic of HAM (see the following discussion).


Intrinsic cord abnormalities due to tumor or abscess, particu-
SPINAL CORD DISEASES ASSOCIATED WITH HIV larly lymphoma, toxoplasmosis, or tuberculosis, should also
be excluded. Human T-lymphotropic virus (HTLV) type 1 or
Extrinsic lesions type 2 infection may be indistinguishable from HAM. Myelitis
Tumors due to syphilis or herpes viruses (varicella-zoster virus [VZV],
Metastatic lymphoma herpes simplex virus [HSV], or CMV) and nutritional disor-
ders should also be considered (Table 19.3).
Other metastases
Infections due to conventional bacteria, Mycobacterium
tuberculosis, or fungi
Clinical Findings
Vertebral osteomyelitis
Epidural or subdural abscess Patients with HAM complain of slowly progressive lower
Epidural or subdural hemorrhage extremity weakness and stiffness, trouble walking, and urinary
frequency and incontinence. Men may note erectile dysfunction.
Intrinsic lesions HAM typically coexists with distal sensory peripheral neuropa-
Tumors thy, with concomitant complaints of lower extremity numbness,
Lymphoma pain, or dysesthesia (175). Neurologic examination shows spas-
Glioma tic paraparesis or paraplegia; gait dysfunction; sensory ataxia;
hyperreflexia, often with absent ankle reflexes in the setting of
Infections concomitant neuropathy; impaired vibration and position senses;
Toxoplasmosis extensor-plantar responses; and sphincter dysfunction (172,175).
M. tuberculosis
Cryptococcosis
Myelopathy Laboratory and Imaging Studies
HTLV types 1 and 2 HAM is a diagnosis of exclusion. MRI of the spinal cord should
Syphilis be undertaken to exclude extrinsic or intrinsic cord lesions. In
HAM, MRI is usually normal. When abnormalities are seen,
HIV-associated myelopathy
they typically involve the thoracic spinal cord with or without
Transverse myelopathy involvement of the cervical cord and include atrophy or increased
Multiple sclerosis intramedullary signal intensity (176,177). Abnormal tibial so-
Stroke matosensory-evoked potentials with prolonged conduction time
are considered by some to be diagnostic (173). Analysis of CSF
Myelitis
is indicated to exclude alternative diagnoses; in HAM, the CSF is
HIV typically acellular with mild to moderate elevation in protein con-
Herpesviruses centration (175). One study showed that CSF HIV-1 RNA con-
Human cytomegalovirus centrations were not increased in patients with HAM; however,
these individuals were all on stable antiretroviral therapy (178).
Varicella-zoster virus
Herpes simplex virus types 1 and 2
Metabolic Treatment and Prevention
Vitamin B12 deficiency
Copper deficiency There is no proven treatment for HAM. Case reports describe
improvement after beginning CART (179182), and this is cur-
Folate deficiency rently the therapy of choice. A controlled trial of L-methionine
Portosystemic shunt (183) and an open-label study of IV immunoglobulin for HAM
showed no benefit (184). The association between advanced
HIV disease and HAM suggests that earlier treatment for HIV
is the best preventative strategy.

References
1. Marra CM, Maxwell CL, Collier AC, et al. Interpreting cerebrospinal fluid 6. Ambinder RF. Epstein-Barr virus associated lymphoproliferations in the
pleocytosis in HIV in the era of potent antiretroviral therapy. BMC Infect AIDS setting. Eur J Cancer. 2001;37(10):12091216.
Dis. 2007;7:37. 7. Lowenthal DA, Straus DJ, Campbell SW, et al. AIDS-related lymphoid neo-
2. Dore GJ, Correll PK, Li Y, et al. Changes to AIDS dementia complex plasia. The Memorial Hospital experience. Cancer. 1988;61(11):23252337.
in the era of highly active antiretroviral therapy. AIDS. 1999;13(10): 8. Remick SC, Diamond C, Migliozzi JA, et al. Primary central nervous
12491253. system lymphoma in patients with and without the acquired immune de-
3. Sacktor N, Lyles RH, Skolasky R, et al. HIV-associated neurologic dis- ficiency syndrome. A retrospective analysis and review of the literature.
ease incidence changes: Multicenter AIDS Cohort Study, 19901998. Medicine. 1990;69(6):345360.
Neurology. 2001;56(2):257260. 9. Pluda JM, Venzon DJ, Tosato G, et al. Parameters affecting the develop-
4. Vivithanaporn P, Heo G, Gamble J, et al. Neurologic disease burden in ment of non-Hodgkins lymphoma in patients with severe human immu-
treated HIV/AIDS predicts survival: a population-based study. Neurology. nodeficiency virus infection receiving antiretroviral therapy. J Clin Oncol.
2010;75(13):11501158. 1993;11(6):10991107.
5. So YT, Beckstead JH, Davis RL. Primary central nervous system lymphoma 10. Goldstein JD, Zeifer B, Chao C, et al. CT appearance of primary CNS lym-
in acquired immune deficiency syndrome: a clinical and pathological study. phoma in patients with acquired immunodeficiency syndrome. J Comput
Ann Neurol. 1986;20(5):566572. Assist Tomogr. 1991;15(1):3944.

Scheld_Ch19.indd 294 2/21/14 5:40 PM


Chapter 19: Human Immunodeficiency Virus 295

11. Ruiz A, Ganz WI, Post MJ, et al. Use of thallium-201 brain SPECT to 37. Langford TD, Letendre SL, Marcotte TD, et al. Severe, demyelinating
differentiate cerebral lymphoma from toxoplasma encephalitis in AIDS pa- leukoencephalopathy in AIDS patients on antiretroviral therapy. AIDS.
tients. AJNR Am J Neuroradiol. 1994;15(10):18851894. 2002;16(7):10191029.
12. Lorberboym M, Wallach F, Estok L, et al. Thallium-201 retention in focal 38. Rackstraw S, Meadway J, Bingham J, et al. An emerging severe leukoen-
intracranial lesions for differential diagnosis of primary lymphoma and non- cephalopathy: is it due to HIV disease or highly active antiretroviral ther-
malignant lesions in AIDS patients. J Nucl Med. 1998;39(8):13661369. apy? Int J STD AIDS. 2006;17(3):205207.
13. Antinori A, Ammassari A, De Luca A, et al. Diagnosis of AIDS-related 39. Costello DJ, Gonzalez RG, Frosch MP. Case records of the Massachusetts
focal brain lesions: a decision-making analysis based on clinical and neuro- General Hospital. Case 18-2011. A 35-year-old HIV-positive woman with
radiologic characteristics combined with polymerase chain reaction assays headache and altered mental status. N Engl J Med. 2011;364(24):23432352.
in CSF. Neurology. 1997;48(3):687694. 40. Zaffiri L, Verma R, Struzzieri K, et al. Immune reconstitution inflammatory
14. Ivers LC, Kim AY, Sax PE. Predictive value of polymerase chain reaction syndrome involving the central nervous system in a patient with HIV infection:
of cerebrospinal fluid for detection of Epstein-Barr virus to establish the a case report and review of literature. New Microbiol. 2013;36(1):8992.
diagnosis of HIV-related primary central nervous system lymphoma. Clin 41. Venkataramana A, Pardo CA, McArthur JC, et al. Immune reconstitution
Infect Dis. 2004;38(11):16291632. inflammatory syndrome in the CNS of HIV-infected patients. Neurology.
15. Corcoran C, Rebe K, van der Plas H, et al. The predictive value of cerebrospi- 2006;67(3):383388.
nal fluid Epstein-Barr viral load as a marker of primary central nervous sys- 42. Berger JR, Kaszovitz B, Post MJ, et al. Progressive multifocal leukoen-
tem lymphoma in HIV-infected persons. J Clin Virol. 2008;42(4):433436. cephalopathy associated with human immunodeficiency virus infection: a
16. Baumgartner JE, Rachlin JR, Beckstead JH, et al. Primary central nervous sys- review of the literature with a report of sixteen cases. Ann Intern Med.
tem lymphomas: natural history and response to radiation therapy in 55 patients 1987;107(1):7887.
with acquired immunodeficiency syndrome. J Neurosurg. 1990;73(2):206211. 43. Berger JR, Pall L, Lanska D, et al. Progressive multifocal leukoencepha-
17. Corn BW, Donahue BR, Rosenstock JG, et al. Performance status and age lopathy in patients with HIV infection. J Neurovirol. 1998;4(1):5968.
as independent predictors of survival among AIDS patients with primary 44. Koralnik IJ, Wuthrich C, Dang X, et al. JC virus granule cell neuronopathy:
CNS lymphoma: a multivariate analysis of a multi-institutional experience. a novel clinical syndrome distinct from progressive multifocal leukoenceph-
Cancer J Sci Am. 1997;3(1):5256. alopathy. Ann Neurol. 2005;57(4):576580.
18. Newell ME, Hoy JF, Cooper SG, et al. Human immunodeficiency virus- 45. Wuthrich C, Dang X, Westmoreland S, et al. Fulminant JC virus encepha-
related primary central nervous system lymphoma: factors influencing sur- lopathy with productive infection of cortical pyramidal neurons. Ann
vival in 111 patients. Cancer. 2004;100(12):26272636. Neurol. 2009;65(6):742748.
19. Nagai H, Odawara T, Ajisawa A, et al. Whole brain radiation alone pro- 46. Mascarello M, Lanzafame M, Lattuada E, et al. Progressive multifocal
duces favourable outcomes for AIDS-related primary central nervous sys- leukoencephalopathy in an HIV patient receiving successful long-term
tem lymphoma in the HAART era. Eur J Haematol. 2010;84(6):499505. HAART. J Neurovirol. 2011;17(2):196199.
20. Skiest DJ, Crosby C. Survival is prolonged by highly active antiretroviral 47. Manfredi R, Piergentili B, Marinacci G, et al. Atypical progressive mul-
therapy in AIDS patients with primary central nervous system lymphoma. tifocal leukoencephalopathy in HIV with a high CD4 count: the use of
AIDS. 2003;17(12):17871793. magnetic resonance imaging plus spectrometry studies. Int J STD AIDS.
21. Hoffmann C, Tabrizian S, Wolf E, et al. Survival of AIDS patients with 2012;23(3):e35e38.
primary central nervous system lymphoma is dramatically improved by 48. Berenguer J, Miralles P, Arrizabalaga J, et al. Clinical course and prognostic
HAART-induced immune recovery. AIDS. 2001;15(16):21192127. factors of progressive multifocal leukoencephalopathy in patients treated with
22. McGowan JP, Shah S. Long-term remission of AIDS-related primary cen- highly active antiretroviral therapy. Clin Infect Dis. 2003;36(8):10471052.
tral nervous system lymphoma associated with highly active antiretroviral 49. Berger JR, Aksamit AJ, Clifford DB, et al. PML diagnostic criteria: consen-
therapy. AIDS. 1998;12(8):952954. sus statement from the AAN Neuroinfectious Disease Section. Neurology.
23. Aboulafia DM, Puswella AL. Highly active antiretroviral therapy as the 2013;80(15):14301438.
sole treatment for AIDS-related primary central nervous system lymphoma: 50. Marzocchetti A, Di Giambenedetto S, Cingolani A, et al. Reduced rate of
a case report with implications for treatment. AIDS Patient Care STDS. diagnostic positive detection of JC virus DNA in cerebrospinal fluid in cases
2007;21(12):900907. of suspected progressive multifocal leukoencephalopathy in the era of po-
24. Travi G, Ferreri AJ, Cinque P, et al. Long-term remission of HIV-associated tent antiretroviral therapy. J Clin Microbiol. 2005;43(8):41754177.
primary CNS lymphoma achieved with highly active antiretroviral therapy 51. Engsig FN, Hansen AB, Omland LH, et al. Incidence, clinical presenta-
alone. J Clin Oncol. 2012;30(10):e119e121. tion, and outcome of progressive multifocal leukoencephalopathy in
25. Raez L, Cabral L, Cai JP, et al. Treatment of AIDS-related primary central HIV-infected patients during the highly active antiretroviral therapy era: a
nervous system lymphoma with zidovudine, ganciclovir, and interleukin 2. nationwide cohort study. J Infect Dis. 2009;199(1):7783.
AIDS Res Hum Retroviruses. 1999;15(8):713719. 52. Khanna N, Elzi L, Mueller NJ, et al. Incidence and outcome of progressive
26. Slobod KS, Taylor GH, Sandlund JT, et al. Epstein-Barr virus-targeted multifocal leukoencephalopathy over 20 years of the Swiss HIV Cohort
therapy for AIDS-related primary lymphoma of the central nervous system. Study. Clin Infect Dis. 2009;48(10):14591466.
Lancet. 2000;356(9240):14931494. 53. Falco V, Olmo M, del Saz SV, et al. Influence of HAART on the clinical
27. Aboulafia DM. Interleukin-2, ganciclovir, and high-dose zidovudine for the course of HIV-1-infected patients with progressive multifocal leukoenceph-
treatment of AIDS-associated primary central nervous system lymphoma. alopathy: results of an observational multicenter study. J Acquir Immune
Clin Infect Dis. 2002;34(12):16601662. Defic Syndr. 2008;49(1):2631.
28. Bossolasco S, Falk KI, Ponzoni M, et al. Ganciclovir is associated with 54. Post MJ, Yiannoutsos C, Simpson D, et al. Progressive multifocal leukoen-
low or undetectable Epstein-Barr virus DNA load in cerebrospinal fluid of cephalopathy in AIDS: are there any MR findings useful to patient manage-
patients with HIV-related primary central nervous system lymphoma. Clin ment and predictive of patient survival? AIDS Clinical Trials Group, 243
Infect Dis. 2006;42(4):e21e25. Team. AJNR Am J Neuroradiol. 1999;20(10):18961906.
29. Aboulafia DM, Ratner L, Miles SA, et al. Antiviral and immunomodu- 55. Lima MA, Bernal-Cano F, Clifford DB, et al. Clinical outcome of long-
latory treatment for AIDS-related primary central nervous system lym- term survivors of progressive multifocal leukoencephalopathy. J Neurol
phoma: AIDS Malignancies Consortium pilot study 019. Clin Lymphoma Neurosurg Psychiatry. 2010;81(11):12881291.
Myeloma. 2006;6(5):399402. 56. Piza F, Fink MC, Nogueira GS, et al. JC virus-associated central nervous system
30. Marretta L, Stocker H, Drauz D, et al. Treatment of HIV-related primary diseases in HIV-infected patients in Brazil: clinical presentations, associated
central nervous system lymphoma with AZT high dose, HAART, interleu- factors with mortality and outcome. Braz J Infect Dis. 2012;16(2):153156.
kin-2 and foscarnet in three patients. Eur J Med Res. 2011;16(5):197205. 57. Marra CM, Rajicic N, Barker DE, et al. A pilot study of cidofovir for progres-
31. Kirk O, Pedersen C, Cozzi-Lepri A, et al. Non-Hodgkin lymphoma in HIV- sive multifocal leukoencephalopathy in AIDS. AIDS. 2002;16(13):17911797.
infected patients in the era of highly active antiretroviral therapy. Blood. 58. Berger JR, Levy RM, Flomenhoft D, et al. Predictive factors for prolonged
2001;98(12):34063412. survival in acquired immunodeficiency syndrome-associated progressive
32. Bower M, Powles T, Nelson M, et al. Highly active antiretroviral therapy multifocal leukoencephalopathy. Ann Neurol. 1998;44(3):341349.
and human immunodeficiency virus-associated primary cerebral lymphoma. 59. Bossolasco S, Calori G, Moretti F, et al. Prognostic significance of JC virus
J Natl Cancer Inst. 2006;98(15):10881091. DNA levels in cerebrospinal fluid of patients with HIV-associated progres-
33. Kreisl TN, Panageas KS, Elkin EB, et al. Treatment patterns and prognosis sive multifocal leukoencephalopathy. Clin Infect Dis. 2005;40(5):738744.
in patients with human immunodeficiency virus and primary central system 60. Gasnault J, Costagliola D, Hendel-Chavez H, et al. Improved survival of HIV-
lymphoma. Leuk Lymphoma. 2008;49(9):17101716. 1-infected patients with progressive multifocal leukoencephalopathy receiving
34. Levy RM, Bredesen DE, Rosenblum ML. Neurological manifestations of early 5-drug combination antiretroviral therapy. PloS One. 2011;6(6):e20967.
the acquired immunodeficiency syndrome (AIDS): experience at UCSF and 61. Yiannoutsos CT, Major EO, Curfman B, et al. Relation of JC virus DNA in
review of the literature. J Neurosurg. 1985;62(4):475495. the cerebrospinal fluid to survival in acquired immunodeficiency syndrome
35. Holland NR, Power C, Mathews VP, et al. Cytomegalovirus encephalitis in patients with biopsy-proven progressive multifocal leukoencephalopathy.
acquired immunodeficiency syndrome (AIDS). Neurology. 1994;44(3, pt 1): Ann Neurol. 1999;45(6):816821.
507514. 62. Garcia De Viedma D, Diaz Infantes M, Miralles P, et al. JC virus load
36. Kriegstein AR, Shungu DC, Millar WS, et al. Leukoencephalopathy and in progressive multifocal leukoencephalopathy: analysis of the correlation
raised brain lactate from heroin vapor inhalation (chasing the dragon). between the viral burden in cerebrospinal fluid, patient survival, and the
Neurology. 1999;53(8):17651773. volume of neurological lesions. Clin Infect Dis. 2002;34(12):15681575.

Scheld_Ch19.indd 295 2/21/14 5:40 PM


296 Part II: Viral Infections and Related Disorders

63. De Luca A, Giancola ML, Ammassari A, et al. The effect of potent antiret- 90. Heaton RK, Clifford DB, Franklin DR Jr, et al. HIV-associated neuro-
roviral therapy and JC virus load in cerebrospinal fluid on clinical outcome cognitive disorders persist in the era of potent antiretroviral therapy:
of patients with AIDS-associated progressive multifocal leukoencephalopa- CHARTER Study. Neurology. 2010;75(23):20872096.
thy. J Infect Dis. 2000;182(4):10771083. 91. Simioni S, Cavassini M, Annoni JM, et al. Cognitive dysfunction in
64. Clifford DB, Nath A, Cinque P, et al. A study of mefloquine treatment HIV patients despite long-standing suppression of viremia. AIDS.
for progressive multifocal leukoencephalopathy: results and exploration of 2010;24(9):12431250.
predictors of PML outcomes. J Neurovirol. 2013;19(4):351318. 92. Bonnet F, Amieva H, Marquant F, et al. Cognitive disorders in HIV-
65. Du Pasquier RA, Kuroda MJ, Zheng Y, et al. A prospective study demon- infected patients: are they HIV-related? AIDS. 2013;27(3):391400.
strates an association between JC virus-specific cytotoxic T lymphocytes 93. Gisslen M, Price RW, Nilsson S. The definition of HIV-associated neu-
and the early control of progressive multifocal leukoencephalopathy. Brain. rocognitive disorders: are we overestimating the real prevalence? BMC
2004;127(pt 9):19701978. Infect Dis. 2011;11:356.
66. Marzocchetti A, Tompkins T, Clifford DB, et al. Determinants of sur- 94. Torti C, Foca E, Cesana BM, et al. Asymptomatic neurocognitive disor-
vival in progressive multifocal leukoencephalopathy. Neurology. 2009; ders in patients infected by HIV: fact or fiction? BMC Med. 2011;9:138.
73(19):15511558. 95. Robertson KR, Smurzynski M, Parsons TD, et al. The prevalence and
67. Khanna N, Wolbers M, Mueller NJ, et al. JC virus-specific immune re- incidence of neurocognitive impairment in the HAART era. AIDS.
sponses in human immunodeficiency virus type 1 patients with progressive 2007;21(14):19151921.
multifocal leukoencephalopathy. J Virol. 2009;83(9):44044411. 96. McArthur JC. HIV dementia: an evolving disease. J Neuroimmunol.
68. Dworkin MS, Wan PC, Hanson DL, et al. Progressive multifocal leukoen- 2004;157(12):310.
cephalopathy: improved survival of human immunodeficiency virus-infected 97. Woods SP, Moore DJ, Weber E, et al. Cognitive neuropsychology of HIV-
patients in the protease inhibitor era. J Infect Dis. 1999;180(3):621625. associated neurocognitive disorders. Neuropsychol Rev. 2009;19(2):
69. Fanjul F, Riveiro-Barciela M, Gonzalez J, et al. Evaluation of progressive mul- 152168.
tifocal leukoencephalopathy treatments in a Spanish cohort of HIV-infected 98. Munoz-Moreno JA, Fumaz CR, et al. Nadir CD4 cell count predicts
patients: do protease inhibitors improve survival regardless of central nervous neurocognitive impairment in HIV-infected patients. AIDS Res Hum
system penetration-effectiveness (CPE) score? HIV Med. 2013;14(5):321325. Retroviruses. 2008;24(10):13011307.
70. Clifford DB, Yiannoutsos C, Glicksman M, et al. HAART improves 99. Ellis RJ, Badiee J, Vaida F, et al. CD4 nadir is a predictor of HIV neu-
prognosis in HIV-associated progressive multifocal leukoencephalopathy. rocognitive impairment in the era of combination antiretroviral therapy.
Neurology. 1999;52(3):623625. AIDS. 2011;25(14):17471751.
71. Gasnault J, Taoufik Y, Goujard C, et al. Prolonged survival without neu- 100. Bhaskaran K, Mussini C, Antinori A, et al. Changes in the incidence and
rological improvement in patients with AIDS-related progressive multi- predictors of human immunodeficiency virus-associated dementia in the era
focal leukoencephalopathy on potent combined antiretroviral therapy. of highly active antiretroviral therapy. Ann Neurol. 2008;63(2):213221.
J Neurovirol. 1999;5(4):421429. 101. McCombe JA, Vivithanaporn P, Gill MJ, et al. Predictors of symptomatic
72. De Luca A, Ammassari A, Pezzotti P, et al. Cidofovir in addition to antiretro- HIV-associated neurocognitive disorders in universal health care. HIV
viral treatment is not effective for AIDS-associated progressive multifocal leu- Med. 2013;14(2):99107.
koencephalopathy: a multicohort analysis. AIDS. 2008;22(14):17591767. 102. Schacker T, Collier AC, Hughes J, et al. Clinical and epidemiologic fea-
73. Tan K, Roda R, Ostrow L, et al. PML-IRIS in patients with HIV infec- tures of primary HIV infection. Ann Intern Med. 1996;125(4):257264.
tion: clinical manifestations and treatment with steroids. Neurology. 103. Davis LE, Hjelle BL, Miller VE, et al. Early viral brain invasion in
2009;72(17):14581464. iatrogenic human immunodeficiency virus infection. Neurology.
74. Harrison DM, Newsome SD, Skolasky RL, et al. Immune reconstitution 1992;42(9):17361739.
is not a prognostic factor in progressive multifocal leukoencephalopathy. 104. Vanhems P, Allard R, Cooper DA, et al. Acute human immunodeficiency
J Neuroimmunol. 2011;238(1-2):8186. virus type 1 disease as a mononucleosis-like illness: is the diagnosis too
75. Safdar A, Rubocki RJ, Horvath JA, et al. Fatal immune restoration disease restrictive? Clin Infect Dis. 1997;24(5):965970.
in human immunodeficiency virus type 1-infected patients with progressive 105. del Saz SV, Sued O, Falco V, et al. Acute meningoencephalitis due to
multifocal leukoencephalopathy: impact of antiretroviral therapy-associated human immunodeficiency virus type 1 infection in 13 patients: clinical
immune reconstitution. Clin Infect Dis. 2002;35(10):12501257. description and follow-up. J Neurovirol. 2008;14(6):474479.
76. Hoffmann C, Horst HA, Albrecht H, et al. Progressive multifocal leuco- 106. Ragin AB, Du H, Ochs R, et al. Structural brain alterations can be
encephalopathy with unusual inflammatory response during antiretroviral detected early in HIV infection. Neurology. 2012;79(24):23282334.
treatment. J Neurol Neurosurg Psychiatry. 2003;74(8):11421144. 107. Valcour V, Chalermchai T, Sailasuta N, et al. Central nervous system
77. DAmico R, Sarkar S, Yusuff J, et al. Immune reconstitution after potent viral invasion and inflammation during acute HIV infection. J Infect Dis.
antiretroviral therapy in AIDS patients with progressive multifocal leuko- 2012;206(2):275282.
encephalopathy. Scand J Infect Dis. 2007;39(4):347350. 108. Peluso MJ, Meyerhoff DJ, Price RW, et al. Cerebrospinal fluid and
78. Martinez JV, Mazziotti JV, Efron ED, et al. Immune reconstitution in- neuroimaging biomarker abnormalities suggest early neurological injury
flammatory syndrome associated with PML in AIDS: a treatable disorder. in a subset of individuals during primary HIV infection. J Infect Dis.
Neurology. 2006;67(9):16921694. 2013;207(11):17031712.
79. Martin-Blondel G, Cuzin L, Delobel P, et al. Is maraviroc beneficial in para- 109. Burdo TH, Lackner A, Williams KC. Monocyte/macrophages and their
doxical progressive multifocal leukoencephalopathy-immune reconstitution role in HIV neuropathogenesis. Immunol Rev. 2013;254(1):102113.
inflammatory syndrome management? AIDS. 2009;23(18):25452546. 110. Ancuta P, Kamat A, Kunstman KJ, et al. Microbial translocation is associ-
80. Hall CD, Dafni U, Simpson D, et al. Failure of cytarabine in progressive ated with increased monocyte activation and dementia in AIDS patients.
multifocal leukoencephalopathy associated with human immunodeficiency PloS One. 2008;3(6):e2516.
virus infection. AIDS Clinical Trials Group 243 Team. N Engl J Med. 111. Ellery PJ, Tippett E, Chiu YL, et al. The CD16 monocyte subset is
1998;338(19):13451351. more permissive to infection and preferentially harbors HIV-1 in vivo.
81. Brickelmaier M, Lugovskoy A, Kartikeyan R, et al. Identification and char- J Immunol. 2007;178(10):65816589.
acterization of mefloquine efficacy against JC virus in vitro. Antimicrob 112. Kusao I, Shiramizu B, Liang CY, et al. Cognitive performance related to HIV-
Agents Chemother. 2009;53(5):18401849. 1-infected monocytes. J Neuropsychiatry Clin Neurosci. 2012;24(1):7180.
82. Elphick GF, Querbes W, Jordan JA, et al. The human polyomavirus, JCV, uses 113. Wesselingh SL, Thompson KA. Immunopathogenesis of HIV-associated
serotonin receptors to infect cells. Science. 2004;306(5700):13801383. dementia. Curr Opin Neurol. 2001;14(3):375379.
83. Cettomai D, McArthur JC. Mirtazapine use in human immunodeficiency 114. Gonzalez-Scarano F, Martin-Garcia J. The neuropathogenesis of AIDS.
virus-infected patients with progressive multifocal leukoencephalopathy. Nat Rev Immunol. 2005;5(1):6981.
Arch Neurol. 2009;66(2):255258. 115. Lindl KA, Marks DR, Kolson DL, et al. HIV-associated neurocognitive
84. Lanzafame M, Ferrari S, Lattuada E, et al. Mirtazapine in an HIV-1 infected disorder: pathogenesis and therapeutic opportunities. J Neuroimmune
patient with progressive multifocal leukoencephalopathy. Le Infezioni in Pharmacol. 2010;5(3):294309.
Medicina. 2009;17(1):3537. 116. Spudich S, Gonzalez-Scarano F. HIV-1-related central nervous system dis-
85. Navia BA, Jordan BD, Price RW. The AIDS dementia complex: I. Clinical ease: current issues in pathogenesis, diagnosis, and treatment. Cold Spring
features. Ann Neurol. 1986;19(6):517524. Harb Perspect Med. 2012;2(6):a007120.
86. Antinori A, Arendt G, Becker JT, et al. Updated research nosology for HIV- 117. Glass JD, Fedor H, Wesselingh SL, et al. Immunocytochemical quanti-
associated neurocognitive disorders. Neurology. 2007;69(18):17891799. tation of human immunodeficiency virus in the brain: correlations with
87. Price RW, Brew B, Sidtis J, et al. The brain in AIDS: central ner- dementia. Ann Neurol. 1995;38(5):755762.
vous system HIV-1 infection and AIDS dementia complex. Science. 118. Everall I, Vaida F, Khanlou N, et al. Cliniconeuropathologic correlates
1988;239(4840):586592. of human immunodeficiency virus in the era of antiretroviral therapy.
88. Chiesi A, Vella S, Dally LG, et al. Epidemiology of AIDS dementia complex J Neurovirol. 2009;15(56):360370.
in Europe. AIDS in Europe Study Group. J Acquir Immune Defic Syndr 119. Wendelken LA, Valcour V. Impact of HIV and aging on neuropsychologi-
Hum Retrovirol. 1996;11(1):3944. cal function. J Neurovirol. 2012;18(4):256263.
89. McArthur JC, Hoover DR, Bacellar H, et al. Dementia in AIDS patients: 120. Becker JT, Kingsley L, Mullen J, et al. Vascular risk factors, HIV se-
incidence and risk factors. Multicenter AIDS Cohort Study. Neurology. rostatus, and cognitive dysfunction in gay and bisexual men. Neurology.
1993;43(11):22452252. 2009;73(16):12921299.

Scheld_Ch19.indd 296 2/21/14 5:40 PM


Chapter 19: Human Immunodeficiency Virus 297

121. Foley J, Ettenhofer M, Wright MJ, et al. Neurocognitive functioning 148. Cysique LA, Waters EK, Brew BJ. Central nervous system antiretroviral
in HIV-1 infection: effects of cerebrovascular risk factors and age. Clin efficacy in HIV infection: a qualitative and quantitative review and impli-
Neuropsychol. 2010;24(2):265285. cations for future research. BMC Neurology. 2011;11:148.
122. Wright EJ, Grund B, Robertson K, Brew BJ, et al. Cardiovascular risk 149. Ciccarelli N, Fabbiani M, Colafigli M, et al. Revised central nervous sys-
factors associated with lower baseline cognitive performance in HIV- tem neuropenetration-effectiveness score is associated with cognitive dis-
positive persons. Neurology. 2010;75(10):864873. orders in HIV-infected patients with controlled plasma viraemia. Antivir
123. McCutchan JA, Marquie-Beck JA, Fitzsimons CA, et al. Role of obesity, Ther. 2013;18(2):153160.
metabolic variables, and diabetes in HIV-associated neurocognitive disor- 150. Winston A, Duncombe C, Li PC, et al. Does choice of combination antiret-
der. Neurology. 2012;78(7):485492. roviral therapy (cART) alter changes in cerebral function testing after 48
124. Marra CM, Zhao Y, Clifford DB, et al. Impact of combination antiretro- weeks in treatment-naive, HIV-1-infected individuals commencing cART?
viral therapy on cerebrospinal fluid HIV RNA and neurocognitive perfor- A randomized, controlled study. Clin Infect Dis. 2010;50(6):920929.
mance. AIDS. 2009;23(11):13591366. 151. Shikuma CM, Nakamoto B, Shiramizu B, et al. Antiretroviral mono-
125. Robertson KR, Su Z, Margolis DM, et al. Neurocognitive effects of treat- cyte efficacy score linked to cognitive impairment in HIV. Antivir Ther.
ment interruption in stable HIV-positive patients in an observational 2012;17(7):12331242.
cohort. Neurology. 2010;74(16):12601266. 152. Price RW, Swanstrom R. Targeting chronic central nervous system HIV
126. Ciccarelli N, Fabbiani M, Di Giambenedetto S, et al. Efavirenz associ- infection. Antivir Ther. 2012;17(7):12271231.
ated with cognitive disorders in otherwise asymptomatic HIV-infected 153. Navia BA, Dafni U, Simpson D, et al. A phase I/II trial of nimodipine for
patients. Neurology. 2011;76(16):14031409. HIV-related neurologic complications. Neurology. 1998;51(1):221228.
127. Zunt JR, Tu RK, Anderson DM, et al. Progressive multifocal leukoen- 154. Clifford DB, McArthur JC, Schifitto G, et al. A randomized clinical trial
cephalopathy presenting as human immunodeficiency virus type 1 (HIV)- of CPI-1189 for HIV-associated cognitive-motor impairment. Neurology.
associated dementia. Neurology. 1997;49(1):263265. 2002;59(10):15681573.
128. Sidtis JJ, Price RW. Early HIV-1 infection and the AIDS dementia complex 155. Schifitto G, Yiannoutsos CT, Ernst T, et al. Selegiline and oxidative stress in
[comment]. Neurology. 1990;40(2):323326. HIV-associated cognitive impairment. Neurology. 2009;73(23):19751981.
129. Price RW, Brew BJ. The AIDS dementia complex. J Infect Dis. 156. Schifitto G, Zhang J, Evans SR, et al. A multicenter trial of selegiline
1988;158(5):10791083. transdermal system for HIV-associated cognitive impairment. Neurology.
130. McArthur JC, Nance-Sproson TE, Griffin DE, et al. The diagnostic utility 2007;69(13):13141321.
of elevation in cerebrospinal fluid beta 2- microglobulin in HIV-1 demen- 157. Schifitto G, Navia BA, Yiannoutsos CT, et al. Memantine and HIV-
tia. Multicenter AIDS Cohort Study. Neurology. 1992;42(9):17071712. associated cognitive impairment: a neuropsychological and proton mag-
131. Ellis RJ, Hsia K, Spector SA, et al. Cerebrospinal fluid human immunodefi- netic resonance spectroscopy study. AIDS. 2007;21(14):18771886.
ciency virus type 1 RNA levels are elevated in neurocognitively impaired in- 158. Zhao Y, Navia BA, Marra CM, et al. Memantine for AIDS dementia com-
dividuals with acquired immunodeficiency syndrome. HIV Neurobehavioral plex: open-label report of ACTG 301. HIV Clin Trials. 2010;11(1):5967.
Research Center Group. Ann Neurol. 1997;42(5):679688. 159. Ho EL, Spudich SS, Lee E, et al. Minocycline fails to modulate cerebrospi-
132. Brew BJ, Pemberton L, Cunningham P, et al. Levels of human immuno- nal fluid HIV infection or immune activation in chronic untreated HIV-1
deficiency virus type 1 RNA in cerebrospinal fluid correlate with AIDS infection: results of a pilot study. AIDS Res Ther. 2011;8:17.
dementia stage. J Infect Dis. 1997;175(4):963966. 160. Nakasujja N, Miyahara S, Evans S, et al. Randomized trial of minocycline
133. McArthur JC, McClernon DR, Cronin MF, et al. Relationship between in the treatment of HIV-associated cognitive impairment. Neurology.
human immunodeficiency virus-associated dementia and viral load in 2013;80(2):196202.
cerebrospinal fluid and brain. Ann Neurol. 1997;42(5):689698. 161. Sacktor N, Miyahara S, Deng L, et al. Minocycline treatment for HIV-
134. Ellis RJ, Moore DJ, Childers ME, et al. Progression to neuropsychologi- associated cognitive impairment: results from a randomized trial.
cal impairment in human immunodeficiency virus infection predicted by Neurology. 2011;77(12):11351142.
elevated cerebrospinal fluid levels of human immunodeficiency virus 162. Simioni S, Cavassini M, Annoni JM, et al. Rivastigmine for HIV-associated
RNA. Arch Neurol. 2002;59(6):923928. neurocognitive disorders: a randomized crossover pilot study. Neurology.
135. McArthur JC, McDermott MP, McClernon D, et al. Attenuated central 2013;80(6):553560.
nervous system infection in advanced HIV/AIDS with combination anti- 163. Wendel KA, McArthur JC. Acute meningoencephalitis in chronic human
retroviral therapy. Arch Neurol. 2004;61(11):16871696. immunodeficiency virus (HIV) infection: putative central nervous system
136. Heaton RK, Franklin DR, Ellis RJ, et al. HIV-associated neurocogni- escape of HIV replication. Clin Infect Dis. 2003;37(8):11071011.
tive disorders before and during the era of combination antiretroviral 164. Mehling M, Drechsler H, Kuhle J, et al. Adaptation of antiretroviral
therapy: differences in rates, nature, and predictors. J Neurovirology. therapy in human immunodeficiency virus infection with central nervous
2011;17(1):316. system involvement. J Neurovirol. 2008;14(1):7884.
137. Cohen WA, Maravilla KR, Gerlach R, et al. Prospective cerebral MR 165. Bogoch II, Davis BT, Venna N. Reversible dementia in a patient with cen-
study of HIV seropositive and seronegative men: correlation of MR find- tral nervous system escape of human immunodeficiency virus. J Infect.
ings with neurologic, neuropsychologic, and cerebrospinal fluid analysis. 2011;63(3):236239.
AJNR Am J Neuroradiol. 1992;13(4):12311240. 166. Canestri A, Lescure FX, Jaureguiberry S, et al. Discordance between
138. Sidtis JJ, Gatsonis C, Price RW, et al. Zidovudine treatment of the AIDS cerebral spinal fluid and plasma HIV replication in patients with neu-
dementia complex: results of a placebo- controlled trial. AIDS Clinical rological symptoms who are receiving suppressive antiretroviral therapy.
Trials Group. Ann Neurol. 1993;33(4):343349. Clin Infect Dis. 2010;50(5):773778.
139. Portegies P, Enting RH, de Gans J, et al. Presentation and course of AIDS 167. Eden A, Fuchs D, Hagberg L, et al. HIV-1 viral escape in cerebrospinal
dementia complex: 10 years of follow-up in Amsterdam, The Netherlands. fluid of subjects on suppressive antiretroviral treatment. J Infect Dis.
AIDS. 1993;7(5):669675. 2010;202(12):18191825.
140. Staprans S, Marlowe N, Glidden D, et al. Time course of cerebrospinal 168. Lescure FX, Moulignier A, Savatovsky J, et al. CD8 encephalitis in
fluid responses to antiretroviral therapy: evidence for variable compart- HIV-infected patients receiving cART: a treatable entity. Clin Infect Dis.
mentalization of infection. AIDS. 1999;13(9):10511061. 2013;57(1):101108.
141. Sacktor NC, Lyles RH, Skolasky RL, et al. Combination antiretroviral 169. Rosenblum M, Scheck AC, Cronin K, et al. Dissociation of AIDS-related
therapy improves psychomotor speed performance in HIV-seropositive vacuolar myelopathy and productive HIV-1 infection of the spinal cord.
homosexual men. Multicenter AIDS Cohort Study (MACS). Neurology. Neurology. 1989;39(7):892896.
1999;52(8):16401647. 170. Tyor WR, Glass JD, Baumrind N, et al. Cytokine expression of mac-
142. Sacktor NC, Skolasky RL, Lyles RH, et al. Improvement in HIV-associated rophages in HIV-1-associated vacuolar myelopathy. Neurology. 1993;
motor slowing after antiretroviral therapy including protease inhibitors. 43(5):10021009.
J Neurovirol. 2000;6(1):8488. 171. Eilbott DJ, Peress N, Burger H, et al. Human immunodeficiency virus type
143. Dore GJ, McDonald A, Li Y, et al. Marked improvement in survival fol- 1 in spinal cords of acquired immunodeficiency syndrome patients with
lowing AIDS dementia complex in the era of highly active antiretroviral myelopathy: expression and replication in macrophages. Proc Natl Acad
therapy. AIDS. 2003;17(10):15391545. Sci U S A. 1989;86(9):33373341.
144. Cysique LA, Vaida F, Letendre S, et al. Dynamics of cognitive change 172. Petito CK, Navia BA, Cho ES, et al. Vacuolar myelopathy pathologically
in impaired HIV-positive patients initiating antiretroviral therapy. resembling subacute combined degeneration in patients with the acquired
Neurology. 2009;73(5):342348. immunodeficiency syndrome. N Engl J Med. 1985;312(14):874879.
145. Crum-Cianflone NF, Moore DJ, Letendre S, et al. Low prevalence of neu- 173. Di Rocco A, Bottiglieri T, Werner P, et al. Abnormal cobalamin-
rocognitive impairment in early diagnosed and managed HIV-infected dependent transmethylation in AIDS-associated myelopathy. Neurology.
persons. Neurology. 2013;80(4):371379. 2002;58(5):730735.
146. Letendre S, Marquie-Beck J, Capparelli E, et al. Validation of the CNS 174. Modi G, Ranchhod J, Hari K, et al. Non-traumatic myelopathy at the
Penetration-Effectiveness rank for quantifying antiretroviral penetration Chris Hani Baragwanath Hospital, South Africathe influence of HIV.
into the central nervous system. Arch Neurol. 2008;65(1):6570. QJM. 2011;104(8):697703.
147. Letendre S. Central nervous system complications in HIV disease: 175. Dal Pan GJ, Glass JD, McArthur JC. Clinicopathologic correlations of
HIV-associated neurocognitive disorder. Top Antivir Med. 2011;19(4): HIV-1-associated vacuolar myelopathy: an autopsy-based case-control
137142. study. Neurology. 1994;44(11):215964.

Scheld_Ch19.indd 297 2/21/14 5:40 PM


298 Part II: Viral Infections and Related Disorders

176. Chong J, Di Rocco A, Tagliati M, et al. MR findings in AIDS-associated 181. Fernandez-Fernandez FJ, de la Fuente-Aguado J, Ocampo-Hermida A, et
myelopathy. AJNR Am J Neuroradiol. 1999;20(8):14121416. al. Remission of HIV-associated myelopathy after highly active antiretro-
177. Shimojima Y, Yazaki M, Kaneko K, et al. Characteristic spinal MRI viral therapy. J Postgrad Med. 2004;50(3):195196.
findings of HIV-associated myelopathy in an AIDS patient. Intern Med. 182. Eyer-Silva WA, Couto-Fernandez JC, Caetano MR, et al. Remission of
2005;44(7):763764. HIV-associated myelopathy after initiation of lopinavir in a patient with
178. Geraci A, Di Rocco A, Liu M, et al. AIDS myelopathy is not associ- extensive previous exposure to highly active antiretroviral therapy. AIDS.
ated with elevated HIV viral load in cerebrospinal fluid. Neurology. 2002;16(17):23672369.
2000;55(3):440442. 183. Di Rocco A, Werner P, Bottiglieri T, et al. Treatment of AIDS-associated
179. Bizaare M, Dawood H, Moodley A. Vacuolar myelopathy: a case report myelopathy with L-methionine: a placebo-controlled study. Neurology.
of functional, clinical, and radiological improvement after highly active 2004;63(7):12701275.
antiretroviral therapy. Int J Infect Dis. 2008;12(4):442444. 184. Cikurel K, Schiff L, Simpson DM. Pilot study of intravenous immu-
180. Staudinger R, Henry K. Remission of HIV myelopathy after highly active noglobulin in HIV-associated myelopathy. AIDS Patient Care STDS.
antiretroviral therapy. Neurology. 2000;54(1):267268. 2009;23(2):7578.

Scheld_Ch19.indd 298 2/21/14 5:40 PM


CHAPTER 20 GUILLAIN-BARR SYNDROME
TONY M. MCGRATH

Since the decline in cases of poliomyelitis, the Guillain-Barr In 1976, following an apparent increase in GBS associ-
syndrome (GBS) has emerged as the most frequent cause of ated with the swine flu vaccine, the National Institute of
acute flaccid paralysis. The spectrum of GBS includes classi- Neurological and Communicative Disorders and Stroke
cal acute inflammatory demyelinating polyneuropathy (AIDP), (NINCDS) was directed by the Centers for Disease Control
acute motor axonal neuropathy (AMAN), acute motor sen- and Prevention (CDC) and the National Institute of Allergy
sory axonal neuropathy (AMSAN), and variants such as Fisher and Infectious Diseases to develop clear diagnostic criteria for
syndrome (1,2). AIDP, the most common form in the Western GBS to assist in epidemiologic studies. These criteria (NINCDS
Hemisphere, is characterized by rapidly progressive, usually criteria) were reported in 1978 (3) and, though intentionally
ascending, and symmetric paralysis of the extremities, variable somewhat restrictive, are now generally accepted. The clinical
involvement of bulbar, facial, and ocular muscles, and loss of section of this chapter contains a summary of these criteria.
deep tendon reflexes (3). Although the disorder is often pre-
ceded by a bacterial or viral illness, no evidence presently exists
to indicate that GBS is caused by direct infection of nervous tis- EPIDEMIOLOGY
sue. Current research implicates an autoimmune mechanism.
Epidemiologic studies of GBS are inherently troublesome (8).
The major difficulty is the lack of definitive diagnostic markers
HISTORY for GBS. Because diagnosis must rely on clinical criteria, inci-
dence may be over- or underestimated if criteria are too broad
Landry is credited with the first modern description of a dis- or stringent or if atypical variants are counted or excluded. Also,
order similar to GBS. In 1859, he reported a patient with par- the results of studies employing dissimilar criteria may not be
esthesias and subjective weakness. Unlike present definitions comparable. A dilemma arises in regard to whether persons with
of GBS, objective weakness did not occur until 1 month after an underlying condition known to produce a neuropathy, such
symptom onset but was then severe and rapidly progressive, as diabetes mellitus, should be included when one is attempting
leading to death 8 days later (4). Although Oslers account in a survey of a pure neuropathy such as GBS. Moreover, GBS dis-
1892 (5) of acute febrile polyneuritis was remarkably similar plays a wide range of clinical severity. Because many epidemio-
to the illness we now term GBS, the development of the lumbar logic studies rely on hospital-based reporting, mild cases not
puncture in the late nineteenth century permitted a more pre- requiring hospitalization could be lost. Despite these difficul-
cise characterization of the disorder and its differentiation from ties, numerous epidemiologic investigations of GBS have been
poliomyelitis and other neuropathies (6). In 1916, Guillain performed. Ten of these are summarized in Table 20.1.
et al. (7) detailed a clinical syndrome in two individuals char- Incidence rates in studies using NINCDS criteria (914,15)
acterized by motor weakness, paresthesias, areflexia with pres- range from 0.4 to 2.0 per 100,000 persons. In the population-
ervation of cutaneous reflexes, and increased cerebrospinal based Olmstead County survey (9), 15% of GBS cases were mild
fluid (CSF) albumin without pleocytosis (albuminocytologic and did not require hospitalization. Therefore, hospital-based
dissociation). Both recovered fully. Similar descriptions soon studies (10,11,13,14) may underestimate the true rate by as
followed, but controversy arose regarding diagnostic criteria. much as 15%. Nevertheless, incidence rates are similar among

TA B L E 2 0 . 1
REGION SURVEYED, PERIOD OF SURVEY, TOTAL, MALE, AND FEMALE GUILLAIN-BARR SYNDROME
INCIDENCE RATE PER 100,000 PEOPLE, AND NUMBER

Average Annual Incidence Rate

Region Surveyed Time Period Total Male Female Cases (n)

Olmstead County, Minnesota (9) 46 yr (19351980) 1.7 2.3 1.2 48


Hordaland, Western Norway (10) 26 yr (19571982) 1.14 1.45 0.83 109
Sardinia (11) 12 yr (19691980) 0.4 0.41 0.39 120
Stockholm, Sweden (12) 19 yr (19731991) 1.84 2.15 1.57 556
Western Australia (13) 6 yr (19801985) 1.35 1.49 1.2 109
Benghazi, Libya (14) 3 yr (19831985) 1.74 1.62 1.86 27
Ontario, Canada (16) 7 yr (19831989) 2.02 1,302
Quebec, Canada (16) 7 yr (19831989) 2.3 1,031
Spain (17) 13 yr (19851997) 0.85 1.14 0.58 337
Ferrara, Italy (15) 8 yr (19942001) 1.97 2.25 1.71 26

299

Scheld_Ch20.indd 299 2/21/14 5:40 PM


300 Part II: Viral Infections and Related Disorders

TA B L E 2 0 . 2 60% of the infections, followed by gastrointestinal ailments and


nonspecific febrile illnesses. Elderly persons with GBS experience
DIAGNOSTIC CRITERIA PROPOSED BY a similar incidence of antecedent infections (27,28). However,
GUILLAIN-BARR SYNDROME STUDY GROUP they may be less likely to complain of preceding gastrointestinal
symptoms (27). As many as 70% of children with GBS experi-
Features required for diagnosis ence an infectious disease within 28 days of neurologic symptom
Progressive motor weakness of more than one extremity onset, with nonspecific upper respiratory illnesses being respon-
Areflexia or marked hyporeflexia sible in nearly 80% (2023). Yet, gastrointestinal prodromic
symptoms may vary. A recent study found that vomiting was
CSF cell counts of no more than 50 monocytes or
more common in children with AIDP compared with AMAN
two polymorphonuclear leukocytes
(28.1% vs. 6.5%), and diarrhea was more common in AMAN
Features strongly supportive of the diagnosis compared with AIDP (32.6% vs. 12.5%) (19). Persons with
Progression for days to a few weeks severe GBS resulting in tetraparesis and prolonged mechanical
Relative symmetry of weakness ventilation are generally no more likely than mildly or moder-
ately affected patients to have an antecedent infection (2931).
Mild sensory signs or symptoms
Neurologic symptoms usually begin within 2 weeks after
Cranial nerve involvement the infectious illness (9,13,16,26,27,30,31) but may occur
Onset of recovery 24 wk after cessation of progression more acutely (31). Shorter periods between the prodromal
Autonomic dysfunction illness and neurologic symptoms seem to follow antecedent
upper respiratory infections, but a longer delay may occur in
Initial absence of fever
patients with preceding gastrointestinal symptoms (32,33).
Elevated CSF protein after 1 wk of symptoms In the majority, an infectious agent responsible for the prodro-
Abnormal electrodiagnostic tests with slowed conduction mal illness is never identified.
or prolonged F-waves Many viruses and bacteria (Table 20.3) have been impli-
cated as antecedent triggering factors in GBS (16,21,28,3236),
Modified from ref. 25. although many of the proposed causal agents and GBS may
simply represent a chance association. Winer et al. (26) tested
the proposition in serologic studies on 100 persons with GBS
and age- and sex-matched controls. Evidence of cytomega-
lovirus (CMV) and C. jejuni infection was significantly more
studies conducted in different decades, ethnic groups, races, and frequent in GBS patients (11% and 14%, respectively) than
geographic areas. These findings are consistent with the hypoth- among controls. Although positive Epstein-Barr virus (EBV),
esis that the triggering agents responsible for GBS are widespread
and numerous, and susceptibility is similar among populations.
Most studies of GBS have revealed higher incidence
among men (911,13) and no significant seasonal differences TA B L E 2 0 . 3
(9,13,16). Incidence increases with advancing age beyond PROPOSED INFECTIOUS ANTECEDENTS OF
40 years, commonly peaking in the seventh and eighth decades GUILLAIN-BARR SYNDROME
(10,11,13,14,16,17). Autumn clustering of GBS following
Campylobacter jejuni infection has occurred (18,19). Viruses Bacteria
Four epidemiologic surveys have examined the incidence
of GBS in children (2023). Two (21,23) used NINCDS crite- Adenovirus Brucella
ria, and two (20,22) used criteria outlined in Table 20.2 (24). Coronavirus Campylobacter jejuni
Although children with clinical variants of GBS were identified,
they were not included in the results. Annual incidence rates of Coxsackievirus Haemophilus influenzae
GBS in children were lower (0.4 to 1.1 per 100,000 children) Cytomegalovirus Helicobacter pylori
than rates obtained in the general population (see Table 20.1). Epstein-Barr virus Listeria monocytogenes
No difference in frequency between boys or girls occurred in
Echo virus Mycoplasma pneumoniae
California (20) and Finland (22), whereas male-to-female ratios
of 1.5:1 were observed in Paraguay (21) and Taiwan (23). Hepatitis A, B, and C Pasteurella tularensis
Although a seasonal preponderance was not apparent in three Herpes simplex Salmonella typhosa
studies (20,22,23), 76% of affected children in Paraguay (21) Herpes zoster Shigella
had onset of symptoms during the summer months.
Human herpesvirus type 6 Yersinia
When childhood GBS in California (20) was analyzed by
age at onset, frequency was significantly greater in 2-year-old Human immunodeficiency virus
children than among other age groups. This finding may reflect Influenza A and B
the higher incidence of triggering infections in this age group. Japanese encephalitis virus
In Paraguay (21), a similar increase in children younger than
Measles
4 years of age was seen, but in Finland (22) and Taiwan (23),
age distribution was evenly dispersed. Mumps
Parainfluenza
Respiratory syncytial virus
Antecedent Events Vaccinia
Clinically apparent antecedent infections occur within 28 days Variola
of onset in approximately 50% of persons with GBS (914,16, West Nile virus
25,26). Minor upper respiratory symptoms account for about

Scheld_Ch20.indd 300 2/21/14 5:40 PM


Chapter 20: Guillain-Barr Syndrome 301

parvovirus B19, and Mycoplasma pneumoniae serologies were occurs in epidemics in endemic areas. Ravi et al. (34) detected
identified in GBS, controls demonstrated similar results. Hankey high titers of IgM antibody specific to JEV in serum and/or CSF
(13) obtained acute and convalescent serum from 62 persons in 62% (21 of 34) of consecutive cases in southern India. None
with GBS and found evidence of recent viral infection in 50%. had signs of encephalitis, and all satisfied NINCDS criteria (see
Parainfluenza virus type 3, CMV, and adenovirus titers were later discussion) for GBS. Clinical course and outcome were
identified in five, six, and seven specimens, respectively, whereas similar in JEV-seropositive and -seronegative individuals. The
EBV, herpes simplex, herpes zoster, parainfluenza type 1 and 2, JEV antigen was demonstrable in the CSF of one person, and
measles, coxsackie, echovirus, coronavirus, and influenza A and virus was isolated from CSF in another. Pathologic examination
B titers were each present in one or two samples. Infection with in one autopsied case demonstrated discrete necrotic lesions at
M. pneumoniae was encountered in five. Despite these various the cerebral graywhite matter junction and demyelination of
reports, convincing evidence of causality exists for only CMV, ventral and dorsal nerve roots. West Nile virus (WNV) was
EBV, human immunodeficiency virus (HIV), herpes zoster, C. reported in an individual with GBS during the 1999 outbreak
jejuni, and M. pneumoniae. Measles-associated GBS may occur, in New York City (42). During the summer of 2002, six cases
but only rarely. Although variola (smallpox) and vaccinia have of acute flaccid paralysis syndrome associated with WNV were
been conclusively linked with GBS, the eradication of smallpox reported in Mississippi and Louisiana (43). The individuals,
and the discontinuation of vaccinia immunization make any however, had asymmetric weakness and CSF pleocytosis, sug-
association with GBS of historic interest only. However, since gesting a polio-like syndrome. Falciparum malaria has also
the September 11, 2001 terrorist attack and the threat of bio- been reported with GBS (44,45). Sokrab et al. (45) reported a
terrorism, the expected increase in smallpox vaccinations could mortality rate of 40% in 10 individuals with acute falciparum
lead to an increase in vaccine-related GBS. malaria and GBS during a seasonal exacerbation.
Dowling and Cook (36) detected immunoglobulin M (IgM) In 1982, Rhodes and Tattersfield (46) reported the first case
antibody directed against CMV in 15% of 220 persons with of GBS following C. jejuni enteritis. Subsequently, C. jejuni
GBS. Those with CMV antibodies were primarily young has been recognized as the most common bacterial antecedent
women (average age, 25.6 years) in whom only 80% had a infection in GBS. Serotyping of C. jejuni is accomplished by
clinically apparent preceding infection. CMV-associated GBS examination of heat-labile antigens (Lior method) and/or heat-
appeared in three separate 10- to 16-week clusters over 3 years. stable antigens (Penner method) (47). Several Penner and Lior
Abnormal liver enzymes were present in 50% of the CMV sero- serotypes of C. jejuni have been isolated from GBS patients, but
positive cases. Evidence of recovery was associated with a fall in Penner serotype 19 and Lior serotype 11 predominate (4858).
CMV IgM antibody titer. No correlation has been demonstrated These serotypes are uncommon in uncomplicated gastroenteri-
between primary infection with CMV and severity or outcome tis (47). Winer et al. (26) and Boucquey et al. (33) found sero-
of GBS (26,33). CMV-associated GBS has occurred following logic evidence of recent C. jejuni infection in approximately
transplant of a CMV-infected heart in a CMV-negative recipi- 14% of GBS, and Mishu et al. (18) and Kaldor and Speed (59)
ent (37) and may occur with CMV reactivation after autolo- reported C. jejuni infection in 37% of GBS. No history of pre-
gous bone marrow or organ transplantation (37,38). ceding gastrointestinal symptoms has been reported in some
EBV (like CMV, a herpesvirus) infection may precede GBS patients with serologic evidence of recent C. jejuni infec-
neurologic symptoms in 2% to 8% of GBS (20,26,28). tion (33,59). The incidence of C. jejuni infection in the United
It may present as hepatitis or mononucleosis or, like CMV, States peaks from August through October (56), whereas the
may be clinically silent prior to GBS onset. Clinical or sub- peak incidence for children in Mexico is from July through
clinical encephalopathy may complicate EBV-associated GBS September (19). Statistically significant summer and autumn
(36). Antecedent primary herpes zoster infection may occur clustering of C. jejuniassociated GBS has occurred (18).
in 3% to 4% of children with GBS (20,23). GBS may also Stool cultures after onset of GBS with C. jejuni seroposi-
develop in association with reactivated herpes zoster infection tivity are often negative (33,49,52,54,59), because C. jejuni
(13,25,30). In contrast, recent herpes simplex virus infection excretion lasts an average of 16 days (60), and GBS symptoms
is uncommonly identified in GBS (34). The most recently iden- may be delayed until 3 weeks after gastrointestinal symptoms.
tified herpesvirus, human herpesvirus type 6 (HHV-6), has Appropriate antibiotic treatment of C. jejuni enteritis does not
been proposed as an etiologic agent in GBS. Antibody against prevent GBS (46,49).
HHV-6 was demonstrated in a significantly greater number of Although Boucquey et al. (33) and Enders et al. (54)
GBS patients than among blood donors in Italy (35). reported no association between C. jejuni seropositivity and
GBS has been described early in the course of HIV infection GBS severity or outcome, severe disease and poor prognosis
(3941). In 1987, HIV was the most common infection associ- in C. jejuniassociated GBS is well recognized (26,46,59,61).
ated with GBS at the University of Miami School of Medicine In one series of 21 C. jejuniinfected persons with GBS, 90%
(41). In Zimbabwe, 55% of 29 consecutive persons with required mechanical ventilation (59). Severe involvement may
GBS were HIV seropositive (39). HIV infection did not pro- occur because of a tendency of C. jejuniassociated GBS to
duce a higher incidence of prodromal illness in GBS. In each damage axons rather than myelin (62). Of 11 children in China
seropositive individual, GBS was the initial manifestation of with AMAN, 90% demonstrated serologic evidence of recent
HIV infection. Generalized lymphadenopathy, coincident en- C. jejuni infection (63). Conversely, Enders et al. (54) reported
cephalopathy, and CSF pleocytosis were significantly more no greater electrophysiologic evidence of axonal involvement
likely with HIV infection. Mean CSF white blood cell counts in in C. jejuniassociated GBS, and although Vriesendorp et al.
HIV-associated GBS were 14/mm3 (range of 0 to 63 cells) (39). (61) demonstrated severe disease and poor recovery more
HIV seropositivity has not been associated with poor recov- commonly in C. jejuniseropositive GBS, electrophysiologic
ery following GBS (40,41). GBS in HIV-infected individuals is evidence of primary axonal involvement was not apparent.
probably secondary to disordered immune regulation charac- The mechanism by which C. jejuni infection triggers GBS
teristic of the infection. is now better understood. Antigenic cross reactivity occurs
Several mosquito-borne infections have been reported with between C. jejuni and components of peripheral nerves (PNs)
GBS. Japanese encephalitis virus (JEV) may be an important an- (64,65). Host factors may determine whether infection with
tecedent infection in endemic areas. JEV is the most frequently C. jejuni produces GBS. Although Hillert et al. (66) found no
recognized cause of mosquito-borne encephalitis and typically common human leukocyte antigen (HLA) class II antigens in

Scheld_Ch20.indd 301 2/21/14 5:40 PM


302 Part II: Viral Infections and Related Disorders

GBS, and Winer et al. (67) demonstrated no association be- not experience an increased frequency of GBS (80). The fac-
tween HLA class I antigen and GBS and only a weak associa- tor associated with the civilian vaccine that was responsible
tion between HLA class II antigen DR2 and severe GBS, Yuki for the increased risk of GBS has never been determined; how-
et al. (52) reported HLA class I antigen B35 in seven Penner se- ever, in 2008, Nachamkin et al. showed that the 1976 swine
rotype 19 C. jejuniassociated cases of GBS. The authors con- flu vaccine contained contaminating moieties which elicited
cluded that the combination of HLA B35 antigen and Penner anti-GM1 antibodies after inoculation into C3H/HeN mice.
serotype 19 C. jejuni infection could precipitate an autoim- Vaccine samples from 11 unopened vaccine lots containing
mune response culminating in GBS. This is due to molecular A/New Jersey/76 swine influenza antigen from three different
mimicry between lipooligosaccharides (LOSs) of C. jejuni and manufactures were found to induce IgG and IgM antibodies
gangliosides in PNs (68). The sialyltransferase gene (cst-II) has to GM1, but no to C. jejuni, after immunization in mice. This
been identified to produce ganglioside-like LOS. The gene con- response was also observed in both the 1991 to 1992 and 2004
sists of 291 amino acids; the 51st determines its enzymatic ac- to 2005 vaccines. No vaccines were found to contain bacterial
tivity. Cst-II (Thr51) produces GM1-like and GD1a-like LOSs, DNA (81).
whereas Cst-II (Asn51) produces GT1a-like and GD1c-like GBS has been reported following other vaccinations, but
LOSs which mimic GQ1b (69,70). clear evidence of causality is lacking. Hankey (13) recorded
Evidence of M. pneumoniae infection has been confirmed recent vaccination in 5% of GBS, and of 100 individuals with
in 1% to 5% of GBS (13,23,26,34). Currently, it is the second GBS studied by Winer et al. (26), 6 had received a vaccination
most commonly identified nonviral pathogen associated with within 3 months of GBS onset. Ten vaccines were involved,
GBS. An influenza-like syndrome may precede the onset of and the rate of vaccination was similar in a control population.
neurologic symptoms. The latter study illustrates the importance of discovering the
Mori et al. (71) reported elevated antiHaemophilus influ- frequency of vaccination in the background population before
enzae antibodies in 6 of 46 (13%) Japanese patients with GBS assigning significance to a case of GBS following vaccination.
but none of the 49 controls. Western blot analysis performed Several reports of GBS following vaccination against polio-
on the H. influenzaepositive patients immunoglobulin G myelitis have been recorded. In 1984, an outbreak of poliomy-
(IgG) confirmed recognition both of lipopolysaccharides of elitis in Finland resulted in an intensified vaccination program
H. influenzae and the ganglioside, GM1 (72). with oral poliovirus vaccine (OPV) (82). Subsequently, an
In 1998, Chiba et al. (73) detected antibodies against unexpected rise in GBS occurred, precipitating epidemiologic
Helicobacter pylori in the CSF from 4 of 7 patients with GBS. surveys attempting to link OPV with GBS. Although an unex-
In 2002, Chiba et al. (74) examined CSF from 13 patients with pectedly high rate of GBS occurred within 10 weeks following
GBS (8 AIDP, 4 AMAN, 1 unexcitable nerve conduction) and OPV administration, a small number of persons were involved,
8 control patients. Six of the AIDP patients had a specific IgG and an unexpectedly high rate of GBS also occurred in the
antibody to the VacA of H. pylori. weeks prior to the vaccination program.
Rabies vaccine and the A/New Jersey/76 swine influenza Five percent to ten percent of GBS cases occur following
vaccine (swine flu vaccine) have been reliably linked with GBS intracranial, thoracic, abdominal, and orthopedic surgical
(32). Early rabies vaccine was made of desiccated brain and spi- procedures (6,13,26,29,30,33,37). This could be secondary to
nal cord of rabies-infected animals. Both acute demyelinating primary CMV infection following blood transfusion, although
encephalomyelitis (ADEM) and GBS sometimes followed vacci- many reports exist of persons who did not receive transfusion.
nation. A proliferative lymphocytic response and antibodies to Other sources of iatrogenic infection associated with surgery
myelin basic protein (MBP), a constituent of central and periph- or hospitalization could be responsible. Possibly, the surgery
eral nervous system myelin, have been demonstrated in persons itself elicits a disordered immune response culminating in GBS.
with rabies vaccineassociated GBS (75). An immune response GBS may also occur during pregnancy or the postpartum
against MBP has been proposed as the cause of ADEM and period (11,13,30,32). Unlike infants of mothers with myas-
GBS in these individuals. The discontinuation of rabies vaccine thenia gravis, infants of mothers with GBS are unaffected.
prepared from infected animal brain and spinal cord has virtu- Whether the pregnancy or an associated infection, such as
ally eliminated rabies vaccineassociated ADEM and GBS. CMV, precipitates GBS is uncertain. Clinically and patho-
In 1976 and 1977, an increase in GBS in the United States logically typical cases of GBS have also been associated with
was temporally associated with the A/New Jersey/76 swine malignancy, especially Hodgkin and non-Hodgkin lymphoma
influenza vaccination campaign. The original CDC study (13,33,83). GBS is rarely associated with other autoimmune
demonstrated a statistically significant increase in the risk of disorders (32).
developing GBS within 6 weeks of swine flu vaccine adminis-
tration in persons 18 years of age and older (76). This study has
been criticized because uniform GBS diagnostic criteria were PATHOLOGY
not used, records were reviewed by nonphysician casework-
ers, no standardized method of record assessment was used, The pathologic findings in GBS are remarkably uniform
and follow-up evaluations were not performed (77). Safrenek irrespective of the antecedent illness or precipitating event.
et al. (78) reviewed all GBS cases in Michigan and Minnesota Lesions usually occur as discrete foci of abnormality and have
from October 1, 1976 through January 31, 1977. Diagnostic been observed along the entire length of PNs. Areas of pref-
criteria were developed, and a neurologist reviewed all medical erential involvement are the nerve roots, sites of ligamentous
records blindly. Although mistakes in the original CDC study entrapment, and distal terminal branches (32). Kanda et al.
were uncovered, these authors also demonstrated a significant (84) noted a relative absence of lesions in the initial few mil-
increase in the relative risk of adults developing GBS within limeters of anterior and dorsal nerve roots. This sparing of
6 weeks of swine flu vaccine. Four manufacturers produced the transitional region between central and peripheral nervous
the swine flu vaccine administered to the civilian population systems (CNS and PNS) may be attributable to the marginal
in 1976. All were associated with an increased risk of GBS blood supply in this location. The pathologic process may
(79). GBS has not been linked to previous or subsequent influ- involve autonomic, motor, and sensory systems simultane-
enza vaccinations. Military personnel who received swine flu ously, but in individual cases, abnormalities may predominate
vaccine developed by the United States military in 1976 did in one. Areas of maximal pathologic change usually parallel

Scheld_Ch20.indd 302 2/21/14 5:40 PM


Chapter 20: Guillain-Barr Syndrome 303

clinical involvement, although this association is imperfect. two types (8894). One type consists of actively phagocytic mac-
Generally, motor involvement exceeds sensory changes, but in rophages that occur in an extratubal location within the endoneu-
most patients, pathologic lesions do not preferentially or more rial space and in an intratubal location beneath the Schwann cell
severely involve the anterior roots or motor nerves (32,8488). basement membrane. Focal lysis of the outermost myelin lamellae
In a subset of GBS patients primarily manifesting motor dys- occurs in the areas contacted by the macrophage. The cytoplas-
function, relative sparing of dorsal roots and sensory nerves mic projections of the macrophage then penetrate through the
has been noted (63,85). The pathologic hallmark of GBS is a myelin gaps, insinuate themselves between the myelin lamellae,
perivenular mononuclear cell infiltrate with segmental demy- and peel away layers of myelin from the axon. These invading,
elination. In 1969, Asbury et al. described multifocal, peri- actively phagocytic macrophages express major histocompatibil-
venular mononuclear cell infiltration involving motor, sensory, ity complex (MHC) class II antigens (86,88,9598).
autonomic, and cranial nerves (85). The invading lymphocytes The manner in which macrophages accomplish myelinoly-
and monocytes were derived from blood. Even in diffusely sis is not understood. Arstila et al. (92) demonstrated increased
involved nerves, inflammatory cell concentration was great- acid phosphatase and acid proteinase activities in GBS lesions.
est around blood vessels of the endoneurium, the connective These lysosomal enzymes may be derived from macrophages.
tissue surrounding individual nerve fibers. Infiltration about However, the macrophage processes observed in GBS lesions are
vessels of the epineurium, the connective tissue sheath of the devoid of organelles, including lysosomes (90). The second type
entire nerve, was present but less commonly observed. of macrophage, the foamy macrophage, is postphagocytic, con-
Small and medium-sized lymphocytes were the predomi- tains myelin debris and lipids, and does not express MHC class
nant inflammatory cells recognized early; macrophages were II antigen (95). In GBS lesions, MHC class II antigen expression
the more prominent cell type in older lesions with considerable has also been observed on endothelial and Schwann cells (98).
myelin destruction. Polymorphonuclear leukocytes were infre- Macrophages have also been observed within the axon cyl-
quently observed and then only in early, extensively affected inder (63). This location might explain some cases of axonal
areas, apparently in response to tissue necrosis. Plasma cells degeneration that apparently ensue in the absence of severe
appeared in some chronic processes. Lesions of various ages demyelination. McKhann et al. (63) reported pathologic findings
were present within specimens from individual cases. Indeed, on 10 autopsies from individuals with acute flaccid paralysis in
reparative processes were apparent in areas still presenting evi- China, a syndrome clinically indistinct from GBS. Although two
dence of ongoing myelin breakdown (85). of the patients had pathologic findings similar to those described
Alterations of the myelin sheath occurred in the areas of inflam- by Asbury et al. (85), five were dominated by selective involve-
matory cell infiltration (85). Retraction at the node of Ranvier ment of motor axons with absence of inflammation and little
was seen in early lesions; more advanced changes included a evidence of demyelination. Numerous macrophages were pres-
complete dissolution of the myelin sheath. Myelin disintegration ent in the areas of axons undergoing wallerian degeneration. The
appears to evolve from the nodal region toward the centrally macrophages were located within both endoneurium and degen-
located Schwann cell. These areas of segmental demyelination erating axons. The authors have designated this entity AMAN.
were bounded by intervals of normal-appearing myelin. Active Immunoglobulin and activated complement deposits have
phagocytosis of myelin was evident. Axonal damage with resul- been described in GBS nerve biopsy specimens. Nyland et al.
tant wallerian degeneration was present almost exclusively in (96) detected activated complement C3b receptor along nerves
areas with extensive inflammation. Asbury et al. (85) concluded in GBS. Positive staining for C3 was observed along nerve fibers,
that axonal degeneration was a secondary phenomenon induced and IgG and IgM were apparent in the region of the myelin
by the severe inflammation and demyelination. sheath. Other investigators have failed to demonstrate immuno-
Schwann cell proliferation was present in lesions from globulin within the myelin sheath (8890,94). Immunoglobulin
patients surviving longer than 2 weeks. Although demon- staining of the endoneurium has occasionally been noted
strated in areas with segmental demyelination but preserved (89,90). Deposition of IgM on perineurium is a normal find-
axons, Schwann cell proliferation was most common in nerves ing (89,90). Deposits of C3d were present in the myelin sheath
that had undergone wallerian degeneration. The proliferat- of a single patient (94). In mice models of GBS, deposition of
ing Schwann cells were arranged in columns, forming a tube complement at the presynaptic motor nerve terminus has been
to guide the regenerating axons. Patients surviving several reported (99,100,101). This in turn leads to a blockage of syn-
months with incomplete recovery of motor function demon- aptic transmission at the neuromuscular junction (102).
strated evidence of imperfect remyelination: myelin sheaths Axonal degeneration in the absence of demyelination or
were thin and pale, internodes were shorter, and Schwann cell inflammation has also been observed by Feasby et al. (88).
numbers were increased (85). Macrophages were not seen within the axon cylinder, and no
Subsequent examinations have supported many of the orig- evidence of inflammation or macrophage-associated demy-
inal findings of Asbury et al., but marked variability in the elination was encountered. Axonal degeneration of motor and
pathologic lesions of GBS is now commonly reported. These sensory nerves was severe. Electrophysiologic studies demon-
reports have described, even in early lesions, less conspicuous strated inexcitability of motor nerves, a pattern seen primar-
or absent lymphocytic infiltrate (63,84,8691). Treatment with ily in axonal degeneration. The authors named this variant
steroids or other immunomodulatory therapies may alter the AMSAN. Prognosis for recovery is poor with axonal degen-
degree of lymphocytic infiltration. Immunohistochemical anal- eration, whether it is a primary event or secondary to intense
ysis of biopsy material has demonstrated T-cell lineage in most inflammation and demyelination.
of the invading lymphocytes with only a few B cells present
(80,83). Although CD4 helper/inducer and CD8 cytotoxic/
suppressor T cells are present, CD4 cells may predominate in PATHOGENESIS OF
early lesions (88). Like Asbury et al. (85), Hanovar et al. (86) GUILLAIN-BARR SYNDROME
were unable to identify the origin of the larger mononuclear
cells present in more advanced lesions. The most compelling evidence for a T-cellmediated mechanism
Pathologic reports of GBS have confirmed the invariable pres- in the pathogenesis of GBS is the finding of T-cell infiltration in
ence of macrophages in areas of demyelination. Electron micro- affected nerves (32,85,87,88,90,91). Another indicator of T-cell
scopic (EM) studies have demonstrated macrophages existing as involvement is the presence of lymphoblasts in blood during

Scheld_Ch20.indd 303 2/21/14 5:40 PM


304 Part II: Viral Infections and Related Disorders

early GBS and a possible correlation between their number and In recent years, the humoral immune system has been
disease severity (103). Increases in HLA-DR antigen and trans- implicated in the pathogenesis of GBS because of (a) the dem-
ferrin receptor, both present with T-cell activation, have been onstration of immunoglobulin and complement deposition in
observed on the surface of T cells in GBS (104108). Serum GBS nerve biopsy specimens (89,90,94,95), (b) the therapeu-
levels of interleukin-2 (IL-2), a T-cell growth factor, and serum- tic effect of plasmapheresis and intravenous immunoglobulin
soluble IL-2 receptor (sIL-2R), shed from activated T cells, are (IVIG) (see later discussion), (c) the ability of GBS sera to cause
increased in GBS (104107). Levels are maximal at onset and demyelination of nerve in tissue culture and possibly in vivo,
decrease during recovery. Serum levels of IL-2 do not correlate and (d) the demonstration of antibodies against PN constituents
with maximal functional disability. However, concentration of in GBS sera. Whole serum or IgM antibody from some patients
sIL-2R may predict severity (107). Activated T cells may me- with early GBS will produce complement-dependent demyelin-
diate injury in GBS by (a) stimulation of B-cell proliferation ation of PN tissue cultures (122124). Electron microscopic
and antibody production against myelin components, (b) mac- studies have disclosed damage to cultured Schwann cells and
rophage recruitment, (c) activation of the complement system, vesicular myelin degeneration after exposure to GBS sera in
or (d) direct cytotoxic damage of myelin or Schwann cells (97). the absence of leukocytes, lymphocytes, or macrophages (122).
Consistent alterations in T-cell subpopulations have not A lesser degree of in vitro demyelination occurs in PN tissue
been demonstrated (104,108110). Dahle et al. (108) noted culture after exposure to control sera, GBS patients during
normal total lymphocyte counts in acute GBS but a decrease in recovery, or persons with other neurologic diseases (123).
the proportion of CD4 helper/inducer T cells and an increase A humoral mechanism in the pathogenesis of GBS has
in the proportion of CD8 cytotoxic/suppressor T cells. Within focused on the detection of antibodies to PN constituents, espe-
the CD4 population, helper/inducer cells predominated over cially gangliosides and related glycolipids. Complement-fixing
suppressor/inducer cells, known to be decreased in diseases antibodies directed against PN myelin are demonstrable in the
of autoimmune mechanism. Winer et al. (110) demonstrated sera of some persons with early GBS (120,125). AntiPN-myelin
decreased numbers of CD8 T cells in GBS patients with sero- antibodies are highest at presentation, significantly increased
logic evidence of recent C. jejuni infection and decreased num- over levels from normal and diseased controls, and decrease
bers of CD4 T cells in GBS patients with serologic evidence with improving clinical symptomatology (125). Production of
of recent CMV infection. Specific activation of T cells against antiPN-myelin antibodies in GBS is predominantly extrathe-
myelin proteins has not been consistently observed with the cal, although measurable amounts are present in CSF (126).
sera from those with GBS. Two series (111,112) found no evi- In GBS, activated complement components are detectable
dence of T-cell sensitivity to myelin P2 protein. Several reports in serum and CSF. AntiPN-myelin antibodies peak prior to
have demonstrated T-cell clonal proliferation against proteins activated complement levels, providing evidence of an antibody-
P2 and P0 and their peptides (113115). However, similar mediated complement attack on PN myelin (127,128).
results have been obtained in normal controls and controls Characterization of antiPN-myelin antibodies has disclosed
with other neurologic diseases (113,115). increased levels of circulating antibody to galactocerebroside
Macrophage involvement in GBS is evidenced by demon- (GalC) (110) and P2 protein (112). Antibody levels to these my-
stration of macrophages in affected nerves in GBS pathologic elin constituents were low and, in the case of P2 protein, only
specimens (32,8588), electron microscopic observation of mac- marginally increased over those of control sera. Koski et al.
rophage-mediated myelin stripping and phagocytosis (8994), (129) discovered antiPN-neutral glycolipid antibodies in GBS
and indications of macrophage activation and their possible sera. Antigenic cross reactivity was demonstrated between this
role as antigen presenters through the surface expression of PN neutral glycolipid and Forssman antigen, a component of
MHC class II antigens. Neopterin is produced by macrophages some viral and bacterial cell membranes.
in response to T-cell activation. Serum levels of neopterin are in- Gangliosides are sialylated glycosphingolipids located on
creased in GBS and fall as clinical involvement improves (116). mammalian cell plasma membranes. Ganglioside function is not
In vitro, monocytes from GBS patients have demonstrated a known precisely, but gangliosides are implicated in cell differ-
greater ability to generate reactive oxygen species, indicative of entiation, cellcell interactions, and receptor functions such as
cytokine activation, than monocytes from controls (104). Thus, binding of viruses, bacterial toxins, hormones, growth factors,
available data indicate that macrophage activation, probably by and interferons (130). IgG and IgA antibodies to GM1, GD1b,
activated T cells or their byproducts, occurs in GBS. and GQ1b have been shown to activate leukocytes, which may
MHC class II marker expression is necessary for antigen pre- contribute to the pathogenesis of GBS (131). The major gangli-
sentation. Invading, actively phagocytic macrophages in GBS osides of PN are GM3 and GD3, and the major ganglioside of
express MHC class II markers on their surface (86,88,89,95). PN myelin is LM1. GM1, although a major component of CNS
Although MHC class II marker has been detected on the Schwann myelin, is only a minor constituent of PN. GM1 has been local-
cell surface in GBS (98), these markers have also been observed ized to both nerve plasma membrane and the myelin sheath and
on the Schwann cell surface in degenerative, toxic, and metabolic appears to accumulate in the paranodal region (132). Antibody
neuropathies (117). Tumor necrosis factor- (TNF-) is a cytokine to gangliosides is present in low amounts in normal human
elaborated by both activated T cells and macrophages. Increased plasma (133). In 1988, Ilyas et al. (134) detected significantly
serum levels of TNF- occur in GBS (118). Although TNF- may increased levels of antibodies to the gangliosides GD1a, GD1b,
play a role in the immune-mediated injury in GBS, recombinant and GT1b and to two associated glycolipids in serum from pa-
TNF- alone will not produce myelin destruction in vitro (119). tients with acute GBS compared to controls.
The mechanism by which activated T cells and macro- High IgG antibody titers against GQ1b, a minor ganglioside
phages gain access to PN is not fully understood but appar- of PN that has been localized to the paranodal region of human
ently involves two adhesion molecules, E-selectin (ELAM-1) ocular motility nerves, was detected in 90% of 41 persons with
and intercellular adhesion molecule-1 (ICAM-1) (97). ELAM-1 Fisher syndrome (FS) (135137), a GBS clinical variant manifest-
is expressed on endothelial cells activated by IL-2, TNF-, and ing primarily as ophthalmoplegia and ataxia. Anti-GQ1b anti-
endotoxin. It aids in the binding of lymphocytes, monocytes, body decreases as clinical symptoms improve and may be absent
eosinophils, and neutrophils to the blood vessel wall. Like IL-2 in people with mild FS (135137). High anti-GQ1b antibody
receptor, ELAM-1 enters the circulation and can be measured titers have also been demonstrated in Bickerstaff brainstem en-
in serum. Elevated ELAM-1 serum levels have been observed cephalitis, a syndrome that bears some clinical resemblances to
acutely in GBS (120,121). FS (138), and in typical GBS with prominent ophthalmoplegia

Scheld_Ch20.indd 304 2/21/14 5:40 PM


Chapter 20: Guillain-Barr Syndrome 305

(137). High-titer anti-GQ1b antibody is not usually detectable The IgG subclasses 1 and 3 predominate in IgG antibodies
in typical GBS lacking ophthalmoplegia or in normal controls to both GM1 and GQ1b. These subclasses are usually seen in
(137). The pathogenetic role of anti-GQ1b antibodies in FS is immune responses to protein antigens rather than to carbohy-
not known. Lipopolysaccharide of C. jejuni isolated from FS drates such as gangliosides. This suggests that the primary anti-
does cross-react with epitopes of anti-GQ1b antibody (56). body response in GBS is against a protein. The antibody simply
Subsequent to the original observation of high antiganglio- cross-reacts with gangliosides (161). The IgG subclass may be
side antibody titers in GBS (134), antiganglioside antibodies a predictor of severity and outcome. Jacobs et al. reported pa-
(AGA) have been detected in 10% to 60% of patients with tients with only IgG1 AGA had a worse prognosis compared
GBS (61,97,139145). The AGA titers may correlate with to patients with both IgG1 and IgG3 AGA. This may be in part
GBS severity and decrease with clinical improvement (143). due to the longer serum half-life of IgG1 (21 days) compared to
Antibodies to many known gangliosides and unidentified glyco- the relative shorter half-life of IgG3 (7 days) (166).
lipid fractions have been demonstrated in association with GBS,
but antibody to GM1 is most frequently cited, especially in cases
with mainly motor symptoms (139,141144,146). Both IgM CLINICAL MANIFESTATIONS
and IgG antibodies to gangliosides are present, but IgM antibod-
ies predominate early (142). The early presence of IgG or IgA An ad hoc committee of the NINCDS proposed diagnostic cri-
antibodies to GM1 has been associated with severe, prolonged teria for GBS in 1978 (3). These criteria, further reaffirmed and
GBS (139) and may predict poor recovery (141,144,147149). elaborated by Asbury in 1980 (167) and Asbury and Cornblath
AGA have also been detected in individuals with hyperreflexia in 1989 (168), are summarized in the following paragraphs
and impaired central motor conduction in association with along with additional comments. Electrodiagnostic criteria are
acute paralysis, acute ataxia, or chronic paralysis (150). discussed in the subsequent section.
Anti-GM1 antibodies have been reported in up to 58% of pa- The diagnosis of GBS is based on (a) progressive motor
tients with GBS showing serologic evidence of preceding C. jejuni weakness of more than one extremity and (b) areflexia,
infection (139,149). Yuki et al. (62) described anti-GM1 anti- although proximal hyporeflexia with distal areflexia may be
body in association with C. jejuni infection in severe GBS with seen. Weakness may precede reflex changes by 2 or 3 days (168)
electrophysiologic evidence of primary axonal degeneration and and typically spreads from the lower extremities to involve the
poor recovery. Immunogenic cross-reactivity between GM1 and upper extremities, trunk, and cranial nerves (6,7). Weakness
C. jejuni isolates from GBS patients has been described (53,57). usually ascends from the thighs to the upper arms, although
GM1 antibodies recognize a carbohydrate sequence that is pres- hands and feet are initially involved in some cases (6). Muscle
ent in several gangliosides and on bacterial cell walls. GM1 gan- wasting may eventually occur in 50% of patients (168).
gliosides may also be important for nerve growth factor signaling. Features strongly supportive of the diagnosis include
Tanaka et al. reported that GM1 antibodies interfered with (a) rapid progression of motor weakness reaching a plateau
the neurotrophic action of nerve growth factor and TrkA auto- in 4 weeksin one study, the average time elapsing from
phosphorylation signaling in PC12 cells, a sympathetic nerve cell onset of weakness to its maximum was 12 days, with 98%
line (151). Nakatani et al. reported GM1 antibodies of patients of patients reaching maximal weakness by 4 weeks (25);
with AMAN inhibit the current action of Cav2.1 (P/Q-type) (b) relative symmetry of motor weakness; (c) mild sensory
voltage-dependent calcium channel in Purkinje cells of rats (152). symptoms. GBS is often heralded by paresthesias in the toes
Elevated titers to GM1 and GD1b occur in C. jejuni infec- and fingers (6). Sensory symptoms are eventually present in
tion without GBS (153). However, research now shows that 70% of GBS patients (9,11,13). Subjective sensory complaints
elevated antibodies to GM1, GD1a, GM1b, and GalNAcGD1a commonly exceed objective sensory findings (6,11). When
occur with higher frequency in AMAN, whereas elevated GQ1b, objective sensory loss occurs, proprioception and vibration
GT1a, GD3, and GD1b antibodies occur in the majority of FS are primarily involved (169), presumably because the fibers
(154). In 32 Japanese children with GBS, Nishimoto et al. (155) involved in their conduction are myelinated to a greater extent
reported the frequency of IgG antibodies to gangliosides: GM1 than are the fibers conducting temperature, pinprick, and light
(34%), GM1b (22%), GD1a (25%), and GalNAc-GD1a (13%). touch; (d) cranial nerve involvement. Data in one study of
In addition, antibodies to ganglioside complexes (GSCs) are 100 consecutive GBS patients revealed facial palsy (frequently
associated with severe GBS. Patients with GBS requiring mechan- bilateral) in 53% of patients, bulbar weakness in 13%, and
ical ventilation may have antibodies to GD1a/GD1b and GD1b/ extraocular muscle palsy in 9% (169). Rarely, all brainstem re-
GT1b complexes, and patients with FS may have antibodies to flexes may be lost simulating brain death; (e) recovery ensues.
GQ1b/GM1 and GQ1b/GD1a GSC (156,157,158,159). GM1/ This usually begins 2 to 4 weeks after cessation of progression
GalNAc-GD1a GSC have also been reported in 10 patients with but may be delayed months (6). Little improvement is seen
GBS (160). Other antibodies associated with GBS have been iden- after 1.5 to 2 years (25,29); and (f) autonomic dysfunction.
tified (Table 20.4) (65,162164). Immunogenic cross reactivity Sympathetic and parasympathetic systems may be involved,
between GalC and M. pneumoniae has also been described (165). producing cardiovascular, gastrointestinal, urinary, pupillary,
and sudomotor symptoms. Autonomic involvement is recog-
nized in about 20% of GBS cases, although the actual figure
TA B L E 2 0 . 4 is probably much higher (13,20,21). Minor cardiovascular
abnormalities in GBS include sinus tachycardia and postural
ANTIBODIES ASSOCIATED WITH GUILLAIN-BARR hypotension. Malignant arrhythmias secondary to autonomic
SYNDROME dysfunction remain a leading cause of death in GBS patients.
Hypertension, hypotension, and sensitivity to vasoactive drugs
GD1a GM1 also occur. Urinary retention, constipation, gastroparesis, and
GD1b GM1b loss of pupillary responses are less frequent (25). Disordered
sympathetic function can produce both excessive sweating and
GD3 GT1a
anhidrosis (170). Impotence may persist in a few men (6).
LM1 GT1b Variant clinical features include (a) fever at the onset of
GQ1b GalNAcGD1a neurologic symptoms; (b) severe sensory loss with pain. Severe
pain may be the presenting complaint in some GBS patients,

Scheld_Ch20.indd 305 2/21/14 5:40 PM


306 Part II: Viral Infections and Related Disorders

particularly in children, in whom the diagnosis of GBS may Clinical and laboratory features that should alert one to the
be difficult initially. Painful symptoms may include myalgias possibility of alternative diagnoses are (a) conspicuous asym-
and arthralgias, paresthesias and dysesthesias, meningismus, metry of extremity weakness; (b) persistent bladder or bowel
radicular and back pain, and abdominal discomfort (9,25,171); dysfunction or bladder or bowel dysfunction at onset; (c) more
(c) progression may continue beyond 4 weeks or show a minor than 50 mononuclear leukocytes/mm3 in CSF or the presence
relapse. Approximately 3% to 9% of patients will have one of polymorphonuclear leukocytes in CSF; (d) protein concen-
or more relapses (9,32); (d) no significant recovery. Complete tration greater than 2.5 g/dL; and (e) distinct sensory level.
recovery occurs in approximately 60% of patients, but mild The differential diagnosis of GBS includes ADEM present-
disability persists in 30%, and significant handicap remains in ing transverse myelitis, or spinal cord compression presenting
5% to 10% (6,9,25,169). Children experience a better outcome as flaccid paralysis, myasthenia gravis, diphtheritic neuropa-
(2023); (e) sphincteric dysfunction may occur; urinary reten- thy, vasculitic neuropathy, lead neuropathy, and arsenic poi-
tion is rare but may occur secondary to autonomic involve- soning. Weakness related to enteroviral infection (including
ment (169); and (f) CNS involvement. Although controversial, poliovirus), botulism, and hysterical paralysis may occasion-
occasional GBS patients demonstrate cerebellar ataxia, exten- ally be confused with GBS. Weakness secondary to enteroviral
sor plantar responses, and ill-defined sensory levels (25). infection is associated with a CSF pleocytosis and electrophys-
CSF findings that are strongly supportive of the diagno- iologic evidence of motor neuron injury. A history of prodro-
sis include (a) elevated CSF protein. Increases in CSF protein mal constipation and poor feeding in infants or the ingestion
concentrations are thought to arise from the severe nerve root of contaminated food in adults, preservation of reflexes in
involvement. CSF protein elevations are usually not present until some patients, and the presence of a presynaptic defect on elec-
after 1 week of symptoms and peak during the nadir and early trophysiologic studies can differentiate weakness secondary to
recovery period (6,11). CSF protein concentrations may reach botulism from GBS. Acute intermittent porphyria is excluded
levels of 2 g/dL, though much lower levels are typical (11,14,32); by the absence of urinary excretion of porphobilinogen and
a CSF protein concentration greater than 2.5 g/dL should alert -aminolevulinic acid. The presence of cells in the CSF should
one to the possibility of cord compression (6). No correlation alert one to the possibility of lymphomatous, carcinomatous,
exists between protein values and mode of onset or progression, or sarcoid meningitis as well as Lyme disease. Extremity and
severity, or outcome of GBS (9,25,32). However, elevated heavy respiratory muscle weakness mimicking GBS can occur 12 to
chain neurofilament (NfH), a marker of axonal damage, may in- 96 hours after organophosphate exposure (176), secondary to
dicate a worse prognosis (172). Papilledema noted in some cases blockade of nicotinic receptors by excess acetylcholine. Basilar
of GBS has been attributed to impaired CSF absorption second- artery occlusion and tick paralysis may mimic MFS (6).
ary to increased protein levels (13,25,169). However, papilledema
and increased intracranial pressure have been seen in GBS with
normal CSF protein, and papilledema is not consistently present ELECTROPHYSIOLOGIC FEATURES
in patients with extremely high protein values (32); and (b) ten or
fewer mononuclear leukocytes per cubic millimeter. Electrophysiologic studies have significantly increased our
CSF variants include (a) normal CSF protein in the period understanding of GBS and improved diagnostic accuracy.
1 to 10 weeks following the onset of neurologic symptoms The symptoms and signs in GBS are attributed to abnormali-
normal CSF protein values may be present in 20% of GBS pa- ties in nerve conduction. Although the diagnostic criteria of
tients (6,9,11,32)and (b) 11 to 50 mononuclear leukocytes GBS described in 1978 (3) report normal studies of nerve
per cubic millimeter. In the presence of HIV infection, CSF conduction in up to 20% of GBS, more recent series have
pleocytosis in GBS is expected. Cornblath et al. (40) noted a identified electrophysiologic abnormalities in 90% to 100%
mean of 23 cells/mm3 in nine HIV-infected persons with GBS. (169,177180). Because of the multifocal nature of pathologic
Clinical variants of GBS include (a) FS with ophthalmo- lesions in GBS, the likelihood of detecting electrophysiologic
plegia, ataxia, and areflexia; (b) polyneuritis cranialis (173); abnormalities increases with the number of nerves tested (177).
(c) pharyngealcervicalbrachial weakness (174); (d) parapare- Generally, three or more motor nerves should be examined
sis with areflexia (32); (e) sensory loss with areflexia (32,168); (168). Early in the course of GBS, nerve conduction may be
(f) pure pandysautonomia (175); (g) pure motor weakness normal or minimally abnormal. Repeat testing may be neces-
(32); (h) AMAN (63); and (i) AMSAN (88). Paraparesis with sary in inconclusive cases. Abnormalities in motor nerve con-
areflexia can be included as a variant of GBS only if spinal duction, sensory nerve conduction, mixed nerve conduction,
cord compression and transverse myelitis have been excluded and late motor responses (H-reflex study and F-wave study)
and electrophysiologic studies are consistent with demyelinat- may occur alone or in any combination.
ing polyneuropathy (32). Sensory loss with areflexia is most In 1990, Asbury and Cornblath (168) proposed electro-
commonly secondary to a neuronopathy associated with diagnostic criteria for demyelinating neuropathies, including
malignancy, particularly small cell carcinoma of the lung and GBS. These criteria require three of the following to be pres-
lymphoma. Rather than a demyelinative process, dorsal roots ent: (a) decreased conduction velocity in two or more motor
are destroyed by intense inflammation (32). Although pure nerves; (b) conduction block or abnormal temporal disper-
pandysautonomia may result from an autoimmune pathogen- sion in one or more motor nervesperoneal nerve between
esis similar to that of GBS, pathologic evidence of demyelin- ankle and below fibular head, median nerve between wrist and
ation is lacking (32,167,175). GBS with motor weakness in elbow, or ulnar nerve between wrist and below elbow; (c) pro-
the absence of subjective or objective sensory loss may occur longed terminal motor latency (TML) in two or more motor
(32). However, such patients should be distinguished from nerves; and (d) absent F-waves or prolonged F-wave latency.
those with AMAN (63), a syndrome clinically similar to a pre- In early GBS, electrophysiologic evidence of demyelination
dominantly motor form of GBS but pathologically and prob- is apparent primarily in proximal nerve plexuses and in the
ably pathogenetically distinct. AMSAN as described by Feasby distal nerve twigs (31,181,182). During the first 5 weeks,
et al. (88) is also pathologically different from GBS and should motor conduction abnormalities are more common than
be considered a separate entity as well. sensory nerve conduction abnormalities (169,178). Slowed
Radiologic studies are generally not helpful in the diagno- motor nerve conduction is present at some point in 60% to
sis of GBS. However, contrast-enhanced magnetic resonance 80% of those with GBS with maximal slowing detected in the
imaging may demonstrate enhancement of nerve roots. third week (167,168,177,180). Electrophysiologic evidence

Scheld_Ch20.indd 306 2/21/14 5:40 PM


Chapter 20: Guillain-Barr Syndrome 307

of motor nerve demyelination (conduction block or temporal Rapidly progressive weakness, respiratory embarrassment or
dispersion) is present in approximately 60% of GBS patients infection, progressive bulbar dysfunction, or cardiovascular
within 2 weeks of illness onset (177,179,183). Markedly abnormalities warrant intensive care monitoring (6,192).
slowed motor conduction velocity has been associated with Respiratory failure occurs in approximately 20%, necessitat-
a poor long-term prognosis (169,182). In a given nerve, con- ing careful respiratory monitoring, especially during the progres-
duction block may be generalized, or predominantly proxi- sive stage of illness. Vital capacity should be measured every 4 to
mal or distal. Abnormal temporal dispersion is evident in only 6 hours, and elective intubation should be performed when the
20% (183). Abnormal TML occurs in 40% of GBS (177,182) vital capacity falls to 15 mL/kg of body weight (6,192195).
and may be present in nerves with normal motor conduction Patients should be observed for clinical signs of respiratory fatigue
velocity between the proximal and distal sites (31). The nadir such as diaphoresis, tachycardia, and paradoxical movement of
of abnormalities of sensory conduction is reached in the fourth abdominal muscles during inspiration. Respiratory rate is vari-
week (169). The delay in sensory electrophysiologic abnor- able in persons with GBS and impending respiratory failure (195).
malities is felt to reflect, in part, secondary involvement of Blood gas studies and oxygen saturation are unreliable predictors
sensory nerves related to intraneural edema accentuated by of respiratory failure. Significant respiratory compromise can be
compression at sites of anatomic vulnerability. present before hypoxia and hypercarbia occur. Oropharyngeal
When the motor nerve is inexcitable, either severe distal de- weakness may warrant earlier intubation (6,192). Newton-John
myelination with complete conduction block or axonal degen- (196) demonstrated fewer pulmonary complications with early
eration is responsible (184,185). Inexcitable motor nerves may assisted ventilation (before the vital capacity fell below 21 mL/
be present in up to 20% of GBS (182). Although some authors kg) compared to late assisted ventilation (after the vital capacity
have reported that inexcitable motor nerves within 2 weeks of fell below 15 mL/kg). The use of immunomodulatory therapies
symptom onset is uniformly associated with a poor long-term (see later discussion) in ventilated GBS patients has decreased the
prognosis (185), Triggs et al. (184) found complete recovery mean time of ventilation to 16 to 24 days from 26 to 48 days
by 1 year in 50% of such individuals. Abnormal temporal dis- (24,197,198). Weaning from mechanical ventilation should not
persion with reduced distal compound motor action potential begin until vital capacity is greater than 7 mL/kg; extubation can
(CMAP) is due to demyelination. Whether secondary to axonal be safely attempted when the vital capacity is 15 mL/kg (197).
loss or severe distal demyelination, the absence or marked re- Because of the prominence of autonomic disturbances in
duction (0% to 20% of the lower limit of normal) of the distal GBS, continuous electrocardiographic and possibly arterial
CMAP amplitude appears to be the strongest electrophysiologic blood pressure monitoring is necessary in those requiring inten-
predictor of prolonged disease and incomplete recovery follow- sive care (6,192,193). The majority of cardiovascular irregu-
ing GBS (31,169,178,182,185). In those with reduced distal larities occur during peak motor weakness, but episodes during
CMAP amplitude, if improvement in CMAP amplitude occurs convalescence have also been reported (170,194,199201).
on sequential studies or if plasma exchange is performed, bet- Cardiovascular dysfunction in GBS include sinus tachycar-
ter outcome results (184). In one series of 23 children in whom dia, postural hypotension, minor ECG changes, hypertension,
reduced distal CMAP amplitude was the most common electro- episodic hypotension, bradyarrhythmia, tachyarrhythmias,
physiologic abnormality, no correlation between distal CMAP and sensitivity to vasoactive drugs (188,194,199,201). Drugs
amplitude and degree of recovery following GBS was evident producing hypotension in GBS include phentolamine, nitro-
(179). However, the absence of a distal CMAP abnormality glycerin, hexamethonium, edrophonium chloride, thiopental
early in GBS does not predict a good outcome (186). sodium, morphine sulfate, and furosemide. Phenylephrine,
Prolonged F-wave latency, indicating abnormal conduction ephedrine, dopamine, and isoproterenol have been associated
in the proximal motor nerve, is the most common electro- with hypertension (170). Carbamazepine precipitated asystole
physiologic abnormality in GBS, occurring in approximately in a man recovering from GBS (200). Atropine and glycopyr-
90% of patients during the course of the illness (169,177). rolate should be used cautiously in GBS because of a risk of
Indeed, prolonged F-wave latency may be the only detectable tachyarrhythmias. Some authors advocate insertion of a car-
electrophysiologic abnormality in some patients (187). F-wave diac pacemaker in any patient with a nonsinus arrhythmia
latency does not predict outcome. (170). Vagal stimulation such as occurs with tracheal suction-
The occurrence of spontaneous fibrillation potentials dur- ing, intubation, extubation, Valsalva, and distention of a hol-
ing EMG in GBS, an indication of muscle denervation, has low viscus may produce hypotension and bradyarrhythmias
also been associated with incomplete recovery following GBS in GBS (192,194). Severe tachyarrhythmias have been termi-
(188,189). It generally appears between the second and fourth nated with verapamil and pindolol (194).
weeks after onset. In some cases, particularly in children, Infection of the urinary tract or lung occurs in almost 25%
the presence of fibrillation on EMG is not predictive of poor of those requiring intensive care (202). Intravenous line or gen-
recovery (178,179). eralized sepsis occurs less commonly. Chest physiotherapy and
In AMAN, sensory and motor nerve conduction velocity incentive spirometry are essential to prevent sputum retention,
and TML are normal, but CMAP is reduced and F-waves are bronchial obstruction, and segmental collapse. In individuals re-
either absent or normal. Unlike GBS with CMAP abnormali- quiring long-term urinary drainage, intermittent catheterization
ties, the outcome in AMAN is good with only mild distal weak- may be less problematic than a chronic indwelling catheter (194).
ness and atrophy present at 1 year (63). Electrophysiologic Gastrointestinal hemorrhage complicates the course of 2%
studies in FS differ from those in typical GBS by predominant to 8% of those in intensive care units (194). The best prophy-
abnormalities in sensory rather than motor nerves (190,191). laxis against gastritis is regular feeding. If feeding is not possible
because of gastric paresis, gastric emptying may be improved
with metoclopramide or cisapride and isotonic, low-fat enteral
TREATMENT formulas, and avoidance of sedatives and narcotics. When
feeding is impractical, sucralfate or magnesium-containing
Despite recent successes of immunomodulatory therapies in antacids may provide adequate gastric bleeding prophylaxis
GBS, meticulous nursing and medical care remain essential. (193). Individuals with GBS have increased nutritional needs
Most individuals with GBS require in-hospital observation. from hypercatabolic states and the metabolic demands of
Patients with mild disease may not require treatment but regenerating myelin and atrophied muscles and must be closely
must be monitored closely until progression has ceased (6). followed (193).

Scheld_Ch20.indd 307 2/21/14 5:40 PM


308 Part II: Viral Infections and Related Disorders

Early physical and occupational therapy in GBS may aid was considered unethical to carry out sham PE, none of the
in the prevention of joint contractures and pressure nerve trials was blinded. These studies included 263 in the PE-treated
palsies and may also hasten the return of limb control, walk- group and 269 treated with conventional methods alone. Only
ing, and balance during the recovery period. Frequent posi- individuals who required assistance walking were included.
tion changes and soft bedding are needed to prevent decubiti The greatest benefit was seen in those who began PE within
(192). Subcutaneous low-dose heparin is commonly employed 1 week of onset. PE 14 days or more after onset observed little
for prophylaxis against deep vein thrombosis (192,194). benefit in those treated. Duration of mechanical ventilation
Successful treatment of pain and dysesthesias associated was significantly shorter, and time to onset of motor recov-
with GBS is highly individualized. Painful symptoms are usu- ery, independent walking, and length of hospitalization were
ally worse at night and may preclude intensive physiotherapy. shorter. Outcome at 6 months and 1 year was better compared
Proper positioning and splinting, socks or gloves, massage, and to those receiving conventional therapy. In one large series (24),
hot or cold packs can provide relief in many (171). Nonsteroidal a 12-year-old boy with GBS died of inflammatory myocarditis
antiinflammatory drugs may alleviate musculoskeletal pain. after two PE treatments. Otherwise, no deaths in the PE-treated
Tricyclic antidepressants and carbamazepine are popular group could be attributed to PE, and the number of deaths in
choices for neurogenic pain but should be used cautiously in each group was similar.
those who have cardiovascular irregularities or urinary retention IVIG has recently proved efficacious in the treatment of
(171,200). Quinine and capsaicin or lidocaine salve have proved GBS. IVIG is pooled human IgG from 5,000 to 10,000 blood
beneficial (171,192). Low-dose methylprednisolone may benefit donors. Side effects of IVIG occur in fewer than 5% of re-
some individuals (192). Narcotic analgesics may be required for cipients and are summarized in Table 20.5 (212). The most
adequate pain relief but should be used cautiously because of common adverse reactions are systemic and include nausea,
secondary hypotension and risk of ileus. Some authors advocate vomiting, headache, fever, chills, myalgias, tachycardia, and
the use of epidurally administered opioids for severe pain (171). hypotension. These reactions are believed to be secondary to
Transcutaneous nerve stimulation has received mixed commen- impurities in the preparation and can usually be managed by
tary as an adjunct to other forms of pain management (171). slowing the infusion rate. Fatal anaphylaxis may occur during
Acute GBS can be a frightening experience. Severe GBS is IVIG administration in IgA-deficient individuals. Preparations
susceptible to anxiety and depression. Reassurance that the with very low levels of IgA should be used in IgA deficiency
disorder is self-limited and that the majority of patients make and in those for whom determination of IgA levels is not pos-
a full recovery is beneficial. Visits from individuals who recov- sible (213). Patients with impaired renal function have rarely
ered from GBS may also be helpful (193,194). developed renal failure following IVIG administration. With
Individuals with GBS are at risk for hyperkalemia fol- current preparations of immune globulin, the remaining
lowing succinylcholine chloride (203). Muscle membrane adverse reactions listed in Table 20.5 are extremely rare (213).
changes after nerve injury result in the proliferation of cholin- The early justification for treatment of GBS with IVIG
ergic receptors. Succinylcholine interacts with the cholinergic was based on its efficacy in other immune-mediated disorders
receptor, producing depolarization with the influx of sodium (214). The mechanism by which IVIG exerts its effectiveness in
and release of potassium. Normally, depolarization does not immune-mediated diseases such as GBS is unknown but could
release enough potassium to increase serum levels. However, be related to the presence of antiidiotypic antibodies (215).
after diffuse PN injury such as occurs in GBS, the increase in Antiidiotypic antibodies are normally circulating antibodies
cholinergic receptors may cause enough potassium release to that bind other antibodies and serve as immunoregulators.
increase serum potassium levels sufficient to produce arrhyth- Treatment with IVIG exposes circulating autoantibodies to
mias and cardiac arrest. It is not known when, if ever, succinyl- antiidiotypic antibodies in IVIG and may result in clearance
choline can be safely administered to patients recovered from of the autoantibodies (215). Other possible beneficial effects
GBS (203). Nondepolarizing neuromuscular blocking agents of IVIG in GBS include inhibition of complement-binding and
may also result in prolonged paralysis (204). cytokine secretion, blockade of antibody receptors, reduction
in autoantibody synthesis, amplification of suppressor cell
activity, and inhibition of lymphocyte proliferation (215).
Immune Modulation Treatment of GBS with IVIG may have several therapeutic
advantages over PE. It is readily available, may be given in any
Immune modulation has now emerged as a promising adjunct
hospital without delay, and can be used in those patients with
to general medical care in GBS. Corticosteroids were used in
cardiovascular instability. Also, IVIG administration does not
the treatment of GBS for many years. Their use was based
remove medications or plasma proteins as PE does and rarely
largely on their antiinflammatory effects and the results of a few
requires placement of a central venous catheter. Treatment
uncontrolled trials. Three randomized, controlled trials showed
cost is comparable to that of PE.
little, if any, benefit from corticosteroids (205207). In fact,
steroids may have a detrimental effect (205). One study com-
paring 21 prednisolone-treated subjects and 19 untreated indi-
viduals demonstrated slower improvement in the treated group TA B L E 2 0 . 5
at 1, 3, and 12 months. Six of the treated groups had disabling
weakness compared to one control. Three relapses occurred in SIDE EFFECTS OF INTRAVENOUS IMMUNE GLOBULIN
the treated group. A nonrandomized study of GBS suggested
Systemic effects
that coincident administration of methylprednisolone and IVIG
was more effective than treatment with IVIG alone (208). Anaphylaxis
Plasma exchange (PE) appeared in the 1980s as a beneficial Hemolytic anemia
treatment for GBS (209). Enthusiasm for its use was based on Viral infection
three sets of observations: (a) case reports showing improve-
Aseptic meningitis
ment following PE; (b) efficacy of PE in chronic inflammatory
demyelinating polyneuropathy; and (c) reports of in vivo demy- Renal failure
elination by GBS sera (25). Four randomized controlled trials Thrombotic events
employing PE have been performed (24,210,211). Because it

Scheld_Ch20.indd 308 2/21/14 5:40 PM


Chapter 20: Guillain-Barr Syndrome 309

TA B L E 2 0 . 6 onset, 29 of 33 examined children had achieved clinical motor


function 1 or 0 (220,223,224,226,227). Two of the remaining
CLINICAL GRADING OF MOTOR FUNCTION four children had no residua at 1 and 2 years (208). These
IN GUILLAIN-BARR SYNDROME reports support the notion that PE can be safely performed in
seriously ill children with GBS and that they benefit from PE
0 Healthy therapy.
1 Minor signs and symptoms but capable of working IVIG is an attractive treatment for childhood GBS for rea-
2 Able to walk 10 meters without assistance sons similar to those in adults. Also, central venous access
required for PE is rarely necessary in IVIG. This is particu-
3 Able to walk 10 meters with assistance
larly advantageous in infants and small children. Sixty-seven
4 Bedridden or chairbound children administered IVIG for GBS have been described
5 Requires assisted ventilation (212,225,227233). All treated children were clinical motor
6 Dead function grade 3 or worse and ranged in age from 1.7 to 16
years. IVIG was administered in doses of 400 mg/kg body
weight on 5 consecutive days, 1 g/kg body weight on 2 con-
secutive days, or a single dose of 2 g/kg body weight. Thirty-
five children were treated within 2 weeks of symptom onset,
A randomized trial comparing IVIG and PE treatment of GBS
1 child was treated 19 (232) and 30 days (230), respectively,
was performed by the Dutch Guillain-Barr Study Group (216).
after onset, and time of treatment was not available in 30 chil-
Individuals with typical GBS of less than 2 weeks duration with
dren (225,229,231). Side effects of IVIG treatment occurred
a clinical motor function grade of 3 or greater (Table 20.6) (24)
in three children. Two complained of headache during treat-
were included. PE of 200 to 250 mL/kg of body weight in five ses-
ment, and one developed macroscopic hematuria. Sixty-five of
sions was performed in 73 subjects, and 400 mg of IVIG/kg body
the 67 children (97%) demonstrated a significant response to
weight was administered on five consecutive days to 74 subjects.
IVIG therapy, usually within 1 week. Improvement attributed
PE was interrupted one or more times in 16% of the PE group,
to IVIG therapy was seen as late as 19 days (212). One of
primarily because of hypotension and difficulties with venous
the nonresponders began IVIG treatment on day 3 of her ill-
access. IVIG infusion was not interrupted in any. Although this
ness but developed respiratory failure on day 7 and required
study did not establish superiority of IVIG over PE in GBS, it
mechanical ventilation for 28 days. She eventually recovered
did demonstrate equal effectiveness. IVIG treatment was signifi-
(227). The course of the other nonresponder was not described,
cantly better in the median time of improvement by one clinical
but all of the children in that series completely recovered in
grade (41 days in the PE group and 27 days in the IVIG group).
4 months (229). Interestingly, both of the nonresponders re-
Significantly fewer subjects receiving IVIG required mechani-
ceived the shorter 2-day course of IVIG rather than the 5-day
cal ventilation compared to the PE-treated group. In the Dutch
course administered in the Dutch Guillain-Barr study (216).
Guillain-Barr study, 8 of 74 (11%) patients treated with IVIG
The results obtained in these 67 IVIG-treated children with
experienced a relapse of symptoms after the treatment ended
GBS are similar to results obtained in the 62 children treated
(217). Clinical deterioration following PE or IVIG may occur be-
with PE. The nonresponder rate in the PE-treated children was
cause early treatment ends before natural progression has ceased
10% versus 3% in the IVIG-treated children. In both groups,
or the pathogenic process is reactivated after treatment is com-
improvement was usually evident within 1 week of begin-
pleted (218). Most authors advocate retreatment only if clinical
ning therapy. Comparisons of the two treatments for time
grade 3 or worse is reached. One trial found no significant differ-
to achieve independent walking and final outcome are not
ences in secondary outcome measures of PE alone compared to
reliable because information is not available for many of the
PE followed by IVIG (219). No data exist to support changing
patients. One of the series compared IVIG administration and
treatment in those who do relapse.
PE in children with GBS (227). This study was not randomized
The treatment of GBS in children is similar to treatment in
or blinded, and the attending neurologist made the treatment
adults. Despite the apparent effectiveness of immunomodula-
choice. Five children underwent PE, and 10 received 1 g of
tory therapies in children with GBS, meticulous nursing and
IVIG/kg body weight on two consecutive days. All children
medical care remain the mainstay of therapy. Children who
receiving PE responded, whereas 9 of 10 children in the IVIG
are too young to perform forced vital capacity measurements
group responded. The PE group improved two functional
must be monitored closely for signs and symptoms of impend-
grades in a mean of 48 days versus 17 days in the IVIG group.
ing respiratory failure.
The outcome at 6 months was similar. Although it appears
No randomized trials have been performed to assess effi-
that IVIG resulted in quicker improvement, the groups were
cacy and safety of PE or IVIG in pediatric GBS. A review of
not equal. Mechanical ventilation was necessary in 60% of
the literature reveals 62 children treated with PE (220227).
the PE group but in only 30% of the IVIG group. Patients in
The children ranged in age from 1.2 to 16 years. Grade 3 or
the IVIG group began treatment one day earlier than those
greater disability (Table 20.6) was present in all treated chil-
in the PE group, and their average age was 2 years younger.
dren. Mild hypotension responsive to fluid administration was
Certainly, one can conclude from the available reports that
the most frequent complication of PE in the 49 children for
both PE and IVIG can favorably alter the course of GBS in
whom information was available (220,221,223,224,226,227).
nonambulatory children, but whether one treatment is supe-
Systemic reactions occurred in four children who received
rior will have to be answered by a randomized trial.
fresh frozen plasma as a replacement fluid. Two children
displayed emotional upset at the end of a PE session, possi-
bly secondary to hypocalcemia (224). Only 1 of 58 reported
children began PE more than 2 weeks after symptom onset PROGNOSIS
(15 days) (224). Fifty-four of the 62 reported children (87%)
receiving PE improved significantly after beginning PE; most Despite the considerable progress made in the treatment of
within 1 week. Two children responded partially, and six did GBS in recent decades, the mortality rate remains 5% to 10%
not respond. One PE nonresponder improved after a single (6,9,14,24,25,28,30,169). In some geographic locations, mor-
IVIG dose of 1.5 g/kg body weight (225). Six months after tality as high as 21% has been recorded (11). Causes of death

Scheld_Ch20.indd 309 2/21/14 5:40 PM


310 Part II: Viral Infections and Related Disorders

directly attributable to GBS include cardiac arrest related to antibodies (139,141,144,147149). Other associated infec-
dysautonomia and respiratory failure (9,14). Mortality sec- tions including HIV are not associated with protracted dis-
ondary to assisted ventilation and paralysis occurs following ease. Increased CSF neuron-specific enolase or S100b protein
accidental disconnection from ventilation, pneumonia, adult levels were seen with longer disease duration (236).
respiratory distress syndrome, septicemia, and hemorrhagic or The most powerful predictor of severe disease and prolonged
thrombotic events. or incomplete recovery appears to be low distal CMAP ampli-
Approximately 60% recover fully. Minor residua such as tude (0% to 20% of normal) (31,169,178,182,183,185,235).
footdrop or mild distal numbness remain in approximately The appearance of denervation on EMG is also predictive of
20%, and 5% to 10% will retain permanent disability poor outcome (188,189). The degree of conduction slowing
(6,9,14,25,33). GBS may recur years after the initial event in does not, however, correlate with maximal clinical disability
5% of individuals (9,23). The lack of uniform recovery has or duration of disease (189).
prompted a search for clinical, laboratory, and electrophysi- In children, GBS may be a more benign disorder com-
ologic features predictive of prognosis. pared to adults, but they often will have severe neuropathic
Most authorities agree that older age, rapid progression pain (237). In Finland (22), 26 of 27 children recovered fully,
to quadriparesis, and need for ventilatory assistance predict and no deaths occurred. No deaths were noted among 72 chil-
severe disease and prolonged or incomplete recovery (6). dren diagnosed in Taiwan, and 75% recovered completely by
Underlying malignancy may also be predictive of poor out- 6 months (23). In Paraguay, 80% achieved full recovery (21).
come. Treatment with PE has a beneficial effect on duration of Conflicting results have been observed with distal CMAP am-
disease and speed of recovery but may not influence the final plitude in children with GBS. Children with inexcitable motor
degree of recovery in GBS (210,211). However, the French nerves or severely reduced distal CMAP (10% of lower limit
Cooperative Group demonstrated a significant increase in the of normal) were more likely to require ventilation and a lon-
percentage of fully recovered GBS patients in the PE versus ger time to become ambulatory than were children with dis-
conventional therapy groups (71% vs. 52%). PE did not influ- tal CMAP amplitude greater than 10% of the lower limit of
ence the incidence of severe residual disability following GBS normal (31). Bradshaw and Jones (179) found no difference
(234). The Dutch Guillain-Barr study found advanced age to between children with reduced distal CMAP amplitude and
be the only clinical predictor of slowed or incomplete recovery other children with GBS. All the children recovered completely.
(216). However, older age has not been associated with poor
outcome in several series (13,27,28,48,235). Beghi et al. (9)
found an association between age and GBS severity only in SUMMARY AND FUTURE
persons older than 70 years of age, and Winer et al. (169) DIRECTIONS
noted poor outcome more often in those older than 40 years
of age. GBS is a demyelinating disorder of PN occurring in all ethnic
In one series, 50% of GBS patients with rapid progression groups and geographic locations. Men and older individuals
to quadriparesis in 2 to 5 days were nonambulatory 2 years appear to be more frequently affected. A variety of preceding
after onset (29). Loss of ambulation in 7 days was predictive events may incite GBS, but minor infections are implicated
of poor outcome in the large multicentered trial comparing most often. Recognition of C. jejuni infection in association
PE and conventional therapy of GBS (235). However, other with GBS, in particular AMAN and FS, has improved our un-
series have not related rapid progression to outcome (9). derstanding of its unique pathogenesis. Both T-cellmediated
A prolonged plateau period before onset of recovery (23 days) and humoral immune responses appear to play important
has also correlated with incomplete recovery (48). roles in the genesis of GBS. The significance of the relation-
The need for ventilatory support may be a negative prog- ship between AGA and GBS has not been fully elucidated.
nostic feature in GBS (6,48,169,235), but Hankey (13) found Electrophysiology is important in both the diagnosis and prog-
no difference in outcome among individuals requiring ventila- nosis of GBS. Despite PE or IVIG treatment of GBS, 5% to
tion shorter or longer than 28 days. 10% of affected individuals will retain permanent disability,
Laboratory features predictive of severe disease and incom- and 5% to 10% will die. With improved understanding of
plete recovery include serologic evidence of C. jejuni infec- the varied causes and pathogenesis of GBS, new and disease-
tion (26,46,59,61) and the early presence of IgG or IgA AGA specific therapies should be developed.

References
1. Hartung HP, Kieseier BC, Kiefer R. Progress in Guillain-Barr syndrome. 9. Beghi E, Kurland LT, Mulder DW, et al. Guillain-Barr syndrome.
Curr Opin Neurol. 2001;14:597604. Clinicoepidemiologic features and effect of influenza vaccine. Arch Neurol.
2. Hartung HP, Willison HJ, Kieseier BC. Acute immunoinflammatory 1985;42:10531057.
neuropathy: update on Guillain-Barr syndrome. Curr Opin Neurol. 10. Larsen JP, Kvale G, Nyland H. Epidemiology of the Guillain-Barr syndrome in
2002;15:571577. the county of Hordaland, Western Norway. Acta Neurol Scand. 1985;71:4347.
3. Criteria for diagnosis of Guillain-Barr syndrome. Ann Neurol. 1978;3: 11. Congia S, Melis M, Carboni MA. Epidemiologic and clinical features of the
565566. Guillain-Barr syndrome in Sardinia in the 19611980 period. Acta Neurol
4. Landry O. Note sur la paralysie ascendante aigue. Gaz Hebd Med Chir. (Napoli). 1989;11:1520.
1859;6:472474, 486, 488. 12. Jiang GX, Cheng Q, Ehrnst A, et al. Guillain-Barr syndrome in Stockholm
5. Osler W. The Principles and Practice of Medicine. New York: Appleton; County, 19731991. Eur J Epidemiol. 1997;13:2532.
1892:777778. 13. Hankey GJ. Guillain-Barr syndrome in Western Australia, 19801985.
6. Ropper AH. The Guillain-Barr syndrome. N Engl J Med. 1992;326: Med J Aust. 1987;146:130133.
11301136. 14. Radhakrishnan K, el-Mangoush MA, Gerryo SE. Descriptive epidemiol-
7. Guillain G, Barr JA, Strohl A. Sur on syndrome de radiculonevrite avec ogy of selected neuromuscular disorders in Benghazi, Libya. Acta Neurol
hyperalbuminose due liquide cephalorachidien sans reaction cellulaire: Scand. 1987;75:95100.
remarques sur les caracteres cliniques et graphiques des reflexes tendineux. 15. Govoni V, Granieri E, Manconi M, et al. Is there a decrease in Guillain-
Bull Soc Med Hop Paris. 1916;40:14621470. Barr syndrome incidence after bovine ganglioside withdrawal in Italy?
8. Alter M. The epidemiology of Guillain-Barr syndrome. Ann Neurol. A population-based study in the local health district of Ferrara, Italy.
1990;27(suppl):S7S12. J Neurol Sci. 2003;216:99103.

Scheld_Ch20.indd 310 2/21/14 5:40 PM


Chapter 20: Guillain-Barr Syndrome 311

16. McLean M, Duclos P, Jacob P, et al. Incidence of Guillain-Barr syndrome 50. Fujimoto S, Yuki N, Itoh T, et al. Specific serotype of Campylobacter jejuni
in Ontario and Quebec, 19831989, using hospital service databases. associated with Guillain-Barr syndrome. J Infect Dis. 1992;165:183.
Epidemiology. 1994;5:443448. 51. Kuroki S, Haruta T, Yoshioka M, et al. Guillain-Barr syndrome associated
17. Cuadrado JI, de Pedro-Cuesta J, Ara JR, et al. Guillain-Barr syndrome in with Campylobacter infection. Pediatr Infect Dis J. 1991;10:149151.
Spain, 19851997: epidemiological and public health views. Eur Neurol. 52. Yuki N, Sato S, Fujimoto S, et al. Serotype of Campylobacter jejuni, HLA,
2001;46:8391. and the Guillain-Barr syndrome. Muscle Nerve. 1992;15:968969.
18. Mishu B, Ilyas AA, Koski CL, et al. Serologic evidence of previous 53. Yuki N, Taki T, Inagaki F, et al. A bacterium lipopolysaccharide that elicits
Campylobacter jejuni infection in patients with the Guillain-Barr syn- Guillain-Barr syndrome has a GM1 ganglioside-like structure. J Exp Med.
drome. Ann Intern Med. 1993;118:947953. 1993;178:17711775.
19. Nachamkin I, Barbosa PA, Ung H, et al. Patterns of Guillain-Barr in chil- 54. Enders U, Karch H, Toyka KV, et al. The spectrum of immune responses to
dren: results from a Mexican population. Neurology. 2007;69:16651671. Campylobacter jejuni and glycoconjugates in Guillain-Barr syndrome and
20. Rantala H, Cherry JD, Shields WD, et al. Epidemiology of Guillain-Barr in other neuroimmunological disorders. Ann Neurol. 1993;34:136144.
syndrome in children: relationship of oral polio vaccine administration to 55. Rees JH, Gregson NA, Rajagopalan G, et al. Absorption of antigan-
occurrence. J Pediatr. 1994;124:220223. glioside Gm1 antibodies in a patient with Guillain-Barr syndrome by
21. Hart DE, Rojas LA, Rosario JA, et al. Childhood Guillain-Barr syndrome Campylobacter jejuni. Ann Neurol. 1994;36:290.
in Paraguay, 1990 to 1991. Ann Neurol. 1994;36:859863. 56. Yuki N, Taki T, Takahashi M, et al. Molecular mimicry between GQ1b
22. Rantala H, Uhari M, Niemela M. Occurrence, clinical manifestations, and ganglioside and lipopolysaccharides of Campylobacter jejuni isolated from
prognosis of Guillain-Barr syndrome. Arch Dis Child. 1991;66:706708; patients with Fishers syndrome. Ann Neurol. 1994;36:791793.
discussion 708709. 57. Aspinall GO, Fujimoto S, McDonald AG, et al. Lipopolysaccharides from
23. Hung KL, Wang HS, Liou WY, et al. Guillain-Barr syndrome in children: Campylobacter jejuni associated with Guillain-Barr syndrome patients
a cooperative study in Taiwan. Brain Dev. 1994;16:204208. mimic human gangliosides in structure. Infect Immun. 1994;62:21222125.
24. Plasmapheresis and acute Guillain-Barr syndrome. The Guillain-Barr 58. Enders U, Karch H, Toyka KV, et al. Reply. Ann Neurol. 1994;35:249.
syndrome Study Group. Neurology. 1985;35:10961104. 59. Kaldor J, Speed BR. Guillain-Barr syndrome and Campylobacter jejuni:
25. Lffel NB, Rossi LN, Mumenthaler M, et al. The Landry-Guillain-Barr a serological study. Br Med J (Clin Res Ed). 1984;288:18671870.
syndrome. Complications, prognosis and natural history in 123 cases. 60. Tauxe RV, Hargrett-Bean N, Patton CM, et al. Campylobacter isolates in the
J Neurol Sci. 1977;33:7179. United States, 19821986. MMWR CDC Surveill Summ. 1988;37:113.
26. Winer JB, Hughes RA, Anderson MJ, et al. A prospective study of 61. Vriesendorp FJ, Mishu B, Blaser MJ, et al. Serum antibodies to GM1,
acute idiopathic neuropathy. II. Antecedent events. J Neurol Neurosurg GD1b, peripheral nerve myelin, and Campylobacter jejuni in patients with
Psychiatry. 1988;51:613618. Guillain-Barr syndrome and controls: correlation and prognosis. Ann
27. Sridharan GV, Tallis RC, Gautam PC. Guillain-Barr syndrome in the Neurol. 1993;34:130135.
elderly. A retrospective comparative study. Gerontology. 1993;39:170175. 62. Yuki N, Yoshino H, Sato S, et al. Acute axonal polyneuropathy associated
28. Winner SJ, Evans JG. Guillain-Barr syndrome in Oxfordshire: clinical with anti-GM1 antibodies following Campylobacter enteritis. Neurology.
features in relation to age. Age Ageing. 1993;22:164170. 1990;40:19001902.
29. Ropper AH. Severe acute Guillain-Barr syndrome. Neurology. 1986;36: 63. McKhann GM, Cornblath DR, Griffin JW, et al. Acute motor axonal neu-
429432. ropathy: a frequent cause of acute flaccid paralysis in China. Ann Neurol.
30. de Jager AE, Sluiter HJ. Clinical signs in severe Guillain-Barr syndrome: 1993;33:333342.
analysis of 63 patients. J Neurol Sci. 1991;104:143150. 64. Nachamkin I, Liu J, Li M, et al. Campylobacter jejuni from patients with
31. Reisin RC, Cersosimo R, Garcia Alvarez M, et al. Acute axonal Guillain- Guillain-Barr syndrome preferentially expresses a GD(1a)-like epitope.
Barr syndrome in childhood. Muscle Nerve. 1993;16:13101316. Infect Immun. 2002;70:52995303.
32. Arnason BGW, Soliven B. Acute inflammatory demyelinating polyra- 65. Willison HJ, Yuki N. Peripheral neuropathies and anti-glycolipid anti-
diculoneuropathy. In: Dyck PJ, Thomas PK, Griffin JW, et al. Peripheral bodies. Brain. 2002;125:25912625.
Neuropathy. Philadelphia: WB Saunders; 1993:14371497. 66. Hillert J, Osterman PO, Olerup O. No association with HLA-DR, -DQ or
33. Boucquey D, Sindic CJ, Lamy M, et al. Clinical and serological studies in a series -DP alleles in Guillain-Barr syndrome. J Neuroimmunol. 1991;31:6772.
of 45 patients with Guillain-Barr syndrome. J Neurol Sci. 1991;104:5663. 67. Winer JB, Briggs D, Welsh K, et al. HLA antigens in the Guillain-Barr
34. Ravi V, Taly AB, Shankar SK, et al. Association of Japanese encephalitis syndrome. J Neuroimmunol. 1988;18:1316.
virus infection with Guillain-Barr syndrome in endemic areas of south 68. Godschalk PC, Kuijf ML, Li J, et al. Structural characterization of
India. Acta Neurol Scand. 1994;90:6772. Campylobacter jejuni lipooligosaccharide outer cores associated with
35. Merelli E, Sola P, Faglioni P, et al. Newest human herpesvirus (HHV-6) in Guillain-Barr and Miller Fisher syndromes. Infect Immun. 2007;75:
the Guillain-Barr syndrome and other neurological diseases. Acta Neurol 12451254.
Scand. 1992;85:334336. 69. Yuki N. Campylobacter sialyltransferase gene polymorphism directs clini-
36. Dowling PC, Cook SD. Role of infection in Guillain-Barr syndrome: cal features of Guillain-Barr syndrome. J Neurochem. 2007;103:150158.
laboratory confirmation of herpesviruses in 41 cases. Ann Neurol. 1981; 70. Yuki N, Shahrizaila N. How do we identify infectious agents that trig-
9(suppl):4455. ger Guillain-Barr syndrome, Fisher syndrome and Bickerstaff brainstem
37. Baldwin RT, Pierce RR, Frazier OH. Guillain-Barr syndrome after heart encephalitis? J Neurol Sci. 2011;302:15.
transplantation. J Heart Lung Transplant. 1992;11:817819. 71. Mori M, Kuwabara S, Miyake M, et al. Haemophilus influenzae infection
38. Myers SE, Williams SF. Guillain-Barr syndrome after autologous bone and Guillain-Barr syndrome. Brain. 2000;123(pt 10):21712178.
marrow transplantation for breast cancer: report of two cases. Bone 72. Mori M, Kuwabara S, Miyake M, et al. Haemophilus influenzae has a GM1
Marrow Transplant. 1994;13:341344. ganglioside-like structure and elicits Guillain-Barr syndrome. Neurology.
39. Thornton CA, Latif AS, Emmanuel JC. Guillain-Barr syndrome associated 1999;52:12821284.
with human immunodeficiency virus infection in Zimbabwe. Neurology. 73. Chiba S, Sugiyama T, Matsumoto H, et al. Antibodies against Helicobacter
1991;41:812815. pylori were detected in the cerebrospinal fluid obtained from patients with
40. Cornblath DR, McArthur JC, Kennedy PG, et al. Inflammatory demyelin- Guillain-Barr syndrome. Ann Neurol. 1998;44:686688.
ating peripheral neuropathies associated with human T-cell lymphotropic 74. Chiba S, Sugiyama T, Yonekura K, et al. An antibody to VacA of
virus type III infection. Ann Neurol. 1987;21:3240. Helicobacter pylori in cerebrospinal fluid from patients with Guillain-Barr
41. Berger JR, Difini JA, Swerdloff MA, et al. HIV seropositivity in Guillain- syndrome. J Neurol Neurosurg Psychiatry. 2002;73:7678.
Barr syndrome. Ann Neurol. 1987;22:393394. 75. Hemachudha T, Griffin DE, Chen WW, et al. Immunologic studies of rabies
42. Ahmed S, Libman R, Wesson K, et al. Guillain-Barr syndrome: an unusual vaccination-induced Guillain-Barr syndrome. Neurology. 1988;38:375378.
presentation of West Nile virus infection. Neurology. 2000;55:144146. 76. Breman JG, Hayner NS. Guillain-Barr syndrome and its relationship to
43. Centers for Disease Control and Prevention. Acute flaccid paralysis syn- swine influenza vaccination in Michigan, 19761977. Am J Epidemiol.
drome associated with West Nile virus infectionMississippi and Louisiana, 1984;119:880889.
JulyAugust 2002. MMWR Morb Mortal Wkly Rep. 2002;51:825828. 77. Kurland LT, Wiederholt WC, Kirkpatrick JW, et al. Swine influenza
44. Kanjalkar M, Karnad DR, Narayana RV, et al. Guillain-Barr syndrome vaccine and Guillain-Barr syndrome. Epidemic or artifact? Arch Neurol.
following malaria. J Infect. 1999;38:4850. 1985;42:10891090.
45. Sokrab TE, Eltahir A, Idris MN, et al. Guillain-Barr syndrome following 78. Safranek TJ, Lawrence DN, Kurland LT, et al. Reassessment of the asso-
acute falciparum malaria. Neurology. 2002;59:12811283. ciation between Guillain-Barr syndrome and receipt of swine influenza
46. Rhodes KM, Tattersfield AE. Guillain-Barr syndrome associated with vaccine in 19761977: results of a two-state study. Expert Neurology
Campylobacter infection. Br Med J (Clin Res Ed). 1982;285:173174. Group. Am J Epidemiol. 1991;133:940951.
47. Hartung HP, Pollard JD, Harvey GK, et al. Immunopathogenesis and treatment 79. Marks JS, Halpin TJ. Guillain-Barr syndrome in recipients of A/New
of the Guillain-Barr syndromePart II. Muscle Nerve. 1995;18:154164. Jersey influenza vaccine. JAMA. 1980;243:24902494.
48. Gruenewald R, Ropper AH, Lior H, et al. Serologic evidence of 80. Johnson DE. Guillain-Barr syndrome in the US Army. Arch Neurol.
Campylobacter jejuni/coli enteritis in patients with Guillain-Barr syn- 1982;39:2124.
drome. Arch Neurol. 1991;48:10801082. 81. Nachamkin I, Shadomy SV, Moran AP, et al. Anti-ganglioside antibody
49. Kuroki S, Saida T, Nukina M, et al. Campylobacter jejuni strains from patients induction by swine (A/NJ/1976/H1N1) and other influenza vaccines:
with Guillain-Barr syndrome belong mostly to Penner serogroup 19 and con- insights into vaccine-associated Guillain-Barr syndrome. J Infect Dis.
tain beta-N-acetylglucosamine residues. Ann Neurol. 1993;33:243247. 2008;198:226233.

Scheld_Ch20.indd 311 2/21/14 5:40 PM


312 Part II: Viral Infections and Related Disorders

82. Kinnunen E, Farkkila M, Hovi T, et al. Incidence of Guillain-Barr syn- 111. Zweiman B, Rostami A, Lisak RP, et al. Immune reactions to P2 protein
drome during a nationwide oral poliovirus vaccine campaign. Neurology. in human inflammatory demyelinative neuropathies. Neurology. 1983;33:
1989;39:10341036. 234237.
83. Klingon GH. The Guillain-Barr syndrome associated with cancer. 112. Hughes RA, Gray IA, Gregson NA, et al. Immune responses to myelin
Cancer. 1965;18:157163. antigens in Guillain-Barr syndrome. J Neuroimmunol. 1984;6:303312.
84. Kanda T, Hayashi H, Tanabe H, et al. A fulminant case of Guillain-Barr 113. Burns J, Krasner LJ, Rostami A, et al. Isolation of P2 proteinreactive
syndrome: topographic and fibre size related analysis of demyelinating T-cell lines from human blood. Ann Neurol. 1986;19:391393.
changes. J Neurol Neurosurg Psychiatry. 1989;52:857864. 114. Taylor WA, Brostoff SW, Hughes RA. P2 specific lymphocyte transfor-
85. Asbury AK, Arnason BG, Adams RD. The inflammatory lesion in idiopathic mation in Guillain-Barr syndrome and chronic idiopathic demyelinating
polyneuritis. Its role in pathogenesis. Medicine (Baltimore). 1969;48: polyradiculoneuropathy. J Neurol Sci. 1991;104:5255.
173215. 115. Khalili-Shirazi A, Hughes RA, Brostoff SW, et al. T cell responses to myelin
86. Honavar M, Tharakan JK, Hughes RA, et al. A clinicopathological study proteins in Guillain-Barr syndrome. J Neurol Sci. 1992;111:200203.
of the Guillain-Barr syndrome. Nine cases and literature review. Brain. 116. Bansil S, Mithen FA, Singhal BS, et al. Elevated neopterin levels in
1991;114(pt 3):12451269. Guillain-Barr syndrome. Further evidence of immune activation. Arch
87. Ropper AH, Adelman L. Early Guillain-Barr syndrome without inflam- Neurol. 1992;49:12771280.
mation. Arch Neurol. 1992;49:979981. 117. Mancardi GL, Cadoni A, Zicca A, et al. HLA-DR Schwann cell reactiv-
88. Feasby TE, Hahn AF, Brown WF, et al. Severe axonal degeneration in ity in peripheral neuropathies of different origins. Neurology. 1988;38:
acute Guillain-Barr syndrome: evidence of two different mechanisms? 848851.
J Neurol Sci. 1993;116:185192. 118. Sharief MK, McLean B, Thompson EJ. Elevated serum levels of
89. Hughes R, Atkinson P, Coates P, et al. Sural nerve biopsies in Guillain- tumor necrosis factor-alpha in Guillain-Barr syndrome. Ann Neurol.
Barr syndrome: axonal degeneration and macrophage-associated demy- 1993;33:591596.
elination and absence of cytomegalovirus genome. Muscle Nerve. 1992; 119. Mithen FA, Colburn S, Birchem R. Human alpha tumor necrosis factor
15:568575. does not damage cultures containing rat Schwann cells and sensory neu-
90. Brechenmacher C, Vital C, Deminiere C, et al. Guillain-Barr syn- rons. Neurosci Res. 1990;9:5963.
drome: an ultrastructural study of peripheral nerve in 65 patients. Clin 120. Hartung HP, Reiners K, Michels M, et al. Serum levels of soluble E-selectin
Neuropathol. 1987;6:1924. (ELAM-1) in immune-mediated neuropathies. Neurology. 1994;44:
91. Carpenter S. An ultrastructural study of an acute fatal case of the Guillain- 11531158.
Barr syndrome. J Neurol Sci. 1972;15:125140. 121. Oka N, Akiguchi I, Kawasaki T, et al. Elevated serum levels of endothelial
92. Arstila AU, Riekkinen PJ, Rinne UK, et al. Guillain-Barr syndrome. leukocyte adhesion molecules in Guillain-Barr syndrome and chronic in-
Neurochemical and ultrastructural study. Eur Neurol. 1971;5:257269. flammatory demyelinating polyneuropathy. Ann Neurol. 1994;35:621624.
93. Prineas JW. Pathology of the Guillain-Barr syndrome. Ann Neurol. 122. Birchem R, Mithen FA, LEmpereur KM, et al. Ultrastructural effects of
1981;9(suppl):619. Guillain-Barr serum in cultures containing only rat Schwann cells and
94. Vallat JM, Leboutet MJ, Jauberteau MO, et al. Widenings of the myelin dorsal root ganglion neurons. Brain Res. 1987;421:173185.
lamellae in a typical Guillain-Barr syndrome. Muscle Nerve. 1994;17: 123. Sawant-Mane S, Clark MB, Koski CL. In vitro demyelination by serum
378380. antibody from patients with Guillain-Barr syndrome requires terminal
95. Griffin JW, Stoll G, Li CY, et al. Macrophage responses in inflammatory complement complexes. Ann Neurol. 1991;29:397404.
demyelinating neuropathies. Ann Neurol. 1990;27(suppl):S64S68. 124. Sawant-Mane S, Estep A 3rd, Koski CL. Antibody of patients with
96. Nyland H, Matre R, Mork S. Immunological characterization of sural Guillain-Barr syndrome mediates complement-dependent cytolysis of rat
nerve biopsies from patients with Guillain-Barr syndrome. Ann Neurol. Schwann cells: susceptibility to cytolysis reflects Schwann cell phenotype.
1981;9(suppl):8086. J Neuroimmunol. 1994;49:145152.
97. Hartung HP, Pollard JD, Harvey GK, et al. Immunopathogenesis and 125. Koski CL, Gratz E, Sutherland J, et al. Clinical correlation with anti-
treatment of the Guillain-Barr syndromePart I. Muscle Nerve. 1995;18: peripheral-nerve myelin antibodies in Guillain-Barr syndrome. Ann
137153. Neurol. 1986;19:573577.
98. Pollard JD, Baverstock J, McLeod JG. Class II antigen expression 126. Rybert B. Extra and intrathecal production of antinerve and antibrain
and inflammatory cells in the Guillain-Barr syndrome. Ann Neurol. antibodies in Guillain-Barr syndrome: evaluation by an antibody index.
1987;21:337341. Neurology. 1984;37:10061009.
99. Willison HJ, Halstead SK, Beveridge E, et al. The role of complement and 127. Hartung HP, Schwenke C, Bitter-Suermann D, et al. Guillain-Barr
complement regulators in mediating motor nerve terminal injury in murine syndrome: activated complement components C3a and C5a in CSF.
models of Guillain-Barr syndrome. J Neuroimmunol. 2008;201202: Neurology. 1987;37:10061009.
172182. 128. Koski CL, Sanders ME, Swoveland PT, et al. Activation of terminal com-
100. Sheikh KA, Zhang G, Gong Y, et al. An anti-ganglioside antibody- ponents of complement in patients with Guillain-Barr syndrome and
secreting hybridoma induces neuropathy in mice. Ann Neurol. 2004;56: other demyelinating neuropathies. J Clin Invest. 1987;80:14921497.
228239. 129. Koski CL, Chou DK, Jungalwala FB. Anti-peripheral nerve myelin
101. Plomp JJ, Willison HJ. Pathophysiological actions of neuropathy-related antibodies in Guillain-Barr syndrome bind a neutral glycolipid of
anti-ganglioside antibodies at the neuromuscular junction. J Physiol. peripheral myelin and cross-react with Forssman antigen. J Clin Invest.
2009;587:39793999. 1989;84:280287.
102. Greenshields KN, Halstead SK, Zitman FM, et al. The neuropathic 130. Yamakawa T, Nagai Y. Glycolipids at the cell surface and their biological
potential of anti-GM1 autoantibodies is regulated by the local glycolipid functions. Trends Biochem Sci. 1978;3:128131.
environment in mice. J Clin Invest. 2009;119:595610. 131. van Sorge NM, Yuki N, Koga M, et al. Ganglioside-specific IgG and
103. Cook SD, Dowling PC, Whitaker JN. The Guillain-Barr syndrome. IgA recruit leukocyte effector functions in Guillain-Barr syndrome.
Relationship of circulating immunocytes to disease activity. Arch Neurol. J Neuroimmunol. 2007;182:177184.
1970;22:470474. 132. Corbo M, Quattrini A, Latov N, et al. Localization of GM1 and Gal
104. Taylor WA, Hughes RA. T lymphocyte activation antigens in Guillain- (beta 1-3) GalNAc antigenic determinants in peripheral nerve. Neurology.
Barr syndrome and chronic idiopathic demyelinating polyradiculoneu- 1993;43:809814.
ropathy. J Neuroimmunol. 1989;24:3339. 133. Mizutamari RK, Wiegandt H, Nores GA. Characterization of anti-
105. Hartung HP, Hughes RA, Taylor WA, et al. T cell activation in Guillain- ganglioside antibodies present in normal human plasma. J Neuroimmunol.
Barr syndrome and in MS: elevated serum levels of soluble IL-2 recep- 1994;50:215220.
tors. Neurology. 1990;40:215218. 134. Ilyas AA, Willison HJ, Quarles RH, et al. Serum antibodies to ganglio-
106. Hartung HP, Reiners K, Schmidt B, et al. Serum interleukin-2 concentra- sides in Guillain-Barr syndrome. Ann Neurol. 1988;23:440447.
tions in Guillain-Barr syndrome and chronic idiopathic demyelinating 135. Chiba A, Kusunoki S, Shimizu T, et al. Serum IgG antibody to ganglio-
polyradiculoneuropathy: comparison with other neurological diseases of side GQ1b is a possible marker of Miller Fisher syndrome. Ann Neurol.
presumed immunopathogenesis. Ann Neurol. 1991;30:4853. 1992;31:677679.
107. Bansil S, Mithen FA, Cook SD, et al. Clinical correlation with serum- 136. Yuki N, Sato S, Tsuji S et al. Frequent presence of anti-GQ1b antibody in
soluble interleukin-2 receptor levels in Guillain-Barr syndrome. Fishers syndrome. Neurology. 1993;43:414417.
Neurology. 1991;41:13021305. 137. Chiba A, Kusunoki S, Obata H, et al. Serum anti-GQ1b IgG antibody is
108. Dahle C, Vrethem M, Ernerudh J. T lymphocyte subset abnormalities associated with ophthalmoplegia in Miller Fisher syndrome and Guillain-
in peripheral blood from patients with the Guillain-Barr syndrome. Barr syndrome: clinical and immunohistochemical studies. Neurology.
J Neuroimmunol. 1994;53:219225. 1993;43:19111917.
109. Hughes RA, Aslan S, Gray IA. Lymphocyte subpopulations and suppres- 138. Yuki N, Sato S, Tsuji S, et al. An immunologic abnormality common to
sor cell activity in acute polyradiculoneuritis (Guillain-Barr syndrome). Bickerstaffs brain stem encephalitis and Fishers syndrome. J Neurol Sci.
Clin Exp Immunol. 1983;51:448454. 1993;118:8387.
110. Winer JB, Gray IA, Gregson NA, et al. A prospective study of acute 139. Gregson NA, Koblar S, Hughes RA. Antibodies to gangliosides in
idiopathic neuropathy. III. Immunological studies. J Neurol Neurosurg Guillain-Barr syndrome: specificity and relationship to clinical features.
Psychiatry. 1988;51:619625. Q J Med. 1993;86:111117.

Scheld_Ch20.indd 312 2/21/14 5:40 PM


Chapter 20: Guillain-Barr Syndrome 313

140. Kusunoki S, Chiba A, Kon K, et al. N-acetylgalactosaminyl GD1a is a tar- 172. Brettschneider J, Petzold A, Sussmuth S, et al. Cerebrospinal fluid bio-
get molecule for serum antibody in Guillain-Barr syndrome. Ann Neurol. markers in Guillain-Barr syndromewhere do we stand? J Neurol.
1994;35:570576. 2009;256:312.
141. Ilyas AA, Mithen FA, Chen ZW, et al. Anti-GM1 IgA antibodies in 173. Shuaib A, Becker WJ. Variants of Guillain-Barr syndrome: Miller Fisher
Guillain-Barr syndrome. J Neuroimmunol. 1992;36:6976. syndrome, facial diplegia and multiple cranial nerve palsies. Can J Neurol
142. Gregson NA, Jones D, Thomas PK, et al. Acute motor neuropathy with Sci. 1987;14:611616.
antibodies to GM1 ganglioside. J Neurol. 1991;238:447451. 174. Ropper AH. Unusual clinical variants and signs in Guillain-Barr
143. Ilyas AA, Mithen FA, Dalakas MC, et al. Antibodies to acidic glycolip- syndrome. Arch Neurol. 1986;43:11501152.
ids in Guillain-Barr syndrome and chronic inflammatory demyelinating 175. Young RR, Asbury AK, Corbett JL, et al. Pure pan-dysautonomia
polyneuropathy. J Neurol Sci. 1992;107:111121. with recovery. Description and discussion of diagnostic criteria. Brain.
144. van den Berg LH, Marrink J, de Jager AE, et al. Anti-GM1 antibodies in 1975;98:613636.
patients with Guillain-Barr syndrome. J Neurol Neurosurg Psychiatry. 176. Schaumburg HH, Garger AR. Human toxic neuropathy due to indus-
1992;55:811. trial agents. In: Poduslo JF, ed. Peripheral Neuropathy. Philadelphia: WB
145. Fredman P, Vedeler CA, Nyland H, et al. Antibodies in sera from patients Saunders; 1993:1541.
with inflammatory demyelinating polyradiculoneuropathy react with 177. Olney RK, Aminoff MJ. Electrodiagnostic features of the Guillain-Barr
ganglioside LM1 and sulphatide of peripheral nerve myelin. J Neurol. syndrome: the relative sensitivity of different techniques. Neurology.
1991;238:7579. 1990;40:471475.
146. Kornberg AJ, Pestronk A, Bieser K, et al. The clinical correlates of high- 178. Ropper AH, Wijdicks EF, Shahani BT. Electrodiagnostic abnormalities
titer IgG anti-GM1 antibodies. Ann Neurol. 1994;35:234237. in 113 consecutive patients with Guillain-Barr syndrome. Arch Neurol.
147. Simone IL, Annunziata P, Maimone D, et al. Serum and CSF anti-GM1 1990;47:881887.
antibodies in patients with Guillain-Barr syndrome and chronic inflam- 179. Bradshaw DY, Jones HR Jr. Guillain-Barr syndrome in children: clini-
matory demyelinating polyneuropathy. J Neurol Sci. 1993;114:4955. cal course, electrodiagnosis, and prognosis. Muscle Nerve. 1992;15:
148. Yuki N, Yamada M, Sato S, et al. Association of IgG anti-GD1a antibody 500506.
with severe Guillain-Barr syndrome. Muscle Nerve. 1993;16:642647. 180. Vajsar J, Taylor MJ, MacMillan LJ, et al. Somatosensory evoked poten-
149. Walsh FS, Cronin M, Koblar S, et al. Association between glycoconjugate tials and nerve conduction studies in patients with Guillain-Barr syn-
antibodies and Campylobacter infection in patients with Guillain-Barr drome. Brain Dev. 1992;14:315318.
syndrome. J Neuroimmunol. 1991;34:4351. 181. Kimura J. Proximal versus distal slowing of motor nerve conduction
150. Oshima A. Central motor conduction in patients with anti-ganglioside velocity in the Guillain-Barr syndrome. Ann Neurol. 1978;3:344350.
antibody associated neuropathy syndromes and hyperreflexia. J Neurol 182. Cornblath DR, Mellits ED, Griffin JW, et al. Motor conduction studies in
Neurosurg Psychiatry. 2002;73:568573. Guillain-Barr syndrome: description and prognostic value. Ann Neurol.
151. Tanaka T, Furutama D, Sakai R, et al. Biological roles on anti-GM1 1988;23:354359.
antibodies in patients with Guillain-Barr syndrome for nerve growth 183. Brown WF, Feasby TE. Conduction block and denervation in Guillain-
factor signaling. Biochem Biophys Acta. 2007;1772:543548. Barr polyneuropathy. Brain. 1984;107(pt 1):219239.
152. Nakatani Y, Hotta S, Utsunomiya I, et al. Cav2.1 voltage-dependent 184. Triggs WJ, Cros D, Gominak SC, et al. Motor nerve inexcitability in
Ca2 channel current is inhibited by serum from select patients with Guillain-Barr syndrome. The spectrum of distal conduction block and
Guillain-Barr syndrome. Neurochem Res. 2009;34:149157. axonal degeneration. Brain. 1992;115(pt 1):12911302.
153. von Wulffen H, Hartard C, Scharein E. Seroreactivity to Campylobacter 185. Brown WF, Feasby TE, Hahn AF. Electrophysiological changes in the
jejuni and gangliosides in patients with Guillain-Barr syndrome. J Infect acute axonal form of Guillain-Barr syndrome. Muscle Nerve. 1993;16:
Dis. 1994;170:828833. 200205.
154. Willison HJ. Gangliosides as targets for autoimmune injury to the nervous 186. van der Mech FG, Meulstee J, Kleyweg RP. Axonal damage in Guillain-
system. J Neurochem. 2007;103(suppl 1):143149. Barr syndrome. Muscle Nerve. 1991;14:9971002.
155. Nishimoto Y, Susuki K, Yuki N. Serologic marker of acute motor axonal 187. Kiers L, Clouston P, Zuniga G, et al. Quantitative studies of F responses in
neuropathy in childhood. Pediatr Neurol. 2008;39:6770. Guillain-Barr syndrome and chronic inflammatory demyelinating poly-
156. Ogawa G, Kaida K, Kusunoki S, et al. Antibodies to ganglioside complexes neuropathy. Electroencephalogr Clin Neurophysiol. 1994;93:255264.
consisting of asialo-GM1 and GQ1b or GT1a in Fisher and Guillain-Barr 188. Eisen A, Humphreys P. The Guillain-Barr syndrome. A clinical and
syndromes. J Neuroimmunol. 2009;214:125127. electrodiagnostic study of 25 cases. Arch Neurol. 1974;30:438443.
157. Kusunoki S, Kaida K, Ueda M. Antibodies against gangliosides and gan- 189. McLeod JG, Walsh JC, Prineas JW, et al. Acute idiopathic polyneu-
glioside complexes in Guillain-Barr syndrome: new aspects of research. ritis. A clinical and electrophysiological follow-up study. J Neurol Sci.
Biochem Biophys Acta. 2008;1780:441444. 1976;27:145162.
158. Kanzaki M, Kaida K, Ueda M, et al. Ganglioside complexes containing 190. Sauron B, Bouche P, Cathala HP, et al. Miller Fisher syndrome: clinical and
GQ1b as targets in Miller Fisher and Guillain-Barr syndrome. J Neurol electrophysiologic evidence of peripheral origin in 10 cases. Neurology.
Neurosurg Psychiatry. 2008;79:11481152. 1984;34:953956.
159. Kaida K, Kamakura K, Ogawa G, et al. GD1b-specific antibody induces 191. Fross RD, Daube JR. Neuropathy in the Miller Fisher syndrome: clinical
ataxia in Guillain-Barr syndrome. Neurology. 2008;71:196201. and electrophysiologic findings. Neurology. 1987;37:14931498.
160. Kaida K, Sonoo M, Ogawa G, et al. GM1/GalNAc-GD1a complex: a target 192. van Doorn PA, van der Mech FGA. Guillain-Barr syndrome. Clin
for pure motor Guillain-Barr syndrome. Neurology. 2008;71:16831690. Immunother. 1994;2:8999.
161. Willison HJ, Veitch J. Immunoglobulin subclass distribution and bind- 193. Hund EF, Borel CO, Cornblath DR, et al. Intensive management and
ing characteristics of anti-GQ1b antibodies in Miller Fisher syndrome. treatment of severe Guillain-Barr syndrome. Crit Care Med. 1993;21:
J Neuroimmunol. 1994;50:159165. 433446.
162. Gong Y, Tagawa Y, Lunn MP, et al. Localization of major gangliosides in the 194. Ropper AH. Intensive care of acute Guillain-Barr syndrome. Can J
PNS: implications for immune neuropathies. Brain. 2002;125:24912506. Neurol Sci. 1994;21:S23S27.
163. OHanlon GM, Bullens RW, Plomp JJ, et al. Complex gangliosides as 195. Chevrolet JC, Deleamont P. Repeated vital capacity measurements as pre-
autoantibody targets at the neuromuscular junction in Miller Fisher dictive parameters for mechanical ventilation need and weaning success in
syndrome: a current perspective. Neurochem Res. 2002;27:697709. the Guillain-Barr syndrome. Am Rev Respir Dis. 1991;144:814818.
164. Lopez PH, Lardone RD, Irazoqui FJ, et al. The origin of anti-GM1 196. Newton-John H. Prevention of pulmonary complications in severe
antibodies in neuropathies: the binding site drift hypothesis. Neurochem Guillain-Barr syndrome by early assisted ventilation. Med J Aust. 1985;
Res. 2002;27:687695. 142:444445.
165. Ang CW, Tio-Gillen AP, Groen J, et al. Cross-reactive anti-galactocerebro- 197. Anderson NA, Kyriakides T, Ferguson IT, et al. Plasma exchange in the
side antibodies and Mycoplasma pneumoniae infections in Guillain-Barr treatment of Guillain-Barr syndrome: experience in a sub-regional refer-
syndrome. J Neuroimmunol. 2002;130:179183. ral centre. Postgrad Med J. 1991;67:154156.
166. Jacobs BC, Koga M, van Rijs W, et al. Subclass IgG to motor ganglio- 198. Smith GD, Hughes RA. Plasma exchange treatment and prognosis of
sides related to infection and clinical course in Guillain-Barr syndrome. Guillain-Barr syndrome. Q J Med. 1992;85:751760.
J Neuroimmunol. 2008;194:181190. 199. Dalos NP, Borel C, Hanley DF. Cardiovascular autonomic dysfunction in
167. Asbury AK. Diagnostic considerations in Guillain-Barr syndrome. Ann Guillain-Barr syndrome. Therapeutic implications of Swan-Ganz moni-
Neurol. 1981;9(suppl):15. toring. Arch Neurol. 1988;45:115117.
168. Asbury AK, Cornblath DR. Assessment of current diagnostic criteria for 200. Hantson P, Ilunga K, Martin N, et al. Cardiac conduction abnormalities
Guillain-Barr syndrome. Ann Neurol. 1990;27(suppl):S21S24. during carbamazepine therapy for neuralgia following Guillain-Barr syn-
169. Winer JB, Hughes RA, Osmond C. A prospective study of acute idio- drome. Acta Neurol Belg. 1993;93:4043.
pathic neuropathy. I. Clinical features and their prognostic value. J Neurol 201. Ropper AH, Wijdicks EF. Blood pressure fluctuations in the dysautono-
Neurosurg Psychiatry. 1988;51:605612. mia of Guillain-Barr syndrome. Arch Neurol. 1990;47:706708.
170. Zochodne DW. Autonomic involvement in Guillain-Barr syndrome: a 202. Ropper AH, Shahani BT. Diagnosis and management of acute areflexic
review. Muscle Nerve. 1994;17:11451155. paralysis with emphasis on Guillain-Barr syndrome. In: Asbury AK,
171. Pentland B, Donald SM. Pain in the Guillain-Barr syndrome: a clinical Gilliat RW. Peripheral Nerve Disorders. A Practical Approach. London:
review. Pain. 1994;59:159164. Butterworth; 1984:4657.

Scheld_Ch20.indd 313 2/21/14 5:40 PM


314 Part II: Viral Infections and Related Disorders

203. Feldman JM. Cardiac arrest after succinylcholine administration in a preg- 221. Niparko N, Goldie WD, Mitchell W, et al. The use of plasmapheresis in
nant patient recovered from Guillain-Barr syndrome. Anesthesiology. the management of Guillain Barr syndrome in pediatric patients. Ann
1990;72:942944. Neurol. 1989;26:448449.
204. Fiacchino F, Gemma M, Bricchi M, et al. Hypo- and hypersensitivity to 222. Epstein MA, Sladky JT. The role of plasmapheresis in childhood Guillain-
vecuronium in a patient with Guillain-Barr syndrome. Anesth Analg. Barr syndrome. Ann Neurol. 1990;28:6569.
1994;78:187189. 223. Khatri BO, Flamini JR, Baruah JK, et al. Plasmapheresis with acute
205. Hughes RA, Newsom-Davis JM, Perkin GD, et al. Controlled trial pred- inflammatory polyneuropathy. Pediatr Neurol. 1990;6:1719.
nisolone in acute polyneuropathy. Lancet. 1978;2:750753. 224. Lamont PJ, Johnston HM, Berdoukas VA. Plasmapheresis in children with
206. Swick HM, McQuillen MP. The use of steroids in the treatment of idio- Guillain-Barr syndrome. Neurology. 1991;41:19281931.
pathic polyneuritis. Neurology. 1976;26:205212. 225. Wang CH, Trifiletti R, Gold AP, et al. Plasma exchange versus intrave-
207. Double blind trial of intravenous methylprednisolone in Guillain- nous immunoglobulin as treatment for childhood onset Guillain Barr
Barr syndrome. Guillain-Barr Syndrome Steroid Trial Group. Lancet. syndrome. Ann Neurol. 1992;32:440.
1993;341:586590. 226. Jansen PW, Perkin RM, Ashwal S. Guillain-Barr syndrome in childhood:
208. Treatment of Guillain-Barr syndrome with high-dose immune globulins natural course and efficacy of plasmapheresis. Pediatr Neurol. 1993;
combined with methylprednisolone: a pilot study. Dutch Guillain-Barr 9:1620.
Study Group. Ann Neurol. 1994;35:749752. 227. Vajsar J, Sloane A, Wood E, et al. Plasmapheresis vs intravenous immu-
209. Cornblath DR, Hughes RA. Guillain-Barr syndrome: the evolution of noglobulin treatment in childhood Guillain-Barr syndrome. Arch Pediatr
therapy. J Neurol. 1994;241:404405. Adolesc Med. 1994;148:12101212.
210. Greenwood RJ, Newsom-Davis J, Hughes RA, et al. Controlled trial of 228. al-Qudah AA. Immunoglobulins in the treatment of Guillain-Barr syn-
plasma exchange in acute inflammatory polyradiculoneuropathy. Lancet. drome in early childhood. J Child Neurol. 1994;9:178180.
1984;1:877879. 229. Shahar EM, Roifman CM, Shorer Z, et al. High dose intravenous serum
211. Osterman PO, Fagius J, Lundemo G, et al. Beneficial effects of plasma gamma globulins are effective in severe pediatric Guillain-Barr syn-
exchange in acute inflammatory polyradiculoneuropathy. Lancet. 1984;2: drome: a prospective follow up study of 23 cases. Ann Neurol. 1994;36:
12961299. 456457.
212. Constantopoulos A, Langona E. Intravenous immune globulin in children 230. McGhee B, Jarjour IT. Single-dose intravenous immune globulin for treat-
with Guillain-Barr syndrome. Neuropediatrics. 1994;25:165. ment of Guillain-Barr syndrome. Am J Hosp Pharm. 1994;51:9799.
213. Duhem C, Dicato MA, Ries F. Side-effects of intravenous immune globu- 231. Notarangelo LD, Duse M, Tiberti S, et al. Intravenous immunoglobulin in two
lins. Clin Exp Immunol. 1994;97(suppl 1):7983. children with Guillain-Barr syndrome. Eur J Pediatr. 1993;152:372374.
214. Vermeulen M, van der Mech FG, Speelman JD, et al. Plasma and gamma- 232. Valle L, Dulac O, Nuyts JP, et al. Intravenous immune globulin is also
globulin infusion in chronic inflammatory polyneuropathy. J Neurol Sci. an efficient therapy of acute Guillain-Barr syndrome in affected children.
1985;70:317326. Neuropediatrics. 1993;24:235236.
215. Thornton CA, Griggs RC. Plasma exchange and intravenous immuno- 233. Suri M, Suri ML, Shahani BT. Intravenous gammaglobulin (IVGG) ther-
globulin treatment of neuromuscular disease. Ann Neurol. 1994;35: apy in children with severe acute inflammatory demyelinating polyneu-
260268. ropathy (AIDP). Neurology. 1993;43:A347.
216. van der Mech FG, Schmitz PI. A randomized trial comparing intrave- 234. Plasma exchange in Guillain-Barr syndrome: one-year follow-up. French
nous immune globulin and plasma exchange in Guillain-Barr syndrome. Cooperative Group on Plasma Exchange in Guillain-Barr Syndrome.
Dutch Guillain-Barr Study Group. N Engl J Med. 1992;326:11231129. Ann Neurol. 1992;32:9497.
217. Kleyweg RP, van der Mech FG. Treatment related fluctuations in 235. McKhann GM, Griffin JW, Cornblath DR, et al. Plasmapheresis and
Guillain-Barr syndrome after high-dose immunoglobulins or plasma- Guillain-Barr syndrome: analysis of prognostic factors and the effect of
exchange. J Neurol Neurosurg Psychiatry. 1991;54:957960. plasmapheresis. Ann Neurol. 1988;23:347353.
218. Ropper AH, Albert JW, Addison R. Limited relapse in Guillain-Barr 236. Mokuno K, Kiyosawa K, Sugimura K, et al. Prognostic value of cerebro-
syndrome after plasma exchange. Arch Neurol. 1988;45:314315. spinal fluid neuron-specific enolase and S-100b protein in Guillain-Barr
219. Hughes RA, Swan AV, Raphal JC, et al. Immunotherapy for Guillain- syndrome. Acta Neurol Scand. 1994;89:2730.
Barr syndrome: a systemic review. Brain. 2007;130:22452257. 237. Korinthenberg R, Schessl J, Kirschner J. Clinical presentation and course
220. Yoshioka M, Kuroki S, Mizue H. Plasmapheresis in the treatment of the of childhood Guillain-Barr syndrome: prospective multicenter study.
Guillain-Barr syndrome in childhood. Pediatr Neurol. 1985;1:329334. Neuropediatrics. 2007;38:1017.

Scheld_Ch20.indd 314 2/21/14 5:40 PM


CHAPTER 21 ACUTE VIRAL MYELITIS
J. DAVID BECKHAM AND KENNETH L. TYLER

Viral infections of the spinal cord occur as part of more exten- evidence of denervation, including the presence of fascicula-
sive infection of the central nervous system (CNS) or periph- tions and fibrillations. Sensory loss in myelitis can be radicular,
eral nervous system. When myelitis occurs in association dermatomal, or both. Depending on the transverse localiza-
with encephalitis or meningitis, the resulting syndromes are tion of the lesion(s), either loss of position and vibration sense
referred to as encephalomyelitis or meningoencephalomyelitis. or loss of pain and temperature may occur. Finding a sensory
Myelitis associated with involvement of spinal nerve roots or level below which sensory functions are lost is a classic hall-
peripheral nerves is referred to as myeloradiculitis or myelo- mark of spinal cord disease. Either relative or absolute spar-
radiculoneuritis. This chapter focuses on acute viral infections ing of sensation in sacral dermatomes (sacral sparing) may
in which spinal cord involvement is the dominant feature. occur when an intramedullary process such as viral myelitis
Acute myelitis associated with rabies virus and HIV infection leaves the most peripheral fibers in the spinothalamic tract
is discussed elsewhere (see Chapters 17 and 19), as are cases of relatively unharmed.
chronic viral myelitis due to infection with retroviruses includ- Patients with acute onset of signs and symptoms sugges-
ing HIV and human T lymphotropic viruses (HTLVs). In addi- tive of spinal cord dysfunction are a medical emergency. Initial
tion to directly infecting and injuring the spinal cord, viruses clinical and laboratory studies should be directed at trying to
can trigger postinfectious immune-mediated tissue injury. identify whether a compressive lesion is present and whether it
Spinal cord involvement is a common but rarely dominant is intramedullary or extramedullary in location. An algorithm
feature of acute disseminated encephalomyelitis (ADEM) (see for the immediate diagnostic approach to patients with acute
Chapter 22). Transverse myelitis (TVM) is an acute syndrome myelopathy is shown in Figure 21.1(2). Table 21.1 summarizes
defined by the nature and extent of the anatomic injury to the key diagnostic tests that may be useful in evaluating a patient
spinal cord, often associated with antecedent viral infections. with suspected acute viral myelitis. In the following sections,
Specific causes of TVM are discussed under the individual vi- viral etiologies of acute myelitis are discussed individually.
ruses involved, and the idiopathic syndrome is briefly reviewed
at the end of this chapter.
The term myelitis means inflammation of the spinal cord
and refers to disease of the spinal cord caused by a direct in-
HERPESVIRUSES
fectious process, a postinfectious process, or another indirect
mechanism of injury. The clinical features are determined to Herpes Simplex Virus
a large degree by the location and extent of the process both
in the craniocaudal and the transverse axes of the spinal cord Both herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2)
rather than by the inciting agent. can cause myelitis. HSV-2 most commonly causes myelitis
The clinical features of myelitis provide important clues in adults and HSV-1 most commonly in children (1,3). The
to the anatomic location of the lesion but do not enable my- clinical presentation ranges from mild forms of disease with
elitis to be separated from other causes of intramedullary full recovery to severe necrotizing myelitis with permanent
spinal cord injury. The clinical features of myelitis caused sequelae. Most cases are monophasic, although about 20%
by different viruses overlap substantially, and identification of patients experience recurrent episodes of myelitis, a feature
of a specific viral etiology typically depends on the results common to infection with several herpesviruses (47). In pa-
of laboratory tests. The characteristic features of myelitis tients with recurrent disease, the interval between recurrences
include variable combinations of weakness; sensory loss; may vary between 1 week and several months, with three or
and bowel, bladder, and sexual dysfunction, typically evolv- more discrete recurrences being noted (5,7). Up to two thirds
ing over days. Apoplectic or hyperacute (hours) evolution of of patients with HSV myelitis have an ascending pattern of
symptoms is occasionally seen in viral myelitis but is more spinal cord involvement, with the remainder having TVM (5).
typical of vascular spinal cord disease (1) (e.g., infarction Most cases of monophasic HSV myelitis are due to
resulting from atherosclerosis, arteritis, emboli or hemor- HSV-2 (5). Patients with HSV-2 myelitis often have a history
rhage, or even venous thrombosis [Foix-Alajouanine syn- of genital herpes. However, in one series, only two of seven
drome]). Viral causes of chronic myelitis or myelopathy in patients with HSV-2 myelitis had known genital herpes, and
which symptoms evolving over weeks or months are largely lesions, when present, often preceded spinal involvement
limited to HIV and HTLV. (5). Clinical features of HSV myelitis include paresis or pa-
Weakness in viral myelitis may be either of the upper motor ralysis, more commonly involving the legs than the arms.
neuron type with associated spasticity, hyperreflexia, and ex- Patients may have either reduced and/or absent tendon
tensor-plantar reflexes or of the lower motor neuron type with reflexes or hyperreflexia with extensor-plantar responses.
flaccid weakness and decreased or absent deep tendon reflexes. Decreased sensation to pain, temperature, and touch is com-
Lower motor neuron involvement in the absence of significant mon and tends to be more severe in sacral dermatomes.
sensory signs or symptoms is often referred to as acute flac- Patients can have decreased anal tone and urinary incon-
cid paralysis or poliomyelitis-like illness, although the latter tinence with overflow. HSV-2 can also cause a lumbosacral
term is best reserved for cases in which pathology is limited radiculomyelitis characterized by urinary retention associ-
to the anterior horns of the spinal cord. Involvement of motor ated with constipation, dull pain in the anogenital region,
neurons in the anterior horns or involvement of the anterior paresthesias, loss of sensation, or flaccid paresis of the leg
roots can result in prominent clinical and electrophysiologic muscles (8).

315

Scheld_Ch21.indd 315 2/21/14 5:40 PM


316 Part II: Viral Infections and Related Disorders

Presentation: Neurologic dysfunction consistent with a spinal cord injury at a specific level

History and physical examination


Confirm acute myelopathy
Elicit time course and extent of deficits
Determine signs, symptoms or prior history suggesting infection, systemic inflammatory disease, vascular/ischemia,
neoplasia, multiple sclerosis, radiation exposure, neuromyelitis optica, or trauma
Determine if recent history of vaccination or systemic illness

First priority: Rule out a compressive etiology

Gadolinium enhanced MRI of the spinal cord within 4 hours

Sufficient to cause Yes Structural abnormality (spondylolisthesis) or


compressive myelopathy? spinal mass

Yes No No

Urgent surgical evaluation Second priority: Define presence/absence of spinal cord inflammation
Consider intravenous
methylprednisolone

Lumbar Puncture
Consider non-inflammatory causes of myelopathy
Ischemia-arterial, venous, watershed or
arteriovenous malformation
Radiation No CSF pleocytosis or gadolinium
Epidural lipomatosis enhancement or elevated igG index
Fibrocartilaginous embolism
Consider early inflammatory myelopathy, false Yes
negative CSF (repeat LP in 27 days)
Third priority: Define extent
of demyelination

Check Brain MRI with gadolinium


and visual evoked potential

Brain/Brain Yes
Site of demyelination? Demyelination?
and optic tract

No
Optic Nerve/Tract

Possible diagnosis Acute Transverse Myelitis (ATM):


Multiple Sclerosis Idiopathic or Disease-Associated
ADEM Possible Neuromyelitis Optica (use standard criteria to distinguish
Disease-Associated ATM (Devics Disease) and initiate appropriate treatment)

FIGURE 21.1 An algorithm illustrating the immediate diagnostic approach to a patient presenting with
a suspected acute myelopathy or myelitis. LP, lumbar puncture. (From Transverse Myelitis Consortium
Working Group. Proposed diagnostic criteria and nosology of acute transverse myelitis. Neurology.
2002;59:499505, with permission.)

Scheld_Ch21.indd 316 2/21/14 5:40 PM


Chapter 21: Acute Viral Myelitis 317

TA B L E 2 1 . 1
POTENTIALLY USEFUL DIAGNOSTIC STUDIES FOR EVALUATING PATIENTS WITH SUSPECTED ACUTE MYELITIS

MRI of spinal cord and brain Serum


CSF examination Complete blood cell and differential, platelet count, hemoglobin
Cell count and differential and hematocrit
Cytology ESR
Glucose C-reactive protein
Protein Serum calcium, albumin
IgG, IgG index BUN, creatine
Electrophoresis and oligoclonal bands Aspartate aminotransferase, alanine aminotransferase, alkaline
Myelin basic protein phosphatase
CSF stains Creatine kinase, aldolase
Gram stain Amylase, lipase
Acid-fast bacilli stain (for mycobacteria) Vitamin B12 level
India ink stain Thyroid function tests
CSF serology Serum protein electrophoresis
VDRL Angiotensin-converting enzyme
Cryptococcal antigen ANA, anti-ds-DNA, anti-SS-A (Ro), anti-SS-B (La), anti-Sm
CSF cultures (Smith), anti-RNP
Bacterial Total complement, C3, C4 complement levels
Fungal Monospot and heterophile tests
Mycobacterial Cold agglutinins (for mycoplasma)
Viral HIV serology
CSF PCR: Mycoplasma IgG, IgM antibodies
HSV-1, HSV-2 Hepatitis A IgG, IgM; hepatitis B serology, hepatitis C serology
EBV RPR, FTA-ABS
VZV Lyme serology (IgG, IgM)
CMV Toxoplasma serology
HHV-6 Miscellaneous test
Enterovirus Chest x-ray
HIV Urinalysis
(WNV) Stool cultures for enteroviruses
(Rabies) Throat cultures for enteroviruses, mumps (selected cases)
(Mycoplasma) Urine cultures for CMV, mumps (selected cases)
(Mycobacterium tuberculosis) PPD
CSF and serum antibody studies EMG/NCV in patients with suspected muscle, peripheral nerve
WNV IgG, IgM or root involvement
HSV IgG, IgM Urodynamic studies in patients with bladder signs or symptoms
CMV IgG, IgM Visual and somatosensory evoked responses
VZV IgG, IgM Special tests for selected cases
EBV VCA IgG, IgM Spinal and/aortic arteriography in cases of suspected infarction
EBV EA IgG or embolus
EBV EBNA IgG Neck skin biopsy with direct fluorescent antibody analysis for
HHV-6 IgG, IgM rabies antigen
HTLV-1 IgG
Measles, mumps, rubella serology (selected cases)
Rabies IgG, IgM (selected cases)
Mycoplasma IgG, IgM complement-fixing antibodies
Borrelia burgdorferi (Lyme)

ANA, antinuclear antibody; ESR, erythrocyte sedimentation rate; FTA-ABS, fluorescent treponemal antibody absorption; PPD, purified protein
derivative; RPR, rapid plasma reagin; VDRL, Venereal Disease Research Laboratory.

The most severe form of HSV myelitis is an acute necro- try, and herpesvirus-like particles have been seen by electron
tizing myelopathy, which occurs predominantly in patients microscopy (913). Some cases have a prominent necrotiz-
with underlying diseases including HIV infection (9), malig- ing arteritis associated with myelomalacia with Cowdry A
nancy (10), and diabetes (11,12), although rare cases occur inclusion-bearing cells seen in the wall of the anterior spinal
in the absence of associated disease (13). All cases to date artery (9).
have been associated with HSV-2. At autopsy, these patients In HSV myelitis, the cerebrospinal fluid (CSF) typi-
often show areas of necrosis in the gray and white matter cally shows a lymphocytic pleocytosis and normal glucose
of the spinal cord associated with perivascular lymphocytic concentration. Patients with recurrent attacks may have a
cuffing. Cowdry type A inclusions are found in neurons, progressive reduction in the degree of pleocytosis with suc-
viral antigen can be demonstrated by immunocytochemis- ceeding attacks (7). Typical cell counts range between 10 and

Scheld_Ch21.indd 317 2/21/14 5:40 PM


318 Part II: Viral Infections and Related Disorders

200 cells/mm3, although rare cases with normal cell counts


have been reported (5,14). The CSF profile in patients with Varicella-Zoster Virus
acute necrotizing myelopathy may show striking pleocyto-
sis with up to 5,750 cells and a predominance of polymor- Myelitis is an unusual complication of varicella-zoster virus
phonuclear (PMN) leukocytes rather than lymphocytes (12). (VZV) infection, with most cases occurring in immuno-
Surprisingly, some reported patients have had no or minimal compromised individuals (1921). Myelitis can also occur
pleocytosis (9,11). The CSF protein concentration is almost in- as a complication of primary varicella infection or chicken-
variably elevated (range, 50 to 430 mg/dL). Oligoclonal bands pox (20,22,23). In immunocompromised patients, common
were found in one of nine patients in one series (5) and have underlying diseases include HIV infection (19,20,21,24,25),
been noted in other case reports (7,15). Hodgkin and non-Hodgkin lymphoma (19), and immunosup-
HSV is only rarely cultured from CSF in patients with pressive therapy (26,27). In a recent review of 31 cases of VZV
myelitis (15), and diagnosis depends on demonstration of HSV myelitis, 55% of patients with VZV myelitis were immuno-
DNA in CSF by polymerase chain reaction (PCR) (46,1517). compromised, and the majority of the remaining patients had
In one series of nine patients, all had PCR amplifiable HSV an underlying comorbidity such as malignancy or autoimmune
DNA in CSF, with six cases due to HSV-2, two cases due to disease (28). Cases of VZV myelitis in immunocompetent pa-
HSV-1, and one case indeterminate. In this series, HSV-2 DNA tients are reported but uncommon (29). Many patients have
was found in six of six cases with ascending myelitis, whereas antecedent zoster, although several cases without rash (myeli-
HSV-1 DNA was found in two of the three patients with nona- tis sine herpete) have been reported (3032). When zoster is
scending TVM (5). In an HSV CSF PCR-negative patient, a present, it can involve virtually any dermatome, but myelitis
presumptive diagnosis can be made by demonstrating intra- is often associated with disseminated zoster in immunocom-
thecal synthesis of HSV-specific antibodies (7). Antibody stud- promised patients (19). Cord dysfunction typically follows the
ies may be particularly useful in cases in which CSF is only onset of zoster with a median of 12 days (range, 5 to 21 days),
available late in infection (e.g., 14 days), when HSV DNA but symptoms of myelitis may follow rash by up to 3 months
is likely to have disappeared. Evidence of intrathecal synthesis or myelitis symptoms may precede the appearance of rash
of HSV-specific antibodies can be based on (a) the presence in (19,24). Cases of TVM following zoster have also occurred
CSF of immunoglobulin M (IgM) anti-HSV antibodies (these in patients who have previously received zoster vaccine (33).
antibodies cross the bloodbrain barrier poorly and their de- Patients with zoster myelitis present with subacute onset
tection in CSF is generally indicative of intrathecal synthesis); of asymmetric leg weakness which progresses to bilateral leg
(b) the detection of HSV-specific oligoclonal bands present weakness with paraparesis in up to 85% of patients (19).
in CSF but not in serum; (c) the comparison of HSV-specific Sensory loss is common (75%), with involvement of pain
immunoglobulin G (IgG) levels in CSF and serum with correc- and temperature more common than position and vibration
tion for bloodbrain barrier leak using either the CSF/serum sense. A Brown-Squard syndrome may occur, with poste-
albumin ratio or the ratio of antibody titers in CSF to that rior column signs (loss of position and vibration sense) ipsi-
in serum for an irrelevant virus (5,14,16,18). Basic labora- lateral to the rash and spinothalamic tract signs (loss of pain
tory studies usually add little to the diagnosis, although some and temperature sense) contralateral (19). Approximately one
patients have elevations in erythrocyte sedimentation rate and third of patients will have a level to all sensory modalities,
C-reactive protein (6,18). urinary incontinence occurs in about 50%, and bowel incon-
Magnetic resonance imaging (MRI) is exceedingly impor- tinence can also occur. In a few cases, VZV myelitis may show
tant for its role in both excluding other potential diagnoses a relapsing and remitting pattern (20,25,34), a feature also
and in establishing the presence of an intramedullary process. seen in some cases of HSV-2 myelitis (see earlier discussion).
The typical appearance of HSV myelitis is of an intramedullary Atypical presentations of VZV myelitis including cases not
fusiform or spindle-shaped area of an increased T2-weighted associated with rash (zoster sine herpete), skin lesions devel-
signal (5,7,14,16). The spinal cord is often enlarged or swollen oping after myelopathy, or lack of correspondence of zoster
in the area of the lesion. Areas of an increased T2-weighted lesions to spinal cord injury level occur more frequently in
signal may also have T1-weighted signal hypointensity. In rare immunocompromised patients (28).
cases, areas are both T1 and T2 hyperintense, a finding sug- In VZV myelitis, CSF shows a mononuclear pleocytosis
gestive of hemorrhagic necrosis (5). Contrast (gadolinium) in about 75% of patients with an increase in PMN predom-
enhancement may be seen in the area of the lesion and the inance in patients with rapid onset and severe disease (19).
adjacent meninges and nerve roots (6,7,16,17). Lesions are Recurrent episodes of CSF PMN pleocytosis with recurrent
most commonly in the upper thoracic and cervical cord (5,6) episodes of VZV myelitis have been reported (30). CSF protein
but can also involve the lower cord including the conus medul- concentration is elevated in 70%, but glucose concentration is
laris and cauda equina (7,1517). almost always normal. It is important to recognize that at least
No controlled clinical trials of antiviral therapy for HSV 50% of immunocompetent patients with a zoster rash in the
myelitis are available. Based on anecdotal reports, patients absence of myelitis have a CSF lymphocytic pleocytosis with
should be treated with intravenous acyclovir for at least cell counts from 5 to 1,440 cells/mm3 and exceeding 100 cells/
14 days (10 mg/kg three times per day). This can be followed mm3 in 30% of those with pleocytosis (35).
by oral antiviral drugs (e.g., valacyclovir at 1 g three times per Approximately 25% of patients with a zoster rash will also
day) until symptoms resolve. The utility of adding steroids is have evidence of CSF anti-VZV IgG and detectable VZV DNA
unproven. When given, steroids should be used only in com- by PCR, but CSF anti-VZV IgM is usually not detectable and
bination with antiviral therapy. A typical regimen involves the CSF VZV IgG index is usually normal, suggesting that
intravenous methylprednisolone (500 to 1,000 mg per day for intrathecal synthesis of VZV-specific antibody does not occur
3 to 5 days) followed by oral prednisone (100 mg per day) in uncomplicated herpes zoster reactivation (35). By contrast,
with doses tapered over 2 weeks (5,7,17). patients with zoster myelitis often have a positive CSF VZV
The prognosis of HSV myelitis is extremely variable. In PCR and intrathecal VZV-specific IgG synthesis, even VZV
one series of nine patients, one third (n 3) made a complete specific CSF oligoclonal bands (36).
recovery, and the remaining two thirds (n 6) had residual Patients with VZV myelitis frequently have abnormalities
sequelae including paraplegia and tetraplegia (5). on spinal MRI (Fig. 21.2). These can include diffuse swelling

Scheld_Ch21.indd 318 2/21/14 5:40 PM


Chapter 21: Acute Viral Myelitis 319

acyclovir alone (20,37) or a combination of acyclovir and


high-dose steroids (31). One patient apparently responded to
oral famciclovir (500 mg every 8 hours), who had not previ-
ously responded to steroids, acyclovir, or foscarnet (37).
Pathologic studies of VZV myelitis are limited (19,25,32).
In a comprehensive review of nine fatal cases, there was
extensive hemorrhagic necrosis with necrotizing vasculitis
and thrombosis in the dorsal root ganglia associated with
Cowdry type A intranuclear inclusions in both ganglion and
satellite cells (19). Almost all cases had abnormalities in the
posterior roots and the posterior horns of the spinal cord, al-
though severity varied. Inclusion bodies were seen in about
50%, and patients could have demyelination and signs of
necrotizing vasculitis and hemorrhagic spinal cord necrosis
(19,32). A similar necrotizing process with demyelination and
Cowdry type A inclusions was described in an AIDS patient
with chronic zoster myelitis (25). In a recent review of five
postmortem cases of VZV myelitis, tissue necrosis and inflam-
matory cell infiltration were the primary findings, but other
pathologic changes were noted as well, including vasculitis,
thrombosis, hemorrhagic transformation, and Cowdry type
A inclusions (28).

FIGURE 21.2 T1-weighted sagittal spinal MRI scans following admin- Cytomegalovirus
istration of gadolinium from a 27-year-old woman with varicella-
zoster myelitis. Enhancing lesions are present at the cervicomedullary
Cytomegalovirus (CMV) involvement of the spinal cord
junction (open arrow) and in the cervical region of the spinal cord
(closed arrows). (From Gilden DH, Beinlich BR, Rubinstien EM, et al. either can result in a pure TVM or can produce myeloradicu-
Varicella zoster myelitis: an expanding spectrum. Neurology. 1994;44: litis, or radiculomyelopathy. Most cases of CMV-associated
18181823, with permission.) myeloradiculopathy occur in HIV-infected patients (4548),
and this can be the initial manifestation of AIDS (49). Myelitis
can be a complication of both adult and pediatric HIV infec-
tion (50). Although CMV myelitis is predominantly a disease
of immunocompromised individuals, there have been isolated
of the cord and areas of high T2-weighted signal with or with- reports of both TVM and myeloradiculopathy in immuno-
out associated T1 hypointensity and contrast enhancement competent individuals (5157). Isolated cases of dual infec-
(20,22,27,32,3739). MRI lesions in the brainstem and cervi- tion of the cord with both HSV and CMV have been reported
cal cord may also occur in immunocompetent patients with in patients with AIDS (58). The incidence of CMV-associated
zoster myelitis (35). myeloradiculopathy has declined dramatically since the
The diagnosis of VZV myelitis is generally suspected introduction of highly active antiretroviral therapy (HAART).
when signs of myelitis develop following a typical zos- Myeloradiculopathy occurs with approximately equal fre-
ter eruption in an immunocompromised patient. Definitive quency as an isolated manifestation of CMV infection and
diagnosis depends on isolation of virus from CSF (24,31), in systemic CMV disease (59). Patients present with rapidly
demonstration of intrathecal synthesis of VZV-specific anti- progressive flaccid paralysis of the legs with hyporeflexia or
body (20,36,37,40), or PCR amplification of VZV DNA from areflexia (60). Urinary retention occurs in almost all patients
CSF (20,27,37,4042). Comparative studies of the sensitiv- (60). Pain, often involving the perianal region and low back,
ity and specificity of these tests are lacking, although clinical is a common initial symptom. Sensory loss usually involves
experience suggests that PCR is likely to be the most sensitive, small-fiber modalities (pain, temperature) more than vibration
and culture the least. Antibody and PCR test results should or proprioception, although both can occur. The symptoms
be viewed as complementary rather than mutually exclusive; are typically progressive, although some patients have a more
as in other viral infections, antibodies typically develop later indolent course.
than detectable nucleic acid. The duration for which VZV One of the unusual features of CMV myeloradiculitis is the
DNA remains detectable in CSF following varicella myelitis common occurrence of a PMN rather than a lymphocytic CSF
is unknown. In patients with relapsing-remitting myelitis, CSF pleocytosis (61,62). This feature is distinctive enough that its
VZV DNA may persist, as two patients had positive CSF PCR presence in an HIV-infected patient with myeloradiculopa-
results 8 and 11 months after onset of disease (20). thy should always suggest the possibility of CMV infection
No controlled clinical trials of treatment for VZV (63). Cell counts exceeding 1,000/mm3 can occur (46,6466),
myelitis are available. Treatment typically involves intrave- although median values of 150 to 650 cells/mm3 are more typi-
nous acyclovir given alone or in combination with steroids cal (60,61). In addition to pleocytosis, the CSF typically shows
(19,31,37,39,4244), although the paucity of cases and the an elevated protein concentration. Severe hypoglycorrhachia
variability in treatment regimens make efficacy difficult to may occur (46), although a CSF glucose concentration of less
access (20). In several patients who received intravenous acy- than 50% of the coincident plasma value has been reported in
clovir after myelopathy was established, treatment seemed to only about 30% of patients (62).
be without effect (24,39,44). By contrast, there are also reports Imaging studies are useful in demonstrating the location
of complete recovery in AIDS patients with VZV myelitis and extent of lesions. Typical findings on spinal MRI include
following acyclovir therapy (10 mg/kg every 8 hours) for 21 to areas of increased T2-weighted signal within the spinal cord,
35 days (42) and improvement in patients treated with either associated with enhancement of the pial lining of the cord,

Scheld_Ch21.indd 319 2/21/14 5:40 PM


320 Part II: Viral Infections and Related Disorders

conus, cauda equine, and lumbosacral nerve roots on contrast- ester of ganciclovir (valganciclovir). Induction and mainte-
enhanced T1-weighted images (66,67). The presence of promi- nance doses of intravenous and oral ganciclovir, intravenous
nent radicular enhancement in association with myelitis occurs foscarnet, and intravenous cidofovir require adjustment in
more often with CMV infection than in other forms of viral patients with renal insufficiency. Neutropenia is the major
myelitis and may provide a clue to the diagnosis. dose-limiting side effect with ganciclovir, and nephrotoxicity
Clinical electrophysiologic studies may be helpful in dem- with foscarnet and cidofovir. Nephrotoxicity can occur with
onstrating the presence of radicular and peripheral nerve ganciclovir but is less common. Neurotoxicity can occur with
involvement in association with myelitis, especially because this ganciclovir and foscarnet and in the case of foscarnet is often
component of the disease may be clinically obscured when the related to electrolyte alterations (especially hypocalcemia).
myelitis is severe. Both slowed conduction velocities consistent Patients whose HIV infection responds to HAART and who
with demyelination and reduced amplitude of motor and sensory consistently (6 months) demonstrate nondetectable HIV
action potentials consistent with axonal injury occur (59,68). viral loads and CD4 cell counts of more than 100 cells/mm3
PCR tests to amplify CMV DNA from CSF are the diagnos- may be able to discontinue maintenance therapy.
tic procedure of choice (59,6972). The diagnostic sensitivity Few detailed pathologic studies of CMV myeloradiculitis
of CSF PCR in CMV myelitis is more than 80%, with a speci- have been performed. There is often a prominent PMN and
ficity of more than 90% (59). It is important to emphasize that mononuclear cell infiltrate involving the sacral spinal cord,
CSF PCR is often positive when cultures are negative (69,72). cauda equina, and lumbosacral nerve roots associated with
Quantitative PCR studies of CMV DNA in CSF suggest that both demyelination and axonal destruction. CMV antigen is
extremely high DNA levels, exceeding 10 million copies of detectable in the spinal cord and in involved roots and endo-
CMV DNA per milliliter of CSF can be found in patients with thelial cell (47,59).
myeloradiculopathy (59,69,73).
CMV may be cultured from CSF in cases of radiculomyeli-
tis (59). The high seroprevalence rate of anti-CMV antibodies Human Herpesvirus-6 and
in serum generally renders serologic studies of limited value. Human Herpesvirus-7
However, the demonstration of specific intrathecal synthesis of
anti-CMV antibodies or the presence of detectable CSF anti- Human herpesvirus type 6 (HHV-6) causes exanthema subitum
CMV IgM antibodies can be considered presumptive evidence (roseola infantum). Most individuals are infected in early child-
for CNS infection. hood (age 6 to 12 months). Although primary HHV-6 infection
Controlled clinical trials of antiviral therapy in CMV- is usually benign, there have been isolated reports of meningo-
associated neurologic disease are lacking, and most reports encephalitis. Myelitis appears to be extremely rare in patients
of treatment of CMV-associated neurologic disease involve with HHV-6 infection. HHV-6 neuroinvasive disease including
isolated cases (59,69,74,75). Response to treatment of my- encephalitis and/or myelitis are most commonly reported as
eloradiculopathy is variable. Improvement or stabilization of a complication in bone marrow transplant recipients (84,85)
symptoms has followed treatment with ganciclovir, foscarnet, and occur with a frequency of about 3% in allogeneic hema-
or a combination of the two agents (60,62,64,68,7680). topoietic stem cell transplantation and up to 16% in patients
Disease has been associated with ganciclovir-resistant strains receiving cord blood transplantation from an unrelated donor
(81,82), and this risk may be enhanced in patients whose dis- (86). Median onset of clinical symptoms is 23 days after trans-
ease developed during ganciclovir treatment for CMV infec- plant, and limbic encephalitis is the most common presenting
tion elsewhere. As a general rule, the presence of myelitis is syndrome with or without associated myelitis. Very rare cases
an extremely poor prognostic indicator, with one review cit- of HHV-6 myelitis in immunocompetent individuals have been
ing mean survival times of 5.4 1.8 weeks in patients not reported (87). HHV-6 may also be responsible for rare cases of
receiving ganciclovir and 14.6 9.4 weeks in those receiving chronic myelitis presenting as spastic paraparesis (88).
ganciclovir therapy (76). There is one reported case of combined encephalitis and
Successful treatment is usually associated with disappear- acute flaccid paralysis in an immunocompetent 19-year-old
ance of CMV DNA from CSF, whereas viral DNA persists man infected with HHV-7 (89). The CSF had a lymphocytic
in nonresponders (74). A typical induction regimen for gan- pleocytosis, an elevated protein concentration, and a nor-
ciclovir involves 14 to 21 days of intravenous therapy with mal glucose. HHV-7 DNA was amplified from CSF by PCR.
doses of 5 mg/kg every 12 hours. Intravenous foscarnet at Serologic studies were also consistent with acute HHV-7 infec-
a dose of 90 mg/kg every 12 hours provides an acceptable tion. Virus was not cultured from either blood or CSF, and
alternative. Patients who fail to respond to either foscarnet serum PCR studies, in contrast to those in CSF, were negative.
or ganciclovir alone may respond to combined therapy with
the two drugs (79). A third agent, cidofovir, has been shown
to be efficacious in treatment of CMV retinitis, but experi- Epstein-Barr Virus
ence with this agent in CMV-associated neurologic disease
is limited (83). A typical dosing regimen is 5 mg/kg intrave- Neurologic complications of Epstein-Barr virus (EBV) infection
nously every week for 2 weeks followed by infusions of 5 mg/ have been estimated to occur in 1% to 5% of patients with
kg every 2 weeks. Fourteen to twenty-one days of ganciclo- severe infectious mononucleosis (90). EBV-associated neuro-
vir or foscarnet therapy is likely to be sufficient for immuno- logic disease can also occur in the absence of, or even precede,
competent patients. In most organ transplant recipients, 14 symptoms of infectious mononucleosis (91,92). CNS and pe-
to 21 days of therapy is also likely to be adequate assuming ripheral nervous system manifestations of EBV infection include
clinical and virologic response has occurred. Maintenance meningoencephalitis, cerebellitis, Guillain-Barr syndrome, and
therapy is usually not required for CMV myelitis in organ TVM. The spinal cord manifestations of EBV infection are
transplant recipients, although maintained vigilance for recur- generally considered among the more unusual neurologic com-
rent disease is essential. In patients with HIV infection, it is plications of EBV infection, although exact data about their
critical that HAART be initiated or optimized concomitantly frequency are not available (9195). Many cases of EBV myeli-
with anti-CMV therapy. HIV-infected patients invariably tis have occurred in apparently immunocompetent individuals
require maintenance therapy with an orally bioavailable valine and often present as a meningoencephalomyeloradiculopathy

Scheld_Ch21.indd 320 2/21/14 5:40 PM


Chapter 21: Acute Viral Myelitis 321

(96,97). Isolated reports of myelitis in immunocompromised elevated in the acute sera, and seroconversion is demonstrable
patients, including a case in a bone marrow transplant recipi- only in a few patients (about 10% to 20%). IgG VCA and
ent, have appeared (98). EBNA antibodies can persist for life, and their presence, in
TVM typically develops 1 to 2 weeks after the onset of the absence of other serologic signs of acute infection, reflect
infectious mononucleosis. It is important to recognize that the past rather than active infection. In rare cases, seroconversion
symptoms of mononucleosis may be mild (e.g., pharyngitis may be delayed for up to 2 months after onset of illness (92),
alone). Patients present with flaccid weakness with absent or further complicating diagnosis.
decreased reflexes (91,92,98101). Most patients have a sen- In patients with EBV myelitis, the CSF typically shows a
sory level, often associated with some radicular sensory signs mild lymphocytic pleocytosis (25 to 500 cells), mildly elevated
and symptoms. Urinary retention is common (101). Less typi- protein concentration, and normal or mildly depressed glu-
cally, the acute onset of paraparesis or tetraparesis is associated cose concentration (91,93,98,99,101104). EBV can be
with spasticity, hyperreflexia, and extensor-plantar reflexes cultured from oropharyngeal washings and circulating lym-
(91). A more indolent course in which weakness is preceded phocytes in patients with infectious mononucleosis. However,
by back pain and radicular sensory symptoms has also been virus is only rarely isolated from CSF in patients with neuro-
reported (91). A lower motor neuron pattern of asymmet- logic disease (105), although EBV DNA has been amplified
ric flaccid weakness resembling poliomyelitis can occur with from brain tissue by PCR (106,107). Virus may be shed by
absence of associated sensory or bladder symptoms (102). asymptomatic patients or as a result of reactivation induced
In some patients, even though myelitis is the dominant fea- during other diseases which limits the specificity of culture.
ture, associated radicular and encephalitic symptoms coexist, In patients with serologic evidence of systemic EBV infec-
and the syndrome has been referred to as encephalomyelora- tion, CSF studies can assist in definitively establishing the
diculopathy (103), encephalomyelitis (101), meningoencepha- presence of associated CNS infection. Demonstration of EBV
lomyelitis, meningomyeloradiculitis, and encephaloradiculitis IgM VCA antibodies in CSF provides evidence of intrathecal
(96). When signs and symptoms suggesting involvement of antibody synthesis and presumptive evidence of associated
multiple areas of the CNS occur, they can appear concomi- CNS infection (99). Sequential studies of CSF EBV-specific
tantly (98) or sequentially (104). One reported patient had IgM have only rarely been performed. In one reported case,
associated bilateral optic neuritis, suggestive of Devic disease CSF IgM was detected at 2 weeks after onset, was equivo-
(neuromyelitis optica [NMO]) (99). cal at 4 weeks, and absent at 8 weeks (99). Intrathecal syn-
Spinal MRI in EBV myelitis typically shows an area of in- thesis of IgG antibodies can be demonstrated by measuring
creased intramedullary T2-weighted and decreased T1-weighted concomitant CSF and serum IgG VCA levels and correcting
signal with enhancement of the lesion and adjacent meninges for CSF/serum albumin ratio (96,104). Amplification of EBV
after administration of gadolinium (91,96,98,99,101). The DNA by PCR from CSF provides strong evidence of CNS
affected area of the spinal cord may appear widened or swol- infection (93,96,98,101,102,104), although cases with nega-
len (91,98). Nerve root enhancement has also been noted in tive CSF PCR and positive serology results have been reported
patients with myeloradiculitis (96). A patient who presented (91,103). The duration for which PCR-amplifiable EBV DNA
with a poliomyelitis-like syndrome had no abnormal intramed- persists in CSF is unknown, although one immunocompro-
ullary signal but did show meningeal enhancement around the mised patient treated with ganciclovir and hyperimmunoglob-
cauda equina (102). Myelitis can occur with a normal MRI ulin was PCR positive after 1 month but became PCR negative
scan (93,103). at 2 months after onset of symptoms (98). A second patient
In patients with a prominent radicular component, results was found to have positive CSF PCRs at 1 and 4 weeks postin-
of clinical electrophysiologic tests may be abnormal with pro- fection, with a negative result at 7 weeks postinfection (108).
longed F-wave latencies on electromyography (EMG) and Semiquantitative PCR can be used to evaluate EBV genome
increased spontaneous activity consistent with denervation copy numbers in different forms of EBV infection, including
(100102). Laboratory studies can provide clues to the diag- CNS disease, and to a more limited degree to evaluate the
nosis. Complete blood cell count may show lymphocytosis and effects of therapy (109,110) Genome copy number has gener-
atypical lymphocytes (93). The presence of significant numbers ally been in the range of 500 to 2,000 copies/mL, although
of atypical lymphocytes in blood or CSF should prompt con- one immunocompromised patient had 100,000 copies/
sideration of EBV infection but can occur with other infections mL (98,110). PCR and antibody tests should be considered
(101). Older serologic tests, including the heterophil antibody complementary, with nucleic acid frequently detected acutely
test (the Paul-Bunnell test), have been largely replaced by com- and then clearing over approximately the first 2 weeks, and
mercial spot and slides tests (e.g., Monospot test) to detect het- antibody production appearing after the first week or two of
erophil antigens. It is important to recognize that heterophil infection.
tests are often negative in patients with EBV-associated TVM Acyclovir inhibits EBV replication and viral shedding
(91,92). Serologic tests measuring antibodies against distinct but does not significantly reduce clinical symptomatol-
virus-specific antigens, including the viral capsid antigens ogy associated with uncomplicated infectious mononucle-
(VCAs), early antigens (EAs), and Epstein-Barr nuclear an- osis (90). Studies of the effects of antiviral therapy in EBV
tigen (EBNA), provide more sensitive and specific confirma- myelitis are anecdotal, and no controlled clinical trials have
tion of diagnosis. Serodiagnosis of EBV infection is made by been performed. Intravenous acyclovir (10 mg/kg three times
demonstrating serum IgM VCA, which is generally present a day for 14 to 21 days) has been associated with clinical
acutely then declines over 1 to 3 months (99). Detection of improvement in some patients (93,101). Whereas ganciclovir
IgM antibodies to VCA is both sensitive and specific for di- is more active in cell culture against EBV than acyclovir, only
agnosis of recent EBV infection and can be found in about anecdotal experience exists. A 16-year-old boy who developed
90% of patients. The presence of IgG antibodies to VCA, IgG TVM following a bone marrow transplantation was success-
antibodies to EA, and no antibodies to EBNA in convalescent fully treated with a combination of ganciclovir (10 mg/kg per
sera provides evidence of recent EBV infection (91,92,99). day intravenously for 4 weeks followed by 60 mg/kg per day
A fourfold increase in titer of anti-VCA IgG antibody between orally for 4 weeks) and CMV hyperimmunoglobulin (400 mg/
acute and convalescent sera is also presumptive evidence of kg three times a week for 1 month then twice weekly for an
acute infection. However, IgG VCA antibody titers are often additional month) (98). The role of steroids as adjunctive

Scheld_Ch21.indd 321 2/21/14 5:40 PM


322 Part II: Viral Infections and Related Disorders

treatment in EBV infections remains controversial, although individuals exposed to circulating vaccine strain viruses and
their use has been advocated for severe complications of often is associated with an undiagnosed immune deficiency
infectious mononucleosis. In one multicenter double-blind (115). A recent case of vaccine-derived poliomyelitis infection
controlled trial, the use of steroids in uncomplicated infectious was recently described in a 44-year-old woman with common
mononucleosis was without clinical benefit and increased variable immune deficiency that was infected when her child
the risk of cardiac and neurologic complications (90,111). was immunized 11.9 years earlier with type 2 vaccine-derived
However, steroids, in combination with acyclovir, have been poliovirus (116). In some cases, low vaccine coverage is associ-
used in individual cases of EBV myelitis, and some patients ated with mosaic recombinant poliovirus lineages that include
have experienced rapid improvement after their institution genetically distinct vaccine-derived strains of poliovirus (117).
(91). Doses have been extremely variable, with one typical An outbreak of vaccine-associated poliomyelitis occurred in
regimen using intravenous methylprednisolone (1,000 mg children in the Dominican Republic and Haiti in 2000/2001
every 12 hours for 7 days) followed by prednisone (60 mg per and involved 21 confirmed cases, with two fatalities (118).
day) for an additional week followed by a tapering dose over All the affected individuals were unvaccinated or incompletely
several months (91). Although studies are limited to isolated vaccinated, with disease being caused by a derivative of the
cases, some reports indicate that the number of genomic copies poliovirus type 1 oral vaccine strain.
of EBV in CSF declines with antiviral therapy (109). Ninety percent to ninety-five percent of patients infected
Pathologic studies of fatal cases of EBV-associated myelitis during an epidemic with poliovirus remain asymptomatic.
are extremely limited. In some patients, both anterior horn cell A small percentage (4% to 8%) develop a minor illness last-
degeneration and inflammatory infiltration of nerve roots have ing 1 to 4 days and characterized by pharyngitis, gastrointesti-
been described (100). Clear prognostic information is diffi- nal symptoms, fever, malaise, and headache. This corresponds
cult to obtain because of the paucity of reported cases. Many to the period during which virus is replicating in the nasophar-
patients make a significant recovery, although mild residual ynx and gastrointestinal tract (119). Only approximately 1%
weakness and sensory loss often persist (91). Other patients to 2% of individuals develop neurologic signs and symptoms
have improved to a more modest degree, with substantial (major illness). Major illness typically begins with fever,
residual weakness and hyperreflexia (92,93,96). malaise, and headache, followed within 24 hours by signs of
meningeal irritation indistinguishable from other forms of
viral meningitis. Within 2 to 5 days of onset of meningitis,
Herpesvirus simiae (Monkey B Virus, weakness appears, often associated with muscle pain and ten-
derness. Infection may predominantly involve the spinal cord
Cercopithecine Herpesvirus-1) (spinal poliomyelitis), brainstem (bulbar poliomyelitis), or
cerebrum (polioencephalitis). Spinal poliomyelitis accounts for
B virus is considered at length in Chapter 14 and is not further
66% to 75% of neurologic cases (120). Weakness manifests
discussed here.
as a flaccid areflexic paralysis, which is typically more severe
proximally than distally, affects the legs more severely than the
arms, is asymmetric, and progresses for 3 to 5 days after onset.
PICORNAVIRUSES (POLIO AND Atrophy appears rapidly, usually within 5 to 7 days, and can
OTHER ENTEROVIRUSES) progress over several weeks.
CSF studies show a lymphocytic pleocytosis, normal or
Poliovirus infection was previously the worlds most com- mildly elevated protein concentration, and normal glucose
mon cause of acute flaccid paralysis. In 1988, an estimated concentration (120). More than 90% of patients have 20 to
350,000 cases of polio occurred worldwide in 125 countries. 300 cells/mm3. During the first 72 hours, PMN cells may pre-
In 1988, the Global Polio Eradication Initiative was launched dominate, but they are subsequently replaced by lymphocytes.
by the World Health Organization (WHO). Thanks to an The results of electrophysiologic tests are consistent with an
aggressive campaign of immunization, the number of cases of anterior horn cell process and include reduced amplitudes of
wild type (nonvaccine-associated) polio has declined by more compound muscle action potentials (CMAPs) with normal
than 99.8% worldwide. In 2013 (through 10 December), sensory amplitudes and no significant alteration in conduction
359 cases of poliovirus have been reported and all were due velocity beyond that explained by the degree of axonal loss.
to serotype 1 viruses. The cases have occurred predominantly MRI studies in classic polio have only rarely been reported but
in countries with ongoing civil strife and weakened gov- show increased T2-weighted signal in the substantia nigra and
ernmental and public health institutions including Somalia spinal cord anterior horns and cord swelling (121,122).
(183), Pakistan (74), Nigeria (50), Syria (17), Kenya (14), The pathologic substrate of spinal poliomyelitis is injury
Afghanistan (11), Ethiopia (6), and Cameroon (4). In 1994, predominantly affecting the motor neurons of the anterior
the WHO was able to certify the region of the Americas as horns (120). Injury is typically more severe in the lumbar and
polio free, followed by the western Pacific region in 2000, cervical enlargements and can extend transversely to involve
and the European region in 2002 (www.polioeradication.org). the posterior and intermediate horns, intermediolateral cell
The last known case of indigenous wild type polio infection column, and even dorsal root ganglia.
in the Western Hemisphere occurred in Peru in 1991 (112). Diagnosis is similar to that of other enteroviruses (EVs) and
Given the reemergence of poliovirus in countries of previous can be made based on virus isolation, amplification of viral
eradication, the American Academy of Pediatrics recommends RNA, or serologic studies. Stool cultures have the highest yield
continued vigilance in the United States to maintain vaccina- for isolation of virus and are often positive for weeks to months
tion with the inactivated poliovirus vaccine (113). A few cases after onset of illness. Amplification of viral RNA from CSF by
of poliomyelitis still occur in several countries in association reverse transcriptase PCR (RT-PCR), using the generally avail-
with the use of the live attenuated (Sabin) polio vaccine able EV probes, confirms the presence of an EV, but not
(114). In 2002, 13 such cases, all from countries in Africa, specifically polio. Poliovirus-specific primers for RT-PCR are
were reported to the WHO. Vaccine-associated poliomyelitis available but are not generally used by hospital diagnostic lab-
can occur at a rate of 1 in 2.7 million vaccines in either vacci- oratories. A fourfold increase in poliovirus-specific neutralizing
nated individuals, nonimmunized, or incompletely immunized or complement-fixing antibody in serum confirms diagnosis.

Scheld_Ch21.indd 322 2/21/14 5:40 PM


Chapter 21: Acute Viral Myelitis 323

There is no specific treatment for spinal poliomyelitis. T2-weighted signal predominantly localized to the gray matter
The experimental antiviral agent pleconaril is unfortunately (121,131), although studies may also be normal (129,132).
not currently available from the manufacturer (ViroPharma) In addition to abnormal intramedullary signal, some patients
but was of benefit in treatment of two of three patients with have cord swelling and gadolinium enhancement of affected
vaccine-associated paralytic poliomyelitis in an uncontrolled areas on T1-weighted images (139).
open-label study (123). Pleconaril acts by integrating into a Diagnosis depends on amplification of enteroviral RNA
hydrophobic pocket on the capsid of picornaviruses such as from CSF by RT-PCR (132), isolation of virus from CSF
polio and thereby inhibiting both viral uncoating and receptor (129,132), or demonstration of a more than fourfold increase
binding (123). Vaccination using either the inactivated Salk or in specific antibody titer between acute and convalescent sera
live attenuated Sabin vaccines or a combination of both pro- (129,131). Isolation of virus from throat or stool provides
vides effective protective immunity and has been the mainstay supportive evidence (137), but because of the potential for
of the worldwide poliovirus eradication campaign. viral shedding for up to several months, cultures from these
Mortality from poliomyelitis was about 8% in the last pre- sites cannot be considered definitive evidence of enteroviral
vaccination polio epidemics in the United States, with deaths CNS infection.
occurring predominantly in patients with bulbar or encepha- No controlled trials of therapy are available. As noted ear-
litic forms of the disease. Patients with spinal poliomyelitis lier, pleconaril, although not currently available, was reported
rarely die. Most patients with weakness show some improve- to be of benefit in two of three treated patients with para-
ment within the first several weeks after onset, with 60% of lytic poliomyelitis associated with the polio vaccine (123). In
eventual recovery being achieved by 3 months and 80% by one adult patient with acute flaccid paralysis due to echovirus
6 months (124). 19, clinical and laboratory evidence of improvement occurred
Twenty-nine percent to sixty-five percent of patients sur- following combined therapy with pleconaril and intravenous
viving paralytic poliomyelitis will subsequently experience immune globulin (IVIG) (138).
new onset of fatigue, weakness, and increasing muscle atro- PCR and in situ PCR (IS-PCR) have been used to search for
phy decades after recovery from their initial illness, termed the evidence of enteroviral infection in patients with amyotrophic
postpolio syndrome (125,126). The most common symptoms lateral sclerosis (ALS). One study reported that 88% of
of postpolio syndrome include generalized fatigue (62% to patients with ALS (vs. 3% of controls) had enteroviral nucleic
89%), weakness in previously affected muscles (54% to 87%), acid detected by IS-PCR in spinal cord specimens. The ampli-
weakness in previously unaffected muscles (33% to 77%), fied RNA had high homology with ECHO virus 7 (140). The
myalgia (39% to 80%), and increasing atrophy (28% to 39%) same group reported isolating enteroviral RNA by RT-PCR
(125,127). Some patients experience new respiratory insuffi- in 60% of spinal cord specimens from Japanese patients with
ciency, bulbar dysfunction, or sleep apnea (125). ALS as compared to 14% of controls. Amplification of nucleic
The pathogenesis of postpolio syndrome remains unknown acid from two cases indicated the sequences had high homol-
and some aspects remain controversial (125,127). There is no ogy with ECHO viruses 7, 9, and 30 (141). Unfortunately,
known therapy for postpolio syndrome that has been shown these studies have not been confirmed by other investigators.
to be of efficacy in randomized controlled trials (128). One important study using real-time RT-PCR failed to detect
any ECHO sequences in 20 spinal cord and 10 motor cortex
samples from patients with ALS (142). As a result, the evi-
Nonpolio Enteroviruses dence linking enteroviral infection to ALS must be considered
unconfirmed and extremely suspect.
Rare cases of myelitis due to EVs other than poliovirus have
been reported. Most cases have been attributed to coxsacki-
evirus A7, A9 (129), B1, B3, and B4 (130133); ECHO virus Enterovirus-71
types 2, 5, 11, 18, 19, and 25 (134139); and EV type 71
(see later discussion). Most patients have been immunocom- EV-71 is endemic worldwide and causes periodic epidemic
petent, although myelitis has been described in a patient outbreaks of both hand, foot, and mouth disease (HFMD) and
with X-linked agammaglobulinemia (132). The clinical syn- neurologic illness (143145). The largest known outbreak to
drome can be indistinguishable from that caused by poliovi- date involved 100,000 to 300,000 cases in Taiwan in 1998
rus. Most patients have asymmetric muscle weakness, usually (146,147). A small outbreak of 45 cases of EV-71 infection,
involving the legs. The deep tendon reflexes are diminished including 7 cases of poliomyelitis-like paralysis, occurred in
or absent, and muscle tone is decreased or flaccid. Weakness the United States in 1987 (148). Overall, the most common
can occur with dramatic suddenness, even evolving over sev- CNS manifestations of EV-71 include aseptic meningitis,
eral hours. Sensory abnormalities are not present in classic brainstem encephalitis, and poliomyelitis-like flaccid paraly-
poliomyelitis. In patients with TVM, the typical picture is sis, with children younger than 4 years of age at higher risk
of a flaccid paralysis either with decreased or absent or less for neurologic complications. Most patients will have a 1- to
commonly with increased deep tendon reflexes combined with 7-day prodromal illness that precedes the onset of neurologic
the presence of a sensory level, urinary retention, and in some disease. Prodromal symptoms include rash, headache, fever,
cases decreased anal sphincter tone (129,134,135,139). coryza, and diarrhea.
Because of their rarity, laboratory studies of patients with EV-71 spinal cord involvement can result in either polio-
nonpolio EV myelitis are limited. CSF usually shows a mild myelitis or TVM (144,145,149152). In some patients, spinal
lymphocytic pleocytosis with normal or mildly elevated pro- cord disease is combined with encephalitis (encephalomyelitis).
tein and normal glucose concentrations (129,137), although The frequency of spinal cord involvement has varied in differ-
cases with entirely normal CSF parameters occur (131,132). ent outbreaks between 1% and 21% (149,153). Sporadic cases
Electrophysiologic studies do not distinguish between ante- of EV-71 poliomyelitis-like illness can also occur (148,152).
rior horn cell disease and a motor axonopathy. In both cases, CSF studies usually show a lymphocytic pleocytosis with
there is evidence of denervation and reduction in amplitude of normal or slightly elevated protein and normal glucose concen-
CMAPs with preserved conduction velocities and normal sen- trations. MRI can show areas of increased T2-weighted signal
sory action potentials (132). MRI can show areas of increased within the cord, cord swelling, and increased signal in the ven-

Scheld_Ch21.indd 323 2/21/14 5:40 PM


324 Part II: Viral Infections and Related Disorders

tral roots and in the conus medullaris associated with dorsal was the presence of weakness in 32%, flaccid paralysis in 11%,
brainstem involvement (154). In patients with poliomyelitis- and 40% of encephalitis cases had hyporeflexia (157).
like illness, increased signal can occur predominantly in the The pathogenesis of the weakness associated with WNV
anterior horns (147,150). Definitive diagnosis depends on iso- is multifactorial. Early reports suggested that some patients
lation of virus from CSF, amplification of viral nucleic acid from had electrophysiologic findings consistent with Guillain-Barr
CSF by RT-PCR, or documentation of seroconversion between syndrome (159). However, most cases of weakness following
acute-phase and convalescent-phase sera. Unfortunately, CSF WNV infection are secondary to a poliomyelitis-like syndrome
viral cultures are only rarely positive. In one recent series, no attributable to WNV-associated injury to anterior horn cells
positive CSF cultures were found among 27 tested patients (160168). Acute flaccid paralysis can vary in extent from a
(146). The sensitivity of CSF RT-PCR is also uncertain. Virus single limb to tetraparesis in some cases with associated se-
is isolated from throat and/or stool cultures in only approxi- vere respiratory impairment (160,161,169). Although WNV
mately 20% of patients, but EV-71 RNA can be amplified from neuroinvasive infection is much more common in adults and
these specimens in approximately 50% of patients, suggesting in the elderly in particular, cases of WNV poliomyelitis are
that RT-PCR is more sensitive than culture (146). also reported in children (170). Uncommon presentations of
No controlled clinical trials of treatment are available. WNV acute flaccid paralysis include isolated upper extremity
IVIG was without effect in one study (103). The antiviral drug brachial monoplegia or diplegia as well as reports of delayed
pleconaril, which has activity against many enteroviral strains, and recurrent limb weakness (171,172).
does not have significant inhibitory activity against EV-71 in Patients with WNV-associated myelitis typically have a CSF
vitro but has not been tested clinically in EV-71 myelitis or lymphocytic or PMN pleocytosis with elevated protein (75 to
CNS infection. It is no longer available in the United States. 234 mg/dL) and normal glucose concentrations (160163).
However, cell counts were normal in 20% of patients in one
series (160,163). When CSF pleocytosis is present, the range
Hepatitis A has typically been between 50 and 350 cells/mm3, although
counts as high as 2,600 cells/mm3 have been reported (160).
TVM is a rare complication of hepatitis A infection. All cases Large studies of MRI changes for WNV myelitis are not avail-
reported to date have been in immunocompetent individu- able. A recent small study of 17 patients with WNV encepha-
als, including both adults and children (57,155,156). Several litis or meningoencephalomyelitis found that 50% of patients
patients have had associated brainstem involvement (57,156). had an abnormal MRI in the deep gray matter or brainstem,
The usual presentation for myelitis is flaccid weakness and a and two patients had increased T2 intensity in the ventral horn
sensory level (57,155,156). CSF shows a lymphocytic pleocyto- of the spinal cord (173). This data is consistent with prior
sis, elevated protein concentration, and normal glucose concen- studies showing that 38% of MRIs were abnormal (162), with
tration (155). Liver function test results are abnormal and can one patient exhibiting enhancement of the cauda equina and
provide an important clue to diagnosis. MRI studies are limited, two patients with areas of increased T2-weighted signal within
but in one report, increased T2-weighted signal was noted in the the cord as well as abnormal gradient and spin-echo signals.
cervical cord in association with cord swelling (155). Diagnosis Electrophysiologic studies in these cases are consistent with
depends on demonstration of seroconversion (155). injury to anterior horn cells or their axons and have shown
reduced motor amplitudes, from 25% to 50% of normal, with
preserved sensory responses, conduction velocities, and distal
latencies. On EMG testing, motor units are normal, but re-
ARBOVIRUSES cruitment is severely reduced (160,162,163).
Pathologic studies on patients with acute poliomyelitis-like
West Nile Virus flaccid paralysis are limited. Histopathologic changes reported
include acute parenchymal and perivascular inflammatory
West Nile virus (WNV) is an arbovirus belonging to the changes in the spinal cord with associated loss of anterior horn
Flaviviridae family and the Japanese encephalitis (JE) virus cells (162,165).
serocomplex, a group that includes JE, St. Louis encephalitis, Diagnosis of WNV infection depends predominantly on
and Murray Valley viruses. WNV was identified as the cause of serology (158). In patients with fever and neurologic manifes-
a cluster of encephalitis cases in New York City in August, 1999 tations, diagnosis of WNV CNS infection can be made by (a)
(157). This marked the first emergence of this virus as a cause detection of anti-WNV IgM in CSF by capture enzyme-linked
of encephalitis in the Western Hemisphere, although major immunosorbent assay (ELISA), (b) demonstration of both IgM
outbreaks of WNV encephalitis had occurred in Romania and IgG antibody in a single serum sample, (c) detection of
(1996), Russia (1999), and Israel (2000) (158). The geographic WNV RNA in CSF by RT-PCR, or (d) isolation of virus from
distribution of the virus spread progressively across the United CSF, blood, or brain tissue. Of these methods, the CSF IgM
States and now causes annual epidemic outbreaks of neuro- assay is the most sensitive and specific. CSF RT-PCR is highly
invasive viral infection throughout the United States. In 2013 specific but considerably less sensitive than serology. Serologic
(through 3 December), 2,318 cases of WNV were reported to cross reactions can occur with other members of the JE virus
the Centers for Disease Control and Prevention (CDC), includ- serocomplex and can often be distinguished by performing
ing 1,171 cases of neuroinvasive disease and 105 deaths (http:// neutralizing antibody tests. IgM antibodies in both serum and
www.cdc.gov/westnile/statsMaps/). Serosurveys following the CSF can persist for 6 months or longer, providing another
initial outbreak in New York suggest that asymptomatic cases potential source of confusion in endemic areas (158).
outnumber symptomatic ones by about 150:1. Symptomatic Currently no known effective treatment of WNV infec-
disease can take the form of West Nile fever, aseptic meningi- tion is available. Both ribavirin and interferon alpha, alone
tis, meningoencephalitis with or without weakness, or acute or in combination, have been utilized in non-controlled stud-
flaccid paralysis. In the initial New York outbreak, 63% had ies. A phase I/II randomized placebo-controlled double-blind
encephalitis, 29% aseptic meningitis, and 8% fever and head- trials to evaluate the efficacy of an Israeli IVIG preparation
ache alone (158). Encephalitis occurred predominantly in older (Collaborative Antiviral Study Group [CASG210]) and a
individuals. One of the striking features in encephalitis cases phase II/III randomized, placebo-controlled, double-blinded

Scheld_Ch21.indd 324 2/21/14 5:40 PM


Chapter 21: Acute Viral Myelitis 325

trial to evaluate the safety and efficacy of a humanized mono- high T2-weighted and reduced T1-weighted signal with asso-
clonal antibody (MGAWN1) were both discontinued due to ciated gadolinium enhancement (179,180,182). Two patients
low enrollment. improved dramatically after intravenous or oral steroid treat-
The prognosis of patients with West Nile flaccid paralysis ment (180,182).
or poliomyelitis is not fully understood, but studies suggest
that morbidity and mortality are substantial (174). A wide
range of presentation and degrees of limb weakness may Tick-Borne Encephalitis Virus
occur. In cases of bulbar involvement and acute flaccid paraly-
sis, the mortality may be as high as 70% (175,176). In gen- Tick-borne encephalitis (TBE) virus is another member of the
eral, respiratory failure is associated with fatality rates greater family Flaviviridae. Human infection can occur either from
than 50%. Of patients who survive West Nile flaccid paralysis, exposure to the virus-carrying tick vector or from ingestion
most strength recovery occurs in the first 6 to 8 months fol- of raw milk or cheese from infected goats, sheep, or cows.
lowing weakness onset. However, initial severity of paralysis Illness is often biphasic, with neurologic symptoms appear-
does not predict strength outcome (175,176). ing following an acute febrile illness and defervescence fol-
lowed by recurrent fever associated with possible neurologic
symptoms. Asymmetric paralysis of single or multiple limbs
Japanese Encephalitis (B) Virus commonly occurs in conjunction with meningoencephalitis,
but myelitis can also occur as the predominant presentation of
JE virus is the most common cause of epidemic viral encepha- infection with either Central European TBE virus or Russian
litis worldwide, typically causing in excess of 50,000 cases per spring-summer encephalitis virus. The most common myelitic
year. As with other arboviruses, asymptomatic cases outnum- presentation is an acute flaccid paralysis resembling paralytic
ber cases of encephalitis by at least 100:1. The virus is endemic poliomyelitis (183185). Diagnosis is made by demonstration
in many parts of Southeast Asia, China, and the Indian sub- of specific IgM antibody in serum or CSF. No specific therapy
continent. Myelitis can occur in conjunction with encephali- is available, although immune globulin has been utilized in
tis or more rarely as the predominant clinical manifestation. non-controlled trials (186). Rare cases of myelitis have also
Most cases of JE virus myelitis present as a poliomyelitis-like occurred following vaccination against TBE virus (187).
acute flaccid paralysis, but cases of acute TVM following JE Symptoms develop days to a few weeks following immuniza-
virus infection are also reported (177). tion and usually take the form of a TVM with weakness, a
In a survey of 22 cases of acute flaccid paralysis in chil- sensory level, and urinary retention. Although patients may
dren occurring in a Vietnamese hospital, 12 (55%) cases initially have a flaccid paralysis with hyporeflexia or areflexia,
were due to JE virus (178). Patients typically presented with a this usually evolves into a hyperreflexic spastic paralysis with
febrile illness followed by acute onset of asymmetric areflexic extensor-plantar responses (187). MRI has been reported to
weakness, typically involving legs more than arms. Seven of show increased T2-weighted signal within the spinal cord
twelve patients had associated acute urinary retention. Muscle (187). One patient improved following immunosuppressive
pains in the affected limbs, back stiffness, and nuchal rigid- therapy with cyclophosphamide (187).
ity were common. Respiratory tract muscle involvement lead-
ing to respiratory failure occurred in one third of patients.
Two patients had findings suggestive of associated brainstem
involvement. Objective sensory findings did not occur in this
ORTHOMYXOVIRUSES AND
series, although two patients had sensory symptoms. Patients PARAMYXOVIRUSES (INFLUENZA,
typically had a CSF lymphocytic pleocytosis with normal or MEASLES, AND MUMPS)
mildly elevated protein concentration and normal glucose
concentration. Electrophysiologic studies in patients with dis- Myelitis has been reported as a rare complication of influ-
tal weakness typically showed reduced amplitude of motor enza A infection (188,189). Patients typically present with
action potentials with normal conduction velocities and distal initial respiratory symptoms and fever. Myelitis manifests
latencies, although study results were often normal in patients as weakness usually with depressed or absent deep tendon
without distal weakness. reflexes, although extensor-plantar responses may be present
Diagnosis of JE virus infection depends on demonstration (189). Cases of TVM associated with H1N1 infection were
of antiJE virus IgG antibodies in serum. Detection of CSF recently reported (190). One case of TVM was reported fol-
antiJE virus IgM is a specific marker of CNS disease, because lowing vaccination with the H1N1 live attenuated influenza
CSF IgM antibodies do not occur in asymptomatic individuals. vaccine; but it was unclear whether TVM was vaccine-related
CSF PCR can be performed by the CDC, and if positive also or a consequence of a concomitant mycoplasma infection
allows for definitive diagnosis of neurologic disease. (191,192). One reported case of influenza myelitis pro-
No specific therapy for JEV myelitis is available. An gressed to complete tetraparesis with associated dysarthria
effective formalin-inactivated JEV vaccine has been avail- and dysphagia (189). CSF shows a pleocytosis with elevated
able since the early 1950s. Neurologic complications follow- protein and normal glucose concentrations. MRI of the spi-
ing vaccination for JE virus are rare. Three cases of myelitis nal cord shows cord swelling with intramedullary increased
associated with the formalin-inactivated killed vaccine have T2- and decreased T1-weighted signal (189). Diagnosis of
been reported (179182). These patients typically present 1 influenza myelitis is based on serology. Demonstration of
to 2 weeks after vaccination with a TVM. Typical symptoms intrathecal synthesis of antibody against influenza A pro-
include paraparesis, a sensory level, and urinary retention. vides strong supportive evidence for direct CNS viral infec-
Weakness is usually initially flaccid and associated with hy- tion (193). TVM has also been reported as a complication
poreflexia or areflexia. CSF studies typically show a pleocy- of seasonal influenza vaccination (193). MRI in one patient
tosis with elevated protein concentration. One patient had a showed fusiform cord enlargement and increased intramed-
PMN predominance (950 cells/mm3, 90% PMN). Elevated ullary T2-weighted signal, but no gadolinium enhancement
CSF myelin basic protein has been detected in several patients on T1-weighted images. Recovery from vaccine-associated
(180,182). MRI scan can show cord swelling, with areas of myelitis can be complete.

Scheld_Ch21.indd 325 2/21/14 5:40 PM


326 Part II: Viral Infections and Related Disorders

Isolated cases of myelitis have been reported in association ties of central motor conduction time are more frequently
with both wild type rubella (164,165) and the live attenuated encountered than abnormal sensory evoked potentials,
vaccine strain (194), measles (195), and mumps (196). although both are abnormal in more than 75% of patients.
Findings related to the caudal region of the cord are typically
more severe than those in the cervical region, although these
OTHER VIRUSES changes vary depending on clinical presentation. Spinal MRI
is abnormal in approximately 90% of patients (219221).
Isolated examples of myelitis caused by other viruses include Common findings include an area of increased T2-weighted
lymphocytic choriomeningitis virus, adenovirus, parvovirus signal hyperintensity in the central region of the cord, occupy-
B19, and hepatitis B (197203). ing two thirds or more of the cords cross-sectional area and
often extending for several segments (219,222). Swelling of
the cord occurs in approximately 50% of patients (219). MRI
TRANSVERSE MYELITIS examination of the brain can be extremely helpful in identify-
ing multifocal demyelination suggestive of either ADEM or
TVM is an anatomic diagnosis that refers to a focal inflamma- multiple sclerosis. CSF oligoclonal bands are also strongly
tory disorder of the spinal cord (myelitis) that affects motor, suggestive of multiple sclerosis. The presence of antibodies
sensory, and autonomic pathways (hence, transverse) directed against the aquaporin-4 water channel is generally
(204208). In adults, the incidence of TVM ranges from 1.3 diagnostic of NMO or NMO-like illness and are not a feature
to 8 cases per million population with a bimodal peak in of idiopathic TVM.
incidence at ages 10 to 19 years and 30 to 39 years (209213). There is no definitive evidence from well-designed clinical
As noted earlier, TVM can be caused by viral infections. It can trials that an effective therapy for TVM exists. Most studies
also occur after an infection or vaccination and is likely due to have suffered from design flaws including lack of random-
a postinfectious or parainfectious immune-mediated response. ization, blinding, or appropriate controls. In many cases, the
The general term TVM should be reserved for those patients rarity of the disorder has resulted in small sample sizes. In
in whom no specific etiology is identified. When a specific eti- several trials using historical controls or no controls, treat-
ology is known, this is best included in the designation (e.g., ment with intravenous methylprednisolone (e.g., 1 g/1.73 m2
EBV TVM). per day for 3 to 5 days) followed by oral prednisone (1 mg/
TVM may selectively involve the conus and epiconus of kg per day for 2 to 3 weeks) was felt to shorten the duration
the spinal cord (214). A progressive myelopathy with evidence of disease and improve outcome (211,223,224). However, in
of spinal cord necrosis with features resembling Devic disease another recent study using historical controls, treatment with
(NMO) may occur (215,216). Relapsing forms of TVM methylprednisolone (500 mg intravenously for 5 days) did not
have also been described (217); distinguishing these cases from alter outcome (225).
predominantly spinal forms of relapsing-remitting multiple Predictors of prognosis in TVM have been studied
sclerosis is problematic. Finally, a subset of patients with a domi- (226,227). In one review of 31 patients, approximately 50%
nant clinical picture of TVM with encephalitic and/or radicular had a good outcome (Barthel score 12) at 6 months. In a
signs and symptoms (encephalomyeloradiculopathy) has recent review of several published studies (2), it was estimated
been reported (218). that one third of patients recover with few or no sequelae, one
Patients develop weakness typically maximal in the legs with third have moderate sequelae, and one third severe sequelae.
50% progressing to complete paraplegia. Virtually all patients The most important predictors of poor outcome were the ini-
have sensory signs and symptoms including paresthesias, numb- tial severity of weakness and evidence of denervation on EMG
ness, or radicular/bandlike dysesthesias (2). Most will have an (225,226). The presence of detectable 14-3-3 protein in CSF is
associated sensory level. Autonomic symptoms are variable and also associated with a poor outcome (228).
can include constipation, bowel or bladder incontinence, and
voiding difficulties, particularly urinary retention (2). Deficits
can develop in as little as 4 hours, although in most patients, ACKNOWLEDGMENTS
progression occurs over days to several weeks. Patients who
take more than 1 month to progress to maximum deficit are Dr. Tyler is supported by grants from the National Institute
unlikely to have TVM. Most patients will have a CSF pleo- of Neurological Disorders and Stroke (NINDS) (NS076512),
cytosis, and it has been suggested that evidence of spinal cord National Institute of Allergy and Infectious Diseases (NIAID)
inflammation, as documented either by CSF pleocytosis, by (AI01064), the Department of Veterans Affairs (BX000963),
abnormal IgG index, or by gadolinium-enhancing lesions, and by the Reuler-Lewin Family Professorship of Neurology
should be part of the diagnostic criteria for TVM (2). at the University of Colorado Health Sciences Center.
Diagnosis of TVM depends on clinical features and sup- Dr. Beckham is supported by the NIAID U54 AI065357 Rocky
portive laboratory study results. In most patients, abnormali- Mountain Regional Center of Excellence.

References
1. Irani DN. Aseptic meningitis and viral myelitis. Neurol Clin. 2008;26: 5. Nakajima H, Furutama D, Kimura F, et al. Herpes simplex virus myeli-
635655, viiviii. tis: clinical manifestations and diagnosis by the polymerase chain reaction
2. Transverse Myelitis Consortium Working Group. Proposed diagnostic crite- method. Eur Neurol. 1998;39:163167.
ria and nosology of acute transverse myelitis. Neurology. 2002;59:499505. 6. Shyu WC, Lin JC, Chang BC, et al. Recurrent ascending myelitis: an
3. Galanakis E, Bikouvarakis S, Mamoulakis D, et al. Transverse myelitis unusual presentation of herpes simplex virus type 1 infection. Ann Neurol.
associated with herpes simplex virus infection. J Child Neurol. 2001;16: 1993;34:625627.
866867. 7. Gobbi C, Tosi C, Stadler C, et al. Recurrent myelitis associated with herpes
4. Nakajima H, Furutama D, Kimura F, et al. Herpes simplex virus type 2 simplex virus type 2. Eur Neurol. 2001;46:215218.
infections presenting as brainstem encephalitis and recurrent myelitis. 8. Eberhardt O, Kuker W, Dichgans J, et al. HSV-2 sacral radiculitis (Elsberg
Intern Med. 1995;34:839842. syndrome). Neurology. 2004;63:758759.

Scheld_Ch21.indd 326 2/21/14 5:40 PM


Chapter 21: Acute Viral Myelitis 327

9. Britton CB, Mesa-Tejada R, Fenoglio CM, et al. A new complication 42. Lionnet F, Pulik M, Genet P, et al. Myelitis due to varicella-zoster virus in
of AIDS: thoracic myelitis caused by herpes simplex virus. Neurology. two patients with AIDS: successful treatment with acyclovir. Clin Infect
1985;35:10711074. Dis. 1996;22:138140.
10. Iwamasa T, Utsumi Y, Sakuda H, et al. Two cases of necrotizing myelopa- 43. Cullis PA, Gilroy J, Cushing R. Herpes zoster myelitis treated with vidara-
thy associated with malignancy caused by herpes simplex virus type 2. Acta bine. J Neurol Neurosurg Psychiatry. 1982;45:94.
Neuropathol. 1989;78:252257. 44. Hwang YM, Lee BI, Chung JW, et al. A case of herpes zoster myelitis: posi-
11. Iwamasa T, Yoshitake H, Sakuda H, et al. Acute ascending necrotizing my- tive magnetic resonance imaging finding. Eur Neurol. 1991;31:164167.
elitis in Okinawa caused by herpes simplex virus type 2. Virchows Arch A 45. Cohen BA, McArthur JC, Grohman S, et al. Neurologic prognosis of
Pathol Anat Histopathol. 1991;418:7175. cytomegalovirus polyradiculomyelopathy in AIDS. Neurology. 1993;43:
12. Wiley CA, VanPatten PD, Carpenter PM, et al. Acute ascending necro- 493499.
tizing myelopathy caused by herpes simplex virus type 2. Neurology. 46. de Gans J, Tiessens G, Portegies P, et al. Predominance of polymorpho-
1987;37:17911794. nuclear leukocytes in cerebrospinal fluid of AIDS patients with cyto-
13. Folpe A, Lapham LW, Smith HC. Herpes simplex myelitis as a cause of megalovirus polyradiculomyelitis. J Acquir Immune Defic Syndr. 1990;3:
acute necrotizing myelitis syndrome. Neurology. 1994;44:19551957. 11551158.
14. Petereit HF, Bamborschke S, Lanfermann H. Acute transverse myelitis 47. Eidelberg D, Sotrel A, Vogel H, et al. Progressive polyradiculopathy in
caused by herpes simplex virus. Eur Neurol. 1996;36:5253. acquired immune deficiency syndrome. Neurology. 1986;36:912916.
15. Ellie E, Rozenberg F, Dousset V, et al. Herpes simplex virus type 2 ascend- 48. Miller RG, Storey JR, Greco CM. Ganciclovir in the treatment of progres-
ing myeloradiculitis: MRI findings and rapid diagnosis by the polymerase sive AIDS-related polyradiculopathy. Neurology. 1990;40:569574.
chain method. J Neurol Neurosurg Psychiatry. 1994;57:869870. 49. Mahieux F, Gray F, Fenelon G, et al. Acute myeloradiculitis due to
16. Kuker W, Schaade L, Ritter K, et al. MRI follow-up of herpes simplex virus cytomegalovirus as the initial manifestation of AIDS. J Neurol Neurosurg
(type 1) radiculomyelitis. Neurology. 1994;52:11021103. Psychiatry. 1990;52:270274.
17. Kusuhara T, Nakajima M, Inoue H, et al. Parainfectious encephalomyelo- 50. Marriage SC, Booy R, Hermione Lyall EG, et al. Cytomegalovirus myeli-
radiculitis associated with herpes simplex virus 1 DNA in cerebrospinal tis in a child infected with human immunodeficiency virus type 1. Pediatr
fluid. Clin Infect Dis. 2002;34:11991205. Infect Dis J. 1990;15:549551.
18. Koskiniemi ML, Vaheri A, Manninen V, et al. Ascending myelitis with high 51. Baig SM, Khan MA. Cytomegalovirus-associated transverse myelitis in a
antibody titer to herpes simplex virus in the cerebrospinal fluid. J Neurol. non-immunocompromised patient. J Neurol Sci. 1989;134:210211.
1982;227:187191. 52. Fux CA, Pfister S, Nohl F, et al. Cytomegalovirus-associated acute
19. Devinsky O, Cho ES, Petito CK, et al. Herpes zoster myelitis. Brain. transverse myelitis in immunocompetent adults. Clin Microbiol Infect.
1991;114(pt 3):11811196. 2003;9:11871190.
20. Gilden DH, Beinlich BR, Rubinstien EM, et al. Varicella-zoster virus myeli- 53. Giobbia M, Carniato A, Scotton PG, et al. Cytomegalovirus-associated
tis: an expanding spectrum. Neurology. 1994;44:18181823. transverse myelitis in a non-immunocompromised patient. Infection. 1999;
21. Chang CC, McLean C, Vujovic O, et al. Fatal acute varicella-zoster virus 27:228230.
hemorrhagic meningomyelitis with necrotizing vasculitis in an HIV-infected 54. Kabins S, Keller R, Naragi S, et al. Viral ascending radiculomyelitis with
patient. Clin Infect Dis. 2009;48:372373. severe hypoglycorrachia. Arch Intern Med. 1976;136:933935.
22. Rosenfeld J, Taylor CL, Atlas SW. Myelitis following chickenpox: a case 55. Miles C, Hoffman W, Lai CW, et al. Cytomegalovirus-associated transverse
report. Neurology. 1993;43:18341836. myelitis. Neurology. 1993;43:21432145.
23. White HH. Varicella myelopathy. N Engl J Med. 1962;266:772773. 56. Rigamonti A, Usai S, Ciusani E, et al. Atypical transverse myelitis due
24. Gomez-Tortosa E, Gadea I, Gegundez MI, et al. Development of to cytomegalovirus in an immunocompetent patient. Neurol Sci. 2005;
myelopathy before herpes zoster rash in a patient with AIDS. Clin Infect 26:351354.
Dis. 1994;18:810812. 57. Tyler KL, Gross RA, Cascino GD. Unusual viral causes of transverse
25. Manian FA, Kindred M, Fulling KH. Chronic varicella-zoster virus myelitis myelitis: hepatitis A virus and cytomegalovirus. Neurology. 1986;36:
without cutaneous eruption in a patient with AIDS: report of a fatal case. 855858.
Clin Infect Dis. 1995;21:986988. 58. Tucker T, Dix RD, Katzen C, et al. Cytomegalovirus and herpes simplex
26. Baethge BA, King JW, Husain F, et al. Herpes zoster myelitis occur- virus ascending myelitis in a patient with acquired immune deficiency syn-
ring during treatment for systemic lupus erythematosus. Am J Med Sci. drome. Ann Neurol. 1985;18:7479.
1989;298:264266. 59. Cinque P, Cleator GM, Weber T, et al. Diagnosis and clinical management
27. Ebo DG, DeClerck LS, Stevens WJ, et al. Herpes zoster myelitis occur- of neurological disorders caused by cytomegalovirus in AIDS patients.
ring during treatment for systemic lupus erythematosus. J Rheumatol. European Union Concerted Action on Virus Meningitis and Encephalitis.
1996;23:548550. J Neurovirol. 1998;4:120132.
28. Hung CH, Chang KH, Kuo HC, et al. Features of varicella zoster virus 60. Anders HJ, Goebel FD. Cytomegalovirus polyradiculopathy in patients
myelitis and dependence on immune status. J Neurol Sci. 2012;318: with AIDS. Clin Infect Dis. 1998;27:345352.
1924. 61. Miller RF, Fox JD, Thomas P, et al. Acute lumbosacral polyradiculopa-
29. Tavazzi E, Minoli L, Ferrante P, et al. Varicella zoster virus meningo- thy due to cytomegalovirus in advanced HIV disease: CSF findings in
encephalo-myelitis in an immunocompetent patient. Neurol Sci. 2008;29: 17 patients. J Neurol Neurosurg Psychiatry. 1996;61:456460.
279283. 62. So YT, Olney RK. Acute lumbosacral polyradiculopathy in acquired
30. Haug A, Mahalingam R, Cohrs RJ, et al. Recurrent polymorphonuclear immunodeficiency syndrome: experience in 23 patients. Ann Neurol. 1994;
pleocytosis with increased red blood cells caused by varicella zoster virus 35:5358.
infection of the central nervous system: case report and review of the litera- 63. Granter SR, Doolittle MH, Renshaw AA. Predominance of neutrophils in
ture. J Neurol Sci. 2010;292:8588. the cerebrospinal fluid of AIDS patients with cytomegalovirus radiculopa-
31. Heller HM, Carnevale NT, Steigbigel RT. Varicella zoster virus transverse thy. Am J Clin Pathol. 1996;105:364366.
myelitis without cutaneous rash. Am J Med. 1990;88:550551. 64. Fuller GN, Gill SK, Guiloff RJ, et al. Ganciclovir for lumbosacral polyra-
32. Meylan PR, Miklossy J, Iten A, et al. Myelitis due to varicella-zoster virus diculopathy in AIDS. Lancet. 1990;335:4849.
in an immunocompromised patient without a cutaneous rash. Clin Infect 65. Jacobson MA, Mills J, Rush J, et al. Failure of antiviral therapy for
Dis. 1995;20:206208. acquired immunodeficiency syndrome-related cytomegalovirus myelitis.
33. LaRovere KL, Raju GP, Gorman MP. Postvaricella acute transverse myelitis Arch Neurol. 1988;45:10901092.
in a previously vaccinated child. Pediatr Neurol. 2008;38:370372. 66. Talpos D, Tien RD, Hesselink JR. Magnetic resonance imaging of AIDS-
34. Baik JS, Kim WC, Heo JH, et al. Recurrent herpes zoster myelitis. J Korean related polyradiculopathy. Neurology. 1991;41:19951997.
Med Sci. 1997;12:360363. 67. Bazan C III, Jackson C, Jinkins JR, et al. Gadolinium-enhanced MRI
35. Haanpaa M, Dastidar P, Weinberg A, et al. CSF and MRI findings in in a case of cytomegalovirus polyradiculopathy. Neurology. 1991;41:
patients with acute herpes zoster. Neurology. 1998;51:14051411. 15221523.
36. Ceroni M, Mazzarello P, Poloni M, et al. Transient intrathecal IgG synthe- 68. Fuller GN. Cytomegalovirus and the peripheral nervous system in AIDS.
sis in herpes zoster myelitis: 2 case reports. Eur Neurol. 1989;29:124127. J Acquir Immune Defic Syndr. 1992;5(suppl 1):S33S36.
37. de Silva SM, Mark AS, Gilden DH, et al. Zoster myelitis: improvement 69. Arribas JR, Clifford DB, Fichtenbaum CJ, et al. Level of cytomegalovirus
with antiviral therapy in two cases. Neurology. 1996;47:929931. (CMV) DNA in cerebrospinal fluid of subjects with AIDS and CMV infec-
38. Friedman DP. Herpes zoster myelitis: MR appearance. AJNR Am J tion of the central nervous system. J Infect Dis. 1995;172:527531.
Neuroradiol. 1992;13:14041406. 70. Clifford DB, Buller RS, Mohammed S, et al. Use of polymerase chain reac-
39. Hirai T, Korogi Y, Hamatake S, et al. Case report: varicella-zoster virus tion to demonstrate cytomegalovirus DNA in CSF of patients with human
myelitisserial MR findings. Br J Radiol. 1996;69:11871190. immunodeficiency virus infection. Neurology. 1993;43:7579.
40. Aizawa H, Suzutani T, Yahara O, et al. A case of varicella-zoster myelopa- 71. Gozlan J, Salord JM, Roullet E, et al. Rapid detection of cytomegalovirus
thy. Acta Neurol Scand. 1996;93:470472. DNA in cerebrospinal fluid of AIDS patients with neurologic disorders.
41. Grant AD, Fox JD, Brink NS, et al. Detection of varicella-zoster virus DNA J Infect Dis. 1992;166:14161421.
using the polymerase chain reaction in an immunocompromised patient 72. Wolf DG, Spector SA. Diagnosis of human cytomegalovirus central nervous
with transverse myelitis secondary to herpes zoster. Genitourin Med. system disease in AIDS patients by DNA amplification from cerebrospinal
1993;69:273275. fluid. J Infect Dis. 1992;166:14121415.

Scheld_Ch21.indd 327 2/21/14 5:40 PM


328 Part II: Viral Infections and Related Disorders

73. Shinkai M, Spector SA. Quantitation of human cytomegalovirus (HCMV) 103. Merelli E, Bedin R, Sola P, et al. Encephalomyeloradiculopathy associated
DNA in cerebrospinal fluid by competitive PCR in AIDS patients with with Epstein-Barr virus: primary infection or reactivation? Acta Neurol
different HCMV central nervous system diseases. Scand J Infect Dis. Scand. 1997;96:416420.
1995;27:559561. 104. Tsutsumi H, Kamazaki H, Nakata S, et al. Sequential development of
74. Cohen BA. Prognosis and response to therapy of cytomegalovirus enceph- acute meningoencephalitis and transverse myelitis caused by Epstein-Barr
alitis and meningomyelitis in AIDS. Neurology. 1996;46:444450. virus during infectious mononucleosis. Pediatr Infect Dis J. 1994;13:
75. Enting R, de Gans J, Reiss P, et al. Ganciclovir/foscarnet for cytomegalo- 665667.
virus meningoencephalitis in AIDS. Lancet. 1992;340:559560. 105. Schiff JA, Schaefer JA, Robinson JE. Epstein-Barr virus in cerebrospi-
76. Anders HJ, Weiss N, Bogner JR, et al. Ganciclovir and foscarnet efficacy nal fluid during infectious mononucleosis encephalitis. Yale J Biol Med.
in AIDS-related CMV polyradiculopathy. J Infect. 1998;36:2933. 1982;55:5963.
77. Decker CF, Tarver JH III, Murray DF, et al. Prolonged concurrent use 106. Imai S, Usui N, Sugiura M, et al. Epstein-Barr virus genomic sequences
of ganciclovir and foscarnet in the treatment of polyradiculopathy due and specific antibodies in cerebrospinal fluid in children with neurologic
to cytomegalovirus in a patient with AIDS. Clin Infect Dis. 1994;19: complications of acute and reactivated EBV infections. J Med Virol. 1993;
548549. 40:278284.
78. Domingo P, Puig M, Iranzo A, et al. Polyradiculopathy due to cytomega- 107. Pedneault L, Katz BZ, Miller G. Detection of Epstein-Barr virus in the
lovirus infection: report of a case in which an AIDS patient responded to brain by the polymerase chain reaction. Ann Neurol. 1992;32:184192.
foscarnet therapy. Clin Infect Dis. 1994;18:10191021. 108. Halsted CC, Chang RS. Infectious mononucleosis and encephalitis: recov-
79. Karmochkine M, Molina JM, Scieux C, et al. Combined therapy with ery of EB virus from spinal fluid. Pediatrics. 1979;64:257258.
ganciclovir and foscarnet for cytomegalovirus polyradiculomyelitis in pa- 109. Meerbach A, Gruhn B, Egerer R, et al. Semiquantitative PCR analysis
tients with AIDS. Am J Med. 1994;97:196197. of Epstein-Barr virus DNA in clinical samples of patients with EBV-
80. Kim YS, Hollander H. Polyradiculopathy due to cytomegalovirus: report associated diseases. J Med Virol. 2001;65:348357.
of two cases in which improvement occurred after prolonged therapy and 110. Weinberg A, Li S, Palmer M, et al. Quantitative CSF PCR in Epstein-
review of the literature. Clin Infect Dis. 1993;17:3237. Barr virus infections of the central nervous system. Ann Neurol. 2002;52:
81. Smith IL, Shinkai M, Freeman WR, et al. Polyradiculopathy associated 543548.
with ganciclovir-resistant cytomegalovirus in an AIDS patient: phenotypic 111. Tynell E, Aurelius E, Brandell A, et al. Acyclovir and prednisolone treat-
and genotypic characterization of sequential virus isolates. J Infect Dis. ment of acute infectious mononucleosis: a multicenter, double-blind,
1996;173:14811484. placebo-controlled study. J Infect Dis. 1996;174:324331.
82. Tokumoto JI, Hollander H. Cytomegalovirus polyradiculopathy caused 112. Robbins FC, de Quadros CA. Certification of the eradication of
by a ganciclovir-resistant strain. Clin Infect Dis. 1993;17:854856. indigenous transmission of wild poliovirus in the Americas. J Infect Dis.
83. Sadler M, Morris-Jones S, Nelson M, et al. Successful treatment of 1997;175(suppl 1):S281S285.
cytomegalovirus encephalitis in an AIDS patient using cidofovir. AIDS. 113. American Academy of Pediatrics Committee on Infectious Disease.
1997;11:12931294. Poliovirus. Pediatrics. 2011;128:805808.
84. Chik KW, Chan PK, Li CK, et al. Human herpesvirus-6 encephalitis after 114. Kew O, Morris-Glasgow V, Landaverde M, et al. Outbreak of polio-
unrelated umbilical cord blood transplant in children. Bone Marrow myelitis in Hispanola associated with type 1 vaccine-derived poliovirus.
Transplant. 2002;29:991994. Science. 2002;296:356359.
85. Drobyski WR, Knox KK, Majewski D, et al. Brief report: fatal encepha- 115. Gumede N, Muthambi V, Schoub BD. Immunodeficiency-associated vac-
litis due to variant B human herpesvirus-6 infection in a bone marrow- cine-derived poliovirus type 3 in infant, South Africa, 2011. Emerg Infect
transplant recipient. N Engl J Med. 1994;330:13561360. Dis. 2012;18:992994.
86. Mori Y, Miyamoto T, Nagafuji K, et al. High incidence of human her- 116. DeVries AS, Harper J, Murray A, et al. Vaccine-derived poliomy-
pes virus 6-associated encephalitis/myelitis following a second unrelated elitis 12 years after infection in Minnesota. N Engl J Med. 2011;364:
cord blood transplantation. Biol Blood Marrow Transplant. 2010;16: 23162323.
15961602. 117. Joffret ML, Jegouic S, Bessaud M, et al. Common and diverse features of
87. Hill AE, Hicks EM, Coyle PV. Human herpes virus 6 and central nervous cocirculating type 2 and 3 recombinant vaccine-derived polioviruses iso-
system complications. Dev Med Child Neurol. 1994;36:651652. lated from patients with poliomyelitis and healthy children. J Infect Dis.
88. Mackenzie IR, Carrigan DR, Wiley CA. Chronic myelopathy associated 2012;205:13631373.
with human herpesvirus-6. Neurology. 1995;45:20152017. 118. Kew O, Morris-Glasgow V, Landaverde M, et al. Outbreak of poliomyeli-
89. Ward KN, White RP, Mackinnon S, et al. Human herpesvirus-7 infection tis in Hispaniola associated with circulating type 1 vaccine-derived polio-
of the CNS with acute myelitis in an adult bone marrow recipient. Bone virus. Science. 2002;296:356359.
Marrow Transplant. 2002;30:983985. 119. Horstmann DM. Epidemiology of poliomyelitis and allied diseases1963.
90. Cohen JI. Epstein-Barr virus infection. N Engl J Med. 2000;343:481492. Yale J Biol Med. 1963;36:526.
91. Caldas C, Bernicker E, Nogare AD, et al. Case report: transverse myelitis 120. Price RW, Plum, F. Poliomyelitis. In: Vinken PJ, Bruyn GW, eds. Infections
associated with Epstein-Barr virus infection. Am J Med Sci. 1994;307: of the Nervous System (Part II). Amsterdam: North Holland Publishing;
4548. 1978:93132.
92. Junker AK, Roland EH, Hahn G. Transverse myelitis and Epstein-Barr 121. Malzberg MS, Rogg JM, Tate CA, et al. Poliomyelitis: hyperintensity
virus infection with delayed antibody responses. Neurology. 1991;41: of the anterior horn cells on MR images of the spinal cord. AJR Am J
15231524. Roentgenol. 1993;161:863865.
93. Clevenbergh P, Brohee P, Velu T, et al. Infectious mononucleosis compli- 122. Choudhary A, Sharma S, Sankhyan N, et al. Midbrain and spinal cord
cated by transverse myelitis: detection of the viral genome by polymerase magnetic resonance imaging (MRI) changes in poliomyelitis. J Child
chain reaction in the cerebrospinal fluid. J Neurol. 1997;244:592594. Neurol. 2010;25:497499.
94. Feinberg WM, Zonis J, Minnich LL. Epstein-Barr virus-associated my- 123. Rotbart HA, Webster AD. Treatment of potentially life-threatening
elopathy in an adult. Arch Neurol. 1984;41:454455. enterovirus infections with pleconaril. Clin Infect Dis. 2001;32:228235.
95. Grose C, Feorino PM. Epstein-Barr virus and transverse myelitis. Lancet. 124. Bodian DH, Hortsmann DM. Polioviruses. In: Horsefall FL, Tamm
1973;1:892. I, eds. Viral and Ricketsial Infections of Man. 4th ed. Philadelphia: JB
96. Majid A, Galetta SL, Sweeney CJ, et al. Epstein-Barr virus myeloradiculi- Lippincott; 1965:430473.
tis and encephalomyeloradiculitis. Brain. 2002;125:159165. 125. Jubelt B, Agre JC. Characteristics and management of postpolio syn-
97. Phowthongkum P, Phantumchinda K, Jutivorakool K, et al. Basal ganglia drome. JAMA. 2000;284:412414.
and brainstem encephalitis, optic neuritis, and radiculomyelitis in Epstein- 126. Gonzalez H, Olsson T, Borg K. Management of postpolio syndrome.
Barr virus infection. J Infect. 2007;54:e141e144. Lancet Neurol. 2010;9:634642.
98. Gruhn B, Meerbach A, Egerer R, et al. Successful treatment of Epstein-Barr 127. Thorsteinsson G. Management of postpolio syndrome. Mayo Clin Proc.
virus-induced transverse myelitis with ganciclovir and cytomegalovirus 1997;72:627638.
hyperimmune globulin following unrelated bone marrow transplantation. 128. Koopman FS, Uegaki K, Gilhus NE, et al. Treatment for postpolio syn-
Bone Marrow Transplant. 1999;24:13551358. drome. Cochrane Database Syst Rev. 2011;(2):CD007818.
99. Corssmit EP, Leverstein-van Hall MA, Portegies P, et al. Severe neuro- 129. Graber D, Fossoud C, Grouteau E, et al. Acute transverse myelitis and
logical complications in association with Epstein-Barr virus infection. coxsackie A9 virus infection. Pediatr Infect Dis J. 1994;13:77.
J Neurovirol. 1997;3:460464. 130. Dery P, Marks MI, Shapera R. Clinical manifestations of coxsackievirus
100. Morgenlander JC. A syndrome of concurrent central and peripheral infections in children. Am J Dis Child. 1974;128:464468.
nervous system involvement due to Epstein-Barr virus infection. Muscle 131. Jadoul C, Van Goethem J, Martin JJ. Myelitis due to coxsackievirus B
Nerve. 1996;19:10371039. infection. Neurology. 1995;45:16261627.
101. Tselis A, Duman R, Storch GA, et al. Epstein-Barr virus encephalomyelitis 132. Katamura K, Hattori H, Kunishima T, et al. Non-progressive viral myeli-
diagnosed by polymerase chain reaction: detection of the genome in the tis in X-linked agammaglobulinemia. Brain Dev. 2002;24:109111.
CSF. Neurology. 1997;48:13511355. 133. Matthews TG, Bailey SC. Transverse myelitisassociation with coxsackie
102. Wong M, Connolly AM, Noetzel MJ. Poliomyelitis-like syndrome B3 infection? Arch Dis Child. 1977;52:518519.
associated with Epstein-Barr virus infection. Pediatr Neurol. 1999;20: 134. Barak Y, Schwartz JF. Acute transverse myelitis associated with ECHO
235237. type 5 infection. Am J Dis Child. 1988;142:128.

Scheld_Ch21.indd 328 2/21/14 5:40 PM


Chapter 21: Acute Viral Myelitis 329

135. Bell EJ, Russell SJ. Acute transverse myelopathy and echo-2 virus infec- 167. Li J, Loeb JA, Shy ME, et al. Asymmetric flaccid paralysis: a neuromus-
tion. Lancet. 1963;2:12261227. cular presentation of West Nile virus infection. Ann Neurol. 2003;53:
136. Johnson DA, Eger AW. Myelitis associated with an echovirus. JAMA. 703710.
1967;201:637638. 168. Gadoth N, Weitzman S, Lehmann EE. Acute anterior myelitis complicat-
137. Kibe T, Fujimoto S, Ishikawa T, et al. Serial MRI findings of benign polio- ing West Nile fever. Arch Neurol. 1979;36:172173.
myelitis. Brain Dev. 1996;18:147149. 169. Ohry A, Karpin H, Yoeli D, et al. West Nile virus myelitis. Spinal Cord.
138. Starlin R, Reed N, Leeman B, et al. Acute flaccid paralysis syndrome 2001;39:662663.
associated with echovirus 19, managed with pleconaril and intravenous 170. Hainline ML, Kincaid JC, Carpenter DL, et al. West Nile poliomyelitis in
immunoglobulin. Clin Infect Dis. 2001;33:730732. a 7-year-old child. Pediatr Neurol. 2008;39:350354.
139. Takahashi S, Miyamoto A, Oki J, et al. Acute transverse myelitis caused 171. Sejvar JJ, Davis LE, Szabados E, et al. Delayed-onset and recurrent
by ECHO virus type 18 infection. Eur J Pediatr. 1995;154:378380. limb weakness associated with West Nile virus infection. J Neurovirol.
140. Berger MM, Kopp N, Vital C, et al. Detection and cellular localiza- 2010;16:93100.
tion of enterovirus RNA sequences in spinal cord of patients with ALS. 172. Zafar SF, Ubogu EE. Subacute brachial diplegia associated with West Nile
Neurology. 2000;54:2025. virus myelitis. Muscle Nerve. 2012;45:900904.
141. Giraud P, Beaulieux F, Ono S, et al. Detection of enteroviral sequences 173. Petropoulou KA, Gordon SM, Prayson RA, et al. West Nile virus
from frozen spinal cord samples of Japanese ALS patients. Neurology. meningoencephalitis: MR imaging findings. AJNR Am J Neuroradiol.
2001;56:17771778. 2005;26:19861995.
142. Walker MP, Schlaberg R, Hays AP, et al. Absence of echovirus sequences 174. Sejvar JJ. The long-term outcomes of human West Nile virus infection.
in brain and spinal cord of amyotrophic lateral sclerosis patients. Ann Clin Infect Dis. 2007;44:16171624.
Neurol. 2001;49:249253. 175. Sejvar JJ, Bode AV, Marfin AA, et al. West Nile Virus-associated flaccid
143. Schmidt NJ, Lennette EH, Ho HH. An apparently new enterovirus iso- paralysis outcome. Emerg Infect Dis. 2006;12:514516.
lated from patients with disease of the central nervous system. J Infect 176. Sejvar JJ, Haddad MB, Tierney BC, et al. Neurologic manifestations and
Dis. 1974;129:304309. outcome of West Nile virus infection. JAMA. 2003;290:511515.
144. Huang CC, Liu CC, Chang YC, et al. Neurologic complications in chil- 177. Verma R, Praharaj HN, Patil TB, et al. Acute transverse myelitis follow-
dren with enterovirus 71 infection. N Engl J Med. 1999;341:936942. ing Japanese encephalitis viral infection: an uncommon complication of a
145. McMinn P, Stratov I, Nagarajan L, et al. Neurological manifestations common disease. BMJ Case Rep. 2012;2012.
of enterovirus 71 infection in children during an outbreak of hand, 178. Solomon T, Kneen R, Dung NM, et al. Poliomyelitis-like illness due to
foot, and mouth disease in Western Australia. Clin Infect Dis. 2001;32: Japanese encephalitis virus. Lancet. 1998;351:10941097.
236242. 179. Fukuda H, Umehara F, Kawahigashi N, et al. Acute disseminated myelitis
146. Li CC, Yang MY, Chen RF, et al. Clinical manifestations and laboratory after Japanese B encephalitis vaccination. J Neurol Sci. 1997;148:
assessment in an enterovirus 71 outbreak in southern Taiwan. Scand J 113115.
Infect Dis. 2002;34:104109. 180. Matsui M, Kawano H, Matsukura M, et al. Acute transverse myelitis
147. Shen WC, Tsai C, Chiu H, et al. MRI of Enterovirus 71 myelitis with after Japanese B encephalitis vaccination in a 4-year-old girl. Brain Dev.
monoplegia. Neuroradiology. 2000;42:124127. 2002;24:187189.
148. Alexander JP Jr, Baden L, Pallansch MA, et al. Enterovirus 71 infec- 181. Ohtaki E, Matsuishi T, Hirano Y, et al. Acute disseminated enceph-
tions and neurologic diseaseUnited States, 19771991. J Infect Dis. alomyelitis after treatment with Japanese B encephalitis vaccine
1994;169:905908. (Nakayama-Yoken and Beijing strains). J Neurol Neurosurg Psychiatry.
149. Chumakov M, Voroshilova M, Shindarov L, et al. Enterovirus 71 isolated 1995;59:316317.
from cases of epidemic poliomyelitis-like disease in Bulgaria. Arch Virol. 182. Ohtaki E, Murakami Y, Komori H, et al. Acute disseminated encephalo-
1979;60:329340. myelitis after Japanese B encephalitis vaccination. Pediatr Neurol. 1992;8:
150. Hayward JC, Gillespie SM, Kaplan KM, et al. Outbreak of poliomy- 137139.
elitis-like paralysis associated with enterovirus 71. Pediatr Infect Dis J. 183. Aendekerk RP, Schrivers AN, Koehler PJ. Tick-borne encephalitis compli-
1989;8:611616. cated by a polio-like syndrome following a holiday in central Europe. Clin
151. Melnick JL. Enterovirus type 71 infections: a varied clinical pattern some- Neurol Neurosurg. 1996;98:262264.
times mimicking paralytic poliomyelitis. Rev Infect Dis. 1984;6(suppl. 2): 184. Bender A, Schulte-Altedorneburg G, Walther EU, et al. Severe tick borne
S387S390. encephalitis with simultaneous brain stem, bithalamic, and spinal cord
152. Samuda GM, Chang WK, Yeung CY, et al. Monoplegia caused by involvement documented by MRI. J Neurol Neurosurg Psychiatry. 2005;
Enterovirus 71: an outbreak in Hong Kong. Pediatr Infect Dis J. 1987;6: 76:135137.
206208. 185. Zambito Marsala S, Francavilla E, Gioulis M, et al. Isolated polio-like
153. Nagy G, Takatsy S, Kukan E, et al. Virological diagnosis of enterovirus syndrome after tick-borne encephalitis presenting with acute hyperck-
type 71 infections: experiences gained during an epidemic of acute CNS emia. Neurol Sci. 2012;33:669672.
diseases in Hungary in 1978. Arch Virol. 1982;71:217227. 186. Dumpis U, Crook D, Oksi J. Tick-borne encephalitis. Clin Infect Dis.
154. Jang S, Suh SI, Ha SM, et al. Enterovirus 71-related encephalomyelitis: 1999;28:882890.
usual and unusual magnetic resonance imaging findings. Neuroradiology. 187. Bohus M, Glocker FX, Jost S, et al. Myelitis after immunisation against
2012;54:239245. tick-borne encephalitis. Lancet. 1993;342:239240.
155. Beeri R, Golan G, Newman D, et al. Transverse myelitis heralding hepati- 188. Owen NL. Myelitis following type A2 influenza. JAMA. 1971;215:
tis A. J Clin Gastroenterol. 1995;20:262263. 19861987.
156. Breningstall GN, Belani KK. Acute transverse myelitis and brainstem 189. Salonen O, Koshkiniemi M, Saari A, et al. Myelitis associated with influ-
encephalitis associated with hepatitis A infection. Pediatr Neurol. enza A virus infection. J Neurovirol. 1997;3:8385.
1995;12:169171. 190. Landau YE, Grisaru-Soen G, Reif S, et al. Pediatric neurologic compli-
157. Nash D, Mostashari F, Fine A, et al. The outbreak of West Nile virus infection cations associated with influenza A H1N1. Pediatr Neurol. 2011;44:
in the New York City area in 1999. N Engl J Med. 2001;344:18071814. 4751.
158. Petersen LR, Marfin AA. West Nile virus: a primer for the clinician. Ann 191. Akkad W, Salem B, Freeman JW, et al. Longitudinally extensive trans-
Intern Med. 2002;137:173179. verse myelitis following vaccination with nasal attenuated novel influenza
159. Ahmed S, Libman R, Wesson K, et al. Guillain-Barre syndrome: an unusual A(H1N1) vaccine. Arch Neurol. 2010;67:10181020.
presentation of West Nile virus infection. Neurology. 2000;55:144146. 192. Ambrose CS, Loop B, Miday R. A case report of transverse myelitis fol-
160. Centers for Disease Control and Prevention. Acute flaccid paralysis lowing influenza vaccination. Arch Neurol. 2011;68:1085; author reply
syndrome associated with West Nile virus infectionMississippi and 10851086.
Louisiana, JulyAugust 2002. MMWR Morb Mortal Wkly Rep. 2002;51: 193. Bakshi R, Mazziotta JC. Acute transverse myelitis after influenza vaccination:
825828. magnetic resonance imaging findings. J Neuroimaging. 1996;6:248250.
161. Glass JD, Samuels O, Rich MM. Poliomyelitis due to West Nile virus. 194. Holt S, Hudgins D, Krishnan KR, et al. Diffuse myelitis associated with
N Engl J Med. 2002;347:12801281. rubella vaccination. Br Med J. 1976;2:10371038.
162. Jeha LE, Sila CA, Lederman RJ, et al. West Nile virus infection: a new 195. Grattan-Smith PJ, Procopis PG, Wise GA, et al. Serious neurological com-
acute paralytic illness. Neurology. 2003;61:5559. plications of measlesa continuing preventable problem. Med J Aust.
163. Leis AA, Stokic DS, Polk JL, et al. A poliomyelitis-like syndrome from 1985;143:385387.
West Nile virus infection. N Engl J Med. 2002;347:12791280. 196. Nussinovitch M, Brand N, Frydman M, et al. Transverse myelitis follow-
164. Al-Shekhlee A, Katirji B. Electrodiagnostic features of acute paralytic ing mumps in children. Acta Paediatr. 1992;81:183184.
poliomyelitis associated with West Nile virus infection. Muscle Nerve. 197. Inoue J, Ueno Y, Kogure T, et al. Analysis of the full-length genome of
2004;29:376380. hepatitis B virus in the serum and cerebrospinal fluid of a patient with
165. Leis AA, Fratkin J, Stokic DS, et al. West Nile poliomyelitis. Lancet Infect acute hepatitis B and transverse myelitis. J Clin Virol. 2008;41:301304.
Dis. 2003;3:910. 198. Scheibe F, Hofmann J, Ruprecht K. Parainfectious myelitis associated with
166. Leis AA, Stokic DS, Webb RM, et al. Clinical spectrum of muscle parvovirus B19 infection. J Neurol. 2010;257:15571558.
weakness in human West Nile virus infection. Muscle Nerve. 2003;28: 199. Kinomoto K, Okamoto Y, Yuchi Y, et al. Acute encephalomyelitis associ-
302308. ated with acute viral hepatitis type B. Intern Med. 2009;48:241243.

Scheld_Ch21.indd 329 2/21/14 5:40 PM


330 Part II: Viral Infections and Related Disorders

200. Stubgen JP. Immune-mediated myelitis associated with hepatitis virus in- 214. Pradhan S, Gupta RK, Kapoor R, et al. Parainfectious conus myelitis.
fections. J Neuroimmunol. 2011;239:2127. J Neurol Sci. 1998;161:156162.
201. Stich O, Herpers M, Keil A, et al. JC virus myelitis without cerebral 215. Hoffman HL. Acute necrotic myelopathy. Brain. 1955;78:377394.
involvement in acute myeloid leukemia. Eur J Neurol. 2011;18:e143e144. 216. Katz JD, Ropper AH. Progressive necrotic myelopathy: clinical course in
202. Linssen WH, Gabreels FJ, Wevers RA. Infective acute transverse myelopa- 9 patients. Arch Neurol. 2000;57:355361.
thy: report of two cases. Neuropediatrics. 1991;22:107109. 217. Tippett DS, Fishman PS, Panitch HS. Relapsing transverse myelitis.
203. Matsui M, Kakigi R, Watanabe S, et al. Recurrent demyelinating trans- Neurology. 1991;41:703706.
verse myelitis in a high titer HBs-antigen carrier. J Neurol Sci. 1996;139: 218. Marrie TJ, Purdy RA, Johnston BL, et al. Encephalomyeloradiculopathy
235237. of infectious or parainfectious etiologya new entity? Clin Infect Dis.
204. al Deeb SM, Yaqub BA, Bruyn GW, et al. Acute transverse myelitis. 1995;20:945953.
A localized form of postinfectious encephalomyelitis. Brain. 1997; 219. Choi KH, Lee KS, Chung SO, et al. Idiopathic transverse myelitis: MR
120(pt 7):11151122. characteristics. AJNR Am J Neuroradiol. 1996;17:11511160.
205. Altrocchi PH. Acute transverse myelopathy. Arch Neurol. 1963;9:111119. 220. Holtas S, Basibuyuk N, Fredriksson K. MRI in acute transverse myelopa-
206. Berman M, Feldman S, Alter M, et al. Acute transverse myelitis: incidence thy. Neuroradiology. 1993;35:221226.
and etiologic considerations. Neurology. 1981;31:966971. 221. Sanders KA, Khandji AG, Mohr JP. Gadolinium-MRI in acute transverse
207. Jeffery DR, Mandler RN, Davis LE. Transverse myelitis. Retrospective myelopathy. Neurology. 1990;40:16141616.
analysis of 33 cases, with differentiation of cases associated with multiple 222. Kalita J, Misra UK. Neurophysiological studies in acute transverse myeli-
sclerosis and parainfectious events. Arch Neurol. 1993;50:532535. tis. J Neurol. 2000;247:943948.
208. Lipton HL, Teasdall RD. Acute transverse myelopathy in adults. A fol- 223. Defresne P, Meyer L, Tardieu M, et al. Efficacy of high dose steroid ther-
low-up study. Arch Neurol. 1973;28:252257. apy in children with severe acute transverse myelitis. J Neurol Neurosurg
209. Dunne K, Hopkins IJ, Shield LK. Acute transverse myelopathy in child- Psychiatry. 2001;71:272274.
hood. Dev Med Child Neurol. 1986;28:198204. 224. Sbire G, Hollenberg H, Meyer L, et al. High dose methylprednisolone in
210. Knebusch M, Strassburg HM, Reiners K. Acute transverse myelitis in severe acute transverse myelopathy. Arch Dis Child. 1997;76:167168.
childhood: nine cases and review of the literature. Dev Med Child Neurol. 225. Kalita J, Misra UK. Is methyl prednisolone useful in acute transverse
1998;40:631639. myelitis? Spinal Cord. 2001;39:471476.
211. Lahat E, Pillar G, Ravid S, et al. Rapid recovery from transverse 226. Kalita J, Misra UK, Mandal SK. Prognostic predictors of acute transverse
myelopathy in children treated with methylprednisolone. Pediatr Neurol. myelitis. Acta Neurol Scand. 1998;98:6063.
1998;19:279282. 227. Ropper AH, Poskanzer DC. The prognosis of acute and subacute trans-
212. Paine RS, Byers RK. Transverse myelopathy in childhood. AMA Am J Dis verse myelopathy based on early signs and symptoms. Ann Neurol.
Child. 1953;85:151163. 1978;4:5159.
213. Frohman EM, Wingerchuk DM. Clinical practice: transverse myelitis. 228. Irani DN, Kerr DA. 14-3-3 protein in the cerebrospinal fluid of patients
N Engl J Med. 2010;363:564572. with acute transverse myelitis. Lancet. 2000;355:901.

Scheld_Ch21.indd 330 2/21/14 5:40 PM


CHAPTER 22 POSTINFECTIOUS
ENCEPHALOMYELITIS
KAREN L. ROOS AND AUGUSTO MIRAVALLE

Postinfectious encephalomyelitis is an acute monophasic dis- Although postinfectious encephalomyelitis can occur sponta-
order of the central nervous system (CNS) that occurs within neously, most often it follows a precipitating event (Table 22.1).
days to weeks of a viral illness or a vaccination. The antecedent As stated, the major preceding event is either a viral infection or a
viral illness is typically either an upper respiratory tract infec- vaccination. Measles, varicella, rubella, mumps, and influenza A
tion or a nonspecific febrile illness. In the past, most cases were and B viruses have all been associated with the development of
associated with the exanthematous diseases (vaccinia, measles, postinfectious encephalomyelitis. The other major inciting event
varicella, and rubella) (1). Although postinfectious encepha- is vaccination, particularly in cases involving enveloped viruses
lomyelitis has a clear temporal relationship with infection such as smallpox (vaccinia virus) and rabies. In addition to viral
or immunization, it is not the result of primary neural tissue infection and vaccination, other less common etiologies associ-
invasion by an organism. Infectious agents are rarely identified ated with postinfectious encephalomyelitis are bacterial infec-
or recovered from neural tissue (2). The disease is instead an tions and autoimmune and hematologic disorders (5,1133).
immune-mediated disease triggered by an infectious agent or
an immunization.
TA B L E 2 2 . 1
HISTORY CAUSES OF POSTINFECTIOUS/POSTVACCINAL
ENCEPHALOMYELITIS
One of the earliest descriptions of postinfectious encephalitis
was recorded in 1790 of a 23-year-old woman who developed Viruses
symptoms of encephalitis following smallpox (3). This was
Measles
followed by reports of several neurologic disorders following
smallpox infection (4). In 1905, a case of encephalitis after Varicella
the jennerian cowpox inoculation was reported in France, Rubella
and another case was observed in the London Hospital in Vaccinia
1912 (5). The disease was recognized as a well-defined entity
in 1922, when 11 fatalities due to postinfectious encephalitis Mumps
were reported in Great Britain (4). Influenza A and B
Paralysis and encephalitis is a well-known complication of Hepatitis C
animal brain tissuederived rabies vaccine. In developed coun- Human immunodeficiency virus
tries, these have been replaced by the use of commercial tissue
culture rabies vaccines, but animal brain tissuederived rabies Vaccinations
vaccines continue to be used in many areas of the world. Smallpox
In 1941, Weston Hurst (6) described a syndrome with similar Rabies
clinical presentation to postinfectious encephalomyelitis but with Measles
a worse prognosis. This entity was characterized by the presence
of petechial hemorrhages around blood vessels, intense numbers Rubella
of polymorphonuclear leukocytes, perivenular demyelination, Oral poliovirus
necrosis, and fibrin deposits on pathologic examination. This dis- Japanese encephalitis
ease was called acute hemorrhagic leukoencephalomyelitis (6). Recombinant hepatitis B
On the basis of the preceding illness, postinfectious encephalo-
myelitis has also been called parainfectious, postexanthematous, Tetanus toxoid
postvaccinal, and postinfluenzal encephalomyelitis. In reference to Bacterial infections
the pathology, this illness is also known as acute disseminated en- Streptococcus pyogenes
cephalomyelitis (ADEM), perivascular myelinoclasis, perivenous Legionella pneumophila
encephalitis, acute demyelinating encephalomyelitis, immune-
mediated or hyperergic encephalomyelitis, and disseminated vas- Legionella cincinnatiensis
culomyelinopathy (7). These terms are used interchangeably. Leptospira interrogans
Rickettsia rickettsii
ETIOLOGY Mycoplasma pneumoniae
Plasmodium falciparum
The incidence of postinfectious encephalomyelitis from differ- Immune disorders
ent causes has been reported to be between 0.4 and 0.8 per
Autologous peripheral blood stem cell
100,000 of population, with a median age of onset of 4.5 to
7.5 years in pediatric studies and 33.5 years in a study of adult Lupus
patients (8). The disease has a seasonal peak in winter and Autoimmune hemolytic anemia
spring, consistent with its putative infectious etiologies (9,10).

331

Scheld_Ch22.indd 331 2/21/14 10:48 PM


332 Part II: Viral Infections and Related Disorders

(24), following leptospirosis (25), typhoid fever (26), and in the


Viral Etiologies recovery phase from Rocky Mountain spotted fever (28).
A postmalaria neurologic syndrome has been described, charac-
Postmeasles encephalomyelitis is the most common CNS com- terized as acute onset of convulsions, acute confusional state, dys-
plication of measles virus infection, with an estimated incidence phasia, acute psychosis, tremor, myoclonus, and ataxia in patients
of 1 to 2 in 1,000 cases of measles. The onset of symptoms of recovering from Plasmodium falciparum malaria. Giemsa-stained
postmeasles encephalomyelitis is variable. Typically, after the smears of peripheral blood must be negative at the time of symp-
rash is fading, fever suddenly returns, associated with head- tom onset, distinguishing this syndrome from cerebral malaria,
aches, vomiting, and signs of meningeal irritation. Headache is which occurs during parasitemia. The development of the syn-
invariably an early feature and often relieved by lumbar punc- drome can be up to 9 weeks (median, 4 days) from eradication
ture. If the spinal cord is involved, there is backache, progres- of the systemic parasitemia (29). Postinfectious encephalomyelitis
sive lower extremity weakness, and urinary retention (13). has been reported as a complication of Mycoplasma pneumoniae
Postinfectious encephalomyelitis as a complication of vari- infection (30), after autologous stem cell transplantation (31), and
cella virus infection is rare and occurs in about 1 in 10,000 cases in association with lupus (32) and autoimmune hemolytic anemia
of chickenpox. The onset of symptoms is usually between 4 and (33). Whether this is simply a chance association or these diseases
14 days after the appearance of the rash, with sudden fever, have a specific role in the pathogenesis is unclear.
ataxia, seizures, drowsiness, stupor, and obtundation (13).
Postinfectious encephalomyelitis is thought to occur in
approximately 1 out of 5,000 children with rubella infection.
Signs and symptoms of neurologic involvement occur within the
CLINICAL PRESENTATION
first week after the onset of the rash. The presentation is usually The presentation of postinfectious encephalomyelitis is usually
very severe with convulsions and sudden loss of consciousness. characterized by abrupt onset of neurologic symptoms days to
Headache and meningeal signs are also common (13). weeks after a viral illness or vaccination. Nevertheless, a clear
Postinfectious encephalomyelitis has been reported in associ- preceding infection or vaccination cannot be found in up to one
ation with hepatitis C virus infection and as a primary manifesta- third of children and half of adults presenting with disease (8,40).
tion of human immunodeficiency virus (HIV) infection (1416). In those cases, systemic symptoms, including fever (43% to 52%),
headache (45% to 58%), malaise, and myalgias may occur shortly
Vaccination before the appearance of neurologic signs and symptoms (41).
Because of the widespread involvement of the optic nerves,
At present, less than 5% of all postinfectious encephalomyeli- brain, and spinal cord, postinfectious encephalomyelitis usu-
tis cases follow immunization. Postvaccinal encephalomyelitis ally presents as a polysymptomatic, monophasic, multifocal
has been associated with immunization for rabies, hepatitis B, neurologic demyelinating disease. Obtundation and depressed
influenza, Japanese B encephalitis, diphtheria/pertussis/tetanus, consciousness, in addition to unilateral or bilateral long tract
measles, mumps, rubella, pneumococcus, polio, smallpox, and signs (85%), acute hemiparesis (76%), and ataxia (59%), are
varicella (Table 22.1) (34). Postvaccinal encephalomyelitis usu- the most common presentations. Cranial nerve deficits may
ally occurs 7 to 14 days after vaccination, but cases have been be present because of involvement of the corticobulbar fibers
reported as early as 1 day and as late as 23 days following vac- to the motor nuclei of the cranial nerves. These signs may be
cination (35). The risk is usually increased directly with increasing associated with an altered level of consciousness ranging from
age of primary vaccination after the first year of life (36). In gen- lethargy to coma (42). Focal or generalized tonic-clonic sei-
eral, postvaccinal encephalomyelitis occurs more frequently in zures and psychosis may also be part of the initial presentation
primary vaccinees than in revaccinees. Complications in revaccin- (43). Postinfectious encephalomyelitis can be distinguished
ees occur in individuals who have not been vaccinated for many clinically from acute viral encephalitis by the predominance of
years, and therefore react like primary vaccinees, or in individuals subcortical white matter involvement. In contrast, viral enceph-
who have acquired immunodeficiency disorders (37). During the alitis usually presents with predominantly cortical features,
1947 smallpox outbreak in New York City, the reported incidence including confusion, aphasia, and convulsions. Other com-
of postvaccinal encephalomyelitis was 1 in 100,000 (35). In 1968, mon presenting signs and symptoms are listed in Table 22.2.
5,594,000 primary smallpox vaccinations and 857,400 revac-
cinations were given in the United States. The overall incidence
of postvaccinal encephalomyelitis was 2.9 per 1 million primary TA B L E 2 2 . 2
vaccinations. None of the revaccinees developed postvaccinal COMMON PRESENTING SYMPTOMS IN
encephalomyelitis. The case-fatality rate of postvaccinal encepha- POSTINFECTIOUS ENCEPHALOMYELITIS
lomyelitis between 1959 and 1966 was approximately 25% in the
United States (38) and 30% to 50% in Europe (4,39). Fever
The incidence of encephalitis associated with the live
Headaches
attenuated measles virus vaccine is thought to be 1.16 per
1,000,000 doses, with most cases occurring in the second week Meningismus
after immunization (19). Postvaccinal encephalomyelitis has Encephalopathy
been associated with the poliovirus vaccine (19), the Japanese Coma
encephalitis vaccine (20), the tetanus toxoid vaccine (21), and
Focal neurologic deficits (hemiparesis, cerebellar ataxia,
the recombinant hepatitis B vaccine (24).
cranial neuropathies)
Seizures
Other Infectious Agents Transverse myelitis
Aphasia
Streptococcus pyogenes has been reported as a causative agent
of postinfectious encephalomyelitis associated with acute glo- Movement disorders
merulonephritis (23). Postinfectious encephalomyelitis has been Psychosis
reported as a complication of Legionella pneumophila infection

Scheld_Ch22.indd 332 2/21/14 10:48 PM


Chapter 22: Postinfectious Encephalomyelitis 333

Interestingly, presenting symptoms may vary in pediatric


versus adult-onset postinfectious encephalomyelitis. Motor PATHOGENESIS
deficits can occur in both adult and pediatric cases. However,
sensory deficits and polyradiculoneuropathies are more fre- The pathology of postinfectious encephalomyelitis can be repro-
quently found in adults, whereas seizures predominate in duced in the animal model of experimental allergic encephalo-
pediatric cases. myelitis. This is a demyelinating disorder of the CNS induced
Even though postinfectious encephalomyelitis usually dis- in animals by immunization with myelin extracts, proteins, or
plays a monophasic disease course, rare cases of relapsing peptides found in myelin. In an effort to reproduce in animal
postinfectious encephalomyelitis have been described. In order models the lesions described in postvaccinal encephalomyelitis,
to fulfill definition of recurrent postinfectious encephalomy- Rivers and colleagues (47) in 1933 injected homogenates of nor-
elitis, the second clinical event should occur at least 3 months mal rabbit brains into monkeys. After 6 months, several mon-
from the initial event, without involvement of new clinical areas keys developed a lymphocytic infiltration and demyelination of
or magnetic resonance imaging (MRI) evidence of dissemina- the CNS tissue (47).
tion in time. It has also been suggested that in order to distin- Experimental allergic encephalomyelitis can be passively
guish recurrent postinfectious encephalomyelitis from multiple transferred to healthy animals by immune lymphocytes (48).
sclerosis (MS), the second event should not occur while the These activated T cells assume a novel functional phenotype
patients is receiving steroid treatment (44). Multiphasic postin- after transfer into a recipient animal that allows them to migrate
fectious encephalomyelitis is a term that has been assigned to the CNS and pass through the bloodbrain barrier. This mi-
to recurrent postinfectious encephalomyelitis cases where the gration briefly precedes the onset of clinical experimental allergic
second event represents a polysymptomatic presentation with encephalomyelitis. There is a minimal interval of 3 days between
involvement of a different anatomic area. In those cases, MRI the intravenous administration of pathogenic T cells and the
must show new areas of involvement with complete or partial onset of clinical experimental allergic encephalomyelitis (49).
resolution of previous lesions (10,44). Long-term clinical and T cells can potentially react with a wide variety of molecular
imaging follow-up has shown the resolution of lesions with structures, but normally they do not react against self-antigens.
no long-lasting neurologic impairments in most of these multi- However, some encephalitogenic CD4 and CD8 T lymphocytes
phasic cases (41). can be found in the blood, thymus, and secondary lymphoid
There is a long-standing controversy about whether a sec- tissues of apparently healthy individuals, but through the action
ond episode of postinfectious encephalomyelitis should be of suppressive cytokines, they usually do not attack the CNS.
called MS and treated accordingly. As a general rule, patients T cells become pathogenic only if activated. One possible sce-
who develop clinical evidence of dissemination in space and nario to explain the development of postinfectious encephalo-
time along with evidence of chronic demyelination in the CNS myelitis is that an infecting microbe expresses a peptide that is
will likely develop MS. The classic scenario is the patient, typi- structurally similar to myelin basic protein (MBP). This epitope
cally a child, who is diagnosed with postinfectious encepha- can trigger the activation of self-reactive T cells by a mechanism
lomyelitis following a viral infection, recovers, and then after known as molecular mimicry. Once activated, these cells can
some time develops recurrent symptoms with or without an multiply and mature into effector T cells, producing mediators
antecedent viral infection. Two criteria are helpful in making and cytokines that can react to normal self-antigens. In addition,
the correct diagnosis: (a) The development of new symptoms some T cells express more than one specific antigen receptor.
representing distinct areas of demyelination not involved in One receptor type could be specific for the myelin antigen and
the original episode favors the diagnosis of relapsing-remitting the other for the microbial antigen. Exposure to the microbial
MS, and (b) the appearance of new lesions on neuroimaging antigen could activate the T cell, which by virtue of its myelin-
supports the diagnosis of MS (45). It is worth remembering that specific alternative receptor could attack the CNS (49).
brain lesions of MS patients usually increase in size and num- Another mechanism proposed to explain the pathogenesis
ber during the course of the illness (41). In addition, chronic of postinfectious encephalomyelitis is the activation of self-
MS lesions appear as black holes on T1-weighted images, reactive immune cells by the release of cytokines by virus-
whereas black holes are not seen on T1-weighted images in mediated death of host cells. Penetration of these self-reactive
patients with postinfectious encephalomyelitis. As patients re- immune cells into the brain or spinal cord leads to the charac-
cover from postinfectious encephalomyelitis, there is evidence teristic pathology of postinfectious encephalomyelitis.
of complete or partial resolution of lesions on neuroimaging. The role of circulating humoral factors in the pathogen-
Oligoclonal bands should not persist in the cerebrospinal fluid esis of postinfectious encephalomyelitis is still unclear, but
(CSF) of patients with postinfectious encephalomyelitis, but several lines of evidence suggest that antibody production by
will either persist or appear over time in the CSF of patients the host may aid in limiting or preventing the disease presum-
with MS (41). ably by binding to MBP and inhibiting the access of autoreac-
Acute hemorrhagic leukoencephalitis is considered a hyper- tive T lymphocytes. Prostaglandins of the E series secreted by
acute form of postinfectious encephalomyelitis and has been blood monocytes and cerebral glial cells inhibit the immune
reported to occur in 2% of pediatric cases (46). On physi- response in experimental allergic encephalomyelitis by down-
cal examination, there may be meningismus, obtundation, regulation of monocytes and T cells and reduce the clinical
and lethargy, in addition to upper motor neuron signs, brain- and histologic abnormalities of experimental allergic encepha-
stem findings, transverse myelitis, and cranial neuropathies. lomyelitis in rats (12).
In general, maximum deficits are reached in the first week
from onset. Recovery usually starts to become clinically evi-
dent after the first week, with complete resolution of deficits DIAGNOSIS
and MRI lesions within 3 months (10). Although the progno-
sis is in general favorable, as high as 30% of patients require The diagnosis of postinfectious encephalomyelitis is based on
intensive care, with an estimated mortality rate of 20% (10). clinical history, the findings on neurologic examination, neu-
Despite the neuropathologic differences between postinfec- roimaging abnormalities of demyelination, and CSF analy-
tious encephalomyelitis and acute hemorrhagic leukoencepha- sis. Several attempts have been made to establish a series of
litis, it is possible that they represent a gradient of severity of clinical features that will increase the likelihood of the diag-
the same pathologic process. nosis of postinfectious encephalomyelitis. The International

Scheld_Ch22.indd 333 2/21/14 10:48 PM


334 Part II: Viral Infections and Related Disorders

TA B L E 2 2 . 3
DIAGNOSTIC CRITERIA OF POSTINFECTIOUS ENCEPHALOMYELITIS

Clinical Features

First clinical attack of inflammatory or demyelinating disease in the CNS


Acute or subacute onset
Affects multifocal areas of the CNS
Polysymptomatic presentation
Must include encephalopathy; acute behavioral change such as confusion or irritability and/or alteration in consciousness ranging
from somnolence to coma
Attack should be followed by improvement on clinical and/or neuroradiologic (MRI) measures.
Sequelae may include residual deficits.
No other etiologies can explain the event.
Relapses (with new or fluctuating symptoms, signs, or MRI findings) occurring within 3 months of the inciting episode are considered
part of the same acute event. In addition, relapses that occur during a steroid taper or within 4 weeks of completing a steroid taper
are considered part of the initial inciting episode.

MRI Features

Large (12 cm in size) multifocal, hyperintense, bilateral, asymmetric lesions in the supratentorial or infratentorial white matter
Rarely, brain MRI shows a single large (12 cm) lesion predominantly affecting white matter.
Gray matter, especially basal ganglia and thalamus, may be involved.
Spinal cord MRI may show confluent intramedullary lesion(s) with variable enhancement, in addition to the abnormalities on brain MRI.
No radiologic evidence of previous destructive white matter changes.

Encephalopathy is a required feature for the diagnosis of postinfectious encephalomyelitis but is not a typical feature of multiple sclerosis. In addition,
a cerebrospinal fluid pleocytosis 50 white blood cells per mm can be observed in postinfectious encephalomyelitis, whereas this finding is highly
atypical for multiple sclerosis.
From Krupp LB, Banwell B, Tenembaum S. Consensus definitions proposed for pediatric multiple sclerosis and related disorders. Neurology.
2007;68(16)(suppl 2):S7S12, with permission.)

Pediatric Multiple Sclerosis Study Group has developed a the diagnosis of postinfectious encephalomyelitis when using
series of criteria for the diagnosis of postinfectious encepha- these guidelines have not been evaluated to date.
lomyelitis (Table 22.3). These criteria have been developed Due to the broad spectrum of differential diagnosis in pa-
based on selected review of the literature and expert panel tients presenting with a monophasic demyelinating CNS disorder
discussion. The specificity, sensitivity, and biologic validity to (Table 22.4), the diagnosis of postinfectious encephalomyelitis

TA B L E 2 2 . 4
DIFFERENTIAL DIAGNOSIS IN PATIENTS PRESENTING WITH A DEMYELINATING
CENTRAL NERVOUS SYSTEM DISORDER

Diagnosis Characteristics

MS Relapses and remissions, optic neuritis, INO, history of fatigue, sensory symptoms, MRI
showing dissemination in time and space, presence of inflammatory markers on CSF
MS variants (Marburg variant, Tumefactive demyelinating lesions on brain MRI, polysymptomatic presentation
Bal concentric sclerosis)
NMO Optic neuritis, acute myelitis, contiguous spinal cord MRI lesion, presence of anti-
aquaporin-4 antibodies
Viral encephalitis (HSV, VZV, EBV, Encephalopathy, seizures, coma, cortical/brainstem symptoms, systemic manifestations
JC, HIV, Japanese encephalitis virus, (fever, vomiting, myalgias, cutaneous rash), lower motor neuron symptoms
WestNile virus) (poliomyelitis like syndromes associated with WNV)
SSPE Behavioral changes, myoclonic jerks, hemiparesis, cogwheel rigidity, dementia
PML/PML-IRIS Progressive cognitive decline, cortical symptoms, seizures, encephalopathy, history of
immunosuppression, widespread enhancement on T2/FLAIR high signal lesions on
brain MRI, lack of spinal cord involvement

INO, internuclear ophthalmoplegia; PML-IRIS, progressive multifocal leukoencephalopathy immune reconstitution inflammatory syndrome.

Scheld_Ch22.indd 334 2/21/14 10:48 PM


Chapter 22: Postinfectious Encephalomyelitis 335

TA B L E 2 2 . 5 Imaging
COMMON PRESENTING SYMPTOMS OF The typical computed tomography findings are areas of dis-
MULTIPLE SCLEROSIS seminated hypoattenuation in the subcortical white matter
of the brain that enhance after the administration of contrast
Optic neuritis (35). They may or may not be surrounded by edema. MRI is
History of fatigue a useful tool to aid in the diagnosis of postinfectious encepha-
lomyelitis. In general, patients with postinfectious encephalo-
Sensory symptoms myelitis have a higher frequency of large confluent bilateral
Lhermitte sign symmetrically oriented diffuse T2/fluid-attenuated inversion
Diplopia/INO recovery (FLAIR) high signal lesions, ventral brainstem lesions,
Uhthoff phenomenon as well as the presence of monophasic pattern of enhancement
(Fig. 22.1) (50). T2-weighted and FLAIR MRI scans demon-
Vertigo strate areas of increased signal in the subcortical white matter,
Gait disturbance brainstem, cerebellum, and periventricular white matter (43).
These may have the appearance of large globular lesions (Fig.
INO, internuclear ophthalmoplegia. 22.1). Periventricular lesions are less frequently seen in postin-
fectious encephalomyelitis than MS, and if present, there is
usually homogeneous contrast enhancement. The lesions are
typically bilateral and asymmetric, vary in size and number,
usually requires a comprehensive, thorough investigation. The and could enhance in a nodular, spotty, ring, or heterogeneous
most common disorders in the differential diagnosis are MS pattern after the administration of intravenous gadolinium.
(both pediatric as well as adult presentation), viral encephali- There may be lesions in gray matter as well. Involvement of the
tis, transverse myelitis, and neuromyelitis optica. Common deep gray matter may help distinguish postinfectious encepha-
symptoms of MS are listed in Table 22.5. Other disorders in- lomyelitis from MS (43). More advanced imaging techniques,
cluding CNS infections, neurometabolic CNS disorders (includ- such as magnetic resonance spectroscopy, have demonstrated
ing mitochondrial disorders), neurosarcoidosis, systemic lupus elevation of lipids and reduction of the myoinositol:creatinine
erythematosus, primary CNS angiitis, NMO, anti-N-methyl- ratio during the acute phase, followed by reduction in lipids
d-aspartate-receptor (anti-NMDA) encephalitis, chronic lym- and increased myoinositol:creatinine ratios in the chronic set-
phocytic inflammation with pontine perivascular enhancement ting (51). Fluorodeoxyglucose positron emission tomography
responsive to steroids (CLIPPERS), Bickerstaff encephalitis, scans usually show marked hypometabolism in the affected
and CNS malignancies should be considered. areas of the brain (45).

A B

C D

FIGURE 22.1 A: Multiple lesions of postinfectious encephalomyelitis on T2-weighted MRI scan. B: The
lesions enhance after the administration of gadoliniumdiethylenetriamine pentaacetic acid due to altered
bloodbrain barrier permeability from the inflammatory process. C: T2-weighted MRI scan showing classic
distribution of lesions seen in an MS patient. There is clear predominance of ovoid-shaped periventricular
lesions, which appear to be perpendicular to the long axis of the lateral ventricle. D: T1-weighted MRI scan of
the same MS patient showing hypointense signal along most of the lesions, suggestive of chronic axonal loss.

Scheld_Ch22.indd 335 2/21/14 10:48 PM


336 Part II: Viral Infections and Related Disorders

TA B L E 2 2 . 6
MAGNETIC RESONANCE IMAGING FEATURES THAT ASSIST IN THE DISTINCTION BETWEEN
MULTIPLE SCLEROSIS AND POSTINFECTIOUS ENCEPHALOMYELITIS

Positive Predictive
MRI Features That Suggest Multiple Sclerosis Diagnosis Sensitivity (%) Specificity (%) Value (%)

Callen criteria (at least 2 out of 3 features suggest MS over ADEM) 75 95 96


Absence of a diffuse bilateral lesion pattern
Presence of T1 hypointensities
Two or more T2/FLAIR periventricular lesions
Barkhof criteria (at least 3 out of 4 features suggest MS over ADEM) 61 91 90
9 T2/FLAIR-weighted lesions or 1 Gd-enhancing lesion
3 periventricular lesions
1 juxtacortical lesion
1 infratentorial lesion

Gd, gadolinium.

A recent review suggested that the use of Callen MS-ADEM and lack of IL-17. However, more concrete studies will need to
criteria resulted in a high sensitivity (75%) and specificity be performed to determine this. In order to facilitate the initial
(95%) for distinguishing MS at first attack from postinfec- steps in the diagnosis of postinfectious encephalomyelitis, we
tious encephalomyelitis (Table 22.6) (52). The authors also propose a list of basic CSF studies that are useful in the differen-
concluded that these MRI-based criteria were predictive of the tial diagnosis of postinfectious encephalomyelitis, MS, and viral
diagnosis of MS versus postinfectious encephalomyelitis even encephalitis (Table 22.7).
in the absence of encephalopathy.

Laboratory Investigations PATHOLOGY


Given the possible similarities between postinfectious encepha-
In rare circumstances, pathologic evaluation of involved tis-
lomyelitis and MS, similar laboratory tests are performed as part
sue can assist in the diagnosis. Consistent with postinfectious
of the workup of both conditions. MS is classically diagnosed
encephalomyelitis clinical presentation, focal, perivenous,
by multiple lesions on MRI as well as the presence of oligoclo-
and subependymal changes dominate the pathologic pattern.
nal IgG bands in the CSF (53). Unfortunately, in the majority
of postinfectious encephalomyelitis cases, there are minor and
nonspecific changes that occur that make diagnosis based on the
standard of care laboratory testing difficult. However, there are TA B L E 2 2 . 7
some distinguishing features that do differentiate this disease
from patients with MS and with healthy controls. These include CEREBROSPINAL FLUID IN THE DIFFERENTIAL
elevated levels of total protein concentration (usually above DIAGNOSIS OF POSTINFECTIOUS
100 mg/dL) and increased cell count (usually more than 50 cells/ ENCEPHALOMYELITIS, MULTIPLE SCLEROSIS,
mm3) (54,55). It has been shown that the presence of certain cyto- AND VIRAL ENCEPHALITIS
kines, such as tumor necrosis factor (TNF)-, interleukin (IL)-2,
IL-4, IL-5, IL-6, IL-8, IL-10, and interferon (IFN)-, are more Cell count with differential
common in postinfectious encephalomyelitis than MS (56,57). Glucose and protein concentration
IL-17 has been reported to be increased in the CSF and blood Myelin basic protein
of MS patients (58,59); however, this is not observed in pa- Oligoclonal bands
tients with postinfectious encephalomyelitis. A study performed
measuring 18 different chemokines in the blood and CSF of IgG index
patients with suspected postinfectious encephalomyelitis, MS, Viral culture
or healthy controls revealed a distinct pattern of increased ex- PCR for HSV-1 DNA
pression of specific chemokines in postinfectious encephalo- PCR for West Nile virus
myelitis patients not observed in patients with MS or controls.
These changes were only observed in the CSF and not in the PCR for HIV RNA
blood. Mean concentration of the chemokines (postinfectious PCR for JC virus DNA
encephalomyelitis, MS, healthy controls) CXCL7 (522 / PCR for EBV DNA
115 vs. 197 / 37 vs. 158 / 53), CCL1 (28.7 / 6.8, PCR for VZV DNA
18.5 / 3.4, 10.0 / 1.7), CCL22 (75.7 / 38.5, 9.4 /
1.8, 4.3 / 0.8), and CCL17 (18.6 / 3.4, 1.8 / 0.6, VZV IgG and IgM
7.2 / 0.6) were distinctly higher in the CSF from postinfec- HSV IgG and IgM
tious encephalomyelitis patients. Based on these data, it may
be possible to distinguish postinfectious encephalomyelitis pa- PCR, polymerase chain reaction.
tients from MS patients based on increased chemokine levels

Scheld_Ch22.indd 336 2/21/14 10:48 PM


Chapter 22: Postinfectious Encephalomyelitis 337

Similar to MS, postinfectious encephalomyelitis lesions are and other nonserotypable herpesviruses), progressive multifo-
mostly in the white matter at the corticalsubcortical junc- cal leukoencephalopathy due to JC virus, and HIV encephali-
tion but may also be seen in the cerebellum, spinal cord, tis. Japanese encephalitis virus classically infects the thalamus,
and brainstem (43,60). Contrary to MS, early postinfectious brainstem, basal ganglia, substantia nigra, spinal cord, cerebral
encephalomyelitis involves infiltration of adaptive immune cortex, and cerebellum. The clinical presentation is character-
cells, mostly T lymphocytes, followed by innate monocytic ized by fever, vomiting, convulsions, and coma. During the acute
cells. Inflammatory lesions in postinfectious encephalomyelitis illness, patients may have movement disorders due to lesions in
spread radially outward from the vessels, whereas in MS, they the basal ganglia and substantia nigra or flaccid paralysis due
border the plaques (43). Additionally, axons in areas of de- to lesions in the spinal cord (61,62). Hyperintense lesions on
myelination are relatively preserved in postinfectious encepha- T2-weighted images in the substantia nigra and thalami can
lomyelitis, whereas in MS, there can be significant axonal be seen in Japanese encephalitis (61,63). The diagnosis is made
loss. The major pathologic distinguishing factor is irregular by demonstrating intrathecal production of specific antibodies.
borders observed in postinfectious encephalomyelitis lesions, West Nile virus encephalitis presents with fever, head-
whereas lesions from MS patients exhibit sharp distinct bor- ache, and confusion and may have associated weakness in a
ders. Postinfectious encephalomyelitis is also distinguished poliomyelitis-like syndrome. The diagnosis is made by either
from acute viral encephalitis by its pathology. Postinfectious the demonstration of West Nile virus nucleic acid in CSF, West
encephalomyelitis is predominantly a disease of white mat- Nile virus immunoglobulin M (IgM) antibody in CSF, or a
ter, but gray matter may also be affected, particularly basal fourfold increase in serum West Nile virus immunoglobulin G
ganglia, thalami, and brainstem (42). Acute viral encephalitis (IgG) antibodies between acute and convalescent sera.
is predominantly a disease of gray matter. The lesions charac- VZV encephalitis presents with headache, malaise, and con-
teristic of postinfectious encephalomyelitis are around small fusion days to weeks after the cutaneous eruption of zoster. The
veins, in the cerebral white matter, brainstem, and spinal cord, neuroimaging abnormalities of VZV encephalitis can be strik-
and are composed of mononuclear cells and lymphocytes. ingly similar to those of postinfectious encephalomyelitis. VZV
Luxol fast blue stains that stain myelin reveal well-demarcated encephalitis may manifest as spherical subcortical white matter
areas of loss of myelin. Staining for axons in the same areas lesions with the typical appearance of demyelination (64).
that have loss of myelin reveals that the axon cylinders are rel- In addition, there may be large and small ischemic and
atively preserved. The neurons in the area show minor changes hemorrhagic infarctions of the cortical and subcortical gray
(4). The degree of the preservation of axon cylinders and the and white matter. The diagnosis is made by demonstrating
extent of the involvement of neurons determine prognosis. either VZV DNA in CSF, VZV IgM antibodies in CSF, or a
positive CSF viral culture.
EBV may cause a meningoencephalitis during the course
DIFFERENTIAL DIAGNOSIS of mononucleosis. Following primary infection, the virus
establishes latent infection in the CNS and can reactivate,
The differential diagnosis is that of inflammatory, demyelinat- causing encephalomyelitis. The clinical presentation includes
ing diseases of the CNS. These include MS, viral encephalitis, fever, headache, focal neurologic deficits, an altered level of
transverse myelitis, and neuromyelitis optica (Devic syndrome) consciousness, and convulsions. This is a monophasic illness
as the most common disorders. Table 22.7 provides a list of with neuroimaging evidence of lesions throughout the CNS.
CSF studies to determine the etiology. The leading disease in Diagnosis is made by demonstrating EBV DNA in CSF.
the differential diagnosis is MS. The first attack of MS can be HSV-1 may reactivate from latent infection in the trigeminal
difficult to differentiate from postinfectious encephalomyelitis. ganglia and present as a brainstem encephalitis instead of caus-
The specific neurologic symptoms are often very helpful in dis- ing the classic presentation of frontoorbital and temporal lobe
tinguishing between the two diseases, as is the history of the dysfunction. There are also reports of HSV DNA in the CSF of
onset of symptoms within 2 to 31 days of a viral illness or vacci- patients with a clinical syndrome of encephalomyelitis and neuro-
nation. Although typical of postinfectious encephalomyelitis, a imaging evidence of multiple hyperintense lesions in the thalamus,
confusional state, headache, a decreased level of consciousness, corpus striatum, pons, and deep white matter on T2-weighted
and convulsions are also quite atypical of MS. MS is more likely MRI scans (65,66). The diagnosis of brainstem encephalitis or
than postinfectious encephalomyelitis to have a monosymp- encephalomyelitis due to HSV-1 is made by the demonstration
tomatic presentation of optic neuritis, a brainstem syndrome, of HSV-1 DNA in CSF by polymerase chain reaction or by the
a cerebellar syndrome, or a transverse myelitis. The presence demonstration of intrathecal production of HSV antibodies.
of bilateral optic neuritis is more suggestive of postinfectious Progressive multifocal leukoencephalopathy is a demyelin-
encephalomyelitis than MS (41). Although both syndromes are ating disease occurring in patients with severe cellular immu-
primarily demyelinating diseases, the lesions of postinfectious nosuppression caused by a reactivation of latent JC virus likely
encephalomyelitis are larger, more extensive, homogeneously acquired during childhood. This disease is progressive to death
enhance with contrast, and can involve gray matter, whereas over the course of a few months. HIV patients and patients re-
the classic lesions of MS are ovoid-shaped, may or may not ceiving immunosuppressive therapies are at higher risk of de-
enhance, and usually involve the periventricular white matter, veloping progressive multifocal leukoencephalopathy (PML).
particularly the trigone and body of the lateral ventricle (41). HIV encephalitis occurs in immunosuppressed individuals and
The Marburg variant of MS is a fulminant form of MS. presents with cognitive, motor, and behavioral abnormalities.
Neuroimaging abnormalities may distinguish the Marburg T2-weighted MRI scans show areas of increased signal inten-
variant from postinfectious encephalomyelitis, but more often sity in the subcortical white matter.
it is the clinical course. Subacute sclerosing panencephalitis (SSPE) is a degenera-
The viral encephalitides that may have a similar presen- tive disease of the brain due to measles virus, which presents
tation and similar neuroimaging and CSF abnormalities to after a latent period of several years or more from acute measles
postinfectious encephalomyelitis include flavivirus infections infection (67). This disease manifests with visual complaints,
(Japanese encephalitis virus, St. Louis encephalitis virus, and behavioral changes, and myoclonic jerks followed by hemipare-
West Nile virus), herpesvirus infections (varicella-zoster virus sis, cogwheel rigidity, and dementia. There are elevated antibody
[VZV], Epstein-Barr virus [EBV], herpes simplex virus [HSV], titers against measles virus in CSF specimens and histopathologic

Scheld_Ch22.indd 337 2/21/14 10:48 PM


338 Part II: Viral Infections and Related Disorders

evidence of extensive demyelination, glial proliferation, and neu- on whether intravenously administered methylprednisolone
ronal and glial intranuclear inclusions (67,68). Although initially therapy should be followed by an oral prednisone taper.
this disease may be a consideration in the differential diagnosis, A randomized, sham-controlled clinical trial of plasma ex-
SSPE is not a monophasic disorder, but a progressive illness. change in patients with either MS (12 patients) or other inflam-
Transverse myelitis is an inflammatory condition of the spi- matory demyelinating disease of the CNS (10 patients) was
nal cord due to a number of infectious and autoimmune eti- performed (72). The 10 patients with inflammatory demyelinat-
ologies. The thoracic cord is most commonly affected. There ing disease other than MS had transverse myelitis, ADEM, neu-
is a progressive weakness of lower extremities over the course romyelitis optica, and focal cerebral demyelinating lesions. The
of several hours to several days. Back pain may be present. patients were randomly assigned to receive either true or sham
Typically, there is a sensory level and bowel and bladder dys- plasma exchange every other day for 2 weeks. All patients had a
function. The maximum deficit is reached by definition within severe clinical deficit and had failed to improve over a period of
4 weeks. Spinal MRI demonstrates swelling of the cord at the 2 weeks from the initiation of high-dose intravenous corticoste-
level of involvement. Devic syndrome refers to the combina- roid therapy. Eight patients who were treated with true plasma
tion of optic neuritis and myelitis and is now more commonly exchange experienced moderate to marked improvement at the
referred to as neuromyelitis optica. end of the 14 days. One patient who was treated with sham
Posttransplantation lymphoproliferative disease and leuko- treatment had a moderate to marked improvement (72).
encephalopathies after chemotherapy and radiotherapy may There are a number of case reports on the use of intrave-
have neuroimaging abnormalities that resemble those of postin- nous immune globulin (IVIG) therapy in ADEM (7375). The
fectious encephalomyelitis but are distinguished by the clinical mechanism of action of IVIG is not completely understood,
setting in which they occur. but IVIG contains a wide spectrum of antibodies that have the
potential to bind and neutralize pathogenic antibodies (73).
These antibodies might bind to MBP and inhibit the access of
TREATMENT AND PREVENTION autoreactive T lymphocytes.
In the initial days of the illness, postinfectious encephalo-
Vaccination of infants against measles, mumps, and rubella has myelitis is often not distinguishable from acute viral encepha-
had a significant effect on decreasing the incidence of postin- litis. A combination of intravenous acyclovir (10 mg/kg every
fectious encephalomyelitis caused by these viruses. Despite the 8 hours) and intravenous methylprednisolone (1,000 mg per
inherent risk of acute neurologic complications following vac- day) can be used until a definitive diagnosis is made. The addi-
cination, the incidence of naturally occurring encephalomyeli- tion of IVIG or plasma exchange may be beneficial either in the
tis is still greater. initial days of therapy or in those patients in whom a diagnosis
Currently, the only agent with proven efficacy in the preven- of postinfectious encephalomyelitis is established, and the course
tion of postvaccinal encephalomyelitis is antivaccinia -globulin is progressive despite intravenous methylprednisolone therapy.
(AGG). The first clinical trial to prevent postvaccinal encepha-
litis was performed in 1956. At the time of primary smallpox
vaccination, 53,630 Dutch military recruits were given an injec- PROGNOSIS
tion of 2 mL of 16% AGG, and 53,044 were given placebo.
The donors of the AGG were healthy volunteers from the Royal The mortality rate of postmeasles encephalomyelitis is 10% to
Netherlands Army and the Royal Netherlands Air Force who 20%, and neurologic sequelae occur in 25% of survivors. The
had recently been vaccinated. Only 3 cases of postvaccinal en- prognosis is related to age, with increasing mortality in those
cephalitis occurred in the treated group, compared with 13 cases older than 16 years, and with the presence and duration of coma
in the control group (69). AGG is not effective in treating post- and convulsions (13). The mortality of postvaricella encephalo-
vaccinal encephalitis once this complication has occurred. myelitis is approximately 10% (13). The mortality rate of post-
Therapeutic recommendations for postinfectious encepha- rubella encephalomyelitis was high, with 20% of children dying
lomyelitis are complicated by the lack of double-blind pla- during the first week (13). Postinfectious encephalomyelitis in
cebo-controlled clinical trials and the fact that postinfectious children that occurs as a complication of an upper respiratory
encephalomyelitis improves spontaneously. Several case series tract infection or nonspecific febrile illness typically has a favor-
suggest that early high-dose corticosteroid therapy is beneficial able prognosis. In adults, however, there are often neurologic
(70,71). Early series used adrenocorticotropic hormone (ACTH) sequelae. As described, prognosis is directly related to the degree
or dexamethasone (1 mg/kg per day), but recent series use in- of pathologic involvement of neurons and axon cylinders. To the
travenously administered methylprednisolone in a daily dose of extent that these are affected by the inflammatory process, there
1,000 mg per day for 3 to 5 days based on the experience with are varying degrees of cognitive deficits, movement disorders
treating an acute exacerbation of MS. No firm guidelines exist (dystonia), and spasticity. These deficits are often chronic.

References
1. Johnson RT. The pathogenesis of acute viral encephalitis and postinfectious 7. Johnson RT, Griffin DE. Postinfectious encephalomyelitis. In: Kennedy PG,
encephalomyelitis. J Infect Dis. 1987;155(3):359364. Johnson RT, eds. Infections of the Nervous System. London: Butterworth;
2. Gurvich EB, Vilesova IS. Vaccinia virus in postvaccinal encephalitis. Acta 1987:209226.
Virol. 1983;27(2):154159. 8. Leake JA, Albani S, Kao AS, et al. Acute disseminated encephalomyelitis in
3. Lucas J. An account of uncommon symptoms succeeding the measles: with childhood: epidemiologic, clinical and laboratory features. Pediatr Infect
the additional remarks on the infection of measles and smallpox. London Dis J. 2004;23(8):756764.
Med J. 1790;11:325331. 9. Menge T, Hemmer B, Nessler S, et al. Acute disseminated encephalomyelitis:
4. Scott TF. Postinfectious and vaccinal encephalitis. Med Clin North Am. an update. Arch Neurol. 2005;62(11):16731680.
1967;51(3):701717. 10. Pohl D, Tenembaum S. Treatment of acute disseminated encephalomyelitis.
5. Rivers TM. Viruses. JAMA. 1929;92:11471152. Curr Treat Options Neurol. 2012;14(3):264275.
6. Hurst EW. Acute hemorrhagic leukoencephalitis: a previously undefined 11. Johnson RT, Griffin DE, Hirsch RL, et al. Measles encephalomyelitisclinical
entity. Med Aust. 1941;2:16. and immunologic studies. N Engl J Med. 1984;310(3):137141.

Scheld_Ch22.indd 338 2/21/14 10:48 PM


Chapter 22: Postinfectious Encephalomyelitis 339

12. Reder AT, Thapar M, Sapugay AM, et al. Prostaglandins and inhibitors of 45. Stve O, Zamvil SS. Pathogenesis, diagnosis, and treatment of acute dis-
arachidonate metabolism suppress experimental allergic encephalomyelitis. seminated encephalomyelitis. Curr Opin Neurol. 1999;12(4):395401.
J Neuroimmunol. 1994;54(12):117127. 46. Barontini F, Di Lollo S, Toscani L. Acute hemorrhagic leukoencephalopa-
13. Gibbons JL, Miller HG, Stanton JB. Para-infectious encephalomyelitis and thy: clinical and CT diagnosis and histopathological confirmation in an
related syndromes: a critical review of the neurological complications of elderly patient. Ital J Neurol Sci. 1984;5(2):215218.
certain specific fevers. Q J Med. 1956;25(100):427505. 47. Rivers TM, Sprunt DH, Berry GP. Observations on Attempts to pro-
14. Sacconi S, Salviati L, Merelli E. Acute disseminated encephalomyelitis duce acute disseminated encephalomyelitis in monkeys. J Exp Med.
associated with hepatitis C virus infection. Arch Neurol. 2001;58(10): 1933;58(1):3953.
16791681. 48. Orgad S, Cohen IR. Autoimmune encephalomyelitis: activation of thy-
15. Narciso P, Galgani S, Del Grosso B, et al. Acute disseminated encephalomy- mus lymphocytes against syngeneic brain antigens in vitro. Science.
elitis as manifestation of primary HIV infection. Neurology. 2001;57(8): 1974;183(4129):10831085.
14931496. 49. Wekerle H. Immune protection of the brainefficient and delicate. J Infect
16. Allen SH, Malik O, Lipman MC, et al. Acute demyelinating encephalomy- Dis. 2002;186(suppl 2):S140S144.
elitis (ADEM) in a patient with HIV infection. J Infect. 2002;45(1):6264. 50. Lu Z, Zhang B, Qiu W, et al. Comparative brain stem lesions on MRI of
17. Johnson RT. Smallpox: the threat of bioterrorism and the risk of the acute disseminated encephalomyelitis, neuromyelitis optica, and multiple
vaccine. Neurology. 2003;60(8):12281229. sclerosis. PLoS One. 2011;6(8):e22766.
18. Fenichel GM. Neurological complications of immunization. Ann Neurol. 51. Ben Sira L, Miller E, Artzi M, et al. 1H-MRS for the diagnosis of acute dis-
1982;12(2):119128. seminated encephalomyelitis: insight into the acute-disease stage. Pediatr
19. Ozawa H, Noma S, Yoshida Y, et al. Acute disseminated encephalomyelitis Radiol. 2010;40(1):106113.
associated with poliomyelitis vaccine. Pediatr Neurol. 2000;23(2):177179. 52. Ketelslegers IA, Neuteboom RF, Boon M, et al. A comparison of MRI cri-
20. Ohtaki E, Matsuishi T, Hirano Y, et al. Acute disseminated encephalomyeli- teria for diagnosing pediatric ADEM and MS. Neurology. 2010;74(18):
tis after treatment with Japanese B encephalitis vaccine (Nakayama-Yoken 14121415.
and Beijing strains). J Neurol Neurosurg Psychiatry. 1995;59(3):316317. 53. Polman CH, Reingold SC, Banwell B, et al. Diagnostic criteria for mul-
21. Boluksbasi O, Ozmenoglu M. Acute disseminated encephalomyelitis asso- tiple sclerosis: 2010 revisions to the McDonald criteria. Ann Neurol.
ciated with tetanus vaccine. Eur Neurol. 1999;41:231232. 2011;69(2):292302.
22. Konstantinou D, Paschalis C, Maraziotis T, et al. Two episodes of leuko- 54. Stuve O, Nessler S, Hartung HP, et al. Acute disseminated encephalo-
encephalitis associated with recombinant hepatitis B vaccination in a single myelitis. Pathogenesis, diagnosis, treatment, and prognosis [in German].
patient. Clin Infect Dis. 2001;33(10):17721773. Der Nervenarzt. 2005;76(6):701707.
23. Ito S, Nezu A, Matsumoto C, et al. Acute disseminated encephalomyelitis 55. Franciotta D, Salvetti M, Lolli F, et al. B cells and multiple sclerosis. Lancet
and poststreptococcal acute glomerulonephritis. Brain Dev. 2002;24(2): Neurol. 2008;7(9):852858.
8890. 56. Ishizu T, Minohara M, Ichiyama T, et al. CSF cytokine and chemokine
24. Sommer JB, Erbguth FJ, Neundorfer B. Acute disseminated encephalomy- profiles in acute disseminated encephalomyelitis. J Neuroimmunol. 2006;
elitis following Legionella pneumophila infection. Eur Neurol. 2000;44(3): 175(12):5258.
182184. 57. Ichiyama T, Shoji H, Kato M, et al. Cerebrospinal fluid levels of cytokines
25. Alonso-Valle H, Muoz R, Hernandez JL, et al. Acute disseminated en- and soluble tumour necrosis factor receptor in acute disseminated encepha-
cephalomyelitis following Leptospira infection. Eur Neurol. 2001;46: lomyelitis. Eur J Pediatr. 2002;161(3):133137.
104105. 58. Graber JJ, Allie SR, Mullen KM, et al. Interleukin-17 in transverse myelitis
26. Krishna KK, Mitra DK, Diwan AG, et al. Acute disseminated encephalomy- and multiple sclerosis. J Neuroimmunol. 2008;196(12):124132.
elitis with typhoid fever. J Assoc Physicians India. 1999;47(10):10171019. 59. Matusevicius D, Kivisakk P, He B, et al. Interleukin-17 mRNA expression
27. Spieker S, Petersen D, Rolfs A, et al. Acute disseminated encephalomyelitis in blood and CSF mononuclear cells is augmented in multiple sclerosis.
following Pontiac fever. Eur Neurol. 1998;40(3):169172. Mult Scler. 1999;5(2):101104.
28. Wei TY, Baumann RJ. Acute disseminated encephalomyelitis after Rocky 60. Mikaeloff Y, Suissa S, Vallee L, et al. First episode of acute CNS inflamma-
Mountain spotted fever. Pediatr Neurol. 1999;21(1):503505. tory demyelination in childhood: prognostic factors for multiple sclerosis
29. Mohsen AH, McKendrick MW, Schmid ML, et al. Postmalaria neurologi- and disability. J Pediatr. 2004;144(2):246252.
cal syndrome: a case of acute disseminated encephalomyelitis? J Neurol 61. Pradhan S, Pandey N, Shashank S, et al. Parkinsonism due to predomi-
Neurosurg Psychiatry. 2000;68(3):388389. nant involvement of substantia nigra in Japanese encephalitis. Neurology.
30. Riedel K, Kempf VA, Bechtold A, et al. Acute disseminated encephalomy- 1999;53(8):17811786.
elitis (ADEM) due to Mycoplasma pneumoniae infection in an adolescent. 62. Johnson RT, Burke DS, Elwell M, et al. Japanese encephalitis: immunocy-
Infection. 2001;29(4):240242. tochemical studies of viral antigen and inflammatory cells in fatal cases.
31. Re A, Giachetti R. Acute disseminated encephalomyelitis (ADEM) after Ann Neurol. 1985;18(5):567573.
autologous peripheral blood stem cell transplant for non-Hodgkins lym- 63. Misra UK, Kalita J. A comparative study of Japanese and herpes simplex
phoma. Bone Marrow Transplant. 1999;24(12):13511354. encephalitides. Electromyogr Clin Neurophysiol. 1998;38(1):4146.
32. Dubreuil F, Cabre P, Smadja D, et al. Acute disseminated encephalomyeli- 64. Weaver S, Rosenblum MK, DeAngelis LM. Herpes varicella zoster enceph-
tis preceding cutaneous lupus [in French]. Rev Med Interne. 1998;19(2): alitis in immunocompromised patients. Neurology. 1999;52(1):193195.
128130. 65. Kusuhara T, Nakajima M, Inoue H, et al. Parainfectious encephalomyelo-
33. Jaing TH, Lin KL, Chiu CH, et al. Acute disseminated encephalomyelitis in radiculitis associated with herpes simplex virus 1 DNA in cerebrospinal
autoimmune hemolytic anemia. Pediatr Neurol. 2001;24(4):303305. fluid. Clin Infect Dis. 2002;34(9):11991205.
34. Stonehouse M, Gupte G, Wassmer E, et al. Acute disseminated encephalo- 66. Nakamura K, Terasaki T, Tsuchiya T, et al. Herpes simplex encephalitis
myelitis: recognition in the hands of general paediatricians. Arch Dis Child. followed by myelopathy [in Japanese]. No To Shinkei. 1993;45:553558.
2003;88(2):122124. 67. Singer C, Lang AE, Suchowersky O. Adult-onset subacute sclerosing
35. Lane JM, Millar JD, Neff JM. Smallpox and smallpox vaccination policy. panencephalitis: case reports and review of the literature. Mov Disord.
Annu Rev Med. 1971;22:251272. 1997;12(3):342353.
36. Feery BJ. Adverse reactions after smallpox vaccination. Med J Aust. 68. Anlar B, Yalaz K, Oktem F, et al. Long-term follow-up of patients with
1977;2(6):180183. subacute sclerosing panencephalitis treated with intraventricular alpha-
37. Katz SL. The case for continuing routine childhood smallpox vaccina- interferon. Neurology. 1997;48(2):526528.
tion in the United States. Am J Epidemiol. 1971;93(4):241244. 69. Nanning W. Prophylactic effect of antivaccinia gamma-globulin against
38. Lane JM, Ruben FL, Abrutyn E, et al. Deaths attributable to smallpox post-vaccinal encephalitis. Bull World Health Organ. 1962;27:317324.
vaccination, 1959 to 1966, and 1968. JAMA. 1970;212(3):441444. 70. Pasternak JF, De Vivo DC, Prensky AL. Steroid-responsive encephalomyeli-
39. Greenberg M. Complications of vaccination against smallpox. Am J Dis tis in childhood. Neurology. 1980;30(5):481486.
Child. 1948;76(5):492502. 71. Straub J, Chofflon M, Delavelle J. Early high-dose intravenous methylpred-
40. Tenembaum S, Chamoles N, Fejerman N. Acute disseminated encephalo- nisolone in acute disseminated encephalomyelitis: a successful recovery.
myelitis: a long-term follow-up study of 84 pediatric patients. Neurology. Neurology. 1997;49(4):11451147.
2002;59(8):12241231. 72. Weinshenker BG. Plasma exchange for severe attacks of inflamma-
41. Dale RC, de Sousa C, Chong WK, et al. Acute disseminated encephalomy- tory demyelinating diseases of the central nervous system. J Clin Apher.
elitis, multiphasic disseminated encephalomyelitis and multiple sclerosis in 2001;16(1):3942.
children. Brain. 2000;123(pt 2):24072422. 73. Marchioni E, Marinou-Aktipi K, Uggetti C, et al. Effectiveness of intra-
42. Hartung HP, Grossman RI. ADEM: distinct disease or part of the MS spec- venous immunoglobulin treatment in adult patients with steroid-resistant
trum? Neurology. 2001;56(10):12571260. monophasic or recurrent acute disseminated encephalomyelitis. J Neurol.
43. Hynson JL, Kornberg AJ, Coleman LT, et al. Clinical and neuroradiologic 2002;249(1):100104.
features of acute disseminated encephalomyelitis in children. Neurology. 74. Nishikawa M, Ichiyama T, Hayashi T, et al. Intravenous immunoglobulin
2001;56(10):13081312. therapy in acute disseminated encephalomyelitis. Pediatr Neurol. 1999;
44. Krupp LB, Banwell B, Tenembaum S. Consensus definitions proposed for 21(2):583586.
pediatric multiple sclerosis and related disorders. Neurology. 2007;68(16) 75. Keliman M BP. Acute disseminated encephalomyelitis: response to intrave-
(suppl 2):S7S12. nous immunoglobulin. J Child Neurol. 1995;10:481483.

Scheld_Ch22.indd 339 2/21/14 10:48 PM


Scheld_Ch22.indd 340 2/21/14 10:48 PM
PART III BACTERIAL AND MYCOPLASMAL
INFECTIONS
CHAPTER 23 PATHOGENESIS AND
PATHOPHYSIOLOGY OF BACTERIAL INFECTIONS
PHILIPP AGYEMAN, DENIS GRANDGIRARD, AND STEPHEN L. LEIB

The brain not only is a normally sterile site but also is pro- This chapter focuses on the pathogenesis of bacterial men-
tected from infection by specialized barriers, including the ingitis and the subsequent development of brain damage.
bony skull and the bloodbrain barrier (BBB). Consequently, Aspects of the pathogenesis of brain abscess are discussed in
infections of the central nervous system (CNS) in general and detail in Chapter 31.
bacterial infections of the CNS in particular are comparatively
rare. In the United States, the incidence of bacterial meningitis
decreased to 1.38 per 100,000 population in 2007 (1). The PATHOGENESIS OF
incidence of brain abscesses is not known precisely but is esti- BACTERIAL MENINGITIS
mated to be approximately 1 per 100,000 population (2,3). In
contrast, the incidence of severe sepsis in the United States in The development of bacterial meningitis progresses through
2007 was 303 cases per 100,000 population (4). four interconnected phases: (a) bacterial colonization and in-
To establish bacterial CNS infections, pathogens must gain vasion of the host with subsequent infection of the CNS, (b)
access either to the subarachnoid space (in the case of meningitis) bacterial multiplication and induction of inflammation in the
or to the brain parenchyma (in the case of brain abscess). Many subarachnoid and ventricular space, (c) progression of inflam-
of the complex processes involved in the transition of pathogens mation with associated pathophysiologic alterations, and (d)
from outside the host into the CNS have been elucidated, par- damage to the CNS (Fig. 23.1).
ticularly for meningitis. Most cases of bacterial meningitis likely
arise from bacteremia, which is caused by invasion of the blood-
stream by the pathogen after colonization of the nasopharyngeal Bacterial Invasion of the Host and
and intestinal mucosa. Similarly, in the pathogenesis of brain
abscess, the hematogenous route is important in a substantial Penetration of the BloodBrain Barrier
number of cases. However, the mechanisms by which pathogens
gain access to the site of infection from the bloodstream likely Figure 23.2 shows the pathogenic steps involved in the devel-
differ substantially between meningitis and brain abscess. This is opment of bacterial meningitis.
suggested by the fact that organisms typically causing bacterial
meningitis (e.g., Streptococcus pneumoniae, Haemophilus influ-
Colonization
enzae, Neisseria meningitidis) very rarely cause brain abscess, Before a meningeal pathogen can cause invasive disease, it has
whereas pathogens typically found in brain abscess (e.g., aerobic to successfully colonize the host first. S. pneumoniae, N. men-
and anaerobic Streptococcus species and Staphylococcus aureus) ingitidis, and H. influenzae type B, which most frequently
are rarely the cause of bacterial meningitis. Further elucidation cause bacterial meningitis in humans, colonize the nasophar-
of the molecular events leading to CNS invasion is likely to ynx and are transmitted from person to person by the respira-
allow a better understanding for these differences. tory route. On the other hand, group B Streptococcus (GBS)
Not all bacterial infections of the CNS are the result of (Streptococcus agalactiae), Escherichia coli, and Listeria
bacteremia. This is most obvious when bacterial meningitis monocytogenes, which cause meningitis at the vulnerable ex-
occurs as a consequence of infection of a cerebrospinal fluid tremes of age and in immunocompromised persons, colonize
(CSF) shunt or when a brain abscess results from breaching the gastrointestinal tract and are transmitted through the oral,
of the skull and meninges by trauma or a neurosurgical pro- vaginal, or fecal-oral route.
cedure. Similarly, in patients in whom brain abscess or men- Colonization of the nasopharyngeal mucosa is established
ingitis originates from a focal infection in the vicinity of the by evasion of mucosal host defense mechanisms, for example,
brain (i.e., sinusitis, otitis media, dental abscess), contiguous ciliary clearance and secretory immunoglobulin A (IgA), suc-
spread rather than bacteremia represents the likely route by cessful competition with other organisms in the aerobic envi-
which the pathogen gains access to the CNS. Again, the patho- ronment of the upper respiratory tract, and adherence to the
gen causing the focal infection seems to determine whether the mucosal epithelium. Specialized surface components, such as
secondary infection is meningitis (in the case of S. pneumoniae components of the polysaccharide capsule, or pili, and the
or H. influenzae) or brain abscess (in the case of streptococci production of bacterial enzymes are crucial for the various
other than S. pneumoniae), suggesting pathogen-dependent steps necessary to establish colonization.
differences in pathogenesis. The mucociliary escalator is the main mechanical defense
Multiple processes occur from the time a pathogen has machinery of the respiratory tract. It constantly propels par-
reached the site of infection until the full manifestations of ticles, including bacteria, to the outside. In order to gain access
the disease have developed. These include the induction of to the mucosa and establish themselves in the respiratory tract,
cytokines and chemokines, activation of inflammatory media- S. pneumoniae, N. meningitidis, and H. influenzae type B have
tors such as nitric oxide (NO), reactive oxygen species (ROS), to overcome the mucociliary escalator. As exemplified by S.
or matrix metalloproteinases (MMPs), recruitment of white pneumoniae, there are several means by which bacteria may
blood cells to the site of infection, and cytotoxic events. Many avoid being trapped and flushed away by mucus. The polysac-
of these events have been analyzed in detail in bacterial menin- charide capsule of S. pneumoniae is almost exclusively nega-
gitis, whereas less is known in the case of brain abscess. tively charged. This could increase electrostatic repulsion from

341

Scheld_Ch23.indd 341 2/21/14 5:41 PM


342 Part III: Bacterial and Mycoplasmal Infections

Nasopharyngeal
colonization

Bloodstream
invasion

Systemic
Bacteremia
hypotension

Meningeal invasion

Bacterial multiplication
and lysis

Ventricular and
Loss of vascular
subarachnoid space Bacterial factors
autoregulation
inflammation

Vasculitis
Breakdown of the CSF outflow Neurotoxic
blood-brain barrier obstruction environment

Vascular spasm,
thrombosis

Cerebral Intracranial Brain


ischemia hypertension edema

Necrosis Apoptosis

Cortical brain damage Hippocampal


e.g., cerebral palsy, brain damage FIGURE 23.1 Selected mechanisms
sensorimotor deficits, e.g., learning disabilities that contribute to the pathogenesis of
cortical blindness bacterial meningitis and the develop-
ment of brain injury.

the likewise negatively charged sialic acid residues of mucus successful colonization of the nasopharynx (9). Ciliostasis has
(5). Additionally, deglycosylation of mucus by secreted exogly- also been documented in H. influenzae infection (10), whereas
cosidases (e.g., neuraminidase A [NanA], beta-galactosidase nasopharyngeal colonization of N. meningitidis entails cyto-
B, etc.) may decrease its viscosity and lessen entrapment toxicity for ciliated epithelial cells (11).
(6). Furthermore, pneumolysin, a major virulence factor of Besides its mechanical action, respiratory mucus also har-
S. pneumoniae that is released by autolysis, inhibits ciliary bors several soluble antibacterial agents. Secretory IgA antibod-
beating (7,8). Pneumolysin has been shown to be necessary for ies inhibit microbial adherence and penetration into the mucosa.

Scheld_Ch23.indd 342 2/21/14 5:41 PM


Chapter 23: Pathogenesis and Pathophysiology of Bacterial Infections 343

Opaque phenotype
Nasopharynx (encapsulated pathogen)

Colonization
Transparent phenotype

Mucin layer

Nasopharyngeal

Host invasion
epithelial
cell layer

Extracellular matrix

Endothelial cells

Survival in the bloodstream


Monocyte

Bloodstream
MMPs
Neutrophils Cytokines
Chemokines
Integrins
Selectins
Blood brain barrier

Infection of the CNS


Brain microvascular
endothelial cells
Basal membrane
Tight
CNS junction
MMPs
Cytokines
Glia cell Chemokines

FIGURE 23.2 Schematic representation of pathogenetic steps involved in the development of bacte-
rial meningitis: (a) bacterial colonization of the nasopharynx and invasion of the host; (b) bacteremia
with subsequent transgression of the bloodbrain barrier (BBB); and (c) bacterial multiplication, induc-
tion of inflammation, and invasion of blood-derived neutrophils into the subarachnoid and ventricular
space. Colonization of the nasopharyngeal mucosa is established by adherence to the mucosal epithelium
and affected by phase variation in colonial morphology. Bacteria then penetrate the mucosal barrier
through or between epithelial cells. The capsule is the primary survival factor in the bloodstream, operat-
ing against circulating antibodies, complement-mediated bacterial killing, and neutrophil phagocytosis.
Pathogens must cross the BBB to induce bacterial meningitis. Once attachment to cerebrovascular endo-
thelial cells has occurred, several strategies can be employed by the pathogen to migrate across the BBB
and gain access to the CSF space. These include (a) paracellular passage by disruption of the intercellular
endothelial connections or endothelial injury; (b) transcellular transport by active or passive transcytosis;
and (c) invasion within white blood cells during diapedesis. The entry of blood-derived neutrophils into
the CSF requires tethering and rolling of leukocytes along the endothelium, which is mediated by the
interaction of members of the selectin family, followed by firm adhesion to endothelium and subsequent
emigration through the vessel wall. The transmigration of leukocytes through vessel walls occurs along
a chemotactic gradient mainly dependent on chemokines. Neutrophils appear to also use certain MMPs,
such as MMP-8 and MMP-9, to digest ECM components and thus facilitate the process of extravasation.

N. meningitidis, S. pneumoniae, and H. influenzae, as well as them from subsequent recognition by intact immunoglobulin
some members of the resident oral flora, secrete highly specific (13). Lysozyme is an important component of the innate im-
endopeptidases that cleave the heavy chain of human IgA1, mune system and abundantly present in the saliva, airway fluid,
including its secretory form (sIgA1), thus separating the mo- and lysosomal granules of neutrophils. Its muramidase activity
nomeric antigen-binding fragments (Fab) from the secondary leads to hydrolyzation of the bacterial cell wall polymer pep-
effector functions of the IgA1 antibody (Fc) (12). IgA1 prote- tidoglycan. Modification of peptidoglycan by N-deacetylation
ases exist as three different classes of proteinases. IgA1 prote- and O-acetylation and the production of lysozyme inhibitors are
ases of H. influenzae and N. meningitidis are genetically related bacterial strategies to fend off lysozyme action (14). The gene
serine proteinases, those of S. pneumoniae are metalloprotein- coding for the enzyme responsible for N-deacetylation, peptido-
ases. Cleavage of IgA1 by proteases impairs specific mucosal glycan N-acetylglucosamine deacetylase A (PdgA), is present in
immunity in the upper respiratory tract, thus allowing bacteria S. pneumoniae and L. monocytogenes (15). Genes encoding for
to colonize. IgA1 proteases may also contribute to the patho- peptidoglycan O-acetyltransferases have also been found in S.
genesis of invasive infections by leading to the coating of bacte- pneumoniae (attenuator of drug resistance [adr]) and N. men-
ria with Fab fragments, which mask epitopes and thus protect ingitidis (peptidoglycan acetylase A [pacA] and pacB) (16). For

Scheld_Ch23.indd 343 2/21/14 5:41 PM


344 Part III: Bacterial and Mycoplasmal Infections

S. pneumoniae, it has been shown that expression of PdgA and pneumococcal carriage and infection induce salivary and
Adr is necessary to provide lysozyme resistance and provide col- serum antibodies against NanA, CbpA, and PsaA, among
onization advantage in the mouse model (17). Finally, apolacto- other pneumococcal antigens (34,35). Also, levels of PsaA and
ferrin, the iron-depleted form of lactoferrin, is bacteriostatic or NanA antibodies in the serum were inversely correlated to fre-
bactericidal for bacteria. The pneumococcal surface protein A quency of respiratory tract infections (35).
(PspA) protects against the bactericidal effect of apolactoferrin, Primary adherence of N. meningitidis depends on the bind-
probably by binding to the active sites of apolactoferrin (18). ing of type 4 pili on the bacterial cell surface to nonciliated
Bacterial adherence to epithelial cells is mediated by adhes- epithelial cells. Thereby, N. meningitidis induces rearrange-
ins on the bacterial surface binding to epithelial cell receptors. ments of the cellular cytoskeleton leading to protrusion of
Based on their structure, fimbrial and afimbrial adhesins can microvilli and formation of cortical plaques in epithelial cells
be distinguished in meningeal pathogens. Fimbriae (or pili) are (10). Clusters of bacteria are then embraced by microvilli,
filamentous structures, first identified in Gram-negative bacte- increasing stability of the adhesion. Further physical proxim-
ria, that protrude from the outer cell membrane. Fimbriae are ity of the bacterial and epithelial cell surfaces accompanied
composed of several subunits that form rodlike structures and by the loss of fimbriae and downregulation of capsule allows
carry a specific adhesive subunit at their tip (19,20). Fimbriae other adhesins to attach to their respective receptors (36).
have been associated with the early steps of adhesion mostly, Most notably, opacity-associated (Opa) proteins, a family of
but not exclusively, in Gram-negative meningeal pathogens. phase-variable outer membrane proteins of N. meningitidis,
Several afimbrial adhesins have been identified in all meningeal bind to carcinoembryonic antigen-related cell adhesion mol-
pathogens and will be discussed with the respective pathogens. ecules (CEACAM), cell surfaceexpressed heparan sulfate
Adherence of pneumococci to mammalian cells and pneu- proteoglycans, and extracellular matrix (ECM) proteins (37).
mococcal virulence is facilitated by regulated expression levels CEACAMs are further involved in the invasion process of
of the polysaccharide capsule in a process called phase varia- N. meningitidis (discussed below). While fimbriae protrude
tion. Capsular phase variation of S. pneumoniae is marked by from the polysaccharide capsule, the capsule interferes with
changes in colonial morphology from opaque to transparent the function of other adhesins of N. meningitidis (38,39).
and correlates with differences in virulence. The transparent, Downmodulation of the capsule or inducing the upregulation
less capsulated phenotype is more capable of colonizing the of adhesion receptors (e.g., CEACAMs) are proposed mecha-
nasopharynx, whereas the opaque phenotype shows increased nisms by which invasive N. meningitidis overcome this disad-
virulence during systemic infections (21,22). Masking of vantage (38).
underlying adhesion molecules by the polysaccharide capsule Although structurally different, fimbriae are also important
is a possible explanation for the inferior colonization capabili- adhesion molecules of H. influenzae (40). They mediate adhe-
ties of opaque strains. Adherence of S. pneumoniae in itself is sion to mucin, ECM, and epithelial cells (41). Besides fimbriae,
thought to involve binding to glycoconjugates on the surface the protein Hsf is a major adhesin of H. influenzae. Hsf is
of the respiratory epithelium, although the exact mechanisms anchored in the outer membrane of H. influenzae and forms
have not been completely elucidated (23). Recognition of these fiber-like structures on the cell surface (41). Similar to N. men-
binding sites is enhanced by cleavage of the terminal sialic acid ingitidis, the outer membrane protein P5 of H. influenzae
by NanA and other glycosidases (24,25). Further putative binds to CEACAM-1 and intercellular adhesion molecule-1
roles of NanA during the colonization process are its involve- (ICAM-1) (42,43).
ment in the formation of biofilm (26) and competition with E. coli and GBS are the main pathogens of neonatal menin-
other meningeal pathogens (27). A specific function as adhe- gitis but, like L. monocytogenes, may also cause meningitis in
sin has been shown for the choline-binding protein A (CbpA; the elderly and immunocompromised persons. Transmission
also called PspC or SpsA), which binds to the polymeric im- from the asymptomatically colonized mother or the environ-
munoglobulin receptor (pIgR) on human mucosal epithelial ment can lead to the colonization of the intestinal tract of the
cells and complement factor H (23). Importance of CbpA infant, the first step in the pathogenesis of neonatal E. coli and
has been demonstrated in an infant rat model where CbpA- GBS meningitis. The food-borne pathogen L. monocytogenes
deficient pneumococci were significantly restricted in their disproportionately affects pregnant women and is transmitted
ability to colonize the nasopharynx (28). CbpA is a member to the fetus through the placenta (44).
of the family of choline-binding proteins. This family of pro- Similar to N. meningitidis, adherence of E. coli to host cells
teins shares phosphorylcholine, a component of lipoteichoic depends primarily on fimbriae. For example, the type I fim-
and teichoic acids, as common cell wall anchor. Interestingly, briae in E. coli harbor the adhesive FimH subunits on their tip
phosphorylcholine also binds to activated platelet-activating (45). FimH binds mannose-containing residues on the host cell
factor receptor (PAFr) on epithelial cells and C-reactive pro- and has been implicated in the tissue tropism of pathogenic
tein (CRP) (29). Inflammation enhances this process, as ex- E. coli (46). Additionally, type S fimbriae are expressed by E.
pression of PAFr on lower respiratory tract epithelial cells, coli that have been isolated in neonatal meningitis and sep-
and thereby PAFr-dependent adhesion and transcytosis of S. sis cases (47). Another heterogenous family of adhesins that
pneumoniae, are increased by rhinovirus, acid, fossil fuel may be assembled into fimbriae-like structures are the Afa/Dr
derived particulate matter, and cigarette smoke (30). On the adhesins that have been reported to adhere to the decay-ac-
other hand, phosphorylcholine expression itself can be modu- celerating factor (DAF) expressed on cells exposed to plasma,
lated by the pneumococcal phosphorylcholine esterase. This complement proteins, CEACAMs, and type IV collagen (19).
may allow the bacterium to limit CRP-binding and consecu- The expression of adhesins on E. coli differs between the dif-
tive complement activation (23). Eventually, cellcell adhesion ferent pathogenic and nonpathogenic subtypes and likely de-
molecules have been identified as important adhesion targets termines the organ tropism. Accordingly, the majority of E.
for viruses, bacteria, and parasites (31). For S. pneumoniae, it coli isolates found in neonatal meningitis exhibits the capsular
has been shown that pneumococcal surface antigen A (PsaA) serotype K1 and a limited repertoire of different O antigens
attaches to E-cadherin, an adhesion receptor it shares with L. (48). The reason for the neurotropism of these specific sero-
monocytogenes (32). CbpA and PsaA are produced by virtu- type combinations is not yet completely understood, although
ally all clinical isolates of S. pneumoniae and contribute to the K1 capsule seems to be the decisive factor for meningitis
their virulence (33). It has also been shown in children that development (49).

Scheld_Ch23.indd 344 2/21/14 5:41 PM


Chapter 23: Pathogenesis and Pathophysiology of Bacterial Infections 345

Akin to neonatal E. coli meningitis, epidemiologic analysis The transmembrane receptor pIgR is necessary for trans-
of GBS in neonates with GBS meningitis shows a predomi- port of IgA and IgM across epithelial cells. After binding of
nance of the capsular serotype III and the clonal complex ST-17 the immunoglobulins at the basolateral surface of the epithe-
(50). Recently, a novel virulence factor has been described in lial membrane, the pIgR-immunoglobulin complex is taken up
bacteria belonging to the ST-17 complex. The hypervirulent into endosomes and transported across the epithelium in vesi-
GBS adhesin is anchored to the cell wall and in vitro signifi- cles to the apical mucosal surface, where it is released into mu-
cantly enhances adhesion to intestinal epithelial cells, resulting cosal secretions (69). S. pneumoniae is capable of exploiting
in increased colonization of the intestine in the in vivo mouse this mechanism for its own transcytosis across mucosal cells.
model (50). Other adhesins involved in the attachment of GBS As previously mentioned, binding of S. pneumoniae to human
to the gastrointestinal epithelium primarily bind to compo- pIgR is mediated by CbpA. This interaction allows S. pneu-
nents of the ECM. For example, the cell surface protein SepB moniae to hijack the recycling pathway of pIgR from the api-
binds to fibronectin but also mediates cleaving of complement cal to the basal cell surface by retrograde transport, thereby
factor C5a. Attachment to laminin is mediated by the adhe- facilitating transmucosal invasion of pneumococci (70,71).
sin Lmb, whereas the surface-anchored protein FbsA binds to The CEACAM proteins, a family of proteins involved in in-
fibrinogen (51). Finally, fimbriae have also been involved in tercellular adhesion, have been identified as target of adhesion
GBS adherence and biofilm formation (52). molecules of N. meningitidis, H. influenzae, and E. coli (72).
For adherence and internalization of L. monocytogenes at Of the CEACAMs that have been associated with bacterial
the intestinal mucosa level, the interaction of the internalin A binding, CEACAM-1 is most widely distributed on different
(InlA) with the cell-junction protein E-cadherin is paramount human cell types. Other CEACAMs that have been shown to
(53,54). E-cadherin is normally located below the tight junc- bind to Opa of N. meningitidis are CEACAM-3, CEACAM-5,
tion and not available as a receptor to intraluminal bacteria. and CEACAM-6 (73). Interestingly, binding to CEACAM-1
However, it has been demonstrated that E-cadherin is pres- does not only allow N. meningitidis to adhere and invade epi-
ent on the luminal surface of mucus-secreting goblet cells and thelial cells by endocytosis (74) but may also downregulate
may be temporarily accessible at the tips of intestinal villi due the host immune response on several levels (72). On the other
to constant shedding of dead enterocytes (55,56). Whereas hand, binding of N. meningitidis to CEACAM-3 on neutro-
adherence to the intestinal epithelium is important for the phils has been shown to enhance engulfment and killing of
pathogenesis of L. monocytogenes in the elderly and immuno- bacteria (73).
compromised persons, transmission of L. monocytogenes to the E-cadherin is the cell surface protein responsible for estab-
neonate occurs via the transplacental route. Histopathologic lishment of adherens junctions between neighboring cells (31).
analysis of placentas from women with fetoplacental listeriosis In the intestine, the interaction of listerial protein InlA with
and in vitro analysis indicate that L. monocytogenes targets E-cadherin leads to the rearrangement of the actin skeleton
E-cadherin on the surface of syncytiotrophoblasts in a similar of the host cell and ensuing engulfment of the bacteria into a
way as in the intestine (57). vacuole that is transported to the basolateral cell surface (56).
Persistence in the vacuole until release into the lamina propria
is in contrast to intracytosolic release and replication of L.
Invasion of the Bloodstream monocytogenes in other human cells. Although interaction of
The functional integrity of the respiratory tract mucosa is criti- InlA with E-cadherin is also involved in the invasion process
cal in protecting the host from bacterial invasion. Viral infec- of L. monocytogenes in the placenta, additionally, binding of
tions of the respiratory tract may lead to a decrease in ciliary InlB to its host receptor Met is necessary (75).
function and associated mechanical clearance of bacteria, up- The exact mechanisms by which E. coli K1 and GBS invade
regulation of different host cell receptors involved in bacterial human mucosal cells have not been elucidated. One adhesin/
adherence, and alteration of host immune response (58,59), invasin that has been identified in E. coli K1 is the Hek outer
and are associated with an increased risk of invasive bacterial membrane protein, which mediates binding to proteoglycan
disease. Epidemic outbreaks of meningitis due to N. meningiti- on the epithelial cell surface (76). Additionally, a role for outer
dis in the sub-Saharan meningitis belt coincide with the dry membrane protein A (OmpA) and IbeB, better studied in BBB
periods of the year, when the protective barrier of the mucous crossing, has been suggested (76). Altogether, akin to other
membrane is compromised (60). meningeal pathogens, this points to an endocytosis-like pro-
Bacteria can penetrate the mucosal barrier through or be- cess which is enabled by rearrangement of the epithelial cell
tween epithelial cells, and both routes seem to be employed cytoskeleton (77).
depending on the pathogen. Presence of a capsule or sialylated Between epithelial cells and mesenchymal cells lies the base-
lipooligosaccharides, factors that are instrumental for the sur- ment membrane, providing a final mechanical barrier before
vival of meningeal pathogens in the bloodstream, hinders the the invading pathogen can disseminate throughout the host.
process of transcytosis (10,61). Most of the identified mecha- Attachment to ECM components is an important property
nisms by which these pathogens directly invade human na- of bacterial infection pathogenesis, and ECM adhesins have
sopharyngeal mucosal cells take advantage of a ubiquitous been identified in most meningeal pathogens (e.g., PavA of
cellular endocytosis mechanisms. In clathrin-mediated endo- S. pneumoniae [23], Opc of N. meningitidis [78], fimbriae of
cytosis, a ligand-receptor interaction initiates the formation of H. influenzae [79], and ScpB of GBS [51]). To cross the base-
a clathrin-coated pit, which eventually forms a vacuole that ment membrane, meningeal pathogens have devised several
engulfs particles (62,63). Additionally, actin rearrangements strategies: degradation of the basement matrix by secretion
are induced and further enhance bacterial internalization. of proteolytic enzymes, use of the hosts plasminogen-plasmin
On the other hand, paracellular migration of bacteria might system, and possibly crossing of the basement membrane in
be facilitated by toll-like receptor (TLR)dependent down- phagocytic cells. Hyaluronidase is an endoglycosidase that
regulation of tight junction components (64) or degrada- degrades hyaluronic acid, a component of the ECM. S. pneu-
tion of intercellular junctions mediated by bacterial enzymes moniae strains demonstrate a strong correlation between
or bacteria-bound plasmin (6568). So far, several host cell hyaluronidase activity and the capacity to induce menin-
factors have been identified as targets of bacterial invasion gitis (80). Intranasal instillation of pneumococci together
mechanisms. with hyaluronidase was followed by meningitis in 50% of

Scheld_Ch23.indd 345 2/21/14 5:41 PM


346 Part III: Bacterial and Mycoplasmal Infections

inoculated mice, whereas mice inoculated without hyaluroni- the antiphagocytic activity of the bacterial capsule. Most effec-
dase failed to develop meningitis (65). Plasminogen is the key tive are antibodies against capsular components, which permit
proenzyme of the fibrinolytic system, which after activation optimal opsonization of bacteria and ensure efficient phago-
becomes the broad-spectrum protease plasmin. Although cytosis by polymorphonuclear (PMN) leukocytes and macro-
fibrin is its main substrate, plasmin can cleave several com- phages. The efficacy of this mechanism is underscored by the
ponents of the basement membrane and ECM (81). Binding more than 95% decrease of H. influenzae type b meningitis after
of plasminogen has been shown for N. meningitidis, S. pneu- the introduction of conjugate vaccines, which induce high anti-
moniae, and H. influenzae (82) and enhances invasiveness of capsular antibody titers even in young children (95).
S. pneumoniae and H. influenzae (67,83,84). Phagocytic cells The first step in the classical complement pathway is the
targeting colonizing pathogens on the mucosal surface move association of C1-complex to antibodies bound to the bacte-
freely between the mucosal surface and the intraluminal space rial surface. The pneumococcal protein PspA has been shown
of the mucosal microvasculature. This movement of phago- to limit this step (96). C1-complex (classical complement path-
cytic cells can be exploited by bacterial pathogens that are able way) or MBL-associated serine protease 2 (MBL pathway) then
to survive in phagocytic cells (85). For L. monocytogenes, it activate the C3-convertase C4b2a, which in turn activates C3
has been shown that dendritic cells are an important transport to C3b. As with any enzyme cascade, activation of the comple-
vehicle from the mucosa to lymphatic organs (86). ment system needs to be balanced carefully to avoid damage
to host cells. Regulators of complement activation include
C4-binding protein (C4BP), factor H, and DAF, which accel-
Intravascular Survival erate the dissolution of C3-convertases (97). E. coli K1, N.
Once in the bloodstream, meningeal pathogens are immediately meningitidis, S. pneumoniae, GBS, and H. influenzae may all
attacked by the host immune defense. To establish successful inhibit the classical and MBL complement pathway by bind-
bacteremia, meningeal pathogens need to overcome the differ- ing of C4BP (98,99). Also, S. pneumoniae, N. meningitidis,
ent effector systems of the immune defense. Clearance of the and GBS can bind factor H and downregulate the alternative
classical meningeal pathogens S. pneumoniae, H. influenzae, complement pathway (100102). In particular, meningococcal
and N. meningitidis relies on antibody-mediated opsonization factor Hbinding protein has received considerable attention
followed by activation of the complement system. In the case because this molecule is a key component of group B menin-
of S. pneumoniae and H. influenzae, this is followed by phago- gococcal vaccines (103,104). The result of C3 activation is the
cytosis and finally intracellular killing, whereas the membrane generation of C5 convertase. C5 convertase cleaves C5 into
attack complex is important for control of N. meningitidis (87). C5a, a strong chemoattractant and anaphylatoxin, and C5b,
In neonates with E. coli and GBS meningitis, immune defense re- member of the membrane attack complex (MAC). GBS ex-
lies especially on innate immunity, with the complement system press an endopeptidase that cleaves C5a (105). The final step
being the first line of defense. The importance of the complement of complement cascade is the generation of the MAC, which
system for the clearance of meningeal pathogens is underlined by mainly affects Gram-negative pathogens. Vitronectin regulates
the fact that patients with impaired complement activation (i.e., this last step of the complement cascade by interfering with
sickle cell disease, anatomical or functional asplenia, or poly- MAC assembly. H. influenzae and N. meningitidis have been
morphisms in the complement components) are predisposed to shown to bind vitronectin through Hsf and Opc, respectively,
invasive pneumococcal infections, whereas patients with defi- and thereby limit bacteriolysis (106). Although evasion of the
ciencies in the terminal complement components are prone to complement system also impedes opsonic phagocytosis, the
invasive infections with N. meningitidis (8891). polysaccharide capsule of the classical meningeal pathogens
The complement system may be activated by the classical additionally allows for direct evasion of bacterial uptake and
pathway, the mannose-binding lectin (MBL) pathway, or the killing by professional phagocytes (107,108).
alternative pathway, all converging at the deposition of acti- In contrast, the intracellular pathogen L. monocytogenes
vated complement factor C3b on the pathogen surface, which depends mainly on its ability to evade intracellular killing in the
catalyzes the downstream effectsopsonization and phago- phagolysosome to evade the host immune system. After uptake
cytosis or bacteriolysisof the complement system (87). The into the cell, L. monocytogenes can rupture the vacuole through
meningeal pathogens have developed several mechanisms to the action of two phospholipases and listeriolysin and escape into
evade deposition of C3b on the cell surface. The polysaccharide the cytosol (109). Concurrently, N. meningitidis and S. pneu-
capsule, although hindering adherence to the mucosal surface moniae have been shown to escape the oxidative burst in leu-
and entry into epithelial cells, is the primary survival factor in kocytes by guarding themselves against ROS by the production
the bloodstream, operating against circulating antibodies, com- of glutathione (110,111). Another important virulence factor of
plement-mediated bacterial killing, and neutrophil phagocytosis L. monocytogenes is the actin assemblyinducing protein ActA,
(49). Accordingly, encapsulation is a shared feature of the major which catalyses polar actin assembly and autophagy escape,
hematogenous meningeal pathogens (H. influenzae, N. menin- thereby enabling intracellular motility and spread to adjacent
gitidis, S. pneumoniae, E. coli K1, and GBS). Sialic acid, which cells (109). Although bacterial clearance by phagocytosis is an
is present on most human cells and blocks activation of com- important early step in listeriosis, the ability of L. monocytogenes
plement, is also found in the capsule of meningeal pathogens, to survive intracellularly in macrophages allows it to travel to
for example, GBS (type III), E. coli (K1), and meningococci target organs, thereby evading innate immune mechanisms.
(groups B and C). Thereby, the polysaccharide capsule may pre- In summary, the intricate defense mechanisms of bacteria
vent complement-mediated bacteriolysis (92). Also, in S. pneu- against the host immune system only need to safeguard the
moniae, the capsule is thought to interfere with the activation survival of one (or a few) organism(s), which may then gain
of the classical complement pathway by limiting binding of im- access to the subarachnoidal space and cause meningitis (112).
munoglobulin and CRP to subcapsular antigens (93). Binding of
immunoglobulin may further be hindered by the action of IgA1
proteases. IgA1-proteases cleave IgA1 in the hinge region and
Meningeal Invasion
liberate monomeric Fab-a fragments, which block the access of To enable efficient neuronal signaling, the CNS is a tightly
intact immunoglobulin G (IgG) or immunoglobulin M (IgM) to regulated space protected from its surroundings by differ-
the pathogen (94). Several host defense mechanisms counteract ent barriers. The boundary to the blood is provided by the

Scheld_Ch23.indd 346 2/21/14 5:41 PM


Chapter 23: Pathogenesis and Pathophysiology of Bacterial Infections 347

BBB, the bloodcerebrospinal fluid barrier (BCSFB), and the pathogens is the ability of bacteria to avoid fusion of the vacu-
arachnoid (113). The BBB is composed of brain endothelial ole with lysosomes as demonstrated for E. coli K1 (129).
cells, closely connected through adherens and tight junctions; Interaction of bacterial adhesins with ECM proteins may
a basal membrane; astrocytic and microglial processes; and enhance invasion of meningeal pathogens. In vitro evidence
pericytes. The close interconnection of endothelial cells, low has been brought forward for the exploitation of the integrin
pinocytic activity, and specific transport mechanisms allow receptormediated mechanism of ECM protein internalization
for a selective permeability (113). Bloodborne pathogens must by N. meningitidis and GBS (122,130). On the other hand, for
cross the BBB or the BCSFB from the bloodstream to induce N. meningitidis, experimental evidence indicates that the for-
bacterial meningitis. Clinical observations and experimental mation of cortical plaques, induced by microcolonies on brain
studies demonstrate a relationship between the magnitude of endothelial cells, and the ensuing reorganization of the intra-
bacteremia and the development of meningitis for most menin- cellular cytoskeleton lead to the opening of intercellular junc-
geal pathogens (89,114,115). However, high bacteremia alone tions followed by paracellular penetration of the BBB (119).
is not sufficient for the development of bacterial meningitis, Receptor-mediated signaling as well as local inflammation
but expression of selective bacterial adhesion and invasion fac- with recruitment of leukocytes may further modify tight junc-
tors is necessary (116). tions and allow bacteria to penetrate paracellularly (119,122).
Attachment to the brain microvascular endothelial cells is Transcellular invasion of brain endothelial cells by L. mono-
facilitated by receptors for meningeal pathogens found on the cytogenes is thought to be mediated by InlA and InlB in similar
endothelium in cerebral capillaries and the choroid plexus. fashion as in the intestine and placenta, although conflicting
The laminin receptor, a cell surface protein involved in binding experimental evidence exists (123,131). Additionally, the ef-
to ECM components, has been suggested as common adhesion fective intracellular trafficking of L. monocytogenes across the
target for S. pneumoniae, N. meningitidis, and H. influenzae BBB in phagocytic cells has been shown in a mouse model
and also supports invasion of cytotoxic necrotizing factor 1 (132,133).
(CNF1) expressing E. coli K1 (117,118). Adhesion to brain In humans, invasion of the CSF by bacteria can occur in-
endothelial cells via the laminin receptor is mediated by CbpA dependently of bacteremia. This happens typically when focal
in pneumococci, outer membrane proteins PilQ and PorA in infections occur in structures in immediate proximity to the
meningococci, and the porin OmpP2 in H. influenzae (117). brain (e.g., otitis media, mastoiditis, sinusitis) or when the in-
Further, fimbriae are important structures in bacterial adhe- tegrity of the skull and meninges surrounding the brain is dis-
sion to human cells (see earlier discussion). Fimbriae are es- rupted (e.g., malformations, trauma, neurosurgery). In a case
sential for binding of N. meningitidis to brain endothelial series of 87 patients with pneumococcal meningitis, more than
cells (119). Binding of E. coli K1 to brain endothelial cells is half of the patients had radiologic signs of otitis or sinusitis
also mediated by FimH on type 1 fimbriae binding to CD48 (134), whereas in a larger observational study, otitis and si-
and OmpA association with glycoproteins on the cell surface nusitis were reported as predisposing conditions in 25% of
(116). For GBS as well, several adhesins are required for bind- patients (135). In a model of pneumococcal respiratory tract
ing to brain endothelial cells, including the pilus tip adhesin infection, meningitis occurred without detectable bacteremia,
PilA (120122). suggesting that pneumococci are able to invade the brain by
Once attachment has occurred, pathogens may employ bypassing the bloodstream (136). Similarly, a galU mutant
several strategies to migrate across the BBB and gain access and its parent pneumococcal strain both caused meningitis fol-
to the CSF space. These include (a) transcellular traversal lowing otitis media infection in gerbils, despite the mutants
by transcytosis, (b) paracellular passage by disruption of the impaired ability to disseminate to the bloodstream following
intercellular endothelial connections or endothelial injury, infection (136).
and (c) invasion within white blood cells during diapedesis. For L. monocytogenes, S. pneumoniae, and N. meningitidis,
Causative organisms of bacterial meningitis have mainly been retrograde access to the CNS via the neural route has also been
shown to migrate into the CSF by transcellular or paracellular documented (123,137,138). However, for L. monocytogenes,
migration (116). However, L. monocytogenes also accesses the this mechanism is likely only important in ruminants. These
CSF space inside leukocytes (123). observations support the notion that meningeal pathogens
Direct invasion of endothelial cells by meningeal pathogens can gain access to the subarachnoid space by several routes.
involves ligand-receptor interactions between bacteria and The relative importance of hematogenous versus nonhematog-
host cells and rearrangement of the endothelial actin cytoskel- enous routes of infection in humans is not exactly known.
eton, which ultimately leads to the uptake of the pathogen in
a vacuole and transcellular transport (116,124). For S. pneu-
moniae, the interaction of phosphorylcholine with PAFr and Meningeal Infections Associated with
expression of NanA are important for invasion of the BBB Foreign Bodies
(125). PAFr is a transmembrane receptor expressed by several
different cell types across the human body and involved in the Pathogenesis of foreign bodyassociated meningeal infection
pathogenesis of different inflammatory diseases, atherogen- differs in several important aspects from bacterial meningitis
esis, embryo implantation, and CNS signaling (126). PAFr is a described earlier. First, at the time of foreign body insertion
G proteincoupled receptor and the ligand-receptor complex and due to local tissue damage, access of bacterial pathogens
can be taken up into a vacuole that either recycles back to to the CSF space is facilitated. Second, bacteria may organize
the cell surface or fuses with lysosomes to terminate signaling on the foreign body in form of biofilm, thereby defying the
(126). Similarly, pneumococci bound to PAFr via phosphoryl- hosts immune system and systemically administered anti-
choline are taken up into vacuoles but may then avoid fusion microbials. Finally, foreign body-associated meningitis fre-
of the vacuole with lysosomes. Instead, they are transported quently brings forth only low-grade inflammation, with fever,
to the basolateral surface of the endothelial cell and released irritability, and shunt malfunction being the most prevalent
out of the cell (127). Likewise, IbeA and CNF1 proteins are clinical findings (139,140). CSF shunt infections develop into
involved in the invasion of brain endothelial cells by E. coli clinical ventriculitis or meningitis only in approximately 30%
K1, ultimately leading to the uptake of E. coli into a vacuole of patients. In patients with hydrocephalus, CSF shunts con-
(116,128). An important aspect of transcytosis of meningeal vey CSF from the lateral cerebral ventricles to the peritoneum

Scheld_Ch23.indd 347 2/21/14 5:41 PM


348 Part III: Bacterial and Mycoplasmal Infections

or right cardiac atrium or transiently to an external collec- by the hematogenous or otogenic route, whereas sole cochle-
tion system. Shunt infection is one of the main complications ostomy surgery did not (161). The exact impact of the foreign
associated with CSF shunts. The reported incidence of CSF body in this setting has not been elucidated. To reduce the
shunt infection is in the range of 5% to 15% (141144), risk of postcochlear implantation meningitis, it is suggested
with pediatric patients being more affected than adults (143). that patients are thoroughly evaluated for accompanying inner
Shunt infections occur primarily as an early postoperative ear malformations, which may increase the risk for CSF leaks,
event, which manifests itself within a few days or weeks after and completion of their vaccination status. At surgery, mea-
surgery (139,142,143). Due to the close association with the sures should be taken to minimize communication between
operative procedure and the prevalence of bacteria belonging the middle and inner ear, as this is thought to be the mode of
to the skin flora, intraoperative inoculation is thought to be bacterial entrance in a sizable portion of meningitis cases in
the most likely cause of infection in early CSF shunt infection. patients with cochlear implant (162).
However, late shunt infections can occur several years after
implantation, likely due to hematogenous infection or associ-
ated with intraabdominal infection (139,143). Shunt coloniza- Bacterial Multiplication in the
tion with bloodborne pathogens such as H. influenzae or S. Cerebrospinal Fluid
pneumoniae has also been found, and one study reported five
cases of meningitis due to N. meningitidis, H. influenzae, and Host defense mechanisms in the CNS are tightly regulated.
S. pneumoniae out of a series of 289 patients with CSF shunt Peripheral immune cells and plasma proteins are largely excluded
followed for 10 years (145). Nevertheless, most shunt infec- from the brain parenchyma by the BBB and BCSFB, and the
tions are caused by staphylococci, in particular S. epidermidis, brain parenchyma additionally features anti-inflammatory
which causes half of all cases (143,144,146). The characteris- properties. Therefore, under physiologic conditions, microglia
tic feature of CSF shunt infection is the adherence of bacteria and astrocytes provide basic immune functions in the brain (rec-
to the inner surface of the shunt tubing. Insertion of a CSF ognition of pathogen-associated molecular patterns [PAMPs],
shunt leads to local inflammation and deposition of ECM pro- initiation of inflammation), whereas peripheral leukocytes cross
teins, that is, fibronectin or fibrinogen, on the shunt surface the BBB only in low numbers and stick to the perivascular space
(147). Binding of ECM proteins via fibrinogen-binding pro- of CNS microvessels (163). In bacterial meningitis, the perivas-
tein (148), fibronectin-binding protein (149), and vitronectin- cular space and the meninges are the interface where cellular
binding proteins (150) enhances adherence of S. epidermidis recognition of PAMPs and danger-associated molecular patterns
to the shunt surface. In an experimental model of catheter- elicits a strong inflammatory reaction and leads to the recruit-
associated infection, presence of a foreign body reduced the ment of peripheral immune cells to the CNS (164,165). In con-
number of bacteria needed to establish infection (151). Once trast to the peripheral blood, where cellular and soluble factors
attached to the shunt surface, S. epidermidis rapidly organizes of the innate immune system are present in abundance and may
into a biofilm, characterized by bacteria embedded in a self- act immediately on bacterial challenge, innate immune defense
generated polysaccharide matrix (152,153). Although shunt in the perivascular and meningeal space needs to be scaled up
infection does elicit an inflammatory response in the CNS first (166,167). This delay in immune activation favors the sur-
(154), staphylococci forming a biofilm are relatively protected vival of pathogens once they have reached the CSF. Bacteria can
from the host immune defense mechanisms (151,155,156). multiply within the CSF almost as efficiently as in vitro (168),
Additionally, bacteria organized in a biofilm have a greatly reaching titers of up to 109 CFU/mL, spread over the entire sur-
increased resistance to systemic antimicrobial therapy. This face of the brain and spinal cord, and extend into the Virchow-
is likely due to the diversity of metabolic states of bacteria Robins space along penetrating vessels. Interestingly, presence
inside the biofilm combined with the physical protection by or absence of leukocytes in the CSF does not affect bacterial
the extracellular polysaccharide matrix (153,157). Penetration multiplication in experimental meningitis models (169,170).
of most antimicrobials through the BBB does not suffice to In contrast, the complement system has been found to be in-
reach the necessary concentrations needed to kill bacteria in- strumental to the control of bacterial multiplication in the CSF,
side a biofilm (158), therefore intraventricular administration supporting leukocyte influx, cytokine expression, and bacterial
of antibiotics and shunt removal are frequently necessary to phagocytosis (171,172). Consistent with this, negative outcome
treat CSF shunt infection. in bacterial meningitis has been associated with depletion of
The pathogenesis of Gram-negative CSF shunt infection complement factors in the CSF (166,173).
is not well understood. Retrograde infection in which bowel Gene expression analyses of S. pneumoniae have shown
perforation may lead to distal colonization of the shunt with that, with exception of capsule genes, classical virulence genes
subsequent retrograde migration to the ventricles is frequently (i.e., encoding for pneumolysin, autolysin, and pyruvate oxi-
suggested as the most likely mechanism for Gram-negative dase) are downregulated in the CSF (174176). In contrast,
shunt infections. upregulation of genes involved in amino acid acquisition/
The several times higher risk of patients after cochlear synthesis and energy metabolism has been documented in
implantation to develop bacterial meningitis was first appreci- S. pneumoniae growing in CSF due to the scarcity of nutrients
ated in 2002. Depending on the type of cochlear implant used, in the CSF (175,176).
there was a 16 to 30 times higher incidence in bacterial menin-
gitis than in the general U.S. population (159). S. pneumoniae
accounted for 60% of the cases with known etiology (159). Induction of Inflammation
Incidence of bacterial meningitis in this special population has
since then decreased, but it is still at least twice as high as in Autolysis or exposure of bacteria to lytic antibiotics in the CSF
the general population (160). Cochlear implants are designed leads to the release of subcapsular components (fragments of
for people with profound bilateral sensorineural deafness in cell wall, lipopolysaccharide, teichoic and lipoteichoic acid,
whom the auditory nerves are intact but hearing aids are inef- peptidoglycans, bacterial DNA, and other cytosolic factors)
fective. In animal studies, the insertion of an electrode array that trigger the inflammatory response in the subarachnoid
into the scala tympani reduced the number of bacteria needed space. Different cells from the tissues lining the CSF (epen-
to induce meningitis in animals challenged with S. pneumoniae dymal and endothelial cells, perivascular macrophages) and

Scheld_Ch23.indd 348 2/21/14 5:41 PM


Chapter 23: Pathogenesis and Pathophysiology of Bacterial Infections 349

brain-resident cells (mostly microglia, but also astrocytes and in CSF samples of patients with bacterial meningitis, and
neuronal cells) are able to recognize these bacterial compo- its concentration is significantly correlated with inflamma-
nents. Distinct and often overlapping sets of specialized extra- tory parameters, TNF- concentrations, and adverse disease
cellular (i.e., TLRs) or intracellular (i.e., NOD-like receptors outcomes (213). IL-1 appears in the CSF of rats as early
[NLR]) receptors are used by these cells to sense the major as 30 minutes after intracisternal injection of H. influenzae
meningeal pathogens (177179). Genetic variations related to lipooligosaccharide (214) and 6 to 12 hours after injection
these receptors are associated with negative outcome during of live S. pneumoniae in infant rats (199). When injected
bacterial meningitis (180182). In response to bacterial stim- directly into the CSF of rabbits, IL-1 triggers a meningeal
uli, cells equipped with these specialized receptors produce inflammation without detectable TNF- activity, suggesting
cytokines, chemokines, and other proinflammatory molecules, an independent mechanism of action, whereas simultaneous
leading to the recruitment of leukocytes to the site of infection administration of TNF- and IL-1 results in a synergistic
(183,184). increase in leukocyte influx into the CSF. TNF- and IL-1
induce other secondary cytokines such as IL-6, IL-8, and
IL-10 (213).
Cytokines and Chemokines IL-6 is produced by monocytes, endothelial cells, and
A large variety of cytokines and chemokines have been de- astrocytes, essentially in response to IL-1. IL-6 is present in
tected in the brain parenchyma or the CSF during bacterial the CSF during meningitis later than TNF- and IL-1 and re-
meningitis in patients or in experimental models (185188). mains present longer than the latter cytokines. Although IL-6
The pattern of cyto-/chemokines produced in response to the can be detected in the CSF of patients with bacterial menin-
different meningeal pathogens has been shown to vary in in gitis, its presence is not correlated with any of the indices of
vitro and in experimental infection models. These differences meningeal inflammation or with severity of disease (215). IL-6
are presumably related to pathogen-specific activation of pat- has a predominantly proinflammatory effect and is a potent
tern recognition receptors (189192). They may contribute to inducer of acute-phase proteins, fever, leukocytosis, and acti-
the differences in outcome observed in patients affected by the vation of the complement and clotting cascades (216). Some
different pathogens. antiinflammatory effects of IL-6 include inhibition of TNF-
Early in the course of bacterial meningitis, tumor necrosis and IL-1 production in vitro and induction of IL-1 recep-
factor- (TNF-), interleukin (IL)-1, and IL-6 are released, tor antagonist (217). Accordingly, knockout mice lacking IL-6
which then trigger, often in synergy, a cascade of inflammatory display an increase in pleocytosis and in the inflammatory
mediators, including other cytokines, chemokines, platelet- response (218).
activating factor (PAF), antimicrobial peptides, prostaglan- IL-8 (CXCL8) was the first identified member of a large
dins, MMPs, NO, and ROS. The expression of TNF- and family of chemokines shown to regulate the migration of leu-
IL-1 is detected first in the ependyma and the meninges and kocyte subsets toward the CNS during bacterial meningitis
later in the parenchyma in experimental meningitis (194). (187). Similar to most chemokines, its primary action is to
Increased CSF concentrations of TNF-, IL-1, IL-6, IL-8, activate and attract leukocytes, mainly neutrophils, to sites of
and IL-10 are characteristic for bacterial meningitis. These cy- inflammation. Cells shown to produce IL-8 upon stimulation
tokines are predominantly proinflammatory with the excep- with TNF, IL-1, or bacterial products include monocyte-mac-
tion of IL-10, which downmodulates the production of TNF- rophages, neutrophils, endothelial cells, astrocytes, microg-
and other proinflammatory cytokines (194196). lia, and neurons. IL-8 is present in the CSF of patients with
High concentrations of TNF- in CSF were detected bacterial meningitis (187) and enhances neutrophil adhesion
within 3 hours after injection of live H. influenzae in an to endothelial cells, a prerequisite for the invasion of leu-
experimental meningitis model (197). Intracisternal injection kocytes into the brain. Belonging to the same chemokine
of S. pneumoniae led to a peak concentration of TNF- in CSF subfamily as IL-8, the concentrations of CXC5 (ENA-78)
approximately 12 to 24 hours after infection, with persistently and CXCL-1 (GRO-) increase in the CSF of patients with
elevated levels for at least 24 hours (198,199). The sustained bacterial meningitis, inducing neutrophil chemotaxis (119).
TNF- activity in the CSF may be explained by continuous To a lesser extent, another subfamily of chemokines, includ-
stimulation by products released from bacteria in the CSF or ing monocyte chemoattractant protein-1 (MCP-1, CCL2),
by a positive feedback loop in the inflammatory cascade (198). MIP-1 (CCL3), and MIP-1 (CCL3) is upregulated in the
TNF- leads to nuclear factor-B (NF-B) activation, which CSF of patients with bacterial meningitis and attracts mono-
regulates the expression of many proinflammatory genes (e.g., nuclear leukocytes (220).
IL-1, TNF-, IL-6, IL-8, macrophage inflammatory protein-1, IL-10 is an antiinflammatory cytokine that inhibits the
inducible NO synthase [iNOS], cyclooxygenase-2, ICAM-1) production of TNF-, IL-1, IL-6, and IL-8 in vitro and
(200,201). attenuates brain edema during meningitis. High levels of
Administration of TNF- into the CSF results in patho- IL-10 have been found in the CSF of patients with bacte-
physiologic changes characteristic of bacterial meningitis, rial meningitis, and in rabbits with experimental meningi-
including BBB breakdown, neutrophil influx, and increase of tis, administration of IL-10 downmodulated subarachnoid
cerebral metabolism, oxygen consumption, and cerebral blood space inflammation (221). In an experimental rat model of
flow (CBF), which in part are mediated through cyclooxygen- pneumococcal meningitis, systemically, but not intrathe-
ase activity (202207). Antibiotics cause rapid lysis of micro- cally, administered IL-10 reduced CSF pleocytosis and the
organisms and an associated brisk release of bacterial cell wall concentration of proinflammatory cytokines (222). The in-
products, resulting in significantly higher TNF- concentra- flammatory responses of IL-10 genedeficient and wild type
tions in CSF shortly after initiation of antimicrobial therapy mice were compared in a mouse model of meningitis induced
(208,209). by intranasal inoculation of S. pneumoniae. Although anti-
The proinflammatory cytokine IL-1 is released by bacterial defense and survival were not influenced, the ab-
mononuclear phagocytes, glial cells, and endothelial cells in sence of IL-10 was associated with an increased immune
the CNS. Upon recognition of intracellular pathogens, the response, as reflected by a more pronounced inflammatory
precursors of IL-1 or IL-18 are activated by caspase-1 in subarachnoid infiltrate and higher concentrations of proin-
the inflammasome complex (179, 210212). IL-1 is present flammatory cytokines and chemokines in brain tissue (223).

Scheld_Ch23.indd 349 2/21/14 5:41 PM


350 Part III: Bacterial and Mycoplasmal Infections

In experimental neonatal E. coli meningitis in mice, IL-10 and an increase in collagen degradation (237). In experi-
favors the immune response by increasing the phagocytic mental meningitis, treatment with MMP inhibitors led to a
activity of immune cells to clear the pathogen more rapidly significant reduction of mortality and seizure incidence and
and reduce the inflammatory response (224). In addition to reduced the extent of cortical damage (236,238). In addi-
IL-10, transforming growth factor- (TGF-) has also been tion to this effect, combined inhibition of MMP and TACE
shown to have antiinflammatory properties in experimental led to a reduction of hippocampal apoptosis and preserved
meningitis by reducing cerebral edema, intracranial pressure learning capacity of animals that recovered from bacterial
(ICP), and CBF (225). Conversely, by specifically blocking meningitis (198).
TGF- signaling on neutrophils and macrophages, neutrophil
influx and bacterial clearance were increased in experimental Neutrophil Invasion
pneumococcal meningitis, which resulted in less secondary
Neutrophil migration into the CSF is a distinctive feature of
brain damage and improved survival (226).
bacterial meningitis and contributes to the deleterious effects
of inflammation on the brain. In response to cytokines, che-
Matrix Metalloproteinases and Related Proteases mokines, and other chemotactic stimuli, neutrophils penetrate
The MMPs comprise endopeptidases that serve as effectors the microvascular basement membrane, leaving the blood-
of cell migration, tissue remodeling, and cytotoxicity by stream to accumulate in the CSF, where they produce the pro-
degradation of ECM components. MMPs are synthesized as found CSF pleocytosis characteristic of bacterial meningitis
inactive zymogens and can be activated by conformational (239241).
changes that disrupt a Zn2 binding cysteine switch. MMPs The entry of blood-derived neutrophils into the CSF is
not only function as effectors of tissue remodeling but facilitated by a cascade of events (Fig. 23.2). The initial step
also interact with the cytokine network. Cytokines such as in the adhesion cascade is the tethering and rolling of leu-
TNF- , IL-1, and IL-2 modulate the expression and regula- kocytes along the endothelium, which is mainly mediated by
tion of MMPs. In return, MMPs and related metalloprotein- the interaction of members of the selectin family and their
ases can act as sheddases or convertases as they transform glycoprotein ligands (242,243). E-selectin and P-selectin,
membrane-bound cytokines, cytokine receptors, and adhe- upregulated on the endothelial cell surface in response to
sion molecules to their soluble forms. TNF- converting proinflammatory stimuli, bind to P-selectin glycoprotein
enzyme (TACE/ADAM-17) is a member of the ADAM (a ligand-1 and other glycosylated ligands (243). The rapid
disintegrin and metalloproteinase) family of membrane pro- formation and breakup of these bonds on the neutrophil sur-
teins and is a highly efficient sheddase of TNF- and TNF face enable the rolling motion along the vessel wall (244).
receptors. These have an integral role in the network of Further, the hydrodynamic drag forces close to the vessel
MMPs and cytokines (227). wall induce the flattening of the rolling neutrophil. This re-
Most MMPs are expressed only on demand through the duces hydrodynamic drag and increases the area where se-
action of cytokines, eicosanoids, growth factors, and com- lectin-ligand bonds may be established (245). Consequently,
ponents of infectious pathogens, among others. The most in mice deficient in P-selectin, partial reduction of neutro-
prominent of these inducers are the proinflammatory cyto- phil influx was observed after cytokine-mediated induction
kines IL-1 and TNF-. MMP-2, in contrast to other MMPs, of sterile meningitis, whereas mice deficient in P-selectin
lacks a classic promoter sequence and is constitutively ex- and E-selectin showed almost complete inhibition of neu-
pressed. Tissue inhibitors of metalloproteinases (TIMPs), trophil influx in the same experimental model (246). The
the specific endogenous inhibitors of MMPs, form com- polysaccharide fucoidin is a selectin blocker that inhibits
plexes with pro- and active forms of MMPs and inhibit the leukocyte rolling and subsequent leukocyte transendothe-
enzymatic activity. Similar to MMPs, TIMPs are also regu- lial migration. Treatment with fucoidin intravenously has
lated by a network of different signaling molecules (228). been shown to attenuate the pleocytosis in experimental
MMPs appear to play a central role in the development of pneumococcal meningitis (247249). Rolling of leukocytes
bacterial meningitis (e.g., breakdown of the BBB, intrathecal along a chemotactic gradient is followed by firm adhesion
production of cytokines, and accumulation of blood-derived to the endothelium. Halting of the rolling motion is induced
leukocytes in the CSF) (229,230). Levels of MMP-1, -3, -7, by the activation of integrins on the neutrophil surface by
-8, -9, and -10 have been found to be upregulated in the chemokines (e.g., IL-8, C5a) (250). Activated leukocyte 1-
CSF of patients with bacterial meningitis, whereas TIMP-1 and 2-integrin (i.e., LFA-1) demonstrate higher affinity to
and TIMP-2 may be slightly upregulated. Interestingly, in members of the immunoglobulin-like superfamily on the
the same patients, TIMP-4 was downregulated in the CSF endothelium, including ICAM-1 and vascular cell adhe-
(231233). Immunohistochemical analysis of the brain tis- sion molecule-1 (VCAM-1) (251). After firm adhesion to
sue of patients with purulent meningoencephalitis revealed the vessel wall, neutrophils crawl, mediated by a different
endothelial cells and infiltrating leukocytes with an increased 2-integrin (i.e., Mac-1), to a suitable transmigration site
staining signal for MMP-9, TIMP-1, and TIMP-2 (234). CSF (252). Transgenic deletion of ICAM-1 in a mouse model of
levels of MMP-9 were significantly higher in those children brain inflammation and trauma led to a significant reduc-
who developed neurologic sequelae than in those who re- tion in the number of granulocytes infiltrating the brain tis-
covered fully, thus identifying high CSF concentrations of sue (253). Also, antibody-mediated blocking of integrins or
MMP-9 as a risk factor for the development of neuronal ICAM-1 was shown to decrease leukocyte influx in experi-
damage (235). mental meningitis models, confirming an important role for
Studies in a rat model of pneumococcal meningitis docu- these molecules in assisting leukocyte entry into inflamed
mented a 100-fold to 1,000-fold transcriptional induction of neural tissue (254256).
MMP-3, -8, -9, -12, -13, and -14, but not of MMP-2 and -7, Finally, the transmigration of leukocytes across the BBB may
in brain parenchymal tissue (198,236). In CSF cells, messen- occur by the paracellular or transcellular route (243,257,258).
ger RNA (mRNA) of MMP-8 and -9 were 10-fold to 100- Intracellular signaling induced by the interaction of integ-
fold increased, whereas MMP-2 and -7 remained at basal rins with their ligands leads to remodeling of endothelial cell
levels (198). The cortical brain damage in this experimental actin cytoskeleton and facilitates paracellular or transcellular
model is associated with changes in MMP-9/TIMP-1 ratio migration of leukocytes (257).

Scheld_Ch23.indd 350 2/21/14 5:41 PM


Chapter 23: Pathogenesis and Pathophysiology of Bacterial Infections 351

Histologic examination shows that the exudate in acute bac-


TARGETS OF DAMAGE terial meningitis consists predominantly of granulocytes,
IN MENINGITIS whereas there is a mixture of lymphocytes, macrophages, and
granulocytes in subacute to chronic forms of meningitis (259).
Figure 23.3 shows the histopathology of experimental pneu- Infiltration of vessel walls by these inflammatory cells, often in
mococcal meningitis and group B streptococcal meningitis. a focal pattern, is observed. Inflammatory alteration of the ves-
sel wall is often associated with permanent thrombosis of the
vessel lumen and vascular occlusion (260). Vascular occlusion
Cerebral Vasculature may also occur in absence of local inflammatory infiltrate (261).
Vascular complications have been shown to be a frequent occur-
Very early in the pathogenesis of meningitis, the cerebral vas- rence in children and adults with bacterial meningitis (262,263).
culature is at the center of processes that lead to the develop- In fatal cases of neonatal meningitis, inflammatory vasculitis is
ment of meningitis and CNS damage. Processes at the level of uniformly present, possibly indicating that the cerebral vascula-
the cerebral vasculature include activation of endothelial cells ture of the neonate is particularly susceptible to inflammatory
by inflammatory mediators produced during bacteremia, dis- damage (264). Some of the severe structural damage in the neo-
ruption of endothelial function by the traversal of meningeal natal brain following meningitis may be related to this suscepti-
pathogens and inflammatory cells from the bloodstream into bility of the vasculature to inflammatory damage.
the CSF, and involvement of the vasculature by the granulo-
cytic inflammation in the subarachnoid space. Important con- Disruption of the BloodBrain Barrier
sequences of these processes are the disruption of the BBB, Under physiologic condition, the BBB and BCSFB act as bar-
brain edema, loss of CBF autoregulation, and focal and global riers to the brain, selectively limiting the entry of macromol-
changes of CBF resulting in cerebral ischemia. ecules and cells. Access of leukocytes to the CNS is therefore
tightly regulated, and in the healthy individual, only very few
Pathology leukocytes transmigrate the BBB and BCSFB (163). In men-
Subarachnoid space inflammation appears as a gray-yellow ingitis, however, the permeability of these brain barriers in-
to green exudate covering the base and convexities of the creases as a result of functionally relevant alterations induced
brain with obvious involvement of cerebral arteries and veins. by the disease process.

FIGURE 23.3 Histopathology of experimental pneumococcal meningitis (AD) and group B


streptococcal meningitis (E). A: Extensive cortical injury consisting of confluent areas of cortical
necrosis. Areas with markedly reduced neuronal density occur in a wedge-shaped distribution
(black arrowhead) suggestive of ischemic damage. The hippocampal dentate gyrus is also shown
(white arrowhead) (cresyl violet, magnification 10). B: Focus of cortical neuronal loss on the
right contains neurons with morphologic features of necrosis, including cell swelling and fading
of cytoarchitecture and is sharply demarcated from preserved neurons on the left (cresyl violet,
magnification 40). C: Subarachnoid space inflammation consisting of bacteria and inflamma-
tory cells extending into the Virchow-Robin space around the penetrating cortical vasculature.
D: Hippocampal dentate gyrus histology of an infant rat suffering from pneumococcal meningitis
at 42 hours after infection. Apoptotic cells are characterized by the presence of round or oval
apoptotic bodies consisting of two or more round, regularly shaped, dark chromatin clumps
(inset). The occurrence of apoptosis is characteristically observed in the inner rim (i.e., the sub-
granular zone) of the dentate gyrus (arrowheads) (cresyl violet, magnification 40). E: Dentate
gyrus of the hippocampus with a sharply demarkated (arrowheads) zone of injury containing
clustered cells with dense and shrunken nuclei demonstrating the appearance of uniformly pyk-
notic nuclei throughout the entire blade (lower right quadrant). (cresyl violet, magnification 40).

Scheld_Ch23.indd 351 2/21/14 5:41 PM


352 Part III: Bacterial and Mycoplasmal Infections

In this respect, enhancement of BBB permeability due to in traumatic brain injury and multiple sclerosis (291,292).
an increase in paracellular leakage has been particularly stud- Interestingly, experimental and clinical evidence does not sup-
ied. Tight junctions and the corresponding intracellular actin port a role for VEGF in the disruption of the BBB in bacterial
cytoskeleton are the most important components of the BBB meningitis (186,291,293).
that regulate paracellular transport pathways. Remodeling of Besides their chemoattractant properties, chemokines also
the actin cytoskeleton, reorganization of tight junctions, and exert a direct effect on BBB permeability. For example, the
enzymatic degradation of tight junctions and basement mem- chemokines IL-8 and MCP-1 lead to cytoskeleton rearrange-
brane components may all lead to increased BBB permeability ments (294).
(257,265). These changes are mediated by direct damage to Massive neutrophil pleocytosis in the CSF space is a hall-
endothelial cells by the infecting pathogen, alterations induced mark feature of bacterial meningitis (see previous discussion).
by signaling and effector molecules released during the inflam- Both the paracellular and transcellular route are accepted for
mation process, and changes induced by the interaction of in- the diapedesis of leukocytes across the BBB (257,258). The
flammatory cells with the brain barriers. effect of neutrophils on BBB permeability may be divided in
In vitro cytotoxic damage to brain endothelial cells has release of effector molecules like MMP-9 and ROS and direct
been seen with S. pneumoniae, mainly mediated by pneu- opening of paracellular pathways. Downstream signaling
molysin (266268), and N. meningitidis, mainly mediated induced by adhesion of leukocytes to ICAM-1 and VCAM
by NO (269). Pneumolysin was also shown to lead to as- induces remodeling of the endothelial cytoskeleton and reor-
trocyte cytoskeleton remodeling in vitro (270), which might ganization of tight junctions, ultimately increasing paracellu-
also support BBB disruption. Nevertheless, the importance of lar permeability of the BBB (257).
direct cytotoxic damage to the brain endothelial cells in vivo As a result of the increased BBB permeability during menin-
is not known. gitis, molecules in the blood, including antibiotics, penetrate at
Different proinflammatory molecules produced by endo- increased rates into the CSF. This facilitates therapy, because
thelial cells, circulating leukocytes, microglia, or astrocytes the CSF/serum ratio of antibiotic concentration is often higher
may induce disruption of the BBB (271). Some of the cyto- than without inflammation (295). Other molecules penetrating
kines that have been associated with disruption of the BBB the BBB may, however, be harmful. It is hypothesized that the
are TNF-, IFN-, IL-1, and IL-6 (218,272). Mechanisms disruption of the BBB leads to alterations in the brains micro-
by which cytokines may increase permeability of endothelial environment that contribute to neuronal demise, for example,
tissue include rearrangement of cellular cytoskeleton and tight by leading to higher concentrations of excitatory amino acids
junction organization, activation of metalloproteinases, and (EAA) (see later discussion) and other potentially harmful
increased generation of ROS. In vitro evidence for a direct molecules present at high concentrations in the bloodstream.
effect of TNF- and IFN- on the endothelial cytoskeleton
and intercellular tight junctions mainly stems from non-CNS
endothelial models (273), whereas direct effects on brain en-
Alterations of Cerebral Blood Flow
dothelial cells are disputed (274276). A direct effect of IL-1 Bacterial meningitis is associated with marked changes in CBF.
on BBB permeability via protein kinase C theta mediated phos- Early in the course of the disease, an increase in blood flow is
phorylation of zona occludens-1 has been demonstrated (277). observed, whereas in advanced meningitis, CBF is reduced
MMP-3 and MMP-9 are, via NF-B upregulation, some (262,279,280,296299). Focal changes in the vasculature, loss of
of the main downstream effector molecules in BBB disrup- CBF autoregulation, ICP, and systemic hypotension may all lead
tion triggered by the cytokines TNF- and IL-1, but MMP-9 to perturbation of CBF in bacterial meningitis. Cerebrovascular
may also be activated by NO or MMP-3 (278). MMPs may changes documented in patients with vascular complications in-
increase BBB permeability by breakdown of basement mem- clude segmental narrowing of vessels and irregularities of vessel
brane and tight junctions. Significantly increased CSF levels of walls (134,300). Additionally, arterial and venous thrombo-
MMP-8 and MMP-9 have consistently been found in human ses are frequently documented (261,301). Vasculitis (301) and
meningitis cases (230232,235). Further, in a rat model of vasospasms (299) likely explain segmental alteration of vessel
meningococcal meningitis, disruption of the BBB, increased wall diameter in meningitis, whereas a procoagulable disposi-
ICP, and CSF pleocytosis were paralleled by the occurrence of tion caused by the bacterial infection may additionally explain
MMP-9 activity in the CSF 6 hours after bacterial challenge vascular thrombosis (261). The middle and anterior cerebral ar-
(229). Besides MMPs, ROS and NO have been identified in teries or the basilar artery are frequently affected during stroke
experimental studies as key mediators in the pathophysiology associated with bacterial meningitis (134,262). Sequelae are
of bacterial meningitis (279282). Both molecular mediators most devastating when large vessels at the base of the brain are
contribute to the development of increased ICP, BBB disrup- affected, resulting, for example, in hemiparesis or quadriparesis
tion, and CSF leukocytosis (283). Inhibition of the biologic (264,302,303).
effects of ROS and NO by radical scavengers in experimental Impairment of CBF autoregulation has been documented
meningitis prevented cerebral damage including BBB break- in bacterial meningitis, both in experimental models and in
down (279,282,284,285). There are several pathways by clinical studies (279,298,299,304308). As a result of loss of
which ROS and NO may induce BBB/BCSFB disruption. Cell CBF autoregulation, systemic hypotension, which may arise in
membrane dysfunction mediated by lipid peroxidation and patients with meningitis as a result of sepsis or severe dehydra-
oxidative damage to cell membrane proteins, tight junction tion, cannot be compensated and may lead to cerebral ischemia.
and cytoskeletal reorganization, and MMP activation may Conversely, systemic hypertension may then augment vaso-
all contribute to BBB disruption (286). In clinical and experi- genic edema and ICP. Elevated ICP has been associated with
mental studies, markers for lipid peroxidation were elevated an increase in CBF in experimental pneumococcal meningitis
in the CSF in bacterial meningitis (287,288). Furthermore, in (309,310). Ultimately, in the rigid framework provided by the
experimental studies, pharmacologic inhibition of lipid perox- cranium, an excessive increase in ICP may limit CBF. Factors
idation reduced ICP and cerebral edema (289), whereas phar- leading to increased ICP in bacterial meningitis are cerebral
macologic ROS inhibition lowered the level of peroxidation edema (see later discussion) and increased blood volume in the
markers in the CSF (280,290). Vascular endothelial growth brain resulting from hyperemia or from venous congestion pre-
factor (VEGF) is also involved in the disruption of the BBB cipitated by thrombotic occlusion of inflamed veins (304).

Scheld_Ch23.indd 352 2/21/14 5:41 PM


Chapter 23: Pathogenesis and Pathophysiology of Bacterial Infections 353

In addition to global cerebral hypoperfusion, brain regions


with focal hypoperfusion caused by vasculitis of large and Inner Ear
small arteries traversing the inflamed subarachnoid space are
frequently observed. This form of vasculitis is thought to be Unilateral or bilateral hearing impairment is the most com-
mainly responsible for ischemic damage leading to perma- mon neurologic sequelae following bacterial meningitis and is
nent neurologic sequelae (Fig. 23.3A and B) (279,303). NO found in 5% to 30% of survivors (323326). S. pneumoniae
plays a crucial albeit complex role in modulating CBF during meningitis is associated with the highest risk of developing
meningitis. In experimental models of bacterial meningitis, sensorineural hearing loss (327330). Onset of hearing loss
inhibition of NO formation by a nonselective, competitive during bacterial meningitis is progressive rather than abrupt
inhibitor of NOS reduced the hyperemia observed early in and the magnitude of hearing loss is related to the duration
the disease (279,311,312). In more advanced disease, inhibi- of untreated infection (324). Hearing loss consecutive to bac-
tion of iNOS significantly increased the extent of ischemia terial meningitis can be transient or permanent. Studies in
and neuronal injury in the brain (296). These studies suggest models of meningitis indicate that hearing loss is the result of
a dynamic role of NO at the level of the cerebral vascula- direct involvement of the inner ear by the inflammation dur-
ture during meningitis. Early in the disease, the vasodilatory ing the acute phase of the disease. However, no close correla-
effect of NO contributes to the hyperemia induced by the tion could be found between the extent of hearing loss and the
subarachnoid space inflammation. Later, when CBF progres- magnitude of CSF pleocytosis or CSF bacterial concentrations
sively declines under the influence of vasoconstrictive factors (324). Magnetic resonance imaging (MRI) studies in humans
(see later discussion), NO produced in the vasculature has with meningitis have documented inflammatory involvement
some protective effects against ischemia. Thus, attempts to of the inner ear, suggesting that the animal models reflect the
downmodulate NO production during meningitis are poten- clinical situation (331). In survivors of meningitis, progressive
tially dangerous, despite the evidence that NO contributes to cochlear ossification and spiral ganglion loss is observed years
some of the potentially harmful changes in bacterial menin- after the disease (332,333).
gitis, such as CSF inflammation, brain edema, and ICP (see
later discussion).
Pathology
ROS also play a critical role in modulating CBF during men- During the acute stage of meningitis, suppurative labyrinthitis
ingitis (280,289,312,313). During the early phases of pneu- is a common feature observed in the human temporal bone
mococcal meningitis in rats, scavenging of O2 and of H2O2 of patients with bacterial meningitis, often accompanied by
by superoxide dismutase and catalase prevented the develop- pus in the perilymphatic duct (334). Similarly, histopathologic
ment of hyperemia and increased ICP (312,313). Generation examination of the temporal bone from rats with bacterial men-
of ROS is localized primarily to the cells of the subarachnoid ingitis showed a dense inflammatory cell infiltrate throughout
and ventricular inflammation and to the cerebral vasculature, the subarachnoid space extending to the inner ear. Studies in
as shown in infant rats with experimental meningitis (280). experimental meningitis revealed that in the earliest stages of
The likely cellular sources of ROS include granulocytes, pneumococcal infection, bacteria and inflammatory cells are
endothelial cells, and activated microglial cells. Interestingly, already present within the cochlea (335,336). Inflammation
the cerebral vasculature shows evidence of marked oxidative in the inner ear is primarily confined to the perilymphatic
alterations during experimental pneumococcal meningitis in space (scala tympani) (337,338). Inflammatory infiltration
infant rats, whereas oxidative damage to the brain paren- of the cochlea was shown to progress via the cochlear aque-
chyma itself has not been yet documented conclusively (314). duct to the perilymphatic space and via the spiral ligament to
The oxidative damage to the vasculature can be inhibited by the endolymphatic space during experimental pneumococcal
treatment with antioxidants. The same antioxidants have been meningitis (336). During this phase, which is characterized by
shown to reduce cerebral ischemic damage in these models high inflammation, damage to the bloodlabyrinth barrier, the
and prevent the decline in CBF (280,281). These data suggest hair cells, and the spiral ganglion is observed (339). Damage
that the cerebral vasculature is exposed to significant oxida- begins at the base of the cochlea and progresses to the apex,
tive stress during meningitis, which in turn contributes to CBF corresponding to hearing loss first at high then at low frequen-
reduction and cerebral ischemia. cies. Studies in experimental meningitis caused by E. coli and
NO and ROS can chemically react and form peroxyni- S. pneumoniae revealed that both pathogens invaded the scala
trite, which is a strong oxidant that exerts cytotoxic effects tympani and triggered the development of lesions including
(283,315). In addition, the reaction leads to the loss of the craters in the apical surface of inner supporting cells and dis-
vasodilatory biologic effect of NO. Nitrotyrosine residues on ruption of the inner hair cell stereocilia. Pneumococcal men-
proteins as a marker for the presence of peroxynitrite were ingitis produced more pronounced lesions and induced breaks
detected in the meninges, the cortical blood vessels penetrating in the junctions between inner hair cells and their adjacent
the subarachnoid space, and inflammatory cells in the brains supporting cells, as well as ballooning and rupture of the api-
of patients with bacterial meningitis and corresponding ani- cal surface of outer hair cells (340). Spiral ganglion neuronal
mal models (283,316). Furthermore, pretreatment with the loss is observed later than the first signs of hair cell damage
peroxynitrite scavenger urate attenuated meningeal inflamma- and hearing loss. In the early stage of inflammation, hearing
tion, BBB disruption, and intracranial hypertension (316). threshold changes are associated with nonlethal structural
Endothelins, potent vasoconstrictor peptides produced in changes in cochlear hair cells and are potentially reversible.
the CNS by vascular endothelial cells, astrocytes, and neurons, Conversely, in more advanced experimental infection, irrepa-
are also involved in CBF dysregulation. Increased endothelin rable ultrastructural inner ear damage, especially the loss of
levels are found in the CSF of patients with bacterial men- spiral ganglion neuronal cells, is associated with severe to pro-
ingitis (317). Endothelin synthesis is triggered by cytokines, found deafness (335,341).The more severe cochlear lesions
that is, TNF- (318320), and inhibited by NO (321). In induced by S. pneumoniae may explain the higher incidence
experimental pneumococcal meningitis, therapy with an endo- of deafness after pneumococcal meningitis. Toxic effects of
thelin antagonist significantly prevented the reduction of CBF the meningeal pathogen (e.g., pneumolysin from S. pneu-
induced by the infection and concomitantly reduced the extent moniae [342]) and inflammatory mediators appear to be
of cerebral ischemia (322). responsible for the cytopathic effects (340,343). In particular,

Scheld_Ch23.indd 353 2/21/14 5:41 PM


354 Part III: Bacterial and Mycoplasmal Infections

the production of ROS seems to be crucial for the pathogen- Pathology


esis of cochlear damage and hearing loss in bacterial meningi-
tis (339). Inducible and endothelial NO synthase expression The neuropathology of bacterial meningitis includes subarach-
and tyrosine nitration have been documented in the cochlea noid space inflammation, inflammatory involvement of the
of rats with pneumococcal meningitis (344). Treatment with cerebral vasculature (see previous discussion), and evidence
peroxynitrite scavengers or antioxidants resulted in reduced of parenchymal brain damage. Damage to the brain paren-
hearing loss and protection of spiral ganglion neuronal cells chyma is evidenced by the presence of brain edema, including
(345,346). Upon microperfusion in cochleas, NO donors have signs of cerebral herniation (see later discussion), by areas of
been demonstrated to damage hair cells and supporting neu- cerebral infarction resulting from ischemia, and by histologic
ronal cells (347,348). Furthermore, in vitro, exposure to NO changes (370374). The latter show loss of neurons, often in
causes irreversible morphologic changes in isolated outer hair a focal pattern (Fig. 23.3A and B), which is most prominent in
cells (349). A direct neurotoxic effect of ROS on hair cells patients who survive acute meningitis for several days before
or spiral ganglion cells seems therefore plausible, but other succumbing to the disease. MRI performed on newborn or
possible points of action may also exist, like damage to the children affected by bacterial meningitis showed infarctions
bloodlabyrinth barrier (339). in the frontal, temporal, and parietal lobes; the basal gan-
Damage to the cochlea and hearing loss is prevented glia; and the thalamus, as well as periventricular white mat-
by antioxidants or NO synthase inhibitors in experimen- ter injuries, leading to cerebral atrophy and hydrocephalus
tal studies (344346). Similarly, strategies to limit the host (263,375377). In adult patients with bacterial meningitis,
inflammatory reaction by using doxycycline (350) or non- the temporal lobe and the limbic system also displayed struc-
lytic antibiotics (351) are promising adjuvant therapies tural changes (367). Neuronal loss is associated with a marked
reducing sensorineural hearing loss. Conflicting results have reaction of astrocytes and microglia (378) as well as axonal
been published concerning the use of dexamethasone in both injury (379,380). Furthermore, meningitis induced by Gram-
experimental and clinical studies. Whereas some experimen- negative bacteria is often characterized by the development of
tal models showed an improvement in hearing capacity by brain abscesses (381,382).
dexamethasone (352354), others did not (355). Similarly, In the dentate gyrus of the hippocampus, special forms of
the beneficial use of dexamethasone in clinical practice is cell death have been observed during meningitis (280,383,384)
inconsistent (356360) and may differ depending on the (Fig. 23.3D and E). The first form of injury is defined as apop-
causative agent, the age of the patient, comorbidity, and tosis based on morphologic criteria in cresyl violetstained
socioeconomic factors. brain sections (condensed, fragmented nuclei) (Fig. 23.3D,
inset) and the detection of fragmented DNA (i.e., TUNEL
stain) (385). Furthermore, activated caspase-3, an effector cas-
pase that is responsible for executing the cell death program,
Central Nervous Tissue has been documented in apoptotic neurons of the hippocam-
pal dentate gyrus in experimental meningitis (384,385). The
specific role of caspase-3 in this form of neuronal apoptosis
Neurologic Sequelae
was documented in infant rats with pneumococcal meningitis,
Apart from the high mortality, bacterial meningitis often where enzymatic caspase-3 activity was significantly increased
causes brain damage of both cortical and subcortical struc- and intracisternal administration of the caspase-3specific
tures. The neurologic sequelae resulting from brain damage inhibitor Ac-DEVD-CHO significantly reduced apoptosis
include hearing impairment (see previous discussion), obstruc- in the hippocampus of infected animals (385). In a different
tive hydrocephalus, and damage to the brain parenchyma approach, caspase-3 activation and hippocampal damage was
with focal sensorimotor deficits, mental retardation, seizure prevented by treatment with a pan-caspase inhibitor, which
disorders, and cortical blindness. In a meta-analysis, hearing exhibits antiinflammatory activity through blocking of inter-
loss was the most frequent sequelae, almost three times more leukin-converting enzyme (386).
frequent than seizures and motor deficits, respectively (323). The apoptotic injury in the hippocampus is of particular
Less frequent were cognitive impairment, hydrocephalus, and significance because experimental data suggest that it is related
visual disturbance. Any sequelae after discharge was present to learning impairment following meningitis (198,387,388).
in 19.9% of patients with bacterial meningitis. The risk for a Such an association is also supported by the observation that
major sequelae after bacterial meningitis (i.e., cognitive deficit, activated caspase-3 and morphologic evidence of apoptosis is
bilateral hearing loss, motor deficit, seizures, visual impair- primarily localized to immature progenitor cells in the subgran-
ment, hydrocephalus) was at least twice as high in African ular zone of the dentate gyrus, cells that have been implicated
and Asian countries compared to European countries (323). in the acquisition of new memory (384,385,389). Damage
Bacterial meningitis caused by S. pneumoniae has consistently to the dentate gyrus of the hippocampus may thus represent
been associated with higher rates of disabilities than infection an anatomic substrate for cognitive impairment and learning
with H. influenzae or N. meningitidis (135,323,361). Lately, disabilities following meningitis (198,387,388,390). As a con-
behavioral and cognitive sequelae in children and adults after sequence of apoptotic damage, hippocampal neurogenesis is
bacterial meningitis have been increasingly and accurately ap- impaired after bacterial meningitis (391). Furthermore, hip-
preciated (362367). This is of special concern in childhood pocampal cell death associated with inflammatory conditions
meningitis, as it was shown that neurologic sequelae of child- results in a dysregulation of hippocampal neurogenesis, with a
hood meningitis may persist for more than 10 years, thus im- transient increase in cell proliferation (392394) and changes
pairing learning during the entire time children attend school in the transcriptional profile in the stem cell population of the
(363,365,368,369). In adults tested 0.5 to 13.5 years after dentate gyrus (i.e., the neurogenic niche) (395).
acute bacterial meningitis, 32% of 155 meningitis survivors Confirming the findings in animal models, brain sections
suffered from relevant impairment of psychomotor perfor- of 20 patients who died from bacterial meningitis showed
mance, speed of cognitive processes, and concentration and apoptotic neurons with immunoreactivity for precursor and
memory functions (366). Overall, these studies emphasize the active forms of caspase-3 in the dentate gyrus (396). In a study
substantial neurologic damage and functional impairment re- in patients surviving meningitis, volumetric measurements
sulting from bacterial meningitis in children and adults. of the hippocampus by MRI techniques showed unilateral

Scheld_Ch23.indd 354 2/21/14 5:41 PM


Chapter 23: Pathogenesis and Pathophysiology of Bacterial Infections 355

and bilateral hippocampal atrophy, potentially reflecting the formation by increasing transcellular influx of water across
apoptotic loss of neurons observed by histopathology (397). the BBB, whereas in vasogenic and interstitial brain edema,
Furthermore, cell proliferation in the hippocampus was also lack of AQP-4 leads to more severe brain edema by limiting
found to be dysregulated in human patients who succumb to outflow of CSF (404,416). In a mouse model of pneumococ-
bacterial meningitis (398). cal meningitis, AQP-4 was significantly upregulated and as-
The second form of hippocampal neuronal damage is sociated with increased cytotoxic brain edema (417). AQP-4
characterized morphologically by uniformly shrunken nuclei, upregulation was also found in the brain of a patient with
forming clusters of damaged cells predominantly in the lower bacterial meningitis (418), but not in the CSF of patients with
blade of the dentate gyrus, spanning the entire width of the bacterial meningitis (419).
dentate gyrus band (Fig. 23.3E). This form of hippocampal Severe brain edema results in a caudal shift of midline struc-
damage is the preferential pattern of neuronal injury observed tures, leading to their entrapment in the tentorial notch or fo-
in experimental meningitis caused by GBS (280,384). Criteria ramen magnum. The most common type of brain herniation
for classical apoptosis, such as formation of apoptotic bodies occurs when a portion of the temporal lobe is displaced (uncal
and positive staining for activated caspase-3, as well as mor- herniation), which compresses cranial nerve III, the midbrain,
phologic criteria for necrosis, for example, cell swelling, loss and the posterior cerebral artery, leading to coma and respi-
of cellular structures, are absent (384). However, this form of ratory arrest. Caudal shifts produce herniation of the para-
neuronal injury may be linked to apoptosis-inducing factor hippocampal gyri and/or cerebellum, which occurs when part
(AIF) (399,400), more reminiscent of ischemia-related neuro- of the cerebellum is displaced through the foramen magnum.
nal damage (401). Therefore, both forms of damage may be These intracranial processes are manifested clinically in altered
temporally distinct and caused by distinct mediators (402). consciousness and postural reflexes. Caudal displacement
of the brainstem causes palsy of the third and sixth cranial
Brain Edema and Cerebral Herniation nerves. Ultimately, these changes can result in decortication or
decerebration with rapid progression to respiratory and car-
The development of cerebral edema is a hallmark of the ce- diac arrest. Cerebral herniation is occasionally precipitated by
rebral involvement during meningitis (403). Cerebral edema lumbar puncture in the setting of brain edema and increased
may be classified as vasogenic, cytotoxic, interstitial, or ICP. The highest risk for cerebral herniation is in patients in
osmotic (404). Cerebral edema in bacterial meningitis may be whom there is a focal brain lesion leading to midline shift and
a combination of all four forms. Vasogenic cerebral edema is intracranial hypertension. In these patients, performance of a
primarily a consequence of increased BBB permeability, which lumbar puncture is contraindicated (420).
leads to extravasation of plasma proteins into the brain pa- Factors other than brain edema and intracranial hyperten-
renchyma (see earlier discussion) and mainly affects the white sion that may lead to acute death from bacterial meningitis
matter (404). Cytotoxic edema results from an increase in include extensive cerebral infarction resulting from vasculitis
intracellular water following intracellular accumulation of and coagulation disturbances and/or circulatory failure result-
osmotically effective ions (i.e., sodium, potassium, or gluta- ing from septic shock (421).
mate). Cytotoxic mechanisms include ischemia and the effect
of EAAs (404406). Cytotoxic edema affects gray and white
matter in the brain. Interstitial edema occurs by an increased
Mediators of Cell Death in Neuronal Tissue
influx of CSF across the ventricular ependyma into the peri- EAAs including glutamate have been proposed as mediators
ventricular white matter, either through increased CSF produc- of brain damage by inducing either apoptosis or necrosis and
tion (increased blood flow in the choroid plexus) or decreased appear to mediate neuronal injury in a variety of brain disor-
resorption secondary to increased CSF outflow resistance ders. In ischemia, the best studied example of EAA-mediated
across the arachnoid villi system of the sagittal sinus (407). In injury, increased concentrations of EAA in the brain intersti-
obstructive hydrocephalus after meningitis, chronic interstitial tial fluid result from an increased release by ischemic neurons
edema may lead to destruction of brain tissue (408). Osmotic and reduced uptake by glial cells (422). Studies in experimen-
edema might be caused by inappropriate secretion of antidi- tal models of bacterial meningitis and inference from clinical
uretic hormone during the course of bacterial meningitis. The studies point to ischemia and EAA as likely mediators of direct
observed hypoosmolality of serum results in a net influx of neuronal toxicity (378,406).
water into the brain (404,409). A role for EAA in mediating neuronal injury in meningi-
Brain edema contributes substantially to the acute fatal tis is supported by the following evidence. Glutamate con-
outcome of bacterial meningitis (134,410,411). The major centrations in the CSF of patients with bacterial meningitis
dangers of extensive brain edema during meningitis are herni- were significantly elevated compared to those in patients with
ation of brain tissue and compression of the brainstem due to viral meningitis and noninflammatory neurologic diseases
increased ICP, which can cause complete cessation of cerebral and correlated with the severity of the disease as scored by
circulation. But increased ICP alone may compromise cerebral the Glasgow Coma Scale (423,424). In a rabbit model, gluta-
perfusion and lead to ischemia. In addition to brain edema mate concentrations in the brain interstitial fluid, as measured
itself, other factors associated with bacterial meningitis can by microdialysis probes, were significantly increased in pneu-
contribute to excessive intracerebral hypertension, including mococcal meningitis (424). The source of the increased EAA
the development of obstructive hydrocephalus, meningitis- concentrations in the brain during meningitis has not been
associated cerebritis, cerebral infarction, cerebral venous identified but may be related to BBB disruption. Furthermore,
thrombosis, and status epilepticus (412,413). in an infant rat model of neonatal meningitis, kynurenic acid,
Impaired regulation of water transport underlies the patho- a nonselective inhibitor of the neurotoxic effect of EAA,
physiology of brain edema. Aquaporins (AQP) are pore-form- significantly attenuated brain injury, both in the cortex and
ing membrane proteins ubiquitously present in living organisms in the hippocampus (378,406). However, adjuvant treat-
(414). AQP-1, AQP-4, and AQP-9 have been documented in ment with dextromethorphan, a noncompetitive N-methyl-
the primate brain and are involved in water homeostasis of d-aspartate (NMDA) receptor antagonist, was found to
the brain (415). The major aquaporin in the brain, AQP-4, aggravate hippocampal apoptosis in the infant rat model of
shows contradictory effects in several forms of brain edema. In pneumococcal meningitis (425). Furthermore, accumula-
cytotoxic edema, AQP-4 was shown to support brain edema tion of endogenous kynurenic acid by combined inhibition

Scheld_Ch23.indd 355 2/21/14 5:41 PM


356 Part III: Bacterial and Mycoplasmal Infections

of kynurenine 3-hydroxylase and kynurenine kinase resulted in its oxidized form, it can inactivate glutamate receptors
in an aggravation of apoptotic neuronal death in the hippo- and reduce the neurotoxicity of EAA, whereas in the reduced
campal dentate gyrus in the same experimental model; in this form, it can react with superoxide to form peroxynitrite,
case, nicotinamide adenine dinucleotide depletion, observed as which can exert cytotoxic effects by damaging DNA, lipid
a result of the intervention, may have caused an energy failure membranes, and proteins (437). Genetic inactivation of iNOS
responsible for the increase in apoptosis (426). results in a marked reduction of caspase-3dependent hippo-
Inflammatory factors such as cytokines, metalloproteinases, campal apoptosis in a mouse model of pneumococcal menin-
and ROS are produced by leukocytes, monocytes, and microg- gitis (438). However, iNOS inhibition by aminoguanidine has
lia as a result of the host immune response. The intensity and been shown to increase neuronal injury in experimental GBS
duration of this response are responsible for the development meningitis (296).
of neuronal damage. Reducing the intensity by the use of anti- MMPs, whose role in CSF inflammation, recruitment of
inflammatory corticosteroids or nonlytic antibiotics may be a granulocytes into the subarachnoid space, and disruption of
therapeutic option (427). Some pathogens have been shown the BBB has been described earlier in this chapter, may also
to interfere with the activation-induced cell death of immune exert direct neurotoxic effects (227) by degrading perineuro-
cells, possibly prolonging in a detrimental way the extent of nal components of the ECM, such as laminin (439,440) or the
inflammation. For example, N. meningitidis inhibits apopto- neural cell adhesion molecule (NCAM) (441). Furthermore,
sis in neutrophils (428) or macrophages (429). Similarly, GBS the modulation of the shedding of death receptors ligands
interfere with TLR-2mediated activation-induced cell death (FasL, TNF-) by MMPs and TIMPs can directly regulate cell
in macrophages and microglia (430). Therefore, modulating death in cells that express these ligands, including neurons
the lifespan of the cells producing neurotoxic factors may be a (278). Given as adjuvant therapy in addition to antibiotics,
beneficial treatment during bacterial meningitis (431). Because MMP inhibitors reduced the extent of cortical damage, and
neutrophils produce different presumably neurotoxic factors combined inhibition of MMP and TACE led to a reduction
(cytokines, metalloproteinases, ROS), often modulating each in hippocampal apoptosis (198,236,238). The adjunctive
other, it is difficult to determine which really directly triggers therapy with MMP-TACE inhibitors also preserved learning
neuronal cell death (432). capacity in experimental animals after recovery from bacterial
The marked increase in ROS production during certain meningitis (198).
pathologic conditions, such as during acute and chronic Apart from host-derived neurotoxic mediators, there is
inflammation, ischemia/reperfusion, or trauma, can lead emerging evidence that bacteria may also directly damage
to oxidative cell and tissue injury. The brain is believed to neurons. An example of such a direct bacterial action is the
be particularly vulnerable to oxidative damage because ability of pneumococci to induce apoptosis in vitro (400).
of its high concentrations of unsaturated fatty acids, high Potential neurotoxic mediators produced by S. pneumoniae
rate of oxygen consumption, and relatively low concentra- include pneumolysin and superoxide. In vitro studies revealed
tions of antioxidants (433). Compatible with an important that pneumolysin and bacteria-derived superoxide initiated
role of ROS in experimental meningitis, the use of radical the neuronal death cascade via damage to mitochondria and
scavengers prevented BBB breakdown, attenuated lipid per- subsequent release of the proapoptotic mitochondrial fac-
oxidation and ischemia in the brain parenchyma, and pro- tors cytochrome c and AIF (442,443). Studies in experimen-
tected neurons from injury in experimental meningitis models tal pneumococcal meningitis in rabbits demonstrated that
(434). Furthermore, the neurotoxic effect of EAAs has been pneumolysin colocalized with apoptotic neurons of the hip-
shown to involve ROS (435). Despite these observations, a di- pocampus, and infection with pneumococcal mutants unable
rect neurotoxic effect of ROS in the brain parenchyma during to produce pneumolysin and superoxide caused significantly
meningitis has been difficult to document conclusively. In the less damage (442). Furthermore, pneumolysin was also im-
infant rat model of pneumococcal meningitis, there was only plicated in the mechanism of cochlear hair cell death in the
a moderate loss of soluble antioxidants (ascorbate, glutathi- rat (444). Pneumolysin was also shown to induce cell damage
one) in the brain parenchyma and CSF and no evidence for in- in endothelial cells in vitro (267,268). Other bacterial toxins
duction of antioxidant defenses (290). Furthermore, most of with the ability to form pores are able to directly induce neu-
the beneficial effects of ROS scavengers in experimental men- ronal damage, such as the beta hemolysin/cytolysin of GBS
ingitis are compatible with a beneficial effect primarily at the (445). But not only the presence of bacteria in the CSF but
level of the cerebral vasculature (314) (see previous discus- also in the blood (bacteremia) has been suggested to influ-
sion). In the same model, marked loss of adenosine triphos- ence the development of neurologic damage during bacterial
phate in the brain parenchyma was positively correlated with meningitis (446).
the extent of neuronal injury (436). Oxidative alterations of
cerebral vasculature and subsequent reduction of CBF leading
to cerebral ischemia are plausible mechanisms for neuronal ACKNOWLEDGMENTS
damage during meningitis.
Theoretically, NO may act as a neuroprotective or neu- This work was supported in part by grant no. 138094 (to SLL)
rotoxic molecule in meningitis depending on its redox state; from the Swiss National Science Foundation.

References
1. Thigpen MC, Whitney CG, Messonnier NE, et al. Bacterial meningitis in 5. Nelson AL, Roche AM, Gould JM, et al. Capsule enhances pneumococ-
the United States, 19982007. N Engl J Med. 2011;364:20162025. cal colonization by limiting mucus-mediated clearance. Infect Immun.
2. Nicolosi A, Hauser WA, Musicco M, et al. Incidence and prognosis of brain 2007;75:8390.
abscess in a defined population: Olmsted County, Minnesota, 19351981. 6. Mook-Kanamori BB, Geldhoff M, van der Poll T, et al. Pathogenesis
Neuroepidemiology. 1991;10:122131. and pathophysiology of pneumococcal meningitis. Clin Microbiol Rev.
3. King N. Brain abscess. Handb Clin Neurol. 2010;96:6574. 2011;24:557591.
4. Lagu T, Rothberg MB, Shieh MS, et al. What is the best method for 7. Steinfort C, Wilson R, Mitchell T, et al. Effect of Streptococcus pneu-
estimating the burden of severe sepsis in the United States? J Crit Care. moniae on human respiratory epithelium in vitro. Infect Immun. 1989;57:
2012;27:414.e1414.e9. 20062013.

Scheld_Ch23.indd 356 2/21/14 5:41 PM


Chapter 23: Pathogenesis and Pathophysiology of Bacterial Infections 357

8. Hirst RA, Sikand KS, Rutman A, et al. Relative roles of pneumolysin and 38. Rowe HA, Griffiths NJ, Hill DJ, et al. Co-ordinate action of bacterial
hydrogen peroxide from Streptococcus pneumoniae in inhibition of epen- adhesins and human carcinoembryonic antigen receptors in enhanced cel-
dymal ciliary beat frequency. Infect Immun. 2000;68:15571562. lular invasion by capsulate serum resistant Neisseria meningitidis. Cell
9. Kadioglu A, Taylor S, Iannelli F, et al. Upper and lower respiratory Microbiol. 2007;9:154168.
tract infection by Streptococcus pneumoniae is affected by pneumoly- 39. St Geme JW III, Cutter D. Influence of pili, fibrils, and capsule on in vitro
sin deficiency and differences in capsule type. Infect Immun. 2002;70: adherence by Haemophilus influenzae type b. Mol Microbiol. 1996;21:2131.
28862890. 40. Weber A, Harris K, Lohrke S, et al. Inability to express fimbriae results in
10. Rao VK, Krasan GP, Hendrixson DR, et al. Molecular determinants of the impaired ability of Haemophilus influenzae b to colonize the nasopharynx.
pathogenesis of disease due to non-typable Haemophilus influenzae. FEMS Infect Immun. 1991;59:47244728.
Microbiol Rev. 1999;23:99129. 41. Cotter SE, Yeo HJ, Juehne T, et al. Architecture and adhesive activity of the
11. Merz AJ, So M. Interactions of pathogenic neisseriae with epithelial cell Haemophilus influenzae Hsf adhesin. J Bacteriol. 2005;187:46564664.
membranes. Annu Rev Cell Dev Biol. 2000;16:423457. 42. Hill DJ, Toleman MA, Evans DJ, et al. The variable P5 proteins of typeable
12. Wani JH, Gilbert JV, Plaut AG, et al. Identification, cloning, and sequenc- and non-typeable Haemophilus influenzae target human CEACAM1. Mol
ing of the immunoglobulin A1 protease gene of Streptococcus pneumoniae. Microbiol. 2001;39:850862.
Infect Immun. 1996;64:39673974. 43. Avadhanula V, Rodriguez CA, Ulett GC, et al. Nontypeable Haemophilus
13. Vitovski S, Read RC, Sayers JR. Invasive isolates of Neisseria meningitidis influenzae adheres to intercellular adhesion molecule 1 (ICAM-1) on respi-
possess enhanced immunoglobulin A1 protease activity compared to colo- ratory epithelial cells and upregulates ICAM-1 expression. Infect Immun.
nizing strains. Faseb J. 1999;13:331337. 2006;74:830838.
14. Callewaert L, Van Herreweghe JM, Vanderkelen L, et al. Guards of the 44. Posfay-Barbe KM, Wald ER. Listeriosis. Semin Fetal Neonatal Med.
great wall: bacterial lysozyme inhibitors. Trends Microbiol. 2012;20: 2009;14:228233.
501510. 45. Le Bouguenec C. Adhesins and invasins of pathogenic Escherichia coli. Int
15. Boneca IG, Dussurget O, Cabanes D, et al. A critical role for peptidoglycan J Med Microbiol. 2005;295:471478.
N-deacetylation in Listeria evasion from the host innate immune system. 46. Sokurenko EV, Chesnokova V, Dykhuizen DE, et al. Pathogenic adaptation
Proc Natl Acad Sci U S A. 2007;104:9971002. of Escherichia coli by natural variation of the FimH adhesin. Proc Natl
16. Davis KM, Weiser JN. Modifications to the peptidoglycan backbone help Acad Sci U S A. 1998;95:89228926.
bacteria to establish infection. Infect Immun. 2011;79:562570. 47. Antao EM, Wieler LH, Ewers C. Adhesive threads of extraintestinal patho-
17. Davis KM, Akinbi HT, Standish AJ, et al. Resistance to mucosal lyso- genic Escherichia coli. Gut Pathog. 2009;1:22.
zyme compensates for the fitness deficit of peptidoglycan modifications by 48. Bonacorsi S, Bingen E. Molecular epidemiology of Escherichia coli causing
Streptococcus pneumoniae. PLoS Pathog. 2008;4:e1000241. neonatal meningitis. Int J Med Microbiol. 2005;295:373381.
18. Bergmann S, Hammerschmidt S. Versatility of pneumococcal surface pro- 49. Kim KS, Itabashi H, Gemski P, et al. The K1 capsule is the critical deter-
teins. Microbiology. 2006;152:295303. minant in the development of Escherichia coli meningitis in the rat. J Clin
19. Pizarro-Cerda J, Cossart P. Bacterial adhesion and entry into host cells. Invest. 1992;90:897905.
Cell. 2006;124:715727. 50. Tazi A, Disson O, Bellais S, et al. The surface protein HvgA mediates group B
20. Proft T, Baker EN. Pili in Gram-negative and Gram-positive bacte- Streptococcus hypervirulence and meningeal tropism in neonates. J Exp Med.
riastructure, assembly and their role in disease. Cell Mol Life Sci. 2010;207:23132322.
2009;66:613635. 51. Maisey HC, Doran KS, Nizet V. Recent advances in understanding the
21. Weiser JN, Austrian R, Sreenivasan PK, et al. Phase variation in pneumo- molecular basis of group B Streptococcus virulence. Expert Rev Mol Med.
coccal opacity: relationship between colonial morphology and nasopharyn- 2008;10:e27.
geal colonization. Infect Immun. 1994;62:25822589. 52. Rinaudo CD, Rosini R, Galeotti CL, et al. Specific involvement of pilus type
22. Kim JO, Weiser JN. Association of intrastrain phase variation in quan- 2a in biofilm formation in group B Streptococcus. PLoS One. 2010;5:e9216.
tity of capsular polysaccharide and teichoic acid with the virulence of 53. Mengaud J, Ohayon H, Gounon P, et al. E-cadherin is the receptor for
Streptococcus pneumoniae. J Infect Dis. 1998;177:368377. internalin, a surface protein required for entry of L. monocytogenes into
23. Hammerschmidt S. Adherence molecules of pathogenic pneumococci. Curr epithelial cells. Cell. 1996;84:923932.
Opin Microbiol. 2006;9:1220. 54. Lecuit M. Understanding how Listeria monocytogenes targets and crosses
24. King SJ. Pneumococcal modification of host sugars: a major contributor to host barriers. Clin Microbiol Infect. 2005;11:430436.
colonization of the human airway? Mol Oral Microbiol. 2010;25:1524. 55. Pentecost M, Otto G, Theriot JA, et al. Listeria monocytogenes invades the
25. Brittan JL, Buckeridge TJ, Finn A, et al. Pneumococcal neuraminidase A: an epithelial junctions at sites of cell extrusion. PLoS Pathog. 2006;2:e3.
essential upper airway colonization factor for Streptococcus pneumoniae. 56. Nikitas G, Deschamps C, Disson O, et al. Transcytosis of Listeria mono-
Mol Oral Microbiol. 2012;27:270283. cytogenes across the intestinal barrier upon specific targeting of goblet cell
26. Parker D, Soong G, Planet P, et al. The NanA neuraminidase of accessible E-cadherin. J Exp Med. 2011;208:22632277.
Streptococcus pneumoniae is involved in biofilm formation. Infect Immun. 57. Lecuit M, Nelson DM, Smith SD, et al. Targeting and crossing of the
2009;77:37223730. human maternofetal barrier by Listeria monocytogenes: role of intern-
27. Shakhnovich EA, King SJ, Weiser JN. Neuraminidase expressed by alin interaction with trophoblast E-cadherin. Proc Natl Acad Sci U S A.
Streptococcus pneumoniae desialylates the lipopolysaccharide of Neisseria 2004;101:61526157.
meningitidis and Haemophilus influenzae: a paradigm for interbacte- 58. McCullers JA. Insights into the interaction between influenza virus and
rial competition among pathogens of the human respiratory tract. Infect pneumococcus. Clin Microbiol Rev. 2006;19:571582.
Immun. 2002;70:71617164. 59. Avadhanula V, Rodriguez CA, Devincenzo JP, et al. Respiratory viruses
28. Rosenow C, Ryan P, Weiser JN, et al. Contribution of novel choline-binding augment the adhesion of bacterial pathogens to respiratory epithelium in a
proteins to adherence, colonization and immunogenicity of Streptococcus viral species- and cell type-dependent manner. J Virol. 2006;80:16291636.
pneumoniae. Mol Microbiol. 1997;25:819829. 60. Greenwood BM. The epidemiology of acute bacterial meningitis in tropi-
29. Cundell DR, Gerard NP, Gerard C, et al. Streptococcus pneumoniae anchor cal Africa. Bacterial meningitis. In: Williams JD, Burnie J, eds. Bacterial
to activated human cells by the receptor for platelet-activating factor. Meningitis. London: Academic Press; 1987:6191.
Nature. 1995;377:435438. 61. Albiger B, Johansson L, Jonsson AB. Lipooligosaccharide-deficient
30. Grigg J. The platelet activating factor receptor: a new anti-infective target Neisseria meningitidis shows altered pilus-associated characteristics. Infect
in respiratory disease? Thorax. 2012;67:840841. Immun. 2003;71:155162.
31. Bonazzi M, Cossart P. Impenetrable barriers or entry portals? The role of 62. Conner SD, Schmid SL. Regulated portals of entry into the cell. Nature.
cell-cell adhesion during infection. J Cell Biol. 2011;195:349358. 2003;422:3744.
32. Anderton JM, Rajam G, Romero-Steiner S, et al. E-cadherin is a recep- 63. Veiga E, Guttman JA, Bonazzi M, et al. Invasive and adherent bacte-
tor for the common protein pneumococcal surface adhesin A (PsaA) of rial pathogens co-Opt host clathrin for infection. Cell Host Microbe.
Streptococcus pneumoniae. Microb Pathog. 2007;42:225236. 2007;2:340351.
33. Crain MJ, Waltman WD II, Turner JS, et al. Pneumococcal surface protein 64. Clarke TB, Francella N, Huegel A, et al. Invasive bacterial pathogens exploit
A (PspA) is serologically highly variable and is expressed by all clinically TLR-mediated downregulation of tight junction components to facilitate
important capsular serotypes of Streptococcus pneumoniae. Infect Immun. translocation across the epithelium. Cell Host Microbe. 2011;9:404414.
1990;58:32933299. 65. Zwijnenburg PJ, van der Poll T, Florquin S, et al. Experimental pneumo-
34. Zhang Q, Bernatoniene J, Bagrade L, et al. Serum and mucosal antibody coccal meningitis in mice: a model of intranasal infection. J Infect Dis.
responses to pneumococcal protein antigens in children: relationships with 2001;183:11431146.
carriage status. Eur J Immunol. 2006;36:4657. 66. Hirst RA, Kadioglu A, OCallaghan C, et al. The role of pneumoly-
35. Lebon A, Verkaik NJ, Labout JA, et al. Natural antibodies against several sin in pneumococcal pneumonia and meningitis. Clin Exp Immunol.
pneumococcal virulence proteins in children during the pre-pneumococcal- 2004;138:195201.
vaccine era: the generation R study. Infect Immun. 2011;79:16801687. 67. Attali C, Durmort C, Vernet T, et al. The interaction of Streptococcus
36. Deghmane AE, Giorgini D, Larribe M, et al. Down-regulation of pili and pneumoniae with plasmin mediates transmigration across endothelial and
capsule of Neisseria meningitidis upon contact with epithelial cells is medi- epithelial monolayers by intercellular junction cleavage. Infect Immun.
ated by CrgA regulatory protein. Mol Microbiol. 2002;43:15551564. 2008;76:53505356.
37. Gray-Owen SD. Neisserial Opa proteins: impact on colonization, dissemi- 68. Soriani M, Santi I, Taddei A, et al. Group B Streptococcus crosses human
nation and immunity. Scand J Infect Dis. 2003;35:614618. epithelial cells by a paracellular route. J Infect Dis. 2006;193:241250.

Scheld_Ch23.indd 357 2/21/14 5:41 PM


358 Part III: Bacterial and Mycoplasmal Infections

69. Mostov KE. Transepithelial transport of immunoglobulins. Annu Rev 98. Agarwal V, Hammerschmidt S, Malm S, et al. Enolase of Streptococcus
Immunol. 1994;12:6384. pneumoniae binds human complement inhibitor C4b-binding pro-
70. Zhang JR, Mostov KE, Lamm ME, et al. The polymeric immunoglobulin tein and contributes to complement evasion. J Immunol. 2012;189:
receptor translocates pneumococci across human nasopharyngeal epithelial 35753584.
cells. Cell. 2000;102:827837. 99. Santi I, Scarselli M, Mariani M, et al. BibA: a novel immunogenic bacterial
71. Elm C, Braathen R, Bergmann S, et al. Ectodomains 3 and 4 of human poly- adhesin contributing to group B Streptococcus survival in human blood.
meric immunoglobulin receptor (hpIgR) mediate invasion of Streptococcus Mol Microbiol. 2007;63:754767.
pneumoniae into the epithelium. J Biol Chem. 2004;279:62966304. 100. Zipfel PF, Skerka C, Hellwage J, et al. Factor H family proteins: on
72. Gray-Owen SD, Blumberg RS. CEACAM1: contact-dependent control of complement, microbes and human diseases. Biochem Soc Trans. 2002;30:
immunity. Nat Rev Immunol. 2006;6:433446. 971978.
73. Sadarangani M, Pollard AJ, Gray-Owen SD. Opa proteins and CEACAMs: 101. Maruvada R, Prasadarao NV, Rubens CE. Acquisition of factor H by a
pathways of immune engagement for pathogenic Neisseria. FEMS novel surface protein on group B Streptococcus promotes complement
Microbiol Rev. 2011;35:498514. degradation. Faseb J. 2009;23:39673977.
74. Griffiths NJ, Bradley CJ, Heyderman RS, et al. IFN-gamma amplifies 102. Lewis LA, Carter M, Ram S. The relative roles of factor H binding pro-
NFkappaB-dependent Neisseria meningitidis invasion of epithelial cells tein, neisserial surface protein A, and lipooligosaccharide sialylation in
via specific upregulation of CEA-related cell adhesion molecule 1. Cell regulation of the alternative pathway of complement on meningococci.
Microbiol. 2007;9:29682983. J Immunol. 2012;188:50635072.
75. Disson O, Grayo S, Huillet E, et al. Conjugated action of two species- 103. Gossger N, Snape MD, Yu LM, et al. Immunogenicity and tolerability
specific invasion proteins for fetoplacental listeriosis. Nature. 2008;455: of recombinant serogroup B meningococcal vaccine administered with or
11141118. without routine infant vaccinations according to different immunization
76. Fagan RP, Lambert MA, Smith SG. The hek outer membrane protein of schedules: a randomized controlled trial. JAMA. 2012;307:573582.
Escherichia coli strain RS218 binds to proteoglycan and utilizes a single 104. Jiang HQ, Hoiseth SK, Harris SL, et al. Broad vaccine coverage pre-
extracellular loop for adherence, invasion, and autoaggregation. Infect dicted for a bivalent recombinant factor H binding protein based vac-
Immun. 2008;76:11351142. cine to prevent serogroup B meningococcal disease. Vaccine. 2010;28:
77. Meier C, Oelschlaeger TA, Merkert H, et al. Ability of Escherichia coli iso- 60866093.
lates that cause meningitis in newborns to invade epithelial and endothelial 105. Jarva H, Jokiranta TS, Wurzner R, et al. Complement resistance mecha-
cells. Infect Immun. 1996;64:23912399. nisms of streptococci. Mol Immunol. 2003;40:95107.
78. Carbonnelle E, Hill DJ, Morand P, et al. Meningococcal interactions with 106. Singh B, Su YC, Riesbeck K. Vitronectin in bacterial pathogenesis: a host
the host. Vaccine. 2009;27(suppl 2):B78B89. protein used in complement escape and cellular invasion. Mol Microbiol.
79. Virkola R, Brummer M, Rauvala H, et al. Interaction of fimbriae of 2010;78:545560.
Haemophilus influenzae type B with heparin-binding extracellular matrix 107. Kolb-Maurer A, Kurzai O, Goebel W, et al. The role of human dendritic
proteins. Infect Immun. 2000;68:56965701. cells in meningococcal and listerial meningitis. Int J Med Microbiol.
80. Kostyukova NN, Volkova MO, Ivanova VV, et al. A study of pathogenic 2003;293:241249.
factors of Streptococcus pneumoniae strains causing meningitis. FEMS 108. Peppoloni S, Ricci S, Orsi CF, et al. The encapsulated strain TIGR4 of
Immunol Med Microbiol. 1995;10:133137. Streptococcus pneumoniae is phagocytosed but is resistant to intracellular
81. Plow EF, Herren T, Redlitz A, et al. The cell biology of the plasminogen killing by mouse microglia. Microbes Infect. 2010;12:9901001.
system. Faseb J. 1995;9:939945. 109. Stavru F, Archambaud C, Cossart P. Cell biology and immunology
82. Knaust A, Weber MV, Hammerschmidt S, et al. Cytosolic proteins con- of Listeria monocytogenes infections: novel insights. Immunol Rev.
tribute to surface plasminogen recruitment of Neisseria meningitidis. 2011;240:160184.
J Bacteriol. 2007;189:32463255. 110. Tala A, Monaco C, Nagorska K, et al. Glutamate utilization promotes
83. Virkola R, Lahteenmaki K, Eberhard T, et al. Interaction of Haemophilus meningococcal survival in vivo through avoidance of the neutrophil oxi-
influenzae with the mammalian extracellular matrix. J Infect Dis. dative burst. Mol Microbiol. 2011;81:13301342.
1996;173:11371147. 111. Nandoskar M, Ferrante A, Bates EJ, et al. Inhibition of human monocyte
84. Bergmann S, Rohde M, Preissner KT, et al. The nine residue plasminogen- respiratory burst, degranulation, phospholipid methylation and bacteri-
binding motif of the pneumococcal enolase is the major cofactor of plas- cidal activity by pneumolysin. Immunology. 1986;59:515520.
min-mediated degradation of extracellular matrix, dissolution of fibrin and 112. Moxon ER, Murphy PA. Haemophilus influenzae bacteremia and men-
transmigration. Thromb Haemost. 2005;94:304311. ingitis resulting from survival of a single organism. Proc Natl Acad Sci
85. Steukers L, Glorieux S, Vandekerckhove AP, et al. Diverse microbial U S A. 1978;75:15341536.
interactions with the basement membrane barrier. Trends Microbiol. 113. Redzic Z. Molecular biology of the blood-brain and the blood-cerebro-
2012;20:147155. spinal fluid barriers: similarities and differences. Fluids Barriers CNS.
86. Pron B, Boumaila C, Jaubert F, et al. Dendritic cells are early cellular tar- 2011;8:3.
gets of Listeria monocytogenes after intestinal delivery and are involved in 114. Kim KS. Strategy of Escherichia coli for crossing the blood-brain barrier.
bacterial spread in the host. Cell Microbiol. 2001;3:331340. J Infect Dis. 2002;186:S220S224.
87. Walport MJ. Complement. First of two parts. N Engl J Med. 115. Moxon ER, Ostrow PT. Haemophilus influenzae meningitis in infant
2001;344:10581066. rats: role of bacteremia in pathogenesis of age-dependent inflammatory
88. Overturf GD. Indications for the immunological evaluation of patients responses in cerebrospinal fluid. J Infect Dis. 1977;135:303307.
with meningitis. Clin Infect Dis. 2003;36:189194. 116. Kim KS. Mechanisms of microbial traversal of the blood-brain barrier.
89. Dietzman DE, Fischer GW, Schoenknecht FD. Neonatal Escherichia coli Nat Rev Microbiol. 2008;6:625634.
septicemiabacterial counts in blood. J Pediatr. 1974;85:128130. 117. Orihuela CJ, Mahdavi J, Thornton J, et al. Laminin receptor initiates
90. Adriani KS, Brouwer MC, Geldhoff M, et al. Common polymorphisms in bacterial contact with the blood brain barrier in experimental meningitis
the complement system and susceptibility to bacterial meningitis. J Infect. models. J Clin Invest. 2009;119:16381646.
2012;66:255262. 118. Kim BY, Kang J, Kim KS. Invasion processes of pathogenic Escherichia
91. Brouwer MC, de Gans J, Heckenberg SG, et al. Host genetic susceptibil- coli. Int J Med Microbiol. 2005;295:463470.
ity to pneumococcal and meningococcal disease: a systematic review and 119. Coureuil M, Join-Lambert O, Lecuyer H, et al. Mechanism of meningeal
meta-analysis. Lancet Infect Dis. 2009;9:3144. invasion by Neisseria meningitidis. Virulence. 2012;3:164172.
92. Vogel U, Hammerschmidt S, Frosch M. Sialic acids of both the capsule and 120. Maisey HC, Hensler M, Nizet V, et al. Group B streptococcal pilus pro-
the sialylated lipooligosaccharide of Neisseria meningitis serogroup B are teins contribute to adherence to and invasion of brain microvascular
prerequisites for virulence of meningococci in the infant rat. Med Microbiol endothelial cells. J Bacteriol. 2007;189:14641467.
Immunol (Berl). 1996;185:8187. 121. Tenenbaum T, Bloier C, Adam R, et al. Adherence to and invasion of
93. Hyams C, Camberlein E, Cohen JM, et al. The Streptococcus pneumoniae human brain microvascular endothelial cells are promoted by fibrino-
capsule inhibits complement activity and neutrophil phagocytosis by mul- gen-binding protein FbsA of Streptococcus agalactiae. Infect Immun.
tiple mechanisms. Infect Immun. 2010;78:704715. 2005;73:44044409.
94. Kilian M, Reinholdt J, Lomholt H, et al. Biological significance of IgA1 122. Banerjee A, Kim BJ, Carmona EM, et al. Bacterial Pili exploit integrin ma-
proteases in bacterial colonization and pathogenesis: critical evaluation of chinery to promote immune activation and efficient blood-brain barrier
experimental evidence. Apmis. 1996;104:321338. penetration. Nat Commun. 2011;2:462.
95. Schuchat A, Robinson K, Wenger JD, et al. Bacterial meningitis in the 123. Drevets DA, Leenen PJ, Greenfield RA. Invasion of the central nervous
United States in 1995. Active Surveillance Team. N Engl J Med. 1997;337: system by intracellular bacteria. Clin Microbiol Rev. 2004;17:323347.
970976. 124. Nassif X, Bourdoulous S, Eugene E, et al. How do extracellular pathogens
96. Li J, Glover DT, Szalai AJ, et al. PspA and PspC minimize immune cross the blood-brain barrier? Trends Microbiol. 2002;10:227232.
adherence and transfer of pneumococci from erythrocytes to macro- 125. Uchiyama S, Carlin AF, Khosravi A, et al. The surface-anchored NanA
phages through their effects on complement activation. Infect Immun. protein promotes pneumococcal brain endothelial cell invasion. J Exp
2007;75:58775885. Med. 2009;206:18451852.
97. Rooijakkers SH, van Strijp JA. Bacterial complement evasion. Mol 126. Honda Z, Ishii S, Shimizu T. Platelet-activating factor receptor. J Biochem.
Immunol. 2007;44:2332. 2002;131:773779.

Scheld_Ch23.indd 358 2/21/14 5:41 PM


Chapter 23: Pathogenesis and Pathophysiology of Bacterial Infections 359

127. Radin JN, Orihuela CJ, Murti G, et al. Beta-arrestin 1 participates in 156. Kristian SA, Birkenstock TA, Sauder U, et al. Biofilm formation induces
platelet-activating factor receptor-mediated endocytosis of Streptococcus C3a release and protects Staphylococcus epidermidis from IgG and com-
pneumoniae. Infect Immun. 2005;73:78277835. plement deposition and from neutrophil-dependent killing. J Infect Dis.
128. Reddy MA, Wass CA, Kim KS, et al. Involvement of focal adhesion kinase 2008;197:10281035.
in Escherichia coli invasion of human brain microvascular endothelial 157. Xu KD, McFeters GA, Stewart PS. Biofilm resistance to antimicrobial
cells. Infect Immun. 2000;68:64236430. agents. Microbiology. 2000;146(pt 3):547549.
129. Kim KJ, Elliott SJ, Di Cello F, et al. The K1 capsule modulates traffick- 158. Bayston R, Ullas G, Ashraf W. Action of linezolid or vancomycin on
ing of E. coli-containing vacuoles and enhances intracellular bacterial biofilms in ventriculoperitoneal shunts in vitro. Antimicrob Agents
survival in human brain microvascular endothelial cells. Cell Microbiol. Chemother. 2012;56:28422845.
2003;5:245252. 159. Reefhuis J, Honein MA, Whitney CG, et al. Risk of bacterial meningitis in
130. Unkmeir A, Latsch K, Dietrich G, et al. Fibronectin mediates Opc- children with cochlear implants. N Engl J Med. 2003;349:435445.
dependent internalization of Neisseria meningitidis in human brain 160. Lalwani AK, Cohen NL. Does meningitis after cochlear implantation
microvascular endothelial cells. Mol Microbiol. 2002;46:933946. remain a concern in 2011? Otol Neurotol. 2012;33:9395.
131. Disson O, Lecuit M. Targeting of the central nervous system by Listeria 161. Wei BP, Shepherd RK, Robins-Browne RM, et al. Threshold shift:
monocytogenes. Virulence. 2012;3:213221. effects of cochlear implantation on the risk of pneumococcal meningitis.
132. Drevets DA, Dillon MJ, Schawang JS, et al. The Ly-6Chigh monocyte Otolaryngol Head Neck Surg. 2007;136:589596.
subpopulation transports Listeria monocytogenes into the brain during 162. Cohen NL, Hirsch BE. Current status of bacterial meningitis after cochlear
systemic infection of mice. J Immunol. 2004;172:44184424. implantation. Otol Neurotol. 2010;31:13251328.
133. Drevets DA, Jelinek TA, Freitag NE. Listeria monocytogenes-infected 163. Ransohoff RM, Brown MA. Innate immunity in the central nervous sys-
phagocytes can initiate central nervous system infection in mice. Infect tem. J Clin Invest. 2012;122:11641171.
Immun. 2001;69:13441350. 164. Pashenkov M, Teleshova N, Kouwenhoven M, et al. Recruitment of
134. Kastenbauer S, Pfister HW. Pneumococcal meningitis in adults: spectrum dendritic cells to the cerebrospinal fluid in bacterial neuroinfections.
of complications and prognostic factors in a series of 87 cases. Brain. J Neuroimmunol. 2002;122:106116.
2003;126:10151025. 165. Engelhardt B, Coisne C. Fluids and barriers of the CNS establish immune
135. van de Beek D, de Gans J, Spanjaard L, et al. Clinical features and privilege by confining immune surveillance to a two-walled castle moat
prognostic factors in adults with bacterial meningitis. N Engl J Med. surrounding the CNS castle. Fluids Barriers CNS. 2011;8:4.
2004;351:18491859. 166. Zwahlen A, Nydegger UE, Vaudaux P, et al. Complement-mediated
136. Marra A, Brigham D. Streptococcus pneumoniae causes experimental opsonic activity in normal and infected human cerebrospinal fluid: early
meningitis following intranasal and otitis media infections via a nonhe- response during bacterial meningitis. J Infect Dis. 1982;145:635646.
matogenous route. Infect Immun. 2001;69:73187325. 167. Stahel PF, Frei K, Fontana A, et al. Evidence for intrathecal synthesis of
137. van Ginkel FW, McGhee JR, Watt JM, et al. Pneumococcal carriage re- alternative pathway complement activation proteins in experimental men-
sults in ganglioside-mediated olfactory tissue infection. Proc Natl Acad ingitis. Am J Pathol. 1997;151:897904.
Sci U S A. 2003;100:1436314367. 168. Small PM, Tuber MG, Hackbarth CJ, et al. Influence of body tempera-
138. Sjolinder H, Jonsson AB. Olfactory nervea novel invasion route of ture on bacterial growth rates in experimental pneumococcal meningitis
Neisseria meningitidis to reach the meninges. PLoS One. 2010;5:e14034. in rabbits. Infect Immun. 1986;52:484487.
139. Vinchon M, Lemaitre MP, Vallee L, et al. Late shunt infection: inci- 169. Ernst JD, Decazes JM, Sande MA. Experimental pneumococcal meningi-
dence, pathogenesis, and therapeutic implications. Neuropediatrics. tis: role of leukocytes in pathogenesis. Infect Immun. 1983;41:275279.
2002;33:169173. 170. Tuomanen EI, Saukkonen K, Sande S, et al. Reduction of inflammation,
140. Odio C, McCracken GH Jr, Nelson JD. CSF shunt infections in pediatrics. tissue damage, and mortality in bacterial meningitis in rabbits treated
A seven-year experience. Am J Dis Child. 1984;138:11031108. with monoclonal antibodies against adhesion-promoting receptors of leu-
141. Reichert MC, Medeiros EA, Ferraz FA. Hospital-acquired meningitis in kocytes. J Exp Med. 1989;170:959969.
patients undergoing craniotomy: incidence, evolution, and risk factors. 171. Tuomanen E, Hengstler B, Zak O, et al. The role of complement in inflam-
Am J Infect Control. 2002;30:158164. mation during experimental pneumococcal meningitis. Microb Pathog.
142. McGirt MJ, Zaas A, Fuchs HE, et al. Risk factors for pediatric ventricu- 1986;1:1532.
loperitoneal shunt infection and predictors of infectious pathogens. Clin 172. Rupprecht TA, Angele B, Klein M, et al. Complement C1q and C3 are
Infect Dis. 2003;36:858862. critical for the innate immune response to Streptococcus pneumoniae in
143. Reddy GK, Bollam P, Caldito G. Ventriculoperitoneal shunt surgery and the central nervous system. J Immunol. 2007;178:18611869.
the risk of shunt infection in patients with hydrocephalus: long-term sin- 173. Goonetilleke UR, Scarborough M, Ward SA, et al. Death is associated
gle institution experience. World Neurosurg. 2012;78:155163. with complement C3 depletion in cerebrospinal fluid of patients with
144. Kockro RA, Hampl JA, Jansen B, et al. Use of scanning electron micros- pneumococcal meningitis. MBio. 2012;3
copy to investigate the prophylactic efficacy of rifampin-impregnated CSF 174. Oggioni MR, Trappetti C, Kadioglu A, et al. Switch from planktonic to
shunt catheters. J Med Microbiol. 2000;49:441450. sessile life: a major event in pneumococcal pathogenesis. Mol Microbiol.
145. Schoenbaum SC, Gardner P, Shillito J. Infections of cerebrospinal fluid 2006;61:11961210.
shunts: epidemiology, clinical manifestations, and therapy. J Infect Dis. 175. Orihuela CJ, Radin JN, Sublett JE, et al. Microarray analysis of pneu-
1975;131:543552. mococcal gene expression during invasive disease. Infect Immun.
146. Filka J, Huttova M, Tuharsky J, et al. Nosocomial meningitis in chil- 2004;72:55825596.
dren after ventriculoperitoneal shunt insertion. Acta Paediatr. 1999;88: 176. Molzen TE, Burghout P, Bootsma HJ, et al. Genome-wide identification of
576578. Streptococcus pneumoniae genes essential for bacterial replication during
147. Del Bigio MR. Biological reactions to cerebrospinal fluid shunt devices: experimental meningitis. Infect Immun. 2011;79:288297.
a review of the cellular pathology. Neurosurgery. 1998;42:319325; 177. Hanamsagar R, Hanke ML, Kielian T. Toll-like receptor (TLR) and
discussion 325326. inflammasome actions in the central nervous system. Trends Immunol.
148. Hartford O, OBrien L, Schofield K, et al. The Fbe (SdrG) protein of 2012;33:333342.
Staphylococcus epidermidis HB promotes bacterial adherence to fibrino- 178. Koedel U. Toll-like receptors in bacterial meningitis. Curr Top Microbiol
gen. Microbiology. 2001;147:25452552. Immunol. 2009;336:1540.
149. Williams RJ, Henderson B, Sharp LJ, et al. Identification of a fibronectin- 179. Hoegen T, Tremel N, Klein M, et al. The NLRP3 inflammasome contributes
binding protein from Staphylococcus epidermidis. Infect Immun. 2002; to brain injury in pneumococcal meningitis and is activated through ATP-
70:68056810. dependent lysosomal cathepsin B release. J Immunol. 2011;187:54405451.
150. Li DQ, Lundberg F, Ljungh A. Characterization of vitronectin-binding 180. Geldhoff M, Mook-Kanamori BB, Brouwer MC, et al. Genetic variation
proteins of Staphylococcus epidermidis. Curr Microbiol. 2001;42: in inflammasome genes is associated with outcome in bacterial meningitis.
361367. Immunogenetics. 2013;65:916.
151. Thurlow LR, Hanke ML, Fritz T, et al. Staphylococcus aureus biofilms 181. Sanders MS, van Well GT, Ouburg S, et al. Toll-like receptor 9 polymor-
prevent macrophage phagocytosis and attenuate inflammation in vivo. phisms are associated with severity variables in a cohort of meningococcal
J Immunol. 2011;186:65856596. meningitis survivors. BMC Infect Dis. 2012;12:112.
152. Peters G, Locci R, Pulverer G. Adherence and growth of coagulase- 182. van Well GT, Sanders MS, Ouburg S, et al. Polymorphisms in Toll-like
negative staphylococci on surfaces of intravenous catheters. J Infect Dis. receptors 2, 4, and 9 are highly associated with hearing loss in survivors
1982;146:479482. of bacterial meningitis. PLoS One. 2012;7:e35837.
153. Costerton JW, Stewart PS, Greenberg EP. Bacterial biofilms: a common 183. Burroughs M, Cabellos C, Prasad S, et al. Bacterial components and the
cause of persistent infections. Science. 1999;284:13181322. pathophysiology of injury to the blood-brain barrier: does cell wall add
154. Snowden JN, Beaver M, Smeltzer MS, et al. Biofilm-infected intracerebro- to the effects of endotoxin in gram-negative meningitis? J Infect Dis.
ventricular shunts elicit inflammation within the central nervous system. 1992;165(suppl 1):S82S85.
Infect Immun. 2012;80:32063214. 184. Mustafa MM, Ramilo O, Saez-Llorens X, et al. Cerebrospinal fluid pros-
155. Vuong C, Voyich JM, Fischer ER, et al. Polysaccharide intercellular adhe- taglandins, interleukin 1 beta, and tumor necrosis factor in bacterial
sin (PIA) protects Staphylococcus epidermidis against major components meningitis. Clinical and laboratory correlations in placebo-treated and
of the human innate immune system. Cell Microbiol. 2004;6:269275. dexamethasone-treated patients. Am J Dis Child. 1990;144:883887.

Scheld_Ch23.indd 359 2/21/14 5:41 PM


360 Part III: Bacterial and Mycoplasmal Infections

185. Tuber MG, Moser B. Cytokines and chemokines in meningeal inflam- 213. Waage A, Halstensen A, Shalaby R. Local production of tumor necrosis
mation: biology and clinical implications. Clin Infect Dis. 1999;28:111; factor, interleukin-l, and interleukin-6 in meningococcal meningitis. J Exp
quiz 12. Med. 1998;170:18591864.
186. Kastenbauer S, Angele B, Sporer B, et al. Patterns of protein expression 214. Helminen M, Vesikari T. Interleukin-1 production in bacterial meningitis.
in infectious meningitis: a cerebrospinal fluid protein array analysis. Scand J Infect Dis. 1990;22:105108.
J Neuroimmunol. 2005;164:134139. 215. Rusconi F, Parizzi F, Garlaschi L, et al. Interleukin 6 activity in infants
187. Zwijnenburg PJ, van der Poll T, Roord JJ, et al. Chemotactic fac- and children with bacterial meningitis. The Collaborative Study on
tors in cerebrospinal fluid during bacterial meningitis. Infect Immun. Meningitis. Pediatr Infect Dis J. 1991;10:117121.
2006;74:14451451. 216. Hirano T, Akira S, Taga T, et al. Biological and clinical aspects of interleu-
188. Klein M, Paul R, Angele B, et al. Protein expression pattern in experimen- kin-6. Immunol Today. 1990;11:443445.
tal pneumococcal meningitis. Microbes Infect. 2006;8:974983. 217. Cohen MC, Cohen S. Cytokine function, a study in biologic diversity.
189. Mogensen TH, Paludan SR, Kilian M, et al. Live Streptococcus pneu- Am J Clin Pathol. 1996;105:589598.
moniae, Haemophilus influenzae, and Neisseria meningitidis activate the 218. Paul R, Koedel U, Winkler F, et al. Lack of IL-6 augments inflamma-
inflammatory response through Toll-like receptors 2, 4, and 9 in species- tory response but decreases vascular permeability in bacterial meningitis.
specific patterns. J Leukoc Biol. 2006;80:267277. Brain. 2003;126:18731882.
190. Fowler MI, Weller RO, Heckels JE, et al. Different meningitis-causing 219. Zwijnenburg PJ, de Bie HM, Roord JJ, et al. Chemotactic activity of
bacteria induce distinct inflammatory responses on interaction with cells CXCL5 in cerebrospinal fluid of children with bacterial meningitis.
of the human meninges. Cell Microbiol. 2004;6:555567. J Neuroimmunol. 2003;145:148153.
191. Tietze K, Dalpke A, Morath S, et al. Differences in innate immune 220. Spanaus KS, Nadal D, Pfister HW, et al. C-X-C and C-C chemokines are
responses upon stimulation with gram-positive and gram-negative bacteria. expressed in the cerebrospinal fluid in bacterial meningitis and mediate
J Periodontal Res. 2006;41:447454. chemotactic activity on peripheral blood-derived polymorphonuclear and
192. Diab A, Zhu J, Linquist L, et al. Haemophilus influenzae and Streptococcus mononuclear cells in vitro. J Immunol. 1997;158:19561964.
pneumoniae induce different intracerebral mRNA cytokine patterns dur- 221. Paris MM, Hickey SM, Trujillo M, et al. The effect of interleukin-10 on
ing the course of experimental bacterial meningitis. Clin Exp Immunol. meningeal inflammation in experimental bacterial meningitis. J Infect Dis.
1997;109:233241. 1997;176:12391246.
193. Kim YS, Honkaniemi J, Sharp FR, et al. Expression of proinflammatory 222. Koedel U, Bernatowicz A, Frei K, et al. Systemically (but not intrathecally)
cytokines tumor necrosis factor-alpha and interleukin-1beta in the brain administered IL-10 attenuates pathophysiologic alterations in experimen-
during experimental group B streptococcal meningitis. Brain Res Mol tal pneumococcal meningitis. J Immunol. 1996;157:51855191.
Brain Res. 2004;128:95102. 223. Zwijnenburg PJG, van der Poll T, Florquin S, et al. Interleukin-10 nega-
194. Arvin B, Neville LF, Barone FC, et al. The role of inflammation and cyto- tively regulates local cytokine and chemokine production but does not
kines in brain injury. Neurosci Biobehav Rev. 1996;20:445452. influence antibacterial host defense during murine pneumococcal menin-
195. Tunkel AR, Wispelwey B, Scheld WM. Bacterial meningitis: recent ad- gitis. Infect Immun. 2003;71:22762279.
vances in pathophysiology and treatment. Ann Intern Med. 1990;112: 224. Mittal R, Gonzalez-Gomez I, Panigrahy A, et al. IL-10 administration
610623. reduces PGE-2 levels and promotes CR3-mediated clearance of Escherichia
196. Mertsola J. Cytokines in the pathogenesis of bacterial meningitis. Trans R coli K1 by phagocytes in meningitis. J Exp Med. 2010;207:13071319.
Soc Trop Med Hyg. 1991;85(suppl 1):1718. 225. Pfister HW, Frei K, Ottnad B, et al. Transforming growth factor beta 2
197. Mustafa MM, Ramilo O, Olsen KD, et al. Tumor necrosis factor in me- inhibits cerebrovascular changes and brain edema formation in the tumor
diating experimental Haemophilus influenzae type B meningitis. J Clin necrosis factor alpha-independent early phase of experimental pneumo-
Invest. 1989;84:12531259. coccal meningitis. J Exp Med. 1992;176:265268.
198. Leib SL, Clements JM, Lindberg RL, et al. Inhibition of matrix metallo- 226. Malipiero U, Koedel U, Pfister W, et al. Bacterial meningitis: the role of
proteinases and tumour necrosis factor alpha converting enzyme as adju- transforming growth factor-beta in innate immunity and secondary brain
vant therapy in pneumococcal meningitis. Brain. 2001;124:17341742. damage. Neurodegener Dis. 2007;4:4350.
199. Barichello T, Pereira JS, Savi GD, et al. A kinetic study of the cytokine/che- 227. Rosenberg GA. Matrix metalloproteinases in neuroinflammation. Glia.
mokines levels and disruption of blood-brain barrier in infant rats after 2002;39:279291.
pneumococcal meningitis. J Neuroimmunol. 2011;233:1217. 228. Leppert D, Lindberg RL, Kappos L, et al. Matrix metalloproteinases: mul-
200. Koedel U, Bayerlein I, Paul R, et al. Pharmacologic interference with NF- tifunctional effectors of inflammation in multiple sclerosis and bacterial
kappaB activation attenuates central nervous system complications in meningitis. Brain Res Rev. 2001;36:249257.
experimental pneumococcal meningitis. J Infect Dis. 2000;182:14371445. 229. Kieseier BC, Paul R, Koedel U, et al. Differential expression of matrix
201. Wellmer A, Gerber J, Ragheb J, et al. Effect of deficiency of tumor necrosis metalloproteinases in bacterial meningitis. Brain. 1999;122:15791587.
factor alpha or both of its receptors on Streptococcus pneumoniae central 230. Paul R, Lorenzl S, Koedel U, et al. Matrix metalloproteinases contribute
nervous system infection and peritonitis. Infect Immun. 2001;69:68816886. to the blood-brain barrier disruption during bacterial meningitis. Ann
202. Tureen J. Effect of recombinant human tumor necrosis factor-alpha on Neurol. 1998;44:592600.
cerebral oxygen uptake, cerebrospinal fluid lactate, and cerebral blood 231. Green JA, Thi Hong Chau T, Farrar JJ, et al. CNS infection, CSF ma-
flow in the rabbit: role of nitric oxide. J Clin Invest. 1995;95:10861091. trix metalloproteinase concentrations, and clinical/laboratory features.
203. Rosenberg GA, Estrada EY, Dencoff JE, et al. Tumor necrosis factor-al- Neurology. 2011;76:577579.
pha-induced gelatinase B causes delayed opening of the blood-brain bar- 232. Tsai HC, Shi MH, Lee SS, et al. Expression of matrix metalloproteinases
rier: an expanded therapeutic window. Brain Res. 1995;703:151155. and their tissue inhibitors in the serum and cerebrospinal fluid of patients
204. Andersson PB, Perry VH, Gordon S. Intracerebral injection of proinflam- with meningitis. Clin Microbiol Infect. 2011;17:780784.
matory cytokines or leukocyte chemotaxins induces minimal myelomono- 233. Lindberg RL, Sorsa T, Tervahartiala T, et al. Gelatinase B [matrix metal-
cytic cell recruitment to the parenchyma of the central nervous system. loproteinase (MMP)-9] and collagenases (MMP-8/-13) are upregulated
J Exp Med. 1992;176:255259. in cerebrospinal fluid during aseptic and bacterial meningitis in children.
205. Liu T, Clark RK, McDonnell PC, et al. Tumor necrosis factor-alpha Neuropathol Appl Neurobiol. 2006;32:304317.
expression in ischemic neurons. Stroke. 1994;25:14811488. 234. Sulik A, Chyczewski L. Immunohistochemical analysis of MMP-9,
206. Glabinski A, Krajewski S, Rafalowska J. Tumor necrosis factor-alpha MMP-2 and TIMP-1, TIMP-2 expression in the central nervous system
induced pathology in the rat brain: characterization of stereotaxic injec- following infection with viral and bacterial meningitis. Folia Histochem
tion model. Folia Neuropathol. 1998;36:5262. Cytobiol. 2008;46:437442.
207. Candelario-Jalil E, Taheri S, Yang Y, et al. Cyclooxygenase inhibi- 235. Leppert D, Leib SL, Grygar C, et al. Matrix metalloproteinase (MMP)-8
tion limits blood-brain barrier disruption following intracerebral injec- and MMP-9 in cerebrospinal fluid during bacterial meningitis: association
tion of tumor necrosis factor-alpha in the rat. J Pharmacol Exp Ther. with blood-brain barrier damage and neurological sequelae. Clin Infect
2007;323:488498. Dis. 2000;31:8084.
208. Friedland IR, Jafari H, Ehrett S, et al. Comparison of endotoxin release 236. Leib SL, Leppert D, Clements J, et al. Matrix metalloproteinases contrib-
by different antimicrobial agents and the effect on inflammation in experi- ute to brain damage in experimental pneumococcal meningitis. Infection
mental Escherichia coli meningitis. J Infect Dis. 1993;168:657662. and Immunity. 2000;68:615620.
209. Lutsar I, Friedland IR, Jafri HS, et al. Factors influencing the anti-inflam- 237. Sellner J, Leib SL. In bacterial meningitis cortical brain damage is associ-
matory effect of dexamethasone therapy in experimental pneumococcal ated with changes in parenchymal MMP-9/TIMP-1 ratio and increased
meningitis. J Antimicrob Chemother. 2003;52:651655. collagen type IV degradation. Neurobiol Dis. 2006;21:647656.
210. Costa A, Gupta R, Signorino G, et al. Activation of the NLRP3 inflamma- 238. Meli DN, Loeffler JM, Baumann P, et al. In pneumococcal meningitis
some by group B streptococci. J Immunol. 2012;188:19531960. a novel water-soluble inhibitor of matrix metalloproteinases and TNF-
211. Opitz B, Puschel A, Schmeck B, et al. Nucleotide-binding oligomeriza- alpha converting enzyme attenuates seizures and injury of the cerebral
tion domain proteins are innate immune receptors for internalized cortex. J Neuroimmunol. 2004;151:611.
Streptococcus pneumoniae. J Biol Chem. 2004;279:3642636432. 239. Tunkel AR, Scheld WM. Pathogenesis and pathophysiology of bacterial
212. Mitchell AJ, Yau B, McQuillan JA, et al. Inflammasome-dependent IFN- meningitis. Clin Microbiol Rev. 1993;6:118136.
gamma drives pathogenesis in Streptococcus pneumoniae meningitis. 240. Korthuis RJ, Anderson DC, Granger DN. Role of neutrophil-endothelial
J Immunol. 2012;189:49704980. cell adhesion in inflammatory disorders. J Crit Care. 1994;9:4771.

Scheld_Ch23.indd 360 2/21/14 5:41 PM


Chapter 23: Pathogenesis and Pathophysiology of Bacterial Infections 361

241. Tuber MG, Sande MA. Pathogenesis of bacterial meningitis: contribu- 269. Constantin D, AlaAldeent D, Murphy S. Transcriptional activation of
tions by experimental models in rabbits. Infection. 1984;12(suppl 1): nitric oxide synthase-2, and NO-induced cell death, in mouse cerebro-
S3S10. vascular endothelium exposed to Neisseria meningitidis. J Neurochem.
242. Springer TA. Traffic signals for lymphocyte recirculation and leukocyte 2002;81:270276.
emigration: the multistep paradigm. Cell. 1994;76:301314. 270. Hupp S, Heimeroth V, Wippel C, et al. Astrocytic tissue remodeling by the
243. Ley K, Laudanna C, Cybulsky MI, et al. Getting to the site of inflam- meningitis neurotoxin pneumolysin facilitates pathogen tissue penetration
mation: the leukocyte adhesion cascade updated. Nat Rev Immunol. and produces interstitial brain edema. Glia. 2012;60:137146.
2007;7:678689. 271. Daneman R. The blood-brain barrier in health and disease. Ann Neurol.
244. Alon R, Hammer DA, Springer TA. Lifetime of the P-selectin-carbohydrate 2012;72:648672.
bond and its response to tensile force in hydrodynamic flow. Nature. 272. Stamatovic SM, Keep RF, Andjelkovic AV. Brain endothelial cell-cell junc-
1995;374:539542. tions: how to open the blood brain barrier. Curr Neuropharmacol.
245. Sundd P, Pospieszalska MK, Ley K. Neutrophil rolling at high shear: 2008;6:179192.
flattening, catch bond behavior, tethers and slings. Mol Immunol. 273. Capaldo CT, Nusrat A. Cytokine regulation of tight junctions. Biochim
2012;55:5969. Biophys Acta. 2009;1788:864871.
246. Tang T, Frenette PS, Hynes RO, et al. Cytokine-induced meningitis is 274. Wong D, Dorovini-Zis K, Vincent SR. Cytokines, nitric oxide, and cGMP
dramatically attenuated in mice deficient in endothelial selectins. J Clin modulate the permeability of an in vitro model of the human blood-brain
Invest. 1996;97:24852490. barrier. Exp Neurol. 2004;190:446455.
247. Granert C, Raud J, Xie X, et al. Inhibition of leukocyte rolling with poly- 275. Chaitanya GV, Cromer WE, Wells SR, et al. Gliovascular and cytokine inter-
saccharide fucoidin prevents pleocytosis in experimental meningitis in the actions modulate brain endothelial barrier in vitro. J Neuroinflammation.
rabbit. J Clin Invest. 1994;93:929936. 2011;8:162.
248. Ostergaard C, Yieng-Kow RV, Benfield T, et al. Inhibition of leuko- 276. Forster C, Burek M, Romero IA, et al. Differential effects of hydrocorti-
cyte entry into the brain by the selectin blocker fucoidin decreases sone and TNFalpha on tight junction proteins in an in vitro model of the
interleukin-1 (IL-1) levels but increases IL-8 levels in cerebrospinal fluid human blood-brain barrier. J Physiol. 2008;586:19371949.
during experimental pneumococcal meningitis in rabbits. Infect Immun. 277. Rigor RR, Beard RS Jr, Litovka OP, et al. Interleukin-1beta-induced
2000;68:31533157. barrier dysfunction is signaled through PKC-theta in human brain
249. Brandt CT, Lundgren JD, Frimodt-Moller N, et al. Blocking of leukocyte microvascular endothelium. Am J Physiol Cell Physiol. 2012;302:
accumulation in the cerebrospinal fluid augments bacteremia and increases C1513C1522.
lethality in experimental pneumococcal meningitis. J Neuroimmunol. 278. Rosenberg GA. Matrix metalloproteinases and their multiple roles in neu-
2005;166:126131. rodegenerative diseases. Lancet Neurol. 2009;8:205216.
250. Campbell JJ, Qin S, Bacon KB, et al. Biology of chemokine and classi- 279. Koedel U, Bernatowicz A, Paul R, et al. Experimental pneumococcal
cal chemoattractant receptors: differential requirements for adhesion- meningitis: cerebrovascular alterations, brain edema, and meningeal
triggering versus chemotactic responses in lymphoid cells. J Cell Biol. inflammation are linked to the production of nitric oxide. Ann Neurol.
1996;134:255266. 1995;37:313323.
251. Chan JR, Hyduk SJ, Cybulsky MI. Chemoattractants induce a rapid and 280. Leib SL, Kim YS, Chow LL, et al. Reactive oxygen intermediates con-
transient upregulation of monocyte alpha4 integrin affinity for vascular tribute to necrotic and apoptotic neuronal injury in an infant rat model
cell adhesion molecule 1 which mediates arrest: an early step in the pro- of bacterial meningitis due to group B streptococci. J Clin Invest.
cess of emigration. J Exp Med. 2001;193:11491158. 1996;98:26322639.
252. Phillipson M, Heit B, Colarusso P, et al. Intraluminal crawling of neutro- 281. Auer M, Pfister LA, Leppert D, et al. Effects of clinically used antioxi-
phils to emigration sites: a molecularly distinct process from adhesion in dants in experimental pneumococcal meningitis. J Infect Dis. 2000;182:
the recruitment cascade. J Exp Med. 2006;203:25692575. 347350.
253. Bohatschek M, Werner A, Raivich G. Systemic LPS injection leads to 282. Mittal R, Gonzalez-Gomez I, Goth KA, et al. Inhibition of inducible
granulocyte influx into normal and injured brain: effects of ICAM-1 defi- nitric oxide controls pathogen load and brain damage by enhancing
ciency. Exp Neurol. 2001;172:137152. phagocytosis of Escherichia coli K1 in neonatal meningitis. Am J Pathol.
254. Weber JR, Angstwurm K, Burger W, et al. Anti ICAM-1 (CD 54) mono- 2010;176:12921305.
clonal antibody reduces inflammatory changes in experimental bacterial 283. Kastenbauer S, Koedel U, Becker BF, et al. Oxidative stress in bacterial
meningitis. J Neuroimmunol. 1995;63:6368. meningitis in humans. Neurology. 2002;58:186191.
255. Saez-Llorens X, Jafari HS, Severien C, et al. Enhanced attenuation of men- 284. Koedel U, Pfister HW. Protective effect of the antioxidant N-acetyl-
ingeal inflammation and brain edema by concomitant administration of L-cysteine in pneumococcal meningitis in the rat. Neurosci Lett.
anti-CD18 monoclonal antibodies and dexamethasone in experimental 1997;225:3336.
Haemophilus meningitis. J Clin Invest. 1991;88:20032011. 285. Kastenbauer S, Koedel U, Becker BF, et al. Experimental meningitis in the
256. Tuomanen EI, Prasad SM, George JS, et al. Reversible opening of the rat: protection by uric acid at human physiological blood concentrations.
blood-brain barrier by anti-bacterial antibodies. Proc Natl Acad Sci Eur J Pharmacol. 2001;425:149152.
U S A. 1993;90:78247828. 286. Pun PB, Lu J, Moochhala S. Involvement of ROS in BBB dysfunction. Free
257. Wittchen ES. Endothelial signaling in paracellular and transcellular leuko- Radic Res. 2009;43:348364.
cyte transmigration. Front Biosci. 2009;14:25222545. 287. Koedel U, Pfister HW. Superoxide production by primary rat cere-
258. von Wedel-Parlow M, Schrot S, Lemmen J, et al. Neutrophils cross the bral endothelial cells in response to pneumococci. J Neuroimmunol.
BBB primarily on transcellular pathways: an in vitro study. Brain Res. 1999;96:190200.
2011;1367:6276. 288. Caksen H, Cemek M, Dede S, et al. Brief clinical study: lipid peroxida-
259. Wilhelm C, Ellner JJ. Chronic meningitis. Neurol Clin. 1986;4:115141. tion and antioxidant status in children with acute purulent meningitis and
260. Cairns H, Russell DS. Cerebral arteritis and phlebitis in pneumococcal encephalitis. Int J Neurosci. 2004;114:105111.
meningitis. J Pathol Bacteriol. 1946;58:649665. 289. Lorenzl S, Koedel U, Frei K, et al. Protective effect of a 21-aminosteroid
261. Vergouwen MD, Schut ES, Troost D, et al. Diffuse cerebral intravas- during experimental pneumococcal meningitis. J Infect Dis. 1995;
cular coagulation and cerebral infarction in pneumococcal meningitis. 172:113118.
Neurocrit Care. 2010;13:217227. 290. Christen S, Schaper M, Lykkesfeldt J, et al. Oxidative stress in brain dur-
262. Klein M, Koedel U, Pfefferkorn T, et al. Arterial cerebrovascular com- ing experimental bacterial meningitis: differential effects of alpha-phenyl-
plications in 94 adults with acute bacterial meningitis. Crit Care. tert-butyl nitrone and N-acetylcysteine treatment. Free Radic Biol Med.
2011;15:R281. 2001;31:754762.
263. Jan W, Zimmerman RA, Bilaniuk LT, et al. Diffusion-weighted imaging in 291. Paul R, Angele B, Popp B, et al. Differential regulation of blood-brain
acute bacterial meningitis in infancy. Neuroradiology. 2003;45:634639. barrier permeability in brain trauma and pneumococcal meningitis-role of
264. Takeoka M, Takahashi T. Infectious and inflammatory disorders of Src kinases. Exp Neurol. 2007;203:158167.
the circulatory system and stroke in childhood. Curr Opin Neurol. 292. Argaw AT, Asp L, Zhang J, et al. Astrocyte-derived VEGF-A drives blood-
2002;15:159164. brain barrier disruption in CNS inflammatory disease. J Clin Invest.
265. Weiss N, Miller F, Cazaubon S, et al. The blood-brain barrier in 2012;122:24542468.
brain homeostasis and neurological diseases. Biochim Biophys Acta. 293. van der Flier M, Coenjaerts FE, Mwinzi PN, et al. Antibody neutralization
2009;1788:842857. of vascular endothelial growth factor (VEGF) fails to attenuate vascular
266. Zysk G, Schneider-Wald BK, Hwang JH, et al. Pneumolysin is the main permeability and brain edema in experimental pneumococcal meningitis.
inducer of cytotoxicity to brain microvascular endothelial cells caused by J Neuroimmunol. 2005;160:170177.
Streptococcus pneumoniae. Infect Immun. 2001;69:845852. 294. Stamatovic SM, Dimitrijevic OB, Keep RF, et al. Inflammation and brain
267. NGuessan PD, Schmeck B, Ayim A, et al. Streptococcus pneumoniae R6x edema: new insights into the role of chemokines and their receptors. Acta
induced p38 MAPK and JNK-mediated caspase-dependent apoptosis in Neurochir Suppl. 2006;96:444450.
human endothelial cells. Thromb Haemost. 2005;94:295303. 295. Owens HM, Destache CJ, Dash AK. Simple liquid chromatographic
268. Bermpohl D, Halle A, Freyer D, et al. Bacterial programmed cell death method for the analysis of the blood brain barrier permeability char-
of cerebral endothelial cells involves dual death pathways. J Clin Invest. acteristics of ceftriaxone in an experimental rabbit meningitis model.
2005;115:16071615. J Chromatogr B Biomed Sci Appl. 1999;728:97105.

Scheld_Ch23.indd 361 2/21/14 5:41 PM


362 Part III: Bacterial and Mycoplasmal Infections

296. Leib SL, Kim YS, Black SM, et al. Inducible nitric oxide synthase and 327. Edmond K, Dieye Y, Griffiths UK, et al. Prospective cohort study of dis-
the effect of aminoguanidine in experimental neonatal meningitis. J Infect abling sequelae and quality of life in children with bacterial meningitis in
Dis. 1998;177:692700. urban Senegal. Pediatr Infect Dis J. 2010;29:10231029.
297. Lu CH, Chang HW, Lui CC, et al. Cerebral haemodynamics in acute bac- 328. Adachi N, Ito K, Sakata H. Risk factors for hearing loss after pediatric
terial meningitis in adults. QJM. 2006;99:863869. meningitis in Japan. Ann Otol Rhinol Laryngol. 2010;119:294296.
298. Moller K, Qvist T, Tofteng F, et al. Cerebral blood flow and metabolism 329. Douglas SA, Sanli H, Gibson WP. Meningitis resulting in hearing loss and
during infusion of norepinephrine and propofol in patients with bacterial labyrinthitis ossificansdoes the causative organism matter? Cochlear
meningitis. Stroke. 2004;35:13331339. Implants Int. 2008;9:9096.
299. Ries S, Schminke U, Fassbender K, et al. Cerebrovascular involvement in 330. Kutz JW, Simon LM, Chennupati SK, et al. Clinical predictors for hearing
the acute phase of bacterial meningitis. J Neurol. 1997;244:5155. loss in children with bacterial meningitis. Arch Otolaryngol Head Neck
300. Pfister HW, Borasio GD, Dirnagl U, et al. Cerebrovascular complications Surg. 2006;132:941945.
of bacterial meningitis in adults. Neurology. 1992;42:14971504. 331. Dichgans M, Jager L, Mayer T, et al. Bacterial meningitis in adults:
301. Pfister HW, Feiden W, Einhaupl KM. Spectrum of complications during demonstration of inner ear involvement using high-resolution MRI.
bacterial meningitis in adults. Results of a prospective clinical study. Arch Neurology. 1999;52:10031009.
Neurol. 1993;50:575581. 332. Caye-Thomasen P, Dam MS, Omland SH, et al. Cochlear ossification in
302. Perry JR, Bilbao JM, Gray T. Fatal basilar vasculopathy complicating bac- patients with profound hearing loss following bacterial meningitis. Acta
terial meningitis. Stroke. 1992;23:11751178. Otolaryngol. 2012;132:720725.
303. Ment LR, Ehrenkranz RA, Duncan CC. Bacterial meningitis as an etiol- 333. Nadol JB Jr, Hsu WC. Histopathologic correlation of spiral ganglion cell
ogy of perinatal cerebral infarction. Pediatr Neurol. 1986;2:276279. count and new bone formation in the cochlea following meningogenic
304. Tureen J, Liu Q, Chow L. Near-infrared spectroscopy in experimental labyrinthitis and deafness. Ann Otol Rhinol Laryngol. 1991;100:712716.
pneumococcal meningitis in the rabbit: cerebral hemodynamics and 334. Merchant SN, Gopen Q. A human temporal bone study of acute bacterial
metabolism. Pediatr Res. 1996;40:759763. meningogenic labyrinthitis. Am J Otol. 1996;17:375385.
305. Tuber MG. Brain edema, intracranial pressure and cerebral blood flow 335. Klein M, Koedel U, Pfister HW, et al. Morphological correlates of acute
in bacterial meningitis. Pediatr Infect Dis J. 1989;8:915917. and permanent hearing loss during experimental pneumococcal meningi-
306. Ashwal S, Tomasi L, Schneider S, et al. Bacterial meningitis in children: tis. Brain Pathol. 2003;13:123132.
pathophysiology and treatment. Neurology. 1992;42:739748. 336. Caye-Thomasen P, Worsoe L, Brandt CT, et al. Routes, dynamics, and cor-
307. Moller K, Skinhoj P, Knudsen GM, et al. Effect of short-term hyperventi- relates of cochlear inflammation in terminal and recovering experimental
lation on cerebral blood flow autoregulation in patients with acute bacte- meningitis. Laryngoscope. 2009;119:15601570.
rial meningitis. Stroke. 2000;31:11161122. 337. Blank AL, Davis GL, VanDeWater TR, et al. Acute Streptococcus pneu-
308. Pedersen M, Brandt CT, Knudsen GM, et al. Cerebral blood flow autoreg- moniae meningogenic labyrinthitis. An experimental guinea pig model
ulation in early experimental S. pneumoniae meningitis. J Appl Physiol. and literature review. Arch Otolaryngol Head Neck Surg. 1994;120:
2007;102:7278. 13421346.
309. Tureen JH, Dworkin RJ, Kennedy SL, et al. Loss of cerebrovascu- 338. Bhatt S, Halpin C, Hsu W, et al. Hearing loss and pneumococcal meningi-
lar autoregulation in experimental meningitis in rabbits. J Clin Invest. tis: an animal model. Laryngoscope. 1991;101:12851292.
1990;85:577581. 339. Klein M, Koedel U, Kastenbauer S, et al. Nitrogen and oxygen molecules
310. Pfister HW, Koedel U, Haberl RL, et al. Microvascular changes during in meningitis-associated labyrinthitis and hearing impairment. Infection.
the early phase of experimental bacterial meningitis. J Cereb Blood Flow 2008;36:214.
Metab. 1990;10:914922. 340. Winter AJ, Comis SD, Osborne MP, et al. A role for pneumolysin but
311. Pfister HW, Koedel U, Dirnagl U, et al. Superoxide dismutase inhibits not neuraminidase in the hearing loss and cochlear damage induced by
brain oedema formation in experimental pneumococcal meningitis. Acta experimental pneumococcal meningitis in guinea pigs. Infect Immun.
Neurochir Suppl (Wien). 1990;51:378380. 1997;65:44114418.
312. Pfister HW, Kodel U, Dirnagl U, et al. Effect of catalase on regional cere- 341. Rappaport JM, Bhatt SM, Kimura RS, et al. Electron microscopic tempo-
bral blood flow and brain edema during the early phase of experimental ral bone histopathology in experimental pneumococcal meningitis. Ann
pneumococcal meningitis. J Infect Dis. 1992;166:14421445. Otol Rhinol Laryngol. 1999;108:537547.
313. Hoffmann OM, Becker D, Weber JR. Bacterial hydrogen peroxide con- 342. Skinner LJ, Beurg M, Mitchell TJ, et al. Intracochlear perfusion of pneu-
tributes to cerebral hyperemia during early stages of experimental pneu- molysin, a pneumococcal protein, rapidly abolishes auditory potentials in
mococcal meningitis. J Cereb Blood Flow Metab. 2007;27:17921797. the Guinea pig cochlea. Acta Otolaryngol. 2004;124:10001007.
314. Schaper M, Gergely S, Lykkesfeldt J, et al. Cerebral vasculature is the 343. Brandt CT, Caye-Thomasen P, Lund SP, et al. Hearing loss and cochlear
major target of oxidative protein alterations in bacterial meningitis. damage in experimental pneumococcal meningitis, with special reference
J Neuropathol Exp Neurol. 2002;61:605613. to the role of neutrophil granulocytes. Neurobiol Dis. 2006;23:300311.
315. Szabo C. Multiple pathways of peroxynitrite cytotoxicity. Toxicol Lett. 344. Kastenbauer S, Klein M, Koedel U, et al. Reactive nitrogen species con-
2003;140141:105112. tribute to blood-labyrinth barrier disruption in suppurative labyrinthitis
316. Kastenbauer S, Koedel U, Becker BF, et al. Pneumococcal meningitis in complicating experimental pneumococcal meningitis in the rat. Brain Res.
the rat: evaluation of peroxynitrite scavengers for adjunctive therapy. Eur 2001;904:208217.
J Pharmacol. 2002;449:177181. 345. Klein M, Koedel U, Pfister HW, et al. Meningitis-associated hearing loss:
317. Koedel U, Gorriz C, Lorenzl S, et al. Increased endothelin levels in cere- protection by adjunctive antioxidant therapy. Ann Neurol. 2003;54:
brospinal fluid samples from adults with bacterial meningitis. Clin Infect 451458.
Dis. 1997;25:329330. 346. Ge NN, Brodie HA, Tinling SP. Long-term hearing loss in gerbils with
318. Skopal J, Turbucz P, Vastag M, et al. Regulation of endothelin release bacterial meningitis treated with superoxide dismutase. Otol Neurotol.
from human brain microvessel endothelial cells. J Cardiovasc Pharmacol. 2008;29:10611067.
1998;31(suppl 1):S370S372. 347. Amaee FR, Comis SD, Osborne MP, et al. Possible involvement of ni-
319. Sibson NR, Blamire AM, Perry VH, et al. TNF-alpha reduces cerebral tric oxide in the sensorineural hearing loss of bacterial meningitis. Acta
blood volume and disrupts tissue homeostasis via an endothelin- and Otolaryngol. 1997;117:329336.
TNFR2-dependent pathway. Brain. 2002;125:24462459. 348. Pai N, Zdanski CJ, Gregory CW, et al. Sodium nitroprusside/nitric oxide
320. Sury MD, Frese-Schaper M, Muhlemann MK, et al. Evidence that causes apoptosis in spiral ganglion cells. Otolaryngol Head Neck Surg.
N-acetylcysteine inhibits TNF-alpha-induced cerebrovascular endothe- 1998;119:323330.
lin-1 upregulation via inhibition of mitogen- and stress-activated protein 349. Jung TT, Llaurado RJ, Nam BH, et al. Effects of nitric oxide on morphol-
kinase. Free Radic Biol Med. 2006;41:13721383. ogy of isolated cochlear outer hair cells: possible involvement in sensori-
321. Brunner F, Bras-Silva C, Cerdeira AS, et al. Cardiovascular endothelins: neural hearing loss. Otol Neurotol. 2003;24:682685.
essential regulators of cardiovascular homeostasis. Pharmacol Ther. 350. Meli DN, Coimbra RS, Erhart DG, et al. Doxycycline reduces mortality
2006;111:508531. and injury to the brain and cochlea in experimental pneumococcal menin-
322. Pfister LA, Tureen JH, Shaw S, et al. Endothelin inhibition improves gitis. Infection and Immunity. 2006;74:38903896.
cerebral blood flow and is neuroprotective in pneumococcal meningitis. 351. Grandgirard D, Burri M, Agyeman P, et al. Adjunctive daptomycin
Ann Neurol. 2000;47:329335. attenuates brain damage and hearing loss more efficiently than rifampin
323. Edmond K, Clark A, Korczak VS, et al. Global and regional risk of in infant rat pneumococcal meningitis. Antimicrob Agents Chemother.
disabling sequelae from bacterial meningitis: a systematic review and 2012;56:42894295.
meta-analysis. Lancet Infect Dis. 2010;10:317328. 352. Rappaport JM, Bhatt SM, Burkard RF, et al. Prevention of hearing loss in
324. Kesser BW, Hashisaki GT, Spindel JH, et al. Time course of hearing loss experimental pneumococcal meningitis by administration of dexametha-
in an animal model of pneumococcal meningitis. Otolaryngol Head Neck sone and ketorolac. J Infect Dis. 1999;179:264268.
Surg. 1999;120:628637. 353. Worsoe L, Brandt CT, Lund SP, et al. Intratympanic steroid prevents
325. Woolley AL, Kirk KA, Neumann AM Jr, et al. Risk factors for hearing long-term spiral ganglion neuron loss in experimental meningitis. Otol
loss from meningitis in children: the Childrens Hospital experience. Arch Neurotol. 2010.
Otolaryngol Head Neck Surg. 1999;125:509514. 354. Kim HH, Addison J, Suh E, et al. Otoprotective effects of dexametha-
326. Oostenbrink R, Maas M, Moons KG, et al. Sequelae after bacterial men- sone in the management of pneumococcal meningitis: an animal study.
ingitis in childhood. Scand J Infect Dis. 2002;34:379382. Laryngoscope. 2007;117:12091215.

Scheld_Ch23.indd 362 2/21/14 5:41 PM


Chapter 23: Pathogenesis and Pathophysiology of Bacterial Infections 363

355. Coimbra RS, Loquet G, Leib SL. Limited efficacy of adjuvant therapy 387. Loeffler JM, Ringer R, Hablutzel M, et al. The free radical scavenger
with dexamethasone in preventing hearing loss due to experimental pneu- alpha-phenyl-tert-butyl nitrone aggravates hippocampal apoptosis and
mococcal meningitis in the infant rat. Pediatr Res. 2007;62:291294. learning deficits in experimental pneumococcal meningitis. The Journal of
356. Brouwer MC, McIntyre P, de Gans J, et al. Corticosteroids for acute bac- Infectious Diseases. 2001;183:247252.
terial meningitis. Cochrane Database Syst Rev. 2010;(9):CD004405. 388. Wellmer A, Noeske C, Gerber J, et al. Spatial memory and learning defi-
357. Peltola H, Roine I, Fernandez J, et al. Hearing impairment in child- cits after experimental pneumococcal meningitis in mice. Neurosci Lett.
hood bacterial meningitis is little relieved by dexamethasone or glycerol. 2000;296:137140.
Pediatrics. 2010;125:e1e8. 389. Grandgirard D, Bifrare YD, Pleasure SJ, et al. Pneumococcal meningitis
358. Manolidis S, Johnson R. Do corticosteroids prevent hearing loss in pedi- induces apoptosis in recently postmitotic immature neurons in the dentate
atric bacterial meningitis? An analysis of the evidence. Ear Nose Throat J. gyrus of neonatal rats. Dev Neurosci. 2007;29:134142.
2006;85:586592. 390. Leib SL, Heimgartner C, Bifrare YD, et al. Dexamethasone aggravates
359. Lebel MH, Freij BJ, Syrogiannopoulos GA, et al. Dexamethasone therapy hippocampal apoptosis and learning deficiency in pneumococcal meningi-
for bacterial meningitis. Results of two double- blind, placebo-controlled tis in infant rats. Pediatric research. 2003;54:353357.
trials. N Engl J Med. 1988;319:964971. 391. Hofer S, Grandgirard D, Burri D, et al. Bacterial meningitis impairs hip-
360. Casella EB, Cypel S, Osmo AA, et al. Sequelae from meningococcal pocampal neurogenesis. J Neuropathol Exp Neurol. 2011;70:890899.
meningitis in children: a critical analysis of dexamethasone therapy. Arq 392. Liebetanz D, Gerber J, Schiffner C, et al. Pre-infection physical exercise
Neuropsiquiatr. 2004;62:421428. decreases mortality and stimulates neurogenesis in bacterial meningitis.
361. Grimwood K, Nolan TM, Bond L, et al. Risk factors for adverse outcomes J Neuroinflammation. 2012;9:168.
of bacterial meningitis. J Paediatr Child Health. 1996;32:457462. 393. Tauber SC, Stadelmann C, Spreer A, et al. Increased expression of BDNF
362. Bedford H, de Louvois J, Halket S, et al. Meningitis in infancy in England and proliferation of dentate granule cells after bacterial meningitis.
and Wales: follow up at age 5 years. BMJ. 2001;323:533536. J Neuropathol Exp Neurol. 2005;64:806815.
363. Anderson V, Anderson P, Grimwood K, et al. Cognitive and executive 394. Gerber J, Bottcher T, Bering J, et al. Increased neurogenesis after experimen-
function 12 years after childhood bacterial meningitis: effect of acute neu- tal Streptococcus pneumoniae meningitis. J Neurosci Res. 2003;73:441446.
rologic complications and age of onset. J Pediatr Psychol. 2004;29:6781. 395. Wittwer M, Grandgirard D, Rohrbach J, et al. Tracking the transcrip-
364. Schmidt H, Heimann B, Djukic M, et al. Neuropsychological sequelae of tional host response from the acute to the regenerative phase of experi-
bacterial and viral meningitis. Brain. 2006;129:333345. mental pneumococcal meningitis. BMC Infect Dis. 2010;10:176.
365. Grimwood K, Anderson P, Anderson V, et al. Twelve year outcomes fol- 396. Nau R, Soto A, Bruck W. Apoptosis of neurons in the dentate gyrus in
lowing bacterial meningitis: further evidence for persisting effects. Arch humans suffering from bacterial meningitis. J Neuropathol Exp Neurol.
Dis Child. 2000;83:111116. 1999;58:265274.
366. Hoogman M, van de Beek D, Weisfelt M, et al. Cognitive outcome 397. Free SL, Li LM, Fish DR, et al. Bilateral hippocampal volume loss in patients
in adults after bacterial meningitis. J Neurol Neurosurg Psychiatry. with a history of encephalitis or meningitis. Epilepsia. 1996;37:400405.
2007;78:10921096. 398. Gerber J, Tauber SC, Armbrecht I, et al. Increased neuronal proliferation
367. Focke NK, Kallenberg K, Mohr A, et al. Distributed, limbic gray matter in human bacterial meningitis. Neurology. 2009;73:10261032.
atrophy in patients after bacterial meningitis. AJNR Am J Neuroradiol. 399. Coimbra RS, Voisin V, de Saizieu AB, et al. Gene expression in cortex and
2012. hippocampus during acute pneumococcal meningitis. BMC Biol. 2006;4:15.
368. Libster R, Edwards KM, Levent F, et al. Long-term outcomes of group B 400. Braun JS, Novak R, Murray PJ, et al. Apoptosis-inducing factor mediates
streptococcal meningitis. Pediatrics. 2012;130:e815. microglial and neuronal apoptosis caused by pneumococcus. J Infect Dis.
369. Bargui F, DAgostino I, Mariani-Kurkdjian P, et al. Factors influencing 2001;184:13001309.
neurological outcome of children with bacterial meningitis at the emer- 401. Thal SE, Zhu C, Thal SC, et al. Role of apoptosis inducing factor (AIF)
gency department. Eur J Pediatr. 2012;171:13651371. for hippocampal neuronal cell death following global cerebral ischemia in
370. Berman PH, Banker BQ. Neonatal meningitis. A clinical and pathological mice. Neurosci Lett. 2011;499:13.
study of 29 cases. Pediatrics. 1966;38:624. 402. Mitchell L, Smith SH, Braun JS, et al. Dual phases of apoptosis in pneu-
371. Arseni C, Nereantiu F. Multiple vascular thrombosis with vasculo- mococcal meningitis. J Infect Dis. 2004;190:20392046.
genic lesions in the course of meningoencephalitis. Confin Neurol. 403. Chan PH, Fishman RA, Wesley MA, et al. Pathogenesis of vasogenic
1972;34:339347. edema in focal cerebral ischemia. Role of superoxide radicals. Adv
372. McDonald R. Purulent meningitis in newborn babies: observations Neurol. 1990;52:177183.
and comments based on a series of 82 patients. Clin Pediatr (Phila). 404. Nag S, Manias JL, Stewart DJ. Pathology and new players in the patho-
1972;11:450454. genesis of brain edema. Acta Neuropathol. 2009;118:197217.
373. Erdohazi M. A clinical and pathological study of 43 cases of hydro- 405. Derugin N, Wendland M, Muramatsu K, et al. Evolution of brain injury
cephalus not associated with spina bifida. Dev Med Child Neurol Suppl. after transient middle cerebral artery occlusion in neonatal rats. Stroke.
1969;20:3137. 2000;31:17521761.
374. Eiben RM. Acute brain swelling (toxic encephalopathy). Pediatr Clin 406. Leib SL, Kim YS, Ferriero DM, et al. Neuroprotective effect of excitatory
North Am. 1967;14:797808. amino acid antagonist kynurenic acid in experimental bacterial meningi-
375. Hernandez MI, Sandoval CC, Tapia JL, et al. Stroke patterns in neonatal tis. J Infect Dis. 1996;173:166171.
group B streptococcal meningitis. Pediatr Neurol. 2011;44:282288. 407. Scheld WM, Dacey RG, Winn HR, et al. Cerebrospinal fluid outflow resis-
376. Jorens PG, Parizel PM, Wojciechowski M, et al. Streptococcus pneu- tance in rabbits with experimental meningitis. Alterations with penicillin
moniae meningoencephalitis with unusual and widespread white matter and methylprednisolone. J Clin Invest. 1980;66:243253.
lesions. Eur J Paediatr Neurol. 2008;12:127132. 408. Del Bigio MR. Neuropathological changes caused by hydrocephalus. Acta
377. Shah DK, Daley AJ, Hunt RW, et al. Cerebral white matter injury in the Neuropathol. 1993;85:573585.
newborn following Escherichia coli meningitis. Eur J Paediatr Neurol. 409. Kaplan SL, Feigin RD. The syndrome of inappropriate secretion of
2005;9:1317. antidiuretic hormone in children with bacterial meningitis. J Pediatr.
378. Tuber MG, Kim YS, Leib SL. Neuronal injury in meningitis. In: Peterson 1978;92:758761.
PK, Remington JS, eds. In Defense of the Brain. Malden, MA: Blackwell 410. Winkler F, Kastenbauer S, Yousry TA, et al. Discrepancies between
Science; 1997:124143. brain CT imaging and severely raised intracranial pressure proven by
379. Nau R, Gerber J, Bunkowski S, et al. Axonal injury, a neglected cause of ventriculostomy in adults with pneumococcal meningitis. J Neurol.
CNS damage in bacterial meningitis. Neurology. 2004;62:509511. 2002;249:12921297.
380. Gerber J, Seitz RC, Bunkowski S, et al. Evidence for frequent focal and dif- 411. Lindvall P, Ahlm C, Ericsson M, et al. Reducing intracranial pressure may
fuse acute axonal injury in human bacterial meningitis. Clin Neuropathol. increase survival among patients with bacterial meningitis. Clin Infect
2009;28:3339. Dis. 2004;38:384390.
381. Renier D, Flandin C, Hirsch E, et al. Brain abscesses in neonates. A study 412. Fishman RA. Brain edema. N Engl J Med. 1975;293:706711.
of 30 cases. J Neurosurg. 1988;69:877882. 413. Brown LW, Feigin RD. Bacterial meningitis: fluid balance and therapy.
382. Cone LA, Leung MM, Byrd RG, et al. Multiple cerebral abscesses because Pediatr Ann. 1994;23:9398.
of Listeria monocytogenes: three case reports and a literature review of 414. Agre P, King LS, Yasui M, et al. Aquaporin water channelsfrom atomic
supratentorial listerial brain abscess(es). Surg Neurol. 2003;59:320328. structure to clinical medicine. J Physiol. 2002;542:316.
383. Zysk G, Bruck W, Gerber J, et al. Anti-inflammatory treatment influences 415. Arcienega, II, Brunet JF, Bloch J, et al. Cell locations for AQP1, AQP4 and
neuronal apoptotic cell death in the dentate gyrus in experimental pneu- 9 in the non-human primate brain. Neuroscience. 2010;167:11031114.
mococcal meningitis. J Neuropathol Exp Neurol. 1996;55:722728. 416. Saadoun S, Papadopoulos MC. Aquaporin-4 in brain and spinal cord oe-
384. Bifrare YD, Gianinazzi C, Imboden H, et al. Bacterial meningitis causes dema. Neuroscience. 2010;168:10361046.
two distinct forms of cellular damage in the hippocampal dentate gyrus in 417. Papadopoulos MC, Verkman AS. Aquaporin-4 gene disruption in mice
infant rats. Hippocampus. 2003;13:481488. reduces brain swelling and mortality in pneumococcal meningitis. J Biol
385. Gianinazzi C, Grandgirard D, Imboden H, et al. Caspase-3 mediates Chem. 2005;280:1390613912.
hippocampal apoptosis in pneumococcal meningitis. Acta Neuropathol. 418. Saadoun S, Papadopoulos MC, Krishna S. Water transport becomes
2003;105:499507. uncoupled from K siphoning in brain contusion, bacterial meningitis,
386. Braun JS, Novak R, Herzog KH, et al. Neuroprotection by a caspase and brain tumours: immunohistochemical case review. J Clin Pathol.
inhibitor in acute bacterial meningitis. Nat Med. 1999;5:298302. 2003;56:972975.

Scheld_Ch23.indd 363 2/21/14 5:41 PM


364 Part III: Bacterial and Mycoplasmal Infections

419. Blocher J, Eckert I, Elster J, et al. Aquaporins AQP1 and AQP4 in the 433. Carney JM, Starke-Reed PE, Oliver CN, et al. Reversal of age-related in-
cerebrospinal fluid of bacterial meningitis patients. Neurosci Lett. crease in brain protein oxidation, decrease in enzyme activity, and loss
2011;504:2327. in temporal and spatial memory by chronic administration of the spin-
420. Hasbun R, Abrahams J, Jekel J, et al. Computed tomography of the head trapping compound N-tert-butyl-alpha-phenylnitrone. Proc Natl Acad Sci
before lumbar puncture in adults with suspected meningitis. N Engl J U S A. 1991;88:36333636.
Med. 2001;345:17271733. 434. Klein M, Koedel U, Pfister HW. Oxidative stress in pneumococcal
421. McMillan DA, Lin CY, Aronin SI, et al. Community-acquired bacterial meningitis: a future target for adjunctive therapy? Prog Neurobiol.
meningitis in adults: categorization of causes and timing of death. Clin 2006;80:269280.
Infect Dis. 2001;33:969975. 435. Spranger M, Schwab S, Krempien S, et al. Excess glutamate levels in the
422. Sugiyama K, Brunori A, Mayer ML. Glial uptake of excitatory amino cerebrospinal fluid predict clinical outcome of bacterial meningitis. Arch
acids influences neuronal survival in cultures of mouse hippocampus. Neurol. 1996;53:992996.
Neuroscience. 1989;32:779791. 436. Ghielmetti M, Ren H, Leib SL, et al. Impaired cortical energy metabolism
423. Spranger M, Krempien S, Schwab S, et al. Excess glutamate in the cerebro- but not major antioxidant defenses in experimental bacterial meningitis.
spinal fluid in bacterial meningitis. J Neurol Sci. 1996;143:126131. Brain Res. 2003;976:139148.
424. Guerra-Romero L, Tauber MG, Fournier MA, et al. Lactate and glu- 437. Simon RP, Beckman JS. Why pus is bad for the brain. Neurology.
cose concentrations in brain interstitial fluid, cerebrospinal fluid, and 2002;58:167168.
serum during experimental pneumococcal meningitis. J Infect Dis. 438. Braun J. Inducible nitric oxide synthase mediates hippocampal cas-
1992;166:546550. pase-3 activation in pneumococcal meningitis. Int J Neurosci. 2009;119:
425. Sellner J, Ringer R, Baumann P, et al. Effect of the NMDA-receptor 455459.
antagonist dextromethorphan in infant rat pneumococcal meningitis. 439. Guo Z, Sun X, He Z, et al. Role of matrix metalloproteinase-9 in apop-
Curr Drug Metab. 2008;9:8388. tosis of hippocampal neurons in rats during early brain injury after
426. Bellac CL, Coimbra RS, Christen S, et al. Inhibition of the kynuren- subarachnoid hemorrhage. Neurol Sci. 2010;31:143149.
ine-NAD pathway leads to energy failure and exacerbates apopto- 440. Murase S, McKay RD. Matrix metalloproteinase-9 regulates sur-
sis in pneumococcal meningitis. J Neuropathol Exp Neurol. 2010;69: vival of neurons in newborn hippocampus. J Biol Chem. 2012;287:
10961104. 1218412194.
427. Grandgirard D, Leib SL. Meningitis in neonates: bench to bedside. Clin 441. Fujita-Hamabe W, Tokuyama S. The involvement of cleavage of neural
Perinatol. 2010;37:655676. cell adhesion molecule in neuronal death under oxidative stress conditions
428. Recher M, Malipiero U, Schaer DJ, et al. Inhibition of meningitis-associ- in cultured cortical neurons. Biol Pharm Bull. 2012;35:624628.
ated neutrophil apoptosis by TNF-alpha depends on functional PI3-kinase 442. Braun JS, Sublett JE, Freyer D, et al. Pneumococcal pneumolysin and
in monocytes. J Leukoc Biol. 2013;93:259266. H(2)O(2) mediate brain cell apoptosis during meningitis. J Clin Invest.
429. Tunbridge AJ, Stevanin TM, Lee M, et al. Inhibition of macrophage apop- 2002;109:1927.
tosis by Neisseria meningitidis requires nitric oxide detoxification mecha- 443. Braun JS, Hoffmann O, Schickhaus M, et al. Pneumolysin causes neuro-
nisms. Infect Immun. 2006;74:729733. nal cell death through mitochondrial damage. Infect Immun. 2007;75:
430. Wennekamp J, Henneke P. Induction and termination of inflammatory 42454254.
signaling in group B streptococcal sepsis. Immunol Rev. 2008;225: 444. Beurg M, Hafidi A, Skinner L, et al. The mechanism of pneumolysin-
114127. induced cochlear hair cell death in the rat. J Physiol. 2005;568:211227.
431. Koedel U, Frankenberg T, Kirschnek S, et al. Apoptosis is essential for 445. Reiss A, Braun JS, Jager K, et al. Bacterial pore-forming cytolysins induce
neutrophil functional shutdown and determines tissue damage in experi- neuronal damage in a rat model of neonatal meningitis. J Infect Dis.
mental pneumococcal meningitis. PLoS Pathog. 2009;5:e1000461. 2011;203:393400.
432. Nguyen HX, OBarr TJ, Anderson AJ. Polymorphonuclear leukocytes 446. Ostergaard C, Leib SL, Rowland I, et al. Bacteremia causes hippocam-
promote neurotoxicity through release of matrix metalloproteinases, re- pal apoptosis in experimental pneumococcal meningitis. BMC Infect Dis.
active oxygen species, and TNF-alpha. J Neurochem. 2007;102:900912. 2010;10:1.

Scheld_Ch23.indd 364 2/21/14 5:41 PM


CHAPTER 24 ACUTE BACTERIAL MENINGITIS
KAREN L. ROOS, ALLAN R. TUNKEL, DIEDERIK VAN DE BEEK, AND W. MICHAEL SCHELD

The meningitis syndrome has been recognized for centu- isolated and described in the last two decades of the nineteenth
ries. Hippocrates realized the important intracranial conse- century. Quincke introduced lumbar puncture (LP) in 1891,
quences of otitic infection, and clear clinical descriptions of and the major CSF alterations associated with meningitis
meningitis have been found dating from the sixteenth cen- (pleocytosis, hypoglycorrhachia, and elevated protein concen-
tury. However, the syndrome of epidemic meningitis with a tration) were well recognized by the turn of the century.
purpuric rash was not identified until 1805, when Viesseux The treatment of bacterial meningitis in the early years of
wrote about an epidemic of malignant purpuric fever sur- this century was dominated by methods for removal of large
rounding Geneva, Switzerland, the first clinical description of volumes of CSF and/or direct instillation of substances (e.g.,
meningococcemia with meningitis. The pathologic hallmark dyes, enzymes) into the SAS. After early leads from European
of the condition, inflammation within the subarachnoid space investigators, the first truly significant therapeutic modality
(SAS), was described in autopsy reports in the French liter- for this disorder on a large scale, the systemic and intrathecal
ature the following year. Danielson and Mann (1) recorded administration of antimeningococcal antisera raised in horses,
the first observations of meningococcemia and meningitis in was documented by Simon Flexner in 1913. Though toxic,
the United States in 1806. Many of these early descriptions antisera therapy reduced the mortality of meningococcal men-
were collated in a treatise by Elisha North of Connecticut in ingitis (from approximately 80% to 30%) during World War
1811 and summarized in references 1 and 2. Then, as now, I and for decades thereafter. The principles of serum therapy
the disease could present dramatically in a fulminant form. were applied by Dr. Hattie Alexander and others to meningitis
The epidemic nature of meningococcemia was frightening to caused by H. influenzae in the 1940s.
physicians and lay persons alike. For example, Dr. Samuel The approach to the patient with bacterial meningitis was
Woodward, of Torrington, Connecticut, wrote the following profoundly altered by the advent of antimicrobial therapy. The
in The American Mercury, Hartford, in 1807: first successful account of the therapy of meningococcal men-
The violent symptoms were great lassitude, with universal pains in
ingitis with an antimicrobial agent in this country was pub-
the muscles, chills; heats, if any, were of short duration; unusual lished by Schwentker et al. (3) in 1937; nine patients survived
prostration of strength; delirium, with severe pain in the head; after receiving subcutaneous and intraspinal injections of sul-
vomiting, with indescribable anxiety of stomach; eyes red and fanilamide, and the sole death occurred after eradication of the
watery, and rolled up, and the head drawn back with spasm; pulse organism from CSF. The introduction of penicillin and other
quick, weak, and irregular; petechiae and vibices all over the body, antimicrobial agents (e.g., streptomycin and chloramphenicol)
and a cadaverous countenance and smell; death often closed the ushered in the modern antimicrobial era, likened to an indus-
scene in ten or fifteen hours after the first attack . . . the body, near trial revolution (4). These developments led to the widespread
the fatal period, and soon after, became as spotted as an adder. . . . belief that serious bacterial infections were solved. Despite
Similarly, the following was written by the Reverend Festus the introduction of myriad new antimicrobial agents and the
Foster of Petersham, Massachusetts, as a letter to the editor of development of newer diagnostic techniques, the mortality
The Worcester Spy, dated March 6, 1810: from meningitis caused by the three major bacterial patho-
gens has not changed appreciably in the last four decades.
I hasten to give you a sketch of the spotted fever in this place.
It made its first appearance about the beginning of January last;
However, the use of the third-generation cephalosporins dur-
but the instances were few and distant from each other, until last ing the 1980s for therapy of gram-negative aerobic bacillary
week. Although it had proved fatal in most instances, seven only meningitis has substantially reduced the mortality of this con-
had died belonging to this town, previous to the 25th of February. dition. Recent years have revealed an explosion of new knowl-
Since that time the disorder has come upon us like a flood of edge on the pathogenesis and pathophysiology of bacterial
mighty waters. We have buried eight persons within the last eight meningitis (see later discussion), with attendant ramifications
days. About twelve or fifteen new cases appeared on Thursday last; on the use of adjunctive therapy (e.g., corticosteroids, nonste-
many of them very sudden and violent. This was the most melan- roidal antiinflammatory agents, and monoclonal antibodies)
choly and alarming day ever witnessed in this place. Seven or eight for this disease.
physicians were continually engaged in the neighborhood north of
the meeting house, and I believe not one half hour passed in the
forenoon without presenting a new case. Pale fear and extreme
anxiety were visible in every countenance. . . .
EPIDEMIOLOGY
It is inconceivable that this fulminant form of meningococ-
cemia had been previously unrecognized, especially given the During 2003 to 2007, approximately 4,100 cases of bacte-
excellent clinical descriptions of rashes in the literature from rial meningitis occurred annually in the United States (5), but
the period. One must speculate that the virulence of menin- this disease is much more common in developing countries
gococci for humans changed in the early nineteenth century. (see later discussion). In addition, the relative frequency with
Meningococci were first isolated in 1887 by Anton which each of the various bacterial species causes meningitis
Weichselbaum in Vienna; they were obtained from the cerebro- is age related. Gram-negative bacilli (principally Escherichia
spinal fluid (CSF) of six patients with meningitis and were ini- coli K1), group B streptococci, other enteric bacilli, and much
tially named Diplococcus intracellularis meningitidis. All three less commonly, Pseudomonas species are the major causative
of the major meningeal pathogens (Neisseria meningitidis, agents during the neonatal period. Meningitis in children and
Streptococcus pneumoniae, and Haemophilus influenzae) were adults is primarily caused by meningococci and pneumococci,

365

Scheld_Ch24.indd 365 2/21/14 5:42 PM


366 Part III: Bacterial and Mycoplasmal Infections

although disease caused by aerobic gram-negative bacilli is in- 55% for household contacts of epiglottitis and meningitis cases,
creasing in frequency, especially in the elderly. N. meningitidis respectively. This prevalence is reflected in the increased risk
is the only major cause of epidemics of bacterial meningitis. for serious Hib disease among household contacts of the index
The development of meningitis depends on a complex array case, which is age dependent: 4% for children 2 years of age,
of factors, including virulence properties of the organisms, the 2% for children 2 to 3 years old, and 0.1% for children 4 to
carrier state, and the hosts humoral immune response. These 5 years of age. The risk for Hib infection among household con-
factors differ among the major pathogens, for which reason epi- tacts is approximately 600-fold greater than the age-adjusted
demiology, carrier state, and host immunity are considered sepa- risk for the population at large and is the basis for chemopro-
rately for each of the three major etiologic agents in this section. phylactic strategies. Carriage is usually asymptomatic and may
The classification of the pathogens, putative virulence fac- occur despite the presence of circulating anticapsular antibod-
tors, and the clinical settings associated with less prevalent ies or effective eradication of meningitis following antibiotic
agents are discussed later (see the section Etiology, later in therapy. The Hib carrier state may persist for weeks to months.
this chapter). The occurrence of Hib meningitis is inversely proportional to
the age-related concentration of type-specific anticapsular anti-
bodies (18). Finnish studies, measuring antipolyribosylribitol
Haemophilus influenzae phosphate (anti-PRP) antibodies by radioimmunoassay, con-
firm the age-related susceptibility to systemic Hib disease:
H. influenzae type b (Hib) was previously the leading cause of 90% of children (3 to 12 months old) had concentrations of
bacterial meningitis in the United States but now accounts for less than 150 ng/mL, whereas adults had higher concentra-
only approximately 7% of cases (5). Meningitis caused by Hib tions (19). These anti-PRP antibodies, in concert with comple-
displays an interesting bimodal seasonal pattern in northern ment, are (a) opsonic and bactericidal against Hib in vitro and
Europe and the northern United States, with peaks in June and (b) protective in vivo. Antibodies to Hib outer membrane pro-
September through November (6,7). teins (Omps) also appear protective, but only against the ho-
The overall annual incidence of serious Hib disease dif- mologous subtype. The anti-PRP response to infection is age
fers between geographic locales and among populations. related, being poor in infants; older children and adults de-
Significant interannual variations in the incidence of meningi- velop higher titers. It is also dependent on PRP concentrations
tis caused by Hib have also been reported within a single geo- and clearance rates. PRP antigenemia may persist for weeks in
graphic area over time (8). This is an important consideration younger children with Hib meningitis, delaying the antibody
in assessing the efficacy of Hib vaccines. Before the advent of response. Approximately 80% of children with Hib meningitis
conjugate vaccines (see later discussion), the overall rate of develop an antibody response within 3 months. The antibody
Hib meningitis in the United States was approximately 60 per response is blunted in children with agammaglobulinemia or
100,000 children younger than 5 years of age (9), greater than immunoglobulin G2 (IgG2) subclass deficiency, as well as in
the figures from other countries in northern Europe. These all children younger than 24 months of age receiving the Hib
incidence rates differ markedly among age-groups (see later PRP vaccine, because this polysaccharide is a poor immunogen
discussion) and in children younger than 6 years. In a 3-year in this age-group (9). The age-related acquisition of protective
nationwide prospective study on pediatric meningitis in Israel, anticapsular antibodies is too rapid to be accounted for by
the incidence of Hib meningitis during the first year of life was the low incidence of carriage or disease caused by Hib alone.
67.1 per 100,000, and in children younger than 5 years of age, Cross-reacting antigens from E. coli and other bacteria within
it was 18.5 per 100,000 (10). Some studies report a higher the gut are postulated to serve as the primary immunogen.
incidence in nonwhites (7,11). For example, the incidence rate Acquisition of Hib (nasopharyngeal carriage) and the con-
for Hib meningitis for the total population of Washington centration of circulating anticapsular antibody are the two
State was 2.2, 3.4, and 13.5 per 100,000 for whites, blacks, main factors that determine risk for disease in most patients.
and Native Americans, respectively (11). In contrast, others Some of these risk factors have been alluded to (e.g., immuno-
have found no differences between rates for blacks and whites globulin or complement deficiency, household contacts of an
younger than 1 year of age (12). index case). Other conditions that also may be important in in-
Before the availability of the current Hib vaccines, 1 in creasing susceptibility to invasive Hib infection include sickle
every 200 children developed invasive Hib disease by 5 years cell anemia, postsplenectomy states, CSF fistulas, chronic pul-
of age. Meningitis caused by Hib in the first 2 months of life monary infections, alcoholism, and probably lower socioeco-
is rare, presumably because of placental transfer of protective nomic status (e.g., Eskimos and American Indians). Day care
concentrations of maternal bactericidal antibody. Most cases outside the home and the presence of young siblings increases
occur between 4 months and 2 years of age. The highest rate the risk for invasive disease, whereas breast-feeding is pro-
of illness occurs in children 6 to 17 months of age; children tective. The risk is highest for children younger than 2 years
older than 2 years of age have a lower incidence (6,8,9,1117). of age in day care but is not apparent for older children, is
Approximately 80% of cases develop in unvaccinated children equally high for those in a family day care setting or those in
younger than 2 years of age in this country, but this propor- a professional day care center (mean group size, 4 and 12 chil-
tion varies by geographic locale. The proportion of cases is ap- dren, respectively), and is significantly higher (p  .02) within
proximately 20% lower in this age-group in northern European the first month of attendance, especially among younger chil-
countries. These differences in age distribution may directly dren (20). The risk ratio doubles with each additional sibling
influence the efficacy of candidate Hib vaccines. Nontypeable younger than 7 years of age and is higher in twins (20). New
strains of H. influenzae are commonly carried in the nasophar- associations have also suggested that the childs previous state
ynx of asymptomatic individuals. Carriage of encapsulated of health, especially a history of otitis media and/or previ-
strains (usually type b) is rare: rates are less than 5% in children ous hospitalization, increases the risk for serious Hib disease.
and less than 1% in adults. However, the carriage rates among Otitis media remains significant, especially for younger chil-
household contacts of an index case are much higher: 20% to dren, even after controlling for confounding variables such
25% overall and more than 50% among children younger than as day care attendance (21). Pharyngitis and otitis media are
5 years of age. This varies with the clinical disease. For exam- associated with Hib meningitis in approximately one half and
ple, carriage rates among children 5 years old are 20% and two thirds of the cases, respectively.

Scheld_Ch24.indd 366 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 367

Because of the bimodal seasonal occurrence of Hib menin- cases in children younger than 5 years of age in Bulgaria in the
gitis, at least in northern latitudes, it has been suggested that mid-1990s (31). In addition, there was a report of emergence of
preceding viral upper respiratory tract infections predispose to cases in Nottingham, United Kingdom of invasive Hib disease
the acquisition of Hib and subsequent disease, but this issue in those previously vaccinated against the disease (32). Several
remains controversial (22). Recent studies comparing the at- explanations were put forth to explain this increase, most nota-
tack rates of meningitis between two ethnic groups living to- bly that the vaccination schedule in the United Kingdom was at
gether in one geographic area (Jews and Bedouins in the Negev 2, 3, and 4 months of age, and no booster dose was given (33).
region of Israel) suggested that community-acquired bacterial In areas in the developing world, declining rates of Hib men-
meningitis is associated more strongly with the type of mor- ingitis have been reported since introduction of Hib conjugate
bidity most prevalent in the region at any given time (e.g., vaccines, with effectiveness ranging from 88% to 94% (3437).
upper respiratory tract or gastrointestinal infections) rather The occurrence of meningitis caused by Hib in individuals older
than any specific type of infection (23). than 6 years of age should prompt efforts to exclude common ac-
There has been a profound reduction (from 76% to 90%) in companying conditions, such as otitis media, sinusitis, epiglottitis,
the incidence of invasive infections caused by Hib in the United CSF leaks, an immunodeficiency state, splenectomy or asplenic
States, specifically in young children (2427), attributed, in states, other parameningeal foci of infection, diabetes mellitus,
part, to the widespread use of conjugate vaccines against Hib and alcoholism (38,39). Although the incidence in children has
that were licensed for routine use in all children beginning at 2 dramatically declined, the incidence of invasive H. influenzae dis-
months of age. In a study of Hib disease rates in Los Angeles ease in adults is more complex. In one population-based study of
County, California, Hib disease was nearly eradicated in a the epidemiology and outcome caused by typeable and nontype-
fully immunized population, demonstrating the importance of able H. influenzae among adults in Utah during 1998 to 2008,
promotion of widespread use of these conjugate vaccines (28). there was an increase in incidence over the study period from 0.14
Similar results have been observed outside the United States. per 100,000 person-years in 1998 to 1.61 per 100,000 person-
In Finland, there has been a marked decrease in the number of years in 2008 (40); patients older than 65 years of age accounted
cases of Hib meningitis from a peak of 43 cases per 100,000 for 51% of the cases and 67% of the deaths.
population in the late 1970s to no cases in 1991 in the greater
Helsinki area (29). Similarly, during a prospective study of bac-
terial meningitis in the northeast Thames region of the United Neisseria meningitidis
Kingdom, there was an 87% decline in the number of cases
of Hib meningitis in 1993 (7 cases) compared with 1991 and Meningococcal infections continue to pose serious problems on
1992 (50 cases) (30). Widespread usage beginning at 2 months all continents. They are influenced by multiple factors, including
of age has nearly eliminated serious invasive Hib disease in chil- geography, season, climate, meningococcal serogroup, and pop-
dren in North America, Western Europe, Japan, and in many ulation demographics (2,41). Although worldwide in distribu-
areas of Latin America. Unfortunately, for reasons of cost, Hib tion, the incidence of epidemic meningococcal meningitis and/
conjugate vaccines are used sparingly in many resource-limited or meningococcemia exhibits high geographic variability. The
settings, and Hib meningitis remains common in children meningitis belt of sub-Saharan Africa represents a classic en-
in these areas. For example, Hib still accounted for 32% of demic area (Fig. 24.1). Although meningococcal infections were

FIGURE 24.1 The meningitis belt of sub-Saharan Africa, comprising areas of Africa that experience
repeated epidemics of serogroup A meningococcal meningitis. (From Centers for Disease Control and
Prevention. CDC Health Information for International Travel 2014. New York: Oxford University Press;
2014.)

Scheld_Ch24.indd 367 2/21/14 5:42 PM


368 Part III: Bacterial and Mycoplasmal Infections

not recorded in the area until the 1880s, large outbreaks still to the dry season. African outbreaks occur during the dry season
occur regularly. Although the precise effects of climatic condi- from December to June. Annual outbreaks in the sub-Saharan
tions on the incidence of meningitis are unresolved, the belt lies meningitis belt tend to peak in late April and early May, when the
within the 300- and 1,100-mm rainfall lines. At least 390,000 dry desert wind (harmattan) has ceased and temperatures are high
cases with 53,000 deaths occurred within the seven countries throughout the day, and terminate abruptly with the onset of the
of the belt in the 10-year period 1951 through 1960. The aver- rainy season (41). Low humidity may alter the pharyngeal muco-
age annual incidence since 1950 has been estimated as approxi- sal barrier, thereby predisposing it to infection. Although the in-
mately 70 cases per 100,000 population by the World Health troduction of a new virulent strain into a susceptible population
Organization (WHO) (41). In 1988, more than 57,000 cases may contribute to the epidemics, many other factorsincluding
of meningococcal disease were reported from the African con- crowding, the presence of other respiratory pathogens, poor hy-
tinent; in 1989, the number of cases increased to more than giene, and poorly defined environmental featurescontribute to
70,000, with more than 40,000 reported from Ethiopia (42). the initiation of a meningococcal epidemic (45).
Because reporting is often delayed and may be incomplete, this Although meningococcal meningitis may be more prevalent
figure likely underestimates the actual number of cases. More in men and boys, the reports are often skewed by the inclusion
than 180,000 cases were reported by the WHO for the regions of military recruits and chronic alcoholics. Meningitis caused
in 1996, the highest yearly total in more than 40 years. Similarly, by N. meningitidis is primarily a disease of children and young
within a 1-month period (October 1974), 4,865 patients with adults: fewer than 10% of cases occur in patients older than
meningococcal meningitis were treated at the major infectious 45 years of age. In the United States and Finland, children younger
diseases hospital in Sao Paulo, Brazil. The overall mean an- than 5 years of age account for approximately 55% of cases dur-
nual incidence of meningitis caused by N. meningitidis reached ing nonepidemic conditions, whereas in Zaria, Nigeria, the peak
370 cases per 100,000 population in the greater Sao Paulo area incidence occurs in 5- to 9-year-olds (43). Major epidemics are
in that year. The attack rate was 517 cases per 100,000 inhabit- heralded by a shift to the right toward older age-groups (i.e.,
ants during a group C epidemic in Upper Volta (now Burkina adolescents instead of children), a predictive feature of epidem-
Faso) in 1979, and recent studies documented an attack rate ics in the meningitis belt identified by prospective surveillance.
of 400 to 450 per 100,000 children up to 8 years of age in the Although meningococcal meningitis is unusual in adults, 33%
Faeroe Islands (43). In contrast, the mean annual attack rate of sporadic meningococcal disease occurred in adults in a 5-year
in the United States (1975 to 1980) was approximately 1.2 population-based study in Atlanta (46). Underlying conditions
per 100,000 persons but was, again, age dependent: 17.1 per such as congestive heart failure, multiple myeloma, and infec-
100,000 in children younger than 1 year of age, 5.2 per 100,000 tion with human immunodeficiency virus (HIV) were prevalent
in 1- to 4-year-old children, and 0.3 per 100,000 among adults. in adults older than 24 years of age with meningococcal infec-
Approximately 2,500 to 3,000 cases of meningococcal infection tion but unusual in the 18- to 24-year-old group.
were reported annually in the United States between 1984 and Large-scale epidemics caused by serogroup A meningo-
2003. In a multistate surveillance project conducted between cocci have occurred at 20- to 30-year intervals throughout the
1989 and 1991 in the United States, the average annual inci- world in the last and this century and continue at approxi-
dence of meningococcal disease was 1.1 per 100,000; 46% of mately 8- to 12-year intervals in the African meningitis belt,
cases occurred in children 2 years of age, and the highest age- where approximately 1% of the population is affected. These
specific incidence was in children younger than 4 months of age strains infrequently cause disease in the United States, but se-
(44). A similar figure of approximately 2 per 100,000 popula- rious outbreaks caused by serogroups A, B, or C continue in
tion was reported from Finland from 1976 through 1980. many areas (Table 24.1). A predominant serogroup circulating
The peak incidence of meningococcal meningitis in indus- in the African meningitis belt since 2000 is W135, an unusual
trialized nations occurs in winter through early spring in both occurrence (see later discussion). Serogroups B and C now
epidemic and endemic periods. Similar seasonal trends may also cause most focal outbreaks and endemic disease in many areas
occur in tropical areas. For example, both the group C and group (see the section Etiology later in this chapter).
A meningococcal epidemics in the Sao Paulo area from 1971 to N. meningitidis disease is exclusive to humans. No inter-
1974 began in May or June, the point of transition from the rainy mediate host, reservoir, or animal-to-human transmission has

TA B L E 2 4 . 1
ETIOLOGY OF BACTERIAL MENINGITIS IN THE NORTHERN HEMISPHERE AMONG PATIENTS AGED 16 YEARS
AND OLDER

Percentage of Total Cases

Organism United States 19621988 Iceland 19751994 Canada 19851995 United States19701998

Haemophilus influenzae 4 5 8 4
Streptococcus pneumoniae 38 20 53 48
Neisseria meningitidis 14 56 2 14
Listeria monocytogenes 11 6 25 7
Othera 20 12 27
Unknown 13 8

a
Includes nonpneumococcal streptococci, gram-negative bacilli, enterococci, Staphylococcus aureus, anaerobes, and diphtheroids.
Adapted from refs. 69, 477, and 478.

Scheld_Ch24.indd 368 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 369

been proved. The nasopharynx is the natural reservoir for causes of neonatal sepsis and meningitis. The prevalence of
meningococci; transmission is facilitated by airborne drop- antimeningococcal capsular antibodies is lowest between
lets or close contact. The definition of close contact has not 6 and 24 months of age, increasing to approximately 70%
been clearly elucidated but generally refers to persons who by early adulthood. The inverse link between occurrence of
have had prolonged (8 hours or longer) contact while in close invasive disease and bactericidal antibody was first docu-
proximity (3 feet or less to the patient) or direct exposure to mented during an outbreak of serogroup C meningococcal
the patients oral secretions (through kissing, mouth-to-mouth meningitis among army recruits in 1968 (48). In this study, the
resuscitation, endotracheal intubation, or endotracheal tube sera of only 5.6% of the recruits who developed meningococ-
management) within 1 week before the onset of the patients cal disease had bactericidal activity before the onset of illness,
symptoms until 24 hours after initiation of appropriate an- compared with 82.2% of control sera. Notably, 5 (38.5%) of
timicrobial therapy (47). Meningococcal colonization may 13 recruits without bactericidal antibody developed systemic
result in an asymptomatic carrier state (which is most com- illness after colonization with the group C strain.
mon) or in endemic, hyperendemic (e.g., a meningitis belt Although recovery from invasive meningococcal disease
between epidemics at 10 to 50 cases per 100,000 per year), generally confers lifelong immunity against the homolo-
or epidemic disease. Although there is no clear relationship gous serogroup, this is not the major immunizing process.
between carriage rate and overt disease, the development of Nasopharyngeal colonization, particularly with serogroups B,
the carrier state and the host immune response are, as for Hib C, or Y, may elicit the development of bactericidal activity, pri-
meningitis, important variables in the epidemiology of menin- marily directed against the colonizing strain but also against
gococcal infections. heterologous organisms within 5 to 12 days of acquisition.
Approximately 6% of the population develops nasopharyn- Colonization with nongroupable meningococci or Neisseria
geal colonization with N. meningitidis yearly. Nasopharyngeal lactamica may elicit protective immunity, especially in young
carriage rates vary with age and the population under study. children. N. lactamica is virtually nonpathogenic, but nasopha-
The carriage rate is markedly influenced by age: 0.5% to 1% ryngeal carriage rates of this organism are highest (4% to 20%)
in children 3 to 48 months old, approximately 5% in adoles- in children between 3 months and 12 years of age, whereas the
cents 14 to 17 years old, and 20% to 40% in young adults. age-adjusted carriage rates for N. meningitidis are only 0.5%
Analogous to Hib, carriage rates are higher in close contacts to 2%. As with Hib, the carriage rates of pathogenic menin-
of an index case. Carriage rates of meningococci of approxi- gococci are too low in children to account for antibody for-
mately 40% have been documented in close family contacts mation, and the importance of other cross-reacting organisms
of meningococcal cases. In closed populations (e.g., military has also been proposed: Bacillus pumilus for group A poly-
barracks during early training), carriage rates of 20% to 60% saccharide and E. coli for group C organisms. Paradoxically,
are commonplace and may reach 90% during epidemics of me- an exuberant IgA response to meningococci may actually
ningococcal disease. Nasopharyngeal carriage usually persists enhance the development of systemic disease. When a large
for weeks to months, similar to Hib carriage. Spread of menin- proportion of induced anticapsular antibodies are of the IgA
gococcal disease is usually carrier mediated (i.e., not spread by class, complement-mediated immune bacteriolysis by IgM is
case-to-case contacts) and largely accounts for the increased blocked, thus enhancing susceptibility to invasive disease. This
risk for disease (500- to 1,000-fold above the background en- peculiar immunologic phenomenon is transient, lasting only a
demic rate) in household contacts of an index case. The organ- few days following asymptomatic nasopharyngeal acquisition
ism is often introduced into the home environment by an adult of N. meningitidis or closely related organisms.
family member, with subsequent transmission to others; infants In addition to antibody, an intact complement system is
are colonized last of all. Although uncharacterized host and also a component of host defense against invasive meningo-
environmental factors contribute to containment of infection coccal disease. Studies of extreme phenotypes have identified
to the nasopharynx (thereby preventing disseminated disease), genetic correlates of increased susceptibility in the complement
host immunity also plays an important role. Nevertheless, system (49). Recurrent or chronic neisserial infections have
those individuals most recently colonized with meningococci been associated with rare isolated deficiencies of late comple-
appear to be at the greatest risk for invasive disease. Secondary ment components (C5, C6, C7, or C8, and perhaps C9), occa-
systemic meningococcal disease often develops within 5 days sionally in concert with failure to produce antimeningococcal
of recognition of the index case, with 70% to 80% of second- antibodies. Recurrent episodes of neisserial infections may
ary cases occurring within 14 days of the primary case. occur in these patients without an increase in susceptibility
As with Hib disease, the age-specific incidence of menin- to other pathogens (50), and screening for complement de-
gococcal infection is inversely proportional to the presence of fects is useful in patients with these syndromes. Complement
serum bactericidal antibodies against serogroups A, B, and deficiency also appears to predispose to meningitis caused by
C. More than 50% of infants possess bactericidal antibody nongroupable meningococci and Neisseria-related bacteria
at birth as a result of transplacental transfer. The specifics of (i.e., Moraxella and Acinetobacter species) (51). In addition,
the antibody response may be responsible for the occurrence complement deficiency or depletion of early components (C1,
of meningococcal meningitis during the neonatal period. The C3, or C4) because of an underlying disease such as nephrotic
group B capsular polysaccharide is a polymer consisting of two syndrome, hepatic failure, systemic lupus erythematosus, pres-
to eight linked sialic acid residues but is immunologically iden- ence of C3 nephritic factor, or multiple myeloma may predis-
tical to the oligosaccharides of several human glycoproteins, pose to the first episode of invasive meningococcal disease.
including brain gangliosides. Immunologic tolerance thus ex- An association between homozygous C4b deficiency (present
ists in this age-group; although IgM antibody can be induced, in approximately 3% of the population) and the develop-
the usual switch to IgG antibody production does not occur ment of childhood meningitis was demonstrated. Up to 30%
(43). Because IgM does not cross the placenta, IgG antibody of patients with invasive meningococcal syndromes display
to the serogroup B polysaccharide is lacking in neonates, con- decreased complement function. Properdin deficiency, or dys-
tributing to the occurrence of group B meningococcal disease function with normal concentrations, also predisposes to me-
in this patient population. In addition, group B meningococcal ningococcal infections; this defect is reversible by vaccination
capsular antigen is identical to the capsular polysaccharides (52). The mortality associated with meningococcal meningitis
of E. coli K1 and certain types of group B streptococci, major in patients with complement component deficiencies is actually

Scheld_Ch24.indd 369 2/21/14 5:42 PM


370 Part III: Bacterial and Mycoplasmal Infections

lower than the general population (3% versus 19%, respec- deficiency or functional or anatomic asplenia and for adoles-
tively). However, since invasive meningococcal infection oc- cents with HIV infection. A two-dose primary series is also
curs in 39% of patients with deficiency of terminal complement recommended for children 9 to 23 months of age at increased
components and about 6% of those with properdin deficiency, risk for invasive meningococcal disease (57). Lack of a vac-
a screening test (e.g., CH50) has been suggested (53) for all cine against serogroup B meningococcus is a significant issue,
individuals with serious meningococcal disease, with further although recent investigations of a multicomponent serogroup
consideration for determination of specific complement and/ B vaccine (4CMenB) demonstrated that this vaccine was im-
or properdin concentrations if documented. Asplenic states munogenic (5861); 84% to 100% of infants who were ad-
increase the risk for serious infections by encapsulated organ- ministered the vaccine developed bactericidal antibodies.
isms, especially Hib or S. pneumoniae but also meningococci.
In genetic casecontrol studies, invasive meningococcal dis-
ease was associated with the IL1RA  2018T C polymor- Streptococcus pneumoniae
phism, with susceptibility increased in homozygous IL1RA 
2018C carriers (49). CFH, SFTPA2, CEACAM3, and Although pneumococcal meningitis occurs in all age-groups,
CEACAM6 polymorphisms were significantly associated with pneumococci remain the most common cause of bacterial
an increased susceptibility to meningococcal disease, whereas meningitis in adults. The annual incidence of pneumococcal
other CEACAM6 and SFTPA2 polymorphisms showed a pro- meningitis has remained relatively stable in the United States
tective effect (49). A genome-wide association study identified for the past three decades until recently with introduction of
variants in the CFH region associated with host susceptibility the heptavalent pneumococcal conjugate vaccine (see later
to meningococcal disease (54). Further studies are needed to discussion). In seven studies analyzing data from diverse geo-
confirm these associations before a definite conclusion can be graphic areas in this country from 1959 to 1978, the annual
drawn on their role because of the limited sample size and lack incidence was 0.3 to 2.3 per 100,000 population, with a mean
of correction for multiple testing. of 1.3 per 100,000. An identical infection rate of 1.3 per
Although all the aforementioned factors (particularly 100,000 persons annually was recorded from the Oklahoma
recent colonization with a pathogenic strain in a nonimmune City area in 1984 and extrapolated from several states and Los
host) undoubtedly contribute to the pathogenesis of meningo- Angeles County in a relatively recent (1995) national survey
coccal disease, the precise determinants contributing to overt conducted by the Centers for Disease Control and Prevention
clinical illness (as opposed to the usual outcome of asymp- (CDC) (62,63); higher rates of invasive pneumococcal disease
tomatic carriage) are poorly defined. Even during epidemics, were reported at the extremes of age (see later discussion),
only 1 in 1,000 to 5,000 colonized patients develop disease in men and boys, and among blacks and American Indians
(43). Various predisposing factors, including crowding, lower as compared with whites. Nearly identical incidence figures
socioeconomic status, and poor general health, have been pro- (1.2 to 1.4/100,000 population annually) were reported from
posed to explain the increased incidence among blacks in the the Gteborg, Sweden area from 1975 through 1980 (64)
United States and among alcoholics in Finland or Alaska (41). and from rebro county in Sweden (1.0/100,000 popula-
However, the influence of such conditions (e.g., overcrowding) tion annually) from 1981 through 1992 (65). Pneumococcal
has not been supported by studies in Nigeria. An antecedent meningitis was, again, more common in men and boys, and
viral infection has been suggested as another predisposing most cases occurred from December through May. However,
factor, because approximately one third of meningococcal higher incidence rates have been reported from other areas.
cases follow symptoms referable to the upper respiratory tract. For example, surveillance studies from 1980 to 1986 among
An outbreak of meningococcal disease followed a large influ- the Alaskan native population in the Yukon-Kusko-Kurin delta
enza epidemic in Texas in 1981, and simultaneous outbreaks region of southwestern Alaska documented an extremely high
of meningococcal and influenza A2 infections have been frequency of invasive pneumococcal disease; the annual rate
described in institutional settings. Although meningococcal for pneumococcal meningitis was 13.2 per 100,000 persons
pneumonia may complicate influenza (e.g., the 1918 to 1919 overall (66). Perhaps more importantly, the annual incidence
pandemic), the role of viral infections in the enhancement of rate was 216 per 100,000 children younger than 2 years
meningococcal dissemination is unproved (22). 18 times higher than that reported from Sweden (64) and 36-
The time from nasopharyngeal acquisition to bloodstream to 37-fold greater than United States rates derived from both
invasion is short (usually approximately 10 days). The incu- passive and active surveillance (7,62,66). These rates of bac-
bation period may also be short, because secondary cases teriologically confirmed invasive pneumococcal disease were
commonly occur within 1 to 4 days of the index case. Once the highest reported for any population worldwide. Although
the organism is bloodborne, more than 90% of meningococcal most cases of invasive pneumococcal disease occurred during
disease is manifested as meningitis and/or meningococcemia. the Arctic summer, pneumococcal meningitis cases clustered in
Although a single case of meningococcemia or meningitis in the winter (66), as described in other reports (7).
a college student engenders alarm, invasive disease due to this The risk for pneumococcal meningitis is age dependent,
organism is no more common in such students when compared with increased incidence rates occurring at the extremes of age.
with nonstudent age-matched 18- to 22-year-old controls but For example, the number of cases per 100,000 persons per year
is increased threefold in freshmen living in dormitories, a tar- in the Gteborg, Sweden area for 1970 through 1980 were as
get of some vaccine recommendations. Active or passive smok- follows: 12.0 for infants younger than 12 months old, 0.4 to
ing and binge alcohol consumption also appear to increase the 0.9 for children and adults 2 to 39 years old, 1.2 to 1.6 for per-
risk of meningococcal disease in this age-group. The first quad- sons 40 to 70 years old, and 2.2 for those older than 70 years
rivalent meningococcal conjugate vaccine against serogroups of age (64). The dramatic incidence among Alaskan native
A, C, Y, and W135 was licensed in January 2005 in the United children younger than 2 years of age (216/100,000 annually)
States for routine use, starting at age 11 to 12 years; recent are noted earlier in this chapter. Similarly, the annual incidence
recommendations are for one vaccine in this age-group, with rates for all invasive pneumococcal infections (including bac-
a booster dose given at age 16 years (55,56). A two-dose pri- teremic pneumonia) in the Oklahoma City area in 1984 were
mary series administered 2 months apart is recommended for 97 per 100,000 for infants younger than 1 year of age and 87
those age 2 to 54 years with persistent complement component per 100,000 for elderly adults older than 80 years of age.

Scheld_Ch24.indd 370 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 371

As with disease caused by the other major meningeal patho- in approximately 20% to 35% of patients with this disease.
gens, pneumococcal meningitis follows recent nasopharyngeal Pneumococci are the most common cause of meningitis in chil-
colonization by a virulent strain. The rate of asymptomatic car- dren with sickle cell anemia and commonly cause meningitis
riage varies with age, environment, geographic locale, and the in other asplenic states or in the setting of primary or acquired
presence of an upper respiratory tract infection. Pneumococci immunodeficiencies (71). S. pneumoniae causes 87% of pyo-
have been isolated from the upper respiratory tract of 5% to genic meningitis cases in sickle cell disease; cases in the 2- to
70% of normal adults; approximately 25% acquire a new 3-year-old group occur at a rate of 12 per 1,000 patient-years.
strain annually. Carriage rates decline with age (30% to 35% The risk for pneumococcal meningitis in this age-group of chil-
for children ages 6 to 11 years and 18% to 19% in adults) and dren with sickle cell anemia is increased 36-fold as compared
are higher in closed populations (e.g., 27% to 58% in schools with that observed in control groups of black children, and it is
and orphanages, 50% to 60% in closed military populations). increased 314-fold over that observed in whites. Pneumococcal
The duration of pneumococcal carriage varies from weeks to meningitis also occurs with increased frequency in persons with
months and is longer in children than in adults. Most carrier the Wiskott-Aldrich syndrome, thalassemia major, childhood
strains in the normal population are of higher numbered cap- nephrotic syndrome, multiple myeloma, and chronic lympho-
sular types and only infrequently are associated with invasive cytic leukemia. Defects in immunoglobulin concentration or
disease (see later discussion). Carriage is prolonged in indi- function, as well as poor alternative complement pathway
viduals with low serum antibody concentrations against the mediated opsonization of pneumococci, are common in many
homologous capsular type before colonization. Spread of this of these disorders. Pneumococcal meningitis is approximately
organism within the family unit is influenced by crowding, the 450-fold more common in patients with acquired immuno-
season (greater in fall and winter), and the presence of pneu- deficiency syndrome (AIDS) when compared with the general
mococcal disease (particularly pneumonia and otitis media). population (71), often despite prophylaxis with trimethoprim-
The precise relationship between pneumococcal carriage and sulfamethoxazole (TMP-SMX) to prevent other opportunistic
the development of protective immunity is poorly defined. infections. In one study of 352 episodes of community-acquired
More than 50% of children develop a rise in type-specific pneumococcal meningitis in adults, 245 (70%) were associ-
antibody concentrations following colonization; this is rarely ated with an underlying disorder and the overall in-hospital
observed in adults, perhaps because of the relatively high an- mortality rate was 30% (72); death in patients younger than
tibody concentrations already present in adults. Nevertheless, 60 years of age was more often caused by neurologic compli-
otitis media often occurs in colonized infants despite the pres- cations; in patients 60 years or older, death was more likely
ence of type-specific antibodies. Antibody concentrations gen- secondary to systemic complications. In children with cochlear
erally decline with time despite persistent carriage of a given implants with positioners who are beyond 24 months after im-
strain (67). In addition, antibody responses to different capsu- plantation, the incidence of bacterial meningitis was 450 cases
lar types vary considerably and are generally poor in infants per 100,000 person-years compared with 0 cases in children
younger than 2 years of age. Specific antibody responses tend without positioners (73); the updated overall incidence was
to be higher after intermittent periods of nasopharyngeal car- 189 cases per 100,000 person-years, with most cases caused
riage than after continuous ones. The antibody response after by S. pneumoniae (74). Outbreaks of pneumococcal meningi-
pneumococcal colonization, its influence on subsequent dis- tis have also been described during African outbreaks of me-
ease, and the impact of other environmental antigens require ningococcal meningitis (75). In children who develop second
further study. episodes of pneumococcal meningitis, screening for congenital
Studies of extreme phenotypes among patients with inva- immunoglobulin deficiencies should be performed (53).
sive pneumococcal disease have identified genetic correlates The rates of pneumococcal meningitis have decreased
of increased susceptibility in the complement system and the in children and adults since introduction of the heptavalent
signalling cascade after toll-like receptor pathways (49). Case pneumococcal conjugate vaccine (from 1.13 to 0.79 cases
control studies showed that invasive pneumococcal disease was per 100,000 between 1998 to 1999 and 2004 to 2005), al-
associated with certain MBL and C3-variant genotypes (49,68). though the increase in disease caused by serotypes not in the
Several factors predispose to pneumococcal meningitis vaccine (19A, 22F, and 35B) is concerning (76). The Advisory
(69,70): Committee on Immunization Practices now recommends use
of the 13-valent pneumococcal conjugate vaccine (77), which
Pneumonia (15% to 25% of patients)
not only offers protection against the serotypes in the heptava-
Otitis media
lent vaccine (4, 6B, 9V, 14, 18C, 19F, 23F) but also protection
Sinusitis
against additional serotypes (serotypes 1, 3, 5, 6A, 7F, 19A).
CSF fistulas, leak
Head injury
Cochlear implants with positioners
Alcoholism, cirrhosis ETIOLOGY
Sickle cell disease, thalassemia major
In a review of 296 episodes of community-acquired meningitis
Other splenic disease
in adults reported from the Massachusetts General Hospital
Wiskott-Aldrich syndrome
from 1962 through 1988, the most common pathogens were
Multiple myeloma
S. pneumoniae (37%), N. meningitidis (13%), and L. mono-
Pneumonia coexists much more commonly with pneumo- cytogenes (10%) (78).
coccal meningitis than with the other two major pathogens. The epidemiology of bacterial meningitis changed dramati-
Acute otitis media is seen in approximately 30% of patients cally in areas that have embraced Hib conjugate vaccines starting
with pneumococcal meningitis; acute sinusitis may also be an in the late 1980s. The influence of these vaccines from the four
important antecedent event. Pneumococci are the most com- largest and most recent surveys conducted in the United States
mon cause of recurrent meningitis in the setting of CSF leaks. (7,62,79) is shown in Table 24.2. Hib was responsible for 45%
Recent or remote head trauma is found in about 10% of pa- to 48% of cases from 1978 through 1986 but only 7% of cases
tients with pneumococcal meningitis. Alcoholism, cirrhosis, in a surveillance study (62) conducted in 1995 in laboratories
and spontaneous bacterial peritonitis are underlying disorders serving all acute-care hospitals in 22 counties from four states

Scheld_Ch24.indd 371 2/21/14 5:42 PM


372 Part III: Bacterial and Mycoplasmal Infections

TA B L E 2 4 . 2
Haemophilus influenzae
ETIOLOGY OF BACTERIAL MENINGITIS IN THE
UNITED STATES, 19781995 Haemophilus species are small, gram-negative, pleomorphic
coccobacilli. They are facultative anaerobes that grow best an-
Percentage of Total Cases aerobically with 5% to 10% CO2 on blood-enriched media.
H. influenzae requires both X factor (hematin) and V factor
19781981 1986 1995 20062007 (nicotinamide adenine dinucleotide, nicotinamide adenine dinu-
Organism (7) (79) (62) (5) cleotide phosphate, or nicotinamide nucleoside) for growth under
aerobic conditions and is nonhemolytic. Chocolate agar is most
Haemophilus 45 48 7 7 commonly employed for the initial isolation of H. influenzae.
influenzae H. influenzae strains are either encapsulated (typeable) or un-
Streptococcus 13 18 47 58 encapsulated (nontypeable). Encapsulated strains are classified
pneumoniae into six types, designated a through f, according to specific reac-
Neisseria 20 14 25 14
tions with antisera directed against epitopes on capsular antigens.
meningitidis
Methods for grouping include counterimmunoelectrophoresis,
latex particle agglutination of culture supernatants, immunofluo-
Streptococcus 3 6 12 18 rescence, and the production of halos surrounding colonies on
agalactiae media containing antisera. Nearly all invasive H. influenzae infec-
Listeria 2 3 6 3 tions are caused by serotype b (Hib). The capsule is a repeating
monocytogenes polymer of PRP, an important virulence determinant of this or-
Other 8 14 ganism (see later discussion). Hib also contains a lipooligosaccha-
ride (here, designated lipopolysaccharide [LPS] for convenience)
Unknown 6
in the outer membrane, an additional virulence determinant. Hib
strains have been further classified into subtypes based on electro-
phoretic mobility differences among Omps. Although the patho-
(10 million population). The incidence of Hib meningitis de- genic role of these Omps is uncertain, subtype analysis is useful
clined from 2.9 to 0.2 per 100,000 population from 1986 to for epidemiologic studies. For example, in a survey of 256 in-
1995, respectively. The median age of patients with bacterial vasive isolates from 22 states representing a variety of clinical
meningitis rose from 15 months to 25 years during this inter- settings, about 70% of cases were caused by strains of three sub-
val, perhaps the most dramatic change in the epidemiology of types (1H, 2L, and 3L) among the 21 Omp subtypes identified. In
any bacterial infection in recent decades. As a result, approxi- contrast, 84% of 80 invasive isolates studied in the Netherlands
mately 70% of bacterial meningitis in the United States is due had the same Omp subtype pattern (type 1, identical to subtype
to pneumococci or meningococci. In the early pivotal trial in 3L in the Granoff classification system), and no strains of subtype
California, invasive (e.g., bacteremia and meningitis) pneumo- 1H, 1L, or 2H were found.
coccal disease was virtually eliminated in infants vaccinated It has been recognized, largely on the basis of multilocus en-
with the heptavalent conjugate vaccine at 2, 4, and 6 months of zyme electrophoretic analysis by Musser et al. (80), that the natu-
age. Although targeted to children, recent data support a herd ral populations of Hib from widely divergent geographic areas are
immunity phenomenon, similar to that experienced with Hib clonal as a consequence of infrequent recombination of chromo-
conjugate vaccines, leading to a decline in pneumococcal bac- somal genes. For example, 32 distinct multilocus enzyme geno-
teremia and/or meningitis in the population (including adults) types, referred to as electrophoretic types (ETs), were apparent
following introduction of conjugate pneumococcal vaccine in among 177 U.S. isolates by analysis of 16 metabolic enzymes, but
infancy. In another surveillance study among residents in eight 73% of invasive disease episodes were caused by strains belonging
surveillance areas representing 17.4 million persons from 1998 to only three ETs. In the largest and most comprehensive analy-
to 2007, the impact of the heptavalent pneumococcal conju- sis (80), 2,209 encapsulated H. influenzae strains (including 1975
gate vaccine was appreciated in which the incidence of men- Hib) from 30 countries on six continents collected over a 40-year
ingitis caused by vaccine serotypes decreased from 0.61 cases period were studied by multilocus electrophoresis of 17 chromo-
per 100,000 population in 1998 to 1999 to 0.05 cases per somally encoded metabolic enzymes, Omp subtyping, and the
100,000 population in 2006 to 2007, although the number of pattern of restriction fragment length polymorphism in the cap
cases of bacterial meningitis caused by nonvaccine serotypes region (the chromosome region responsible for capsular expres-
increased by 61% (5); the mean age of all patients with menin- sion). On the basis of allele profiles at the enzyme loci, 280 distinct
gitis increased from 30.3 years in 1998 to 1999 to 41.9 years ETs in two phylogenetic divisions were identified: the population
in 2006 to 2007. However, despite the declining incidence of structure is definitely clonal. Currently, nearly all invasive disease
bacterial meningitis in the United States, the overall case-fatality worldwide is caused by nine clones of Hib. One genetically distinct
rates did not change significantly (15.7% in 1998 to 1999 com- clone complex occurs with considerable frequency worldwide,
pared with 14.3% in 2006 to 2007; p  .50). but marked geographic variation occurs for other clones or clone
As shown in Table 24.1, the responsible organisms for families. Based on an extensive analysis, it appears that this distri-
community-acquired bacterial meningitis in adults older than bution of clones on an intercontinental scale is largely accounted
16 years of age are somewhat different, but S. pneumoniae, for by the patterns of racial/ethnic composition and historical de-
N. meningitidis, and L. monocytogenes predominate. Similar mographic movements of the human host populations (80).
recent (e.g., after 2000) surveys for resource-limited settings
are lacking and sorely needed to guide vaccine strategies in
this age-group. Neisseria meningitidis
In the following sections, each of the major meningeal
pathogens is considered, including relevant classification Neisseria species are nonspore-forming, nonmotile, oxidase-
schemes. In addition, the potential virulence characteristics of positive, gram-negative cocci (measuring approximately 0.8 
each organism are briefly discussed. 0.6 m) that usually appear as biscuit- or kidney-shaped

Scheld_Ch24.indd 372 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 373

diplococci on smears of infected fluids. Because other organ- with an increase in both the incidence and the mortality of
isms (e.g., Moraxella species) are similar morphologically, meningococcal disease in Canada (82). This and four other
identification rests on biochemical or immunologic tech- closely related clonal strains were also implicated in a marked
niques. All Neisseria species are oxidase positive, and sugar increase in outbreaks of serogroup C meningococcal disease in
fermentation reactions are usually sufficient for speciation the United States (83). These studies suggested the emergence of
within the genus. Gonococci ferment glucose (but not malt- a clonal group of virulent serogroup C meningococcal strains
ose or lactose) to acid, whereas meningococci ferment both in North America, leading to an increase in the rate of menin-
glucose and maltose to acid. N. lactamica, a related organ- gococcal disease in some regions, an increased number of out-
ism occasionally present in throat cultures, ferments glucose, breaks, and a higher case-fatality rate (84).
maltose, and lactose. Maltose-negative variants of meningo- In an analysis from the United States (85), part of an active
cocci have been isolated; these strains may be differentiated and ongoing laboratory-based population-based surveillance for
from gonococci by fluorescent antibody tests, coagglutination meningococcal infection from 1992 to 1996, the serogroup dis-
tests, or electrophoretic analysis of hexokinase isoenzymes. tribution for the three most common isolates was as follows:
These strains usually acquire the ability to ferment maltose C, 35%; B, 32%; and Y, 26%. Similar results were noted in
on subculture; true maltose-negative variants are rare, but New York City through 2000 (86). In contrast, serogroup B
this property is genetically linked to sulfadiazine resistance. accounted for 75% of meningococcal isolates in Italy (87).
Meningococci grow rapidly on blood, chocolate, gonococcal, More than 98% of cases of invasive meningococcal disease
or enriched Mueller-Hinton agar in a moist, 3% to 10% CO2 are sporadic (88). From 1998 to 2007, a total of 2,262 cases
environment at 35C to 37C. A modified Thayer-Martin me- of meningococcal disease were reported to the Active Bacterial
dium is employed for meningococcal isolation from contami- Core surveillance sites, with an annual incidence of 0.53 cases
nated sites, as in detection of the carrier state. The transoral per 100,000 population (89). The incidence decreased from
approach for obtaining specimens is more practical and is at 0.92 cases per 100,000 population in 1998 to 0.33 cases per
least as sensitive as older transnasal approaches for carrier 100,000 population in 2007; the incidence decreased to this
detection. Because this organism is susceptible to drying and historic low before the introduction of the quadrivalent menin-
chilling, all specimens should be inoculated promptly. gococcal conjugate vaccine. Infants younger than 1 year of age
As in other gram-negative bacteria, the ultrastructural char- had the highest incidence (5.38 cases per 100,000 population),
acteristics of meningococci are complex. The surface compo- although the distribution of serogroups is also different in this
nents include capsular polysaccharide, fimbriae or pili, LPS, and population (serogroup B, 3.08 per 100,000 population; sero-
Omp; several of these structures are important virulence deter- group C, 0.53 per 100,000 population; serogroup Y, 1.50 per
minants. Meningococci are classified by serogroups based on 100,000 population). A recent outbreak of serogroup C disease
structural differences among capsular polysaccharides and ag- was reported in New York City among men who have sex with
glutination reactions with specific antisera, and they are further men (90). During the outbreak of meningococcal disease coin-
defined by serotypes based on analysis of the Omp. Capsular ciding with the Hajj pilgrimage in March 2000, the attack rate
polysaccharide detected by positive agglutination with refer- of W135 disease was 25 cases per 100,000 pilgrims (91); all out-
ence antisera is uniformly present among invasive isolates, but break-associated isolates of serogroup W135 were members of a
20% to 50% or more of carrier strains are unencapsulated single clone of the hypervirulent ET-37 complex, which occurred
(nongroupable). The serogroups have important epidemiologic as the result of expansion of a clone that had been in circulation
and prevention-related implications. Thirteen serogroups are since 1970 (92). A high incidence of serogroup X cases was re-
recognized (43), designated as follows: A, B, C, D, H, I, K, ported in Niger (93), representing 51% of 1,139 confirmed cases
L, X, Y, Z, 29E, and W135. Most meningococcal disease is of meningococcal meningitis in 2006; serogroup X disease also
caused by organisms in serogroups A, B, C, and Y, although emerged in Togo and Burkina Faso during 2006 to 2010 (94).
the proportion of cases caused by serogroup W135 is increas- Serotypes within a serogroup of N. meningitidis are clas-
ing. Although serious outbreaks of serogroup A, B, or C disease sified largely on analysis of Omp profiles in the cell enve-
have occurred worldwide, most focal outbreaks and endemic lope. At least 20 serotypes are recognized (95), resulting in
disease in many countries are caused by serogroups B and C. a classification scheme that is useful in epidemiologic studies.
For example, in the United States from 1975 to 1980, the dis- Physicochemical characterization of the Omps, which might
tribution of serogroups among 12,980 cases was as follows: be candidate antigens for vaccines, has led to the designation
B, 56%; C, 19%; Y, 11%; W135, 10%; and A, 3%. Similar fig- of five major classes. Class 2 and 3 proteins are the major po-
ures were reported for the period 1978 to 1981 (7). Serogroup rins responsible for aqueous channels in the outer membrane.
W135 disease, especially among adults, has been increasing Class 5 proteins are surface exposed and may have a role in
in the United States and apparently elsewhere (e.g., Senegal) virulence, but the function of class 1 and 4 proteins is poorly
since 1981. Group B organisms, especially B:15:P1.16, have defined. The serotype designation has important epidemiologic
emerged as important pathogens in northern Europe, caus- uses and may identify virulence characteristics among menin-
ing serious local outbreaks peaking in the 10- to 20-year-old gococci. For example, serotype 2 (2a, 2b) strains are respon-
group. The continued high prevalence of serogroup B menin- sible for approximately 50% of serogroup B disease (in which
gococcal disease has important implications because of the lack 15 serotypes have been described), followed by serotypes 15
of an effective, widely available vaccine against this serogroup. and 9; in contrast, serotypes 4 and 6 have been isolated only
An outbreak of serogroup C disease occurred in California in from carriers. Serotype 2 is also responsible for about 80% of
1987 (81). Serious outbreaks caused by serogroup A continue invasive serogroup C disease and is an important marker for
in Nepal, Saudi Arabia, Chad, Sudan, Burkina Faso, and else- serogroups Y and W135 pathogenic strains as well (95). All
where. Approximately 40,000 cases of serogroup A disease serogroup A meningococci, in contrast, are homogeneous with
occurred in Ethiopia in the spring of 1989 (Fig. 24.1). respect to Omp and show no homology to serotypes within
There have been increases in the incidence of serogroup other serogroups. Serotype analysis has also been linked to
C disease in North America to equal or surpass the incidence of certain clinical characteristics (96). However, this technique
disease caused by serogroup B (42). From 1985 through 1992, a is available only in research or reference laboratories. In addi-
clonal serogroup C meningococcal strain (designated ET-15 and tion, at least eight immunotypes of meningococci, classified by
defined by multilocus enzyme electrophoresis) was associated differences among LPS subtypes, are known to exist and may

Scheld_Ch24.indd 373 2/21/14 5:42 PM


374 Part III: Bacterial and Mycoplasmal Infections

play a role in pathogenesis and disease expression. LPS im- virulence, probably because of its antiphagocytic properties
munotype analysis may also be useful in the further character- that allow the organism to escape host phagocytic clearance
ization of the epidemiology of meningococcal disease (41,43). mechanisms within the bloodstream and/or CSF. Pili are pro-
The recent availability of monoclonal antibodies to detect tein surface appendages composed of identical pilin-repeating
variations in Omp and/or LPS will improve the resolution of subunits. Pili from meningococci and gonococci are morpho-
these typing systems for epidemiologic analysis. logically and chemically similar. Cross-reacting antibodies bind
In addition to classification by serogroup, serotype, and to a short peptide sequence (residues 69 to 94) of gonococcal
LPS subtype, many reports have focused on multilocus enzyme pili that is essential for receptor-binding function. Fresh menin-
electrophoresis for the characterization of the chromosomal gococcal isolates from carriers and cases contain 7 to 40 pili
genome of isolates and for estimation of the genetic relatedness per diplococcus. Pili are important mediators of meningococ-
among strains (97). These studies, similar to the analysis of cal adhesion to human nonciliated columnar nasopharyngeal
Hib (80), have identified the clonal nature of N. meningitidis epithelial cells, an important early step in the development of
and have been useful for epidemiologic purposes. For example, the carrier state. Extracellular proteases that cleave the IgA1
electrophoretic variation (defined as 17% genetic distance heavy chain in the hinge region are elaborated by pathogenic
between isolates) in seven metabolic enzymes and two Omp in Neisseria species (i.e., meningococci and gonococci). Although
423 isolates of serogroup A strains recovered from 23 epidem- the role of IgA proteases in the pathogenesis of disease is con-
ics or outbreaks occurring in 38 countries on six continents troversial, these enzymes are produced by only a few organisms
over a 70-year period since 1915 identified 21 clones (des- (e.g., N. meningitidis, Neisseria gonorrhoeae, H. influenzae,
ignated A I-1 through A IV-4) containing 34 ETs (97). This S. pneumoniae, and Streptococcus sanguinis), many of which
technique has been useful for delineating similarities and dif- are important meningeal pathogens and may promote invasion
ferences among isolates from cases and carriers before, during, at the pharyngeal portal of entry. Meningococcal LPS resem-
and after epidemics (98). It has also been useful for tracing bles H. influenzae LPS by a lack of the O-antigenic polysac-
movements of epidemic strains geographically over time (81). charide side chains found in enteric bacilli despite a smooth
For example, following an epidemic in Nepal (1983 to 1984), phenotype and proven virulence. LPS is clearly important in
a single clonal complex of serogroup A meningococci (III-1, the genesis of an array of the clinical manifestations of menin-
representing 11 closely related ETs) was introduced into Saudi gococcemia and/or meningitis (see later discussion). Although
Arabia in 1987 by Muslim pilgrims traveling to Mecca for the the specific role for Omp in meningococcal virulence is unclear,
annual hajj (98). The same strain was then introduced into these organisms release substantial amounts of cell surface ma-
sub-Saharan Africa following the hajj, causing an explosive terial (in the form of outer membrane vesicles containing Omp
outbreak in Chad in 1988. An analysis of 109 isolates of se- and LPS) during growth in vitro and in vivo in the absence of
rogroup B meningococci in Norway has also revealed differ- cell lysis, a process exacerbated by antimicrobial agents. These
ences among carriers and cases (99). Although 78 ETs were outer membrane vesicles, or blebs, represent relevant vehicles
identified, 91% of the cases of systemic disease in 1984 were for central nervous system (CNS) tissue damage during menin-
caused by strains from the ET-5ET-37 complex, whereas gococcal infection (see later discussion). Tissue invasion may
these isolates were recovered from only 7% and 9%, respec- also be facilitated by the ability of meningococci to obtain iron
tively, of healthy carriers. The most common clonal complex from transferrin.
found among carriers was never isolated from patients with
invasive disease, suggesting a low virulence potential for these
clones. Two clones, ET-15 and ET-508, have been associated
with outbreaks of meningococcal infection in North America Streptococcus pneumoniae
(82,83,100). Clonal analysis will undoubtedly continue to
Pneumococci are nonspore-forming, nonmotile, small (ap-
contribute important information on the epidemiology of me-
proximately 0.8 m), gram-positive streptococci that typically
ningococcal infection, and it may prove useful in an analysis
appear as lancet-shaped diplococci with the tapered ends in
of virulence properties. As noted earlier, serogroup W135 has
juxtaposition in clinical specimens. They tend to associate in
been circulating, particularly among pilgrims to Mecca during
pairs rather than in short chains, although the latter morphol-
the hajj since 2000 in Africa, the Middle East, and worldwide
ogy is facilitated by broth culture. Pneumococci are facultative
(attack rate 25/100,000); all isolates were members of a single
anaerobes that flourish in a variety of supplemented artificial
hypervirulent ET-37 complex (91,92). This occurred through
media. Optimal growth occurs in various media supplemented
the expansion of a clone that had been circulating since 1970.
with serum or blood and glucose, at a pH level of 7.8 and a
Vaccination with quadrivalent vaccine in hajj pilgrims from
temperature of 37C in an enriched CO2 environment. The
several areas (e.g., North America, Western Europe) appears
organisms are catalase negative and relatively fastidious.
to have reduced transmission of W135 meningococci to close
Colonies on blood agar are initially dome shaped, but they
contacts upon return (101,102).
become umbilicated with time as a result of the activity of au-
The putative meningococcal virulence characteristics are as
tolytic enzymes (l-alaninemuramylamidase). Pneumococci are
follows:
-hemolytic (i.e., a greenish discoloration surrounds colonies
Capsular polysaccharide on blood agar), although
-hemolysis occurs under anaero-
Pili bic conditions. S. pneumoniae must be separated from other
IgA protease -hemolytic streptococci in the laboratory; this is usually ac-
LPS (endotoxin) complished with a disk susceptibility test using the unique sus-
Omps ceptibility of pneumococci to Optochin (ethylhydrocupreine
Outer membrane vesicles, or blebs hydrochloride). Pneumococci, but not other streptococci, are
Metabolic pathways (e.g., iron) also sensitive to bile or bile salts (e.g., 10% deoxycholate),
but the bile solubility test is now rarely performed in hospital
As noted earlier, all isolates from invasive infections are en- laboratories.
capsulated (serogroup positive), but 20% to 90% of isolates Pneumococci are classified within serotypes on the basis of
from carriers are unencapsulated (nontypeable). The capsu- antigenic differences among capsular polysaccharides. These
lar polysaccharide appears to be essential for meningococcal capsular substances are complex polysaccharides that form

Scheld_Ch24.indd 374 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 375

hydrophilic gels on the surface of the organism. At present, a marker of neurovirulence. In addition to the K1 capsule,
at least 90 serotypes have been identified, classified by two multiple other potential virulence factors for bloodbrain bar-
systems of nomenclature (American and Danish). Capsular rier (BBB) traversal and meningitis have been documented in
polysaccharides are identified by agglutination; the Neufeld E. coli from phylogenetic groups A, D, and B2 (e.g., O45:K1
quellung reaction, characterized by capsular swelling and in- and O18:K1 strains) (103).
creased refraction in the presence of antisera, also useful for Aerobic gram-negative bacilli have become increasingly
identifying pneumococci in clinical specimens (e.g., CSF); pre- important in patients with bacterial meningitis (104). Beyond
cipitation; and counterimmunoelectrophoresis. It is important the neonatal period, aerobic gram-negative bacillary men-
to emphasize that cross reactions exist between individual ingitis occurs in three major clinical settings: head trauma
pneumococcal serotypes, as well as with other bacteria (e.g., (approximately 30% of cases, especially in conjunction with
E. coli, Klebsiella pneumoniae, Hib, and S. sanguinis). For CSF rhinorrhea or otorrhea); after neurosurgical procedures
example, serotype 14 pneumococcal capsular polysaccharide (approximately 50% of cases); and in association with other
cross-reacts with type III group B streptococci and with certain conditions (approximately 20% of cases), including strongy-
human ABO blood group isoantigens. loidiasis, gram-negative bacteremia, ruptured brain abscess,
Capsular polysaccharide is essential for pneumococcal vir- or impairment of host defense mechanisms (e.g., steroids or
ulence, and a few serotypes are associated with most invasive AIDS). Many of these infections are nosocomial in origin,
infections. Encapsulated organisms (smooth colonies on agar) although community-acquired gram-negative bacillary menin-
are virulent for humans and experimental animals, whereas gitis is increasing in frequency, particularly in the elderly older
unencapsulated (rough colonies on agar) strains are avirulent. than 71 years and in debilitated, alcoholic, or diabetic adults
Capsular polysaccharide enhances virulence through its anti- (105107). The most common causes of gram-negative aero-
phagocytic properties, as in Hib and meningococci. bic bacillary meningitis beyond the first month of life include
Of the more than 90 known pneumococcal serotypes, only Klebsiella species (about 40% of cases), E. coli (roughly 15%
a few (usually the lower number types rather than the higher to 30%), and Pseudomonas aeruginosa (about 10% to 20%).
number types commonly found in the carrier state) account In a recent study from Korea of 91 adult patients with noso-
for most invasive pneumococcal infections. The predominant comial meningitis, Acinetobacter species accounted for 32.5%
capsular types were types 1, 2, and 3 in the preantibiotic era. of cases (108).
A different pattern has emerged in the past 40 years, and it dif-
fers between adults and children. A few capsular types cause
the majority of bacteremic cases among adults: types 1, 3, 4, 7,
8, 9, 12, and 14, and less commonly types 6, 18, and 19 (these Streptococcus agalactiae
are not listed in rank order). Approximately 65% of cases are
Group B streptococci are the most common cause of invasive
caused by eight serotypes, although no single serotype predom-
neonatal disease in the United States, accounting for approxi-
inates among bacteremic adults. Capsular types 14, 6, 18, 19,
mately 11,000 cases of meningitis and/or bacteremia yearly.
23, 1, 4, and 9 cause approximately 85% of serious infections
The incidence of group B streptococcal neonatal infections has
in children, a pattern different from that observed in adults.
remained relatively stable in this country until recently (109).
Perhaps more important with respect to this discussion, there
The use of antimicrobial agents (e.g., ampicillin) in pregnant
is a very strong correlation between serotypes causing pneu-
women with vaginal colonization with group B streptococci
monia (or bacteremia) and those responsible for pneumococ-
at the time of delivery has led to a decline in the incidence
cal meningitis (69). For example, 76% to 86% of blood and
of neonatal invasive group B streptococcal disease, perhaps,
CSF isolates were included among the 14 serotypes represented
as suggested by one study, with an increase in ampicillin-
in the original pneumococcal vaccine. However, marked geo-
resistant E. coli early-onset sepsis in low-birth-weight infants
graphic variations exist among pneumococcal serotypes caus-
(110). However, it appears to be increasing in frequency in the
ing invasive disease, and the serotype distribution may change
developing world.
over time in a given locale. Furthermore, some serotypes may
Group B streptococci are classified into six main serotypes
be more virulent than others and associated with less favor-
(designated Ia, Ib/c, Ia/c, II, III, and IV) based on the expression
able outcomes, including death. Older studies of pneumococ-
of type-specific capsular polysaccharide antigens and various
cal meningitis in the 1950s (summarized by Scheld [69]) noted
surface proteins as additional antigenic markers (111); addi-
an increased mortality with disease caused by serotypes 2, 8,
tional candidate serotypes are under evaluation. Although all
and 12. Although type 12 caused only 2.5% of cases, it was
serotypes have been isolated from neonates with invasive dis-
responsible for 30.7% of the deaths. Similarly, serotype 6 was
ease, type III is responsible for the vast majority of meningeal
associated with four of eight deaths in a more recent survey.
infections, suggesting a high virulence potential and/or CNS
The role of other cell surface components (such as
tropism for this serotype. The chromosomal genetic diversity
C-polysaccharide antigens, cell wall antigens, M- or R-protein
of S. agalactiae was studied and the clonal nature of the native
antigens) and putative toxins (such as hemolysin, neur-
bacterial populations was again demonstrated (112). A collec-
aminidase, purpura-producing principle) in the pathogenesis
tion of 128 isolates representing all six serotypes, including
and pathophysiology of pneumococcal meningitis remains
44 type III isolates from invasive episodes (18 recovered from
poorly defined. Nevertheless, CSF inflammation is induced
CSF), were subjected to multilocus enzyme electrophoresis,
by pneumococcal cell wall and its constituents (particularly
an analysis based on electrophoretically demonstrable allelic
lipoteichoic acid) but not by purified pneumococcal capsular
profiles at 11 metabolic enzyme loci, all encoded at the chro-
polysaccharide (see later discussion).
mosome level. Nineteen distinct ETs were identified in two
primary phylogenetic divisions, each representing a multilocus
clonal genotype. A single ET (ET-1), comprising 40 isolates of
Gram-Negative Bacilli serotype III group B streptococci, formed the first phylogenetic
division. These strains produced greater amounts of neur-
Approximately 84% of cases of neonatal meningitis and sepsis aminidase and were more virulent than the other type III iso-
attributable to E. coli are caused by strains bearing the K1 lates found in several of the 18 ETs in the second division. This
capsular polysaccharide antigen; this capsular type serves as newly evolved clone (ET-1) is responsible for most invasive

Scheld_Ch24.indd 375 2/21/14 5:42 PM


376 Part III: Bacterial and Mycoplasmal Infections

disease episodes caused by group B streptococci type III in the corticosteroid therapy, and immunosuppressed adults (e.g.,
United States (112). Overall, approximately 52% of group B renal transplant recipients) (120122). In one recent study,
streptococcal meningitis cases in the United States occur dur- patients with chronic lymphocytic leukemia had a greater than
ing the first month of life. In one recent review of 444 cases 1,000-fold risk of acquiring listeriosis (123). Other predispos-
of neonatal bacterial meningitis over a 7-year period, group ing conditions include diabetes mellitus, liver disease, chronic
B streptococcus was the most common etiology in early- renal disease, collagen-vascular diseases, pregnancy, and con-
onset (occurring between day 0 to 4 of birth) and late-onset ditions associated with iron overload. Listeria meningitis has
(occurring between day 5 and 28 of birth) disease, respon- also been reported with use of antitumor necrosis factor-
sible for 77% and 50% of cases, respectively (113). In the (TNF- ) agents, such as infliximab in patients with Crohn dis-
United States, the overall mortality rate ranges from 7% to ease (124) and ulcerative colitis (125), and etanercept in a pa-
27%. Survivors of group B streptococcal meningitis also have tient with adult Still disease (126). Meningitis can also occur
substantial long-term morbidity (114), indicating the need for in immunocompetent children and adults (127,128). In non-
ongoing developmental follow-up and the development of pre- perinatal cases, the route of transmission is often unknown.
ventive strategies (see later discussion). At least 1% of normal individuals excrete the organism in
In addition to the common conditions of neonatal men- their stools, but contacts of symptomatic patients have much
ingitis and postpartum fever and/or bacteremia in parturi- higher excretion rates (approximately 25%). Nevertheless, the
ent women caused by group B streptococci, these organisms true carriage rate, its duration, and its relationship to invasive
also cause serious infections in adults, including meningi- disease are poorly defined. Although often considered a zoo-
tis (115,116). Risk factors in adults include age older than nosis, most patients do not report animal exposure. Reports
60 years, diabetes mellitus, parturition, cardiac disease, colla- have emphasized food-borne transmission of L. monocy-
gen-vascular diseases, malignancy, alcoholism, hepatic failure, togenes, a route of transmission that accounts for the over-
renal failure, corticosteroid therapy, decubitus ulcers, neuro- whelming majority of sporadic cases. Many foods have been
genic bladder, previous stroke, and AIDS. No underlying ill- implicated, including coleslaw, Mexican-style cheese, raw veg-
nesses were found in 43% of patients in one review (116). etables, seafood, pasteurized milk, Swiss cheese, raw hot dogs,
undercooked chicken, alfalfa sprouts, cantaloupe, diced celery,
hog head cheese (a meat jelly made from hog heads and feet),
Listeria monocytogenes and processed meats, thus pointing to the intestinal tract as
the usual portal of entry (120,122,129134). In one outbreak,
L. monocytogenes remains an important cause of neonatal
57 cases of listeriosis occurred in western Switzerland in as-
meningitis; the source is the genital tract or subclinical infec-
sociation with the consumption of soft cheese (135); 40% of
tion of the mother. Although L. monocytogenes may cause
these cases were meningitis and 39% were meningoencepha-
meningitis in normal adults, most patients are diabetic, alco-
litis. Some studies report a higher frequency of listeriosis in
holic, elderly, or immunosuppressed. L. monocytogenes had
summer, opposite to the seasonal pattern seen with most
been the major cause of bacterial meningitis among renal
other forms of bacterial meningitis. However, the incidence
transplant recipients, but this is decreasing in frequency as a
of invasive listeriosis has been decreasing, likely a result of a
result of the use of TMP-SMX prophylaxis.
decrease in the prevalence of L. monocytogenes contamina-
L. monocytogenes is widespread. Although clusters of nos-
tion of ready-to-eat food (136); this has been associated with
ocomial cases and focal outbreaks are reported, most cases
a decrease in nonperinatal listeriosis-associated deaths (137).
of human listeriosis are sporadic. The incidence of L. mono-
L. monocytogenes is a gram-positive, nonspore-forming,
cytogenes infections is difficult to quantitate. Many countries
catalase-positive, aerobic rod that may be difficult to culture
in northern Europe have reported annual incidence figures of
on initial isolation but that, once grown, passes readily on a
approximately 2 to 3 per million. After a large (142 cases)
variety of laboratory media. The organisms may appear coc-
food-borne outbreak in Los Angeles County, California, in
coid on Gram stains of clinical specimens, particularly CSF,
1985, mandatory reporting of L. monocytogenes isolates by
and are often mistaken for pneumococci. More importantly,
clinical laboratories was instituted. During the first year of
L. monocytogenes resembles diphtheroids and may thus be
active surveillance, 94 cases of listeriosis were reported (117),
dismissed as a contaminant, a grave error. The presence of
for an annual crude incidence of 11.7 per million persons, sim-
-hemolysis and a characteristic tumbling motility at room
ilar to figures (11.3 per million annually) reported from France
temperature are used to separate L. monocytogenes from
in 1984. Listeriosis is undoubtedly underreported.
similar diphtheroid-like organisms. A hemolytic and cytolytic
Approximately one third of cases in the United States are
toxin (listeriolysin 0) of 52 kd appears essential for virulence;
in neonates and/or their mothers (39% in the Los Angeles
the toxin is expressed under conditions of low pH and low iron
survey) (117). The proportion of perinatal infections in Europe
concentration and may facilitate phagolysosomal disruption
is higher. Among the nonperinatal cases, various risk factors
and growth within mononuclear phagocytes. At least 11 sero-
for listeriosis were identified, including immunosuppression as
types are recognized, but more than 90% of invasive infections
a documented history of steroid ingestion or chemotherapy
are caused by three serotypes: Ia, Ib, and IVb. The rate of un-
(35% of cases, the single most important risk factor); age
favorable outcome among adults with Listeria meningitis was
older than 75 years; renal disease; cancer; alcoholism and/or
recently found to increase over a 14-year period from 27% to
cirrhosis; and AIDS. Nevertheless, serious Listeria infections,
61%, with the emerging L. monocytogenes serotype ST6 iden-
including meningitis, remain uncommon in patients with
tified as the main factor leading to a poorer prognosis (138).
AIDS (118,119), but diagnosis of Listeria meningitis in any-
one younger than 50 years of age should prompt testing for
HIV, if not done previously. Of the nonperinatal cases, only Staphylococcus epidermidis
2 of 57 had no definable underlying risk factor; 21 of 57 had
meningitis (117). As stated earlier, L. monocytogenes remains Coagulase-negative staphylococci are very rare causes of bac-
a distinctly unusual cause of meningitis in developing coun- terial meningitis in children and adults, except in the setting
tries. Listerial infection is most common in infants younger of an indwelling CSF shunt, where these organisms are the
than 1 month of age (up to 10% of cases), adults older than most prevalent pathogen. Therefore, this group is discussed
60 years of age, alcoholics, cancer patients, those receiving elsewhere in this volume.

Scheld_Ch24.indd 376 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 377

or surgery; gastrointestinal disease; head and neck cancer;


Staphylococcus aureus suppurative pharyngitis; CSF shunts; and immunosuppression
(particularly corticosteroid administration) (145,146). Most
Meningitis caused by Staphylococcus aureus is relatively cases arise from spread of infection secondary to a contigu-
unusual; this organism was responsible for 0.8% to 8.8% ous focus of disease; anaerobic meningitis rarely complicates
of cases in various surveys (139,140). S. aureus is the second bacteremia from a distant extracranial focus.
most common cause of CSF shunt infections, accounting for A variety of bacterial species are responsible for anaerobic
12% to 36% of cases. This organism is also frequently isolated meningitis. Only nine cases of Bacteroides fragilis meningitis
from patients with nosocomial meningitis and is responsible unaccompanied by a brain abscess had been reported in the
for approximately 20% of such cases. Although secondary modern era through 1987 (147). Seven occurred in premature
meningitis in the setting of infective endocarditis is an uncom- infants or neonates (median age, 20 days), thereby complicat-
mon event, most of these infections are caused by S. aureus. ing congenital defects or gastrointestinal disease such as necro-
Other important associated conditions have been recognized, tizing enterocolitis (148). Meningitis caused by Fusobacterium
including head trauma, neurosurgical procedures, various ab- species, usually Fusobacterium necrophorum, generally occurs
scesses (cerebral, epidural, oral, abdominal), sinusitis, osteo- in older children (median age, about 5 years) or adults as a
myelitis, decubitus ulcers, pneumonia, cellulitis, injection drug complication of acute or chronic otitis media (148). A vari-
use, malignancy, and infected intravascular grafts or shunts. ety of anaerobic gram-positive cocci have been isolated in a
Several other predisposing factors have been proposed. In a few cases, particularly peptostreptococci. Meningitis caused
retrospective review of 28 cases of S. aureus meningitis seen by Clostridium species almost always develops following head
from 1972 to 1982 at three North Carolina teaching hospi- trauma or a neurosurgical procedure. For example, in a sum-
tals, 22 occurred beyond the neonatal period (140). Among mary of 17 cases caused by Clostridium perfringens (149), only
the adult patients (n  20; mean age 52 years), 45% had an 3 were not associated with CNS trauma or surgery. Although
underlying condition (diabetes mellitus, malignancy, renal the disease course is highly variable, some cases of clostridial
failure, immunosuppression), 35% had had head trauma or meningitis are characterized by intracranial gas formation,
undergone neurosurgery (ventriculoperitoneal shunt, crani- visible on plain skull radiographs or computed tomography
otomy), and about 20% developed meningitis in association (CT); CSF white blood cell (WBC) concentrations exceeding
with endocarditis or a paraspinal infection. Mortality was 20,000/mm3; and death within hours of presentation. A few
high (50% in adults), especially when S. aureus meningitis cases of meningitis caused by Actinomyces species (in the
complicated a distant extracranial focus of infection (five of absence of brain abscess formation) and Propionibacterium
the six patients with purulent meningitis during active endo- acnes (usually subacute with a predominantly monocytic CSF
carditis died) (140). The prognosis for S. aureus infections of pleocytosis) have also been reported. In approximately one
CSF shunts is more favorable. In a review of clinical and bac- eighth of patients, the infection is mixed, with anaerobic plus
teriologic data from 61 postoperative and 43 hematogenous aerobic or microaerophilic organisms recovered from CSF.
cases of S. aureus meningitis from Denmark, postoperative
cases had a lower mortality rate (18%) than cases resulting
from hematogenous spread (56%); hematogenous S. aureus Unusual Etiologic Agents
meningitis had a higher mortality rate related to age, pres-
ence of shock, and infections with strains of phage type 95 CNS infections caused by higher bacteria (e.g., Mycobacterium
(141). Hospital-acquired cases are often caused by methicil- species, Nocardia species, Actinomyces species), spirochetes
lin-resistant strains (142). In one series from 1999 to 2008 (e.g., Treponema pallidum, Borrelia burgdorferi, Leptospira
(143), S. aureus accounted for approximately 5% of cases of species), Brucella species, and so on, are discussed elsewhere in
culture-proven bacterial meningitis in adults; since 2005, more this volume. A plethora of bacteria have been documented as the
than 75% of all cases were caused by methicillin-resistant cause of meningitis in isolated case reports or in small numbers
Staphylococcus aureus (MRSA) and 52% (11 of 21 cases) of of patients, including group A streptococci usually in associa-
hematogenous cases were seen in injection drug users. In a tion with pharyngitis, otitis media, and/or sinusitis (150); non-
multicenter review of 86 cases of MRSA meningitis in adults pneumococcal -hemolytic streptococci such as Streptococcus
(144), the infection was nosocomial in 93% of cases; in those mitis (151); enterococci (152,153); Streptococcus gallolyticus;
patients with postoperative meningitis, the most common pre- diphtheroids (although P. acnes is an important etiologic agent
disposing conditions were the presence of CSF devices, neuro- in patients with CSF shunt and drain infections); N. gonor-
surgery, CSF leaks, and head trauma. rhoeae (approximately 30 cases reported in the past 20 years);
Neisseria subflava; Gardnerella vaginalis (one case report);
many members of the Enterobacteriaceae in addition to E. coli
Anaerobic Bacteria and Salmonella species; Flavobacterium meningosepticum;
Haemophilus species other than H. influenzae; and many oth-
Meningitis caused by anaerobic bacteria is rare, accounting ers. Fewer than 0.5% of adult cases of bacterial meningitis
for fewer than 1% of pyogenic cases, except following the in- are caused by group C streptococci but may occur in humans
traventricular rupture of a brain abscess. Anaerobic meningitis after contact with domestic animals (especially horses) or their
may be underrecognized because CSF is not routinely cultured unpasteurized products (154,155); however, mortality is high,
anaerobically. Enriched media and proper transport of CSF to perhaps because of the unpredictable susceptibility of this
the laboratory, which are essential for isolation of anaerobes, organism to
-lactam agents.
are not uniformly performed. Only five cases caused by strict Despite the frequency with which the viridans strepto-
anaerobes were reported among 18,642 patients analyzed by cocci cause bacteremia, they are unusual causes of meningitis
the CDC in one study (7). (0.3% to 5% of culture-proven cases) (156). Streptococcus
Anaerobic meningitis is associated with a variety of clinical salivarius meningitis has been reported following spinal an-
conditions, including rupture of brain abscess or extension to esthesia (157,158) and myelogram procedures (159), sup-
the surface of the brain; chronic otitis, mastoiditis, or sinus- porting the importance of appropriate infection control
itis; head trauma; neurosurgical procedures (e.g., craniotomy, practices (i.e., masks, proper aseptic technique, and safe
laminectomy); congenital dural defects; abdominal trauma injection practice) in those who perform spinal procedures.

Scheld_Ch24.indd 377 2/21/14 5:42 PM


378 Part III: Bacterial and Mycoplasmal Infections

Streptococcus suis is the most frequent cause of bacterial


meningitis in southern Vietnam and is associated with sig-
nificant morbidity attributable to hearing loss (160); the pig
is the natural reservoir of this microorganism and the main
source of human infection. Risk factors for S. suis meningitis
include eating high-risk dishes (such as undercooked pig
blood or pig intestine) popular in parts of Asia, occupational
exposure to pigs, and exposures to pig or pork in the pres-
ence of skin injuries (161).
Polymicrobial bacterial meningitis, with simultaneous re-
covery of two or more bacterial species from CSF, is unusual.
Mixed infections account for about 1% of bacterial meningitis
cases. In a review of 34 series encompassing 11,281 cases of
bacterial meningitis, 116 cases (1%) were mixed (162). This
condition appears to be evolving in the antibiotic era. Before
1950, nearly all cases occurred in children and were caused
by combinations of bacteria commonly associated with men-
ingitis (the three major meningeal pathogens). Since 1950,
most cases have occurred in adults, with a wider spectrum of
etiologic agents, particularly gram-negative aerobic bacilli. FIGURE 24.2 Infiltration of the meninges by neutrophils.
Approximately one third of cases were nosocomially acquired.
Common predisposing conditions in the older population af-
fected since 1950 include contiguous foci of infection, tumors
in close proximity to the neuraxis (e.g., head and neck), rectal and the pathophysiology of associated complications have sub-
carcinoma, or fistulous communications with the CNS. The sequently allowed for a more complete understanding of the
mortality rate is 63% for cases occurring after 1950. Several pathologic processes operating in this disorder.
cases of meningitis caused by mixed bacterial and mycobacte- Bacteria reach the meninges through one of the following
rial or fungal agents have also been reported. pathways: (a) hematogenous dissemination from a distant
Simultaneous isolation of viruses and bacteria from the site (e.g., nasopharynx, lung, skin, and genitourinary tract);
CSF is rare; only seven well-documented cases were reported (b) spread from an adjacent suppurative focus of infection
prior to 1988 (163). However, in a 1-year retrospective review (e.g., otitis media, sinusitis, and mastoiditis); and (c) a con-
from the Ohio State University published in 1986, 5 (2.8%) of genital or an acquired structural defect (176).
176 children with CSF enteroviral isolates also had bacterial Once bacteria gain access to the SAS, an inflammatory
meningitis (163). Conversely, CSF samples from 5 (4.8%) of process ensues (Fig. 24.2). Neutrophils migrate into the SAS,
105 children with bacterial meningitis also grew an entero- producing a purulent exudate. On gross examination, the exu-
virus. All the patients presented in late summer at the peak date has a gray-yellow or yellow-green appearance (Fig. 24.3).
of the enterovirus season, and each case was caused by a dif-
ferent bacterial pathogen. Because the CSF formula was in-
distinguishable from that of patients with typical bacterial
meningitis, and because the clinical course and response to
therapy were similar to those of patients with typical bacte-
rial meningitis, this condition may be underrecognized, as CSF
viral cultures are rarely performed when bacterial meningitis
is the likely diagnosis.

PATHOGENESIS AND
PATHOPHYSIOLOGY
Despite the availability of effective antimicrobial therapy,
bacterial meningitis continues to be a potentially fatal ill-
ness. Several investigators have examined the pathogenic and
pathophysiologic mechanisms operating in meningitis, with
the aim of improving the outcome of patients with this disor-
der. These pathogenetic and pathophysiologic mechanisms are
discussed in detail in Chapter 23 and are detailed in a number
of excellent reviews (164174).

PATHOLOGY
Adams et al. (175) described the pathology of bacterial menin-
gitis in 1948 based on examination of autopsy material from
patients with H. influenzae meningitis. Experimental models of FIGURE 24.3 Purulent exudate in subarachnoid space over cerebral
bacterial meningitis, knowledge of host defense mechanisms, hemispheres.

Scheld_Ch24.indd 378 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 379

FIGURE 24.6 Purulent exudate (arrow) covering spinal cord and


nerve roots.

FIGURE 24.4 Purulent exudate in the cerebral sulci. neutrophil in eradicating infection at this stage is unknown.
The presence of free-living bacteria in the exudate suggests
that phagocytosis by neutrophils is incomplete as a result of
deficient opsonic activity in CSF; however, low CSF leukocyte
concentrations in the presence of high CSF bacterial concen-
It is most abundant in the cisterns at the base of the brain trations have been associated with a poor prognosis in both
and over the convexities of the hemispheres in the rolandic experimental and human meningitis (178). These observa-
and sylvian sulci (175) (Fig. 24.4). The tendency for exudate tions suggest that the neutrophils have a beneficial role in par-
to accumulate in the cisterns at the base of the brain is ex- tial control of the early stages of the infection. The presence of
plained by the anatomy of the SAS, which is deepest at the large numbers of neutrophils in the SAS and vessel walls may,
base of the brain. The various cisterns are expansions of the however, also be detrimental to the host, as is discussed in the
SAS, with the largest of these areas lying between the cerebel- previous section.
lum and medulla and extending downward below the foramen Within the first 48 to 72 hours of infection, there is
magnum, the so-called cisterna magna or cerebellomedullary evidence of inflammation in the walls of the small and
cistern (177). Purulent exudate accumulates in this cistern and medium-sized subarachnoid arteries (Fig. 24.8). The endo-
extends into the other basal cisterns and onto the posterior thelial cells swell and multiply, narrowing the lumen. The
surface of the spinal cord (Figs. 24.5 and 24.6). The exudate adventitia is infiltrated by neutrophils, and neutrophils and
also extends into the arachnoidal sheaths of the cranial nerves lymphocytes form a layer beneath the intima (Fig. 24.9).
and into the perivascular spaces of the cortex. A small amount Subintimal arterial infiltration by neutrophils and lympho-
of exudate may be found in the ventricular fluid and attached cytes is relatively unique to infection of the meninges, and
to the ventricular walls and choroid plexus; thus, the appear- it may be related to the anatomy of the meningeal arter-
ance of the ventricular fluid is usually cloudy by the end of the ies. It is only rarely observed in inflammatory processes in
first week of the infection (175). other organs (175). The adventitia of the subarachnoid ves-
Microscopic examination of the subarachnoid exudate sels, as they enter the brain parenchyma, is formed by the
in the early stages of infection demonstrates large numbers arachnoid membrane. As arteries and veins enter the brain
of neutrophils and bacteria (lying either free in the exudate parenchyma, they carry with them a sleeve of arachnoid im-
or within neutrophils) (Fig. 24.7) (176). The role of the mediately surrounding the vessels and a sleeve of pia mater

FIGURE 24.5 Purulent exudate surrounding the temporal poles, FIGURE 24.7 Diplococci. Red-stained cells are polymorphonuclear
optic chiasm, and pons. leukocytes.

Scheld_Ch24.indd 379 2/21/14 5:42 PM


380 Part III: Bacterial and Mycoplasmal Infections

FIGURE 24.8 Aggregation of white blood cells in the arterial FIGURE 24.10 Inflammatory cell extension from the subarachnoid
intima. space into a space of Virchow-Robin. Note the infiltrate around the
arterial wall and on the intimal surface.

external to this. Between these two layers lies an extension by macrophages, which are derived from meningeal histio-
of the SAS, known as the perivascular space or Virchow- cytes. Lymphocytes and fibroblasts proliferate in the exudate.
Robin space, which is filled with CSF (177) (Fig. 24.10). Microscopic changes in the brain parenchyma may also be
Because the vessel wall is enveloped by the arachnoid mem- present. The nuclei of neurons and glial cells become shrunken,
brane, it is affected early by any inflammatory process in pyknotic, and darkly staining (Fig. 24.12). Rod-shaped mi-
the meninges. However, as shown in animal models of croglial cells and astrocytes increase in number in the cerebral
bacterial meningitis, the arachnoid membrane generally and cerebellar cortex, brainstem, and spinal cord (Fig. 24.13).
remains intact. Astrocytic processes become swollen (Fig. 24.14). There is a loss
The meningeal veins become distended and develop mural of myelinated fibers in the subcortical white matter, cerebellum,
inflammation during bacterial meningitis. There may be and brainstem (175). Similar morphologic changes are seen in
focal necrosis of the vessel wall, along with mural thrombus ischemic and hypoxic cortical injury, suggesting that ischemia
formation in the lumen of the vein or in the dural sinus (175) and/or hypoxia may contribute to the pathologic changes from
(Fig. 24.11). Hemorrhagic cortical infarction is the result of bacterial meningitis at this stage.
cortical venous and dural sinus thrombosis. Also at the end of the first week of the infection, there
Toward the end of the first week of meningeal infection, is infiltration of the subependymal tissues and perivascular
there is a change in the cellular composition of the subarachnoid spaces by neutrophils and lymphocytes. The ependymal and
exudate. Neutrophils begin to degenerate and are removed subependymal tissues become edematous, and cells begin to

FIGURE 24.9 Small artery with inflammatory


infiltration of the vessel wall and mural thrombus.

Scheld_Ch24.indd 380 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 381

FIGURE 24.11 Inflammatory infiltrate in the wall of a cerebral vein


with thrombosis. Note inflammatory cells in thrombus. FIGURE 24.14 Reactive gemistocytic astrocytes (arrow).

die; desquamation of the ependymal lining also occurs. Rod-


shaped microglial cells and swollen astrocytes proliferate and
overgrow the remnants of the ependymal lining. An inflamma-
tory infiltrate in the walls of the subependymal arteries may
occlude the vessel, leading to tissue necrosis (175).
As the infection progresses, the subarachnoid exudate
continues to accumulate. In some areas, it may become sev-
eral millimeters thick (Fig. 24.15). Toward the end of the sec-
ond week, the exudate separates into two layers. The outer
layer, just beneath the arachnoid membrane, is composed of
neutrophils and fibrin. The inner layer, which is contiguous
with the pia, is composed of lymphocytes, plasma cells, and
macrophages (175). As the subarachnoid exudate continues
to accumulate, the flow of CSF may become obstructed.
The dynamics involved in obstruction of CSF flow are as
follows: the bulk of CSF is formed by the choroid plexus
in the lateral and third ventricles, and it flows through the
FIGURE 24.12 Shrunken red neurons. cerebral aqueduct into the fourth ventricle. CSF leaves the
fourth ventricle through the midline foramen of Magendie
and the lateral foramina of Luschka to reach the SAS (177).
When the foramina of Magendie and Luschka are blocked by
exudate, the spinal fluid cannot circulate to the convexities of

FIGURE 24.15 Purulent exudate in the sylvian fissure and covering


FIGURE 24.13 Marked proliferation of microglial cells (arrow). the cerebellum in the brain of a premature infant with meningitis.

Scheld_Ch24.indd 381 2/21/14 5:42 PM


382 Part III: Bacterial and Mycoplasmal Infections

in the subcortical white matter. Cytotoxic edema is caused


by an accumulation of intracellular water and sodium with
subsequent swelling of cells. Membrane polyunsaturated fatty
acids and other toxic factors released from leukocytes contrib-
ute to the development of cytotoxic edema (69). Interstitial
edema is a result of obstruction to CSF resorption, as discussed
earlier in this chapter.
Cerebral edema leads to an increase in ICP, and increased
ICP adversely affects cerebral perfusion pressure (CPP), defined
as the difference between the systemic mean arterial pressure
(MAP) and the ICP: CPP  MAP ICP (180). Cerebral blood
flow may be maintained at normal or near-normal levels in the
presence of increased ICP, provided that the CPP is maintained
at a range of at least 50 to 60 mm Hg. As ICP continues to rise,
or if systemic arterial pressure decreases, cerebral ischemia and
infarction may result.
Experimental evidence exists for a loss of autoregulation
of cerebral blood flow in bacterial meningitis (181). This is
FIGURE 24.16 Hydrocephalus. another potential contributing factor to the development of
cerebral ischemia in this infection. Cerebral blood flow is
normally constant within a range of mean systemic arterial
the brain, where it is normally absorbed. The flow of CSF is
pressure from 50 to 150 mm Hg. When autoregulation is
blocked at the level of the fourth ventricle, resulting in non-
disturbed, systemic hypotension results in decreased cerebral
communicating or obstructive hydrocephalus (Fig. 24.16).
blood flow and cerebral ischemia (182).
From the fourth ventricle, CSF normally flows to the SAS at
Cerebral edema may lead to herniation of brain tissue.
the base of the brain. From here, CSF flows up over the convex-
Herniation may compress intracerebral arteries, leading to
ity of the hemispheres to be absorbed by the arachnoid villi in
ischemia and infarction; it may also compress the surface of
the intracranial venous sinuses (177). The presence of a fibrino-
the brain against the dura, leading to necrosis of brain tis-
purulent exudate in the SAS interferes with the absorption of
sue. Tonsillar herniation, the downward displacement of the
CSF by the arachnoid villi. This obstruction to CSF resorption
cerebellar tonsils through the foramen magnum, can result in
resulting from inflammatory changes in the arachnoid granula-
apnea, hemodynamic instability, coma, and death (177,183).
tions results in communicating hydrocephalus. When the sub-
The pathologic lesions described are typical of meningi-
arachnoid exudate has been present for several weeks, there
tis caused by bacteria, but some distinctions among lesions
are (a) marked fibrosis of the arachnoid villi and (b) pockets of
caused by H. influenzae, N. meningitidis, and S. pneumoniae
exudate walled off by adhesions between the arachnoid mem-
infection have been observed. The subarachnoid exudate
brane and dura (175). These fibrotic changes produce further
in H. influenzae meningitis is very thick and purulent, with
mechanical obstruction to the resorption of CSF by the arach-
loculated pockets of pus in the basilar cisterns and cerebral
noid villi. The end results are (a) transependymal movement of
sulci. In contrast, the exudate in pneumococcal meningitis
CSF from the ventricular system into the brain parenchyma and
tends to be more extensive over the convexities of the hemi-
(b) the development of interstitial cerebral edema.
spheres than in the basilar cisterns (Fig. 24.18). In menin-
The development of diffuse cerebral edema and increased
ICP further complicates the pathologic changes already
described. Cerebral edema is defined as an increase in the
volume of the brain resulting from an increased water con-
tent (179) (Fig. 24.17). The cerebral edema in meningitis is
a combination of vasogenic, cytotoxic, and interstitial edema
(170). Vasogenic edema is a result of increased permeabil-
ity of brain capillaries with the subsequent accumulation of
water and protein molecules in the extracellular space, mainly

FIGURE 24.17 Specimen demonstrating the characteristic changes of FIGURE 24.18 Purulent exudate over the cerebral convexities sec-
cerebral edema-flattened gyri and narrowed ventricles. ondary to pneumococcal meningitis.

Scheld_Ch24.indd 382 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 383

Bacteremia also contributes to the pathologic features of


this disease. Bacteremia is present in 30% to 90% of cases
of bacterial meningitis. It can be either the primary event
leading to development of meningitis or a secondary event
arising from the clearance of bacteria from the SAS through
the arachnoid villi to the bloodstream. Pneumococcal cell
walls activate the alternative complement pathway, with
the generation of chemotactic peptides in the systemic
circulation and in CSF. The principal component of this
activity is C5a, a highly chemotactic peptide that is a
stimulus for an intense CSF accumulation of neutrophils.
By this process, neutrophils also become sequestered in
the pulmonary vascular bed, leading to cardiopulmonary
dysfunction, neutrophil-mediated vascular damage, and the
development of the acute respiratory distress syndrome and
thereby further contributing to the morbidity and mortality
of meningitis (69).

FIGURE 24.19 Petechial hemorrhages in gray matter secondary to


meningococcal meningitis.
CLINICAL MANIFESTATIONS
Neonates
gococcal meningitis, the pathologic changes depend on the
severity and duration of the infection. In acute fulminating Clinical clues to the presence of meningitis in neonates are
meningococcemia, death may occur before pus can accumu- temperature instability (hypothermia or hyperthermia), list-
late in the SAS. At autopsy, severe hyperemia and swelling lessness, high-pitched crying, fretfulness, lethargy, refusal to
of cerebral tissue are evident, with petechial hemorrhages feed, weak suck, irritability, jaundice, vomiting, diarrhea, or
in the gray and white matter (Fig. 24.19) and in the sub- respiratory distress (184,185). Nuchal rigidity is not typically
ependymal regions of the lateral ventricles. A hemorrhagic found in the neonate. A change in the childs affect or state
ependymitis is typical of severe lethal meningococcal infec- of alertness is one of the most important signs of meningitis.
tion (Fig. 24.20). The presence of pus in the SAS may be evi- A bulging fontanelle (seen in one third of cases) usually occurs
dent only by microscopic examination (183). The pathologic late during the course of illness; seizures are observed in 40%
changes in meningococcal meningitis of longer duration are of neonates with bacterial meningitis.
similar to those described for meningitis caused by pyogenic
organisms in general.
Cranial and spinal nerve deficits, focal neurologic deficits, Infants and Children
seizure disorders, and subdural effusions are well-recognized
complications of meningitis. The cranial and spinal nerve The symptoms and signs of acute bacterial meningitis in
deficits are usually transient and caused by exudate in the infants and children depend on the age of the child, duration
SAS surrounding the nerves. Focal neurologic deficits and sei- of illness, and host response to infection (186); the clinical fea-
zure activity arise from cortical and subcortical ischemia and tures can be subtle, variable, nonspecific, and even absent. The
infarction (bland and hemorrhagic), which are the result of initial symptoms of bacterial meningitis in infants and chil-
inflammation and thrombosis in arteries and veins. Subdural dren may be any of the following: fever, stiff neck, headache,
effusions are relatively common in the course of bacterial lethargy, irritability, nausea, vomiting, and photophobia
meningitis in children; they are the result of an increase in (Table 24.3). In children 1 to 4 years of age, fever (94%), vom-
the permeability of the thin-walled capillaries and veins in iting (82%), and nuchal rigidity (77%) are the most common
the inner layer of the dura, with leakage of fluid into the initial symptoms.
subdural space. Although the symptoms are nonspecific, the combination
of one or more of these symptoms with signs of meningeal
irritation should suggest the diagnosis of meningitis. The classic
signs of meningeal irritation are nuchal rigidity and Brudzinski
and Kernig signs. Brudzinski actually described several signs of
meningeal irritation, including the nape-of-the-neck sign, the
identical contralateral reflex sign, and a reciprocal contralateral
reflex sign, as well as others (187,188). The nape-of-the-neck
sign is Brudzinskis best-known sign and is universally recog-
nized today as Brudzinski sign. The nape-of-the-neck sign
is positive when passive flexion of the neck results in flexion
of the hips and knees. The identical contralateral reflex sign
is elicited with the patient in the supine position by passively
flexing the hip and knee on one side. This sign is positive when
the contralateral leg flexes with this maneuver. The reciprocal
contralateral reflex sign is positive when the leg that has flexed
in response to passive flexion of the other leg begins to extend
spontaneously, resembling a little kick. The identical and re-
FIGURE 24.20 Hemorrhagic ventriculitis complicating meningitis. ciprocal contralateral reflex signs are not elicited as often as the

Scheld_Ch24.indd 383 2/21/14 5:42 PM


384 Part III: Bacterial and Mycoplasmal Infections

TA B L E 2 4 . 3 meningitis, 70 of viral meningitis, and 12 of unknown etiol-


ogy. Lethargy was more common in children with bacterial
SYMPTOMS AND SIGNS OF BACTERIAL MENINGITIS meningitis than in children with viral meningitis: 50% of the
BY AGE-GROUP children with bacterial meningitis were lethargic, and 32%
of the children with viral meningitis were lethargic (p .14).
Age-Group Symptoms Signs Although vomiting is a symptom of meningeal irritation, it
is a nonspecific symptom in children. Vomiting occurred in
Child Fever Nuchal rigidity 336 children, 84 of whom had bacterial or viral meningitis.
Lethargy or altered level Purpuric or Fever was present in every child with meningitis. The tempera-
of consciousness petechial rash ture elevations were higher in bacterial meningitis than in viral
Headache Seizures meningitis; 80% of the children with bacterial meningitis had
Irritability Ataxia temperatures of 38.8C or higher. In children with viral men-
Nausea and vomiting Focal neurologic ingitis, 40% had temperatures of 38.8C or greater (193). The
Respiratory symptoms deficits, including possibility of meningitis in a child who does not or cannot
Photophobia cranial nerve complain of headache or stiff neck and who does not have
palsies meningeal signs should be suspected when fever accompanies
Adult Fever Nuchal rigidity changes in behavior, changes in mental status, or new onset
Headache Altered level of of seizures.
Lethargy, confusion, or consciousness In one recent review of children aged 2 months to 15 years
coma Seizures who presented with suspected meningitis, the classic clinical
Nausea and vomiting Focal neurologic signs had limited value in establishing the diagnosis (194).
Photophobia deficits, including Clinical examination revealed nuchal rigidity in 65% of
Respiratory symptoms cranial nerve those with meningitis; Brudzinski and Kernig signs were elic-
palsies ited in 51% and 27% of those with meningitis, respectively.
Elderly Fever Nuchal rigidity Therefore, physicians should have a low threshold for LP in
Confusion or coma Altered level of patients at high risk for bacterial meningitis, given the serious
Headache consciousness nature of this disease.
Respiratory symptoms Seizuresstatus In a review of 1,064 cases of bacterial meningitis in infants
epilepticus and children, there were no signs of meningeal irritation in
16 patients (1.5%). Eight patients were older than 2 years
of age. LP was performed because of unexplained fever as-
sociated with an altered level of consciousness, behavioral
changes, seizure activity, or petechial skin lesions. Meningitis
was caused by N. meningitidis in seven patients, H. influenzae
nape-of-the-neck sign (188). The manner in which Kernig sign in six, S. pneumoniae in two, and Salmonella enteritidis in one.
is elicited and the interpretation of the results of the maneuver Most patients had a peripheral leukocytosis with a left shift.
as it is done today are different from those originally described The peripheral WBC count was greater than 10,000 cells/mm3
by Kernig (189). The maneuver as described by Kernig was in 12 patients and greater than 20,000 cells/mm3 in 7 patients
performed with the patient in a seated position while the phy- (195). The results of this review suggest that although menin-
sician attempted to extend the knee passively. In the presence gitis may occasionally occur without meningeal signs, there
of meningitis, knee extension was resisted so that a contrac- will usually be other signs or symptoms of intracranial infec-
ture of the extremities was maintained (188,189). Today the tion and a peripheral leukocytosis.
sign is elicited with the patient in a supine position. The thigh Observational data that are useful in predicting the pres-
is flexed on the abdomen, with the knee flexed. The leg is then ence of serious illness (e.g., meningitis) in a febrile child in-
passively extended. When meningeal inflammation is present, clude the following: (a) quality of cry, (b) reaction to parent
the patient resists leg extension (190). Nuchal rigidity and stimulation, (c) level of consciousness, (d) color, (e) hydra-
Brudzinski and Kernig signs are observed in fewer than 50% tion, and (f) response to social stimulation. These six items
of children with bacterial meningitis. were identified as significant and independent predictors of
The possibility of bacterial meningitis should be considered serious illness from a list of 14 observational items, scored
in every child with fever, vomiting, nuchal rigidity, and lethargy by pediatricians, for 312 febrile children 24 months of age or
or an altered mental status. In a review of 110 cases of culture- younger (196). The quality of the cry in a child with a serious
proven bacterial meningitis in children, fever (38.5C) was illness was weak, moaning, or high pitched. A healthy child
the most common symptom, being present in 94% of patients. was either not crying or had a strong cry with normal tone.
The absence of fever, particularly hypothermia, was associated Reaction to parental stimulation was judged based on the par-
with a worse prognosis, perhaps related to the slower rate of ent holding the child, talking to the child, or giving the child
bacterial replication in CSF when temperatures are elevated. a bottle. The child with a serious illness did not stop crying or
Apart from fever, the most common symptoms were (a) vomit- barely responded to stimulation by its parent. Consciousness
ing (82%) and nuchal rigidity (77%) in 1- to 4-year-old chil- was impaired in children with serious illnesses. They were le-
dren and (b) headache (92%) in children ages 5 to 12 years. thargic, stuporous, or obtunded. Sick children were described
Vomiting and nuchal rigidity were present in 80% of the as pale, cyanotic, or ashen. Signs of dehydration were present.
children who were 12 months or older. Nuchal rigidity is a The response to social stimulation was judged according to
classic sign of meningitis but can be absent early in the course whether the child would smile when talked to or smiled at.
of this illness; therefore, the absence of nuchal rigidity should Sick children did not respond to social stimulation. These six
not exclude the diagnosis of bacterial meningitis (191). items, when used together, had a specificity of 88% and a sen-
In a review of 709 LPs done on children in an outpatient sitivity of 77% for the presence of serious illness. When com-
setting in which there was a concern for meningitis, the CSF bined with history and physical examination, the sensitivity of
was abnormal in 16% (192). There were 30 cases of bacterial the six-item model increased to 92%. If all six of the observa-

Scheld_Ch24.indd 384 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 385

tion items were normal in a child, the probability of that child an LP is very low. If, however, there are clinical signs of menin-
having a serious illness was only 4.7% (196). gitis, an LP is indicated. If the seizure has a focal onset or there
The possibility of meningitis in a febrile child may also is a focal neurologic deficit on examination, CT is indicated
be suggested by the tempo of the illness. The presentation of before LP is performed. All children with new-onset febrile
meningitis in children is that of either a subacute infection or convulsions in whom LP is not performed should be reexam-
an acute fulminant illness. Children with a subacute presenta- ined 1 to 4 hours after the initial examination to be sure that
tion have fever, lethargy, and nuchal rigidity that progresses serious disease is not present (203).
over 1 to several days and is usually preceded by an upper The presence of a diffuse erythematous maculopapular
respiratory tract infection or otitis media (197). Children rash may be an early manifestation of meningococcemia or
with meningitis may also present with an illness that has been may represent a viral illness. The presence of a purpuric or
progressive over 24 to 72 hours or a fulminant illness that petechial rash on the trunk and lower extremities is sugges-
develops over several hours. tive of meningococcemia, although petechiae are sometimes
Children with a more rapidly progressive illness have signs seen in echovirus type 9 meningitis, acute staphylococcal en-
and symptoms of meningeal irritation and increased ICP on docarditis, and rarely pneumococcal or H. influenzae men-
initial presentation. CSF pressures exceeding 300 mm H2O ingitis (198,204). Petechiae are found in the skin, mucous
are common in acute bacterial meningitis, and ICPs exceeding membranes, or conjunctivae, but never in the nailbeds, of pa-
500 to 600 mm H2O are not unusual (197,198). Increased tients with meningococcemia; they usually fade in 3 or 4 days
ICP in bacterial meningitis in children is the consequence, in (205). Petechiae and/or purpura occurs in 50% to 75% of
part, of vasogenic and cytotoxic cerebral edema, altered CSF children with meningococcal meningitis. Children with fulmi-
resorption, and the inappropriate secretion of antidiuretic nating meningococcal septicemia may have the Waterhouse-
hormone (199) (see Chapter 23). The clinical manifestations Friderichsen syndrome, characterized by the following: (a)
of increased ICP include (a) an altered level of consciousness; sudden onset of a febrile illness, (b) large petechial hemor-
(b) dilated, poorly reactive or nonreactive pupils; (c) abnor- rhages in the skin and mucous membranes, (c) cardiovascu-
malities of ocular motility; (d) pathologically brisk lower ex- lar collapse, and (d) disseminated intravascular coagulation.
tremity reflexes; and (e) bradycardia and hypertension, also Of all patients with a meningococcal infection, 10% to 20%
known as Cushing reflex. The development of elevated ICP have a fulminant meningococcal septicemia (206) (Color
should be anticipated and monitored in a child with bacte- Figs. 24.21 to 24.26).
rial meningitis. The absence of papilledema does not exclude Focal neurologic signs, such as cranial nerve palsies with
the presence of increased ICP. Papilledema is rarely observed abnormalities of ocular motility, hemiparesis, visual field
early in the course of increased ICP and is usually not evident defects, and ataxia, may occur early or late in the course of
until increased ICP has been present for at least several hours bacterial meningitis in approximately 15% of children (197).
(197,198). For this reason, the presence of papilledema at the Cranial nerve palsies likely develop as the nerve becomes
time of the initial presentation should raise suspicion of a focal enveloped by exudate in the arachnoidal sheath surrounding
intracranial process such as a brain abscess or other localized the nerve. Alternatively, cranial nerve palsies may be a sign
mass lesion, and it is an indication for CT prior to LP. of increased ICP. The presence of bilateral palsies of the sixth
Seizures occur in 30% to 40% of children with acute bacte- cranial nerve, manifested as weakness of lateral rectus muscles,
rial meningitis, usually during the first 3 days of illness (200). is a well-recognized sign of increased ICP.
In one review of 52 cases of H. influenzae meningitis in chil-
dren, seizures occurred in 44% (23 cases) (198). There has
been a long-standing controversy about whether to do an LP
in febrile children with new-onset seizures. The vast majority
of children who present with a new-onset seizure associated
with fever and who have a normal neurologic examination do
not have meningitis. One series reviewed the results of LP per-
formed on 328 children presenting with their first febrile con-
vulsion. None of the children had meningeal signs. Meningitis
was diagnosed by LP in four children (1.2%). Three of the
children had viral meningitis, and one had H. influenzae men-
ingitis. All four children were younger than 18 months of age.
All the children in this series who were older than 18 months
of age had unequivocal signs of meningitis (201). A similar ob-
servation was made in a review of LP performed on 304 chil-
dren for evaluation of new-onset seizures associated with
fever. There were 15 cases of meningitis, and in only one case
were there no meningeal signs. In that case, the child had viral
meningitis and recovered fully without treatment (202). These
studies suggest that LP should not necessarily be routinely per-
formed in children for evaluation of simple febrile convulsions
in the absence of meningeal signs.
Convulsive seizure associated with fever is a problem
unique to young children. A simple febrile seizure, as defined
by the Consensus Development Meeting on Long-term
Management of Febrile Seizures (1980), occurs between ages
3 months and 5 years in association with fever and is of brief
duration (15 minutes), nonfocal, nonrepetitive, and without
associated neurologic deficits. If the seizure fits this definition FIGURE 24.21 Early appearance of rash in a patient with acute me-
and the child is awake and alert after the seizure, the yield of ningococcemia.

Scheld_Ch24.indd 385 2/21/14 5:42 PM


386 Part III: Bacterial and Mycoplasmal Infections

FIGURE 24.22 Close up of rash in a patient with acute meningo- FIGURE 24.23 Fulminant petechial/purpuric rash in patient with
coccemia. meningococcemia.

FIGURE 24.24 Examples of severe purpura fulminans and peripheral gangrene in two patients with
acute meningococcemia.

FIGURE 24.26 Gross pathologic specimen showing intraventricular


FIGURE 24.25 Gross pathologic specimen depicting adrenal hemor- hemorrhage from a patient with acute meningococcemia and dissemi-
rhage in fulminant acute meningococcemia (Waterhouse-Friderichsen nated intravascular coagulation. Note marked right-to-left herniation
syndrome). (fatal in this case) and periventricular white matter edema.

Scheld_Ch24.indd 386 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 387

Hemiparesis may be caused by vasculitis and cerebral with a few exceptions. The most common bacterial cause of
infarction or may be a sign of the presence of a large sub- meningitis in adults is S. pneumoniae. The clinical presenta-
dural effusion. Subdural effusions commonly develop in the tion of meningitis caused by S. pneumoniae is somewhat dif-
course of bacterial meningitis in children and are not usually ferent from that of meningitis caused by the meningococcus
associated with clinical symptomatology. Subdural effusions or H. influenzae. Adults with pneumococcal meningitis usu-
develop when the infection in the adjacent SAS leads to an ally have an altered mental status on admission and rapidly
increase in the permeability of the thin-walled capillaries and become stuporous or comatose. In addition, recurrent seizure
veins in the inner layer of the dura. The result is leakage of al- activity and focal neurologic deficits are more common in the
bumin-rich fluid into the subdural space, usually a self-limited early stages of pneumococcal meningitis than in meningo-
process. When the inflammatory process subsides, fluid for- coccal or H. influenzae meningitis. These and other factors
mation usually ceases and the fluid in the subdural space is re- may contribute to the continued high mortality rate in pneu-
sorbed (197). Some subdural effusions are, however, clinically mococcal meningitis. S. pneumoniae is associated with the
significant. The development of a hemiparesis or increased highest mortality rate of the major meningeal pathogens. For
ICP may be the consequence of an enlarging subdural effusion example, a mortality of 36.8% was reported from one series
causing mass effect. The presence of a prolonged fever in a of 55 cases of community-acquired pneumococcal meningitis
child with a subdural effusion suggests that the effusion has (212). Death is more likely to occur in patients of advanced
become infected. age, in the absence of meningismus, and in the presence of
Ataxia may be the presenting sign of bacterial meningitis pneumonia, other extraneural complications, or a prolonged
in a child. Ataxia is a sign of vestibular dysfunction, and in duration of illness prior to therapy (7 days). Nevertheless, it
this clinical setting, it suggests the presence of labyrinthitis. appears that most deaths occur later in the disease course as
In most children, it is a transient symptom; however, it has a consequence of cardiorespiratory insufficiency (213). They
implications for prognosis, because auditory and vestibular are not usually caused by early overwhelming CNS damage.
disturbances usually occur together. As such, ataxia is associ- A classic description of the clinical presentation of bac-
ated with postmeningitic hearing loss (207). terial meningitis in adults was presented by Carpenter and
Children with meningitis may develop hyponatremia and Petersdorf (211) in 1962. This review includes 209 cases of
the syndrome of inappropriate secretion of antidiuretic hor- bacterial meningitis: 53 cases were caused by meningococci,
mone (SIADH) (208). The symptoms of hyponatremia and/ 63 by pneumococci, 35 by H. influenzae, and 58 by other bac-
or SIADH are lethargy, stupor, confusion, and/or seizure ac- terial organisms. A reliable history of the onset of symptoms
tivity. When the following criteria are met, the diagnosis of was obtained in 134 patients. Thirty-six patients (27%) had
SIADH should be considered: (a) serum sodium level less sudden onset of headache, confusion, lethargy, and loss of con-
than 135 mEq/L, (b) serum osmolality less than 270 mOsm/L, sciousness and sought hospitalization within the first 24 hours.
(c) urine osmolality greater than two times the serum osmolal- Only 3 of these 36 patients had symptoms of respiratory tract
ity, (d) urine sodium greater than 30 mEq/L, and (e) no clinical infection. In contrast, 71 (53%) of the 134 patients had more
evidence of hypovolemia or dehydration (209). SIADH is not slowly progressive symptoms of meningitis for 1 to 7 days be-
the only cause of hyponatremia in children with bacterial men- fore admission. Of these, 26 patients (37%) had respiratory
ingitis. Hyponatremia may also develop when fluid therapy is symptoms. In 27 (20%) of the 134 patients, an infection in
too aggressive or as a result of the syndrome of cerebral salt the respiratory tract developed 1 to 3 weeks before the first
casting. Regardless of the reason for hyponatremia, the serum symptom of meningitis. The results of physical examination
sodium level should be monitored frequently in every child for meningeal signs were recorded in 199 cases. Either nu-
with bacterial meningitis. This is discussed in greater detail chal rigidity, Kernig signs, or Brudzinski signs were present in
later in this chapter (see the section Therapy). 161 patients (81%). Studies suggest that the latter two signs
Ocular complications, including fundal abnormalities, cra- are less common in adults with meningitis than previously
nial nerve palsies (see earlier discussion), pupillary dysfunc- reported (214). Level of consciousness on admission was re-
tion, and corneal or conjunctival lesions, are actually quite corded in 191 patients (96%); only 9 (5%) patients were alert,
common during the course of bacterial meningitis in children, 48 (24%) were lethargic, 44 (22%) were confused, and the re-
but only one case of transient cataract formation has been mainder were obtunded or comatose (211). Patients with me-
reported in association with meningococcal meningitis. ningococcal meningitis were most often alert, and those with
Bacterial meningitis is at least 30-fold more common in chil- pneumococcal meningitis were more often obtunded.
dren with cochlear implants to address profound hearing loss Geiseler et al. (215) made observations of altered con-
than in controls (210). The major pathogen is S. pneumoniae sciousness at presentation in bacterial meningitis, similar to
and risk factors include use of a positioner to improve trans- those of Carpenter and Petersdorf (211). They recorded level
mission of the electrical signal by pushing the electrode against of consciousness at the time of admission in 1,289 patients
the medial wall of the cochlea (voluntarily withdrawn from with community-acquired bacterial meningitis. Overall, 230
the market by the manufacturer in July 2002); and joint ra- (17.8%) were alert, 672 (52.1%) were irritable or lethargic,
diographic evidence of inner ear malformation and CSF leak. 262 (20.3%) were stuporous or obtunded, and 125 (9.7%)
More cases occurred within 30 days of surgery, but sporadic were comatose and/or convulsing (215). In adults, as in chil-
cases were observed 24 months after implantation (210). dren, lethargy or an altered mental status is the strongest indi-
cator of bacterial meningitis.
Durand et al. (78) reviewed the charts of all cases of acute
Adults bacterial meningitis in individuals 16 years or older at the
Massachusetts General Hospital from 1962 through 1988.
The typical presentation of bacterial meningitis in an adult The most common pathogen of community-acquired menin-
is that of an upper respiratory tract infection during which gitis was S. pneumoniae. Ninety-five percent of patients had
a meningeal symptom, such as headache, nuchal rigidity, fever (temperature 37.7C) on presentation. Neck stiffness
vomiting, or an altered level of consciousness, develops (211) was present in 88% of patients on initial physical exami-
(Table 24.3). The clinical signs and symptoms of bacterial nation. At the time of admission, 61 (22%) were alert, 142
meningitis in an adult are very similar to those in children, (51%) were confused or lethargic, 61 (22%) were responsive

Scheld_Ch24.indd 387 2/21/14 5:42 PM


388 Part III: Bacterial and Mycoplasmal Infections

only to pain, and 17 (6%) were unresponsive to all stimuli. only 57% had meningismus and only 21% complained of
On presentation or during the first 24 hours, 81 (29%) had headache. Pneumococci were the most common etiologic
focal seizures or focal neurologic findings. The most common agent, and these cases were often (58%) accompanied by
predisposing factors for community-acquired meningitis were pneumonia, sinusitis, or otitis media; the overall mortality was
pneumonia, sinusitis, acute otitis media, altered immune state, approximately 40%.
alcoholism, and diabetes mellitus (78). Most elderly patients with meningitis have nuchal rigidity,
In another review of 696 episodes of community-acquired that is, resistance to passive flexion of the neck. Resistance to
bacterial meningitis, the triad of fever, neck stiffness, and passive movement of the neck is a common physical finding
altered mental status was present in only 44% of episodes in elderly patients because of the presence of cervical spon-
(216), although almost all patients (95%) presented with at dylosis. It is important to be able to distinguish between the
least two of four symptoms (i.e., headache, fever, stiff neck, cervical rigidity of cervical spondylosis and nuchal rigidity
and altered mental status). Cerebrovascular complications resulting from meningitis. In nuchal rigidity, the neck resists
may manifest at any time during the course of pneumococcal flexion but can be passively rotated from side to side. In ri-
meningitis. In an observational study of 696 patients with gidity consequent to cervical spinal disease, lateral rotation,
community-acquired bacterial meningitis, cerebral infarction extension, and flexion of the neck are all associated with
occurred in 174 (25%) episodes and was seen in 128 (36%) of resistance. Similarly, hypertonicity of the neck muscles result-
352 patients with pneumococcal meningitis (217). ing from disease of basal ganglia, such as parkinsonism, can be
Pneumonia is present on admission in 25% to 50% of adults distinguished from true nuchal rigidity.
with pneumococcal meningitis (211). Acute and chronic oti- Specific comments should be made about the presentation
tis media are also predisposing conditions for pneumococcal of nontraumatic, spontaneous gram-negative bacillary men-
meningitis. In one series of 178 patients with pneumococcal ingitis in the elderly or debilitated patient. In these patients,
meningitis, acute otitis media was present in 59 (33.1%) (215). the classic signs and symptoms of meningitis may be subtle
When pneumonia or otitis media is not present, the possibility at initial presentation. They may have only low-grade fever
of a dural sinus fistula should be considered. S. pneumoniae and altered mental status without headache or nuchal rigid-
is the most common causative agent in meningitis following ity; however, patients with spontaneous gram-negative bacil-
head trauma (basilar skull fracture) or meningitis associated lary meningitis tend to have a rapidly progressive fulminant
with a structural defect (either congenital or traumatic in course associated with bacteremia, shock, and coma (223).
origin) that creates a communication between the paranasal The elderly patient with gram-negative meningitis may rapidly
sinuses, nasopharynx, or middle ear and the SAS (218). become comatose after presenting with what at first appeared
In adults aged 15 to 60 years, underlying host factors may to be a minor illness. Once coma develops, nuchal rigidity may
increase the risk for meningitis while simultaneously blunt- not be present, because this sign is lost during deep coma.
ing its presenting signs. Such predisposing factors include In a recent 30-year study of 185 patients 65 years of age
malignancy, alcoholism, sickle cell disease, diabetes, organ and older, the diagnosis of community-acquired bacterial
transplantation, splenectomy, high-dose steroid therapy, and meningitis was more difficult because of the absence of char-
long-term dialysis. In these clinical settings, the symptoms of acteristic meningeal signs (224); compared with adult patients
meningitis may include altered sensorium, persistent head- younger than 65 years of age, the older patients showed greater
ache, or new-onset seizures. Fever or nuchal rigidity may not neurologic severity with a high number presenting with coma
develop (219). on admission, seizures, and hemiparesis.
The occurrence of H. influenzae meningitis in an adult
should prompt consideration of the presence of (a) otitis
media, (b) paranasal sinusitis, (c) other parameningeal foci of Posttraumatic Meningitis
infection, (d) CSF leak from previous head trauma, or (e) a
Bacterial meningitis may develop following a traumatic head
concurrent pneumonia, pharyngitis, or immunodeficiency dis-
injury that produces a dural fistula between the SAS and the
ease. The clinical presentation of H. influenzae meningitis in
nasal cavity, paranasal sinuses, or middle ear. The infection
adults is typical of bacterial meningitis and includes headache,
may develop shortly after the injury or may not occur until
fever, altered mental status, and nuchal rigidity (220).
months to years later (225). Traumatic head injury is the most
common cause of recurrent meningitis in the adult (220).
Elderly Conversely, congenital fistulous connections to the CNS, often
via the middle ear in association with Mondini dysplasia, are
Meningitis should be suspected in every elderly patient who the most common underlying process in children with recur-
is febrile and either disoriented, stuporous, or comatose rent bacterial meningitis (226). An immunodeficiency state
(Table 24.3). In a review of 54 cases of bacterial meningitis may also be instrumental in the development of this syndrome.
in the elderly, confusion was present in 92% (12 of 13) of the A dural fistula develops when the force of the injury is suf-
patients with pneumococcal meningitis and in 78% (7 of 9) of ficient to fracture bone and tear the dura. The most common
those with gram-negative meningitis on initial presentation. site for dural fistula is in the anterior cranial fossa, in the area
This review compared the clinical presentation of bacterial of the cribriform plate. Here, the bone is very thin and the
meningitis in the elderly (patients aged 50 years and older) dura is tightly adherent to the bone. A fracture in this area
with that in younger patients (aged 15 to 49 years). On initial allows CSF to leak through torn arachnoid and dura into the
presentation, the incidence of more severe abnormalities of nasal cavity, resulting in CSF rhinorrhea (225). There may also
mental status in the older age-group with bacterial meningitis be loss of the sense of smell, or anosmia. CSF rhinorrhea can
was statistically different from that of the younger group, and be distinguished from nasal secretions by testing the fluid for
concurrent pneumonia was present more often in the older
2-transferrin.
patients than in the younger group (221). In another review Physical signs indicating a basilar skull fracture with the
of CNS infections in patients older than 65 years of age at potential for development of a dural fistula and meningitis in-
the Mount Sinai Hospital in New York from 1970 through clude periorbital ecchymoses, bruising behind the ear (Battle
1985, 28 cases of bacterial meningitis were identified (222). sign), hemotympanum, and/or blood in the external auditory
Although fever (often of low grade) was uniformly present, canal (225).

Scheld_Ch24.indd 388 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 389

In most patients, CSF rhinorrhea ceases spontaneously.


Approximately one in four patients with CSF rhinorrhea de- Immunosuppressed Hosts
velops meningitis (225); the reported frequency ranges from
9% to 36%. Surgery is indicated in patients who develop The risk for development of bacterial meningitis in an im-
meningitis with persistent rhinorrhea. The management of munocompromised patient depends on a number of factors,
meningitis occurring in this setting and the approach to dem- such as the underlying disease and its treatment, the duration
onstration of the location of the dural fistula is discussed later of immunosuppression, and the type of immune abnormality
in this chapter. (232). There are four major types of host defense abnormali-
ties in the immunocompromised patient: (a) defects in T-
lymphocytemacrophage function (cell-mediated immunity);
(b) defects in humoral immunity; (c) defects in the number and
Meningitis Following function of neutrophils; and (d) loss of splenic function from
Neurosurgical Procedures surgery, disease, or radiotherapy, resulting in the inability to
remove encapsulated bacteria. Knowledge of the type of host
Meningitis complicating a neurosurgical procedure, such as a defense abnormality is often helpful in predicting the infecting
craniotomy, is usually insidious in onset and difficult to dis- organism (233,234).
tinguish clinically from the neurologic abnormalities expected Patients with defects in cell-mediated immunity include
in the postoperative period. Although an altered level of con- (a) those with lymphomas, particularly Hodgkin disease;
sciousness and signs of meningeal irritation may be expected (b) organ transplant recipients; (c) patients treated with daily
in the postoperative period, the presence of fever or prolonged corticosteroid therapy; and (d) patients with AIDS. These
obtundation should prompt an examination of the CSF. patients are most susceptible to CNS infection by microorgan-
Approximately 60% to 70% of all cases of meningitis com- isms that are intracellular parasites, the eradication of which
plicating a neurosurgical procedure, with the exception of a depends on an intact T-lymphocytemacrophage system (233).
shunting procedure, are caused by gram-negative bacilli (223). L. monocytogenes is a causative organism of bacterial menin-
The remainder is caused by staphylococci, predominantly gitis in patients with defective cell-mediated immunity due to
S. aureus. In the postneurosurgical patient, K. pneumoniae, hematologic malignancies, organ transplantation, pregnancy,
Acinetobacter baumannii, and E. coli are the most common chronic corticosteroid therapy, alcoholism, and advanced age
infecting gram-negative organisms. Craniotomy for trauma (235). The incidence of L. monocytogenes meningitis in HIV-
or for tumor represents the most common surgical proce- infected individuals is low due to pneumocystis prophylaxis
dure associated with postoperative gram-negative bacterial with TMP-SMX. The clinical presentation of L. monocyto-
meningitis (227). genes meningitis includes fever and headache, as well as an
Although the surgical insertion and subsequent constant increased tendency for focal neurologic deficits and seizures
presence of an indwelling ventriculoperitoneal (VP) or ventric- during the initial presentation. Meningitis caused by this
uloatrial (VA) shunt catheter for decompression of hydroceph- organism may also present with a clinical picture suggestive of
alus allow bacteria to enter the CSF space, signs of meningitis an acute brainstem disorder or rhombencephalitis, with signs
usually do not accompany these infections in the early stages. of ataxia, cranial nerve deficits, and nystagmus (236).
The bacteria involved in early shunt infection gain entry to Patients with defective humoral immunity are unable to
the lumen of the shunt from a contaminated wound or from mount an antibody response to bacterial infection, and they
the patients skin surface at the time of operation (228). The are, therefore, unable to control infection caused by encap-
initial symptoms of shunt infection are nonspecific and include sulated bacteria. Patients with this type of host defense ab-
fever, nausea, vomiting, and lethargy. Fever is the most com- normality include those with chronic lymphocytic leukemia,
mon manifestation of shunt infection. Virtually all patients multiple myeloma, or Hodgkin disease following chemother-
have temperatures greater than 37.8C, and most have tem- apy and radiotherapy, among others. These patients are at par-
peratures of 38.8C or more (229). Fever is often the sole ticular risk for meningitis caused by S. pneumoniae, Hib, and
manifestation of infection in patients with VA shunts, whereas less commonly N. meningitidis. The presentation of meningitis
patients with infected VP shunts are more likely to present in these patients is often that of a fulminant illness resulting in
with signs of shunt malfunction and/or signs of inflammation death in several hours.
around the shunt reservoir or along the course of the tub- Patients with splenectomy may develop (a) overwhelming
ing (229,230). Signs of shunt malfunction are secondary to bacteremia and fulminant meningitis with the same organ-
progressive hydrocephalus and, in children, include enlarg- isms, resulting from loss of the filtering function of the splenic
ing cranial circumference, tense nonpulsatile fontanelle, and sinusoids in removing encapsulated bacteria from the blood-
papilledema. Signs of shunt malfunction may be associated stream, and (b) a reduced ability to produce IgM opsonizing
with signs of meningitis. Results of examination of CSF from antibodies (233,234).
the lumbar area may be negative; therefore, CSF should be Patients with neutropenia are at particular risk for meningitis
obtained by aspiration from the infected shunt reservoir. caused by P. aeruginosa and members of the Enterobacteriaceae
Infections of the CNS may also develop when subcuta- family (234). The clinical presentation of bacterial meningitis in
neous CSF reservoirs, such as Ommaya and Rickham reser- patients with neutropenia may be subtle, consisting of low-grade
voirs, are placed for therapeutic purposes. These and other fever and lethargy or a change in headache pattern (233). Signs
types of indwelling intraventricular catheters may lead to of meningeal irritation depend on the hosts ability to mount
meningitis with coagulase-negative staphylococci, S. aureus, an inflammatory response; therefore, in the neutropenic patient
Corynebacterium species, or gram-negative bacilli. Infections they are often minimal.
usually occur within the first 3 months after insertion of the
device and, as with infections of VP or VA shunts, are prob-
ably the consequence of contamination by skin flora during DIFFERENTIAL DIAGNOSIS
implantation or subsequent use for therapeutic purposes.
In these patients, signs of meningitis are usually not present, Although the diagnosis of meningitis is usually made by
but most will complain of fever, lethargy, headache, or nausea examination of the CSF, the decision to perform spinal fluid
and vomiting (231). analysis is based on the clinical presentation. When the signs

Scheld_Ch24.indd 389 2/21/14 5:42 PM


390 Part III: Bacterial and Mycoplasmal Infections

and symptoms suggest meningitis, and the decision is made developed during or after treatment for frontal sinusitis, mas-
to examine the CSF, the next step is to be certain that a focal toiditis, or otitis media. If the abscess is large, mild alterations
intracranial mass lesion does not exist that may predispose of consciousness may occur; however, focal neurologic defi-
to brain herniation following LP. If the history and neuro- cits, seizures, and signs of increased ICP do not develop until
logic examination suggest a focal mass lesion, then LP should the infection has extended into the subdural space or a deeper
be delayed until a neuroimaging procedure, either a cranial intraparenchymal complication has occurred (244).
CT scan, without and with contrast enhancement, or a cranial The decision to delay LP until CT or MRI scan is obtained
magnetic resonance imaging (MRI) scan is obtained. may be made when the patient does not appear to be seriously
LP is relatively contraindicated in the presence of a focal ill or when there is uncertainty about the findings of neurologic
mass lesion because of the danger of brain herniation. However, examination. Patients with viral meningitis usually do not ap-
it has become common practice to delay LP until a CT or MRI pear as ill as patients with bacterial meningitis and often have
scan has been obtained despite the absence of focal neurologic had symptoms for several days. When the history suggests a
deficits by history or examination. The time involved in waiting focal onset to the headache or a transient neurologic symptom,
for a CT or MRI scan significantly delays treatment, and delay LP is best delayed until a CT or MRI scan has been obtained.
in treatment is the most critical factor in determining morbid- The initial symptoms of viral meningitis are fever, headache,
ity and mortality in bacterial meningitis. Therefore, if a CT or lethargy, myalgias, and nuchal rigidity. There are several distin-
MRI scan is to be performed, antimicrobial therapy should be guishing clinical features of viral meningitis: (a) viral meningi-
initiated promptly, pending results. In the absence of an altered tis has a more insidious onset and a slower progression than
level of consciousness, focal neurologic signs, and/or papill- bacterial meningitis; (b) patients with viral meningitis often
edema, an LP can be safely performed without first obtaining complain of an incapacitating headache that is not relieved by
a CT or MRI scan. Although CT is commonly performed be- analgesics, but they are otherwise awake and alert; (c) the fever
fore LP in adults with suspected meningitis, the vast majority is usually higher in bacterial meningitis than in viral meningitis;
of scans are unnecessary and unlikely to reveal abnormalities, and (d) although generalized malaise may be present, stupor,
and clinical characteristics can be used rapidly to exclude pa- obtundation, and coma do not occur in viral meningitis (219).
tients that are unlikely to have abnormal findings on CT (237). Altered level of consciousness, focal neurologic deficits, and
Focal infectious lesions that have clinical presentations similar new-onset seizure activity are symptoms of a viral encephalitis,
to those of meningitis and that can result in significant morbid- meningoencephalitis, or bacterial meningitis. The presentation
ity if LP is unknowingly performed include brain abscess, sub- of herpes simplex virus (HSV) encephalitis is often subacute
dural empyema, and epidural abscess. The clinical presentation and on examination is characterized by (a) fever, confusion,
of each of these disorders has similarities and distinguishing or a change in behavior; (b) new-onset seizure activity; and/or
features when compared with that of meningitis. (c) focal neurologic deficits. A history of hemicranial headache
The most common symptom of a brain abscess is a hemicra- of several days duration, preceding the onset of the confusional
nial or generalized headache, generally seen in 70% to 75% of state, is a classic presentation of this illness. HSV has a predi-
patients (238242). A brain abscess presents as an expanding lection for the temporal and orbitofrontal areas; therefore, a
intracranial mass lesion rather than as an infectious process; as change in mentation or behavior is a common finding (249).
such, fever is present in only 45% to 50% of patients and usually Signs and symptoms of meningitis represent the most com-
is not prominent. More than 50% of patients have focal neuro- mon neurologic presentation of Lyme disease. Patients have
logic deficits, and one third of patients present with new-onset headache, stiff neck, low-grade fever, a unilateral or bilateral
focal or generalized seizure activity. The findings on neurologic (in 25% of cases) facial nerve palsy, or a radiculitis. The char-
examination are related both to the site of the abscess and to acteristic skin lesion of Lyme disease, erythema migrans (EM),
the presence of raised ICP caused by an expanding mass lesion. precedes the symptoms of meningitis in approximately 80% to
Hemiparesis is the most common sign of a frontoparietal lobe 90% of patients. Signs and symptoms of meningitis occur weeks
abscess. A disturbance of language or behavior or an upper hom- to a few months after the initial infection, or they may be the
onymous quadrantanopia is the sign of a temporal lobe abscess. first manifestation of the disease without antecedent EM (250).
Ataxia is the most common sign of a cerebellar abscess. Nuchal The presence of a rash with meningitis suggests meningococ-
rigidity rarely occurs until the abscess has ruptured into the ven- cemia. As has been discussed, the classic lesions associated with
tricle or until infection has spread to the SAS. Sudden worsening fulminating meningococcal septicemia are large petechial hem-
of the headache, accompanied by new-onset meningismus, may orrhages in the skin and mucous membranes. Between 50% and
signify rupture of the abscess into the ventricular space (243). 75% of children with meningococcal meningitis have a purpu-
Most patients with a subdural empyema initially complain ric or petechial rash, principally on the trunk and lower ex-
of headache that is localized to the side of the subdural in- tremities. Petechiae are found in the skin, mucous membranes,
fection. The headache becomes increasingly more severe and and conjunctivae, but not in the nailbeds, in meningococcemia.
generalized, and it is followed by an alteration in the level of Petechiae are also sometimes seen on the trunk and extremi-
consciousness. Fever, chills, and nuchal rigidity are present in ties in echovirus type 9 meningitis, acute staphylococcal endo-
most cases. Focal neurologic deficits are present in 80% to carditis, and rarely pneumococcal or H. influenzae meningitis
90% of patients, and they include hemiparesis or hemiplegia, except in asplenic patients (198,204). Petechiae may be found
paralysis of horizontal gaze to the side opposite the lesion, in the nailbeds in acute staphylococcal endocarditis. Petechial
and focal or generalized seizures (244). The diagnosis should rashes should be promptly examined microscopically in the
be considered in patients with acute bacterial sinusitis in com- initial evaluation of meningococcemia after aspiration or after
bination with severe intractable headache, varying degrees of making a touch preparation on a glass slide; approximately
altered level of consciousness, focal neurologic deficits, and/or 70% of these preparations will reveal the organisms, usually
signs of meningeal irritation (245,246). The presentation of a within vacuolated neutrophils. In fulminant meningococcemia,
posterior fossa subdural empyema includes severe headache, the organisms may be visualized in the peripheral blood smear.
vomiting, marked nuchal rigidity, cranial nerve deficits, and Although the sensitivity of this method is low, this simple test
pupillary abnormalities (247); cerebellar signs were elicited in should always be performed in suspected meningococcemia.
only 40% of patients in one study (248). Headache, fever, rash, and altered mental status are symp-
A typical presentation of an intracranial epidural abscess toms of rickettsial infections (see Chapter 27) and, as such, enter
is an unrelenting hemicranial headache and fever that have into the differential diagnosis of meningitis. A petechial rash

Scheld_Ch24.indd 390 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 391

is characteristic of Rocky Mountain spotted fever (RMSF), by pallor, unstable blood pressure, diaphoresis, tachycardia, and
which is caused by Rickettsia rickettsii. The rash of typhus is a arrhythmias. A leukocytosis of 15,000 to 30,000 cells/mm3, with
faint macular-papular pink rash (251). The rash of RMSF con- a shift to the left, is common. Liver function abnormalities are
sists initially of 1- to 5-mm pink macules that are often noted usually seen, but the most specific laboratory abnormality in this
first on the wrist and ankles and then spread centrally to the disorder is marked elevation in the serum creatine kinase (CK)
chest, face, and abdomen. The rash of RMSF usually does not concentration, usually exceeding 10,000 IU/L (253).
involve the mucous membranes. Petechial lesions in the axillae Signs of a posterior fossa tumor are stiff neck, cranial nerve
and around the ankles, accompanied by lesions on the palms abnormalities, gait disturbance, vomiting, cerebellar deficits,
and soles of the feet, are characteristic of RMSF, but this clas- and occasionally an altered level of consciousness.
sic pattern is often absent. The macules will initially blanch A cranial CT or MRI scan and examination of the CSF
with pressure, but after a few days they become fixed and turn will narrow the differential diagnosis. The possibility of the
dark red or purple. Diagnosis can be made by biopsy of the presence of increased ICP should be considered before LP.
lesions and staining of the specimen with fluorescent antibod- Increased ICP is an expected complication of bacterial menin-
ies to R. rickettsii (252). A negative result does not exclude gitis and is not a contraindication to LP.
RMSF, because sensitivity of this test is only 70%. The clinical signs of increased ICP are (a) a dilated, nonreac-
The characteristic rash caused by an enterovirus consists of tive pupil; (b) drowsiness; (c) abnormalities of ocular motility,
erythematous macules and papules on the face, neck, trunk, the most common of which are the consequence of unilateral or
and to a lesser degree the extremities. Rarely, the rash associ- bilateral palsies of the sixth cranial nerve; and (d) bradycardia
ated with enteroviral infection may become petechial in nature. and hypertension, the Cushing reflex. Pupillary dilation is usu-
The rash of Lyme disease, EM, begins as a red macule or ally secondary to parenchymal midbrain distortion from either
papule at the site of the tick bite. It then expands centrifugally raised ICP or transtentorial herniation. Drowsiness or stupor
as an erythematous lesion with central clearing. This may be is often the first sign of increasing ICP and is caused by inter-
the only lesion, or the disease may disseminate to form mul- ference with the reticular activating system in the brainstem.
tiple secondary ringlike lesions. Symptoms of meningitis may If raised ICP appears likely and a focal intracranial mass lesion
develop while the skin lesions are still present, or they may has been excluded by CT or MRI scan, LP can usually be safely
begin 1 to 6 months after the skin lesions have resolved (250). performed. When the decision is made to delay LP until a CT or
Noninfectious neurologic disorders that have clinical presen- MRI scan has been obtained, blood cultures should be obtained
tations similar to those of meningitis are subarachnoid hemor- and intravenous antibiotics and dexamethasone therapy begun
rhage, neuroleptic malignant syndrome, and posterior fossa while awaiting results of CT or MRI scan (see later discussion).
tumors. The classic presentation of a subarachnoid hemorrhage Intravenous antibiotics usually do not sterilize the CSF in the time
is the sudden onset of a severe, excruciating headache, or a sud- it takes to obtain a CT or MRI scan and spinal fluid. Blood cul-
den transient loss of consciousness followed by a severe head- tures may identify the infecting organism in 50% to 80% of cases
ache. Most patients complain of vomiting. Syncope accompanies of bacterial meningitis (although this frequency varies with the
the explosive onset of headache in about 50% of cases. Nuchal causative organism), and they are more often positive in patients
rigidity develops within a few hours of the onset of a subarach- who have not received prior treatment with oral antibiotics (189).
noid hemorrhage and is usually associated with a change in the
level of consciousness. Low-grade fever may develop within sev-
eral days. When an intracranial aneurysm ruptures into the brain LABORATORY DIAGNOSIS
parenchyma, a focal neurologic deficit is usually present. A uni-
lateral palsy of the third nerve, with a dilated, nonreactive pupil,
is suggestive of third nerve compression by an aneurysm at the Cerebrospinal Fluid
junction of the posterior communicating artery and the internal
carotid artery. The triad of headache, neck stiffness, and vomiting The typical CSF findings of bacterial and aseptic meningitis
should raise suspicion of a warning leak from an aneurysm (252). are compared in Table 24.4.
The symptoms of neuroleptic malignant syndrome (NMS) are
fever, generalized lead-pipe rigidity (including cervical rigidity),
Opening Pressure
fluctuating level of consciousness (ranging from agitation to stupor The first step in examination of the CSF is measurement of
and coma), and autonomic instability. The latter is characterized the opening pressure with an air-water manometer. This step

TA B L E 2 4 . 4
TYPICAL CEREBROSPINAL FLUID FINDINGS IN BACTERIAL VERSUS ASEPTIC MENINGITIS

CSF Parameter Bacterial Meningitis Aseptic Meningitis

Opening pressure 180 mm H2O Normal or slightly elevated


Glucose 40 mg/dl 45 mg/dl
CSF-to-serum glucose ratio 0.4 0.6
Protein 50 mg/dl Normal or elevated
White blood cells 10 to 10,000/mm3neutrophils predominate 502,000/mm3lymphocytes predominate
Gram stain Positive in 7090% of untreated cases Negative
Lactate 3.8 mmol/L Normal
C-reactive protein 100 ng/mL Minimal
Limulus lysate assay Positive indicates gram-negative meningitis Negative

Scheld_Ch24.indd 391 2/21/14 5:42 PM


392 Part III: Bacterial and Mycoplasmal Infections

is often neglected, but knowledge of the presence of raised to 5 mononuclear cells (lymphocytes and monocytes)/mm3; a
ICP is important in management of the patient. Normal CSF WBC count of greater than 10 cells indicates a pathologic pro-
pressure, with the patient in the lateral recumbent position, is cess such as infection. Normal CSF does not contain PMN leu-
usually defined as less than 180 mm H2O (254). However, nor- kocytes; however, following centrifugation, an occasional PMN
mal opening pressure can be as high as 250 mm H2O in obese leukocyte may be seen. It has been stressed that for the CSF to
patients (255). CSF pressure should not be measured with the be considered normal, no more than a single PMN leukocyte
patient in a seated position. If the spinal needle is inserted with should be seen in the differential count, accompanied by a total
the patient seated, the patient should then be moved to a lat- WBC count of less than 5 cells/mm3 (254,256). However, most
eral recumbent position and the opening pressure recorded. CSF differential counts are now performed on cytocentrifuged
Elevated CSF pressure in the range of 200 to 500 mm H2O is specimens in hospital laboratories. In these preparations, a few
common in bacterial meningitis. PMN leukocytes may be seen even in the absence of disease
(i.e., when minimal blood contamination is present) or in as-
Appearance sociation with a high peripheral leukocyte concentration (258).
A traumatic puncture or an intracerebral or subarachnoid
Normal CSF is clear. The presence of more than 200 WBCs/ hemorrhage introduces RBCs and WBCs into the CSF. The
mm3, more than 400 red blood cells (RBCs)/mm3, bacteria correction factor for the WBC count in the presence of blood
(105 CFU/mL), or an elevated protein concentration makes in the CSF is as follows: (a) If the peripheral RBC and WBC
the fluid appear cloudy or turbid. When the LP is traumatic counts are normal, then 1 WBC/700 RBCs can be subtracted
and the initial CSF sample appears bloody, the fluid should from the total WBC count in CSF; and (b) in the presence of
clear as flow continues. Xanthochromia refers to a yellow or an abnormal peripheral WBC or RBC count, the following
yellow-orange color in the supernatant of centrifuged spinal formula can be used (258), although valid studies on the diag-
fluid; it may be used to distinguish CSF that is bloody second- nostic accuracy in meningitis are lacking:
ary to subarachnoid hemorrhage from CSF that is bloody as
a result of a traumatic LP. When CSF is bloody secondary to True WBC (CSF)  Actual WBC (CSF) WBC (Blood)
traumatic LP, the supernatant of the centrifuged fluid is clear.  RBC (CSF)/RBC (Blood)
In subarachnoid hemorrhage, the supernatant is xanthochro-
mic within 2 hours after the hemorrhage. Elevated CSF pro- Generalized seizures may induce a transient CSF pleocyto-
tein concentrations (150 mg/dL) also cause xanthochromia sis consisting predominantly of PMN leukocytes. However, to
(254,256) and are the usual reason for xanthochromia in attribute a CSF pleocytosis to seizure activity, the following
bacterial meningitis. criteria should be met: (a) The fluid should be clear and color-
less, (b) the opening pressure should be normal, (c) the CSF
Glucose glucose concentration should be normal, (d) the WBC count
should not exceed 80 cells/mm3, (e) there should be no men-
The normal CSF glucose concentration is greater than 45 mg/ ingeal signs or other evidence of infection, and (f) Gram stain
dL. A glucose concentration of less than 40 mg/dL occurs in ap- results should be negative (259). Even if these conditions are
proximately 58% of patients with bacterial meningitis (257). met, patients should usually be treated with antibiotics until
However, the CSF glucose may be falsely low in the presence the results of bacterial cultures are known. There also remains
of hypoglycemia, or it may be erroneously interpreted as nor- the possibility that a viral encephalitis, with a predominance
mal in the presence of CNS infection when the serum glucose of PMN leukocytes in CSF, is the cause of the seizure activity.
is elevated. An accurate interpretation of the CSF glucose con- In large reported series of bacterial meningitis, in 90% of
centration is done by determining the CSF-to-serum glucose cases, there are greater than 100 WBCs/mm3 in the CSF, and
ratio. A normal CSF-to-serum glucose ratio is about 0.6 (257). in 65% to 70% of cases, there are greater than 1,000 WBCs/
Values less than 0.31 are an indication of low CSF glucose, mm3 (211,215,260,261). The differential count usually shows
and they are observed in approximately 70% of patients with a predominance of PMN leukocytes. In about 10% of cases of
bacterial meningitis (257). A decreased CSF-to-serum glucose bacterial meningitis, there may be a predominance of lympho-
ratio is also consistent with fungal or tuberculous meningitis, cytes early in the infection, especially if the total WBC concen-
carcinomatous meningitis, mumps encephalitis, subarachnoid tration is less than 1,000/mm3. In one series, 32% (13 of 41) of
hemorrhage in 15% to 20% of patients, and several other patients with bacterial meningitis with a CSF WBC concentra-
conditions (258). tion of 1,000 cells/mm3 or less had a predominance of lympho-
cytes (262). In addition, in about 20% to 75% of patients with
Protein viral meningitides, the CSF may initially have a predominance
Any process that disrupts the BBB results in an elevated of PMN leukocytes, with an eventual shift (over the course of
CSF protein concentration. Values greater than 50 mg/dL in several hours) to a monocytic predominance. This has led to
CSF obtained from the lumbar SAS, as well as ventricular CSF controversy about the necessity for repeated LP and the time
protein concentrations greater than 15 mg/dL, are considered period in which a repeated LP should be obtained to demon-
abnormal. When the LP is traumatic and there is blood in strate a shift in cell type. It is our feeling that a repeated LP is
the CSF, the true protein concentration is corrected by sub- usually not necessary unless there is further clinical deteriora-
tracting 1 mg of protein per deciliter for every 1,000 RBCs in tion. In the presence of a lymphocytic pleocytosis, the results
CSF (258). of CSF chemistries, Gram stain, and other tests (see later dis-
cussion) suggest the diagnosis. If bacterial meningitis is sus-
pected, even though there is a predominance of lymphocytes,
White Blood Cell Count the patient should be treated with antibiotics until the results
The CSF abnormalities characteristic of bacterial meningitis in- of bacterial cultures are known.
clude a polymorphonuclear (PMN) pleocytosis, a low glucose
concentration, and an elevated protein concentration. The
Gram Stain and Culture
CSF should be examined promptly after it is obtained, because
WBCs in the CSF begin to disintegrate after about 90 minutes. Examination of CSF by Gram stain allows for rapid, accurate
The normal WBC count in the CSF of adults and children is 0 identification of the infecting organism. If the CSF is cloudy,

Scheld_Ch24.indd 392 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 393

smears should be obtained from fresh, uncentrifuged fluid for phosphokinase) and fibrin-degradation products; however,
Gram stain. If the CSF is clear, smears should be obtained the elevations are nonspecific. Tests for these compounds are
from the centrifuged sediment. The Gram stain is positive in rarely, if ever, performed in hospital laboratories.
identifying the organism in 60% to 90% of cases of bacterial
meningitis (257,263). However, the probability of detecting Combinations of Cerebrospinal Fluid Tests
bacteria on a Gram-stained specimen depends on the number
of organisms present. Most smears will be positive when the Although many of the aforementioned tests commonly per-
CSF bacterial concentration is greater than 105 CFU/mL. Only formed on CSF may suggest the diagnosis of bacterial men-
25% of smears are positive when the bacterial concentration ingitis, none is irrefutable evidence of this disease, except a
is 103 CFU/mL or less (197). positive culture and/or positive stains. Combinations of test
results with clinical parameters may permit a more accurate
assessment of the probability of bacterial versus viral
Cerebrospinal Fluid Lactate meningitis. This approach appears to have merit, as empha-
The lactic acid concentration in CSF has been reported to be sized in a retrospective review of 422 patients with acute
useful in differentiating between bacterial and viral meningitis, meningitis at Duke University (271). The following CSF pa-
especially in those patients who have been partially treated with rameters were individual predictors of bacterial meningitis
antibiotics prior to examination of the CSF, as well as in those with greater than 99% certainty: glucose level of less than
patients with low CSF WBC concentrations. In a European 34 mg/dL; CSF-to-blood glucose ratio less than 0.23; leuko-
study, the lactic acid in CSF was measured in 50 patients with cyte counts more than 2,000/mm3 or neutrophil counts more
acute bacterial meningitis. In 46 patients (92%), the CSF lac- than 1,180/mm3. Although any one of these results predicted
tate concentration was 3.5 mmol/L or greater. The investigators bacterial meningitis with high probability, none could rule it
in this study concluded that CSF lactate was useful in the diag- out. A multiple regression model using four parameters (age,
nosis of acute bacterial meningitis if it was 3.5 mmol/L or more month of onset, CSF-to-blood glucose ratio, and CSF neu-
(264). Other studies have demonstrated that in most cases of trophil concentration) proved highly reliable in separating
acute bacterial meningitis, the CSF lactate concentration is bacterial from viral meningitis. Although the model requires
3.8 mmol/L or more (254). Although the sensitivity of the CSF further validation, a nomogram is included in the article and
lactate level is high for bacterial meningitis, its specificity is low. should be consulted for more precise analysis of gram-negative
In a review of the lactic acid concentrations in 493 samples cases (271).
of CSF from 434 adults with various CNS conditions, the A number of other studies have examined combinations
lactate concentration was greater than 35 mg/dL in 50 cases. of clinical features, with or without test results, to develop
Only 19 of the 50 cases of infective meningitis were caused by models in an attempt to accurately predict the likelihood of
either bacterial or viral pathogens. Although the lactic acid bacterial meningitis compared to other potential etiologies
concentration was elevated in most cases of bacterial menin- (most often viruses) (272). In several retrospective studies of
gitis in this study, the CSF samples with elevated lactic acid immunocompetent patients older than 1 month of age with
concentrations had cell counts and chemistries suggestive of acute bacterial or viral meningitis, a CSF glucose concentra-
bacterial meningitis; therefore, the elevated lactate concentra- tion less than 34 mg/dL, a CSF-to-blood glucose ratio less than
tion provided little additional information (265). In this re- 0.23, a CSF protein concentration greater than 220 mg/dL,
view, as in others, an elevated CSF lactic acid concentration more than 200 leukocytes per cubic millimeter of CSF, and
was nonspecific. Other causes of elevated CSF lactate concen- more than 1,180 neutrophils per cubic millimeter of CSF were
trations include recurrent seizure activity, cerebral ischemia, found to be individual predictors of bacterial rather than viral
hypocapnia, closed head injury, neoplasms, and craniotomy meningitis, with 99% certainty or better. Many other pre-
(254,265). Although the source of the CSF lactate is debated, diction models have been developed. In a recently published
cerebral hypoxia/ischemia, anaerobic glycolysis, vascular metaanalysis of bacterial meningitis score validation stud-
compromise, and metabolism of the CSF leukocytes are all ies in which 5,312 patients were identified from eight stud-
potentially important factors (266). ies, 4,896 (92%) had sufficient clinical data to calculate the
Additional studies have examined whether elevated CSF Bacterial Meningitis Score, which identifies children with CSF
lactate concentrations are useful in differentiating bacterial pleocytosis who are at very low risk of bacterial meningitis
from nonbacterial meningitis in patients who have not received (low-risk features were negative CSF Gram stain, CSF abso-
prior antimicrobial therapy (267). Two metaanalyses, one in- lute neutrophil count 1,000 cells/mm3, CSF protein 80 mg/
cluding 25 studies with 1,692 patients (adults and children) dL, and peripheral absolute neutrophil count 10,000 cells/
(268) and the other including 31 studies with 1,885 patients mm3) (273). The combined sensitivity was 99.3%, specificity
(269), concluded that the CSF lactate concentration is useful 62.1%, and negative predictive value 99.7%, indicating that
in the differentiation of bacterial from aseptic meningitis. In this scoring system could be used to assist clinical decision
another study of adult patients, the CSF lactate concentration making for the management of children with CSF pleocytosis.
had a negative predictive value 99% and positive predictive If used, these models should be limited to the age cohort in
value 82% for bacterial meningitis when it was 3.8 mmol/L or which they were developed. Despite the positive results of this
higher (270). However, in patients who received antimicrobial metaanalysis and other studies, clinical judgment should con-
therapy prior to LP, CSF lactate concentrations had a substan- tinue to be used in decisions about the need for administration
tially lower sensitivity compared to those who had not been of empirical therapy in patients with suspected bacterial men-
treated with antimicrobial agents (269), such that the useful- ingitis (272). Prediction models may be most useful in doubt-
ness of CSF lactate in patients pretreated with antimicrobial ful cases, when they can be used to suggest a reconsideration
therapy is probably limited. of the diagnosis.

Other Tests Partially Treated Meningitis


The CSF concentration of several other substances also in- The effect of oral antibiotic therapy on CSF analysis was studied
creases in the presence of bacterial meningitis, including in two prospective studies of 281 children with Hib meningitis.
various enzymes (e.g., lactate dehydrogenase and creatine Ninety-four (33%) children had been treated with more

Scheld_Ch24.indd 393 2/21/14 5:42 PM


394 Part III: Bacterial and Mycoplasmal Infections

than one dose of antibiotics within 1 week before admission. The limulus amebocyte lysate test can detect minute quantities
Compared with results in untreated children, the results of CSF of endotoxin (e.g., 10 ng/mL) in the CSF. It is reported to have a
analysis in children pretreated with antibiotics showed sig- sensitivity of 77% to 100%, with some studies reporting sensi-
nificant decreases in the percentage of neutrophils (p  .03), tivities of 97% to 99% for detecting gram-negative endotoxin.
protein concentration (p  .001), and percentage with a posi- It has been recommended as a useful method for the detection of
tive Gram stain or culture (p  .05). Differences in total WBC gram-negative bacterial meningitis (279). It is occasionally em-
count, glucose concentration, CSF-to-serum glucose ratio, and ployed in the setting of an abnormal CSF following neurosurgery
blood culture results were not statistically significant. When or head trauma. Nevertheless, the results of the test rarely change
adjustment was made for duration of illness before admission, patient management because physicians should employ antimi-
only the difference in CSF protein concentration remained sta- crobials with activity against gram-negative aerobic bacilli in this
tistically significant (p  .01) between children who were pre- clinical setting, even if the limulus lysate test result is negative.
treated as compared with untreated children. The duration of
illness preceding admission was significantly longer in children
who had been treated with antibiotics compared with that in C-Reactive Protein and Procalcitonin
untreated children. These observations suggested that the natu-
ral progression of illness in the pretreated group was less rapid The C-reactive protein (CRP) is an acute-phase reactant that, when
than that in the untreated group and possibly accounted for the present in concentrations greater than 100 ng/mL in CSF, is quite
differences in numbers of WBCs and bacteria in the pretreated sensitive for differentiating bacterial from viral meningitis. The
group, in whom infection was less fulminant (274). CRP response is minimal in viral meningitis. CRP concentrations
Intravenous antibiotic therapy, even for as long as several in CSF may be elevated in other CNS inflammatory or necrotic
days prior to initial LP, does not markedly alter the chemical conditions and thus are not specific for bacterial meningitis; how-
or morphologic characteristics of the CSF in cases of bacterial ever, when cell counts and chemistries suggest meningitis, the
meningitis (197). CSF was examined in 68 children with acute CRP concentration is useful in distinguishing between bacterial
bacterial meningitis on admission and 44 to 68 hours after meningitis and viral meningitis (280,281). In this circumstance,
intravenous antibiotic therapy. Initial antibiotic therapy in all a negative CSF CRP result excludes bacterial meningitis with
cases consisted of ampicillin (200 mg/kg daily in six divided 99% certainty. Because CRP is produced in the liver, serum CRP
doses) in combination with chloramphenicol (100 mg/kg daily may be useful in differentiating bacterial from viral meningitis
in four divided doses). In those cases in which meningococci, as well. A normal serum CRP has a negative predictive value of
pneumococci, or group A streptococci were isolated from about 99% for acute bacterial meningitis. Thus, in patients where
CSF culture, aqueous penicillin G (400,000 U/kg daily in six the CSF Gram stain is negative and the differential diagnosis is
divided doses) was substituted. In 65 children with meningi- between acute bacterial (or partially treated bacterial) and viral
tis caused by Hib, pneumococci, group A streptococci, and meningitis, a normal serum CRP concentration excludes bacterial
meningococci, intravenous antibiotic therapy did not signifi- meningitis with about 99% certainty and these patients may be
cantly alter the CSF protein, glucose, or WBC concentrations. safely observed in the absence of antibacterial therapy.
However, bacteria were not evident on smear and did not A normal serum procalcitonin (another acute-phase reac-
grow in culture from CSF obtained after intravenous antibi- tant) concentration has nearly identical predictive value to the
otic therapy of this duration (275). CRP (282284). In one study, a serum procalcitonin concen-
In general, bacteria should not be seen on Gram stain or tration of more than 0.2 ng/mL had a sensitivity and specificity
grow in culture from CSF examined 24 hours after treatment of up to 100% in the diagnosis of bacterial meningitis (285),
has begun with appropriate antibiotic therapy. The CSF glu- although false-negative results have been reported (286). In
cose concentration approaches normality by the third day of another study, serum procalcitonin, at a cutoff of 0.28 ng/mL,
antibiotic therapy in 80% of patients, but it may remain low had a sensitivity of 95%, specificity of 100%, negative predic-
for as long as 10 days. The CSF protein concentration remains tive value of 100%, and positive predictive value of 97% in the
elevated for at least 10 days. The WBC count in CSF remains diagnosis of bacterial meningitis (270). In patients with menin-
elevated in more than 50% of cases after a standard 7- to gitis in whom the CSF Gram stain is negative and analysis of
10-day course of antibiotic therapy, but it typically decreases other parameters is inconclusive, serum concentrations of CRP
when compared with the value obtained prior to therapy or or procalcitonin that are normal or below the limit of detection
early in the course of bacterial meningitis (256). have a high negative predictive value in the diagnosis of bacte-
rial meningitis, so that these patients (i.e., with a presumptive
Rapid Diagnostic Tests diagnosis of viral meningitis) can be carefully observed with-
Several techniques have been developed for the rapid detec- out initiation of antimicrobial therapy (278,284).
tion of bacterial antigens in the CSF, including the latex
agglutination test, the staphylococcal or other coagglutina-
tion tests, and counterimmunoelectrophoresis, among many Other Diagnostic Markers
others (276). These techniques use serum containing bacterial
antibodies or commercially available antisera directed against Other markers that have been studied as markers for acute
the capsular polysaccharide to detect the presence of bacte- bacterial meningitis in children and adults include CSF
rial antigens in CSF. Counterimmunoelectrophoresis requires concentrations of cortisol, heparin-binding protein, soluble
specialized equipment and expertise and is rarely performed triggering receptor expressed on myeloid cells 1, interleukin-6,
in hospital laboratories. Due to unacceptably poor sensitivity interleukin-12, interleukin-1
, tumor necrosis factor- , com-
of latex agglutination for the diagnosis of bacterial meningitis plement component B, and complement component 3 (272).
in CSF samples with a negative Gram stain (277), this test Most of these studies included low numbers of patients, limit-
has been abandoned in most hospital laboratories and cannot ing their generalizability. In one study, heparin-binding protein
be routinely recommended, although might be considered for had a sensitivity of 100% and specificity of 99.2% in the dif-
patients who have been pretreated with antimicrobial therapy ferentiation of bacterial from aseptic meningitis (287).
and when CSF Gram stain and culture results are negative, An immunochromatographic test for detection of S. pneu-
where available (278). moniae in CSF was found to be 100% sensitive and specific for

Scheld_Ch24.indd 394 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 395

diagnosing pyogenic pneumococcal meningitis (288), although CT scan contributes very little to the diagnosis of meningitis.
more studies are needed to demonstrate the usefulness of this The diagnosis is made by the clinical presentation and analysis
test in the diagnosis of pneumococcal meningitis; the overall of the CSF. The extent of meningeal enhancement on CT also
sensitivity of the test is 95% to 100% (289). does not influence management or prognosis. The value of CT
in suspected bacterial meningitis is in the exclusion of other CNS
pathologic processes and in the investigation of the complications
Polymerase Chain Reaction of this infection, including (a) prolonged fever for several days
after the initiation of antibiotic therapy, (b) fever that develops
Polymerase chain reaction (PCR) assays that use specific bacterial after an afebrile period during therapy (secondary fever), (c) pro-
primers to detect the nucleic acid of S. pneumoniae, N. meningit- longed obtundation or coma, (d) new or recurrent seizure activ-
idis, H. influenzae, E. coli, S. agalactiae, and L. monocytogenes ity, (e) signs of increased ICP, and (f) focal neurologic deficits.
in CSF are available. These assays are reported to be highly sensi- The most common causes of prolonged fever in patients
tive (290295). In clinical practice, culture results are often re- with bacterial meningitis are subdural effusions, drug fever,
ported before the results of the PCR assay are known limiting the and concomitant arteritis or pneumonia. In published series,
usefulness of this assay. A broad-range bacterial PCR that can 9% to 13% of patients with Hib or with pneumococcal or me-
be performed in 2 hours and that can detect small numbers of ningococcal meningitis had fever for 10 days or longer after the
viable and nonviable organisms in CSF has been developed. This initiation of appropriate antibiotic therapy. In approximately
could be useful as a screening test for bacterial meningitis and 25% of these patients, the fever was attributed to the presence
in patients who have been treated with antimicrobial therapy in of a subdural effusion. The most common causes of secondary
whom CSF culture is often negative (296); in this study, the test fever are nosocomial infections and subdural effusions (304).
characteristics for broad-based bacterial PCR demonstrated a Although the intracranial complications of meningitis are dem-
sensitivity of 100%, a specificity of 98.2%, a positive predictive onstrated well by CT scan, the results of the CT scan rarely
value of 98.2%, and a negative predictive value of 100%. In influence the management of children with meningitis and pro-
another study with use of a multiplex PCR assay for detection of longed fever in the absence of other clinical features suggesting
N. meningitidis, S. pneumoniae, and Hib, the overall specificity CNS complications (305). In one review of 107 children with
and positive predictive value were 100% and the negative pre- bacterial meningitis who underwent CT scan, one or more ab-
dictive value was 99.1% to 99.5% (297). Multiplex assays for normalities were found in 52% of cases (306). However, the
detecting genes of meningeal pathogens were 100% specific for majority of findings did not require specific intervention.
detecting its target organisms or serogroups, and the lower limit Subdural effusions are a relatively common complication
of detection was similar to that for the singleplex assays (298). of bacterial meningitis, being reported in 20% to 50% of in-
In another study, the sensitivity of broad-range PCR was higher fants and children with meningitis. Only a small percentage is
than that of culture (59% versus 43%), whereas the specificity clinically significant (307310). In most cases, the fluid in the
was 97% for both methods of diagnosis (299). Therefore, broad- subdural space is sterile and is resorbed when the inflammatory
based bacterial PCR can be used to detect the most common process subsides; however, when a subdural effusion is demon-
microorganisms in only one test and has adequate sensitivity strated by CT in a patient with prolonged fever, the possibility of
and excellent specificity (272). The broad-based bacterial PCR the development of a subdural empyema is raised. Subdural effu-
can be done within 2 hours in most industrialized countries, sions are typically low-density collections of fluid adjacent to the
although they are scarce in resource-poor countries. PCR may inner border of the skull that are hypodense to brain and nearly
be particularly useful in patients with bacterial meningitis who isodense to spinal fluid (Fig. 24.27). They are often bilateral and
have received prior antimicrobial therapy and are more likely to may flatten and displace the frontal horns posteriorly. When
have negative CSF cultures (300). The sensitivity and specificity a subdural effusion becomes purulent, its density on CT scan
of PCR in CSF for the diagnosis of pneumococcal meningitis are
92% to 100% and 100%, respectively (289). Real-time PCR has
also been used for the diagnosis of L. monocytogenes menin-
goencephalitis (301). Problems with false-positive results arise
when using PCR, although further refinements in this technique
may lead to its usefulness in the diagnosis of bacterial meningi-
tis, particularly when CSF Gram stain and cultures are negative.
Another potential application of PCR is rapid detection of the in
vitro susceptibility of meningeal pathogens to specific antimicro-
bial agents. In one report, a novel real-time PCR-hybridization
assay was developed for the rapid detection of penicillin sus-
ceptibility in S. pneumoniae; when applied to 24 pneumococcal
DNA-positive CSF extracts, penicillin-sensitive S. pneumoniae
was detected in all instances (302). Further studies may establish
the usefulness of this rapid technique in allowing clinicians to
decide on the use of specific antimicrobial therapy in patients
with bacterial meningitis (see later discussion).

Neuroimaging
In the acute stage of bacterial meningitis, the CT scan may be
normal or it may demonstrate enhancement of the meninges and
ependyma with widening of the cisterns at the base of the brain
and the cortical sulci, a result of the accumulation of purulent
exudate in the basal cisterns and over the convexities of the hemi- FIGURE 24.27 Contrast-enhanced CT scan demonstrating subdural
spheres (303). However, the presence of these abnormalities on effusion.

Scheld_Ch24.indd 395 2/21/14 5:42 PM


396 Part III: Bacterial and Mycoplasmal Infections

A B

FIGURE 24.28 A: CT scan demonstrating extraaxial fluid collection suggestive of subdural effusion as
opposed to subdural empyema. B: After administration of contrast, the medial border of the subdural
fluid collection enhances; therefore, it is a subdural empyema.

appears higher than that of CSF. After the administration of an (c) loss of sulcal markings; and (d) lack of visualization of the
intravenous contrast agent, there is significant enhancement, perimesencephalic, suprasellar, or quadrigeminal cisterns (303)
when the effusion is an empyema, at the border between the ex- (Fig. 24.30). The CT appearance of communicating hydroceph-
traaxial fluid collection and the underlying cortex (Fig. 24.28). alus is an enlargement of the entire ventricular system, including
Sterile subdural effusions do not typically demonstrate contrast the fourth ventricle, with periventricular lucencies surrounding
enhancement of the medial border (303) (Fig. 24.29). the frontal horns (Fig. 24.31). The latter abnormality repre-
The possibility of raised ICP secondary to diffuse cerebral sents transependymal movement of CSF from the ventricular
edema or obstructive or communicating hydrocephalus should system into the brain parenchyma as a result of blockage in
be considered in patients with a progressive or prolonged the normal CSF resorption pathways (303). The development
alteration of consciousness. The CT abnormalities consistent of an obstructive hydrocephalus secondary to blockage of CSF
with diffuse cerebral edema include (a) loss of differentiation flow by exudate at the foramina of Magendie and Luschka has
between gray matter and white matter; (b) compression of the CT appearance of dilated lateral and third ventricles, with
the ventricles, giving the frontal horns a slitlike appearance; nonvisualization of the fourth ventricle.

A B

FIGURE 24.29 A: CT scan of subdural fluid collection before contrast enhancement. B: CT scan after
contrast enhancement. The medial border of the subdural fluid collection does not enhance; therefore,
it is a subdural effusion.

Scheld_Ch24.indd 396 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 397

A B

FIGURE 24.30 A and B: CT scans demonstrating diffuse cerebral edema. There is loss of differentiation
between gray matter and white matter, and the sulci are markedly less prominent.

The development of seizure activity and/or focal neurologic Following the administration of contrast, cortical infarctions
symptoms and signs during the course of meningitis are clear-cut have a gyriform, nodular, or ring pattern of enhancement (306)
indications for neuroimaging. The cause of these abnormalities (Fig. 24.32). Hemorrhagic infarctions are characteristically as-
may be cerebritis, brain abscess, cortical infarction, enlarging sociated with hyperdense areas on noncontrasted scans (303).
subdural effusions, or empyema. Areas of cerebritis can easily be MRI scanning, like CT, is useful for evaluating the compli-
missed by CT scan. When they are visualized by CT, they appear cations of bacterial meningitis. Subdural empyemas, cortical
as low-density lesions on the noncontrasted scan; after contrast ad- infarctions, and areas of cerebritis are more readily imaged by
ministration, they are surrounded by an inhomogeneous halo. MRI than by CT, but in a sick patient an MRI scan is more dif-
There may also be diffusion of contrast medium into the low- ficult to obtain than a CT scan. It is considerably more difficult
density center of an area of cerebritis. As the abscess matures and to manage a critically ill patient in an isolated MRI scanner
a capsule is formed, it becomes a low-density lesion with a sharply suite than in the CT scanner.
demarcated dense ring of contrast enhancement, surrounded by a The extent and degree of leptomeningeal enhancement from
variable hypodense region of edema (311) (see Chapter 25). bacterial meningitis are well demonstrated by MRI scan after the
Cortical infarctions complicating bacterial meningitis are the intravenous administration of the paramagnetic contrast agent
result of vasculitis. The CT appearance of a cortical infarction is gadolinium (Fig. 24.33). Paramagnetic contrast agents produce
that of a hypodense lesion that conforms to a vascular territory. local alterations in magnetic environments that directly affect the
MRI signal obtained from protons. The image that is obtained
after the administration of the contrast agent visualizes this ef-
fect on proton relaxation. The contrast agent itself is not visual-
ized. Areas of active breakdown in the BBB are enhanced when
scans are obtained after the administration of gadolinium (312).
Pathologic examination of animals with experimental bacterial
meningitis demonstrated that areas of contrast enhancement on
both CT and MRI scans correlated with inflammatory cell infiltra-
tion, and gadolinium-enhanced T1-weighted MRI scans revealed
inflammatory meningeal and ependymal lesions more effectively
than did contrast-enhanced CT. Unenhanced T1- and T2-weighted
MRI scans did not detect meningeal inflammation (313).
Subdural effusions can sometimes be distinguished from
subdural empyemas by their MRI appearance. Subdural effu-
sions are low-protein collections; therefore, they appear isoin-
tense to spinal fluid on MRI (Fig. 24.34). Subdural empyemas
are more proteinaceous and therefore appear to have higher
signal intensity than CSF on T2-weighted MRI scans (314).
MRI is superior to CT scan in visualizing a cortical infarc-
tion. Ischemia and/or infarction are common causes of focal
neurologic deficits in bacterial meningitis. On T2-weighted
MRI images, areas of infarction appear as areas of abnor-
mal, increased signal intensity. An acute infarction is often
FIGURE 24.31 CT scan demonstrating enlargement of the entire not visualized on CT scan within the first 24 hours unless the
ventricular system, including the fourth ventricle, characteristic of infarction is large or associated with edema and mass effect.
communicating hydrocephalus. MRI is the most sensitive modality for these complications,

Scheld_Ch24.indd 397 2/21/14 5:42 PM


398 Part III: Bacterial and Mycoplasmal Infections

A B

C D

FIGURE 24.32 A and B: CT scan before and after contrast enhancement. High ventricular level. C and D:
CT scan before and after contrast enhancement. Low ventricular level. Hypodense lesion characteristic
of cortical infarction in a patient with Hib meningitis. Following the administration of contrast, there is
a gyriform pattern of enhancement.

particularly with regard to infarction, especially when seen Although no prospective data are available on the timing of
on diffusion-weighted imaging, and ventriculitis (315). MR administration of antimicrobial therapy in patients with bac-
angiography and perfusion-weighted imaging may show vas- terial meningitis, a retrospective cohort study in patients with
cular complications, including focal stenosis and irregularity community-acquired bacterial meningitis demonstrated that a
of major intracranial arteries. CT angiography is more sensi- delay in initiation of antimicrobial therapy after patient ar-
tive than MR angiography for demonstrating focal stenosis of rival in the emergency department was associated with an ad-
small cerebral arteries. verse clinical outcome when the patients condition advanced
to a high stage of prognostic severity (317), thus supporting
the assumption that treatment of bacterial meningitis before
INITIAL MANAGEMENT it advances to a high level of clinical severity improves clini-
cal outcome. This concept has also been supported by two
The initial management of a patient with presumed bacterial retrospective studies: one demonstrated a reduction in mortal-
meningitis is to obtain blood cultures, initiate antimicrobial ity with early administration of antimicrobial therapy (318),
and dexamethasone therapy if indicated, and obtain spinal and the other showed a benefit in terms of neurologic out-
fluid analysis to determine whether the CSF formula is con- come and survival in patients who received antimicrobial
sistent with that diagnosis (see earlier discussion) (278,316). therapy before the patients level of consciousness deteriorated
Empirical antimicrobial therapy should be initiated based on to a score lower than 10 on the Glasgow Coma Scale (319).
the patients age and underlying disease status (Table 24.5). In another retrospective case study, delay in administration

Scheld_Ch24.indd 398 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 399

TA B L E 2 4 . 5
EMPIRICAL THERAPY FOR PURULENT MENINGITIS

Predisposing Factor Antimicrobial Therapy

Age
Neonate Ampicillin plus cefotaxime; or
ampicillin plus an aminoglycoside
Infant (123 mo) Vancomycin plus a third- or fourth-
generation cephalosporina
Children and adults Vancomycin plus a third- or fourth-
(250 yr) generation cephalosporina,b
Adults 50 yr Vancomycin plus ampicillin plus
a third- or fourth-generation
cephalosporina
Immunocompromised Vancomycin plus ampicillin plus
state either cefepime or meropenem
Basilar skull fracture Vancomycin plus a third- or fourth-
generation cephalosporina
Head trauma; Vancomycin plus either ceftazidime
postneurosurgery or meropenem
FIGURE 24.33 Gadoliniumdiethylenetriamine pentaacetic acid a
enhanced T1-weighted magnetic resonance scan demonstrating diffuse Cefotaxime or ceftriaxone or cefepime.
b
meningeal enhancement in a patient with meningitis. Add ampicillin if meningitis caused by Listeria monocytogenes is
suspected.

of antimicrobial therapy was associated with death; in the


multivariate analysis, a delay of longer than 6 hours in antimi- initiation of antimicrobial therapy, with the potential for in-
crobial administration after presentation conferred an 8.4-fold creased morbidity and mortality in patients with bacterial men-
greater risk of death (320). An additional retrospective cohort ingitis. Therefore, emergency empirical antimicrobial therapy
study of 286 patients with community-acquired bacterial men- and adjunctive dexamethasone therapy if indicated, after ob-
ingitis confirmed these results, in which early and adequate taining blood cultures, should be initiated before sending the
administration of antimicrobial therapy related to onset of patient to the CT scanner. Although CSF cultures may be sterile
overt signs of meningitis was independently associated with after the initiation of antimicrobial therapy, pretreatment blood
favorable outcome (odds ratio [OR]  11.19) (321). cultures and the CSF formula or Gram stain will provide evi-
Some patients should have a noncontrast CT scan of the head dence for or against a diagnosis of bacterial meningitis. In one
performed before LP to rule out the presence of brain shift (as a retrospective review of 177 patients (39 of whom had received
result of an intracranial mass lesion or generalized brain edema) prior antimicrobial therapy) with CSF cultureproven bacterial
because of the potential risk of herniation (278). However, the meningitis (322), the combination of blood culture and CSF
time involved in waiting for a CT scan significantly delays the Gram stain, with or without latex agglutination, identified the
causative bacterium in 92% of patients. Although some clini-
cians routinely order CT scans of the head before performance
of an LP in adults with suspected bacterial meningitis, this is not
necessary in most patients. In a study of 301 patients with bac-
terial meningitis (237), the clinical features at baseline that were
associated with an abnormal finding on CT scan of the head
were an age of at least 60 years, immunocompromised status,
a history of CNS disease, a history of seizure within 1 week be-
fore presentation, and neurologic abnormalities (an abnormal
level of consciousness, an inability to answer two consecutive
questions correctly or to follow two consecutive commands,
gaze palsy, abnormal visual fields, facial palsy, arm drift, leg
drift, and abnormal language). It is reasonable to proceed with
LP without CT scan of the head if the patient does not meet any
of the following criteria: new-onset seizures, an immunocom-
promised state, signs that are suspicious for space-occupying
lesions (papilledema or focal neurologic signs, not including
cranial nerve palsy), or moderate to severe impairment of con-
sciousness (278,323). Although the decision to perform a CT
before LP must be individualized, these guidelines are useful
in determining the patient groups that are more likely to have
abnormal findings on neuroimaging studies.
Once the infecting meningeal pathogen is isolated and sus-
FIGURE 24.34 T2-weighted magnetic resonance scan demonstrating ceptibility testing known, antimicrobial therapy can be modi-
subdural effusion. fied for optimal treatment (Tables 24.6 and 24.7) (278,316).

Scheld_Ch24.indd 399 2/21/14 5:42 PM


400 Part III: Bacterial and Mycoplasmal Infections

TA B L E 2 4 . 6
RECOMMENDED ANTIMICROBIAL THERAPY FOR ACUTE BACTERIAL
MENINGITIS

Microorganisma Antimicrobial Therapy

Haemophilus influenzae type b Third-generation cephalosporinb


Neisseria meningitidis Third-generation cephalosporinb
Streptococcus pneumoniae Vancomycin plus a third- or fourth-generation cephalosporinb
Listeria monocytogenes Ampicillin
Streptococcus agalactiae Ampicillin or penicillin G or a third-generation cephalosporin

a
Pathogen presumptively identified by positive Gram stain.
b
Cefotaxime or ceftriaxone or cefepime.

TA B L E 2 4 . 7
SPECIFIC ANTIMICROBIAL THERAPY FOR ACUTE MENINGITIS

Microorganism Standard Therapy Alternative Therapiesa

Bacteria
Haemophilus influenzae

-Lactamase negative Ampicillin Ceftriaxone, cefotaxime, cefepime,
chloramphenicol, aztreonam, fluoroquinolone

-Lactamase positive Ceftriaxone or cefotaxime Cefepime, chloramphenicol, aztreonam,
fluoroquinolone
Neisseria meningitidis
Penicillin MIC 0.1 g/mL Penicillin G or ampicillin Ceftriaxone, cefotaxime, chloramphenicol
Penicillin MIC 0.11.0 g/mL Ceftriaxone or cefotaxime Chloramphenicol, fluoroquinolone, meropenem
Streptococcus pneumoniae
Penicillin MIC 0.06 g/mL Penicillin G or ampicillin Ceftriaxone, cefotaxime, chloramphenicol
Penicillin MIC 0.12 g/mL b

Ceftriaxone or cefotaxime MIC 1.0 g/mL Ceftriaxone or cefotaxime Meropenem, cefepime


Ceftriaxone or cefotaxime MIC 1.0 g/mL Vancomycinc plus Vancomycin plus moxifloxacind
ceftriaxone or cefotaxime
Enterobacteriaceaee Ceftriaxone or cefotaxime Aztreonam, fluoroquinolone, trimethoprim-
sulfamethoxazole, meropenem, ampicillin
Pseudomonas aeruginosa Ceftazidimef or cefepimef Aztreonamf, fluoroquinolonef, meropenemf
Acinetobacter baumanniie Meropenem Colistin (usually formulated as colistimethate
sodium)b, polymyxin Bb
Listeria monocytogenes Ampicillin or penicillin Gf Trimethoprim-sulfamethoxazole
Streptococcus agalactiae Ampicillin or penicillin Gf Ceftriaxone, cefotaxime, vancomycin
Staphylococcus aureus
Methicillin-sensitive Nafcillin or oxacillin Vancomycin, linezolid, daptomycin
Methicillin-resistant Vancomycinc Trimethoprim-sulfamethoxazole, linezolid, daptomycin
Staphylococcus epidermidis Vancomycinc Linezolid

a
There may not be clinical data to support all recommendations for alternative antimicrobial agents in patients with bacterial meningitis, but specific
agents are suggested based on CSF penetration in experimental animal models and in vitro activity against the offending pathogen.
b
Might also need to be administered by the intraventricular or intrathecal routes.
c
Addition of rifampin may be considered; see text for indications.
d
Would recommend moxifloxacin given its excellent CSF penetration and in vitro activity against S. pneumoniae, although there are no clinical data to
support its usefulness in patients with pneumococcal meningitis; if used, many authorities would combine moxifloxacin with vancomycin or a third-
generation cephalosporin such as cefotaxime or ceftriaxone.
e
Choice of a specific antimicrobial agent must be guided by in vitro susceptibility testing.
f
Addition of an aminoglycoside should be considered.
Adapted from van de Beek D, Brouwer MC, Thwaites GE, et al. Advances in treatment of bacterial meningitis. Lancet. 2012;380:16931702.

Scheld_Ch24.indd 400 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 401

TA B L E 2 4 . 8 Recommended dosages of antimicrobial agents for children


with infections of the CNS are shown in Table 24.8, and those
RECOMMENDED DOSES OF ANTIBIOTICS FOR for adults are presented in Table 24.9. In addition, certain
CHILDREN patients should receive adjunctive dexamethasone therapy
when presenting with suspected or proven bacterial meningitis
Dosing (185,186,278,316). This is discussed in more detail in the sec-
Antibiotic Daily Dose Interval (hr) tion Adjunctive Therapy.
Penicillin G 250,000400,000 U/kg 46
Ampicillin 300 mg/kg 46 TREATMENT
Chloramphenicol 75100 mg/kg 6
Ceftriaxone 80100 mg/kg 1224a General Principles of Therapy
Cefotaxime 225300 mg/kg 68
Ceftazidime 150 mg/kg 8 Bacteriologic cure of meningitis is defined as the eradica-
tion of bacteria from CSF. Effective antimicrobial therapy of
Cefepime 150 mg/kg 8 bacterial meningitis depends on attaining adequate bacteri-
Meropenem 120 mg/kg 8 cidal activity in the CSF. Several factors, largely elucidated
Nafcillin 200 mg/kg 6 in experimental animal models of meningitis, determine
Vancomycinb 60 mg/kg 6 whether bactericidal activity is achieved, including (a) the
ability of an antibiotic to penetrate the BBB, (b) the activ-
Rifampin 1020 mg/kg 1224 ity of the antibiotic within purulent CSF, and (c) the rate of
Gentamicin 7.5 mg/kg 8 metabolism of an antibiotic and its rate of clearance from
Amikacin 2030 mg/kg 8 CSF (324328).
Trimethoprim- 1020 mg/kg (based 612
The BBB poses physiologic restrictions, allowing only
sulfamethoxazole on trimethoprim)
highly lipid-soluble substances or substances transported by
carrier-mediated facilitated diffusion to traverse it under nor-
a
mal conditions (324,329). The ability of an antibiotic to pen-
If a once-daily regimen is used, we recommend that on the first day,
a dose of 80 mg/kg be given at diagnosis, at 12 and 24 hr, and then
etrate the BBB depends on several factors: (a) degree of lipid
every 24 hr thereafter. solubility, (b) degree of ionization at physiologic pH, (c) pro-
b
Maintain serum trough concentrations of 15 to 20 g/mL. tein binding in serum, (d) molecular size and structure of the
antibiotic, and (e) status of the BBB. The BBB acts physiolog-
ically like a lipid bilayer. In general, the greater the lipid solu-
bility of an antibiotic, the better its penetration into CSF. For
example, chloramphenicol is a highly lipophilic substance
TA B L E 2 4 . 9 that easily penetrates the BBB. The
-lactam antibiotics have
poor lipid solubility, which limits their entry into CSF under
RECOMMENDED DOSES OF ANTIBIOTICS normal conditions (324). The un-ionized form of a drug pos-
FOR BACTERIAL MENINGITIS IN ADULTS sesses greater lipid solubility than the ionized form. Thus,
(15 YEARS OF AGE AND OLDER) a lesser degree of ionization at the pH of serum and CSF
increases entry of antibiotics into CSF by increasing their
Antibiotic Total Daily Dose (Dosing Interval)
lipid solubility. Penicillin G has a high degree of ionization
Penicillin G 2024 miU (every 4 h) at the pH of plasma and CSF. This, combined with its low
lipid solubility, may explain the poor penetration of penicil-
Ampicillin 12 g (every 4 h) lin G across intact meninges. The normal plasma-to-CSF pH
Ceftriaxone 4 g (every 1224 h)a gradient is approximately 0.1 pH unit (330). The plasma-
Cefotaxime 812 g (every 46 h) to-CSF pH gradient is altered, however, by purulent men-
Ceftazidime 6 g (every 8 h)b ingitis. The accumulation of lactate in CSF during bacterial
meningitis decreases the pH of CSF, increases the gradient,
Cefepime 6 g (every 8 h) and enhances the penetration of some antibiotics into CSF.
Meropenem 6 g (every 8 h) Conversely, as metabolic acidosis develops, the pH gradi-
Vancomycin 3060 mg/kg (every 812 h)c ent is reversed and the penetration of antibiotics into CSF is
Nafcillin, oxacillin 912 g (every 4 h)d reduced (324).
Protein binding and molecular size limit the ability of an
Chloramphenicol 4 g (every 6 h)e antibiotic to enter the CSF. Only the free nonprotein-bound
Rifampin 600 mg (every 24 h) portion of an antibiotic in serum can enter the CSF; therefore,
Gentamicin, tobramycin 5 mg/kg (every 8 h) highly protein-bound antibiotics have lower CSF concentra-
Amikacin 15 mg/kg (every 8 h) tions than antibiotics with a lower degree of protein binding,
other factors being equal. Increased binding to plasma pro-
Trimethoprim- 1020 mg/kg (every 612 h) teins reduces the amount of antibiotic penetration into CSF;
sulfamethoxazole (based on trimethoprim) however, it is the concentration of free antibiotic in CSF rela-
tive to its minimum bactericidal concentration (MBC) that de-
a
Actual dose studied was 50 mg/kg, every 12 h. termines its therapeutic effectiveness (329).
b
Not enough patients studied to make firm recommendation. Although the pharmacokinetics of an antibiotic greatly in-
c
Maintain serum trough concentrations of 15 to 20 g/mL.
d
Higher doses associated with leukopenia. fluence its ability to penetrate the BBB, the most important
e
Use 6 g/d for pneumococcal meningitis. factor appears to be the presence of meningeal inflamma-
Data from refs. 479 and 181. tion. A moderate degree of meningeal inflammation results
in a marked increase in the penetrability of most antibiotics.

Scheld_Ch24.indd 401 2/21/14 5:42 PM


402 Part III: Bacterial and Mycoplasmal Infections

In many instances, altered BBB permeability is essential


for an antibiotic to be effective in bacterial meningitis. Antimicrobial Therapy for Specific Organisms
The morphologic alterations of the BBB observed in an adult
rat model of experimental meningitis consist of an early and Neisseria meningitidis
sustained increase in pinocytotic vesicles with a progressive Penicillin G and ampicillin are the preferred antibiotics
separation of intercellular tight junctions in the cerebral mi- for the treatment of meningitis caused by N. meningitidis
crovasculature (331). These features may contribute to anti- (197,219,334). A 7-day course of therapy is adequate for
microbial entry into purulent CSF. most cases of uncomplicated meningococcal meningitis. There
Once the antibiotic penetrates into CSF, several factors are reports of strains of N. meningitidis resistant to penicil-
influence its ability to eradicate the infection: (a) sufficient lin (335). However,
-lactamaseproducing isolates are still
concentrations of free active drug must be achieved in CSF, rare (336). Penicillin-resistant strains that do not produce
-
because this form of the antibiotic is necessary for bactericidal lactamase appear, instead, to have a reduced affinity for peni-
effect. The high protein concentrations in purulent CSF limit cillin-binding proteins (e.g., PBP-2 and PBP-3) and have been
the concentration of free, unbound antibiotic; (b) an antibi- reported from Spain, the United Kingdom, and other countries
otic must achieve concentrations in CSF in vivo exceeding the (335,337339). In Spain, the number of relatively penicillin-
MBC of the infecting organism by 10- to 20-fold for optimal resistant meningococcal isolates reached 20% in 1989 (340).
efficacy (173); (c) the bactericidal activity of an antibiotic However, in the United States in 1991, MICs of penicillin of
may be diminished by the coadministration of a bacteriostatic 0.125 g/mL were noted for only 3 of 100 isolates submit-
agent. For example, chloramphenicol inhibits the bactericidal ted to the CDC (341). Routine susceptibility testing of menin-
effect of aminoglycosides against gram-negative aerobic bacilli gococcal isolates is recommended. Meningococcal meningitis
within the CSF (332). Conversely, antimicrobial combinations caused by the relatively penicillin-resistant strains has, how-
may exert an enhanced, synergistic improvement in the rate ever, been successfully managed with penicillin therapy, and
of bactericidal activity within the CSF in vivo (e.g., ampicil- thus the clinical significance of this partial resistance is unclear
lin plus gentamicin versus L. monocytogenes or S. agalactiae); at present (173). Nevertheless, this situation must be carefully
(d) early in the course of bacterial meningitis, there may be monitored, because meningococci showing relative resistance
very large numbers of bacteria in CSF (i.e., 108 CFU/mL). to penicillin (i.e., MICs in the range of 0.1 to 1.0 g/mL) are
Some antibioticsin particular the
-lactam antibiotics increasing in incidence worldwide. In Ontario, Canada, the
demonstrate an inoculum effect in vitro, such that the mini- prevalence of invasive meningococcal disease caused by strains
mum inhibitory concentration (MIC) increases dramatically with decreased in vitro susceptibility to penicillin was much
as the inoculum of the test strain is increased from 105 to higher (21.7%) in 2006 (342), although it did not change in
107 CFU/mL under standardized in vitro conditions (324). frequency between 2000 and 2006. Cefotaxime or ceftriaxone
The inoculum effect may explain the failure of certain anti- should be used when relatively penicillin-resistant strains of
biotics in vivo, as the in vitro activity of an antibiotic is rou- meningococci are isolated and when a patient is allergic to
tinely determined in standard growth media using a bacterial penicillin (343). Because of potent in vitro activity and ease
concentration of 105 CFU/mL (325); (e) an antibiotic must of administration (e.g., every 12 hours and perhaps effective
remain physically stable in the presence of bacterial inacti- at every 24 hours), ceftriaxone may well be the drug of choice
vating enzymes, such as
-lactamase and chloramphenicol for serious meningococcal infection, including meningitis.
acetyltransferase (329). Chloramphenicol is also generally effective and widely used in
The effectiveness of an antibiotic in eradicating bacterial developing countries.
meningitis is also determined by its rate of metabolism and
the activity of its metabolites. For example, cephalothin is
metabolized in vivo to desacetylcephalothin, which is less ac-
Streptococcus pneumoniae
tive in vitro than the parent compound (324). In contrast, the Initial therapy of pneumococcal meningitis includes a combi-
metabolite of cefotaxime (desacetylcefotaxime) is as active in nation of a third- or fourth-generation cephalosporin (either
vitro as the parent compound. Antibiotics are removed from ceftriaxone, cefotaxime, or cefepime) plus vancomycin until
CSF either by simple resorption through arachnoid villi or by the results of antimicrobial susceptibility testing are known.
an energy-dependent active transport process that removes The Clinical and Laboratory Standards Institute has recently
the antibiotic from the CSF to the intravascular compartment redefined the in vitro susceptibility breakpoints for pneumo-
across the epithelium of the choroid plexus. This exit pump coccal isolates from patients with meningitis as either suscep-
is inhibited by weak organic acids, such as salicylates and pro- tible or resistant, with intravenous penicillin breakpoints of
benecid, and to some extent by meningitis itself.
-Lactam 0.06 g/mL or lower and 0.12 g/mL or greater, respectively
antibiotics are cleared from the CSF by this process. The third- (344). A pneumococcal isolate with an MIC for cefotaxime
generation cephalosporins (e.g., ceftriaxone and cefotaxime) or ceftriaxone of less than 0.5 g/mL is considered suscep-
possess decreased affinity (as compared with penicillin G) for tible, 1.0 g/mL intermediate, and more than 2.0 g/mL
the choroid plexus exit pump, so that they remain in CSF resistant (345). In the United States, approximately 34% of
for a longer time (329). pneumococcal isolates are penicillin nonsusceptible (MICs
There have also been investigations to determine whether in the intermediate and resistant ranges) and approximately
continuous infusion of antimicrobial therapy improves out- 14% are resistant to ceftriaxone (346). The mechanisms by
come in patients with bacterial meningitis. In one study of which S. pneumoniae develop resistance to
-lactam antibi-
723 African children with bacterial meningitis randomly as- otics (penicillin and extended-spectrum cephalosporins) is
signed to receive bolus or continuous infusion of cefotaxime through alterations of one or more penicillin-binding proteins
for the first 24 hours of therapy, 272 children died, but the (347). Alterations in the penicillin-binding proteins lead to a
mode of administration did not significantly affect the propor- decrease in their affinity for
-lactam antibiotics and thus a
tion of children who died or were severely disabled at the time decreased susceptibility to the antibiotic (346). In Brazil, peni-
of hospital discharge (333); children with pneumococcal men- cillin resistance was mainly detected in isolates of serotypes 14
ingitis given continuous cefotaxime infusion were significantly (61%), 23F (16%), 6B (10%), and 19F (3%) (348). Results
less likely to die or have sequelae, however. of recent surveillance studies in the United States show that

Scheld_Ch24.indd 402 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 403

the prevalence of penicillin-nonsusceptible S. pneumoniae Malawi during 2004 to 2006 (363) and in 43% of isolates in
ranges from 25% to more than 50% (349); rates are as high Papua New Guinea (364).
as 60% in some parts of Latin America and as high as 80% Fluoroquinolones have shown efficacy in some small series
in some countries in Asia. Factors reported to predispose to or randomized trials in patients with gram-negative meningi-
resistance include the patients age (younger than 10 or older tis. In recent years, in response to drug-resistant pneumococci
than 50 years); immunosuppression; prolonged hospital stay; in particular, newer fluoroquinolones with improved activ-
children in day care settings; infection by serotypes 14 and ity against gram-positive cocci have been introduced. These
23; and frequent, prolonged, or prophylactic use of antimicro- agents (e.g., gatifloxacin, moxifloxacin, gemifloxacin, and
bial therapy. However, penicillin nonsusceptible strains have garenoxacin) penetrate well into CSF and have produced ex-
been isolated even when no risk factors or comorbidities are cellent results in experimental models of multidrug-resistant
identified (350). pneumococcal meningitis, including vancomycin-tolerant
In view of the increasing number of strains resistant to strains (365370). Pharmacodynamic analysis suggests that a
penicillin, all CSF isolates of S. pneumoniae should be tested Cmax CSF-to-MBC ratio of at least 5 and CSF concentrations
for sensitivity to penicillin and the third-generation cephalo- above the MBC for the test strain for the entire dosing interval
sporins by in vitro susceptibility testing. A third- or fourth- are necessary for optimal bactericidal activity. Furthermore,
generation cephalosporin (i.e., cefotaxime or ceftriaxone newer antipneumococcal fluoroquinolones demonstrate syn-
or cefepime) is recommended for strains of pneumococci ergistic activity in vitro and in vivo in experimental models of
resistant to penicillin (MIC 0.12 g/mL) but sensitive to pneumococcal meningitis and combination therapy appears to
the third-generation cephalosporins (MIC 1 g/mL). For prevent quinolone resistance among pneumococci (370). A
-
strains resistant to penicillin and the cephalosporins, van- lactam (e.g., ceftriaxone)potent antipneumococcal quinolone
comycin plus a third- or fourth-generation cephalosporin is regimen is very promising (371,372) and may well supplant
the antimicrobial regimen of choice (351,352). The addition the currently favored ceftriaxone (or cefotaxime or cefepime)
of rifampin or a ceftriaxone-rifampin regimen has also been (373)vancomycin in the future, pending evaluation in ran-
recommended by some authorities, but rifampin may dem- domized controlled trials.
onstrate indifference or slight antagonism when combined
with
-lactam agents in standardized in vitro assays (353),
although rifampin, without bacteriolytic activity, may protect
Gram-Negative Bacilli
against neuronal damage (354). Although concerns have been The results of clinical trials in patients with gram-negative
raised about use of vancomycin in patients with pneumococ- bacillary meningitis favor the use of a third- (or fourth-)
cal meningitis who are also receiving adjunctive dexametha- generation cephalosporin over conventional aminoglycoside-
sone, appropriate CSF concentrations of vancomycin may be containing regimens (219,223,374). Cefotaxime, ceftizoxime,
attained as long as appropriate dosages of vancomycin are ceftriaxone, and ceftazidime penetrate well into inflamed CSF
used. In a study of 14 patients, administration of intravenous and are highly active against gram-negative enteric bacilli
vancomycin (at a continuous infusion of 60 mg/kg per day, (223,375). Cure rates of 78% to 94% have been achieved with
after a 15 mg/kg loading dose) led to mean serum and CSF the cephalosporins, compared with previous mortality rates of
vancomycin concentrations of 25.5 g/mL and 7.2 g/mL, 40% to 90% with predominantly aminoglycoside-containing
respectively (355). These data indicate that appropriate CSF regimens (173).
concentrations can be attained when appropriate doses are However, given the emergence of strains of gram-negative
used. Trough serum concentrations of 15 to 20 g/mL are bacilli that are resistant to the third-generation cephalosporins
recommended (356). Intrathecal or intraventricular vancomy- (376), the use of other intravenous agents, with or without in-
cin is a reasonable consideration in patients not responding to traventricular antimicrobials, may need to be considered and
parenteral therapy. several have been used in patients with meningitis caused by
Vancomycin-resistant strains of pneumococci have not aerobic gram-negative bacilli (278). In general, the aforemen-
been seen, but strains of S. pneumoniae tolerant to vancomy- tioned third-generation cephalosporins appear to be equally
cin have been reported. Tolerance is the ability of a bacteria to efficacious for the treatment of gram-negative bacillary menin-
survive in the presence of an antibiotic, neither growing nor gitis, with the exception of meningitis caused by P. aeruginosa.
being eradicated by the antibiotic. Tolerance may be a precur- Ceftazidime or cefepime is recommended when P. aeruginosa
sor for the development of antimicrobial resistance because it is suspected (377). Clinical trials suggest the efficacy of ceftazi-
creates survivors of antibiotic therapy (357359). dime alone for Pseudomonas meningitis, but a combination of
Imipenem has been utilized in the therapy of penicillin- ceftazidime and an aminoglycoside may be used if response is
resistant pneumococcal meningitis, although its proconvulsant delayed (173).
activity may limit its usefulness; meropenem, a carbapenem Although clinical experience is scant, the fluoroquinolones
with less seizure proclivity than imipenem, may be an effec- have demonstrated efficacy in animal models of gram-negative
tive alternative (360). However, in a study of 20 cefotaxime- bacillary meningitis. Intravenous pefloxacin has shown good
resistant S. pneumoniae isolates (361), 4 were of intermediate efficacy with bacteriologic eradication from the CSF in nine of
susceptibility and 13 were resistant to meropenem, suggest- ten patients with gram-negative aerobic bacillary meningitis
ing that meropenem may not be a useful alternative agent for failing conventional therapy in one study (378). However, the
the treatment of pneumococcal isolates that are highly resis- quinolones should be considered only for gram-negative bacil-
tant to penicillin and cephalosporins. Chloramphenicol is one lary meningitis caused by multiresistant strains or in patients
agent that has been studied for the treatment of pneumococcal unresponsive to standard therapies (379).
meningitis. However, clinical failures with chloramphenicol Although extended-spectrum penicillins (e.g., ticarcillin-
have been reported in patients with penicillin-resistant iso- clavulanate, temocillin) and aztreonam have proved effec-
lates, probably because of the poor bactericidal activity of tive in the therapy of experimental models of gram-negative
chloramphenicol against these strains; 20 of 25 children had an bacillary meningitis in animals, their use is not recommended
unsatisfactory outcome (i.e., death, serious neurologic deficit, because the clinical experience with third-generation cephalo-
poor clinical response) in one study (362). Chloramphenicol sporins in humans is far more extensive. Meropenem has been
resistance was also found in 27% of pneumococcal isolates in successfully used in patients with gram-negative meningitis

Scheld_Ch24.indd 403 2/21/14 5:42 PM


404 Part III: Bacterial and Mycoplasmal Infections

(including P. aeruginosa), and further investigations may rapid sterilization of the CSF at follow-up LP at approxi-
confirm its efficacy in the therapy of bacterial meningitis (380). mately 24 hours (2% versus 12% positive cultures; p  .11);
The worldwide database on the use of meropenem in the ther- (b) less moderate to profound sensorineural hearing loss at
apy of bacterial meningitis is quite extensive and encouraging, the 2-month follow-up examination (4% versus 17%; p 
including cases of gram-negative bacillary meningitis failing .05); and (c) reversible biliary pseudolithiasis on serial ab-
third-generation cephalosporin therapy (e.g., Enterobacter dominal ultrasonography (16 of 35 versus 0 of 35; p  .001)
species). (391). In another comparative trial, ceftriaxone also led to a
For empirical treatment of Acinetobacter meningitis, more rapid clinical response as compared with cefuroxime
intravenous meropenem with or without an aminogly- (392). The third-generation cephalosporins, cefotaxime and
coside administered by the intraventricular or intrathe- ceftriaxone, are clearly preferable to cefuroxime in the treat-
cal route has been recommended (381); if the organism is ment of Hib meningitis. Resistance of Hib to the third-gen-
later found to be resistant to carbapenems, colistin (usu- eration cephalosporins and fluoroquinolones in vitro has not
ally formulated as colistimethate sodium) or polymyxin B been described (393). A combination of chloramphenicol and
should be substituted for meropenem and may also need to ampicillin was at one time the recommended therapy for Hib
be administered by the intraventricular or intrathecal route meningitis. The use of a third-generation cephalosporin, ei-
(382). Intravenous colistin (5 mg/kg per day) was success- ther cefotaxime or ceftriaxone, has the following advantages
fully used to treat a patient with meningitis caused by a over therapy with a combination of ampicillin plus chlor-
multidrug-resistant A. baumannii (383); intrathecal colistin amphenicol: (a) the need to monitor serum chloramphenicol
was also efficacious in other cases of meningitis caused by concentrations is eliminated; (b) the potential toxicities of
this same multidrug-resistant organism (384,385) and in- chloramphenicol are avoided; (c) the number of daily doses
trathecal polymyxin E has also been used in a patient with of antibiotics is decreased (199); (d) approximately 29% of
Acinetobacter meningitis (386). In a summary of treatment Hib strains causing meningitis in the United States are re-
of multidrug-resistant A. baumannii, a total of 14 patients sistant to ampicillin, through the production of
-lactamase,
were treated for CNS infection (ventriculitis or meningitis) although a smaller number of strains are resistant to ampicil-
with colistin given intravenously and/or either intrathecally lin because of reduced affinity for penicillin-binding proteins.
or intraventricularly (387); sterilization was achieved in all An increasing number of Hib strains are resistant to chloram-
cases and cure in 13 of 14 cases. In the presence of meningi- phenicol, through the production of chloramphenicol acetyl-
tis, CSF concentrations of colistin were shown to be 0.5 g/ transferase (199). More than 50% of ampicillin-resistant H.
mL (34% to 67% of serum concentrations) (388). Two cases influenzae CSF isolates from Spain are also chloramphenicol
of A. baumannii meningitis were also successfully treated resistant (394). Thus, in some countries, Hib isolates resistant
with tigecycline (389). to both ampicillin and chloramphenicol are common (199).
DNA coding for both the
-lactamase enzyme and the chlor-
amphenicol acetyltransferase enzyme can reside on plasmids,
Haemophilus influenzae type b although chromosomally mediated resistance, as, for exam-
A third-generation cephalosporin, either cefotaxime or cef- ple, to trimethoprim, has been described (395). Therefore,
triaxone, is recommended for the initial therapy of H. influ- any
-lactamasepositive Hib isolate should be tested for sus-
enzae meningitis (334). There are few differences between ceptibility to chloramphenicol. In addition, therapy with the
cefotaxime and ceftriaxone for therapy of bacterial menin- third-generation cephalosporins may result in a more rapid
gitis. Both are generally very active against the major men- sterilization of the CSF as compared with therapy with ampi-
ingeal pathogens, rapidly sterilize CSF cultures, and are safe cillin plus chloramphenicol.
and effective (390). The long half-life of ceftriaxone allows The pharmacokinetics of chloramphenicol are highly vari-
for twice-daily (or even once-daily) administration of this able among individuals; therefore, serum concentrations of this
antibiotic. Several studies have documented that once-daily antibiotic must be monitored to ensure therapeutic concentra-
administration of ceftriaxone is safe and efficacious for the tions while avoiding potential toxic concentrations, especially
treatment of bacterial meningitis (336). However, this is not in infants. Therapeutic serum concentrations are in the range
yet recommended as standard therapy for adults; a twice- of 15 to 25 g/mL, obtained 60 to 120 minutes after the com-
daily dose is preferred (173). Ceftriaxone has shown prom- pletion of an intravenous or oral dose. Concentrations in ex-
ise as once-daily therapy for completion of the therapeutic cess of 30 g/mL are associated with an increased incidence of
course in the home setting in stable children with menin- bone marrow suppression, and levels exceeding 50 to 80 g/
gitis following an uncomplicated hospital stay. A 7- to 10- mL may depress myocardial contractility (396). The pharma-
day course of antibiotics is generally recommended for Hib cology of chloramphenicol is altered in patients in shock or
meningitis. with liver disease. In either clinical situation, excessive serum
Despite initial enthusiasm, cefuroxime, a second- concentrations of chloramphenicol could potentially decrease
generation cephalosporin, is not recommended for the treat- cardiac contractility. Chloramphenicol should, therefore, be
ment of Hib meningitis. The in vitro bactericidal activity of avoided in patients with these conditions (396).
this drug has been shown to be inferior to that of the third- When chloramphenicol is used in combination with phe-
generation cephalosporins, and there have been reports of an nobarbital and phenytoin, serum concentrations of all three
unusually high incidence of positive Gram stain and cultures drugs must be monitored. Chloramphenicol inhibits hepatic
in CSF obtained several days into treatment. In a prospective, microsomal enzymes and therefore prolongs the half-life
multicenter study, 106 children with acute bacterial menin- of phenytoin in serum, resulting in toxic concentrations of
gitis were randomly assigned to receive either ceftriaxone phenytoin. Phenytoin, conversely, interferes with hepatic
or cefuroxime (391). Delayed sterilization of CSF was more metabolism of chloramphenicol, resulting in toxic serum con-
common among six patients given cefuroxime than in one centrations. Phenobarbital induces hepatic microsomal en-
patient given ceftriaxone (p  .112). When all children with zymes, increases chloramphenicol metabolism, and decreases
positive CSF cultures (Hib, N. meningitidis, S. pneumoniae, S. serum chloramphenicol concentrations. These drug interac-
agalactiae) were included in the analysis, ceftriaxone therapy, tions interfere with the eradication of the infection as well as
as compared with cefuroxime therapy, resulted in (a) more with the management of seizure activity (263,397).

Scheld_Ch24.indd 404 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 405

As discussed, fluoroquinolone-resistant H. influenzae similar antibacterial activity to vancomycin in an experimen-


have not emerged, and these agents penetrate well into the tal model of MRSA meningitis (407), and daptomycin plus
CSF. Trovafloxacin was compared with ceftriaxone ( van- rifampin has been successfully used in patients with MRSA
comycin) in a multicenter, randomized comparative trial con- meningitis (408410).
ducted in 11 countries and enrolling children 3 months to
12 years of age (398). The major pathogens were Hib, 39%; Anaerobes
N. meningitidis, 32%; and S. pneumoniae, 21%. The overall
A combination of chloramphenicol and penicillin G has been
efficacy was similar in both groups (prompt CSF sterilization
recommended for meningitis caused by anaerobes. Penicillin G
in 94% to 96%); fluoroquinolones may be an excellent al-
has excellent activity against most anaerobes, with the excep-
ternative for Hib meningitis in patients with
-lactam allergy.
tion of Bacteroides fragilis. Chloramphenicol is active against
most B. fragilis isolates. Analogous to the experience in patients
Streptococcus agalactiae with brain abscess, we prefer metronidazole for the therapy of
Penicillin G or ampicillin or a third-generation cephalospo- the rare cases of anaerobic meningitis. Penicillin G should be
rin has been standard therapy for neonatal meningitis caused used in addition pending culture results. Metronidazole is bac-
by group B streptococci (GBS) and is the recommended ther- tericidal against virtually all strict anaerobic organisms and
apy for treatment of S. agalactiae meningitis in adults (278). penetrates into the CSF and brain well.
Additionally, the number of penicillin-resistant strains of
GBS appears to be increasing (399). Infection with strains of
S. agalactiae resistant to tetracycline, erythromycin, lincomy- Empirical Antimicrobial Therapy by
cin, and clindamycin has been reported; therefore, the use of Age-Group and Underlying Condition
these penicillin substitutes in patients with GBS infections is
not recommended (399). Neonates
The therapy of GBS meningitis in patients with life-threatening
penicillin allergy presents a problem. If third-generation cephalo- Enteric gram-negative bacilli, streptococci (in particular
sporins must be avoided, then vancomycin or teicoplanin may be S. agalactiae), and L. monocytogenes are the most com-
tried, but clinical experience is almost nonexistent. mon causative organisms of bacterial meningitis in neonates.
A third-generation cephalosporin plus ampicillin is recom-
Listeria monocytogenes mended as initial therapy in this age-group.
Ampicillin is the drug of choice (often combined with gentami- Children
cin during the initial phase of treatment) for meningitis caused
by L. monocytogenes. In addition, in a recent retrospective A combination of cefotaxime or ceftriaxone or cefepime and
review of patients with listeriosis (58% with primary bactere- vancomycin has become the antibiotic of choice for the initial
mia and 42% with meningitis), differences in mortality were treatment of acute meningitis in children in whom the etio-
not seen in those treated with ampicillin or with the combina- logic agent has not been identified. The empirical therapy of
tion of ampicillin and gentamicin. An alternative agent in a bacterial meningitis in children should include coverage for
penicillin-allergic patient is TMP-SMX, which is bactericidal S. pneumoniae, Hib, and N. meningitidis, which is provided
against Listeria in vitro. In one retrospective series, therapy by the third-generation cephalosporins; vancomycin is added
with TMP-SMX plus ampicillin was associated with a lower for pneumococcal meningitis pending in vitro susceptibility
failure rate and fewer neurologic sequelae than the combi- testing. The recommended doses of these antibiotics are listed
nation of ampicillin plus an aminoglycoside (122), although in Table 24.8.
more data are needed before this combination can be recom-
mended. Oral therapy with TMP-SMX has been used in some Adults (Ages 15 to 50 Years)
patients with Listeria meningitis and may be considered in S. pneumoniae and N. meningitidis are the causative organ-
patients who demonstrate a rapid clinical response to intrave- isms of approximately 85% of cases of bacterial meningitis in
nous therapy and in whom good adherence is expected (400). otherwise healthy adults (411). Empirical therapy of meningi-
The third-generation cephalosporins are inactive against this tis in adults should, therefore, be directed toward these organ-
organism (173). Intravenous vancomycin is not efficacious, al- isms. Ceftriaxone (4 g per day in divided doses every 12 hours)
though intraventricular vancomycin was successful in one case or cefotaxime (up to 8 to 12 g per day in divided doses every
of recurrent L. monocytogenes meningitis (401). Meropenem 4 to 6 hours) or cefepime (6 g per day in divided doses every
may be a useful alternative as it is highly active against listeriae. 8 hours) is effective therapy for meningitis caused by either
of these organisms (412); vancomycin should be added until
Staphylococci the results of antimicrobial susceptibility testing are known.
The recommended doses of these antibiotics are listed in
Meningitis caused by S. aureus (MSSA) is treated with nafcil-
Table 24.9. All CSF isolates of pneumococci and meningococci
lin or oxacillin (139,140,402,403). Vancomycin is the drug of
should be tested for penicillin or cephalosporin resistance.
choice for methicillin-resistant staphylococci and for patients
allergic to penicillin. The CSF should be monitored during
therapy, and if the spinal fluid continues to yield viable or-
Older Adults
ganisms after 48 hours of intravenous treatment, then either The most common organisms causing meningitis in adults
intrathecal or intraventricular vancomycin, 20 mg once daily older than 50 years are S. pneumoniae and enteric gram-
(in adults), can be added (402404). The role of adjunctive negative bacilli; however, meningitis caused by Listeria and
rifampin therapy is unclear, although the addition of rifampin H. influenzae is increasingly recognized. For initial therapy of
or TMP-SMX should be considered in patients not responding meningitis in elderly patients, either ceftriaxone or cefotaxime
to therapy and if the organism is susceptible (144). Linezolid or cefepime plus vancomycin, in combination with ampicillin,
has been used successfully in some patients with MRSA CNS is recommended (219,221,263). Meropenem may be an attrac-
infections (405,406). Daptomycin has been shown to have tive candidate for monotherapy in this age-group in the future.

Scheld_Ch24.indd 405 2/21/14 5:42 PM


406 Part III: Bacterial and Mycoplasmal Infections

Duration of Therapy type of immune abnormality. Knowledge of the latter helps pre-
dict the infecting organism (233,234). Patients with defects in
The standards for the duration of therapy of bacterial men- cell-mediated immunity are most susceptible to CNS infections
ingitis have been derived from clinical experience rather than by microorganisms that are intracellular parasites, the eradica-
rigid scientific analysis (413). They are basically empirical. tion of which depends on an intact T-lymphocytemacrophage
Although shorter courses of therapy may be equally effica- system. L. monocytogenes is the most common cause of bac-
cious, we recommend the following duration of treatment terial meningitis in patients with defective cell-mediated im-
as general guidelines, not rigid standards, when the etiologic munity (234). Patients with defective humoral immunity are
agent is known: N. meningitidis, 5 to 7 days; H. influenzae, 7 unable to mount an antibody response to a bacterial infection,
to 10 days; S. pneumoniae, 10 to 14 days; GBS, 14 to 21 days; and they are therefore unable to control infection caused by
and gram-negative aerobic bacilli and L. monocytogenes, 3 encapsulated bacteria. These patients are at particular risk for
to 4 weeks. Nevertheless, it must be stressed that the patients meningitis caused by S. pneumoniae, Hib, and, less commonly,
response, as assessed by clinical and laboratory parameters, N. meningitidis. Patients with neutropenia are at particular
is the most important criterion in the decision to terminate risk for meningitis caused by P. aeruginosa and members of the
therapy within this discretionary range. In a double-blind ran- Enterobacteriaceae family (233). The choice of antibiotic for
domized trial of 5 or 10 days of therapy with ceftriaxone for empirical treatment of bacterial meningitis in the immunosup-
bacterial meningitis in children beyond the neonatal period, pressed patient should be made based on the type of immune
it was determined that ceftriaxone could be discontinued in abnormality.
those patients who were stable after 5 days of treatment (414),
although the uncertainties around organism-specific data
(especially for S. pneumoniae) and the need for clinical judg-
ment at day 5 should lead to caution in reducing treatment
Adjunctive Therapy
duration (415). As is discussed in Chapter 23, the generation of bacterial cell
wall components in CSF during treatment of meningitis with
Meningitis Following Trauma antibiotics contributes to increased inflammation in the SAS
S. pneumoniae is the most common cause of meningitis follow- (18). Bacterial cell wall components stimulate the release of
ing traumatic head injury in association with the formation of inflammatory cytokines in the CNS, such as TNF, IL-1, and
a dural sinus fistula (218). H. influenzae is a less common, but prostaglandins (173). It may be possible to reduce the inflam-
also important, pathogen in this setting. A third- or fourth- matory response in the SAS and thus improve the outcome
generation cephalosporin plus vancomycin is recommended of this infection by administering antiinflammatory agents in
for empirical treatment of meningitis in patients with closed conjunction with antibiotics (172,418).
head injury. The regimen can subsequently be modified based TNF is a macrophage-secreted hormone that is released in
on the results of CSF cultures. response to bacterial endotoxin. The injection of small doses
of purified endotoxin into healthy volunteers causes the
Meningitis Following appearance of elevated serum concentrations of TNF within
Neurosurgical Procedures 90 minutes after the infusion, accompanied by symptoms
of headache, fever, rigors, and myalgia (419). Endogenous
The most common organisms causing meningitis in the patient TNF release has been observed in patients with sepsis and
who has undergone a neurosurgical procedure, with the ex- in those with meningococcemia. Damas et al. (420) detected
ception of a shunting procedure, are gram-negative bacilli and very high serum concentrations of TNF (mean, 701 339
staphylococci (223). Initial therapy of meningitis in the post- pg/mL; normal, 75 15 pg/mL) in patients in septic shock.
neurosurgical patient should be directed against gram-negative Waage et al. (421) found elevated serum TNF concentra-
bacilli, but also against P. aeruginosa and S. aureus (219). A tions in patients with meningococcal disease. The patients
third- or fourth-generation cephalosporin is recommended for with the highest concentrations (0.1 ng/mL) died. Ming
the treatment of gram-negative bacillary meningitis (223,374). et al. (422) found elevated concentrations of TNF in CSF
Ceftazidime or cefepime should be used. Cefepime is a fourth- during bacterial meningitis in both mice and humans. None
generation cephalosporin that is also active against pseudo- of the CSF samples from patients with viral (echovirus, cox-
monads. Vancomycin should be added until infection with sackievirus, or mumps virus) meningitis or other neurologic
staphylococci is excluded. diseases (e.g., multiple sclerosis) in this study contained
Coagulase-negative staphylococci and S. aureus are the measurable concentrations of TNF (422). This suggests that
most common pathogens causing CSF shunt infections. the presence of TNF in CSF may be specific for bacterial
Unless the organism is clearly susceptible to methicillin, meningitis (173).
vancomycin is recommended for shunt infections caused by TNF induces IL-1 release from endothelial cells and mac-
staphylococci (139,402,403). Therapy of methicillin-resis- rophages (423). IL-1 represents a family of polypeptides that
tant staphylococcal shunt infections should include a com- are both beneficial and detrimental to the host. The primary
bination of intravenous vancomycin and either oral rifampin sources of IL-1 are monocytes and macrophages, but IL-1
or intrashunt or intraventricular vancomycin (403,416). is also produced by brain astrocytes and microglia. IL-1
Although cefuroxime may enter ventricular fluid in the pres- is a potent chemoattractant for neutrophils, monocytes, B
ence of an infected CSF shunt, the concentrations are quite cells, and T cells; it has an important role in B-cell prolifera-
variable (417). This agent is not, therefore, recommended for tion and antibody production, as well as in T-cell activation
shunt infections. (424). IL-1 may, however, also be detrimental to the host.
IL-1 released into tissue induces a proliferative response.
Immunosuppressed Hosts
IL-1 released by astrocytes into brain tissue may contribute
As has been discussed, the risk for development of bacterial to brain gliosis and scar formation (424). IL-1 increases the
meningitis in an immunocompromised patient depends on a concentration of metabolites of arachidonic acidmost no-
number of factors, such as (a) the underlying disease and its tably PGE2 and leukotriene B4, which are potent mediators
treatment, (b) the duration of immunosuppression, and (c) the of inflammation (423).

Scheld_Ch24.indd 406 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 407

Possible therapeutic approaches to decrease the harmful ef- The results of a double-blind placebo-controlled trial of
fects of TNF and/or IL-1 might include (a) drugs or procedures 200 infants and children with bacterial meningitis demon-
to decrease their production, block their biologic activity, or strated a beneficial effect of dexamethasone therapy in reduc-
enhance removal from the circulation, (b) passive immuni- ing the incidence of sensorineural hearing loss. Patients were
zation with antibodies against TNF and IL-1, and (c) drugs treated with ceftriaxone or cefuroxime, with either dexa-
that interfere with IL-1induced arachidonic acid metabolites. methasone (0.15 mg/kg every 6 hours for 4 days) or placebo.
Corticosteroids are highly effective in reducing IL-1 produc- Of 84 patients in the placebo-treated group, 13 (15.5%) had
tion in vitro and in vivo. Many of the biologic activities of moderate or more severe bilateral hearing loss as compared
IL-1 are inflammatory; aspirin, acetaminophen, and nonste- with 3 (3.3%) of 92 of the dexamethasone-treated children
roidal antiinflammatory agents can reduce fever, muscle PGE2 (p  .01) (430). The beneficial effects of dexamethasone were
production, leukocyte chemotaxis, and so on. Therapeutic observed only in the children receiving concurrent cefuroxime
concentrations of nonsteroidal antiinflammatory agents and (which may be suboptimal therapy) and not in those treated
antipyretic blood levels of aspirin do not, however, reduce IL-1 with ceftriaxone, thus rendering interpretation difficult.
production, IL-1mediated lymphocyte activation, or IL-1 Dexamethasone appeared to be of particular benefit in chil-
synthesis of acute-phase proteins (424). dren with milder cases of Hib meningitis (430).
Passive immunization with monoclonal antibodies directed Similar trends suggesting a beneficial effect of dexametha-
against TNF and IL-1 may be a future therapeutic option. sone were observed in a third randomized, placebo-controlled
Beutler et al. (425) passively immunized mice with antiserum trial by the Dallas group for children receiving cefuroxime
to murine TNF and protected them from the lethal effects of (431). Once again, patients receiving dexamethasone became
gram-negative bacteremia. A major limitation, however, of afebrile sooner, and the CSF glucose concentration rose more
monoclonal antibody therapy for meningitis is the BBB. Even rapidly during the first day of therapy. Although the small
during active inflammation, the BBB is an effective barrier of sample size precluded a significant result from an analysis of
antibody penetration into the CSF. To achieve sufficient anti- hearing loss, the data combined with the data of the previous
body concentrations within the CSF, it would be necessary to study (430) continued to reveal an advantage for corticosteroid
produce serum concentrations of antibodies at least 20- to 100- therapy (432). The use of cefuroxime, a suboptimal agent (see
fold higher than the expected protective concentration in serum earlier discussion) (338), in approximately 160 of 260 patients
or to administer the antibody by intrathecal injection (426). in these trials had led some investigators to question the rou-
Despite aggressive supportive care and the administration tine use of dexamethasone as adjunctive therapy based on
of appropriate antimicrobial agents, the outcome for patients the results of these clinical trials (433). A metaanalysis of 11
with fulminant meningococcemia is often poor. Serum TNF randomized clinical trials (as of 1988) of dexamethasone for
concentrations correlate directly with outcome in this condi- adjunctive therapy confirmed benefit for H. influenzae type b
tion. Activated protein C (drotrecogin alfa activated) reduces meningitis, especially for hearing outcomes, and suggested
mortality in patients with severe sepsis (427) but has been benefit for pneumococcal meningitis in children if begun with
withdrawn from the worldwide market. Plasmapheresis has or before parenteral antibiotics (434).
been attempted, although on an extremely limited scale, in Another trial, from Costa Rica, randomized infants and
meningococcemia and may lead to a rapid decrease in serum children with bacterial meningitis to receive cefotaxime with
TNF concentrations and/or improved mortality and morbidity either dexamethasone or placebo (435). In this study, the
(428). Despite the lack of a controlled clinical trial, this ap- dexamethasone or placebo was administered 15 to 20 minutes
proach definitely deserves further study. before the first dose of cefotaxime in an attempt to attenu-
Clinical trials suggest a beneficial effect from dexametha- ate the SAS inflammatory response maximally. When patients
sone in the treatment of bacterial meningitis in children and were monitored for a mean of 15 months, those who had re-
adults. In a prospective, randomized trial, 429 patients with ceived adjunctive dexamethasone had a significantly decreased
bacterial meningitis were treated with either (a) dexametha- incidence of one or more neurologic sequelae, although there
sone, ampicillin, and chloramphenicol or (b) ampicillin and was only a trend in reduction of audiologic impairment.
chloramphenicol only. Dexamethasone was administered in- A review of the medical records of 97 infants and children
tramuscularly with the first dose of antibiotic, at a dose of with pneumococcal meningitis (treated from 1984 to 1990)
8 mg to children younger than 12 years and 12 mg to adults demonstrated a beneficial effect of dexamethasone therapy in
every 12 hours for 3 days. There were 56 cases of Hib men- infants and children with fulminant meningeal infection, as
ingitis, 106 cases of pneumococcal meningitis, and 267 cases defined by laboratory studies, altered level of consciousness,
of meningococcal meningitis. The case-fatality rate was sig- and the presence of septic shock and cerebrovascular instabil-
nificantly lowered in patients with pneumococcal meningitis ity. They accounted for two thirds of the deaths and had a
receiving dexamethasone; only 7 of 52 patients died, com- significantly increased incidence of seizures and permanent bi-
pared with 22 of 54 patients not receiving dexamethasone lateral moderate or greater hearing loss. Of the survivors, 1 of
(p  .01). Dexamethasone therapy also significantly reduced 8 steroid-treated patients, as compared with 7 of 13 nonste-
the incidence of hearing loss in patients with pneumococcal roid-treated patients, had moderate or severe bilateral hearing
meningitis. None of the 45 surviving patients in the dexa- loss (436). However, this was a retrospective review and there
methasone-treated group developed hearing loss, whereas were no data on differences in outcome with regard to specific
4 of 32 patients treated with antibiotics alone became deaf antibiotic used.
(p  .05) (429). However, there were no significant differences In a prospective, placebo-controlled double-blind trial of
between groups in time to afebrility or improvement in CSF dexamethasone (given at a dosage of 0.4 mg/kg every 12 hours
parameters, there was no documentation of possible adverse for 2 days) in 115 children with acute bacterial meningitis,
effects, an extraordinarily high percentage of patients pre- Hib was the infecting organism in 30 (55%) of 55 patients
sented in a comatose state, most patients (370 of 429) received in the placebo group, and in 37 (62%) of 60 patients in the
inadequate therapy for 3 to 5 days before hospitalization, the dexamethasone-treated group. N. meningitidis was the infect-
antibiotics were administered intramuscularly, and no differ- ing organism in 12 (22%) of 55 patients in the placebo group
ences in mortality were noted in patients with meningococcal and in 16 (27%) of 60 patients in the dexamethasone-treated
or Hib meningitis. group. At follow-up examination 3, 9, and 15 months after

Scheld_Ch24.indd 407 2/21/14 5:42 PM


408 Part III: Bacterial and Mycoplasmal Infections

discharge, 3 (5%) of 60 dexamethasone-treated patients had adult patients who received dexamethasone, the routine use
one or more neurologic or audiologic sequelae, compared with of adjunctive dexamethasone (given concomitant with or just
9 (16%) of 55 placebo recipients (p  .065) (437). prior to the first dose of an antimicrobial agent for maximal
Other studies questioned the routine use of adjunctive attenuation of the SAS inflammatory response) is warranted
dexamethasone in infants and children with bacterial meningi- in most adults with pneumococcal meningitis (448). A recent
tis. In one trial, there were no significant reductions in audio- study demonstrated a favorable trend toward reduced rates for
logic and neurologic sequelae with adjunctive dexamethasone death and hearing loss and no evidence that dexamethasone
therapy, although dexamethasone was given within 24 hours was harmful in patients with meningococcal meningitis (449).
of antimicrobial therapy (median, 11 hours) and the study Adjunctive dexamethasone should not be used in patients
was stopped prematurely because the standard of care became who have already received antimicrobial therapy for several
early administration of dexamethasone (438). Similarly, in the hours. Despite these positive benefits in terms of morbidity
second trial, adjunctive dexamethasone was not associated and mortality, there were some concerns regarding cognitive
with significant improvements in neurologic sequelae, devel- long-term outcome in patients treated with dexamethasone.
opmental outcome, or unilateral or bilateral deafness (439). However, a follow-up study of 87 eligible patients in which
Dexamethasone was given within 4 hours of the first anti- 46 were treated with adjunctive dexamethasone and 41 with
microbial, and there was a lack of follow-up for 13% of the placebo, neuropsychologic evaluation showed no significant
study population. In a very large study from Malawi, dexa- differences between patients treated with dexamethasone or
methasone again failed to improve outcome in children with placebo (450). In an evaluation of 357 episodes of pneumo-
bacterial meningitis, but the evaluation of hearing loss was coccal meningitis from 2006 to 2009 in the Netherlands since
suboptimal and still suggested a steroid benefit (440). implementation of adjunctive dexamethasone on a large scale
The American Academy of Pediatrics recommends con- basis, the prognosis has improved with mortality rates de-
sideration of dexamethasone therapy in infants and children creasing from 30% to 20% (451).
2 months and older with proven or suspected bacterial meningi- Despite these positive benefits, the routine use of adjunc-
tis. A daily dose of 10 to 12 mg/m2 (0.6 mg/kg) in four divided tive dexamethasone in patients with bacterial meningitis in the
doses is recommended for 3 to 4 days (441). Therapy for 2 days developing world has been controversial. In one randomized,
may also be efficacious (437,442). Children should, however, be double-blind, placebo-controlled study in adolescents and
carefully monitored for potential complications of corticosteroid adults in Vietnam with confirmed bacterial meningitis (452),
use, specifically gastrointestinal hemorrhage and hyperglycemia. patients who received adjunctive dexamethasone experienced
The concomitant use of an intravenous H2 receptor antagonist is a significant reduction in the risk of death at 1 month (relative
recommended to prevent gastrointestinal tract bleeding. If cor- risk [RR], 0.43) and the risk of death or disability at 6 months
ticosteroids are used, they should definitely be administered (RR, 0.56); the highest proportion of cases in this study were
early, that is, before or simultaneously with the first dose(s) caused by S. suis, followed by S. pneumoniae. In contrast, in
of parenteral antimicrobial agents. This is particularly impor- a randomized, double-blind, placebo-controlled study from
tant, because administration of currently available bacteriolytic Malawi, there were no significant differences in mortality at
agents (e.g., ceftriaxone) leads to rapid release of free endotoxin 40 days in the intention-to-treat analysis (56% in the dexa-
from gram-negative organisms into CSF, with an attendant ex- methasone group versus 53% in the placebo group) or when
aggeration of the hosts inflammatory response (443). the analysis was restricted to patients with proven pneumococ-
The results of a prospective, randomized, double-blind trial cal meningitis (53% in the dexamethasone group versus 50%
of adjunctive dexamethasone therapy for bacterial meningitis in the placebo group) (453). However, in this trial, almost
in 301 adults in five European countries over 9 years demon- 90% of the patients were infected with HIV and most likely
strated that dexamethasone improves the outcome in adults had advanced disease; delayed presentation was also associ-
with acute bacterial meningitis. The benefits were most striking ated with a poorer outcome, although adjusting for this factor
in the patients with pneumococcal meningitis (444). In another in the analysis had no effect. These data suggest that adjunc-
clinical trial, patients with pneumococcal meningitis who were tive dexamethasone is not beneficial in resource-poor coun-
treated with dexamethasone had a lower fatality rate than tries where a substantial number of patients are infected with
those that were not treated with dexamethasone (429). There HIV (454). In a Cochrane metaanalysis of 24 studies involving
has been concern that dexamethasone would decrease the pen- 4,041 participants, adjunctive dexamethasone did not reduce
etration of vancomycin into the CSF. In a prospective study of overall mortality, but there was a trend to lower mortality in
11 adults with community-acquired pneumococcal meningitis adults; corticosteroids were associated with lower rates of se-
that were treated with a combination of dexamethasone and vere hearing loss, any hearing loss, and neurologic sequelae,
vancomycin at a dose of 15 mg/kg every 8 hours or 7.5 mg/kg although these benefits were only seen in studies from high-
every 6 hours, there were four therapeutic failures (445). The income countries (455). In a subgroup analysis based on caus-
dose of vancomycin was well below the recommended dose of ative microorganism, corticosteroids reduced severe hearing
60 mg/kg per day. In a prospective randomized clinical trial of loss in patients with H. influenzae meningitis and mortality in
the bactericidal activity of vancomycin against cephalosporin- patients with S. pneumoniae meningitis.
resistant pneumococci in CSF of children with acute bacterial The use of adjunctive dexamethasone is of particular con-
meningitis, vancomycin in a dose of 60 mg/kg per day pen- cern in patients with pneumococcal meningitis caused by
etrated reliably into the CSF when the children were treated penicillin- and cephalosporin-resistant strains, in which case
concomitantly with dexamethasone (0.6 mg/kg per day divided patients may require antimicrobial therapy with vancomycin
into four doses for 4 days) (446). The recommended dosage of (173,278). A diminished CSF inflammatory response after
dexamethasone for adults is 8 to 10 mg intravenously every dexamethasone administration might significantly reduce
8 hours for 2 to 4 days. Dexamethasone therapy should not vancomycin penetration into CSF and delay CSF sterilization,
adversely affect the outcome of viral meningitis (447). as shown in an experimental rabbit model of penicillin-and
Many other clinical trials were undertaken to determine the cephalosporin-resistant pneumococcal meningitis. This re-
effects of adjunctive dexamethasone on outcome in patients sult was confirmed in another rabbit model of pneumococ-
with bacterial meningitis (173,278). On the basis of previous cal meningitis in which significantly lower CSF vancomycin
data, and the apparent absence of serious adverse outcomes in concentrations and differences in bacterial killing were found

Scheld_Ch24.indd 408 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 409

in the dexamethasone-treated rabbits. However, CSF van- TA B L E 2 4 . 1 0


comycin penetration was not reduced by dexamethasone in
a study in children (446), and in another study in which a TREATMENT OF INCREASED INTRACRANIAL
continuous infusion of vancomycin was used (60 mg/kg per PRESSURE
day), adequate CSF concentrations (7.2 g/mL) were achieved
despite the concomitant administration of adjunctive dexa- 1. Head of the bed elevated 30 degrees
methasone (355). CSF concentrations of ceftriaxone are not 2. Hyperventilation to maintain Paco2 between 27 and
significantly altered in animals or patients treated with adjunc- 30 mm Hga
tive dexamethasone (456,457). In contrast, in an experimental 3. Mannitol
rabbit model of cephalosporin-resistant pneumococcal men-
a. Children: 0.52.0 g/kg infused over 30 min and repeated
ingitis (458), concomitant use of dexamethasone with ceftri-
as necessary
axone resulted in higher CSF bacterial counts and a higher
number of therapeutic failures. For any patient receiving b. Adults: 1.0-g/kg bolus injection or 0.25 g/kg every 23 hr
adjunctive dexamethasone who is not improving as expected 4. Pentobarbital
or who has a pneumococcal isolate for which the cefotaxime a. Initial dose: 510 mg/kg at a rate of 1 mg/kg/min
or ceftriaxone minimal inhibitory concentration (MIC) is
b. Maintenance dosage: 13 mg/kg/hr
2.0 g/mL or greater, a repeat LP 36 to 48 hours after initia-
tion of antimicrobial therapy is recommended to document the a
sterility of CSF (278). In the study cited earlier, only 78 (72%) Although hyperventilation is beneficial (for about 1224 hr) in low-
ering intracranial pressure, caution is advised, as Paco2 values below
of 108 CSF cultures that were positive for S. pneumoniae were 25 mm Hg may produce cerebral ischemia.
submitted for in vitro susceptibility testing, and all were sus-
ceptible to penicillin (444), a finding that is unusual in many
areas of the world. In patients with pneumococcal meningitis
caused by strains that are highly resistant to penicillin or ceph- ICP exceeding 20 mm Hg is abnormal and should be
alosporins, careful observation and follow-up are critical to treated; however, outcome may be improved if pressures
determine whether use of adjunctive dexamethasone is associ- greater than 15 mm Hg are treated. The rationale for treating
ated with adverse clinical outcome in these patients (278,448). the smaller elevations in pressure is to avoid large elevations,
In addition to corticosteroids, several other adjunctive ap- or so-called plateau waves, that can lead to herniation and
proaches to the therapy of bacterial meningitis may be useful irreversible brainstem injury (180,263). Plateau waves are sus-
(418,459). These include (a) bactericidal but nonbacteriolytic tained elevations in ICP that may occur spontaneously or as
antibiotics to reduce endotoxin and other injurious substance the result of small increases in cerebral blood volume from hy-
(e.g., outer membrane vesicle) release into CSF (a theoretical poxia, fever, or intratracheal suctioning. When ICP is already
but as yet impractical method); (b) nonsteroidal antiinflam- high, plateau waves may be reached quickly and lead to brain
matory agents; (c) other prostaglandin inhibitors; (d) anti death (182,263). The treatment of increased ICP is outlined in
endotoxin-binding agents; (e) monoclonal antibodies directed Table 24.10.
against complement factor 5 (460), endotoxin, cytokines, or Nonetheless, in one study of 15 patients with bacterial
leukocyteendothelium adhesion molecules; (f) pentoxifylline; meningitis in whom intracranial pressure was measured (462),
(g) cytokine antagonists; (h) nitric oxide synthase (NOS) in- intracranial pressure was successfully lowered in most patients
hibitors (i.e., aminoguanidine); (i) thalidomide, by blocking by a broad range of measures, which consisted of sedation,
TNF release from microglia (461); (j) osmotic dehydrating steroids, normal fluid and electrolyte homeostasis, blood
agents (e.g., mannitol, glycerol); (k) scavengers of peroxyni- transfusion, albumin infusion, decrease of MAP, treatment
trite; and (l) inhibitors of matrix metalloproteinases (MMPs). with a prostacyclin analog, and eventually thiopental, ven-
triculostomy, and dihydroergotamine. In nonsurvivors, mean
intracranial pressure was significantly higher and CPP was
Treatment of Complications markedly lower than in survivors despite treatment; however,
this was not a comparative study and the results should be
interpreted with caution.
Raised Intracranial Pressure Elevating the head of the bed 30 degrees reduces the
ICP is usually increased in bacterial meningitis; therefore, this ICP. Turning the head to the side (particularly to the left)
complication should be anticipated and treated promptly. The or hyperextending the neck may trigger an increase in ICP.
clinical signs of increased ICP are (a) an altered level of con- Intratracheal suctioning or endotracheal intubation may in-
sciousness ranging from drowsiness to coma; (b) a dilated, crease ICP (182,263,463).
poorly reactive, or nonreactive pupil; (c) abnormalities of Hyperosmolar agents, such as mannitol, decrease ICP by
ocular motility; and (d) bradycardia and hypertensionthe decreasing cerebral edema. Mannitol remains almost entirely
Cushing reflex. Increasing ICP may be associated with only in the extracellular intravascular space, making this compart-
one or a combination of these clinical signs. Papilledema does ment hyperosmolar to brain tissue. The result is movement of
not develop until increased ICP has been present for several water from brain tissue into the intravascular space. Mannitol
hours; therefore, the absence of papilledema should not be can be given either as a bolus intravenous injection of 1 g/kg
used to exclude the presence of increased ICP. Increased ICP over 10 to 15 minutes or in small frequent doses of 0.25 g/
may lead to herniation. Signs of impending herniation in- kg every 2 to 3 hours. A bolus injection can be repeated at 3-
clude (a) midposition, nonreactive pupils; (b) unequal or di- to 4-hour intervals to maintain the serum osmolality between
lated, nonreactive pupils; (c) skew deviation or dysconjugate 315 and 320 mOsm/L (180,182,263,463).
eye movements; (d) decorticate or decerebrate posturing; and Dexamethasone appears to be beneficial in reducing ICP
(e) bradycardia and abnormal respiratory patterns. and cerebral edema in animal models of bacterial meningitis
Patients who are awake and alert can be watched clinically (464,465). However, its efficacy for reducing cerebral edema
for signs of advancing increased ICP. Patients who are stupor- in patients with bacterial meningitis has not been established.
ous or comatose may benefit from an ICP monitoring device. Steroids are known to be beneficial in reducing cerebral edema

Scheld_Ch24.indd 409 2/21/14 5:42 PM


410 Part III: Bacterial and Mycoplasmal Infections

surrounding tumors. In this situation, cerebral edema is largely is continuous for 90 minutes or longer can cause permanent
vasogenic in origin. Steroids reduce vasogenic edema by re- neurologic sequelae.
ducing the permeability of cerebral capillary endothelial cells For early termination of seizure activity, a short-acting an-
(466). There is experimental evidence to suggest that steroids ticonvulsant with a rapid onset of action (such as lorazepam
reduce interstitial edema in meningitis (467). Cerebral edema or diazepam) is recommended. Lorazepam is administered
in meningitis is a combination of vasogenic, cytotoxic, and intravenously in 1- to 4-mg doses in adults and in an initial
interstitial edema (170). The evidence that steroids decrease dose of 0.05 mg/kg in children. Lorazepam has a duration
vasogenic and interstitial edema suggests a role for cortico- of action three to four times longer than that of diazepam in
steroids in the management of this complication, which may adults (441); 4 mg of lorazepam is therapeutically equivalent
contribute to raised ICP. to 10 mg of diazepam (470). Diazepam is administered in a
The use of steroids to reduce cerebral edema in meningitis dose of 0.25 to 0.4 mg/kg (maximum, 10 mg) at a rate of 1 to
also has disadvantages. Steroids decrease inflammation in the 2 mg per minute. The 10-mg dose may be repeated up to three
meninges. As has been discussed, a moderate degree of inflam- times at intervals of 15 to 20 minutes (471). Diazepam has
mation in the meninges is required for the CSF penetration a half-life of 15 minutes; therefore, the blood level decreases
of many antibiotics. By reducing meningeal inflammation, the rapidly. A long-acting anticonvulsant should be administered
concentration of antibiotics in CSF is reduced. This may be immediately after lorazepam or diazepam. The long-acting
most important several days into treatment, when meningeal anticonvulsant of choice in children and adults is phenytoin
inflammation has been reduced substantially by antibiotic or fosphenytoin. Phenytoin is administered in a dose of 18 to
treatment. Steroids should, therefore, be discontinued within 20 mg/kg at a rate no faster than 50 mg per minute. Phenytoin
approximately 4 days of treatment (441). can prolong the QT interval or lead to hypotension. If either
High-dose barbiturate therapy is useful when other modal- of these side effects is observed, the rate of administration is
ities have failed to control ICP. Barbiturates decrease the cere- decreased. Fosphenytoin is a water-soluble prodrug of phe-
bral metabolic demand for oxygen and thus decrease cerebral nytoin that is converted to phenytoin by nonspecific phos-
blood flow. The result is a decrease in ICP. Pentobarbital is phatases. Doses of fosphenytoin are expressed as phenytoin
administered in an initial dose of 5 to 10 mg/kg at a rate equivalents. Infusion side effects are less common with fos-
of 1 mg/kg per minute, followed by a dose of 1 to 3 mg/ phenytoin than with phenytoin (472). Fosphenytoin is admin-
kg per hour. This therapy requires an intracranial monitor- istered in a dose of 18 to 20 mg/kg at a rate no faster than
ing device or an electroencephalogram (EEG) to monitor 150 mg per minute. Phenytoin is very effective in controlling
cerebral activity, because the clinical examination is severely convulsions without depressing consciousness or respiration
limited by the depressive effects of barbiturate. Pentobarbital (263). Intravenous phenytoin reaches peak brain and blood
is administered until the ICP is reduced below 20 mm Hg concentrations within 15 minutes (473). The half-life of phe-
or until the EEG demonstrates a suppression-burst pattern. nytoin following a loading dose is approximately 36 hours
Recommended serum concentrations of pentobarbital to re- (470). Serum concentrations greater than 25 g/mL are usu-
duce ICP are 20 to 40 g/dL. A Swan-Ganz catheter should ally necessary to terminate status epilepticus. If an 18- to
be in place to monitor cardiac output. High-dose barbiturates 20-mg/kg dose of phenytoin fails to control seizure activity, an
are associated with significant cardiac toxicity, including de- additional 500 mg of phenytoin can be given. A maintenance
creased cardiac output, decreased contractile force, arrhyth- dose of 100 mg every 6 hours (in adults) should be started
mias, and hypotension. Pentobarbital is the recommended after the loading dose.
barbiturate when barbiturate coma is desired, because this If fosphenytoin fails to control seizure activity, the patient
drug has a relatively short half-life. The half-life of pento- can be treated with intravenous levetiracetam or valproic
barbital is 24 hours, compared with the longer half-life of acid or intubated, mechanically ventilated, and treated with
phenobarbital (5 days). The use of pentobarbital allows for phenobarbital. For adults, phenobarbital is administered
a more rapid reversal of barbiturate coma than does the use intravenously at a rate of 100 mg per minute until seizure
of phenobarbital. Pentobarbital coma is maintained until the activity stops, or to a loading dose of 20 mg/kg (263). The
ICP has been below 20 mm Hg for 24 hours. The dosage of loading dose of phenobarbital in children is 20 mg/kg, ad-
barbiturate is then slowly decreased to prevent a rebound in- ministered intravenously at a rate of 30 mg per minute (474).
crease in ICP (263,463,468). The most common adverse effects of phenobarbital loading
are hypotension and respiratory depression. If these compli-
Seizures cations are managed and seizure activity continues, an addi-
tional 10 mg/kg can be given (474). The primary reason for
Seizures occur in 30% to 40% of children with acute
failure to control seizures is that anticonvulsants are admin-
bacterial meningitis (200). They occur in more than 30% of
istered in subtherapeutic doses or that the rate of administra-
adults with pneumococcal meningitis in the first few days of
tion is too slow.
illness (218). In a nationwide prospective study on adults with
The combination of phenytoin and phenobarbital controls
community-acquired bacterial meningitis, seizures occurred
seizure activity in the vast majority of patients. When they fail
in 17% of patients and were associated with severe CNS and
to do so, general anesthesia with pentobarbital can be tried.
systemic inflammation, structural CNS lesions, pneumococ-
The dose of pentobarbital is the same for children and adults:
cal meningitis, and predisposing conditions (469). The high
a loading dose of 3 to 5 mg/kg and a maintenance dose of 1 to
associated mortality rate warrants a low threshold for start-
2 mg/kg per hour (263,474). In the past, paraldehyde or a
ing anticonvulsant therapy in those with clinical suspicion
continuous intravenous diazepam drip was used to treat status
of a seizure. If not managed quickly and aggressively, status
epilepticus; however, paraldehyde is no longer available, and a
epilepticus may develop. Severe or prolonged seizure activity
diazepam drip is no longer recommended.
can produce permanent damage resulting from anoxic isch-
emic changes in areas of the temporal lobe, cerebellum, and
thalamus (263,468). The increased energy requirements of
Fluid Management
discharging neurons cannot be met by cerebral blood flow Most children with bacterial meningitis are hyponatremic
during sustained seizure activity. The result is ischemic necro- (serum sodium concentration 135 mEq/L) early in the course
sis and loss of cortical neurons (441). Status epilepticus that of their illness (441). Fifty percent of children have evidence of

Scheld_Ch24.indd 410 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 411

SIADH on admission to the hospital (208). Restriction of flu- cases, maintenance rates (1,500 to 1,700 mL/m2 daily) will be
ids to correct serum sodium is potentially important, because reached by 36 to 48 hours after admission. These recommen-
the degree and duration of hyponatremia correlate with the dations do not apply to the child who is admitted in shock or
development of neurologic sequelae. However, a rigid adher- who is severely dehydrated (441).
ence to fluid restriction, a time-honored practice in the treat-
ment of hyponatremia in children with bacterial meningitis, is Subdural Effusion
no longer recommended because of the adverse effects of hy- Most subdural effusions do not need intervention and are asso-
povolemia on cerebral perfusion pressure. A Cochrane review ciated with no permanent deficits (199). Routine subdural para-
on fluid therapy for acute bacterial meningitis concluded that centesis should be avoided. Only the rare effusion becomes an
there is some evidence that supports maintaining intravenous empyema or is large enough to have a mass effect. In either in-
fluids rather than restricting them in the first 48 hours, in set- stance, serial imaging of the fluid collection by CT or MRI scan
tings with high mortality rates and where patients present late. will allow for early detection of these complications. Although
However, where children present early and mortality rates are rare, subdural empyema must be considered in patients with
lower, there is insufficient evidence to guide practice. community-acquired bacterial meningitis and otitis or sinusitis,
The initial rate of intravenous fluid administration should focal neurologic deficits, or epileptic seizures. S. pneumoniae is
be approximately three fourths of normal maintenance re- the predominant causative organism and neurosurgical inter-
quirements, or about 1,000 to 1,200 mL/m2 daily. A 5% vention should be regarded as first-choice therapy in patients
dextrose solution with one-fourth to one-half normal saline with empyema causing midline shift and focal neurologic ab-
and 20 to 40 mEq/L potassium is recommended. The serum normalities or a decreased level of consciousness (476).
sodium concentration and urine specific gravity should be
measured every 6 to 12 hours (441). The mean duration of
hyponatremia in children with Hib meningitis in one study PREVENTION
was 20 hours (range, 0 to 240 hours) (475). The volume of
fluids administered can be gradually increased when the The chemoprophylaxis and immunoprophylaxis of bacterial
serum sodium concentration rises above 135 mEq/L. In most infections of the CNS are considered in detail in Chapter 51.

References

1. Danielson L, Mann EA. The first American account of cerebrospinal men- 17. Ward JI, Lum MK, Hall DB, et al. Invasive Haemophilus influenzae type b
ingitis. [Reprinted from Adams D, ed. Medical and agricultural register for disease in Alaska: background epidemiology for a vaccine efficacy trial.
the years 1806 and 1807, vol 1, no 5. Boston: Manning & Loring.] Rev J Infect Dis. 1986;153:1726.
Infect Dis. 1983;5:969972. 18. Fothergill LD, Wright J. Influenzal meningitis: relation of age incidence
2. North E. Concerning the epidemic of spotted fever in New England to bactericidal power of blood against causal organism. J Immunol.
[reprinted excerpts]. Rev Infect Dis. 1980;2:811816. 1933;24:273284.
3. Schwentker FF, Gelman S, Long PH. The treatment of meningococcic men- 19. Peltola H, Kayhty H, Virtanen M, et al. Prevention of Haemophilus influen-
ingitis with sulfanilamide. JAMA. 1937;108:14071408. zae type b bacteremic infections with the capsular polysaccharide vaccine.
4. Scheld WM, Mandell GL. Landmark perspective. Sulfonamides and menin- N Engl J Med. 1984;310:15611565.
gitis. JAMA. 1984;251:791794. 20. Takala AK, Eskola J, Palmgren J, et al. Risk factors of invasive
5. Thigpen MC, Whitney CG, Messonnier NE, et al. Bacterial meningitis in Haemophilus influenzae type b disease among children in Finland.
the United States, 19982007. N Engl J Med. 2011;364:20162025. J Pediatr. 1989;115:694701.
6. Takala AK, Eskola J, Peltola H, et al. Epidemiology of invasive Haemophilus 21. Cochi SL, Fleming DW, Hightower AW, et al. Primary invasive Haemophilus
influenzae type b disease among children in Finland before vaccination with influenzae type b disease: a population-based assessment of risk factors.
Haemophilus influenzae type b conjugate vaccine. Pediatr Infect Dis J. J Pediatr. 1986;108:887896.
1989;8:297302. 22. Krasinski K, Nelson JD, Butler S, et al. Possible association of mycoplasma
7. Schlech WF III, Ward JI, Band JD, et al. Bacterial meningitis in the United and viral respiratory infections with bacterial meningitis. Am J Epidemiol.
States, 1978 through 1981. The National Bacterial Meningitis Surveillance 1987;125:499508.
Study. JAMA. 1985;253:17491754. 23. Rosenthal J, Dagan R, Press J, et al. Differences in the epidemiology of
8. Sherry B, Emanuel I, Kronmal RA, et al. Interannual variation of childhood community-acquired bacterial meningitis between two eth-
the incidence of Haemophilus influenzae type b meningitis. JAMA. nic populations cohabiting in one geographic area. Pediatr Infect Dis J.
1989;261:19241929. 1988;7:630633.
9. Cochi SL, Broome CV, Hightower AW. Immunization of US children with 24. Michaels RH, Ali O. A decline in Haemophilus influenzae type b meningi-
Hemophilus influenzae type b polysaccharide vaccine. A cost-effectiveness tis. J Pediatr. 1993;122:407409.
model of strategy assessment. JAMA. 1985;253:521529. 25. Adams WG, Deaver KA, Cochi SL, et al. Decline of childhood
10. Dagan R, Isaachson M, Lang R, et al. Epidemiology of pediatric meningi- Haemophilus influenzae type b (Hib) disease in the Hib vaccine era. JAMA.
tis caused by Haemophilus influenzae type b, Streptococcus pneumoniae, 1993;269:221226.
and Neisseria meningitidis in Israel: a 3-year nationwide prospective 26. Murphy TV, White KE, Pastor P, et al. Declining incidence of Haemophilus
study. Israeli Pediatric Bacteremia and Meningitis Group. J Infect Dis. influenzae type b disease since introduction of vaccination. JAMA.
1994;169:912916. 1993;269:246248.
11. Ostroy PR. Bacterial meningitis in Washington state. West J Med. 27. Broadhurst LE, Erickson RL, Kelley PW. Decreases in invasive Haemophilus
1979;131:339343. influenzae diseases in US Army children, 1984 through 1991. JAMA.
12. Santosham M, Kallman CH, Neff JM, et al. Absence of increasing inci- 1993;269:227231.
dence of meningitis caused by Haemophilus influenza type b. J Infect Dis. 28. Vadheim CM, Greenberg DP, Eriksen E, et al. Eradication of Haemophilus
1979;140:10091012. influenzae type b disease in southern California. Kaiser-UCLA Vaccine
13. Fraser DW, Darby CP, Koehler RE, et al. Risk factors in bacterial meningi- Study Group. Arch Pediatr Adolesc Med. 1994;148:5156.
tis: Charleston County, South Carolina. J Infect Dis. 1973;127:271277. 29. Peltola H, Kilpi T, Anttila M. Rapid disappearance of Haemophilus influ-
14. Granoff DM, Basden M. Haemophilus influenzae infections in Fresno enzae type b meningitis after routine childhood immunisation with conju-
County, California: a prospective study of the effects of age, race, and con- gate vaccines. Lancet. 1992;340:592594.
tact with a case on incidence of disease. J Infect Dis. 1980;141:4046. 30. Urwin G, Yuan MF, Feldman RA. Prospective study of bacterial meningitis
15. Peltola H, Virtanen M. Systemic Haemophilus influenzae infection in in North East Thames region, 19913, during introduction of Haemophilus
Finland. Clin Pediatr (Phila). 1984;23:275280. influenzae vaccine. BMJ. 1994;309:14121414.
16. Takala AK. Epidemiologic characteristics and risk factors for invasive 31. Kojouharova M, Gatcheva N, Setchanova L, et al. Childhood bacterial
Haemophilus influenzae type b disease in a population with high vaccine meningitis in Bulgaria: a population-based retrospective study in six re-
efficacy. Pediatr Infect Dis J. 1989;8:343346. gions during 199296. Int J Infect Dis. 2003;7:109112.

Scheld_Ch24.indd 411 2/21/14 5:42 PM


412 Part III: Bacterial and Mycoplasmal Infections

32. Garner D, Weston V. Effectiveness of vaccination for Haemophilus influen- 60. Santolaya ME, ORyan ML, Valenzuela MT, et al. Immunogenicity and
zae type b. Lancet. 2003;361:395396. tolerability of a multicomponent meningococcal serogroup B (4CMenB)
33. Steinhoff M, Goldblatt D. Conjugate Hib vaccines. Lancet. 2003;361: vaccine in healthy adolescents in Chile: a phase 2b/3 randomised, observer-
360361. blind, placebo-controlled study. Lancet. 2012;379:617624.
34. Gessner BD, Sutanto A, Linehan M, et al. Incidences of vaccine-preventable 61. Stephens DS. Prevention of serogroup B meningococcal disease. Lancet.
Haemophilus influenzae type b pneumonia and meningitis in Indonesian 2012;379:592594.
children: hamlet-randomised vaccine-probe trial. Lancet. 2005;365:4352. 62. Schuchat A, Robinson K, Wenger JD, et al. Bacterial meningitis in the
35. Cowgill KD, Ndiritu M, Nyiro J, et al. Effectiveness of Haemophilus United States in 1995. Active Surveillance Team. N Engl J Med. 1997;337:
influenzae type b conjugate vaccine introduction into routine childhood 970976.
immunization in Kenya. JAMA. 2006;296:671678. 63. Istre GR, Tarpay M, Anderson M, et al. Invasive disease due to
36. Daza P, Banda R, Misoya K, et al. The impact of routine infant immuni- Streptococcus pneumoniae in an area with a high rate of relative penicillin
zation with Haemophilus influenzae type b conjugate vaccine in Malawi, resistance. J Infect Dis. 1987;156:732735.
a country with high human immunodeficiency virus prevalence. Vaccine. 64. Burman LA, Norrby R, Trollfors B. Invasive pneumococcal infections:
2006;24:62326239. incidence, predisposing factors, and prognosis. Rev Infect Dis. 1985;7:
37. Adegbola RA, Secka O, Lahai G, et al. Elimination of Haemophilus 133142.
influenzae type b (Hib) disease from The Gambia after the introduction of 65. Kragsbjerg P, Kllman J, Oleen P. Pneumococcal meningitis in adults. Scand
routine immunisation with a Hib conjugate vaccine: a prospective study. J Infect Dis. 1994:659666.
Lancet. 2005;366:144150. 66. Davidson M, Schraer CD, Parkinson AJ, et al. Invasive pneumococ-
38. Takala AK, Eskola J, van Alphen L. Spectrum of invasive Haemophilus cal disease in an Alaska native population, 1980 through 1986. JAMA.
influenzae type b disease in adults. Arch Intern Med. 1990;150:25732576. 1989;261:715718.
39. Farley MM, Stephens DS, Brachman PS Jr, et al. Invasive Haemophilus 67. Gray BM, Dillon HC Jr. Epidemiological studies of Streptococcus pneu-
influenzae disease in adults. A prospective, population-based surveil- moniae in infants: antibody to types 3, 6, 14, and 23 in the first two years
lance. CDC Meningitis Surveillance Group. Ann Intern Med. 1992;116: of life. J Infect Dis. 1988;158:948955.
806812. 68. Adriani KS, Brouwer MC, Geldhoff M, et al. Common polymorphisms in
40. Rubach MP, Bender JM, Mottice S, et al. Increasing incidence of invasive the complement system and susceptiblity to bacterial meningitis. J Infect.
Haemophilus influenzae disease in adults, Utah, USA. Emerg Infect Dis. 2013;66:255262.
2011;17:16451650. 69. Scheld WM. Bacterial meningitis in the patient at risk: intrinsic risk factors
41. Peltola H. Meningococcal disease: still with us. Rev Infect Dis. 1983;5: and host defense mechanisms. Am J Med. 1984;76:193207.
7191. 70. Musher DM. Infections caused by Streptococcus pneumoniae: clini-
42. Reido FX, Plikaytis BD, Broome CV. Epidemiology and prevention of cal spectrum, pathogenesis, immunity, and treatment. Clin Infect Dis.
meningococcal disease. Pediatr Infect Dis J. 1995;14:643657. 1992;14:801807.
43. Scheld WM. Meningococcal diseases. In: Warren KS, Mahmoud AAF, eds. 71. Godeau B, Bachir D, Schaeffer A, et al. Severe pneumococcal sepsis and
Tropical and Geographical Medicine. 2nd ed. New York: McGraw-Hill; meningitis in human immunodeficiency virus-infected adults with sickle cell
1990:798814. disease. Clin Infect Dis. 1992;15:327329.
44. Jackson LA, Wenger JD. Laboratory-based surveillance for meningococcal 72. Weisfelt M, van de Beek D, Spanjaard L, et al. Clinical features, complica-
disease in selected areas, United States, 19891991. MMWR CDC Surveill tions, and outcome in adults with pneumococcal meningitis: a prospective
Summ. 1993;42:2130. case series. Lancet Neurol. 2006;5:123129.
45. Schwartz B, Moore PS, Broome CV. The global epidemiology of meningo- 73. Biernath KR, Reefhuis J, Whitney CG, et al. Bacterial meningitis among
coccal disease. Clin Microbiol Rev. 1989;2:S118S124. children with cochlear implants beyond 24 months after implantation.
46. Stephens DS, Hajjeh RA, Baughman WS, et al. Sporadic meningococcal Pediatrics. 2006;117:284289.
disease in adults: results of a 5-year population-based study. Ann Intern 74. Wei BP, Robins-Browne RM, Shepherd RK, et al. Can we prevent cochlear
Med. 1995;123:937940. implant recipients from developing pneumococcal meningitis? Clin Infect
47. Gardner P. Clinical practice. Prevention of meningococcal disease. N Engl Dis. 2008;46:e1e7.
J Med. 2006;355:14661473. 75. Greenwood B. Pneumococcal meningitis epidemics in Africa. Clin Infect
48. Goldschneider I, Gotschlich EC, Artenstein MS. Human immunity Dis. 2006;43:701703.
to the meningococcus. I. The role of humoral antibodies. J Exp Med. 76. Hsu HE, Shutt KA, Moore MR, et al. Effect of pneumococcal conjugate
1969;129:13071326. vaccine on pneumococcal meningitis. N Engl J Med. 2009;360:244256.
49. Brouwer MC, de Gans J, Heckenberg SG, et al. Host genetic susceptibil- 77. Nuorti JP, Whitney CG; Centers for Disease Control and Prevention.
ity to pneumococcal and meningococcal disease: a systematic review and Prevention of pneumococcal disease among infants and childrenuse of
meta-analysis. Lancet Infect Dis. 2009;9:3144. 13-valent pneumococcal conjugate vaccine and 23-valent pneumococcal
50. Ross SC, Densen P. Complement deficiency states and infection: epidemiol- polysaccharide vaccinerecommendations of the Advisory Committee on
ogy, pathogenesis and consequences of neisserial and other infections in an Immunization Practices (ACIP). MMWR Recomm Rep. 2010;59:118.
immune deficiency. Medicine (Baltimore). 1984;63:243273. 78. Durand ML, Calderwood SB, Weber DJ, et al. Acute bacterial meningitis in
51. Fijen CA, Kuijper EJ, Tjia HG, et al. Complement deficiency predisposes adults. A review of 493 episodes. N Engl J Med. 1993;328:2128.
for meningitis due to nongroupable meningococci and Neisseria-related 79. Wenger JD, Hightower AW, Facklam RR, et al. Bacterial meningitis in the
bacteria. Clin Infect Dis. 1994;18:780784. United States, 1986: report of a multistate surveillance study. The Bacterial
52. Sjoholm AG, Kuijper EJ, Tijssen CC, et al. Dysfunctional properdin in a Meningitis Study Group. J Infect Dis. 1990;162:13161323.
Dutch family with meningococcal disease. N Engl J Med. 1988;319:3337. 80. Musser JM, Kroll JS, Granoff DM, et al. Global genetic structure and mo-
53. Overturf GD. Indications for the immunological evaluation of patients lecular epidemiology of encapsulated Haemophilus influenzae. Rev Infect
with meningitis. Clin Infect Dis. 2003;36:189194. Dis. 1990;12:75111.
54. Davila S, Wright VJ, Khor CC, et al. Genome-wide association study identi- 81. Moore PS, Reeves MW, Schwartz B, et al. Intercontinental spread of
fies variants in the CFH region associated with host susceptibility to menin- an epidemic group A Neisseria meningitidis strain. Lancet. 1989;2:
gococcal disease. Nat Genet. 2010;42:772776. 260263.
55. Centers for Disease Control and Prevention Advisory Committee on 82. Whalen CM, Hockin JC, Ryan A, et al. The changing epidemiology of
Immunization Practices. Revised recommendations of the Advisory Committee invasive meningococcal disease in Canada, 1985 through 1992. Emergence
on Immunization Practices to vaccinate all persons aged 1118 years of a virulent clone of Neisseria meningitidis. JAMA. 1995;273:390394.
with meningococcal conjugate vaccine. MMWR Morb Mortal Wkly Rep. 83. Jackson LA, Schuchat A, Reeves MW, et al. Serogroup C meningococcal
2007;56:794795. outbreaks in the United States. An emerging threat. JAMA. 1995;273:
56. Centers for Disease Control and Prevention. Updated recommendations 383389.
for use of meningococcal conjugate vaccinesAdvisory Committee on 84. Harrison LH. The worldwide prevention of meningococcal infection. Still
Immunization Practices (ACIP), 2010. MMWR Morb Mortal Wkly Rep. an elusive goal. JAMA. 1995;273:419421.
2011;60:7276. 85. Rosenstein NE, Perkins BA, Stephens DS, et al. The changing epidemiology
57. Centers for Disease Control and Prevention. Recommendation of the of meningococcal disease in the United States, 19921996. J Infect Dis.
Advisory Committee on Immunization Practices (ACIP) for use of quad- 1999;180:18941901.
rivalent meningococcal conjugate vaccine (MenACWY-D) among children 86. Moura AS, Pablos-Mendez A, Layton M, et al. Epidemiology of menin-
aged 9 through 23 months at increased risk for invasive meningococcal gococcal disease, New York City, 19892000. Emerg Infect Dis. 2003;9:
disease. MMWR Morb Mortal Wkly Rep. 2011;60:13911392. 355361.
58. Vesikari T, Esposito S, Prymula R, et al. Immunogenicity and safety of an 87. Mastrantonio P, Stefanelli P, Fazio C, et al. Serotype distribution, antibi-
investigational multicomponent, recombinant, meningococcal serogroup otic susceptibility, and genetic relatedness of Neisseria meningitidis strains
B vaccine (4CMenB) administered concomitantly with routine infant and recently isolated in Italy. Clin Infect Dis. 2003;36:422428.
child vaccinations: results of two randomised trials. Lancet. 2013;381: 88. Bilukha OO, Rosenstein N; National Center for Infectious Diseases,
825835. Centers for Disease Control and Prevention. Prevention and control of
59. Snape MD, Pollard AJ. The beginning of the end for serogroup B meningococcal disease. Recommendations of the Advisory Committee on
meningococcus? Lancet. 2013;381:785787. Immunization Practices (ACIP). MMWR Recomm Rep. 2005;54:121.

Scheld_Ch24.indd 412 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 413

89. Cohn AC, MacNeil JR, Harrison LH, et al. Changes in Neisseria meningit- 118. Decker CF, Simon GL, DiGioia RA, et al. Listeria monocytogenes infec-
idis disease epidemiology in the United States, 19982007: implications for tions in patients with AIDS: report of five cases and review. Rev Infect
prevention of meningococcal disease. Clin Infect Dis. 2010;50:184191. Dis. 1991;13:413417.
90. Centers for Disease Control and Prevention. Notes from the field: sero- 119. Berenguer J, Solera J, Diaz MD, et al. Listeriosis in patients infected with
group C invasive meningococcal disease among men who have sex with human immunodeficiency virus. Rev Infect Dis. 1991;13:115119.
menNew York City, 20102012. MMWR Morb Mortal Wkly Rep. 120. Lorber B. Listeriosis. Clin Infect Dis. 1997;24:19; quiz 1011.
2013;61:1048. 121. Mylonakis E, Hohmann EL, Calderwood SB. Central nervous system
91. Wilder-Smith A, Goh KT, Barkham T, et al. Hajj-associated outbreak infection with Listeria monocytogenes. 33 years experience at a gen-
strain of Neisseria meningitidis serogroup W135: estimates of the attack eral hospital and review of 776 episodes from the literature. Medicine
rate in a defined population and the risk of invasive disease developing in (Baltimore). 1998;77:313336.
carriers. Clin Infect Dis. 2003;36:679683. 122. Clauss HE, Lorber B. Central nervous system infection with Listeria
92. Mayer LW, Reeves MW, Al-Hamdan N, et al. Outbreak of W135 me- monocytogenes. Curr Infect Dis Rep. 2008;10:300306.
ningococcal disease in 2000: not emergence of a new W135 strain but 123. Goulet V, Hebert M, Hedberg C, et al. Incidence of listeriosis and related
clonal expansion within the electophoretic type-37 complex. J Infect Dis. mortality among groups at risk of acquiring listeriosis. Clin Infect Dis.
2002;185:15961605. 2012;54:652660.
93. Boisier P, Nicolas P, Djibo S, et al. Meningococcal meningitis: unprec- 124. Kamath BM, Mamula P, Baldassano RN, et al. Listeria meningitis after
edented incidence of serogroup X-related cases in 2006 in Niger. Clin treatment with infliximab. J Pediatr Gastroenterol Nutr. 2002;34:410412.
Infect Dis. 2007;44:657663. 125. Chuang MH, Singh J, Ashouri N, et al. Listeria meningitis after inf-
94. Delrieu I, Yaro S, Tamekloe TA, et al. Emergence of epidemic Neisseria liximab treatment of ulcerative colitis. J Pediatr Gastroenterol Nutr.
meningitidis serogroup X meningitis in Togo and Burkina Faso. PLoS 2010;50:337339.
One. 2011;6:e19513. 126. La Montagna G, Valentini G. Listeria monocytogenes meningitis in a patient
95. Frasch CE, Zollinger WD, Poolman JT. Serotype antigens of Neisseria receiving etanercept for Stills disease. Clin Exp Rheumatol. 2005;23:121.
meningitidis and a proposed scheme for designation of serotypes. Rev 127. Zuniga M, Aguado JM, Vada J. Listeria monocytogenes meningitis in pre-
Infect Dis. 1985;7:504510. viously healthy adults: long-term follow-up. Q J Med. 1992;85:911915.
96. Spanjaard L, Bol P, de Marie S, et al. Association of meningococcal sero- 128. Ben Shimol S, Einhorn M, Greenberg D. Listeria meningitis and ven-
types with the course of disease: serotypes 2a and 2b in the Netherlands, triculitis in an immunocompetent child: case report and literature review.
19591981. J Infect Dis. 1987;155:277282. Infection. 2012;40:207211.
97. Olyhoek T, Crowe BA, Achtman M. Clonal population structure of 129. Schuchat A, Deaver KA, Wenger JD, et al. Role of foods in sporadic lis-
Neisseria meningitidis serogroup A isolated from epidemics and pandem- teriosis. I. Case-control study of dietary risk factors. The Listeria Study
ics between 1915 and 1983. Rev Infect Dis. 1987;9:665692. Group. JAMA. 1992;267:20412045.
98. Crowe BA, Wall RA, Kusecek B, et al. Clonal and variable properties of 130. Pinner RW, Schuchat A, Swaminathan B, et al. Role of foods in sporadic
Neisseria meningitidis isolated from cases and carriers during and after an listeriosis. II. Microbiologic and epidemiologic investigation. The Listeria
epidemic in the Gambia, West Africa. J Infect Dis. 1989;159:686700. Study Group. JAMA. 1992;267:20462050.
99. Caugant DA, Kristiansen BE, Froholm LO, et al. Clonal diversity of 131. Gottlieb SL, Newbern EC, Griffin PM, et al. Multistate outbreak of liste-
Neisseria meningitidis from a population of asymptomatic carriers. Infect riosis linked to turkey deli meat and subsequent changes in US regulatory
Immun. 1988;56:20602068. policy. Clin Infect Dis. 2006;42:2936.
100. Kellerman SE, McCombs K, Ray M, et al. Genotype-specific carriage of 132. Centers for Disease Control and Prevention. Outbreak of invasive liste-
Neisseria meningitidis in Georgia counties with hyper- and hyposporadic riosis associated with the consumption of hog head cheeseLouisiana,
rates of meningococcal disease. J Infect Dis. 2002;186:4048. 2010. MMWR Morb Mortal Wkly Rep. 2011;60:401405.
101. Lingappa JR, Al-Rabeah AM, Hajjeh R, et al. Serogroup W-135 meningo- 133. Centers for Disease Control and Prevention. Multistate outbreak of
coccal disease during the Hajj, 2000. Emerg Infect Dis. 2003;9:665671. listeriosis associated with Jensen Farms cantaloupeUnited States,
102. Hahne S, Handford S, Ramsay M. W135 meningococcal carriage in Hajj AugustSeptember 2011. MMWR Morb Mortal Wkly Rep. 2011;60:
pilgrims. Lancet. 2002;360:20892090. 13571358.
103. Bonacorsi S, Clermont O, Houdouin V, et al. Molecular analysis and 134. Gaul LK, Farag NH, Shim T, et al. Hospital-acquired listeriosis outbreak
experimental virulence of French and North American Escherichia coli caused by contaminated diced celeryTexas, 2010. Clin Infect Dis.
neonatal meningitis isolates: identification of a new virulent clone. J Infect 2013;56:2026.
Dis. 2003;187:18951906. 135. Bula CJ, Bille J, Glauser MP. An epidemic of food-borne listeriosis in west-
104. Unhanand M, Mustafa MM, McCracken GH Jr, et al. Gram-negative ern Switzerland: description of 57 cases involving adults. Clin Infect Dis.
enteric bacillary meningitis: a twenty-one-year experience. J Pediatr. 1995;20:6672.
1993;122:1521. 136. Voetsch AC, Angulo FJ, Jones TF, et al. Reduction in the incidence of
105. Leonard MK, Murrow JR, Jurado R, et al. Salmonella meningitis in adults invasive listeriosis in foodborne diseases active surveillance network sites,
infected with HIV: case report and review of the literature. Am J Med Sci. 19962003. Clin Infect Dis. 2007;44:513520.
2002;323:266268. 137. Bennion JR, Sorvillo F, Wise ME, et al. Decreasing listeriosis mortality in
106. Chang WN, Tsai YC, Chien CC, et al. Frequent association with neuro- the United States, 19902005. Clin Infect Dis. 2008;47:867874.
surgical conditions in adult Proteus mirabilis meningitis: report of five 138. Koopmans MM, Brouwer MC, Bijlsma MW, et al. Listeria monocyto-
cases. Clin Neurol Neurosurg. 2002;104:121124. genes sequence type 6 and increased rate of unfavorable outcome in men-
107. Reichert MC, Medeiros EA, Ferraz FA. Hospital-acquired meningitis in ingitis: epidemiologic cohort study. Clin Infect Dis. 2013;57:247253.
patients undergoing craniotomy: incidence, evolution, and risk factors. 139. Schlesinger LS, Ross SC, Schaberg DR. Staphylococcus aureus meningitis: a
Am J Infect Control. 2002;30:158164. broad-based epidemiologic study. Medicine (Baltimore). 1987;66:148156.
108. Kim HI, Kim SW, Park GY, et al. The causes and treatment outcomes 140. Kim JH, van der Horst C, Mulrow CD, et al. Staphylococcus aureus men-
of 91 patients with adult nosocomial meningitis. Korean J Intern Med. ingitis: review of 28 cases. Rev Infect Dis. 1989;11:698706.
2012;27:171179. 141. Jensen AG, Espersen F, Skinhoj P, et al. Staphylococcus aureus menin-
109. Dillon HC Jr, Khare S, Gray BM. Group B streptococcal carriage and gitis. A review of 104 nationwide, consecutive cases. Arch Intern Med.
disease: a 6-year prospective study. J Pediatr. 1987;110:3136. 1993;153:19021908.
110. Stoll BJ, Hansen N, Fanaroff AA, et al. Changes in pathogens caus- 142. Arda B, Yamazhan T, Sipahi OR, et al. Meningitis due to methicillin-
ing early-onset sepsis in very-low-birth-weight infants. N Engl J Med. resistant Staphylococcus aureus (MRSA): review of 10 cases. Int J
2002;347:240247. Antimicrob Agents. 2005;25:414418.
111. Henrichsen J, Ferrieri P, Jelinkova J, et al. Nomenclature of antigens of 143. Aguilar J, Urday-Cornejo V, Donabedian S, et al. Staphylococcus aureus
group B streptococci. Int J Syst Bacteriol. 1984;34:500. meningitis: case series and literature review. Medicine (Baltimore).
112. Musser JM, Mattingly SJ, Quentin R, et al. Identification of a high-virulence 2010;89:117125.
clone of type III Streptococcus agalactiae (group B Streptococcus) causing 144. Pintado V, Pazos R, Jimenez-Mejias ME, et al. Methicillin-resistant
invasive neonatal disease. Proc Natl Acad Sci U S A. 1989;86:47314735. Staphylococcus aureus meningitis in adults: a multicenter study of
113. Gaschignard J, Levy C, Romain O, et al. Neonatal bacterial meningitis: 86 cases. Medicine (Baltimore). 2012;91:1017.
444 cases in 7 years. Pediatr Infect Dis J. 2011;30:212217. 145. Heerema MS, Ein ME, Musher DM, et al. Anaerobic bacterial meningitis.
114. Libster R, Edwards KM, Levent F, et al. Long-term outcomes of group B Am J Med. 1979;67:219227.
streptococcal meningitis. Pediatrics. 2012;130:e8e15. 146. Law DA, Aronoff SC. Anaerobic meningitis in children: case report and
115. Farley MM, Harvey RC, Stull T, et al. A population-based assessment review of the literature. Pediatr Infect Dis J. 1992;11:968971.
of invasive disease due to group B Streptococcus in nonpregnant adults. 147. Feder HM Jr. Bacteroides fragilis meningitis. Rev Infect Dis. 1987;9:
N Engl J Med. 1993;328:18071811. 783786.
116. Dunne DW, Quagliarello V. Group B streptococcal meningitis in adults. 148. Tarnvik A. Anaerobic meningitis in children. Eur J Clin Microbiol.
Medicine (Baltimore). 1993;72:110. 1986;5:271274.
117. Mascola L, Sorvillo F, Neal J, et al. Surveillance of listeriosis in Los 149. Long JG, Preblud SR, Keyserling HL. Clostridium perfringens meningi-
Angeles County, 19851986. A first years report. Arch Intern Med. tis in an infant: case report and literature review. Pediatr Infect Dis J.
1989;149:15691572. 1987;6:752754.

Scheld_Ch24.indd 413 2/21/14 5:42 PM


414 Part III: Bacterial and Mycoplasmal Infections

150. van de Beek D, de Gans J, Spanjaard L, et al. Group a streptococcal men- 185. Saez-Llorens X, McCracken GH Jr. Bacterial meningitis in children.
ingitis in adults: report of 41 cases and a review of the literature. Clin Lancet. 2003;361:21392148.
Infect Dis. 2002;34:e32e36. 186. Kim KS. Acute bacterial meningitis in infants and children. Lancet Infect
151. Bignardi GE, Isaacs D. Neonatal meningitis due to Streptococcus mitis. Dis. 2010;10:3242.
Rev Infect Dis. 1989;11:8688. 187. Brudzinski J. Un signe nouveau sur les membres infrieurs dans les mningites
152. Stevenson KB, Murray EW, Sarubbi FA. Enterococcal meningitis: report chez les infants (signe de la nuque). Arch Med Enfants. 1909;12:745752.
of four cases and review. Clin Infect Dis. 1994;18:233239. 188. Verghese A, Gallemore G. Kernigs and Brudzinskis signs revisited. Rev
153. Knoll BM, Hellmann M, Kotton CN. Vancomycin-resistant Enterococcus Infect Dis. 1987;9:11871192.
faecium meningitis in adults: case series and review of the literature. Scand 189. Kernig VM. ber ein krankheits symptom der acuten meningitis.
J Infect Dis. 2013;45:131139. St Petersburg Med Wochenschr. 1882;7:398.
154. Rajasekhar A, Clancy CJ. Meningitis due to group C Streptococcus: a case 190. DeMyer W. Examination of the patient who has a disorder of conscious-
report and review of the literature. Scand J Infect Dis. 2010;42:571578. ness. In: DeMyer W, ed. Technique of the Neurologic Examination.
155. Minces LR, Brown PJ, Veldkamp PJ. Human meningitis from 3rd ed. New York: McGraw-Hill; 1980:359404.
Streptococcus equi subsp. zooepidemicus acquired as zoonoses. Epidemiol 191. Valmari P, Peltola H, Ruuskanen O, et al. Childhood bacterial meningitis:
Infect. 2011;139:406410. initial symptoms and signs related to age, and reasons for consulting a
156. Lu CH, Chang WN, Chang HW. Adults with meningitis caused by viri- physician. Eur J Pediatr. 1987;146:515518.
dans streptococci. Infection. 2001;29:305309. 192. Fontana A, Kristensen F, Dubs R, et al. Production of prostaglandin E and
157. Centers for Disease Control and Prevention. Bacterial meningitis after in- an interleukin-1 like factor by cultured astrocytes and C6 glioma cells.
trapartum spinal anesthesiaNew York and Ohio, 20082009. MMWR J Immunol. 1982;129:24132419.
Morb Mortal Wkly Rep. 2010;59:6569. 193. Gururaj VJ, Russo RM, Allen JE, et al. To tap or not to tap. What are the
158. Rubin L, Sprecher H, Kabaha A, et al. Meningitis following spinal anesthe- best indicators for performing a lumbar puncture in an outpatient child?
sia: 6 cases in 5 years. Infect Control Hosp Epidemiol. 2007;28:11871190. Clin Pediatr (Phila). 1973;12:488493.
159. Hsu J, Jensen B, Arduino M, et al. Streptococcal meningitis following 194. Amarilyo G, Alper A, Ben-Tov A, et al. Diagnostic accuracy of clinical
myelogram procedures. Infect Control Hosp Epidemiol. 2007;28:614617. symptoms and signs in children with meningitis. Pediatr Emerg Care.
160. Mai NT, Hoa NT, Nga TV, et al. Streptococcus suis meningitis in adults in 2011;27:196199.
Vietnam. Clin Infect Dis. 2008;46:659667. 195. Geiseler PJ, Nelson KE. Bacterial meningitis without clinical signs of men-
161. Nghia HD, Tu le TP, Wolbers M, et al. Risk factors of Streptococcus suis ingeal irritation. South Med J. 1982;75:448450.
infection in Vietnam. A case-control study. PLoS One. 2011;6:e17604. 196. McCarthy PL, Sharpe MR, Spiesel SZ, et al. Observation scales to identify
162. Downs NJ, Hodges GR, Taylor SA. Mixed bacterial meningitis. Rev serious illness in febrile children. Pediatrics. 1982;70:802809.
Infect Dis. 1987;9:693703. 197. Klein JO, Feigin RD, McCracken GH Jr. Report of the Task Force on
163. Sferra TJ, Pacini DL. Simultaneous recovery of bacterial and viral patho- Diagnosis and Management of Meningitis. Pediatrics. 1986;78:959982.
gens from cerebrospinal fluid. Pediatr Infect Dis J. 1988;7:552556. 198. Dodge PR, Swartz MN. Bacterial meningitisa review of selected aspects.
164. Gerber J, Nau R. Mechanisms of injury in bacterial meningitis. Curr Opin II. Special neurologic problems, postmeningitic complications and clinico-
Neurol. 2010;23:312318. pathological correlations. N Engl J Med. 1965;272:10031010.
165. Kim KS. Current concepts on the pathogenesis of Escherichia coli men- 199. Kaplan SL, Fishman MA. Update on bacterial meningitis. J Child Neurol.
ingitis: implications for therapy and prevention. Curr Opin Infect Dis. 1988;3:8293.
2012;25:273278. 200. Dunn EW. Neurologic complications of bacterial meningitis. In: Dunn
166. Koedel U, Klein M, Pfister HW. New understandings on the pathophysiol- DW, Epstein LG, eds. Decision Making in Child Neurology. Toronto: BC
ogy of bacterial meningitis. Curr Opin Infect Dis. 2010;23:217223. Decker; 1987:162163.
167. Koedel U, Scheld WM, Pfister HW. Pathogenesis and pathophysiology of 201. Rutter N, Smales OR. Role of routine investigations in children presenting
pneumococcal meningitis. Lancet Infect Dis. 2002;2:721736. with their first febrile convulsion. Arch Dis Child. 1977;52:188191.
168. Mook-Kanamori BB, Geldhoff M, van der Poll T, et al. Pathogenesis 202. Lorber J, Sunderland R. Lumbar puncture in children with convulsions
and pathophysiology of pneumococcal meningitis. Clin Microbiol Rev. associated with fever. Lancet. 1980;1:785786.
2011;24:557591. 203. Nelson KB, Ellenberg JH. Febrile seizures. In: Dreifuss FE, ed. Pediatric
169. Quagliarello V, Scheld WM. Bacterial meningitis: pathogenesis, patho- Epileptology. Boston: John Wright; 1983:173198.
physiology, and progress. N Engl J Med. 1992;327:864872. 204. Tunkel AR, Scheld WM. Acute meningitis. In: Mandell GL, Bennett JE,
170. Scheld WM, Koedel U, Nathan B, et al. Pathophysiology of bacte- Dolin R, eds. Principles and Practice of Infectious Diseases. 4th ed.
rial meningitis: mechanism(s) of neuronal injury. J Infect Dis. 2002; New York: Churchill Livingstone; 1995:831865.
186(suppl 2):S225S233. 205. Harter DH, Jubelt B. Bacterial infections. In: Rowland LP, ed. Merritts
171. Tunkel AR. Bacterial Meningitis. Philadelphia: Lippincott Williams & Textbook of Neurology. Philadelphia: Lea & Febiger; 1984:5372.
Wilkins; 2001. 206. Frederiks JAM. Meningococcal meningitis. In: Vinken PJ, Bruyn GW,
172. Tunkel AR, Scheld WM. Pathogenesis and pathophysiology of bacterial Klawans HL, et al, eds. Handbook of Clinical Neurology. Amsterdam:
meningitis. Clin Microbiol Rev. 1993;6:118136. Elsevier; 1988:2140.
173. Tunkel AR, Wispelwey B, Scheld WM. Bacterial meningitis: recent ad- 207. Kaplan SL, Feigin RD. Clinical presentations, prognostic factors and
vances in pathophysiology and treatment. Ann Intern Med. 1990;112: diagnosis of bacterial meningitis. In: Sande MA, Smith AL, Root RK, eds.
610623. Bacterial Meningitis. New York: Churchill Livingstone; 1985:8394.
174. van der Flier M, Geelen SP, Kimpen JL, et al. Reprogramming the host 208. Kaplan SL, Feigin RD. The syndrome of inappropriate secretion of
response in bacterial meningitis: how best to improve outcome? Clin antidiuretic hormone in children with bacterial meningitis. J Pediatr.
Microbiol Rev. 2003;16:415429. 1978;92:758761.
175. Adams RD, Kubik CS, Bonner FJ. The clinical and pathological aspects of 209. Kanakriyeh M, Carvajal HF, Vallone AM. Initial fluid therapy for children
B influenzal meningitis. Arch Pediatr. 1948;65:408441. with meningitis with consideration of the syndrome of inappropriate anti-
176. Escourolle R, Poirier J. Pathology of infectious diseases. In: Escourolle diuretic hormone. Clin Pediatr (Phila). 1987;26:126130.
R, Poirier J, eds. Manual of Basic Neuropathology. Philadelphia: WB 210. Reefhuis J, Honein MA, Whitney CG, et al. Risk of bacterial meningitis in
Saunders; 1978:105120. children with cochlear implants. N Engl J Med. 2003;349:435445.
177. Walton J. The cerebrospinal fluid (CSF): anatomy and physiology. In: 211. Carpenter RR, Petersdorf RG. Risk of bacterial meningitis in children
Walton J, ed. Brains Disease of the Nervous System. Vol 1. 9th ed. with cochlear implants. Am J Med. 1962;33:262275.
Oxford: Oxford University Press; 1985:6471. 212. Stanek RJ, Mufson MA. A 20-year epidemiological study of pneumococcal
178. Scheld WM. Pathogenesis and pathophysiology of pneumococcal men- meningitis. Clin Infect Dis. 1999;28:12651272.
ingitis. In: Sande MA, Smith AL, Root RK, eds. Bacterial Meningitis. 213. Bruyn GA, Kremer HP, de Marie S, et al. Clinical evaluation of pneumo-
London: Churchill Livingstone; 1985:3769. coccal meningitis in adults over a twelve-year period. Eur J Clin Microbiol
179. Miller JD, Adams JH. The pathophysiology of raised intracranial pres- Infect Dis. 1989;8:695700.
sure. In: Adams JH, Corsellis JAN, Duchen LW, eds. Greenfields 214. Thomas KE, Hasbun R, Jekel J, et al. The diagnostic accuracy of Kernigs
Neuropathology. 4th ed. New York: Wiley Medical; 1984:5384. sign, Brudzinskis sign, and nuchal rigidity in adults with suspected
180. Dacey RG. Monitoring and treating increased intracranial pressure. meningitis. Clin Infect Dis. 2002;35:4652.
Pediatr Infect Dis J. 1987;6:11611163. 215. Geiseler PJ, Nelson KE, Levin S, et al. Community-acquired purulent
181. Sande MA, Scheld WM, McCracken GH. Summary of a workshop on the meningitis: a review of 1,316 cases during the antibiotic era, 19541976.
pathophysiology of bacterial meningitis: implications for new manage- Rev Infect Dis. 1980;2:725745.
ment strategies. Pediatr Infect Dis J. 1987;6:11671171. 216. van de Beek D, de Gans J, Spanjaard L, et al. Clinical features and
182. Ropper AH. Raised intracranial pressure in neurologic disease. Semin prognostic factors in adults with bacterial meningitis. N Engl J Med.
Neurol. 1984;4:397407. 2004;351:18491859.
183. Rorke LB, Pitts FW. Purulent meningitis: the pathologic basis of clinical 217. Schut ES, Lucas MJ, Brouwer MC, et al. Cerebral infarction in adults with
manifestations. Clin Pediatr. 1963;2:6471. bacterial meningitis. Neurocrit Care. 2012;16:421427.
184. Bell WE. Bacterial meningitis in children: selected aspects. Pediatr Clin 218. Kaufman BA, Tunkel AR, Pryor JC, et al. Meningitis in the neurosurgical
North Amer. 1992;39:651658. patient. Infect Dis Clin North Am. 1990;4:677701.

Scheld_Ch24.indd 414 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 415

219. Keroack MA. The patient with suspected meningitis. Emerg Med Clin 253. Guze BH, Baxter LR Jr. Current concepts. Neuroleptic malignant syn-
North Am. 1987;5:807826. drome. N Engl J Med. 1985;313:163166.
220. Spagnuolo PJ, Ellner JJ, Lerner PI, et al. Haemophilus influenzae meningitis: 254. Dougherty JM, Roth RM. Cerebral spinal fluid. Emerg Med Clin North
the spectrum of disease in adults. Medicine (Baltimore). 1982;61:7485. Am. 1986;4:281297.
221. Gorse GJ, Thrupp LD, Nudleman KL, et al. Bacterial meningitis in the 255. Corbett JJ, Mehta MP. Cerebrospinal fluid pressure in normal obese
elderly. Arch Intern Med. 1984;144:16031607. subjects and patients with pseudotumor cerebri. Neurology. 1983;33:
222. Behrman RE, Meyers BR, Mendelson MH, et al. Central nervous system 13861388.
infections in the elderly. Arch Intern Med. 1989;149:15961599. 256. Conly JM, Ronald AR. Cerebrospinal fluid as a diagnostic body fluid. Am
223. Lefrock JL, Smith BR. Gram-negative bacillary meningitis in adults. J Med. 1983;75:102108.
In: Vinken PJ, Bruyn GW, Klawans HL, eds. Handbook of Clinical 257. Marton KI, Gean AD. The spinal tap: a new look at an old test. Ann
Neurology. Amsterdam: Elsevier; 1988:103115. Intern Med. 1986;104:840848.
224. Cabellos C, Verdaguer R, Olmo M, et al. Community-acquired bacte- 258. Hayward RA, Oye RK. Are polymorphonuclear leukocytes an abnormal
rial meningitis in elderly patients: experience over 30 years. Medicine finding in cerebrospinal fluid? Results from 225 normal cerebrospinal
(Baltimore). 2009;88:115119. fluid specimens. Arch Intern Med. 1988;148:16231624.
225. Tunkel AR, Scheld WM. Acute infectious complications of head injury. 259. Schmidley JW, Simon RP. Postictal pleocytosis. Ann Neurol. 1981;9:
In: Braackman R, ed. Handbook of Clinical NeurologyHead Injury. 8184.
Amsterdam: Elsevier; 1990:317326. 260. Karandanis D, Shulman JA. Recent survey of infectious meningitis in
226. Kline MW. Review of recurrent bacterial meningitis. Pediatr Infect Dis J. adults: review of laboratory findings in bacterial, tuberculous, and aseptic
1989;8:630634. meningitis. South Med J. 1976;69:449457.
227. Berk SL, McCabe WR. Meningitis caused by gram-negative bacilli. Ann 261. Swartz MN, Dodge PR. Bacterial meningitisa review of selected aspects.
Intern Med. 1980;93:253260. I. General clinical features, special problems and unusual meningeal reac-
228. Bayston R, Lari J. A study of the sources of infection in colonised shunts. tions mimicking bacterial meningitis. N Engl J Med. 1965;272:898902.
Dev Med Child Neurol. 1974;16:1622. 262. Powers WJ. Cerebrospinal fluid lymphocytosis in acute bacterial meningi-
229. Schoenbaum SC, Gardner P, Shillito J. Infections of cerebrospinal fluid tis. Am J Med. 1985;79:216220.
shunts: epidemiology, clinical manifestations, and therapy. J Infect Dis. 263. Roos KL, Scheld WM. The management of fulminant meningitis in the
1975;131:543552. intensive care unit. Crit Care Clin. 1988;4:375392.
230. Forward KR, Fewer HD, Stiver HG. Cerebrospinal fluid shunt infections. 264. Lindquist L, Linne T, Hansson LO, et al. Value of cerebrospinal fluid anal-
A review of 35 infections in 32 patients. J Neurosurg. 1983;59:389394. ysis in the differential diagnosis of meningitis: a study in 710 patients with
231. Browne MJ, Dinndorf PA, Perek D, et al. Infectious complications suspected central nervous system infection. Eur J Clin Microbiol Infect
of intraventricular reservoirs in cancer patients. Pediatr Infect Dis J. Dis. 1988;7:374380.
1987;6:182189. 265. Lannigan R, MacDonald MA, Marrie TJ, et al. Evaluation of cerebrospi-
232. Tunkel AR, Scheld WM. Central nervous system infection in the compro- nal fluid lactic acid levels as an aid in differential diagnosis of bacterial
mised host. In: Rubin RH, Young LS, eds. Clinical Approach to Infection and viral meningitis in adults. J Clin Microbiol. 1980;11:324327.
in the Compromised Host. 3rd ed. New York: Plenum Press; 1994: 266. Kolmel HW, von Maravic M. Correlation of lactic acid level, cell count
163210. and cytology in cerebrospinal fluid of patients with bacterial and non-
233. Rubin RH, Hooper DC. Central nervous system infections in the compro- bacterial meningitis. Acta Neurol Scand. 1988;78:69.
mised host. Med Clin North Am. 1985;69:281293. 267. Genton B, Berger JP. Cerebrospinal fluid lactate in 78 cases of adult men-
234. Armstrong D, Wong B. Central nervous system infections in immunocom- ingitis. Intensive Care Med. 1990;16:196200.
promised hosts. Ann Rev Med. 1982;33:293308. 268. Huy NT, Thao NT, Diep DT, et al. Cerebrospinal fluid lactate concentra-
235. Hussein AS, Shafran SD. Acute bacterial meningitis in adults. A 12-year tion to distinguish bacterial from aseptic meningitis: a systemic review and
review. Medicine (Baltimore). 2000;79:360368. meta-analysis. Crit Care. 2010;14:R240.
236. John JF. Listeria monocytogenes. In: Vinken PJ, Bruyn GW, Klawans 269. Sakushima K, Hayashino Y, Kawaguchi T, et al. Diagnostic accuracy of
HL, eds. Handbook of Clinical Neurology. Amsterdam: Elsevier; 1988: cerebrospinal fluid lactate for differentiating bacterial meningitis from
89101. aseptic meningitis: a meta-analysis. J Infect. 2011;62:255262.
237. Hasbun R, Abrahams J, Jekel J, et al. Computed tomography of the head 270. Viallon A, Desseigne N, Marjollet O, et al. Meningitis in adult patients
before lumbar puncture in adults with suspected meningitis. N Engl J with a negative direct cerebrospinal fluid examination: value of cytochem-
Med. 2001;345:17271733. ical markers for differential diagnosis. Crit Care. 2011;15:R136.
238. Kaplan K. Brain abscess. Med Clin North Am. 1985;69:345360. 271. Spanos A, Harrell FE Jr, Durack DT. Differential diagnosis of acute men-
239. Wispelwey B, Scheld WM. Brain abscess. Clin Neuropharmacol. ingitis. An analysis of the predictive value of initial observations. JAMA.
1987;10:483510. 1989;262:27002707.
240. Hakan T, Ceran N, Erdem I, et al. Bacterial brain abscesses: an evaluation 272. Brouwer MC, Thwaites GE, Tunkel AR, et al. Dilemmas in the diagno-
of 96 cases. J Infect. 2006;52:359366. sis of acute community-acquired bacterial meningitis. Lancet. 2012;380:
241. Tonon E, Scotton PG, Gallucci M, et al. Brain abscess: clinical aspects of 16841692.
100 patients. Int J Infect Dis. 2006;10:103109. 273. Nigrovic LE, Malley R, Kuppermann N. Meta-analysis of bacterial men-
242. Carpenter J, Stapleton S, Holliman R. Retrospective analysis of 49 cases ingitis score validation studies. Arch Dis Child. 2012;97:799805.
of brain abscess and review of the literature. Eur J Clin Microbiol Infect 274. Kaplan SL, Smith EO, Wills C, et al. Association between preadmission
Dis. 2007;26:111. oral antibiotic therapy and cerebrospinal fluid findings and sequelae
243. Lee TH, Chang WN, Su TM, et al. Clinical features and predictive factors caused by Haemophilus influenzae type b meningitis. Pediatr Infect Dis.
of intraventricular rupture in patients who have bacterial brain abscesses. 1986;5:626632.
J Neurol Neurosurg Psychiatry. 2007;78:303309. 275. Blazer S, Berant M, Alon U. Bacterial meningitis. Effect of antibiotic treat-
244. Silverberg AL, DiNubile MJ. Subdural empyema and cranial epidural ment on cerebrospinal fluid. Am J Clin Pathol. 1983;80:386387.
abscess. Med Clin North Am. 1985;69:361374. 276. Gray LD, Fedorko DP. Laboratory diagnosis of bacterial meningitis. Clin
245. Hicks CW, Weber JG, Reid JR, et al. Identifying and managing intracranial Microbiol Rev. 1992;5:130145.
complications of sinusitis in children: a retrospective series. Pediatr Infect 277. Tarafdar K, Rao S, Recco RA, et al. Lack of sensitivity of the latex aggluti-
Dis J. 2011;30:222226. nation test to detect bacterial antigen in the cerebrospinal fluid of patients
246. DeMuri GP, Wald ER. Complications of acute bacterial sinusitis in children. with culture-negative meningitis. Clin Infect Dis. 2001;33:406408.
Pediatr Infect Dis J. 2011;30:701702. 278. Tunkel AR, Hartman BJ, Kaplan SL, et al. Practice guidelines for the man-
247. Morgan DW, Williams B. Posterior fossa subdural empyema. Brain. agement of bacterial meningitis. Clin Infect Dis. 2004;39:12671284.
1985;108(pt 4):983992. 279. Dwelle TL, Dunkle LM, Blair L. Correlation of cerebrospinal fluid endo-
248. Madhugiri VS, Sastri BV, Bhagavatula ID, et al. Posterior fossa subdural toxinlike activity with clinical and laboratory variables in gram-negative
empyema in childrenmanagement and outcome. Childs Nerv Syst. 2011; bacterial meningitis in children. J Clin Microbiol. 1987;25:856858.
27:137144. 280. Gray BM, Simmons DR, Mason H, et al. Quantitative levels of C-reactive
249. Barnes DW, Whitley RJ. CNS diseases associated with varicella zoster protein in cerebrospinal fluid in patients with bacterial meningitis and
virus and herpes simplex virus infection. Pathogenesis and current ther- other conditions. J Pediatr. 1986;108:665670.
apy. Neurol Clin. 1986;4:265283. 281. Abramson JS, Hampton KD, Babu S, et al. The use of C-reactive protein
250. Reik L. Lyme disease. In: Scheld WM, Whitley RJ, Durack DT, eds. from cerebrospinal fluid for differentiating meningitis from other central
Infections of the Central Nervous System. 2nd ed. New York: Lippincott- nervous system diseases. J Infect Dis. 1985;151:854858.
Raven; 1996:685718. 282. Sormunen P, Kallio MJ, Kilpi T, et al. C-reactive protein is useful in distin-
251. Hornick RB. The typhus group. In: Wyngaarden JB, Smith LH, eds. Cecil guishing Gram stain-negative bacterial meningitis from viral meningitis in
Textbook of Medicine. Philadelphia: WB Saunders; 1988:17381742. children. J Pediatr. 1999;134:725729.
252. Mohr JP, Kistler JP, Zabramski JM, et al. Intracranial aneurysms. In: 283. Shimetani N, Shimetani K, Mori M. Levels of three inflammation mark-
Barnett HJM, Mohr JP, Stein BM, et al, eds. Stroke: Pathophysiology, ers, C-reactive protein, serum amyloid A protein and procalcitonin, in the
Diagnosis, and Management. New York: Churchill Livingstone; 1986: serum and cerebrospinal fluid of patients with meningitis. Scand J Clin
643677. Lab Invest. 2001;61:567574.

Scheld_Ch24.indd 415 2/21/14 5:42 PM


416 Part III: Bacterial and Mycoplasmal Infections

284. Nathan BR, Scheld WM. The potential roles of C-reactive protein and 312. McNamara MT. Paramagnetic contrast media for magnetic resonance im-
procalcitonin concentrations in the serum and cerebrospinal fluid in the aging of the central nervous system. In: Brant-Zawadzki MB, Normal D,
diagnosis of bacterial meningitis. Curr Clin Top Infect Dis. 2002;22: eds. Magnetic Resonance Imaging of the Central Nervous System. New
155165. York: Raven Press; 1987:97105.
285. Viallon A, Zeni F, Lambert C, et al. High sensitivity and specificity of 313. Mathews VP, Kuharik MA, Edwards MK, et al. Dyke award. Gd-DTPA-
serum procalcitonin levels in adults with bacterial meningitis. Clin Infect enhanced MR imaging of experimental bacterial meningitis: evaluation
Dis. 1999;28:13131316. and comparison with CT. AJR Am J Roentgenol. 1989;152:131136.
286. Schwarz S, Bertram M, Schwab S, et al. Serum procalcitonin lev- 314. Sze G, Zimmerman RD. The magnetic resonance imaging of infections
els in bacterial and abacterial meningitis. Crit Care Med. 2000;28: and inflammatory diseases. Radiol Clin North Am. 1988;26:839859.
18281832. 315. Nickerson JP, Richner B, Santy K, et al. Neuroimaging of pediatric intracra-
287. Saha SK, Darmstadt GL, Yamanaka N, et al. Rapid diagnosis of pneu- nial infectionpart 1: techniques and bacterial infections. J Neuroimaging.
mococcal meningitis: implications for treatment and measuring disease 2012;22:e42e51.
burden. Pediatr Infect Dis J. 2005;24:10931098. 316. van de Beek D, Brouwer MC, Thwaites GE, et al. Advances in treatment
288. Werno AM, Murdoch DR. Medical microbiology: laboratory diagnosis of of bacterial meningitis. Lancet. 2012;380:16931702.
invasive pneumococcal disease. Clin Infect Dis. 2008;46:926932. 317. Aronin SI, Peduzzi P, Quagliarello VJ. Community-acquired bacterial
289. Moisi JC, Saha SK, Falade AG, et al. Enhanced diagnosis of pneumococ- meningitis: risk stratification for adverse clinical outcome and effect of
cal meningitis with use of the Binax NOW immunochromatographic test antibiotic timing. Ann Intern Med. 1998;129:862869.
of Streptococcus pneumoniae antigen: a multisite study. Clin Infect Dis. 318. Miner JR, Heegaard W, Mapes A, et al. Presentation, time to antibiotics,
2009;48(suppl 2):S49S56. and mortality of patients with bacterial meningitis at an urban county
290. Cherian T, Lalitha MK, Manoharan A, et al. PCR-enzyme immunoassay medical center. J Emerg Med. 2001;21:387392.
for detection of Streptococcus pneumoniae DNA in cerebrospinal fluid 319. Lu CH, Huang CR, Chang WN, et al. Community-acquired bacte-
samples from patients with culture-negative meningitis. J Clin Microbiol. rial meningitis in adults: the epidemiology, timing of appropriate an-
1998;36:36053608. timicrobial therapy, and prognostic factors. Clin Neurol Neurosurg.
291. Backman A, Lantz P, Radstrom P, et al. Evaluation of an extended diag- 2002;104:352358.
nostic PCR assay for detection and verification of the common causes of 320. Proulx N, Frechette D, Toye B, et al. Delays in the administration of anti-
bacterial meningitis in CSF and other biological samples. Mol Cell Probes. biotics are associated with mortality from adult acute bacterial meningitis.
1999;13:4960. QJM. 2005;98:291298.
292. Kristiansen BE, Ask E, Jenkins A, et al. Rapid diagnosis of meningococcal 321. Lepur D, Barsic B. Community-acquired bacterial meningitis in adults:
meningitis by polymerase chain reaction. Lancet. 1991;337:15681569. antibiotic timing in disease course and outcome. Infection. 2007;35:225231.
293. Ni H, Knight AI, Cartwright K, et al. Polymerase chain reaction for diag- 322. Coant PN, Kornberg AE, Duffy LC, et al. Blood culture results as determi-
nosis of meningococcal meningitis. Lancet. 1992;340:14321434. nants in the organism identification of bacterial meningitis. Pediatr Emerg
294. Jaton K, Sahli R, Bille J. Development of polymerase chain reaction as- Care. 1992;8:200205.
says for detection of Listeria monocytogenes in clinical cerebrospinal fluid 323. van de Beek D, de Gans J, Tunkel AR, et al. Community-acquired bacte-
samples. J Clin Microbiol. 1992;30:19311936. rial meningitis in adults. N Engl J Med. 2006;354:4453.
295. Radstrom P, Backman A, Qian N, et al. Detection of bacterial DNA in 324. Scheld WM. Theoretical and practical considerations of antibiotic therapy
cerebrospinal fluid by an assay for simultaneous detection of Neisseria for bacterial meningitis. Pediatr Infect Dis. 1985;4:7483.
meningitidis, Haemophilus influenzae, and streptococci using a semin- 325. Papastamelos AG, Tunkel AR. Antibacterial agents in infections of the cen-
ested PCR strategy. J Clin Microbiol. 1994;32:27382744. tral nervous system and eye. Infect Dis Clin North Am. 1995;9:615637.
296. Saravolatz LD, Manzor O, VanderVelde N, et al. Broad-range bacterial 326. Tunkel AR, Scheld WM. Therapy of bacterial meningitis: principles and
polymerase chain reaction for early detection of bacterial meningitis. Clin practice. Infect Control Hosp Epidemiol. 1989;10:565569.
Infect Dis. 2003;36:4045. 327. Sinner SW, Tunkel AR. Antimicrobial agents in the treatment of bacterial
297. Tzanakaki G, Tsopanomichalou M, Kesanopoulos K, et al. Simultaneous meningitis. Infect Dis Clin North Am. 2004;18:581602, ix.
single-tube PCR assay for the detection of Neisseria meningitidis, 328. Nau R, Sorgel F, Eiffert H. Penetration of drugs through the blood-
Haemophilus influenzae type b and Streptococcus pneumoniae. Clin cerebrospinal fluid/blood-brain barrier for treatment of central nervous
Microbiol Infect. 2005;11:386390. system infections. Clin Microbiol Rev. 2010;23:858883.
298. Wang X, Theodore MJ, Mair R, et al. Clinical validation of multiplex 329. Reed MD. Current concepts in clinical therapeutics: bacterial meningitis
real-time PCR assays for detection of bacterial meningitis pathogens. in infants and children. Clin Pharm. 1986;5:798809.
J Clin Microbiol. 2012;50:702708. 330. Barlow CF. Clinical aspects of the bloodbrain barrier. Ann Rev Med.
299. Welinder-Olsson C, Dotevall L, Hogevik H, et al. Comparison of 1964;15:187205.
broad-range bacterial PCR and culture of cerebrospinal fluid for diag- 331. Quagliarello VJ, Long WJ, Scheld WM. Morphologic alterations of the
nosis of community-acquired bacterial meningitis. Clin Microbiol Infect. blood-brain barrier with experimental meningitis in the rat. Temporal se-
2007;13:879886. quence and role of encapsulation. J Clin Invest. 1986;77:10841095.
300. Singhi SC, Mohankumar D, Singhi PD, et al. Evaluation of polymerase 332. Cherubin CE, Marr JS, Sierra MF, et al. Listeria and gram-negative bacil-
chain reaction (PCR) for diagnosing Haemophilus influenzae b meningi- lary meningitis in New York City, 19721979. Frequent causes of menin-
tis. Ann Trop Paediatr. 2002;22:347353. gitis in adults. Am J Med. 1981;71:199209.
301. Le Monnier A, Abachin E, Beretti JL, et al. Diagnosis of Listeria mono- 333. Pelkonen T, Roine I, Cruzeiro ML, et al. Slow initial beta-lactam infu-
cytogenes meningoencephalitis by real-time PCR for the hly gene. J Clin sion and oral paracetamol to treat childhood bacterial meningitis: a ran-
Microbiol. 2011;49:39173923. domised, controlled trial. Lancet Infect Dis. 2011;11:613621.
302. Kearns AM, Graham C, Burdess D, et al. Rapid real-time PCR for deter- 334. McCracken GH Jr, Nelson JD, Kaplan SL, et al. Consensus report: anti-
mination of penicillin susceptibility in pneumococcal meningitis, includ- microbial therapy for bacterial meningitis in infants and children. Pediatr
ing culture-negative cases. J Clin Microbiol. 2002;40:682684. Infect Dis J. 1987;6:501505.
303. Enzmann DR. Acute meningitis. In: Enzmann DR, ed. Imaging of 335. Van Esso D, Fontanals D, Uriz S, et al. Neisseria meningitidis strains with
Infections and Inflammations of the Central Nervous System: Computed decreased susceptibility to penicillin. Pediatr Infect Dis J. 1987;6:438439.
Tomography, Ultrasound, and Nuclear Magnetic Resonance. New York: 336. Cabellos C, Viladrich PF, Verdaguer R, et al. A single daily dose of ceftri-
Raven Press; 1984:188211. axone for bacterial meningitis in adults: experience with 84 patients and
304. Lin TY, Nelson JD, McCracken GH Jr. Fever during treatment for bacterial review of the literature. Clin Infect Dis. 1995;20:11641168.
meningitis. Pediatr Infect Dis J. 1984;3:319322. 337. Dillon JR, Pauze M, Yeung KH. Spread of penicillinase-producing and
305. Kline MW, Kaplan SL. Computed tomography in bacterial meningitis of transfer plasmids from the gonococcus to Neisseria meningitidis. Lancet.
childhood. Pediatr Infect Dis J. 1988;7:855857. 1983;1:779781.
306. Friedland IR, Paris MM, Rinderknecht S, et al. Cranial computed tomo- 338. Sutcliffe EM, Jones DM, el-Sheikh S, et al. Penicillin-insensitive meningo-
graphic scans have little impact on management of bacterial meningitis. cocci in the UK. Lancet. 1988;1:657658.
Am J Dis Child. 1992;146:14841487. 339. Woods CR, Smith AL, Wasilauskas BL, et al. Invasive disease caused by
307. Cabral DA, Flodmark O, Farrell K, et al. Prospective study of computed Neisseria meningitidis relatively resistant to penicillin in North Carolina.
tomography in acute bacterial meningitis. J Pediatr. 1987;111:201205. J Infect Dis. 1994;170:453456.
308. Benson P, Nyhan WL, Shimizu H. The prognosis of subdural effusions 340. Saez-Nieto JA, Lujan R, Berron S, et al. Epidemiology and molecular basis
complicating pyogenic meningitis. J Pediatr. 1960;58:670682. of penicillin-resistant Neisseria meningitidis in Spain: a 5-year history
309. Syrogiannopoulos GA, Nelson JD, McCracken GH Jr. Subdural collec- (19851989). Clin Infect Dis. 1992;14:394402.
tions of fluid in acute bacterial meningitis: a review of 136 cases. Pediatr 341. Jackson LA, Tenover FC, Baker C, et al. Prevalence of Neisseria men-
Infect Dis. 1986;5:343352. ingitidis relatively resistant to penicillin in the United States, 1991.
310. Goodman JM, Mealey J Jr. Postmeningitic subdural effusions: the syn- Meningococcal Disease Study Group. J Infect Dis. 1994;169:438441.
drome and its management. J Neurosurg. 1969;30:658663. 342. Brown EM, Fisman DN, Drews SJ, et al. Epidemiology of invasive
311. Britt RH, Enzmann DR, Yeager AS. Neuropathological and computer- meningococcal disease with decreased susceptibility to penicillin in
ized tomographic findings in experimental brain abscess. J Neurosurg. Ontario, Canada, 2000 to 2006. Antimicrob Agents Chemother. 2010;
1981;55:590603. 54:10161021.

Scheld_Ch24.indd 416 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 417

343. Perez Trallero E, Garcia Arenzana JM, Ayestaran I, et al. Comparative 369. Pfister M, Zhang L, Hammarlund-Udenaes M, et al. Modeling of trans-
activity in vitro of 16 antimicrobial agents against penicillin-susceptible me- fer kinetics at the serum-cerebrospinal fluid barrier in rabbits with ex-
ningococci and meningococci with diminished susceptibility to penicillin. perimental meningitis: application to grepafloxacin. Antimicrob Agents
Antimicrob Agents Chemother. 1989;33:16221623. Chemother. 2003;47:138143.
344. Centers for Disease Control and Prevention. Effects of new penicillin 370. Kuhn F, Cottagnoud M, Acosta F, et al. Cefotaxime acts synergistically
susceptibility breakpoints for Streptococcus pneumoniaeUnited States, with levofloxacin in experimental meningitis due to penicillin-resistant
20062007. MMWR Morb Mortal Wkly Rep. 2008;57:13531355. pneumococci and prevents selection of levofloxacin-resistant mutants in
345. National Committee for Clinical Laboratory Standards. Performance vitro. Antimicrob Agents Chemother. 2003;47:24872491.
Standards for Antimicrobial Susceptibility Testing. Tenth Informational 371. Tunkel AR, Scheld WM. Treatment of bacterial meningitis. Curr Infect
Supplement (Aerobic Dilution). Wayne, PA: National Committee for Dis Rep. 2002;4:716.
Clinical Laboratory Standards; 2000. 372. Cottagnoud P, Tauber MG. Fluoroquinolones in the treatment of meningitis.
346. Kaplan SL, Mason EO Jr. Mechanisms of pneumococcal antibiotic resis- Curr Infect Dis Rep. 2003;5:329336.
tance and treatment of pneumococcal infections in 2002. Pediatr Ann. 373. Saez-Llorens X, ORyan M. Cefepime in the empiric treatment of menin-
2002;31:250260. gitis in children. Pediatr Infect Dis J. 2001;20:356361.
347. Jacobs MR. Drug-resistant Streptococcus pneumoniae: rational antibiotic 374. Landesman SH, Corrado ML, Shah PM, et al. Past and current roles for
choices. Am J Med. 1999;106:19S25S; discussion 48S52S. cephalosporin antibiotics in treatment of meningitis. Emphasis on use in
348. Barroso DE, Godoy D, Castineiras TM, et al. Beta-lactam resistance, se- gram-negative bacillary meningitis. Am J Med. 1981;71:693703.
rotype distribution, and genotypes of meningitis-causing Streptococcus 375. Cherubin CE, Eng RH, Norrby R, et al. Penetration of newer cephalospo-
pneumoniae, Rio de Janeiro, Brazil. Pediatr Infect Dis J. 2012;31:3036. rins into cerebrospinal fluid. Rev Infect Dis. 1989;11:526548.
349. Appelbaum PC. Resistance among Streptococcus pneumoniae: implica- 376. ONeill E, Humphreys H, Phillips J, et al. Third-generation cephalosporin
tions for drug selection. Clin Infect Dis. 2002;34:16131620. resistance among Gram-negative bacilli causing meningitis in neurosurgi-
350. Meyer CN, Samuelsson IS, Galle M, et al. Adult bacterial meningitis: cal patients: significant challenges in ensuring effective antibiotic therapy.
aetiology, penicillin susceptibility, risk factors, prognostic factors and J Antimicrob Chemother. 2006;57:356359.
guidelines for empirical antibiotic treatment. Clin Microbiol Infect. 377. Norrby SR. Role of cephalosporins in the treatment of bacterial meningi-
2004;10:709717. tis in adults. Overview with special emphasis on ceftazidime. Am J Med.
351. Bradley JS, Kaplan SL, Klugman KP, et al. Consensus: management of 1985;79:5661.
infections in children caused by Streptococcus pneumoniae with decreased 378. Segev S, Barzilai A, Rosen N, et al. Pefloxacin treatment of menin-
susceptibility to penicillin. Pediatr Infect Dis J. 1995;14:10371041. gitis caused by gram-negative bacteria. Arch Intern Med. 1989;149:
352. Tunkel AR, Scheld WM. Acute bacterial meningitis. Lancet. 1995;346: 13141316.
16751680. 379. Tunkel AR, Scheld WM. Treatment of bacterial meningitis. In: Wolfson
353. Giron KP, Gross ME, Musher DM, et al. In vitro antimicrobial effect against JS, Hooper DC, eds. Quinolone Antimicrobial Agents. Washington, DC:
Streptococcus pneumoniae of adding rifampin to penicillin, ceftriaxone, or American Society for Microbiology; 1993:381395.
1-ofloxacin. Antimicrob Agents Chemother. 1995;39:27982800. 380. Klugman KP, Dagan R. Carbapenem treatment of meningitis. Scand J
354. Gerber J, Pohl K, Sander V, et al. Rifampin followed by ceftriaxone for Infect Dis Suppl. 1995;96:4548.
experimental meningitis decreases lipoteichoic acid concentrations in ce- 381. Kim BN, Peleg AY, Lodise TP, et al. Management of meningitis due to
rebrospinal fluid and reduces neuronal damage in comparison to ceftriax- antibiotic-resistant Acinetobacter species. Lancet Infect Dis. 2009;9:
one alone. Antimicrob Agents Chemother. 2003;47:13131317. 245255.
355. Ricard JD, Wolff M, Lacherade JC, et al. Levels of vancomycin in cerebro- 382. van de Beek D, Drake JM, Tunkel AR. Nosocomial bacterial meningitis.
spinal fluid of adult patients receiving adjunctive corticosteroids to treat N Engl J Med. 2010;362:146154.
pneumococcal meningitis: a prospective multicenter observational study. 383. Jimenez-Mejias ME, Pichardo-Guerrero C, Marquez-Rivas FJ, et al.
Clin Infect Dis. 2007;44:250255. Cerebrospinal fluid penetration and pharmacokinetic/pharmacodynamic
356. Rybak M, Lomaestro B, Rotschafer JC, et al. Therapeutic monitoring of parameters of intravenously administered colistin in a case of multidrug-
vancomycin in adult patients: a consensus review of the American Society resistant Acinetobacter baumannii meningitis. Eur J Clin Microbiol Infect
of Health-System Pharmacists, the Infectious Diseases Society of America, Dis. 2002;21:212214.
and the Society of Infectious Diseases Pharmacists. Am J Health Syst 384. Vasan W, Desmery P, Ilutovich S, et al. Intrathecal use of colistin. J Clin
Pharm. 2009;66:8298. Microbiol. 2000;38:3523.
357. McCullers JA, English BK, Novak R. Isolation and characterization of 385. Cascio A, Conti A, Sinardi L, et al. Post-neurosurgical multidrug-resistant
vancomycin-tolerant Streptococcus pneumoniae from the cerebrospinal Acinetobacter baumannii meningitis successfully treated with intrathecal
fluid of a patient who developed recrudescent meningitis. J Infect Dis. colistin. A new case and a systematic review of the literature. Int J Infect
2000;181:369373. Dis. 2010;14:e572e579.
358. Novak R, Henriques B, Charpentier E, et al. Emergence of vancomycin 386. Benifla M, Zucker G, Cohen A, et al. Successful treatment of Acinetobacter
tolerance in Streptococcus pneumoniae. Nature. 1999;399:590593. meningitis with intrathecal polymyxin E. J Antimicrob Chemother.
359. Liu HH, Tomasz A. Penicillin tolerance in multiply drug-resistant natural 2004;54:290292.
isolates of Streptococcus pneumoniae. J Infect Dis. 1985;152:365372. 387. Katragkou A, Roilides E. Successful treatment of multidrug-resistant
360. Bradley JS, Scheld WM. The challenge of penicillin-resistant Streptococcus Acinetobacter baumannii central nervous system infections with colistin.
pneumoniae meningitis: current antibiotic therapy in the 1990s. Clin J Clin Microbiol. 2005;43:49164917.
Infect Dis. 1997;24(suppl 2):S213S221. 388. Antachopoulos C, Karvanen M, Iosifidis E, et al. Serum and cerebrospinal
361. Buckingham SC, Davis Y, English BK. Pneumococcal susceptibility to fluid levels of colistin in pediatric patients. Antimicrob Agents Chemother.
meropenem in a mid-south childrens hospital. South Med J. 2002;95: 2010;54:39853987.
12931296. 389. Tutuncu EE, Kuscu F, Gurbuz Y, et al. Tigecycline use in two cases with
362. Friedland IR, Klugman KP. Failure of chloramphenicol therapy in penicil- multidrug-resistant Acinetobacter baumannii meningitis. Int J Infect Dis.
lin-resistant pneumococcal meningitis. Lancet. 1992;339:405408. 2010;14(suppl 3):e224e226.
363. Cornick JE, Everett DB, Broughton C, et al. Invasive Streptococcus 390. McCracken GH Jr. Current management of bacterial meningitis in infants
pneumoniae in children, Malawi, 20042006. Emerg Infect Dis. 2011; and children. Pediatr Infect Dis J. 1992;11:169174.
17:11071109. 391. Schaad UB, Suter S, Gianella-Borradori A, et al. A comparison of ceftriax-
364. Manning L, Laman M, Greenhill AR, et al. Increasing chloramphenicol one and cefuroxime for the treatment of bacterial meningitis in children.
resistance in Streptococcus pneumoniae isolates from Papua New Guinean N Engl J Med. 1990;322:141147.
children with acute bacterial meningitis. Antimicrob Agents Chemother. 392. Lebel MH, Hoyt MJ, McCracken GH Jr. Comparative efficacy of ceftri-
2011;55:44544456. axone and cefuroxime for treatment of bacterial meningitis. J Pediatr.
365. Cottagnoud P, Acosta F, Cottagnoud M, et al. Efficacies of BMS 284756 1989;114:10491054.
against penicillin-sensitive, penicillin-resistant, and quinolone-resistant 393. Jorgensen JH. Update on mechanisms and prevalence of antimicrobial re-
pneumococci in experimental meningitis. Antimicrob Agents Chemother. sistance in Haemophilus influenzae. Clin Infect Dis. 1992;14:11191123.
2002;46:184187. 394. Campos J, Garcia-Tornel S, Gairi JM, et al. Multiply resistant
366. Cottagnoud P, Acosta F, Cottagnoud M, et al. Gemifloxacin is efficacious Haemophilus influenzae type b causing meningitis: comparative clinical
against penicillin-resistant and quinolone-resistant pneumococci in exper- and laboratory study. J Pediatr. 1986;108:897902.
imental meningitis. Antimicrob Agents Chemother. 2002;46:16071609. 395. Campos J, Chanyangam M, deGroot R, et al. Genetic relatedness of anti-
367. Takahata M, Yamada H, Morita T, et al. Evaluation of T-3811ME (BMS- biotic resistance determinants in multiply resistant Hemophilus influenzae.
284756), a new des-F(6)-quinolone, for treatment of meningitis caused J Infect Dis. 1989;160:810817.
by penicillin-resistant Streptococcus pneumoniae in rabbits. Antimicrob 396. Kaplan SL. Haemophilus influenzae meningitis. In: Vinken PJ, Bruyn GW,
Agents Chemother. 2002;46:17601765. Klawans HL, eds. Handbook of Clinical Neurology. Amsterdam: Elsevier;
368. Rodriguez-Cerrato V, McCoig CC, Saavedra J, et al. Garenoxacin 1988:5970.
(BMS-284756) and moxifloxacin in experimental meningitis caused 397. Krasinski K, Kusmiesz H, Nelson JD. Pharmacologic interactions among
by vancomycin-tolerant pneumococci. Antimicrob Agents Chemother. chloramphenicol, phenytoin and phenobarbital. Pediatr Infect Dis.
2003;47:211215. 1982;1:232235.

Scheld_Ch24.indd 417 2/21/14 5:42 PM


418 Part III: Bacterial and Mycoplasmal Infections

398. Saez-Llorens X, McCoig C, Feris JM, et al. Quinolone treatment for pedi- 428. Drapkin MS, Wisch JS, Gelfand JA, et al. Plasmapheresis for fulminant
atric bacterial meningitis: a comparative study of trovafloxacin and ceftri- meningococcemia. Pediatr Infect Dis J. 1989;8:399400.
axone with or without vancomycin. Pediatr Infect Dis J. 2002;21:1422. 429. Girgis NI, Farid Z, Mikhail IA, et al. Dexamethasone treatment for
399. Lerner PI, Gopalakrishna KV, Wolinsky E, et al. Group B Streptococcus bacterial meningitis in children and adults. Pediatr Infect Dis J. 1989;8:
(S. agalactiae) bacteremia in adults: analysis of 32 cases and review of the 848851.
literature. Medicine (Baltimore). 1977;56:457473. 430. Lebel MH, Freij BJ, Syrogiannopoulos GA, et al. Dexamethasone therapy
400. Gunther G, Philipson A. Oral trimethoprim as follow-up treat- for bacterial meningitis. Results of two double-blind, placebo-controlled
ment of meningitis caused by Listeria monocytogenes. Rev Infect Dis. trials. N Engl J Med. 1988;319:964971.
1988;10:5355. 431. Lebel MH, Hoyt MJ, Waagner DC, et al. Magnetic resonance imaging
401. Richards SJ, Lambert CM, Scott AC. Recurrent Listeria monocyto- and dexamethasone therapy for bacterial meningitis. Am J Dis Child.
genes meningitis treated with intraventricular vancomycin. J Antimicrob 1989;143:301306.
Chemother. 1992;29:351353. 432. McCracken GH Jr, Lebel MH. Dexamethasone therapy for bacterial men-
402. Roos KL, Scheld WM. Staphylococcal infections of the central nervous ingitis in infants and children. Am J Dis Child. 1989;143:287289.
system. In: Archer GL, Crossley K, eds. Staphylococci and Staphylococcal 433. Kaplan SL. Dexamethasone for children with bacterial meningitis. Should
Diseases. New York: Churchill Livingstone; 2013. it be routine therapy? Am J Dis Child. 1989;143:290292.
403. Everett ED, Strausbaugh LJ. Antimicrobial agents and the central nervous 434. McIntyre PB, Berkey CS, King SM, et al. Dexamethasone as adjunctive
system. Neurosurgery. 1980;6:691714. therapy in bacterial meningitis. A meta-analysis of randomized clinical
404. Gump DW. Vancomycin for treatment of bacterial meningitis. Rev Infect trials since 1988. JAMA. 1997;278:925931.
Dis. 1981;3(suppl):S289S292. 435. Odio CM, Faingezicht I, Paris M, et al. The beneficial effects of early
405. Higa T, Tasaka T, Kubo Y, et al. Successful treatment of meningoencepha- dexamethasone administration in infants and children with bacterial men-
litis caused by methicillin-resistant Staphylococcus aureus with intrave- ingitis. N Engl J Med. 1991;324:15251531.
nous linezolid in an allogeneic cord blood stem cell transplant recipient. 436. Kennedy WA, Hoyt MJ, McCracken GH Jr. The role of corticosteroid
Scand J Infect Dis. 2008;40:990992. therapy in children with pneumococcal meningitis. Am J Dis Child.
406. Sipahi OR, Bardak S, Turhan T, et al. Linezolid in the treatment of meth- 1991;145:13741378.
icillin-resistant staphylococcal post-neurosurgical meningitis: a series of 437. Schaad UB, Lips U, Gnehm HE, et al. Dexamethasone therapy for bac-
17 cases. Scand J Infect Dis. 2011;43:757764. terial meningitis in children. Swiss Meningitis Study Group. Lancet.
407. Bardak-Ozcem S, Turhan T, Sipahi OR, et al. Daptomycin versus vanco- 1993;342:457461.
mycin in treatment of methicillin-resistant Staphylococcus aureus men- 438. King SM, Law B, Langley JM, et al. Dexamethasone therapy for bacterial
ingitis in an experimental rabbit model. Antimicrob Agents Chemother. meningitis: better never than late? Can J Infect Dis. 1994;5:210215.
2013;57:15561558. 439. Wald ER, Kaplan SL, Mason EO Jr, et al. Dexamethasone therapy for
408. Lee DH, Palermo B, Chowdhury M. Successful treatment of methicillin- children with bacterial meningitis. Meningitis Study Group. Pediatrics.
resistant staphylococcus aureus meningitis with daptomycin. Clin Infect 1995;95:2128.
Dis. 2008;47:588590. 440. Molyneux EM, Walsh AL, Forsyth H, et al. Dexamethasone treatment in
409. Wallace MR, Sander AW, Licitra C, et al. Methicillin-resistant childhood bacterial meningitis in Malawi: a randomised controlled trial.
Staphylococcus aureus meningitis with daptomycin. Infect Dis Clin Pract. Lancet. 2002;360:211218.
2009;17:6970. 441. Kaplan SL, Fishman MA. Supportive therapy for bacterial meningitis.
410. Kelesidis T, Humphries R, Ward K, et al. Combination therapy with dap- Pediatr Infect Dis J. 1987;6:670677.
tomycin, linezolid, and rifampin as treatment option for MRSA meningi- 442. Syrogiannopoulos GA, Lourida AN, Theodoridou MC, et al.
tis and bacteremia. Diagn Microbiol Infect Dis. 2011;71:286290. Dexamethasone therapy for bacterial meningitis in children: 2- versus
411. McCabe WR. Empiric therapy for bacterial meningitis. Rev Infect Dis. 4-day regimen. J Infect Dis. 1994;169:853858.
1983;5(suppl 1):S74S83. 443. Arditi M, Ables L, Yogev R. Cerebrospinal fluid endotoxin levels in chil-
412. Saez-Llorens X, Castano E, Garcia R, et al. Prospective randomized dren with H. influenzae meningitis before and after administration of in-
comparison of cefepime and cefotaxime for treatment of bacterial men- travenous ceftriaxone. J Infect Dis. 1989;160:10051011.
ingitis in infants and children. Antimicrob Agents Chemother. 1995;39: 444. de Gans J, van de Beek D; European Dexamethasone in Adulthood
937940. Bacterial Meningitis Study Investigators. Dexamethasone in adults with
413. Radetsky M. Duration of treatment in bacterial meningitis: a historical bacterial meningitis. N Engl J Med. 2002;347:15491556.
inquiry. Pediatr Infect Dis J. 1990;9:29. 445. Viladrich PF, Gudiol F, Linares J, et al. Evaluation of vancomycin for ther-
414. Molyneux E, Nizami SQ, Saha S, et al. 5 versus 10 days of treatment apy of adult pneumococcal meningitis. Antimicrob Agents Chemother.
with ceftriaxone for bacterial meningitis in children: a double-blind ran- 1991;35:24672472.
domised equivalence study. Lancet. 2011;377:18371845. 446. Klugman KP, Friedland IR, Bradley JS. Bactericidal activity against ceph-
415. Snape MD, Kelly DF. Fine with five? Shorter antibiotic courses for child- alosporin-resistant Streptococcus pneumoniae in cerebrospinal fluid of
hood meningitis. Lancet. 2011;377:18091810. children with acute bacterial meningitis. Antimicrob Agents Chemother.
416. Gardner P, Leipzig T, Phillips P. Infections of central nervous system 1995;39:19881992.
shunts. Med Clin North Am. 1985;69:297314. 447. Waagner DC, Kennedy WA, Hoyt MJ, et al. Lack of adverse effects
417. Edwards MS, Baker CJ, Butler KM, et al. Penetration of cefuroxime of dexamethasone therapy in aseptic meningitis. Pediatr Infect Dis J.
into ventricular fluid in cerebrospinal fluid shunt infections. Antimicrob 1990;9:922923.
Agents Chemother. 1989;33:11081110. 448. Tunkel AR, Scheld WM. Corticosteroids for everyone with meningitis?
418. Tunkel AR. Bacterial meningitis: non-antibiotic modes of therapy. Curr N Engl J Med. 2002;347:16131615.
Opin Infect Dis. 1993;6:638643. 449. Heckenberg SG, Brouwer MC, van der Ende A, et al. Adjunctive
419. Saez-Llorens X, Ramilo O, Mustafa MM, et al. Molecular pathophysiol- dexamethasone in adults with meningococcal meningitis. Neurology.
ogy of bacterial meningitis: current concepts and therapeutic implications. 2012;79:15631569.
J Pediatr. 1990;116:671684. 450. Weisfelt M, Hoogman M, van de Beek D, et al. Dexamethasone and
420. Damas P, Reuter A, Gysen P, et al. Tumor necrosis factor and interleukin-1 long-term outcome in adults with bacterial meningitis. Ann Neurol.
serum levels during severe sepsis in humans. Crit Care Med. 1989;17: 2006;60:456468.
975978. 451. Brouwer MC, Heckenberg SG, de Gans J, et al. Nationwide implementa-
421. Waage A, Halstensen A, Espevik T. Association between tumour necrosis tion of adjunctive dexamethasone therapy for pneumococcal meningitis.
factor in serum and fatal outcome in patients with meningococcal disease. Neurology. 2010;75:15331539.
Lancet. 1987;1:355357. 452. Nguyen TH, Tran TH, Thwaites G, et al. Dexamethasone in Vietnamese
422. Ming WJ, Bersani L, Mantovani A. Tumor necrosis factor is chemo- adolescents and adults with bacterial meningitis. N Engl J Med.
tactic for monocytes and polymorphonuclear leukocytes. J Immunol. 2007;357:24312440.
1987;138:14691474. 453. Scarborough M, Gordon SB, Whitty CJ, et al. Corticosteroids for
423. Tracey KJ, Lowry SF, Cerami A. Cachectin: a hormone that triggers acute bacterial meningitis in adults in sub-Saharan Africa. N Engl J Med.
shock and chronic cachexia. J Infect Dis. 1988;157:413420. 2007;357:24412450.
424. Dinarello CA. An update on human interleukin-1: from molecular biology 454. Greenwood BM. Corticosteroids for acute bacterial meningitis. N Engl J
to clinical relevance. J Clin Immunol. 1985;5:287297. Med. 2007;357:25072509.
425. Beutler B, Milsark IW, Cerami AC. Passive immunization against cachectin/ 455. Brouwer MC, McIntyre P, Prasad K, et al. Corticosteroids for acute bacte-
tumor necrosis factor protects mice from lethal effect of endotoxin. rial meningitis. Cochrane Database Syst Rev. 2013;6:CD004405.
Science. 1985;229:869871. 456. Gaillard JL, Abadie V, Cheron G, et al. Concentrations of ceftriaxone in
426. Gigliotti F, Lee D, Insel RA, et al. IgG penetration into the cerebrospinal cerebrospinal fluid of children with meningitis receiving dexamethasone
fluid in a rabbit model of meningitis. J Infect Dis. 1987;156:394398. therapy. Antimicrob Agents Chemother. 1994;38:12091210.
427. Bernard GR, Vincent JL, Laterre PF, et al. Efficacy and safety of recom- 457. Buke AC, Cavusoglu C, Karasulu E, et al. Does dexamethasone affect cef-
binant human activated protein C for severe sepsis. N Engl J Med. 2001; triaxone [corrected] penetration into cerebrospinal fluid in adult bacterial
344:699709. meningitis. Int J Antimicrob Agents. 2003;21:452456.

Scheld_Ch24.indd 418 2/21/14 5:42 PM


Chapter 24: Acute Bacterial Meningitis 419

458. Cabellos C, Martinez-Lacasa J, Tubau F, et al. Evaluation of com- 468. Meldrum BS, Vigouroux RA, Brierley JB. Systemic factors and epileptic brain
bined ceftriaxone and dexamethasone therapy in experimental cepha- damage. Prolonged seizures in paralyzed, artificially ventilated baboons.
losporin-resistant pneumococcal meningitis. J Antimicrob Chemother. Arch Neurol. 1973;29:8287.
2000;45:315320. 469. Zoons E, Weisfelt M, de Gans J, et al. Seizures in adults with bacterial
459. Tuomanen E. Partner drugs: a new outlook for bacterial meningitis. Ann meningitis. Neurology. 2008;70:21092115.
Intern Med. 1988;109:690692. 470. Leppik I. Status epilepticus. Paper presented at: the American Academy of
460. Woehrl B, Brouwer MC, Murr C, et al. Complement component 5 con- Neurology Meeting; April 1983; San Diego, CA.
tributes to poor disease outcome in humans and mice with pneumococcal 471. Dreifuss FE. Status epilepticus. In: Dreifuss FE, ed. Pediatric Epileptology:
meningitis. J Clin Invest. 2011;121:39433953. Classification and Management of Seizures in the Child. Boston: John
461. Peterson PK, Hu S, Sheng WS, et al. Thalidomide inhibits tumor necrosis Wright; 1983:221230.
factor-alpha production by lipopolysaccharide- and lipoarabinomannan- 472. Lowenstein DH, Alldredge BK. Status epilepticus. N Engl J Med.
stimulated human microglial cells. J Infect Dis. 1995;172:11371140. 1998;338:970976.
462. Lindvall P, Ahlm C, Ericsson M, et al. Reducing intracranial pressure may 473. Treiman DM, Delgado-Escueta AV. Status epilepticus. In: Thompson
increase survival among patients with bacterial meningitis. Clin Infect RA, Green JR, eds. Critical Care of Neurological and Neurosurgical
Dis. 2004;38:384390. Emergencies. New York: Raven Press; 1980:5399.
463. Marshall LF, Marshall SB. Medical management of intracranial pressure. 474. Dunn DW. Status epilepticus. In: Dunn DW, Epstein LG, eds. Decision
In: Cooper PR, ed. Head Injury. 2nd ed. Baltimore: Lippincott Williams Making in Child Neurology. Toronto: BC Decker; 1987:124.
& Wilkins; 1987:177196. 475. Kaplan SL, Mason EO Jr, Mason SK, et al. Prospective comparative
464. Tauber MG, Khayam-Bashi H, Sande MA. Effects of ampicillin and cor- trial of moxalactam versus ampicillin or chloramphenicol for treat-
ticosteroids on brain water content, cerebrospinal fluid pressure, and ment of Haemophilus influenzae type b meningitis in children. J Pediatr.
cerebrospinal fluid lactate levels in experimental pneumococcal meningi- 1984;104:447453.
tis. J Infect Dis. 1985;151:528534. 476. Jim KK, Brouwer MC, van der Ende A, et al. Subdural empyema in bacterial
465. Syrogiannopoulos GA, Olsen KD, Reisch JS, et al. Dexamethasone in meningitis. Neurology. 2012;79:21332139.
the treatment of experimental Haemophilus influenzae type b meningitis. 477. Sigurdardottir B, Bjornsson OM, Jonsdottir KE, et al. Acute bacterial men-
J Infect Dis. 1987;155:213219. ingitis in adults. A 20-year overview. Arch Intern Med. 1997;157:425430.
466. Fishman RA. Brain edema and disorders of intracranial pressure. In: 478. McMillan DA, Lin CY, Aronin SI, et al. Community-acquired bacterial
Rowland LP, ed. Merritts Textbook of Neurology. 8th ed. Philadelphia: meningitis in adults: categorization of causes and timing of death. Clin
Lea & Febiger; 1989:262266. Infect Dis. 2001;33:969975.
467. Scheld WM, Dacey RG, Winn HR, et al. Cerebrospinal fluid outflow resis- 479. Tauber MG, Burroughs M, Niemoller UM, et al. Differences of pathophys-
tance in rabbits with experimental meningitis. Alterations with penicillin iology in experimental meningitis caused by three strains of Streptococcus
and methylprednisolone. J Clin Invest. 1980;66:243253. pneumoniae. J Infect Dis. 1991;163:806811.

Scheld_Ch24.indd 419 2/21/14 5:42 PM


CHAPTER 25 MYCOPLASMAL AND
UREAPLASMAL INFECTIONS
ARI BITNUN AND SUSAN RICHARDSON

Mycoplasmas and ureaplasmas (class Mollicutes) are the them wide notoriety for contamination of continuous cell
smallest known bacteria (1,2). They are adapted to life in lines. The small genome size also translates into less syn-
humans, animals, insects, and plants and cannot survive free thetic capacity and the need for specially enriched media for
living in nature due to their dependence on host cells for laboratory cultivation (6).
nutrients. Of the more than 200 known species, about 17 have This chapter covers CNS diseases attributable to M. pneu-
been associated with human mucous membrane colonization. moniae (Mp), M. hominis (Mh), and Ureaplasma species (Uu).
ThreeMycoplasma pneumoniae, Mycoplasma hominis, and Because the presence of infection or disease in sites other than
Ureaplasma urealyticum/Ureaplasma parvumaccount for the nevous system can be helpful in diagnosis, these are discussed
most reports of central nervous system (CNS) disease in as well. In the section on Mp, information related to Guillain-
humans. Rare cases of CNS disease due to other species have Barr syndrome is included due to overlapping pathogenetic
been reported, most often in association with immunocom- mechanisms. The section on diagnosis includes some basic in-
promising or other predisposing conditions (35). formation on laboratory detection and collection of specimens
Mycoplasmas and ureaplasmas have a cell volume less as a resource for interacting with the clinical laboratory.
than 5% that of a typical bacillus such as Escherichia coli
and one-sixth the number of genes (about 800,000 base pairs
for M. pneumoniae vs. 4.6 million base pairs for E. coli)
(1,6). It is thought that they evolved from gram-positive
MYCOPLASMA PNEUMONIAE
eubacteria by a process of degenerative evolution typified
by gradual reduction in genome size (6,7). Due to the lack Etiology
of a cell wall, they are pleomorphic in shape, are highly sus-
ceptible to adverse environmental conditions, and are resis- Mp is primarily an extracellular pathogen of the respiratory
tant to -lactam and glycopeptide antibiotics. Because they tract. It was first isolated in culture from the sputum of a patient
do not synthesize folic acid, they are also resistant to the with atypical pneumonia in 1944 (8). Initially referred to as
sulfonamides. Their small size allows them to pass through the Eaton agent, it was classified as a pleuropneumonia-like
0.45-m pore filters commonly used to filter-sterilize media. organism in 1961 and received its current taxonomic name in
This trait, coupled with challenges in detection, has earned 1963 (9). Mp is filamentous in structure (Fig. 25.1), measuring

FIGURE 25.1 Scanning electron microscopy of Mycoplasma pneumoniae cells. There are large aggre-
gates of cells shown; the isolated spindle and filamentous shapes in between are individual cells with
typical morphology. (Courtesy J. Carr, Centers for Disease Control and Prevention.)

420

Scheld_Ch25.indd 420 2/21/14 5:43 PM


Chapter 25: Mycoplasmal and Ureaplasmal Infections 421

1 to 2 m in diameter and 0.1 to 0.2 m in width. It attaches


to human epithelial cells primarily by means of a highly spe- Pathogenesis of Neurologic Disease
cialized organelle that contains a network of adhesion proteins
The pathogenesis of Mp-associated neurologic disease is
including the P1 and P30 adhesins (1,6,10). Gliding motility
incompletely understood. Three broad mechanisms have been
plays an important role in its ability to colonize fully differenti-
proposed with varying levels of supportive evidence (30,31):
ated mucus-producing tissues of the respiratory tract (11,12).
(a) direct invasion of the brain parenchyma; (b) autoimmu-
It is capable of invading and surviving within cells in vitro
nity, or other immune-mediated processes; and (c) vascular
(13,14), but whether this occurs to a meaningful extent in vivo
occlusion. Neurotoxin-mediated CNS disease, as seen with
is not known. Human-to-human transmission requires close
Mycoplasma gallisepticum in turkeys and Mycoplasma neuro-
contact and occurs by the droplet contact route.
lyticum in mice (32,33), has not been demonstrated in humans
with Mp-associated neurologic disease.
Several lines of evidence support the concept of direct infec-
Epidemiology tion of the brain by Mp. Mp has, albeit rarely, been cultured
from or detected by various indirect microbiologic techniques
Respiratory tract infection due to Mp is extremely common; at autopsy from the brain of several fatal cases of suspected
the incidence of pneumonia is highest in school-aged children Mp neurologic disease (Fig. 25.2) (3437). It has also been
(15,16). The proportion of pneumonia cases attributable to cultured from or detected in the cerebrospinal fluid (CSF) by
Mp increases from about 20% in children between 10 and polymerase chain reaction (PCR) of over 50 subjects, 80%
16 years of age to about 50% in young adults (1719). Many of whom had meningitis, meningoencephalitis, or encepha-
infections, however, are subclinical, particularly in those litis (28,3853). A cerebrovascular vasculopathy character-
younger than 5 years of age (20,21). There is no particular ized by antimycoplasmal immunostaining of endothelial cells
seasonal distribution. Epidemics occur every 3 to 7 years su- and electron microscopic demonstration of Mycoplasma-like
perimposed on low level but constant endemicity (17). structures within endothelial cells has been observed (54,55).
The incidence of neurologic disease attributable to Mp Finally, by analogy to other species, there is conclusive evi-
has been estimated at 0.1% or less (22). Among hospitalized dence from both natural and experimental infection of rodents,
patients with serologically confirmed Mp infection, the inci- turkeys, and alligators by Mycoplasma pulmonis, M. gallisep-
dence is higher, between 1% and 10% (2325). Neurologic ticum, and Mycoplasma alligatoris, respectively, for the direct
complications are most often sporadic, but clusters of cases infection hypothesis (5658).
have been observed during epidemics of respiratory tract Autoimmunity due to antigenic mimicry and production
disease (26,27). Children account for 50% to 70% of those of antineuronal antibodies has been implicated in the patho-
diagnosed with neurologic complications of Mp (23,28,29). genesis of Mp-associated neurologic syndromes, including
Furthermore, in Europe and North America, Mp is a lead- postinfectious encephalitis, acute disseminated encephalomy-
ing cause of acute encephalitis in children, responsible for elitis (ADEM), transverse myelitis, and Guillain-Barr syn-
5% to 13% of all cases (23,28,29). There is no apparent sex drome (30,31). Glycolipid epitopes of Mp cross-react with
predilection (29). several gangliosides, and anti-galactocerebroside antibodies

FIGURE 25.2 Histopathology from fatal cases of Mycoplasma pneumoniaeassociated polyradiculo-


neuropathy in previously healthy 45-year-old man. M. pneumoniae was detected in tracheobronchial
secretion by PCR and M. pneumoniae serology was reactive in serum by the complement fixation test
with a titer of 1,280. A: Subcortical cerebral white matter with numerous perivascular foci of demyelin-
ation and necrosis (hematoxylin and eosin stain, original magnification 40); B: Immunohistochemical
evidence of M. pneumoniae antigen inside macrophages present in the perivascular inflammatory
infiltrate (immunohistochemical assay performed by using the monoclonal antiM. pneumoniae antibody
and naphthol fast red as counterstain, original magnification 50). (From Stamm B, Moschopulos M,
Hungerbuehler H, et al. Neuroinvasion by Mycoplasma pneumoniae in acute disseminated encephalomy-
elitis. Emerg Infect Dis. 2008;14(4):641643, with permission.)

Scheld_Ch25.indd 421 2/21/14 5:43 PM


422 Part III: Bacterial and Mycoplasmal Infections

(anti-GalC) have been demonstrated in the sera of individuals suggests this cytokine may play an important role in the patho-
with postinfectious neurologic syndromes due to Mp (5962). genesis of the late-onset form of disease (31,65).
In one study, serum anti-GalC was detected in 3 of 3 indi- The pathogenesis of Mp-associated stroke is poorly under-
viduals with postinfectious CNS disease attributed to Mp stood (30,31). A procoagulable state has been implicated in
as compared to 8 of 32 with Mp infection restricted to the a minority of cases (6670), whereas in others, a systemic or
respiratory tract and 2 of 52 healthy controls (61). Among focal vasculitis appeared to be responsible (71,72). The obser-
patients enrolled in several Guillain-Barr syndrome treat- vation that the mean interval between the onset of respiratory
ment trials in Europe, 11 of 16 subjects with anti-GalC had illness and stroke is about 10 days (range 3 to 21 days) is con-
serologic evidence of Mp infection; anti-GalC was present sistent with an immune-mediated process (30).
not only in 78% of Mp-associated Guillain-Barr syndrome
patients but also in 58% of Mp infections without neurologic
disease (63). The antigen mimicry hypothesis is further sup- Clinical Manifestations
ported by the observation that the anti-GalC activity in serum
of patients with Mp-associated Guillain-Barr syndrome is Respiratory and Spectrum of Nonneurologic
inhibited by preincubation with Mp antigens (59,60). Among
subjects with Mp-associated encephalitis in the California
Systemic Manifestations
Encephalitis Project, anti-GalC was detected in the CSF of Mp is primarily a pathogen of the respiratory tract. Clinical
50% of subjects in whom there was neuroimaging evidence of disease is typified by an insidious onset of fever, headache,
demyelination (64). Whether anti-GalC causes disease or is an malaise, sore throat, and dry cough. In most, disease remains
epiphenomenon is uncertain. confined to the upper respiratory tract. Progression to tracheo-
With respect to Mp encephalitis, current data suggest that bronchitis or pneumonia occurs in fewer than 10% of cases.
pathogenesis is multifactorial, involving both direct infection Walking pneumonia, the hallmark of Mp respiratory disease,
and immunologically mediated processes. Several investiga- derives its name from the relatively mild clinical disposition of
tors have observed that Mp DNA is detected in serum or CSF most of those afflicted. Rales and wheezes may occasionally be
of subjects with a prodrome of less than or equal to 7 days heard, but physical examination is often unrevealing despite
but not in those with a longer prodrome (28,48,49). This sug- radiographic evidence of pneumonia. Widespread mottled,
gests two distinct patterns of Mp encephalitis: (a) an early- diffuse nodular densities are characteristic (Fig. 25.3); pleu-
onset syndrome caused by direct invasion and (b) a late-onset ral effusions occur in 5% to 20% of cases. Although rare,
syndrome in which the presence of the organism in the CSF fulminantand even fataldisease can occur.
or brain is not necessary to cause disease. The CSF cytokine Extrapulmonary manifestations of Mp infection have been
profile of both early- and late-onset forms of Mp encephalitis, described for almost every organ system (1,73). Neurologic
consisting of elevated interleukin-6 (IL-6) and IL-8 and nor- complications, followed by those affecting the skin and
mal interferon- and tumor necrosis factor-, suggests a dif- mucous membranes, are most common. The prototypical
ferent pathophysiology to that associated with other bacterial dermatologic complication, erythema multiforme minor and
and viral pathogens (31,65). The elevated proinflammatory Stevens-Johnson syndrome, are seen most often in children
cytokine IL-18 in late-onset, but not early-onset, encephalitis and young adults (7476); in some cases, isolated mucosal

A B

FIGURE 25.3 Posteroanterior (A) and lateral (B) chest radiographs of a 20-year-old patient with culture-
proven Mycoplasma pneumoniae pneumonia. Streaky, nodular densities are present in the right middle
and left lower lobe areas. (From Clyde WA Jr. Infections of the respiratory tract due to Mycoplasma
pneumoniae. In: Chernick V, Kendig E, eds. Disorders of the Respiratory Tract in Children. 5th ed.
Philadelphia: WB Saunders; 1990:403412, with permission.)

Scheld_Ch25.indd 422 2/21/14 5:43 PM


Chapter 25: Mycoplasmal and Ureaplasmal Infections 423

involvement can occur (75). Myalgia, arthralgia, and polyar- profile, and EEG and neuroimaging findings. Current or
thropathy are relatively common; septic arthritis is restricted recent upper or lower respiratory tract infection or pneu-
primarily to those with immunocompromising conditions monia, whether of undetermined cause or confirmed to be
(77,78). Cardiac complications include myocarditis, pericar- due to Mp, should raise the index of suspicion for Mp as
ditis, and pericardial effusion. Other extrapulmonary com- a possible cause. It is worth emphasizing again, however,
plications include acute glomerulonephritis, tubulointerstitial that the absence of respiratory disease does not preclude
nephritis, immunoglobulin (Ig) A nephropathy, renal failure, the diagnosis, particularly in young children. The presence
autoimmune hemolytic anemia, thrombocytopenic purpura, of nonneurologic extrapulmonary features compatible with
and intravascular coagulation (1). Mp may also serve as a clinical clue. The presence of cold
hemagglutinins, often used as a quick screen for Mp infec-
Neurologic Manifestations tion, should not be relied upon due to poor sensitivity and
The clinical manifestations of Mp-associated neurologic dis- specificity (167).
ease are protean and generally indistinguishable from those Microbiologic diagnosis of Mp infection is based on
due to other viral and bacterial pathogens. Syndromes ascribed serology and detection of the pathogen by culture or PCR
to Mp, selected key clinical features, strength of the microbio- in clinical samples. Culture is of limited use and not rou-
logic evidence supporting the association, and references for tinely available in most clinical laboratories due to its
each are provided in Table 25.1. It is important to empha- low sensitivity compared to PCR and because it is labor
size that for most of the neurologic syndromes associated with intensive, expensive, and requires up to 3 to 12 weeks of
Mp infection, proof of causality is lacking. The association incubation for positive results. The sensitivity of PCR (1 to
is strongest for encephalitis, meningitis, meningoencephalitis, 10 colony forming units) is 100-fold higher than culture.
ADEM, transverse myelitis, Guillain-Barr syndrome, and As Mp neurologic disease can be either due to direct infec-
acute striatal necrosis. tion of the brain or immunologically mediated, PCR testing
A history of respiratory tract infection preceding or or culture of both CSF and respiratory samples is recom-
accompanying neurologic symptoms is an important clue to mended. In one prospective 5-year study, Mp was detected
the diagnosis of Mp-associated neurologic disease. However, in the CSF of 6 of 11 (54%) children and in the respira-
the absence of a respiratory illness does not preclude Mp as a tory tract of 5 of 11 (46%) children with probable Mp en-
cause; between 35% and 75% of those with Mp-associated cephalitis (28). CSF samples intended for Mp PCR should
acute encephalitis have no history of respiratory symptoms be tested or frozen promptly because the stability of Mp
(23,28,29,40,7981). In those with respiratory symptoms, DNA is adversely impacted by storage at room or refrigera-
tracheobronchitis or pneumonia is more typical of adolescents tor temperatures (168). An important potential limitation
and adults, whereas children younger than 5 years of age often of both culture and PCR is that a positive result from a
have upper respiratory tract disease characterized by rhinitis, respiratory sample may reflect infection acquired as much
mild cough, or sore throat. In individuals with demyelinating as 3 to 7 months earlier and therefore may not be relevant
conditions, bilateral striatal necrosis, Bickerstaff brainstem to the acute illness (15,16,169171). For this reason, com-
encephalitis, opsoclonus myoclonus syndrome, and stroke, bining PCR testing with acute and convalescent serologic
respiratory symptoms are almost universal, preceding neuro- testing is encouraged.
logic symptom onset by 1 to 4 weeks. Numerous commercial serologic assays are available for
Acute encephalitis is the most common neurologic compli- the diagnosis of Mp infection (172). Most use a crude culture
cation attributable to Mp. Common clinical features include extract, which contains glycolipid antigens that cross-react
fever (50% to 100%), reduced or altered consciousness (45% with other mycoplasmal, bacterial, human, or plant antigens
to 100%), symptoms or signs of meningeal irritation (20% (173). Better performance (sensitivity and specificity) has been
to 80%), seizures (40% to 60%), focal neurologic deficits observed when recombinant antigens are used or the antigen
(20% to 60%), and ataxia (10% to 25%) (28,40,79). A mild is enriched for cytadhesin protein P1, although these assays
lymphocytic pleocytosis averaging less than 100 cells/L or a are generally not commercially available (174,175). In the ap-
slightly elevated CSF protein is demonstrated in 30% to 60% propriate clinical context, detection of IgM or IgA in acute
of cases (28,40,79,81). Nonspecific electroencephalographic sera, seroconversion from negative to positive, or a fourfold
(EEG) abnormalities, such as diffuse slowing or findings indica- rise in titer between acute and convalescent sera is indicative
tive of an epileptic focus, are observed in 80% to 100% of cases of acute infection (1,6). A negative result, however, does not
(28,40,79). Periodic lateralizing epileptiform discharges (28) exclude Mp infection; in a study of 12 commercially available
and extreme spindles (82) have been seen rarely. Computed kits in a predominantly adult population with PCR-proven
tomography or magnetic resonance imaging abnormalities sug- Mp respiratory tract infection, anti-Mp IgM was detected in
gestive of focal edema, ischemia, or inflammation are evident only 16% to 42% of acute sera and 32% to 84% of convales-
in 35% to 60% of cases (Figs. 25.4 to 25.6) (28,40,81). Mp cent sera (176). Negative serology has also been observed in
encephalitis is a severe entity with a mortality of up to 10% patients with culture or PCR-proven CNS disease due to Mp
(23,79,81) and residual sequelae that include cognitive impair- (28,34,38).
ment, seizure disorder, or focal motor deficits in 40% to 60% False-positive serologic results are also a significant
of survivors (28,79,81,83). In a study of 462 children with concern. For a cohort of children with encephalitis in
encephalitis, those due to Mp were seven times more likely to whom the prevalence of Mp infection is about 7% (28),
die or have severe neurologic sequelae than other children in a false-positive serologic test rate of approximately 50%
the cohort, second only to herpes simplex virus (84). is expected for an assay with a sensitivity of 90% and a
specificity of 94% (30). The observation that 80% of chil-
dren with encephalitis and reactive Mp IgM in acute sera, in
Diagnosis whom the organism was not detected by PCR in either CSF
or respiratory samples, had compelling evidence implicating
Mp should be considered in the differential diagnosis of other pathogens as the cause of encephalitis, reinforce this
any subject presenting with one of the syndromes associ- observation and the downside of relying solely on serology
ated with Mp regardless of clinical manifestations, CSF for diagnosis (28).

Scheld_Ch25.indd 423 2/21/14 5:43 PM


424 Part III: Bacterial and Mycoplasmal Infections

TA B L E 2 5 . 1
SELECTED CLINICAL FEATURES OF MYCOPLASMA PNEUMONIAEASSOCIATED
NEUROLOGIC SYNDROMES

Clinical Syndrome Selected Clinical and Investigative Features

Encephalitis and Most common neurologic syndrome associated with Mp; children account for 60%80% of cases
meningoencephalitis (23,29)
(24,25,28,34,35,37, Protean clinical features generally indistinguishable from encephalitis due to other etiologies; death or
3941,4345, neurologic sequelae occur in 40%60% (23,28,29,79,83,91)
4749,53,79,83, Respiratory symptoms prior to or concurrent with neurologic symptom onset in 25%65% of cases
85100) (23,28,29,79,83)
Mp detected in CSF or brain tissue by PCR or culture in significant number of cases
(28,34,35,39,41,4345,4749,52,53,8587,100)
Detection of Mp in blood or CSF most often in absence of prodrome, or prodrome 7 days in duration;
in those with a prodrome 7 days at time of neurologic symptom onset, Mp often detected in the
respiratory tract, but not in blood or CSF (28,48,49)
Antibiotic therapy is recommended; corticosteroids may be of benefit (101)
Meningitis Intermediate in frequency; majority of cases in children and young adults
(2325,29,52,94,102) Clinical features and CSF findings typical of aseptic meningitis; benign clinical course
Onset usually within 7 days of respiratory symptom onset
Implicated on basis of serology alone in most cases; occasionally detected in CSF by PCR or culture (52)
Potential benefit of antimicrobial therapy unknown
Acute disseminated Intermediate in frequency; no age predilection
encephalomyelitis Polysymptomatic with encephalopathy, focal neurologic deficits, and neuroimaging evidence of
(28,36,46,51,103113) multifocal asymmetric demyelination presenting 14 weeks after respiratory illness
Mp implicated on basis of serology alone in most reported cases; occasionally detected by PCR in
respiratory samples (28,36,108) or CSF (46,51)
Anti-ganglioside antibodies demonstrated occasionally (105)
Primary treatment with corticosteroids, IVIG, or plasmapheresis; role of antimicrobial therapy
uncertain
Acute hemorrhagic Rare; fewer than 10 reports; no age predilection
leukoencephalitis Hyperacute form of ADEM characterized by rapid progression from confusion and muscle weakness
(Hurst disease) to stupor and coma 14 weeks after respiratory tract illness
(114117) Mp implicated on the basis of serology alone in all reported cases
Treatment with plasmapheresis (115,116) or corticosteroids (114,116,117) with partial response in some
Neurologic sequelae common in survivors (114,116)
Transverse myelitis Moderate in frequency (3550 reported cases); most cases in children and young adults
(38,42,111, Focal motor, sensory, or autonomic deficits and neuroimaging evidence of focal spinal cord
118120) demyelination 14 weeks after respiratory illness
Mp implicated on basis of serology alone in most reported cases; occasionally detected by PCR in
respiratory samples (38) or CSF (38,42)
Primary treatment with corticosteroids, IVIG, or plasmapheresis; role of antimicrobial therapy
uncertain
Bickerstaff brainstem Rare; 2 case reports involving children; no reported adult cases
encephalitis (121,122) External ophthalmoplegia, ataxia, and altered consciousness 12 weeks after respiratory illness
Mp implicated on the basis of serology and, in one case, detection of Mp in the respiratory tract by
PCR (121)
Elevated anti-GQ1b antibody titers in serum
Treatment with IVIG or immune adsorption associated with full recovery
Acute bilateral striatal Intermediate in frequency (1020 reported cases); children and adolescents, no reported adult cases
encephalitis (striatal Encephalopathy and extrapyramidal features most often 14 weeks after respiratory tract illness (124)
necrosis) (123140) Mp implicated on the basis of serology in most cases; detected by PCR in respiratory sample of one
case (124) and CSF of one case (123)
Anti-GM1 ganglioside antibody demonstrated in one case (136)
Treatment with corticosteroids (135,136), IVIG (124,136,140), or plasmapheresis (135) associated
with apparent response in some
Gradual but full recovery in most cases; residual movement disorder in some (127,129,132,137)
Acute bilateral thalamic Rare; fewer than 5 reports all children
necrosis (141143) Acute onset of encephalopathy and symmetric thalamic abnormality on imaging within 7 days of
respiratory symptom onset
Mp implicated on the basis of serology alone in all reported cases
Residual sequelae such as dystonia and tremor

(continued)

Scheld_Ch25.indd 424 2/21/14 5:43 PM


Chapter 25: Mycoplasmal and Ureaplasmal Infections 425

TA B L E 2 5 . 1
SELECTED CLINICAL FEATURES OF MYCOPLASMA PNEUMONIAEASSOCIATED
NEUROLOGIC SYNDROMES (CONTINUED)

Clinical Syndrome Selected Clinical and Investigative Features

Opsoclonus-myoclonus Rare; fewer than 10 reports, predominantly involving children


syndrome (144148) Opsoclonus, myoclonus, and ataxia 14 weeks after respiratory tract illness
Mp implicated on the basis of serology and selectively PCR detection in respiratory tract samples (145)
Anti-glutamate receptor antibodies detected in CSF of one case (148)
Corticosteroid and/or IVIG prescribed in all cases; dramatic response to plasmapheresis in one case (146)
Full recovery in all reported cases
Guillain-Barr syndrome Moderate in frequency; Mp implicated in 5%15% of GBS cases (149152)
(GBS) (83,149155) Onset of neurologic symptoms 14 weeks after respiratory illness
Mp implicated on basis of serology alone in most reported cases; occasionally detected by PCR or
culture in respiratory samples (153,155157) or CSF (153)
Anti-ganglioside antibodies often detected (152,157)
Primary treatment with IVIG or plasmapheresis; role of antimicrobial therapy uncertain
Cerebellar ataxia Rare; reported in both children and adults
(39,98,158,159) Onset of cerebellar manifestations 13 weeks after respiratory illness; in some, but not all, associated
with a more generalized encephalitic picture (39,158)
Mp implicated on the basis of serology in most cases; detected in throat sample and CSF in one case (39)
Potential benefit of antimicrobial therapy or immune-modulating therapies unknown
Stroke (50,6672, Rare; no age predilection
160164) Neurologic symptoms usually develop 13 weeks after onset of respiratory illness
Hypothesized to be related to hypercoagulable state (6670) or vasculitis (71,72)
Mp implicated on the basis of serology alone in most reported cases; detected in respiratory tract
sample and CSF in one reported case (50)
Antiphospholipid and anticardiolipin antibodies demonstrated in some cases (50,70)
Potential benefit of antimicrobial therapy, corticosteroids, or IVIG unknown
Cerebral vasculopathy Rare; 4 reported cases, all adults
(54,55,165,166) Limited evidence implicating Mp; diagnosis based on immunostaining and electron microscopy
Subacute presentation with cognitive decline, weakness, ataxia

PCR, polymerase chain reaction; IVIG, intravenous immune globulin; ADEM, acute disseminated encephalomyelitis.

FIGURE 25.4 Neuroimages of an 8-year-old girl who presented with acute left-sided hemiplegia sub-
sequent to a 5-day history of fever, headache, and vomiting; Mycoplasma pneumoniae was detected in
the CSF by PCR. A: Magnetic resonance angiogram shows prominence of the vessels of the right middle
cerebral artery distribution in keeping with loss of autoregulation (arrows). The caliber of the arteries
in the left middle cerebral artery distribution is normal. B: T2-weighted coronal image (TR5700, TE90)
shows extensive cortical swelling and increased signal intensity within the gray matter of the right frontal
and temporal lobes. The white matter is relatively spared. (From Bitnun A, Ford-Jones E, Blaser S, et al.
Mycoplasma pneumoniae ecephalitis. Semin Pediatr Infect Dis. 2003;14:96107, with permission.)

Scheld_Ch25.indd 425 2/21/14 5:43 PM


426 Part III: Bacterial and Mycoplasmal Infections

FIGURE 25.5 Magnetic resonance image demonstrating pronounced


edema of the cortical gray matter in the left hemisphere with exten-
sion to involve the subjacent white matter. The ventricles are large, FIGURE 25.6 Magnetic resonance image of a 10-year-old female
but there is no periventricular edema. This 14-month-old female pre- with Mycoplasma pneumoniaeinduced ADEM. On this T2-weighted
sented with a right-sided focal seizure and encephalopathy following axial (TR2800, TE90) image, multiple foci of increased signal inten-
a respiratory illness of 7 days duration. Mycoplasma pneumoniae sity within deep gray structures and white matter are present. Upper
was implicated as the likely culprit on the basis of a complement respiratory symptoms of 2 weeks duration had resolved 1 to 2 weeks
fixation titer of 1:128. (From Bitnun A, Ford-Jones E, Blaser S, prior to the onset of encephalitis. M. pneumoniae was implicated on
et al. Mycoplasma pneumoniae ecephalitis. Semin Pediatr Infect Dis. the basis of positive serology (positive IgM by enzyme immunoassay
2003;14:96107, with permission.) and acute/convalescent complement fixation titers of 1:128/1:512)
and detection of the organism by PCR in the throat. (From Bitnun
A, Ford-Jones E, Blaser S, et al. Mycoplasma pneumoniae ecephalitis.
Semin Pediatr Infect Dis. 2003;14:96107, with permission.)
Treatment
The most appropriate therapeutic interventions for suspected (23,25,52,53,61,83,87,112). In adults and children 8 years of
or proven CNS disease due to Mp depend largely on the patho- age or older, in whom direct infection of the brain is proven
genesis of the syndrome in question. Key considerations are or possible, antibiotics with good in vitro and in vivo activ-
the need for antimicrobial therapy and/or immune-modulating ity against Mp capable of traversing the bloodbrain bar-
therapies such as corticosteroids, intravenous immune globulin rier and achieving therapeutic levels within the CNS such
(IVIG), or plasmapheresis (177,178). Because controlled trials as azithromycin, doxycycline, or a fluoroquinolone such as
evaluating these treatments have not been conducted, recom- moxifloxacin or levofloxacin are preferred (100,180184).
mendations are based on anecdotal evidence from case reports Chloramphenicol is another option as it achieves excellent
and case series and, in reference to immunologically mediated CSF levels (184) and has good activity against Mp, but the
entities such as ADEM, transverse myelitis, and Guillain-Barr risk of idiosyncratic bone marrow aplasia is of concern. For
syndrome, extrapolated from data relating to these syndromes those with immunologically mediated conditions in whom the
irrespective of the infectious trigger. A multidisciplinary care CSF tests negative for Mp by PCR, eradication of the patho-
approach is important; for patients with increased intracranial gen from the respiratory tract using erythromycin or clarithro-
pressure, urgent neurosurgical consultation should be sought mycin may be appropriate. In children younger than 8 years
as hemicraniectomy may be lifesaving (179). of age, doxycycline and tetracycline should be avoided due to
Antibiotic therapy should be considered for all patients the risk of enamel hypoplasia and irreversible darkening of
with neurologic disease attributed to Mp, despite the pau- permanent teeth.
city of efficacy data, because of the potential for significant Immune-modulating therapies are frequently used in the
neurologic sequelae. It should be noted, however, that the management of demyelinating conditions. Corticosteroids
impact of such therapy on outcome is unknown; full recov- and IVIG, used alone or in combination, have been associated
ery has been observed with (38,39,41,44,51,52,61,83,85,8 with temporal clinical improvement in some (28,46,51,110
6,97,100,112,155) and without (47,48,61,106) antibiotic 112,114,118120,185), but not all (36,103,107,186), patients
therapy, as has an apparent lack of response to such therapy with ADEM and transverse myelitis. IVIG is preferred in

Scheld_Ch25.indd 426 2/21/14 5:43 PM


Chapter 25: Mycoplasmal and Ureaplasmal Infections 427

Guillain-Barr syndrome (155,156,187). Plasma exchange is


reserved for patients (with all three conditions) who fail to OTHER MYCOPLASMA SPECIES
respond to corticosteroids or IVIG (103,109,114).
Corticosteroids may also have a role in the treatment of Mh and Uu are rare causes of CNS disease but neverthe-
Mp-associated encephalitis. In a case report and retrospective less account for the vast majority of mycoplasmal CNS dis-
review of severe Mp encephalitis, treatment with corticoste- ease not attributable to Mp. Both are commensals of the
roids was associated with complete or near-complete recovery genitourinary tract. The Ureaplasma genus contains 14 dis-
in 78% of cases (101). By comparison, full recovery was evi- tinct serovars divided into two separate species: U. parvum
dent in only 52% of children not treated with steroids (79). (serovars 1, 3, 6, and 14) and U. urealyticum (serovars 2, 4,
A prolonged taper over several weeks may be warranted in 5, and 7 through 13) (188). It is not known if there are any
severe cases to prevent symptomatic relapse (101). The role differences between the two species with respect to pathoge-
of immune-modulating therapies in less common forms of nicity, and at present, they can only be differentiated by sero-
Mp neurologic disease is uncertain. Corticosteroids and IVIG, typing or PCR (2,189). For purposes of this review, they are
sometimes in combination, have been associated with temporal considered together. Rare case reports of CNS disease due to
clinical improvement in several patients with Mp-associated Mycoplasma salivarium, Mycoplasma faucium, Mycoplasma
bilateral striatal necrosis (124,135,136) and opsoclonus myoc- genitalium, and Mycoplasma maculosum have been reported
lonus syndrome (145,147,148). IVIG and immune adsorption (35,190,191). A summary of the salient features of CNS
therapy have been used with apparent success in Bickerstaff infections due to mycoplasmal species other than Mp is pro-
brainstem encephalitis (121,122). vided in Table 25.2.

TA B L E 2 5 . 2
CENTRAL NERVOUS SYSTEM INFECTIONS ATTRIBUTABLE TO MYCOPLASMA AND UREAPLASMA SPECIES
OTHER THAN MYCOPLASMA PNEUMONIAE

Species Syndrome Salient Clinical Features

Mycoplasma Meningitis and Sole pathogen in reported cases; diagnosis by culture or molecular detection
hominis meningoencephalitis techniques
(192205) Premature and term neonates predominantly; older individuals with head trauma
or neurosurgical procedures (192,205) or immunocompromising conditions (192)
Targeted antibiotic therapy recommended, although recovery without such
therapy noted in some neonates (200,201)
Brain abscess Sole pathogen in most reported cases, polymicrobial in others (190); diagnosis
(190,192,206209) by culture or molecular detection techniques
Predominantly premature and term neonates; rare reports of adults and older
children (190,192,206)
Most adult cases associated with head trauma or neurosurgical procedures
(192,206)
Ureaplasma Meningitis Sole pathogen in most reported cases; diagnosis by culture or molecular
urealyticum/ (200,201,210220) detection techniques
Ureaplasma Premature neonates predominantly; one reported case of immunocompromised
parvum adult following a neurosurgical procedure (221)
Targeted antibiotic therapy recommended, although recovery without such
therapy noted in many neonates (200,201,214)
Mycoplasma Brain abscess (5) Polymicrobial; detected in culture or by PCR
salivarium Poor dental hygiene in 55-year-old male with history of alcohol abuse and
prior pulmonary tuberculosis; second case involved 26-year-old male with no
underlying conditions
Antibiotic therapy included moxifloxacin in both cases
Residual neurologic deficit in one case
Mycoplasma Brain abscess (190,191) Polymicrobial; to date, detection only using molecular techniques
faucium Immunologically normal adults; association with sinusitis, dental abnormalities (191)
Need for targeted antimicrobial therapy uncertainfull recovery with antibiotic
therapy not targeting M. faucium reported in all cases (191)
Mycoplasma Brainstem encephalitis (4) Five-year-old previously healthy girl; full recovery
genitalium Detected by PCR in CSF
Mycoplasma Meningitis (3) Fifteen-year-old with common variable immune deficiency
maculosum Repeatedly detected by PCR and cultured from CSF
Likely acquired from pet dog (M. maculosum is a canine Mycoplasma species)
Failed treatment with doxycycline and ciprofloxacin; responded to prolonged
course of valnemulin hydrochloride (pleuromutilin antibiotic)

PCR, polymerase chain reaction; CSF, cerebrospinal fluid

Scheld_Ch25.indd 427 2/21/14 5:43 PM


428 Part III: Bacterial and Mycoplasmal Infections

Gram stain of a colony sample should lead to consideration of


MYCOPLASMA HOMINIS mycoplasmal infection. Molecular diagnosis using PCR offers
superior sensitivity and improved turnaround time compared
Epidemiology to culture (231,232). Serology is of no value in diagnosis.

Mh colonizes the genital tract of 0% to 20% and 10% to 30%


of otherwise healthy sexually active men and women, respec- Treatment
tively (222224). Higher rates occur in men with urethritis
and in women with bacterial vaginosis, urethritis, or cervicitis Mh is usually susceptible to the tetracyclines (tetracycline,
(225,226). Between 5% and 10% of newborns are asymptom- doxycycline, minocycline), chloramphenicol, lincosamides
atically colonized at the time of birth (227,228). Colonization (clindamycin), quinupristin-dalfopristin, and selectively to the
is transient, only rarely being detected in prepubertal children fluoroquinolones (233). Tetracyclines are considered the drugs
beyond 3 months of age (228). Colonization rates increase of choice (192,193). Doxycycline has been used most exten-
with the onset of sexual activity (222,229,230). sively, including in neonates despite the potential for bone and
dental toxicity (194,196,197,199,201,208). Chloramphenicol
has been used alone or in combination with other antibiotics
Clinical Features (193,195,197199,203,209); in one case, it had to be discon-
tinued due to presumed toxicity that included hypotension and
Mh has been associated with nongonococcal urethritis in both pancytopenia (199). Several recent Mh meningitis cases have
men and women and with bacterial vaginosis and pelvic in- been successfully treated with moxifloxacin or gatifloxacin
flammatory disease in women. Wound infections subsequent to (193,202,206). The in vitro efficacy of these fourth-generation
trauma or surgery and septic arthritis in association with a pros- fluoroquinolones is superior to that of the older generation
thetic joint or an immunocompromised state such as malignancy fluoroquinolones ciprofloxacin and levofloxacin (193,233).
and hypogammaglobulinemia have been reported (3,192,205). Potential advantages of the fluoroquinolones include their bac-
The majority of CNS infections due to Mh occur in neo- tericidal activity and excellent CSF penetration (193,202,206).
nates (193). Premature and term infants have both been af- A 2- to 3-week course of antibiotic therapy is usually sufficient
fected. Meningitis and meningoencephalitis are the most for Mh meningitis. In those with brain abscesses, 6 weeks of
common entities (193204); brain abscess and subdural em- such therapy should be given; surgical drainage needs to be
pyema also occur (207209). Colonization of the respiratory considered in all cases.
or genitourinary tract during the birthing process followed by It is paramount that susceptibility testing be performed on
hematogenous dissemination is the likely pathogenesis. Onset all sterile site isolates of Mh due to increasing rates of antibi-
of symptoms in reported cases ranged from day 1 to day 32 of otic resistance (234). For example, over a 20-year period in
life, the majority during the first 2 weeks of life (193). Clinical Germany, resistance to tetracycline, doxycycline, and cipro-
and laboratory features are indistinguishable from those due floxacin rose from 2.1% to 14.5%, 0% to 14.5%, and 0% to
to other bacterial pathogens. CSF pleocytosis, elevated CSF 15.4%, respectively (234).
protein, and hypoglycorrhachia have all been observed (193);
a predominance of polymorphonuclear leukocytes is charac-
teristic. Adverse outcome is seen in approximately 50% of UREAPLASMA SPECIES
cases, death in 28%, and neurologic sequelae in 28% (193). (UREAPLASMA UREALYTICUM
In older children and adults, Mh has been implicated as a
cause of brain abscess subsequent to neurosurgical procedures AND UREAPLASMA PARVUM)
(192,206) and meningitis in the context of immune suppres-
sion related to cancer chemotherapy (192). Fever and focal Epidemiology
neurologic manifestations are typical. Mh has also been de-
tected by multiple 16S ribosomal DNA sequencing in poly- Asymptomatic colonization of the genital tract occurs in 40%
microbial brain abscesses (190); the significance of Mh in to 80% of sexually active women (2,222,225); among sexually
pathogenesis of these brain abscesses and, by extension, the active men, approximately 25% to 50% are similarly colonized
need for Mh-targeted antimicrobial therapy is unknown. (223,225). Approximately half of newborn infants of colonized
mothers acquire the infection perinatally, the vast majority
asymptomatically (235237). Colonization rates decline during
Diagnosis early infancy, and in children 2 to 13 years of age, the organism
can rarely be detected (228,230). As is the case with Mh, genital
Because of the rarity of Mh CNS infection and the difficulty in colonization rates increase with onset of sexual activity (230).
culturing the organism, diagnosis requires a high index of sus-
picion. It should be considered in any neonate with meningitis,
encephalitis, or brain abscess when CSF cultures are negative Clinical Features
and there is no clinical response to standard neonatal sepsis
treatment, such as a combination of -lactams (e.g., ampicillin In adults, Uu is primarily associated with nongonococcal ure-
plus cefotaxime) or a -lactam with an aminoglycoside. thritis in men and complications of pregnancy in women (238).
As the organism is extremely sensitive to environmental con- Maternal urogenital colonization with Uu is associated with
ditions, specimens should ideally be inoculated into appropri- histologic chorioamnionitis, stillbirth, preterm birth, and ad-
ate transport media, such as 10B or SP4 broth, at the bedside. verse neonatal outcomes including neonatal pneumonia, bron-
The preferred culture medium is SP4 broth and agar (pH 7.5) chopulmonary dysplasia, intraventricular hemorrhage, and
supplemented with arginine. Mh may occasionally grow as lower psychomotor development scores at 2 years of age (239).
pinpoint translucent colonies after 2 to 3 days incubation on High levels of the proinflammatory cytokines IL-1, tumor
conventional blood or chocolate agar plates. Failure of these necrosis factor-, IL-6, and IL-8 in amniotic fluid correlate
colonies to enlarge and the inability to visualize organisms by with preterm premature rupture of membranes (pPROM) and

Scheld_Ch25.indd 428 2/21/14 5:43 PM


Chapter 25: Mycoplasmal and Ureaplasmal Infections 429

isolation of Uu, which suggests that Uu is truly pathogenic in 24 to 48 hours. Uu is distinguished from other Mycoplasma
this setting. Extragenital non-CNS infections reported rarely species by its ability to hydrolyze urea. As is the case for Mh,
in older children and adults include postoperative wound and PCR is increasingly used in diagnosis, and serology is of no
prosthetic joint infections and systemic infections such as septic diagnostic value.
arthritis in patients with common variable immune deficiency
and other immunocompromising conditions (3,240243).
CNS disease due to Uu occurs primarily in premature Treatment
infants (200,201,208,210220). Meningitis is the most com-
mon syndrome. There is one report of a brain abscess, from In contrast with Mh, Uu is usually susceptible to erythromycin
which both Uu and Mh were isolated, in a 3-week-old term and other macrolides but is relatively resistant to clindamycin
infant (208). An association between Uu infection and intra- (2). Macrolides are considered the treatment of choice for
ventricular hemorrhage has been noted in some (201,220), infections outside the CNS, whereas the tetracyclines, chlor-
but not all, studies (244). In one study, the risk of grade 3 to amphenicol, or a fluoroquinolone are preferred for CNS in-
4 intraventricular hemorrhage among very-low-birth-weight fections. Doxycycline and chloramphenicol exhibit excellent
premature infants was fivefold higher when Uu was detected in vitro efficacy (233), and no treatment failures have been
in serum by PCR and serum IL-1 was elevated; of note, reported with their use (212). The fluoroquinolones have been
U. parvum was the only Uu species detected in the serum of used less often but appeared to be effective (210,212,245).
these patients (220). Despite these suggestive data, a causal Most cases of meningitis have been treated with erythromycin
link is uncertain because very-low-birth-weight premature in- in combination with doxycycline, chloramphenicol, or a fluo-
fants are at high risk of intraventricular hemorrhage for many roquinolone (201,208,211214,218,219,245,246). Persistent
reasons irrespective of Uu colonization or infection status. CSF cultures while on erythromycin monotherapy and prompt
In adults, CNS disease due to Uu is exceedingly rare. The only sterilization following the addition of doxycycline, chloram-
published report was that of a 38-year-old male who developed phenicol, or ciprofloxacin has been observed (212,213,218).
Uu (biovar 2) meningitis 10 weeks after renal transplantation and Susceptibility testing of sterile site isolates is required as resis-
4 weeks after placement of a ventricular drain for management tance to the tetracyclines, chloramphenicol, and the fluoroqui-
of increased intracranial pressure (221). He was successfully nolones can occur (2,211,245,247).
treated with a combination of doxycycline and chloramphenicol. Approximately two thirds of neonates with Uu meningi-
tis recover despite not receiving specific antibiotic therapy,
perhaps reflecting the low virulence of the organism and
Diagnosis containment of the infection by the immune system (212).
Nevertheless, it is recommended that symptomatic patients, in
Unlike Mh, Uu cannot be cultured from routine bacterio- whom Uu is isolated from the CSF, should receive targeted an-
logic media. Its specific transport and culture requirements tibiotic therapy (2,212). A treatment duration of 2 to 3 weeks
are similar to those of Mh (239). Growth usually occurs in is sufficient in most cases.

References
1. Waites KB, Talkington DF. Mycoplasma pneumoniae and its role as a 14. Meseguer MA, Alvarez A, Rejas MT, et al. Mycoplasma pneumoniae:
human pathogen. Clin Microbiol Rev. 2004;17:697728, table of contents. a reduced-genome intracellular bacterial pathogen. Infect Genet Evol.
2. Waites KB, Katz B, Schelonka RL. Mycoplasmas and ureaplasmas as neo- 2003;3:4755.
natal pathogens. Clin Microbiol Rev. 2005;18:757789. 15. Foy HM, Kenny GE, McMahan R, et al. Mycoplasma pneumoniae pneumo-
3. Heilmann C, Jensen L, Jensen JS, et al. Treatment of resistant Mycoplasma nia in an urban area. Five years of surveillance. JAMA. 1970;214:16661672.
infection in immunocompromised patients with a new pleuromutilin anti- 16. Foy HM. Infections caused by Mycoplasma pneumoniae and possible carrier
biotic. J Infect. 2001;43:234238. state in different populations of patients. Clin Infect Dis. 1993;17(suppl 1):
4. Sakata H, Hayashi T, Hirano Y, et al. Brain stem encephalitis due to S37S46.
Mycoplasma genitalium [in Japanese]. Kansenshogaku Zasshi. 1993;67: 17. Foy HM, Kenny GE, Cooney MK, et al. Long-term epidemiology of infec-
500504. tions with Mycoplasma pneumoniae. J Infect Dis. 1979;139:681687.
5. Orsted I, Gertsen JB, Schonheyder HC, et al. Mycoplasma salivarium iso- 18. Feikin DR, Moroney JF, Talkington DF, et al. An outbreak of acute respira-
lated from brain abscesses. Clin Microbiol Infect. 2011;17:10471049. tory disease caused by Mycoplasma pneumoniae and adenovirus at a fed-
6. Atkinson TP, Balish MF, Waites KB. Epidemiology, clinical manifestations, eral service training academy: new implications from an old scenario. Clin
pathogenesis and laboratory detection of Mycoplasma pneumoniae infec- Infect Dis. 1999;29:15451550.
tions. FEMS Microbiol Rev. 2008;32:956973. 19. Mogabgab WJ. Mycoplasma pneumoniae and adenovirus respiratory ill-
7. Maniloff J. Phylogeny of mycoplasmas. In: Maniloff J, McElhaney R, nesses in military and university personnel, 19591966. Am Rev Respir
Finch L, et al, eds. Mycoplasmas: Molecular Biology and Pathogenesis. Dis. 1968;97:345358.
Washington, DC: American Society of Microbiology; 1992:549559. 20. Foy HM, Grayston JT, Kenny GE, et al. Epidemiology of Mycoplasma
8. Eaton MD, Meiklejohn G, van Herick W. Studies on the etiology of pri- pneumoniae infection in families. JAMA. 1966;197:859866.
mary atypical pneumonia: a filterable agent transmissible to cotton rats, 21. Dorigo-Zetsma JW, Wilbrink B, van der Nat H, et al. Results of molecular
hamsters, and chick embryos. J Exp Med. 1944;79:649668. detection of Mycoplasma pneumoniae among patients with acute respira-
9. Chanock RM. Mycoplasma pneumoniae: proposed nomenclature for atyp- tory infection and in their household contacts reveals children as human
ical pneumonia organism (Eaton agent). Science. 1963;140:662. reservoirs. J Infect Dis. 2001;183:675678.
10. Waites KB, Balish MF, Atkinson TP. New insights into the pathogenesis 22. Yesnick L. Central nervous system complications of primary atypical pneu-
and detection of Mycoplasma pneumoniae infections. Future Microbiol. monia. AMA Arch Intern Med. 1956;97:9398.
2008;3:635648. 23. Ponka A. Central nervous system manifestations associated with sero-
11. Jordan JL, Chang HY, Balish MF, et al. Protein P200 is dispensable for logically verified Mycoplasma pneumoniae infection. Scand J Infect Dis.
Mycoplasma pneumoniae hemadsorption but not gliding motility or coloniza- 1980;12:175184.
tion of differentiated bronchial epithelium. Infect Immun. 2007;75:518522. 24. Sterner G, Biberfeld G. Central nervous system complications of
12. Krunkosky TM, Jordan JL, Chambers E, et al. Mycoplasma pneumoniae Mycoplasma pneumoniae infection. Scand J Infect Dis. 1969;1:203208.
host-pathogen studies in an air-liquid culture of differentiated human air- 25. Lind K, Zoffmann H, Larsen SO, et al. Mycoplasma pneumoniae infection
way epithelial cells. Microb Pathog. 2007;42:98103. associated with affection of the central nervous system. Acta Med Scand.
13. Yavlovich A, Tarshis M, Rottem S. Internalization and intracellular survival 1979;205:325332.
of Mycoplasma pneumoniae by non-phagocytic cells. FEMS Microbiol 26. Tong CY, Menson E, Lin JP, et al. Prevalence of Mycoplasma encephalitis.
Lett. 2004;233:241246. Lancet Infect Dis. 2011;11:425426.

Scheld_Ch25.indd 429 2/21/14 5:43 PM


430 Part III: Bacterial and Mycoplasmal Infections

27. Walter ND, Grant GB, Bandy U, et al. Community outbreak of 57. Hill A. Mycoplasma pulmonis infection in the brains of rodents. J Infect
Mycoplasma pneumoniae infection: school-based cluster of neurologic dis- Dis. 1983;147:540555.
ease associated with household transmission of respiratory illness. J Infect 58. Brown DR, Nogueira MF, Schoeb TR, et al. Pathology of experimental
Dis. 2008;198:13651374. mycoplasmosis in American alligators. J Wildl Dis. 2001;37:671679.
28. Bitnun A, Ford-Jones EL, Petric M, et al. Acute childhood encephalitis and 59. Susuki K, Odaka M, Mori M, et al. Acute motor axonal neuropathy
Mycoplasma pneumoniae. Clin Infect Dis. 2001;32:16741784. after Mycoplasma infection: evidence of molecular mimicry. Neurology.
29. Koskiniemi M. CNS manifestations associated with Mycoplasma pneu- 2004;62:949956.
moniae infections: summary of cases at the University of Helsinki and 60. Kusunoki S, Shiina M, Kanazawa I. Anti-Gal-C antibodies in GBS subse-
review. Clin Infect Dis. 1993;17(suppl 1):S52S57. quent to Mycoplasma infection: evidence of molecular mimicry. Neurology.
30. Bitnun A, Richardson SE. Mycoplasma pneumoniae: innocent bystander 2001;57:736738.
or a true cause of central nervous system disease? Curr Infect Dis Rep. 61. Nishimura M, Saida T, Kuroki S, et al. Post-infectious encephalitis with
2010;12:282290. anti-galactocerebroside antibody subsequent to Mycoplasma pneumoniae
31. Narita M. Pathogenesis of neurologic manifestations of Mycoplasma pneu- infection. J Neurol Sci. 1996;140:9195.
moniae infection. Pediatr Neurol. 2009;41:159166. 62. Kusunoki S, Chiba A, Hitoshi S, et al. Anti-Gal-C antibody in auto-
32. Jordan F. Avian mycoplasmas. In: Tully J, Whitcomb R, eds. The Mycoplasmas. immune neuropathies subsequent to Mycoplasma infection. Muscle Nerve.
New York: Academic Press; 1979:148. 1995;18:409413.
33. Findlay G, Klieneberger E, Maccallum F, et al. Rolling disease: new syn- 63. Ang CW, Tio-Gillen AP, Groen J, et al. Cross-reactive anti-galactocerebro-
drome in mice associated with pleuropulmonary-like organism. Lancet. side antibodies and Mycoplasma pneumoniae infections in Guillain-Barre
1938;2:15111153. syndrome. J Neuroimmunol. 2002;130:179183.
34. Koletsky RJ, Weinstein AJ. Fulminant Mycoplasma pneumoniae infection. 64. Christie LJ, Honarmand S, Yagi S, et al. Anti-galactocerebroside testing
Report of a fatal case, and a review of the literature. Am Rev Respir Dis. in Mycoplasma pneumoniae-associated encephalitis. J Neuroimmunol.
1980;122:491496. 2007;189:129131.
35. Launes J, Paetau A, Linnavuori K, et al. Direct invasion of the brain paren- 65. Narita M, Tanaka H, Togashi T, et al. Cytokines involved in CNS manifesta-
chyma by Mycoplasma pneumoniae. Acta Neurol Scand. 1997;95:374. tions caused by Mycoplasma pneumoniae. Pediatr Neurol. 2005;33:105109.
36. Stamm B, Moschopulos M, Hungerbuehler H, et al. Neuroinvasion by 66. Fu M, Wong KS, Lam WW, et al. Middle cerebral artery occlusion after re-
Mycoplasma pneumoniae in acute disseminated encephalomyelitis. Emerg cent Mycoplasma pneumoniae infection. J Neurol Sci. 1998;157:113115.
Infect Dis. 2008;14(4):641643. 67. Mulder LJ, Spierings EL. Stroke in a young adult with Mycoplasma pneu-
37. Powers JM, Johnson MD. Mycoplasmal panencephalitis: a neuropatho- moniae infection complicated by intravascular coagulation. Neurology.
logic documentation. Acta Neuropathol. 2012;124:143148. 1987;37:14301431.
38. Abele-Horn M, Franck W, Busch U, et al. Transverse myelitis associated 68. Ode B, Cronberg S. Infection and intracranial arterial thrombosis. Lancet.
with Mycoplasma pneumoniae infection. Clin Infect Dis. 1998;26:909912. 1976;2:863864.
39. Abramovitz P, Schvartzman P, Harel D, et al. Direct invasion of the central 69. Ryu JS, Kim HJ, Sung IY, et al. Posterior cerebral artery occlusion after
nervous system by Mycoplasma pneumoniae: a report of two cases. J Infect Mycoplasma pneumoniae infection associated with genetic defect of
Dis. 1987;155:482487. MTHFR C677T. J Child Neurol. 2009;24:891894.
40. Christie LJ, Honarmand S, Talkington DF, et al. Pediatric encephalitis: what 70. Senda J, Ito M, Atsuta N, et al. Paradoxical brain embolism induced by
is the role of Mycoplasma pneumoniae? Pediatrics. 2007;120:305313. Mycoplasma pneumoniae infection with deep venous thrombus. Intern
41. Dionisio D, Valassina M, Mata S, et al. Encephalitis caused directly by Med. 2010;49:20032005.
Mycoplasma pneumoniae. Scand J Infect Dis. 1999;31:506509. 71. Ovetchkine P, Brugieres P, Seradj A, et al. An 8-y-old boy with acute stroke
42. Goebels N, Helmchen C, Abele-Horn M, et al. Extensive myelitis associ- and radiological signs of cerebral vasculitis after recent Mycoplasma pneu-
ated with Mycoplasma pneumoniae infection: magnetic resonance imaging moniae infection. Scand J Infect Dis. 2002;34:307309.
and clinical long-term follow-up. J Neurol. 2001;248:204208. 72. Perez C, Montes M. Cutaneous leukocytoclastic vasculitis and encephali-
43. Inoue S, Ohfu M, Morooka T, et al. A case of encephalitis due to tis associated with Mycoplasma pneumoniae infection. Arch Intern Med.
Mycoplasma pneumoniae: detection of specific DNA from cerebrospinal 2002;162:352354.
fluid and elevation of interleukin-6 [in Japanese]. Kansenshogaku Zasshi. 73. Sanchez-Vargas FM, Gomez-Duarte OG. Mycoplasma pneumoniaean
1998;72:771775. emerging extra-pulmonary pathogen. Clin Microbiol Infect. 2008;14:105117.
44. Kasahara I, Otsubo Y, Yanase T, et al. Isolation and characterization of 74. Levy M, Shear NH. Mycoplasma pneumoniae infections and Stevens-
Mycoplasma pneumoniae from cerebrospinal fluid of a patient with pneu- Johnson syndrome. Report of eight cases and review of the literature. Clin
monia and meningoencephalitis. J Infect Dis. 1985;152:823825. Pediatr. 1991;30:4249.
45. Lin WC, Lee PI, Lu CY, et al. Mycoplasma pneumoniae encephalitis in 75. Schalock PC, Dinulos JG. Mycoplasma pneumoniae-induced Stevens-
childhood. J Microbiol Immunol Infect. 2002;35:173178. Johnson syndrome without skin lesions: fact or fiction? J Am Acad
46. Matsumoto N, Takahashi S, Toriumi N, et al. Acute disseminated encepha- Dermatol. 2005;52:312315.
lomyelitis in an infant with incontinentia pigmenti. Brain Dev. 2009;31: 76. Lyell A, Gordon AM, Dick HM, et al. Mycoplasmas and erythema multi-
625628. forme. Lancet. 1967;2:11161118.
47. Nagayama Y, Sakurai N, Tamai K, et al. Isolation of Mycoplasma pneu- 77. OSullivan MV, Isbel NM, Johnson DW, et al. Disseminated pyogenic
moniae from pleural fluid and/or cerebrospinal fluid: report of four cases. Mycoplasma pneumoniae infection in a renal transplant recipient, detected
Scand J Infect Dis. 1987;19:521524. by broad-range polymerase chain reaction. Clin Infect Dis. 2004;39:
48. Narita M, Itakura O, Matsuzono Y, et al. Analysis of mycoplasmal central e98e99.
nervous system involvement by polymerase chain reaction. Pediatr Infect 78. Roifman CM, Rao CP, Lederman HM, et al. Increased susceptibility to
Dis J. 1995;14:236237. Mycoplasma infection in patients with hypogammaglobulinemia. Am J
49. Narita M, Matsuzono Y, Itakura O, et al. Survey of mycoplasmal bacte- Med. 1986;80:590594.
remia detected in children by polymerase chain reaction. Clin Infect Dis. 79. Lehtokoski-Lehtiniemi E, Koskiniemi ML. Mycoplasma pneumoniae
1996;23:522525. encephalitis: a severe entity in children. Pediatr Infect Dis J. 1989;8:
50. Padovan CS, Pfister HW, Bense S, et al. Detection of Mycoplasma pneu- 651653.
moniae DNA in cerebrospinal fluid of a patient with M. pneumoniae infec- 80. Domenech C, Leveque N, Lina B, et al. Role of Mycoplasma pneumoniae
tion-associated stroke. Clin Infect Dis. 2001;33:E119E121. in pediatric encephalitis. Eur J Clin Microbiol Infect Dis. 2009;28:9194.
51. Riedel K, Kempf VA, Bechtold A, et al. Acute disseminated encephalomy- 81. Daxboeck F, Blacky A, Seidl R, et al. Diagnosis, treatment, and prognosis
elitis (ADEM) due to Mycoplasma pneumoniae infection in an adolescent. of Mycoplasma pneumoniae childhood encephalitis: systematic review of
Infection. 2001;29:240242. 58 cases. J Child Neurol. 2004;19:865871.
52. Socan M, Ravnik I, Bencina D, et al. Neurological symptoms in patients 82. Heatwole CR, Berg MJ, Henry JC, et al. Extreme spindles: a distinc-
whose cerebrospinal fluid is culture- and/or polymerase chain reaction- tive EEG pattern in Mycoplasma pneumoniae encephalitis. Neurology.
positive for Mycoplasma pneumoniae. Clin Infect Dis. 2001;32:E31E35. 2005;64:10961097.
53. Tjhie JH, van de Putte EM, Haasnoot K, et al. Fatal encephalitis caused 83. Thomas NH, Collins JE, Robb SA, et al. Mycoplasma pneumoniae infec-
by Mycoplasma pneumoniae in a 9-year-old girl. Scand J Infect Dis. 1997; tion and neurological disease. Arch Dis Child. 1993;69:573576.
29:424425. 84. Rautonen J, Koskiniemi M, Vaheri A. Prognostic factors in childhood acute
54. Rhodes RH, Monastersky BT, Tyagi R, et al. Mycoplasmal cerebral vas- encephalitis. Pediatr Infect Dis J. 1991;10:441446.
culopathy in a lymphoma patient: presumptive evidence of Mycoplasma 85. Suzuki K, Matsubara S, Uchikata M, et al. A case of Mycoplasma pneu-
pneumoniae microvascular endothelial cell invasion in a brain biopsy. moniae pneumonia associated with meningoencephalitis [in Japanese].
J Neurol Sci. 2011;309:1825. Nihon Kyobu Shikkan Gakkai Zasshi. 1982;20:10841089.
55. Zu-Rhein GM, Powers JM. Correspondence regarding: a novel micro- 86. Narita M, Matsuzono Y, Togashi T, et al. DNA diagnosis of central ner-
angiopathy with endothelial cell atypia and multifocal white matter lesions: vous system infection by Mycoplasma pneumoniae. Pediatrics. 1992;90:
a direct mycopasmal infection? J Neuropathol Exp Neurol. 2011;70:1151. 250253.
56. Chin RP, Daft BM, Meteyer CU, et al. Meningoencephalitis in commer- 87. Ieven M, Demey H, Ursi D, et al. Fatal encephalitis caused by Mycoplasma
cial meat turkeys associated with Mycoplasma gallisepticum. Avian Dis. pneumoniae diagnosed by the polymerase chain reaction. Clin Infect Dis.
1991;35:986993. 1998;27:15521553.

Scheld_Ch25.indd 430 2/21/14 5:43 PM


Chapter 25: Mycoplasmal and Ureaplasmal Infections 431

88. Lerer RJ, Kalavsky SM. Central nervous system disease associated with 118. Tezer H, Kara A, Haliloglu G, et al. Mycoplasma pneumoniae-associated
Mycoplasma pneumoniae infection: report of five cases and review of the transverse myelitis with unexpected rapid response to macrolide therapy:
literature. Pediatrics. 1973;52:658668. a case report. Turk J Pediatr. 2008;50:585588.
89. Dorff B, Lind K. Two fatal cases of meningoencephalitis associated with 119. Yis U, Kurul SH, Cakmakci H, et al. Mycoplasma pneumoniae: nervous
Mycoplasma pneumoniae infection. Scand J Infect Dis. 1976;8:4951. system complications in childhood and review of the literature. Eur J
90. Weinblatt ME, Caplan ES. Fatal Mycoplasma pneumoniae encephalitis in Pediatr. 2008;167:973978.
an adult. Arch Neurol. 1980;37:321. 120. Weng WC, Peng SS, Wang SB, et al. Mycoplasma pneumoniaeassociated
91. Koskiniemi M, Vaheri A. Effect of measles, mumps, rubella vaccination transverse myelitis and rhabdomyolysis. Pediatr Neurol. 2009;40:128130.
on pattern of encephalitis in children. Lancet. 1989;1:3134. 121. Steer AC, Starr M, Kornberg AJ. Bickerstaff brainstem encephalitis
92. Bruch LA, Jefferson RJ, Pike MG, et al. Mycoplasma pneumoniae in- associated with Mycoplasma pneumoniae infection. J Child Neurol.
fection, meningoencephalitis, and hemophagocytosis. Pediatr Neurol. 2006;21:533534.
2001;25:6770. 122. Kikuchi M, Tagawa Y, Iwamoto H, et al. Bickerstaffs brainstem encepha-
93. Hulihan JF, Bebin EM, Westmoreland BF. Bilateral periodic lateralized litis associated with IgG anti-GQ1b antibody subsequent to Mycoplasma
epileptiform discharges in Mycoplasma encephalitis. Pediatr Neurol. pneumoniae infection: favorable response to immunoadsorption therapy.
1992;8:292294. J Child Neurol. 1997;12:403405.
94. Hodges GR, Fass RJ, Saslaw S. Central nervous system disease as- 123. Saitoh S, Wada T, Narita M, et al. Mycoplasma pneumoniae infection may
sociated with Mycoplasma pneumoniae infection. Arch Intern Med. cause striatal lesions leading to acute neurologic dysfunction. Neurology.
1972;130:277282. 1993;43:21502151.
95. Fernandez CV, Bortolussi R, Gordon K, et al. Mycoplasma pneumoniae 124. Sakoulas G. Brainstem and striatal encephalitis complicating Mycoplasma
infection associated with central nervous system complications. J Child pneumoniae pneumonia: possible benefit of intravenous immunoglobulin.
Neurol. 1993;8:2731. Pediatr Infect Dis J. 2001;20:543545.
96. Kumar RK. Mycoplasma encephalitisshould we be thinking more about 125. Al-Mateen M, Gibbs M, Dietrich R, et al. Encephalitis lethargica-
it? J Paediatr Child Health. 1998;34:305. like illness in a girl with Mycoplasma infection. Neurology. 1988;38:
97. de Lalibera IB, Silveira Gde A, Toma RK, et al. Meningoencephalitis 11551158.
associated with Mycoplasma pneumoniae. Einstein (Sao Paulo). 2012;10: 126. Sasaki M, Matsuda H, Omura I, et al. Transient seizure disappearance
100102. due to bilateral striatal necrosis in a patient with intractable epilepsy.
98. Simpkins A, Strickland SM, Oliver J, et al. Complete resolution of advanced Brain Dev. 2000;22:5055.
Mycoplasma pneumoniae encephalitis mimicking brain mass lesions: 127. Donovan MK, Lenn NJ. Postinfectious encephalomyelitis with localized
report of two pediatric cases and review of literature. Neuropathology. basal ganglia involvement. Pediatr Neurol. 1989;5:311313.
2012;32:9199. 128. Kim JS, Choi IS, Lee MC. Reversible parkinsonism and dystonia follow-
99. Lin JJ, Lin KL, Chiu CH, et al. Antineuronal antibodies and infectious ing probable Mycoplasma pneumoniae infection. Mov Disord. 1995;10:
pathogens in severe acute pediatric encephalitis [published online ahead 510512.
of print November 8, 2012]. J Child Neurol. 129. Green C, Riley DE. Treatment of dystonia in striatal necrosis caused by
100. Esposito S, Tagliabue C, Bosis S, et al. Levofloxacin for the treatment of Mycoplasma pneumoniae. Pediatr Neurol. 2002;26:318320.
Mycoplasma pneumoniae-associated meningoencephalitis in childhood. 130. Larsen PD, Crisp D. Acute bilateral striatal necrosis associated
Int J Antimicrob Agents. 2011;37:472475. with Mycoplasma pneumoniae infection. Pediatr Infect Dis J. 1996;15:
101. Carpenter TC. Corticosteroids in the treatment of severe Mycoplasma 11241126.
encephalitis in children. Crit Care Med. 2002;30:925927. 131. van Buiren M, Uhl M. Images in clinical medicine. Bilateral striatal
102. Cano-Sanchez A, Cano-Gracia H, Fernandez-Pardo J, et al. Aseptic men- necrosis associated with Mycoplasma pneumoniae infection. N Engl J
ingitis caused by Mycoplasma pneumoniae in a 19-year-old woman. Eur J Med. 2003;348:720.
Clin Microbiol Infect Dis. 1999;18:228229. 132. Termine C, Uggetti C, Veggiotti P, et al. Long-term follow-up of an
103. Gupta A, Kimber T, Crompton JL, et al. Acute disseminated encephalomy- adolescent who had bilateral striatal necrosis secondary to Mycoplasma
elitis secondary to Mycoplasma pneumoniae. Intern Med J. 2009;39:6869. pneumoniae infection. Brain Dev. 2005;27:6265.
104. Tan MJ, Chattophadyay AK, Griffiths PD, et al. Acute central and 133. Okumura K, Aizaki K, Tsuru T. Case of acute brainstem and striatal
peripheral demyelination associated with Mycoplasma pneumoniae. encephalopathy associated with Mycoplasma pneumoniae infection
Pediatr Neurol. 2003;29:239241. [in Japanese]. No To Hattatsu. 2011;43:471475.
105. Kumada S, Kusaka H, Okaniwa M, et al. Encephalomyelitis subsequent to 134. El Hafidi N, Allouch B, Benbrahim F, et al. Mycoplasma pneumoniae
Mycoplasma infection with elevated serum anti-Gal C antibody. Pediatr encephalitis associated with basal ganglia necrosis [in French]. Rev
Neurol. 1997;16:241244. Neurol. 2012;168:4952.
106. Kornips HM, Verhagen WI, Prick MJ. Acute disseminated encephalo- 135. Coon EA, Patterson MC. Teaching neuroimages: call it as you see it: evo-
myelitis probably related to a Mycoplasma pneumoniae infection. Clin lution of bilateral striatal necrosis. Neurology. 2012;78:e123.
Neurol Neurosurg. 1993;95:5963. 136. Fusco C, Bonini E, Soncini G, et al. Transient basal ganglia and thalamic
107. Fisher RS, Clark AW, Wolinsky JS, et al. Postinfectious leukoencepha- involvement following Mycoplasma pneumoniae infection associated
litis complicating Mycoplasma pneumoniae infection. Arch Neurol. with antiganglioside antibodies. J Child Neurol. 2010;25:102933.
1983;40:109113. 137. Zambrino CA, Zorzi G, Lanzi G, et al. Bilateral striatal necrosis associ-
108. Bouziri A, Khaldi A, Menif K, et al. Acute disseminated encephalomyeli- ated with Mycoplasma pneumoniae infection in an adolescent: clinical
tis associated with Mycoplasma pneumoniae infection in children: 2 case and neuroradiologic follow up. Mov Disord. 2000;15:10231026.
reports [in French]. Tunis Med. 2010;88:125128. 138. Noseda G, Harpey JP, Brandel JP, et al. Acute basal ganglia necrosis
109. Hagiwara H, Sakamoto S, Katsumata T, et al. Acute disseminated with favorable course during Mycoplasma encephalitis [in French]. Arch
encephalomyelitis developed after Mycoplasma pneumoniae infection Pediatr. 1996;3:11071110.
complicating subclinical measles infection. Intern Med. 2009;48:479483. 139. Brandel JP, Vidailhet M, Noseda G, et al. Mycoplasma pneumoniae
110. Aydin A, Atasever S, Cakmakci H. Acute disseminated encephalomy- postinfectious encephalomyelitis with bilateral striatal necrosis. Mov
elitis presenting with bilateral transient amaurosis. Pediatr Neurol. Disord. 1996;11:333336.
2005;32:6063. 140. Mariotti P, Nociti V, Stefanini MC, et al. Pneumonias link with the head
111. Tsiodras S, Kelesidis T, Kelesidis I, et al. Mycoplasma pneumoniae- and heart. Lancet. 2010;376:388.
associated myelitis: a comprehensive review. Eur J Neurol. 2006;13: 141. Ashtekar CS, Jaspan T, Thomas D, et al. Acute bilateral thalamic necro-
112124. sis in a child with Mycoplasma pneumoniae. Dev Med Child Neurol.
112. Behan PO, Feldman RG, Segerra JM, et al. Neurological aspects of myco- 2003;45:634637.
plasmal infection. Acta Neurol Scand. 1986;74:314322. 142. Wang HS. Acute bilateral thalamic necrosis in a child with Myco-
113. Pellegrini M, OBrien TJ, Hoy J, et al. Mycoplasma pneumoniae infec- plasma pneumoniae. Dev Med Child Neurol. 2004;46:214; author reply
tion associated with an acute brainstem syndrome. Acta Neurol Scand. 214215.
1996;93:203206. 143. Neilson DE, Adams MD, Orr CM, et al. Infection-triggered familial or
114. Pfausler B, Engelhardt K, Kampfl A, et al. Post-infectious central and recurrent cases of acute necrotizing encephalopathy caused by muta-
peripheral nervous system diseases complicating Mycoplasma pneu- tions in a component of the nuclear pore, RANBP2. Am J Hum Genet.
moniae infection. Report of three cases and review of the literature. Eur 2009;84:4451.
J Neurol. 2002;9:9396. 144. Chemli J, Ketata S, Dalhoumi A, et al. Opsoclonus-myoclonus syndrome
115. Ryan LJ, Bowman R, Zantek ND, et al. Use of therapeutic plasma associated with Mycoplasma pneumoniae infection [in French]. Arch
exchange in the management of acute hemorrhagic leukoencephalitis: Pediatr. 2007;14:10031006.
a case report and review of the literature. Transfusion. 2007;47:981986. 145. Huber BM, Strozzi S, Steinlin M, et al. Mycoplasma pneumoniae
116. Catalan M, Naccarato M, Grandi FC, et al. Acute hemorrhagic leukoen- associated opsoclonus-myoclonus syndrome in three cases. Eur J Pediatr.
cephalitis with atypical features. Neurol Sci. 2009;30:5557. 2010;169:441445.
117. Renault F, Arzimanoglou A, Rancurel G, et al. Hurst-type acute leuko- 146. Mesraoua B, Abbas M, DSouza A, et al. Adult opsoclonus-myoclonus
encephalitis with complete recovery in a child [in French]. Presse Med. syndrome following Mycoplasma pneumoniae infection with dramatic
1985;14:19591961. response to plasmapheresis. Acta Neurol Belg. 2011;111:136138.

Scheld_Ch25.indd 431 2/21/14 5:43 PM


432 Part III: Bacterial and Mycoplasmal Infections

147. Nunes JC, Bruscato AM, Walz R, et al. Opsoclonus-myoclonus syndrome 176. Beersma MF, Dirven K, van Dam AP, et al. Evaluation of 12 commer-
associated with Mycoplasma pneumoniae infection in an elderly patient. cial tests and the complement fixation test for Mycoplasma pneumoniae-
J Neurol Sci. 2011;305:147148. specific immunoglobulin G (IgG) and IgM antibodies, with PCR used as
148. Shiihara T, Takahashi Y. Correspondence: a further case of opsoclonus- the gold standard. J Clin Microbiol. 2005;43:22772285.
myoclonus syndrome associated with Mycoplasma pneumoniae infection. 177. Tsiodras S, Kelesidis I, Kelesidis T, et al. Central nervous system
Eur J Pediatr. 2010;169:639; author reply 641. manifestations of Mycoplasma pneumoniae infections. J Infect. 2005;51:
149. Hughes RA, Cornblath DR. Guillain-Barr syndrome. Lancet. 2005; 343354.
366:16531666. 178. Bitnun A, Ford-Jones E, Blaser S, et al. Mycoplasma pneumoniae ecepha-
150. Jacobs BC, Rothbarth PH, van der Meche FG, et al. The spectrum of litis. Semin Pediatr Infect Dis. 2003;14:96107.
antecedent infections in Guillain-Barr syndrome: a case-control study. 179. Schwab S, Junger E, Spranger M, et al. Craniectomy: an aggressive treat-
Neurology. 1998;51:11101115. ment approach in severe encephalitis. Neurology. 1997;48:412417.
151. Schessl J, Luther B, Kirschner J, et al. Infections and vaccinations pre- 180. Jaruratanasirikul S, Hortiwakul R, Tantisarasart T, et al. Distribution of
ceding childhood Guillain-Barr syndrome: a prospective study. Eur J azithromycin into brain tissue, cerebrospinal fluid, and aqueous humor of
Pediatr. 2006;165:605612. the eye. Antimicrob Agents Chemother. 1996;40:825826.
152. Sinha S, Prasad KN, Jain D, et al. Preceding infections and anti-ganglio- 181. Lutsar I, McCracken GH Jr, Friedland IR. Antibiotic pharmacodynamics in
side antibodies in patients with Guillain-Barr syndrome: a single centre cerebrospinal fluid. Clin Infect Dis. 1998;27:11171127, quiz 11281129.
prospective case-control study. Clin Microbiol Infect. 2007;13:334337. 182. Wolff M, Boutron L, Singlas E, et al. Penetration of ciprofloxacin into
153. Fleischhauer P, Huben U, Mertens H, et al. Demonstration of Mycoplasma cerebrospinal fluid of patients with bacterial meningitis. Antimicrob
pneumoniae in cerebrospinal fluid in acute polyneuritis [in German]. Agents Chemother. 1987;31:899902.
Dtsch Med Wochenschr. 1972;97:678682. 183. Yim CW, Flynn NM, Fitzgerald FT. Penetration of oral doxycycline into
154. Ponka A, von Bonsdorff M, Farkkila M. Polyradiculitis associated with the cerebrospinal fluid of patients with latent or neurosyphilis. Antimicrob
Mycoplasma pneumoniae reversed by plasma exchange. Br Med J (Clin Agents Chemother. 1985;28:347348.
Res Ed). 1983;286:475476. 184. Kramer PW, Griffith RS, Campbell RL. Antibiotic penetration of the
155. Meseguer MA, Aparicio M, Calvo A, et al. Mycoplasma pneumoniae anti- brain. A comparative study. J Neurosurg. 1969;31:295302.
gen detection in Guillain-Barr syndrome. Eur J Pediatr. 1998;157:1034. 185. Hsing J, Welgampola M, Kiernan MC. Reversible myeloradiculopathy
156. Lacroix LE, Galetto A, Haenggeli CA, et al. Delayed recognition of due to Mycoplasma pneumoniae. J Clin Neurosci. 2007;14:6164.
Guillain-Barr syndrome in a child: a misleading respiratory distress. 186. Csabi G, Komaromy H, Hollody K. Transverse myelitis as a rare, seri-
J Emerg Med. 2010;38:e59e61. ous complication of Mycoplasma pneumoniae infection. Pediatr Neurol.
157. Arakawa H, Yuhara Y, Todokoro M, et al. Immunoadsorption therapy 2009;41:312313.
for a child with Guillain-Barr syndrome subsequent to Mycoplasma in- 187. Ginestal RC, Plaza JF, Callejo JM, et al. Bilateral optic neuritis and
fection: a case study. Brain Dev. 2005;27:431433. Guillain-Barre syndrome following an acute Mycoplasma pneumoniae
158. Steele JC, Gladstone RM, Thanasophon S, et al. Acute cerebellar ataxia infection. J Neurol. 2004;251:767768.
and concomitant infection with Mycoplasma pneumoniae. J Pediatr. 188. Robertson JA, Stemke GW, Davis JW Jr, et al. Proposal of Ureaplasma
1972;80:467469. parvum sp. nov. and emended description of Ureaplasma urealyticum
159. Coleman RJ, Brown JS, Butler P, et al. Cerebellar syndrome with hydro- (Shepard et al. 1974) Robertson et al. 2001. Int J Syst Evol Microbiol.
cephalus due to Mycoplasma pneumoniae infection. Postgrad Med J. 2002;52:587597.
1990;66:554556. 189. Waites KB, Schelonka RL, Xiao L, et al. Congenital and opportunistic
160. Leonardi S, Pavone P, Rotolo N, et al. Stroke in two children with infections: Ureaplasma species and Mycoplasma hominis. Semin Fetal
Mycoplasma pneumoniae infection. A causal or casual relationship? Neonatal Med. 2009;14:190199.
Pediatr Infect Dis J. 2005;24:843845. 190. Al Masalma M, Armougom F, Scheld WM, et al. The expansion of the
161. Parker P, Puck J, Fernandez F. Cerebral infarction associated with microbiological spectrum of brain abscesses with use of multiple 16S
Mycoplasma pneumoniae. Pediatrics. 1981;67:373375. ribosomal DNA sequencing. Clin Infect Dis. 2009;48:11691178.
162. Visudhiphan P, Chiemchanya S, Sirinavin S. Internal carotid artery occlu- 191. Al Masalma M, Lonjon M, Richet H, et al. Metagenomic analysis of
sion associated with Mycoplasma pneumoniae infection. Pediatr Neurol. brain abscesses identifies specific bacterial associations. Clin Infect Dis.
1992;8:237239. 2012;54:202210.
163. Lee CY, Huang YY, Huang FL, et al. Mycoplasma pneumoniae-associated 192. Madoff S, Hooper DC. Nongenitourinary infections caused by Mycoplasma
cerebral infarction in a child. J Trop Pediatr. 2009;55:272275. hominis in adults. Rev Infect Dis. 1988;10:602613.
164. Dowd AB, Grace R, Rees WD. Cerebral infarction associated with 193. Hata A, Honda Y, Asada K, et al. Mycoplasma hominis meningitis in a
Mycoplasma pneumoniae infection. Lancet. 1987;2:567. neonate: case report and review. J Infect. 2008;57:338343.
165. Zu-Rhein GM, Lo SC, Hulette CM, et al. A novel cerebral microangiopathy 194. Alonso-Vega C, Wauters N, Vermeylen D, et al. A fatal case of Mycoplasma
with endothelial cell atypia and multifocal white matter lesions: a direct hominis meningoencephalitis in a full-term newborn. J Clin Microbiol.
mycoplasmal infection? J Neuropathol Exp Neurol. 2007;66:11001117. 1997;35:286287.
166. Arthur G, Margolis G. Mycoplasma-like structures in granulomatous 195. Gewitz M, Dinwiddie R, Rees L, et al. Mycoplasma hominis. A cause of
angiitis of the central nervous system. Case reports with light and electron neonatal meningitis. Arch Dis Child. 1979;54:231233.
microscopic studies. Arch Pathol Lab Med. 1977;101:382387. 196. Hjelm E, Jonsell G, Linglof T, et al. Meningitis in a newborn infant caused
167. Jacobs E. Serological diagnosis of Mycoplasma pneumoniae infections: by Mycoplasma hominis. Acta Paediatr Scand. 1980;69:415418.
a critical review of current procedures. Clin Infect Dis. 1993;17(suppl 1): 197. Kirk N, Kovar I. Mycoplasma hominis meningitis in a preterm infant.
S79S82. J Infect. 1987;15:109110.
168. Schwartz SB, Zhang M, Thacker WL, et al. Mycoplasma pneumoniae in 198. Knausz M, Niederland T, Dosa E, et al. Meningo-encephalitis in a neonate
encephalitis. In: 100th Meeting of the American Society of Microbiology. caused by maternal Mycoplasma hominis treated successfully with chlor-
Chicago: ASM; 1999. amphenicol. J Med Microbiol. 2002;51:187188.
169. Biberfeld G, Sterner G. A study of Mycoplasma pneumoniae infections in 199. McDonald JC, Moore DL. Mycoplasma hominis meningitis in a prema-
families. Scand J Infect Dis. 1969;1:3946. ture infant. Pediatr Infect Dis J. 1988;7:795798.
170. Nilsson AC, Bjorkman P, Persson K. Polymerase chain reaction is superior 200. Waites KB, Duffy LB, Crouse DT, et al. Mycoplasmal infections of cere-
to serology for the diagnosis of acute Mycoplasma pneumoniae infection brospinal fluid in newborn infants from a community hospital population.
and reveals a high rate of persistent infection. BMC Microbiol. 2008;8:93. Pediatr Infect Dis J. 1990;9:241245.
171. Thurman KA, Walter ND, Schwartz SB, et al. Comparison of laboratory 201. Waites KB, Rudd PT, Crouse DT, et al. Chronic Ureaplasma urealyticum
diagnostic procedures for detection of Mycoplasma pneumoniae in com- and Mycoplasma hominis infections of central nervous system in preterm
munity outbreaks. Clin Infect Dis. 2009;48:12441249. infants. Lancet. 1988;1:1721.
172. Waites KB, Thacker WL, Talkington DF. The value of culture and serol- 202. Watt KM, Massaro MM, Smith B, et al. Pharmacokinetics of moxifloxa-
ogy for the detection of Mycoplasma pneumoniae infections in the clini- cin in an infant with Mycoplasma hominis meningitis. Pediatr Infect
cal laboratory in the age of molecular diagnostics. Clin Microbiol Newsl. Dis J. 2012;31:197199.
2001;23:123129. 203. Wealthall SR. Mycoplasma meningitis in infants with spina bifida. Dev
173. Kenny GE, Kaiser GG, Cooney MK, et al. Diagnosis of Mycoplasma Med Child Neurol Suppl. 1975:117122.
pneumoniae pneumonia: sensitivities and specificities of serology with 204. Wolthers KC, Kornelisse RF, Platenkamp GJ, et al. A case of Mycoplasma
lipid antigen and isolation of the organism on soy peptone medium for hominis meningo-encephalitis in a full-term infant: rapid recovery
identification of infections. J Clin Microbiol. 1990;28:20872093. after start of treatment with ciprofloxacin. Eur J Pediatr. 2003;162:
174. Montagnani F, De Paolis F, Beghetto E, et al. Use of recombinant chimeric 514516.
antigens for the serodiagnosis of Mycoplasma pneumoniae infection. Eur 205. Meyer RD, Clough W. Extragenital Mycoplasma hominis infections in
J Clin Microbiol Infect Dis. 2010;29:13771386. adults: emphasis on immunosuppression. Clin Infect Dis. 1993;17(suppl. 1):
175. Tabassum I, Chaudhry R, Chourasia BK, et al. Identification of an S243S249.
N-terminal 27 kDa fragment of Mycoplasma pneumoniae P116 protein 206. McCarthy KL, Looke DF. Successful treatment of post-neurosurgical
as specific immunogen in M. pneumoniae infections. BMC Infect Dis. intracranial Mycoplasma hominis infection using gatifloxacin. J Infect.
2010;10:350. 2008;57:344346.

Scheld_Ch25.indd 432 2/21/14 5:43 PM


Chapter 25: Mycoplasmal and Ureaplasmal Infections 433

207. Chong JS, Tseung SY, Lam HS, et al. Successful treatment of multiple 229. Hammerschlag MR, Alpert S, Rosner I, et al. Microbiology of the vagina
subdural empyemata caused by Mycoplasma hominis in a newborn. in children: normal and potentially pathogenic organisms. Pediatrics.
Neonatology. 2009;95:179182. 1978;62:5762.
208. Rao RP, Ghanayem NS, Kaufman BA, et al. Mycoplasma hominis and 230. Foy H, Kenny G, Bor E, et al. Prevalence of Mycoplasma hominis
Ureaplasma species brain abscess in a neonate. Pediatr Infect Dis J. and Ureaplasma urealyticum (T strains) in urine of adolescents. J Clin
2002;21:10831085. Microbiol. 1975;2:226230.
209. Siber GR, Alpert S, Smith AL, et al. Neonatal central nervous system 231. Petrikkos GL, Hadjisoteriou M, Daikos GL. PCR versus culture in the
infection due to Mycoplasma hominis. J Pediatr. 1977;90:625627. detection of vaginal Ureaplasma urealyticum and Mycoplasma hominis.
210. Biran V, Dumitrescu AM, Doit C, et al. Ureaplasma parvum meningitis in Int J Gynaecol Obstet. 2007;97:202203.
a full-term newborn. Pediatr Infect Dis J. 2010;29:1154. 232. Ferandon C, Peuchant O, Janis C, et al. Development of a real-time PCR
211. Chung HY, Chung JW, Chun SH, et al. A case of erythromycin-resistant targeting the yidC gene for the detection of Mycoplasma hominis and
Ureaplasma urealyticum meningitis in a premature infant [in Korean]. comparison with quantitative culture. Clin Microbiol Infect. 2011;17:
Korean J Lab Med. 2007;27:4649. 155159.
212. Clifford V, Tebruegge M, Everest N, et al. Ureaplasma: pathogen or 233. Kenny GE, Cartwright FD. Susceptibilities of Mycoplasma hominis, M.
passenger in neonatal meningitis? Pediatr Infect Dis J. 2010;29:6064. pneumoniae, and Ureaplasma urealyticum to GAR-936, dalfopristin,
213. Hentschel J, Abele-Horn M, Peters J. Ureaplasma urealyticum in the cere- dirithromycin, evernimicin, gatifloxacin, linezolid, moxifloxacin, quinu-
brospinal fluid of a premature infant. Acta Paediatr. 1993;82:690693. pristin-dalfopristin, and telithromycin compared to their susceptibilities
214. Neal TJ, Roe MF, Shaw NJ. Spontaneously resolving Ureaplasma urea- to reference macrolides, tetracyclines, and quinolones. Antimicrob Agents
lyticum meningitis. Eur J Pediatr. 1994;153:342343. Chemother. 2001;45:26042608.
215. Ollikainen J, Hiekkaniemi H, Korppi M, et al. Ureaplasma urealyticum 234. Krausse R, Schubert S. In-vitro activities of tetracyclines, macrolides,
cultured from brain tissue of preterm twins who died of intraventricular fluoroquinolones and clindamycin against Mycoplasma hominis and
hemorrhage. Scand J Infect Dis. 1993;25:529531. Ureaplasma ssp. isolated in Germany over 20 years. Clin Microbiol Infect.
216. Ollikainen J, Hiekkaniemi H, Korppi M, et al. Ureaplasma urealyticum 2010;16:16491655.
infection associated with acute respiratory insufficiency and death in pre- 235. Sanchez PJ, Regan JA. Vertical transmission of Ureaplasma urealyticum in
mature infants. J Pediatr. 1993;122:756760. full term infants. Pediatr Infect Dis J. 1987;6:825828.
217. Sethi S, Sharma M, Narang A, et al. Isolation pattern and clinical outcome 236. Sanchez PJ, Regan JA. Vertical transmission of Ureaplasma urea-
of genital Mycoplasma in neonates from a tertiary care neonatal unit. lyticum from mothers to preterm infants. Pediatr Infect Dis J. 1990;9:
J Trop Pediatr. 1999;45:143145. 398401.
218. Shaw NJ, Pratt BC, Weindling AM. Ureaplasma and Mycoplasma 237. Syrogiannopoulos GA, Kapatais-Zoumbos K, Decavalas GO, et al.
infections of the central nervous system in preterm infants. Lancet. Ureaplasma urealyticum colonization of full term infants: perinatal acqui-
1989;2:15301531. sition and persistence during early infancy. Pediatr Infect Dis J. 1990;9:
219. Stahelin-Massik J, Levy F, Friderich P, et al. Meningitis caused by 236240.
Ureaplasma urealyticum in a full term neonate. Pediatr Infect Dis J. 1994; 238. Volgmann T, Ohlinger R, Panzig B. Ureaplasma urealyticum-harmless
13:419421. commensal or underestimated enemy of human reproduction? A review.
220. Viscardi RM, Hashmi N, Gross GW, et al. Incidence of invasive urea- Arch Gynecol Obstet. 2005;273:133139.
plasma in VLBW infants: relationship to severe intraventricular hemor- 239. Viscardi RM. Ureaplasma species: role in diseases of prematurity. Clin
rhage. J Perinatol. 2008;28:759765. Perinatol. 2010;37:393409.
221. Geissdorfer W, Sandner G, John S, et al. Ureaplasma urealyticum menin- 240. Lucke K, Kuster SP, Bertea M, et al. A deep sternal wound infection caused
gitis in an adult patient. J Clin Microbiol. 2008;46:11411143. by Ureaplasma urealyticum. J Med Microbiol. 2010;59:12541256.
222. McCormack WM, Almeida PC, Bailey PE, et al. Sexual activity and vagi- 241. Skoldenberg OG, Rysinska AD, Neander G, et al. Ureaplasma urealyti-
nal colonization with genital mycoplasmas. JAMA. 1972;221:13751377. cum infection in total hip arthroplasty leading to revision. J Arthroplasty.
223. McCormack WM, Lee YH, Zinner SH. Sexual experience and urethral 2010;25:1170.e111170.e13.
colonization with genital mycoplasmas. A study in normal men. Ann 242. Cordtz J, Jensen JS. Disseminated Ureaplasma urealyticum infection in a
Intern Med. 1973;78:696698. hypo-gammaglobulinaemic renal transplant patient. Scand J Infect Dis.
224. Tully JG, Taylor-Robinson D, Rose DL, et al. Evaluation of culture media 2006;38:11141117.
for the recovery of Mycoplasma hominis from the human urogenital tract. 243. Asmar BI, Andresen J, Brown WJ. Ureaplasma urealyticum arthritis
Sex Transm Dis. 1983;10:256260. and bacteremia in agammaglobulinemia. Pediatr Infect Dis J. 1998;17:
225. Keane FE, Thomas BJ, Gilroy CB, et al. The association of Mycoplasma 7376.
hominis, Ureaplasma urealyticum and Mycoplasma genitalium with bac- 244. Goldenberg RL, Andrews WW, Goepfert AR, et al. The Alabama
terial vaginosis: observations on heterosexual women and their male part- Preterm Birth Study: umbilical cord blood Ureaplasma urealyticum and
ners. Int J STD AIDS. 2000;11:356360. Mycoplasma hominis cultures in very preterm newborn infants. Am J
226. Schlicht MJ, Lovrich SD, Sartin JS, et al. High prevalence of genital Obstet Gynecol. 2008;198:43.e15.
mycoplasmas among sexually active young adults with urethritis or cer- 245. Singh J, Arrieta A, Lang DJ. Neonate with chronic meningitis and hydro-
vicitis symptoms in La Crosse, Wisconsin. J Clin Microbiol. 2004;42: cephalus. Pediatr Infect Dis J. 2003;22:10251026, 10301031.
46364640. 246. Garland SM, Murton LJ. Neonatal meningitis caused by Ureaplasma
227. Grattard F, Soleihac B, De Barbeyrac B, et al. Epidemiologic and molecu- urealyticum. Pediatr Infect Dis J. 1987;6:868870.
lar investigations of genital mycoplasmas from women and neonates at 247. Xie X, Zhang J. Trends in the rates of resistance of Ureaplasma urealyti-
delivery. Pediatr Infect Dis J. 1995;14:853858. cum to antibiotics and identification of the mutation site in the quinolone
228. Foy HM, Kenny GE, Levinsohn EM, et al. Acquisition of mycoplasmata resistance-determining region in Chinese patients. FEMS Microbiol Lett.
and T-strains during infancy. J Infect Dis. 1970;121:579587. 2006;259:181186.

Scheld_Ch25.indd 433 2/21/14 5:43 PM


CHAPTER 26 BARTONELLA INFECTIONS,
INCLUDING CAT-SCRATCH DISEASE
MICHAEL GILADI, MOSHE EPHROS, AND DAVID F. WELCH

The bacterial genus Bartonella is named for Dr. A. L. Barton, nonhuman wild and domestic mammals, including rodents,
who described the erythrocyte-adherent Bartonella bacilli- cervids, and cattle, without associated identifiable human ill-
formis in 1909. It is the etiologic agent of Oroya fever, an ness (13). Comparison of phylogenetic data, inferred mainly
acute bacteremic infection characterized by sepsis and hemo- from 16S rDNA, 16S-23S rRNA intergenic spacer, citrate
lysis, and of verruga peruana, principally a cutaneous nodular synthase and 60-kd heat shock protein gene sequences have
vascular eruption representing chronic infection. The previ- identified six evolutionary clusters within the genus Bartonella
ously suspected link between the two conditions was tragi- (14). Cat-scratch disease (CSD)causing B. henselae are classi-
cally confirmed in 1885 by Daniel Carrin, a medical student fied into two serotypes, Houston-1 and Marseille, which cor-
who injected himself with bloody material from a verruga and respond to two genotypes based on 16S rRNA gene sequences,
subsequently died of Oroya fever. This form of bartonellosis is genotype I and genotype II. The significance of this and other
thus known as Carrin disease and South American bartonel- classifications with respect to pathogenesis and clinical mani-
losis because it is limited to the Andean mountain regions of festations has not been established (15,16).
Peru, Ecuador, and Colombia. It affects the local population
and, rarely, travelers to these countries. Bartonellosis garnered
little attention outside its endemic zone in recent years until EPIDEMIOLOGY
related bacteria, then named Rochalimaea species, were found
to be important pathogens, primarily in patients with acquired Infections with B. bacilliformis are geographically limited to
immunodeficiency syndrome (AIDS). middle altitudes of the Andes mountains, probably because of
the distribution of species of the genus Lutzomyia (formerly
Phlebotomus), its sandfly vectors. B. quintana is globally dis-
TAXONOMY tributed; there have been reports of focal, but widely separated,
outbreaks of trench fever, also known as quintan or 5-day
The now supplanted genus Rochalimaea was formerly clas- fever. Outbreaks commonly have been associated with condi-
sified with Bartonella in the order Rickettsiales and con- tions of poor sanitation and personal hygiene that predispose
sisted of only two species, Rochalimaea (Rickettsia) vinsonii, to exposure to the human body louse Pediculus humanus, the
the Canadian vole agent, and Rochalimaea (Rickettsia) only identified vector of B. quintana. Although B. quintana
quintana, the agent of trench fever. The latter is a debilitating has also been identified in the human head louse P. humanus
but self-limited human illness so named after it affected many capitis, there is no strong evidence that head lice are vectors
military personnel in World War I. Except for sporadic out- of this organism between human hosts (17). Nonhuman verte-
breaks, trench fever had all but disappeared from the clinical brate reservoirs have not yet been identified for either B. bacil-
scene in recent decades. However, the 1990s saw the reemer- liformis or B. quintana.
gence of R. quintana as a pathogen of considerable interest Cats bacteremic with B. henselae constitute the major res-
(15), coincident with the discovery of two related species ervoir of this pathogen. B. henselae has been documented
that also cause human disease, originally named Rochalimaea to cause bacteremia (18,19) in seemingly healthy domestic
henselae and Rochalimaea elizabethae (69). cats, including some that have been specifically associated
Bartonella species are alphaproteobacteria, which also with bacillary angiomatosis (19) or typical CSD (18) in their
contains Afipia, Agrobacterium, and Brucella. Unlike mem- human contacts. B. henselae bacteremia has been globally
bers of the order Rickettsiales, Bartonella species have been reported among pet, impounded, or stray cats. Rates of
cultured on cell-free media. Sequencing of 16S ribosomal RNA bacteremia can vary and may be as high as 89% (1825).
(rRNA) genes to determine phylogenetic relationships among Other animals, particularly dogs, have been implicated as a
these organisms revealed high levels of relatedness between possible reservoir for B. henselae, but reports are anecdotal
B. bacilliformis and the former Rochalimaea species (10) and and evidence essentially circumstantial. Fleas and ticks are
confirmed that all of them are more closely related to Brucella arthropod vectors of B. henselae, based on epidemiologic as-
and Agrobacterium than to members of the rickettsiae. Based sociations (26,27) and reports of identification of B. henselae
on DNA hybridization and 16S rRNA similarity, the former by both culture and DNA amplification from cat-associated
Rochalimaea species were combined with Bartonella in 1993 fleas (18,19). Cat-to-cat transmission by infected fleas has
(11), and the members of the family Bartonellaceae were been shown to occur (21), although evidence of cat-to-human
removed from the order Rickettsiales. A further proposal was transmission by fleas is lacking. Like B. quintana, B. henselae
made in 1995 to merge into the genus Bartonella a number infection is globally endemic. However, regional variations in
of species of the genus Grahamella, which are intraeryth- the prevalence of either B. henselae or B. quintana may occur.
rocytic pathogens of rodents, birds, fish, and other animals Transmission to humans has been linked to cats by serologic
(12). Recently, an increasing number of Bartonella species and epidemiologic studies (27,28), its recovery from cases of
have been identified and characterized. Currently, the genus human lymphadenitis consistent with CSD (18,29), and the
Bartonella consists of at least 27 recognized species or subspe- identification of B. henselae DNA by polymerase chain reac-
cies, of which at least 13 have been recognized as confirmed or tion (PCR) in CSD lymphadenitis and other affected tissues
potential human pathogens. Others have been isolated from (3033).

434

Scheld_Ch26.indd 434 2/21/14 5:44 PM


Chapter 26: Bartonella Infections, Including Cat-Scratch Disease 435

B. quintana or B. henselae bacteremia in HIV-infected per-


OVERVIEW OF CLINICAL/ sons is often characterized by insidious development of fatigue,
PATHOLOGIC MANIFESTATIONS malaise, body aches, weight loss, progressively higher and lon-
ger recurring fevers, and sometimes headache. Hepatomegaly
may occur. Although there is evidence implicating Bartonella
Oroya Fever and Verruga Peruana species in some cases of HIV-associated encephalopathy, menin-
goencephalitis, and neuropsychiatric disease, lumbar puncture
Bartonella bacilliformis during acute bacteremia in HIV-infected persons can be unre-
vealing (6,9). Rarely, fever without localizing symptoms or signs
Oroya fever, the bacteremic illness of primary B. bacilliformis
in association with B. henselae bacteremia has been reported in
infection, develops 2 to 14 weeks (mean 3 weeks) after in-
immunocompetent patients (6,9). Aseptic meningitis concurrent
oculation by the sandfly Lutzomyia verrucarum (34). Bacteria
with bacteremia has been documented in an immunocompetent
invade blood vessel endothelium, proliferate, and upon re-
host (26). B. henselae bacteremia can evolve into long-term
entry into blood vessels replicate and destroy erythrocytes.
asymptomatic persistence (26).
Microvascular thrombosis results in end-organ ischemia. In its
Both B. quintana and B. henselae are considered important
milder form, the febrile illness often remits in a week. When
pathogens of endocarditis, accounting for approximately 3%
abrupt in onset, high fever, chills, diaphoresis, headache, and
of all patients with infective endocarditis, and a much larger
mental status changes are associated with a rapidly developing
proportion, up to 28%, of patients with culture-negative
severe hemolytic anemia (3538). Lymphadenopathy, throm-
endocarditis (1,4,4951). B. elizabethae, B. vinsonii subspe-
bocytopenia, severe myalgia and arthralgia, and complications
cies berkhoffii, B. alsatica, and B. koehlerae have rarely been
such as delirium, coma, dyspnea, and angina can occur during
isolated from patients with endocarditis (8,5254). Patients
this stage. Without antimicrobial treatment, mortality rates
with B. quintana endocarditis often have been homeless and
up to 40% to 80% have been reported (39,40); however, a
alcoholic, whereas patients with B. henselae endocarditis have
disease milder in severity and with a low (0.7%) case-fatality
commonly reported being in contact with a cat. The typical
rate may occur (41). Convalescence is associated with a de-
clinical presentation is that of subacute bacterial endocarditis,
cline of fever and disappearance of bacteria on blood smears
including neurologic manifestations such as stroke. In a ret-
as well as increased susceptibility to intercurrent opportunistic
rospective study of 101 patients with Bartonella endocarditis,
infections such as salmonellosis (42) or toxoplasmosis (43).
embolic phenomena were reported in 43% of patients. A sig-
Usually within months of acute infection, verruga peruana
nificant number of patients were afebrile at presentation, 12 of
may become evident. This late-stage manifestation of infec-
the 101 patients died, 2 relapsed, and 76 underwent valvular
tion is characterized by crops of nodular skin lesions; mucosal
surgery (55).
and internal lesions can also occur. Their histology typically
contains neovascular proliferation with occasional bacte-
ria evident in interstitial spaces. B. bacilliformis invasion of
endothelial cells, which was described by Rocha-Lima and Bacillary Angiomatosis and Peliosis
believed to be the etiology of cytoplasmic inclusions, is rare
(44). Verruga peruana lesions may develop at one site while
receding at another. They may persist for months to years and
Bartonella quintana and Bartonella henselae
eventually become fibrotic with involution. Asymptomatic Bacillary angiomatosis (BA), also termed epithelioid angio-
persistent bacteremia with B. bacilliformis infection can occur matosis or bacillary epithelioid angiomatosis, is a disorder of
in up to 15% of survivors of acute infection (45) who may neovascular proliferation originally described involving skin
serve as the organisms reservoir. and regional lymph nodes of HIV-infected persons (5658).
It has since been demonstrated to involve a variety of inter-
nal organs (5961), including the brain (62), and to occur
Bacteremic Illness and Endocarditis in other immunocompromised (59,63) and immunocompe-
tent hosts (6466). B. quintana infections have a predilection
Bartonella quintana, Bartonella henselae, for causing subcutaneous and deep soft tissue disease and
lytic bone lesions, whereas B. henselae infections are associ-
and Other Bartonella Species ated with lymph node disease and parenchymal peliosis of
The natural course of trench fever includes a spectrum of self- the liver and/or spleen. B. henselae and B. quintana (2,9,67)
limited clinical patterns (46). Incubation may span days to equally cause BA of the skin, the most common manifesta-
weeks before the typical sudden onset of fever. The febrile ill- tion of this illness. Risk factors for B. quintana infection
ness may be brief (lasting 4 to 5 days), prolonged (uninterrupted are low income, homelessness, and body louse infestation,
for 2 to 6 weeks), or most commonly, paroxysmal (three to five whereas B. henselae infection is associated with cat or cat
episodes, each of about 5 days). Fever may be accompanied fleas contact (67).
by other nonspecific symptoms and signs such as headache, Skin lesions often arise in crops, but their timing and gross
vertigo, retroorbital pain, conjunctival injection, nystagmus, appearance can vary. They can be remarkably similar to the
myalgia, arthralgia, hepatosplenomegaly, rash, leukocytosis, lesions of verruga peruana. However, most cases of BA have
and albuminuria. B. quintana has reemerged as a cause of been identified outside the region of endemic B. bacilliformis.
bacteremic illness (designated also as urban trench fever) in Thus, the most important differential diagnoses are Kaposi sar-
human immunodeficiency virus (HIV)uninfected homeless coma and pyogenic granuloma. Studies addressing the poten-
patients with chronic alcoholism (5,47,48). B. quintana was tial association between pyogenic granuloma and Bartonella
also identified in body lice from these patients. Clinical char- infection have resulted in conflicting results (68,69). The his-
acteristics include headache, sweats, severe leg pain, and low tologic distinction of BA from other neovascular tumors has
platelet counts. Many bacteremic patients were afebrile and been clearly described (70,71).
some were asymptomatic. Chronic bacteremia, as indicated by Bacillary peliosis (BP), originally described involving the
positive blood cultures up to 78 weeks, and intermittent bac- liver and sometimes spleen in HIV-infected persons (72), has
teremia were found to occur. since been identified in other immunosuppressed persons

Scheld_Ch26.indd 435 2/21/14 5:44 PM


436 Part III: Bacterial and Mycoplasmal Infections

and found to involve lymph nodes as well (59,73). Involved


organs contain numerous blood-filled, partially endothelial NEUROLOGIC MANIFESTATIONS
celllined cystic structures and surrounding clumps of bacilli OF BARTONELLA INFECTIONS
(identified by Warthin-Starry silver staining) in the midst of
inflammatory cells.
Associated with Oroya Fever
Cat-Scratch Disease Acute B. bacilliformis infection (Oroya fever) has long been
recognized to have associated neurologic manifestations (34).
Acute onset of severe headache is usually coincident with
Bartonella henselae (Possibly Bartonella clarridgeiae, onset of fever and development of hemolysis. Characteristics
Bartonella quintana, and Afipia felis) of the meningoencephalitis that occurs in about 1 in 10 cases
B. henselae is the major etiologic agent of CSD (18,19,2832). include diffuse neurologic impairment that may result in sei-
B. clarridgeiae, B. quintana, and Afipia felis have rarely been asso- zures, hallucinations, delirium, and/or reduced consciousness,
ciated with CSD in humans (7476). The various manifestations which can progress to obtundation and coma. Diminished
that comprise CSD have been recognized over the past 100 years, level of consciousness is associated with a poorer prognosis.
but the syndrome per se was not really defined until 1950 (77). Meningeal findings without encephalitis also can occur and
In typical CSD (about 90% of cases), a cutaneous papule or vice versa. Less commonly, localized findings in the form of
pustule usually develops within a week after an animal contact cranial or spinal nerve palsies occur, with the latter sometimes
(more commonly a kitten) at a site of inoculation (usually a causing a meningomyelitis with flaccid or spastic paralysis.
scratch or bite) (7880). Regional adenopathy (mostly involv- Cerebrospinal fluid (CSF) protein elevation and mild mono-
ing head, neck, or upper extremity) develops in 1 to 7 weeks nuclear leukocytosis can occur, and bacteria may be identi-
(Fig. 26.1). About one third to one half of patients have fever, fied within these leukocytes. Peripheral nerve palsies that
and about one sixth develop lymph node suppuration. The occur during the later eruptive verruga stage are usually due to
histopathology of nodes includes a mixture of nonspecific in- granulomatous inflammatory lesions within peripheral nerves;
flammatory reactions including granulomas and stellate necro- such impediments are usually chronic but associated with
sis. Bacilli may be demonstrable by Warthin-Starry staining. gradual resolution.
Atypical CSD (about 10%) occurs as extranodal or compli- The often fatal course of Oroya fever has allowed his-
cated disease in the absence or presence of lymphadenopathy topathologic correlation with clinically evident neurologic
and includes Parinaud oculoglandular syndrome, encephalop- manifestations (92,93). Most of the neurologic manifesta-
athy, neuroretinitis and other neurologic syndromes, fever of tions appear to be the result of the vascular endothelial cell
unknown origin, hepatic and splenic abscesses, granulomatous damage that develops. In the leptomeninges, capillary and
hepatitis, debilitating myalgia, arthritis or arthralgia (affect- venous congestion and thrombosis are common, associated
ing mostly females older than age 20 years), osteomyelitis and with microhemorrhages, adventitial proliferation, perivascu-
other musculoskeletal manifestations, and erythema nodosum lar edema, and rarely, formation of new microvasculature.
(79,8184). Other manifestations and syndromes (e.g., pneu- As a consequence of profound hemolytic anemia (hemoglo-
monitis, myocarditis, and thrombocytopenia) have also been bin concentration 4 g/L has been reported in Oroya fever)
associated with CSD (8588). and microvascular thrombosis, resulting in ischemia, there
In most cases, whether typical or atypical, spontaneous can be subacute neuronal degeneration. Reactive glial prolif-
resolution occurs in 2 to 4 months. The prolonged course eration is usually diffuse but in some cases may be nodular.
of CSD lymphadenopathy, which is often accompanied by Occasionally, granuloma-like nodules composed of microglial
fever, night sweats, weight loss, and liver or spleen involve- cells and histiocytes, named verrucomas, are found in the cho-
ment, may resemble lymphoma or other malignant processes. roid plexus, ependyma, and brain parenchyma. Ultimately, the
Consequently, this may lead to unnecessary, extensive, costly, meningoencephalitis associated with Carrin disease appears
and sometimes invasive diagnostic procedures (8991). to be more a consequence of the damage done to the hosts
microvasculature, complicated by the associated profound
anemia, than the result of a primary neurotropic affinity on
the part of B. bacilliformis.

Associated with Trench Fever


and Other Manifestations of
Bartonella quintana Infection
Although trench fever is often associated with headache, specific
neurologic manifestations of this form of B. quintana infection
are uncommon. Few cases of B. quintana infection with distinct
central nervous system (CNS) pathology have been described.
A 19-year-old HIV-uninfected patient with hypogammaglobu-
linemia presented with fever, left hemiparesis, slurred speech,
urinary incontinence, blurred vision, and behavioral changes,
which developed over 2 months due to a necrotizing granulo-
matous process involving the right thalamus and surrounding
tissues. B. quintana was identified in brain tissue, bone marrow,
and serum specimens from this patient by PCR and nucleotide
FIGURE 26.1 Cat-scratch disease lymphadenopathy in a 10-year-old sequencing. B. quintana DNA was also amplified from the CSF
previously healthy male. of an 8-year-old immunocompetent child with encephalitis and

Scheld_Ch26.indd 436 2/21/14 5:44 PM


Chapter 26: Bartonella Infections, Including Cat-Scratch Disease 437

axillary adenitis (94). A previously healthy 16-month-old girl to occur in its absence. Persistent generalized headache is a
was admitted to the intensive care unit with encephalopathy common part of the history, but fever is an inconsistent find-
complicated by Guillain-Barr syndrome and hydrocephalus, ing. Patients may become restless and combative. Nearly half
which necessitated placement of a ventriculoperitoneal shunt. of patients develop seizures, which may range from focal to
She had serologic and molecular evidence of central nervous generalized and from brief and self-limited to status epilep-
system infection by B. quintana (95). ticus. Short-term anticonvulsant therapy may be required,
as may be supportive therapy in the face of obtundation or
coma. Nuchal rigidity, pathologic reflexes, or pupillary dila-
Associated with Cat-Scratch Disease tion may be present transiently. Neurologic deficits such as
aphasia, cranial nerve palsy, paresis, hemiplegia, and ataxia
The most commonly recognized neurologic manifestations are usually self-limited, although time to resolution may span
associated with B. henselae infection are those of CSD, pre- weeks to months to as long as a year. Persistence of intel-
dominantly encephalopathy and neuroretinitis. Isolated cranial lectual impairment, ataxia, and seizures have been reported
and peripheral neuropathies (e.g., facial palsy), polyneuropa- (81,112,118,125,128), as well as rare cases of death due to
thy, transverse myelitis, and other manifestations uncommonly CSD meningoencephalitis in two previously healthy children,
occur (96145). CSD vertebral osteomyelitis may rarely present aged 4 and 6 years (157,158).
with neurologic complications, including intraspinal extension Laboratory studies in the setting of CSD encephalopathy do
(146150). Encephalopathy probably occurs in 2% to 4% of not add specific positive diagnostic findings to the clinical pic-
all recognized CSD cases, although estimates range from 1% ture, but they serve to exclude other processes. CSF measure-
to 7% (115). Extrapolating from an estimated U.S. CSD case ments fit no consistent pattern, except that hypoglycorrhachia
rate of 9.3 per year per 100,000 population, 2% to 4% would is rare. Elevation of CSF protein concentration and pleocytosis
represent between 500 and 1,000 annual CSD encephalopathy with lymphocytic predominance occur in only about one third
cases in the United States. The California Encephalitis Project of patients (but not necessarily in the same patients) (151).
reported Bartonella species as the causative agent in 7 (2%) of Peripheral blood leukocytosis occurs as well in only about one
334 patients with encephalitis, making it the most common bac- third of patients. CSF cultures have been consistently negative.
teria associated with encephalitis (151). In contrast, Bartonella Studies of the brain with computed tomography (CT) and/
cases were found neither among 203 patients with encephali- or magnetic resonance imaging (MRI) usually show no abnor-
tis in a multicenter prospective study from England nor among malities. Transient nonspecific abnormalities are occasionally
253 patients in a national prospective study conducted in France identified, but a few cases of persistent structural abnormali-
(152,153). However, the diagnosis of Bartonella encephalitis in ties have been reported (128,159). Electroencephalography
the latter study may have been underrepresented because the during the acute phase of CSD encephalopathy commonly
authors excluded survivors hospitalized for more than 5 days reveals diffuse slowing, yet another nonspecific feature that
because these patients were assumed to have aseptic meningi- resolves with clinical recovery. Brain biopsy is usually not in-
tis rather than encephalitis. Patients with encephalitis due to dicated because of the self-limited nature of CSD encephalopa-
Bartonella species may have a fulminant presentation but often thy, and thus little is known about the histologic correlates
recover fully within several days after onset and thus could have of the clinical manifestations. At autopsy of a fatality due to
been excluded from the study (154). Such diagnoses can easily CSD encephalomeningitis, there was marked cerebral edema
be overlooked if the clinician fails to obtain an adequate history. with no gross evidence of acute meningitis. Microscopic ex-
With domestic cats representing the single largest category of amination revealed multiple granulomatous lesions, meningi-
companion animals in the United States, the importance of an tis, and encephalitis. Warthin-Starry silver stain of the brain
accurate history regarding animal exposure cannot be overem- and liver revealed pleomorphic rod-shaped bacilli consistent
phasized when evaluating a patient with findings consistent with with B. henselae infection. Analysis of brain tissue with PCR
one of these syndromes. A common pitfall in history taking is to confirmed the presence of B. henselae DNA (157). Histologic
inquire about a cat scratch or a cat bite rather than cat contact, examination of the second fatality showed extensive diffuse
as a significant proportion of CSD patients report cat contact perivascular lymphocytic infiltrates with microglial nodules
without injury. Though less established, one must also keep an scattered throughout the frontal, parietal, and occipital lobes
open mind to the possibility of transmission of B. henselae from and the pons. In some foci, the nodules appeared vaguely
other animals such as dogs. granulomatous (158). Biopsy of concurrent lymphadenopathy,
CSD encephalopathy remains predominantly a clinical when done, reveals features typical of CSD.
diagnosis, now subject to laboratory confirmation by tech- The neuroretinitis associated with CSD (96,108,116,117,
niques described later in this chapter (predominantly anti- 120,121,124,129,133,160164) has been confirmed by se-
body testing). Adolescents and adults may represent a greater rology and culture to be related to B. henselae infection.
proportion of cases of CSD encephalopathy than they do of Neuroretinitis in association with B. henselae bacteremia (96),
CSD overall (81). Encephalitis was also reported to be more aseptic meningitis (165), and encephalopathy (166) have been
common in elderly patients (older than 60 years of age) with reported in patients with CSD. Chorioretinitis and multiple
CSD than in younger patients (155). The pathogenesis of CSD hypodense areas within the spleen and liver parenchyma have
encephalopathy and other CNS manifestations associated been described in a 10-year-old previously healthy boy several
with CSD remains unclear. Whether these rare complications weeks after a cat scratch (167). Although long-term prognosis
are attributable to direct invasion of the CNS by B. henselae or is usually good, some individuals may develop mild postin-
to other mechanisms such as vasculitis or immune response is fectious optic neuropathy, and few may develop permanent
unknown. B. henselae has been shown to infect feline microg- visual disturbances. Vitrectomy is only rarely indicated. With
lial cells in vitro and survive intracellularly for up to 4 weeks; the refinement of techniques for identification of Bartonella
however, no ultrastructural abnormalities were identified infection, diagnostic accuracy has improved, broadening
within infected brain cells by electron microscopy (156). the spectrum of CSD-associated retinal manifestations and
In most patients, encephalopathy usually follows lymph- identifying new Bartonella species as possible pathogens
adenopathy, by a period of days up to 2 months, although it in neuroretinitis. B. grahamii was identified by PCR ampli-
has also been reported to precede lymph node involvement or fication and sequence analysis in the intraocular fluid of an

Scheld_Ch26.indd 437 2/21/14 5:44 PM


438 Part III: Bacterial and Mycoplasmal Infections

Associated with HIV Infection


Intracerebral bacillary angiomatosis was first recognized in
1990 (62) in a man with AIDS. In 1994, antibody and DNA
amplification evidence of Bartonella infection was reported
in the setting of neurologic manifestations complicating HIV
infection (182), and epidemiologic studies have confirmed an
association between the presence of serum anti-Bartonella anti-
bodies and increased risk of development of neuropsychologic
decline or dementia over 5 years. An estimated 4% of new
cases of HIV-associated dementia or neuropsychologic decline
might be ascribed to Bartonella infections and therefore are
potentially treatable with antibiotics. Subsequent small num-
bers of case reports have added anecdotal evidence suggesting
the potential utility of antimicrobial therapy in reversing
Bartonella-associated neuropsychiatric abnormalities.

FIGURE 26.2 Left eye neuroretinitis in a previously healthy 35-year-old


male cat owner with cat-scratch disease. Disc edema with macular exu- Chronic Neurologic and
dates in a star formation (macular star; arrow) are seen. Macular star Neurocognitive Manifestations
retinopathy is thought to result from leakage of lipid-containing exudate
from capillaries in the optic head with subsequent extension into the A few recent studies, originating mostly from one group of
subretinal space and macular region. (Courtesy Ehud Zamir, MD.) investigators, reported the detection of various Bartonella
species, including B. henselae, B. vinsonii subsp. berkhoffii,
B. koehlerae, Candidatus B. melophagi, or coinfection with
HIV-seronegative patient with bilateral neuroretinitis and be- more than one Bartonella spp. in blood samples, using a novel
havioral changes, and B. elizabethae infection was diagnosed enrichment blood culture technique, followed by PCR and
serologically in another patient with neuroretinitis (168,169). DNA sequencing. Patients were apparently immunocompe-
The typical clinical scenario of CSD neuroretinitis, a pro- tent individuals who presented with various chronic neuro-
cess first described as Leber idiopathic stellate retinopathy logic or neurocognitive syndromes, including seizures, ataxia,
(116,170), is that of painless, fairly sudden loss of visual acuity, memory loss, tremors, fatigue, insomnia, headache, arthralgia,
usually unilaterally, and sometimes preceded by an influenza- myalgia, hallucinations, and other symptoms. Many individu-
like syndrome or development of regional lymphadenopathy. als had extensive arthropod and animal exposure. It has been
Neuroretinitis is characterized by papilledema often associated suggested that because the duration of illness in these patients
with macular exudates in a star formation (Fig. 26.2). In a ret- ranged from months to many years, these Bartonella species
rospective study among 24 patients with CSD with 35 affected may induce a chronic intravascular, persistent, or relapsing
eyes, isolated foci of retinitis or choroiditis were the most com- infection. More studies are needed to evaluate the role of these
mon ocular manifestation identified in 83% of eyes and 83% pathogens in patients with chronic neurologic and neurocog-
of patients. Optic disk swelling was the second most common nitive dysfunction (183188).
finding (46% of eyes, 63% of patients), followed by a macu-
lar star (43% of eyes, 63% of patients) and vascular-occlusive
events (14% of eyes, 21% of patients). Final visual acuity was Infections Caused by Other Species
20/25 or better in 26 (74%) of 35 eyes and was similar in both
treated and untreated patients (161). Optic disk edema asso- B. vinsonii subsp. arupensis was isolated from the blood of
ciated with peripapillary serous retinal detachment has been a 62-year-old cattle rancher who was admitted to a hospital
described as an early sign of ocular CSD. The typical macular with acute onset of confusion, emotional liability, difficulty in
star may or may not follow these early manifestations (171). walking, facial numbness, slurred speech, diplopia, headache,
Other types of ocular CSD manifestations include optic neu- and myalgias. He was discharged 7 days later with a signifi-
ritis, anterior uveitis, panuveitis, vitreitis, pars planitis, focal cant improvement in neurologic symptoms (189).
retinal vasculitis, retinal white spot syndrome, branch retinal A case of meningitis attributed to Bartonella washoensis,
arteriolar or venular occlusions, central retinal artery and vein isolated from a patients blood using routine blood culture sys-
occlusion, focal choroiditis, vitreous and retinal hemorrhages, tem, was described in a 47-year-old previously healthy woman
and a process associated with peripapillary angiomatosis with exposure to pet and farm animals. Oropsylla montana
(172181). The pathophysiology of neuroretinitis is thought fleas were implicated as the vector for disease transmission in
to be leakage of lipid-containing exudate from capillaries in the this case (190).
optic head with subsequent extension into the subretinal space
and macular region. This type of process has been recognized
as a secondary phenomenon in most circumstances, occur- LABORATORY CONFIRMATION
ring in association with traumatic injuries of the eye or brain, OF CLINICAL DIAGNOSIS
ocular vascular disturbances, toxins, autoimmune states (e.g.,
Behet syndrome), or a variety of infections (e.g., influenza- The laboratory diagnosis of Bartonella-associated diseases can
like syndromes, syphilis, leptospirosis, tularemia, tuberculosis, be achieved through modified conventional bacteriologic cul-
psittacosis, endemic mycoses, and parasites). Thus, although ture methods, co-culture with endothelial cells, immunosero-
this process can be considered characteristic of CSD neuroreti- logic or immunocytochemical means, or DNA amplification.
nitis, it is not pathognomonic and many other causes must be These approaches are described in current diagnostic micro-
included in the differential diagnosis. biology references (191). Serologic testing has become the

Scheld_Ch26.indd 438 2/21/14 5:44 PM


Chapter 26: Bartonella Infections, Including Cat-Scratch Disease 439

mainstay of diagnosis, particularly when the involved tissue is diagnosis, replacing the skin test that was poorly standardized
less accessible for biopsy sampling such as CSD patients with and carried a potential risk for transmission of infectious agents.
CNS infection. Early lymph node biopsy or fine-needle aspira- An immunofluorescence assay (IFA) and several enzyme immu-
tion (before spontaneous resolution of lymphadenitis occurs) noassays (EIAs) have been described for B. henselae and B. quin-
for histopathology, PCR, and culture should be encouraged tana detection. They have been used primarily to demonstrate
in patients with serious complications such as encephalitis or anti-Bartonella antibodies in persons with CSD (28,129,200)
neuroretinitis, when CSD is a likely diagnosis and B. henselae and in some cases of HIV-associated aseptic meningitis, en-
serology is negative or equivocal. cephalopathy, or neuropsychiatric disease (96,182). The IFA
described by Regnery et al. (28) in 1992 generally performs
well, but it was not designed for detecting immunoglobulin M
Detection and Identification of the Agent (IgM) antibodies. In studies in which this assay was performed
at the U.S. Centers for Disease Control and Prevention, with
Bartonella species usually do not grow under the conditions reciprocal titers of more than 64 as the cutoff value, both sen-
used for standard bacteriologic cultures. If culture is attempted, sitivity (84% to 95%) and specificity (94% to 98%) were high.
freshly prepared media provide optimal recovery. Heart infu- It has been less consistent in studies performed in Europe (201).
sion agar with 5% to 10% defibrinated rabbit or horse blood A high seroprevalence of antiB. henselae immunoglobulin G
supports better growth of most strains than chocolate or 5% (IgG) in Europe due to exposure to nonB. henselae species
sheep blood agars. Plates sealed after 24 hours of incubation may result in inferior performance of the IFA compared with
to preserve moisture content usually can be incubated up to the United States. EIA studies in some HIV-infected persons
30 days without notable deterioration. Even when these tech- with encephalopathy or neuropsychiatric findings have demon-
niques are strictly applied, recovery of B. henselae from lymph strated antibodies reactive with a formalin-fixed whole bacterial
nodes and other specimens of patients with CSD is extremely cell antigen preparation of B. henselae but have not satisfacto-
rare. Moderate success in the recovery of isolates has been rily demonstrated B. henselae specificity (182). An EIA using
achieved using alternate techniques (192,193). The combina- N-lauroyl-sarcosineinsoluble outer membrane antigen from
tion of enrichment culture and PCR amplification may opti- agar-grown B. henselae to test sera from 84 patients with CSD
mally detect Bartonella species with respect to sensitivity and a defined by PCR (82 patients) or skin test (6 patients) determined
clinically relevant time frame, although this approach is gener- the EIA sensitivity to be 75% for antiB. henselae IgG alone,
ally not available in the majority of clinical laboratories. 48% for IgM alone, and 85% overall when positive IgG, IgM,
Colonies of Bartonella species are sticky, autoadherent, and or both were accepted as diagnostic. EIA specificity was 98%
of two morphologic types: (a) irregular, raised, whitish, rough, (200). The same EIA was used to study antibody kinetics in pa-
and dry appearing or (b) smaller, circular, tan, and moist tients with CSD. AntiB. henselae IgM remained positive for
appearing. Both types are often present in the same culture. 3 months or less and its presence indicated acute disease. IgG
The degree of colonial heterogeneity varies by species, with titers also declined over time but may last for longer than 2 years.
B. henselae typically having a greater proportion of rough colo- Serologic cross reactivity is among the limitations of both
nies than B. quintana. Repeated subcultures cause most strains IFA and EIA. Neither adequately discriminates between anti
of B. henselae to revert to smooth cultures. Gram stain of a col- B. henselae and antiB. quintana antibodies. Cross reactivity
ony reveals small, gram-negative, slightly curved rods (which has also been demonstrated between Bartonella and others in-
may mimic Haemophilus, Campylobacter, or Helicobacter), cluding Coxiella burnetii and Chlamydia species. Data regard-
and a wet mount usually demonstrates twitching motility. ing the clinical significance of such cross reactivity are limited,
B. bacilliformis and B. clarridgeiae possess flagella, whereas and this represents a potential concern because all of these
B. henselae, B. elizabethae, and to lesser extent, B. quintana microorganisms are causative agents of endocarditis and may
have twitching motility believed due to pili. Presumptive identi- have similar clinical presentations.
fication of B. henselae or B. quintana can be made on the basis
of these features, plus a lengthy (7-day) period of incubation
before appearance, negative catalase and oxidase reactions, Treatment of Bartonella-Related
and absence of acid production from carbohydrates. Neurologic Manifestations
Confirmatory identification is usually by referral to a labo-
ratory experienced with Bartonella species. Newer methods Antibiotic regimens for Bartonella infections have been de-
have been applied to identification of Bartonella species in- termined empirically, based on clinical experience, as well as
cluding mass spectrometry (194), in addition to PCR-based published case reports and clinical studies, which are mostly
and DNA hybridization techniques that can be used to dis- uncontrolled with limited follow-up (202). One of the most
tinguish species and for direct detection in clinical material intriguing aspects of nonB. bacilliformis infections is the
(30,31,90,195199). DNA fragments of B. henselae can be clinical observation that antimicrobial therapy seems to have
amplified from various clinical specimens including fresh a much greater impact on immunocompromised patients with
lymph nodes and other tissues (including brain), CSF, freshly systemic disease (e.g., patients with AIDS with bacteremia
aspirated pus, minute amounts of tissue obtained from lymph and BA) than on immunocompetent patients with localized
node fine-needle aspiration, and paraffin-embedded material. disease (e.g., patients with CSD with regional lymphadenitis).
Molecular subtyping of strains can be performed using PCR- This perhaps reflects the different pathologic processes in-
based sequence analysis (15) or restriction fragment length volved. The recommended therapy for acute B. bacilliformis
polymorphism, and repetitive extragenic palindromic PCR. infection, Oroya fever, is either chloramphenicol (0.5 g PO/IV
four times a day for 2 weeks) plus another antibiotic, (prefer-
ably a -lactam), or ciprofloxacin 0.5 g twice a day for 10
Serology days. CNS involvement with impaired consciousness neces-
sitates parenteral therapy. Because of ease of administration,
Because culture of Bartonella species remains technically dif- low cost, and observed clinical effectiveness, the initial ther-
ficult with a low success rate, alternative means of diagnosis apy of choice for uncomplicated bacteremia and BA caused by
are important. Serology is the most commonly used test for B. henselae or B. quintana in immunocompromised patients

Scheld_Ch26.indd 439 2/21/14 5:44 PM


440 Part III: Bacterial and Mycoplasmal Infections

is oral erythromycin (e.g., 0.5 g four times a day as stearate). of antimicrobials in the treatment of other manifestations of
Other macrolides, doxycycline (100 mg twice daily), or other CSD, and because of the severe clinical manifestations of this
tetracyclines may serve as alternatives (67,202,203). complication, antimicrobial therapy is prudent. Doxycycline
The role of antimicrobial therapy for CSD-associated neu- with or without addition of rifampin seems to be a reason-
roretinitis is controversial. In a small retrospective case series, able choice in these cases (203). Dramatic clinical improve-
doxycycline and rifampin appeared to shorten the course of ment following treatment with high-dose steroids has been
disease and hasten visual recovery in seven patients with CSD described in a 4-year-old previously healthy child with CSD
neuroretinitis compared to historic cases. The demonstration encephalopathy. Another 12-year-old child with CSD and
of B. henselae bacteremia associated with neuroretinitis adds brainstem encephalopathy with basal ganglia impairment who
weight to the argument for treatment with antibiotics (96,160). was admitted with profound coma recovered after treatment
There is no definite evidence of the utility of antibiotic therapy with high-dose methylprednisolone and antibiotics; however,
in shortening or altering the course of CSD encephalopathy. because of the self-limited nature of this complication, the role
However, because of anecdotal reports of apparent efficacy of steroids in this setting is difficult to evaluate (204,205).

References
1. Drancourt M, Mainardi JL, Brouqui P, et al. Bartonella (Rochalimaea) quin- 20. Chomel BB, Abbott RC, Kasten RW, et al. Bartonella henselae prevalence
tana endocarditis in three homeless men. N Engl J Med. 1995;332:419423. in domestic cats in California: risk factors and association between bactere-
2. Koehler JE, Quinn FD, Berger TG, et al. Isolation of Rochalimaea species mia and antibody titers. J Clin Microbiol. 1995;33:24452450.
from cutaneous and osseous lesions of bacillary angiomatosis. N Engl J 21. Chomel BB, Kasten RW, Floyd-Hawkins K, et al. Experimental transmis-
Med. 1992;327:16251631. sion of Bartonella henselae by the cat flea. J Clin Microbiol. 1996;34:
3. Larson AM, Dougherty MJ, Nowowiejski DJ, et al. Detection of Bartonella 19521956.
(Rochalimaea) quintana by routine acridine orange staining of broth blood 22. Brunetti E, Fabbi M, Ferraioli G, et al. Cat-scratch disease in Northern Italy:
cultures. J Clin Microbiol. 1994;32:1492496. atypical clinical manifestations in humans and prevalence of Bartonella
4. Spach DH, Callis KP, Paauw DS, et al. Endocarditis caused by Rochalimaea infection in cats. Eur J Clin Microbiol Infect Dis. 2013;32:531534.
quintana in a patient infected with human immunodeficiency virus. J Clin 23. Azzag N, Haddad N, Durand B, et al. Population structure of Bartonella
Microbiol. 1993;31:692694. henselae in Algerian urban stray cats. PloS One. 2012;7:e43621.
5. Spach DH, Kanter AS, Dougherty MJ, et al. Bartonella (Rochalimaea) 24. Rolain JM, Locatelli C, Chabanne L, et al. Prevalence of Bartonella clar-
quintana bacteremia in inner-city patients with chronic alcoholism. N Engl ridgeiae and Bartonella henselae in domestic cats from France and detec-
J Med. 1995;332:424428. tion of the organisms in erythrocytes by immunofluorescence. Clin Diagn
6. Slater LN, Welch DF, Hensel D, et al. A newly recognized fastidious gram- Lab Immunol. 2004;11:423425.
negative pathogen as a cause of fever and bacteremia. N Engl J Med. 25. Gurfield AN, Boulouis HJ, Chomel BB, et al. Epidemiology of Bartonella
1990;323:15871593. infection in domestic cats in France. Vet Microbiol. 2001;80:185198.
7. Regnery RL, Anderson BE, Clarridge JE III, et al. Characterization of a 26. Lucey D, Dolan MJ, Moss CW, et al. Relapsing illness due to Rochalimaea
novel Rochalimaea species, R. henselae sp. nov., isolated from blood of a henselae in immunocompetent hosts: implication for therapy and new epi-
febrile, human immunodeficiency virus-positive patient. J Clin Microbiol. demiological associations. Clin Infect Dis. 1992;14:683688.
1992;30:265274. 27. Tappero JW, Mohle-Boetani J, Koehler JE, et al. The epidemiology of bacil-
8. Daly JS, Worthington MG, Brenner DJ, et al. Rochalimaea elizabethae lary angiomatosis and bacillary peliosis. JAMA. 1993;269:770775.
sp. nov. isolated from a patient with endocarditis. J Clin Microbiol. 28. Regnery RL, Olson JG, Perkins BA, et al. Serological response to
1993;31:872881. Rochalimaea henselae antigen in suspected cat-scratch disease. Lancet.
9. Welch DF, Pickett DA, Slater LN, et al. Rochalimaea henselae sp. nov., 1992;339:14431445.
a cause of septicemia, bacillary angiomatosis, and parenchymal bacillary 29. Dolan MJ, Wong MT, Regnery RL, et al. Syndrome of Rochalimaea hense-
peliosis. J Clin Microbiol. 1992;30:275280. lae adenitis suggesting cat scratch disease. Ann Intern Med. 1993;118:
10. Relman DA, Lepp PW, Sadler KN, et al. Phylogenetic relationships among 331336.
the agent of bacillary angiomatosis, Bartonella bacilliformis, and other 30. Waldvogel K, Regnery RL, Anderson BE, et al. Disseminated cat-scratch
alpha-proteobacteria. Mol Microbiol. 1992;6:18011807. disease: detection of Rochalimaea henselae in affected tissue. Eur J Pediatr.
11. Brenner DJ, OConnor SP, Winkler HH, et al. Proposals to unify the gen- 1994;153:2327.
era Bartonella and Rochalimaea, with descriptions of Bartonella quin- 31. Anderson B, Sims K, Regnery R, et al. Detection of Rochalimaea hense-
tana comb. nov., Bartonella vinsonii comb. nov., Bartonella henselae lae DNA in specimens from cat scratch disease patients by PCR. J Clin
comb. nov., and Bartonella elizabethae comb. nov., and to remove the Microbiol. 1994;32:942948.
family Bartonellaceae from the order Rickettsiales. Int J Syst Bacteriol. 32. Goral S, Anderson B, Hager C, et al. Detection of Rochalimaea henselae
1993;43:777786. DNA by polymerase chain reaction from suppurative nodes of children
12. Birtles RJ, Harrison TG, Saunders NA, et al. Proposals to unify the genera with cat-scratch disease. Pediatr Infect Dis J. 1994;13:994997.
Grahamella and Bartonella, with descriptions of Bartonella talpae comb. 33. Le HH, Palay DA, Anderson B, et al. Conjunctival swab to diagnose ocular
nov., Bartonella peromysci comb. nov., and three new species, Bartonella cat scratch disease. Am J Ophthalmol. 1994;118:249250.
grahamii sp. nov., Bartonella taylorii sp. nov., and Bartonella doshiae sp. 34. Ricketts WE. Carrins disease: a study of the incubation period in thirteen
nov. Int J Syst Bacteriol. 1995;45:18. cases. Am J Trop Med. 1947;27:657659.
13. Breitschwerdt EB, Kordick DL. Bartonella infection in animals: carriership, 35. Strong RP, Tyzzer EE, Brues CT, et al. Report of the First Expedition to
reservoir potential, pathogenicity, and zoonotic potential for human infec- South America, 1913. Cambridge, MA: Harvard University Press; 1915.
tion. Clin Microbiol Rev. 2000;13:428438. 36. Ricketts WE. Bartonella bacilliformis anemia (Oroya fever); a study of
14. Houpikian P, Raoult D. Molecular phylogeny of the genus Bartonella: what 30 cases. Blood. 1948;3:10251049.
is the current knowledge? FEMS Microbiol Lett. 2001;200:17. 37. Reynafarje C, Ramos J. The hemolytic anemia of human bartonellosis.
15. Zeaiter Z, Fournier PE, Raoult D. Genomic variation of Bartonella hense- Blood. 1961;17:562578.
lae strains detected in lymph nodes of patients with cat scratch disease. 38. Maguina C, Garcia PJ, Gotuzzo E, et al. Bartonellosis (Carrions disease) in
J Clin Microbiol. 2002;40:10231030. the modern era. Clin Infect Dis. 2001;33:772779.
16. Arvand M, Feil EJ, Giladi M, et al. Multi-locus sequence typing of 39. Schultz MG. A history of bartonellosis (Carrions disease). Am J Trop Med
Bartonella henselae isolates from three continents reveals hypervirulent and Hyg. 1968;17:503515.
feline-associated clones. PloS One. 2007;2:e1346. 40. Ricketts WE. Clinical manifestations of Carrions disease. Arch Intern Med
17. Bonilla DL, Kabeya H, Henn J, et al. Bartonella quintana in body lice and (Chic). 1949;84:751781.
head lice from homeless persons, San Francisco, California, USA. Emerg 41. Kosek M, Lavarello R, Gilman RH, et al. Natural history of infection
Infect Dis. 2009;15:912915. with Bartonella bacilliformis in a nonendemic population. J Infect Dis.
18. Demers DM, Bass JW, Vincent JM, et al. Cat-scratch disease in Hawaii: 2000;182:865872.
etiology and seroepidemiology. J Pediatr. 1995;127:2326. 42. Cuadra M. Salmonellosis complication in human bartonellosis. Tex Rep
19. Koehler JE, Glaser CA, Tappero JW. Rochalimaea henselae infection. Biol Med. 1956;14:97113.
A new zoonosis with the domestic cat as reservoir. JAMA. 1994;271: 43. Pinkerton H, Weinman D. Toxoplasma infection in man. Arch Pathol.
531535. 1940;30:374392.

Scheld_Ch26.indd 440 2/21/14 5:44 PM


Chapter 26: Bartonella Infections, Including Cat-Scratch Disease 441

44. Arias-Stella J, Lieberman PH, Erlandson RA, et al. Histology, immunohis- 74. Kordick DL, Hilyard EJ, Hadfield TL, et al. Bartonella clarridgeiae, a newly
tochemistry, and ultrastructure of the verruga in Carrions disease. Am J recognized zoonotic pathogen causing inoculation papules, fever, and lymph-
Surg Pathol. 1986;10:595610. adenopathy (cat scratch disease). J Clin Microbiol. 1997;35:18131818.
45. Dooley JR. Bartonellosis In: Binford CH, Connor DH, eds. Pathology 75. Margileth AM, Baehren DF. Chest-wall abscess due to cat-scratch disease
of Tropical and Extraordinary Diseases. Washington, DC: Armed Forces (CSD) in an adult with antibodies to Bartonella clarridgeiae: case report
Institute of Pathology; 1976:190193. and review of the thoracopulmonary manifestations of CSD. Clin Infect
46. McNee JW, Renshaw A. Trench fever: a relapsing fever occurring with Dis. 1998;27:353357.
the British forces in France. Br Med J. 1916;1:225234. 76. Giladi M, Avidor B, Kletter Y, et al. Cat scratch disease: the rare role of
47. Brouqui P, Lascola B, Roux V, et al. Chronic Bartonella quintana bactere- Afipia felis. J Clin Microbiol. 1998;36:24992502.
mia in homeless patients. N Engl J Med. 1999;340:184189. 77. Debre R, Lamy M, Jammet ML, et al. La maladie des griffes de chat. Sem
48. Foucault C, Barrau K, Brouqui P, et al. Bartonella quintana bacteremia Hop. 1950;26:18951904.
among homeless People. Clin Infect Dis. 2002;35:684689. 78. Moriarty RA, Margileth AM. Cat scratch disease. Infect Dis Clin North
49. Raoult D, Fournier PE, Drancourt M, et al. Diagnosis of 22 new cases of Am. 1987;1:575590.
Bartonella endocarditis. Ann Intern Med. 1996;125:646652. 79. Carithers HA. Cat-scratch disease. An overview based on a study of
50. Houpikian P, Raoult D. Blood culture-negative endocarditis in a reference 1,200 patients. Am J Dis Child. 1985;139:11241133.
center: etiologic diagnosis of 348 cases. Medicine (Baltimore). 2005;84: 80. Margileth AM. Cat scratch disease. Adv Pediatr Infect Dis. 1993;8:121.
162173. 81. Carithers HA, Margileth AM. Cat-scratch disease. Acute encephalopathy
51. Fournier PE, Thuny F, Richet H, et al. Comprehensive diagnostic strategy and other neurologic manifestations. Am J Dis Child. 1991;145:98101.
for blood culture-negative endocarditis: a prospective study of 819 new 82. Anderson BE, Neuman MA. Bartonella spp. as emerging human patho-
cases. Clin Infect Dis. 2010;51:131140. gens. Clin Microbiol Rev. 1997;10:203219.
52. Roux V, Eykyn SJ, Wyllie S, et al. Bartonella vinsonii subsp. berkhoffii as 83. Giladi M, Maman E, Paran D, et al. Cat-scratch disease-associated
an agent of afebrile blood culture-negative endocarditis in a human. J Clin arthropathy. Arthritis Rheum. 2005;52:36113617.
Microbiol. 2000;38:16981700. 84. Maman E, Bickels J, Ephros M, et al. Musculoskeletal manifestations of
53. Avidor B, Graidy M, Efrat G, et al. Bartonella koehlerae, a new cat- cat scratch disease. Clin Infect Dis. 2007;45:15351540.
associated agent of culture-negative human endocarditis. J Clin Microbiol. 85. Abbasi S, Chesney PJ. Pulmonary manifestations of cat-scratch disease; a case
2004;42:3462468. report and review of the literature. Pediatr Infect Dis J. 1995;14:547548.
54. Raoult D, Roblot F, Rolain JM, et al. First isolation of Bartonella alsatica from 86. Wesslen L, Ehrenborg C, Holmberg M, et al. Subacute Bartonella infection
a valve of a patient with endocarditis. J Clin Microbiol. 2006;44:278279. in Swedish orienteers succumbing to sudden unexpected cardiac death or
55. Raoult D, Fournier PE, Vandenesch F, et al. Outcome and treatment of having malignant arrhythmias. Scand J Infect Dis. 2001;33:429438.
Bartonella endocarditis. Arch Intern Med. 2003;163:226230. 87. Meininger GR, Nadasdy T, Hruban RH, et al. Chronic active myocarditis
56. Stoler MH, Bonfiglio TA, Steigbigel RT, et al. An atypical subcutaneous following acute Bartonella henselae infection (cat scratch disease). Am J
infection associated with acquired immune deficiency syndrome. Am J Clin Surg Pathol. 2001;25:12111214.
Pathol. 1983;80:714718. 88. Borker A, Gardner R. Severe thrombocytopenic purpura as a complica-
57. Cockerell CJ, Whitlow MA, Webster GF, et al. Epithelioid angiomatosis: tion of cat scratch disease. Clin Pediatr (Phila). 2002;41:117118.
a distinct vascular disorder in patients with the acquired immunodeficiency 89. Ghez D, Bernard L, Bayou E, et al. Bartonella henselae infection
syndrome or AIDS-related complex. Lancet. 1987;2:654656. mimicking a splenic lymphoma. Scand J Infect Dis. 2001;33:935936.
58. LeBoit PE, Berger TG, Egbert BM, et al. Epithelioid haemangioma-like 90. Gilad J, Wolak A, Borer A, et al. Isolated splenic cat scratch disease in an
vascular proliferation in AIDS: manifestation of cat scratch disease bacillus immunocompetent adult woman. Clin Infect Dis. 2003;36:e10e13.
infection? Lancet. 1988;1:960963. 91. Markaki S, Sotiropoulou M, Papaspirou P, et al. Cat-scratch disease
59. Slater LN, Welch DF, Min KW. Rochalimaea henselae causes bacillary presenting as a solitary tumour in the breast: report of three cases. Eur J
angiomatosis and peliosis hepatis. Arch Intern Med. 1992;152:602606. Obstet Gynecol Reprod Biol. 2003;106:175178.
60. Koehler JE, LeBoit PE, Egbert BM, et al. Cutaneous vascular lesions and 92. Trelles JO, Palomino L, Trelles L. Neurologic form of Carrions disease.
disseminated cat-scratch disease in patients with the acquired immunode- Clinical-anatomical study of 9 cases [in Spanish]. Rev Neuropsiquiatr.
ficiency syndrome (AIDS) and AIDS-related complex. Ann Intern Med. 1969;32:245306.
1988;109:449455. 93. Trelles JO. Neurological disorders in Peru. In: Spillane JD, ed. Tropical
61. Kemper CA, Lombard CM, Deresinski SC, et al. Visceral bacillary epithe- Neurology. New York: Oxford University Press; 1973:387396.
lioid angiomatosis: possible manifestations of disseminated cat scratch dis- 94. Parrott JH, Dure L, Sullender W, et al. Central nervous system infection
ease in the immunocompromised host: a report of two cases. Am J Med. associated with Bartonella quintana: a report of two cases. Pediatrics.
1990;89:216222. 1997;100:403408.
62. Spach DH, Panther LA, Thorning DR, et al. Intracerebral bacillary 95. Mantadakis E, Spanaki AM, Psaroulaki A, et al. Encephalopathy com-
angiomatosis in a patient infected with human immunodeficiency virus. plicated by Guillain-Barre syndrome and hydrocephalus and associated
Ann Intern Med. 1992;116:740742. with acute Bartonella quintana infection. Pediatr Infect Dis J. 2007;26:
63. Relman DA, Loutit JS, Schmidt TM, et al. The agent of bacillary angioma- 860862.
tosis. an approach to the identification of uncultured pathogens. N Engl J 96. Wong MT, Dolan MJ, Lattuada CP Jr, et al. Neuroretinitis, aseptic menin-
Med. 1990;323:15731580. gitis, and lymphadenitis associated with Bartonella (Rochalimaea) hense-
64. Cockerell CJ, Bergstresser PR, Myrie-Williams C, et al. Bacillary epithe- lae infection in immunocompetent patients and patients infected with
lioid angiomatosis occurring in an immunocompetent individual. Arch human immunodeficiency virus type 1. Clin Infect. 1995;21:352360.
Dermatol. 1990;126:787790. 97. Stevens H. Cat-scratch fever encephalitis. AMA Am J Dis Child.
65. Tappero JW, Koehler JE, Berger TG, et al. Bacillary angiomatosis and 1952;84:218222.
bacillary splenitis in immunocompetent adults. Ann Intern Med. 1993;118: 98. Debre R, Van Bogaert L, Thieffry S, et al. Accidents nerveux de la maladie
363365. des griffes du chat. Bull Acad Natl Med. 1952;136:454459.
66. Zarraga M, Rosen L, Herschthal D. Bacillary angiomatosis in an 99. Jambor J, Emura E. Benign inoculation lymphoreticulosis (cat-scratch
immunocompetent child: a case report and review of the literature. Am J disease); report of two cases with positive skin tests. AMA Arch Derm
Dermatopathol. 2011;33:513515. Syphilol. 1953;67:439442.
67. Koehler JE, Sanchez MA, Garrido CS, et al. Molecular epidemiology of 100. Thompson TE Jr, Miller KF. Cat scratch encephalitis. Ann Intern Med.
Bartonella infections in patients with bacillary angiomatosis-peliosis. 1953;39:146151.
N Engl J Med. 1997;337:18761883. 101. Weinstein L, Meade RH 3rd. The neurological manifestations of cat
68. Lee J, Lynde C. Pyogenic granuloma: pyogenic again? Association between scratch disease. Am J Med Sci. 1955;229:500505.
pyogenic granuloma and Bartonella. J Cutan Med Surg. 2001;5:467470. 102. Paxson EM, McKay RJ Jr. Neurologic symptoms associated with cat
69. Levy I, Rolain JM, Lepidi H, et al. Is pyogenic granuloma associated with scratch disease. Pediatrics. 1957;20:1322.
Bartonella infection? J Am Acad Dermatol. 2005;53:10651066. 103. Gair DR, Walls WL. Encephalitis in cat scratch disease; report of two
70. Cockerell CJ, LeBoit PE. Bacillary angiomatosis: a newly characterized, cases. J Fla Med Assoc. 1957;44:491492.
pseudoneoplastic, infectious, cutaneous vascular disorder. J Am Acad 104. Smith RE, Darling RM. Encephalopathy of cat-scratch disease. AMA J
Dermatol. 1990;22:501512. Dis Child. 1960;99:107108.
71. LeBoit PE, Berger TG, Egbert BM, et al. Bacillary angiomatosis. The his- 105. Steiner MM, Vuckovitch D, Hadawi SA. Cat-scratch disease with enceph-
topathology and differential diagnosis of a pseudoneoplastic infection in alopathy. Case report and review of the literature. J Pediatr. 1963;62:
patients with human immunodeficiency virus disease. Am J Surg Pathol. 514520.
1989;13:909920. 106. Brooksaler F. Cat scratch disease with encephalopathy. Am J Dis Child.
72. Perkocha LA, Geaghan SM, Yen TS, et al. Clinical and pathological fea- 1964;107:185187.
tures of bacillary peliosis hepatis in association with human immunodefi- 107. Pollen RH. Cat-scratch encephalitis. Neurology. 1968;18:10311033.
ciency virus infection. N Engl J Med. 1990;323:15811586. 108. Sweeney VP, Drance SM. Optic neuritis and compressive neuropathy as-
73. Leong SS, Cazen RA, Yu GS, et al. Abdominal visceral peliosis associ- sociated with cat scratch disease. Can Med Assoc J. 1970;103:13801381.
ated with bacillary angiomatosis. Ultrastructural evidence of endothelial 109. Lyon LW. Neurologic manifestations of cat-scratch disease. Report of a
destruction by bacilli. Arch Pathol Lab Med. 1992;116:866871. case and review of the literature. Arch Neurol. 1971;25:2327.

Scheld_Ch26.indd 441 2/21/14 5:44 PM


442 Part III: Bacterial and Mycoplasmal Infections

110. Torres JR, Sanders CV, Strub RL, et al. Cat-scratch disease causing revers- 147. Vermeulen MJ, Rutten GJ, Verhagen I, et al. Transient paresis associated
ible encephalopathy. JAMA. 1978;240:16281629. with cat-scratch disease: case report and literature review of vertebral
111. Gadoth N, Oren A, Keynan A, et al. Cat-scratch disease presenting as osteomyelitis caused by Bartonella henselae. Pediatr Infect Dis J. 2006;25:
status epilepticus. A case report. Isr J Med Sci. 1979;15:162164. 11771181.
112. Selby G, Walker GL. Cerebral arteritis in cat-scratch disease. Neurology. 148. Hussain S, Rathore MH. Cat scratch disease with epidural extension
1979;29:14131418. while on antimicrobial treatment. Pediatr Neurosurg. 2007;43:164166.
113. Miller P, Bell WE. Cat-scratch disease with encephalopathy. Clin Pediatr 149. Tasher D, Armarnik E, Mizrahi A, et al. Cat scratch disease with cervical
(Phila). 1980;19:233234. vertebral osteomyelitis and spinal epidural abscess. Pediatr Infect Dis J.
114. Pickerill RG, Milder JE. Transverse myelitis associated with cat-scratch 2009;28:848850.
disease in an adult. JAMA. 1981;246:28402841. 150. Al-Rahawan MM, Gray BM, Mitchell CS, et al. Thoracic vertebral os-
115. Pampe D, Holt RM. Cat scratch disease with reversible encephalopathy. teomyelitis with paraspinous mass and intraspinal extension: an atypical
Tex Med. 1984;80:3839. presentation of cat-scratch disease. Pediatr Radiol. 2012;42:116119.
116. Dreyer RF, Hopen G, Gass JD, et al. Lebers idiopathic stellate neuroreti- 151. Glaser CA, Gilliam S, Schnurr D, et al. In search of encephalitis etiologies:
nitis. Arch Ophthalmol. 1984;102:11401145. diagnostic challenges in the California Encephalitis Project, 19982000.
117. Brazis PW, Stokes HR, Ervin FR. Optic neuritis in cat scratch disease. Clin Infect Dis. 2003;36:731742.
J Clin Neuroophthalmol. 1986;6:172174. 152. Granerod J, Ambrose HE, Davies NW, et al. Causes of encephalitis
118. Lewis DW, Tucker SH. Central nervous system involvement in cat scratch and differences in their clinical presentations in England: a multicentre,
disease. Pediatrics. 1986;77:714721. population-based prospective study. Lancet Infect Dis. 2010;10:835844.
119. Melis K, Bochner A, Vandenberghe P, et al. Cat-scratch disease with 153. Mailles A, Stahl JP, Steering Committee and Investigators Group. Infectious
reversible encephalopathy. Eur J Pediatr. 1989;149:2425. encephalitis in France in 2007: a national prospective study. Clin Infect Dis.
120. Bar S, Segal M, Shapira R, et al. Neuroretinitis associated with cat scratch 2009;49:18381847.
disease. Am J Ophthalmol. 1990;110:703705. 154. Glaser C, Bloch KC. Encephalitis: why we need to keep pushing the enve-
121. Chrousos GA, Drack AV, Young M, et al. Neuroretinitis in cat scratch lope. Clin Infect Dis. 2009;49:18481850.
disease. J Clin Neuroophthalmol. 1990;10:9294. 155. Ben-Ami R, Ephros M, Avidor B, et al. Cat-scratch disease in elderly
122. Yagupsky P, Sofer S. Cat-scratch encephalopathy presenting as status epi- patients. Clin Infect Dis. 2005;41:969974.
lepticus and lymphadenitis. Pediatr Emerg Care. 1990;6:4345. 156. Munana KR, Vitek SM, Hegarty BC, et al. Infection of fetal feline
123. Harvey RA, Misselbeck WJ, Uphold RE. Cat-scratch disease: an unusual brain cells in culture with Bartonella henselae. Infect Immun. 2001;69:
cause of combative behavior. Am J Emerg Med. 1991;9:5253. 564569.
124. Ulrich GG, Waecker NJ Jr, Meister SJ, et al. Cat scratch disease associ- 157. Gerber JE, Johnson JE, Scott MA, et al. Fatal meningitis and encephalitis
ated with neuroretinitis in a 6-year-old girl. Ophthalmology. 1992;99: due to Bartonella henselae bacteria. J Forensic Sci. 2002;47:640644.
246249. 158. Fouch B, Coventry S. A case of fatal disseminated Bartonella henselae
125. Revol A, Vighetto A, Jouvet A, et al. Encephalitis in cat scratch disease with infection (cat-scratch disease) with encephalitis. Arch Pathol Lab Med.
persistent dementia. J Neurol Neurosurg Psychiatry. 1992;55:133135. 2007;131:15911594.
126. Slota MC, OConnor K. Recognizing and treating cat scratch disease with 159. Singhal AB, Newstein MC, Budzik R, et al. Diffusion-weighted mag-
encephalopathy in children. Crit Care Nurse. 1992;12:3942. netic resonance imaging abnormalities in Bartonella encephalopathy.
127. Xu DL, Wang Z, Song YJ. Cat-scratch disease encephalopathy. Chin Med J Neuroimaging. 2003;13:7982.
J (Engl). 1994;107:104106. 160. Reed JB, Scales DK, Wong MT, et al. Bartonella henselae neuro-
128. Hahn JS, Sum JM, Lee KP. Unusual MRI findings after status epilepticus retinitis in cat scratch disease. Diagnosis, management, and sequelae.
due to cat-scratch disease. Pediatr Neurol. 1994;10:255258. Ophthalmology. 1998;105:459466.
129. Golnik KC, Marotto ME, Fanous MM, et al. Ophthalmic manifestations 161. Solley WA, Martin DF, Newman NJ, et al. Cat scratch disease: posterior
of Rochalimaea species. Am J Ophthalmol. 1994;118:145151. segment manifestations. Ophthalmology. 1999;106:15461553.
130. Centers for Disease Control and Prevention. Encephalitis associated with 162. Ullrich K, Saha N, Lake S. Neuroretinitis following bull ant sting. BMJ
cat scratch diseaseBroward and Palm Beach Counties, Florida, 1994. Case Rep. 2012;2012.
MMWR Morb Mortal Wkly Rep. 1994;43:909, 915916. 163. Irshad FA, Gordon RA. Bartonella henselae neuroretinitis in a 15-year-old
131. Hadley S, Albrecht MA, Tarsy D. Cat-scratch encephalopathy: a cause girl with chronic myelogenous leukemia. J AAPOS. 2009;13:602604.
of status epilepticus and coma in a healthy young adult. Neurology. 164. Donnio A, Jean-Charles A, Merle H. Macular hole following Bartonella
1995;45:196. henselae neuroretinitis. Eur J Ophthalmol. 2008;18:456458.
132. Salgado CD, Weisse ME. Transverse myelitis associated with probable 165. Pinto VL Jr, Curi AL, Pinto Ada S, et al. Cat scratch disease complicated
cat-scratch disease in a previously healthy pediatric patient. Clin Infect. with aseptic meningitis and neuroretinitis. Braz J Infect Dis. 2008;12:
2000;31:609611. 158160.
133. Thompson PK, Vaphiades MS, Saccente M. Cat-scratch disease present- 166. Smith RA, Scott B, Beverley DW, et al. Encephalopathy with retinitis due
ing as neuroretinitis and peripheral facial palsy. J Neuroophthalmol. to cat-scratch disease. Dev Med Child Neurol. 2007;49:931934.
1999;19:240241. 167. Belvisi V, Tieghi T, Grenga PL, et al. Bartonella henselae infection present-
134. Walter RS, Eppes SC. Cat scratch disease presenting with peripheral facial ing with ocular and hepatosplenic manifestations in an immunocompetent
nerve paralysis. Pediatrics. 1998;101:E13. child. Pediatr Infect Dis J. 2012;31:882883.
135. Puligheddu M, Giagheddu A, Genugu F, et al. Epilepsia partialis continua 168. Kerkhoff FT, Bergmans AM, van Der Zee A, et al. Demonstration of
in cat scratch disease. Seizure. 2004;13:191195. Bartonella grahamii DNA in ocular fluids of a patient with neuroretinitis.
136. Rondet B, Sarret C, Lacombe P, et al. Neurological symptoms with J Clin Microbiol. 1999;37:40344038.
Bartonella henselae infection: report on 2 pediatric cases [in French]. Arch 169. OHalloran HS, Draud K, Minix M, et al. Lebers neuroretinitis in
Pediatr. 2012;19:823826. a patient with serologic evidence of Bartonella elizabethae. Retina.
137. Marienfeld CB, Dicapua DB, Sze GK, et al. Expressive aphasia as a pre- 1998;18:276278.
sentation of encephalitis with Bartonella henselae infection in an immu- 170. Carroll DM, Franklin RM. Lebers idiopathic stellate retinopathy. Am J
nocompetent adult. Yale J Biol Med. 2010;83:6771. Ophthalmol. 1982;93:96101.
138. Cherinet Y, Tomlinson R. Cat scratch disease presenting as acute encepha- 171. Wade NK, Levi L, Jones MR, et al. Optic disk edema associated with peri-
lopathy. Emerg Med J. 2008;25:703704. papillary serous retinal detachment: an early sign of systemic Bartonella
139. Brenneis C, Scherfler C, Engelhardt K, et al. Encephalitis lethargica fol- henselae infection. Am J Ophthalmol. 2000;130:327334.
lowing Bartonella henselae infection. J Neurol. 2007;254:546547. 172. Gray AV, Reed JB, Wendel RT, et al. Bartonella henselae infection as-
140. Baylor P, Garoufi A, Karpathios T, et al. Transverse myelitis in 2 patients sociated with peripapillary angioma, branch retinal artery occlusion, and
with Bartonella henselae infection (cat scratch disease). Clin Infect Dis. severe vision loss. Am J Ophthalmol. 1999;127:223224.
2007;45:e42e45. 173. Ormerod LD, Dailey JP. Ocular manifestations of cat-scratch disease.
141. Stockmeyer B, Schoerner C, Frangou P, et al. Chronic vasculitis and Curr Opin Ophthalmol. 1999;10:209216.
polyneuropathy due to infection with Bartonella henselae. Infection. 174. Pinna A, Puglia E, Dore S. Unusual retinal manifestations of cat scratch
2007;35:107109. disease. Int Ophthalmol. 2011;31:125128.
142. Massei F, Gori L, Taddeucci G, et al. Bartonella henselae infection associ- 175. Font RL, Del Valle M, Mitchell BM, et al. Cat-scratch uveitis confirmed
ated with Guillain-Barre syndrome. Pediatr Infect Dis J. 2006;25:9091. by histological, serological, and molecular diagnoses. Cornea. 2011;30:
143. Dyachenko P, Ziv M, Raz R, et al. Cat scratch disease encephalopathy in 468471.
an immunocompetent patient. Eur J Intern Med. 2005;16:610611. 176. Curi AL, Machado D, Heringer G, et al. Cat-scratch disease: ocular mani-
144. Angibaud G, Balague JP, Lafontan JF. Bartonella hensalae encephalopathy festations and visual outcome. Int Ophthalmol. 2010;30:553558.
[in French]. Presse Med. 2005;34:297298. 177. Terrada C, Bodaghi B, Conrath J, et al. Uveitis: an emerging clinical
145. Rocha JL, Pellegrino LN, Riella LV, et al. Acute hemiplegia associated form of Bartonella infection. Clin Microbiol Infect. 2009;15(suppl 2):
with cat-scratch disease. Braz J Infect Dis. 2004;8:263266. 132133.
146. Abdel-Haq N, Abuhammour W, Al-Tatari H, et al. Disseminated cat 178. Drancourt M, Berger P, Terrada C, et al. High prevalence of fastidious bac-
scratch disease with vertebral osteomyelitis and epidural abscess. South teria in 1520 cases of uveitis of unknown etiology. Medicine (Baltimore).
Med J. 2005;98:11421145. 2008;87:167176.

Scheld_Ch26.indd 442 2/21/14 5:44 PM


Chapter 26: Bartonella Infections, Including Cat-Scratch Disease 443

179. Martinez-Osorio H, Calonge M, Torres J, et al. Cat-scratch disease 193. Duncan AW, Maggi RG, Breitschwerdt EB. A combined approach for
(ocular bartonellosis) presenting as bilateral recurrent iridocyclitis. Clin the enhanced detection and isolation of Bartonella species in dog blood
Infect Dis. 2005;40:e43e45. samples: pre-enrichment liquid culture followed by PCR and subculture
180. Khurana RN, Albini T, Green RL, et al. Bartonella henselae infection onto agar plates. J Microbiol Methods. 2007;69:273281.
presenting as a unilateral panuveitis simulating Vogt-Koyanagi-Harada 194. Fournier PE, Couderc C, Buffet S, et al. Rapid and cost-effective identi-
syndrome. Am J Ophthalmol. 2004;138:10631065. fication of Bartonella species using mass spectrometry. J Med Microbiol.
181. Gray AV, Michels KS, Lauer AK, et al. Bartonella henselae infection 2009;58:11541159.
associated with neuroretinitis, central retinal artery and vein occlusion, 195. Avidor B, Kletter Y, Abulafia S, et al. Molecular diagnosis of cat scratch
neovascular glaucoma, and severe vision loss. Am J Ophthalmol. 2004;137: disease: a two-step approach. J Clin Microbiol. 1997;35:19241930.
187189. 196. Mouritsen CL, Litwin CM, Maiese RL, et al. Rapid polymerase chain
182. Schwartzman WA, Patnaik M, Barka NE, et al. Rochalimaea antibodies in reaction based detection of the causative agent of cat scratch disease
HIV-associated neurologic disease. Neurology. 1994;44:13121316. (Bartonella henselae) in formalin-fixed, paraffin-embedded samples. Hum
183. Breitschwerdt EB, Maggi RG, Duncan AW, et al. Bartonella species in Pathol. 1997;28:820826.
blood of immunocompetent persons with animal and arthropod contact. 197. George TI, Manley G, Koehler JE, et al. Detection of Bartonella henselae
Emerg Infect Dis. 2007;13:938941. by polymerase chain reaction in brain tissue of an immunocompromised
184. Breitschwerdt EB, Maggi RG, Nicholson WL, et al. Bartonella sp. bacte- patient with multiple enhancing lesions. Case report and review of the
remia in patients with neurological and neurocognitive dysfunction. J Clin literature. J Neurosurg. 1998;89:640644.
Microbiol. 2008;46:28562861. 198. Diederen BMW, Vermeulen MJ, Verbakel H, et al. Evaluation of an inter-
185. Maggi RG, Kosoy M, Mintzer M, et al. Isolation of Candidatus Bartonella nally controlled real-time polymerase chain reaction assay targeting the
melophagi from human blood. Emerg Infect Dis. 2009;15:6668. groEL gene for the detection of Bartonella spp. DNA in patients with
186. Breitschwerdt EB, Maggi RG, Farmer P, et al. Molecular evidence of peri- suspected cat-scratch disease. Eur J Clin Microbiol. 2007;26:629633.
natal transmission of Bartonella vinsonii subsp. berkhoffii and Bartonella 199. Diaz MH, Bai Y, Malania L, et al. Development of a novel genus-specific
henselae to a child. J Clin Microbiol. 2010;48:22892293. real-time PCR assay for detection and differentiation of Bartonella species
187. Breitschwerdt EB, Maggi RG, Robert Mozayeni B, et al. PCR amplifica- and genotypes. J Clin Microbiol. 2012;50:16451649.
tion of Bartonella koehlerae from human blood and enrichment blood 200. Giladi M, Kletter Y, Avidor B, et al. Enzyme immunoassay for the diagno-
cultures. Parasit Vectors. 2010;3:76. sis of cat-scratch disease defined by polymerase chain reaction. Clin Infect
188. Breitschwerdt EB, Mascarelli PE, Schweickert LA, et al. Hallucinations, Dis. 2001;33:18521858.
sensory neuropathy, and peripheral visual deficits in a young woman in- 201. Sander A, Posselt M, Oberle K, et al. Seroprevalence of antibodies to
fected with Bartonella koehlerae. J Clin Microbiol. 2011;49:34153417. Bartonella henselae in patients with cat scratch disease and in healthy
189. Welch DF, Carroll KC, Hofmeister EK, et al. Isolation of a new subspecies, controls: evaluation and comparison of two commercial serological tests.
Bartonella vinsonii subsp. arupensis, from a cattle rancher: identity with Clin Diagn Lab Immunol. 1998;5:486490.
isolates found in conjunction with Borrelia burgdorferi and Babesia microti 202. Rolain JM, Brouqui P, Koehler JE, et al. Recommendations for treatment
among naturally infected mice. J Clin Microbiol. 1999;37:25982601. of human infections caused by Bartonella species. Antimicrob Agents and
190. Probert W, Louie JK, Tucker JR, et al. Meningitis due to a Bartonella Chemother. 2004;48:19211933.
washoensis-like human pathogen. J Clin Microbiol. 2009;47:23322335. 203. Spach DH, Koehler JE. Bartonella-associated infections. Infect Dis Clin
191. Maggi RG, Kemp VAJ, Chomel BB, et al. Bartonella. In: Versalovic J, North Am. 1998;12:137155.
Carroll KC, Funke G, et al, eds. Manual of Clinical Microbiology. 204. Weston KD, Tran T, Kimmel KN, et al. Possible role of high-dose corti-
Washington, DC: ASM Press; 2011:786798. costeroids in the treatment of cat-scratch disease encephalopathy. J Child
192. Fournier PE, Robson J, Zeaiter Z, et al. Improved culture from lymph Neurol. 2001;16:762763.
nodes of patients with cat scratch disease and genotypic character- 205. Genizi J, Kasis I, Schif A, et al. Effect of high-dose methyl-prednisolone on
ization of Bartonella henselae isolates in Australia. J Clin Microbiol. brainstem encephalopathy and basal ganglia impairment complicating cat
2002;40:36203624. scratch disease. Brain Dev. 2007;29:377379.

Scheld_Ch26.indd 443 2/21/14 5:44 PM


CHAPTER 27 RICKETTSIOSES, ANAPLASMOSES,
AND Q FEVER
DIDIER RAOULT

Bacteria in the order Rickettsiales are gram-negative micro- fever group (SFG) that includes 24 species of organisms patho-
organisms that grow in association with eukaryotic cells. They genic for humans (Table 27.1). Rickettsiae are associated with
do not grow in axenic media but require living hosts such as arthropods. They are mainly transmitted to humans by bites
cell cultures, embryonated eggs, or susceptible animals. With from infected arthropods, but infections from aerosols of infected
the exception of Rickettsia prowazekii, the agent of epidemic insect feces and blood transfusions have also been described
typhus, and possibly Rickettsia felis in sub-Saharan Africa, (1). Ixodid or hard ticks are the vectors of SFG rickettsiae and
these bacteria infect humans incidentally as zoonoses. On the have a specific geographic distribution. Mites are the vectors
basis of molecular phylogeny, the bacteria causing rickettsial of Rickettsia akari (worldwide) and Orientia tsutsugamushi
diseases have been reclassified (Table 27.1). Rickettsioses are (linked to Asia), lice are the vectors of R. prowazekii, and fleas
emerging infectious diseases, with many new rickettsial dis- are the vectors of R. typhi and perhaps mosquitoes for Rickettsia
eases having been described in the past 15 years. Rickettsioses felis (2) (Table 27.2).
can be grouped as follows: Q fever, ehrlichioses, and diseases The main clinical signs and symptoms of rickettsioses include
caused by Rickettsia and Orientia species. fever, headache, a rash that is maculopapular or sometimes pete-
The genus Rickettsia is subdivided into the typhus group, chial or vesicular, inoculation eschars at the site of the arthropod
containing Rickettsia typhi and R. prowazekii, and the spotted bite, and local lymphadenopathies. Neurologic involvement is

TA B L E 2 7 . 1
GENETIC CLASSIFICATION OF RICKETTSIAE PATHOGENIC FOR HUMANS

Genus Group Species Serotype

Alpha proteobacteria
Rickettsiaceae Rickettsia Typhus R. prowazekii
R. typhi
Spotted fever R. conorii Malish
Isral
Astrakhan
Indian
R. rickettsii
R. sibirica
R. mongolotimonae
R. slovaca
R. honei
R. japonica
R. heilongjianghensis
R. aeschlimannii
R. helvetica
R. australis
R. felis
R. akari
R. parkeri
R. africae
R. raoultii
R. philipii (364D)
R. massiliae
Orientia Scrub typhus O. tsutsugamushi
Anaplasmataceae Ehrlichia E. chaffeensis
E. ewingii
Ehrlichia sp. Wisconsin
Anaplasma A. phagocytophilum
Neorickettsia N. sennetsu
Wolbachia W. pipientis
Gamma proteobacteria
Coxiella C. burnetii

444

Scheld_Ch27.indd 444 2/21/14 6:58 PM


Chapter 27: Rickettsioses, Anaplasmoses, and Q Fever 445

TA B L E 2 7 . 2
SPOTTED FEVERS

Year of
Regional Micro-
Arthropod Eschar Lymph High Fatality organism
Disease Organism Host Geographic Area Rash Tche Noire Node Fever Rate Isolation

Rocky Mountain R. rickettsii Dermacentor America (north, Yes, may be Very rare No Yes High 1919
spotted fever andersoni, central, and purpuric
Dermacentor south)
variabilis,
Amblyomma
cajennense
Mediterranean R. conorii Rhipicephalus Mediterranean, Yes, papular, Common No Yes Moderate 1932
spotted fever, sanguineus Africa, Asia may be
Kenya tick purpuric
typhus
Siberian tick R. sibirica Dermacentor Siberia, China Yes Common No Yes Low 1946
typhus nuttalli
Rickettsial pox R. akari Liponyssoides Worldwide Yes Common Yes Yes Low 1946
sanguineus vesicular
Queensland tick R. australis Ixodes Eastern Yes, may be Common ? Yes Moderate 1950
typhus holocyclus Australia vesicular
Israel spotted R. conorii idem R. Isral, south Yes Rare No Yes Low 1974
fever serotype conorii Europe
israel
Flinders Island R. honei Ixodes Flinders Island, Yes Common Yes Yes Low 1991
spotted fever granulosus Eastern
Australia,
Thailand
Astrakhan R. conorii R. pusillus Caspian sea, Yes Rare No Yes Low 1991
fever serotype Balkans
astrakhan
Japanese R. japonica Ixodes ricinus Japan (China ?) Yes Common No Yes Low 1992
spotted fever
African tick- R. africae Amblyomma Sub-Saharan, Yes (half of Frequently Yes No Low 1992
bite fever hebraum, Africa, cases); multiple
Amblyomma West Indies may be
variegatum vesicular
Lymphangitis- R. sibirica Hyalomma Mongolia, Africa, Yes Common Yes Yes Low 1996
associated mongolotimo- species, Mediterranean
rickettsiosis nae Rhipicephalus area
pusillus
Scalp eschar R. slovaca Dermacentor Europe, Pakistan Very rare May be Yes, No Low 1997
and neck R. raoultii marginatus, Europe, Asia, erythem- painful Unknown 1999
lymphadenop- Dermacentor North Africa atous
athy (SENLAT), reticularis
tick-borne
lymphadenop-
athy (TIBOLA)
Chinese spotted R. heilonji- China Yes Common No Yes ? 1998
fever anghensis
Unnamed R. helvetica Ixodes ricinus Europe, Asia No, Yes No No Sudden 2000
erythema death
Flea-borne R. felis Ctenocephalides Worldwide Sometimes Sometimes Unknown Yes Unknown 2001
spotted fever felis
Unnamed R. aeschlimannii Hyaloma Africa Yes Yes Yes Yes Unknown 2002
marginatum
Indian tick typhus R. conorii Rhipicephalus India Yes Yes No Yes ? 2002
serotype
Indian

(continued)

Scheld_Ch27.indd 445 2/21/14 6:58 PM


446 Part III: Bacterial and Mycoplasmal Infection

TA B L E 2 7 . 2
SPOTTED FEVERS (CONTINUED)

Year of
Regional Micro-
Arthropod Eschar Lymph Fatality organism
Disease Organism Host Geographic Area Rash Tche Noire Node High Fever Rate Isolation

American R. parkeri Amblyomma North, Central, Yes Common Yes Low to Unknown 1939
boutonneuse maculatum, and South moderate
fever Amblyomma America
triste

Spotted fever R. massiliae Rhipicephalus North and South Yes Common No Yes Unknown 1992
species America,
Europe, North
and sub-Saharan
Africa
Spotted fever R. philipii Dermacentor North America No Yes Yes Yes Unknown 1975
364 D occidentalis

mainly associated with severe forms of disease where it is part their epidemiology and geographic distribution. Rickettsia
of a multiple organ dysfunction syndrome (MODS). The main species were divided into three taxonomic groups:
pathologic mechanism in rickettsioses is a vasculitis following
infection of the vascular endothelial cells (Fig. 27.1). Apart from The typhus group comprises R. typhi, the agent of flea-
the known pathogens, many other rickettsial strains have been borne, murine typhus, and R. prowazekii causing louse-
found in arthropods, in particular ticks (1), but their roles as borne epidemic typhus, Brill-Zinsser disease, and American
human pathogens have yet to be determined. sylvatic typhus linked to flying squirrels.
The first name given to a rickettsial disease, typhus, is in- The SFG rickettsiae, which cause tick-transmitted diseases
dicative of central nervous system (CNS) involvement because such as Rocky Mountain spotted fever (RMSF) (caused by
it derives from (typhos) meaning cerebral confusion. R. rickettsii), Mediterranean spotted fever (MSF) (Rickettsia
Rickettsioses are seasonal and the arthropod host determines conorii), and African tick-bite fever (Rickettsia africae);

FIGURE 27.1 Rickettsia rickettsii in a human


endothelial cell. Five rickettsiae are present, vis-
ible as electron-dense structures surrounded by
a capsular halo in the peripheral cytoplasm, at
1-, 6-, 8-, 11-, and 12-oclock positions. Note
that the rickettsia at the 1-oclock position is
clearly a rod-shaped bacterium with a cell wall.
Electron microscope; magnification 11,400.
(Courtesy Dr. D. Silverman, Department of
Microbiology and Immunology, University of
Maryland at Baltimore.)

Scheld_Ch27.indd 446 2/21/14 6:58 PM


Chapter 27: Rickettsioses, Anaplasmoses, and Q Fever 447

TA B L E 2 7 . 3
RICKETTSIOSES
TRANSMISSION OF RICKETTSIAL DISEASES

Tick borne (tick is both vector and reservoir) Historical Background and
Transmitted through tick saliva after tick bite (depending Emerging Rickettsioses
on prevalence of local tick):
R. rickettsii, R. conorii, R. sibirica, R. africae Epidemic typhus was differentiated from typhoid in the six-
R. australis, R. honei, R. slovaca, R. mongolotimonae teenth century (4) because of the existence of a rash in the rick-
R. aeschlimannii, R. japonica, R. heilongjianghensis, ettsial disease. Some authors suspected an American origin for
R. parkeri? R. philipii (364D)? etc. typhus because of the American sylvatic flying squirrel reservoir.
Mite borne (mite is both vector and reservoir) At the beginning of the twentieth century, Ch Nicolle proved
Transmitted through mite bite: the role of lice in typhus transmission, and Ricketts (5) proved
R. akari (mouse mite) that the wood tick, Dermacentor andersoni, was involved in
O. tsutsugamushi (chiggers) the transmission of RMSF in Montana. In the Old World in
1910, Conor reported the first cases of MSF in Tunis. The role
Flea borne (flea is both vector and reservoir) of Rhipicephalus sanguineus was established in 1930 (2).
Transmitted through flea feces by autoinoculation or by Recently, several new tick-borne diseases have been identi-
aerosol: fied. Of the rickettsial diseases, only five that are transmitted by
R. felis (cat fleas) ticks were known before 1991 (1), and subsequently, many new
R. typhi (rat fleas) diseases have been reported, including Astrakhan (6); Flinders
Louse borne (louse is only vector) Island spotted fever (7); African tick-bite fever (8); Japanese
Transmitted through infected feces by autoinoculation or spotted fever (9); American Boutonneuse Fever (Rickettsia
by aerosol: parkeri); and, in Europe, infections caused by Rickettsia sibirica
R. prowazekii mongolotimonae (10), Rickettsia slovaca (11), Rickettsia raoul-
tii, Rickettsia helvetica (12), and Rickettsia aeschlimannii (13)
(Table 27.2).
New rickettsial species have frequently been found when
arthropod vectors have been studied, that is, ticks, mites, or
insects (fleas, lice, etc.). For example, C. burnetii, R. africae,
a mite transmitted disease, rickettsialpox (R. akari); and a R. conorii caspia, R. felis, R. sibirica mongolotimonae, R.
spotted fever caused by R. felis that may be transmitted by slovaca, R. helvetica, and R. aeschlimannii were first isolated
fleas or mosquitoes (Tables 27.2 and 27.3). from arthropods and later from people.
The scrub typhus group includes O. tsutsugamushi, a mite Recently, new diseases have been discovered using combina-
(chigger)-borne disease. tions of isolation, serologic testing, and polymerase chain reac-
Coxiella burnetii, the agent of Q fever, belongs to the tion (PCR) amplification. However, to definitively establish a
-Proteobacteria phylum. It causes a zoonosis in many disease, one should obtain an isolate of the presumptive agent
mammal species, including domestic animals and wild un- from clinical patients. When this is not possible, an association
gulates, and is excreted in birth products and milk. Humans of serology, immunologic detection of antigens in tissues, and
are usually infected by aerosols and may develop an acute PCR amplification of two different target genes may be used
primary infection eventually followed by a chronic disease to establish an etiologic relationship between a disease and an
when predisposing factors are present. Many acute infec- organism. Morphologic recognition of rickettsia-like bodies
tions cause neurologic signs; meningitis, meningoencephali- by electron microscopy is of little value (14). Microorganisms
tis, and Guillain-Barr syndrome have been associated with found in ticks (15) should be suspected of being human patho-
C. burnetii. gens, because isolates of unknown pathogenicity have errone-
Ehrlichioses and anaplasmoses are zoonoses (3). Ehrlichia ously been considered nonpathogenic for years (1).
species grow in blood cells in a cytoplasmic vacuole, forming
clusters or morulae. The bacteria in this large group are in-
creasingly recognized as potential human pathogens, and their Diseases
current taxonomy has been changed recently to match current
phylogenic knowledge. Four genera are described, as follows:
Rocky Mountain Spotted Fever
One apparently associated with a helminth vector and named
Neorickettsia contains only one human pathogen recognized Epidemiology. RMSF is the most severe of the rickettsioses.
in Japan and Laos (Neorickettsia sennetsu); two are trans- It is caused by R. rickettsii and is currently the main tick-
mitted by ticks, Ehrlichia (including the three human patho- transmitted rickettsiosis recognized in America along with R.
gens, Ehrlichia chaffeensis, Ehrlichia ewingii, and Ehrlichia parkeri (with R. africae in West Indies). It was described first
sp. Wisconsin) and Anaplasma (with Anaplasma phagocyto- in the nineteenth century in the Western United States but is
philum being the only currently identified human pathogen). now known to be prevalent in 44 states in the United States
The fourth genus, Wolbachia, has one recognized species, and in Central and South America (Brazil, Argentina, Costa
Wolbachia pipientis, which could be associated with arthro- Rica, Columbia, and Mexico).
pods or nematodes. When associated with filaria, in human The main tick vectors are Dermacentor andersoni (the
filariasis, it appears to be a pathogenic factor and a treatment Rocky Mountain spotted wood tick) in Western United
target. The two most common ehrlichioses, the human mono- States and Dermacentor variabilis (the American dog tick) in
cytic ehrlichiosis (HME) caused by E. chaffeensis and human the East, the Midwest, and the South. In Central and South
granulocytic anaplasmosis (HGA) caused by A. phagocyto- America, Amblyomma cajennense is the major identified vector.
philum may cause neurologic pathology such as meningitis or The duration of attachment is critical for infections, and trans-
meningoencephalitis. mission is unlikely when the tick feeds for less than 20 hours.

Scheld_Ch27.indd 447 2/21/14 6:58 PM


448 Part III: Bacterial and Mycoplasmal Infection

The tick bite is painless and frequently goes unnoticed. The epi- TA B L E 2 7 . 5
demiology of RMSF undergoes yearly variations that are largely
unexplained but probably mainly due to variations in tick activ- DIFFERENTIAL DIAGNOSIS OF ROCKY MOUNTAIN
ity and human encounters with ticks. There are 500 to 1,000 SPOTTED FEVER
cases annually in the United States, with 90% reported from
April to September, during late spring and summer. The disease With a rash
is relatively more prevalent in children younger than 10 years. Measles
Rubeola
Clinical Manifestations. The incubation period of RMSF Adenovirus
ranges from 2 to 14 days. Initially, patients have a sudden onset Coxsackie virus
of fever and headache, nonspecific signs with a broad list of Dengue
possible differential diagnoses. This has led to the recommenda- HIV infection
tion that in areas endemic for RMSF, primary care physicians Infectious mononucleosis
should treat individuals presenting with unexplained fever, with Mycoplasma pneumoniae
or without other manifestations of RMSF, as if they had the Disseminated Neisseria gonorrhoeae infection
disease (16,17). A rash usually appears 3 to 5 days after the Staphylococcus aureus
onset of symptoms but is not seen in 10% of patients, especially Streptococcus pyogenes
in dark-skinned patients. It is diffuse and involves palms and Treponema pallidum (especially in patients with AIDS)
soles; it is maculopapular and can be purpuric. Skin necrosis or Purpura fulminans (Meningococci, Haemophilus,
gangrene involving the digits or limbs may be observed in severe Pneumococci, Capnocytophaga canimorsus or various
cases (18). Renal failure is common in severe cases (19) and is Babesia sp. in splenectomized patients)
related to acute tubular necrosis or hypovolemia. Pulmonary With central nervous system involvement
manifestations of RMSF range from cough to respiratory dis- Meningitis
tress from pulmonary edema. Retinal abnormalities seen on Enteroviruses
funduscopic examination include venous engorgement, retinal Meningococci
edema, hemorrhages, papilledema, and arterial occlusion. HIV
Abnormalities in routine laboratory screens are nonspe- Mumps
cific; the white blood cell (WBC) count is generally normal, Pneumococci
anemia is uncommon, but thrombocytopenia is observed in Leptospirosis
one third to one half of patients. Hyponatremia and eleva- Listeriosis
tions in serum transaminases (e.g., aspartate aminotransferase Lyme disease
[AST]) occur commonly. Encephalitis and meningoencephalitis
Analysis of fatal cases of RMSF has shown that older age, Eastern or western equine encephalitis
hospitalization, and lack of treatment or treatment with chlor- St. Louis encephalitis
amphenicol are significantly correlated with death. Treatment West Nile virus
with tetracyclines, however, lowered the mortality rate (20). Japanese encephalitis
Glucose-6-phosphatase dehydrogenase (G6PD) deficiency is a Other viral encephalitidies (e.g., Nipah, Hendra) etc.
risk factor for fulminant disease (21). Herpes simplex virus
Cytomegalovirus (in patients with AIDS)
Neurologic Involvement. The frequency and the severity of Q fever
neurologic abnormalities depend on the severity of the illness Leptospirosis
(22). Neurologic complications are often the cause of death. Any multiorgan dysfunction syndrome
Headache is often severe and diffuse or bifrontal (2328) Toxic shock syndrome
(Table 27.4). In one study, 23% of patients had serious CNS S. pyogenes

TA B L E 2 7 . 4
NEUROLOGIC FINDINGS IN ROCKY MOUNTAIN complications, including stupor, seizures, delirium, ataxia,
SPOTTED FEVER focal neurologic deficits, papilledema, and coma (19). Coma
is more likely to occur in fatal cases (28). Cranial and periph-
Finding Occurrence
eral nerve abnormalities can occur, with hearing loss being the
most common. Neck stiffness, which is common, is usually
Headache 80%
related to neck muscle myalgia. The incidence of meningeal
Stupor 20% signs is about 20% and a diagnosis of bacterial or viral men-
Meningismus 20% ingitis is often considered (27) (Table 27.5). It was found that
Neck stiffness 20% 21 (66%) of 32 patients with RMSF who had undergone a
lumbar puncture had abnormalities of the cerebrospinal fluid
Ataxia 10%20%
(CSF) and abnormal CSF findings have now been documented
Coma 10%20% in 63 patients (19). There was a pleocytosis in 38%, the pro-
Seizures 10% tein concentration was increased in 35%, and there was a
Decreased hearing 10% moderate decrease in glucose concentration in 8%. The WBC
count in the CSF is rarely more than 100 cells/mm3. Usually
Meningitis 10%
lymphocytes predominate.
Focal neurologic signs 2% A wide variety of focal neurologic deficits have been described
Papilledema 2% including cranial nerve palsies, aphasia, and ataxia, hemiplegia,
complete paralysis, spasticity, and neurogenic bladder (29).

Scheld_Ch27.indd 448 2/21/14 6:58 PM


Chapter 27: Rickettsioses, Anaplasmoses, and Q Fever 449

FIGURE 27.2 Cerebral petechiae in RMSF. These lesions are com- FIGURE 27.4 Plantar rash in a patient with Mediterranean spotted fever.
monly found in the gray matter, but in this specimen, they are promi-
nent in the white matter. (From Binford CH, Connor DH. Pathology
of Tropical and Extraordinary Diseases. Washington, DC: Armed
Forces Institute of Pathology; 1976, with permission.) and a rash, R. massiliae infecting Rhipicephalus ticks has been
found to cause spotted fever in Europe, R. amblyommii has
been linked to fever and rash in Southeastern United States,
Patients with encephalitis may have Rocky Mountain spotless
and a spotted fever with inoculation eschar in California was
fever (30) and two patients have been described who developed
linked to a new rickettsia (Rickettsia philipii 364D). Finally,
a rash only after a brain biopsy was obtained (22). Therefore,
R. africae is prevalent in West Indies and reported in American
in endemic areas, doxycycline may be added to acyclovir in the
travelers to Africa. However, these diseases are generally mild
treatment of encephalitis. There is very little information on
with rare involvement of the CNS.
computed tomography (CT) findings in RMSF, but low-density
areas and edema have been described (29). The electroencepha-
logram (EEG) usually shows diffuse cortical dysfunction, espe-
Other Tick-Borne Rickettsioses
cially in comatose patients (19). Tick-transmitted rickettsioses have limited geographic distri-
The gross pathologic findings in RMSF involving the CNS butions that are determined by their tick vectors. In Europe,
include edema and hemorrhage (Fig. 27.2). Microscopically, MSF is caused by R. conorii and is also known as bouton-
perivascular accumulations of inflammatory cells, glial nod- neuse fever, Marseilles fever, Astrakhan fever, Israeli spotted
ules, and arteriolar thrombonecrosis with small infarcts have fever, Indian tick typhus, or Kenyan tick typhus. It resembles
been seen. Microorganisms are readily demonstrated in the RMSF but has specific features. The disease is milder, but a
endothelial cells of blood vessels using either immunofluores- fatality rate of 1.5% to 2.5% of hospitalized patients is still
cence (31) or immunoperoxidase techniques (Fig. 27.3). found. A malignant form of the disease with purpuric rash,
Residual neurologic deficits are common following RMSF. shock, and MODS has been described in alcoholic, diabetic,
They include learning disabilities, deafness, behavioral distur- human immunodeficiency virus (HIV)infected, and old, or
bance, depression, transverse myelitis, aphasia, and impair- debilitated patients. The typical clinical presentation is that
ment of fine motor skills (20,3235). It was reported in one of a patient with fever, a rash (which may involve the palms
series that 1 to 8 years after recovery, 57% of patients had and soles [Fig. 27.4]), and a tche noire, that is, a black es-
neurologic abnormalities, including headaches, mild defects in char at the site of the tick bites. The tche noire is found in
intellectual functioning, and EEG abnormalities (34). 50% to 80% of patients. The rash is often papular, which led
In America, it was believed that RMSF for decades was the to one of the names of the disease (boutonneuse fever). Israel
only tick-borne rickettsiosis, but a discrepancy appears in the tick-bite fever and Astrakhan fever appear milder than typical
increasing number of reported cases and a lowering fatality MSF, and a tche noire is often lacking.
rate (35). Because several tick-borne SF were identified such as R. slovaca and R. raoultii causes a newly described dis-
R. parkeri that causes a mild disease with an inoculation eschar ease that appears to be common in Europe. The tick vectors,

FIGURE 27.3 A characteristic endovasculitic lesion from the


skin of a patient with RMSF, showing mononuclear infiltration
of the blood vessel wall with hemorrhage and thrombus forma-
tion. Similar lesions are found throughout the body, including the
brain. (From Walker DH. Rickettsial and chlamydial diseases.
In: Kissane JM, ed. Andersons Pathology. 9th ed. St. Louis: CV
Mosby; 1990:348361, with permission.)

Scheld_Ch27.indd 449 2/21/14 6:58 PM


450 Part III: Bacterial and Mycoplasmal Infection

Dermacentor marginatus and Dermacentor reticulatus, feed reported in confirmed MSF) in half of the cases. An aphthous
mainly in the cold months and tend to attach on the scalp of stomatitis can be associated with the disease. In Asia, Japanese
people, preferring haired sites. The disease is more prevalent in spotted fever (caused by Rickettsia japonica), Siberian tick
children and women, contrary to other tick-borne rickettsio- typhus (caused by Rickettsia sibirica), and infections caused
ses. It is rarely exanthematic, with the typical clinical picture by R. mongolotimonae comprise commonly and ropelike lym-
including an erythematous skin lesion ranging from 2 to 8 cm phangitis between the inoculation eschar and a lymph node.
in diameter at the site of the tick bite on the scalp and regional Rickettsia australis (Queensland tick typhus) and Rickettsia
lymphadenopathy (which may be painful). Rarely, patients honei (Flinders Island spotted fever) cause diseases resembling
have fever and a rash, and a deep postinfectious asthenia and MSF, but their rash can be vesicular. The neurologic involve-
residual alopecia at the site of the tick bite can be observed. ment of the other tick-borne spotted fevers is comparable
A case of meningoencephalitis has been reported. The occur- to that of RMSF. MSF has been associated with meningitis,
rence of this rickettsiosis without rash may stimulate research meningoencephalitis, deafness, stupor, and coma (often with
on other new rickettsial diseases with only localized manifesta- MODS) (37). Meningoencephalitis was reported in two pa-
tions (36). This disease has been named successively TIBOLA tients with Japanese spotted fever (38).
(for tick-borne disease), DEBONEL (for Dermacentor-borne The spotted fever caused by R. felis is a new incompletely
eschar and lymphadenopathy) and SENLAT (scalp eschar and defined disease. The bacterium is found in fleas in the United
neck lymphadenopathy after tick bite) (36a). States, Peru, Europe, and Africa and in mosquitoes in Africa.
In Africa, African tick-bite fever may be the most com- It has been grown only recently (39,40). Seven cases have been
mon tick-borne rickettsiosis worldwide. It is due to R. africae, reported from Texas, Mexico, Brazil, France, and Germany.
which is transmitted by African ticks, Amblyomma hebraeum, All had fever, six of seven had a rash, and an inoculation es-
and Amblyomma variegatum. They usually feed on ungulates char was present in some patients (39,41). A diagnosis can
but also feed readily on human beings and cause a high preva- be based on serology using specific R. felis antigen or PCR
lence of infection in rural Africa (60% of tested patients ex- of blood or skin biopsies. The most effective treatment has
hibit antibodies) and in travelers. More than 50% of patients not been established, but the bacterium is highly susceptible
have more than one tche noire (Fig. 27.5), which are to doxycycline (42). Recently, it was reported commonly
most often on the lower limbs and are often associated with associated with fever in Kenya and Senegal where malaria is
regional lymphadenopathy in the groin. The disease differs endemic, causing as much as 6% of fevers (2a). The infection
from the other milder rickettsioses in that fever is frequently in children in Africa consists of fever and vesicular rash and
absent; a rash is present in only half of the patients with the was named Yaaf (42a).
disease; and the rash may be vesicular (which has never been
Rickettsialpox (Rickettsia akari)
Rickettsialpox was first described in New York City, where it is
still prevalent (43). The recent terrorism events (of September
11, 2001) have led physicians to pay increased attention to skin
eschars (as might be seen with anthrax) and vesicular rashes
(which might be seen with smallpox), and this has led to an
increase in the cases of rickettsialpox diagnosed in New York
City between 2001 and 2002 (44). R. akari, the causal agent,
is an SFG rickettsia transmitted by the bite of the mouse mite
(Liponyssoides sanguineus). Serologically, it cross-reacts with
other SFG rickettsia. The prevalence of infections is probably
underestimated, although cases have also been reported from
Arizona, Utah, and Ohio and a high seroprevalence was found
in intravenous drug users in Baltimore. Cases have also been
diagnosed in Russia, Ukraine, Slovenia, and Korea (1). Ten
days after the mite bite, the beginning of the illness is marked
by fever, headache, and myalgia. A careful examination will
reveal an inoculation eschar and regional lymphadenopathy.
Two to six days later, a rash appears consisting of 5 to 40
macular then papular to vesicular (or even pustular) lesions.
The name of the disease was derived from the latter, and it is
often mistaken for chickenpox. The disease is usually mild (1).

Epidemic Typhus
Typhus is transmitted by the human body louse, which lives
in clothes and multiplies rapidly when cold weather and lack
of hygiene allow it to. Its prevalence reflects the low socio-
economic status of certain members of a society (45) and
rises during war, in poor countries in refugee camps, and
in homeless people in rich countries, including the United
States and Europe. Recent reports of cases have been made in
Burundi (46), Rwanda, Russia, Peru, the United States, and
Algeria (47).
Humans are the reservoirs of R. prowazekii and lice are
the vectors. They are infected by ingesting R. prowazekii in a
FIGURE 27.5 Inoculation eschar in a patient with African tick-bite fever. blood meal. The organism multiplies in the gut and is released

Scheld_Ch27.indd 450 2/21/14 6:58 PM


Chapter 27: Rickettsioses, Anaplasmoses, and Q Fever 451

in feces (48), where R. prowazekii can survive for weeks.


Patients are infected by aerosols or by inoculation of infected
feces into the skin during scratching. Patients who recover
from typhus may have latent infections and relapse years later
with stress. The relapsing form is named Brill-Zinsser disease
and is associated with bacteremia that might lead to lice be-
coming infected and the start of a new outbreak (1). In the
United States, the eastern flying squirrel (Glaucomys volans)
and its fleas, lice, and mites can be infected. They constitute a
sylvatic reservoir and generate cases of domestic typhus (49).
Typhus begins abruptly with fever, headache, and myalgias.
Coughing is also common, as is neurologic involvement, ev-
idenced by stupor, confusion, or coma (Figs. 27.6 to 27.8).
A rash is observed in 20% to 80% of patients but may be
difficult to observe in dark-skinned people. It usually starts
in the axilla and then spreads. It is usually macular but may
be purpuric in severe cases. Diarrhea and jaundice are often
reported. Splenomegaly is found only infrequently. In severe
cases, shock is observed and the spontaneous fatality rate is
20% to 30%. Abnormal laboratory results might include leu-
kopenia, thrombocytopenia, anemia, and increased serum he-
patic enzymes. The disease should be considered in patients
with high fever and confusion and in patients exposed to lice.
In tropical countries, epidemic typhus might be confused with
typhoid, malaria (50), hemorrhagic fevers, and dengue. This
could have fatal consequences because the prescribed treat-
ments for typhoid (-lactams, co-trimoxazole, and quino-
lones) are ineffective in typhus. In people with lice, it can be
confused with trench fever and relapsing fever, in which the FIGURE 27.7 Illustration of a typhus nodule of the cerebral cortex in
same treatment can be prescribed (46). a patient with epidemic typhus. Note the proliferative character of the
Brill-Zinsser disease is the late relapsing form of typhus that is lesion and the absence of necrosis. (rom Wolbach SB, Todd JL, Palfrey
frequently undiagnosed as a rash and recent exposure to lice are FW. The Etiology and Pathology of Typhus. Cambridge, MA: The
Belknap Press of Harvard University Press; 1922. Copyright 1922
commonly not present (51). Interviewing the patient may reveal
by the President and Fellows of Harvard College.)
previous exposure to lice, associated, or not, with a previous
diagnosis of typhus. The disease is mild and the prognosis is good.
Sylvatic typhus in the United States is caused by an R. Murine Typhus
prowazekii variant and is a milder disease. The most promi-
Murine typhus is associated with rat and opossum fleas.
nent clinical features are neurologic, with meningitis being the
Humans are infected when contaminated flea feces are in-
clinical manifestation (49). Few cases have been described and
oculated into the skin during scratching at the sites of flea
nearly all occurred in areas east of the Mississippi, where the
bites. The disease is more prevalent in hot and humid areas,
eastern flying squirrel is found. In these areas, cases may be
specifically when rats proliferate. In the United States, 50 to
observed in winter and sylvatic typhus should be considered in
100 cases are reported yearly, mainly in southern California
patients with a rash.
and southern Texas. Recently, cases have been described in
Mexico, Indonesia, Southern Europe, and Africa (52,53).
The incubation period ranges from 8 to 16 days. The dis-
ease begins abruptly with fever, myalgias, arthralgias, nausea,
vomiting, and headache. A discrete rash is observed in 40% to
50% of patients, on average, 6 days after the onset of signs. It
is detected less often in dark-skinned patients. The rash usu-
ally consists of pink macules but may become maculopapular
(54). It begins in the axilla and generalizes to the trunk but
usually does not involve the face, palms, and soles. It can be-
come purpuric in severe cases. One third of patients have a
cough and one fourth of the patients have nonspecific intersti-
tial pneumonia sometimes associated with pleural effusion. In
severe forms, respiratory distress might require intubation and
artificial ventilation. Neurologic symptoms range from confu-
sion and stupor to coma and seizures in severe forms. Cerebral
hemorrhages may occur. Digestive involvement can manifest
as vomiting, abdominal pain, jaundice, and in severe cases,
hematemesis (52,55).
Laboratory abnormalities include leukopenia, which may
FIGURE 27.6 A hemorrhagic lesion in the superficial cerebral cortex
in a patient with epidemic typhus. Macrophages have ingested ex-
be followed by leukocytosis. There might also be thrombo-
travasated red blood cells. (From Wolbach SB, Todd JL, Palfrey FW. cytopenia and anemia, especially when hemolysis is observed
The Etiology and Pathology of Typhus. Cambridge, MA: The Belknap (often in patients with G6PD deficiency). A moderate increase
Press of Harvard University Press; 1922. Copyright 1922 by the in serum liver enzymes is common. In patients with severe dis-
President and Fellows of Harvard College.) ease, hyponatremia and hypoalbuminemia are observed.

Scheld_Ch27.indd 451 2/21/14 6:58 PM


452 Part III: Bacterial and Mycoplasmal Infection

FIGURE 27.8 Two examples of typhus nodules in


the gray matter of a patient with epidemic typhus.
Such lesions typically show marked inflammatory
reaction with infiltrating macrophages and lympho-
cytes. (From Ash JE, Spitz S. Pathology of Tropical
Diseases. Philadelphia: WB Saunders; 1945, with
permission.)

The prognosis is usually favorable, but 10% of pa- typhus is not more severe in HIV-infected patients and an
tients require intensive care and 1% die. The neurologic HIV-suppressive factor appears to be produced during infec-
complications of murine typhus usually manifest during tion. Relapses of the disease may occur (61,62). Diagnosis
the second week of illness (56). Aseptic meningitis occurs may be difficult because the clinical presentation is often
in 2% to 5% of patients (56). Meningoencephalitis, which nonspecific and identifying epidemiologic factors is criti-
is rare, occurs in older patients. Such patients usually have cal. Infectious mononucleosis is commonly confused with
seizures. Papilledema rarely occurs (57). The CSF WBC scrub typhus.
count rarely exceeds 150 cells/mm3 and mononuclear cells
predominate (56).

Scrub Typhus (Orientia tsutsugamushi) Diagnostics


Scrub typhus is transmitted by the bite of trombiculid mite
larvae infected by O. tsutsugamushi. It is prevalent in a tri-
Serology
angle extending between northern Japan, eastern Australia, Serologic assays are the simplest diagnostic tests to perform
and eastern Russia, and including the Far East, China, and (Table 27.6). The Weil-Felix test was the first to be used and
the Indian subcontinent. Altogether, 1 billion people may be involves antigens from three Proteus strains: Proteus vulgaris
exposed. Seasonality is determined by emergence of larvae. In OX-2, P. vulgaris OX-19, and Proteus mirabilis OXK. It was
temperate zones, it occurs mainly in autumn and to a lesser used widely to detect rickettsioses based on serologic cross re-
extent in spring. O. tsutsugamushi species have a wide hetero- actions (63), but the test lacks sensitivity and specificity.
genicity. The more common serotypes are Kato, Karp, Gilliam, Today, the most commonly used serologic test is microim-
and Kawasaki (58). munofluorescence (MIF). It is reliable but does not allow dif-
The incubation period in rural or urban scrub typhus is ferentiation of infections within the different SFG rickettsiae
10 days or more. The onset of signs is usually sudden and (64,65). The Western blot immunoassay (63) can be used to
associated with fever, headache, and myalgias. With care- differentiate between infections with the various SFG rick-
ful examination, an inoculation eschar may be found at the ettsiae, provided that acute-phase serum samples are used.
site of the mite bite and draining lymph nodes may be ten- The test detects two types of antigens, high-molecular-weight
der. Generalized lymphadenopathies and rash may be ob- outer membrane protein (rOmpA and rOmpB) and lipo-
served. The symptoms vary according to organ involvement. polysaccharide-like antigen. The proteins are species specific
Neuromeningeal symptoms are relatively common. Severe (63,66) and provide the basis for rickettsial serotyping (67).
forms can occur, associated with septic shock. If serum samples are collected very early in infection, strong
The fatality rate in untreated patients is 6% to 10%. In homologous reactions are often observed, making a spe-
a study of scrub typhus in Thailand, 9 (13%) of 72 patients cific diagnosis possible (68). However, because such serum
presented with meningitis or encephalitis syndromes, or both samples are not always available, more specific methods are
(59). One of the nine had cerebellitis and another had papill- needed. Cross-absorption studies are useful, especially if com-
edema (59). Focal neurologic signs may predominate (60). The plemented by Western blotting (68). This is particularly the
CSF contains mainly mononuclear WBCs (59). Laboratory case with typhus, in which in 50% of patients, serum samples
abnormalities may include leukopenia, thrombocytopenia, have the identical antibody titers to both R. prowazekii and
and increased levels of hepatic enzymes. Interestingly, scrub R. typhi (69).

Scheld_Ch27.indd 452 2/21/14 6:58 PM


Chapter 27: Rickettsioses, Anaplasmoses, and Q Fever 453

TA B L E 2 7 . 6
DIAGNOSTIC TESTS FOR RICKETTSIAL DISEASES

Smears Serology Immunodetection PCR Culture Xenodiagnosis

Rickettsioses
RMSF (R. rickettsii) NA MIF (cross reaction Yes (skin biopsy) Yes (skin) Yes Tick by PCR
with other MSF)
MSF (R. conorii) NA MIF (cross reaction Yes (skin biopsy) Yes (skin) Yes Tick by PCR
with other MSF)
R. africae NA MIF (often late) Yes (skin biopsy) Yes (skin, blood) Yes Tick by PCR
R. slovaca NA MIF (often negative) NA Yes (skin, Yes (blood) Tick by PCR
lymph nodes)
R. akari NA MIF Yes (skin) Yes Yes (skin) No
R. felis NA MIF NA Yes (blood) NA NA
R. prowazekii NA MIF (cross reactions Yes (skin biopsy) Yes (blood) Yes (blood) Louse by PCR
with R. typhi)
R. typhi NA MIF (cross NA? Yes (blood) Yes NA
reactions with
R. prowazekii)
O. tsutsugamushi NA MIF (use strains: Yes Yes (skin, Yes NA
Kato, Gillian, blood)
Karp, Kawasaki)
Ehrlichioses
E. chaffeensis Yes (morula in MIF (cross reactions Yes (in tissues) Yes Yes ?
monocytes) with E. ewingii)
A. phagocytophilum Yes (morula in MIF Yes (in tissues) Yes Yes NA
PMN)
E. ewingii Yes (morula in MIF (cross NA Yes NA NA
PMN) reactions with
E. chaffeensis)
Q fever
C. burnetii NA MIF (phase II Yes (valve, Yes (valve) Yes NA
and phase I for placenta)
chronic diseases)

NA, not applicable.

PCR-Based Detection of Rickettsiae Rickettsial DNA can also be detected in ticks (7578), fleas
(40), or mosquitoes (2a) and lice (47) by PCR-based amplifica-
PCR amplification can be used to detect rickettsiae in blood, tion methods (79). Detection strategies based on recognition of
skin biopsies, skin swabbing, and arthropods (70,71). Blood sequences within the 16S rRNA gene (80,81) and those encod-
should be centrifuged and PCRs for rickettsiae should be car- ing a 17-kd protein (8284), citrate synthase (41,46,85), and
ried out on the leukocyte-rich buffy coat. Although heparinized the rOmpB (46,86) and rOmpA (for SFG rickettsiae) (10,87)
blood can be used for isolation of rickettsiae into cell culture, it have been described. Because none of the PCR assays to date
is preferable to use blood collected in ethylenediaminetetraac- is specific for individual rickettsial species, reaction products
etate (EDTA) or sodium citrate for PCR amplification, because must be further analyzed to identify the species detected.
heparin inhibits PCR and is difficult to neutralize. PCR am- Approaches involving either restriction endonuclease analysis
plifications must be performed before the initiation of antibi- or base sequence determination have been described.
otic treatment and before antibodies become detectable. Fresh
tissues are preferred for PCRs, but paraffin-embedded tissues
and even slide-fixed specimens may be used (72). Although not
Immunologic Detection of Rickettsiae
always present, the tche noire is the most useful sample to Skin biopsy specimens have been used in the diagnosis of rick-
biopsy or to swab (72a) to detect SFG rickettsiae by PCR assay ettsioses since the early work of Woodward et al. (88). Samples
(73), and using this technique, we have characterized several can be tested fresh or after fixation and paraffin embedding.
species in our laboratory (10,74). PCR amplification of tche Samples obtained at autopsy can be tested in the same manner
noires or blood samples can be very useful because they enable as skin biopsy specimens (89,90).
earlier diagnoses of infections than cell culture or serology. The use of methods incorporating specific polyclonal anti-
PCR-based methods for the detection of rickettsiae are attrac- bodies or monoclonal antibodies allows the detection of rick-
tive because they not only circumvent the need for culture but ettsiae in blood or other tissues (91). This approach enables
also possibly offer a more sensitive and specific alternative. diagnosis of infections in patients before they seroconvert and

Scheld_Ch27.indd 453 2/21/14 6:58 PM


454 Part III: Bacterial and Mycoplasmal Infection

hence early administration of specific treatment. The method monitored. Mechanical ventilation is used in cases of respira-
can also be used to diagnose rickettsial infections in retrospec- tory distress, and antiseizure drugs in patients with seizures. In
tive studies using fixed tissues. patients with gangrene of the extremities, amputation may be
necessary. Glucocorticoids have not proven useful.
Isolation of Rickettsiae Prevention is based on the prevention of tick, louse, and
flea bites by using repellents and/or protective garments. It is
Isolation of rickettsiae is difficult and dangerous and should
also useful to check for ticks after exposure. Careful examina-
be attempted only in specialized laboratories. Rickettsiae have
tion of the scalp, groin, and axilla is recommended. Ticks can
been isolated by several methods. Although laboratory animals,
be removed with forceps and the skin should be disinfected.
originally guinea pigs and subsequently rats and voles, and
embryonated eggs have been widely used, cell culture is the most
commonly used system for primary isolation. Tick or mamma-
lian cell lines can be used. We have used a microculture system to
ANAPLASMOSES
isolate rickettsiae from human blood and other sources (9294). Anaplasmataceae species comprise four genera. They multiply
The shell vial assay was adapted from a commercially available in their hosts blood cells, including red blood cells (in animals
method for cytomegalovirus culture and early antigen detection. Anaplasma marginale), platelets (in dogs Ehrlichia platys), mono-
Isolation of rickettsiae by cell culture is now performed routinely cytes (E. chaffeensis), and neutrophils (A. phagocytophilum). Two
in our laboratory from heparinized blood (leukocyte-rich buffy are tick-transmitted diseases: Anaplasma and Ehrlichia, whereas
coat) and skin biopsies collected from patients before they have two are helminth-borne diseases: Neorickettsia and Wolbachia.
received antibiotic therapy and from arthropods (93,94). The index case of modern ehrlichioses was reported in the
United States in 1987 (100). The patient developed a fever after
being bitten by a tick in Arkansas, despite receiving chlorampheni-
Treatment col. The patient had, on blood smears, morulae in polymorpho-
nuclear (PMN) cells and antibodies to Ehrlichia canis. Since then,
Doxycycline is the main drug used to treat rickettsioses. In three human erhlichial agents have been discovered (3). The first
vitro, SFG rickettsiae are susceptible to doxycycline, chlor- agent of human ehrlichiosis identified in the United States was
amphenicol, clarithromycin, ketolides, fluoroquinolones, and E. chaffeensis. This infects monocytes and causes HME. The sec-
rifampicin (42,95). Typhus group rickettsiae are also suscep- ond is A. phagocytophilum causing HGA and infects PMN. The
tible to erythromycin (96). third is E. ewingii, an agent transmitted by A. americanum, which
Doxycycline is the main drug used to treat RMSF, and it is prevalent in Arkansas and infects neutrophils. It cross-reacts
should be prescribed in suspected cases. It is used in both children serologically with E. chaffeensis and infects mainly immunocom-
and adults, but not in pregnant women and allergic patients. In promised people. It has yet to be cultured and has been identified in
rickettsioses such as MSF, murine typhus, and epidemic typhus, dogs and patients by PCR. A recently identified Ehrlichia sp. was
a single day of treatment with doxycycline (200 mg) is effec- detected in four patients in Minnesota (100a) and is yet unnamed.
tive, but this has not yet been determined for RMSF. The drug
should be given orally except in patients with gastric intoler-
ance or coma, in whom it should be given intravenously. The Human Monocytic Ehrlichiosis
usual dose is 200 mg daily in two doses. The required duration (Ehrlichia chaffeensis)
of treatment in RMSF is not yet fully determined, but because
of the lack of relapse, it can be stopped 3 days after apyrexia.
In pregnant women, chloramphenicol is the only available
Epidemiology
alternative to doxycycline, but the drug has been shown to be E. chaffeensis has been isolated or identified by PCR mainly
less effective than doxycycline in treating RMSF (97). Failure in the United States, in southeastern, south central, mid-
has been reported when patients with murine typhus or epi- Atlantic States, and California. Its vector is the American tick,
demic typhus were treated with quinolones (98), and the drug A. americanum (lone-star tick) (Table 27.7), and white-tailed
should be avoided in these diseases despite its in vitro activity. deer are the mammalian reservoir. Immature ticks are infected
Preliminary reports indicate the new macrolides such as azithro- by feeding on blood from persistently bacteremic reservoirs.
mycin and clarithromycin are effective in the treatment of chil- E. chaffeensis is transmitted transtadially in the tick and infects
dren with MSF (99). Severely ill patients should be treated in its next host (deer or human) during its next blood meal. The dis-
intensive care units and fluid administration should be carefully ease epidemiology reflects the ticks biology because most cases

TA B L E 2 7 . 7
EHRLICHIOSES: GEOGRAPHIC DISTRIBUTION

Agent Disease Vector Geographic Distribution

E. chaffeensis Human monocytic ehrlichiosis Amblyoma americanum South central, southeastern, mid-Atlantic,
coastal states (United States)
A. phagocytophilum Human granulocytic Ixodes ricinus Ixodes Europe, eastern and northern United States
anaplasmosis scapularis
E. ewingii Unnamed A. americanum South, central, southeastern, mid-Atlantic,
coastal states (United States)
Ehrlichia sp. Wisconsin Unnamed I. scapularis Minnesota or Wisconsin (United States)
N. sennetsu Japanese monocytic ehrlichiosis Fluke of the gray mulet? Japan

Scheld_Ch27.indd 454 2/21/14 6:58 PM


Chapter 27: Rickettsioses, Anaplasmoses, and Q Fever 455

are contracted in the south of the United States, in rural areas


where the tick is most prevalent, and from April to September
when the ticks are most active. In highly endemic areas, the
incidence of HME can reach 100 cases per 100,000 inhabitants.
Older patients are more susceptible to infection. Case reports
from South America and Asia have been provided (100b).

Clinical Findings
The incubation period is 7 to 10 days, and tick exposure is re-
ported by 80% of patients. Common presenting signs are fever,
headache, nausea, malaise, and anorexia. If untreated, patients
may develop severe signs and require intensive care. Digestive
tract involvement is common with nausea, vomiting, diarrhea,
and abdominal pain. CNS infection manifests in many forms,
from confusion to coma (101). A rash is observed in 30% of
patients, and lymphadenopathies in 25%. In severe forms, shock
may be observed with hypotension, tachycardia, respiratory
distress, seizures, renal insufficiency, myocardial failure, and
coma. CNS involvement is common (3). Patients may develop FIGURE 27.9 Human granulocytic anaplasmosis: Anaplasma phago-
confusion, photophobia, stupor, hallucinations, and eventually cytophylum. (May-Grunwald-Giemsa [MGG] stain 60.)
seizures and coma. Changes in mental status, ataxia, blurred
vision, and cranial palsy have also been reported (101). CSF
examination often reveals pleocytosis, with a predominance of also vectors of Lyme disease, and the epidemiology of the
lymphocytes or neutrophils, and elevated protein levels. diseases is similar. Concurrent infections may also occur. The
Full blood cell counts typically show leukopenia, due temporal distribution of the disease parallels that of nymph
to lymphopenia and neutropenia, and thrombocytopenia. stage activity, with two peaks in spring and autumn. Ticks
Coagulopathies may be observed in severe forms. Increases in are infected while feeding on bacteremic rodents. Deer play
AST, alanine aminotransferase (ALT), and lactate dehydroge- a major role as hosts of adult ticks and reservoirs. In highly
nase are common. The prognosis of HME improves with early endemic areas in the United States, incidences of infection can
antibiotic treatment, but the fatality rate is still high, at 2.5%. In reach 50 per 100,000 inhabitants per year. Men are at higher
people concurrently infected with HIV, the disease may be most risk of infection than women or children (3).
severe, and in a recent study, 6 of 13 patients with coinfections
died. The diagnosis of HME should be strongly considered in Clinical Findings
patients with a history of tick exposure and unexplained fever. The incubation period is around 10 days and a history of tick
exposure is reported by 80% of patients. Signs of the disease fre-
Laboratory Diagnosis quently begin abruptly with fever, headache, malaise, and myal-
Careful examination of blood and CSF smears may help to gias that may be particularly severe. Rash is found in fewer than
identify typical morulae. Treatment should be started in any 10% of patients. Visceral involvement may be observed and diges-
suspected case. Diagnosis can be confirmed by culture, but tive symptoms such as nausea, vomiting, and diarrhea have been
PCR is used more commonly. A confirmatory PCR using a sec- reported. Neurologic symptoms may include confusion, meningi-
ond target gene is useful. Most cases are currently diagnosed tis, and meningoencephalitis. Brachial plexopathy and demyelin-
serologically by demonstrating a fourfold or greater increase ating polyneuropathy have also been reported (102104).
in antibody titers or by seroconversion. The reference tech- The prognosis is favorable in most patients, even without
nique is immunofluorescence antibody (IFA). A single titer of specific therapy, but some patients may develop septic shock.
25 is indicative of the diagnosis. There are cross-reactive anti- Most deaths are the consequence of induced immunosuppres-
bodies among Ehrlichia species and with A. phagocytophilum. sion and patients may die of invasive aspergillosis, candidiasis,
cryptococcosis, and herpes esophagitis (104a).
Treatment Laboratory findings often include thrombocytopenia with
leukopenia (lymphopenia and/or neutropenia). Increased
Doxycycline is the recommended treatment in adults and chil- serum transaminases are also common. A diagnosis can be
dren. Only rifampin also shows in vitro activity, but the drug made by the observation of morulae within PMN cells in blood
has been used only in a few pregnant patients. The treatment smears. Culture from blood is possible (Fig. 27.9) (105), but
is given empirically for 2 weeks, or it should be continued for PCR is used more commonly to identify A. phagocytophilum.
3 to 5 days after fever subsides (101a). Most cases are diagnosed by serology using IFA. Treatment
is similar to that of HME except that in vitro A. phagocyto-
philum is susceptible to fluoroquinolones. Response to these
Human Granulocytic Anaplasmosis agents has not been tested in patients.
(Anaplasma phagocytophilum)
Human Ewingii Ehrlichiosis
Epidemiology
(Ehrlichia ewingii)
A. phagocytophilum was identified in ungulates in the 1930s
as an agent causing chronic neutropenia. The first human case Canine granulocytic ehrlichiosis, reported in the United States in
was recognized in 1990 and the disease has now been reported 1972, is caused by the yet uncultured E. ewingii. The bacterium
from America and Europe. It is transmitted by Ixodes scapularis has been characterized by amplification and sequencing of the
(eastern North America), I. pacificus (western North America), 16S rRNA gene. E. ewingii is vectored by A. americanum, which
I. ricinus (Europe), and I. persulcatus (Asia). These ticks are also transmits E. chaffeensis. Of 60 cases of human ehrlichioses

Scheld_Ch27.indd 455 2/21/14 6:58 PM


456 Part III: Bacterial and Mycoplasmal Infection

in Missouri in 1999, 4 were caused by E. ewingii (105); sub-


sequently 4 other cases have been reported by the Centers for
Disease Control and Prevention (106,107). The disease is prev-
alent in immunocompromised hosts (7/8), those also infected
with HIV or receiving immunosuppressive drugs. Patients, who
report tick exposure, present with fever, thrombocytopenia, leu-
kopenia, and various symptoms including meningitis. Morulae
may be seen in PMN cells in blood smears. The prognosis in
reported cases has been good and patients respond dramatically
to doxycycline. Patients have antibodies that cross-react with
E. chaffeensis and PCR of blood samples has been shown to be
useful in the diagnosis of the disease. Infections with E. ewingii
should be considered when ehrlichiosis is suspected in immuno-
compromised patients exposed to A. americanum ticks.

Other Anaplamoses FIGURE 27.10 Typical lesions on liver biopsy during acute Q fever:
a central lipid vacuole surrounded by a fibrinoid ring and an inflam-
matory granuloma with mainly macrophages and neutrophils. This
Neorickettsia senetsu histologic feature is called the doughnut aspect.
This bacterium is associated with fish flukes and can cause
fever and mononucleosis syndrome in humans eating raw fish.
First described in Japan, it has been reported since in Korea and a giant outbreak was observed in the Netherlands (109a).
and recently in Laos (107a). The current geographic distribution is largely unknown. As
C. burnetii is considered a potential bioterrorism agent, diag-
Wolbachia nostic tools have been developed rapidly in the United States.
Adult men are the predominant group (110).
Wolbachia can infect the nematode causing filariasis in humans
in which they control the reproduction rate of the worm. They
cause some of the inflammatory responses of filariasis specifi- Clinical Manifestations
cally at the initiation of treatment. Doxycycline has been shown
useful in the treatment of filariasis following this discovery, In several series, 60% of infected patients seroconvert without
which is much less toxic than other current therapies (107b). apparent disease, 38% experience self-limiting mild disease
(111), and only 2% require exhaustive diagnostic procedures
Candidatus Neoehrlichia mikurensis and hospitalization. Months to years after initial infection,
This rare disease found in Europe and Asia can cause fever in 0.2% to 0.5% of patients develop cardiovascular infection
immunocompromised hosts. manifestations, usually those suffering from immunosuppres-
sion or a cardiac valve or vascular lesion (109a,112).
Patients diagnosed with acute infections present with a variety
Q FEVER of symptoms. Most common are isolated prolonged fever (14%)
(113), pneumonia (37%), and as the only symptom in 17%.
Q fever is a worldwide zoonosis caused by C. burnetii. Ungulates Hepatitis is found in 60% of patients and is the only sign in
and pets are the major reservoirs. The infection in humans is 40%. Some hepatitis cases are associated with an inflammatory
variable in severity, clinical expression, and natural course and syndrome and autoantibodies and may be resistant to antibiotic
may be acute or chronic. C. burnetii, a gram-negative bacte- treatment. When liver biopsies are performed, typical lesions are
rium, multiplies in acidic vacuoles within the monocytes of its lipid vacuoles surrounded by a fibrinoid ring in the form of a
host. Growing in vitro, C. burnetii becomes a deleted avirulent doughnut (Fig. 27.10). Patients may have a rash in 1.5% of cases.
mutant, which is named phase II and is useful in serologic diag- Patients may also have other cardiovascular manifestations such
nosis of acute infections (108). Coxiella organisms survive for as pericarditis or more rarely myocarditis (1% to 2%).
long periods in the environment and can be wind borne.
C. burnetii infection is incompletely eliminated after acute
infection in some hosts, for example, immunocompromised Neurologic Manifestations
patients and those with cardiac valve lesions. C. burnetii
organisms continue to multiply despite high antibody titers Severe headache is the most common CNS symptom. The neu-
and cause chronic infection. In patients with endocarditis, rologic manifestations of Q fever (Table 27.8) also include toxic
interleukin-10 (an antiinflammatory cytokine) is increased and confusional states, encephalitis, dementia, cerebellar symp-
monocytes are unable to control growth of C. burnetii (109). toms, cranial nerve palsies, psychoses, and motor and sensory
neuropathies (22,114). A few cases of encephalitis, meningo-
encephalitis, and encephalomyelitis have been reported late in
Epidemiology the course of acute Q fever (115117). Meningoencephalitis is
present in approximately 1% of patients with Q fever (118).
C. burnetii infects a wide range of animals, especially ungulates Seizures and coma may accompany the meningoencephali-
and cats, which are the most important vectors for people who tis of Q fever. A study (114) reported an incidence of neuro-
are usually infected by aerosols, or less frequently by ingesting logic complications among 22% of 103 patients, in which 46
milk products. Interhuman spread of infections during sexual had acute Q fever, 5 chronic Q fever, and 52 past infections.
intercourse, parturition, and blood transfusions has been re- Six of the forty-five patients with acute Q fever had residual
ported (108). In the past few years, major outbreaks have been neurologic impairment, weakness, blurred vision, recurrent
related to sheep and goats in the United States and Canada (108) meningismus, residual paresthesias, and sensory loss involving

Scheld_Ch27.indd 456 2/21/14 6:58 PM


Chapter 27: Rickettsioses, Anaplasmoses, and Q Fever 457

TA B L E 2 7 . 8 oligoamnios, intrauterine fetal death (IUFD), and premature


delivery (123a,123b).
NEUROLOGIC SYMPTOMS AND SYNDROMES Patients with Q fever endocarditis have a very chronic
ASSOCIATED WITH Q FEVER infection with low-grade fever, progressive deterioration of
valve function, and progressive heart failure. Fever is inter-
Symptoms/Syndromes Situation mittent and vegetations are frequently absent with echocar-
diography. If not diagnosed, the disease progressively worsens
Meningitis (aseptic) Acute Q fever and emboli (mainly cerebral) may be observed associated with
Encephalitis Acute Q fever renal insufficiency, splenomegaly, and hepatomegaly. Digital
Meningoencephalitis Acute Q fever clubbing may also be observed (108).
Guillain-Barr syndrome Acute Q fever Laboratory abnormalities include increases in hepatic
enzymes and leukopenia and thrombocytopenia, which are
Miller-Fisher syndrome Acute Q fever common. Circulating anticoagulant associated with antiphos-
Myelitis Acute Q fever pholipid may be observed, as may antismooth muscle
Fever and coma Acute Q fever antibodies. With endocarditis, antinuclear antibodies, micro-
Fever and seizures Acute Q fever hematuria, and serum rheumatoid factor are often found.
Cranial nerve palsy Acute Q fever
Pseudo herpetic encephalitis Acute Q fever
Diagnosis
Acute dementia Acute Q fever
Cerebellitis Acute Q fever Diagnosis is based mainly on serology. Direct detection of
Optic neuritis Acute Q fever
C. burnetii by culture, PCR, or immunochemistry in valve,
liver, or blood samples is useful, but serology by IFA is the
Hemiplegia Endocarditis reference method. Two antigens (phase I and phase II) can
Behavioral disturbance Postinfection be used in the IFAs. Acute Q fever is diagnosed when there is
Chronic fatigue Postinfection seroconversion or a fourfold increase is seen to phase II an-
tigen. A single serum sample with immunoglobulin G (IgG)
antibodies more than 200 and immunoglobulin M (IgM)
more than 50 against phase II is also diagnostic (124). In
the left leg (114). Behavioral disturbance, cerebellar symp- chronic Q fever, antibodies are at higher titers and directed
toms, cranial nerve palsies, extrapyramidal disease, and the against both phase I and II. IgG to phase I at a titer of 800
Miller-Fisher variant of the Guillain-Barr syndrome (areflexia or 1,600 is diagnostic of chronic infection, as is IgA of more
and ophthalmoparesis) have been reported as complications of than 100.
acute Q fever. The most common residual disorder of Q fever
meningitis is a disturbance of vision (119). In the series re-
ported by Derrick (120), 1 (0.3%) of 273 patients examined Treatment
had encephalomyelitis. In a report of neurologic involvement
in acute Q fever in 29 patients, we found 17 with meningoen- In vitro, several antimicrobials are bacteriostatic with mini-
cephalitis or encephalitis, 8 with meningitis and myelitis, and mum inhibitory concentrations compatible with clinical use.
4 with peripheral neuropathy (121). A review of 16 patients These include co-trimoxazole, doxycycline, rifampin, fluoro-
with Q fever meningoencephalitis (116) revealed that 8 pa- quinolones, and newer macrolides (but not erythromycin). All
tients had an elevated CSF WBC level, ranging from 18 to these compounds have been used with anecdotal success. Only
1,392 cells/mm3. In all but one case, mononuclear cells pre- doxycycline for acute Q fever (108) and co-trimoxazole for a
dominated. The EEG showed nonspecific abnormalities in five Q fever during pregnancy (125) have been proven effective.
of the six patients tested. None of these compounds is bactericidal in vitro, but doxy-
PostQ fever chronic fatigue was first reported in Australia. cycline together with chloroquine (which alkalinize the acidic
Patients presented with prolonged fatigue, arthralgia, myalgia, pH of the vacuole where C. burnetii organisms multiply) is
muscle fasciculation, blurred vision, sweats, and enlarged pain- bactericidal and has been proven the best antibiotic therapy
ful lymph nodes. A casecontrol study was conducted more for chronic Q fever (125a).
recently in the United Kingdom in 102 patients from a 1989 Treatment is straightforward in patients with acute Q fever,
Q fever outbreak, and pneumonia patients were used as con- but when patients spontaneously resolve, treatment is of un-
trols (122). Chronic fatigue syndrome was found more often in certain benefit. Doxycycline, the most effective antibiotic,
patients convalescing from acute Q fever. In some patients, the should be given for up to 2 weeks. The use of fluoroquino-
symptoms persisted for many years. Thus, human C. burnetii lones for patients with neurologic involvement was suggested
infection may induce a persistent debilitating syndrome in because of the good CSF penetration by the drugs, but this
convalescing patients, as is occasionally observed in patients is not supported by convincing clinical data. Some patients
with chronic typhoid fever or chronic brucellosis. However, with hepatitis do not respond well to antimicrobial therapy
this clinical manifestation is extremely rare in my experience because of an excessive immunologic response. They do, how-
in France. Whether this reflects differences in strains or other ever, improve rapidly with a short course of glucocorticoids
specificities is unknown (123). In the Netherlands, many cases (108). In pregnant women, it has been shown that taking
have been reported since the beginning of a large outbreak; co-trimoxazole for the duration of pregnancy avoids unfavor-
preliminary data suggest that behavioral psychotherapy may able outcomes (125).
be helpful in these cases. Bactericidal drugs are necessary in patients with endo-
Prognosis is usually favorable even without treatment, carditis (126,127). In vitro, antibiotic efficacy is impaired
except in certain patients. In pregnant women, with or with- by the low pH of the vacuole in which C. burnetii organ-
out symptoms, Q fever compromises the pregnancy, includ- isms reside. Hydroxychloroquine increases the pH of this
ing spontaneous abortion, intrauterine growth retardation, vacuole and restores the bactericidal effect of doxycycline.

Scheld_Ch27.indd 457 2/21/14 6:58 PM


458 Part III: Bacterial and Mycoplasmal Infection

In patients with endocarditis, the recommended treatment or more (127,128). The outcome could be favorable if there
is the concurrent use of doxycycline (200 mg daily) and OH is a four-time decrease of phase I IgG and IgA and the com-
chloroquine (600 mg daily, then adjusted to reach a 1-mg/ plete disappearance of IgM specific to phase II antigen with
mL plasma concentration). This regimen is given for 18 to no biologic or clinical sign of disease progression (125a).
36 months, depending on serologic responses. The major Serologic monitoring for at least 5 years seems appropriate
problem with this treatment is photosensitivity and expo- given the risk of relapse.
sure to the sun should be avoided. Alternative treatment is Prevention is based on veterinary control in animals. A Q fever
the concurrent use of doxycycline and ofloxacin for 3 years vaccine is available in Australia.

References
1. Raoult D, Roux V. Rickettsioses as paradigms of new or emerging infec- 26. Hattwick MA, OBrien RJ, Hanson BF. Rocky Mountain spotted fever:
tious diseases. Clin Microbiol Rev. 1997;10:694719. epidemiology of an increasing problem. Ann Inter Med. 1976;84:732739.
2. Raoult D, Olson JG. Emerging rickettsioses. In: Scheld WM, Craig WA, 27. Kirk JL, Fine DP, Sexton DJ, et al. Rocky Mountain spotted fever: a clinical
Hughes JM, eds. Emerging Infections 3. Washington, DC: ASM Press; review based on 48 confirmed cases, 19431986. Medicine (Baltimore).
1999:1735. 1990;69:3545.
2a. Socolovschi C, Pages F, Raoult D. Rickettsia felis in Aedes albopictus, the 28. Helmick CG, Bernard KW, DAngelo LJ. Rocky mountain spotted fever:
Asian tiger mosquito. Emerg Infect Dis. 2012;18(10):16871689. clinical, laboratory, and epidemiological features of 262 cases. J Infect Dis.
3. Dumler JS, Walker DH. Tick-borne ehrlichioses. Lancet Infect Dis. 1984;150:480488.
2001;1:2128. 29. Horney LF, Walker DH. Meningoencephalitis as a major manifestation of
4. Hansen W, Freney J. Le typhus pidmique, sa transmission et la dcou- Rocky Mountain spotted fever. South Med J. 1988;81:915918.
verte de lagent tiologique. Lyon Pharmaceutique. 1996;47:130138. 30. Westerman EL. Rocky Mountain spotless fever: a dilemma for the clinician.
5. Ricketts HT. Some aspects of Rocky Mountain spotted fever as shown by Arch Intern Med. 1982;142:11061107.
recent investigations. Rev Infect Dis. 1991;13:12271240. 31. Dimmitt DC, Fishbein DB, Dawson JE. Human ehrlichiosis associated
6. Tarasevich IV, Makarova V, Fetisova NF, et al. Astrakhan fever: new spot- with cerebrospinal fluid pleocytosis: a case report. Am J Med. 1989;87:
ted fever group rickettsiosis. Lancet. 1991;337:172173. 677678.
7. Stewart RS. Flinders Island spotted fever: a newly recognised endemic focus 32. Harrell GT. Rickettsial involvement of the central nervous system. Med
of tick typhus in Bass Strait. Part 1. Clinical and epidemiological features. Clin North Am. 1953;37:395422.
Med J Aust. 1991;154:9499. 33. Wright L. Intellectual sequelae of Rocky Mountain spotted fever. J Abnorm
8. Kelly PJ, Matthewman LA, Beati L, et al. African tick-bite fever: a new Psychol. 1972;80:315316.
spotted fever group rickettsiosis under an old name. Lancet. 1992;340: 34. Rosenblum MJ, Masland RL, Harrell GT. Residual effects of rickettsial
982983. disease on the central nervous system. Arch Intern Med. 1952;90:444455.
9. Mahara F, Koga K, Sawada S, et al. The first report of the rickettsial infec- 35. Raoult D, Parola P. Rocky Mountain spotted fever in the USA: a benign dis-
tions of spotted fever group in Japan: three clinical cases [in Japanese]. ease or a common diagnostic error? Lancet Infect Dis. 2008;8(10):587589.
Kansenshogaku Zasshi. 1985;59:11651171. 36. Raoult D, Lakos A, Fenollar F, et al. Spotless rickettsiosis caused by
10. Raoult D, Brouqui P, Roux V. A new spotted-fever-group rickettsiosis. Rickettsia slovaca and associated with Dermatocentor ticks. Clin Infect
Lancet. 1996;348:412. Dis. 2002;34:13311336.
11. Raoult D, Berbis P, Roux V, et al. A new tick-transmitted disease due to 36a. Angelakis E, Pulcini C, Waton J, et al. Scalp eschar and neck lymphade-
Rickettsia slovaca. Lancet. 1997;350:112113. nopathy caused by Bartonella henselae after tick bite. Clin Infect Dis.
12. Nilsson K, Lindquist O, Pahlson C. Association of Rickettsia helvetica 2010;50(4):549551.
with chronic perimyocarditis in sudden cardiac death. Lancet. 1999;354: 37. Raoult D, Weiller PJ, Chagnon A, et al. Mediterranean spotted fever: clini-
11691173. cal, laboratory and epidemiological features of 199 cases. Am J Trop Med
13. Raoult D, Fournier PE, Abboud P, et al. First documented human Rickettsia Hyg. 1986;35:845850.
aeschlimannii infection. Emerg Infect Dis. 2002;8:748749. 38. Araki M, Takatsuka K, Kawamura J, et al. Japanese spotted fever involving
14. Walker DH, Valbuena GA, Olano JP. Pathogenic mechanisms of diseases the central nervous system: two case reports and a literature review. J Clin
caused by Rickettsia. Ann N Y Acad Sci. 2003;990:111. Microbiol. 2002;40:38743876.
15. Parola P, Raoult D. Ticks and tickborne bacterial diseases in humans: an 39. Richter J, Fournier PE, Petridou J, et al. Rickettsia felis infection acquired
emerging infectious threat. Clin Infect Dis. 2001;32:897928. in Europe and documented by polymerase chain reaction. Emerg Infect
16. Rizzolo PJ, Addison L. Rocky Mountain spotted fever vs. viral syndrome: Dis. 2002;8:207208.
a management approach in a primary care setting. N C Med J. 1989;50: 40. Rolain JM, Franc M, Davoust B, et al. Molecular detection of Bartonella
181183. quintana, B. clarridgeiae and Rickettsia felis in cat fleas from France.
17. Fischer JJ. Rocky Mountain spotted fever: when and why to consider the Emerg Infect Dis. 2003;9(3):338342.
diagnosis. Postgrad Med. 1990;87:109118. 41. Schriefer ME, Sacci JB Jr, Dumler JS, et al. Identification of a novel rickett-
18. Walker DH. Rickettsia rickettsii and other spotted fever group rickett- sial infection in a patient diagnosed with murine typhus. J Clin Microbiol.
siae (rocky mountain spotted fever and other spotted fevers). In: Mandell 1994;32:949954.
GL, Bennett JE, Dolin R, eds. Principles and Practice of Infectious 42. Rolain JM, Sthul L, Maurin M, et al. Evaluation of antibiotic susceptibili-
Diseases. 7th ed. Philadelphia: Elsevier Churchill Livingstone; 2009: ties of three rickettsial species including Rickettsia felis by a quantitative
chap 187. PCR DNA assay. Antimicrob Agents Chemother. 2002;46:27472751.
19. Duval J, Kaplowitz LG, Fisher JJ, et al. Rocky mountain spotted fever: a 42a. Mediannikov O, Fenollar F, Bassene H, et al. Description of yaaf, the
clinical dilemma. In: Remington JB, Swartz HN, eds. Current Clinical vesicular fever caused by acute Rickettsia felis infection in Senegal. J Infect.
Topics in Infectious Diseases. 2nd ed. New York: McGraw Hill; 1981: 2013;66(6):536540.
89108. 43. Brettman LR, Lewin S, Holzman RS, et al. Rickettsialpox: report of an out-
20. Botelho-Nevers E, Socolovschi C, Raoult D, et al. Treatment of Rickettsia break and a contemporary review. Medicine (Baltimore). 1981;60:363372.
spp. infections: a review. Expert Rev Anti Infect Ther. 2012;10(12): 44. Paddock CD, Zaki SR, Koss T, et al. Rickettsialpox in New York City: a
14251437. persistent urban zoonosis. Ann N Y Acad Sci. 2003;990:2636.
21. Walker DH, Hawkins HK, Hudson P. Fulminant Rocky Mountain 45. Raoult D, Roux V. The body louse as a vector of reemerging human dis-
spotted fever: its pathologic characteristics associated with glucose-6- eases. Clin Infect Dis. 1999;29:888911.
phosphate dehydrogenase deficiency. Arch Pathol Lab Med. 1983;107: 46. Raoult D, Roux V, Ndihokubwaho JB, et al. Jail fever (epidemic typhus)
121125. outbreak in Burundi. Emerg Infect Dis. 1997;3:357360.
22. Marrie TJ, Raoult D. Rickettsial infections of the central nervous system. 47. Fournier PE, Ndihokubwaho JB, Guidran J, et al. Prevalence of human
Semin Neurol. 1992;12:213224. pathogens in body and head lice. Emerg Infect Dis. 2002;18:15151518.
23. Weber DJ, Walker DH. Rocky Mountain spotted fever. Infect Dis Clin 48. Fang R, Houhamdi L, Raoult D. Detection of Rickettsia prowazekii in
North Am. 2002;5:1935. body lice and their feces by using monoclonal antibodies. J Clin Microbiol.
24. Hazard GW, Ganz RN, Nevin RW, et al. Rocky Mountain spotted fever in 2002;40:33583363.
the eastern United States. N Engl J Med. 1969;280:5762. 49. Massung RF, Davis LE, Slater K, et al. Epidemic typhus meningitis in the
25. Sexton DJ, Burgdorfer W. Clinical and epidemiologic features of Rocky southwestern United States. Clin Infect Dis. 2001;32:979982.
Mountain spotted fever in Mississippi, 19331973. South Med J. 1975;68: 50. Raoult D, Ndihokubwayo JB, Tissot-Dupont H, et al. Outbreak of epidemic
15291535. typhus associated with trench fever in Burundi. Lancet. 1998;352:353358.

Scheld_Ch27.indd 458 2/21/14 6:58 PM


Chapter 27: Rickettsioses, Anaplasmoses, and Q Fever 459

51. Stein A, Purgus R, Olmer M, et al. Brill-Zinsser disease in France. Lancet. 84. Balayeva NM, Eremeeva ME, Tissot-Dupont H, et al. Genotype charac-
1999;353:1936. terization of the bacterium expressing the male-killing trait in the ladybird
52. Dumler JS, Taylor JP, Walker DH. Clinical and laboratory features of beetle Adalia bipunctata with specific rickettsial molecular tools. Appl
murine typhus in South Texas, 1980 through 1987. JAMA. 1991;266: Environ Microbiol. 1995;61:14311347.
13651370. 85. Wood DO, Williamson LR, Winkler HH, et al. Nucleotide sequence of
53. Parola P, Vogelaers D, Roure C, et al. Murine typhus in travelers returning the Rickettsia prowazekii citrate synthase gene. J Bacteriol. 1987;169:
from Indonesia. Emerg Infect Dis. 1998;4:677680. 35643572.
54. Tselentis Y, Edlinger E, Alexious G, et al. An endemic focus of murine 86. Gilmore RD, Cieplak W, Policastro PF, et al. The 120 kilodalton outer
typhus in Europe. J Infect. 1986;13:9192. membrane protein (rOmpB) of Rickettsia rickettsii is encoded by an un-
55. Gikas A, Doukakis S, Pediaditis J, et al. Murine typhus in Greece: epide- usually long open reading frame: evidence for protein processing from a
miological, clinical, and therapeutic data from 83 cases. Trans R Soc Trop large precursor. Mol Microbiol. 1991;5:23612370.
Med Hyg. 2002;96:250253. 87. Roux V, Fournier PE, Raoult D. Differentiation of spotted fever group
56. Silpapojakul K, Ukkchoke C, Krisanapan S. Rickettsial meningitis and rickettsiae by sequencing and analysis of restriction fragment length
encephalitis. Arch Intern Med. 1991;151:17531757. polymorphism of PCR amplified DNA of the gene encoding the protein
57. Manor E, Politi F, Marmor A, et al. Papilledema in endemic typhus. Am J rOmpA. J Clin Microbiol. 1996;34:20582065.
Ophthalmol. 1977;84:559562. 88. Woodward TE, Pedersen CE Jr, Oster CN, et al. Prompt confirmation of
58. Kawamura A, Tanaka H, Tamura A. Tsutsugamushi Diseases. Tokyo, Rocky Mountain spotted fever: identification of rickettsiae in skin tissues.
Japan: University of Tokyo Press; 1995. J Infect Dis. 1976;136(3):297301.
59. Weiss E. The biology of rickettsiae. Annu Rev Microbiol. 1982;36:345370. 89. Walker DH, Cain BG, Olmstead PM. Laboratory diagnosis of Rocky
60. Kim DE, Lee SH, Park KI, et al. Scrub typhus encephalomyelitis with prom- Mountain spotted fever by immunofluorescent demonstration of
inent focal neurologic signs. Arch Neurol. 2000;57:17701772. Rickettsia in cutaneous lesions. Am J Clin Pathol. 1978;69:619623.
61. Watt G, Kantipong P, Jongsakul K, et al. Doxycycline and rifampicin for 90. Walker DH, Gay RM, Valdes-Dapena M. The occurrence of eschars in
mild scrub-typhus infections in northern Thailand: a randomised trial. Rocky Mountain spotted fever. J Am Acad Dermatol. 1981;4:571576.
Lancet. 2000;356:10571061. 91. Lepidi H, Fournier PE, Raoult D. Histologic features and immunodetection
62. Watt G, Kantipong P, de Souza M, et al. HIV-1 suppression during acute of African tick-bite fever eschar. Emerg Infect Dis. 2006;12(9):13321337.
scrub-typhus infection. Lancet. 2000;356:475479. 92. Bacellar F, Regnery RL, Nuncio MS, et al. Genotypic evaluation of rickett-
63. Raoult D, Dasch GA. Immunoblot cross-reactions among Rickettsia, sial isolates recovered from various species of ticks in Portugal. Epidemiol
Proteus spp. and Legionella spp. in patients with Mediterranean spotted Infect. 1995;114:169178.
fever. FEMS Immunol Med Microbiol. 1995;11:1318. 93. Marrero M, Raoult D. Centrifugation-shell vial technique for rapid detec-
64. Hechemy KE, Raoult D, Eisemann C, et al. Detection of antibod- tion of Mediterranean spotted fever rickettsia in blood culture. Am J Trop
ies to Rickettsia conorii with a latex agglutination test in patients with Med Hyg. 1989;40:197199.
Mediterranean spotted fever. J Infect Dis. 1986;153:132135. 94. Espejo-Arenas E, Raoult D. First isolates of Rickettsia conorii in Spain
65. Hechemy KE, Michaelson EE. Recent advances in the immunoserology of using a centrifugation-shell vial assay. J Infect Dis. 1989;159:11581159.
Rickettsioses. J Med Microbiol. 1985;1314:511. 95. Rolain JM, Maurin M, Vestris G, et al. In vitro susceptibilities of 27 rick-
66. Teysseire N, Raoult D. Comparison of Western immunoblotting and micro- ettsiae to 13 antimicrobials. Antimicrob Agents Chemother. 1998;42:
immunofluorescence for diagnosis of Mediterranean spotted fever. J Clin 15371541.
Microbiol. 1992;30:455460. 96. Raoult D, Drancourt M. Antimicrobial therapy of rickettsial diseases.
67. Philip RN, Casper EA, Burgdorfer W, et al. Serologic typing of rickett- Antimicrob Agents Chemother. 1991;35:24572462.
siae of the spotted fever group by micro immunofluorescence. J Immunol. 97. Holman RC, Paddock CD, Curns AT, et al. Analysis of risk factors for
1978;121:19611968. fatal rocky mountain spotted fever: evidence for superiority of tetracy-
68. Raoult D, Fournier PE, Fenollar F, et al. Rickettsia africae, a tick-borne patho- clines for therapy. J Infect Dis. 2001;184:14371444.
gen in travelers to sub-Saharan Africa. N Engl J Med. 2001;344:15041510. 98. Zanetti G, Francioli P, Tagan D, et al. Imported epidemic typhus. Lancet.
69. La Scola B, Rydkina L, Ndihokubwayo JB, et al. Serological differentiation 1998;352:1709.
of murine typhus and epidemic typhus using cross-adsorption and Western 99. Rolain JM, Raoult D. Treatment of infections caused by intracellular
blotting. Clin Diagn Lab Immunol. 2000;7:612616. pathogens. In: Hooper D, Rubinstein E, eds. Quinolone Antimicrobial
70. Roux V, Raoult D. Body lice as tools for diagnosis and surveillance of Agents. 3rd ed. Boston: American Society for Microbiology; 2002.
reemerging diseases. J Clin Microbiol. 1999;37:596599. 100. Maeda K, Markowitz N, Hawley RC, et al. Human infection with
71. La Scola B, Raoult D. Diagnosis of Mediterranean spotted fever by cul- Ehrlichia canis, a leukocytic Rickettsia. N Engl J Med. 1987;316:853856.
tivation of Rickettsia conorii from blood and skin samples using the 100a. Pritt BS, Sloan LM, Johnson DK, et al. Emergence of a new pathogenic
centrifugation-shell vial technique and by detection of R. conorii in cir- Ehrlichia species, Wisconsin and Minnesota, 2009. N Engl J Med.
culating endothelial cells: a 6-year follow-up. J Clin Microbiol. 1996;34: 2011;365(5):422429. doi:10.1056/NEJMoa1010493.
27222727. 100b. Yabsley MJ. Natural history of Ehrlichia chaffeensis: vertebrate hosts and
72. Stein A, Raoult D. A simple method for amplification of DNA from paraf- tick vectors from the United States and evidence for endemic transmission
fin-embedded tissues. Nucleic Acids Res. 1992;20:52375238. in other countries. Vet Parasitol. 2010;167:136.
72a. Mouffok N, Parola P, Raoult D. Murine typhus, Algeria. Emerg Infect Dis. 101. Ratnasamy N, Everett ED, Roland WE, et al. Central nervous system
2008;14(4):676678. manifestations of human ehrlichiosis. Clin Infect Dis. 1996;23:314319.
73. Williams WJ, Radulovic S, Dasch GA, et al. Identification of Rickettsia 101a. Dumler JS, Madigan JE, Pusterla N, et al. Ehrlichioses in humans: epide-
conorii infection by polymerase chain reaction in a soldier returning from miology, clinical presentation, diagnosis, and treatment. Clin Infect Dis.
Somalia. Clin Infect Dis. 1994;19:9399. 2007;45(suppl 1):S45S51.
74. Brouqui P, Harle JR, Delmont J, et al. African tick bite fever: an imported 102. Bakken JS, Krueth J, Wilson-Nordskog C, et al. Clinical and laboratory
spotless rickettsiosis. Arch Intern Med. 1997;157:119124. characteristics of human granulocytic ehrlichiosis. JAMA. 1996;275:
75. Sanogo O, Zeaiter Z, Caruso G, et al. Detection of Bartonella henselae 199205.
in ticks Ixodes ricinus (Acari: Ixodida) removed from humans in Belluno 103. Bakken JS, Erlemeyer SA, Kanoff RJ, et al. Demyelinating polyneuropa-
Province, Italy. Emerg Infect Dis. 2003;9:329332. thy associated with human granulocytic ehrlichiosis. Clin Infect Dis.
76. Shpynov S, Parola P, Rudakov N, et al. Detection and identification of spot- 1998;27:13231324.
ted fever group rickettsiae in Dermatocentor ticks from Russia and central 104. Horowitz HW, Marks SJ, Weintraub M, et al. Brachial plexopathy
Kazakhstan. Eur J Clin Microbiol Infect Dis. 2001;20:903905. associated with human granulocytic ehrlichiosis. Neurology. 1996;46:
77. Gage K, Schrumpf ME, Karstens RH, et al. DNA typing of rickettsiae in 10261029.
naturally infected ticks using a polymerase chain reaction/restriction frag- 104a. Demma LJ, Holman RC, McQuiston JH, et al. Epidemiology of human
ment length polymorphism system. Am J Trop Med Hyg. 1994;50:247260. ehrlichiosis and anaplasmosis in the United States, 20012002. Am J Trop
78. Gage KL, Gilmore RD, Karstens RH, et al. Detection of Rickettsia rickettsii Med Hyg. 2005;73:400409.
in saliva, hemolymph and triturated tissues of infected Dermacentor ander- 105. Buller RS, Arens M, Hmiel SP, et al. Ehrlichia ewingii, a newly recognized
soni ticks by polymerase chain reaction. Mol Cell Probes. 1992;6:333341. agent of human ehrlichiosis. N Engl J Med. 1999;341:148155.
79. Higgins JA, Azad AF. Use of polymerase chain reaction to detect bacteria in 106. Yabsley MJ, Varela AS, Tate CM, et al. Ehrlichia ewingii infection in white-
arthropods: a review. J Med Entomol. 1995;32:213222. tailed deer (Odocoileus virginianus). Emerg Infect Dis. 2002;8:668671.
80. Roux V, Raoult D. Phylogenetic analysis of the genus Rickettsia by 16S 107. Masters EJ, Storch GA, Sumner JW. Ehrlichia ewingii in an immunocom-
rDNA sequencing. Res Microbiol. 1995;146:385396. petent adult. Mo Med. 2009;106(4):301303.
81. Pretzman C, Ralph D, Stothard DR, et al. 16S rRNA gene sequence of 107a. Newton PN, Rolain JM, Rasachak B, et al. Sennetsu neorickettsiosis: a
Neorickettsia helminthoeca and its phylogenetic alignment with members probable fish-borne cause of fever rediscovered in Laos. Am J Trop Med
of the genus Ehrlichia. Int J Syst Bact. 1995;45:207211. Hyg. 2009;81:190194.
82. Anderson BE, Tzianabos T. Comparative sequence analysis of a genus- 107b. Mand S, Debrah AY, Klarmann U, et al. Doxycycline improves filarial
common rickettsial antigen gene. J Bacteriol. 1989;171:51995201. lymphedema independent of active filarial infection: a randomized con-
83. Baird RW, Stenos J, Stewart R, et al. Genetic variation in Australian spotted trolled trial. Clin Infect Dis. 2012;55(5):621630.
fever group rickettsiae. J Clin Microbiol. 1996;34:15261530. 108. Maurin M, Raoult D. Q fever. Clin Microbiol Rev. 1999;12:518553.

Scheld_Ch27.indd 459 2/21/14 6:58 PM


460 Part III: Bacterial and Mycoplasmal Infection

109. Raoult D, Mege JL, Marrie TJ. Q fever: queries remaining after decades of 121. Bernit E, Pouget J, Janbon F, et al. Neurological involvement in acute
research. In: Scheld M, Craig WA, Hugues JM, eds. Emerging Infections. Q fevera report of 29 cases and review of the literature. Arch Intern
5th ed. Washington, DC: ASM Press; 2001:2956. Med. 2002;162:693700.
109a. Raoult D. Chronic Q fever: expert opinion versus literature analysis and 122. Ayres JG, Flint N, Smith EG, et al. Post-infection fatigue syndrome fol-
consensus. J Infect. 2012;65(2):102108. lowing Q fever. Q J Med 1998;91:105123.
110. Maltezou HC, Raoult D. Q fever in children. Lancet Infect Dis. 123. Raoult D. Q fever: still a mysterious disease. Q J Med 2002;95:491492.
2002;2:686691. 123a. Angelakis E, Million M, DAmato F, et al. Q fever and pregnancy: dis-
111. Dupuis G, Peter O, Luthy R, et al. Serological diagnosis of Q fever endo- ease, prevention, and strain specificity. Eur J Clin Microbiol Infect Dis.
carditis. Eur Heart J. 1986;7:10621066. 2013;32(3):361368. doi:10.1007/s10096-012-1750-3.
112. Fenollar F, Fournier PE, Carrieri MP, et al. Risks factors and prevention of 123b. Carcopino X, Raoult D, Bretelle F, et al. Q Fever during pregnancy: a
Q fever endocarditis. Clin Infect Dis. 2001;33:312316. cause of poor fetal and maternal outcome. Ann N Y Acad Sci. 2009;1166:
113. Raoult D, Tissot-Dupont H, Foucault C, et al. Q fever 19851998 7989. doi:10.1111/j.1749-6632.2009.04519.x.
clinical and epidemiologic features of 1,383 infections. Medicine. 124. Tissot-Dupont H, Thirion X, Raoult D. Q fever serology: cutoff deter-
2000;79:109123. mination for microimmunofluorescence. Clin Diagn Lab Immunol.
114. Reilly S, Northwood JL, Caul EO. Q fever in Plymouth, 19721988: a 1994;1:189196.
review with particular reference to neurological manifestations. Epidemiol 125. Raoult D, Fenollar F, Stein A. Q fever during pregnancydiagnosis, treat-
Infect. 1990;105:391408. ment, and follow-up. Arch Intern Med. 2002;162:701704.
115. Somma-Moreira RE, Caffarena RM, Somma S, et al. Analysis of Q fever 125a. Million M, Thuny F, Richet H, et al. Long-term outcome of Q fever endo-
in Uruguay. Rev Infect Dis. 1987;9:386387. carditis: a 26-year personal survey. Lancet Infect Dis. 2010;10(8):527535.
116. Marrie TJ. Pneumonia and meningo-encephalitis due to Coxiella burnetii. 126. de Lamballerie X, Charrel RN, Attoui H, et al. Classification of hepatitis
J Infect. 1985;11:5961. C virus variants in six major types based on analysis of the envelope 1 and
117. Powell O. Q fever: clinical features in 72 cases. Australas Ann Med. nonstructural 5B genome regions and complete polyprotein sequences.
1960;9:214223. J Gen Virol. 1997;78(pt 1):4551.
118. Drancourt M, Raoult D, Xeridat B, et al. Q fever meningoencephalitis in 127. Raoult D, Houpikian P, Tissot-Dupont H, et al. Treatment of Q fever
five patients. Eur J Epidemiol. 1991;7:134138. endocarditis: comparison of two regimens containing doxycycline and
119. Ferrante MA, Dolan MJ. Q Fever meningoencephalitis in a soldier return- ofloxacin or hydroxychloroquine. Arch Intern Med. 1999;159:167173.
ing from the Persian Gulf war. Clin Infect Dis. 1993;16:489496. 128. Levy PY, Drancourt M, Etienne J, et al. Comparison of different antibi-
120. Derrick EH. The course of infection with Coxiella burnetii. Med J Aust. otic regimens for therapy of 32 cases of Q fever endocarditis. Antimicrob
1973;1:10511057. Agents Chemother. 1991;35:533537.

Scheld_Ch27.indd 460 2/21/14 6:58 PM


CHAPTER 28 WHIPPLES DISEASE
MATTHIAS MAIWALD AND DAVID A. RELMAN

In a seminal autopsy report in 1907, George H. Whipple pro- to areas of pathology (14,28). Finally, after several decades of
vided a thorough and articulate description of the disease that unsuccessful attempts, the bacterium was isolated in culture,
now bears his name (1). His publication illustrated nearly all first from a heart valve specimen (29) and subsequently from
the key features that define this clinical entity. However, there other clinical specimen types, including intestinal tissue (30)
was no mention of neurologic manifestations, and no postmor- and cerebrospinal fluid (CSF) (31).
tem examination of the central nervous system (CNS). More The Whipples disease bacterium (T. whipplei) is rod-
than 50 years later, the incidence and significance of CNS in- shaped, measures about 0.2 m by 1 to 2 m, and is sur-
volvement in Whipples disease began to be appreciated (2). rounded by a 20-nm-thick cell wall. The outermost layer
We now know that Whipples disease is a systemic disorder surrounding the bacterium consists of a symmetric membrane
that typically affects the gastrointestinal tract and that the CNS that morphologically resembles those of eukaryotic origin
is one of the most common sites of extraintestinal involvement, (25). An inner electron-dense layer within the wall reacts
along with the lymphatic system and the heart (3,4). In addi- strongly with periodic acidSchiff (PAS) reagent and accounts
tion, the CNS is the most common site of relapse after anti- for the PAS-positive staining pattern of the bacteria and their
microbial treatment of non-CNS disease (5). Furthermore, it remnants within macrophage vacuoles. The organism stains
has been suggested that early CNS infection occurs in most weakly gram positive and is not acid fast. Examples of typi-
or all patients with Whipples disease, although only a subset cal morphology, when viewed by electron microscopy, are
of patients develop symptomatic or radiologically apparent shown in Figure 28.1 and in several references (24,25,27).
CNS disease (6). Characterization of the bacterium at the molecular level was
Classical Whipples disease primarily affects middle-aged initially achieved by broad-range 16S ribosomal DNA (rDNA)
men and usually presents with arthralgias, abdominal pain, polymerase chain reaction (PCR): a unique bacterial 16S ribo-
fever, diarrhea, malabsorption, and weight loss (4,712). A fas- somal RNA (rRNA) gene sequence was found directly in af-
tidious bacterium, Tropheryma whipplei, is the causal agent of fected tissues (13,14). Phylogenetic analysis indicated that the
the disease; it is an actinomycete (class Actinobacteria) on the bacterium belongs to the actinomycetes (13,14); however, its
basis of molecular phylogenetic analysis (1316). Whipples relationships to the other known actinomycetes are relatively
disease can present with protean manifestations, especially in distant (15). The name Tropheryma whipplei (initially pro-
the setting of symptomatic CNS involvement. Thus, delays posed as Tropheryma whippelii) is based on the Greek words
or failures in the diagnosis of this disease are common. The trophe, nourishment, and eryma, barrier, because of the mal-
prognosis of CNS Whipples disease is generally serious and is absorption in classical Whipples disease and to honor George
worsened by delays in, or withholding of appropriate antimi- Whipple as the discoverer of the illness (14,16,32).
crobial therapy (5,17). Cultivation of T. whipplei in the laboratory was finally
This chapter focuses on the CNS aspects of Whipples disease. successful in prolonged co-culture with human fibroblast
Relatively little attention is given to the systemic (non-CNS) fea-
tures of this disorder. These have been well described in a num-
ber of other reviews (4,10,1821). Because it is an extension of
the CNS, a discussion of Whipples disease involving the eye is
included (see the section Clinical Manifestations).

ETIOLOGY
George Whipple concluded from histology and special stains
that the subject of his case report suffered from a disorder
of lipid metabolism (1). Although he observed bacilliform
structures in silver-stained sections of a lymph node, he did
not interpret this finding as related to the pathogenesis of the
disease. However, beginning in the 1950s and 1960s, evidence
started to accumulate that Whipples disease is caused by a
distinct species of bacterium. First, bacteria with a charac-
teristic uniform morphology are consistently seen by electron
microscopy in pathologically affected tissues (2225). These
organisms are usually numerous, are recognized and phago-
cytosed by tissue macrophages, and undergo binary fission in
areas of pathology. Second, patients with clinical manifesta-
tions of Whipples disease respond to antibacterial therapy FIGURE 28.1 Electron micrograph of Whipple bacillus undergoing
(4,26). Third, intact bacteria disappear with clinical response binary fission within the vitreous of a patient with chronic uveitis.
to antibiotics and reappear during clinical relapse (27). (From Rickman LS, Freeman WR, Green WR, et al. Brief report: uve-
Fourth, specific bacterial DNA sequences from T. whipplei itis caused by Tropheryma whippelii [Whipples bacillus]. N Engl J
are consistently detected in and have been shown to hybridize Med. 1995;332:363366, with permission.)

461

Scheld_Ch28.indd 461 2/21/14 8:10 PM


462 Part III: Bacterial and Mycoplasmal Infections

cells (29). In the initial culture setup, a bacterial generation differences in strain distribution have been noticed, so far, no
time of about 18 days was observed (29), but subsequent re- specific associations between different T. whipplei strains and
ports estimated the doubling time at between 28 hours and particular disease manifestations or carrier states have been
4 days (31,33,34). In any case, this is among the slowest identified (36).
growth rates ever reported for a medically relevant bacterium.
Subsequently, a cell-free culture medium was designed, con-
sisting of tissue culture medium supplemented with amino EPIDEMIOLOGY
acids which the bacterium cannot synthesize on its own (34),
and additional strainsboth in cell culture and in cell-free me- Classical Whipples disease with intestinal involvement is a
diumhave been cultivated from various other clinical speci- rare entity. It has been estimated that about 20 to 30 cases of
men types (35,36). Of relevance for the neurologic aspects of Whipples disease are reported each year in the literature (4)
Whipples disease is the successful isolation of T. whipplei from and that the total number of published cases since Whipples
CSF specimens; this includes CSF from neurologically symp- original description is less than 2,000 (12). Available epidemi-
tomatic as well as asymptomatic patients and from a patient ologic data indicate that the disease almost exclusively affects
in the treatment-free period after prolonged antibiotic therapy Caucasians, that the mean age at diagnosis is 50 to 60 years,
(31,37). These findings indicate that T. whipplei is viable and that there is a strong proclivity for males, and that people in
presumably is able to replicate in the CNS. Nevertheless, at the farming trades and other outdoor professions are propor-
the present time, culture is not a suitable tool for routine diag- tionately overrepresented (4,1012,58). However, available
nostic purposes in the workup of possible Whipples disease. incidence estimates almost certainly underrepresent the true
Two T. whipplei isolates from culture, one from CSF and number of cases and tend to exclude cases with nonclassical
another from an infected heart valve, served as the basis for pathology or purely extraintestinal disease. In fact, recent re-
genome sequencing projects (38,39). It is now established that ports indicate that the involvement of T. whipplei in culture-
the genome of T. whipplei is quite small, slightly less than 1 negative endocarditis without intestinal manifestations may be
Mbp, and bears the features of a reduced-genome organism more common than would be expected from the incidence of
that is dependent on a host for synthesis of amino acids and classical Whipples disease, suggesting that endocarditis might
for energy metabolism. At the same time, there is a relative follow a different epidemiologic pattern (59,60). Other re-
abundance of predicted surface molecules, and this includes ports implicate T. whipplei in acute transient illnesses, such as
a unique protein family, termed the WiSP family (for T. whip- episodes of febrile illnesses in Senegal (61) and acute diarrhea
plei surface proteins). The genome also possesses a number of in children in France (56), although the causative role of the
built-in mechanisms to create genetic and antigenic varia- organism in these instances is uncertain.
tion, and these are predicted to generate considerable variabil- Epidemiologic assessment of CNS Whipples disease is
ity among the organisms surface molecules. It is hypothesized hampered by frequent uncertainties surrounding the diagnosis
that these features are linked to immune evasion and thus to of cases. Often, investigators diagnose CNS Whipples disease
the organisms ability to sustain a chronic infection. on the basis of CNS clinical manifestations plus confirmed di-
The natural occurrence and reservoirs of T. whipplei are agnosis of Whipples disease pathology at other anatomic sites,
a topic of ongoing research and debate. The first record of typically in the small intestines or mesenteric lymph nodes. In
the bacteriums occurrence outside diseased human tissues was cases with positive histopathology in the small intestine and
provided by the detection of specific sequences in wastewater confirmed by either electron microscopy or a well-validated
from sewage treatment plants (40), suggesting that T. whip- PCR test, the diagnosis is firm (4,18,62). However, there is
plei may have an environmental reservoir and reside within an increasing number of reports and case series of presumed,
polymicrobial communities. Several diagnostic PCR studies isolated CNS Whipples disease in the absence of other organ
only rarely found positive results in pathologically unaffected manifestations (6366). In a considerable fraction of these
human tissues (4144). However, several other studies found published cases, the diagnosis has been made solely on the
T. whippleispecific DNA sequences in saliva, gastric juice, in- basis of positive PAS staining results and/or insufficiently vali-
testinal biopsies, and stool of asymptomatic persons (4549), dated PCR tests. As discussed in the section Diagnosis and
prompting debate as to the reliability and generalizability of Differential Diagnosis (later in this chapter), such diagnostic
these findings (20,50). More recent studies have reported posi- approaches lack the necessary specificity to be able to confirm
tivity rates of 2% to 4% in stool and 0.2% in saliva of healthy CNS Whipples disease.
adults and 8% in stool and 2% in saliva of sewage workers, Current literature suggests that the demographics of CNS
both in France, and the results were confirmed by a second Whipples disease are roughly similar to those of Whipples
PCR assay (48,49). Further evidence was provided with the disease cases involving all sites, although relevant informa-
detection of T. whipplei in stool and saliva of asymptomatic tion is limited. In the large review by Dobbins (4), among
household members of patients with Whipples disease, includ- 28 patients with CNS Whipples disease for whom an age is
ing some of the same bacterial genotypes as in the respective listed, the mean is 40 years. A published series of 11 cases
patients (51). As a consequence, it has been postulated that of CNS Whipples disease lists a 9:2 male:female ratio and a
there is an asymptomatic carrier state with T. whipplei and mean age at onset of 49 years (67). Another review of CNS
that the organism is a commensal bacterium of humans that Whipples disease (68) provides a mean age of 50 years and a
causes Whipples disease only in a subset of predisposed indi- male:female ratio of 8:2 among a group of 70 patients with
viduals (20,36,50). predominant but not exclusive neurologic presentation, and
Differences between T. whipplei strains were first detected a mean age of 42 years and a male:female ratio of about 1:1
in the 16S-23S rRNA intergenic spacer sequence, and up to among a group of 21 patients with presumed isolated neuro-
seven different spacer sequence types have been described logic manifestations.
(5254). Subsequent analyses found several highly variable Among several published case series of systemic and/
genomic sequences (HVGS) that formed the basis for an im- or classical intestinal Whipples disease, the proportion of
proved strain typing system (55); this genotyping system has patients with neurologic manifestations ranges between
been used in several epidemiologic investigations in France and slightly less than 10% and slightly more than 40% (712).
in Senegal (51,56,57). However, although some geographical Three case series (810) list neurologic symptoms as the

Scheld_Ch28.indd 462 2/21/14 8:10 PM


Chapter 28: Whipples Disease 463

presenting symptomsalthough that does not mean sole or Also, there is no significantly increased incidence of Whipples
isolated CNS diseasein 4% of cases each. The overall in- disease in patients with known primary immunodeficiencies
cidence and the proportion of truly isolated CNS Whipples or immunosuppressive therapies and instead only reports that
disease are too rare and too small to allow for any meaningful immunosuppressive therapy may hasten the onset of clinically
estimates. Patients who are symptomatic may be only a small apparent Whipples disease in patients with prodromal articular
subset of those with bacterial infection in the CNS or with manifestations (84,85). Surprisingly, one sequence-based study
CNS pathology. This follows from two types of observations. of the lung microbiome (i.e., the lungs microbial content and
First, a PCR-based study (69) demonstrated positive results composition) (86) found T. whipplei DNA in HIV-infected in-
for T. whipplei in CSF in 7 out of 10 (70%) neurologically dividuals. Furthermore, in 11 of the 82 HIV-positive subjects,
asymptomatic patients at the time of diagnosis of intestinal T. whipplei sequences dominated the microbial community
Whipples disease. Second, autopsy cases from the preantibi- (50% of sequence reads). The clinical significance of these
otic era had a high proportion of pathologic findings in the findings is unclear because none of the individuals suffered
brain. Sieracki et al. (2) described extensive CNS involvement from respiratory complaints or known lung pathology, but
at autopsy in two patients with Whipples disease who never these findings also suggest that T cellbased immunity is impor-
reported CNS symptoms. In a series of postmortem examina- tant in controlling the organism.
tions on patients with Whipples disease described by Enzinger Based on the detection of T. whipplei in acute self-limited
and Helwig (3), 10 of 11 patients had characteristic brain illnesses and also in asymptomatic people, it has been proposed
lesions associated with this disease. It has been suggested that T. whipplei may be transmitted from infected hosts or
that isolated CNS manifestations as primary presentations the environment, possibly in early childhood; may cause acute
of Whipples disease may be increasing in frequency and that primary infections in some; may go on to be carried by some;
this may be attributed to the incidental use of antibiotics that and may cause classical Whipples disease only in those who
suppress early, presymptomatic non-CNS disease but do not lack the immune response to contain the organism (87,88).
penetrate well enough into the CNS to halt progression there However, at this point, available data are not sufficient to be
(70). Again, the numbers of isolated CNS cases are insuffi- able to confirm these proposals. Taken together, there is good
cient for a careful analysis of this issue, and increasing disease evidence that immunologic factors play an important role in
awareness may be an alternative explanation. pathogenesis, but the abnormalities identified so far are com-
paratively modest and mainly manifest as immunologic dys-
regulation between a Th1 and Th2 response.
PATHOGENESIS AND Current concepts hold that the disease process is initiated
PATHOPHYSIOLOGY by translocation of the bacterium from the intestinal lumen
into the intestinal mucosa, possibly via invasion of intestinal
The pathogenesis of Whipples disease and of T. whipplei in- epithelial cells or through epithelial intercellular junctions,
fection is an evolving field. A number of immunologic abnor- followed by crossing of the basement membrane and replica-
malities have been identified in patients with Whipples disease, tion in the intestinal lamina propria (24,89). There, the bac-
and these are presumed to play a role in pathogenesis by way teria are ingested by macrophages and appear to spread via
of increasing host susceptibility to infection. However, so far blood vessels or via lymphatics into mesenteric lymph nodes
these abnormalities are all of a quantitative nature: No clear and into the systemic circulation. Infected blood monocytes
genetic defects or dichotomous presence or absence of any are possible vehicles for dissemination to other organs (90).
defined phenotypic markers have been described. Identified Uncertainty still exists concerning the preferred compartment
abnormalities include (a) diminished ability of macrophages for bacterial multiplication. Earlier electron microscopic stud-
of Whipples disease patients to degrade intracellular micro- ies indicated that most intact bacterial structures in untreated
organisms (7173); (b) reduced interleukin-12 production by intestinal Whipples disease are extracellular, just beneath the
patients peripheral blood monocytes upon stimulation with intestinal basement membrane, whereas ingested bacteria
bacterial antigens (74); (c) dysregulation of mononuclear cell in macrophages exist both with intact shapes and in vary-
function such that the components of a T helper 1 (Th1) im- ing stages of degradation (24,25). In situ hybridization local-
mune response are reduced and those of a T helper 2 (Th2) ized the 16S rRNA of T. whipplei (corresponding to intact
immune response increased (75); (d) diminished Th1 reactiv- bacteria) to the extracellular spaces of the intestinal lamina
ity of peripheral and duodenal CD4 T cells upon stimulation propria, most concentrated underneath the basement mem-
with specific T. whipplei antigens (76); (e) a transcriptional brane, but not in macrophages (28). However, some investi-
pattern of M2/alternatively activated macrophageswhich gators also report intracellular replication in a monocyte and
is associated with a Th2 immune responseas detected in macrophage cell culture model and enhanced replication after
duodenal tissue of one patient with Whipples disease (77); addition of interleukin-16 (91).
(f) a lack of an inflammatory response and a predominance Once T. whipplei disseminates, the brain is one of the fa-
of an M2/alternative activation phenotype in peripheral and vored sites for metastatic infection. The mechanisms by which
duodenal macrophages, in combination with reduced nitrite the bacteria cross the bloodbrain barrier and infect brain
production in duodenal tissue and an impaired capacity of pe- parenchyma are unknown. Entry by extracellular bacteria
ripheral monocytes to perform oxidative burst upon exposure or by bacteria within monocytes both are possibilities. The
to T. whipplei (78); and (g) a paradoxically higher immu- perivascular and subependymal distribution of macrophage
noglobulin G (IgG) antibody response against T. whipplei in nodules suggests bloodborne bacterial dissemination. The
asymptomatic carriers as compared to patients, indicating a distribution of microorganisms and histologic lesions within
diminished serologic response to infection (79,80). the CNS is relatively widespread and therefore argues against
There are a few reports of secondary or opportunistic infec- highly localized microbial tropism. The size and multifocal
tions in patients with Whipples disease (81), but apart from nature of the microglial nodules in CNS Whipples disease
Giardia duodenalis infection, which has been reported in 8% is compatible with neurologic deficits described in patients.
and 12% of patients, respectively, in two case series (82,83), For example, microscopic lesions in the oculomasticatory
these are not commonly observed phenomena, and humoral segments of the midbrain and upper pons might explain the
immune responses against other pathogens are largely intact (4). unusual features of oculomasticatory myorhythmia (OMM)

Scheld_Ch28.indd 463 2/21/14 8:10 PM


464 Part III: Bacterial and Mycoplasmal Infections

or oculofacial-skeletal myorhythmia (OFSM), both findings


that are nearly pathognomonic for CNS Whipples disease
(9294). However, the degrees to which CNS manifestations
are directly due to the bacteria or to immunologic or cellular
responses are not known. In cases with extensive CNS dis-
ease, bacteria are found within neurons and in macrophages
(see the next section), and neuronal loss and reactive astrocy-
tosis are seen during later stages of the disease process.

PATHOLOGY
In 1936, Ford and Walsh (95) described the case of a 47-year-
old man who died after a 10-month history of sleep distur-
bance, progressive paralysis of ocular movements, keratitis,
bulbar palsies, and what would later be called OMM (92). They
did not recognize this as a case of Whipples disease. Notable
findings at postmortem examination included cerebral atrophy, FIGURE 28.2 Histology of CNS Whipples disease (periodic acid
Schiff [PAS] reaction). Clumps or plaques of PAS-positive material
swelling of basal ganglia, and widespread degenerative changes within neuropil of the cortical gray matter. The most intense stain-
of the cerebral gray matter with encephalitis, especially involv- ing involves macrophages (arrows). Less intense lacy staining patterns
ing the corticobulbar tracts. In some areas, there was neuronal may correspond to the location of extracellular bacteria (magnifica-
cell death; other neurons were swollen with a granular blu- tion 200). (Courtesy Dr. Donald Regula, Department of Pathology,
ish purple cytoplasm. More than 50 years later, these tissues Stanford University School of Medicine, Stanford, California.)
were reexamined with a variety of histologic techniques, in-
cluding the PAS stain (96). The encephalitis involved extensive
mononuclear cell infiltration of gray and white matter with
perivascular cuffing, microglial proliferation, and marked as- with the PAS reagents (Figs. 28.2 and 28.3). Reactive and hy-
trocytosis. Foamy macrophages formed nodules and contained pertrophic astrocytes are common at the periphery of these
PAS-positive accumulations. Some neurons and astrocytes were nodules; lymphocytes and plasma cells are uncommon. Dual
PAS positive as well. These are all features that are typical of staining with PAS reagent and antibodies directed against
Whipples disease pathology in the brain. In addition, electron glial fibrillary-associated protein (GFAP) indicates astrocytes
microscopy confirmed the presence of bacillary structures in (Fig. 28.3). In cases with more extensive involvement, individ-
the cerebral cortex as well as in retinal tissue. ual PAS-positive cells are more diffusely distributed and may
Other cases of CNS involvement in Whipples disease have infiltrate overlying subarachnoid space (100) and white mat-
probably gone unrecognized (2,3). For example, 3 of the ter (Fig. 28.4). High-power examination of PAS-stained tissue
34 patients with Whipples disease that were reported in the reveals intracellular and extracellular material with variable
literature between 1907 and 1948 had symptoms referable to appearance, from lacy strands to darker specks and larger ir-
the CNS at the time of death, including confusion and somno- regular granules or clumps (Fig. 28.5). Occasionally, a bacilli-
lence (97); however, CNS Whipples disease was not formally form particle can be discerned. In areas with more substantial
discussed before 1960. The development of PAS reagents and collections of PAS-positive material, there tends to be neuro-
electron microscopy greatly facilitated this discussion. Sieracki nal loss and demyelination with vacuolization. Smaller (mi-
et al. (2) were the first to address CNS pathology in Whipples croscopic) microglial nodules have been described that stain
disease directly. Two postmortem examinations revealed nu- less intensely with the PAS reagents (99). Some reports have
merous and widespread periventricular, subependymal, and described microinfarcts that occur most often in the frontal
subcortical nodules, containing PAS-positive sickle-form or occipital lobes (100); it is hypothesized that such infarcts
particle-containing (SPC) cells. These SPC cells corresponded may be caused by small emboli released from cardiac valvular
to the same foamy macrophages that had been traditionally vegetations that were found commonly in Whipples disease
associated with Whipples disease in intestinal tissues. They patients in earlier autopsy series (3,4). The phagocytes associ-
noted less consistent involvement of the choroid plexus and ated with these infarcts are not PAS positive.
leptomeninges. In the autopsy review by Enzinger and Helwig Bacilli were first seen in the brain in 1969 with electron
(3) of Whipples disease 3 years later, 10 of 11 cases in which microscopy and resembled those previously described in non-
CNS tissue was available displayed the characteristic lesions CNS cases of Whipples disease (101,102). The distribution of
described earlier. bacteria and bacterial remnants corresponds well with areas
The gross and microscopic pathology of CNS Whipples of PAS-positive staining. Large numbers of extracellular bac-
disease has been summarized by a number of investigators teria can be detected within neuropil and near macrophages.
(98101). In many cases, chalky yellowish white 1- to 2-mm Intact intracellular bacteria, as well as bacilli undergoing vari-
nodules are distributed diffusely throughout the cortical and ous stages of degradation, are found within the widely distrib-
subcortical, cerebral and cerebellar gray matter, and in sub- uted PAS-positive macrophages (98,101,103105). Evidence
ependymal locations. They demonstrate a special predilection of bacillary binary fission suggests that microbial replication
for temporal, periventricular, and periaqueductal gray matter, does take place within the CNS (98,101). Macrophages with
hippocampus, hypothalamus, and basal ganglia. Less com- degraded bacilli found at the center of older lesions stain more
mon sites include the cerebellum and the thalamic and olivary strongly with PAS than macrophages with intact bacilli at the
nuclei. They tend to be perivascular. Other common gross periphery (99). Late stages of bacterial degradation produce
pathologic findings in CNS Whipples disease include cortical intracellular accumulations of serpiginous bacterial mem-
atrophy and ventricular dilation. branes that create the large, homogeneous, and more strongly
On microscopic examination, nodules are aggregates of staining PAS-positive granules. Whipples disease pathol-
primarily macrophages (microglia) that stain strongly positive ogy can be focal; involvement of the brain, as with the small

Scheld_Ch28.indd 464 2/21/14 8:10 PM


Chapter 28: Whipples Disease 465

FIGURE 28.3 Cellular distribution of periodic acidSchiff (PAS)positive material in CNS Whipples
disease (dual staining with PAS and antibody directed against glial fibrillary-associated protein [GFAP]).
Left: PAS-positive material is localized to GFAP-negative cells (macrophages) in this section of cortical
gray matter. GFAP-positive cells are predominantly astrocytes (arrows) (magnification 400). Right:
Astrocytes (brown) are seen in the vicinity of extracellular and intracellular PAS-positive material (magni-
fication 600). (Courtesy Dr. Lysia Forno, Department of Pathology, VA Palo Alto Health Care System,
Palo Alto, California.)

intestine, may be patchy (106); thus, a negative random biopsy the bloodbrain barrier wellwere typically used for treat-
result does not rule out the disease (see also section Diagnosis ment. In one review of 88 mostly tetracycline-treated patients,
and Differential Diagnosis). 31 relapsed after initial treatment, and relapses in 13 patients
(15% of all patients) affected the CNS (5). CNS relapses
generally have a poor prognosis, and some are refractory to
CLINICAL MANIFESTATIONS renewed antibiotic treatment (4,5,17,107). However, more re-
cent case series (10,12,108,109) show a much lower incidence
Neurologic manifestations of Whipples disease occur in three of CNS relapses, particularly when initial intravenous induc-
scenarios: (a) accompanied by intestinal and/or other systemic tion therapy with -lactams and/or oral long-term treatment
disease manifestations at the time of diagnosis, particularly with trimethoprim-sulfamethoxazole (TMP-SMX)which
when the disease is diagnosed at an advanced stage; (b) in the cross the bloodbrain barrier better than tetracyclineshave
setting of clinical relapse after antibiotic treatment of intestinal been used.
or other manifestations; and (c) as primary or isolated neuro- Presentation with isolated CNS Whipples disease is an
logic disease without other apparent manifestations. Among extremely rare clinical event. However, this may be difficult
these scenarios, neurologic relapse after treatment (b) is most to differentiate from CNS disease combined with other sys-
commonly encountered. This scenario was particularly com- temic manifestations; for example, a history of migratory
mon before the 1980s, when tetracyclineswhich do not cross arthralgias or chronic cough might represent extraintestinal

FIGURE 28.5 Histology of CNS Whipples disease (periodic acid


FIGURE 28.4 Histology of CNS Whipples disease (periodic acid Schiff reaction). High-powered view of neurons with faint positive
Schiff reaction). Less intense lacy staining patterns may correspond to material (arrow). Neurons are less commonly involved than are mac-
the location of extracellular bacteria (arrow) (magnification 1,500). rophages (microglia), except in severe disease (magnification 2,000).
(Courtesy Dr. Donald Regula, Department of Pathology, Stanford (Courtesy Dr. Donald Regula, Department of Pathology, Stanford
University School of Medicine, Stanford, California.) University School of Medicine, Stanford, California.)

Scheld_Ch28.indd 465 2/21/14 8:10 PM


466 Part III: Bacterial and Mycoplasmal Infections

Whipples disease, but these complaints are quite nonspecific TA B L E 2 8 . 1


and may not be viewed as significant during the time leading
up to more prominent manifestations. Even cases with clas- CLINICAL MANIFESTATIONS OF CNS WHIPPLES
sic features of Whipples disease may be missed because of DISEASE
the drawn-out time course of the illness. Nonetheless, there
are a number of notable reports in this context. Some reports More common
represent primary presentation with isolated CNS Whipples Dementia (or cognitive impairment)
disease where examination of other organs yielded negative Ophthalmoplegia
results (65,99,110,111). Several other cases represent primary
Myoclonus
neurologic presentation where other organs were not inves-
tigated (112,113) or where involvement of other organs was Altered level of consciousness
also found but was not prominent (92,96,98,114119). A Psychiatric abnormalities
few published cases represent CNS relapses years after initial Hypothalamic dysfunction
treatment, at a time when the non-CNS findings had become
Ataxia
completely negative (120122). Several combined case reports
and literature reviews specifically dedicated to primary or iso- Less common
lated CNS Whipples disease have been published (6365); the OMM or OFSMa
most recent one by Mohamed et al. (65) discusses 24 cases. Seizures
However, these published cases should be interpreted with
Aphasia
caution, because a significant number of these diagnoses were
based solely on PAS-positive histologic findings that have Cortical visual defect
not been confirmed with either electron microscopy or well- Dysarthria
validated PCR assays. As discussed (see Epidemiology and Tinnitus
Diagnosis and Differential Diagnosis), PAS-positive histo-
Meningitis
logic findings are not entirely specific for Whipples disease.
Another report (37) summarizes 20 cases (authors own and a
cases from the literature) that are described as T. whipplei Oculomasticatory myorhythmia (OMM) or oculofacial-skeletal myo-
rhythmia (OFSM); both may be pathognomonic.
chronic encephalitis, but the case definition used would also fit
the description of primary or isolated CNS Whipples disease
as commonly used by other authors; similar caution concern-
ing the certainty of diagnoses is also indicated with this series.
Common neurologic manifestations of Whipples disease are
dementia (or cognitive impairment), ophthalmoplegia, myoclo- Less common clinical manifestations of CNS Whipples
nus, altered level of consciousness, psychiatric abnormalities, hy- disease include aphasia, paresis, impaired vision due to cor-
pothalamic dysfunction, and ataxia (4,37,67,68,93) (Table 28.1). tical lesions, seizures, dysarthria, auditory impairment, tin-
The first three are considered a characteristic triad that should nitus, vertigo, trigeminal neuralgia, hydrocephalus, and
suggest the diagnosis of CNS Whipples disease, although only a meningitis (68,93,110,116,125128). Syndromes and pathol-
minority of patients (approximately 15%) has all three signs to- ogy suggestive of cerebrovascular stroke have been reported
gether (4,93). Headache is a common but nonspecific complaint. in patients with Whipples disease (129,130). In addition to
The dementia is slowly progressive and manifest by memory im- cognitive impairment, behavioral disturbances may be promi-
pairment, confusion, personality change, paranoia, emotional la- nent (131). In the setting of disease relapse with neurologic
bility, and depression. Approximately 71% of patients with CNS presentation, the most common clinical features are demen-
Whipples disease show signs of cognitive impairment, and nearly tia, ataxia, hypothalamic dysfunction, ophthalmoplegia, and
half of these patients also demonstrate psychiatric disturbances seizures (4,5). Although uncommon, the spinal cord and
(93). Patients have sometimes been misdiagnosed with Alzheimer peripheral nerves may also be involved in Whipples disease
disease. Nearly all cases of ophthalmoplegia are supranuclear, (68,110,132,133), and although not directly affecting the
usually in the form of vertical volitional ophthalmoplegia with nervous system, infectious spondylodiscitis with T. whipplei
preserved involuntary extraocular movements in response to (134,135) and myopathy (136) have also been reported.
head movements. OMM and OFSM constitute a peculiar mix- OMM was originally described by Ford and Walsh (95)
ture of synchronized eye movements and myoclonus that are re- in 1936 and by Van Bogaert et al. (137) in 1963, but the
ported in about 20% of patients; both may be pathognomonic diagnosis of Whipples disease was not made until reexamina-
for CNS Whipples disease (see later discussion) (93), but other tion of these cases years later (96,104). Though rare, OMM
forms of myoclonus can occur without eye involvement or in a may be unique to Whipples disease and for this reason war-
dyssynchronous fashion. In a particularly severe case of myoc- rants appreciation by clinicians. OMM is manifest by smooth
lonus, a patient suffered from synchronized 1-Hz jerks of the convergent-divergent pendular oscillations of the eyes with
right face, pharynx, arm, diaphragm, and calf, causing her entire synchronous rhythmic (approximately one per second, 1 Hz)
body to shake (114). Segmental myoclonus involving the facial contractions of the jaw (92,93,138,139). It can be continuous
nerve is also described in the setting of Whipples disease (123). during the day, persist during sleep, and remain unaffected by
Hypothalamic signs reported in this disease include polydipsia, environmental stimuli (92). In one patient, it led to permanent
polyphagia, insomnia, and hypersomnia (111,120,121,124). bruxism (140). Similar clinical findings, such as convergent
In their review of 28 patients, including 12 with CNS manifes- nystagmus, may be related to OMM and are equally sugges-
tations, Fleming and colleagues (8) found headache, diplopia, tive of Whipples disease. In addition, OMM may be a form
depression, confusion, and other forms of altered personality (or subset) of OFSM (93,141143), which is also associated
to be common at the time of presentation. A subsequent review with Whipples disease and is manifest by synchronous rhyth-
of CNS Whipples disease cases from the same institution (67) mic movements of the face and extremity muscles. It can also
found cerebellar ataxia (6 of 11 cases, 55%) to be more common involve eyelids, palate, tongue, and cervical muscles and is also
than described in other series. seen rarely in ocular pontocerebellar atrophy.

Scheld_Ch28.indd 466 2/21/14 8:10 PM


Chapter 28: Whipples Disease 467

Early observations on the natural course of untreated steatorrhea, increased erythrocyte sedimentation rate and
Whipples disease suggested three clinical phases (3,7): (a) an C-reactive protein, and low serum carotene levels are relatively
initial prolonged phase with insidious onset, marked by ar- common in patients with Whipples disease (3,4,7,8,1012).
thralgias, fatigue, occasional cough, and anemia; (b) a sec- Standard CSF analysis may be unremarkable or may display
ond phase characterized by abdominal pain, diarrhea, fever, mild abnormalities such as elevated pressure, elevated protein
lymphadenopathy, and weight loss; and (c) a terminal phase concentration, and mild pleocytosis (4,68,93,128). The CSF
marked by cachexia, congestive heart failure, and death. In glucose content is almost always normal. The electroencepha-
retrospect, CNS findings were often evident but unrecognized logram (EEG) may be unremarkable or show diffuse slowing
during the second phase. Nonetheless, even today, the diagno- (65,68,115).
sis of Whipples disease is made in some cases only after death. Neuroimaging studies have not revealed any specific or
Untreated CNS disease usually leads to progressive dementia, characteristic patterns for CNS Whipples disease; imaging re-
decline in level of consciousness, coma, and death within a sults may even be normal (10,68,93,152). Computed tomog-
period of months to several years. raphy (CT) scans may reveal cortical atrophy and ventricular
The eye is a site of significant and probably underappre- dilation; low-density, contrast-enhancing mass lesions are also
ciated involvement by Whipples disease (93,116,144149). well described (105,110,126,153). Magnetic resonance imag-
Ocular manifestations include (a) CNS disturbances affecting ing (MRI) probably offers greater sensitivity than CT scan-
the eye, as described earlier, and including ophthalmoplegia, ning in detecting CNS Whipples disease pathology (68,93).
cranial nerve III, IV, or VI palsies, OMM/OFSM, pupillary Various reports describe lesions with increased T2-weighted
abnormalities, and ptosis (93); and (b) direct peripheral in- signal intensity in the subcortical white matter, hypothalamus,
volvement of the eye and ocular structures, mostly by infec- uncus, medial temporal lobes, and amygdala with variable de-
tious processes (147,148). The latter, in general, occurs in grees of enhancement (103,111,112,132,139,154) (Fig. 28.6).
individuals with preexisting evidence of gastrointestinal and/ Adler and Galetta (139) provide gadolinium-enhanced T1-
or neurologic involvement; however, a few reported cases of weighted images with evidence of bloodbrain barrier leakage
ocular disease have been diagnosed without or with minimal that resolved with treatment. Positron emission tomography
extraocular manifestations. In one of these cases, T. whipplei (PET) scanning may also be used to detect areas of pathology
DNA was detected in the duodenal mucosa, despite normal with increased glucose uptake (65). Focal lesions as detected
duodenal histology (144). The most common eye manifes- by neuroimaging may be targeted for stereotactic brain biop-
tations of Whipples disease are visual loss, uveitis, vitreitis, sies, if this becomes necessary (65,105,110,112,113).
retinitis, optic neuritis, and papilledema. Anterior chamber Diagnostic testing includes studies based on histology, cy-
findings have included keratitis with corneal precipitates and tology, electron microscopy, and PCR. Histology is the tra-
iris nodules. These latter findings may suggest a diagnosis of ditional diagnostic approach for Whipples disease and may
sarcoidosis. Ocular disease is usually bilateral. Histologic or reveal PAS-positive inclusions in macrophages in intestinal,
cytologic examination may reveal typical PAS-positive mac- brain, and other tissues (62). Some investigators supplement
rophages (SPC cells) within the vitreous humor, lens capsule, histologic examination with immunohistochemistry using T.
and inner retina (116,145,149,150). Whipples bacilli have whippleispecific antibodies, although this test is not widely
also been visualized with electron microscopy, and their DNA available (37). Cytologic examination, involving PAS stain-
can be detected in vitreous fluid and ocular tissues by PCR ing of a cytocentrifuge pellet, is useful for CSF and vitreous
(144,149,150). In a large series of microbiologic investigations fluid (69,120,149,153). When positive, cytology shows mono-
on 1,520 anterior chamber and vitreous fluid specimens from nuclear cells with PAS-positive inclusions, also termed sickle-
cases of uveitis of unknown etiology in France, infections were form particle-containing cells (Fig. 28.7). Electron microscopy,
diagnosed in 147 cases, and among other fastidious bacteria, which has been in use since the 1960s, serves as the traditional
possible T. whipplei infection was diagnosed by PCR in 8 cases confirmatory test for PAS-positive histologic findings (24,25).
and definite T. whipplei infection in 2 cases (151). Thus, when PCR testing has been available since the 1990s; various assays
investigating inflammatory eye manifestations suspected to have targeted sequences such as the 16S rRNA gene, the 16S-
be due to Whipples disease, a combination of PAS staining 23S rDNA intergenic spacer, and genomic repetitive sequences
and PCR may be a useful diagnostic approach. Antimicrobial (14,41,49,155157). In recent years, most PCR assays have
treatment usually arrests progression of ocular disease and been transferred to a real-time platform (44,158,159). PCR
may lead to clinical remission (149). can serve as a confirmatory or supplementary test for intes-
tinal biopsies and as a primary or supplementary test for ex-
traintestinal tissues or body fluids (19,21). The possibility of
DIAGNOSIS AND DIFFERENTIAL serology for diagnostic purposes has been explored but has
DIAGNOSIS not yet reached a stage of wider availability for routine diag-
nostic use (29,79,80).
The diagnosis of CNS Whipples disease remains a clinical In the setting of suspected CNS Whipples disease, a rea-
challenge, mainly because of its rarity and because most signs sonable primary approach is to pursue a diagnosis with upper
and symptomsexcept presumably OMM and OFSMcan gastrointestinal endoscopy and small intestinal biopsies, in
also be observed in other clinical entities. The diagnosis re- combination with lumbar puncture to obtain CSF for cytology
quires astute attention to the overall clinical history and and/or PCR testing (21). Cytologic examination requires co-
physical findings, and this includes CNS findings as well as ordination with the receiving laboratory because CSF must be
other relevant details, such as joint problems, low-grade fever, fresh (preferably 1 hour old) (69). Some investigators have
unusual skin pigmentation, changes in bowel habits, weight proposed PCR testing of saliva and stool for first-line screen-
loss, and abdominal and peripheral lymphadenopathy. Any ing to diagnose classical intestinal Whipples disease, but this
diagnostic workup for possible CNS Whipples disease should approach lacks sensitivity for extraintestinal manifestations
always include upper gastrointestinal endoscopy, including and may be positive in asymptomatic carriers (49). Secondary
multiple (about five) small intestinal biopsies, as well as an as- approaches should be guided by clinical judgment and should
sessment of a patients lymph nodes. Results of routine labora- target suspected pathologically affected sites, such as lymph
tory studies are often nonspecific. Anemia, hypoalbuminemia, nodes, ocular fluids, or brain. Like the intestinal mucosa, brain

Scheld_Ch28.indd 467 2/21/14 8:10 PM


468 Part III: Bacterial and Mycoplasmal Infections

A B
FIGURE 28.6 Magnetic resonance images revealing posterior subcortical white matter lesions in a
59-year-old man with biopsy-proved Whipples disease of the duodenum and brain, before (A) and after
(B) 10 months of antibiotic therapy. (From Cooper GS, Blades EW, Remler BF, et al. Central nervous sys-
tem Whipples disease: relapse during therapy with trimethoprim-sulfamethoxazole and remission with
cefixime. Gastroenterology. 1994;106:782786, with permission.)

FIGURE 28.7 Light microscopic images from cytologic examination of CSF from patients with Whipples
disease, using the periodic acidSchiff (PAS) stain. A: Typical sickle-form particle-containing (SPC) cell
from a patient before antibiotic therapy. B: Cell with PAS-positive material in the cytoplasm that does not
resemble that of typical SPC cells (indeterminate finding) from a patient undergoing antibiotic therapy.
Original magnification: A, 196; B, 158. (From von Herbay A, Ditton HJ, Schuhmacher F, et al.
Whipples disease: staging and monitoring by cytology and polymerase chain reaction analysis of cerebro-
spinal fluid. Gastroenterology. 1997;113:434441, with permission.)

Scheld_Ch28.indd 468 2/21/14 8:10 PM


Chapter 28: Whipples Disease 469

involvement may be localized and/or patchy, and therefore higher frequency (4 to 5 Hz) than those seen in CNS Whipples
brain biopsies should be guided by neurophysiologic examina- disease. Isolated myorhythmias are also sometimes seen in
tion and imaging results (68,93). Apart from positive results brainstem vascular disease and in cerebellar degeneration sec-
for standard brain histology (using the PAS stain) and electron ondary to chronic alcoholism. Creutzfeldt-Jakob disease and
microscopy, T. whipplei rDNA has been amplified from brain CNS Whipples disease share some features, including patient
tissue (37,65); however, the common practice of prolonged age at onset, dementia, and myoclonus. Subacute sclerosing
fixation may hinder PCR-based amplification from archival panencephalitis can also resemble CNS Whipples disease;
specimens, and controls for the presence of amplifiable DNA however, the former typically occurs in a younger age-group.
are a necessity (160). Although a rare occurrence, PCR may CNS Whipples disease can be distinguished from slow virus
detect T. whipplei DNA in histologically unaffected sites, such infections or prion diseases on the basis of brain pathology.
as the duodenal mucosa in a case of T. whipplei uveitis (144). OMM and OFSM are mimicked by at least three neuro-
Also, apart from being useful for establishing a diagnosis, PCR logic clinical entities, although generally, they are differenti-
and cytologic examination of CSF initially and at regular in- ated from other forms of pendular nystagmus by their smooth
tervals during treatment may be useful for staging and moni- continuous nature, high amplitude, and slow frequency.
toring disease in order to detect and respond to the threat of First, OFSM occurs rarely in ocular pontocerebellar atrophy.
CNS relapses in classical and CNS Whipples disease (69). Second, oculopalatal myoclonus can appear similar to OMM,
Reactivity to the PAS stain is not specific for Whipples although there are no convergent ocular oscillations in oculo-
disease. The PAS reagent stains other microorganisms, includ- palatal myoclonus, and there is no olivary pseudohypertrophy
ing a number of actinomycetes and fungi, as well as nonmi- in Whipples disease. Third, OMM can be confused with con-
crobial glycoprotein and some cellular debris (4,18,62). As a vergence nystagmus of Parinaud, but the latter is episodic and
consequence, it is necessary to confirm PAS-positive findings is usually provoked by attempted upward gaze (92).
by electron microscopy (showing bacteria of the typical size
and shape of T. whipplei) or by PCR. Some investigators have
indeed documented instances of PAS-positive brain histol- TREATMENT
ogy that were not confirmed to be due to Whipples disease
(37,160). Two microorganisms to consider in the differential Therapy for Whipples disease has been largely guided by
diagnosis of PAS-positive findings are Mycobacterium avium empirical observations and retrospective literature reviews
complex and Histoplasma capsulatum, both of which are (5,810,108). Before the 1980s, when tetracyclines were com-
found within macrophages and both of which may involve the monly used for treatment, relapse rates were high and CNS
brain. These organisms are particularly relevant in the setting relapses occurred frequently (5). Even though recent case se-
of HIV infection; in such instances, attempts should be made ries show much lower relapse rates (10,12,109), CNS relapses
to distinguish these organisms from T. whipplei by culture, continue to pose a real and very serious threat to patients.
PCR, and other staining procedures. In this context, it is rel- In two remarkable published cases, CNS relapse occurred 8
evant that CNS and other systemic T. whipplei infections have and 12 years, respectively, after treatment and presumed cure
been documentedalthough rarelyin patients with AIDS of intestinal Whipples disease (121,122). Earlier case series
(161,162), and Whipples diseaselike syndromes can be in- showed that patients who received initial intravenous induc-
duced by M. avium complex and Rhodococcus equi in patients tion therapy with penicillin and streptomycin followed by oral
with AIDS (163,164). maintenance therapy with tetracyclines (the Duke regimen)
Similarly, PCR testing requires attention to detail; false- had a more favorable clinical outcome (5). Supplemented by
positive results may arise from PCR contamination as well the fact that TMP-SMX shows better clinical outcomes than
as from amplification of nontarget sequences (20,165,166). tetracycline-based regimens (108) and even penetrates a rela-
When PCR results are solely judged by agarose gel electropho- tively intact bloodbrain barrier well, several recent recom-
resis, bands of presumably the right size may be seen that nev- mendations for treatment suggest initial intravenous induction
ertheless do not correspond to T. whipplei DNA (M. Maiwald therapy for 2 weeks with a -lactamcontaining regimen (e.g.,
and D. A. Relman, unpublished results). As a consequence, either penicillin G plus streptomycin, a third-generation ceph-
it is recommended that one employ strict anti-contamination alosporin, or a carbapenem) followed by oral maintenance
practices and use PCR assays that have been validated with ad- therapy with TMP-SMX for at least 1 year (1921).
equate positive and negative clinical samples, that one include Despite the use of these induction and maintenance regi-
adequate negative as well as inhibition controls, and confirm mens, instances of treatment failure, including worsening
the identity of PCR products by sequencing. Some investiga- of CNS symptoms, have been observed under TMP-SMX
tors propose confirmation of positive results by a second PCR therapy, and this includes cases of apparently acquired TMP-
with a different target sequence (157). SMX resistance in T. whipplei (103,108,139,168170). Some
The differential diagnosis of CNS Whipples disease in- patients with worsening CNS symptoms have benefitted from
cludes other forms of subacute or chronic encephalopathy a change in regimen or from cycles of intravenous salvage
(e.g., Wernicke encephalopathy, heavy-metal poisoning, and therapy. Relevant case reports describe intravenous treatment
storage diseases in children), Alzheimer disease, Parkinson periods with ceftriaxone (69,139); switches to the oral third-
disease, prion diseases, cerebral vascular infarcts, CNS vascu- generation cephalosporin, cefixime (103); and addition of ri-
litis, paraneoplastic syndromes, neurosyphilis and other CNS fampin, which is also known for excellent CNS penetration, to
infections, and chronic demyelinating disorders (167). In ad- the regimen (171).
dition, hypothalamic presentations of Whipples disease may In vitro studies with cultivated T. whipplei have shown that
resemble Kleine-Levin syndrome (120). A history of alcohol- in co-culture with fibroblasts, the organism appears susceptible
ism and a response to thiamine may help to identify Wernicke to doxycycline, macrolides, penicillins, rifampin, teicoplanin,
encephalopathy. Some presentations of CNS Whipples disease and TMP-SMX and variably susceptible to imipenem (172); in
may resemble multiple sclerosis (MS), but supranuclear oph- axenic medium, the organism also appears susceptible to cef-
thalmoplegia, myoclonus, and seizures are unusual in MS. triaxone and vancomycin (173). Furthermore, genome analy-
Also, MS may be associated with myorhythmias and rarely sis has suggested that T. whipplei is inherently resistant to the
with pendular ocular oscillations, but they are usually of a trimethoprim component in TMP-SMX (174). Thus, some

Scheld_Ch28.indd 469 2/21/14 8:10 PM


470 Part III: Bacterial and Mycoplasmal Infections

have proposed that the TMP-SMX combination can be safely strategies. However, once diagnosed, it is important to treat
replaced by sulfadiazine alone (175); the latter also penetrates with prolonged courses of antibiotics that penetrate the blood
the bloodbrain barrier well. In vitro studies have further sug- brain barrier in order to prevent CNS relapses.
gested that the combination of doxycycline and hydroxychlo-
roquine is bactericidal for T. whipplei, presumably because
hydroxychloroquine enhances tetracycline activity by raising FUTURE DIRECTIONS
the pH value of phagolysosomes (172). Encouraging initial
treatment results in patients have been reported with this regi- Although significant progress has been made in the last 10
men (19,176). As a consequence, it has been suggested that to 20 years, a number of important questions about the
patients with CNS involvement may benefit from treatment pathogenesis, clinical spectrum, diagnosis, and treatment of
with doxycycline plus hydroxychloroquine, plus either TMP- Whipples disease remain to be answered. Further informa-
SMX or sulfadiazine (175). To improve on CNS penetration tion concerning the prevalence and clinical significance of
of the tetracycline-chloroquine combination, one group (177) T. whipplei in healthy carriers, in the respiratory tract of
treated a patient with persistent infection of the CSF using a HIV-infected individuals (86), and in nonhuman reservoirs is
combination of minocycline and chloroquine (minocycline has needed. The association of T. whipplei with acute self-limiting
better CNS penetration than doxycycline) and successfully disease is intriguing and warrants investigation of causality
eradicated T. whipplei from the CSF. A randomized clinical and possible clearance of the organism. Further explora-
trial of initial intravenous ceftriaxone followed by 1 year of tion of possible predisposing immunologic host factors for
oral TMP-SMX versus initial meropenem followed by 1 year Whipples disease is needed, as is additional work on patho-
of TMP-SMX in 40 patients (109) did not show any signifi- genetic mechanisms. The first randomized clinical trial of an-
cant difference between the two regimens, and apart from one tibiotic treatment (109) may invite investigators to conduct
patient with persistent T. whipplei in the CSF, there were no additional trials, such as with different drug combinations or
relapses within a median observation period of 89 months. treatment durations; however, the limited number of avail-
Some patients may experience clinical deterioration, often able patients with disease will continue to pose challenges for
accompanied by fever and other systemic manifestations, efforts to achieve statistical significance. Diagnostic strategies
weeks to months after initiation of treatment. In these pa- will undoubtedly be refined; in this context, it will be interest-
tients, the deterioration has been attributed to a phenomenon ing to see whether the paradoxical serologic response of
akin to immune reconstitution inflammatory syndrome (IRIS), patients (79,80) may be further used for the development of
as described in patients with HIV infection (178). This phe- a diagnostic test.
nomenon may be clinically serious. Although patients often The clinical significance of T. whipplei strain diversity
respond favorably to corticosteroid treatment, one patient remains another open question; currently, there appears to
apparently failed steroid treatment and responded instead to be no link between different strains and disease manifesta-
thalidomide (179). tions. The cultivation of T. whipplei in 2000 (29) and the
In summary, the treatment of Whipples disease, and its sequencing of its genome in 2003 (38,39) were big steps for-
CNS manifestations in particular, remains difficult. It is unclear ward and provided the basis for advanced strain typing ca-
how much weight should be assigned to in vitro susceptibility pacities (55). With remarkable advances in DNA sequencing
data. One should continue to rely on clinical studies or case technologies and capabilities, it seems clear that many more
series as a major source of information on which to base treat- T. whipplei genome sequences will be generated for both
ment decisions. Initial intravenous therapy with a -lactam, research and clinical purposes. These efforts will certainly
either alone or in combination with another agent, and subse- facilitate further advances in diagnostics, understanding of
quent extended oral treatment with drugs that penetrate well pathogenesis, and treatment for this unusual organism and
into the CNS appear to be most effective. disease.

PREVENTION ACKNOWLEDGMENTS
Whipples disease in its classic form is a rare disorder. Our We thank Drs. Donald Regula and Lysia Forno, Department of
present understanding of the risk factors for this disease is Pathology, Stanford University School of Medicine, Stanford,
insufficient to allow for any recommendations for preventive California, for histopathologic photomicroscopy.

References
1. Whipple GH. A hitherto undescribed disease characterized anatomically 7. Maizel H, Ruffin JM, Dobbins WO III. Whipples disease: a review of
by deposits of fat and fatty acids in the intestinal and mesenteric lymphatic 19 patients from one hospital and a review of the literature since 1950.
tissues. Bull Johns Hopkins Hosp. 1907;18:382391. Medicine (Baltimore). 1970;49:175205.
2. Sieracki JC, Fine G, Horn RC Jr, et al. Central nervous system involve- 8. Fleming JL, Wiesner RH, Shorter RG. Whipples disease: clinical, biochemi-
ment in Whipples disease. J Neuropathol Exp Neurol. 1960;19: cal, and histopathologic features and assessment of treatment in 29 pa-
7075. tients. Mayo Clin Proc. 1988;63:539551.
3. Enzinger FM, Helwig EB. Whipples disease. A review of the literature and 9. von Herbay A, Otto HF. Whipples disease: a report of 22 patients. Klin
report of fifteen patients. Virchows Archiv. 1963;336:238269. Wochenschr. 1988;66:533539.
4. Dobbins WO III. Whipples Disease. Springfield, IL: Charles C. Thomas; 10. Vital Durand D, Lecomte C, Cathbras P, et al. Whipple disease: clinical
1987. review of 52 cases. Medicine (Baltimore). 1997;76:170184.
5. Keinath RD, Merrell DE, Vlietstra R, et al. Antibiotic treatment and relapse 11. Lagier JC, Lepidi H, Raoult D, et al. Systemic Tropheryma whip-
in Whipples disease. Long-term follow-up of 88 patients. Gastroenterology. plei: clinical presentation of 142 patients with infections diagnosed
1985;88:18671873. or confirmed in a reference center. Medicine (Baltimore). 2010;89:
6. Dobbins WO III. Whipples disease. Mayo Clin Proc. 1988;63:623624. 337345.

Scheld_Ch28.indd 470 2/21/14 8:10 PM


Chapter 28: Whipples Disease 471

12. Ojeda E, Cosme A, Lapaza J, et al. Whipples disease in Spain: a clinical 43. Maiwald M, von Herbay A, Persing DH, et al. Tropheryma whippelii
review of 91 patients diagnosed between 1947 and 2001. Rev Esp Enferm DNA is rare in the intestinal mucosa of patients without other evidence of
Dig. 2010;102:108123. Whipple disease. Ann Intern Med. 2001;134:115119.
13. Wilson KH, Blitchington R, Frothingham R, et al. Phylogeny of the 44. Fenollar F, Fournier PE, Raoult D, et al. Quantitative detection of
Whipples disease-associated bacterium. Lancet. 1991;338(8765):474475. Tropheryma whipplei DNA by real-time PCR. J Clin Microbiol.
14. Relman DA, Schmidt TM, Macdermott RP, et al. Identification of the un- 2002;40:11191120.
cultured bacillus of Whipples disease. N Engl J Med. 1992;327:293301. 45. Ehrbar HU, Bauerfeind P, Dutly F, et al. PCR-positive tests for Tropheryma
15. Maiwald M, Ditton HJ, von Herbay A, et al. Reassessment of the phylo- whippelii in patients without Whipples disease. Lancet. 1999;
genetic position of the bacterium associated with Whipples disease and 353(9171):2214.
determination of the 16S-23S ribosomal intergenic spacer sequence. Int J 46. Street S, Donoghue HD, Neild GH. Tropheryma whippelii DNA in saliva
Syst Bacteriol. 1996;46:10781082. of healthy people. Lancet. 1999;354(9185):11781179.
16. La Scola B, Fenollar F, Fournier PE, et al. Description of Tropheryma 47. Amsler L, Bauernfeind P, Nigg C, et al. Prevalence of Tropheryma whip-
whipplei gen. nov., sp. nov., the Whipples disease bacillus. Int J Syst Evol plei DNA in patients with various gastrointestinal diseases and in healthy
Microbiol. 2001;51:14711479. controls. Infection. 2003;31:8185.
17. Schnider PJ, Reisinger EC, Gerschlager W, et al. Long-term follow-up in 48. Fenollar F, Trani M, Davoust B, et al. Prevalence of asymptomatic
cerebral Whipples disease. Eur J Gastroenterol Hepatol. 1996;8:899903. Tropheryma whipplei carriage among humans and nonhuman primates.
18. Marth T, Raoult D. Whipples disease. Lancet. 2003;361(9353):239246. J Infect Dis. 2008;197:880887.
19. Fenollar F, Puchal X, Raoult D. Whipples disease. N Engl J Med. 49. Fenollar F, Laouira S, Lepidi H, et al. Value of Tropheryma whipplei quan-
2007;356:5566. titative polymerase chain reaction assay for the diagnosis of Whipple dis-
20. Schneider T, Moos V, Loddenkemper C, et al. Whipples disease: new as- ease: usefulness of saliva and stool specimens for first-line screening. Clin
pects of pathogenesis and treatment. Lancet Infect Dis. 2008;8:179190. Infect Dis. 2008;47:659667.
21. Maiwald M, von Herbay A, Relman DA. Whipples disease. In: Sleisenger MH, 50. Desnues B, Al Moussawi K, Fenollar F. New insights into Whipples
Feldman M, Friedman LS, et al, eds. Sleisenger and Fordtrans Gastrointestinal disease and Tropheryma whipplei infections. Microbes Infect. 2010;12:
and Liver Disease: Pathophysiology, Diagnosis, Management. 9th ed. 11021110.
Philadelphia: Saunders/Elsevier; 2010:18331842. 51. Fenollar F, Keita AK, Buffet S, et al. Intrafamilial circulation of Tropheryma
22. Chears WC, Ashworth CT. Electron microscopic study of the intestinal whipplei, France. Emerg Infect Dis. 2012;18:949955.
mucosa in Whipples disease. Demonstration of encapsulated bacilliform 52. Hinrikson HP, Dutly F, Nair S, et al. Detection of three different types
bodies in the lesion. Gastroenterology. 1961;41:129138. of Tropheryma whippelii directly from clinical specimens by sequenc-
23. Yardley JH, Hendrix TR. Combined electron and light microscopy in ing, single-strand conformation polymorphism (SSCP) analysis and type-
Whipples disease. Bull Johns Hopkins Hosp. 1961;109:8098. specific PCR of their 16S-23S ribosomal intergenic spacer region. Int J Syst
24. Dobbins WO III, Kawanishi H. Bacillary characteristics in Whipples Bacteriol. 1999;49:17011706.
disease: an electron microscopic study. Gastroenterology. 1981;80: 53. Maiwald M, von Herbay A, Lepp PW, et al. Organization, structure,
14681475. and variability of the rRNA operon of the Whipples disease bacterium
25. Silva MT, Macedo PM, Moura Nunes JF. Ultrastructure of bacilli and the (Tropheryma whippelii). J Bacteriol. 2000;182:32923297.
bacillary origin of the macrophagic inclusions in Whipples disease. J Gen 54. Geissdrfer W, Wittmann I, Rllinghoff M, et al. Detection of a new 16S-
Microbiol. 1985;131:10011013. 23S rRNA spacer sequence variant (type 7) of Tropheryma whippelii in
26. Paulley JW. A case of Whipples disease (intestinal lipodystrophy). a patient with prosthetic aortic valve endocarditis. Eur J Clin Microbiol
Gastroenterology. 1952;22:128133. Infect Dis. 2001;20:762763.
27. Trier JS, Phelps PC, Eidelman S, et al. Whipples disease: light and electron 55. Li W, Fenollar F, Rolain JM, et al. Genotyping reveals a wide heterogeneity
microscope correlation of jejunal mucosal histology with antibiotic treat- of Tropheryma whipplei. Microbiology. 2008;154:521527.
ment and clinical status. Gastroenterology. 1965;48:684707. 56. Raoult D, Fenollar F, Rolain JM, et al. Tropheryma whipplei in children
28. Fredricks DN, Relman DA. Localization of Tropheryma whippelii with gastroenteritis. Emerg Infect Dis. 2010;16:776782.
rRNA in tissues from patients with Whipples disease. J Infect Dis. 57. Keita AK, Bassene H, Tall A, et al. Tropheryma whipplei: a common bacte-
2001;183:12291237. rium in rural Senegal. PLoS Negl Trop Dis. 2011;5(12):e1403.
29. Raoult D, Birg ML, LaScola B, et al. Cultivation of the bacillus of Whipples 58. von Herbay A, Otto HF, Stolte M, et al. Epidemiology of Whipples dis-
disease. N Engl J Med. 2000;342:620625. ease in Germany: analysis of 110 patients diagnosed in 1965-95. Scand J
30. Raoult D, La Scola B, Lecocq P, et al. Culture and immunological detection Gastroenterol. 1997;32:5257.
of Tropheryma whippelii from the duodenum of a patient with Whipple 59. Fournier PE, Thuny F, Richet H, et al. Comprehensive diagnostic strategy
disease. JAMA. 2001;285:10391043. for blood culture-negative endocarditis: a prospective study of 819 new
31. Maiwald M, von Herbay A, Fredricks DN, et al. Cultivation of Tropheryma cases. Clin Infect Dis. 2010;51:131140.
whipplei from cerebrospinal fluid. J Infect Dis. 2003;188:801808. 60. Geissdrfer W, Moos V, Moter A, et al. High frequency of Tropheryma
32. Maiwald M, Relman DA. Tropheryma. In: Whitman WB, Goodfellow M, whipplei in culture-negative endocarditis. J Clin Microbiol. 2012;50:
Kmpfer P, et al, eds. Bergeys Manual of Systematic Bacteriology. Vol 5: 216222.
The Actinobacteria. 2nd ed. New York: Springer; 2012:717726. 61. Fenollar F, Mediannikov O, Socolovschi C, et al. Tropheryma whip-
33. Masselot F, Boulos A, Maurin M, et al. Molecular evaluation of antibi- plei bacteremia during fever in rural West Africa. Clin Infect Dis.
otic susceptibility: Tropheryma whipplei paradigm. Antimicrob Agents 2010;51:515521.
Chemother. 2003;47:16581664. 62. von Herbay A. Morbus Whipple. Histologische Diagnostik nach der
34. Renesto P, Crapoulet N, Ogata H, et al. Genome-based design of a cell- Entdeckung von Tropheryma whippelii. Pathologe. 2001;22:8288.
free culture medium for Tropheryma whipplei. Lancet. 2003;362(9382): 63. Szer T, Demirkan N, Tahta K, et al. Whipples disease confined to the cen-
447449. tral nervous system: case report and review of the literature. Scand J Infect
35. Fenollar F, Birg ML, Gauduchon V, et al. Culture of Tropheryma whipplei Dis. 1999;31:411414.
from human samples: a 3-year experience (1999 to 2002). J Clin Microbiol. 64. Panegyres PK, Edis R, Beaman M, et al. Primary Whipples disease of the
2003;41:38163822. brain: characterization of the clinical syndrome and molecular diagnosis.
36. Keita AK, Raoult D, Fenollar F. Tropheryma whipplei as a commensal bac- QJM. 2006;99:609623.
terium. Future Microbiol. 2013;8:5771. 65. Mohamed W, Neil E, Kupsky WJ, et al. Isolated intracranial Whipples
37. Fenollar F, Nicoli F, Paquet C, et al. Progressive dementia associated with diseasereport of a rare case and review of the literature. J Neurol Sci.
ataxia or obesity in patients with Tropheryma whipplei encephalitis. BMC 2011;308:18.
Infect Dis. 2011;11:171. 66. Sung VW, Lyerly MJ, Fallon KB, et al. Isolated CNS Whipple disease with
38. Bentley SD, Maiwald M, Murphy LD, et al. Sequencing and analysis of the normal brain MRI and false-positive CSF 14-3-3 protein: a case report and
genome of the Whipples disease bacterium Tropheryma whipplei. Lancet. review of the literature. Brain Behav. 2012;2:838843.
2003;361(9358):637644. 67. Matthews BR, Jones LK, Saad DA, et al. Cerebellar ataxia and central ner-
39. Raoult D, Ogata H, Audic S, et al. Tropheryma whipplei twist: a vous system Whipple disease. Arch Neurol. 2005;62:618620.
human pathogenic Actinobacteria with a reduced genome. Genome Res. 68. Gerard A, Sarrot-Reynauld F, Liozon E, et al. Neurologic presentation of
2003;13:18001809. Whipple disease: report of 12 cases and review of the literature. Medicine
40. Maiwald M, Schuhmacher F, Ditton HJ, et al. Environmental occurrence (Baltimore). 2002;81:443457.
of the Whipples disease bacterium (Tropheryma whippelii). Appl Environ 69. von Herbay A, Ditton HJ, Schuhmacher F, et al. Whipples disease: stag-
Microbiol. 1998;64:760762. ing and monitoring by cytology and polymerase chain reaction analysis of
41. von Herbay A, Ditton HJ, Maiwald M. Diagnostic application of a poly- cerebrospinal fluid. Gastroenterology. 1997;113:434441.
merase chain reaction assay for the Whipples disease bacterium to intesti- 70. Riggs JE. The evolving natural history of neurologic involvement in
nal biopsies. Gastroenterology. 1996;110:17351743. Whipple disease: a hypothesis. Arch Neurol. 1988;45:830.
42. Pron B, Poyart C, Abachin E, et al. Diagnosis and follow-up of Whipples 71. Bjerknes R, Laerum OD, Degaard S. Impaired bacterial degradation by
disease by amplification of the 16S rRNA gene of Tropheryma whippelii. monocytes and macrophages from a patient with treated Whipples disease.
Eur J Clin Microbiol Infect Dis. 1999;18:6265. Gastroenterology. 1985;89:11391146.

Scheld_Ch28.indd 471 2/21/14 8:10 PM


472 Part III: Bacterial and Mycoplasmal Infections

72. Bjerknes R, Odegaard S, Bjerkvig R, et al. Whipples disease. 103. Cooper GS, Blades EW, Remler BF, et al. Central nervous system Whipples
Demonstration of a persisting monocyte and macrophage dysfunction. disease: relapse during therapy with trimethoprim-sulfamethoxazole and
Scand J Gastroenterol. 1988;23:611619. remission with cefixime. Gastroenterology. 1994;106:782786.
73. Bai JC, Sen L, Diez R, et al. Impaired monocyte function in patients suc- 104. De Jonghe P, Martin JJ, Budka H, et al. Cerebral manifestations of
cessfully treated for Whipples disease. Acta Gastroenterol Latinoam. Whipples disease. Acta Neurol Belg. 1979;79:305313.
1996;26:8589. 105. Johnson L, Diamond I. Cerebral Whipples disease. Diagnosis by brain
74. Marth T, Neurath M, Cuccherini BA, et al. Defects of monocyte in- biopsy. Am J Clin Pathol. 1980;74:486490.
terleukin 12 production and humoral immunity in Whipples disease. 106. Moorthy S, Nolley G, Hermos JA. Whipples disease with minimal intes-
Gastroenterology. 1997;113:442448. tinal involvement. Gut. 1977;18:152155.
75. Marth T, Kleen N, Stallmach A, et al. Dysregulated peripheral and muco- 107. Schmitt BP, Richardson H, Smith E, et al. Encephalopathy complicat-
sal Th1/Th2 response in Whipples disease. Gastroenterology. 2002;123: ing Whipples disease: failure to respond to antibiotics. Ann Intern Med.
14681477. 1981;94:5152.
76. Moos V, Kunkel D, Marth T, et al. Reduced peripheral and mucosal 108. Feurle GE, Marth T. An evaluation of antimicrobial treatment for
Tropheryma whipplei-specific Th1 response in patients with Whipples Whipples disease. Tetracycline versus trimethoprim-sulfamethoxazole.
disease. J Immunol. 2006;177:20152022. Dig Dis Sci. 1994;39:16421648.
77. Desnues B, Lepidi H, Raoult D, et al. Whipple disease: intestinal infiltrat- 109. Feurle GE, Junga NS, Marth T. Efficacy of ceftriaxone or meropenem
ing cells exhibit a transcriptional pattern of M2/alternatively activated as initial therapies in Whipples disease. Gastroenterology. 2010;138:
macrophages. J Infect Dis. 2005;192:16421646. 478486.
78. Moos V, Schmidt C, Geelhaar A, et al. Impaired immune functions of 110. Halperin JJ, Landis DM, Kleinman GM. Whipple disease of the nervous
monocytes and macrophages in Whipples disease. Gastroenterology. system. Neurology. 1982;32:612617.
2010;138:210220. 111. Adams M, Rhyner PA, Day J, et al. Whipples disease confined to the
79. Bonhomme CJ, Renesto P, Nandi S, et al. Serological microarray for a para- central nervous system. Ann Neurol. 1987;21:104108.
doxical diagnostic of Whipples disease. Eur J Clin Microbiol Infect Dis. 112. Wroe SJ, Pires M, Harding B, et al. Whipples disease confined to the CNS
2008;27:959968. presenting with multiple intracerebral mass lesions. J Neurol Neurosurg
80. Fenollar F, Amphoux B, Raoult D. A paradoxical Tropheryma whipplei Psychiatry. 1991;54:989992.
western blot differentiates patients with Whipple disease from asymptom- 113. Mendel E, Khoo LT, Go JL, et al. Intracerebral Whipples disease diag-
atic carriers. Clin Infect Dis. 2009;49:717723. nosed by stereotactic biopsy: a case report and review of the literature.
81. Meier-Willersen HJ, Maiwald M, von Herbay A. Morbus Whipple in Neurosurgery. 1999;44:203209.
Assoziation mit opportunistischen Infektionen. Dtsch Med Wochenschr. 114. Stoupel N, Monseu G, Pardoe A, et al. Encephalitis with myoclonus in
1993;118:854860. Whipples disease. J Neurol Neurosurg Psychiatry. 1969;32:338343.
82. von Herbay A, Maiwald M, Ditton HJ, et al. Histology of intesti- 115. Finelli PF, McEntee WJ, Lessell S, et al. Whipples disease with predomi-
nal Whipples disease revisited: a study of 48 patients. Virchows Arch. nantly neuroophthalmic manifestations. Ann Neurol. 1977;1:247252.
1996;429:335343. 116. Grtner J. Whipples disease of the central nervous system, associated
83. Fenollar F, Lepidi H, Gerolami R, et al. Whipple disease associated with with ophthalmoplegia externa and severe asteroid hyalitis. A clinicopath-
giardiasis. J Infect Dis. 2003;188:828834. ologic study. Doc Ophthalmol. 1980;49:155187.
84. Kneitz C, Suerbaum S, Beer M, et al. Exacerbation of Whipples disease as- 117. Ludwig B, Bohl J, Haferkamp G. Central nervous system involvement in
sociated with infliximab treatment. Scand J Rheumatol. 2005;34:148151. Whipples disease. Neuroradiology. 1981;21:289293.
85. Mahnel R, Kalt A, Ring S, et al. Immunosuppressive therapy in Whipples 118. Buckle MJ, Ellis RW, Bone M, et al. Neurologically presenting
disease patients is associated with the appearance of gastrointestinal man- Whipple disease: case report and review of the literature. J Clin Pathol.
ifestations. Am J Gastroenterol. 2005;100:11671173. 2008;61:11401141.
86. Lozupone C, Cota-Gomez A, Palmer BE, et al. Widespread colonization 119. Panegyres PK. Diagnosis and management of Whipples disease of the
of the lung by Tropheryma whipplei in HIV infection. Am J Respir Crit brain. Pract Neurol. 2008;8:311317.
Care Med. 2013;187(10):11101117. 120. Feurle GE, Volk B, Waldherr R. Cerebral Whipples disease with negative
87. Greub G. A new piece added to the Whipple puzzle: Tropheryma whip- jejunal histology. N Engl J Med. 1979;300:907908.
plei primary infection with bacteremia and cough. Clin Infect Dis. 121. Lieb K, Maiwald M, Berger M, et al. Insomnia for 5 years. Lancet.
2010;51:522524. 1999;354(9194):1966.
88. Moos V, Schneider T. Changing paradigms in Whipples disease and 122. Frazier JL, Quinones-Hinojosa A. Isolated Whipple disease of the brain
infection with Tropheryma whipplei. Eur J Clin Microbiol Infect Dis. resembling a tumour. Acta Neurochir (Wien). 2009;151:173175.
2011;30:11511158. 123. Stamboulis E, Kararizou E, Manta P, et al. Segmental myoclonus
89. Dobbins WO, Ruffin JM. A light- and electron-microscopic study of bac- in Whipples disease. Electromyogr Clin Neurophysiol. 1995;35:
terial invasion in Whipples disease. Am J Pathol. 1967;51:225242. 113116.
90. Raoult D, Lepidi H, Harle JR. Tropheryma whipplei circulating in blood 124. Prss H, Katchanov J, Zschenderlein R, et al. A patient with cerebral
monocytes. N Engl J Med. 2001;345:548. Whipple disease with gastric involvement but no gastrointestinal symp-
91. Desnues B, Raoult D, Mege JL. IL-16 is critical for Tropheryma whipplei toms: a consequence of local protective immunity? J Neurol Neurosurg
replication in Whipples disease. J Immunol. 2005;175:45754582. Psychiatry. 2007;78:896898.
92. Schwartz MA, Selhorst JB, Ochs AL, et al. Oculomasticatory myorhyth- 125. Thompson DG, Ledingham JM, Howard AJ, et al. Meningitis in Whipples
mia: a unique movement disorder occurring in Whipples disease. Ann disease. BMJ. 1978;2(6129):1415.
Neurol. 1986;20:677683. 126. Feldman M, Hendler RS, Morrison EB. Acute meningoencephalitis
93. Louis ED, Lynch T, Kaufmann P, et al. Diagnostic guidelines in central after withdrawal of antibiotics in Whipples disease. Ann Intern Med.
nervous system Whipples disease. Ann Neurol. 1996;40:561568. 1980;93:709711.
94. Lynch T, Fahn S, Louis ED, et al. Oculofacial-skeletal myorhythmia in 127. Anderson M. Neurology of Whipples disease. J Neurol Neurosurg
Whipples disease. Mov Disord. 1997;12:625626. Psychiatry. 2000;68:25.
95. Ford FR, Walsh FB. Clinical observations upon the importance of the 128. Aksamit AJ. Whipples disease of the central nervous system. Handb Clin
vestibular reflexes in ocular movements. Bull Johns Hopkins Hosp. Neurol. 2010;96:231237.
1936;58:8088. 129. Naegeli B, Bannwart F, Bertel O. An uncommon cause of recurrent
96. Knox DL, Green WR, Troncoso JC, et al. Cerebral ocular Whipples dis- strokes: Tropheryma whippelii endocarditis. Stroke. 2000;31:20022003.
ease: a 62-year odyssey from death to diagnosis. Neurology. 1995;45: 130. Peters G, du Plessis DG, Humphrey PR. Cerebral Whipples disease
617625. with a stroke-like presentation and cerebrovascular pathology. J Neurol
97. Plummer K, Russi S, Harris WH Jr, et al. Lipophagic intestinal granulo- Neurosurg Psychiatry. 2002;73:336339.
matosis (Whipples disease). Clinical and pathologic study of thirty-four 131. Manzel K, Tranel D, Cooper G. Cognitive and behavioral abnormali-
cases, with special reference to clinical diagnosis and pathogenesis. Arch ties in a case of central nervous system Whipple disease. Arch Neurol.
Intern Med. 1950;86:280310. 2000;57:399403.
98. Silbert SW, Parker E, Horenstein S. Whipples disease of the central ner- 132. Kremer S, Besson G, Bonaz B, et al. Diffuse lesions in the CNS revealed
vous system. Acta Neuropathol. 1976;36:3138. by MR imaging in a case of Whipple disease. AJNR Am J Neuroradiol.
99. Romanul FC, Radvany J, Rosales RK. Whipples disease confined to the 2001;22:493495.
brain: a case studied clinically and pathologically. J Neurol Neurosurg 133. Schrter A, Brinkhoff J, Gnthner-Lengsfeld T, et al. Whipples disease
Psychiatry. 1977;40:901909. presenting as an isolated lesion of the cervical spinal cord. Eur J Neurol.
100. Smith WT, French JM, Gottsman M, et al. Cerebral complications of 2005;12:276279.
Whipples disease. Brain. 1965;88:137150. 134. Altwegg M, Fleisch-Marx A, Goldenberger D, et al. Spondylodiscitis
101. Schochet SS Jr, Lampert PW. Granulomatous encephalitis in Whipples dis- caused by Tropheryma whippelii. Schweiz Med Wochenschr. 1996;126:
ease. Electron miscroscopic observations. Acta Neuropathol. 1969;13:111. 14951499.
102. de Groodt-Lasseel M, Martin JJ. Ultrastructural study of central ner- 135. Weber U, Morf MH, Gubler JG, et al. Spondylodiscitis as the first mani-
vous system lesions in Whipples disease [in French]. Pathol Biol (Paris). festation of Whipples diseasea removal worker with chronic low back
1969;17:121132. pain. Clin Rheumatol. 2003;22:443446.

Scheld_Ch28.indd 472 2/21/14 8:10 PM


Chapter 28: Whipples Disease 473

136. Swash M, Schwartz MS, Vandenburg MJ, et al. Myopathy in Whipples did not have intestinal Whipples disease. J Clin Microbiol. 2004;42:
disease. Gut. 1977;18:800804. 44874493.
137. Van Bogaert L, Lafon R, Pages P, et al. Sur une encphalite subaigunon 159. Sloan LM, Rosenblatt JE, Cockerill FR III. Detection of Tropheryma
classable, principalement caractrise par des myorhythmies oculo-facio- whipplei DNA in clinical specimens by LightCycler real-time PCR. J Clin
cervicales. Rev Neurol (Paris). 1963;109:443453. Microbiol. 2005;43:35163518.
138. Grotta JC, Pettigrew LC, Schmidt WA, et al. Oculomasticatory myorhyth- 160. Taskn K, Schulz T, Elgjo K, et al. Diagnostic utility of the polymerase
mia. Ann Neurol. 1987;22:395396. chain reaction in 2 cases of suspected Whipple disease. Arch Intern Med.
139. Adler CH, Galetta SL. Oculo-facial-skeletal myorhythmia in Whipple dis- 1998;158:801803.
ease: treatment with ceftriaxone. Ann Intern Med. 1990;112:467469. 161. Jankovic J. Whipples disease of the central nervous system in AIDS. N
140. Tison F, Louvetgiendaj C, Henry P, et al. Permanent bruxism as a manifes- Engl J Med. 1986;315:10291030.
tation of the oculo-facial syndrome related to systemic Whipples disease. 162. Maiwald M, Meier-Willersen HJ, Hartmann M, et al. Detection of
Mov Disord. 1992;7:8285. Tropheryma whippelii DNA in a patient with AIDS. J Clin Microbiol.
141. Hausser-Hauw C, Roullet E, Robert R, et al. Oculo-facio-skeletal myo- 1995;33:13541356.
rhythmia as a cerebral complication of systemic Whipples disease. Mov 163. Wang HH, Tollerud D, Danar D, et al. Another Whipple-like disease in
Disord. 1988;3:179184. AIDS? N Engl J Med. 1986;314:15771578.
142. Amarenco P, Roullet E, Hannoun L, et al. Progressive supranuclear palsy 164. Roth RI, Owen RL, Keren DF, et al. Intestinal infection with
as the sole manifestation of systemic Whipples disease treated with pe- Mycobacterium avium in acquired immune deficiency syndrome (AIDS).
floxacine. J Neurol Neurosurg Psychiatry. 1991;54:11211122. Histological and clinical comparison with Whipples disease. Dig Dis Sci.
143. Simpson DA, Wishnow R, Gargulinski RB, et al. Oculofacial-skeletal 1985;30:497504.
myorhythmia in central nervous system Whipples disease: additional case 165. Rolain JM, Fenollar F, Raoult D. False positive PCR detection of
and review of the literature. Mov Disord. 1995;10:195200. Tropheryma whipplei in the saliva of healthy people. BMC Microbiol.
144. Rickman LS, Freeman WR, Green WR, et al. Uveitis caused by Tropheryma 2007;7:48.
whippelii (Whipple bacillus). N Engl J Med. 1995;332:363366. 166. Goyo D, Camacho A, Gomez C, et al. False-positive PCR detection
145. Font RL, Rao NA, Issarescu S, et al. Ocular involvement in Whipples of Tropheryma whipplei in cerebrospinal fluid and biopsy samples
disease: light and electron microscopic observations. Arch Ophthalmol. from a child with chronic lymphocytic meningitis. J Clin Microbiol.
1978;96:14311436. 2009;47:37833784.
146. Knox DI, Bayless TM, Yardley JH, et al. Whipples disease presenting with 167. Barrett AM. Is it Alzheimers disease or something else? 10 disor-
ocular inflammation and minimal intestinal symptoms. Johns Hopkins ders that may feature impaired memory and cognition. Postgrad Med.
Med J. 1968;123:175182. 2005;117:4753.
147. Avila MP, Jalkh AE, Feldman E, et al. Manifestations of Whipples disease 168. Levy M, Poyart C, Lamarque D, et al. Whipples disease: acquired resis-
in the posterior segment of the eye. Arch Ophthalmol. 1984;102:384 tance to trimethoprim-sulfamethoxazole. Am J Gastroenterol. 2000;95:
390. 23902391.
148. Chan RY, Yannuzzi LA, Foster CS. Ocular Whipples disease: earlier de- 169. Bakkali N, Fenollar F, Biswas S, et al. Acquired resistance to trime-
finitive diagnosis. Ophthalmology. 2001;108:22252231. thoprim-sulfamethoxazole during Whipple disease and expression of the
149. Touitou V, Fenollar F, Cassoux N, et al. Ocular Whipples disease: thera- causative target gene. J Infect Dis. 2008;198:101108.
peutic strategy and long-term follow-up. Ophthalmology. 2012;119: 170. Fenollar F, Rolain JM, Alric L, et al. Resistance to trimethoprim/sul-
14651469. famethoxazole and Tropheryma whipplei. Int J Antimicrob Agents.
150. Williams JG, Edward DP, Tessler HH, et al. Ocular manifestations of 2009;34:255259.
Whipple disease: an atypical presentation. Arch Ophthalmol. 1998;116: 171. Singer R, von Herbay A, Willig F. Successful treatment of cerebral
12321234. Whipples disease with rifampicin. Med Klin. 1995;90:117118.
151. Drancourt M, Berger P, Terrada C, et al. High prevalence of fastidious bac- 172. Boulos A, Rolain JM, Raoult D. Antibiotic susceptibility of Tropheryma
teria in 1520 cases of uveitis of unknown etiology. Medicine (Baltimore). whipplei in MRC5 cells. Antimicrob Agents Chemother. 2004;48:
2008;87:167176. 747752.
152. Verhagen WI, Huygen PL, Dalman JE, et al. Whipples disease and the 173. Boulos A, Rolain JM, Mallet MN, et al. Molecular evaluation of antibiotic
central nervous system: a case report and a review of the literature. Clin susceptibility of Tropheryma whipplei in axenic medium. J Antimicrob
Neurol Neurosurg. 1996;98:299304. Chemother. 2005;55:178181.
153. Wiethlter H, Dichgans J. Diagnosis of cerebral Whipples disease by ce- 174. Cannon WR. Whipples disease, genomics, and drug therapy. Lancet.
rebrospinal fluid cytology. Arch Psychiatr Nervenkr. 1982;231:283287. 2003;361(9372):1916.
154. Black DF, Aksamit AJ, Morris JM. MR imaging of central nervous 175. Bakkali N, Fenollar F, Rolain JM, et al. Comment on: therapy for
system Whipple disease: a 15-year review. AJNR Am J Neuroradiol. Whipples disease. J Antimicrob Chemother. 2008;61:968969.
2010;31:14931497. 176. Fenollar F, Raoult D. Infection: how should classic Whipple disease be
155. Hinrikson HP, Dutly F, Altwegg M. Homogeneity of 16S-23S ribosomal managed? Nat Rev Gastroenterol Hepatol. 2010;7:246248.
intergenic spacer regions of Tropheryma whippelii in Swiss patients with 177. Feurle GE, Moos V, Schneider T, et al. The combination of chloroquine
Whipples disease. J Clin Microbiol. 1999;37:152156. and minocycline, a therapeutic option in cerebrospinal infection of
156. Fenollar F, Fournier PE, Robert C, et al. Use of genome selected repeated Whipples disease refractory to treatment with ceftriaxone, meropenem
sequences increases the sensitivity of PCR detection of Tropheryma whip- and co-trimoxazole. J Antimicrob Chemother. 2012;67:12951296.
plei. J Clin Microbiol. 2004;42:401403. 178. Feurle GE, Moos V, Schinnerling K, et al. The immune reconstitution
157. Edouard S, Fenollar F, Raoult D. The rise of Tropheryma whipplei: a 12- inflammatory syndrome in Whipple disease: a cohort study. Ann Intern
year retrospective study of PCR diagnoses in our reference center. J Clin Med. 2010;153:710717.
Microbiol. 2012;50:39173920. 179. Lagier JC, Fenollar F, Lepidi H, et al. Successful treatment of immune
158. Dreier J, Szabados F, von Herbay A, et al. Tropheryma whipplei infec- reconstitution inflammatory syndrome in Whipples disease using thalido-
tion of an acellular porcine heart valve bioprosthesis in a patient who mide. J Infect. 2010;60:7982.

Scheld_Ch28.indd 473 2/21/14 8:10 PM


CHAPTER 29 TUBERCULOUS MENINGITIS
DOROTHEE HEEMSKERK, JEREMY FARRAR, AND MAXINE CAWS

Yea, I have known inflammations, Imposthumes, whelks, scirrhus the disease in four different stages, according to the pulse of the
Tumors growing to the Meninges, with the Skull, and other patient. He emphasized that the terminology Dropsy upon the
Diseases of an evil conformation, excited in the Membranes of brain (-
[hydrocephalus]  [on]  

the Brain; by which at first for a long time, frequent headache, [the brain] as described by Hippocrates [460 to 377 bc] in his
and most cruel, and then afterwards a sleepy and deadly distemper
hath been induced; the cause of the Disease not detected, but
De Morbis Popularibus) is in these cases incorrect, because the
after Death by the Anatomy; and indeed it is to be suspected that accumulated fluid is not found between the skull and the dura
inveterate and pertinacious pains in the Head, which return, and mater but most frequently in the ventricles. The antediluvian
daily become more tormentive, in spight of all Remedies depend technique proposed by Hippocrates to make a perforation in the
upon some such invincible Cause. upper part of the cranium to evacuate the fluid Whytt concluded
was of no use (3). This publication gave an impetus to scientists
Thomas Willis (16211675), from De Anima Brutorum
to study this condition systematically and many contributions
(1672) (1)
followed with different views on the origin of the disease. Some
considered the arachnoid the seat of the pest, others lesions in the
brain parenchyma, the ventricles, until finally Penn in 1825, who
HISTORY thought the origin was in the pia mater, called it meningitis (1).
The term tubercular meningitis was first used in 1836
Tuberculosis by P.H. Green in the Lancet. Green introduced the term tu-
bercular meningitis to describe the condition of the cerebral
Tuberculosis (TB) has been a part of everyday human life since membranes, which were affected by tubercular lesions in nine
ancient times. There is evidence of TB in man dating back to tenths of the cases in a series of 45 children, who at the same
4000 bc, but the disease may have been present even earlier. time had tubercular deposits in the lungs or the abdomen.
Before the discovery of the causative agent (Mycobacterium Green argued that these findings were a more essential
tuberculosis), TB, in its many forms, has had many different characteristic of the disease than the accumulation of cerebro-
syndromal descriptions: phthisis, consumption, scrofula, Pott spinal fluid (CSF) (4,5). The condition was uniformly lethal.
disease, or others less well known such as yaksma (Indian)
Elucidating Pathogenesis
and Chaky Onkay (Incan). The term tubercle was first used
by Franciscus de la Boe, also known as Sylvius of Leyden By the end of the nineteenth century, attempts were made
(16141672). He stated that tubercles were often seen in the to relieve symptoms from raised intracranial pressure and
lungs of consumptives (2). He was also accredited with dis- hydrocephalus. Walter Wynter (1860 to 1945) had devised a
covering a cleft in the brain consequently named the sylvian crude technique to puncture the lumbar subarachnoid space.
fissure, which we now know to be a preferential site for He successively performed this archaic form of lumbar punc-
exudates formed in tuberculous meningitis (TBM). ture on four patients with TBM to relieve symptoms but
with short-lived improvement (widening of the pupils and
The Origin of Tuberculous Meningitis temporary improvement of sensorium), but all four patients
died (6,7). Morton (1891) describes the clinical findings of
The earliest descriptions of intracranial TB date back to the a series of patients during illness and the pathologic findings
seventeenth century. Physicians frequently used the term acute in the brain postmortem to further explore whether there
hydrocephalus or dropsy of the brain for a condition in would be a rationale for Wynters procedure. Although failing
children of which the etiology was unknown but presented with to clearly associate the extent of the hydrocephalus to the
fever, headache, vomiting, and rapid death. Some of the his- observed clinical picture, he concluded:
torical descriptions vividly illustrate the despair of both patient
and doctor; some pathologic descriptions were punctilious and The operation does no harm, and as the patient is already comatose
very archetypal. no anaesthetic is required. But in any efforts we may make to
remove the more serious symptoms of tuberculous meningitis by
Nec minus a phlegmone et abcessu quam hujasmodi meningitis et draining the intraventricular fluid we must remember it is nearly
tuberculis, cephalgiae lethales et incurabiles oriuntur (Sometimes always only part of general tuberculosis, which may, and prob-
the headaches, fatal and incurable, follow abscesses and swellings ably will, prove fatal in other ways, though if in this rapidly fatal
of the envelopes of the brain, as well as placques and tubercles of meningitis we can prolong life it may be some time longer before
these membranes). (Willis, 1672 [1]) the general tuberculosis does its deadly work. (8)
Willis was far ahead of his time as it was not until 150 years Tuberculous meningitis remained universally fatal.
later that the tubercles found upon autopsy were regarded as a It was not Wynter but Heinrich Quincke who began to
distinguishing aspect of the clinical syndrome, which was only popularize the lumbar puncture for both therapeutic and
then proposed to be tubercular meningitis. diagnostic purposes in the late nineteenth and early twentieth
Attempts were made to define this disease entity based century. With the discovery of M. tuberculosis by Robert Koch
on clinical and autopsy findings, but due to the multiform in 1892 and the development of x-rays by Wilhelm Roentgen
presentation both clinically and pathologically, consensus was in 1895, diagnosis before death was now achievable, although
not reached until Robert Whytt (1714 to 1766) lifted the disease effective treatment remained elusive.
out of obscurity with his treatise Observations on the dropsy in The disease was still thought to develop in a manner analo-
the brain. He gives a detailed account of 20 patients, dividing gous to other meningitides until a meticulous serial autopsy

474

Scheld_Ch29.indd 474 2/21/14 8:21 PM


Chapter 29: Tuberculous Meningitis 475

study of 82 patients by Rich and McCordock (9) in 1933 to 1947) expanded on these ideas and developed the Mantoux
provided the basis of what still now is considered to be the diagnostic test in 1907, which is still in use today. During 1902
establishment of pathogenesis. In guinea pigs and rabbits, to 1920, Albert Calmette and his assistant Camille Gurin de-
Rich and McCordock (9) could only provoke inflammation veloped a vaccine by serial passage of Mycobacterium bovis
of the meninges by the direct inoculation of bacilli into the (which can cause TB in both cattle and humans). After 13 years
central nervous system (CNS) and not by peripheral injec- and 230 passages, the bacille Calmette-Gurin (BCG) strain
tion and consequent hematogenous spread. In human autopsy was considered attenuated and was first used as a vaccine in
studies, they described that the tubercles found in the brain humans in 1921 (2).
were seldom of the same age as those found in other organs The accomplishments in the first half of the twentieth cen-
and that vasculitis found in the brain was rather a process tury in diagnosis, treatment, and understanding of pathogenesis
originating from the adventitia inward, more likely to be the seemed to offer the opportunity of eradication. However, the
result of a focus within the brain rather than caused by di- next 60 years has been fraught with setbacks and TB remains
rect hematogenous spread of bacilli. These findings led them a huge global public health problem. The first cases of AIDS
to form a coherent hypothesis in which they postulated that, came to light in the early 1980s. Since then, it has become clear
after inhalation of the pathogen, a short-lived bacteremia fol- that the interaction of TB and HIV has had a severe impact
lowed, during which bacilli spread throughout the body and on both pandemics, while complicating the management of
seeded the surface of the brain, forming small granulomas both diseases. HIV-infected individuals are more susceptible
known as Rich foci. These can exist without causing symp- to both active TB disease and all extrapulmonary forms of TB.
toms for an unknown period but may rupture; upon release The HIV epidemic has therefore generated significant increases
of the mycobacteria, meninges become inflamed, giving rise to in the number of adults presenting with TBM in high HIV
a multitude of possible pathologic tuberculous conditions in prevalence areas. Drug-resistant strains of M. tuberculosis
the CNS (9). have continued to increase in prevalence, including multidrug-
resistant (MDR) and extensively drug-resistant (XDR) TB,
Treatment Development which are now present in every region of the world. Huge
advances have been made in unravelling immunology, and
The discovery of the causative pathogen was met with eu- genetics has deepened our understanding of the immunopa-
phoria, as new hopes were raised of a cure. Experimental thology underlying disease; however, vaccination, diagnosis,
treatment was attempted with tuberculin, originally a glyc- and treatment are still reliant on antiquated techniques, which
erin extract of M. tuberculosis developed by Koch, but with are inadequate to control the continued pandemic.
catastrophic results. Other therapies raised hopes but proved
ineffective, including sanocrysin (gold therapy) (10). In 1944,
the first effective antituberculous agent, streptomycin, was dis- BURDEN OF DISEASE
covered by Salman Waksman, a discovery for which he would
win the Nobel Prize for Medicine in 1950. By 1948, dozens of
cases of successfully treated TBM with intrathecal and intra- Tuberculosis Epidemiology
muscular streptomycin were reported in the literature. Rich
and Samuels (11) reviewed these cases while giving a striking In the nineteenth century, TB was highly prevalent in Europe,
account of a case involving a 2-year-old boy who at the height with an annual incidence estimated to be greater than 1,000
of his disease was in a vegetative state but within 6 months per 100,000. Mortality was high because patients were only
recovered with residual weakness of his left arm and minor treated with bed rest or ineffective therapies and it is thought
mental impairment. Many publications followed on successful that over 50% of sufferers died. In the time of Robert Koch,
streptomycin treatment. However, by 1950, numerous strepto- one out of seven Germans died of TB. In the late nineteenth and
mycin resistance reports appeared (1217). Paraaminosalicylic early twentieth century, incidence declined in more developed
acid (PAS) was added to the regimen; although a weak antitu- countries due to economic development, improved living con-
berculous agent, it prevented development of resistance. With ditions, hygiene, and the introduction of sanatoria in which
the introduction of isoniazid (1952), a major improvement in patients were isolated from the general population. After the
treatment of all forms of TB was seen. It became clear that introduction of antimycobacterial treatment, the decline in
intrathecal administration was no longer necessary. With the incidence and mortality accelerated. The initial response was
introduction of pyrazinamide (1954) and rifampicin (1963), euphoric, and expectations to extinguish this blazing epidemic
treatment regimens for all presentations of drug-susceptible were high.
TB could be shortened to 6 to 8 months. The relative con- Despite the initial achievements during the twentieth cen-
tributions of the first-line TB drugs to the efficacy of TBM tury, global TB burden remains enormous. Worldwide, approx-
treatment were comprehensively reviewed by Donald in 2010 imately 2.4 billion people are infected with M. tuberculosis, of
(18). Fifty years on these drugs isoniazid, pyrazinamide, rifam- whom 10% will develop active disease during their lifetime.
picin, and streptomycin remain the mainstay of treatment for People infected with HIV with latent TB are 20 to 30 times as
the vast majority of patients with TB globally. It is difficult to likely to develop active forms of TB (19).
think of another common infectious disease whose treatment A systematic analysis for the global burden of disease study
regimen has remained largely unchanged for over 50 years. in 2010 including mortality data from 187 countries from
1980 to 2010 ranked TB as the 10th leading cause of death
globally. Of the 52.8 million deaths of all causes globally in
Vaccine Development 2010, 1.2 million were attributable to TB (20). According
Despite failing to find a cure, Koch continued experimenting to the World Health Organization (WHO), 8.7 million new
with tuberculin hoping to develop an effective vaccine; how- cases were reported in 2011 and an estimated 1.4 million died
ever, it proved not to be effective. Clemens van Pirquet (1874 (21). The Millennium Development Goals aimed to achieve a
to 1929), after observing a hypersensitivity reaction to a sec- 50% reduction in prevalence and death rates of TB relative to
ond inoculation with smallpox vaccine, was led to the idea that 1990 levels by the year 2015. According to the WHO, from
Kochs tuberculin might cause a similar reaction in patients 1990 to 2011, a reduction of 41% in mortality was observed.
previously exposed to mycobacteria. Charles Mantoux (1877 According to the WHO, if the current trend is preserved, the

Scheld_Ch29.indd 475 2/21/14 8:21 PM


476 Part III: Bacterial and Mycoplasmal Infections

set target for 2015 will be met (21). However, this is depen- TA B L E 2 9 . 1
dent on the continued commitment of national TB programs
and policy makers. The TB epidemic is highly pluriform, with TUBERCULOUS MENINGITIS SEVERITY GRADING
22 developing countries carrying more than 80% of the bur- ACCORDING TO MEDICAL RESEARCH COUNCIL 1948
den of TB. Demographic factors such as poverty, crowding,
and malnutrition play an important role, as does availability Early Patients with mainly nonspecific symptoms, with
of good quality TB drugs. The impact of the HIV pandemic little or no clinical signs of meningitis, with no
is illustrated by the huge increase in the contribution of HIV/ pareses, in good general condition, and fully
TB to cause of death patterns among young adult men and conscious. Diagnosis established mainly on
women; by 2010, HIV/TB and injuries combined caused more findings in cerebrospinal fluid (CSF).
than half of deaths among men aged 20 to 39 years (20). Advanced Patients obviously extremely ill, deeply
Together with HIV, MDR and XDR TB continue to fuel the comatose, or with gross pareses
pandemic (21). The goal to eliminate TB as a public health Medium Patients in a condition between those of the first
problem by 2050 seems quixotic without renewed and sus- two groups
tained commitment from international donors and the global
health community. From Medical Research Council. Streptomycin treatment of tubercu-
lous meningitis. Lancet. 1948;1:582596.

Burden of Tuberculosis of the Central


Nervous System
adults, permanent neurologic disability affects over half of the
The burden of extrapulmonary TB is tightly associated with survivors (30). Co-infection with HIV has a major impact on
the general pandemic and accounts for more than 10% of all mortality. In Vietnam, mortality for the corresponding grade
TB cases (21). Of these, about 5% are forms of TB in the CNS. groups in a trial of immediate versus delayed antiretroviral
These estimates are crude because diagnosis of extrapulmonary therapy (ART) for HIV-associated adult TBM was stage I,
TB in general and CNS TB in particular is challenging, and 40%; stage II, 52%; and stage III, 75% (31). In HIV-infected
according to WHO definitions, a patient who has both signs children in India, 6-month mortality has also been reported
of pulmonary and extrapulmonary TB should be classified as to be drastically higher than in HIV-negative children (36%
having pulmonary TB (21). vs. 10%) (32). These mortality figures are a reflection of the
In endemic settings, TB is often the leading cause of child- severe immunosuppression in these HIV-infected patients,
hood bacterial meningitis (22). In 2009, 7% of all annual illustrating the importance of commitment of policymakers to
cases of bacterial meningitis and septicemia in the United invest in accessible integrated TB/HIV care.
Kingdom were caused by TB, being the third leading cause
after meningococcal and pneumococcal disease (23). With The Influence of HIV
the introduction of meningococcal C (1999) and pneumococ-
cal (2006) vaccines in the routine immunization schedule in The HIV epidemic has undoubtedly fuelled the TB epidemic.
the United Kingdom (and in the absence of an effective TB Worldwide, an estimated 34 million people are infected with
vaccine), TBM may well become the lead cause. HIV (33). An estimated one third of these patients are thought
In high TB-burden countries, young children aged 0 to to be co-infected with M. tuberculosis. HIV-infected patients
4 years are mostly affected, however, rarely, younger than with latent TB have a 50% lifetime risk of progressing to active
3 months of age (24). The age range from 5 to 15 years old is forms of TB versus HIV-negative patients with latent TB who
often referred to as the favored age as this population has have a 10% lifetime risk of developing disease (34). HIV pa-
the lowest rate of TB of all forms (25). In countries with a low tients are more likely to develop extrapulmonary forms of TB,
prevalence of TB, most cases of TBM are in adults. Commonly including TBM, and have a higher mortality. Diagnosis of TB
reported risk factors are alcoholism, diabetes, recent cortico- can be more challenging in HIV patients due to a less specific
steroid use, malignancy, and immunosuppression (26). The presentation, wider differential diagnosis, and lower sensitiv-
advent of HIV has dramatically changed the dynamics of the ity of sputum smear microscopy. However, in contrast, CSF
TB pandemic. smear is more likely to be positive in HIV-infected individuals
with TBM due to higher bacillary loads (35). Patients with low
Mortality and Morbidity CD4 levels and smear-negative results may have atypical CSF
findings, immunologic tests are not reliable, and neuroimag-
Mortality among TBM cases remains high and varies depend-
ing may not reveal typical lesions and includes a wider differ-
ing on age, risk group (HIV), stage of disease upon diagno-
ential diagnosis (36). Polypharmacy may prove problematic,
sis, drug sensitivity of the infecting organism, time between
with higher toxicity from combined ART and antitubercu-
onset of symptoms and initiation of effective antibiotics,
lous regimens and drug interactions. ART-naive patients who
and the sophistication of health care infrastructure and fa-
commence ART treatment during their treatment of TBM may
cilities. Mortality in children ranges between 10% and 20%
present with immune reconstitution inflammatory syndrome
(24,25,27,28). Neurologic sequelae are frequent and reported
(IRIS), which may be particularly detrimental if it presents
in more than half of surviving children. In a retrospective sur-
intracranially (37).
vey of 554 children in South Africa, 74% of patients suffered
long-term disabilities or death (13%), including hearing or
vision impairment (14% and 16% respectively), motor deficits
Drug Resistance
(44%), and cognitive impairment (77%) (29). WHO estimates 20% of M .tuberculosis infections worldwide
For HIV-uninfected adult patients presenting early (stage I are now resistant to at least one first-line drug (38). MDR
disease), mortality is approximately 20% (30). Mortality TBM is resistant to at least rifampicin and isoniazid, the two
increases with delayed presentation and advanced stage of most effective first-line agents. MDR TBM has been associated
disease (Table 29.1). For patients in stage II, mortality is with very high mortality in both children and adults (3941).
approximately 30% and in stage III, 55%. As in children, in Drug resistance in TBM is rarely diagnosed in time to make

Scheld_Ch29.indd 476 2/21/14 8:21 PM


Chapter 29: Tuberculous Meningitis 477

appropriate treatment adjustments. The difficulties of access Neurologic sequelae most frequently described in adults are
to rapid TB drug susceptibility testing (DST) in much of the cognitive impairment, motor deficits, cranial nerve palsy, and
world are compounded by the rarity of a positive isolation optic atrophy (47). A 5-year follow-up on Vietnamese adults
of M. tuberculosis from the CSF. Drug resistance prevalence with TBM who took part in a randomized controlled trial on
among TBM cases will generally follow similar patterns to the effect of dexamethasone on survival could only demonstrate
those observed regionally for pulmonary TB. an overall benefit of dexamethasone up to 2 years following
Isoniazid resistance in the absence of concomitant rifam- treatment. Five years after treatment completion, there was
picin resistance is more prevalent than MDR TB; 7% of M. no difference in overall survival or disability outcome in both
tuberculosis strains globally are now resistant to isoniazid (38). groups. In the group receiving dexamethasone, 48.4% (vs.
Because isoniazid is the most effective drug in decreasing the 52.7% in placebo group) of patients had died at 5 years (31%
bacterial load in the first two days of anti-TB treatment, early at 9 months), 6.8% (vs. 7.4% in the placebo group) were se-
recognition of resistance is of great importance, particularly in verely disabled, 17.2% (vs. 14.8% in placebo group) had inter-
TBM cases where rapid killing of bacilli is likely to be crucial. In mediate disability, and only 27.6% (vs. 25.1% in the placebo
Vietnam, isoniazid resistance alone or combined with resistance group) had good outcome (48). However, the beneficial effect
to other drugs was found in a third of culture-positive samples of dexamethasone was preserved at 5 years for patients with
in adult HIV patients with TBM, with 4.3% MDR TBM (40). stage I TBM at presentation, demonstrating that, contrary to
In a cohort with predominantly HIV-negative patients, isonia- preceding medical wisdom, patients with stage I TBM are the
zid resistance was found in 37.1% of samples of which 21% group who gain the greatest benefit from corticosteroids.
were MDR (overall MDR rate was 5.6%) (39). Mortality from
MDR TBM was 100% in the absence of available second-line
therapy. In children with a culture-proven diagnosis of TBM, Vaccination with Bacilli Calmette-Guerin and
MDR TB or rifampicin monoresistance was identified in 5% Protection Against Tuberculous Meningitis
of cases in South Africa. Multidrug resistance, not surprisingly,
was associated with very high mortality (83%) (42). The controversies surrounding the protection that the BCG vac-
Isoniazid resistance without rifampicin resistance also has a cine confers to adult pulmonary and meningeal TB still exist. In
significant impact on mortality. An initial analysis of a cohort adults, reported efficacy against all forms has ranged from as
of TBM patients in Vietnam failed to find a significant im- high as 80% to 0%. It is proposed that the vaccine establishes
pact of isoniazid resistance (/ streptomycin resistance) on immunity by inducing effector memory T cells in the lungs that
mortality (43), but a larger study in Vietnamese HIV-positive gradually wane after 10 to 15 years rather than building a lon-
patients showed a significant reduction in survival (adjusted ger lasting central memory (49). Various theories have been
hazard ratio [HR], 1.78; 95% confidence interval [CI], 1.18 to proposed for the differences observed in efficacy, including dif-
2.66) among patients infected with isoniazid-resistant, rifam- ferences in the circulating M. tuberculosis strains, BCG vaccine
picin-susceptible strains compared to those with fully suscep- strains, or preimmunization exposure to environmental myco-
tible isolates (40). In a retrospective cohort study in the United bacteria. Nonetheless, consensus on the benefit of prevention of
States, researchers also found a significant increase in risk of severe forms of childhood TB including TBM and miliary TB is
death associated with isoniazid resistance (odds ratio [OR], established. The vaccine is thought to be 52% to 86% protec-
1.61; 95% CI, 1.08 to 2.40) (44). Among Vietnamese HIV- tive against developing the severe complications of childhood
positive adult patients, the adjusted hazard ratio for mortality TB such as miliary TB and TBM (50). It has been estimated that
of MDR TBM patients compared to patients with isolates sus- the 100.5 million BCG vaccinations given to infants in 2002
ceptible to all agents or streptomycin monoresistant was 5.21 would have prevented 29,729 cases of TBM in children during
(95% CI, 2.38 to 11.42) (Fig. 29.1). their first 5 years of life, or 1 case for every 3,435 vaccinations,
and 11,486 cases of miliary TB, or 1 case for every 9,314 vac-
Long-Term Disability cinations. Based on these data, it is considered a cost-effective
intervention in Southeast Asia, Africa, and the Western Pacific,
Very little is known about long-term outcome and disability where TB infection rate and vaccine coverage are highest (50).
for both children and adults. Antituberculous chemotherapy In BCG-vaccinated children in India who do develop TBM, the
has been unsuccessful in completely preventing long-term clinical spectrum of disease does not seem to be ameliorated
sequelae; in many cases, diagnosis may be too late, but in (51). The BCG is the most widely used vaccination globally.
others, significant neurologic damage occurs subsequent to
initiation of treatment. Especially in children, neurocogni-
tive impairment can jeopardize development, education, and
quality of life and place a great burden on families, schooling,
IMMUNOPATHOGENESIS
and medical systems. A recent long-term follow-up study on
a South African cohort of pediatric TBM patients who were Mycobacterium tuberculosis
severely ill on presentation (stage II or III) reported only 20%
of children to be functionally normal at follow-up (median Transmission of TB occurs when a person inhales mycobacteria-
6 years after TBM treatment completion). The main areas laden droplet nuclei. One to five bacilli are sufficient to cause in-
of functional deficit were cognitive impairment (80%), poor fection. M. tuberculosis is the causative agent of almost all cases
scholastic progress (43%), and emotional disturbance (40%). of TBM. M. tuberculosis is an obligate aerobic, intracellular
A smaller proportion of children (25%) had evidence of bacterium. On the cell surface, it has a waxy coating. The high
motor impairment (45). In a Danish nationwide, population- lipid content of the cell wall renders these bacilli imperceptible
based cohort study with up to 30 years follow-up, TBM was to the Gram stain. The organisms are slow growing with a gen-
associated with an almost twofold increased long- term risk of eration time of 15 to 20 hours, contrasting with that of some
dying compared to a background population (mortality risk pyogenic bacteria, such as Streptococcus pneumoniae, Neisseria
ratio [MRR], 1.79; 95%, CI, 1.09 to 2.95). In this study, the meningitides, and Staphylococcus aureus, with generation times
underlying cause of long-term death was most frequently TBM of less than an hour. The complex antigenic structure of the
itself rather than secondary to the most commonly reported cell wall includes polysaccharides, proteins, peptides, lipids,
neurologic sequelae (46). and glycolipids with specific immunologic properties. Other

Scheld_Ch29.indd 477 2/21/14 8:21 PM


478 Part III: Bacterial and Mycoplasmal Infections

A Survival by resistance group B Survival by resistance category


1.00 1.00
Survival Probability

Survival Probability
0.75 0.75

Sensitive/SM mono-resistant
Fully sensitive
0.50 0.50
SM mono-resistant
INH+/-SM resistance
0.25 INH mono-resistant 0.25

MDR INH+SM resistant MDR

0.00 0.00
0 3 6 9 0 3 6 9
Months Since Randomization Months Since Randomization
No. at risk No. at risk
Fully sensitive 98 52 41 35 Sensitive 126 66 53 47
STR mono-resistant 28 14 12 12 INH+/-STR resistance 52 25 11 8
INH mono-resistant 12 6 2 2 MDR 8 0 0 0
INH+STR resistant 40 19 9 6
MDR 8 0 0 0

C Survival by lineage D Survival by lineage of patients infected with


fully susceptible M. tuberculosis strains only
1.00 1.00
Survival Probability

Survival Probability

0.75 0.75
East Asian/Beijing

0.50 East Asian/Beijing 0.50

Euro-American Euro-American
0.25 0.25
Indo-Oceanic
Indo-Oceanic
0.00 0.00
0 3 6 9 0 3 6 9
Months Since Randomization Months Since Randomization
No. at risk No. at risk
Indo-Oceanic 18 5 3 3 Indo-Oceanic 14 3 2 2
Euro-American 22 10 6 5 Euro-American 14 7 6 5
Beijing 82 43 30 26 Beijing 55 33 26 23

FIGURE 29.1 Survival by drug-resistance pattern of pathogen of an HIV-positive cohort. SM,


streptomycin; INH, isoniazid; MDR, multidrug resistant. (Tho DQ, Trk ME, Yen NT. Influence of
antituberculosis drug resistance and Mycobacterium tuberculosis lineage on outcome in HIV-associated
tuberculous meningitis. Antimicrob Agents Chemother. 2012;56[6]:30743079.)

antigens are contained within the cytoplasm. These molecules eradicate M. tuberculosis, T cells are recruited to the site of
determine the characteristic immune response to tuberculous infection and generate a chronic inflammation and granuloma
infection and its resultant pathology. formation in order to contain the infection (52). The granu-
loma is pivotal to tuberculous disease and characterized by the
formation of central necrosis of the lesion, often referred to as
Macrophages and Granuloma Formation caseation. In solid necrosis, mycobacterial growth is inhibited,
and the infection can be contained and can remain dormant.
In pulmonary TB, the alveolar macrophage has a central role Granuloma with liquefied central necrosis provides an optimal
in the initial innate immune response to M. tuberculosis as environment for extracellular mycobacteria and may rupture,
well as in initiating the adaptive T-cell immunity. If upon allowing the spread of bacilli to other parts of the lung, the
recognition and ingestion of bacteria, the macrophages fail to bloodstream, or the exterior environment (53).

Scheld_Ch29.indd 478 2/21/14 8:21 PM


Chapter 29: Tuberculous Meningitis 479

The onset of the adaptive immune response to M. tuberculosis


is delayed compared to other infections. This delay allows for Central Nervous System Pathology
an exponential growth of the mycobacteria before it is slowed
or arrested by the host defenses (54). For the adaptive immune Unfortunately, current postmortem studies are scant (64,65)
response in TB, CD4 T cells are essential. CD4 T cells exert but would contribute a great deal to our understanding of the
their protective effect by the production of cytokines, primar- disease. Rabbit models are thought to mimic human disease;
ily interferon- (IFN-) and tumor necrosis factor-
(TNF-
). however, immunologic interpretation is imperfect. Murine
IFN- is a central protective cytokine in mycobacterial infection studies are not of use in directly studying neuropathogenesis
proposed to protect by mediating the induction of nitric oxide in TBM although are widely used in understanding suscep-
synthase (NOS), enhancing the microbicidal system within mac- tibility and protective responses to mycobacteria (66). Our
rophages (55). TNF-
plays a key role in granuloma formation current knowledge on pathogenesis of CNS TB is based on
and macrophage induction and has immunoregulatory prop- the hypothesis promulgated over 70 years ago. Even though
erties (53). Other cytokines involved in mycobacterial disease later researchers have expressed criticism (67), their view
control are interleukin (IL)-1B, IL-6, IL-12, IL-15, IL-18 (proin- has been carried forward by most recent experts; in short,
flammatory) and IL-10, IL-4, and transforming growth factor- small intracranial granulomas are formed after seeding of ba-
(TGF-) (antiinflammatory) (53,56). In addition to the complex cilli in the brain during a short-lived bacteremia. These Rich
regulatory meshwork of cells and immune mediators that con- foci may rupture during periods of relative immunosuppres-
trol the innate and adaptive immune response, different compo- sion or other unknown stimuli, releasing mycobacteria in the
nents may be negatively influenced by microbe-specific virulence subarachnoid space or ventricular space which may give rise to
factors. The virulence of the mycobacteria is thought be founded the various forms of CNS TB: meningitis, tuberculoma, tuber-
in their ability to regulate macrophage chemotaxis, necrosis, and culous abscess, encephalitis, or spinal cord TB. Incorporating
apoptosis, facilitating a beneficial environment within the mac- more recent pathologic data, Donald and colleagues (68) have
rophage or extracellularly in the granuloma and consequently proposed the following classification of pathogenesis in TBM:
their own proliferation and possibly also facilitating egress to 1. The hematogenous dissemination of bacilli from the pri-
other sites of infection, including the CNS (57). mary complex establishes a cortical or meningeal focus.
Soon after its establishment, this proceeds to caseate
and discharge its contents into the subarachnoid space.
Immune Response in Tuberculous Meningitis In young children, this hematogenous dissemination is
particularly likely to take the form of miliary TB.
TNF-
is thought to have a crucial but controversial role in
2. In a small minority of cases, hematogenous dissemination
pathogenesis of TBM. TNF-
is a proinflammatory cytokine
may establish a caseating focus in the choroid plexus or in
produced by monocytes, macrophages, and dendritic cells upon
the walls of the ventricles from which TBM may develop.
stimulation with mycobacteria or mycobacterial products and
3. Hematogenous dissemination at the time of primary infection,
plays an essential role in granuloma formation and mainte-
or later, establishes a cortical or meningeal focus. This is ini-
nance (58). Whereas in pulmonary TB, TNF-
-neutralizing
tially controlled but may, at any time thereafter, undergo case-
drugs can lead to progression to (fatal) disease, in TBM, an
ation and discharge its contents into the subarachnoid space.
elevated production of TNF-
has been proposed to be associ-
4. A caseous process extends from adjacent structures such as
ated with more severe disease, but it has not been established
the vertebrae or middle ear to involve the CNS (very rare).
if this is merely a marker of advanced disease or if dispro-
portionate TNF-
responses mediate disease progression. It is As a result of this infection, a dense gelatinous fibrinocellular
probable that protective immunity is dependent on a delicate leptomeningeal exudate is formed. Microscopically, this exudate
balance between pro- and antiinflammatory factors and that contains small and large mononuclear cells, including epitheli-
individuals with responses at the extremities of the spectrum oid cells, which also act as macrophages. The exudate typically
are both at risk of more severe disease. centers around the interpeduncular fossa. When substantial, the
In vitro infection of microglial cells (macrophages of the exudate may extend anteriorly to the suprasellar region, and it
brain) results in the production of robust amounts of TNF- may extend through the prepontine cistern and surround the

, IL-6, IL-1B, CCL2, CCL5, and CXCL10 (59). In rabbits spinal cord and cerebellum, often into the sylvian fissures. It can
infected intracisternally with M. bovis, oral thalidomide envelope and compress cranial nerves and arteries. Vasculitis
treatment led to reduction of TNF-
levels and clinical may develop, giving rise to ischemic events. Hydrocephalus can
improvement (60). In vivo in TBM, TNF-
levels show a peak develop by blockage of CSF circulation when exudates cover
in the early phase of disease but drop soon after initiation the choroid plexus and the basal subarachnoid cisterns around
of treatment (61). Some researchers found an association in the midbrain and pons or when tuberculoma cause narrowing
disease severity and CSF levels of IFN- and TNF-
in both of the aqueduct and third ventricle (66,69). Exudate, vasculi-
HIV-positive and HIV-negative patients (62). This was not tis, and hydrocephalus can cause changes in brain parenchyma.
supported in a Vietnamese cohort, in which the only cytokine Border-zone encephalitis describes a tissue reaction com-
independently associated with severe disease was IL-6. HIV monly seen in brain tissue adjacent to zones of thick adher-
co-infection was associated with attenuated levels of several ent exudate. The brain tissue softens, showing signs of edema,
immune mediators in CSF, in whom low levels of IFN- did perivascular infiltration, and microglial reaction (66,70). In the
show an association with death, implying a protective role for following paragraphs, we will briefly discuss other forms of
IFN-. Interestingly, the addition of dexamethasone to treat- CNS TB; the rest of the chapter will focus on TBM.
ment was not associated with an attenuation of inflammatory
indices but did lead to a decreased mortality rate (61).
The brain provides a unique immunologic environment to Tuberculoma in the Central Nervous System
pathogens. We know most of the inflammatory mediators are
produced locally at the site of infection. Immunologic studies CNS tuberculoma can be encountered separate from TBM, as
usually involve cells from peripheral blood or CSF. Compared to it is estimated only 10% of patients with tuberculoma develop
the infected tissue, even in pulmonary TB, low proportions of M. meningitis (71,72). Conversely, in some radiologic studies,
tuberculosisspecific effector cells are found in the blood (63). tuberculomas were observed either at presentation or developed

Scheld_Ch29.indd 479 2/21/14 8:21 PM


480 Part III: Bacterial and Mycoplasmal Infections

during treatment in over 60% of patients with TBM (73). In Stroke was reported in 13% to 57% of Indian patients with
endemic areas, tuberculoma represents the cause of up to 30% TBM (85). In Vietnam, serial MRI revealed stroke in only 9%
of patients presenting with intracranial masses. Tuberculoma of patients upon diagnosis, but after 60 days of treatment,
can be solitary or multiple, with some reporting hundreds of 41% of patients had developed stroke (73). This may be even
lesions in one patient, dubbed tuberculomatosis cerebri (71). more prevalent in children, with infarcts reported in 76% of
In general, tuberculomas in the context of TBM, although de- systematically scanned children in South Africa (86). Most in-
pending on the location, are not associated with worse out- farcts occur in the region of the arteria cerebri media (middle
come and will resolve on antituberculous treatment (74). cerebral artery [MCA]), particularly in the medial lenticulostri-
Rarely, tuberculomas coalesce and liquefy to cause tuberculous ate and thalamoperforating vessels, causing the characteristic
cerebral abscess, which may necessitate surgery (75). basal ganglia infarcts (84). It is thought that this vascular in-
volvement follows the distribution of the meningeal exudates,
which causes local vasculitis, particularly at the base of the
Tuberculous Encephalopathy brain and along the sylvian fissures. The proposed mechanisms
by which the vessels occlude resulting in ischemia are multi-
Tuberculous encephalopathy (TBE) was first described in 1966 form. Meticulous histopathologic descriptions of the vascular
in Indian children who presented with symptoms of a diffuse changes in TBM were published by Hektoen (87) in the late
cerebral involvement (coma, convulsions, movement disorders) nineteenth century. He concluded that the changes could be ei-
in the context of disseminated TB but normal cerebrospinal find- ther explained by an endarteritis with subendothelial tubercles,
ings (76). As there was no clear evidence of TB infection within proposed to be caused by direct hematogenous invasion of ba-
the brain, the authors proposed an alternative pathogenetic im- cilli in the vessel wall, or tuberculous proliferation affected the
mune-/hypersensitivity-mediated explanation for this syndrome, arteries from the adventitia inward to reach the media and the
pathologically characterized by white matter, myelin loss with intima (87). Currently, largely based on the findings of Rich
commensurate axonal loss, and focal necrosis. The principal and McCordock (9), the widely accepted view is that the in-
pathogenetic factor in the group of cases was said to be an allergic flammation is spread from outward (adventitia) in rather than
cerebral edema leading to an edematous leukoencephalopathy reversed. Other stenosing or damaging mechanisms are thought
similar to acute disseminated encephalomyelitis (ADEM) (77). to be intimal proliferation, vessel wall necrosis, or vasospasms.
However, use of steroids for these patients had proven ineffec- The role of vessel thrombosis is unclear. There is some evidence
tive. Careful review of the original publication and the literature that strokes early in the course of disease are caused by vaso-
of the following 40 years led South African experts to reappraise spasms and later strokes involve proliferative intimal disease,
this entity in 2007, concluding that the patient population joined raising the prospect for therapeutic interventions for the pre-
under the umbrella of TBE was clinically and histopathologi- vention of thrombotic and vasospasm-associated stroke (9,84).
cally heterogenous. According to the authors, other plausible
factors may have caused the typical findings of TBE, including
hypoxic ischemia and toxic or drug-related complications of TB The Role of Miliary Tuberculosis
infection (78). TBE has not been reported in adults.
Rich and McCordock (9) did not assign a role for miliary TB
in the pathogenesis of TBM as the Rich foci found in their
Tuberculosis of the Spinal Cord subjects were often older than the miliary lesions, and there-
fore military TB was deemed to be an incidental occurrence
Tuberculous radiculomyelitis (TBRM) has been used as a generic rather than part of the etiology. Even though this is the view
term to include arachnoiditis, intramedullary tuberculoma, and carried forward in most textbooks, some researchers claim
spinal cord complications of TBM (79). Currently, it is thought that, particularly in children and possibly in immunocompro-
that TBRM may develop in alternative ways, either (1) as a pri- mised patients, the disseminated state of miliary TB increases
mary lesion, (2) as an extension from TBM, or (3) secondary to the likelihood of the formation of leptomeningeal granuloma
vertebral TB (80). Intramedullary tuberculomas are rare. The (Rich foci) (68). This may well explain the frequent asso-
pathogenesis is parallel to CNS TB: via bloodborne seeding of ciation of TBM with miliary TB (Fig. 29.2). In children, the
bacilli, granuloma formation follows, drainage persists into the
subarachnoid space which may lead to an inflammatory reac-
tion in the pia-arachnoid, which may lead to secondary men-
ingitis (81). Clinical presentation may vary, usually presenting
with symptoms of a subacute intramedullary space-occupying
lesion (82). Early recognition of this form of CNS tuberculoma
is important because early surgical intervention and decom-
pression followed by long-term antituberculous chemotherapy
may significantly improve morbidity. Accurate diagnosis can be
helped by magnetic resonance imaging (MRI) (83).

Vascular Events
Secondary to the infection with M. tuberculosis and resultant
intracerebral immunologic response, stroke may develop. In the
late nineteenth and early twentieth century, vascular involvement
in TBM was recognized and extensively studied. Still the vas-
cular events in TBM are a pressing subject in TBM research,
because the damage caused by stroke is often irreversible and the
occurrence of stroke is associated with worse outcome (84,85). FIGURE 29.2 Chest x-ray showing miliary TB of the lungs.

Scheld_Ch29.indd 480 2/21/14 8:21 PM


Chapter 29: Tuberculous Meningitis 481

prevalence of miliary TB of the leptomeninges is much higher TA B L E 2 9 . 2


than in adults. Younger children most often develop both TBM
and miliary TB within 3 months of primary infection. Children PRESENTING CLINICAL FEATURES IN ADULTS AND
with concomitant miliary TB and TBM are also significantly OLDER CHILDREN
younger than those with TBM only (24). The immune system
of these young children and the immunocompromised may SYMPTOM FREQUENCY/RANGE
not be robust enough to prevent an overwhelming bacteremia,
exemplified by the miliary character of disease. In these indi- Headache 50%80%
viduals, TBM may be the result of a more direct spread of the Fever 60%95%
pathogen to the meninges and subarachnoid space. Vomiting 30%60%
Photophobia 5%10%
CLINICAL PRESENTATION Anorexia/weight loss 60%80%

CLINICAL SIGN
General Symptoms on Presentation
Neck stiffness 40%80%
TBM typically presents in a subacute manner. Presenting signs,
Confusion 10%30%
symptoms, CSF findings, and frequencies according to the British
Infection Society guidelines are shown in Table 29.2. In adults, Coma 30%60%
the majority of patients present with fever, headache, nuchal Cranial nerve palsy 30%50%
rigidity, vomiting, meningism, abnormal mental stage, and pho- VI 30%40%
tophobia (75,88,89). Weight loss, night sweats, lethargy, and
III 5%15%
cough have also be reported (90). The mean duration of symp-
toms is typically more than 5 days. A longer duration of history VII 10%20%
is associated with worse symptomatology on presentation. Hemiparesis 10%20%
In 1948, the British Medical Research Council first pub- Paraparesis 5%10%
lished a classification of TBM patients according to the severity
Seizures adults 5%
of disease (see Table 29.1). Over the years, these stage groups
have been refined and are published in different formats. In Children 50%
general, both adult and pediatric patients in stage I are fully
conscious and may have nonspecific symptoms; in stage II, CEREBROSPINAL FLUID
patients will have signs of meningitis, lethargy, or cranial FINDINGS
nerve palsies; in stage III, TBM is accompanied by stupor,
severe illness, gross paralysis, or paresis, convulsions, and or Clear appearance 80%90%
involuntary movements (51). In clinical practice and research, Opening pressure 25 cm H2O 50%
it would be more useful to have a Glasgow Coma Scale (GCS) Leucocyte count (103/mL) 51,000
guided staging system. An example is shown in Table 29.3.
Typical findings upon neurologic examination are VI cere- Neutrophils 10%70%
bral nerve palsy (present in up to 40% of patients) but also Lymphocytes 30%90%
nerves III and VII are often involved (5% to 20%). Hemiparesis Protein (g/L) 0.453.0
and paraparesis may be present upon presentation (in 5% to
Lactate (mmol/L) 5.010.0
20% of patients) (75,88) but may also develop during treat-
ment secondary to infarction. Seizures are rarely a presenting Glucose CSF/Blood 0.5 95%
symptom in adults, however in children are reported in around
50% of patients (28,29).Visual disturbances, decreased vision, Thwaites G, Fisher M, Hemingway C, et al. British Infection Society
and diplopia may have a variety of causes, including primary guidelines for the diagnosis and treatment of tuberculosis of the central
nervous system in adults and children. J Infect. 2009;59(3):167187.
involvement of optic nerve by tuberculous lesion leading to
optic neuritis, optochiasmatic arachnoiditis (OCA), and
tuberculoma in the chiasmatic region or in the optic pathways
(91). More often, visual disturbances are secondary to raised
intracranial pressure or ethambutol toxicity. A rare cause of TA B L E 2 9 . 3
visual loss in TBM is neuroretinitis (92). Tuberculoma can
cause a wide array of neurologic symptoms associated with MEDICAL RESEARCH COUNCIL GRADING SYSTEM
space-occupying lesions depending on their location within the FOR TUBERCULOUS MENINGITIS
CNS. Urinary retention is common which may indicate spinal
cord involvement. Movement disorders are associated with TBM Grade Diagnostic Criteria
basal ganglia involvement, mostly tremor, but also chorea,
Grade I Glasgow coma score 15, no focal neurology
ballismus, and myoclonus have been reported (88).
The initial symptoms of TBM may be nonspecific, but Grade II Glasgow coma score 1114 or
within the context of prolonged symptoms, a previous history Glasgow coma score 15 with focal neurology
of TB, or a chest x-ray consistent with recent or past TB Grade III Glasgow coma score 10
infection, this history must raise heightened suspicion with the
treating physician. In many textbooks, TBM is described as
From Heemskerk D, Day J, Chau TT, et al. Intensified treatment
chronic or subacute meningitis; however, this terminology is with high dose rifampicin and levofloxacin compared to standard
unhelpful. Once a patient with TBM seeks medical care, he treatment for adult patients with tuberculous meningitis (TBM-IT):
or she should be treated as a medical emergency as with any protocol for a randomized controlled trial. Trials. 2011;12:25.
other meningitis.

Scheld_Ch29.indd 481 2/21/14 8:21 PM


482 Part III: Bacterial and Mycoplasmal Infections

Children may have a more protracted and nonspecific pathogen, paradoxical response can be diagnosed clinically;
presentation. In TB-endemic settings, it is often the most com- however, incorrect diagnosis, drug resistance, and cerebral
mon cause of childhood bacterial forms of meningitis (22). infarction may be alternative causes of deterioration despite
Prodromal symptoms include fever, headache, anorexia, and treatment. Early recognition of these alternative causes is
vomiting in older children, whereas failure to thrive, poor important as they warrant urgent intervention.
appetite, vomiting, and sleep disturbances are more com- In TBM, vascular events are most often ischemic in nature.
mon in younger children or infants. Cough and weakness Vascular involvement is more frequently seen in chronic men-
may also be reported (28,29).As these symptoms are non- ingitides than in treated acute bacterial forms of meningitis
specific, children tend to present to the hospital only when (96). Other infective causes of stroke in tropical regions may
the clinical situation has deteriorated and they are already in include malaria, syphilis, Chagas disease, cysticercosis. Events
the later stages of disease (29). Similar to adults, when ini- can go unnoticed, as they occur silently or in severely ill pa-
tial nonspecific symptoms are associated with a history of tients already in deep coma. The most common signs of TBM-
recent contact with a case of documented TB, TBM should associated stroke are mono- or hemiplegia but also may present
be suspected. A prolonged history of more than 5 days, focal as lowered consciousness, disorders of movement, seizures,
neurologic deficit, and abnormal movements have been found cranial nerve palsies, papilledema, and decerebration (84).
to be independently predictive of TBM (27). Examination Clearly, neurologic deterioration also can have its origin in a tu-
may reveal nonspecific signs of meningism, failure to thrive, berculoma, cerebral edema, or infiltrating exudate (97). Unlike
and in younger children, a bulging fontanel or increased head hypertensive or atherothrombotic stroke, transient ischemic at-
circumference. Funduscopy may reveal signs of papilledema tacks (TIAs) and lacunar lesions are rare in TBM (84). Patients
or retinal involvement. In a retrospective study of 554 South may present with symptoms of stroke but more often develop
African children, 97% presented in stage II or III, with a stroke later in the course of disease, typically during the first
median duration of symptoms of 9 days. Meningeal irritation weeks of treatment (73,84). It is not currently possible to pre-
was the most frequent finding (98%). Convulsions (47%) and dict which patients will develop stroke and it is associated with
mono-/para-/quadri-/hemiplegia (63%) were also frequently higher mortality and morbidity. Initial imaging studies may not
observed. Cranial nerve palsies were less common (27%) than be sensitive enough to detect the early changes of an ischemic
in adults, as was raised intracranial pressure (23%) (29). event. Antithrombotic therapies such as aspirin and dipyridam-
The emergence of HIV has changed the epidemiology of TB ole which prevent or reduce the incidence of stroke may im-
and particularly the clinical outcome of disease. HIV-infected prove outcomes in TBM and warrant further clinical study.
patients are more likely to develop extrapulmonary forms of
the disease. TBM is considered an AIDS-defining condition.
Research studies do not suggest that HIV greatly alters the DIAGNOSIS
clinical presentation of TBM, especially in patients with higher
CD4 counts; the clinical presentation may mirror that seen
in HIV-negative individuals. Patients with lower CD4 counts Clinical Case Definition
may have a more atypical course of disease, with less specific
Early recognition of TBM is pivotal, because prompt initiation
and more subtle signs and wider differential diagnosis, render-
of treatment greatly increases chances of survival and reduces
ing diagnosis more challenging (36). Therefore, HIV-infected
disability. However, early symptoms are nonspecific and
patients may present later during the course of disease, with
diagnostic confirmation has hardly improved since the early
altered consciousness, and consequently more often in the
twentieth century. Ziehl-Neelsen smear for acid-fast bacilli is
advanced stages of disease (35). HIV patients are more likely
central to diagnosis because it gives rapid results; however, the
to have impaired cognition, generalized lymphadenopathy,
reported sensitivity is low. Sensitivity estimates depend on the
and hepatosplenomegaly (32). A retrospective study compar-
criteria used for gold standard and range widely from 10%
ing clinical presentation and outcome in children with and
to 60%. This wide variation is likely to depend on many fac-
without HIV infection surprisingly found that HIV-uninfected
tors including laboratory performance, workload, technician
children were more likely to present with a decreased level
diligence and experience, time from taking the sample to stain-
of consciousness; this may be related to the poor immune
ing in the laboratory, and volume of CSF examined. Liquid
response in immunocompromised children. Similar to adults,
culture of M. tuberculosis is considered the gold standard for
HIV-infected children had a longer history of being unwell,
diagnosis, but due to the slow growing nature of mycobacteria,
poorer nutritional state, more commonly had accompanying
the time to a positive result may range from 2 to 8 weeks. This
hepatosplenomegaly, lymphadenopathy, and otorrhea (93).
renders the test ineffective for clinical decision making regard-
Despite the similarity in presentation, outcome is significantly
ing treatment initiation, although a positive result can confirm
worse for HIV-infected adults and children with TBM.
the decision to continue therapy (although a negative result
should not automatically lead to stopping) and provides an
Progression During Treatment isolate for drug susceptibility evaluations. A suggestive history
Paradoxical responses during antituberculous treatment are must raise clinical suspicion. A clinical diagnostic algorithm
frequently reported despite appropriate chemotherapy with based on prospective data and validated against a second
susceptible bacilli. This can be encountered in all tissues but data set is available based on clinical and laboratory features
most often in the lungs, lymph nodes, and the brain (94). In the (98,99). Different clinical algorithms are published through-
brain, tuberculoma may develop or enlarge during treatment out the literature; those of most use are based on simple
for pulmonary TB, TBM, or miliary TB. Either discovered on clinical and laboratory criteria and can be used in resource-
routine brain imaging or accompanied by worsening of symp- limited settings, where disease burden is highest. Table 29.4
toms, signs of a space-occupying lesion, or convulsions. This shows an example.
generally occurs within 1 to 4 months of starting treatment, In order to address the heterogeneity in clinical diagno-
often after initial improvement. Antituberculous therapy sis among published research studies of TBM, a consensus
should be continued; the addition of systemic corticoste- score-based case definition, based on expert opinion, has been
roids may be considered (95). Within the context of a sound published for use in the research context, with an alternative
diagnosis and microbiologic confirmation of a susceptible scoring if imaging is not available. This case definition is not

Scheld_Ch29.indd 482 2/21/14 8:21 PM


Chapter 29: Tuberculous Meningitis 483

TA B L E 2 9 . 4 In children, CSF modifications are similar to adults; however,


smear and culture are less sensitive.
DIAGNOSTIC CRITERIA OF TUBERCULOUS In HIV-positive patients, atypical findings are encoun-
MENINGITIS USED IN CLINICAL TRIALS tered in a considerable proportion of patients, which may
lead to delay in diagnosis and treatment. Normal levels of
Classification Diagnostic Criteria lactate, glucose, protein, and WCC are more often reported
in HIV; even completely normal CSF findings can be found
Definite TBM Clinical meningitis plus acid-fast bacilli seen particularly in patients with severe immunosuppression (CD4
in the CSF or count 50 cells/L). In the context of HIV, neutrophils often
Mycobacterium tuberculosis cultured from predominate in the CSF cell population (37).
the CSF Essential to the search for acid-fast bacilli is the volume of
Probable TBM Clinical meningitis plus one of the following the CSF sample, the time spent on microscopic examination,
criteria: and the efficiency with which the microbiology, biochemistry,
Radiographic evidence of pulmonary and hematology laboratories use the precious CSF sample.
tuberculosis Increasing the volume (to a minimum of 6 mL) and slide
Acid-fast bacilli seen in sputum or examination time to a standard time (preferably 30 minutes)
gastric fluid can improve the yield to more than 60% of clinically diag-
Evidence of extrapulmonary tuberculosis nosed cases (101). Particularly for pediatric patients, it can be
CT or MRI brain scan features difficult to obtain large volumes of CSF, but drawing of larger
consistent with TBM volumes should be encouraged to improve the confirmation
Possible TBM Clinical meningitis plus 2 of the following rate, where not contraindicated. It should be remembered that
criteria: the major safety issue relates to the decision on whether to
History of previous tuberculosis perform a lumbar puncture; the volume of CSF then taken is
Illness duration 5 days of secondary consequence. Hence having made the decision
Glasgow coma score 15 to perform a CSF, it is only ethical to then take a volume of
Focal neurologic signs CSF that will give a good chance of improving the care of the
patient. The CSF should be concentrated prior to examina-
and 2 of the following criteria: tion of the deposit, either by centrifugation or filtration (102).
Yellow CSF Direct smear examination of CSF is rarely positive.
50% lymphocytes in the CSF
CSF glucose 50% blood glucose

From Heemskerk D, Day J, Chau TT, et al. Intensified treatment


Chest X-Ray
with high dose rifampicin and levofloxacin compared to standard
treatment for adult patients with tuberculous meningitis (TBM-IT): The chest x-ray may reveal active or previous infection with
protocol for a randomized controlled trial. Trials. 2011;12:25. M. tuberculosis. In children, signs of primary infection may
be noted on chest x-ray. In adults, the chest x-ray is often
normal, but all typical lesions can be found including apical
scarring, calcified Ghon complex, upper lobe infiltration, and
intended to determine treatment decisions and may be less nodular and cavitating disease. Miliary TB is frequently asso-
practical in the clinical setting. In particular, it should not be ciated with TBM (see Fig. 29.2) found on chest x-ray in 25%
used to exclude a diagnosis of TBM (100) (Table 29.5). to 50% of adults and 15% to 25% of children with TBM
Without the context of a suggestive clinical history, TBM (103,104).
can mimic other chronic meningoencephalitides. Notably, in
HIV-infected patients, the chief obstacle in clinical practice
is the distinction from other chronic forms of meningitis, in Mycobacterial Culture
particularly cryptococcal meningitis, cerebral toxoplasmosis,
cytomegalovirus encephalitis, and CNS lymphoma. Because As for CSF smear, sensitivity of culture of M. tuberculosis
diagnostic yield from smear is generally low, careful exclu- from the CSF is increased by using a larger volume of CSF,
sion of other diagnoses is imperative. As soon as suspicion which should be concentrated prior to inoculation of the
is raised, history taking and additional investigations should deposit wherever possible. Previously, Lowenstein-Jensen
be directed toward the exposure to TB and ruling out other (LJ) media and later agar media (Middlebrook 7H10, 7H11)
treatable causes. were recommended; however, liquid culture techniques show
increased sensitivity and more rapid turnaround times for the
isolation of mycobacteria and should be used where possible.
Cerebrospinal Fluid Analysis Commercial liquid culture systems include BACTEC MGIT
960 system (Becton Dickinson Microbiology Systems, Sparks,
Fundamental to diagnosis is the lumbar puncture and conse- MD) and MB/BacT system (BioMrieux, Durham, NC) and
quent CSF analysis. CSF pressure is raised (20 cm H2O) in have reduced the time to results for both isolation and DST of
approximately 50% of adults and 40% to 75% of children mycobacteria (105). CSF cultures generally become positive
(10 cm H2O). Typically, the CSF is straw colored. The between 10 and 21 days on commercial liquid culture systems,
results can be equivocal. The white cell count (WCC) is generally although late positives may occur after 35 days due to the low
lower (10 to 1,000  106 cells/mL) than in bacterial meningitis, bacillary load (106,107).
predominantly lymphocytic, with a low serum to CSF glucose The microscopically observed drug susceptibility
ratio (50%). Lactate is usually raised, reflecting intracerebral (MODS) assay is a noncommercial liquid culture technique
metabolic disturbances or ischemic processes. Raised total protein with minimal technical requirements that can detect both
levels (0.5 g/L) are an indication of bloodbrain barrier disrup- mycobacteria and drug resistance (108). In TBM, MODS
tion or increased intracerebral production of immunoglobulins. culture has shown comparable sensitivity to mycobacteria

Scheld_Ch29.indd 483 2/21/14 8:21 PM


484 Part III: Bacterial and Mycoplasmal Infections

TA B L E 2 9 . 5
UNIFORM CASE DEFINITION

CLINICAL CRITERIA DIAGNOSTIC SCORE

Maximum category score  6

Symptom duration of more than 5 days 4


Systemic symptoms suggestive of tuberculosis (one or more of the following): 2
weight loss (or poor weight gain in children), night sweats, or persistent cough
for more than 2 weeks
History of recent (within past year) close contact with an individual with 2
pulmonary tuberculosis or a positive TST or IGRA (only in children younger
than 10 years of age)
Focal neurologic deficit (excluding cranial nerve palsies) 1
Cranial nerve palsy 1
Altered consciousness 1

CSF CRITERIA Maximum category score  4

Clear appearance 1
Cells: 10500/L 1
Lymphocytic predominance (50%) 1
Protein concentration greater than 1 g/L 1
CSF-to-plasma glucose ratio of less than 50% or an absolute CSF glucose 1
concentration less than 2  2 mmol/L

CEREBRAL IMAGING CRITERIA Maximum category score  6

Hydrocephalus 1
Basal meningeal enhancement 2
Tuberculoma 2
Infarct 1
Precontrast basal hyperdensity 2

EVIDENCE OF TUBERCULOSIS ELSEWHERE Maximum category score  4

Chest radiograph suggestive of active tuberculosis: signs of tuberculosis  2; 2/4


miliary tuberculosis  4
CT/ MRI/ultrasound evidence for tuberculosis outside the CNS 2
AFB identified or Mycobacterium tuberculosis cultured from another sourcethat 4
is, sputum, lymph node, gastric washing, urine, blood culture
Positive commercial M. tuberculosis NAAT from extraneural specimen 4
Exclusion of alternative diagnoses
An alternative diagnosis must be confirmed microbiologically (by stain, TST, tuberculin skin test. IGRA, interferon-
culture, or NAAT when appropriate), serologically (e.g., syphilis), or release assay; NAAT, nucleic acid amplification
histopathologically (e.g., lymphoma). The list of alternative diagnoses that test. AFB, acid-fast bacilli. The individual
should be considered, dependent on age, immune status, and geographical points for each criterion (1, 2, or 4 points)
region, include pyogenic bacterial meningitis, cryptococcal meningitis, were determined by consensus and by
syphilitic meningitis, viral meningoencephalitis, cerebral malaria, parasitic considering their quantified diagnostic value
or eosinophilic meningitis (Angiostrongylus cantonensis, Gnathostoma as defined in studies.
spinigerum, toxocariasis, cysticercosis), cerebral toxoplasmosis and bacterial
brain abscess (space-occupying lesion on cerebral imaging), and malignancy
(e.g., lymphoma)

(continued)

Scheld_Ch29.indd 484 2/21/14 8:21 PM


Chapter 29: Tuberculous Meningitis 485

TA B L E 2 9 . 5
UNIFORM CASE DEFINITION (CONTINUED)

Clinical entry criteria


Symptoms and signs of meningitis include one or more of the following: headache, irritability, vomiting, fever, neck stiffness,
convulsions, focal neurologic deficits, altered consciousness, or lethargy.
Tuberculous meningitis classification
Definite tuberculous meningitis
Patients should fulfill criterion A or B:
A. Clinical entry criteria plus one or more of the following: acid-fast bacilli seen in the CSF, Mycobacterium tuberculosis
cultured from the CSF, or a CSF-positive commercial nucleic acid amplification test.
B. Acid-fast bacilli seen in the context of histologic changes consistent with tuberculosis in the brain or spinal cord with
suggestive symptoms or signs and CSF changes or visible meningitis (on autopsy).
Probable tuberculous meningitis
Clinical entry criteria plus a total diagnostic score of 10 or more points (when cerebral imaging is not available) or 12 or more
points (when cerebral imaging is available) plus exclusion of alternative diagnoses. At least 2 points should either come from
CSF or cerebral imaging criteria.
Possible tuberculous meningitis
Clinical entry criteria plus a total diagnostic score of 69 points (when cerebral imaging is not available) or 611 points
(when cerebral imaging is available) plus exclusion of alternative diagnoses. Possible tuberculosis cannot be diagnosed or
excluded without doing a lumbar puncture or cerebral imaging.
Nontuberculous meningitis
Alternative diagnosis established, without a definitive diagnosis of tuberculous meningitis or other convincing signs of dual
disease

From Marais S, Thwaites G, Schoeman JF, et al. Tuberculous meningitis: a uniform case definition for use in clinical research. Lancet Infect Dis.
2010;10(11):803812.

growth indicator tube (MGIT) for CSF culture, however with drug to a failing regimen should be applied. Experience from
a median turnaround time of 6 days versus 15.5 days (106). pulmonary TB and pharmacokinetic data support the use of a
This study did not evaluate direct DST using MODS or MGIT. fluoroquinolone (levofloxacin, gatifloxacin, or moxifloxacin)
MODS is increasingly used in low-resource countries for the (113,114). Ciprofloxacin should not be used because it is less
diagnosis of pulmonary MDR TB; however, it is still not active against M. tuberculosis; resistance develops rapidly and
widely used for TBM, although that should change. WHO has may cause selection of strains resistant to more active fluoro-
endorsed both commercial and noncommercial liquid culture quinolones (113).
systems for TB diagnosis (109).
MDR TB is defined as M. tuberculosis with resistance
to at least rifampicin and isoniazid. XDR TB is defined as Tuberculin Skin Testing
M. tuberculosis with resistance to at least isoniazid and rifam-
picin, any fluoroquinolone, and at least one of three injectable The Mantoux tuberculin skin test (TST) is performed by
second-line drugs (amikacin, capreomycin, or kanamycin). intracutaneously injecting a small dose of purified protein
Whereas MDR pulmonary TB can be effectively treated with derivative (PPD). After 48 to 72 hours, an induration of 15 mm
second-line antituberculous drugs, evidence on effective treat- or more is considered positive in all persons (115). The value
ment of MDR TBM is limited to small series or single case of this diagnostic test is highly dependent on the background
reports and mortality is extremely high (39). The majority of prevalence of TB, age of the patient, and the co-infection with
deaths from TBM occur within the first month of treatment HIV. Up to 10% to 15% of immunocompetent children with
(30,31,110). If the CSF is culture positive for mycobacteria, culture-documented TB do not initially show TST reactivity.
molecular diagnostic tests can be used to establish resistance to Sensitivity can be decreased by host factors, such as young
rifampicin or isoniazid (111,112). If only conventional pheno- age, poor nutrition, immunosuppression, other viral infections
typic DST is available, the results of the drug susceptibility test (such as measles, varicella, and influenza), recent TB infec-
will be returned to the treating physician at the earliest after tion, and disseminated TB diseases (116). In culture-proven
4 to 6 weeks but more often after at least 8 weeks of treat- TBM patients in Egypt, only 19% of patients had tubercu-
ment. The majority of TBM patients infected with rifampicin- lin positivity on admission. The yield improved when the test
resistant strains will have succumbed before the decision can was repeated after 60 days, with 62% positivity (117). Severe
be made to change to a second-line regimen; for this reason, immunosuppression will also suppress skin test reactivity
molecular tests for rifampicin resistance should be applied in (118). False-positive TST results may also occur in BCG-
all cases of TBM, but especially where there is a high suspicion vaccinated individuals and those exposed to environmental
of MDR TBM (previous treatment history, known exposure nontuberculous mycobacteria (116). In the United Kingdom,
to an MDR or chronic TB case, HIV patient, or failure to Heaf testing was preferred to Mantoux. It follows the same
respond to first-line therapy). Currently, there is no evidence principles as the Mantoux test, however a special Heaf gun
base to support treatment regimens for patients with MDR with multiple small needles is used, which is thought to be less
TBM, but the principle of TB treatment to never add a single painful in children and has an easier readout (119).

Scheld_Ch29.indd 485 2/21/14 8:21 PM


486 Part III: Bacterial and Mycoplasmal Infections

In-Tube test (QFT-GIT) uses enzyme-linked immunosorbent assay


Developments in Diagnostics (ELISA) technique to measure cell-mediated immunity in response
to proteins specific for M. tuberculosis complex. When assessed
Biochemical tests for TBM in India, the whole blood IGRA had sensitivity of 44.4%
Biochemical tests can detect features of the infecting organism or and specificity of 62.5% for diagnosing TBM with positive pre-
products of the host immune response. Of the biochemical tests, dictive value (PPV) of 72.7%. IGRA on CSF had 88% indetermi-
adenosine deaminase (ADA) assays are still of interest world- nate results in TBM patients (131).
wide, particularly in low-income settings. The test is relatively
cheap and easy to perform. It is an attractive candidate for the Nucleic Acid Amplification Tests
diagnosis of TBM because it has been shown to be of value in Amplification of mycobacterial DNA by polymerase chain
the distinction of tuberculous pleural effusions (120). ADA is re- reaction (PCR) has turned away attention from immunologic
leased by T cells during cell-mediated immune response to bacilli techniques. Systematic review on commercial nucleic acid
(121). A recent metaanalysis reviewed 10 publications on ADA amplification tests (NAATs) for TBM summarized the perfor-
for the diagnosis of TBM and concluded that the mean values of mance as having potential to rule in or confirm diagnosis (speci-
the sensitivity and specificity of the ADA assays were respectively ficity 98%), but low sensitivity (56%) precludes the use of these
79% and 91% compared to culture as the gold standard, with tests to rule out (132). NAAT should be used as an adjunct to
an inability particularly to distinguish TBM from bacterial men- conventional microscopy, culture, and clinical algorithms.
ingitis (122). However, publication bias and spectrum bias in The most recent advance in diagnosis is the GeneXpert MTB/
included cases may have resulted in overestimation of diagnostic RIF (Cepheid, Sunnyvale, CA), an easy-to-use desktop machine
accuracy. Inclusion of inappropriate controls will result in an that simultaneously detects the presence of M. tuberculosis and
overestimation of diagnostic accuracy, particularly if the test is rifampicin resistance using real-time PCR, giving results within
poor at discriminating the common differential diagnoses; raised 2 hours. Risk of contamination and consequent false-positive
levels may also be seen in sarcoidosis, meningeal lymphoma, results is reduced by the use of sealed disposable cartridges.
subarachnoid hemorrhage, and neurobrucellosis. Although The rapid turnover time may lead to an important improve-
ADA may be of use within diagnostic algorithms, the probability ment in the management of MDR TB. The test has been en-
that a patient has TBM with a negative result is still too high to dorsed by the WHO in 2010 for the use in pulmonary TB (112).
rule out the diagnosis using ADA values. Furthermore, in HIV- GeneXpert is more sensitive than smear, compared to culture.
infected individuals, the test is not of use (123). In smear-negative, culture-positive sputum samples, detection
of M. tuberculosis showed 76.9% sensitivity and 99.0% speci-
ficity in a decentralized low-income setting. Sensitivity for
Immunoassays rifampicin resistance was 94.4% and specificity 98.3%, regard-
Various immunoassays have been evaluated for diagnosis of TBM, less of HIV infection (133). These results are clearly promis-
with highly variable sensitivity and specificity between studies, ing and roll out of the GeneXpert MTB/RIF test has been
again commonly due to spectrum bias among controls. WHO impressive globally (http://who.int/tb/laboratory/mtbrifrollout/
issued the first ever negative policy recommendation against en/). The GeneXpert test has been shown to be more sensitive
using existing commercial serologic assays for the diagnosis of than smear on extrapulmonary samples in reports combining
pulmonary TB in 2011, following a systematic evaluation (124). analysis of all extrapulmonary samples with a small number
Immunoassays may not distinguish between acute infection and of CSF samples (134137). In June 2013, the WHO Strategic
previous exposure, and cross reactivity of antibodies may further and Technical Advisory Group (STAG) reviewed the evidence
decrease specificity. The detection of lipoarabinomannan (LAM) for use of GeneXpert on extrapulmonary samples. 15 pub-
has been evaluated for the diagnosis of TB and current interest is lished and 7 unpublished studies, involving 5,922 samples,
heightened since the development of a point of care lateral flow were included in the review which endorsed GeneXpert for use
urine assay, which has shown value in the diagnosis of pulmonary on extrapulmonary samples, including CSF. The report recom-
TB in HIV patients with severe immunosuppression (CD4 100 mended Xpert MTB/RIF should be used in preference to con-
cells/L) (125,126). However, this is a relatively distinct subset ventional microscopy and culture as the initial diagnostic test
of patients and a CD4 assay is first required to determine if the in testing cerebrospinal fluid specimens from patients presumed
LAM test is applicable. LAM is a cell wall component of M. tu- to have TB meningitis (strong recommendation given the ur-
berculosis, has immunoregulatory and antiinflammatory effects, gency of rapid diagnosis, very low quality of evidence). Pooled
and serves as a virulence factor of the mycobacteria (127). Earlier sensitivity and specificity against a clinical gold standard were
studies on ELISA directed toward detection of IgG antibodies to estimated to be 55.6% and 98.8%, respectively (138).
LAM antigens have reported promising sensitivity and specificity Data from 265 consecutive patients with suspected TBM
in small studies, but highly variable results are published for dif- presenting at Hospital for Tropical Diseases, Ho Chi Minh City,
ferent clinical populations with TB, reflecting the spectrum bias Vietnam showed GeneXpert detected M. tuberculosis in 56.7%
in evaluation of serologic tests for TB. A standardized LAM an- (n  97/171 [95% CI, 48.9; 64.2%]) of pretreatment CSF sam-
tigen detection ELISA test, showing results within 2 to 3 hours ples of patients clinically diagnosed with TBM (specificity was
(Clearview TB ELISA, Inverness Medical Innovations, Waltham, 100%, n  94/94). Detection was only marginally lower than
MA), has been assessed on CSF of 150 patients in an HIV high- MGIT culture at 60.8% (n  104/171) and similar to detection
prevalence setting. With a disappointingly low sensitivity of 31%, rates in CSF reported for other commercial nucleic acid ampli-
but specificity of 94%, it has the potential to be a rapid rule-in fication tests, with the advantage that rifampicin resistance is
test for TBM for HIV-infected patients with advanced immuno- also detected by GeneXpert (139). Three cases of rifampicin re-
suppression when used in combination with a clinical prediction sistance were detected and confirmed by MGIT DST. Although
rule (128), but optimized CSF smear is likely to have a higher di- the numbers of MDR cases are too small to draw robust conclu-
agnostic yield. Other immunologic tests that have been evaluated sions regarding accuracy in CSF, rifampicin resistance detection
are interferon- release assays (IGRA), including peripheral blood by GeneXpert MTB/RIF has been shown to be accurate in spu-
IgG ELISPOT response to PPD, CFP-10, and ESAT-6; however, all tum samples. For MDR TBM, this test will be of great impor-
showed poor sensitivity in patients with TBM (129,130). One of tance, as detection of drug resistance within hours rather than
the commercially available IGRA tests, QuantiFERON-TB Gold days will allow research into optimal treatment of MDR TBM.

Scheld_Ch29.indd 486 2/21/14 8:21 PM


Chapter 29: Tuberculous Meningitis 487

Further multicenter evaluation of this novel GeneXpert assay


may lead to significant improvement in the diagnosis and treat-
ment of TBM in general and MDR TBM in particular.

Central Nervous System Imaging


Radiologic results can add additional evidence for diagnosis while
also potentially serving as the missing link between epidemiol-
ogy, etiology, and pathophysiology, especially given the paucity of
postmortem studies. Presenting findings are pluriform and during
treatment, progressive changes may appear. Contrast-enhanced
MRI is the modality of choice because it has a higher resolution
over computed tomography (CT) scanning. However, in many
endemic settings, MRI is too expensive or not readily available.
The typical initial findings are hydrocephalus, basal menin-
geal enhancement, and tuberculoma. Hydrocephalus (Fig. 29.3)
is the most common finding. Hydrocephalus is also seen in bacte-
rial meningitis, although less frequent (140). Generally, in TBM,
hydrocephalus is of the communicating type (141). Occasionally,
obstructive hydrocephalus can be diagnosed by imaging, when
narrowing of the aqueduct of Sylvius is noted or when a
parenchymal mass is demonstrated obstructing the flow of CSF.
If neurosurgical interventions (shunting, endoscopic third ven-
triculostomy) are available and contemplated, air encephalogra-
phy can be helpful in excluding patients with communicating FIGURE 29.4 Basal meningeal enhancement in an adult with TBM.
hydrocephalus from undergoing unnecessary procedures (142). Gadolinium-enhanced T1-weighted magnetic resonance image show-
Basal meningeal enhancement (Fig. 29.4) in the advanced stage ing TBM-associated basal meningeal enhancement extending toward
the sylvian fissures.
can, although rarely, be demonstrated on noncontrast CT scans,
when obliteration of the interpeduncular cisterns is observed. After
contrast administration, there is typically diffuse enhancement of
have been reported to occur in 5% to 40% of patients presenting
the basal subarachnoid cisterns; occasionally, meningeal enhance-
with TBM (74,144). The typical features on MRI imaging de-
ment is seen over the cerebral convexities, the sylvian fissures, and
pend on whether the granuloma is noncaseating, caseating with
the tentorium (143). In the early stages, CT or MRI imaging with-
a solid center, or caseating with a liquid center. The noncaseat-
out the use of contrast may show little or no abnormalities.
ing tuberculoma is usually hypointense on T1-weighted images
Tuberculomas (Fig. 29.5) are characteristic for TBM and can
and hyperintense on T2-weighted images and shows homoge-
involve the parenchyma of the brain, spinal cord, subarachnoid,
neous enhancement after contrast administration. The caseating
and subdural or epidural space; may be multiple or solitary; and
tuberculoma with solid caseation looks hypointense or isoin-
tense on T1-weighted images and isointense to hypointense on
T2-weighted images, with rim enhancement on contrast admin-
istration. Caseating granuloma with a liquid center looks hypoin-
tense on T1-weighted images and hyperintense on T2-weighted
images, with rim enhancement after contrast administration (74).
On imaging, a (tubercular) abscess may be difficult to distinguish
from tubercular granulomas with a liquid center. Tuberculous
abscesses are generally more rapidly progressive, larger, have a
thinner wall, and can be loculated and irregular in shape.
During treatment, tuberculoma may develop paradoxically;
however, in a prospective serial MRI study, this phenomenon was
observed in 60% of patients, suggesting that it is part of the natural
course of disease on treatment rather than being discordant (73).
Ischemic events most commonly occur during treatment rather
than being a presenting sign, mostly located in the basal ganglia
(Fig. 29.6), which is in line with the severe basal infection, exu-
dates formation, and consequent vasculitis. In a serial MRI study,
dexamethasone reduced the proportion of patients who developed
infarcts during treatment; however, the difference was not statisti-
cally significant, possibly due to insufficient sample size (73).
Using contrast-enhanced MRI in children in Turkey, pre-
senting findings were meningeal enhancement (90.9%), hydro-
cephalus (63.6%), infarction (45.5%), tuberculomas (27.2%),
cranial nerve involvement (27.2%), and severe cortical atro-
phy (9.1%) (145). Retrospectively, miliary involvement of
the leptomeninges was present in a very large (88%) propor-
FIGURE 29.3 Hydrocephalus associated with TBM. Gadolinium- tion of young South African children scanned for TBM (146).
enhanced T1-weighted magnetic resonance image showing gross However, rare miliary CNS involvement is also reported in some
hydrocephalus associated with TBM. adult patients (Fig. 29.7). This discrepancy between adults and

Scheld_Ch29.indd 487 2/21/14 8:21 PM


488 Part III: Bacterial and Mycoplasmal Infections

FIGURE 29.5 Tuberculoma in an adult with TBM. Gadolinium-enhanced T1-weighted MRI showing
multiple tuberculoma in the cerebral peduncle extending to the hypothalamus.

young children may be an indication that direct hematogenous in HIV patients (36). Infarcts, tuberculous abscess, tubercu-
spread to the meninges plays a larger role in the young, pos- lous encephalitis, and optochiasmatic arachnoiditis on imag-
sibly due to immature innate and adaptive immunity. ing have all been associated with poor outcome.
The same diagnostic imaging criteria apply to children with With the advent of newer more sensitive imaging tech-
HIV infection, however cortical atrophy is a more common niques, our ability to detect abnormalities will improve.
finding (86) (Fig. 29.8). In adults with AIDS, radiologic find- Magnetic resonance angiography (MRA) may have a role in
ings are reported to be similar to patients without HIV; how- predicting the chance of infarction in TBM (148). 3-Tesla
ever, the differential diagnosis will include other opportunistic magnetic resonance neurography imaging may carry improved
infections and primary or metastatic lymphoma of the CNS sensitivity and ability to detect cranial nerve involvement, for
(32,37,147). Some have observed that hydrocephalus and example. Still, the obvious obstacles in low-resource settings,
meningeal enhancement are a less common radiologic finding where rapid diagnosis is needed the most, remain.

FIGURE 29.6 Basal ganglia infarct in an adult with TBM. CT image FIGURE 29.7 Miliary lesions in an adult with TBM. Gadolinium-
without contrast showing infarction of the left capsula interna and enhanced T1-weighted MRI showing multiple miliary lesions and
caudate head. small tuberculoma in an adult patient with TBM.

Scheld_Ch29.indd 488 2/21/14 8:21 PM


Chapter 29: Tuberculous Meningitis 489

A B C

D E F

G H I

FIGURE 29.8 Common MRI findings in children with TBM. A: Contrast-enhanced axial T1-weighted
image reveals thick leptomeningeal enhancement in suprasellar cistern extending into interpeduncular
cistern. B: Contrast-enhanced coronal T1-weighted image reveals leptomeningeal enhancement with
marked basal leptomeningeal enhancement. C: Axial T1-weighted image reveals several enhancing
nodules in parenchyma. D: Axial T2-weighted image reveals infarction of right caudate head and puta-
men. E: Gadolinium-enhanced T1 axial imaging showing extensive basal leptomeningeal enhancement
in suprasellar, interpeduncular, and ambient cisterns. F: Unenhanced sagittal T1-weighted image reveals
massive hydrocephalus. G: Contrast-enhanced axial T1-weighted image reveals thick leptomeningeal
enhancement in suprasellar cistern extending into interpeduncular and ambient cisterns and small tu-
berculoma in left brainstem. H: Contrast-enhanced axial T1-weighted image reveals basal meningeal
enhancement and both dilated temporal horns. I: Contrast-enhanced coronal T1-weighted image reveals
thick leptomeningeal enhancement of interhemispheric fissure. (continued)

Scheld_Ch29.indd 489 2/21/14 8:21 PM


490 Part III: Bacterial and Mycoplasmal Infections

J K L

FIGURE 29.8 (continued) J: Contrast-enhanced axial T1-weighted image shows tuberculoma in the left
temporal lobe. K: Contrast-enhanced axial T1-weighted image shows small tuberculomata in brainstem
and basal meningeal enhancement of both sylvian fissures. L: Contrast-enhanced axial T1-weighted
image shows small tuberculoma in left brain peduncle and temporal lobe, both dilated temporal horns
and meningeal enhancement of left sylvian fissure. (Courtesy of Dr. Nguyen Duc Bang, Pham Ngoc Thach
Hospital for Tuberculosis and Lung Diseases, Ho Chi Minh City, Vietnam.)

similar; however, for children, often higher dosages are used


TREATMENT (Table 29.6).
In this section, we would like to put emphasis on the
The treatment of TBM may be divided into four comple- statement that the CNS should be regarded as a unique
mentary areas: specific antituberculous therapy, adjunctive therapeutic compartment (152) and pharmacokinetic and
immunomodulatory therapy, anticoagulant therapy, and man- pharmacodynamic data should be considered in the construc-
agement of intracranial pressure. In addition, treatment of tion of more effective treatment schedules. The majority of
HIV-associated TBM requires consideration of drug interac- deaths from TBM occur in the first 2 months of treatment,
tions and IRIS. indicating that effective antimycobacterial killing is most
critical in the intensive phase. However, prevention of relapse
Specific Antituberculous Treatment and the prevention of emerging resistance are additionally im-
portant principles of effective multidrug treatment.
Treatment guidelines for TBM treatment are not uniform. The ability of the different first-line antituberculous drugs
In general, global guidelines recommend 9 to 12 months to penetrate the CSF is variable and few of the second-line
treatment with rifampicin, isoniazid, pyrazinamide, and strep- drugs are effective in reaching the brain. A summary of
tomycin (or ethambutol) in the intensive phase, followed by a antimycobacterial activity and CSF penetration of the first-line
combination of rifampicin and isoniazid in the continuation drugs used in the intensive phase is appropriate. We will also
phase (75,149151). These treatment regimens are based on briefly review some of the second-line agents with favorable
the early trials in pulmonary TB involving the introduction CSF levels. Of note, little is known about the levels of antimy-
of the new first-line antituberculous drugs. The drug dos- cobacterial drugs in the brain tissue. Drugs may need to over-
ages and duration of treatment recommended for TBM are come both bloodCSF barrier, consisting of the choroid plexus
derived from pulmonary regimens and are not based on phar- and the lining epithelial cells, and the bloodbrain barrier, made
macokinetic principles. Regimens for children and adults are up of endothelial tight junctions in capillaries and surrounding

TA B L E 2 9 . 6
RECOMMENDED DAILY DOSAGE OF FIRST-LINE ANTITUBERCULOUS DRUGS IN
CHILDREN AND ADULTS WITH TUBERCULOUS MENINGITIS

Drug Daily Dose Children (12 years) Daily Dose Adult

Isoniazid (INH) 10 mg/kg (range 615 mg/kg) 5 mg/kg (range 46 mg/kg)


Rifampicin (RIF) 15 mg/kg (range 1020 mg/kg) 10 mg/kg (range 812 mg/kg)
Pyrazinamide (PZA) 35 mg/kg (range 3040 mg/kg) 25 mg/kg (range 2030 mg/kg)
Streptomycin (SM) 17.5 mg/kg (range 1520 mg/kg) 15 mg/kg (range 1218 mg/kg)
Ethambutol (EMB) 20 mg/kg (range 1525 mg/kg) 15 mg/kg (range 1520 mg/kg)

From Donald PR. The chemotherapy of tuberculous meningitis in children and adults. Tuberculosis
(Edinb). 2010;90(6):375392.

Scheld_Ch29.indd 490 2/21/14 8:21 PM


Chapter 29: Tuberculous Meningitis 491

glial cells. The level of drugs in the two compartments, CSF in preventing resistance when used with companion drugs.
and brain tissue, may not be equal. A third hurdle may be It is less efficient in eradicating slow-growing organisms.
the penetration and action of drugs in the relatively anaerobic Some advocate the administration of higher doses because
conditions within the tuberculoma. resistance is increasingly abundant and higher intracerebral
Cmax may lead to killing of strains with low-level resistance,
Streptomycin and Ethambutol which do not carry mutations in the katG gene. Additionally,
the N-acetyltransferase-2 genotype of an individual (NAT2)
The initial drug to be introduced for TBM treatment was influences the EBA of isoniazid at a given dose, and faster
streptomycin. This aminoglycoside must be given intramus- isoniazid acetylators consistently have a lower EBA (154).
cularly. Streptomycin is a protein synthesis inhibitor. The Conversely, slow acetylators may have increased susceptibility
minimal inhibitory concentration (MIC) in fully susceptible to hepatotoxicity (159).
clinical isolates is in the range 0.5 to 2.0 g/mL (153). With
the commonly used dosages in adults, the early bactericidal
activity (EBA; generally defined as the fall in counts per mil-
Rifampicin
liliter sputum per day during the first 2 days of treatment) of Rifampicin is a key drug in the treatment for TBM, illustrated
streptomycin, however, is low (0.1 log10 colony-forming unit by the high mortality in MDR TBM patients compared to
[CFU]/mL sputum per day) (154). With a poor penetration in isolated isoniazid resistance. In contrast, in a review of the lit-
CSF, the contribution of streptomycin to multidrug regimens erature, Donald (18) found little effect on mortality in adults
for TBM is probably very limited. In the absence of meningeal after the introduction of rifampicin and pyrazinamide to the
inflammation, penetration does not occur. However, in menin- TBM treatment schedule but a significant effect on survival in
gitis, the penetration can be up to 20% of simultaneous serum children. Still, based on the drug resistance data, rifampicin
levels. In the early course of disease, when bloodbrain barrier seems to have a pivotal role in treatment; it may well be that
disruption is prominent, levels above the MIC may be found in doses in adults are not sufficient to reach adequate levels in the
CSF (3 to 16 g/mL). However, when clinical improvement is CSF. An Indonesian phase 2 clinical trial, establishing the safety
noted, CSF levels barely reach the MIC (0 to 1.25 to 4 g/mL) of high-dose intravenous (13 mg/kg/day) rifampicin with or
(155). Because TBM may reflect a general state of dissemi- without (high dose) moxifloxacin, did not show increased tox-
nated TB, it appears appropriate to maintain streptomycin in icity and moreover showed a 50% reduction in mortality for
treatment schedules, however at the cost of increased toxicity. patients receiving high-dose rifampicin (160). However prom-
For HIV patients, ethambutol should be substituted for strep- ising, this trial was not powered for a clinical outcome, and the
tomycin because injection should be avoided when possible in results of a current randomized controlled trial comparing an
HIV-infected individuals. Ethambutol is bacteriostatic against intensified 2-month regimen of high-dose (15 mg/kg/day) oral
actively growing TB bacilli by obstructing the formation of rifampicin and levofloxacin are awaited (161).
the bacterial cell wall. Ethambutol is slightly more efficient Rifampicin inhibits bacterial RNA synthesis by inhibiting
in penetrating the CSF, with levels compared to serum in the RNA polymerase. Rifampicin is highly bound to plasma pro-
range of 0% to 54%. In healthy adults, despite an oral dose of teins, which leaves only 20% of total drug freely diffusible. This
50 mg/kg which was twice the usual therapeutic dose, and in is reflected in the ability of rifampicin to penetrate the CSF, with
the presence of proportionally high blood levels, ethambutol a CSF/plasma ratio of maximally 20% found in early TBM and
did not appear in the CSF of healthy adults. After oral doses no drug detectable in CSF in the absence of meningeal inflam-
of 18.6 to 25 mg/kg levels, ethambutol did appear in the CSF mation (155). After an oral dosage of approximately 11 mg/kg,
of patients with active meningitis (0.74 to 1.98 g/mL) (155). serum Cmax averaging 11.5 g/mL were obtained at 2 hours.
Still, with an MIC of 0.5 to 2.0 g/mL, there may be a limited Rifampicin penetrated very slowly into the CSF, and concentra-
role for ethambutol in TBM treatment. Both streptomycin and tions only slightly in excess of its MIC against M. tuberculosis
ethambutol have been shown ineffective in sterilizing sputum (approximately 0.3 g/mL) maintained throughout the period
in pulmonary TB, so their role in TBM may be limited to re- (158). In general, low serum levels of rifampicin are reported,
sistance prevention. It is possible that by increasing the dose particularly in HIV-positive patients, in whom absorption of
of these drugs, therapeutic levels could be achieved in the CSF, all TB drugs may be impaired (162). It is suggested that ri-
but this would come at the cost of increased toxicity. fampicin serum concentrations 2 hours post dose between 8
and 24 g/mL are required for optimal treatment of pulmo-
Isoniazid nary TB. Serum levels below 4 g/mL are defined as very low
(163). In Indonesia, 70% of TB patients had 2-hour plasma
After the introduction of isoniazid, a major improvement concentrations (Cmax) below 4 g/mL (164). Increasing the
was seen in the outcome for patients with all grades of TBM. dose of rifampicin from 10 mg/kg to 13 mg/kg led to a dis-
Isoniazid exerts its antimycobacterial activity by inhibiting proportionate (65%) increase in plasma levels and significantly
the synthesis of mycolic acid required for the mycobacterial increased the proportion of patients with rifampicin peak
cell wall. It is the most bactericidal TB drug and kills ap- plasma concentrations above the reference value of 8 g/mL
proximately 95% of rapidly multiplying organisms in sputum (165). The EBA and bactericidal activity of rifampicin (0.2 to
samples within 48 hours (156). Isoniazid has the highest EBA 0.6 log10 CFU/mL sputum per day) may be enhanced with an
of the first-line TB drugs ranging from 0.4 to 0.8 log10 CFU/ increased dose.
mL sputum per day (154). MIC in liquid media is low: 0.02
to 0.04 g/mL (153). A Cmax of 3 to 5 g/mL is needed for
optimal action against sensitive M. tuberculosis and isoniazid-
Pyrazinamide
resistant strains with relatively low MICs (157). It has good The mechanism of action of pyrazinamide is not completely
penetration in the CSF in both children and adults. Peak levels understood. It is known for its ability to kill semidormant
are reached at approximately 6 hours after dose (155). With M. tuberculosis bacilli in low pH milieu that are not killed by
an oral dosage of about 9 mg/kg, isoniazid rapidly diffused the other TB drugs, possibly by disrupting membrane energetics
into the CSF. By 4 hours, mean CSF isoniazid concentrations and inhibiting membrane transport function in M. tuberculosis
measured were 3.2 g/mL, well over the MIC and in the range (166). In pulmonary TB, addition of pyrazinamide to a
of optimal Cmax for sensitive strains (158). Isoniazid is effective 6-month regimen significantly reduced relapse rate to less than

Scheld_Ch29.indd 491 2/21/14 8:21 PM


492 Part III: Bacterial and Mycoplasmal Infections

5% (18,156). Pyrazinamide is very efficient in penetrating the Ethionamide may be a valuable drug for the management of
CSF. Generally high levels are found comparable to those in both susceptible and drug-resistant TBM. Ethionamide is a
serum (167). The EBA in the first few days of treatment is low structural analog of isoniazid. If isoniazid resistance is con-
but at days 4 to 14 matches that of rifampicin and isoniazid ferred by mutations in the KatG gene, these isolates may still
and is probably also active against extracellular bacilli (18). be sensitive to ethionamide; however, inhA mutations for
Because rifampicin penetration in CSF is limited and isoniazid isoniazid resistance confer cross-resistance to ethionamide
resistance is frequent, the role of pyrazinamide in TBM should (173,174). Cycloserine is only moderately bactericidal and
not be underestimated. has an MIC of 25 to 75 g/mL; however, penetration in CSF
is good (157). None of the second-line drugs have been sub-
jected to clinical trials for the use in TBM treatment with the
Drug-Resistant Tuberculous Meningitis exception of moxifloxacin and levofloxacin (160,161).
Of the new agents, a diarylquinoline (bedaquiline or Sirturo,
The general principles for treating multidrug-resistant TB previously known as TMC207) was approved for the treat-
are as follows: (a) use at least three previously unused drugs, ment of MDR TB by the U.S. Food and Drug Administration
one of which should be a fluoroquinolone; (b) streptomycin in December 2012. It has a novel mode of action specifically in-
resistance does not confer resistance to other aminoglycosides, hibiting mycobacterial adenosine triphosphate (ATP) synthase
therefore amikacin or kanamycin can be used; and (c) treat (175). When added to a background regimen of second-line
for at least 18 months (168). There is no uniform guideline drugs in patients with MDR pulmonary TB, TMC207 led to
for treatment of drug-resistant TBM. No clinical trials have more rapid sputum culture conversion and possibly prevented
been conducted. Our ability to diagnose drug-resistant TBM resistance formation in companion drugs without adding to
more rapidly with PCR-based techniques will warrant an toxicity of the regimen (176). Its MIC against M. tuberculosis
expeditious introduction of protocols for practitioners in is very low (0.06 g/mL) (177). The EBA in the first 3 days
all settings. Such guidelines should be based on our existing of treatment is not optimal, but from days 4 to 7, TMC207
pharmacokinetic and pharmacodynamic knowledge in order induced similar reduction in CFUs to rifampicin and isoniazid
to minimize toxicity and maximize efficacy. Most second-line over the same period (178). The ability of this new compound
TB drugs have limited bactericidal capacity and do not diffuse to cross the bloodbrain barrier has not been established yet;
easily to the CSF (41). however, it has favorable pharmacokinetic properties and is an
attractive candidate for future studies in patients with TBM.
In view of the severity of TBM, generally, higher toxicity
of treatment may be acceptable if accompanied by improved
Second-Line Agents clinical outcomes. The most common reason for treatment
interruption is hepatotoxicity. Especially isoniazid, rifampi-
Of the second-line drugs, fluoroquinolones are an attractive
cin, and pyrazinamide are implicated in drug-induced hepa-
option for the treatment of TB meningitis because of their
titis (DIH); however, when carefully monitored, gross hepatic
demonstrable in vitro activity, intracellular penetration, toler-
failure can be prevented. Streptomycin can cause oto- and
ability, good bioavailability, and ease of administration. With
nephrotoxicity, and ethambutol optic neuritis. Cardiac tachyar-
the exception of ciprofloxacin, the mycobactericidal activity is
rhythmias are reported for the fluoroquinolones, moxifloxacin
comparable to that of isoniazid. The EBA of levofloxacin, gati-
in particular, but are very rare. Key to favorable outcome is
floxacin, and moxifloxacin in pulmonary TB were compared
early initiation of effective anti-TB treatment. Clinical trials
to that of isoniazid by Johnson and colleagues (169). This
are needed to develop more effective treatment guidelines for
study reported levofloxacin to have the greatest EBA, compa-
both drug-susceptible and resistant TBM. General manage-
rable to that of isoniazid. The EBA 02 of INH (0.67 log10
ment guidelines have been published by the British Infection
cfu/ml/day) was greater than that of moxifloxacin and gati-
Society and may be helpful for clinicians to make individual
floxacin [both 400 mg daily] (0.33 and 0.35 log10 cfu/ml/day,
treatment decisions (Fig. 29.9) (75).
respectively), but not of levofloxacin 1000 mg daily (0.45 log10
cfu/ml/day) (169). The diffusion to the cerebral compartment
is excellent. A pharmacokinetic study comparing ciprofloxa- Adjunctive Treatment
cin, levofloxacin, and gatifloxacin in patients with TBM found
levofloxacin to have excellent CSF penetration, with a ratio
of area under the curve (AUC) in CSF to AUC in plasma of
Corticosteroids
75%. This compared favorably with gatifloxacin (35%) and Global guidelines now recommend the use of corticosteroids
ciprofloxacin (14%) (170). For moxifloxacin, the penetration as an adjunct to treatment. Addition of dexamethasone to the
has been reported to be 71% and 82% of plasma levels with antibiotic regimen used in TBM has been proven to reduce
doses of 800 mg and 400 mg, respectively (171). mortality in TBM in both adults and children (30,179).
In serum, median free AUC024/actual MIC90 in plasma was A Cochrane review concluded that the adjunctive use of
180.99 for levofloxacin, 179.77 for gatifloxacin, and 58.35 for corticosteroids reduce the risk of death (risk ratio [RR], 0.78;
moxifloxacin. Cmax was 15.55 g/mL for levofloxacin, 4.51 g/ 95% CI, 0.67 to 0.91; 1140 participants, 7 trials) in patients
mL for gatifloxacin, and 6.13 g/mL for moxifloxacin (172). with TBM. Data on disabling residual neurologic deficit
These pharmacokinetic data suggest that the fluoroquinolones, showed that corticosteroids additionally reduce the risk of
levofloxacin and moxifloxacin in particular, have the poten- death or disabling residual neurologic deficit (RR, 0.82; 95%
tial to be highly effective sterilizing agents in the treatment of CI, 0.70 to 0.97; 720 participants, 3 trials) (180). This has
TBM. not been proven or refuted for HIV-positive patients. The
Of the remaining second-line drugs, only ethionamide and beneficial effect of steroids may not be homogenously distrib-
cycloserine have considerable ability to diffuse to the CSF. In uted among different patient populations. Across the different
TBM, serum to CSF penetration ratio of 40% to 100% were severity groups, the beneficial effect of dexamethasone may
reported, with peak CSF concentrations ranging from 1.0 to be more pronounced in the less severe grades (30,48). More
2.6 g/mL achieved 3 hours after administration of a 250-mg recently, it has been reported that the immune response can
dose (155). The reported MIC was 0.25 to 0.5 g/mL (157). be modulated through the leukotriene A4 hydrolase (LTA4H)

Scheld_Ch29.indd 492 2/21/14 8:21 PM


Chapter 29: Tuberculous Meningitis 493

FIGURE 29.9 Management of TBM (British Infection Society guidelines).

Scheld_Ch29.indd 493 2/21/14 8:21 PM


494 Part III: Bacterial and Mycoplasmal Infections

gene. Indirectly, this gene regulates TNF-


levels, and pro- Immune Reconstitution with Antiretroviral Therapy
moter polymorphisms can lead to either a hyperinflammatory
state, inadequate inflammation, or an intermediate response. The mortality for HIV-infected patients with TBM is alarmingly
Patients with TBM, who were homozygous for the LTA4H- high. CD4 counts of patients with TBM are characteristically
high polymorphism (T/T), had higher levels of TNF and high very low (35). For these patients, the question of timing of initi-
leukocytes in CSF. Among patients not receiving glucocorti- ation of ART is critical. Initiation should be as early as possible
coids, mortality was highest among patients with a T/T geno- to support the immune response; however, toxicity, pharmaco-
type. However, these patients showed the greatest reduction in kinetic interactions, IRIS, and pill burden may complicate early
mortality in the group treated with adjunctive dexamethasone, treatment. Two recent clinical trials on initiation of ART in
suggesting that the dexamethasone may reduce excessive in- HIV-associated TB showed that severely immunocompromised
flammatory response in this group. By contrast, patients who (CD4 200 cells/L) patients with (mostly pulmonary) TB
were homozygous for the polymorphism causing low expres- may benefit from early initiation of ART with lower rates of
sion of LTA4H (C/C) showed increased mortality when treated AIDS-defining illnesses or death, but at the cost of a higher oc-
with dexamethasone, probably due to further suppression of currence of IRIS (187,188). In TBM, mortality is much higher
an inadequate immune response. The patients who were het- and intracranial IRIS may be detrimental (189). A randomized
erozygous at this locus (C/T) had the lowest mortality and the controlled trial in Vietnam showed no reduction in mortality
use of dexamethasone did not appear to influence mortality in with immediate initiation of ARTs; instead, the study suggested
this group (181). that it may be safer to defer the initiation of ART to 8 weeks
These results suggest that disease severity in humans can be of TBM treatment in patients with TBM, as delayed treatment
caused either by an exaggerated or a deficient immune response was associated with fewer adverse events (31). TBM IRIS has
and, perhaps more importantly, that treatment with cortico- been reported to be a frequent occurrence in ART-naive pa-
steroids may only benefit those with a certain predisposition tients who started ART treatment 2 weeks after initiation of
to hyperinflammation and may be detrimental in those with TBM treatment (16/34, 47%). High CSF neutrophil counts,
deficient immune responses. This needs prospective assessment culture positivity, and a combination of high CSF TNF-
and
and if proven may necessitate more tailored use of corticoste- low IFN- concentrations on presentation was associated with
roids according to individual genotyping or phenotyping of development of IRIS (190). For a more elaborate review and
the immune response in the future. guideline of ART and TB drug interactions, we refer to the
National Institutes of Health (NIH) guidelines (191).
Thalidomide
Aspirin and Dipyridamole
Thalidomide is a drug with immunomodulatory properties
through the inhibition of TNF-
(182). In animal studies, Stroke is associated with poor outcome in TBM. Infiltrative
TNF-
levels in the CSF produced during TBM were shown and proliferative vasculitis and necrotizing vessel pathologies
to correlate with the extent of pathogenesis, although this has have been implicated in the pathogenesis. The relative contri-
been difficult to replicate in humans with TBM (84). In rabbits bution of thrombosis to the development of ischemic events is
infected intracranially with TB, an antitubercular regimen unknown. Tuberculous thrombophlebitis has been described
that included thalidomide led to reduction of TNF-
levels in earlier pathologic studies (64,87). Severe pulmonary TB is
in CSF and prevented death (183). The same group has used characterized by impaired fibrinolysis and a hypercoagulable
a thalidomide analogue (IMiD3) for the treatment of experi- state (192). In children with TBM, changes were found in
mental animals infected with TBM as an adjunct to standard procoagulation, antithrombotic factors, fibrinolysis, platelet
antituberculous treatment. IMiD3 has comparable immuno- counts, and vascular endothelium functions, all contributing
modulatory action to thalidomide, but unlike thalidomide, to an increased risk of thrombosis (193). Aspirin has been sub-
IMiD3 is not teratogenic. The additional use of IMiD3 in jected to clinical trials in TBM because it is antithrombotic and
rabbits resulted in marked improvement in survival, reduced possibly neuroprotective. However, a clinical trial in 146 chil-
CSF leukocytosis, lower levels of TNF, and attenuated inflam- dren with TBM showed no significant effect on either mortality
mation of the meninges on histologic examination. The ben- or neurologic deficits with low-dose or high-dose aspirin regi-
eficial effect on survival and severity of symptoms of IMiD3 mens (194). One open-label randomized study in 118 adults
was markedly greater than that of thalidomide in this animal on the role of aspirin showed a beneficial effect on mortality
model (184). and MRI results (195). In this study, some patients selectively
Conversely, increases of TNF-
concentrations have been received corticosteroids, which may have biased the results.
reported during thalidomide treatment, raising more ques- Larger appropriately randomized studies are needed to estab-
tions about the interaction between TNF-
and thalidomide. lish the role of aspirin (and dipyridamole) in the management
A clinical study in 15 adults with TB, including HIV-positive of TBM-related ischemic events. In an intensive neurosurgi-
and HIV-negative patients, showed a reduction in disease cal care setting in South Africa, cerebral tissue oxygenation
severity with the use of thalidomide, which was correlated was monitored in two children. A decline in oxygenation was
with an increase in TNF-
levels. By contrast, those patients reversed by aggressive therapy with oxygen, fluid resuscita-
with poor outcome who were not designated to receive tion, inotropic support, and blood transfusion, which possibly
thalidomide continued to demonstrate clinical progression of prevented infarction (196). More profound insights into the
the disease and remained with low levels of TNF-
and type pathogenesis of TBM vascular disease are necessary to guide
1 cytokines (185). rational therapeutic interventions.
In children with TBM, a randomized placebo controlled
study of adjunctive thalidomide was stopped early because of
an increased risk of death and adverse events in the treatment Raised Intracranial Pressure and
arm (186). The role of TNF-
in TB is complex and has yet Hydrocephalus
to be fully elucidated. Currently, there is no role of TNF-

inhibition in the treatment for TBM; however, the potential Furosemide with or without acetazolamide can be used for
role for IMiD3 in the management of TBM in patients may be treating communicating hydrocephalus in TBM to decrease
a subject of future pharmacokinetic research. CSF production by the choroid plexus. Some institutions favor

Scheld_Ch29.indd 494 2/21/14 8:21 PM


Chapter 29: Tuberculous Meningitis 495

daily lumbar punctures with intracranial pressure monitoring and Indo-Oceanic strains. HIV-negative patients infected
through manometry to assess the response to medical therapy. with the Beijing-strain had shorter duration of illness before
An external ventricular drain, ventriculoperitoneal shunting, presentation to hospital and fewer CSF leukocytes, suggesting
or endoscopic third ventriculostomy may be indicated for that mycobacterial genotype may affect disease progression
patients not responding to conservative measures and noncom- and the nature of the intracerebral inflammatory response
municating hydrocephalus (197). Patients for these procedures (214). Studies in South African children have also shown an
should be carefully selected, as the success rate depends on the association between Beijing genotype and extrapulmonary TB
correct diagnosis, the severity of disease, and the expertise of (208). A recent retrospective cohort study of TBM patients
the neurosurgical teams. Outcome is better in patients with in Thailand supported the theory that the Beijing genotype is
early intervention in the better grades (198). Some institutions the most pathogenic strain of M. tuberculosis and associated
use osmotic agents, such as mannitol or hypertonic saline, with TBM, whereas the Euro-American lineage was much less
however there have been no studies establishing the effective- commonly linked with TBM. Results showed that modern
ness in TB of the CNS. sublineages of Beijing genotype were associated with higher
CSF WCC and more severe (stage III) disease but not with
mortality rate (215). Conversely, among HIV-positive patients
Hyponatremia in Vietnam, those infected with the modern Beijing lineage
strains had lower mortality than patients infected with the
TBM accompanied by hyponatremia is associated with a worse ancient Indo-Oceanic lineage (HR, 0.29; 95% CI, 0.14; 0.61)
outcome. Mostly, it is caused by the syndrome of inappropri- (214). This contradictory finding might be explained by the
ate antidiuretic hormone secretion (SIADH) or cerebral salt- proinflammatory properties of the modern Beijing lineages,
wasting (CSW) syndrome. CSW is probably underdiagnosed which although detrimental to the immunocompetent host
in TBM, and the distinction between the two entities is often may be conversely protective in the immunocompromised. It is
difficult in endemic settings. Treatment should be directed to known that protective immunity in TB disease is a delicate bal-
avoid both hypoosmolality and hypovolemia. Fluid restriction ance of pro- and antiinflammatory cascades. However, studies
can be detrimental and is not recommended. Generally, it is in other regions have found no association between Beijing
advised to treat all hyponatremic patients cautiously with genotype and dissemination (216218). Within each region,
hypertonic saline with or without fludrocortisones (197,199). the M. tuberculosis genotypes circulating vary significantly as
Correction should be gradual in order to avoid pontine do the genotyping divisions applied to analysis and therefore
myelinolysis. the pathogen populations being compared are not identical.
Wider studies synthesizing global data are required to estab-
lish definitive interpretation.
Pathogen Genetics and Virulence
M. tuberculosis strains were shown to vary in virulence in the Host-Genetics
1960s when Mitchison et al. conducted experimental infection
of Guinea pigs with strains from South India and Britain It has long been known that there is a genetic element to TB
(200,201). Many epidemiologic in vitro and in vivo stud- susceptibility. Of those exposed to M. tuberculosis, approxi-
ies have since attempted to establish differences in virulence mately 10% establish an infection and, of those, 10% will
with regard to dissemination and disease severity but no develop active disease while the remaining individuals will
consensus has yet been established due to often contradic- harbor a latent infection. Approximately 1% of active TB
tory findings and the difficulty of interpretation (202,203). cases develop into TBM. Susceptibility to the different forms
There is wide variation in experimental technique in vitro of TB (latent, active, disseminated) is a complex interplay
or the comparative strains in both laboratory and epidemio- between host genetics, pathogen genetics, and environmental
logic studies. Evolutionarily, TBM represents a dead end for factors (such as smoking, malnutrition, comorbidities).
the pathogen and therefore propensity to cause TBM is not Protective immunity to TB depends on innate immunity
directly advantageous but may be a by-product of increased and an effective TH1 response, as evidenced by the dramatic
pulmonary virulence causing greater transmission (204). The increase in susceptibility shown by HIV-infected individuals.
standard laboratory strain, H37Rv, which was the first M. Many candidate genes have been proposed to be associated
tuberculosis strain sequenced in 1997 (205), is laboratory with susceptibility to pulmonary TB (219222), but fewer
adapted and exhibits low virulence in vitro and in animal studies have looked at the association between host genetics
models in comparison with almost all clinical strains (206), and susceptibility to severe and disseminated disease. The
therefore demonstration of increased virulence in comparison toll-like (TLR) receptor pathway has been implicated in TB
with H37RV does not represent a hypervirulent strain. progression. The human TLR family has 12 members that can
Analysis of large sequence polymorphisms within the recognize pathogen-associated molecular patterns (PAMPs)
M. tuberculosis genome has defined six major global lineages, and upon activation initiate an innate immune response,
which are largely geographically restricted within the cytokine production, and the formation of the adaptive
eponymous region; Indo-Oceanic, Euro-American, East Asian, immune response. The TLRs known to be involved in M.
East African-Indian, and two Mycobacterium africanum lin- tuberculosis recognition are TLR2, TLR4, TLR9, and possibly
eages. Within the modern East-Asian lineage, the Beijing TLR8 (223). One of the few host genetic studies to investigate
genotype has been defined by its characteristic spoligotype susceptibility factors for TBM as distinct from pulmonary TB
and extensively investigated, as it has been associated with showed an association between a polymorphism in the TLR2
increased virulence and drug resistance in several regions gene (SNP T597C) and the development of TBM and miliary
(207212). Introduction of the Beijing genotype into some TB, indicating the TLR2 pathway plays a role in the ability to
regions also resulted in rapid increases in prevalence of this disseminate of M. tuberculosis (214). However, this polymor-
strain (212). In Vietnam, among TBM cases, it shows sig- phism is itself synonymous, and the causative single nucleotide
nificant association with HIV status, drug resistance, and polymorphism (SNP) in linkage disequilibrium has not yet
multidrug resistance (213) compared with Euro-American been identified. Further studies investigating the interaction

Scheld_Ch29.indd 495 2/21/14 8:21 PM


496 Part III: Bacterial and Mycoplasmal Infections

between host and pathogen in TBM susceptibility have shown need for the development of a rapid, sensitive, point of care
that individuals with the TLR2 polymorphism are more likely diagnostic that can be used in decentralized settings. This will
to be infected with the Beijing genotype of M. tuberculosis lead to a reduction in frequently encountered disastrous treat-
and that this association is strongest in those with TBM (214). ment delays.
Polymorphisms in the toll-interleukin 1 receptor domain-con- Currently, the TB drug pipeline has 10 compounds in the
taining adaptor protein (TIRAP) gene have also been shown clinical development phase for both MDR and drug-sensitive
to be associated with susceptibility to TBM in Vietnam and TB (http://www.newtbdrugs.org/pipeline.php). Because the
South Africa (224,225). TIRAP is a protein further down the pipeline is aimed to create affordable, tolerable, active new
TLR pathway, which mediates signals from the TLR receptors, antimycobacterials for pulmonary TB, CSF penetration is
activating macrophages and dendritic cells. not necessarily an attribute that these candidate compounds
Recently a SNP in the promoter region of the LTA4H gene have been developed for. Assessing any new drug that emerges
has been identified to play a role in susceptibility to myco- from the pipeline will need to be subjected to well-designed,
bacteria which is discussed in more detail in the section on adequately powered clinical trials for TBM, which are
corticosteroid treatment. Future research on host genetic resource consuming and difficult to achieve and will require
susceptibility to TB and TBM will need to explore the poten- multisite involvement. Other areas of future research for TBM
tial of genetically tailored adjunctive therapies. treatment should include the benefit of adjunctive drugs such
as aspirin or immune-modulating drugs, which may reduce
sequelae. In general, future trials should be aimed at efficiency
CONCLUSION by using innovative trial designs, making more efficient use
of existing drugs, and by basing the trial rationale on existing
CNS TB is a devastating form of TB. In resource-limited pharmacokinetic/pharmacodynamic data, in particular, the
settings, it places a high burden on patients, their fami- ability to penetrate the brain. Protection conferred by BCG
lies, society, and health care systems. Disease incidence is a vaccine is insufficient. The TB vaccine pipeline has 11 vac-
direct reflection of the TB epidemic and complicated by the cine candidates entered in clinical trials. (http://www.tbvi
MDR/XDR TB and HIV epidemic. The current develop- .eu/fileadmin/user_upload/Documenten/News/TB_Vaccine_
ments in TBM research have advanced our understanding Pipeline_2011_FINAL03042012.pdf). Most are preexpo-
of the disease; however, they have not led to acceptable im- sure vaccines aimed to prevent TB disease intended to either
provements in clinical outcomes. Early diagnosis and treat- replace BCG (recombinant live vaccines) or to be given after
ment initiation are essential to a good outcome. The recent BCG prime as boosters (either protein adjuvant formulations
endorsement of the GeneXpert test for pulmonary TB may or recombinant viral carriers) (226). Recently, MVA85A, a
impact the management of MDR TBM in particular, which booster vaccine for BCG, was subjected to the first efficacy
is highly lethal, and offers the potential for improved diag- trial since BCG; however, it failed to prove a statistically sig-
nostics for TBM. However, more robust evidence must be nificant effect (227). The development of an effective vaccine
generated to validate the specificity for rifampicin resistance has the potential to greatly influence the TB epidemic, how-
on CSF, because withholding rifampicin from treatment regi- ever requiring renewed funding incentives, appropriately
mens for sensitive strains may be detrimental. Little is known structured trials, and eventually government commitment to
about the optimal second-line treatment regimen for MDR support vaccination strategies. It is a less popular notion, but
TBM and this may pose TB programs and clinicians with a fundamental to the control of both the TB and HIV pandemics
problem when confronted with a rifampicin-resistant positive is the commitment of policymakers to address socioeconomic
result on GeneXpert, without an elaborate drug sensitivity issues, such as poverty, crowding, lack of education, failing
spectrum. Both first- and second-line treatment will need to health care infrastructure, and lack of access to health care
be optimized for TBM according to the ability of antimyco- factors significantly contributing to the perpetuation of these
bacterial drugs to penetrate the brain. Still, there is a pressing epidemics.

References

1. Ruhrah J. The history of tuberculous meningitis. Med Library Hist J. 14. Kwapinski J. [Streptomycin resistance of Mycobacterium tuberculosis
1904;2(3):160165. cultured in cerebro-spinal fluid]. Gruzlica. 1950;18(34):437447.
2. Madkour M. Tuberculosis. New York: Springer-Verlag: 2004:928. 15. Long ER. [Resistance of Mycobacterium tuberculosis to streptomycin]. Rev
3. Whytt R. Observations on the Dropsy in the Brain. Edinburgh: J. Balfour; 1768. Bras Tuberc Doencas Torac. 1950;18(130):440454.
4. Meindl JL, Meindl CO. Tuberculous meningitis in the 1830s. Lancet. 16. Muratore A. [Study of streptomycin resistance of the bacterial flora of the
1982;1(8271):554555. tuberculous meningitis departments and its relation with the insurgence
5. Green P. Researches into the diseases of children, conducted on the known of supervening meningitis during streptomycin therapy]. Riv Clin Pediatr.
principles of anatomy and pathology. Lancet. 1836;25(643):492495. 1950;48(5):291304.
6. Pearce JM. Walter Essex Wynter, Quincke, and lumbar puncture. J Neurol 17. Pothmann FJ, Fehr KO. [Resistance of the tubercle bacillus to streptomycin
Neurosurg Psychiatry. 1994;57(2): 179. and to streptomycin in combination with TB I in tuberculous meningitis].
7. Wynter W. Four cases of tubercular meningitis in which paracentesis was Beitr Klin Tuberk Spezif Tuberkuloseforsch. 1950;103(5):422430.
performed for the relief of fluid pressure. Lancet. 1891;1:981982. 18. Donald PR. The chemotherapy of tuberculous meningitis in children and
8. Morton CA. The pathology of tuberculous meningitis with reference to its adults. Tuberculosis (Edinb). 2010;90(6):375392.
treatment by tapping the subarachnoid space of the spinal cord. Br Med J. 19. Centers for Disease Control and Prevention. HIV and TB Factsheet.
1891;2(1607):840841. Atlanta: Centers for Disease Control and Prevention; 2011:2.
9. Rich AR, McCordock HA. The pathogenesis of tuberculous meningitis. 20. Lozano R, Naghavi M, Foreman K. Global and regional mortality from
Bull John Hopkins Hosp. 1933;52:237. 235 causes of death for 20 age groups in 1990 and 2010: a systematic anal-
10. Keers RY. The gold rush 1925-35. Thorax. 1980;35(12):884889. ysis for the Global Burden of Disease Study 2010. Lancet. 2012;380(9859):
11. Rich CB, Samuels AJ. Tuberculous meningitis treated with streptomycin. 20952128.
Can Med Assoc J. 1948;58(3):282284. 21. World Health Organization. Global Tuberculosis Control: WHO Report
12. Anderson W, Kaake M. In vivo development of streptomycin resistance by 2011. Geneva: World Health Organization; 2011:258.
the tubercle bacillus. Can Med Assoc J. 1950;62(1):5961. 22. Wolzak NK, Cooke ML, Orth H, et al. The changing profile of pediatric
13. de Partearroyo FR, Morante AF. [Streptomycin resistance of the tubercle meningitis at a referral centre in Cape Town, South Africa. J Trop Pediatr.
bacillus]. Arch Tisiol Mal Appar Respir. 1950;5(8):639648. 2012;58(6):491495.

Scheld_Ch29.indd 496 2/21/14 8:21 PM


Chapter 29: Tuberculous Meningitis 497

23. Meningitis Research Foundation. Meningitis and septicaemia: UK facts and 55. Flynn JL, Chan J, Triebold KJ, et al. An essential role for interferon
figures. http://www.meningitis.org/facts. Accessed June 6, 2014. gamma in resistance to Mycobacterium tuberculosis infection. J Exp Med.
24. van den Bos F, Terken M, Ypma L, et al. Tuberculous meningitis and 1993;178(6):22492254.
miliary tuberculosis in young children. Trop Med Int Health. 2004;9(2) 56. Cooper AM. Cell-mediated immune responses in tuberculosis. Annu Rev
309313. Immunol. 2009;27:393422.
25. Starke JR. Tuberculosis of the central nervous system in children. Semin 57. Ramakrishnan L. Revisiting the role of the granuloma in tuberculosis. Nat
Pediatr Neurol. 1999;6(4):318331. Rev Immunol. 2012;12(5):352366.
26. Thwaites G, Chau TT, Mai NT, et al. Tuberculous meningitis. J Neurol 58. Miller EA, Ernst JD. Illuminating the black box of TNF action in tubercu-
Neurosurg Psychiatry. 2000;68(3):289299. lous granulomas. Immunity. 2008;29(2):175177.
27. Principi N, Esposito S. Diagnosis and therapy of tuberculous meningitis in 59. Rock RB, Hu S, Gekker G, et al. Mycobacterium tuberculosis-induced
children. Tuberculosis (Edinb). 2012;92(5):377383. cytokine and chemokine expression by human microglia and astrocytes:
28. Farinha NJ, Razali KA, Holzel H. Tuberculosis of the central nervous effects of dexamethasone. J Infect Dis. 2005;192(12):20542058.
system in children: a 20-year survey. J Infect. 2000;41(1):6168. 60. Tsenova L, Bergtold A, Freedman VH, et al. Tumor necrosis factor alpha
29. van Well GT, Paes BF, Terwee CB, et al. Twenty years of pediatric tubercu- is a determinant of pathogenesis and disease progression in mycobacterial
lous meningitis: a retrospective cohort study in the Western Cape of South infection in the central nervous system. Proc Natl Acad Sci U S A. 1999;
Africa. Pediatrics. 2009;123(1):e1e8. 96(10):56575662.
30. Thwaites GE, Nguyen DB, Nguyen HD, et al. Dexamethasone for the 61. Thwaites GE, Simmons CP, Than Ha Quyen N, et al. Pathophysiology and
treatment of tuberculous meningitis in adolescents and adults. N Engl J prognosis in Vietnamese adults with tuberculous meningitis. J Infect Dis.
Med. 2004;351(17):17411751. 2003;188(8):11051115.
31. Torok ME, Yen NT, Chau TT, et al. Timing of initiation of antiretroviral 62. Patel VB, Bhigjee AI, Bill PL, et al. Cytokine profiles in HIV seropositive
therapy in human immunodeficiency virus (HIV)associated tuberculous patients with tuberculous meningitis. J Neurol Neurosurg Psychiatry.
meningitis. Clin Infect Dis. 2011;52(11):13741383. 2002;73(5):598599.
32. Vinnard C, Macgregor RR. Tuberculous meningitis in HIV-infected 63. Brighenti S, Andersson J. Local immune responses in human tuberculo-
individuals. Curr HIV/AIDS Rep. 2009;6(3):139145. sis: learning from the site of infection. J Infect Dis . 2012;205(suppl 2):
33. World Health Organization. Global HIV/AIDS Response: Epidemic S316S324.
Update and Health Sector Progress Towards Universal Access. Progress 64. Poltera AA. Thrombogenic intracranial vasculitis in tuberculous meningi-
report 2011. Geneva: World Health Organization; 2010:233. tis. A 20-year post mortem survey. Acta Neurol Belg. 1977;77(1):1224.
34. World Health Organization. TB/HIV: A Clinical Manual. Geneva: World 65. Poltera AA, Templeton AC. Intracranial tuberculosis in Uganda: a post-
Health Organization; 2010:212. mortem survey. Afr J Med Sci. 1973;4(4):343349.
35. Torok ME, Chau TT, Mai PP, et al. Clinical and microbiological features 66. Rock RB Olin M, Baker CA, et al. Central nervous system tuberculo-
of HIV-associated tuberculous meningitis in Vietnamese adults. PLoS One. sis: pathogenesis and clinical aspects. Clin Microbiol Rev. 2008;21(2):
2008;3(3):e1772. 243261.
36. Garg RK, Sinha MK. Tuberculous meningitis in patients infected with 67. Sagawa I, Miyano T, Saito H, et al. The pathogenesis of tuberculous menin-
human immunodeficiency virus. J Neurol. 2011;258(1):313. gitis; a criticism of Richs focus-theory. Acta Tuberc Jpn. 1954;4(1):1218.
37. Marais S, Pepper DJ, Marais BJ, et al. HIV-associated tuberculous menin- 68. Donald PR, Schaaf HS, Schoeman JF. Tuberculous meningitis and miliary
gitisdiagnostic and therapeutic challenges. Tuberculosis (Edinb). 2010; tuberculosis: the Rich focus revisited. J Infect. 2005;50(3):193195.
90(6):367374. 69. Dastur DK, Manghani DK, Udani PM. Pathology and pathogenetic
38. World Health Organization. Multidrug and Extensively Drug-Resistant TB mechanisms in neurotuberculosis. Radiol Clin North Am. 1995;33(4):
(M/XDR-TB): 2010 Global Report on Surveillance and Response. Geneva: 733752.
World Health Organization; 2010:77. 70. Schaaf HS, Zumla A. Tuberculosis: A Comprehensive Clinical Reference.
39. Thwaites GE, Lan NT, Dung NH, et al. Effect of antituberculosis drug Saunders: Stellenbosch, South Africa; 2009:101.
resistance on response to treatment and outcome in adults with tuberculous 71. Wasay M, Kheleani BA, Moolani MK, et al. Brain CT and MRI findings in
meningitis. J Infect Dis. 2005;192(1):7988. 100 consecutive patients with intracranial tuberculoma. J Neuroimaging.
40. Tho DQ, Trk ME, Yen NT, et al. Influence of antituberculosis drug 2003;13(3):240247.
resistance and mycobacterium tuberculosis lineage on outcome in HIV- 72. Arseni C. Two hundred and one cases of intracranial tuberculoma treated
associated tuberculous meningitis. Antimicrob Agents Chemother. 2012; surgically. J Neurol Neurosurg Psychiatry. 1958;21(4):308311.
56(6):30743079. 73. Thwaites GE, Macmullen-Price J, Tran TH, et al. Serial MRI to determine
41. Patel VB, Padayatchi N, Bhigjee AI, et al. Multidrug-resistant tubercu- the effect of dexamethasone on the cerebral pathology of tuberculous
lous meningitis in KwaZulu-Natal, South Africa. Clin Infect Dis. 2004; meningitis: an observational study. Lancet Neurol. 2007;6(3):230236.
38(6):851856. 74. Anuradha HK, Garg RK, Sinha MK, et al. Intracranial tuberculomas in
42. Seddon JA, Visser DH, Bartens M, et al. Impact of drug resistance on clini- patients with tuberculous meningitis: predictors and prognostic signifi-
cal outcome in children with tuberculous meningitis. Pediatr Infect Dis J. cance. Int J Tuberc Lung Dis. 2011;15(2):234239.
2012;31(7):711716. 75. Thwaites G, Fisher M, Hemingway C, et al. British Infection Society guide-
43. Thwaites GE, Chau TT, Caws M, et al. Isoniazid resistance, mycobacterial lines for the diagnosis and treatment of tuberculosis of the central nervous
genotype and outcome in Vietnamese adults with tuberculous meningitis. system in adults and children. J Infect. 2009;59(3):167187.
Int J Tuberc Lung Dis. 2002;6(10):865871. 76. Dastur DK, Udani PM. The pathology and pathogenesis of tuberculous
44. Vinnard C, Winston CA, Wileyto EP, et al. Isoniazid resistance and death encephalopathy. Acta Neuropathol. 1966;6(4):311326.
in patients with tuberculous meningitis: retrospective cohort study. BMJ. 77. Udani PM, Dastur DK. Tuberculous encephalopathy with and without
2010;341:c4451. meningitis. Clinical features and pathological correlations. J Neurol Sci.
45. Schoeman J, Wait J, Burger M, et al. Long-term follow up of childhood 1970;10(6):541561.
tuberculous meningitis. Dev Med Child Neurol. 2002;44(8):522526. 78. Lammie GA, Hewlett RH, Schoeman JF, et al. Tuberculous encephalopa-
46. Christensen AS, Roed C, Omland LH, et al. Long-term mortality in patients thy: a reappraisal. Acta Neuropathol. 2007;113(3):227234.
with tuberculous meningitis: a Danish nationwide cohort study. PLoS One. 79. Wadia NH, Dastur DK. Spinal meningitides with radiculo-myelopathy.
2011;6(11):e27900. 1. Clinical and radiological features. J Neurol Sci. 1969;8(2):239260.
47. Kalita J, Misra UK, Ranjan P. Predictors of long-term neurological sequelae 80. Hernandez-Albujar S, Arribas JR, Royo A, et al. Tuberculous radicu-
of tuberculous meningitis: a multivariate analysis. Eur J Neurol. 2007; lomyelitis complicating tuberculous meningitis: case report and review.
14(1):3337. Clin Infect Dis. 2000;30(6):915921.
48. Torok ME, Nguyen DB, Tran TH, et al. Dexamethasone and long-term 81. Cascino J, Dibble JB. Tuberculoma of spinal cord. J Am Med Assoc. 1956;
outcome of tuberculous meningitis in Vietnamese adults and adolescents. 162(5):461462.
PLoS One. 2011;6(12):e27821. 82. Ramdurg SR, Gupta DK, Suri A, et al. Spinal intramedullary tuberculosis:
49. Orme IM. The Achilles heel of BCG. Tuberculosis (Edinb). 2010;90(6): a series of 15 cases. Clin Neurol Neurosurg. 2009;111(2):115118.
329332. 83. Lu M. Imaging diagnosis of spinal intramedullary tuberculoma: case
50. Trunz BB, Fine P, Dye C. Effect of BCG vaccination on childhood reports and literature review. J Spinal Cord Med. 2009;33(2):159162.
tuberculous meningitis and miliary tuberculosis worldwide: a meta-analysis 84. Lammie GA, Hewlett RH, Schoeman JF, et al. Tuberculous cerebrovascular
and assessment of cost-effectiveness. Lancet. 2006;367(9517):11731180. disease: a review. J Infect. 2009;59(3):156166.
51. Khemiri M, Bagais A, Becher SB, et al. Tuberculous meningitis in Bacille 85. Misra UK, Kalita J, Maurya PK. Stroke in tuberculous meningitis. J Neurol
Calmette-Guerin-vaccinated children: clinical spectrum and outcome. Sci. 2011;303(12):2230.
J Child Neurol. 2012;27(6):741746. 86. Dekker G, Andronikou S, van Toorn R, et al. MRI findings in children with
52. Flynn JL, Chan J, Lin PL. Macrophages and control of granulomatous tuberculous meningitis: a comparison of HIV-infected and non-infected
inflammation in tuberculosis. Mucosal Immunol. 2011;4(3):271278. patients. Childs Nerv Syst. 2011;27(11):19431949.
53. van Crevel R, Ottenhoff TH, van der Meer JW. Innate immunity to 87. Hektoen L. The vascular changes of tuberculous meningitis, especially the
Mycobacterium tuberculosis. Clin Microbiol Rev. 2002;15(2):294309. tuberculous endarterities. J Exp Med. 1896;1(1):112163.
54. Urdahl KB, Shafiani S, Ernst JD. Initiation and regulation of T-cell responses 88. Thwaites GE, Tran TH. Tuberculous meningitis: many questions, too few
in tuberculosis. Mucosal Immunol. 2011;4(3):288293. answers. Lancet Neurol. 2005;4(3):160170.

Scheld_Ch29.indd 497 2/21/14 8:21 PM


498 Part III: Bacterial and Mycoplasmal Infections

89. Hosoglu S, Ayaz C, Geyik MF, et al. Tuberculous meningitis in adults: an 117. Kilpatrick ME, Girgis NI, Tribble D, et al. The value of the tuberculin skin
eleven-year review. Int J Tuberc Lung Dis. 1998;2(7):553557. test in patients with tuberculous meningitis. J Egypt Public Health Assoc.
90. Girgis NI, Sultan Y, Farid Z, et al. Tuberculosis meningitis, Abbassia Fever 1996;71(12):18.
Hospital-Naval Medical Research Unit No. 3-Cairo, Egypt, from 1976 to 118. Kerkhoff AD, Kranzer K, Samandari T, et al. Systematic review of TST
1996. Am J Trop Med Hyg. 1998;58(1):2834. responses in people living with HIV in under-resourced settings: implica-
91. Gourie-Devi M. Optochiasmatic arachnoiditis and neurotuberculosis: tions for isoniazid preventive therapy. PLoS One. 2012;7(11):e49928.
prognostic indicators and therapeutic strategies. Neurol India. 2010;58(5): 119. Heaf F. The multiple-puncture tuberculin test. Lancet. 1951;2(6674):
714715. 151153.
92. Garg RK, Paliwal V, Malhotra HS. Tuberculous optochiasmatic arach- 120. Ocana I, Martinez-Vazquez JM, Segura RM, et al. Adenosine deaminase
noiditis: a devastating form of tuberculous meningitis. Expert Rev Anti in pleural fluids. Test for diagnosis of tuberculous pleural effusion. Chest.
Infect Ther. 2012;9(9):719729. 1983;84(1):5153.
93. van der Weert EM, Hartgers NM, Schaaf HS, et al. Comparison of diagnostic 121. Gupta BK, Bharat A, Debapriya B, et al. Adenosine deaminase levels
criteria of tuberculous meningitis in human immunodeficiency virus-infected in CSF of tuberculous meningitis patients. J Clin Med Res. 2010;2(5):
and uninfected children. Pediatr Infect Dis J. 2006;25(1):6569. 220224.
94. Smith H. Paradoxical responses during the chemotherapy of tuberculosis. 122. Xu, Jiang RH, Li L, et al. Diagnostic value of adenosine deaminase in
J Infect. 1987;15(1):13. cerebrospinal fluid for tuberculous meningitis: a meta-analysis. Int J
95. Das A, Das SK, Mandal A, et al. Cerebral tuberculoma as a manifestation Tuberc Lung Dis. 2010;14(11):13821387.
of paradoxical reaction in patients with pulmonary and extrapulmonary 123. Katti MK. Pathogenesis, diagnosis, treatment, and outcome aspects of
tuberculosis. J Neurosci Rural Pract. 2012;3(3):350354. cerebral tuberculosis. Med Sci Monit. 2004;10(9):RA215RA229.
96. Somer T, Finegold SM. Vasculitides associated with infections, immuniza- 124. World Health Organization. Commercial Serodiagnostic Tests for
tion, and antimicrobial drugs. Clin Infect Dis. 1995;20(4):10101036. Diagnosis of Tuberculosis. Expert Group Meeting Report 2010. Geneva:
97. Udani, PM, Parekh UC, Dastur DK Neurological and related syndromes World Health Organization; 2010.
in CNS tuberculosis. Clinical features and pathogenesis. J Neurol Sci. 125. Lawn SD. Point-of-care detection of lipoarabinomannan (LAM) in urine
1971;14(3):341357. for diagnosis of HIV-associated tuberculosis: a state of the art review.
98. Thwaites GE, Chau TT, Stepniewska K, et al. Diagnosis of adult tu- BMC Infect Dis. 2012;12:103.
berculous meningitis by use of clinical and laboratory features. Lancet. 126. Lawn SD, Kerkhoff AD, Vogt M, et al. Clinical significance of lipoarabi-
2002;360(9342):12871292. nomannan detection in urine using a low-cost point-ofcare diagnostic
99. Torok ME, Nghia HD, Chau TT, et al. Validation of a diagnostic algo- assay for HIV-associated tuberculosis. Aids. 2012;26(13):16351643.
rithm for adult tuberculous meningitis. Am J Trop Med Hyg. 2007;77(3): 127. Chan J, Fan XD, Hunter SW, et al. Lipoarabinomannan, a possible
555559. virulence factor involved in persistence of Mycobacterium tuberculosis
100. Marais S, Thwaites G, Schoeman JF, et al. Tuberculous meningitis: a within macrophages. Infect Immun. 1991;59(5):17551761.
uniform case definition for use in clinical research. Lancet Infect Dis. 128. Patel VB, Singh R, Connolly C, et al. Comparison of a clinical prediction
2010;10(11):803812. rule and a LAM antigen-detection assay for the rapid diagnosis of TBM in
101. Thwaites GE, Chau TT, Farrar JJ. Improving the bacteriological diagnosis a high HIV prevalence setting. PLoS One. 2010;5(12):e15664.
of tuberculous meningitis. J Clin Microbiol. 2004;42(1):378379. 129. Park SC, Lee BI, Cho SN, et al. Diagnosis of tuberculous meningitis by
102. Kumar P, Srivatsava MV, Singh S, et al. Filtration of cerebrospinal detection of immunoglobulin G antibodies to purified protein derivative
fluid improves isolation of mycobacteria. J Clin Microbiol. 2008;46(8): and lipoarabinomannan antigen in cerebrospinal fluid. Tuber Lung Dis.
28242825. 1993;74(5):317322.
103. Zarabi M, Sane S, Girdany BR. The chest roentgenogram in the early 130. Simmons CP, Thwaites GE, Quyen NT, et al. Pretreatment intracerebral
diagnosis of tuberculous meningitis in children. Am J Dis Child. and peripheral blood immune responses in Vietnamese adults with tuber-
1971;121(5):389392. culous meningitis: diagnostic value and relationship to disease severity
104. Munt PW. Miliary tuberculosis in the chemotherapy era: with a clinical re- and outcome. J Immunol. 2006;176(3):20072014.
view in 69 American adults. Medicine (Baltimore). 1972;51(2):139155. 131. Vidhate MR, Singh MK, Garg RK, et al. Diagnostic and prognostic value
105. Piersimoni C, Scarparo C, Callegaro A, et al. Comparison of MB/Bact of Mycobacterium tuberculosis complex specific interferon gamma release
alert 3D system with radiometric BACTEC system and Lowenstein-Jensen assay in patients with tuberculous meningitis. J Infect. 2011;62(5):400403.
medium for recovery and identification of mycobacteria from clinical 132. Pai M, Flores LL, Pai N, et al. Diagnostic accuracy of nucleic acid
specimens: a multicenter study. J Clin Microbiol. 2001; 39(2):651657. amplification tests for tuberculous meningitis: a systematic review and
106. Caws M, Dang TM, Torok E, et al. Evaluation of the MODS culture meta-analysis. Lancet Infect Dis. 2003;3(10):633643.
technique for the diagnosis of tuberculous meningitis. PLoS One. 133. Boehme CC, Nicol MP, Nabeta P, et al. Feasibility, diagnostic accu-
2007;2(11):e1173. racy, and effectiveness of decentralised use of the Xpert MTB/RIF test
107. Venkataswamy MM, Rafi W, Nagarathna S, et al. Comparative evaluation for diagnosis of tuberculosis and multidrug resistance: a multicentre
of BACTEC 460TB system and Lowenstein-Jensen medium for the isola- implementation study. Lancet. 2011;377(9776):14951505.
tion of M. tuberculosis from cerebrospinal fluid samples of tuberculous 134. Ioannidis P, Papaventsis D, Karabela S, et al. Cepheid GeneXpert MTB/
meningitis patients. Indian J Med Microbiol. 2007;25(3):236240. RIF assay for Mycobacterium tuberculosis detection and rifampin
108. World Health Organization. Non-Commercial Culture and Drug- resistance identification in patients with substantial clinical indications
Susceptibility Testing Methods for Screening of Patients at Risk of Multi- of tuberculosis and smear-negative microscopy results. J Clin Microbiol.
Drug Resistant TuberculosisPolicy Statement. Geneva: World Health 2011;49(8):30683070.
Organization; 2010. 135. Hillemann D, Rsch-Gerdes S, Boehme C, et al. Rapid molecular detection
109. World Health Organization. Use of Liquid TB Culture and Drug of extrapulmonary tuberculosis by the automated GeneXpert MTB/RIF
Susceptibility Testing (DST) in Low and Middle Income Countries. system. J Clin Microbiol. 2011;49(4):12021205.
Summary Report of the Expert Group Meeting on the Use of Liquid 136. Tortoli E, Russo C, Piersimoni C, et al. Clinical validation of Xpert MTB/
Culture Media. Geneva: World Health Organization; 2007. RIF for the diagnosis of extrapulmonary tuberculosis. Eur Respir J.
110. Schoeman CJ. The epidemiology and outcome of childhood tuber- 2012;40(2):442447.
culous meningitis. The Pelonomi Hospital experience. S Afr Med J. 137. Vadwai V, Boehme C, Nabeta P, et al. Xpert MTB/RIF: a new pillar in diagno-
1990;78(5):245247. sis of extrapulmonary tuberculosis? J Clin Microbiol. 2011;49(7):25402545.
111. World Health Organization. Molecular Line Probe Assay for Rapid 138. World Health Organization. Automated Real-time Nucleic Acid Amplification
Screening of Patients at Risk of Multidrug-Resistant Tuberculosis Technology for Rapid and Simultaneous Detection of Tuberculosis and
(MDR-TB). Policy Statement. Geneva: World Health Organization; Rifampicin Resistance: Xpert MTB/RIF System for the Diagnosis of
2008. Pulmonary and Extrapulmonary TB in Adults and Children: Policy Update.
112. World Health Organization. Automated Real Time Nucleic Acid Geneva, Switzerland: World Health Organization; 2013.
Amplification Technology for Rapid and Simultaneous Detection of 139. Nhu NT, Heemskerk D, Thu do DA, et al. Evaluation of GeneXpert
Tuberculosis and Rifampicin Resistance: Expert MTB/RIF system. Policy MTB/RIF for diagnosis of tuberculous meningitis. J Clin Microbiol. 2014;
Statement. Geneva: World Health Organization; 2011. 52(1):226233.
113. Takiff H, Guerrero E. Current prospects for the fluoroquinolones as 140. Kasanmoentalib ES, Brouwer MC, van der Ende A, et al. Hydrocephalus
first-line tuberculosis therapy. Antimicrob Agents Chemother. 2011; in adults with community-acquired bacterial meningitis. Neurology.
55(12):54215429. 2010;75(10):918923.
114. Pranger AD, Alffenaar JW, Aarnoutse RE. Fluoroquinolones, the corner- 141. Schoeman JF, Van Zyl LE, Laubscher JA, et al. Serial CT scanning in child-
stone of treatment of drug-resistant tuberculosis: a pharmacokinetic and hood tuberculous meningitis: prognostic features in 198 cases. J Child
pharmacodynamic approach. Curr Pharm Des. 2011;17(27):29002930. Neurol. 1995;10(4):320329.
115. Centers for Disease Control and Prevention. Targeted Tuberculin Testing 142. Figaji AA, Fieggen AG, Peter JC. Air encephalography for hydrocephalus
and Treatment of Latent Tuberculosis Infection. Atlanta: Centers for in the era of neuroendoscopy. Childs Nerv Syst. 2005;21(7):559565.
Disease Control and Prevention; 2000. 143. Bernaerts A, Vanhoenacker FM, Parizel PM, et al. Tuberculosis of the cen-
116. Shingadia D. The diagnosis of tuberculosis. Pediatr Infect Dis J. 2012; tral nervous system: overview of neuroradiological findings. Eur Radiol.
31(3):302305. 2003;13(8):18761890.

Scheld_Ch29.indd 498 2/21/14 8:21 PM


Chapter 29: Tuberculous Meningitis 499

144. Garcia-Monco JC. Central nervous system tuberculosis. Neurol Clin. 172. Peloquin CA, Hadad DJ, Molino LP, et al. Population pharmacokinetics
1999;17(4):737759. of levofloxacin, gatifloxacin, and moxifloxacin in adults with pulmonary
145. Uysal G, Kse G, Gven A, et al. Magnetic resonance imaging in diagnosis tuberculosis. Antimicrob Agents Chemother. 2008;52(3):852857.
of childhood central nervous system tuberculosis. Infection. 2001; 173. Morlock GP, Metchock B, Sikes D, et al. ethA, inhA, and katG loci of
29(3):148153. ethionamide-resistant clinical Mycobacterium tuberculosis isolates.
146. Janse van Rensburg P, Androniku S, van Toorn R, et al. Magnetic reso- Antimicrob Agents Chemother. 2003;47(12):37993805.
nance imaging of miliary tuberculosis of the central nervous system in 174. Zhang Y, Yew WW. Mechanisms of drug resistance in Mycobacterium
children with tuberculous meningitis. Pediatr Radiol. 2008;38(12): tuberculosis. Int J Tuberc Lung Dis. 2009;13(11):13201330.
13061313. 175. Andries K, Verhasselt P, Guillemont J, et al. A diarylquinoline drug ac-
147. Villoria MF, Fortea F, Moreno S, et al. MR imaging and CT of central tive on the ATP synthase of Mycobacterium tuberculosis. Science.
nervous system tuberculosis in the patient with AIDS. Radiol Clin North 2005;307(5707):223227.
Am. 1995;33(4):805820. 176. Diacon AH, Donald PR, Pym A. Randomized pilot trial of eight weeks
148. Kalita J, Prasad S, Maurya PK, et al. MR angiography in tuberculous of bedaquiline (TMC207) treatment for multidrug-resistant tuberculosis:
meningitis. Acta Radiol. 2012; 53(3):324329. long-term outcome, tolerability, and effect on emergence of drug resis-
149. American Thoracic Society, Centers for Disease Control and Prevention, tance. Antimicrob Agents Chemother. 2012;56(6):32713276.
Infectious Diseases Society of America. Treatment of tuberculosis. 177. Veziris N, Ibrahim M, Lounis N, et al. Sterilizing activity of second-line
MMWR Recomm Rep. 2003;52(RR-11):177. regimens containing TMC207 in a murine model of tuberculosis. PLoS
150. World Health Organization. Global Tuberculosis Control. Geneva: World One. 2011;6(3):e17556.
Health Organization; 2010. 178. Rustomjee R, Diacon AH, Allen J, et al. Early bactericidal activity and
151. World Health Organization. Treatment of Tuberculosis: Guidelines for pharmacokinetics of the diarylquinoline TMC207 in treatment of pulmo-
National Programmes. Geneva: World Health Organization; 2010:108. nary tuberculosis. Antimicrob Agents Chemother. 2008;52(8):28312835.
152. Berning SE, Cherry TA, Iseman MD. Novel treatment of meningitis 179. Schoeman JF, Van Zyl LE, Laubscher JA, et al. Effect of corticosteroids on
caused by multidrug-resistant Mycobacterium tuberculosis with intrathe- intracranial pressure, computed tomographic findings, and clinical outcome in
cal levofloxacin and amikacin: case report. Clin Infect Dis. 2001;32(4): young children with tuberculous meningitis. Pediatrics. 1997;99(2):226231.
643646. 180. Prasad K, Singh MB. Corticosteroids for managing tuberculous meningi-
153. Rastogi N, Labrousse V, Goh KS. In vitro activities of fourteen antimi- tis. Cochrane Database Syst Rev. 2008;(1):CD002244.
crobial agents against drug susceptible and resistant clinical isolates of 181. Tobin D, Roca FJ, Oh SF, et al. Host genotype-specific therapies can
Mycobacterium tuberculosis and comparative intracellular activities optimize the inflammatory response to mycobacterial infections. Cell.
against the virulent H37Rv strain in human macrophages. Curr Microbiol. 2012;148(3):434446.
1996;33(3):167175. 182. Klausner JD, Freedman VH, Kaplan G. Thalidomide as an anti-TNF-alpha
154. Donald PR, Diacon AH. The early bactericidal activity of anti-tubercu- inhibitor: implications for clinical use. Clin Immunol Immunopathol.
losis drugs: a literature review. Tuberculosis (Edinb). 2008;88(suppl 1): 1996;81(3):219223.
S75S83. 183. Tsenova L, Sokol K, Freedman VH, et al. A combination of thalidomide
155. Barling RW, Selkon JB. The penetration of antibiotics into cerebrospinal plus antibiotics protects rabbits from mycobacterial meningitis-associated
fluid and brain tissue. J Antimicrob Chemother. 1978;4(3):203227. death. J Infect Dis. 1998;177(6):15631572.
156. Mitchison DA. Role of individual drugs in the chemotherapy of tubercu- 184. Tsenova L, Mangaliso B, Muller G, et al. Use of IMiD3, a thalidomide
losis. Int J Tuberc Lung Dis. 2000;4(9):796806. analog, as an adjunct to therapy for experimental tuberculous meningitis.
157. Donald PR. Cerebrospinal fluid concentrations of antituberculosis agents Antimicrob Agents Chemother. 2002;46(6):18871895.
in adults and children. Tuberculosis (Edinb). 2010;90(5):279292. 185. Gori A, Rossi MC, Marchetti G, et al. Clinical and immunological benefit
158. Ellard GA, Humphries MJ, Allen BW. Cerebrospinal fluid drug concentra- of adjuvant therapy with thalidomide in the treatment of tuberculosis dis-
tions and the treatment of tuberculous meningitis. Am Rev Respir Dis. ease. AIDS. 2000;14(12):18591861.
1993;148(3):650655. 186. Schoeman JF, Springer P, van Rensburg AJ, et al. Adjunctive thalidomide
159. Donald PR, Parkin DP, Seifart HI, et al. The influence of dose and therapy for childhood tuberculous meningitis: results of a randomized
N-acetyltransferase-2 (NAT2) genotype and phenotype on the pharma- study. J Child Neurol. 2004;19(4):250257.
cokinetics and pharmacodynamics of isoniazid. Eur J Clin Pharmacol. 187. Abdool Karim SS, Naidoo K, Grobler A, et al. Integration of antiretroviral
2007;63(7):633639. therapy with tuberculosis treatment. N Engl J Med. 2011;365(16):14921501.
160. Ruslami R, Ganiem AR, Dian S, et al. Intensified regimen containing 188. Blanc FX, Sok T, Laureillard D, et al. Earlier versus later start of antiret-
rifampicin and moxifloxacin for tuberculous meningitis: an open-label, roviral therapy in HIV-infected adults with tuberculosis. N Engl J Med.
randomised controlled phase 2 trial. Lancet Infect Dis. 2012;13(1): 2011;365(16):14711481.
2735. 189. Torok ME, Farrar JJ. When to start antiretroviral therapy in HIV-
161. Heemskerk D, Day J, Chau TT, et al. Intensified treatment with high dose associated tuberculosis. N Engl J Med. 2011;365(16):15381540.
rifampicin and levofloxacin compared to standard treatment for adult pa- 190. Marais SS, Meintjes G, Pepper DJ, et al. Frequency, severity, and predic-
tients with tuberculous meningitis (TBM-IT): protocol for a randomized tion of tuberculous meningitis immune reconstitution inflammatory syn-
controlled trial. Trials. 2011;12:25. drome. Clin Infect Dis. 2013;56(3):450460.
162. Tappero JW, Bradford WZ, Agerton TB, et al. Serum concentrations 191. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for
of antimycobacterial drugs in patients with pulmonary tuberculosis in the Use of Antiretroviral Agents in HIV-1-Infected Adults and Adolescents.
Botswana. Clin Infect Dis. 2005;41(4):461469. Washington, DC: U.S. Department of Health and Human Services; 2012:240.
163. Peloquin CA, Jaresko GS, Yong CL, et al. Population pharmacokinetic 192. Robson, SC, White NW, Aronson I, et al. Acute-phase response and
modeling of isoniazid, rifampin, and pyrazinamide. Antimicrob Agents the hypercoagulable state in pulmonary tuberculosis. Br J Haematol.
Chemother. 1997;41(12):26702679. 1996;93(4):943949.
164. van Crevel R, Alisjahbana B, de Lange WC, et al. Low plasma concentra- 193. Schoeman J, Mansvelt E, Springer P, et al. Coagulant and fibrinolytic sta-
tions of rifampicin in tuberculosis patients in Indonesia. Int J Tuberc Lung tus in tuberculous meningitis. Pediatr Infect Dis J. 2007;26(5):428431.
Dis. 2002;6(6):497502. 194. Schoeman JF, Janse van Rensburg A, Laubscher JA, et al. The role of aspirin
165. Ruslami R, Nijland H, Aarnoutse R, et al. Evaluation of high- versus in childhood tuberculous meningitis. J Child Neurol. 2011;26(8):956962.
standard-dose rifampin in Indonesian patients with pulmonary tuberculo- 195. Misra UK, Kalita J, Nair PP. Role of aspirin in tuberculous meningitis: a
sis. Antimicrob Agents Chemother. 2006;50(2):822883. randomized open label placebo controlled trial. J Neurol Sci. 2010;293
166. Zhang y, Wade MM, Scorpio A, et al. Mode of action of pyrazinamide: (12):1217.
disruption of Mycobacterium tuberculosis membrane transport and 196. Figaji AA, Sandler SI, Fieggen AG, et al. Continuous monitoring and in-
energetics by pyrazinoic acid. J Antimicrob Chemother. 2003;52(5): tervention for cerebral ischemia in tuberculous meningitis. Pediatr Crit
790795. Care Med. 2008;9(4):e25e30.
167. Ellard GA, Humphries MJ, Gabriel M, et al. Penetration of pyrazinamide 197. Figaji AA, Fieggen AG. The neurosurgical and acute care management
into the cerebrospinal fluid in tuberculous meningitis. Br Med J (Clin Res of tuberculous meningitis: evidence and current practice. Tuberculosis
Ed). 1987;294(6567):284285. (Edinb). 2010;90(6):393400.
168. Jassal M, Bishai WR. Extensively drug-resistant tuberculosis. Lancet 198. Rajshekhar V. Management of hydrocephalus in patients with tubercu-
Infect Dis. 2009;9(1):1930. lous meningitis. Neurol India. 2009;57(4):368374.
169. Johnson JL, Hadad DJ, Boom WH, et al. Early and extended early bacteri- 199. Murthy JM. Management of intracranial pressure in tuberculous menin-
cidal activity of levofloxacin, gatifloxacin and moxifloxacin in pulmonary gitis. Neurocrit Care. 2005;2(3):306312.
tuberculosis. Int J Tuberc Lung Dis. 2006; 10(6):605612. 200. Bhatia AL, Csillag A, Mitchison DA, et al. The virulence in the guinea-pig
170. Thwaites GE, Bhavnani SM, Chau TT, et al. Randomized pharmacokinetic of tubercle bacilli isolated before treatment from South Indian patients
and pharmacodynamic comparison of fluoroquinolones for tuberculous with pulmonary tuberculosis. 2. Comparison with virulence of tubercle
meningitis. Antimicrob Agents Chemother. 2011;55(7):32443253. bacilli from British patients. Bull World Health Organ. 1961;25:313322.
171. Alffenaar JW, van Altena R, Bkkerink HJ, et al. Pharmacokinetics of 201. Naganathan N, Mahadev B, Challu VK, et al. Virulence of tubercle ba-
moxifloxacin in cerebrospinal fluid and plasma in patients with tubercu- cilli isolated from patients with tuberculosis in Bangalore, India. Tubercle.
lous meningitis. Clin Infect Dis. 2009;49(7):10801082. 1986;67(4):261267.

Scheld_Ch29.indd 499 2/21/14 8:21 PM


500 Part III: Bacterial and Mycoplasmal Infections

202. Coscolla M, Gagneux S Does M. tuberculosis genomic diversity explain 215. Faksri K, Drobniewski F, Nikolayevskyy V, et al. Epidemiological trends
disease diversity? Drug Discov Today Dis Mech. 2010;7(1):e43e59. and clinical comparisons of Mycobacterium tuberculosis lineages in Thai
203. Comas I, Gagneux S. A role for systems epidemiology in tuberculosis TB meningitis. Tuberculosis (Edinb). 2011;91(6):594600.
research. Trends Microbiol. 2011;19(10):492500. 216. Click ES, Moonan PK, Winston CA, et al. Relationship between
204. Brites D, Gagneux S. Old and new selective pressures on Mycobacterium Mycobacterium tuberculosis phylogenetic lineage and clinical site of
tuberculosis. Infect Genet Evol. 2011;12(4):678685. tuberculosis. Clin Infect Dis. 2012;54(2):211219.
205. Cole ST, Brosch R, Parkhill J, et al. Deciphering the biology of 217. Glynn JR, Alghamdi S, Mallard K, et al. Changes in Mycobacterium
Mycobacterium tuberculosis from the complete genome sequence. Nature. tuberculosis genotype families over 20 years in a population-based study
1998;393(6685):537544. in Northern Malawi. PLoS One. 2010;5(8):e12259.
206. Sreevatsan S, Pan X, Stockbauer KE, et al. Restricted structural gene 218. Hasan Z, Tanveer M, Kanji A, et al. Spoligotyping of Mycobacterium
polymorphism in the Mycobacterium tuberculosis complex indicates tuberculosis isolates from Pakistan reveals predominance of Central
evolutionarily recent global dissemination. Proc Natl Acad Sci U S A. Asian Strain 1 and Beijing isolates. J Clin Microbiol. 2006;44(5):
1997;94(18):98699874. 17631768.
207. Chatterjee A, DSouza D, Vira T, et al. Strains of Mycobacterium tubercu- 219. Awomoyi AA, Marchant A, Howson JM, et al. Interleukin-10, polymor-
losis from western Maharashtra, India, exhibit a high degree of diversity phism in SLC11A1 (formerly NRAMP1), and susceptibility to tuberculo-
and strain-specific associations with drug resistance, cavitary disease, and sis. J Infect Dis. 2002;186(12):18081814.
treatment failure. J Clin Microbiol. 2010;48(10):35933599. 220. Bellamy R. Identifying genetic susceptibility factors for tuberculosis
208. Hesseling AC, Marais BJ, Kirchner HL, et al. Mycobacterial genotype in Africans: a combined approach using a candidate gene study and a
is associated with disease phenotype in children. Int J Tuberc Lung Dis. genome-wide screen. Clin Sci (Lond). 2000;98(3):245250.
2010;14(10):12521258. 221. Bellamy R. Susceptibility to mycobacterial infections: the importance of
209. Nahid P, Bliven EE, Kim EY, et al. Influence of M. tuberculosis lineage host genetics. Genes Immun. 2003;4(1):411.
variability within a clinical trial for pulmonary tuberculosis. PLoS One. 222. Spira A, Carroll JD, Liu G, et al. Apoptosis genes in human alveolar
2010;5(5):e10753. macrophages infected with virulent or attenuated Mycobacterium tuber-
210. van Crevel R, Parwati I, Sahiratmadja E, et al. Infection with culosis: a pivotal role for tumor necrosis factor. Am J Respir Cell Mol
Mycobacterium tuberculosis Beijing genotype strains is associated with Biol. 2003;29(5):545551.
polymorphisms in SLC11A1/NRAMP1 in Indonesian patients with tuber- 223. Kleinnijenhuis J, Oosting M, Joosten LA, et al. Innate immune recognition
culosis. J Infect Dis. 2009;200(11): 16711674. of Mycobacterium tuberculosis. Clin Dev Immunol. 2011;2011:12.
211. Visser ME, Stead MC, Walzl G, et al. Baseline predictors of sputum culture 224. Dissanayeke SR, Levin S, Pienaar S, et al. Polymorphic variation in TIRAP
conversion in pulmonary tuberculosis: importance of cavities, smoking, is not associated with susceptibility to childhood TB but may determine
time to detection and W-Beijing genotype. PLoS One. 2012;7(1):e29588. susceptibility to TBM in some ethnic groups. PLoS One. 2009;4(8):e6698.
212. Parwati I, van Crevel R, van Soolingen D. Possible underlying mecha- 225. Hawn TR, Dunstan SJ, Thwaites GE, et al. A polymorphism in Toll-
nisms for successful emergence of the Mycobacterium tuberculosis Beijing interleukin 1 receptor domain containing adaptor protein is associated
genotype strains. Lancet Infect Dis. 2010;10(2):103111. with susceptibility to meningeal tuberculosis. J Infect Dis. 2006;194(8):
213. Caws Thwaites G, Stepniewska K, et al. Beijing genotype of Mycobacterium 11271134.
tuberculosis is significantly associated with human immunodeficiency 226. Kaufmann SH, Hussey G, Lambert PH New vaccines for tuberculosis.
virus infection and multidrug resistance in cases of tuberculous meningi- Lancet. 2010;375(9731):21102119.
tis. J Clin Microbiol. 2006;44(11):39343939. 227. Tameris MD, Hatherill, M, Landry BS. Safety and efficacy of MVA85A,
214. Caws M, Thwaites G, Dunstan S, et al. The influence of host and bacterial a new tuberculosis vaccine, in infants previously vaccinated with
genotype on the development of disseminated disease with Mycobacterium BCG: a randomised, placebo-controlled phase 2b trial. Lancet. 2013;
tuberculosis. PLoS Pathog. 2008;4(3):e1000034. 381(9871):10211028.

Scheld_Ch29.indd 500 2/21/14 8:21 PM


CHAPTER 30 INFECTIONS DUE TO
NONTUBERCULOUS MYCOBACTERIA
JEANA L. BENWILL AND RICHARD J. WALLACE, JR.

Mycobacteria other than Mycobacterium tuberculosis have caused disseminated disease in patients with advanced HIV
established themselves as human pathogens. By 1960, most infection before the advent of effective antiretroviral therapy
of the common species of nontuberculous mycobacteria (ART). Among the slowly growing mycobacteria: MAC,
(NTM) had been described, including Mycobacterium kansa- M. kansasii, M. genavense, M. malmoense, and M. simiae
sii, Mycobacterium avium, Mycobacterium intracellulare, and have been identified with central nervous system (CNS) dis-
Mycobacterium fortuitum (1). More than 140 species have now ease. In addition to the slowly growing NTM, the RGM
been recognized and characterized in the genus Mycobacterium organisms that have been associated with CNS disease have
(25), more than 70 of them in the past 20 years (4,5). Much been M. fortuitum group (primarily M. fortuitum) and the
of this taxonomic explosion relates to the use and availability M. abscessus complex. This chapter summarizes the epidemi-
of 16S ribosomal (rRNA) gene sequencing, 65 kDa heat shock ology, diagnosis, and treatment of each of the NTM reported
protein (hsp65), -subunit of RNA polymerase (rpoB), and to cause CNS disease.
large public DNA databases (25).
Two general groups of NTM can be delineated on the
basis of microbiologic, clinical, and epidemiologic character- GENERAL OVERVIEW
istics: the slowly growing and rapidly growing mycobacteria
(RGM). Characteristics of the species included in these groups NTM are generally free-living organisms that are ubiquitous
are shown in Table 30.1 (see later discussion). The incidence in the environment. They have been recovered from surface
of disease due to NTM has increased substantially in the past water, household water and biofilms, soil, and domestic and
20 years, in part because of better knowledge of its diseases, wild animals (16). Studies have demonstrated that mycobac-
in part because of improved laboratory techniques, and in teria easily become aerosolized from aqueous sources and that
part because of the pandemic of human immunodeficiency more easily aerosolized strains also more commonly cause pul-
virus (HIV). The most commonly recovered species or spe- monary infections.
cies complex is M. avium complex (MAC), which commonly During the 1950s, the ability of NTM to cause human disease
became increasingly clear as a consequence of several landmark
publications. In 1959, Runyon (7) proposed a classification sys-
tem that divided human isolates of NTM into four groups on
TA B L E 3 0 . 1 the basis of growth rates, colony morphology, and pigmentation
CLASSIFICATION OF MYCOBACTERIAL SPECIES in the presence and absence of light. This allowed mycobacterial
COMMONLY CAUSING HUMAN DISEASE laboratories to more readily identify individual species of NTM,
allowing clearer characterization of distinct diseases and syn-
Mycobacterium tuberculosis complex dromes associated with these organisms. In 1979, Wolinsky (1)
Mycobacterium tuberculosis
published an exhaustive review of the clinical and laboratory
features of these organisms. This provided one of the first refer-
Mycobacterium bovis ence sources for physicians to review the clinical significance of
Mycobacterium africanum the wide variety of species grouped as NTM and the clinical dis-
Mycobacterium leprae ease syndromes they produce. The number of cases of NTM dis-
ease has dramatically increased, as has knowledge about these
Slowly growing mycobacteria
mycobacteria, in the more than 30 years since this publication
Mycobacterium kansasii appeared. Subsequent major reviews that have updated newer
Mycobacterium marinum species since the study by Wolinsky (1) in 1959 were by Wayne
Mycobacterium avium complex and Sramek (2) in 1992, Brown-Elliott and Wallace (3) in 2002,
and Tortoli (4,5) in 2003 and 2006.
Mycobacterium avium
The frequency of disease due to the different species of NTM
Mycobacterium intracellulare is unknown. By the early 1990s, laboratories informally reported
Mycobacterium malmoense that more than 50% of evaluated mycobacterial species were
Mycobacterium simiae NTM. Much of this increase is related to the high incidence of
disseminated MAC disease in patients with acquired immunode-
Mycobacterium ulcerans
ficiency syndrome (AIDS). Among 41,439 cases of AIDS reported
Mycobacterium xenopi to the Centers for Disease Control and Prevention (CDC) from
Rapidly growing mycobacteria 1981 to 1987, disseminated NTM infection occurred in 5.5%,
Mycobacterium fortuitum group and 96% of these were due to MAC (8). As a consequence, CNS
infections due to NTM have increased in frequency.
Mycobacterium chelonae
Prior to describing NTM infections of the CNS in patients
Mycobacterium abscessus complex with HIV, it may be useful to briefly review disseminated NTM
Mycobacterium mucogenicum infection in these patients. With the exception of rare cases
that might occur by direct extension or from trauma, infection

501

Scheld_Ch30.indd 501 2/21/14 5:45 PM


502 Part III: Bacterial and Mycoplasmal Infections

of the CNS is likely to occur from hematogenous dissemina- The authors attribute the increase of NTM disease during the
tion. Therefore, these patients may approximate the patient 2000 to 2003 period to increased awareness and diagnostic
population from which CNS disease arises. Horsburgh and consideration of NTM (12).
Selik (8) summarized the epidemiology of disseminated NTM The review by Wolinsky (1) serves as a convenient and au-
infection in the pre-ART era in the United States from 1981 thoritative starting point for a discussion of NTM infections
to 1987 based on data reported to the CDC. As stated pre- of the CNS prior to 1979. One case each of meningitis due to
viously, disseminated NTM infection was reported in 5.5% M. kansasii, MAC, and M. gordonae were described. The pa-
(2,269 cases) of patients with AIDS over this 7-year period and tient with M. kansasii was a 34-year-old woman who died of
96% of these were MAC. The remaining 4% included M. kan- disseminated infection. The patient with MAC was a 13-year-
sasii (2.9%), M. gordonae (0.6%), and M. fortuitum complex old boy with disseminated disease who had MAC isolated from
(0.3%). The number of patients with NTM disease increased CSF and had granulomatous meningitis on postmortem exami-
in parallel with the incidence of AIDS, and the overall propor- nation. The patient with M. gordonae infection was a hydroce-
tion in the pre-ART era remained nearly constant at 5.5%. phalic infant who had undergone multiple shunt procedures with
Significant regional variations in this figure, from 3.9% to infection of the peritoneal fluid and CSF. Wolinsky (1) also men-
7.8%, were noted. NTM disease occurs primarily in patients tioned one case of MAC meningitis from his own experience.
with AIDS, generally in patients with fewer than 50 CD4 In addition, he noted two instances in which other pathogens
cells/L (9). Patients with disseminated NTM infection had were also present: M. kansasii was isolated from the meninges at
significantly shorter survival (median, 7.4 months) than pa- postmortem examination in a 2-year-old with miliary and men-
tients with AIDS without disseminated NTM infection (me- ingeal M. tuberculosis, and M. intracellulare was isolated from
dian, 13.3 months). Disseminated NTM infection is diagnosed the CSF of a 46-year-old woman with cryptococcal meningitis
after AIDS in nearly one half (47.2%) of patients. In short, the (a subsequent smear of the CSF for acid-fast bacilli [AFB] was
early AIDS epidemic has been accompanied by an epidemic of positive, but cultures were negative with the patient receiving
disseminated NTM disease as severely immunocompromised chemotherapy). Because two pathogens were isolated, the role
patients were exposed to these ubiquitous but relatively less of NTM in these patients diseases was difficult to determine.
virulent organisms. With the advent of ART, however, the re- The following sections of this chapter review the epidemiol-
covery rate of MAC and other NTM in patients with AIDS has ogy, microbiology, and chemotherapy of the NTM implicated
fallen dramatically since the mid to late 1990s. in CNS disease and summarizes reported cases to date. The
In the absence of HIV infection, on the other hand, the dis- chapter is organized by species of mycobacterium divided into
tribution of infections due to the different NTM species was re- slowly and rapidly growing species. Several case series de-
ported by Swiss investigators who reviewed all 513 HIV-negative scribed more than one species. Investigators from Brooklyn,
patients from whom NTM were isolated at the University of New York described 11 years of experience at Kings County
Zurich from 1983 to 1988 (9). In this study, 34 patients were Hospital with NTM from 1980 through 1990 infections of the
thought to have clinical disease due to these infections, 21 had CNS in patients with AIDS (13). Sixteen CSF cultures were
pulmonary disease, 10 had soft tissue disease, and 1 had dis- positive for NTM. Fifteen grew MAC and one grew M. fortui-
seminated disease. There were no instances of CNS involve- tum group. Three patients concurrently had CNS toxoplas-
ment. The most common isolates were MAC in 16 patients mosis. Ten of the fifteen died during hospitalization. Japanese
and M. kansasii in 9 patients. M. terrae complex, M. fortuitum workers accumulated 109 cases of NTM disease by nationwide
group, M. marinum, and M. malmoense were also involved. investigation from 1955 to 1965; 6 involved the CNS, but the
NTM disease in children before the AIDS era was reviewed NTM isolates were not speciated (14). A report from Malaysia
by Lincoln and Gilbert (10). In contrast to adults, cervical identified a 9-month-old admitted with meningitis and CSF
lymphadenitis due to M. scrofulaceum and MAC, as well as from which AFB with yellow-orange colonies were cultured
cutaneous disease due to M. marinum and M. ulcerans, pre- from a cisternal puncture but not from a lumbar puncture (15).
dominated. Twelve cases of disseminated disease were re- Wolinskys view of these reports raised the important issue of
ported. Six patients were reported with definite CNS disease acceptable criteria for the diagnosis and reporting of CNS dis-
and one additional case was highly probable. Of these seven ease due to NTM. Given a compatible clinical situation, isola-
patients, M. kansasii was the pathogen in three patients (one tion of multiple colonies of NTM in pure culture from repeated
was a dual infection with M. tuberculosis), scotochromogens specimens of CNS tissue or fluid would constitute strong evi-
were reported in two patients, and MAC in two patients. dence for NTM as a cause of the patients CNS disease (16).
Three patients were Asian and two patients each were from Clinical and microbiologic response to specific therapy and the
Europe and the United States. Cerebrospinal fluid (CSF) re- absence of other pathogens would virtually seal the diagnosis.
sults reported in five of seven patients were typical of myco-
bacterial infection, that is, lymphocytic pleocytosis, elevated
protein level, and depressed glucose levels. One definite case, SLOWLY GROWING
however, had a neutrophilic pleocytosis. Treatment was not
detailed and four patients died. A 2-year prospective surveil-
NONTUBERCULOUS
lance study of children in Netherlands with NTM demon- MYCOBACTERIA
strated that MAC was the most common pathogen, but no
disseminated or CNS disease was reported (11). Jensen et al. Mycobacterium avium Complex
(12) conducted a retrospective review of mycobacterial dis-
ease in HIV-infected children in Spain from 1997 to 2008. Of
1,307 HIV-infected children, 42 had mycobacterial disease
Epidemiology
of which 22 had NTM. Nine had disseminated disease, but MAC includes ubiquitous, free-living organisms (1). MAC iso-
CNS involvement was not reported. Overall, a reduction of lates are readily recovered from natural reservoirs including
mycobacterial infection decreased from 5.9 to 1.6 events per soil and water, and organisms similar to those causing human
1,000 HIV-infected children-year. NTM disease rates were disease have been recovered from aerosols near natural waters.
2.3, 3.4, and 0.3 per 1,000 HIV-infected children-year for Recent environmental studies have shown that the major reser-
1997 to 1999, 2000 to 2002, and 2003 to 2008, respectively. voir for M. avium is household water and pipe biofilms but that

Scheld_Ch30.indd 502 2/21/14 5:45 PM


Chapter 30: Infections Due to Nontuberculous Mycobacteria 503

M. intracellulare is absent and presumably acquired from envi- description of AIDS from Los Angeles (25), all five eventually
ronmental exposure outside the household (17). The geographic developed disseminated MAC disease and two of the five had
distribution of the organism, however, is not uniform. A survey CNS disease due to MAC (26).
of skin-test reactivity within the United States to an antigen pre- Hawkins et al. (27) reported the experience of the Memorial
pared from MAC was published in 1969 (18). The skin-test Sloan-Kettering Cancer Center with disseminated MAC infec-
reagent, called PPD-B or Battey antigen (Battey State Hospital, tion in patients with AIDS from 1981 to 1984. Of 366 patients
Rome, Georgia), was applied to approximately 275,000 naval with AIDS, 67 (18.3%) were diagnosed with disseminated MAC
recruits who had lived their entire lives in a single county. Skin- infection. CNS involvement was not identified before death in
test reactivity was most common among recruits from the any of these patients. Autopsies were performed in 45 of these
southeastern and Gulf Coast states. In these areas, more than patients, and 12 (39%) of 31 whose brain tissue was cultured
70% of individuals had been exposed to or infected with MAC had positive cultures for MAC from brain tissue. In another re-
or an antigenically similar organism. MAC lung disease appears port from the same institution focusing on the neuropathology
to be relatively more prevalent in these same areas. of AIDS, Snider et al. (28) described 50 patients (of 160 patients
The incidence of MAC infections in selected groups of HIV- with AIDS from 1980 to 1983) who developed neurologic dys-
infected patients before the advent of effective ART was measured function. Twenty patients had complete autopsies. MAC was
by Nightingale et al. (9). They followed 1,006 patients at one cultured from the brains of two patients with disseminated
institution during a 3-year period with monthly blood cultures MAC disease, and AFB were seen microscopically in the brain
from the date of AIDS diagnosis and determined the incidence of tissue in one of these. AFB were seen in brain tissue of a third
MAC bacteremia to be 21% at 1 year and 43% at 2 years (9). patient with disseminated MAC disease, but cultures were nega-
The incidence of MAC bacteremia was inversely related to CD4 tive. These patients had impaired cognition, psychomotor retar-
lymphocyte counts, occurring in only 3% of patients with CD4 dation, fevers, sweats, anorexia, malaise, and gastrointestinal
lymphocytes of 100 to 199 cells/L increasing progressively to complaints. CSF showed a mild pleocytosis (7 to 36 white blood
39% for patients with fewer than 10 CD4 lymphocytes/L. cells [WBCs]/L, 90% polymorphonuclear [PMN] in one pa-
tient, and 70% lymphocytes in a second) but normal protein and
Pathogenesis glucose concentrations. Pathologically, two had gray and white
matter changes of subacute encephalitis with nodular collections
MAC is likely to disseminate to the CNS hematogenously from
of microglial cells, without inflammatory changes and reactive
a respiratory or gastrointestinal source. In rare cases, CNS dis-
astrocytosis and small foci of demyelination in the white matter.
ease may occur following trauma, direct extension from an
Levy and Bredesen (29) reviewed the neurologic manifesta-
adjacent focus of infection such as the paranasal or mastoid
tions of 1,286 patients with AIDS in San Francisco, of whom
sinuses and with implantable foreign material (1922).
474 had diseases affecting the CNS. Included were four patients
Early experience with MAC disease in HIV-infected pa-
with MAC infection of the CNS. Computed tomographic (CT)
tients indicated that heavy bacteremia, widespread disease,
scan of the head revealed single hypodense lesions in two cases, a
and a high tissue burden of organisms were typical. Subsequent
single contrast-enhancing lesion in one, and no abnormalities in
study suggests that these early observations focused on patients
one. Biopsy of the contrast-enhancing lesion revealed toxoplas-
with relatively far advanced disease. Torriani et al. (23) studied
mosis and lymphoma, in addition to MAC infection. In the CSF,
44 patients with MAC bacteremia who eventually had com-
there was a mild pleocytosis (12 to 15 WBCs/L) and mild el-
plete autopsies over 5 years in California. No tissue involve-
evations of protein but normal glucose values. CSF culture grew
ment could be identified in 30% of patients, whereas 70% of
MAC in two patients. Brain biopsy yielded MAC in one patient,
patients had one to nine different tissue sites involved (median
and postmortem examination identified MAC in the CNS in the
of four). CNS involvement was seen in one patient who had
fourth patient. In reviewing the published literature to that point,
both cerebral and spinal cord infection with MAC. The likeli-
the authors identified 14 cases of CNS infection with MAC.
hood of detectable tissue involvement increased directly in pro-
Most patients had disseminated MAC infection before present-
portion to the length of survival after the first positive blood
ing with diffuse encephalitis. Meningitis, cranial neuropathy, and
culture: 43% in patients dying within 2 months, 72% among
peripheral neuropathy were also reported in association with
patients dying in 2 to 10 months, and 90% among patients
MAC infection of the CNS. Survival was uniformly poor.
dying after 10 months. This suggested that mycobacteremia
In these severely immunocompromised patients, more than
preceded disseminated MAC disease. These workers hypoth-
one opportunistic pathogen may be isolated from the CNS es-
esized that mycobacteria infect a mucosal surface (gut or lung),
pecially in the era of more sensitive tests like polymerase chain
multiply locally, and eventually enter the bloodstream and dis-
reaction (PCR) assays. Two AIDS patients were reported by
seminate, seeding other organs and tissues. CNS involvement is
Sharma et al. (32) in which one patient had altered sensorium,
likely to be a natural consequence of this pathogenic sequence.
fever, and meningismus who in the CSF had 20 WBCs/L
Some understanding of the pathogenesis of CNS infection by
(100% lymphocytes) and mild elevation of protein with nor-
MAC in immunocompromised patients is provided by a murine
mal glucose. CSF culture and multiplex PCR grew M. avium
model reported by Wu et al. (24). Following intravenous ad-
and M. tuberculosis. The second patient had prior pulmonary
ministration of M. avium, numerous mycobacteria were pres-
tuberculosis (TB) and had severe headache. CSF contained
ent in macrophages within granulomatous lesions in the brain
110 WBCs/L (70% lymphocytes), 600 mg/dL protein, and
by 6 months. Interestingly, none of the mice exhibited clinical
glucose 63 mg/dL. Cultures of CSF were negative for M. avium
signs of meningitis or encephalitis, a finding similar to many
and M. tuberculosis, but multiplex PCR was positive for both
HIV-infected patients with CNS involvement with M. avium.
species. In addition, CSF cryptococcal antigen was positive.
Mycobacterium avium Complex Infection of the At Kings County Hospital in New York, Jacob et al. (13)
reviewed all positive CSF cultures for NTM over 11 years,
Central Nervous System in HIV-Infected Patients from 1980 to 1990. Of the 16 patients with positive cultures,
CNS disease due to MAC in HIV-infected patients has been 14 occurred from 1987 to 1990, 15 were MAC, and 1 was
documented through case reports, case series, and reviews of M. fortuitum group (see later discussion). Twelve patients had
medical records associated with laboratory isolates of MAC a previous diagnosis of AIDS and the remaining four had at
(Table 30.2). Interestingly, of the five patients in the initial least one major risk factor for HIV infection. All 15 patients

Scheld_Ch30.indd 503 2/21/14 5:45 PM


504 Part III: Bacterial and Mycoplasmal Infections

TA B L E 3 0 . 2
REPORTED MYCOBACTERIUM AVIUM COMPLEX INFECTIONS OF THE CENTRAL NERVOUS SYSTEM IN
PATIENTS WITH AIDS

No. of Age State or


Reference Cases (yr) Sex Country Diagnosis Culture Source Source Population of Cases

23 1 NR NR CA Encephalomyelitis Blood, PM: 44 pts. with blood cultures positive for


brain, spinal MAC and autopsies at University of
cord California, San Diego, 19881992
26 2 30 M CA Meningoencephalitis CSF, PM: brain Initial report of AIDS from Los Angeles,
30 M CA NR PM: brain 5 pts.
27 12a NR NR NY NR PM: brain 67 pts. with disseminated MAC of
366 total pts. with AIDS at Sloan-
Kettering, 19811985, 12/45 with
PM cultures of brain but 0/50 with
cultures antemortem
28 3b 39 M NY Encephalitis PM 8 pts. with disseminated MAC of
50 pts. with CNS involvement at
Sloan-Kettering, 19801983
29 4 NR NR CA Encephalitis Brain biopsy 482 pts. with CNS involvement
NR NR CA Meningitis PM of 1,286 total pts. with AIDS
at University of California, San
NR NR CA Meningitis CSF
Francisco to 1986
NR NR CA Meningitis CSF
30 3 NR NR Italy Encephalitis NR 30 pts. who developed disseminated
MAC of 350 pts. followed
prospectively 19851993
31 1 NR NR France Rhombencephalitis CSFa Single case report
32 2 58, 38 M India Meningitis CSF Pts. with positive CSF for NTM and
TB. 1 pt. with positive CSF culture
for NTM and TB
33 2 NR NR CA NR CSF 100 consecutive MAC isolates from
89 pts. at San Francisco General
Hospital
13 15 33 4 9M:6F NY Meningitis CSF, PM 16 pts. with NTM isolated from CSF at
Kings County Hospital 19801990
34 1 38 M TX Meningitis CSF 32 pts. who had CSF cytology at M.D.
Anderson 19821986
35 2 Adult NR France Encephalitis PM 125 autopsied pts. at 1 hospital
19821986
36 2 Adult NR Zaire Meningitis CSF 43 pts. with CNS manifestations of 104
consecutive HIV-positive admitted
pts. in Kinshasa
37 1 42 M Germany Meningoencephalitis CSF Single case report
38 1 32 M Australia Epidural abscess Abscess Single case report
39 1 Adult Epidural abscess Single case report
40 2 Adult 1M:1F Spain Meningoencephalitis; CSF Pts. with positive CSF cultures for NTM
brain abscess between 19821994
41 1 Adult M Australia Brain abscess Brain Single case report
42 1 34 F France Brain abscess Brain biopsy Single case report
43 1 40 M France Brain abscess Brain biopsy Single case report
44 1 38 M Canada Brain abscess Brain biopsy Single case report
45 1 33 M Michigan Brain abscesses Brain biopsy Single case report
46 1 42 F India Brain abscess Abscess Single case report
47 1 23 M Taiwan Meningoencephalitis CSF Single case report
and myelitis

NR, not reported; PM, postmortem cultures and/or histopathology; pts., patients; M, male; pt., patient; TB, Mycobacterium tuberculosis; F, female.
a
Cytomegalovirus also isolated from CSF.
b
Cases may overlap.

Scheld_Ch30.indd 504 2/21/14 5:45 PM


Chapter 30: Infections Due to Nontuberculous Mycobacteria 505

FIGURE 30.1 Magnetic resonance scan of the brain and spinal cord demonstrating numerous tiny nod-
ules scattered diffusely in an AIDS patient with MAC isolated from CSF. (From Lee YC, Lu CL, Lai CC,
et al. Mycobacterium avium complex infectionrelated immune reconstitution inflammatory syndrome
of the central nervous system in an HIV-infected patient: case report and review. J Microbiol Immunol
Infect. 2013;46:6872, with permission.)

with MAC disease had widespread dissemination, and in 3 pa- an interesting case of diffuse nodules of the brain and spinal
tients, autopsies demonstrated extensive MAC disease of CNS, cord (Fig. 30.1) of a patient with AIDS whose treatment for
bone marrow, liver, gastrointestinal tract, and lymph nodes, disseminated MAC had been discontinued after immune re-
but no other CNS disease. constitution. MAC was isolated from blood and CSF cultures
The number of additional cases of CNS disease due to MAC and responded well with reinitiation of MAC drug therapy.
over the years has declined dramatically with better ART and Most cases in the literature describe one to three brain lesions,
the decline in cases of disseminated MAC (3346). However, but Verma and Dhamija (45) reported a case of disseminated
MAC involving the CNS has been increasingly reported with M. avium-intracellulare infection in an AIDS patient with mul-
immune reconstitution (41,42,44,47). Lee et al. (47) reported tiple brain lesions (Fig. 30.2).

FIGURE 30.2 Magnetic resonance scan of the brain with multiple ring-enhancing lesions with MAC isolated
from brain biopsy. (From Verma R, Dhamija R. Disseminated Mycobacterium avium-intracellulare infection
presenting as multiple ring-enhancing lesions on brain MRI. Mayo Clin Proc. 2009;84:394, with permission.)

Scheld_Ch30.indd 505 2/21/14 5:45 PM


506 Part III: Bacterial and Mycoplasmal Infections

Mycobacterium avium Complex Infection of the isolated from the CSF. The second patient had chronic myelog-
Central Nervous System Not Associated with enous leukemia (CML) in blast crisis and MAC was isolated in
pure culture with high colony counts on both slants.
HIV Infection These cases raise the critical issues of interpreting positive
MAC infection of the CNS has also been reported in the ab- CSF cultures of NTM and reasonable criteria to diagnose NTM
sence of HIV infection, both in patients with another immu- infection of the CNS. These authors suggest that to ascribe
nocompromising process and in patients without identifiable CNS disease to NTM, multiple colonies of the NTM should
immunologic deficits (Table 30.3). The frequency with which be demonstrated on repeated cultures of CSF in the absence of
this occurs is difficult to determine because it is relatively rare, other pathogens. None of these three patients met these crite-
generally not reported, and no surveillance system for NTM ria, but the high colony counts and pure culture of MAC from
infection exists. At Mt. Sinai Hospital in New York, mycobac- the single CSF specimen of the second patient make contamina-
teria were recovered from CSF specimens from 21 patients be- tion unlikely. However, these criteria may focus too narrowly
tween 1970 and 1983 (16). Three isolates were NTM and 19 on CSF alone as a specimen source. For example, isolation of
were M. tuberculosis. Two patients had high colony counts of MAC from a biopsy of brain, meninges, aspirate of a brain ab-
MAC recovered on both inoculated slants. The third had a sin- scess, or from a postmortem specimen in an appropriate clini-
gle colony of M. gordonae, probably a contaminant, isolated cal setting would be strong microbiologic evidence of MAC
from otherwise normal CSF. The first patient was previously disease. In addition, these criteria do not allow for the pos-
healthy and presented with meningitis clinically. Her CSF had sibility of dual infection with NTM and another pathogen. A
471 WBCs/L (84% lymphocytes), 234 mg/dL of protein, and case report illustrates these points (48). A 48-year-old woman
41 mg/dL of glucose. Both MAC and M. tuberculosis were presented with fever, a shift in her leukocyte differential toward

TA B L E 3 0 . 3
REPORTED MYCOBACTERIUM AVIUM COMPLEX INFECTIONS OF THE CENTRAL NERVOUS SYSTEM IN
PATIENTS WITHOUT HIV INFECTION

No. of State or Diagnosis


Reference Cases Age Sex Country (Culture Source) Comorbidity Source Population of Cases

1 1 NR NR U.S.A. Meningitis (NR) Immunosuppressed Single case reported in extensive


review of NTM
16 2 58 M New York Meningitis (CSF) CML in blast crisis, 21 pts. with mycobacterial
40 F New York Meningitis (CSF) M. tuberculosis meningitis (19 TB), Mt. Sinai
(CSF), previously Hospital, New York City
healthy 19701983
48 1 48 F U.S.A. Meningitis, brain C. neoformans (CSF), Single case report
abscess (CSF) previously healthy
49 1 31 F Switzerland Meningitis, brain Previously healthy Single case report
abscess (CSF) (? corticosteroids)
50 NR NR Colorado Meningitis (NR) Disseminateda MAC 13 pts. with disseminated MAC
at National Jewish Hospital,
Denver, 19401984
51 1 47 F Japan Meningitis (CSF) Disseminated MAC, Single case report
CML, pulmonary
alveolar proteinosis
52 1 50 M Brain abscess (tissue) Systemic lupus Single case report
53 1 38 M Brain abscess (tissue) Sarcoidosis Single case report
54 1 52 M U.S.A. Brain abscess (tissue) Lung carcinoma s/p Single case report
lobectomy and
radiotherapy
55 1 63 M Canada Brain abscess (tissue) Sarcoidosis Single case report
56 1 29 M Ohio Bone marrow, liver Hodgkin disease Single case report
57 1 73 F Japan Meningitis (CSF) Previously healthy Single case report
59 1 38 M U.S.A. Brain abscess (tissue) Sarcoidosis, Single case report
corticosteroids
107 1 5 F Denmark Meningitis (CSF) NR Single case report
107 1 13 M U.S.A. Meningitis (CSF, PM) Disseminated MAC Single case reported in textbook
on NTM

NR, not reported; M, male; F, female; PM, postmortem.


a
Granulomatous meningitis at PM examination.

Scheld_Ch30.indd 506 2/21/14 5:45 PM


Chapter 30: Infections Due to Nontuberculous Mycobacteria 507

immature forms, and a posterior fossa mass lesion. A ventricu- Four interesting cases of brain abscess mimicking spindle
loatrial shunt was placed and CSF from this procedure con- cell tumors have been described. Di Patre et al. (52) reported
tained 32 mg/dL of glucose, 38 mg/dL of protein, and 83 WBCs/ a 50-year-old patient with systemic lupus erythematosus who
L (all mononuclear). Two days later, CSF obtained by lumbar presented with a dural-based meningioma-like mass in the
puncture (opening pressure 300 mm) had 14 mg/dL of glucose, right frontal lobe. The mass resembled a meningioma with fas-
107 mg/dL of protein, and 44 WBCs/L (75% mononuclear). cicular arrangement of spindle cells without caseation or giant
India ink and culture demonstrated Cryptococcus neoformans. cells but by AFB stains consisted of large numbers of AFB in
The patient was treated with amphotericin B and 5-fluorocyto- histiocytes (52). A very similar case was reported by Morrison
sine and gradually improved. After 6 weeks, CSF from the orig- et al. (53) of a 38-year-old man with sarcoidosis who also
inal lumbar puncture grew M. intracellulare. Lumbar puncture presented with a spindle cell pseudotumor. It had minimal
was repeated and several AFB were seen on direct smear, but necrosis but was loaded with AFB. Arkun et al. (54) reported
no further cultures grew the organism. Isoniazid and rifampin a 52-year-old with history of lung carcinoma, left upper lobec-
were added to her therapeutic regimen and she eventually re- tomy, and radiotherapy with complex, multiloculated, ring-
covered. She was free of relapse at a 3-year follow-up but had enchancing cystic lesion of the left tentorium with compression
significant neurologic sequelae. The role of M. intracellulare in of the fourth ventricule resulting in hydrocephalus. The tis-
producing disease in this patient was difficult to determine, but sue consisted of spindle cells arranged in bands, fascicles, and
based on careful evaluation of the circumstances, the authors ill-defined nodules with a single giant cell. Ziehl-Neelsen and
concluded that the organism was unlikely to be a contaminant. Fite staining was positive, and MAC was identified by DNA
Another case of MAC infection of the CNS in a previously probe (54). Sadek et al. (55) described a case with underlying
healthy person was reported by Uldry et al. (49). A 31-year-old sarcoidosis with headache and word-finding difficulties with
woman presented with a 2-year history of progressive head- a left frontal ring-enchancing lesion. Tissue revealed multiple
aches that suddenly became worse associated with lethargy, foci of spindle cell pseudotumor formation with large num-
confusion, vomiting, and a grand mal seizure. On examination, ber of AFB. Similar pseudotumors have been described with
the patient had depressed consciousness, a stiff neck, decorti- M. tuberculosis in immunosuppressed patients, but these are
cate posturing, and bilateral flexor-plantar reflexes. CT of the some of the first to be described due to NTM.
head showed hydrocephalus due to compression of the fourth Gyure et al. (56) presented the case of a patient with un-
ventricle and meningeal enhancement with contrast. A ventric- derlying Hodgkin disease who presented with signs of me-
uloatrial shunt was placed and the CSF contained 453 WBCs/ ningoencephalitis that included seizures, confusion, nuchal
L (80% lymphocytes). The patient was treated with isoniazid, rigidity, and extensor-plantar responses. The patient died and
rifampin, and pyrazinamide for TB and steroids for sarcoid- at autopsy was found to have focal aggregates of lymphocytes,
osis, but all cultures were negative. Immunologic evaluation macrophages, and AFB located predominantly in a perivas-
was negative. The patient improved clinically, and repeated cular location. Cultures of CSF and brain were positive for
immunologic parameters, HIV serology, and chest radiography MAC. Okada and Yoshioka (57) reported a case of acute dis-
remained normal. Over the subsequent 2 years, however, the seminated encephalomyelitis associated with meningitis in a
patient fluctuated clinically, the CSF profile remained abnor- 73-year-old female who had fever, meningism, disorientation,
mal, and cultures were repeatedly negative. After 2 years, head- left eye eversion, muscle weakness, and ataxic gait. Magnetic
ache recurred and a left hemiparesis developed. Repeated CT resonance imaging (MRI) showed multifocal and asymmetric
scans showed a right temporal mass with surrounding edema. increased T2 signals of the white and cortical graywhite mat-
Craniotomy revealed a hemorrhagic necrotic mass, which on ter junction of cerebral hemispheres, cerebellum, and brain-
microscopic examination had granulomas, multinucleated giant stem. CSF contained WBCs of 1,295/L (60% PMNs) and
cells, and few AFB. The tissue grew MAC. This case is notewor- protein of 60 mg/dL with normal glucose. M. intracellulare
thy because it illustrates the difficulty of diagnosing and treating was isolated from CSF by PCR. The patient was treated with
MAC infection of the CNS. However, an alternative explana- rifampicin, streptomycin, clarithromycin, and steroids with
tion is possible, based on the long duration of disease before the resolution of symptoms.
diagnosis of MAC. The patient may have had an undiagnosed Immune defects involving interleukins (IL) 10 and 12 and
cause for her CNS disease initially and then developed a MAC tumor necrosis factor- (TNF-) and interferon- (IFN-) are
cerebral abscess as a consequence of chronic steroid therapy. risk factors for pulmonary and disseminated NTM. Browne
Additional cases of CNS infection due to MAC have been re- et al. (58) reported adult-onset immunodeficiency in Thai and
ported in the context of disseminated MAC disease by workers Taiwanese patients associated with autoantibodies to IFN-.
at National Jewish Hospital (Denver, Colorado)in a review One hundred five patients had NTM isolated, with 52 having
of 13 cases of disseminated MAC infection in HIV-negative disseminated disease, but no details of CNS involvement were
patients from 1940 through 1984 (50). The criteria for dissem- reported. Two cases of immunodeficiency with CNS NTM in-
inated disease by which they selected cases required isolation volvement have been reported. Dickerman et al. (59) reported
of MAC from at least one nonpulmonary site and microbio- a case of a 38-year-old male with sarcoidosis who underwent
logic or histopathologic evidence of granulomatous disease at MRI due to possible seizure activity which demonstrated right
a second anatomically distinct site. By this case definition, one frontal, left parietal, and right cerebellar lesions. M. avium was
patient with meningeal involvement was identified. No details cultured from resected frontal and parietal lesions. Despite
were provided. It is now presumed that all these patients had therapy, the cerebellar lesion increased in size and total resec-
a genetic abnormality that predisposed them to NTM disease. tion of the cerebellar mass was performed. Histopathology
One case of disseminated MAC infection with CNS in- showed encapsulated abscess with mixture of plasma cells,
volvement was reported by Japanese workers in a patient with lymphocytes, and macrophages with AFB. The patient was
pulmonary alveolar proteinosis and CML (51). A 47-year-old found to have low concentrations of IFN- and TNF-. The
woman with CML who had been treated with busulfan for case described by Sadek et al. (55) as previously discussed on
2 years developed a recurrent cough and had an abnormal chest further analysis showed TNF- and IL-12 defect in response
radiogram. AFB smears of expectorated sputum were positive. to MAC antigens but high levels of IFN-. These cases demon-
Transbronchial lung biopsy revealed pulmonary alveolar pro- strate the complex and fascinating IL, TNF, and IFN immune
teinosis. Cultures of sputum, bone marrow, and CSF grew MAC. response in NTM disease.

Scheld_Ch30.indd 507 2/21/14 5:45 PM


508 Part III: Bacterial and Mycoplasmal Infections

Laboratory with AIDS (6871) and in MAC lung disease in HIV-negative


patients (60,6365). Their usefulness and role in the therapy
MAC is readily identified in the laboratory by acid-fast smear of MAC disease of the CNS have not been studied, but they
and culture using techniques for CSF and tissue standard- offer exciting potential for therapy of a very difficult disease.
ized for TB. The organisms grow well in broth medium and Treatment of MAC lung disease is continued until cultures
on Middlebrook 7H10 agar, typically producing small, flat remain consistently negative for at least 12 months.
transparent smooth colonies on agar that often have a pale For the treatment of MAC in AIDS, a joint panel between
yellow color. Their colony morphology readily distinguishes the CDC, the National Institutes of Health, and the HIV
them from M. tuberculosis. Commercial nucleic acid probes Medicine Association of the Infectious Diseases Society of
(AccuProbe, Gen-Probe, Inc.) are available that identify America published recommendations (72). The basic princi-
MAC isolates with greater than 99% accuracy 1 day after the ples for therapy included the necessity of regimens containing
colonies have grown, a technique used in most laboratories. at least two drugs, one of which should be either clarithromy-
M. avium and M. intracellulare are separate species, but their cin or azithromycin. Most commonly, ethambutol and either
separation has no clinical value for the individual patient with rifampin or rifabutin would be the second and third drugs.
lung disease and, hence, is generally not done. Specific DNA Clofazimine was once considered among the possible agents
probes that recognize only M. avium or M. intracellulare are to treat these infections. However, studies now show increased
commercially available, however, MAC is also readily identi- mortality when this drug is used in this setting, so it should
fied by high-performance liquid chromatography (HPLC) by not be used for treatment of disseminated MAC. Treatment
analysis of mycolic acid patterns, a technique used by many should be continued for at least 12 months and discontinuing
large state laboratories and the CDC. treatment is based on the absence of ongoing MAC disease
Antimicrobial susceptibility testing of NTM based on the and appropriate and sustained immune response due to ART.
American Thoracic Society (ATS) and the 2011 Clinical and Treatment of CNS infection is complicated by the lack
Laboratory Standards Institute (CLSI) guidelines currently of therapeutic data and by the preference for parenteral
recommend reporting primary susceptibility to only clarithro- therapy in critically ill patients with altered mental status.
mycin with secondary testing of linezolid and moxifloxacin Clarithromycin, rifampin, and ethambutol penetrate relatively
(60,61). It recognized that the only drug for MAC for which well into the CNS. With inflamed meninges, the aminoglyco-
susceptibility testing has been shown to be predictive of clini- sides and the newer quinolones penetrate into the CNS to a
cal outcome was clarithromycin. Studies have shown that un- limited extent. Intrathecal therapy with aminoglycosides has
treated strains of MAC were all clarithromycin susceptible, been well described in other conditions, especially meningitis
with minimum inhibitory concentrations (MICs) of less than due to gram-negative bacteria. At present, the standard agents
8 g/mL, whereas strains that relapsed or failed therapy had for treatment of pulmonary or disseminated MAC disease
one of two mutations in the 23S rRNA gene, with MICs gener- would appear to be the best available agents for MAC CNS
ally more than 32 g/mL. Susceptibility to other drugs such as disease.
ethambutol, rifampin, rifabutin, streptomycin, and amikacin
provided no information about clinical responses to therapy.
However, recently, Brown-Elliott et al. (62) proposed amika-
cin MIC breakpoints reporting 96.2% of clinical isolates had
Mycobacterium kansasii
MICs of less than 32 g/mL and prolonged exposure corre- The major reservoir for M. kansasii is likely commercial or
lated with development of 16S rRNA mutation with MICs household water, and clinical disease predominates along the
more than 64 g/mL. southeastern and southern coastal states and the central plains
states. Unlike other NTM, M. kansasii has never been found
Treatment in soil or natural water supplies but has been recovered con-
sistently from tap water in cities where M. kansasii infection
Therapy of disease due to MAC, like susceptibility testing, is is endemic. Previous studies in Texas show that M. kansasii
more difficult and more controversial than that for M. tuber- disease is concentrated in urban areas, supporting a pos-
culosis. There have been few prospective controlled treatment sible association between clinical disease and potable water
trials (6367). Treatment of CNS disease must be extrapo- supplies. M. kansasii infection causes pulmonary nodular
lated from cumulative experience and data on treatment of disease in patients with bronchiectasis, fibrocavitary pulmo-
MAC lung disease in HIV-negative patients and disseminated nary disease resembling TB, and, rarely, disseminated disease
disease in patients with AIDS. Recommendations for therapy in immunocompromised hosts including patients with AIDS
generally follow ATS guidelines last updated in 2007 (60). The (1,8). The most common extrapulmonary sites are lymph
regimen recommended by the ATS for the treatment of MAC nodes, bone marrow, bone, joints, and skin. Only 10 cases
lung disease is a three-drug regimen. The daily regimen con- of CNS disease have been described, 7 of whom occurred in
sists of clarithromycin (500 mg twice daily) or azithromycin patients with AIDS.
(250 mg daily), rifampin (600 mg), and ethambutol (15 mg/kg
daily). The same three drugs can also be given intermittently. Mycobacterium kansasii Infections of the
The doses then are clarithromycin at 1,000 mg or azithromy-
cin 500 mg, ethambutol at 25 mg/kg, and rifampin at 600 mg
Central Nervous System
given three times weekly (Monday, Wednesday, and Friday). The 10 cases of CNS disease due to M. kansasii that have
Rifabutin at 150 mg can be substituted for rifampin in the been reported are listed in Table 30.4. The first description of
daily regimen, and at 300 mg in the three-times-weekly regi- M. kansasii disease in the CNS was reported by Wood et al.
men for severe disease. Aminoglycoside therapy with either (73) in 1956 in the context of disseminated M. kansasii infec-
streptomycin or amikacin is usually reserved for cavitary, ex- tion and was just discussed. In the second report of M. kansasii
tensive disease or macrolide-resistant cases. Patients requiring disease of the CNS in the literature (1966), both M. kansasii
long-term parenteral therapy should receive a dose of 8 to and M. tuberculosis were isolated from a 2-year-old girl with
10 mg/kg two to three times per week. meningitis (74). The patient presented with signs of meningi-
These macrolide-based regimens appear to have good ac- tis and her father was subsequently discovered to have active
tivity in the therapy of disseminated MAC disease in patients pulmonary TB. Lumbar puncture on two occasions had

Scheld_Ch30.indd 508 2/21/14 5:45 PM


Chapter 30: Infections Due to Nontuberculous Mycobacteria 509

TA B L E 3 0 . 4
REPORTED MYCOBACTERIUM KANSASII INFECTIONS OF THE CENTRAL NERVOUS SYSTEM

Reference No. of State/ Diagnosis


No. Cases Age Sex Country (Culture Source) Comorbidity Source Population of Cases

73 1 34 F U.S.A. Meningitis (CSF) Disseminated M. 15 pts. with M. kansasii


kansasii pregnancy infection, not HIV infected
74 1 2 F KY Meningitis (PM) M. tuberculosis Single case report
meningitis (CSF)
76 1 NR NR DC Meningitis (CSF) AIDS, disseminated 12 AIDS pts. with disseminated
M. kansasii M. kansasii at George
Washington University Hospital
77 1 NR NR TX Meningitis (CSF) AIDS, C. neoformans 9 pts. with disseminated
(CSF), MAC, M. kansasii of 1,100 AIDS
(CSF, blood) pts. at Parkland Hospital,
Dallas
79 1 NR NR NY Meningitis (CSF) AIDS 121 AIDS pts. with neurologic
complications in New York City
80 1 Adult M TX Brain abscess (tissue) AIDS, HCV 12 AIDS pts. with M. kansasii
infection at UTMB Galveston
19902001
81 1 22 F NY Meningitis (CSF) AIDS 86 AIDS pts. with NTM at Kings
County Hospital, Brooklyn
19811990
82 1 40 F GA Brain abscess AIDS, disseminated Single case report
(abscess aspirate) M. kansasii
83 1 42 M FL Brain abscess AIDS, disseminated Single case report
(abscess aspirate) M. kansasii
84 1 13 M U.S.A. Meningitis (CSF, PM) Disseminated Single case report
M. kansasii

F, female; pts., patients; NR, not reported; M, male; HCV, hepatitis C virus; PM, postmortem examination.

pleocytosis (200 to 313 WBCs/L, with a lymphocytic pre- AIDS during 1990 to 2001. Twelve patients were identified
dominance), elevated protein level (59 mg/dL), and depressed with only one patient having CNS involvement. Little details
glucose levels (22 to 27 mg/dL). The second specimen had one were provided, but brain biopsy revealed granulomatous in-
AFB on direct microscopy and grew M. tuberculosis. Despite flammation with AFB and the patient recovered on therapy.
treatment with isoniazid and streptomycin, the patient died. At The sixth case of M. kansasii CNS disease was reported
autopsy, the CSF grew M. tuberculosis, but the leptomeninges by Shafer and Sierra (81) in 1992. They summarized the ex-
grew M. kansasii in pure culture. This highly unusual report, perience with NTM (excluding MAC and M. gordonae) over
the first of its kind, raises the possibility of mixed infection by a 10-year period (1981 to 1990) at Kings County Hospital
M. tuberculosis and M. kansasii. in Brooklyn, New York. Seven patients had M. kansasii. Six
In patients with advanced HIV infection, M. kansasii often of the seven M. kansasii isolates were from respiratory speci-
produces disseminated infection. Among patients with AIDS mens. The seventh was isolated from the CSF of a 22-year-old
reported to the CDC from 1981 to 1987, disseminated M. kan- HIV-infected Haitian woman who had a brain lesion visual-
sasii was reported in 0.44% of patients from areas endemic for ized by contrast-enhanced CT scan but no other details of this
M. kansasii and in 0.08% in patients from nonendemic areas case were provided.
(8). Several retrospective studies have examined disseminated In 1992, Gordon and Blumberg (82) reported a 40-year-old
M. kansasii infection in HIV-infected patients (7580). In two woman with a 5-day history of right-sided headaches and left-
of these reports, M. kansasii was isolated from CSF in one sided weakness. Physical examination disclosed fever, oral thrush,
patient each, but few clinical details were provided (79,80). right Horner syndrome, and gait ataxia. CD4 lymphocyte count
A report from New York reported 121 patients with AIDS was 118 cells/L. CT scan of the head revealed two ring-enhanc-
of whom one had M. kansasii isolated from the CSF (79). ing lesions in the right frontal lobe and thalamus. Fine-needle as-
Symptoms included fever, weight loss, hypoadrenalism, and piration of the right frontal lobe mass revealed focal necrosis and
hematemesis. Lumbar puncture was performed before treat- perivascular inflammation, but no organisms were visible micro-
ment and three times over the next month because of neuro- scopically. Cultures grew only M. kansasii. The patient survived
logic deterioration. All CSF specimens had normal cell counts, 20 weeks after the initiation of treatment.
protein values, and glucose values, and negative cultures until A mass lesion with associated meningitis due to M. kansasii
the fourth specimen grew M. kansasii several weeks after the infection was reported from Florida in 1993 (83). A 42-year-
patients death. Smith et al. (80) reported a retrospective re- old man with AIDS presented with 2 weeks of headaches,
view of the pathologic features of M. kansasii in patients with 1 week of confusion and fever, and 1 day of loss of balance.

Scheld_Ch30.indd 509 2/21/14 5:45 PM


510 Part III: Bacterial and Mycoplasmal Infections

CT of the head revealed a 4-cm mass in the right occipitopa- grouped pre-ART era and ART era. Clarithromycin-treated
rietal region, which enhanced with intravenous contrast. CSF patients survived a median of 8 months (2 vs. 10 months) lon-
contained no cells, no organisms microscopically, and normal ger compared to those treated without clarithromycin.
glucose level, but protein level was 690 mg/dL. Aspiration of In place of isoniazid, which has marginal activity, sulfa-
the intracranial mass yielded pus, which was smear positive methoxazole-containing regimens have been used successfully
for AFB. Blood, sputum, and CSF cultures grew M. kansasii in the treatment of rifampin-resistant disease, but these studies
and PCR of the abscess aspirate was positive for M. kansa- antedated the availability of clarithromycin (91,92). In a study
sii. Treatment with isoniazid, rifampin, and ethambutol led of 40 patients, Ahn et al. (84) demonstrated that the addition
to resolution of symptoms and negative blood cultures within of streptomycin at 1 g twice weekly for the first 3 months to a
1 month and clearing of the lesion on CT by 3 months. 12-month regimen of the recommended three-drug daily com-
Thus, disseminated M. kansasii disease occurs in a small bination resulted in a long-term success rate of 97%.
but consistent fraction of patients with AIDS, and CNS in- Treatment for CNS disease should be based on optimal
volvement occurs rarely. The majority of published CNS infec- regimens for this infection at other body sites where sufficient
tions with M. kansasii have occurred in HIV patients. CNS numbers of cases provide a more solid understanding of the
disease may be a manifestation of hematogenous spread of this chemotherapy of this disease, combined with pharmacologic
microorganism. principles of the treatment of CNS infections. One additional
drug that offers potential for treatment of CNS disease due
Laboratory to M. kansasii is linezolid (Zyvox). All isolates of M. kansa-
sii have MICs of less than 4 g/mL (peak serum levels are
M. kansasii is seen on smear and recovered in culture by tech- 15 to 20 g/mL) (93) and the drug has excellent CNS pen-
niques designed for M. tuberculosis. It grows readily in broth, etration. Because of the large number of drugs available for
as well as on Middlebrook 7H10 agar and Lowenstein-Jensen M. kansasii and the cost of linezolid, there is no reported
agar. The organism typically produces rough large colonies that experience with other clinical forms of M. kansasii or with
turn bright yellow with exposure to light (photochromogen). A CNS disease. Linezolid has proven effective for the treatment
species-specific DNA probe for M. kansasii has been described of Mycobacterium chelonae (94) and Nocardia species (95),
and is commercially available (AccuProbe, Gen-Probe, Inc.). including patients with brain abscesses (95).
The organism is readily identified by biochemical tests as well
as by HPLC. On acid-fast smears, M. kansasii typically ap-
pears as a large, long bacillus that has unusual beading when
stained with Ziehl-Neelsen or Kinyoun stains. This often al- Mycobacterium simiae
lows an initial suspicion that one is dealing with this organism,
as opposed to M. tuberculosis. This species of mycobacteria was first reported in 1965 when it
Current 2011 CLSI recommendations for susceptibility was isolated from Macacus rhesus monkeys (96). M. simiae are
testing of M. kansasii are for rifampin and clarithromycin only slow-growing organisms that may produce pigmented and non-
given the concentration problems with isoniazid and to do ad- pigmented colonies. It has been rarely associated with human
ditional testing for other drugs such as amikacin, ethambutol, disease but has been described to cause lung disease in the set-
ciprofloxacin, moxifloxacin, linezolid, rifabutin, and sulfon- ting of structural lung disease (bronchiectasis). M. simiae is usu-
amides only if the isolate is rifampin resistant (61). ally reported from Israel and the southwestern United States
based on clinical samples (60). The epidemiologic source of
M. simiae is not well established but has been reported in sev-
Treatment eral pseudo-outbreaks in the southwestern United States attrib-
Treatment of M. kansasii disease of the CNS is extrapolated uted to hospital water and has been recovered from tap water
from the treatment of M. kansasii lung disease in HIV-negative sources derived from large aquifers (97). Four cases of CNS
patients and from the case reports summarized herein. infection of M. simiae have been reported. Valero et al. (98)
Treatment of M. kansasii was dramatically improved by the reported 137 clinical isolates of M. simiae in 75 patients in San
additions of rifampin and ethambutol to the pharmacopeia, Antonio, Texas over an 11-year period (1983 to 1993). One
as the success rate for regimens containing these two drugs hundred twenty-eight (93%) isolates were from respiratory
approach 100% (84,85). The current 2007 ATS recommenda- sources, four blood, one skin, one urine, one lymph node, and
tion for treatment of pulmonary disease due to M. kansasii one bone marrow. Only one case involved the CNS, a 38-year-
includes isoniazid (300 mg per day), ethambutol (15 mg/kg old man with AIDS who presented with fever, night sweats,
per day), and rifampin (600 mg per day) for a duration that and falls. CT of the head revealed three brain lesions with brain
includes 12 months of negative cultures (60). biopsy reported as B-cell lymphoma that was AFB stain posi-
However, the British Thoracic Society reported 155 pa- tive with no granulomas. Tissue culture grew M. simiae, but the
tients who were treated with ethambutol and rifampicin (86). patient refused therapy and 10 weeks after brain biopsy died.
Sputum conversion was reported in 99.4% of patients. In ad- The second case of M. simiae occurred in a 40-year-old
dition, Griffith et al. (87) reported 15 patients treated with man with AIDS who developed right hemiparesis, progressive
standard MAC therapy three times weekly. The sputum con- truncal ataxia, vertigo, and horizontal binocular diplopia with
version and long-term success rates were excellent with no fever and weight loss (99). MRI demonstrated multiple en-
treatment failures or relapses, but the study was too small to hancing isointense nodular lesions at the left cerebral peduncle
change the current ATS recommendations. Thus, clarithromy- and medulla involving the leptomeninges. Lumbar puncture
cin in combination with rifampin and ethambutol is believed on two occasions revealed 10 to 20 mononuclear cells/L,
to be the best regimen based on excellent in vitro activity, mini- glucose of 47 to 72 mg/dL, and protein of 49 to 135 mg/
mal toxicity, clinical performance against other NTM, and an- dL. Partial surgical resection of the medullary lesion revealed
ecdotal experience (78,88,89). For patients with HIV disease dense proliferation of spindle cells with histiocytes, lympho-
on ART regimens for which rifampicins are contraindicated, cytes, and mononuclear cells. Ziehl-Neelsen staining was AFB
a macrolide may substitute for the rifampicin if the ART regi- positive and tissue culture grew M. simiae that was confirmed
men cannot be adjusted (63). Tompkins and Witzig (90) retro- using 16S rRNA PCR gene sequencing. M. haemophilum was
spectively reviewed M. kansasii infection in 137 HIV patients also isolated from blood cultures, and the authors attribute

Scheld_Ch30.indd 510 2/21/14 5:45 PM


Chapter 30: Infections Due to Nontuberculous Mycobacteria 511

the negative tissue culture for M. haemophilum due to lack of increased intracranial pressure necessitating a ventriculoperito-
hemin or ferric ammonium citrate in culture medium that is neal (VP) shunt. The patient underwent several shunt revisions
essential for its growth (61). He received isoniazid, rifampin, complicated with the development of ascites. CSF specimens
pyrazinamide, ethambutol, and clarithromycin for treatment grew M. gordonae, and shunt valve was loaded with AFB.
and improved with only residual neurologic deficits. This is The meningitis and ascites resolved with shunt removal and
the fifth case of CNS NTM infection forming spindle cell pseu- antimycobacterial therapy. The second case of M. gordonae
dotumors (5255,99). meningitis occurred in a 23-year-old woman, who underwent
Balkis et al. (100) reported the third case of M. simiae in a VP shunt following excision of a cerebellar medulloblastoma
an 83-year-old HIV-negative man who developed fever, confu- 9 years prior, who presented with fever, hematuria, and progres-
sion, and agitation 2 days after surgery for a right hip fracture sive renal insufficiency (105). Nine months before admission,
secondary to a fall who had complained of feeling ill for she had been placed on prednisone for presumed hypersensi-
several weeks prior to the fall. Bilateral interstitial infiltrates tivity pneumonitis and hepatitis. Cultures of lung biopsy ob-
were seen on chest radiography. His level of consciousness tained 9 months prior revealed 2 growth of M. gordonae. CSF
deteriorated along with respiratory failure requiring intuba- was tapped from the shunt and showed numerous AFB. CSF
tion. Brain CT scan showed small vessel ischemic disease and analysis showed normal glucose and slightly elevated protein.
CSF specimen showed 1 WBC/L, normal glucose and pro- M. gordonae were cultured from CSF, urine, renal biopsy, and
tein levels. Bronchoalveolar lavage was performed and 1 week the shunt that had been removed.
later grew M. simiae in culture. One week after lumbar punc- Although one cannot generalize from two reported cases,
ture, CSF culture grew M. simiae. Both isolates were identi- it remains prudent to consider a mycobacterial etiology in
fied using 16S rRNA gene sequencing. The patient developed chronically febrile patients who have had shunting procedures
cardiopulmonary arrest and died. The authors contend that including infection of the shunt itself. Treatment with antimy-
because cultures were recovered from two different sterile sites cobacterial agents and removal of the foreign body was effec-
and no other mycobacteria was isolated in any other culture tive in these two cases.
during hospitalization, M. simiae was a true pathogen.
The final case of M. simiae was reported in a 39-year-old
with a 4-month history of fever, night sweats, and weight loss Mycobacterium genavense
who was diagnosed with AIDS (101). Physical exam revealed
hepatosplenomegaly, and liver biopsy showed AFB with mini- This species of mycobacteria was first reported by Bottger et al.
mal disruption of the liver tissue and was treated empirically (106) in 1992 who described it in 18 patients with AIDS present-
for TB with rifampicin, isoniazid, ethambutol, and pyrazin- ing with fever, diarrhea, and marked weight loss. Two subsequent
amide. CSF specimen showed no WBCs, normal protein, reports on M. genavense described similar symptoms in patients
and glucose. After hospital discharge, blood and CSF culture with AIDS, with a 68% mortality by 12 months (107,108).
grew M. simiae and antimycobacterial therapy was changed In terms of CNS disease, two cases have been reported.
to moxifloxacin, rifabutin, ethambutol, and azithromycin and Berman et al. (109) reported a 50-year-old man with AIDS. The
ART was started 10 days later. Three months after ART, fever patient was admitted with a 1-month history of memory prob-
and malaise returned with negative microbiologic workup, lems, confusion, and episodic tremor of the right arm, as well
and immune reconstitution inflammatory syndrome was diag- as a 2-week history of right-sided weakness and paresthesias.
nosed and resolved with steroid therapy. The patient had mild right-sided motor and sensory deficits and
M. simiae is highly drug resistant, including ethambutol and slurred speech. CT revealed cerebral edema and a mass effect in
rifabutin. The best antimycobacterial drugs are the macrolides the left parietal region. The patient was treated empirically for
(clarithromycin or azithromycin), trimethoprim-sulfamethoxazole toxoplasmosis with pyrimethamine and sulfadiazine but had a
(TMP-SMX), moxifloxacin, and amikacin (102). grand mal seizure after 1 week. MRI at that time showed a
left parietal mass abutting the dura with meningeal enhance-
ment and a large zone of surrounding edema. CT-guided needle
Mycobacterium gordonae biopsy revealed a patchy lymphocytic infiltrate without granu-
loma formation but with clusters of AFB on Ziehl-Neelsen stain.
M. gordonae organisms are ubiquitous in the environment, Treatment was started with five drugs to cover M. tuberculosis
but their recovery has been most closely identified with water. and MAC, and then was changed to clarithromycin, cipro-
M. gordonae are slow-growing, rarely pathogenic organisms floxacin, and ethambutol when the organism was identified as
that are occasionally encountered in the microbiology labora- M. genavense. The patient did well clinically and repeat imag-
tory and usually considered an environmental contaminant. ing demonstrated complete resolution at 12 months.
Weinberger et al. (103) reviewed all reported cases of disease Uchino et al. (110) reported a 50-year-old man with primary
due to M. gordonae to 1992 and identified 23 cases, of which immunodeficiency who presented with dysarthria, aphasia, and
13 had sufficient information to be judged definite. Eight had right hemiparesis. MRI revealed multiple intracranial masses in
pulmonary disease, seven had soft tissue infection of an ex- the subcortical regions. CSF cultures were negative for NTM.
tremity, five had disseminated disease, three had peritonitis, Brain biopsy was perfomed, which confirmed M. genavense
and one had corneal infection. The CNS was involved in two infection. These reports, as well as the previous reports on
cases, both of whom involved CSF shunts (see following sec- M. genavense, describe significant difficulties culturing this organ-
tion). In most of these patients, the infection was treated with ism using standard techniques for mycobacteria. Identification to
three antimycobacterial drugs including isoniazid, rifampin, the species level, in most cases, requires molecular methods.
ethambutol, and/or an aminoglycoside. Overall, 6 of 24 pa-
tients died. These cases and all reported cases of M. gordonae
infection involving the CNS were in the premacrolide era, and Mycobacterium terrae Complex
no isolates were confirmed by molecular methods.
The first case was a hydrocephalic child who had two ven- The M. terrae complex initially included only M. terrae and
triculoatrial shunts placed at 18 days and 7 months of age (104). M. nonchromogenicum. The species has now been expanded
The child had an elevated CSF protein level (106 mg/dL) and to multiple other species, with the most common isolate being

Scheld_Ch30.indd 511 2/21/14 5:45 PM


512 Part III: Bacterial and Mycoplasmal Infections

M. arupense. The M. terrae complex are rare, slowly grow-


ing nonpigmented mycobacteria that are present in tap water
presumed to be nonpathogenic in the lung but are a recognized
cause of posttraumatic tenosynovitis and local osteomyelitis
of the extremities. Two cases involving the CNS have been
reported (111,112). Woods and Washington (111) reviewed
the microbiology and clinical aspects of NTM and described
the Cleveland Clinic Foundations experience with NTM,
from 1982 to 1985. Of 140 patients with mycobacteria iso-
lated from clinical specimens, 93 were NTM, primarily MAC
(124 specimens). M. gordonae (75 specimens), M. kansasii
(59 specimens), and M. fortuitum group (58 specimens) ac-
counted for most of the remainder. Whereas most of these iso-
lates were from respiratory tract specimens, one patient who
had an acoustic neuroma removed several months previously
developed meningitis due to M. terrae complex. Details of the
case were not published. Lai et al. (112) described a 65-year-old
HIV-negative man who had radiotherapy for nasopharyngeal
carcinoma 6 years prior to presentation. He was admitted with
a 2-month history of headache and 5-day history of fever, nau-
sea, and poor appetite. Physical exam revealed meningismus
and CT disclosed dilated ventricles with diminished sulci. Two
lumbar punctures were performed revealing pleocytosis, 140
to 488 WBCs/L (77% to 81% PMNs), elevated protein of
131 to 267 mg/dL, and glucose 11 to 37 mg/dL. Microbiologic FIGURE 30.3 Magnetic resonance imaging, convalescent phase
CSF analysis showed negative Gram stain and culture, AFB axial fluid-attenuated inversion recovery (FLAIR) demonstrates dif-
stain, and cryptococcal antigen. Fever and headache persisted, fuse white matter high signal from a patient that M. malmoense and
M. interjectum was identified from CSF. (From ODwyer JP, OConnor
and MRI revealed leptomeningeal enhancement of the right
JG, McDermott H, et al. Meningoencephalitis associated with non-
temporal region and premedullary areas of the brain stem. tuberculous mycobacteria [published online November 22, 2009].
Empirical therapy for TB meningitis was initiated, but neuro- BMJ Case Rep, with permission.)
logic status worsened to coma. Repeat CT demonstrated hy-
drocephalus necessitating external ventricular drainage. Five
weeks after CSF AFB cultures were obtained, both yielded
treatment, encephalopathy improved, enabling the patient to
M. nonchromogenicum.
be discharged home. Specific antimycobacterial therapy was
not described further.
Mycobacterium malmoense
M. malmoense is a slow-growing pigmented species first re- Mycobacterium triplex
covered in Malmo, Sweden. It can cause a variety of diseases
M. triplex is a rare slowly growing nonpigmented species.
such as chronic pulmonary disease or cervical lymphadenitis
Most clinical cases have had underlying immune deficien-
in children. It is second only to MAC in many areas of north-
cies, especially advanced HIV disease. Zeller et al. (115)
ern Europe as a cause of NTM disease and is rarely seen in
described a 41-year-old man from Nigeria with AIDS with
the United States. Two cases with CNS disease have been re-
positive cultures of blood, sputum, and ascitic fluid with the
ported. Florakis et al. (113) described a patient with chronic
organism shown to be related to M. triplex by partial 16S
mastoiditis who was found to have pituitary insufficiency sec-
rRNA gene sequencing. The patient developed signs of left
ondary to mycobacterial infection due to M. malmoense. The
hemiballism-hemichorea. An MRI showed a lesion in the
disease was proven by surgical biopsy and 16S rRNA gene
subthalamic lesion. Cultures of CSF also grew M. triplex.
sequencing. The second case was a 49-year-old woman with
The patient initially responded to antimycobacterial therapy
a 6-month history of deteriorating vision, anorexia, anxiety,
but did poor clinically and died 5 months after the start of
and poor memory with a change in behavior manifested as
the therapy.
disinhibition and aggression (114). Physical exam revealed
Thus, it appears that less commonly encountered species of
poor cognitive performance, bilateral papilledema, and gen-
mycobacteria also cause CNS disease on occasion. Diagnosis
eral hyperreflexia and extensor plantar reflexes. CSF analy-
of mycobacterial disease of the CNS requires, ideally, that the
sis revealed pleocytosis, 37 WBCs/L (monocytes), elevated
involved tissue be examined microbiologically for AFB and
protein of 949 mg/dL, and 1:3 CSF:serum glucose ratio with
histopathologically specifically for mycobacteria and chronic
negative AFB stain. MRI demonstrated periventricular white
(often granulomatous) inflammation. Treatment of CNS dis-
matter changes and enhancement in the basal ganglia. CSF
ease due to these organisms must be based on published find-
culture grew M. malmoense and M. interjectum based on
ings and clinical experience, largely in other organs, as well as
the Hain PCR method. Empirical therapy with rifampicin,
principles of pharmacotherapy of CNS disease.
isoniazid, ethambutol, and pyrazinamide with steroids was
initiated. Over the next 3 months, the clinical course wors-
ened with development of myoclonus, vision loss, sleepwake
Transverse Myelopathy and Radiculomyelopathy
cycle disturbance, and agitation. Repeat CSF analysis revealed One study described two cases of transverse myelopathy and
elevated protein of 541 mg/dL and bilateral optic atrophy by radiculomyelopathy associated with pulmonary disease due to
funduscopic examination. MRI demonstrated development of M. kansasii and M. malmoense (116). The fact that only one
secondary white matter changes (Fig. 30.3). After 8 weeks of such report exists suggests it is a rare event with NTM.

Scheld_Ch30.indd 512 2/21/14 5:45 PM


Chapter 30: Infections Due to Nontuberculous Mycobacteria 513

lumbar discectomy (119). CSF was sampled repeatedly over a


RAPIDLY GROWING 10-month period. Two initial cultures of CSF were positive for
NONTUBERCULOUS M. fortuitum group and eight subsequent cultures during a 10-
month period were negative with the patient on treatment. CSF
MYCOBACTERIA leukocyte counts ranged from approximately 100 to 800 cells/
L, peaking 1 month after presentation. Dbridement of the
RGM include four clinically relevant species, including M. for-
infected site revealed granulomatous reaction with AFB in
tuitum, M. mucogenicum, M. chelonae, and the M. absces-
the bony tissue. Treatment was primarily with doxycycline,
sus complex. The proper taxonomy and nomenclature of
amikacin, and capreomycin once the identity of the organism
M. abscessus complex that now includes M. abscessus (sensu
was established. The next case involved a 57-year-old woman
stricto), M. massiliense, and M. bolletti will be a continual and
who developed recurrent fever and headache for 1 year fol-
dynamic process as more detailed genomic sequencing occurs.
lowing surgery for meningioma (120). Repeated CSF analysis
The RGM are generally considered environmental saprophytes
was indicative of bacterial meningitis with negative microbio-
widely distributed in nature, including tap water (1,60).
logic cultures. One year after surgery, repeat CSF culture grew
M. fortuitum group, which was treated with clarithromycin
and levofloxacin. Symptoms and CSF analysis improved on
Mycobacterium fortuitum therapy.
The remaining two cases involved children. A 9-year-old
The M. fortuitum group includes a variety of species that in- female underwent tympanomastoidectomy and 1 month later
clude M. fortuitum, M. porcinum, M. senegalense, M. mag- developed a mastoid fluid collection (20). Fluid collection was
eritense, M. peregrinum, and M. houstonense. M. fortuitum drained and culture grew M. fortuitum group with subsequent
was originally described as a cause of a postinjection site development of osteomyelitis of the right temporal bone with
abscess and principally causes primary skin and soft tissue meningeal enhancement by MRI. Lumbar puncture was nega-
infections, surgical wound infections, and central venous cath- tive for organisms. The patient underwent a modified radical
eterrelated infections (117). Rarely, other infections such as mastoidectomy and histopathology revealed granulomas with
pulmonary disease, prosthetic valve endocarditis, and cervical central necrosis, chronic inflammation with lymphocytes, and
lymphadenitis can occur. AFB stain negative. Tissue sent to the CDC for culture and
The relative frequency of CNS infection due to the M. for- susceptibilities identified M. fortuitum/M. peregrinum group
tuitum group is unknown but is probably rare. Most cases with reported sensitivity to doxycycline and meropenem. The
were reported before the recognition of the earlier noted new patient was treated with doxycycline and meropenem and
species, which require molecular methods for identification. 1 month later developed fever, headache, and back pain. MRI
Hence, all early isolates are listed as M. fortuitum group un- demonstrated a subdural fluid collection in the cerebellopon-
less clearly delineated from these other species by molecular tine angle that was surgically excised. Histopathology revealed
methods. Reported experience with this disease is summa- inflammatory tissue and negative stain for AFB. Quinn and
rized in Table 30.5. It has been associated with spinal cathe- Steele (21) reported a 12-year-old male with Arnold Chiari
ters, ventricular shunts, and mastoiditis. Indirect information type I malformation diagnosed at 8 years of age who under-
in this regard can be gleaned from reported cases such as the went posterior cranial fossa decompression with dural graft-
1983 report by Wallace et al. (117). During a 4-year period, ing. Fever and severe headaches followed immediately after
these workers were involved in the care of 125 patients with surgery with CSF showing lymphocytosis with negative bac-
disease due to RGM. One of these patients had M. fortuitum terial and fungal cultures. Two weeks postoperatively, he de-
meningitis. veloped a CSF leak requiring surgical revision, but fever and
Hand and Sanford (118) reported the first case of a CNS headaches continued, in addition to nausea, vomiting, and
infection due to the M. fortuitum group. They described an photophobia. Three weeks postoperatively, he developed per-
8-year-old boy with 12 days of fever, stiff neck, back and leg sonality changes that progressed to deteriorating mental status
pain, lethargy, and disturbed gait. Eight days before admis- and 5 weeks postoperatively, CT demonstrated obstructive
sion, a lumbar puncture revealed 3,025 WBCs/L (74% PMN hydrocephalus. Ventriculostomy with subsequent VP shunt
cells), 237 mg/dL of protein, and 51 mg/dL of glucose. Smears placement was performed. CSF obtained during ventriculos-
and cultures were negative, and the boy was diagnosed with tomy demonstrated AFB and he was empirically treated for
aseptic meningitis. On admission, the boy was febrile and TB. When AFB was identified as M. fortuitum group, antimy-
had a stiff neck. Repeated lumbar puncture was essentially cobacterial therapy was changed to amikacin, cefoxitin, and
unchanged. On the sixth hospital day, however, attempted clarithromycin. The VP was not removed due to the patient
lumbar puncture yielded frank pus. Myelography revealed being shunt dependent. Over the next 2 months, fevers, de-
a complete block to contrast material at T12. At surgery, a creased appetite, and abdominal pain with VP obstruction
cauda equina abscess was drained and an Ommaya reservoir requiring multiple revision and externalization of the shunt
was placed. Cultures of the pus obtained at attempted lumbar occurred. Ventricular and peritoneal fluid grew M. fortuitum
puncture and the material from surgery grew an M. fortuitum group several times. Up to this point, the patient had received
group, which was resistant to all antibiotics tested. Multiple 2 months of cefoxitin, 4 months of amikacin and clarithro-
subsequent cultures of CSF and reservoir grew M. fortuitum mycin, 1 month of ciprofloxacin, and 3 weeks of merope-
group and the reservoir was eventually removed. The organ- nem. Based on in vitro susceptibility, he was continued on
ism based on in vitro susceptibility testing was only sensitive ciprofloxacin, clarithromycin, and TMP-SMX with minimal
to oxacillin and kanamycin, and the patient was treated with planned therapy of 6 months. However, subsequent chest CT
these antibiotics for 25 days. Six months after discharge, the demonstrated multiple pulmonary nodules, and CSF grew
patient was ambulating without crutches but with persistent M. fortuitum group. The authors concluded that the VP
gait disturbance. shunt seeded the vasculature resulting in pulmonary infection.
Four cases of M. fortuitum group CNS infection have been Antimycobacterial therapy was changed to linezolid, azithro-
reported occurring after surgical procedures (20,21,119,120). mycin, amikacin, ciprofloxacin, and TMP-SMX with clinical
Meningitis developed in a 19-year-old woman following improvement. After 12 months from initial presentation, the

Scheld_Ch30.indd 513 2/21/14 5:45 PM


514 Part III: Bacterial and Mycoplasmal Infections

TA B L E 3 0 . 5
REPORTED MYCOBACTERIUM FORTUITUM GROUP INFECTIONS OF THE CENTRAL NERVOUS SYSTEM

Reference No. of Age State or Source Population


No. Cases (yr) Sex Country Diagnosis (Culture Source) Comorbidity of Cases

117,126 1 10 M Texas Subdural empyema Chronic otitis media 125 pts. with RGM
meningitis, posterior disease over 4 years
fossa abscess, mastoiditis, (most from Texas);
(CSF, abscess) case summarized in
detail in reference (126)
118 1 8 M Texas Cauda equina abscess, Motor vehicle accident 6 pts. with RGM disease
meningitis (abscess, CSF) over 3 years, Dallas
119 1 19 F Louisiana Meningitis, deep wound Lumbar discectomy 8 pts. with RGM
infection (CSF, biopsy) disease over 6 years,
New Orleans
120 1 57 F Japan Meningitis Meningioma Single case report
20 1 9 F Michigan Mastoiditis, temporal bone Otitis media, Single case report
osteomyelitis, subdural myringotomy,
fluid collection tympanoplasty
21 1 12 M Louisiana Meningitis Arnold Chiari Single case report
malformation,
ventriculoperitoneal
shunt
121 1 60 F Hong Kong Ventriculitis (CSF) Ventriculoatrial shunt Single case report
122 1 13 F Florida Meningitis Ventriculoperitoneal Single case report
shunt
123 1 Adult U.S.A. Ventriculitis (CSF) Spinal epidural catheter Single case report
22 1 60 M North Meningitis, infected pump Intrathecal drug Single case report
Carolina delivery device
124 1 16 M Spain Meningitis, infected wound Motor vehicle accident Single case report
(CSF, wound)
125 1 Adult M Texas Meningitis, dural abscess Motor vehicle accident 123 pts. with RGM
nonpulmonary disease
13 1 28 M ? Meningitis AIDS Single case report
127 1 28 M Texas Meningitis AIDS, disseminated Single case report
disease
128 2 28 M Thailand ? Hydrocephalus 103 pts. with NTM
disease in Thailand
54 F
129 1 33 F Germany Meningoencephalomyelitis None Single case report
130 1 Meningitis Prosthetic valve Single case report
endocarditis
131 1 NR NR India Brain abscess (abscess) NR 50 pts. with brain
abscess admitted to
neurosurgical unit

M, male; pts., patients; F, female; NR, not reported.

shunt was obstructed again and CSF remained culture positive time of shunt placement, the CSF was normal except for the
for M. fortuitum group. Intraventricular injection of amikacin presence of blood. From the time of shunt placement, how-
(4 mg) and linezolid (1 mg) were initiated due to failed oral ever, the patient had persistent fever. Routine studies of the
and intravenous therapy. Based on new in vitro susceptibil- CSF were normal except for a slightly elevated protein level.
ity, antimycobacterial therapy with ciprofloxacin, TMP-SMX, AFB smears, however, were positive and cultures grew M. for-
and twice-weekly intraventricular amikacin and linezolid was tuitum group. The CSF studies were repeated and the results
initiated with subsequent sterile CSF cultures. were the same. The only drugs to which the organism was
Two additional cases involving ventricular shunts have been susceptible in vitro were amikacin and ofloxacin. Treatment
reported. Chan et al. (121) described a 60-year-old woman in with these antibiotics was ineffective, and the patient deterio-
which a ventriculoatrial shunt was inserted for obstructive hy- rated until the shunt was removed. Intraventricular amikacin
drocephalus from spontaneous cerebellar hematoma. At the (20 mg on alternate days) was initiated through a Rickham

Scheld_Ch30.indd 514 2/21/14 5:45 PM


Chapter 30: Infections Due to Nontuberculous Mycobacteria 515

reservoir. After 1 week of intraventricular plus systemic ther- negative, had hydrocephalus, and M. fortuitum group was iso-
apy, the patient defervesced. Smears and cultures of the CSF lated from the CSF.
became negative after 3 weeks of this therapy, and therapy was Kell et al. (129) reported M. fortuitum group CNS infection
continued for 10 weeks. The other case involved a VP shunt in a 33-year-old HIV-negative patient with recurrent double
for hydrocephalus (122). Two additional reports involving an vision, paresthesia, and vertigo for 2 years following an appen-
epidural spinal catheter and intrathecal drug delivery device dectomy. MRI demonstrated multiple-enhancing leptomenin-
have been described (22,123). geal and intraparenchymal lesions in the cervical spinal cord
Two cases of M. fortuitum group meningitis followed local and brainstem. CSF contained 100 WBC/L (lymphocytes),
penetrating trauma. In 1984, Santamaria-Jauregui et al. (124) CSF/serum glucose ratio of 0.4, total protein of 2,500 mg/dL,
reported a 16-year-old boy who developed relapsing M. for- and positive AFB stain. However, microbiologic cultures and
tuitum group meningitis following a motor vehicle accident. PCR were repeatedly negative. The authors assumed infec-
A minor skin wound in the right sacral region from the ac- tion was due to slowly growing NTM, and antimycobacterial
cident necessitated excision and drainage 3 days later. Two therapy was initiated with clarithromycin, ethambutol, and ri-
weeks later, the boy developed fever, headache, and a stiff fabutin. The patient improved until several months later when
neck. Lumbar puncture revealed cloudy CSF with 1,085 WBC Lhermitte phenomenon, neck stiffness, recurrent hiccups, and
(80% neutrophils) with normal biochemistry and negative unsteady gait developed. MRI demonstrated increased num-
microbiologic analysis. Radiographic studies were negative bers of lesions disseminated in the basilar cisterns, subarach-
including head CT scan and myelography. The patient was noidal space of the vermis, and meninges and lumbar puncture
treated with two courses of multidrug antibacterial therapy proved disease progression. Therapy was changed to etham-
but relapsed both times when the drugs were discontinued. butol, isoniazid, and levofloxacin with improved symptoms.
Repeated lumbar puncture after the second relapse showed the Two months later, this patient developed cranial nerve pal-
CSF contained 2,100 WBCs/L (75% neutrophils), 100 mg/dL sies and MRI revealed hydrocephalus requiring VP shunt and
of protein, and 45 mg/dL of glucose. The patient developed therapy was changed to rifampicin, ethambutol, isoniazid, and
signs of meningitis and repeated CSF examination showed levofloxacin for 1 month without improvement. Open biopsy
1,200 WBCs (75% PMN), glucose level of 10 mg/dL, and pro- (approximately 1 year after initial presentation) of a menin-
tein level of 1.5 g/dL. CSF from both relapses grew M. fortui- geal lesion at C5/C6 demonstrated caseating granulomas with
tum group. The original sacral wound became fluctuant and negative Ziehl-Neelsen stain for AFB. Tissue by PCR analy-
pus from the wound grew M. fortuitum group. An abscess sis identified M. fortuitum group. Tissue culture remained
and fistula that reached the sacrum were surgically excised and negative (on chemotherapy). Amikacin and meropenem were
drained and a piece of glass was found in the dbrided tissue. started when M. fortuitum group was identified, and later, ti-
However, the fistula could not be demonstrated to communi- gecycline was added and amikacin was stopped. The patient
cate with the spinal canal. M. fortuitum group was isolated had improved at 18 months and therapy was continued.
from the dbrided tissue and glass fragment. Treatment with The remaining two reported cases of M. fortuitum group
isoniazid and trimethoprim-sulfamethoxazole produced clini- CNS infection included a patient with fatal prosthetic valve
cal improvement and normalization of the CSF. Wallace et al. endocarditis with secondary meningitis (130). A study from
(125) reported a similar case that also followed a motor vehi- Hyderabad, India reviewed 50 cases of brain abscesses that
cle accident and recovered after prolonged antibiotic therapy were diagnosed by CT scan and confirmed surgically in all cases
and multiple surgeries. (131). One specimen grew M. fortuitum group but no other
Dalovisio et al. (126) reported 10 patients with disease due microorganisms. No other details of this case were published.
to M. fortuitum group and M. chelonae. One was a 10-year-old
boy with chronic otitis media. A CT scan of the head showed
a right cerebellopontine angle mass. At surgery, the patient
Laboratory
had a subdural empyema and abscess, which grew M. fortui- RGM are the most difficult of the NTM to be recognized by
tum group. The patient was treated with erythromycin, iso- standard staining and culture techniques used for M. tubercu-
niazid, and kanamycin for 2 weeks without success. Clinical losis. The organisms do not always stain well with the Ziehl-
deterioration led to reexploration of the posterior fossa but Neelsen or Kinyoun method and may not be recognized readily
only partial resection of the abscess due to extensive disease. with the fluorochrome method. For example, it is not uncom-
Subsequent treatment with doxycycline and ethionamide for mon to see a sputum sample that is AFB smear negative but
several months did not resolve the fever, and after 8 months, with a heavy growth of an RGM species such as M. abscessus,
he developed obstructive hydrocephalus requiring VP shunt suggesting that the organisms did not stain. Though not per-
placement. A culture obtained at that time grew M. fortuitum tinent to CSF specimens, RGM are also highly susceptible to
group susceptible to amikacin. The patient was treated with the decontamination procedures that are used to remove other
amikacin with gradual improvement, and all signs of the in- bacteria from specimens. Hence, quantitation of the amount
fection eventually cleared; however, the patient developed per- of RGM organisms present by smear or culture can be difficult
manent partial hearing loss from the amikacin. The authors and should be assessed carefully. Separation of M. fortuitum
concluded that aggressive surgical dbridement and treatment from the M. chelonae and M. abscessus group is done fairly
with antibacterial drugs based on in vitro susceptibility results readily in the laboratory if molecular methods are used (132).
(often including amikacin and doxycycline) were critical com- M. fortuitum is the more drug susceptible of the RGM
ponents of successful therapy. group, and essentially all isolates are susceptible or interme-
Two patients with AIDS developed M. fortuitum group diate in vitro to achievable serum levels of amikacin, imipe-
meningitis with no precipitating cause (13,127). Both were nem, sulfonamides, and fluoroquinolones, and approximately
28-year-old men with far advanced HIV disease presenting with 50% are susceptible to doxycycline and cefoxitin (102). Most
clinical meningitis. One patient was lost to follow-up (13) and isolates are susceptible to linezolid (133), an oxazolidinone
one patient died with proof of the meningitis at autopsy (127). that has excellent CNS penetration with high serum and CSF
Saritsiri et al. (128) reviewed NTM infection of 103 patients levels (95). Macrolide therapy for the RGM group is compli-
in a Thai hospital. Seventy-one patients were HIV-infected cated by inducible erythromycin ribosomal methylase (erm)
and only two cases involved the CNS. Both patients were HIV gene that confers resistant by methylating an adenine in the

Scheld_Ch30.indd 515 2/21/14 5:45 PM


516 Part III: Bacterial and Mycoplasmal Infections

peptidyltransferase regions of the 23S rRNA (102). Erm gene bronchiectasis. The natural epidemiology and pathogenicity of
sequencing and extended incubation with clarithromycin of M. massiliense and M. bolletti is less known.
the mycobacteria for 14 days, compared to routine 3-day in- Eleven cases of CNS disease with M. abscessus complex have
cubation, will detect inducible resistance. been reported. Maniu et al. (137) described a 59-year-old woman
with a stab wound to the neck 6 months before her admission
Treatment with meningitis. Despite multiple antibiotics that included
clarithromycin, the patient died of her disease. A 27-year-old
In general, patients with serious M. fortuitum group disease
man with secondary pulmonary alveolar proteinosis associated
are given parenteral therapy with amikacin plus cefoxitin or
with myelodysplastic syndrome developed a subcutaneous ab-
imipenem combined with one or more oral agents and then
scess and meningitis after video-assisted thoracic surgery (138).
are treated with oral agents alone. For most diseases, the total
The patient died of graft versus host disease and M. abscessus
duration of therapy has been 6 months. Success rates with
complex sepsis after transplantation. Liebeskind et al. (139) de-
M. fortuitum group skin and soft tissue infections and in lung
scribed a 35-year-old man with 5 months of fever, fatigue, night
disease are quite high. Wallace et al. (125) prospectively stud-
sweats, anorexia, jaundice, and difficulty concentrating found to
ied treatment outcomes of 76 patients with extrapulmonary
have M. abscessus complex bacteremia with a vegetation of the
disease due to M. fortuitum group who were treated on the
posterior leaflet of the mitral valve. Lumbar puncture revealed
basis of in vitro drug susceptibility results once these became
80 WBC/L, glucose 41 mg/dL, and total protein 88 mg/dL.
available. Thirty-three patients had extensive or serious disease
Punctate lesions of the left basal ganglia, right thalamus, and
requiring inpatient combination chemotherapy and surgical d-
diploic space of the right parietal bone were seen on MRI.
bridement (31 patients). Almost all patients were treated suc-
Ciprofloxacin, amikacin, and meropenem for antimycobacte-
cessfully despite that these studies antedated the availability of
rial therapy was started. On hospital day 18, CSF cultures from
clarithromycin, the fluorinated quinolones, and imipenem. No
admission grew M. abscessus complex and therapy based on in
single regimen for M. fortuitum group CNS disease has been es-
vitro susceptibilities was switched to clarithromycin, imipenem,
tablished. Multiple drugs would appear to have potential; and
and amikacin. During hospitalization, the patient had intermit-
three to four drugs should probably be used, at least initially.
tent tonic-clonic seizures and on day 44 had a seizure; repeat
MRI revealed left frontoparietal hemorrhage and subsequently
developed brain herniation and respiratory distress on day
Mycobacterium chelonae 69 and died. Autopsy demonstrated disseminated granulomas
with AFB in meninges and subcortical structures.
M. chelonae was previously grouped as the M. chelonae/ab- Talati et al. (140) reported a 28-year-old woman with left
scessus complex and M. abscessus was not officially recognized eye enucleation and ocular prosthesis placement after trauma
as a separate species until 1992. Thus, literature prior to that 10 years previously. The patient underwent prosthesis replace-
date makes the nomenclature difficult to interpret. M. chelonae ment due to failure and within 1 year developed extrusion of
causes predominately skin, soft tissue, and bone infection and the implant with purulent drainage. The prosthesis was removed
rarely pulmonary disease. M. chelonae is more likely to cause and surgical cultures grew M. abscessus complex. The patient
disseminated cutaneous disease compared to the M. fortuitum was placed on clarithromycin and presented several months later
group (117,134,135). Wound infections and abscesses are gener- with headache, meningismus, and nausea. MRI revealed en-
ally the consequence of trauma, surgery, or immunosuppression. hancement of the left orbit, inferior portion of the left cavernous
Wallace et al. (134) characterized clinical disease due to sinus, and infratemporal fossa. CSF analysis showed pleocytosis,
100 isolates of M. chelonae associated with extrapulmonary dis- 411 WBC/L, glucose 30 mg/dL, and total protein 111 mg/dL
ease. No case involved CNS disease. One case of M. chelonae CNS with negative bacterial and fungal cultures. The patient was
infection has been reported involving a 39-year-old man who de- treated with amikacin and meropenem for 2 weeks followed by
veloped a spontaneous extradural abscess with associated psoas cefoxitin monotherapy for 6 weeks. Fever and headaches per-
abscess due to M. chelonae who recovered after multiple surgeries sisted, and an Ommaya reservoir was placed for intraventricular
and prolonged antibiotic therapy over a 33-month period (136). amikacin therapy and cefoxitin was changed to tigecycline. After
M. chelonae organisms are more susceptible based on in 1 month of therapy, seizures developed and MRI demonstrated a
vitro susceptibility to tobramycin than to amikacin, and it is cerebral abscess surrounding the Ommaya reservoir, prompting
the preferred aminoglycoside for this species. Macrolide ther- shunt removal. At that time, therapy was changed to cefoxitin
apy, either with clarithromycin or azithromycin, is an option and tigecycline. At this point, the patient had received 4 months
for treatment because M. chelonae does not possess an erm of intravenous antibiotic therapy with persistent symptoms.
gene. M. chelonae are uniformly resistant to cefoxitin, and ap- Repeat CSF analysis revealed 450 WBC/L (82% PMN) and
proximately 60% of isolates are susceptible to imipenem; how- total protein 548 mg/dL. Repeat MRI (Fig. 30.4) demonstrated
ever, susceptibility testing with imipenem is problematic due basilar meningitis, resolving brain abscess, and multiple micro-
to lack of reproducibility (102). Linezolid penetrates into the abscesses. CSF cultures grew M. abscessus complex. The patient
CNS, and approximately 50% of isolates are susceptible (102). was treated with a variety of antibiotics including amikacin, tige-
cycline, linezolid, meropenem, moxifloxacin, and co-trimoxazole
and received approximately 21 months of therapy with marked
Mycobacterium abscessus Complex improvement of her symptoms and functional status.
Lee et al. (141) reviewed 15 patients with CNS NTM infec-
The M. abscessus complex includes M. abscessus (sensu tion in a Taiwanese hospital from 2000 to 2010. M. abscessus
stricto), M. massiliense, and M. bolletti. The proper taxonomy complex was identified in 8 patients, MAC in 6 patients, and
surrounding these closely related RGM is currently in debate. M. kansasii in 1 patient. The MAC and M. kansasii patients
M. abscessus (sensu stricto) is predominately isolated in the were not described in detail. Six out of the 8 patients with
southeastern United States and has similar endemic geographic M. abscessus complex had preserved isolates that were ana-
distribution as MAC. It is the predominate RGM causing pul- lyzed using 23S rRNA and erm (41) PCR gene sequencing. All
monary disease (approximately 80%) and similar to MAC is six isolates were determined to be M. massiliense. Underlying
usually superimposed on preexisting lung disease, especially disease identified 2 patients to have otomastoiditis, 1 patient

Scheld_Ch30.indd 516 2/21/14 5:45 PM


Chapter 30: Infections Due to Nontuberculous Mycobacteria 517

FIGURE 30.4 Magnetic resonance imaging of the brain of patient with M. abscessus complex. The top
left panel is an axial section FLAIR image showing marked nodular enhancement in the orbit and intra-
temporal fossa. The bottom left panel is a second axial section flair image showing an abscess in the fron-
tal lobe also caused by M. abscessus complex infection. The follow-up images show significant interval
improvement in nodular basilar disease and decrease in size and enhancement of the frontal lobe abscess.
(From Talati NJ, Rouphael N, Kuppalli K, et al. Spectrum of CNS disease caused by rapidly growing
mycobacteria. Lancet Infect Dis. 2008;8:390398, with permission.)

Scheld_Ch30.indd 517 2/21/14 5:45 PM


518 Part III: Bacterial and Mycoplasmal Infections

chronic otitis media, and 4 patients steroid use. Five patients de- cerebral thrombophlebitis. The patient died 6 days after ad-
veloped infection postneurosurgery, and 2 patients had dissemi- mission with multiorgan failure. Similar to the first patient,
nated disease. Clarithromycin-based therapy combined with extensive microbiologic analysis only yielded a positive CSF
imipenem, amikacin, levofloxacin, and moxifloxacin were the PCR product of M. mucogenicum identified by 16S rRNA and
most common regimens with treatment courses ranging from rpoB gene sequencing, which was confirmed with CSF culture.
4 months to 1 year. Five patients died. The final case reported of M. mucogenicum CNS infection
was similar to the previous two cases and was also fatal (144).
Treatment A 42-year-old man presented with 3-week history of head-
M. abscessus (sensu stricto) is the most drug-resistant organ- ache, fever, chills, dizziness, and myalgias. Physical exam dem-
ism of the RGM which makes treatment difficult. Most iso- onstrated neck tenderness and mild sixth cranial nerve palsy.
lates are susceptible to amikacin. Approximately 50% are CT revealed a hypodense cystic area on the posterior aspect
sensitive to linezolid and imipenem based on in vitro suscepti- of right internal capsule. Lumbar puncture showed leuko-
bilities (102). Seventy percent are susceptible to cefoxitin, and cytosis with monocytic predominance 84%, glucose 27 mg/
commonly, tigecycline therapy is used, but there are no estab- dL, and protein of 457 mg/dL. Over 6 days, neurologic status
lished breakpoints. M. abscessus (sensu stricto) has a func- worsened and repeat CT demonstrated sudden dilatation of
tional erm gene in contrast to M. massiliense and M. bolletti the lateral and third ventricules. On day 7, the patient had
causing macrolide-based therapy a rare exception. In contrast, no spontaneous respirations and corneal or gag reflexes with
M. massiliense and M. bolletti lack a functional erm gene, fixed, dilated pupils. Electroencephalogram yielded an isoelec-
and macrolide-based therapy is the main treatment. These tric line and death occurred on day 9. Microbiologic analysis
organisms otherwise have similar in vitro susceptibilities as was negative. Autopsy revealed a brain with extensive, dif-
M. abscessus (sensu stricto) (102). No single regimen has fuse lymphoplasmatic leptomeningeal infiltrates with multi-
been established for CNS infection with M. abscessus (sensu nucleated giants cells, noncaseating granulomas, and necrosis.
stricto), M. massiliense, or M. bolletti. DNA was extracted from 35 different parts of the encephalon
and 2 samples from the temporal lobe by PCR, rpoB, and re-
striction fragment length polymorphism yielded M. mucogeni-
Mycobacterium mucogenicum cum. The authors conclude that M. mucogenicum was not a
contaminant because the water in the autopsy room was used
M. mucogenicum, formerly known as a Mycobacterium as a negative control and only 2 out of 35 samples of brain
chelonaelike organism, is an RGM common in tap water. It tissue isolated M. mucogenicum.
has been associated with outbreaks linked to water contami-
nation and central venous catheter infections remain the most
common clinical infection due to this organism. Four cases of Mycobacterium neoaurum
CNS infection due to M. mucogenicum have been reported.
Wallace et al. (142) reviewed 87 isolates of which 20 (23%) M. neoaurum is a RGM rarely associated with human infec-
were determined to be clinically significant. Only two cases tion. One case of CNS involvement has been reported (145).
involved the CNS with limited clinical data. The one case with A 63-year-old HIV-negative woman with rheumatoid arthritis
clinical disease occurred in an AIDS patient who had a lum- who had a stroke 10 months previously presented with rapidly
bar puncture 3 weeks prior for an unrelated reason. M. mu- functional decline over 2 months and died. Autopsy revealed
cogenicum was isolated from multiple CSF cultures that were multiple bilateral infarcts of the cortices, pons, thalamus,
also AFB smear positive. Two fatal cases of M. mucogenicum middle temporal gyrus, and putamen. Infarcts were associated
were reported by Adkambi et al. (143). The first patient was a with a thick exudate characterized by granulomatous inflam-
23-year-old man with 2-week history of headache, vertigo, and mation with caseation necrosis and foreign bodytype giant
neck pain who presented with fever. A CSF specimen revealed cells. Ziehl-Neelsen stain was negative. PCR on brain tissue
24 WBC/L (26% PMN), normal glucose and protein lev- identified M. mucogenicum by 16S rDNA gene sequencing.
els, and negative Ziehl-Neelsen stain. The patient died 3 days
after admission. Microbiologic analysis was negative except
for positive CSF PCR analysis yielding M. mucogenicum using Mycobacterium tokaiense
16S rRNA and rpoB gene sequencing. CSF culture confirmed
M. mucogenicum. The second case was an 82-year-old man Kondo et al. (146) reported a 36-year-old man found to have
with diabetes and chronic renal failure with femoral vascu- a pituitary stalk lesion who underwent complete surgical re-
lar graft who was admitted for fever, cough, and confusion. section. Histologic examination revealed caseous necrosis,
Chest radiography demonstrated alveolar pneumonia. CSF multinucleated giant cells with negative Ziehl-Neelsen stain-
analysis demonstrated no WBCs, normal glucose level, and ing with cultures. M. tokaiense was identified by 16S rDNA
elevated protein level of 67 mg/dL. CT scan showed extensive gene sequencing.

References
1. Wolinsky E. Nontuberculous mycobacteria and associated diseases. Am 5. Tortoli E. The new mycobacteria: an update. FEMS Immunol Med
Rev Respir Dis. 1979;119:107159. Microbiol. 2006;48:159178.
2. Wayne LG, Sramek HA. Agents of newly recognized or infrequently 6. Goslee S, Wolinsky E. Water as a source of potentially pathogenic
encountered mycobacterial diseases. Clin Microbiol Rev. 1992;5:125. mycobacteria. Am Rev Respir Dis. 1976;113:287292.
3. Brown-Elliott BA, Wallace RJ Jr. Clinical and taxonomic status of 7. Runyon EH. Anonymous mycobacteria in pulmonary disease. Med Clin
pathogenic nonpigmented or late-pigmenting rapidly growing mycobacte- North Am. 1959;43:273290.
ria. Clin Microbiol Rev. 2002;15:716746. 8. Horsburgh CR Jr, Selik RM. The epidemiology of disseminated nontuber-
4. Tortoli E. Impact of genotypic studies on mycobacterial taxonomy: the new culous mycobacterial infection in the acquired immunodeficiency syndrome
mycobacteria of the 1990s. Clin Microbiol Rev. 2003;16:319354. (AIDS). Am Rev Resp Dis. 1989;139:47.

Scheld_Ch30.indd 518 2/21/14 5:45 PM


Chapter 30: Infections Due to Nontuberculous Mycobacteria 519

9. Nightingale SD, Byrd LT, Southern PM, et al. Incidence of Mycobacterium 38. Rotstein AH, Stuckey SL. Mycobacterium avium complex spinal epidural
avium-intracellulare complex bacteremia in human immunodeficiency abscess in an HIV patient. Australas Radiol. 1999;43:554557.
virus-positive patients. J Infect Dis. 1992;165:10821085. 39. Moulignier A, Eliasewicz M, Mikol J, et al. Spinal cord compression due to
10. Lincoln EM, Gilbert LA. Disease in children due to mycobacteria other concomitant primary lymphoma and Mycobacterium avium-intracellulare
than Mycobacterium tuberculosis. Am Rev Respir Dis. 1972;105:683714. infection of the paravertebral muscles in an AIDS patient. Eur J Clin
11. Haverkamp MH, Arend SM, Lindeboom JA, et al. Nontuberculous myco- Microbiol Infect Dis. 1996;15:891893.
bacterial infection in children: a 2-year prospective surveillance study in the 40. Flor A, Capdevila JA, Martin N, et al. Nontuberculous mycobacterial
Netherlands. Clin Infect Dis. 2004;39:450456. meningitis: report of two cases and review. Clin Infect Dis. 1996;23:
12. Jensen J, Alvaro-Meca A, Micheloud D, et al. Reduction in mycobacte- 12661273.
rial disease among HIV-infected children in the highly active antiretroviral 41. Murray R, Mallad S, Health C, et al. Cerebral Mycobacterium avium
therapy era (19972008). Pediatr Infect Dis J. 2012:31:278283. infection in an HIV-infected patient following immune reconstitution and
13. Jacob CN, Henein SS, Heurich AE, et al. Nontuberculous mycobacterial cessation therapy for disseminated Mycobacterium avium complex infec-
infection of the central nervous system in patients with AIDS. South Med J. tion. Eur J Clin Microbiol Infect Dis. 2001;20:199201.
1993;86:638640. 42. Lascaux AS, Philippe L, Deforges L, et al. Late cerebral relapse of a
14. Yamamoto M, Sudo K, Taga M, et al. A study of diseases caused by atypi- Mycobacterium avium complex disseminated infection in an HIV-infected
cal mycobacteria in Japan. Am Rev Respir Dis. 1967;96:779787. patient after cessation of antiretroviral therapy. AIDS. 2003;17:14101411.
15. Chan KE, Pathmanathan T. Meningitis due to atypical mycobacteria. Med 43. Berger P, Lepidi H, Drougoul-Vey MP, et al. Mycobacterium avium brain
J Malaya. 1961;15:113116. abscess at the initiation of highly active antiretroviral therapy. Eur J Clin
16. Klein NC, Damsker B, Hirschman SZ. Mycobacterial meningitis: retro- Microbiol Infect Dis. 2004;23:142144.
spective analysis from 1970 to 1983. Am J Med. 1985;79:2934. 44. Fortin C, Rouleau D. Cerebral Mycobacterium avium abscesses: late
17. Wallace RJ Jr, Iakhiaeva E, Williams MD, et al. Absence of Mycobacterium immune reconstitution syndrome in an HIV-1-infected patient receiving
intracellulare and presence of Mycobacterium chimaera in household water highly active antiretroviral therapy. Can J Infect Dis Med Microbiol.
and biofilm samples of patients in the United States with Mycobacterium 2005;16:187189.
avium complex respiratory disease. J Clin Microbiol. 2013;51:17471752. 45. Verma R, Dhamija R. Disseminated Mycobacterium avium-intracellulare
18. Edwards LB, Acquaviva FA, Livesay VT, et al. An atlas of sensitivity to infection presenting as multiple ring-enhancing lesions on brain MRI.
tuberculin, PPD-B, and histoplasmin in the United States. Am Rev Respir Mayo Clin Proc. 2009;84:394.
Dis. 1969;99:1132. 46. Karne SS, Sangle SA, Kiyawat DS, et al. Mycobacterium avium-
19. Naguib MT, Byers JM, Slater LN. Paranasal sinus infection due to atypi- intracellulare brain abscess in HIV-positive patient. Ann Indian Acad
cal mycobacteria in two patients with AIDS. Clin Infect Dis. 1994;19: Neurol. 2012;15:5455.
789791. 47. Lee YC, Lu CL, Lai CC, et al. Mycobacterium avium complex infection-
20. McAvoy MJ, Carron MA, Poulik J, et al. Sequelae of rapid growing related immune reconstitution inflammatory syndrome of the central
mycobacteria otomastoiditis in a child. Arch Otolaryngol Head Neck Surg. nervous system in an HIV-infected patient: case report and review.
2009;135:602604. J Microbiol Immunol Infect. 2013;46:6872.
21. Quinn JV, Steele RW. Ventriculitis following a neurosurgic procedure. Clin 48. Gentry RH, Farrar WE, Mahvi TA, et al. Simultaneous infection of the
Pediatr. 2002;41:439441. central nervous system with Cryptococcus neoformans and Mycobacterium
22. Aliabadi H, Osenbach RK. Intrathecal drug delivery device infec- intracellulare. South Med J. 1977;70:865868.
tion and meningitis due to Mycobacterium fortuitum: a case report. 49. Uldry PA, Bogousslavsky J, Regli F, et al. Chronic Mycobacterium avium
Neuromodulation. 2008;11:311314. infection of the central nervous system in a nonimmunosuppressed woman.
23. Torriani FJ, McCutchan JA, Bozzette SA, et al. Autopsy findings in AIDS Eur J Neurol. 1992;32:285288.
patients with Mycobacterium avium complex bacteremia. J Infect Dis. 50. Horsburgh CR Jr, Mason UG III, Farhi DC, et al. Disseminated infection
1994;170:16011605. with Mycobacterium avium-intracellulare. Medicine. 1985;64:3648.
24. Wu HS, Kolonoski P, Chancy YY, et al. Invasion of the brain and chronic 51. Miyake S, Ohdama S, Kumagai T, et al. A case of pulmonary alveo-
central nervous system infection after systemic Mycobacterium avium lar proteinosis and disseminated atypical mycobacteriosis complicating
complex infection in mice. Infect Immunol. 2000;68:29792984. chronic myelogenous leukemia. Jpn J Chest Dis. 1992;30:17701776.
25. Gottlieb M, Schroff R, Schanker H, et al. Pneumocystis carinii pneumonia 52. Di Patre PL, Radziszewski W, Martin NA, et al. A meningioma-mimicking
and mucosal candidiasis in previously healthy homosexual men. N Engl J tumor caused by Mycobacterium avium complex in an immunocompro-
Med. 1981;305:14251431. mised patient. Am J Surg Pathol. 2000;24:136139.
26. Zakowski P, Fligiel S, Berlin OGW, et al. Disseminated Mycobacterium 53. Morrison A, Gyure KA, Stone J, et al. Mycobacterial spindle cell pseudo-
avium-intracellulare infection in homosexual men dying of acquired tumor of the brain: a case report and review of the literature. Am J Surg
immunodeficiency. JAMA. 1982;248:29802982. Pathol. 1999;23:12941299.
27. Hawkins CC, Gold JWM, Whimbey E, et al. Mycobacterium avium 54. Arkun K, Gordon DA, Lincoln C, et al. Atypical mycobacterial brain
complex infections in patients with the acquired immunodeficiency abscess presenting as a spindle cell lesion in an immunocompetent patient.
syndrome. Ann Intern Med. 1986;105:184188. Clin Neuropathol. 2012;31:155158.
28. Snider WD, Simpson DM, Nielsen S, et al. Neurological complications 55. Sadek M, Yue FY, Lee EY, et al. Clinical and immunologic features of an
of acquired immune deficiency syndrome: analysis of 50 patients. Ann atypical intracranial Mycobacterium avium complex (MAC) infection com-
Neurol. 1983;14:403418. pared with those of pulmonary MAC infections. Clin Vaccine Immunol.
29. Levy RM, Bredesen DE. Central nervous system dysfunction in acquired 2008;15:15801589.
immunodeficiency syndrome. In: Rosenblum ML, Levy RM, Bredesen DE, 56. Gyure KA, Prayson RA, Estes ML, et al. Symptomatic Mycobacterium
eds. AIDS and the Nervous System. New York: Raven Press; 1988:2963. avium complex infection of the central nervous system: a case report and
30. Monforte ADA, Gori A, Vago L, et al. Atypical mycobacterial disease review of the literature. Arch Pathol Lab Med. 1995;119:836839.
findings at autopsy in a cohort of 350 AIDS patients in Milan, Italy. J Infect 57. Okada H, Yoshioka K. Acute disseminated encephalomyelitis associated
Dis. 1995;172:901902. with meningitis due to Mycobacterium intracellulare. Inter Med. 2010;
31. Duong M, Piroth L, Chavnet P, et al. A case of rhombencephalitis with 49:21132116.
isolation of cytomegalovirus and Mycobacterium avium complex in a 58. Browne SK, Burbelo PD, Chetchotisakd P, et al. Adult-onset immunodefi-
woman with AIDS. AIDS. 1994;8:13561357. ciency in Thailand and Taiwan. N Engl J Med. 2012;367:725734.
32. Sharma K, Sharma A, Modi M, et al. PCR detection of co-infection with 59. Dickerman RD, Stevens QE, Rak R, et al. Isolated intracranial infection
Mycobacterium tuberculosis and Mycobacterium avium in AIDS patients with Mycobacterium avium complex. J Neurosurg Sci. 2003;47:101105.
with meningitis. J Med Microbiol. 2012;61:17891791. 60. Griffith DE, Aksamit T, Brown-Elliott BA, et al. An official ATS/IDSA
33. Cregan P, Yajko DM, Ng VL, et al. Use of DNA probes to detect statement: diagnosis, treatment, and prevention of nontuberculous myco-
Mycobacterium intracellulare and X mycobacteria among clinical bacterial disease. Am J Respir Crit Care Med. 2007;175:367416.
isolates of Mycobacterium avium complex. J Infect Dis. 1992;166: 61. Clinical and Laboratory Standards Institute. Susceptibility Testing of
191194. Mycobacteria, Nocardia, and Other Aerobic Actinomycetes; Approved
34. Katz RL, Alappattu C, Glass JP, et al. Cerebrospinal fluid manifestations of Standard. 2nd ed. Philadelphia: Clinical and Laboratory Standards
the neurologic complications of human immunodeficiency virus infection. Institute; 2011.
Acta Cytol. 1989;33:233244. 62. Brown-Elliott BA, Iakhiaeva E, Griffith DE, et al. In vitro activity of amika-
35. Gray F, Geny C, Lionnet F, et al. Etude neuropathologique de 135 cas cin against isolates of Mycobacterium avium complex with proposed MIC
adultes de syndrome dimmuno-deficience acquise (SIDA). Ann Pathol. breakpoints and finding of a 16S rRNA gene mutation in treated isolates.
1991;11:236247. J Clin Microbiol. 2013;51:33893394.
36. Perriens JH, Mahamudi M, Luabeya MK. Neurological complications 63. Ahn CH, Ahn SS, Anderson RA, et al. A four-drug regimen for initial
of HIV-1-seropositive internal medicine inpatients in Kinshasa, Zaire. treatment of cavitary disease caused by Mycobacterium avium complex.
J Acquir Immune Defic Syndr. 1992;5:333340. Am Rev Respir Dis. 1986;134:438441.
37. Malessa R, Diener HC, Olbricht T, et al. Successful treatment of meningo- 64. Wallace RJ Jr, Brown BA, Griffith DE, et al. Initial clarithromycin mono-
encephalitis caused by Mycobacterium avium intracellulare in AIDS. Clin therapy for Mycobacterium avium-intracellulare complex lung disease. Am
Invest. 1994;72:850852. Rev Resp Dis. 1994;149:13351341.

Scheld_Ch30.indd 519 2/21/14 5:45 PM


520 Part III: Bacterial and Mycoplasmal Infections

65. Griffith DE, Wallace RJ Jr, Brown BA, et al. Azithromycin monotherapy for 91. Wallace RJ Jr, Dunbar D, Brown B, et al. Rifampin-resistant
HIV() patients with Mycobacterium avium intracellulare (MAI) complex Mycobacterium kansasii. Clin Infect Dis. 1994;18:736743.
lung disease. Am J Respir Crit Care Med. 1995;151:A477. 92. Ahn CH, Wallace RJ Jr, Steele LC, et al. Sulfonamide-containing regimens
66. Griffith DE, Brown BA, Murphy DT, et al. Initial (6 month) results of for disease caused by rifampin-resistant Mycobacterium kansasii. Am Rev
three-times-weekly azithromycin in treatment regimens for Mycobacterium Resp Dis. 1987;135:1016.
avium complex lung disease in human immunodeficiency virus-negative 93. Brown-Elliott BA, Crist CJ, Mann LB, et al. In vitro activity of linezolid
patients. J Infect Dis. 1998;178:121126. against slowly growing nontuberculous mycobacteria. Antimicrob Agents
67. Griffith DE, Brown-Elliott BA, Wallace RJ Jr. Thrice-weekly clarithromycin- Chemother. 2003;47:17361738.
containing regimen for treatment of Mycobacterium kansasii lung disease: 94. Brown-Elliott BA, Wallace RJ Jr, Blinkhorn R, et al. Successful treatment
results of a preliminary study. Clin Infect Dis. 2003:37:11781182. of disseminated Mycobacterium chelonae infection with linezolid. Clin
68. Dautzenberg B, Truffot C, Legris S, et al. Activity of clarithromycin against Infect Dis. 2001;33:14331434.
Mycobacterium avium infection in patients with the acquired immune 95. Moylett EH, Pacheco SE, Brown-Elliott BA, et al. Clinical experience
deficiency syndrome. Am Rev Respir Dis. 1991;144:564569. with linezolid for the treatment of Nocardia infection. Clin Infect Dis.
69. Ruf B, Schrmann D, Mauch H, et al. Effectiveness of the macrolide 2003;36:313318.
clarithromycin in the treatment of Mycobacterium avium complex infection 96. Karassova V, Weissfeiler J, Krasznay E. Occurrence of atypical myco-
in HIV-infected patients. Infection. 1992;20:267272. bacteria in Macacus rhesus. Acta Microbiol Acad Sci Hung. 1965;12:
70. Gordin FM, Sullam PM, Shafran SD, et al. A randomized, placebo- 275282.
controlled study of rifabutin added to a regimen of clarithromycin and 97. Conger NG, OConnell RJ, Laurel VL, et al. Mycobacterium simiae
ethambutol for treatment of disseminated infection with Mycobacterium outbreak associated with a hospital water supply. Infect Control Hosp
avium complex. Clin Infect Dis. 1999;28:10801085. Epidemiol. 2004;25:10501055.
71. Dautzenberg B, Saint Marc T, Meyohas MC, et al. Clarithromycin and 98. Valero G, Peters J, Jorgensen JH, et al. Clinical isolates of Mycobacterium
other antimicrobial agents in the treatment of disseminated Mycobacterium simiae in San Antonio, Texas. An 11-year review. Am J Respir Crit Care
avium infections in patients with acquired immunodeficiency syndrome. Med. 1995;152:15551557.
Arch Intern Med. 1993;153:368372. 99. Phowthongkum P, Puengchitprapai A, Udomsantisook N, et al. Spindle
72. Panel on Opportunistic Infections in HIV-Infected Adults and Adolescents. cell pseudotumor of the brain associated with Mycobacterium haemophi-
Guidelines for the prevention and treatment of opportunistic infections in lum and Mycobacterium simiae mixed infection in a patient with AIDS:
HIV-infected adults and adolescents: recommendations from the Centers the first case report. Int J Infect Dis. 2008;12:421424.
for Disease Control and Prevention, the National Institutes of Health, 100. Balkis MM, Kattar MM, Araj GF, et al. Fatal disseminated Mycobacterium
and the HIV Medicine Association of the Infectious Diseases Society of simiae in a non-HIV patient. Int J Infect Dis. 2009;13:e286e287.
America. http://aidsinfo.nih.gov/contentfiles/adult_oi.pdf. Published 2013. 101. Phillips DR, Kirshan H, Watson J. First UK report of successful treatment of
Updated June 17, 2013. Accessed September 1, 2013 Mycobacterium simiae and immune reconstitution inflammatory syndrome
73. Wood LE, Buhler VB, Pollak A. Human infection with the yellow in an HIV-seropositive patient. Sex Transm Infect. 2008;84:271272.
acid fast bacillus: a report of fifteen additional cases. Am Rev Resp Dis. 102. Brown-Elliott BA, Nash KA, Wallace RJ Jr. Antimicrobial susceptibility
1956;73:917921. testing, drug resistance mechanisms, and therapy of infections with non-
74. Huempfner HR, Kingsolver WR, Deuschle KW. Tuberculous meningitis tuberculous mycobacteria. Clin Microbiol Rev. 2012;25:545582.
caused by both Mycobacterium tuberculosis and atypical mycobacteria. 103. Weinberger M, Berg SL, Feuerstein IM, et al. Disseminated infection
Am Rev Resp Dis. 1966;94:612614. with Mycobacterium gordonae: report of a case and critical review of the
75. Bamberger DM, Driks MR, Gupta MR, et al. Mycobacterium kansasii literature. Clin Infect Dis. 1992;14:12291239.
among patients infected with human immunodeficiency virus in Kansas 104. Gonzales EP, Crosby RM, Walker SH. Mycobacterium aquae infection
City. Clin Infect Dis. 1994;18:395400. in a hydrocephalic child (Mycobacterium aquae meningitis). Pediatrics.
76. Symington JS, Parenti DM, Keiser JF, et al. Disseminated Mycobacterium 1971;48:974977.
kansasii infection in HIV-infected patients. In: Programs and Abstracts of 105. Turner DM, Ramsey PG, Ojemann GA, et al. Disseminated Mycobacterium
the 1994 IDSA Annual Meeting. gordonae infection associated with glomerulonephritis. West J Med.
77. Carpenter JL, Parks JM. Mycobacterium kansasii infections in patients positive 1985;142:391393.
for human immunodeficiency virus. Rev Infect Dis. 1991;13:789796. 106. Bottger EC, Teske A, Kirschner P, et al. Disseminated Mycobacterium
78. Weinroth SE, Pincetl P, Tuazon CU. Disseminated Mycobacterium kansa- genavense infection in patients with AIDS. Lancet. 1992;340:7680.
sii infection presenting as pneumonia and osteomyelitis of the skull in a 107. Wald A, Coyle MB, Carlson LC, et al. Infection with a fastidious
patient with AIDS. Clin Infect Dis. 1994;18:261262. Mycobacterium resembling Mycobacterium simiae in seven patients with
79. Koppel BS, Wormser GP, Tuchman AJ, et al. Central nervous system AIDS. Ann Intern Med. 1992;117:586589.
involvement in patients with acquired immune deficiency syndrome (AIDS). 108. Pechere M, Emler S, Wald A, et al. Infection with Mycobacterium ge-
Acta Neurol Scand. 1985;71:337353. navense: clinical features in 44 cases. Berlin: Program and abstract of the
80. Smith MB, Molina CP, Schnadig VJ, et al. Pathologic features of 9th International Conference on AIDS, 1993. Abstract no. WSB102.
Mycobacterium kansasii infection in patients with acquired immunodefi- 109. Berman SF, Kim RC, Haghighat D, et al. Mycobacterium genavense
ciency syndrome. Arch Pathol Lab Med. 2003;127:554560. infection presenting as a solitary brain mass in a patient with AIDS: case
81. Shafer RW, Sierra MF. Mycobacterium xenopi, Mycobacterium fortuitum, report and review. Clin Infect Dis. 1994;19:11521154.
Mycobacterium kansasii and other nontuberculous mycobacteria in an area 110. Uchino H, Terasaka S, Yamaguchi S, et al. Multiple infectious intracranial
of endemicity for AIDS. Clin Infect Dis. 1992;15:161162. lesions of Mycobacterium genavense in an immunocompromised patient.
82. Gordon SM, Blumberg HM. Mycobacterium kansasii brain abscess in a Brain Nerve. 2011:63:7983.
patient with AIDS. Clin Infect Dis. 1992;14:78990. 111. Woods GL, Washington II JA. Mycobacteria other than Mycobacterium
83. Bergen GA, Yangco BG, Adelman HM. Central nervous system infection tuberculosis: review of microbiologic and clinical aspects. Rev Infect Dis.
with Mycobacterium kansasii. Ann Intern Med. 1993;118:396. 1987;9(2):275294.
84. Ahn CH, Lowell JR, Ahn SS, et al. Short-course chemotherapy for 112. Lai CC, Chen HW, Liu WL, et al. Fatal meningitis caused by
pulmonary disease caused by Mycobacterium kansasii. Am Rev Respir Dis. Mycobacterium nonchromogenicum in a patient with nasopharyngeal
1983;128:10481050. carcinoma. Clin Infect Dis. 2008;46:325326.
85. Campbell IA. A study of 9-month treatment with rifampicin and etham- 113. Florakis D, Kontogeorgos G, Anapliotou M, et al. Isolated pituitary
butol in pulmonary infection with M. kansasii (abstract). Bull Int Union granuloma by atypical Mycobacterium in an immunosuppressed woman.
Against Tuberc Lung Dis. 1986;61:433. Clin Endocrinol. 2002;56:123126.
86. Research Committee British Thoracic Society. Mycobacterium kansasii 114. ODwyer JP, OConnor JG, McDermott H, et al. Meningoencephalitis
pulmonary infection: a prospective study of the results of nine months of associated with non-tuberculous mycobacteria [published online ahead of
treatment with rifampicin and ethambutol. Thorax. 1994;49:442445. print November 22, 2009]. BMJ Case Rep.
87. Griffith DE, Brown-Elliott BA, Wallace RJ Jr. Thrice-weekly clarithro- 115. Zeller V, Nardi A-L, Truffot-Pernot C, et al. Disseminated infection
mycin containing regimen for treatment of Mycobacterium kansasii with a mycobacterium related to Mycobacterium triplex with central
lung disease: results of a preliminary study. Clin Infect Dis. 2003;37: nervous system involvement associated with AIDS. J Clin Microbiol.
11781182. 2003;41:27852787.
88. Brown BA, Wallace RJ Jr, Onyi GO. Activities of clarithromycin against 116. White VLC, Al-Shahi R, Gamble E, et al. Transverse myelopathy and
eight slowly growing species of nontuberculous mycobacteria, deter- radiculomyelopathy associated with pulmonary atypical Mycobacterium
mined by using a broth microdilution MIC system. Antimicrob Agents infections. Thorax. 2001;56:158160.
Chemother. 1992;36:19871990. 117. Wallace RJ Jr, Swenson JM, Silcox VA, et al. Spectrum of disease due to
89. Biehle J, Cavalieri SJ. In vitro susceptibility of Mycobacterium kansasii to rapidly growing mycobacteria. Rev Infect Dis. 1983;5(4):657679.
clarithromycin. Antimicrob Agents Chemother. 1992;36:20392041. 118. Hand WL, Sanford JP. Mycobacterium fortuituma human pathogen.
90. Tompkins JC, Witzig RS. Mycobacterium kansasii in HIV patients: clar- Ann Int Med. 1970;73:971977.
ithromycin and antiretroviral effects. Int J Tuberc Lung Dis. 2007;11: 119. Dalovisio JR, Pankey GA. Problems in diagnosis and therapy of
331337. Mycobacterium fortuitum infections. Am Rev Resp Dis. 1978;117:625630.

Scheld_Ch30.indd 520 2/21/14 5:45 PM


Chapter 30: Infections Due to Nontuberculous Mycobacteria 521

120. Fujikawa K, Suenaga A, Motomura M, et al. A case of Mycobacterium 134. Wallace RJ Jr, Brown BA, Onyi GO. Skin, soft tissue, and bone infections
fortuitum meningitis following surgery for meningioma. Rinsho due to Mycobacterium chelonae subspecies chelonae: importance of
Shinkeigaku. 2006;46:480484. prior corticosteroid therapy, frequency of disseminated infections, and
121. Chan KH, Mann KS, Seto WH. Infection of a shunt by Mycobacterium resistance to oral antimicrobials other than clarithromycin. J Infect Dis.
fortuitum: case report. Neurosurgery. 1991;29:472474. 1992;166:405412.
122. Midani S, Rathore MH. Mycobacterium fortuitum infection of ventri- 135. Ingram CW, Tanner DC, Durack DT, et al. Disseminated infection with
culo-peritoneal shunt. South Med J. 1999;92:705707. rapidly growing mycobacteria. Clin Infect Dis. 1993;16:462471.
123. Madaras-Kelly KJ, DeMasters TA, Stevens DL. Mycobacterium fortuitum 136. Suttner NJ, Adhami Z, Aspoas AR. Mycobacterium chelonae lumbar
meningitis associated with an epidural catheter: case report and a review spinal infection. Br J Neurosurg. 2001;15:265269.
of the literature. Pharmacotherapy. 1999;19:661666. 137. Maniu CV, Hellinger WC, Chu S-Y, et al. Failure treatment of chronic
124. Santamaria-Jauregui J, Sanz-Hospital J, Berenguer J, et al. Meningitis Mycobacterium abscessus meningitis despite adequate clarithromycin
caused by Mycobacterium fortuitum. Am Rev Resp Dis. 1984;130: levels in cerebrospinal fluid. Clin Infect Dis. 2001;33:745748.
136137. 138. Asai Y, Ouchi H, Ohosima T, et al. A case of secondary pulmonary alveo-
125. Wallace RJ Jr, Swenson JM, Silcox VA, et al. Treatment of non-pulmonary lar proteinosis associated with myelodysplastic syndrome, complicated
infections due to Mycobacterium fortuitum and Mycobacterium chelonei: with disseminated M. abscessus infection. Nihon Kokyuki Gakkai Zasshi.
based on in vitro susceptibilities. J Infect Dis. 1985;152:500514. 2009;47:11201125.
126. Daloviso JR, Pankey GA, Wallace RJ Jr, et al. Clinical usefulness of ami- 139. Liebeskind DS, Ostrzega N, Wasterlain CG, et al. Neurologic mani-
kacin and doxycycline in the treatment of infection due to Mycobacterium festations of disseminated infection with Mycobacterium abscessus.
fortuitum and Mycobacterium chelonei. Rev Infect Dis. 1981;3: Neurology. 2001;27:810813.
10681074. 140. Talati NJ, Rouphael N, Kuppalli K, et al. Spectrum of CNS disease caused
127. Smith MB, Boyars MC, Woods GL. Fatal Mycobacterium fortuitum by rapidly growing mycobacteria. Lancet Infect Dis. 2008;8:390398.
meningitis in a patient with AIDS. Clin Infect Dis. 1996;23:13271328. 141. Lee MR, Cheng A, Lee YC, et al. CNS infections caused by Mycobacterium
128. Saritsiri S, Udomsantisook N, Suankratay C. Nontuberculous mycobacte- abscessus complex: clinical features and antimicrobial susceptibilities of
rial infections in King Chulalongkorn Memorial Hospital. J Med Assoc isolates. J Antimicrob Chemother. 2012;67:222225.
Thai. 2006;89:20352046. 142. Wallace RJ Jr, Silcox VA, Tsukamura M, et al. Clinical significance,
129. Kell CA, Hunfeld KP, Wagner S, et al. Chronic meningoencephalomy- biochemical features, and susceptibility patterns of sporadic isolates of
elitis due to Mycobacterium fortuitum in an immunocompetent patient. the Mycobacterium chelonae-like organism. J Clin Microbiol. 1993:31:
J Neurol. 2008;255:18471849. 32313239.
130. Kuruvila MT, Mathews P, Jesudason M, et al. Mycobacterium fortuitum 143. Adkambi T, Foucault C, La Scola B, et al. Report of two fatal cases of
endocarditis and meningitis after balloon mitral valvotomy. J Assoc Mycobacterium mucogenicum central nervous system infection in immu-
Physicians India. 1999;42:10221023. nocompetent patients. J Clin Microbiol. 2006;44:837840.
131. Lakshmi V, Rao RR, Dinakar I. Bacteriology of brain abscess 144. Sariol CA, Galib Y, Pantoja P, et al. Fatal granulomatous meningoenceph-
observations on 50 cases. J Med Microbiol. 1993;38:187190. alitis associated to Mycobacterium mucogenicum-like microorganism: a
132. Steingrube VA, Gibson JL, Brown BA, et al. PCR amplification and case report. P R Health Sci J. 2009;28:276280.
restriction endonuclease analysis of a 65-Kilodalton heat shock protein 145. Heckman GA, Hawkins C, Morris A, et al. Rapidly progressive dementia
gene sequence for taxonomic separation of rapidly growing mycobacteria. due to Mycobacterium neoaurum meningoencephalitis. Emerg Infect Dis.
J Clin Microbiol. 1995;33:149153. 2004;10:924927.
133. Wallace RJ Jr, Brown-Elliott BA, Ward SC, et al. Activities of linezolid 146. Kondo A, Mori K, Iwata J, et al. Caseous necrotic granuloma in the
against rapidly growing mycobacteria. Antimicrob Agents Chemother. pituitary stalk due to nontuberculous mycobacteria (Mycobacterium
2001;45:764767. tokaiense) infectioncase report. Neurol Med Chir. 2006;46:8083.

Scheld_Ch30.indd 521 2/21/14 5:45 PM


CHAPTER 31 BRAIN ABSCESS
MATTHIAS KLEIN, HANS-WALTER PFISTER, ALLAN R. TUNKEL, AND W. MICHAEL SCHELD

DEFINITION EPIDEMIOLOGY
Brain abscess is a focal intracerebral infection that begins as The incidence of brain abscess is estimated at 0.3 to 1.3 cases
a localized area of cerebritis and develops into a collection per 100,000 people per year (7,8) with a male-to-female ratio
of pus surrounded by a well-vascularized capsule. It con- of 2:1 to 4:1 (4,9,10). One important factor in the incidence
tinues to be a diagnostic and therapeutic challenge to the of brain abscess (as is true for most infectious diseases) seems
clinician. to be the general health status of the population: In a study on
973 patients from South Africa with brain abscess from 1983
to 2002, the incidence dropped by 50% from 1983 to 2002,
HISTORY mainly due to improvements of socioeconomic standards and
increased availability of primary health care services (9). In the
The first reference to a brain abscess is attributed to total population, the incidence of brain abscess is relatively
Hippocrates in the fifth century bc (1). He described a low; however, the risk is markedly elevated in certain patient
syndrome of purulent otorrhea and fever associated with groups (Table 31.1).
cerebral symptoms and stressed that the chances of survival
depended on the external draining of purulent material:
Chills, pain, and fever throughout the head, especially in ETIOLOGY
the ear, temples, and bregma. . . . If anyone moves him, he
vomits copiously and easily. . . . If, in this patient, a wa- In the preantibiotic era, analysis of intracranial pus revealed
tery discharge breaks out through the nostrils or ears, it runs Staphylococcus aureus in 25% to 30% of patients, strepto-
out mixed with pus, and he recovers; if not, he usually dies cocci in 30%, coliforms in 12%, and no growth in approxi-
in seven days. The Corpus Hippocraticum even advocated mately 50% (11,12). With proper attention to techniques, the
surgical therapy: Then incise the head at the bregma; bore role of anaerobic agents in brain abscess has become appar-
right through to the brain, and heal the wound as you would ent. In one earlier study (13), 14 of 18 abscesses grew anaer-
one made by sawing. The first documented successful drain- obes on culturepredominantly streptococci in 66%, with
age of a brain abscess (of otitic origin) is attributed to the Bacteroides species in 60%. Series from the United Kingdom
French surgeon S.F. Morand in 1752 (2). In 1872, the army have stressed the role of anaerobic bacteria in brain abscesses,
surgeon J. F. Weeds reported on the successful drainage of a especially of otitic origin (11,12). The results of seven studies
traumatic frontal brain abscess and reviewed the American (in adults and children) demonstrate a particularly important
and European medical literature (2). Of 214 cases of brain role for streptococci, Enterobacteriaceae (in particular Proteus
abscess on record, only 4 cases resulted in recovery. Weeds species), and Bacteroides species (Table 31.2). The organisms
noted that not a single case recovered without the abscess and their frequency of isolation in exclusively pediatric-based
being opened and its contents evacuated, either by nature or series of brain abscess are similar to these figures except in
by the knife of the surgeon. In 1893, Sir William MacEwen the neonatal setting: In a large study on neonatal brain ab-
published his famous monograph Pyogenic Infective Diseases scess, Proteus mirabilis was identified in 27 (90%) of 30 cases,
of the Brain and Spinal Cord (2). Nineteen patients oper- Escherichia coli in two, and Serratia marcescens in another
ated on for a cerebral or cerebellar abscess were reported in newborn (14).
that work; eighteen of these recovered and only one patient The location of a given brain abscess or its predisposing
died. This publication established MacEwen as the father cause often suggests the most likely etiologic agents (Table 31.3).
of modern brain abscess management. Diagnostic delay was
the major obstacle in the success of therapeutic intervention,
as Victor Horsley put it in 1888, These cases are fortunately Bacteria
simple, perhaps the simplest, examples of intracranial sur-
gery, but there remains yet the method of diagnosis (3). The streptococci represent a diverse group of organisms;
In the twentieth century, refined surgical techniques, intro- however, those most frequently isolated in patients with brain
duction of antibiotics, and finally the advent of computed abscess belong to the Streptococcus milleri group (Streptococcus
tomography (CT) have contributed to a significant improve- anginosus, Streptococcus constellatus, Streptococcus interme-
ment in the mortality and morbidity rates associated with dius), which has a recognized predilection for causing focal
brain abscess (e.g., 4% mortality from 1981 to 1986 in a suppurative disease (1517). Streptococci from this group
study by Mampalam and Rosenblum [4]). Among the major are often microaerophilic. Streptococcus pneumoniae, despite
problems in the twenty-first century are postoperative intra- being the leading cause of bacterial meningitis in adults, is
cranial infections due to antimicrobial-resistant bacteria and only rarely cultured from patients with brain abscesses (18).
growing numbers of immunosuppressed patients who have S. aureus is the most common cause of traumatic and post-
an increased risk of developing a brain abscess, mostly due to operative brain abscesses, often isolated in pure culture (9).
atypical pathogens such as fungi, and excessive mortality After craniotomy, S. aureus accounted for almost 50% and
rates of up to 97% (5,6). Further progress in the therapy of Staph. epidermidis for 5% of intracranial infections in a large
brain abscess is therefore necessary. prospective study of 2,944 patients (19) and the proportion

522

Scheld_Ch31.indd 522 2/21/14 5:46 PM


Chapter 31: Brain Abscess 523

TA B L E 3 1 . 1
PATIENTS WITH AN INCREASED RISK OF DEVELOPING A BRAIN ABSCESS (SELECTED STUDIES WITH HIGH
NUMBERS OF PATIENTS)

Underlying Condition/Predisposing Factor Risk of Developing a Brain Abscess Reference No.

Acute or chronic sinusitis 1.7% 281


Chronic otitis media 0.14% 25
Lifetime risk for a 30-year-old patient: 0.5% 282
Penetrating craniocerebral trauma 3.0% and 4.2% after missile or fragment injury 283, 284
Craniotomy 0.58% risk of developing abscess or empyema 19
Cyanotic congenital heart disease 2.1% 285
Single pulmonary AVM 3.1% and 7.7% 152, 286
Multiple pulmonary AVMs 6.6% and 16.4% 152, 286
Diffuse pulmonary AVMs 37.5% 280
Acute infective endocarditis 4.1% and 7.1% 287, 147
Solid organ transplant recipients Heart or heartlung transplantation: 1.05% and 2.16% 5, 288, 289
Kidney transplantation: 0.36% 5
Liver transplantation: 0.63%, 0.64%, and 2.4% 5, 290
Allogeneic, syngeneic, and autologous BMT 1.77% and 4.0% (77% patients with toxoplasmosis, 23% 291, 292
and peripheral blood SCT with fungal brain abscesses, results of a German studya)
Allogeneic BMT 0.9% and 1.6% 293, 294
Allogeneic hematopoietic SCT 2.7% (toxoplasmosis, results of a German studya) 295
Human immunodeficiency virus infection 0.22% (United States), 1.0% (Francea), and 2.2% 296298
(on antiretroviral therapy) (Germanya) incidence of toxoplasmosis per year

AVM, arteriovenous malformation; BMT, bone marrow transplant; SCT, stem cell transplant.
a
The incidence of toxoplasmic encephalitis is lower in the United States due to the lower endemicity (seropositivity) compared with Europe (292).

TA B L E 3 1 . 2
BACTERIOLOGIC RESULTS OF ABSCESS PUS CULTURES FROM 491 PATIENTS REPORTED IN SEVEN STUDIES
FROM THE UNITED STATES (122), GREAT BRITAIN (28,154), SWITZERLAND (132), SWEDEN (299), CHINA (218),
AND TAIWAN (300)

Etiologic Pathogen Frequency (%) Etiologic Pathogen Frequency (%)

Streptococci 40.3 Pasteurellaceae 5.5


Unspecified aerobic streptococci 9.0 Haemophilus species 4.1
Viridans streptococcia 15.5 Eikenella species 0.6
Streptococcus pneumoniae 1.4 Pasteurella species 0.4
Unspecified anaerobic streptococci 14.5 Actinobacillus actinomycetemcomitans 0.4
Staphylococci 12.6 Bacteroides speciesc 21.8
Staphylococcus aureus 11.4 Fusobacterium species 4.9
Coagulase-negative staphylococci 1.2 Propionibacterium species 2.4
Enterococci 1.2 Corynebacterium species 1.8
Peptostreptococci 5.9 Pseudomonas species 1.2
Enterobacteriaceae 25.3 Veillonella species 0.6
Proteus speciesb 12.9 Actinomyces species 0.6
Escherichia coli 6.3 Lactobacillus species 0.6
Klebsiella species 4.1 Clostridium species 0.4
Citrobacter species 0.8 Nocardia asteroides 0.4
Salmonella species 0.6 Prevotella intermedia 0.2
Enterobacter species 0.4 Acinetobacter species 0.2
Serratia marcescens 0.2 Bacillus species 0.2

Cultures were positive in 402 cases (82%). A single organism was detected in 72% of these patients and 28% had mixed cultures. The percentages refer
to the total number of positive cultures (n 402). Because of the presence of mixed cultures, the total of the percentage numbers exceeds 100%.
a
Mostly Streptococcus milleri.
b
Mostly Proteus mirabilis or Proteus fragilis.
c
Mostly Bacteroides fragilis or Bacteroides melaninogenicus.

Scheld_Ch31.indd 523 2/21/14 5:46 PM


524 Part III: Bacterial and Mycoplasmal Infections

TA B L E 3 1 . 3
MOST PROBABLE LOCATION AND MICROBIAL FLORA OF BRAIN ABSCESSES ACCORDING TO THE SOURCE OF
INFECTION OR UNDERLYING CONDITION

Underlying Condition Most Probable Site of Abscess Most Common Microbial Flora References

Paranasal sinus Frontal lobe Streptococci (in particular, S. milleri), 11, 20, 120,
Bacteroides species, Staph. aureus, 121, 154,
Haemophilus species 281, 301, 302
Otogenic infection Temporal lobe, cerebellum Proteus species, streptococci, Bacteroides 20, 24, 28, 154,
species, Pseudomonas species, Staph. aureus 303306
Odontogenic infection Frontal lobe Streptococci, staphylococci, Actinomyces 23, 28, 307
species, Actinobacillus species, Bacteroides
species, Fusobacterium species
Bacterial endocarditis Frequently multiple abscesses, Staph. aureus, viridans streptococci 148, 227
any lobe can be involved
Pulmonary infection (abscess, Frequently multiple abscesses, Streptococci, staphylococci, Bacteroides species, 227
empyema, bronchiectasis) any lobe can be involved Fusobacterium species, Enterobacteriaceae
Right-to-left shunt (congenital Frequently multiple abscesses, Streptococci, staphylococci, Peptostreptococcus 161, 162, 285,
cyanotic heart disease, any lobe can be involved species, Haemophilus species 302, 308
pulmonary arteriovenous
malformation)
Penetrating trauma or Depends on site of wound Staph. aureus (methicillin-resistant Staph. 19, 28, 116,
postoperative aureus), Staph. epidermidis, streptococci, 154, 277
Enterobacteriaceae (multiresistant strains),
Clostridium species
Immunosuppressed patients: Frequently multiple abscesses, Aspergillus species, Candida species, Nocardia 5, 6, 289, 290,
e.g., bone marrow, stem cell, any lobe can be involved species, Toxoplasma gondii, Cryptococcus 293295, 309
or solid-organ recipients neoformans, Listeria monocytogenes,
Mycobacterium species
Patients with acquired Frequently multiple abscesses, Toxoplasma gondii, Mycobacterium species, 90, 92, 203,
immunodeficiency syndrome any lobe can be involved Cryptococcus neoformans, Aspergillus 310, 311
species, Candida species, Nocardia species,
L. monocytogenes

of methicillin-resistant Staph. aureus (MRSA) was very high predisposing factors like meningitis, tumor, ischemia, trauma,
(41%). In a recent study from Argentina, MRSA was even intracranial hematoma, and human immunodeficiency virus
identified in 10% of 80 patients with brain abscess (15). (HIV) infection (3638). Enterobacter species, though an-
Enterococcus faecalis has been reported as a rare cause of oto- other rare cause of brain abscess (39), must be considered in
genic, metastatic (e.g., due to endocarditis), meningitis-associated, postoperative cases (27). Serratia marcescens is another rare
and neonatal brain abscess (2022). Peptostreptococci, which are cause of neonatal brain abscess (40,41). In adults, only few
a common oropharyngeal flora, have been occasionally found in cases with opportunistic S. marcescens brain abscess are re-
odontogenic, otorhinogenic, postoperative, and metastatic (e.g., ported (42,43). In general, Enterobacteriaceae must be taken
following esophageal dilation) brain abscess (20,23). into account especially in postoperative deep brain infections
Enterobacteriaceae are usually found in mixed cultures. (9,19,35).
Proteus species are the most commonly isolated organism; Aggregatibacter aphrophilus and Aggregatibacter paraphro-
they exhibit a particular association with otogenic disease philus are most often isolated in brain abscesses due to
(17,24,25) and neonatal brain abscess (26). Brain abscesses Haemophilus species (16,17,44,45). Brain abscess secondary
due to Proteus species have also been reported after neuro- to H. influenzae is rare (46). Eikenella corrodens, which be-
surgical procedures (27). E. coli is sometimes found in oto- longs to the oropharyngeal flora, has been occasionally detected
genic or metastatic brain abscesses (28). It is also a cause of in particular in otorhinogenic or odontogenic brain abscess
neonatal brain abscesses (29). In two studies from Taiwan (47,48). Pasteurella multocida brain abscess has been reported
(30,31), Klebsiella species (mostly Klebsiella pneumoniae, as a rare complication of animal scratches or bites (4951).
less often Klebsiella oxytoca) were common (some with a Actinobacillus actinomycetemcomitans is sometimes encoun-
gas-forming appearance) in patients with debilitating dis- tered in polymicrobial odontogenic brain abscesses, often in
eases (in particular, diabetes mellitus). Citrobacter species, conjunction with Actinomyces infection (52,53).
especially Citrobacter diversus, are often implicated in brain Pseudomonas aeruginosa is mainly associated with oto-
abscesses that arise as a complication of neonatal meningitis genic brain abscess but has also been detected as a postop-
(32,33), but they are very rare in adults with brain abscesses erative, posttraumatic, or as a hematogenous complication
(34). Salmonella (S. typhi, S. typhimurium, and S. enteritidis (9,20,35,54). Anaerobes are significant causative pathogens
of group B, C, or D) brain abscess is also uncommon (35). of brain abscess, often found in mixed culture (16,35).
This disease is most likely to occur in adults with certain Bacteroides and Fusobacterium species are most frequently

Scheld_Ch31.indd 524 2/21/14 5:46 PM


Chapter 31: Brain Abscess 525

isolated. Brain abscesses due to Clostridium species (typically intracranial infections may be due to M. tuberculosis (9,30),
with a gas-forming appearance) may develop in particular reaching 56% in a Saudi Arabian study (75). In addition to
after penetrating head trauma with soil or fecal contamination M. tuberculosis, other mycobacteria were observed to cause
of the wound. Clostridium infections must also be considered focal CNS lesions; these include Mycobacterium bovis (bacille
postoperatively (55). In particular, cases due to Clostridium Calmette-Gurin) in an immunocompromised child (76),
welchii, Clostridium perfringens, Clostridium septicum, Mycobacterium kansasii in patients with acquired immuno-
Clostridium tertium, and Clostridium bifermentans have been deficiency syndrome (AIDS) (77), and Mycobacterium avium
reported (5658). Propionibacterium acnes has also gained at- complex in patients with and without AIDS (78).
tention as a cause of postoperative and posttraumatic brain
abscess, being reported to occur up to 10 years after surgery
(5961). Few cases of a brain abscess due to corynebacteria Yeasts and Fungi
have been reported (6264). Actinomyces species are anaero-
bic human commensals of low pathogenicity, which have been Fungi have assumed an increasing role as the etiologic agents
repeatedly recovered from brain abscesses (65). Two thirds of of focal intracranial infections. Many pathogenic fungi, such
the patients with central nervous system (CNS) actinomyco- as Cryptococcus neoformans, Candida species, Sporothrix
sis have an obvious primary site of infection distant from the schenckii, Aspergillus species, and Zygomycetes species are
CNS, most commonly the lung or cervicofacial region. When ubiquitous (79). Others are geographically restricted, such as
no obvious source is apparent, occult dental infection is often Coccidioides immitis (southwest United States, northern Mexico,
suspected (66). and Argentina), Histoplasma capsulatum (greater Mississippi
Especially in immunocompromised hosts with brain abscess, Valley, along the U.S.Mexico border, in multiple regions of
Listeria monocytogenes or Nocardia species can be found as Central and South America, and in scattered areas around the
causative pathogens (more than 90% of patients with listerial world), and Blastomyces dermatitidis (midwestern and mideast-
brain abscess and 34% with nocardial brain abscess are im- ern United States, along the St. Lawrence River, and in certain
munocompromised [9,30,35,67]). N. asteroides was isolated countries in Africa, the Middle East, and India). Most cases occur
in 98% of nocardial brain abscesses; single cases were due to in immunocompromised patients, and mortality remains high.
N. farcinica and N. caviae (67). N. asteroides infections of the Cerebral aspergillosis occurs in 10% to 20% of all cases
brain often had evidence of a pulmonary portal of entry (38% of invasive aspergillosis (Fig. 31.1) and only rarely is the
of cases). Bacillus cereus (68,69), Rhodococcus equi (70,71), brain the sole site of infection (80). Without therapy, mor-
and Gordona terrae (72) are other recognized pathogens of tality is high, but survival rates of up to 31% are reported
brain abscess in immunocompromised hosts. In approximately in patients receiving voriconazole therapy (81,82). Among
5% to 10% of cases of meningitis due to Mycobacterium tuber- the most important risk factors for invasive aspergillosis are
culosis, tuberculous granulomas (tuberculomas) can be found immunosuppressive therapy (including high-dose steroids),
(73,74); they usually resolve with medical therapy. When the hematologic malignancies, solid-organ and bone marrow
caseous core of a tuberculoma liquefies, a tuberculous abscess transplantation, and HIV infection (80,83). A report of brain
results; tuberculous brain abscesses are usually larger and less abscesses in bone marrow transplant recipients observed
common than tuberculomas, may be multiloculated, and often that 33 (58%) of 62 cases were due to Aspergillus species
have greater mass effect and edema (73). Depending on the (mostly Aspergillus fumigatus, with a few cases of Aspergillus
prevalence of tuberculosis, a considerable proportion of focal flavus and Aspergillus terreus) (6). Fungi of the Zygomycetes

FIGURE 31.1 Cerebral aspergillosis. A: Space-occupying brain lesion (asterisk) with contrast enhance-
ment on MRI in a 60-year-old patient. B: Histologic evaluation revealed a predominantly granulocytic
infiltration with the detection of Aspergillus fumigatus (arrow).

Scheld_Ch31.indd 525 2/21/14 5:46 PM


526 Part III: Bacterial and Mycoplasmal Infections

(formerly Phycomycetes) class can also invade the CNS. or without a scolex (109). Other helminthic infections that occa-
Rhino-(orbital-)cerebral mucormycosis usually affects immu- sionally lead to focal intracranial lesions include schistosomiasis
nocompromised patients, in particular patients with diabetes (110), echinococcosis (111), paragonimiasis (112), trichinosis
mellitus, hematologic malignancies, or stem-cell and solid (113), sparganosis (114), and infections due to Angiostrongylus
organ transplantation (8486). Isolated cerebral mucormy- cantonensis (115) or Strongyloides stercoralis (116).
cosis has been observed mostly in injection drug users with
or without AIDS (87), but also in patients with hematologic
malignancies (88) or pharmacologic immunosuppression PATHOGENESIS
(89). Candidiasis is the fungal infection most often observed
at autopsy to be involving the CNS (90). Cases of diffuse Brain abscesses occur most commonly in association with one
cerebritis with miliary microabscesses, large parenchymal ab- of three distinct clinical settings: (a) a contiguous focus of
scesses, meningitis, and cerebrovascular complications have infection, (b) cranial trauma or neurosurgical procedures, or
been described (90). Risk factors for the development of CNS (c) hematogenous spread from a distant focus. No predispos-
candidiasis are immunosuppression, treatment with antibiot- ing factors are recognized in approximately 10% to 25% of
ics or corticosteroids, intravascular catheters, recent abdomi- reported cases (Table 31.4).
nal surgery, prematurity (in neonates), recent neurosurgery or
cerebrospinal fluid (CSF) diversion, and injection drug abuse
(90). Cryptococcus neoformans usually causes meningitis Contiguous Infectious Foci
when it invades the CNS, but mass lesions due to this organ-
ism have been observed. Neuroradiologic studies have shown Most patients with brain abscess demonstrate a contiguous
cryptococcoma (granuloma) in 11% of patients with crypto- focus of infection, usually sinusitis or otitis.
coccal meningitis studied by CT (91) and in 50% of patients
studied by magnetic resonance imaging (MRI) (92). Otitis/Mastoiditis
Scedosporium apiospermum (formerly known as Allescheria
The incidence of otogenic brain abscess (Tables 31.1 and 31.4)
boydii, Pseudallescheria boydii, and Petriellidium boydii) has re-
seems to decrease in developed countries. However, in areas
ceived increased attention as a potential cause of CNS infection
where otitis media continues to be neglected or where therapy
(93). In addition to immunosuppression, near drowning in pol-
is delayed, intracranial complications of this process still present
luted water and rarely direct inoculation (orbital trauma, lum-
a particularly serious threat (117). Chronic otitis media and/or
bar puncture) were reported as predisposing conditions (94,95).
mastoiditis leads to intracranial extension much more often
Cladosporium trichoides (Xylohypha bantianum, Cladosporium
than acute disease. In particular, cholesteatoma is a common
bantianum) is the most common cause of cerebral phaeohypho-
associated finding in otogenic brain abscesses, being found in
mycosis, accounting for 28 of 53 reviewed cases (96).
38% to 98% of patients (24,25). Most otogenic brain abscesses
are solitary lesions. The majority of otogenic brain abscesses
(55% to 75%) are located in the temporal lobe (24,25). The cer-
Protozoa and Helminths ebellum is the next most commonly affected area (20% to 30%),
and it has been observed that approximately 90% of cerebellar
Various protozoa and helminths may cause brain abscesses, abscesses are secondary to otogenic infections (118). Otogenic
with their incidence varying with geographic location. intracranial suppuration most often develops in patients younger
Toxoplasmosis is one of the most common parasitic infec- than 30 years and shows a male predominance (24,25).
tions of the brain, particularly in the setting of AIDS (97) (see
later discussion) but also in patients after bone marrow trans- Sinusitis
plantation (98,99). Entamoeba histolytica infections rarely
Sinugenic brain abscesses are almost exclusively located in
involve the brain (100). The lesions are usually multiple and
the frontal lobe (Fig. 31.2), reflecting the distribution of the
are most often associated with another focal site of infec-
associated sinusitis (119121). Most cases are in the setting of
tion, usually hepatic. Free-living amebas (Naegleria species,
frontoethmoidal disease followed by maxillary disease (122).
Acanthamoeba species, and Balamuthia species) are prefer-
entially neurotropic (101,102). CNS infection with Naegleria
fowleri typically presents as acute fulminant meningoenceph- TA B L E 3 1 . 4
alitis (primary amebic meningoencephalitis) in immunocom-
petent children and adults after exposure to polluted water UNDERLYING CONDITIONS IN 1,730 PATIENTS WITH
(water-related sport activities in the tropical and subtropical BRAIN ABSCESS (AFTER [20,28,122,126,127,154,312])
climates) (102). Balamuthia mandrillaris and Acanthamoeba
species cause granulomatous amebic encephalitis, which has Underlying Condition Mean Frequency (Range)
a subacute or chronic course and is usually seen in immu-
nocompromised individuals (in particular, in patients with Otitis/mastoiditis 33% (1466%)
AIDS) (102,103). Amebic CNS infections are usually fatal. Hematogenous spread 23% (1734%)
Neurocysticercosis (due to Taenia solium larvae) is the Trauma/neurosurgery 14% (323%)
most common parasitic infection of the human nervous system Sinusitis 9% (120%)
(104107). It is endemic in many parts of the world, particularly
Latin America, Africa, and Asia, and still relatively common in Dental infection 2% (013%)
Portugal, Spain, and Eastern European countries. In many de- Unknown (cryptogenic) 17% (1025%)
veloped countries with high rates of immigration from endemic Othersa 3% (09%)
areas, neurocysticercosis is an imported disease (107,108).
Cysticercosis is probably the single most important cause of ac- a
Other causes were in particular meningitis and infections of the
quired epilepsy in the developing world (109). Focal brain lesions scalp or skull. The total is 101% because of the presence of multiple
in neurocysticercosis usually present as (solid-enhancing) granu- predisposing conditions in some patients.
lomas, calcified nodules, or (ring-enhancing) cystic lesions with

Scheld_Ch31.indd 526 2/21/14 5:46 PM


Chapter 31: Brain Abscess 527

FIGURE 31.2 Sinusitis and brain abscess. MRI of a brain abscess (asterisk) located in the frontal lobe
caused by frontal sinusitis (arrow). A: T2-weighted sequence. B: T1-weighted with contrast. C: Diffusion-
weighted imaging.

Sphenoid sinusitis, despite its relative rarity (approximately due to P. mirabilis or C. diversus. Neonatal meningitis due to
3% of all sinusitis cases), has a relatively high rate of intra- Enterobacter species or Serratia species is also often compli-
cranial complications (123). Intracranial suppuration second- cated by brain abscesses (39,40). Although P. mirabilis is a rare
ary to sinusitis predominates in men in their second or third cause of neonatal meningitis, 27 of 30 neonatal brain abscesses
decade of life (119,120). diagnosed over a 12-year period were due to this organism (14).
Additionally, abscess formation has been associated with more
Dental Infection than 70% of cases of C. diversus meningitis in the infant (129).
The increased propensity for these two organisms to cause a
Dental infections have less often been documented as a predis-
brain abscess is incompletely understood (130,131).
posing cause of a brain abscess in the past (Table 31.4), but a
recent study indicated an important role for dental infection as
a source of brain abscess in up to 12% of patients (16). Given
the frequency of dental infections, however, intracranial compli-
Cranial Trauma and Neurosurgical
cations of this process are rare. The site of an associated brain Procedures
abscess is most commonly the frontal lobe, but temporal lobe
localization can also occur by direct extension. A large majority Penetrating Cranial Injury
of intracranial infections in this setting follow a recent tooth ex-
The risk of a brain abscess is increased in the setting of a penetrat-
traction or dental manipulation. Bacteremia has been observed
ing cranial injury. Combat series reported a risk of 3% to 4.2%
in 100% of patients after dental extraction, in 70% after den-
of developing a brain abscess after missile or fragment injuries
tal scaling, in 55% after third molar surgery, and in 20% after
(Table 31.1). Risk factors were extensive brain injury, coma,
endodontic treatment (124). Another study reported that 9% of
trajectory through an air sinus, wound infections, CSF fistula,
children with extensive tooth decay were bacteremic before treat-
inadequate initial dbridement, retained (bone or metallic) frag-
ment (125). Hygiene procedures such as brushing of the teeth
ments, and incomplete dural closure. Several series have noted
increased the prevalence of bacteremia to 40%, and anesthetic
the role of head trauma as the cause of brain abscess in civilians.
and surgical procedures increased it to 97%. In both studies, the
Thus, head trauma was reported as an underlying cause of brain
bacteria isolated from the blood were mostly facultative species
abscess in 3% to 11% of patients in most series (4,28,132,133).
indigenous to dental plaque, in particular viridans streptococci
Whereas the incidence of brain trauma as the cause for cerebral
(124,125). Many cases of cryptogenic brain abscess are believed
abscess formation declined in a study from the United Kingdom
to be secondary to asymptomatic dental foci of infection.
(16), it was found to be the most common cause (33%) for brain
abscess in 973 patients from South Africa (9), reflecting the inci-
Facial and Scalp Infections
dence of brain trauma (due to accidents or violence) in a society.
Facial and scalp infections were reported in 1% to 4% of pa- Various types of cranial trauma have been implicated, including
tients with brain abscess (126,127). Septic thrombosis of the compound depressed skull fractures (133,134), dog bites (135),
cavernous sinus is a possible link, and Staph. aureus is the injuries due to rooster pecking (136), and cranial penetration
most common pathogen in this setting (128). with lawn darts, pencil tips, or paint brushes (137,138). Several
cases of brain abscess as a complication of cervical traction with
Meningitis tongs or halo fixation have been described (139,140). Associated
pin-site cellulitis is usually observed.
Brain abscess rarely complicates bacterial meningitis; however,
it should be strongly considered as an associated possibility in
Craniotomy
the neonate with purulent meningitis, particularly when men-
ingitis is due to gram-negative organisms (29). The vast major- Brain abscess after neurosurgery is common (Fig. 31.3).
ity of neonatal brain abscesses that complicate meningitis are In a series of 2,944 patients who underwent neurosurgery,

Scheld_Ch31.indd 527 2/21/14 5:46 PM


528 Part III: Bacterial and Mycoplasmal Infections

FIGURE 31.3 Brain abscess complicating


brain surgery. MRI of a brain abscess
(asterisk) in a 28-year-old patient who
had been operated on a cyst of the lateral
ventricle by occipital craniotomy (arrow).
A: Fluid-attenuated inversion recovery
(FLAIR). B: T1-weighted imaging with con-
trast enhancement. Pseudomonas aerugi-
nosa and coagulase-negative staphylococci
were identified as causative pathogens.

neurosurgical site infections developed in 117 (4%) (19). These abscesses are more often multiple and multiloculated as
Forty-four patients (1.5%) had incisional infections (30 with compared with those that have an origin from a contiguous
scalp infections and 14 with bone flap osteitis), and seventy- focus of infection. Thirty-seven percent to 50% of multiple
three (2.5%) had deep wound infections (meningitis in 56 brain abscesses were of hematogenous origin in several studies
and mass lesions in 17 patients). Given the high number of (142144), whereas metastatic spread accounted only for
neurosurgical operations at certain centers, it was reported 17% to 34% of all brain abscesses (Table 31.4).
among the most frequent causes for brain abscess in some
series (16,141). Risk factors for deep wound infections were a Pulmonary Infections
Glasgow Coma Scale score of less than 10 on admission, total Pyogenic lung diseases, especially lung abscess and bronchi-
hair removal, emergency surgery, CSF drainage, CSF leak- ectasis, accounted for 7% to 18% of total brain abscess cases
age, and early subsequent operations. Interestingly, wound in four studies (4,9,122,127). Underlying chronic pulmonary
contamination, surgical duration, and the absence of antimi- diseases such as cystic fibrosis, however, are surprisingly infre-
crobial prophylaxis were not significant risk factors for deep quently complicated by a brain abscess (145). When observed,
wound infections (19). The authors, therefore, hypothesized it most often occurs in older adolescents or adults (146).
that the risk of contaminating CSF or brain parenchyma be- The organisms cultured from the sputum only occasionally
gins during surgery but persists in the postoperative period matched those cultured from the brain abscess and in some
if a CSF drainage or leakage is present. This hypothesis was patients, associated sinus disease was found. These observa-
supported by a predominance of hospital-acquired antibiotic- tions suggest that the lung is not necessarily the source of brain
resistant bacteria in deep wound infections. abscess in patients with cystic fibrosis and that contiguous
spread of infection should also be considered.
Hematogenous Spread Endocarditis
Hematogenous brain abscesses often share the following char- Brain abscess occurs in 5% to 7% of patients in the setting
acteristics: (a) a distant focus of infection, most often within of bacterial endocarditis (Table 31.1) (147,148), despite the
the chest; (b) location in the distribution of the middle cerebral presence of persistent bacteremia (see Chapter 34). The high-
artery (Fig. 31.4), (c) initial location at the gray matterwhite est incidence of brain abscess (5.4%) was reported in infective
matter junction, (d) poor encapsulation, and (e) high mortality. endocarditis of the left-sided valves due to Staph. aureus (148).

FIGURE 31.4 Pathology of brain abscess. A: Deep white matter abscess (asterisk) adjacent to the body of
the lateral ventricle. B: Multiple brain abscesses at the junction between gray and white matter (arrows).
C: Gross photograph of a large, chronic temporal lobe abscess (asterisk). The location of this abscess is
immediately above the petrous temporal bone.

Scheld_Ch31.indd 528 2/21/14 5:46 PM


Chapter 31: Brain Abscess 529

FIGURE 31.5 Brain abscess in endocarditis. MRI of Staphylococcus aureus endocarditisassociated brain
abscess in a 75-year-old patient. A: The FLAIR-sequence shows perifocal edema (arrow) and (B) contrast
enhancement indicates the capsule of the abscess (arrow). C: The diffusion-weighted imaging shows
increased signal intensity within the abscess cavity (arrow).

The development of a brain abscess depends on the level and Other Foci of Metastatic Spread
duration of the bacteremia, the virulence of the organism, and
the occurrence of preceding emboli. A greater incidence of Other distant foci of infection may be associated with brain
brain abscess or meningitis is observed in cases of acute, rather abscess and have included wound and skin infections, osteomy-
than subacute bacterial endocarditis (Fig. 31.5). elitis, pelvic infection, cholecystitis, and other forms of intraab-
dominal sepsis. Brain abscess has also been described as an
unusual complication of esophageal dilation of caustic strictures
Right-to-Left Shunt and endoscopic ligation of varices (164,165). Brain abscess in
Abnormal venous-arterial channels which increase the risk of these cases is probably due to bacteria of the oropharyngeal
developing a brain abscess probably because of septic micro- flora that entered into the circulation through lesions of the mu-
emboli that can escape the pulmonary capillary filter (right- cous membrane. Transient bacteremia develops in up to 100%
to-left shunt) include pulmonary arteriovenous malformations of cases of esophageal dilation (mostly due to viridans strepto-
(AVMs), congenital cardiac defects, patent ductus arterio- cocci, Staph. aureus, and Staph. epidermidis) and in up to 50%
sus, and patent foramen ovale (PFO) (149,150). Hereditary of cases involving sclerotherapy (mostly due to Pseudomonas
hemorrhagic telangiectasia (Rendu-Osler-Weber disease) is species and -hemolytic streptococci) (164,166).
complicated by brain abscess, with striking regularity (151).
Approximately 25% of patients with this condition have
pulmonary AVMs and 70% of the patients with pulmonary
Location
AVMs have hereditary hemorrhagic telangiectasia (152). The In approximate decreasing order of frequency, a solitary abscess
incidence of brain abscess seems to increase with the num- may involve the following brain regions: frontal, temporal,
ber of pulmonary AVMs (Table 31.1). Clinical findings, such parietal, and occipital lobes and cerebellum (Table 31.5). This
as cyanosis, clubbing, polycythemia, and hypoxemia, may
be absent (asymptomatic AVMs). The diagnostic workup of
patients with brain abscess should therefore include a search TA B L E 3 1 . 5
for pulmonary AVMs (153). Congenital cyanotic heart disease
(CCHD) was found in 0% to 10% of patients with brain ab- LOCALIZATION OF BRAIN ABSCESSES (ACCORDING
scess (122,154). Some pediatric series suggest that CCHD is TO SELECTED STUDIES FROM THE COMPUTED
the most common cause of brain abscess in children (150,155). TOMOGRAPHY ERA [20,122,132,154,218,313])
In many studies, 50% or more of all brain abscesses were at-
tributed to CCHD in this age-group (155160), and in two Localization Mean Frequency (Range)
large series of brain abscess in those with cyanotic heart dis-
Frontal lobe 37% (1949%)
ease, more than 50% of patients were younger than 10 years
of age (161,162). Tetralogy of Fallot is consistently the most Temporal lobe 24% (1841%)
commonly cited defect (161,162). Transposition of the great Parietal lobe 22% (746%)
arteries, double outlet right ventricle, single ventricle, tricuspid Occipital lobe 10% (326%)
atresia, Ebstein disease, and Eisenmenger complex were the
Cerebellum 7% (010%)
next most frequently observed disorders in the largest series
(161). Several reports showed PFO in patients with brain ab- Sella 1% (04%)
scesses (163). However, the prevalence of PFO in adults is high Basal ganglia 1% (07%)
(20% to 35%). Therefore, a population study will be neces-
sary to document whether the prevalence of PFO is higher in 79% (range 6290%) of the 495 patients had single and 21% (range
patients with brain abscess with a presumed spread from a 1038%) had multiple abscesses. Therefore, the total exceeds 100%.
distant infectious focus than in the general population.

Scheld_Ch31.indd 529 2/21/14 5:46 PM


530 Part III: Bacterial and Mycoplasmal Infections

FIGURE 31.6 Brainstem abscess. Brainstem abscess


which presents (A) as a ring-enhancing lesion (arrow)
on T1-weighted image with contrast enhancement.
B: On diffusion-weighted imaging, the lesion appears
hyperintense (asterisk). Fusobacterium nucleatum
was isolated from specimen collected by stereotactic
biopsy.

distribution reflects the associated, often contiguous, focus of cochlear, and vestibular aqueducts) or between temporal su-
infection (Table 31.3). Intrasellar, brainstem, basal ganglia, ture lines. Hematogenous dissemination is occasionally im-
and thalamic abscesses are rare. plicated with contiguous foci, particularly in cases of sinus
or odontogenic origin. None of these hypotheses explains the
Pituitary Abscesses relative rarity of intracranial infection with sinusitis or oti-
tis, how bacteria traverse an intact dura, or what determines
Pituitary abscess is caused either by hematogenous seeding of
the form of intracranial complication that eventually evolves
the pituitary gland or by direct extension of an adjacent infec-
(e.g., meningitis, epidural abscess, subdural empyema, or
tion, such as meningitis, sphenoid sinusitis, cavernous sinus
brain abscess) in the individual with the same predisposing
thrombophlebitis, or a contaminated CSF fistula (167). Besides
condition.
infection by pyogenic bacteria, pituitary infections have been
The development of a brain abscess in the setting of neo-
caused by Treponema pallidum, M. tuberculosis, Entamoeba
natal meningitis deserves special comment. The pathogenesis
histolytica, and a variety of fungi, including Histoplasma
appears to follow the initiation of a necrotizing vasculitis
capsulatum, Blastomyces dermatitidis, Sporothrix schenckii,
(particularly in the small penetrating vessels), leading to sub-
Candida species, and Aspergillus species (167,168).
sequent hemorrhagic necrosis and liquefaction of the subcor-
tical white matter, an area highly susceptible to changes in
Brainstem Abscesses cerebral perfusion (174). Others disagree with this formula-
Brainstem abscesses (Fig. 31.6) often arise from hematogenous tion, contending that the evidence favors an initial ventricu-
spread or, less frequently, in association with a contiguous infec- litis followed by ependymal disruption and subsequent direct
tion, such as otitis (169,170). Brainstem abscesses have a rela- extension of the infection into the brain parenchyma. Similar
tively poor prognosis. This is primarily because their anatomic findings have been noted in an infant rat model of Citrobacter
location can lead to catastrophic neurologic complications and diversus meningitis and brain abscess (175) and support the
often preclude definitive drainage. Brainstem abscesses tend to latter hypothesis.
elongate in the brainstem rather than expand laterally (170). In hematogenous cases, the polycythemia and systemic hy-
Therefore, the clinical findings can be confusing. poxia observed in CCHD and hereditary hemorrhagic telangi-
ectasia increase blood viscosity, with an associated reduction
Abscesses in the Thalamus and Basal Ganglia in brain capillary flow, perhaps leading to microinfarction and
Inflammatory lesions (especially when solitary) of the thala- reduced tissue oxygenation in the brain. Septic microemboli
mus and basal ganglia are also somewhat unusual (Table 31.5). that escape the pulmonary capillary filter through right-to-left
Most often they are hematogenous (171) and they are fre- shunts during bacteremic episodes might then seed already
quently associated with hydrocephalus, most likely due to close compromised brain tissue. This hypothesis is in good agree-
proximity to the ventricles (172). ment with the development of abscesses in focal brain damage
due to, for example, brain tumor, hemorrhage, or ischemia.
In 1997, a review reported 15 cases of abscess formation in
General Pathogenetic Considerations brain neoplasms (176). Six of the brain abscesses developed in
a meningioma, glioma, or craniopharyngioma and were asso-
The final common pathway for brain abscess development in ciated with bacteremia due to Salmonella typhi, Staph. aureus,
the preceding conditions appears to require a compromised Bacteroides oralis, or E. coli (most of the other abscesses de-
area of brain. Experimental data suggest that infection is veloped in pituitary tumors or craniopharyngiomas and were
extremely difficult to establish in normal brain tissue (173). attributed to contiguous spread from an adjacent infected
Brain abscess may develop from a contiguous infection via di- sinus). It was proposed that tumor-associated bloodbrain
rect extension through areas of associated osteitis or osteomy- barrier breaching favored the development of hematogenous
elitis and retrograde thrombophlebitic spread via diploic or brain abscesses as eight such cases of abscess development at
emissary veins into the intracranial compartment. Additional the site of an intracerebral hemorrhage were reported (21).
possibilities in the case of otogenic infection include spread A similar mechanism has been proposed for the observation of
through preexisting channels (the internal auditory canal, brain abscess formation in ischemic brain tissue (177,178) and

Scheld_Ch31.indd 530 2/21/14 5:46 PM


Chapter 31: Brain Abscess 531

after endovascular treatments of cerebral aneurysms, AVMs,


or dural arteriovenous fistulas (38,179,180).
The immune response that is elicited to combat the invad-
ing pathogen is an essential part of abscess formation as it also
destroys surrounding normal brain tissue (181). This is sug-
gested by the finding that lesions of the brain are much greater
than the area of bacterial growth in experimental brain ab-
scesses (182,183). The most extensive work on the pathogen-
esis of brain abscess has been performed in a mouse model of
S. aureus brain abscess. S. aureus has been shown to activate
parenchymal microglia and astrocytes (in a toll-like receptor
2 [TLR2], TLR4, and MyD88-dependent fashion [184,185]).
In turn, they produce a variety of cytokines and chemokines
such as interleukin-1, tumor necrosis factor- (TNF-), and
macrophage inflammatory protein-2 (MIP-2) (186189) that
attract peripheral inflammatory cells. If the initial immune re-
sponse is unable to clear the infection (which can be related
to the presence of bacterial virulence factors such as -toxins
in S. aureusrelated brain abscess or the result of an insuffi-
cient immune responses), a continuing inflammatory response
is initiated and upheld. Among the stimuli for such an on-
going inflammation are bacterial toxins (pathogen-associated
molecular patterns [PAMPs]) and endogenous agents that are
set free as a consequence of tissue damage (damage-associated
molecular patterns [DAMPs]) and stimulate the immune re-
sponse (189,190). As a consequence, the development of a
chronic disruption of the bloodbrain barrier and final abscess
formation is found. These experimental findings support the
idea that an intervention with both antiinfective and antiin-
flammatory compounds might be an effective strategy to mini-
mize damage to brain parenchyma in brain abscess (181). FIGURE 31.8 Histology of advanced brain abscess. A: Photomicrograph
of an abscess wall during a later stage of abscess development. Thick
collagen fibers (arrows) are seen within the capsule wall. B: Trichrome
PATHOLOGY stain photomicrograph of abscess wall. Thick collagen fibers (arrows)
are seen above the inflammatory mass.

Histopathologic Stages of Brain Abscess


Development In the second stage of late cerebritis (days 4 to 9), develop-
ment of a well-formed necrotic center reaches its maximum
The evolution of a brain abscess includes four histopathologic size. Additionally, fibroblasts appear, setting the stage for
stages (191). This staging process, described in animal mod- capsule formation and a marked increase in neovascularity at
els of brain abscess, correlates well with human brain abscess the periphery of the necrotic zone. These newly formed capil-
evolution (192). laries often lack tight junctions and leak proteinaceous fluid.
The first stage is an early cerebritis (days 1 to 3 following Surrounding this is the beginning of a reactive astrocyte re-
intracerebral inoculation in animals), which progresses to a sponse, along with persistent white matter edema.
perivascular inflammatory response surrounding the developing The third stage, early capsule formation (days 10 to 13),
necrotic center by the third day. Profound edema in the sur- is characterized by a slight decrease in the size of the necrotic
rounding white matter develops concurrently (Fig. 31.7). center (Fig. 31.8). At this point, there is a well-developed layer

FIGURE 31.7 Histology of early brain abscess. A: Wall of brain abscess. Normal white matter is seen
to the left, and there is an intermediate area of edematous brain (asterisk) adjacent to the abscess wall.
B: Higher magnification showing periabscess inflammatory exudates (asterisk). There is relatively little
encapsulation at this early stage. C: Higher magnification of abscess wall. Note the combination of
increased gliosis and inflammatory cells (asterisk) adjacent to the abscess.

Scheld_Ch31.indd 531 2/21/14 5:46 PM


532 Part III: Bacterial and Mycoplasmal Infections

of fibroblasts, with significantly more reticulin deposition on oxygen concentration, the offending organism, and the hosts
the cortical side than on the ventricular side of the lesion. immune response.
Outside this developing capsule is a region of persistent cere-
britis and neovascularity, with a further increase in reactive
astrocytes. CLINICAL MANIFESTATIONS
These processes continue in the fourth and final stage, late
capsule formation (day 14 and later). The capsule continues
to thicken, with an abundance of reactive collagen present by Signs and Symptoms
the third week.
Some studies have criticized the preceding models util- The clinical manifestations of a brain abscess (Table 31.6)
ity in describing a uniform mode of brain abscess evolution can vary greatly among patients. Additionally, symptoms
(193). These authors, using the same dog model and inocu- from the primary site of infection (i.e., otitis, sinusitis, or
lum, could not detect viable organisms in the brain lesions distant suppurative foci) may predominate. The evolution
after 3 days and, in all animals, the lesions spontaneously of symptoms of a brain abscess may range from indolent to
resolved. Further work is necessary to reconcile this debate. fulminant; the average duration of symptoms until hospital
Nonetheless, a brain abscess model in rats supports the pre- admission is 11 to 12 days (range, hours to several weeks)
viously mentioned histologic progression (194). Two other (122,132). The classic triad of fever, headache, and a focal
experimental models of brain abscess using organisms other neurologic deficit is present in less than 50% of patients.
than -hemolytic streptococci also indicate that this view In most patients, the prominent clinical manifestations of
of abscess evolution is either overly stereotyped or affected brain abscess are due to an expanding intracerebral mass
by inadequate growth of the microorganisms. In a model rather than infection; neurologic signs, such as headache and
of Bacteroides fragilis (inoculated with Staph. epidermi- hemiparesis, predominate in patients with multiloculated
dis) brain abscess (195), the same stages of evolution were brain abscess as compared with patients with uniloculated
observed; however, the early and late capsule stages could brain abscess (199). The clinical presentation of patients with
not be differentiated, because of a delay in encapsulation cerebellar abscesses is different, because signs of raised in-
when compared to abscesses following streptococcal chal- tracranial pressure (ICP) (e.g., headache, vomiting, and pap-
lenge. These abscesses enlarged more quickly, were prone illedema, 90%), fever (90%), meningismus (70%), and
to early ventricular rupture (25%), and exhibited incom- depressed consciousness (50%) are particularly common
plete encapsulation. This suggests the extreme virulence of (118,200). Cerebellar signs are present in 25% to 50% of
B. fragilis, an important pathogen in brain abscess, in brain patients, whereas hemipareses are evident in only 10% of pa-
tissue when part of a mixed infection. In an experimental tients. The clinical presentation of frontal lobe abscesses is
model using S. aureus (196), there were quantitative and often dominated by headache, drowsiness, inattention, and
qualitative differences in abscess evolution when compared a generalized deterioration in mental function. Hemiparesis
with inoculation of -streptococci. S. aureus inoculation with unilateral motor signs and a motor speech disorder are
resulted in larger lesions, earlier ependymitis, and delayed the most common focal findings. A temporal lobe abscess
progress toward healing, with longer periods required for may present with early ipsilateral headache. If the abscess
the infection to reach a stable size. The same approximate is in the dominant hemisphere, an aphasia or dysphasia may
stages were present, but their separation was also not as be present. An upper homonymous hemianopia may also be
distinct as previously described. The white matter appeared demonstrated and may be the only sign of a right temporal
more susceptible to destruction than the gray matter, and lobe abscess. Intrasellar abscesses often simulate a pituitary
the spread of infection was centrifugal, rather than along tumor, presenting with headache (90%), abnormal pitu-
particular white matter tracts. In contradistinction, a model itary function (50%), and visual field defects (50%) (167).
of E. coli brain abscess exhibited expansion of the abscess The clinical presentation of brainstem abscess is usually a
along white matter tracts (197). In addition, results from rapidly progressing neurologic deficit involving cranial nerves
the S. aureus experimental model raised some questions and long tracts.
regarding the previously held assumption that the capsule
serves to contain infection. The abscess reached its maxi-
mum size in the late cerebritis stage, before any significant
capsule formation had taken place, suggesting that the host
was able to contain the infection before capsule formation. TA B L E 3 1 . 6
Also, even in the late capsule stage, inflammation, necrosis,
PRESENTING SIGNS AND SYMPTOMS IN 968
and edema extended well beyond the well-formed capsule.
PATIENTS WITH BRAIN ABSCESS (AFTER [20,122,126,
Two observations regarding encapsulation deserve special
127,218,234,299])
attention: (a) capsule formation is usually more complete on
the cortical side of the abscess than on the ventricular side Sign or Symptom Frequency Range (Mean)
(191), and (b) encapsulation is less extensive in abscesses
resulting from hematogenous spread than in those arising Headache 5597% (77%)
from a contiguous focus of infection (198). The preferential
Impaired consciousness 2891% (53%)
deposition of collagen on the outer edge of the abscess is
thought to be due to the better vascularization of the gray Fever 3262% (53%)
matter (197). This discrepancy probably explains the pro- Nausea with vomiting 3585% (51%)
pensity for abscesses to rupture medially rather than into the Papilledema 956% (39%)
subarachnoid space. Similarly, cerebral ischemia from a sep-
Hemiparesis 2344% (36%)
tic embolus might impede optimal collagen formation (198).
Therefore, brain abscess formation is a continuum from Seizures 1335% (24%)
cerebritis to a collagen-encapsulated necrotic focus; however, Neck stiffness 541% (23%)
its maturity is dependent on many factors, including local

Scheld_Ch31.indd 532 2/21/14 5:46 PM


Chapter 31: Brain Abscess 533

FIGURE 31.9 Cerebral metastasis as differential diagnosis of brain abscess. Cystic lesion in a 66-year-
old patient that appears (A) hyperintense on T2-weighted imaging (asterisk) and (B) shows only a very
distinct contrast enhancement of the rim (arrow). C: Diffusion-weighted imaging is negative (asterisk).
The lesion was diagnosed as a metastasis of a pulmonary adenocarcinoma.

diagnosis is primary CNS lymphoma, followed by brain ab-


Differential Diagnosis scesses due to pathogens other than Toxoplasma (Table 31.3),
metastatic tumors, and cerebrovascular disease (202).
Because of their nonspecific presentation and frequent lack of
fever, brain abscesses can be confused with several other pro-
cesses. The major differential diagnoses of a brain abscess in-
clude primary (glioblastoma) or metastatic malignancies of the
APPROACH TO A DIAGNOSIS
CNS (Figs. 31.9 and 31.10). Furthermore, differential diagnoses
may include other infectious diseases, including viral encephali- Laboratory Investigations
tis (especially due to herpes simplex virus), subdural empyema
and epidural abscess, or mycotic aneurysms in the setting of in- Examination of the blood is rarely helpful in the diagnosis of a
fective endocarditis. Cerebral venous sinus thrombosis, cerebral brain abscess (Table 31.7). The serum C-reactive protein (CRP)
infarction, resolving hematoma, arterial infarction in the stage values are more commonly elevated than the blood leukocyte
of luxury perfusion, AVM, neurosarcoidosis, and tumefactive counts or erythrocyte sedimentation rate (ESR). However, up to
multiple sclerosis may also be confused with a brain abscess, 23% of patients in previous studies had a normal CRP serum con-
both symptomatically and occasionally radiographically (201). centration. Blood cultures are infrequently positive (about 10%) at
In patients with AIDS, the most common cause of intracranial the time of presentation. However, blood cultures were reported to
mass lesions is toxoplasmosis; the most important differential be positive in 86% of patients with listerial brain abscesses (203).

FIGURE 31.10 Glioblastoma multiforme as differential diagnosis of brain abscess. Two cerebral lesions
in a 56-year-old HIV-positive patient with a CD4 count of 1000 cells/L and negative anti-Toxoplasma
antibodies. A: The lesions are hyperintense on the T2-weighted image (arrows) and (B) show ring-shaped
contrast enhancement (arrows). C: Diffusion-weighted imaging is isointense in the center of the lesion
(arrows). Glioblastoma multiforme was diagnosed after biopsy.

Scheld_Ch31.indd 533 2/21/14 5:46 PM


534 Part III: Bacterial and Mycoplasmal Infections

TA B L E 3 1 . 7
APPROACH TO A DIAGNOSIS OF BRAIN ABSCESS AND POSSIBLE UNDERLYING CONDITIONS

Method Possible Results References

Cranial CT/MRI Cerebritis: focal area of edema (high signal on T2-, low signal on T1-weighted 190, 220, 261
with contrast images, hypodense on CT) with mass effect (sulcal effacement or ventricular
administration compression) and minimal/heterogeneous enhancement
Abscess: necrotic center (pus) is usually isodense with CSF on CT and
hyperintense on MRI; capsule can be iso- or hyperintense on T1- and
hypointense on T2-weighted images; edema around the abscess cavity; mass
effect, ring-enhancement on CT or MRI
Complications: edema, hemorrhage, hydrocephalus, ventricular rupture?
Evidence for source of infection: sinusitis, otitis, mastoiditis, fracture, osseus
dehiscence, congenital malformation, intracranial foreign body?
Diffusion-weighted Abscess fluid is hyperintense on DW MRI (neoplastic cystic brain lesions are 190
(DW) MRI hypointense); apparent diffusion coefficient values are decreased in brain
abscesses (and increased in neoplastic cystic brain lesions)
Stereotactic abscess Microscopy; aerobic and anaerobic cultures: positive in 84% (mean, range 28, 122, 132, 154, 218, 299
aspiration 6997%); fungal, mycobacterial, and nocardial cultures (on indication)
Blood cultures Aerobic and anaerobic cultures: positive in 3 (11%) of 27 patients in one study 122
Laboratory studies Blood leukocyte count: elevated in 47% (mean, range 3057%) 4, 122, 132, 218, 299
Erythrocyte sedimentation rate: elevated in 59% (mean, range 4865%) 4, 218, 299
Serum C-reactive protein: elevated in 82% (mean, range 7790%) 204, 314316
Serologic test for HIV (on indication)
Echocardiography Right-to-left shunt, valvular vegetations (endocarditis)
Chest x-ray/CT of Pulmonary infection: abscess, pneumonia, empyema, bronchiectasis,
the chest aspergilloma
Pulmonary arteriovenous malformation
Dental (panoramic) Dental infection
radiography

Lumbar puncture is also of very limited value for the neoplasms (201). Cranial CT is widely available and often fa-
diagnosis, most often reveals nonspecific CSF alterations, and cilitates early detection of brain abscess. For the detection of
carries a considerable risk of cerebral herniation; therefore, it a brain abscess, the CT must be performed with intravenous
should not be performed in patients with a space-occupying contrast administration. During early brain abscess, a nonen-
brain abscess. Cerebral herniation developed immediately hancing hypodensity is usually found. However, it has to be
after lumbar puncture in 2.1% of patients (mean, range 0% kept in mind that in the earlier phases of brain abscess, CT
to 4.2%) (126,127,132,204,205), and neurologic deteriora- may show only low attenuation abnormalities with mass effect
tion was observed in 13.5% of 193 patients with brain abscess (Fig. 31.11) or even fail to detect a lesion at all (Fig. 31.14).
who received a lumbar puncture (9). Positive CSF cultures, This is especially true in brainstem abscesses. During later
however, were reported in only 6% of patients (mean, range stages, when bloodbrain barrier breaching progresses, con-
0% to 10%) (4,122,127,132), unless there has been a rup- trast enhancement and finally a thick ring-enhancing lesion is
ture of abscesses into the ventricles or subarachnoid space, in found on CT, representing the development of an inflamma-
which case up to 20% may be positive (122). The CSF cell tory capsule (Figs. 31.12 and 31.13). During later stages, the
count can range from 0 to more than 100,000 cells/L, the contrast-enhancing ring becomes thinner. Features thought to
protein level from normal to more than 500 mg/dL, and CSF discriminate abscesses from malignant tumors (thinner, more
glucose concentration can be normal or severely depressed regular contrast-enhancing rim and homogeneous enhance-
(122,132). ment of the capsule after infusion of contrast medium) do not
always permit a precise diagnosis.
The value of MRI in differentiating brain abscess from
Neuroimaging Studies other cystic lesions of the brain, such as tumor, has increased
in the past years. Compared to CT, MRI is more sensitive in
Neuroimaging is the most important first step in establish- the detection of early cerebritis and small satellite lesions.
ing the diagnosis of brain abscess. Cranial CT (Table 31.7, Furthermore, MRI allows estimation of the degree of central
Figs. 31.11 to 31.14) and in particular MRI (Table 31.7, necrosis, ring enhancement, and surrounding cerebral edema
Figs. 31.2, 31.3, 31.5, and 31.15 to 31.17) are very sensitive more accurately than CT. MRI is also better suited for the de-
neuroimaging modalities for the detection of brain abscesses tection of brain abscess in the brainstem. In contrast to tumors,
and yield precise anatomic information. However, this is not increased signal in diffusion-weighted imaging (DWI) at re-
paralleled by an equivalent specificity; a similar appearance is duced apparent diffusion coefficient (ADC) values are found
occasionally seen with other cystic brain lesions, in particular in brain abscesses (Figs. 31.9 and 31.10) (206209). In a study

Scheld_Ch31.indd 534 2/21/14 5:46 PM


Chapter 31: Brain Abscess 535

FIGURE 31.11 Frontotemporoparietal space-occupying brain abscess. A: Unenhanced CT scan showing


a large right frontotemporoparietal mass lesion (asterisk) that is causing marked right-to-left shift of mid-
line structures and effacement of the ipsilateral lateral ventricle. B: Contrast-enhanced CT scan showing
a thin-rimmed, homogeneous enhancement (arrow) surrounding an area of decreased attenuation within
the center of the abscess. C: Unenhanced CT scan, performed 6 weeks after surgical aspiration of the
abscess and antibiotic therapy, showing resolution of the mass effect. D: Enhanced scan showing minimal
contrast enhancement in the temporal lobe (arrow).

FIGURE 31.12 Multiloculated brain abscesses in sepsis. Multiple


cerebral abscesses due to hematogenous spread of Staphylococcus
aureus in a patient with sepsis. Lesions are seen in periventricular
locations (A, white arrow), in the centrum semiovale (B, white
arrow), and at the junction between gray and white matter (B,
dotted arrow).

FIGURE 31.13 Occipital lobe abscess. A: Unen-


hanced axial CT scan showing a hypodense occip-
ital lesion (arrow). B: After contrast enhancement,
a ring-enhancing lesion (arrow) was found on
T1-weighted MRI. Furthermore, an additional
cortical contrast enhancing lesion was found
(arrowhead).

Scheld_Ch31.indd 535 2/21/14 5:46 PM


536 Part III: Bacterial and Mycoplasmal Infections

FIGURE 31.14 Presentation of differ-


ent stages of brain abscess on CT. On
CT, during the early stage of a brain
abscess, (A) a hypodense area is seen
(asterisk) with (B) only slight contrast
enhancement (arrow). Three weeks
later, (C) the hypodense area has in-
creased in size (asterisk) and (D) the
abscess wall can be seen after admin-
istration of contrast (arrow). Causative
pathogen: Staphylococcus aureus.

FIGURE 31.15 Presentation of brain abscess on CT and MRI. Parietooccipital brain abscess in a 42-year-
old female appears (A) as a hypodense lesion (asterisk) on native CT. On MRI, (B) the edema (arrow) sur-
rounding the abscess appears hyperintense and (C) contrast enhancement visualizes the capsule (arrow).
D: On diffusion-weighted imaging, the abscess cavity appears hyperintense (arrow). Streptococcus inter-
medius and Aggregatibacter aphrophilus were identified as causative pathogens.

Scheld_Ch31.indd 536 2/21/14 5:46 PM


Chapter 31: Brain Abscess 537

perfusion-weighted MR measurements (of the peripheral re-


gions of cystic lesions of the brain) to distinguish brain abscess
from tumor (214). Possibly, a combination of MR spectros-
copy, DTI, and perfusion-weighted MR might be helpful to
further increase specificity of MR in diagnosing brain abscess
in the future.

Biopsy
The most important diagnostic measure in the establishment
of the diagnosis of brain abscess is histologic and microbio-
logic assessment of tissue from the suspicious lesion. This
not only allows differentiation between tumor and abscess
but also identification of the causative pathogen. Thus, a
specimen obtained from biopsy of cystic lesions should (a) be
sent for histologic evaluation and (b) be evaluated by Gram
stain and aerobic, anaerobic, mycobacterial, and fungal cul-
tures (Fig. 31.18). Furthermore, staining for mycobacteria,
Nocardia, and fungi should be performed. In recent studies,
16S ribosomal DNA polymerase chain reaction amplification
was shown to increase the number of bacterial species isolated
FIGURE 31.16 Nocardia farcinica brain abscess. T1-weighted MRI from brain abscesses (215217), but further data are needed
with contrast shows multiple parenchymal ring-enhancing lesions in to determine its use in identification of important causative
a 39-year-old patient after kidney transplantation. Nocardia farcinica pathogens in patients with brain abscess. Identification of the
was isolated from brain biopsy material. causative organism(s) is especially crucial for further therapy
(see the following discussion).
of 147 cystic lesions of the brain in 115 patients, the sensitivity
and specificity of DWI for the differentiation of brain abscesses
from non-abscesses were 96% each (208). This does not hold TREATMENT
true for all cases of cerebral toxoplasmosis where MRI, and
especially DWI, findings differ depending on the stage of the Treatment of brain abscess is based on the following principles:
disease (Fig. 31.17) (210). Although less readily available, an (a) surgical therapy of brain abscess, (b) antibiotic therapy, and
increased fractional anisotropy (FA) was found in the cavity of (c) surgical therapy of the primary infectious focus. Therefore,
abscesses using diffusion tensor imaging (DTI), allowing dis- optimal medical care of brain abscess requires the cooperation
crimination of abscesses from tumors (211). Using magnetic between an infectious disease specialist, neurologist, neurora-
resonance (MR) spectroscopy, resonances for acetate, lactate, diologist, neurosurgeon, microbiologist, and sometimes an ear,
alanine, succinate, and pyruvate as well as valine, leucine, nose, and throat specialist, surgeon, dentist, or maxillary sur-
and isoleucine were found elevated in brain abscess but not in geon. Cranial CT or MRI allows a precise localization of the
tumors (212,213). Recent studies suggest an additional role of abscess or, when multiple abscesses are present, identification of

FIGURE 31.17 Cerebral toxoplasmosis. A: T2-weighted image shows a left frontal lesion (asterisk) with
large edema causing midline shift (arrow). A smaller hyperintense lesion was found in the occipital lobe
(dotted arrow). B: The large frontal lesion shows contrast enhancement of the capsule wall (arrow).
C: Diffusion-weighted imaging is isointense at the area of the frontal lesion (asterisk) and hyperintense
at the small occipital T2-weighted hyperintense lesion (arrow). Toxoplasma gondii was identified as
causative pathogen in this HIV-positive 35-year-old patient (CD4 count: 50 cells/L).

Scheld_Ch31.indd 537 2/21/14 5:46 PM


538 Part III: Bacterial and Mycoplasmal Infections

FIGURE 31.18 Diagnostic abscess aspiration. A 56-year-old HIV-positive patient with a CD4 count
of 40 cells/L presented with fever and somnolence. A: T1-weighted MRI showed multiple contrast-
enhancing lesions that (B) were hyperintense on diffusion-weighted imaging. The largest lesion (asterisk)
was chosen for diagnostic stereotactic aspiration. After computed calculation of the most suitable approach
to avoid bleeding complications, (C) a stereotactic frame (pound sign) was attached to the patients head.
The skin at the area for the calculated access (arrowhead) was disinfected and prepared for a 1-cm long in-
cision. D: A burr hole of 2 mm in diameter was drilled. A perfect position of the drill (arrow) was ensured
through attachment of the drill to the stereotactic frame. E: Guided through a precalculated position on
the stereotactic frame, the abscess was carefully punctured with a needle (arrowheads) measuring 1 mm
in diameter. F, G: The cavity of the lesion was found liquid, and fluid was aspirated (o). Microbiologic
studies of the specimen revealed Aspergillus fumigatus. (CG courtesy of Prof. Dr. Friedrich-Wilhelm
Kreth, Department of Neurosurgery, Ludwig Maximilians University of Munich.)

the lesion that is most suited for stereotactic aspiration. In addi-


tion, sources of infection (e.g., otitis or sinusitis) can be detected. Surgical Therapy
Randomized controlled studies of therapies for brain abscess do
not exist. Treatment recommendations are, therefore, based on The timing of surgery and type of surgical procedure selected
retrospective analyses and clinical experience. Although doubted remain controversial and prospective studies are lacking.
by some authors (132), some major series have demonstrated Usually, aspiration or excision is used. Retrospective series
that the administration of preoperative antibiotics reduces the failed to detect any significant difference in mortality (or
yield of positive cultures (4,218). However, antimicrobial ther- functional neurologic outcome) between patients treated by
apy can be withheld until surgery only if the operation can be aspiration or excision (4,28,218,219). Nevertheless, the type
performed within a short time (hours) and the abscess does not of care (stereotactic aspiration plus medical treatment vs. op-
show a significant mass effect (risk of herniation). The risks of eration plus medical treatment vs. medical treatment alone)
stereotactic aspiration are probably less than the risks of incor- in these studies was often not based on clear and explicit
rect diagnosis and choice of antibiotics; therefore, the decision to data. However, there is consensus that bacterial brain abscess
use empirical therapy alone should be made with great caution. should be treated by surgery if larger than 2.5 cm and causing

Scheld_Ch31.indd 538 2/21/14 5:46 PM


Chapter 31: Brain Abscess 539

significant mass effect. However, it remains unclear what therefore, abnormal results of clotting studies or thrombocy-
type of operation should be favored. Interestingly, outcome topenia are particular contraindications for burr-hole aspira-
was more associated with clinical presentation and the speed tion and must be corrected.
of operation rather than the type of operation (14,132,220).
The choice of surgical treatment should, therefore, depend Excision
on the patients clinical status, the location and accessibility
Open craniotomy and total excision of the abscess may be-
of the abscess(es), the techniques available, and the surgeons
come necessary in all lesions that fail to respond to repeated
expertise.
aspirations and medical therapy (Fig. 31.20). Excision is also
necessary in brain abscesses secondary to foreign bodies, and
Stereotactic Aspiration some authors advocate that abscesses exhibiting gas by CT or
Because stereotactic aspiration causes less tissue damage than on plain film should also be completely excised, because an
excision, CT (or MRI)-guided stereotactic aspiration through open craniotomy will allow for the detection and treatment of
a burr hole is usually considered the method of choice in pa- an extracranial communication (224). Furthermore, patients
tients with brain abscess (Fig. 31.18). Stereotactic aspiration is with abscesses in the posterior cranial fossa are often prefera-
particularly valuable for deep-seated abscesses (brainstem, cer- bly treated by excision (223,225,226). Patients with posttrau-
ebellum, and basal ganglia), multiple abscesses, and abscesses matic or postoperative brain abscess have also been considered
in critical locations (Figs. 31.6 and 31.19), where high precision candidates for excision by most authorities (222,225).
is necessary (221223). In addition, patients who are poor can-
didates for general anesthesia might better be treated by aspi-
ration rather than operation. With large superficial abscesses Medical Therapy
of the cerebral hemispheres, CT-guided freehand burr-hole
aspiration may be performed as an emergency measure (in those Antibiotic therapy, usually in combination with surgery, is
with impending cerebral herniation). It requires less preparation important in the management of brain abscess. Because the
but is less precise than stereotactic aspiration. chance to detect the causative pathogen from specimens col-
Compared with excision, the disadvantage of aspiration lected during aspiration or excision of the abscess decreases
is that repeated aspirations or even excision may become in a time-dependent manner after antibiotic therapy has been
necessary. Another considerable risk is intracranial bleeding; started, immediate surgery followed by empirical antibiotic

FIGURE 31.19 Successful aspiration of


a cerebellar brain abscess. The 38-year-
old patient with a history of long-
standing injection drug abuse presented
with impaired consciousness and fever.
A: Diffusion-weighted MRI showed a
hyperintense lesion in the cerebellum
(asterisk) B: Histologic studies of brain
biopsy specimen showed a massive infil-
tration of inflammatory cells (arrows),
predominantly granulocytes (hematoxylin
and eosin [H&E] staining). C: Gram stain
revealed gram-positive cocci that pre-
sented in clusters (arrows). Microbiologic
investigations identified Streptococcus
anginosus. D: Follow-up MRI 18 days
after aspiration showed disappearance of
the hyperintensity on diffusion-weighted
imaging. (B, C courtesy of Prof. Dr. Armin
Giese, Center for Neuropathology and
Prion Research, Ludwig Maximilians
University of Munich.)

Scheld_Ch31.indd 539 2/21/14 5:46 PM


540 Part III: Bacterial and Mycoplasmal Infections

FIGURE 31.20 Open surgery in temporal brain abscess. A: T1-weighted MRI with contrast enhance-
ment shows right temporal brain abscess (asterisk) in a 52-year-old patient. Initial stereotactic aspiration
failed to identify a causative pathogen (possibly due to pretreatment of the patient). Despite broad antibi-
otic therapy, the size of the abscess increased, requiring open surgery. During surgery, (B) a hard abscess
wall (asterisk) was found. For orientation, please note the sylvian vein (black arrow). C: After the abscess
was opened, the hard abscess wall can be seen from inside (arrow) as well as pus (asterisk) inside the
abscess cavity. (B, C courtesy of Dr. Jan Mehrkens, Department of Neurosurgery, Ludwig Maximilians
University of Munich.)

therapy is recommended. However, if surgery is delayed, and cloxacillin were low. Metronidazole attains high concen-
empirical antibiotic therapy needs to be started before sur- trations in brain abscess pus (230), often exceeding serum
gery. Despite a lack of data, medical therapy alone might be concentrations after a dose of 400 to 800 mg every 8 hours.
an option in certain situations. However, by not performing Because of these results and the bactericidal activity of metro-
surgery, confirmation of the diagnosis and identification of the nidazole against strict anaerobes, this agent is an important
causative pathogen is important to be able to select the cor- component of antimicrobial regimens for brain abscesses.
rect antimicrobial therapy. In consequence, only few patients Vancomycin attained acceptable concentrations (for aerobic
are treated by medical therapy alone; for example, only 19 of streptococci and staphylococci) in the single brain abscess
973 patients received nonsurgical treatment in a study from studied (231). Cefotaxime, ceftazidime, ceftriaxone, and
South Africa (9). In retrospective case series, candidates that ampicillin/sulbactam were shown to be efficient in the treat-
seemed suitable for antibiotic therapy alone were patients with ment or to have an adequate penetration into brain abscesses
small abscesses (2.5 cm), in good clinical condition (Glasgow (232236). Imipenem/cilastatin has also been demonstrated to
Coma Scale 12), and in whom the etiology of the abscess penetrate at sufficient levels into brain abscess pus and to be
was established (170,227). Medical therapy alone can also effective (237240). However, because of the reported neu-
be considered in patients with a lesion in the cerebritis stage rotoxicity (increased incidence of seizures) of imipenem, me-
(which are much more likely to respond to antibiotic therapy ropenem is preferred; a successful outcome using this agent in
alone because of lack of a capsule) or surgically inaccessible the treatment of a brain abscess due to a multidrug-resistant
abscesses (in particular brainstem abscesses) (170). These pa- Enterobacter cloacae was shown (241). In the search for
tients should be treated with high-dose broad-spectrum intra- newer antibiotics, linezolid has been shown to be a promis-
venous antibiotics for at least 6 to 8 weeks and perhaps longer. ing agent for the treatment of CNS infections, having good
If the patient shows neurologic deterioration or serial CT penetration into the CSF (242,243). Quinolones, which have
scans show that the abscess does not decrease in size within been used with success, should be used with caution in the
several weeks or even grows, a surgical procedure should be treatment of cerebral abscesses, because they can also lower
reconsidered to confirm the diagnosis, collect material for cul- seizure thresholds (244).
ture, and remove as much pus as possible (220,228).
Empirical Antimicrobial Therapy
Suitable Antimicrobial Agents The antimicrobial regimens commonly recommended for the
Randomized controlled studies that compared the efficacy of therapy of brain abscess are empirical and reflect the consider-
different antibiotic regimens are lacking in patients with brain ations already noted, as well as the in vitro activity of the com-
abscess. Antibiotics used for the treatment of brain abscess ponent agent(s) against the usual pathogens. No controlled
should be administered intravenously, be active against the trials on the relative efficacy of various regimens have been
pathogens that have been identified as causative agents of the performed. Interpretation of the success or failure of a given
abscess or are likely in a given clinical scenario, penetrate into regimen is further confused by the various surgical procedures,
the abscess fluid (and into the site of the underlying infection) which may be used concomitantly.
in adequate concentrations, and have bactericidal activity In patients with community-acquired brain abscesses
(220). Additional therapeutic concerns include the effect of the (Tables 31.8 and 31.9), third-generation cephalosporins,
abscess environment on the activity of the antibiotic and the such as ceftriaxone or cefotaxime, which are directed against
possibility of negative drug interactions when multiple agents streptococci and a broad range of gram-negative bacteria are
are administered. In 32 patients with brain abscess, penicillin preferred for first-line empirical therapy. In addition, cover-
G was detectable consistently if the dose exceeded 24 million age of anaerobes (e.g., by metronidazole) and staphylococci
units daily (in adults) (229). Fusidic acid entered the brain (e.g., by flucloxacillin, nafcillin, or vancomycin) should be
abscess readily, but concentrations of various cephalosporins considered in appropriate circumstances. If meropenem is

Scheld_Ch31.indd 540 2/21/14 5:46 PM


Chapter 31: Brain Abscess 541

TA B L E 3 1 . 8
EMPIRICAL ANTIMICROBIAL THERAPY OF BRAIN ABSCESS

Underlying Condition Recommended Antimicrobial Regimen

Community acquired (immunocompetent patients) Third-generation cephalosporina,b metronidazole


Postoperative or posttraumatic Meropenem (or cefepime) vancomycin

a
Ceftriaxone or cefotaxime; for Pseudomonas species: ceftazidime aminoglycoside, meropenem aminoglycoside, cefepime aminoglycoside,
or ciprofloxacin.
b
If staphylococci are suspected: plus vancomycin (alternatives: rifampicin, nafcillin). Linezolid (1200 mg per day in 2 divided doses [600 mg IV every
12 hours] intravenously in adults) has an activity comparable to that of vancomycin but has a much better bloodCSF penetration (however, clinical
reports on the use of linezolid in CNS infections are very limited). It needs to be kept in mind that administration of linezolid for the prolonged periods
required for brain abscess therapy may lead to significant side effects such as bone marrow suppression, optic neuropathy, and polyneuropathy.

used, the addition of metronidazole is not necessary because TA B L E 3 1 . 9


meropenem is active against anaerobic bacteria itself. Patients RECOMMENDED DOSAGES FOR ANTIMICROBIAL
with otogenic brain abscesses should be treated initially with THERAPY OF BRAIN ABSCESSES IN ADULTS
ceftazidime or cefepime, because these cephalosporins are also (DOSING MAY NEED ADJUSTMENT IN PATIENTS
effective against Pseudomonas aeruginosa (Table 31.8). WITH UNDERLYING RENAL OR LIVER DISEASE)
Because patients with posttraumatic brain abscess or noso-
comial brain abscess are at risk for infection with MRSA and Antimicrobial Agent Dosage
multiresistant Enterobacteriaceae, meropenem or cefepime
plus vancomycin is the recommended empirical treatment in Amphotericin B 0.61.25 (to 1.5) mg/kg i.v.a
such cases (Table 31.8). Bone marrow, stem cell, and solid deoxycholate q24h
organ transplant recipients with brain abscesses often require
Liposomal amphotericin B 5 mg/kg i.v. q24h
initial polypragmatic therapy including meropenem, vanco-
mycin, voriconazole, and anti-Toxoplasma therapy to treat a Ampicillin 2 g i.v. q4h
wide variety of potential infectious pathogens. In all cases, the Cefepime 2 g i.v. q8h
empirical treatment should be modified or extended, if labora- Cefotaxime 34 g i.v. q8h
tory, clinical, or radiologic findings are suggestive of a specific
Ceftazidime 2 g i.v. q8h
pathogen. After culture results are obtained, therapy needs
to be modified according to the identified pathogen(s) and in Ceftriaxone 2 g i.v. q12h
vitro susceptibility testing. Clindamycin 600 mg i.v. q6h
The usual treatment of choice for CNS candidiasis is the Nafcillin 2 g i.v. q4h
combination of intravenous liposomal amphotericin B and oral
Fluconazole 400800 mg i.v. or p.o. q24h
flucytosine (90,245). Cryptococcal CNS disease, best studied in
patients with cryptococcal meningitis, is usually treated with a Flucytosine 25 mg/kg p.o. q6h
combination therapy consisting of liposomal amphotericin B Fosfomycin 5 g i.v. q8h
and flucytosine (induction therapy). After 2 weeks, consolida- Itraconazole 200400 mg p.o.b q12h
tion therapy with fluconazole (400 mg per day) should be given
Linezolid 600 mg i.v. q12h
for an additional 8 to 10 weeks. Then, immunosuppressed pa-
tients should receive fluconazole (200 to 400 mg per day) for an Meropenem 2 g i.v. q8h
additional 6 to 12 months (suppression) (246). In HIV-positive Metronidazole 500 mg i.v. q68h
patients, suppressive therapy is continued until CD4 cells are Nafcillin 2 g i.v. q4h
maintained above 150 to 200 cells/L for more than 6 months
in patients on antiretroviral therapy. CNS coccidioidomycosis Penicillin G 4 106 U i.v. q4h
should be treated with fluconazole or, alternatively, itraconazole Pyrimethamine 50 mg p.o. q24hc
(247), and patients with CNS blastomycosis should be treated Sulfadiazine 68 g p.o. q24h
with liposomal amphotericin B (248). CNS histoplasmosis Rifampin 10 mg/kg i.v. (up to 600 mg) q24h
should be treated initially with amphotericin B for 3 months,
then with itraconazole for 12 months (249). In the treatment of Trimethoprim 160 mg i.v. q68h
cerebral mucormycosis, amphotericin B is the only antifungal ( sulfamethoxazole)
agent with proven in vivo activity (250). Actinomycosis of the Voriconazole 4 mg/kg i.v. q12hd
CNS is often treated with high-dose intravenous penicillin for 2 Vancomycin 15 mg/kg i.v.e q812h
to 6 weeks, followed by oral therapy with amoxicillin, ampicil-
lin, or penicillin V for 6 to 12 months. Doxycycline, erythro- i.v., intravenously; p.o., orally.
mycin, and clindamycin can be given alternatively according to a
High dosages are recommended in particular for the treatment of
sensitivity test results. aspergillosis.
b
Instillation of antibiotics into the abscess cavity during Possible alternative to amphotericin B; poor penetration of the
aspiration has been employed, but the efficacy of this practice bloodCSF barrier.
c
Loading dose: 50200 mg p.o., 1015 mg/day folinic acid should be
has never been established. Antibiotics given in this manner given to decrease bone marrow suppression from pyrimethamine.
may diffuse into surrounding brain tissue in high concentra- d
Loading dose 2 6 mg/kg on the first day.
tions, causing seizures. It should, therefore, be used with cau- e
Need to monitor serum concentrations and maintain at 1520 g/mL.
tion and is not recommended.

Scheld_Ch31.indd 541 2/21/14 5:46 PM


542 Part III: Bacterial and Mycoplasmal Infections

Management of Nocardial Brain Abscesses Management of Patients with Multiple Abscesses


A study of 131 patients with nocardial brain abscesses (67) For patients with multiple brain abscesses, it has been recom-
recommended that (a) if an extraneural nocardial infection mended that all abscesses larger than 2.5 cm in diameter or
(present in 70% of patients) is documented in a clinically sta- with significant mass effect should be stereotactically aspirated
ble immunocompetent patients with a brain abscess less than (or excised) (220,221). If the abscesses are all smaller than
2 cm in diameter, empirical treatment with trimethoprim- 2.5 cm in diameter, the largest and/or most accessible lesion
sulfamethoxazole should be given (possible alternatives are should be aspirated for diagnostic purposes. In the case of en-
meropenem, ceftriaxone, ampicillin, linezolid, vancomycin, largement of an abscess after a 2-week interval or a failure of
or amikacin) and (b) if the patient deteriorates clinically, an abscess to diminish in size after 3 to 4 weeks of antibiotics,
or if the abscess does not decrease in size within 4 weeks, further surgical drainage is recommended.
stereotactic aspiration should be performed to confirm the
diagnosis and to decompress the lesion. However, several re- Duration of Antimicrobial Therapy and Follow-up
cent reviews challenged this view and recommended that an
In general, the appropriate duration of antimicrobial therapy
open or stereotactic biopsy should be performed early in the
for brain abscess is not completely clear, but duration of antibi-
patients course, because it facilitates an early and accurate
otic therapy for shorter than 4 weeks are not reported. Usually,
diagnosis (251253). Another series of 11 patients confirmed
4 to 8 weeks of intravenous therapy is reported suitable, pro-
that aspiration is an effective treatment of nocardial brain
vided that the pathogens are susceptible, a clear clinical and
abscesses with considerable mass effect (254). Nine of these
radiographic response is observed, and the treatment is well
patients were treated only with aspiration and long-term
tolerated by the patient. Some favor an additional 2 to 4 weeks,
chemotherapy (intravenous chemotherapy for 4 to 6 weeks
others a several months course of oral antimicrobial therapy
and subsequent oral drug therapy for 6 to 12 months; nine
to prevent relapse. However, clinical evidence for that recom-
patients were treated with trimethoprim-sulfamethoxazole)
mendation is lacking. Drugs used for oral treatment include
and two patients underwent craniotomy and lesion excision.
trimethoprim-sulfamethoxazole, amoxicillin/clavulanic acid,
Four of the nine patients needed only one aspiration, three
moxifloxacin, linezolid, and rifampin (220), although rifampin
patients needed two aspirations, and three or four aspirations
should not be used as single-agent therapy. Follow-up should
were necessary in one patient each. There were no fatalities
include clinical examinations and serial CT scans or MRI to
in this series.
document resolution of the abscess (Fig. 31.19). However, even
a cured brain abscess may continue to exhibit areas of con-
Management of Cerebral Aspergillosis trast enhancement on neuroimaging for several months after
completion of successful therapy (256). Follow-up DWI might
A prospective unblinded study on 277 patients with inva-
be of help in assessing treatment responses in brain abscess in
sive aspergillosis showed that voriconazole therapy was as-
the future (257).
sociated with a lower mortality than amphotericin B during
the first 12 weeks of treatment but the number of patients
with cerebral aspergillosis was low (n 5 in each treat-
Corticosteroids
ment group) (82). Nevertheless, in a retrospective study of The use of corticosteroids in brain abscess is problematic
81 patients with cerebral aspergillosis, treatment with vori- because it might interfere with the CNS penetration of anti-
conazole was associated with a complete or partial response biotics (258). In summary, there is no data favoring a routine
in 35% of patients (81). In comparison, cerebral aspergil- use of steroids in the treatment of brain abscess. Adjunctive
losis was reported to be fatal in more than 90% of patients therapy with corticosteroids should, therefore, be restricted
despite therapy with amphothericin B. Thus, voriconazole to patients with a progressive neurologic deterioration or
is recommended as the treatment of choice for cerebral as- impending cerebral herniation and radiologic evidence that the
pergillosis. Furthermore, multifactorial analysis revealed abscess is causing significant cerebral edema and mass effect.
that additional neurosurgical treatment such as craniotomy, Furthermore, in patients with abscesses in areas where edema
abscess resection, abscess drainage, or shunting/placement might quickly become critical (cerebellum), the use of cortico-
of an Ommaya reservoir were associated with improved steroids should be considered. Whenever corticosteroids are
outcome (81). To date, whether excision or aspiration is used, a rather quick reduction of dosage is recommended once
the better treatment for cerebral aspergillosis is not clear. the patient has had a clinical response.
However, an early diagnostic stereotactic aspiration, if fea-
sible, is warranted in cases of suspected cerebral aspergil-
losis (80,255). TREATMENT OF COMPLICATIONS
The most important complications of brain abscesses are (a)
Management of Brain Abscess in Patients with AIDS
seizures, (b) obstructive hydrocephalus, (c) intraventricular
The following procedures have been proposed for the evalu- rupture of the abscess resulting in ventriculitis and/or menin-
ation and treatment of intracranial lesions in patients with gitis, and (d) brain edema.
AIDS (202): (a) empirical treatment for toxoplasmosis should
be instituted in all cases (pyrimethamine plus sulfadiazine
or pyrimethamine plus clindamycin), except when a single Seizures
intracranial mass lesion accompanies negative serology for
toxoplasmosis; and (b) a stereotactic biopsy is indicated if On admission, seizures are reported in up to 34% of patients
serology is negative or the patient deteriorates clinically or with brain abscess (9,16,32,259). Acute seizures should be ter-
radiologically during antitoxoplasmic therapy, although large minated with administration of intravenous benzodiazepines,
lesions may require immediate surgical decompression. In such as lorazepam or midazolam, or by intravenous fosphe-
HIV-infected children, proceeding directly to stereotactic bi- nytoin. For the prevention of further seizures, carbamazepine
opsy may be considered, because toxoplasmosis is rare in this and fosphenytoin are recommended; in addition to phenytoin,
patient population. valproate and levetiracetam are available for intravenous

Scheld_Ch31.indd 542 2/21/14 5:46 PM


Chapter 31: Brain Abscess 543

administration. Patients without seizures but with epileptic a beneficial effect of corticosteroids on outcome (122,132).
discharges on the electroencephalogram (EEG) should also be However, the analysis is complicated because corticosteroids
treated with antiepileptic drugs. Interestingly, a reduced fre- are given mostly to severely obtunded and comatose patients
quency of seizures in brain abscess was reported in patients who are known to have a dismal prognosis (4,218). The use of
treated with aspiration in comparison to those treated with corticosteroids should be restricted to patients with a progres-
excision, but studies were retrospective (260). The prophylac- sive neurologic deterioration or impending cerebral herniation
tic treatment of patients without EEG abnormalities, however, and radiologic evidence that the abscess is causing significant
still is a matter of debate, and there are no conclusive studies cerebral edema and mass effect. Prolonged use of corticoste-
of seizure prophylaxis in patients with brain abscess. For brain roids cannot be recommended in view of animal experimental
abscesses, some authors propose seizure prophylaxis for all findings of delayed capsule formation and reduced bacterial
patients with a duration of at least 1 year (190,227). Others clearance. Therefore, high-dose corticosteroid therapy (e.g.,
recommend reevaluation of initially treated patients by neu- intravenous dexamethasone at 8 mg every 8 hours) should be
rologic and EEG examination several weeks after treatment given when necessary and should then be tapered off over a few
of the abscess is initiated and withdrawal of the medication if days after the patients condition has stabilized. Severe brain
no seizures occurred and EEG does not show signs typical for edema and impending cerebral herniation may necessitate fur-
epilepsy (261). In patients with EEG abnormalities that are ther measures to reduce the increased ICP, such as osmotherapy
indicating epileptic activity, however, the treatment should be (e.g., mannitol), sedation, and moderate hyperventilation.
continued.

Meningitis
Obstructive Hydrocephalus
A brain abscess can rarely complicate bacterial meningitis
Unilateral or bilateral external ventricular drainage (in cases and vice versa; also, both can develop from contiguous or
with obstruction at the foramen of Monro, especially in cases hematogenous spread from an extradural focus. The associa-
with basal ganglia or thalamic abscess [171]) may become tion of brain abscess and meningitis is particularly common
necessary in brain abscesses complicated by obstructive hy- in neonates. In older children and adults with brain abscess,
drocephalus. Placement of an external ventricular drain is es- meningitis is probably the result of rupture of the abscess into
pecially important in patients with abscess formation in the the ventricles or into the subarachnoid space in most cases.
posterior cranial fossa. In a series of 77 patients with cerebel- Meningitis is also more common in patients with a brain
lar abscesses, hydrocephalus was diagnosed in 79.2% (118). abscess due to L. monocytogenes: 38% of these patients were
In these patients, hydrocephalus was found to be a significant reported to have positive CSF cultures (203).
adverse prognostic factor. With a policy of aggressive CSF di- Lumbar puncture should not be performed in patients
version (immediate ventricular drainage in any patient with with a proven brain abscess. However, one should be aware
a cerebellar abscess and an overt or incipient hydrocephalus, of meningitis as a complication of brain abscess, because
even in patients that were fully conscious), the authors were meningitis-associated systemic and CNS complications (such
able to reduce the mortality from 29% to 11.6% (118). Besides as vascular complications) may contribute to an adverse out-
acute hydrocephalus, persistent disturbances of CSF circulation come. For the management of acute bacterial meningitis and
are common in patients with brain abscess: 10 of 20 survivors its complications, see Chapter 24.
of intraventricular rupture of brain abscesses in the study by
Takeshita et al. (262) required permanent CSF shunting.
Hyponatremia
Ventricular Rupture Like other intracranial diseases, a brain abscess can be com-
plicated by hyponatremia; its incidence in patients with brain
The encapsulation of a brain abscess often is more complete on abscess, however, is unknown. Two different syndromes caus-
the cortical than on the ventricular side (191). Thus, brain ab- ing hyponatremia are particularly important, the syndrome of
scesses are more likely to rupture into the ventricles rather than inappropriate antidiuretic hormone secretion (SIADHS) and
laterally into the subarachnoid space (262). Intraventricular that of cerebral salt wasting (CSW). SIADHS is character-
rupture of brain abscesses affects approximately 15% to 25% ized by water retention, leading to dilutional hyponatremia.
of patients with brain abscess (127,262,263) and results in ven- By comparison, CSW is due to primary natriuresis, leading to
triculitis, meningitis, and disturbances of the CSF circulation, hypovolemia and sodium deficit. SIADHS should be treated by
causing an additional increase of ICP. Accordingly, intraven- volume restriction; the treatment of CSW consists of sodium
tricular rupture of brain abscess is associated with an unfa- and water replacement (264).
vorable outcome (127,263). Because intraventricular rupture
of brain abscesses often affects drainage of CSF, placement of
external ventricular drains (sometimes even bilaterally) might Hemorrhage
be necessary in such patients.
Bleeding into the abscess cavity, the surrounding brain paren-
chyma, into the ventricular system, or the subarachnoid space
Brain Edema has been reported as a rare complication of brain abscesses
(265267). It may lead to a stroke-like presentation and the
The edema around the abscess cavity is usually very pro- neuroradiologic diagnosis of the brain abscess may be con-
nounced; it may be greater in volume than the abscess itself fused by a hemorrhage. The underlying mechanism is not
(201). To reduce the surrounding edema, corticosteroids are clear. However, the bleeding seems likely to occur from newly
frequently recommended. Controlled prospective studies on formed fragile blood vessels in the wall of the abscess cavity.
adjunctive therapy with corticosteroids for brain abscesses There are no specific recommendations for the treatment of
have not been performed. Retrospective studies failed to show hemorrhage associated with brain abscess.

Scheld_Ch31.indd 543 2/21/14 5:46 PM


544 Part III: Bacterial and Mycoplasmal Infections

authors observed a reduced frequency of seizures (272,274),


PROGNOSIS but others did not (4,273).

Mortality
Other Sequelae
The mortality of brain abscess was 40% to 80% in the pre-
antibiotic era with a decline in numbers after the introduc- Other major sequelae of brain abscess are focal neurologic def-
tion of penicillin (268). An adverse prognosis previously was icits: Major deficits such as pareses or aphasia were evident in
associated with (a) delayed or missed diagnosis, (b) poor 18%, and minor deficits such as visual field defects, eye muscle
localization, (c) multiple, deep, or multiloculated lesions, palsies, or unilateral deafness were reported in another 9%
(d) ventricular rupture, (e) coma on admission, (f) fungal eti- of the survivors in one long-term follow-up study (272) and
ology, and (g) inappropriate antibiotics. Additional negative intellectual impairment (in the aforementioned study, 18% of
factors often cited are extremes of age, large abscesses, pres- the survivors were found to be severely mentally handicapped
ence of metastatic abscesses, and rapidly progressive disease [272]). A follow-up evaluation of 32 children treated for brain
(4,132,218,262,269). Since the introduction of CT scanning, abscess noted that intellectual impairment was more common
the mortality has decreased substantially (4,16,270). Thus, in those younger than 5 years at the time of diagnosis and
in recent studies, the mortality of brain abscess was between that behavioral abnormalities predominated in older children
10% and 19% (9,1517,30). Easier detection of underlying (275). This disturbing effect on mental function was con-
conditions (e.g., sinusitis or osseous dehiscence), monitoring firmed in a second study of neonatal brain abscess, as only 4 of
of the therapeutic progress, and recognition of complications 17 children followed up for more than 2 years in one study
by CT and MRI have probably contributed to the improved had an intelligence quotient at or above 80 (276).
prognosis. Furthermore, CT-guided stereotactic aspiration has
greatly improved in particular the treatment of deep-seated
and brainstem abscesses. PROPHYLAXIS OF BRAIN ABSCESS
Prophylactic measures that may be relevant to neurologists, neu-
Seizures rosurgeons, and infectious disease specialists are in particular
antibiotic prophylaxis after penetrating craniocerebral injuries
Among the long-term complications of brain abscess, seizures or craniotomy and the secondary prevention in patients with
have been particularly well studied. Older follow-up stud- pulmonary AVMs, cyanotic heart disease, and AIDS. For pen-
ies showed a prevalence of 50% (mean, range 34% to 72%) etrating craniocerebral injuries, recommendations exist that
(269,271273). However, most of the patients included in these broad-spectrum antibiotic prophylaxis should be given as soon
studies had been treated in the pre-CT era and the figures might as possible after the injury and be continued for 5 days postop-
be lower today. By comparison, studies in the CT era (though eratively (dbridement of the scalp wound and of the intracranial
with fewer patients and much shorter follow-up) reported injury as far as it is feasible without further neural damage) (277).
seizures in 12% of the survivors (mean, range 10% to 16%) A metaanalysis of the efficacy of prophylactic antibiot-
(4,132,234). Seizures were reported to have manifested within ics for craniotomy reported a near fourfold reduction of the
1 year after discharge in 86% (269), within 2 years in 78% wound infection rate (superficial and deep wound infections
(272), or in 41% within 1 year and in 76% of patients within were included as wound infections) by antibiotic prophylaxis
4 years (273); however, intervals of more than 10 years have (278). A large study on 6,243 patients with craniotomy noted
also been observed. Also, the amount of spike and sharp waves a reduction of incision infection through introduction of anti-
on EEG showed a marked increase during the first 4 years both biotic prophylaxis from 8.4% to 4.6% (279).
in patients with and in those without seizures (273). Neither sei- A retrospective analysis of patients with diffuse pulmonary
zures nor EEG abnormalities before treatment of the abscess are AVMs suggested that a combination of antibiotic prophy-
predictive for a later epilepsy (269). Seizures are common fol- laxis for procedures associated with bacteremia and occlusion
lowing frontal, parietal, or temporal abscesses, but they are rare (transcatheter embolotherapy) of the larger AVMs is effective in
after abscesses located in the cerebellum or the occipital lobe preventing brain abscess (280). For patients with CCHD, cor-
(272,273). The impact of the surgical method on the frequency rective surgery should be considered; patients with uncorrected
of seizures is not clear. After burr-hole aspiration alone com- cyanotic congenital heart disease should be administered anti-
pared with craniotomy and total excision of the abscess, some biotic prophylaxis for procedures associated with bacteremia.

References
1. Viale GL, Deseri S, Gennaro S, et al. A craniocerebral infectious disease: case 7. Nicolosi A, Hauser WA, Musicco M, et al. Incidence and prognosis of brain
report on the traces of Hippocrates. Neurosurgery. 2002;50:13761378. abscess in a defined population: Olmsted County, Minnesota, 19351981.
2. Canale DJ. William Macewen and the treatment of brain abscesses: revis- Neuroepidemiology. 1991;10:122131.
ited after one hundred years. J Neurosurg. 1996;84:133142. 8. Duel P, Siboni K, Jensen TG. Intracranial abscesses in Odense Hospital.
3. Habib AA, Mozaffar T. Brain abscess. Arch Neurol. 2001;58:13021304. Survey of bacteriology, epidemiology, and treatment with antibiotics,
4. Mampalam TJ, Rosenblum ML. Trends in the management of bacterial 19631989. Dan Med Bull. 1991;38:407410.
brain abscesses: a review of 102 cases over 17 years. Neurosurgery. 1988;23: 9. Nathoo N, Nadvi SS, Narotam PK, et al. Brain abscess: management and out-
451458. come analysis of a computed tomography era experience with 973 patients.
5. Selby R, Ramirez CB, Singh R, et al. Brain abscess in solid organ transplant World Neurosurg. 2011;75:716726.
recipients receiving cyclosporine-based immunosuppression. Arch Surg. 10. Menon S, Bharadwaj R, Chowdhary A, et al. Current epidemiology of
1997;132:304310. intracranial abscesses: a prospective 5 year study. J Med Microbiol. 2008;57:
6. Hagensee ME, Bauwens JE, Kjos B, et al. Brain abscess following mar- 12591268.
row transplantation: experience at the Fred Hutchinson Cancer Research 11. de Louvois J, Gortavai P, Hurley R. Bacteriology of abscesses of the central
Center, 19841992. Clin Infect Dis. 1994;19:402408. nervous system: a multicentre prospective study. Br Med J. 1977;2:981984.

Scheld_Ch31.indd 544 2/21/14 5:46 PM


Chapter 31: Brain Abscess 545

12. de Louvois J. The bacteriology and chemotherapy of brain abscess. 45. Chen CC, Wang SJ, Fuh JL. Isolation of Haemophilus parahaemolyticus in a
J Antimicrob Chemother. 1978;4:395413. patient with cryptogenic brain abscess. Scand J Infect Dis. 2001;33:385386.
13. Heineman HS, Braude AI. Anaerobic infection of the brain. Observations on 46. Feldman WE, Schwartz, J. Haemophilus influenzae type b brain abscess
eighteen consecutive cases of brain abscess. Am J Med. 1963;35:682697. complicating meningitis: case report. Pediatrics. 1983;72:473475.
14. Renier D, Flandin C, Hirsch E, et al. Brain abscesses in neonates. A study 47. Sane SM, Faerber EN, Belani KK. Respiratory foreign bodies and Eikenella
of 30 cases. J Neurosurg. 1988;69:877882. corrodens brain abscess in two children. Pediatr Radiol. 1999;29:327330.
15. Landriel F, Ajler P, Hem S, et al. Supratentorial and infratentorial brain 48. Ozum U, Karadag O, Egilmez R, et al. A case of brain abscess due to
abscesses: surgical treatment, complications and outcomesa 10-year Entamoeba species, Eikenella corrodens and Prevotella species. Br J
single-center study. Acta Neurochir (Wien). 2012;154:903911. Neurosurg. 2008;22:596598.
16. Sharma R, Mohandas K, Cooke RP. Intracranial abscesses: changes in 49. Weber DJ, Wolfson JS, Swartz MN, et al. Pasteurella multocida infections.
epidemiology and management over five decades in Merseyside. Infection. Report of 34 cases and review of the literature. Medicine (Baltimore).
2009;37:3943. 1984;63:133154.
17. Roche M, Humphreys H, Smyth E, et al. A twelve-year review of cen- 50. Klein DM, Cohen ME. Pasteurella multocida brain abscess following per-
tral nervous system bacterial abscesses; presentation and aetiology. Clin forating cranial dog bite. J Pediatr. 1978;92:588589.
Microbiol Infect. 2003;9:803809. 51. Spadafora R, Pomero G, Delogu A, et al. A rare case of neonatal sepsis/
18. Grigoriadis E, Gold WL. Pyogenic brain abscess caused by Streptococcus meningitis caused by Pasteurella multocida complicated with status epilep-
pneumoniae: case report and review. Clin Infect Dis. 1997;25:11081112. ticus and focal cerebritis. Pediatr Med Chir. 2011;33:199202.
19. Korinek AM. Risk factors for neurosurgical site infections after craniotomy: 52. Zijlstra EE, Swart GR, Godfroy FJ, et al. Pericarditis, pneumonia and brain
a prospective multicenter study of 2944 patients. Neurosurgery. 1997;41: abscess due to a combined ActinomycesActinobacillus actinomycetem-
10731079. comitans infection. J Infect. 1992;25:8387.
20. Lu CH, Chang WN, Lin YC, et al. Bacterial brain abscess: microbio- 53. Kuijper EJ, Wiggerts HO, Jonker GJ, et al. Disseminated actinomycosis due
logical features, epidemiological trends and therapeutic outcomes. QJM. to Actinomyces meyeri and Actinobacillus actinomycetemcomitans. Scand
2002;95:501509. J Infect Dis. 1992;24:667672.
21. Inamasu J, Kagami H, Nakamura Y, et al. Brain abscess developing at the 54. Lee JH, Kim DG. Brain abscess related to metal fragments 47 years after
site of preceding intracerebral hemorrhage. J Neurol. 2002;249:221223. head injury. Case report. J Neurosurg. 2000;93:477479.
22. Mylona E, Vadala C, Papastamopoulos V, et al. Brain abscess caused by 55. Cohen JE, Mierez R, Tsai EC. Postcraniotomy gas-containing brain abscess:
Enterococcus faecalis following a dental procedure in a patient with heredi- a neurosurgical emergency. Case report. Surg Neurol. 1999;51:568570.
tary hemorrhagic telangiectasia. J Clin Microbiol. 2012;50:18071809. 56. Tekkok IH, Higgins MJ, Ventureyra EC. Posttraumatic gas-containing
23. Li X, Tronstad L, Olsen I. Brain abscesses caused by oral infection. Endod brain abscess caused by Clostridium perfringens with unique simultane-
Dent Traumatol. 1999;15:95101. ous fungal suppuration by Myceliophthora thermophila: case report.
24. Sennaroglu L, Sozeri B. Otogenic brain abscess: review of 41 cases. Neurosurgery. 1996;39:12471251.
Otolaryngol Head Neck Surg. 2000;123:751755. 57. Cheng YT, Huang CT, Leu HS, et al. Central nervous system infection
25. Kangsanarak J, Fooanant S, Ruckphaopunt K, et al. Extracranial and due to Clostridium septicum: a case report and review of the literature.
intracranial complications of suppurative otitis media. Report of 102 cases. Infection. 1997;25:171174.
J Laryngol Otol. 1993;107:9991004. 58. Lew JF, Wiedermann BL, Sneed J, et al. Aerotolerant Clostridium tertium
26. Saez-Llorens XJ, Umana MA, Odio CM, et al. Brain abscess in infants and brain abscess following a lawn dart injury. J Clin Microbiol. 1990;28:
children. Pediatr Infect Dis J. 1989;8:449458. 21272129.
27. Rau CS, Chang WN, Lin YC, et al. Brain abscess caused by aerobic Gram- 59. Chu RM, Tummala RP, Hall WA. Focal intracranial infections due to
negative bacilli: clinical features and therapeutic outcomes. Clin Neurol Propionibacterium acnes: report of three cases. Neurosurgery. 2001;49:
Neurosurg. 2002;105:6065. 717720.
28. Alderson D, Strong AJ, Ingham HR, et al. Fifteen-year review of the mor- 60. Chung S, Kim JS, Seo SW, et al. A case of brain abscess caused by
tality of brain abscess. Neurosurgery. 1981;8:16. Propionibacterium acnes 13 months after neurosurgery and confirmed by
29. Krajewski R, Stelmasiak Z. Brain abscess in infants. Childs Nerv Syst. 16S rRNA gene sequencing. Korean J Lab Med. 2011;31:122126.
1992;8:279280. 61. Kranick SM, Vinnard C, Kolson DL. Propionibacterium acnes brain abscess
30. Tsou TP, Lee PI, Lu CY, et al. Microbiology and epidemiology of brain appearing 10 years after neurosurgery. Arch Neurol. 2009;66:793795.
abscess and subdural empyema in a medical center: a 10-year experience. 62. Altmann G, Bogokovsky B. Brain abscess due to Corynebacterium haemo-
J Microbiol Immunol Infect. 2009;42:405412. lyticum. Lancet. 1973;1:378379.
31. Liliang PC, Lin YC, Su TM, et al. Klebsiella brain abscess in adults. 63. Wooster SL, Qamruddin A, Clarke R, et al. Brain abscess due to
Infection. 2001;29:8186. Corynebacterium xerosis. J Infect. 1999;38:5556.
32. Auvichayapat N, Auvichayapat P, Aungwarawong S. Brain abscess in 64. Dykhuizen RS, Douglas G, Weir J, et al. Corynebacterium afermentans
infants and children: a retrospective study of 107 patients in northeast subsp. lipophilum: multiple abscess formation in brain and liver. Scand J
Thailand. J Med Assoc Thai. 2007;90:16011607. Infect Dis. 1995;27:637639.
33. Kline MW. Citrobacter meningitis and brain abscess in infancy: epidemiol- 65. Smego RA. Actinomycosis of the central nervous system. Rev Infect Dis.
ogy, pathogenesis, and treatment. J Pediatr. 1988;113:430434. 1987;9:855865.
34. Booth LV, Palmer JD, Pateman J, et al. Citrobacter diversus ventriculitis 66. Dailey AT, LeRoux PD, Grady MS. Resolution of an actinomycotic abscess
and brain abscesses in an adult. J Infect. 1993;26:207209. with nonsurgical treatment: case report. Neurosurgery. 1993;32:134136.
35. Lakshmi V, Umabala P, Anuradha K, et al. Microbiological spectrum of 67. Mamelak AN, Obana WG, Flaherty JF, et al. Nocardial brain abscess:
brain abscess at a tertiary care hospital in South India: 24-year data and treatment strategies and factors influencing outcome. Neurosurgery.
review. Patholog Res Int. 2011;2011:583139. 1994;35:622631.
36. Shanley DJ, Holmes SM. Salmonella typhi abscess in a craniopharyngioma: 68. Psiachou-Leonard E, Sidi V, Tsivitanidou M, et al. Brain abscesses result-
CT and MRI. Neuroradiology. 1994;36:3536. ing from Bacillus cereus and an Aspergillus-like mold. J Pediatr Hematol
37. Aliaga L, Mediavilla JD, Lopez, dlO, et al. Nontyphoidal Salmonella Oncol. 2002;24:569571.
intracranial infections in HIV-infected patients. Clin Infect Dis. 1997;25: 69. Mori T, Tokuhira M, Takae Y, et al. Successful non-surgical treatment of
11181120. brain abscess and necrotizing fasciitis caused by Bacillus cereus. Intern
38. Kirollos RW, Bosma JJ, Radhakrishnan J, et al. Endovascularly treated Med. 2002;41:671673.
cerebral aneurysm using Guglielmi detachable coils acting as a nidus for 70. Corne P, Rajeebally I, Jonquet O. Rhodococcus equi brain abscess in an
brain abscess formation secondary to Salmonella bacteremia: case report. immunocompetent patient. Scand J Infect Dis. 2002;34:300302.
Neurosurgery. 2002;51:234237. 71. Kohl O, Tillmanns HH. Cerebral infection with Rhodococcus equi in a
39. Lai KK. Enterobacter sakazakii infections among neonates, infants, chil- heart transplant recipient. J Heart Lung Transplant. 2002;21:11471149.
dren, and adults. Case reports and a review of the literature. Medicine 72. Drancourt M, Pelletier J, Cherif AA, et al. Gordona terrae central ner-
(Baltimore). 2001;80:113122. vous system infection in an immunocompetent patient. J Clin Microbiol.
40. Ries M, Deeg KH, Heininger U, et al. Brain abscesses in neonatesreport 1997;35:379382.
of three cases. Eur J Pediatr. 1993;152:745746. 73. Garcia-Monco JC. Central nervous system tuberculosis. Neurol Clin.
41. Lam AH, Berry A, deSilva M, et al. Intracranial Serratia infection in pre- 1999;17:737759.
term newborn infants. AJNR Am J Neuroradiol. 1984;5:447451. 74. Ozates M, Kemaloglu S, Gurkan F, et al. CT of the brain in tuberculous
42. Huang CR, Lu CH, Chien CC, et al. Protean infectious types and frequent meningitis. A review of 289 patients. Acta Radiol. 2000;41:1317.
association with neurosurgical procedures in adult Serratia marcescens 75. Bahemuka M, Babiker MA, Wright SG, et al. The pattern of infection of
CNS infections: report of two cases and review of the literature. Clin the nervous system in Riyadh: a review of 121 cases. Q J Med. 1988;68:
Neurol Neurosurg. 2001;103:171174. 517524.
43. Faisal W, Burnton G, Imlay-Gillespie L, et al. Cerebral abscesses and septic 76. Coppes MJ, Olivieri NF, Howes M, et al. Mycobacterial brain abscess pos-
pulmonary emboli due to Serratia marcescens infection arising from a sibly due to bacille Calmette-Guerin in an immunocompromised child. Clin
Portacath. J Microbiol Immunol Infect. 2010;43:538541. Infect Dis. 1992;14:662665.
44. Kao PT, Tseng HK, Su SC, et al. Haemophilus aphrophilus brain abscess: 77. Gordon SM, Blumberg HM. Mycobacterium kansasii brain abscess in a
a case report. J Microbiol Immunol Infect. 2002;35:184186. patient with AIDS. Clin Infect Dis. 1992;14:789790.

Scheld_Ch31.indd 545 2/21/14 5:46 PM


546 Part III: Bacterial and Mycoplasmal Infections

78. Uldry PA, Bogousslavsky J, Regli F, et al. Chronic Mycobacterium avium 113. Bia FJ, Barry M. Parasitic infections of the central nervous system. Neurol
complex infection of the central nervous system in a nonimmunosup- Clin. 1986;4:171206.
pressed woman. Eur Neurol. 1992;32:285288. 114. Kudesia S, Indira DB, Sarala D, et al. Sparganosis of brain and spinal
79. Davis LE. Fungal infections of the central nervous system. Neurol Clin. cord: unusual tapeworm infestation (report of two cases). Clin Neurol
1999;17:761781. Neurosurg. 1998;100:148152.
80. Denning DW. Invasive aspergillosis. Clin Infect Dis. 1998;26:781803. 115. Purohit AK, Dinakar I, Sundaram C, et al. Angiostrongylus cantonensis
81. Schwartz S, Ruhnke M, Ribaud P, et al. Improved outcome in central abscess in the brain. J Neurol Neurosurg Psychiatry. 1991;54:10151016.
nervous system aspergillosis, using voriconazole treatment. Blood. 116. Masdeu JC, Tantulavanich S, Gorelick PP, et al. Brain abscess caused by
2005;106:26412645. Strongyloides stercoralis. Arch Neurol. 1982;39:6263.
82. Herbrecht R, Denning DW, Patterson TF, et al. Voriconazole versus am- 117. Samuel J, Fernandes CM, Steinberg JL. Intracranial otogenic complica-
photericin B for primary therapy of invasive aspergillosis. N Engl J Med. tions: a persisting problem. Laryngoscope. 1986;96:272278.
2002;347:408415. 118. Nadvi SS, Parboosing R, van, DJ. Cerebellar abscess: the significance of
83. Kleinschmidt-DeMasters BK. Central nervous system aspergillosis: a 20- cerebrospinal fluid diversion. Neurosurgery. 1997;41:6166.
year retrospective series. Hum Pathol. 2002;33:116124. 119. Singh B, Van Dellen J, Ramjettan S, et al. Sinogenic intracranial complica-
84. Gamaletsou MN, Sipsas NV, Roilides E, et al. Rhino-orbital-cerebral tions. J Laryngol Otol. 1995;109:945950.
mucormycosis. Curr Infect Dis Rep. 2012;14:423434. 120. Jones NS, Walker JL, Bassi S, et al. The intracranial complications of
85. DiBartolo MA, Kelley PS. Rhino-orbital-cerebral mucormycosis (ROCM): rhinosinusitis: can they be prevented? Laryngoscope. 2002;112:5963.
a comprehensive case review. Aviat Space Environ Med. 2011;82:913916. 121. Gallagher RM, Gross CW, Phillips CD. Suppurative intracranial compli-
86. Mbarek C, Zribi S, Khamassi K, et al. Rhinocerebral mucormycosis: five cations of sinusitis. Laryngoscope. 1998;108:16351642.
cases and a literature review. B-ENT. 2011;7:189193. 122. Chun CH, Johnson JD, Hofstetter M, et al. Brain abscess. A study of
87. Hopkins RJ, Rothman M, Fiore A, et al. Cerebral mucormycosis associ- 45 consecutive cases. Medicine (Baltimore). 1986;65:415431.
ated with intravenous drug use: three case reports and review. Clin Infect 123. Lew D, Southwick FS, Montgomery WW, et al. Sphenoid sinusitis.
Dis. 1994;19:11331137. A review of 30 cases. N Engl J Med. 1983;309:11491154.
88. Zarei M, Morris J, Aachi V, et al. Acute isolated cerebral mucormyco- 124. Heimdahl A, Hall G, Hedberg M, et al. Detection and quantitation by
sis in a patient with high grade non-Hodgkins lymphoma. Eur J Neurol. lysis-filtration of bacteremia after different oral surgical procedures. J Clin
2000;7:443447. Microbiol. 1990;28:22052209.
89. Sethi PK, Khanna L, Batra A, et al. Central nervous system fungal infec- 125. Roberts GJ, Holzel HS, Sury MR, et al. Dental bacteremia in children.
tions: Observations from a large tertiary hospital in northern India. Clin Pediatr Cardiol. 1997;18:2427.
Neurol Neurosurg. 2012;114(9):12321237. 126. Yang SY. Brain abscess: a review of 400 cases. J Neurosurg. 1981;55:
90. Sanchez-Portocarrero J, Perez-Cecilia E, Corral O, et al. The central ner- 794799.
vous system and infection by Candida species. Diagn Microbiol Infect 127. Nielsen H, Gyldensted C, Harmsen A. Cerebral abscess. Aetiology and
Dis. 2000;37:169179. pathogenesis, symptoms, diagnosis and treatment. A review of 200 cases
91. Popovich MJ, Arthur RH, Helmer E. CT of intracranial cryptococcosis. from 19351976. Acta Neurol Scand. 1982;65:609622.
AJR Am J Roentgenol. 1990;154:603606. 128. Ebright JR, Pace MT, Niazi AF. Septic thrombosis of the cavernous
92. Tien RD, Chu PK, Hesselink JR, et al. Intracranial cryptococcosis in sinuses. Arch Intern Med. 2001;161:26712676.
immunocompromised patients: CT and MR findings in 29 cases. AJNR 129. Graham DR, Band JD. Citrobacter diversus brain abscess and meningitis
Am J Neuroradiol. 1991;12:283289. in neonates. JAMA. 1981;245:19231925.
93. Castiglioni B, Sutton DA, Rinaldi MG, et al. Pseudallescheria boydii 130. Kline MW, Mason EO Jr, Kaplan SL. Characterization of Citrobacter
(Anamorph Scedosporium apiospermum) infection in solid organ trans- diversus strains causing neonatal meningitis. J Infect Dis. 1988;157:
plant recipients in a tertiary medical center and review of the literature. 101105.
Medicine (Baltimore). 2002;81:333348. 131. Li J, Musser JM, Beltran P, et al. Genotypic heterogeneity of strains
94. Panichpisal K, Nugent K, Sarria JC. Central nervous system pseudallesche- of Citrobacter diversus expressing a 32-kilodalton outer membrane
riasis after near-drowning. Clin Neurol Neurosurg. 2006;108:348352. protein associated with neonatal meningitis. J Clin Microbiol. 1990;28:
95. Nesky MA, McDougal EC, Peacock JE Jr. Pseudallescheria boydii brain 17601765.
abscess successfully treated with voriconazole and surgical drainage: case 132. Seydoux C, Francioli P. Bacterial brain abscesses: factors influencing mor-
report and literature review of central nervous system pseudallescheriasis. tality and sequelae. Clin Infect Dis. 1992;15:394401.
Clin Infect Dis. 2000;31:673677. 133. Patir R, Sood S, Bhatia R. Post-traumatic brain abscess: experience of
96. Heney C, Song E, Kellen A, et al. Cerebral phaeohyphomycosis caused by 36 patients. Br J Neurosurg. 1995;9:2935.
Xylohypha bantiana. Eur J Clin Microbiol Infect Dis. 1989;8:984988. 134. Stephanov S. Brain abscesses from neglected open head injuries: experience
97. Porter SB, Sande MA. Toxoplasmosis of the central nervous system in with 17 cases over 20 years. Swiss Surg. 1999;5:288292.
the acquired immunodeficiency syndrome. N Engl J Med. 1992;327: 135. Sutton LN, Alpert G. Brain abscess following cranial dog bite. Clin
16431648. Pediatr (Phila). 1984;23:580.
98. Mele A, Paterson PJ, Prentice HG, et al. Toxoplasmosis in bone marrow 136. Berkowitz FE, Jacobs DW. Fatal case of brain abscess caused by rooster
transplantation: a report of two cases and systematic review of the litera- pecking. Pediatr Infect Dis J. 1987;6:941942.
ture. Bone Marrow Transplant. 2002;29:691698. 137. Tiffany KK, Kline MW. Mixed flora brain abscess with Pseudomonas
99. Cibickova L, Horacek J, Prasil P, et al. Cerebral toxoplasmosis in an paucimobilis after a penetrating lawn dart injury. Pediatr Infect Dis J.
allogeneic peripheral stem cell transplant recipient: case report and review 1988;7:667669.
of literature. Transpl Infect Dis. 2007;9:332335. 138. Tay JS, Garland JS. Serious head injuries from lawn darts. Pediatrics.
100. Campbell S. Amebic brain abscess and meningoencephalis. Semin Neurol. 1987;79:261263.
1993;13:153160. 139. Niazi T, Quinones-Hinojosa A, Schmidt MH. Management of brain
101. Yoder JS, Straif-Bourgeois S, Roy SL, et al. Primary amebic meningoen- abscesses associated with halo fixation. Neurosurg Focus. 2008;24:E14.
cephalitis deaths associated with sinus irrigation using contaminated tap 140. Ray A, Iyer RV, King AT. Cerebral abscess as a delayed complication of
water. Clin Infect Dis. 2012;55:e79e85. halo fixation. Acta Neurochir (Wien). 2006;148:10151016.
102. Martinez AJ, Visvesvara GS. Free-living, amphizoic and opportunistic 141. Roche M, Redmond RM, ONeill S, et al. A case of multiple cerebral
amebas. Brain Pathol. 1997;7:583598. abscesses due to infection with Cladophialophora bantiana. J Infect.
103. Deol I, Robledo L, Meza A, et al. Encephalitis due to a free-living amoeba 2005;51:e285e288.
(Balamuthia mandrillaris): case report with literature review. Surg Neurol. 142. Su TM, Lan CM, Tsai YD, et al. Multiloculated pyogenic brain abscess:
2000;53:611616. experience in 25 patients. Neurosurgery. 2008;62(suppl 2):556561.
104. Carpio A. Neurocysticercosis: an update. Lancet Infect Dis. 2002;2: 143. Basit AS, Ravi B, Banerji AK, et al. Multiple pyogenic brain abscesses:
751762. an analysis of 21 patients. J Neurol Neurosurg Psychiatry. 1989;52:
105. White AC. Neurocysticercosis: updates on epidemiology, pathogenesis, 591594.
diagnosis, and management. Annu Rev Med. 2000;51:187206. 144. Sharma BS, Khosla VK, Kak VK, et al. Multiple pyogenic brain abscesses.
106. Del Brutto OH. A review of cases of human cysticercosis in Canada. Can Acta Neurochir (Wien). 1995;133:3643.
J Neurol Sci. 2012;39:319322. 145. Fenton ME, Cockcroft DW, Gjevre JA. Intracerebral abscess: a complica-
107. Del Brutto OH. Neurocysticercosis in Western Europe: a re-emerging tion of severe cystic fibrosis lung disease. Can Respir J. 2008;15:4547.
disease? Acta Neurol Belg. 2012;112(4):335343. 146. Cooper DM, Russell LE, Henry RL. Cerebral abscess as a complication of
108. Pittella JE. Neurocysticercosis. Brain Pathol. 1997;7:681693. cystic fibrosis. Pediatr Pulmonol. 1994;17:390392.
109. Del Brutto OH, Rajshekhar V, White AC Jr, et al. Proposed diagnostic 147. Sonneville R, Mirabel M, Hajage D, et al. Neurologic complications
criteria for neurocysticercosis. Neurology. 2001;57:177183. and outcomes of infective endocarditis in critically ill patients: the
110. Pittella JE. Neuroschistosomiasis. Brain Pathol. 1997;7:649662. ENDOcardite en REAnimation prospective multicenter study. Crit Care
111. Taratuto AL, Venturiello SM. Echinococcosis. Brain Pathol. 1997;7: Med. 2011;39:14741481.
673679. 148. Tornos P, Almirante B, Mirabet S, et al. Infective endocarditis due to
112. Kusner DJ, King CH. Cerebral paragonimiasis. Semin Neurol. 1993;13: Staphylococcus aureus: deleterious effect of anticoagulant therapy. Arch
201208. Intern Med. 1999;159:473475.

Scheld_Ch31.indd 546 2/21/14 5:46 PM


Chapter 31: Brain Abscess 547

149. Horiuchi Y, Kato Y, Dembo T, et al. Patent foramen ovale as a risk factor 183. Baldwin AC, Kielian T. Persistent immune activation associated with a
for cryptogenic brain abscess: case report and review of the literature. mouse model of Staphylococcus aureus-induced experimental brain ab-
Intern Med. 2012;51:11111114. scess. J Neuroimmunol. 2004;151:2432.
150. Ciurea AV, Stoica F, Vasilescu G, et al. Neurosurgical management of 184. Garg S, Nichols JR, Esen N, et al. MyD88 expression by CNS-resident
brain abscesses in children. Childs Nerv Syst. 1999;15:309317. cells is pivotal for eliciting protective immunity in brain abscesses. ASN
151. Mathis S, Dupuis-Girod S, Plauchu H, et al. Cerebral abscesses in heredi- Neuro. 2009;1:e00007.
tary haemorrhagic telangiectasia: a clinical and microbiological evalua- 185. Stenzel W, Soltek S, Sanchez-Ruiz M, et al. Both TLR2 and TLR4 are
tion. Clin Neurol Neurosurg. 2012;114:235240. required for the effective immune response in Staphylococcus aureus-
152. Moussouttas M, Fayad P, Rosenblatt M, et al. Pulmonary arteriove- induced experimental murine brain abscess. Am J Pathol. 2008;172:
nous malformations: cerebral ischemia and neurologic manifestations. 132145.
Neurology. 2000;55:959964. 186. Kielian T, Hickey WF. Proinflammatory cytokine, chemokine, and cellu-
153. Memon KA, Cleveland KO, Gelfand MS. Brain abscesses associated with lar adhesion molecule expression during the acute phase of experimental
right-to-left shunts in adults. Am J Med Sci. 2012;343:334336. brain abscess development. Am J Pathol. 2000;157:647658.
154. ODonoghue MA, Green HT, Shaw MD. Cerebral abscess on Merseyside 187. Stenzel W, Soltek S, Miletic H, et al. An essential role for tumor necro-
19801988. J Infect. 1992;25:163172. sis factor in the formation of experimental murine Staphylococcus au-
155. Wong TT, Lee LS, Wang HS, et al. Brain abscesses in childrena coopera- reus-induced brain abscess and clearance. J Neuropathol Exp Neurol.
tive study of 83 cases. Childs Nerv Syst. 1989;5:1924. 2005;64:2736.
156. Tekkok IH, Erbengi A. Management of brain abscess in children: review 188. Gupta RK, Nath K, Prasad A, et al. In vivo demonstration of neuroin-
of 130 cases over a period of 21 years. Childs Nerv Syst. 1992;8:411416. flammatory molecule expression in brain abscess with diffusion tensor
157. Aebi C, Kaufmann F, Schaad UB. Brain abscess in childhoodlong-term imaging. AJNR Am J Neuroradiol. 2008;29:326332.
experiences. Eur J Pediatr. 1991;150:282286. 189. Hanamsagar R, Torres V, Kielian T. Inflammasome activation and
158. Domingo Z, Peter JC. Brain abscess in childhood. A 25-year experience. IL-1/IL-18 processing are influenced by distinct pathways in microglia.
S Afr Med J. 1994;84:1315. J Neurochem. 2011;119:736748.
159. Ersahin Y, Mutluer S, Guzelbag E. Brain abscess in infants and children. 190. Muzumdar D, Jhawar S, Goel A. Brain abscess: an overview. Int J Surg.
Childs Nerv Syst. 1994;10:185189. 2011;9:136144.
160. Hirsch JF, Roux FX, Sainte-Rose C, et al. Brain abscess in childhood. 191. Britt RH, Enzmann DR, Yeager AS. Neuropathological and computer-
A study of 34 cases treated by puncture and antibiotics. Childs Brain. ized tomographic findings in experimental brain abscess. J Neurosurg.
1983;10:251265. 1981;55:590603.
161. Takeshita M, Kagawa M, Yato S, et al. Current treatment of brain 192. Britt RH, Enzmann DR. Clinical stages of human brain abscesses on
abscess in patients with congenital cyanotic heart disease. Neurosurgery. serial CT scans after contrast infusion. Computerized tomographic, neu-
1997;41:12701278. ropathological, and clinical correlations. J Neurosurg. 1983;59:972989.
162. Prusty GK. Brain abscesses in cyanotic heart disease. Indian J Pediatr. 193. Kurzydlowski H, Wollenschlager C, Venezio FR, et al. Reevaluation of
1993;60:4351. an experimental streptococcal canine brain abscess model. J Neurosurg.
163. Kawamata T, Takeshita M, Ishizuka N, et al. Patent foramen ovale as 1987;67:717720.
a possible risk factor for cryptogenic brain abscess: report of two cases. 194. Flaris NA, Hickey WF. Development and characterization of an experimen-
Neurosurgery. 2001;49:204206. tal model of brain abscess in the rat. Am J Pathol. 1992;141:12991307.
164. Schlaeffer F, Riesenberg K, Mikolich D, et al. Serious bacterial infections 195. Britt RH, Enzmann DR, Placone RC, et al. Experimental anaerobic brain
after endoscopic procedures. Arch Intern Med. 1996;156:572574. abscess. Computerized tomographic and neuropathological correlations.
165. Laviv Y, Ben-Daviv U, Vated M, et al. Brain abscess following endoscopic J Neurosurg. 1984;60:11481159.
ligation of esophageal varicose veins. Acta Neurochir (Wien). 2010;152: 196. Enzmann DR, Britt RR, Obana WG, et al. Experimental Staphylococcus
733734. aureus brain abscess. AJNR Am J Neuroradiol. 1986;7:395402.
166. Bautista-Casasnovas A, Castro-Gago M. Multiple brain abscesses follow- 197. Neuwelt EA, Lawrence MS, Blank NK. Effect of gentamicin and dexameth-
ing esophageal dilation. Childs Nerv Syst. 1996;12:1. asone on the natural history of the rat Escherichia coli brain abscess model
167. Vates GE, Berger MS, Wilson CB. Diagnosis and management of pituitary with histopathological correlation. Neurosurgery. 1984;15:475483.
abscess: a review of twenty-four cases. J Neurosurg. 2001;95:233241. 198. Wood JH, Doppman JL, Lightfoote WE, et al. Role of vascular prolif-
168. Berger SA, Edberg SC, David G. Infectious disease in the sella turcica. Rev eration on angiographic appearance and encapsulation of experimental
Infect Dis. 1986;8:747755. traumatic and metastatic brain abscesses. J Neurosurg. 1978;48:264273.
169. Hall WA. Infectious lesions of the brain stem. Neurosurg Clin N Am. 199. Su TM, Lan CM, Tsai YD, et al. Multiloculated pyogenic brain abscess:
1993;4:543551. experience in 25 patients. Neurosurgery. 2003;52:10751079.
170. Carpenter JL. Brain stem abscesses: cure with medical therapy, case 200. Hsu CW, Lu CH, Chuang MJ, et al. Cerebellar bacterial brain abscess:
report, and review. Clin Infect Dis. 1994;18:219226. report of eight cases. Acta Neurol Taiwan. 2011;20:4752.
171. Lutz TW, Landolt H, Wasner M, et al. Diagnosis and management of 201. Falcone S, Post MJ. Encephalitis, cerebritis, and brain abscess: patho-
abscesses in the basal ganglia and thalamus: a survey. Acta Neurochir physiology and imaging findings. Neuroimaging Clin N Am. 2000;10:
(Wien). 1994;127:9198. 333353.
172. Chang YT, Lu CH, Chuang MJ, et al. Supratentorial deep-seated bacterial 202. American Academy of Neurology. Evaluation and management of
brain abscess in adults: clinical characteristics and therapeutic outcomes. intracranial mass lesions in AIDS. Report of the Quality Standards
Acta Neurol Taiwan. 2010;19:178183. Subcommittee of the American Academy of Neurology. Neurology.
173. Molinari GF, Smith L, Goldstein MN, et al. Brain abscess from septic cere- 1998;50:2126.
bral embolism: an experimental model. Neurology. 1973;23:12051210. 203. Eckburg PB, Montoya JG, Vosti KL. Brain abscess due to Listeria mono-
174. Foreman SD, Smith EE, Ryan NJ, et al. Neonatal Citrobacter meningi- cytogenes: five cases and a review of the literature. Medicine (Baltimore).
tis: pathogenesis of cerebral abscess formation. Ann Neurol. 1984;16: 2001;80:223235.
655659. 204. Schliamser SE, Backman K, Norrby SR. Intracranial abscesses in adults:
175. Kline MW, Kaplan SL, Hawkins EP, et al. Pathogenesis of brain abscess an analysis of 54 consecutive cases. Scand J Infect Dis. 1988;20:19.
formation in an infant rat model of Citrobacter diversus bacteremia and 205. Tattevin P, Bruneel F, Regnier B. Cranial CT before lumbar puncture in
meningitis. J Infect Dis. 1988;157:106112. suspected meningitis. N Engl J Med. 2002;346:12481251.
176. Nassar SI, Haddad FS, Hanbali FS, et al. Abscess superimposed on 206. Leuthardt EC, Wippold FJ, Oswood MC, et al. Diffusion-weighted MR
brain tumor: two case reports and review of the literature. Surg Neurol. imaging in the preoperative assessment of brain abscesses. Surg Neurol.
1997;47:484488. 2002;58:395402.
177. Arentoft H, Schonheyder H, Schonemann NK. Cerebral Salmonella 207. Tomar V, Yadav A, Rathore RK, et al. Apparent diffusion coefficient with
typhimurium abscess in a patient with a stroke. Infection. 1993;21: higher b-value correlates better with viable cell count quantified from the
251253. cavity of brain abscess. AJNR Am J Neuroradiol. 2011;32:21202125.
178. Chen ST, Tang LM, Ro LS. Brain abscess as a complication of stroke. 208. Reddy JS, Mishra AM, Behari S, et al. The role of diffusion-weighted
Stroke. 1995;26:696698. imaging in the differential diagnosis of intracranial cystic mass lesions:
179. Mourier KL, Bellec C, Lot G, et al. Pyogenic parenchymatous and nidus a report of 147 lesions. Surg Neurol. 2006;66:246250.
infection after embolization of an arteriovenous malformation. An unusual 209. Ohba S, Ushioda T, Nakagawa T, et al. Diffusion magnetic resonance
complication. Case report. Acta Neurochir (Wien). 1993;122:130133. imaging for enhanced visualization of malignant cerebral tumors and
180. Zurin AA, Ushikoshi S, Houkin K, et al. Cerebral abscess as an unusual abscesses. Neurol India. 2011;59:674678.
complication of coil embolization in a dural arteriovenous fistula. Case 210. Schroeder PC, Post MJ, Oschatz E, et al. Analysis of the utility of diffusion-
report. J Neurosurg. 1997;87:109112. weighted MRI and apparent diffusion coefficient values in distinguishing
181. Kielian T. Immunopathogenesis of brain abscess. J Neuroinflammation. central nervous system toxoplasmosis from lymphoma. Neuroradiology.
2004;1:16. 2006;48:715720.
182. Kielian T, Cheung A, Hickey WF. Diminished virulence of an alpha-toxin 211. Toh CH, Wei KC, Ng SH, et al. Differentiation of brain abscesses from
mutant of Staphylococcus aureus in experimental brain abscesses. Infect necrotic glioblastomas and cystic metastatic brain tumors with diffusion
Immun. 2001;69:69026911. tensor imaging. AJNR Am J Neuroradiol. 2011;32:16461651.

Scheld_Ch31.indd 547 2/21/14 5:46 PM


548 Part III: Bacterial and Mycoplasmal Infections

212. Lai PH, Hsu SS, Ding SW, et al. Proton magnetic resonance spectroscopy 244. Wessalowski R, Thomas L, Kivit J, et al. Multiple brain abscesses caused
and diffusion-weighted imaging in intracranial cystic mass lesions. Surg by Salmonella enteritidis in a neonate: successful treatment with cipro-
Neurol. 2007;68(suppl 1):S25S36. floxacin. Pediatr Infect Dis J. 1993;12:683688.
213. Nath K, Agarwal M, Ramola M, et al. Role of diffusion tensor imaging 245. Rex JH, Walsh TJ, Sobel JD, et al. Practice guidelines for the treatment
metrics and in vivo proton magnetic resonance spectroscopy in the differ- of candidiasis. Infectious Diseases Society of America. Clin Infect Dis.
ential diagnosis of cystic intracranial mass lesions. Magn Reson Imaging. 2000;30:662678.
2009;27:198206. 246. Saag MS, Graybill RJ, Larsen RA, et al. Practice guidelines for the man-
214. Chiang IC, Hsieh TJ, Chiu ML, et al. Distinction between pyogenic brain agement of cryptococcal disease. Infectious Diseases Society of America.
abscess and necrotic brain tumour using 3-tesla MR spectroscopy, diffu- Clin Infect Dis. 2000;30:710718.
sion and perfusion imaging. Br J Radiol. 2009;82:813820. 247. Galgiani JN, Ampel NM, Catanzaro A, et al. Practice guideline for the
215. Al MM, Armougom F, Scheld WM, et al. The expansion of the microbio- treatment of coccidioidomycosis. Infectious Diseases Society of America.
logical spectrum of brain abscesses with use of multiple 16S ribosomal Clin Infect Dis. 2000;30:658661.
DNA sequencing. Clin Infect Dis. 2009;48:11691178. 248. Chapman SW, Bradsher RW Jr, Campbell GD Jr, et al. Practice guidelines
216. Al MM, Lonjon M, Richet H, et al. Metagenomic analysis of brain for the management of patients with blastomycosis. Infectious Diseases
abscesses identifies specific bacterial associations. Clin Infect Dis. Society of America. Clin Infect Dis. 2000;30:679683.
2012;54:202210. 249. Wheat J, Sarosi G, McKinsey D, et al. Practice guidelines for the man-
217. Keller PM, Rampini SK, Bloemberg GV. Detection of a mixed infection in agement of patients with histoplasmosis. Infectious Diseases Society of
a culture-negative brain abscess by broad-spectrum bacterial 16S rRNA America. Clin Infect Dis. 2000;30:688695.
gene PCR. J Clin Microbiol. 2010;48:22502252. 250. Adler DE, Milhorat TH, Miller JI. Treatment of rhinocerebral mucormy-
218. Yang SY, Zhao CS. Review of 140 patients with brain abscess. Surg cosis with intravenous interstitial, and cerebrospinal fluid administration
Neurol. 1993;39:290296. of amphotericin B: case report. Neurosurgery. 1998;42:644648.
219. Takeshita M, Kagawa M, Izawa M, et al. Current treatment strategies and 251. Lin YJ, Yang KY, Ho JT, et al. Nocardial brain abscess. J Clin Neurosci.
factors influencing outcome in patients with bacterial brain abscess. Acta 2010;17:250253.
Neurochir (Wien). 1998;140:12631270. 252. Kilincer C, Hamamcioglu MK, Simsek O, et al. Nocardial brain abscess:
220. Arlotti M, Grossi P, Pea F, et al. Consensus document on controver- review of clinical management. J Clin Neurosci. 2006;13:481485.
sial issues for the treatment of infections of the central nervous system: 253. Fleetwood IG, Embil JM, Ross IB. Nocardia asteroides cerebral abscess
bacterial brain abscesses. Int J Infect Dis. 2010;14(suppl 4):S79S92. in immunocompetent hosts: report of three cases and review of surgical
221. Mamelak AN, Mampalam TJ, Obana WG, et al. Improved management recommendations. Surg Neurol. 2000;53:605610.
of multiple brain abscesses: a combined surgical and medical approach. 254. Lee GY, Daniel RT, Brophy BP, et al. Surgical treatment of nocardial brain
Neurosurgery. 1995;36:7685. abscesses. Neurosurgery. 2002;51:668671.
222. Bhand AA. Brain abscessdiagnosis and management. J Coll Physicians 255. Stevens DA, Kan VL, Judson MA, et al. Practice guidelines for diseases
Surg Pak. 2004;14:407410. caused by Aspergillus. Infectious Diseases Society of America. Clin Infect
223. Kocherry XG, Hegde T, Sastry KV, et al. Efficacy of stereotactic aspira- Di. 2000;30:696709.
tion in deep-seated and eloquent-region intracranial pyogenic abscesses. 256. Dobkin JF, Healton EB, Dickinson PC, et al. Nonspecificity of ring enhance-
Neurosurg Focus. 2008;24:E13. ment in medically cured brain abscess. Neurology. 1984;34:139144.
224. Young RF, Frazee J. Gas within intracranial abscess cavities: an indication 257. Nath K, Ramola M, Husain M, et al. Assessment of therapeutic response
for surgical excision. Ann Neurol. 1984;16:3539. in patients with brain abscess using diffusion tensor imaging. World
225. Cavusoglu H, Kaya RA, Turkmenoglu ON, et al. Brain abscess: analysis Neurosurg. 2010;73:6368.
of results in a series of 51 patients with a combined surgical and medical 258. Kourtopoulos H, Holm SE, Norrby SR. The influence of steroids on the
approach during an 11-year period. Neurosurg Focus. 2008;24:E9. penetration of antibiotics into brain tissue and brain abscesses. An experi-
226. Tseng JH, Tseng MY. Brain abscess in 142 patients: factors influencing mental study in rats. J Antimicrob Chemother. 1983;11:245249.
outcome and mortality. Surg Neurol. 2006;65:557562. 259. Hakan T, Ceran N, Erdem I, et al. Bacterial brain abscesses: an evaluation
227. Mathisen GE, Johnson JP. Brain abscess. Clin Infect Dis. 1997;25:763779. of 96 cases. J Infect. 2006;52:359366.
228. Rosenblum ML, Mampalam TJ, Pons VG. Controversies in the manage- 260. Morgan H, Wood MW, Murphey F. Experience with 88 consecutive cases
ment of brain abscesses. Clin Neurosurg. 1986;33:603632. of brain abscess. J Neurosurg. 1973;38:698704.
229. de Louvois J, Hurley R. Inactivation of penicillin by purulent exudates. 261. Nau R, Behnke-Mursch J, Bhler R, et al. Hirnabszess. In: Kommission
Br Med J. 1977;1:9981000. Leitlinien der Deutschen Gesellschaft fr Neurologie, ed. Leitlinien fr
230. Ingham HR, Selkon JB. Metronidazole and brain abscess. Lancet. 1982;2: Diagnostik und Therapie in der Neurologie. Stuttgart: Thieme Verlag;
613614. 2008:408413.
231. Levy RM, Gutin PH, Baskin DS, et al. Vancomycin penetration of a 262. Takeshita M, Kawamata T, Izawa M, et al. Prodromal signs and clinical
brain abscess: case report and review of the literature. Neurosurgery. factors influencing outcome in patients with intraventricular rupture of
1986;18:632636. purulent brain abscess. Neurosurgery. 2001;48:310316.
232. Sjolin J, Eriksson N, Arneborn P, et al. Penetration of cefotaxime and 263. Choudhari KA. Prodromal signs and clinical factors influencing out-
desacetylcefotaxime into brain abscesses in humans. Antimicrob Agents come in patients with intraventricular rupture of purulent brain abscess.
Chemother. 1991;35:26062610. Neurosurgery. 2001;49:481483.
233. Sjolin J, Lilja A, Eriksson N, et al. Treatment of brain abscess with cefo- 264. Harrigan MR. Cerebral salt wasting syndrome. Crit Care Clin. 2001;17:
taxime and metronidazole: prospective study on 15 consecutive patients. 125138.
Clin Infect Dis. 1993;17:857863. 265. Rhode V, van Oosterhout A, Mull M, et al. Subarachnoid haemorrhage
234. Green HT, ODonoghue MA, Shaw MD, et al. Penetration of ceftazidime as initial symptom of multiple brain abscesses. Acta Neurochir (Wien).
into intracranial abscess. J Antimicrob Chemother. 1989;24:431436. 2000;142:205208.
235. Skrap M, Melatini A, Vassallo A, et al. Stereotactic aspiration and 266. Sudhakar KV, Agrawal S, Rashid MR, et al. MRI demonstration of haem-
drainage of brain abscesses. Experience with 9 cases. Minim Invasive orrhage in the wall of a brain abscess: possible implications for diagnosis
Neurosurg. 1996;39:108112. and management. Neuroradiology. 2001;43:218222.
236. Akova M, Akalin HE, Korten V, et al. Treatment of intracranial abscesses: 267. Baumann CK. Multiple bilateral cerebral abscesses with hemorrhage.
experience with sulbactam/ampicillin. J Chemother. 1993;5:181185. J Neurosci Nurs. 1997;29:48, 1314.
237. Gombert ME, duBouchet L, Aulicino TM, et al. Antimicrobial syner- 268. Ballantine HJ, White JC. Brain abscess. Influence of the antibiotic on
gism in the therapy of experimental cerebral nocardiosis. J Antimicrob therapy and morbidity. N Engl J Med. 1953;248:1419.
Chemother. 1989;24:3943. 269. Koszewski W. Epilepsy following brain abscess. The evaluation of possible
238. Krone A, Schaal KP, Brawanski A, et al. Nocardial cerebral abscess cured risk factors with emphasis on new concept of epileptic focus formation.
with imipenem/amikacin and enucleation. Neurosurg Rev. 1989;12: Acta Neurochir (Wien). 1991;113:110117.
333340. 270. Szyfter W, Kruk-Zagajewska A, Borucki L, et al. Evolution in manage-
239. Asensi V, Carton JA, Maradona JA, et al. Imipenem therapy of brain ment of otogenic brain abscess. Otol Neurotol. 2012;33:393395.
abscesses. Eur J Clin Microbiol Infect Dis. 1996;15:653657. 271. Calliauw L, de Praetere P, Verbeke L. Postoperative epilepsy in subdural
240. Asensi V, Carton JA, Maradona JA, et al. Therapy of brain abscess with suppurations. Acta Neurochir (Wien). 1984;71:217223.
imipenema safe therapeutic choice? J Antimicrob Chemother. 1996;37: 272. Nielsen H, Harmsen A, Gyldensted C. Cerebral abscess. A long-term
200203. follow-up. Acta Neurol Scand. 1983;67:330337.
241. Meis JF, Groot-Loonen J, Hoogkamp-Korstanje JA. A brain abscess 273. Legg NJ, Gupta PC, Scott DF. Epilepsy following cerebral abscess. A clini-
due to multiply-resistant Enterobacter cloacae successfully treated with cal and EEG study of 70 patients. Brain. 1973;96:259268.
meropenem. Clin Infect Dis. 1995;20:1567. 274. Leys D, Christiaens JL, Derambure P, et al. Management of focal intra-
242. Ntziora F, Falagas ME. Linezolid for the treatment of patients with central cranial infections: is medical treatment better than surgery? J Neurol
nervous system infection. Ann Pharmacother. 2007;41:296308. Neurosurg Psychiatry. 1990;53:472475.
243. Myrianthefs P, Markantonis SL, Vlachos K, et al. Serum and cerebrospi- 275. Buonaguro A, Colangelo M, Daniele B, et al. Neurological and behav-
nal fluid concentrations of linezolid in neurosurgical patients. Antimicrob ioral sequelae in children operated on for brain abscess. Childs Nerv Syst.
Agents Chemother. 2006;50:39713976. 1989;5:153155.

Scheld_Ch31.indd 548 2/21/14 5:46 PM


Chapter 31: Brain Abscess 549

276. Jenson HB, Levy SR, Duncan C, et al. Treatment of multiple brain ab- 296. Sacktor N, Lyles RH, Skolasky R, et al. HIV-associated neurologic dis-
scesses caused by Bacillus cereus. Pediatr Infect Dis J. 1989;8:795798. ease incidence changes: Multicenter AIDS Cohort Study, 19901998.
277. Bayston R, de Louvois J, Brown EM, et al. Use of antibiotics in penetrating Neurology. 2001;56:257260.
craniocerebral injuries. Infection in Neurosurgery Working Party of British 297. Abgrall S, Rabaud C, Costagliola D. Incidence and risk factors for toxo-
Society for Antimicrobial Chemotherapy. Lancet. 2000;355:18131817. plasmic encephalitis in human immunodeficiency virus-infected patients
278. Kanter MC, Hart RG. Neurologic complications of infective endocarditis. before and during the highly active antiretroviral therapy era. Clin Infect
Neurology. 1991;41:10151020. Dis. 2001;33:17471755.
279. Korinek AM, Baugnon T, Golmard JL, et al. Risk factors for adult 298. Maschke M, Kastrup O, Esser S, et al. Incidence and prevalence of neu-
nosocomial meningitis after craniotomy: role of antibiotic prophylaxis. rological disorders associated with HIV since the introduction of highly
Neurosurgery. 2006;59:126133. active antiretroviral therapy (HAART). J Neurol Neurosurg Psychiatry.
280. Faughnan ME, Lui YW, Wirth JA, et al. Diffuse pulmonary arteriovenous 2000;69:376380.
malformations: characteristics and prognosis. Chest. 2000;117:3138. 299. Svanteson B, Nordstrom CH, Rausing A. Non-traumatic brain abscess.
281. Clayman GL, Adams GL, Paugh DR, et al. Intracranial complications Epidemiology, clinical symptoms and therapeutic results. Acta Neurochir
of paranasal sinusitis: a combined institutional review. Laryngoscope. (Wien). 1988;94:5765.
1991;101:234239. 300. Lu C, Chang W, Chang H. Adult bacterial meningitis in Southern Taiwan:
282. Nunez DA, Browning GG. Risks of developing an otogenic intracranial epidemiologic trend and prognostic factors. J Neurol Sci. 2000;182:3644.
abscess. J Laryngol Otol. 1990;104:468472. 301. Lerner DN, Choi SS, Zalzal GH, et al. Intracranial complications of sinus-
283. Rish BL, Caveness WF, Dillon JD, et al. Analysis of brain abscess after pene- itis in childhood. Ann Otol Rhinol Laryngol. 1995;104:288293.
trating craniocerebral injuries in Vietnam. Neurosurgery. 1981;9:535541. 302. Press OW, Ramsey PG. Central nervous system infections associated with
284. Taha JM, Haddad FS, Brown JA. Intracranial infection after mis- hereditary hemorrhagic telangiectasia. Am J Med. 1984;77:8692.
sile injuries to the brain: report of 30 cases from the Lebanese conflict. 303. Ingham HR, Slekon JB, Roxby CM. Bacteriological study of otogenic
Neurosurgery. 1991;29:864868. cerebral abscesses: chemotherapeutic role of metronidazole. Br Med J.
285. Fischbein CA, Rosenthal A, Fischer EG, et al. Risk factors of brain abscess 1977;2:991993.
in patients with congenital heart disease. Am J Cardiol. 1974;34:97102. 304. Kangsanarak J, Navacharoen N, Fooanant S, et al. Intracranial com-
286. Swanson KL, Prakash UB, Stanson AW. Pulmonary arteriovenous fistulas: plications of suppurative otitis media: 13 years experience. Am J Otol.
Mayo Clinic experience, 19821997. Mayo Clin Proc. 1999;74:671680. 1995;16:104109.
287. Heiro M, Nikoskelainen J, Engblom E, et al. Neurologic manifestations 305. Habib RG, Girgis NI, Abu el Ella AH, et al. The treatment and outcome of
of infective endocarditis: a 17-year experience in a teaching hospital in intracranial infections of otogenic origin. J Trop Med Hyg. 1988;91:8386.
Finland. Arch Intern Med. 2000;160:27812787. 306. Kurien M, Job A, Mathew J, et al. Otogenic intracranial abscess: con-
288. Munoz P, Valerio M, Palomo J, et al. Infectious and non-infectious neuro- current craniotomy and mastoidectomychanging trends in a developing
logic complications in heart transplant recipients. Medicine (Baltimore). country. Arch Otolaryngol Head Neck Surg. 1998;124:13531356.
2010;89:166175. 307. Renton TF, Danks J, Rosenfeld JV. Cerebral abscess complicating den-
289. Hall WA, Martinez AJ, Dummer JS, et al. Central nervous system tal treatment. Case report and review of the literature. Aust Dent J.
infections in heart and heart-lung transplant recipients. Arch Neurol. 1996;41:1215.
1989;46:173177. 308. Kagawa M, Takeshita M, Yato S, et al. Brain abscess in congenital
290. Bonham CA, Dominguez EA, Fukui MB, et al. Central nervous system cyanotic heart disease. J Neurosurg. 1983;58:913917.
lesions in liver transplant recipients: prospective assessment of indica- 309. Bleggi-Torres LF, de Medeiros BC, Werner B, et al. Neuropathological find-
tions for biopsy and implications for management. Transplantation. ings after bone marrow transplantation: an autopsy study of 180 cases.
1998;66:15961604. Bone Marrow Transplant. 2000;25:301307.
291. Uckan D, Cetin M, Yigitkanli I, et al. Life-threatening neurological com- 310. Clifford DB. Focal brain lesions in people with HIV. Curr Treat Options
plications after bone marrow transplantation in children. Bone Marrow Neurol. 1999;1:167172.
Transplant. 2005;35:7176. 311. Ammassari A, Cingolani A, Pezzotti P, et al. AIDS-related focal brain
292. Maschke M, Dietrich U, Prumbaum M, et al. Opportunistic CNS infection lesions in the era of highly active antiretroviral therapy. Neurology.
after bone marrow transplantation. Bone Marrow Transplant. 1999;23: 2000;55:11941200.
11671176. 312. Arseni C, Ciurea AV. Brain abscesses. Comments with reference to 810 cases.
293. Antonini G, Ceschin V, Morino S, et al. Early neurologic complications Zentralbl Neurochir. 1981;42:7783.
following allogeneic bone marrow transplant for leukemia: a prospective 313. Miller ES, Dias PS, Uttley D. CT scanning in the management of intracra-
study. Neurology. 1998;50:14411445. nial abscess: a review of 100 cases. Br J Neurosurg. 1988;2:439446.
294. Graus F, Saiz A, Sierra J, et al. Neurologic complications of autologous 314. Grimstad IA, Hirschberg H, Rootwelt K. 99mTc-hexamethylpropyle-
and allogeneic bone marrow transplantation in patients with leukemia: neamine oxime leukocyte scintigraphy and C-reactive protein levels in the
a comparative study. Neurology. 1996;46:10041009. differential diagnosis of brain abscesses. J Neurosurg. 1992;77:732736.
295. Roemer E, Blau IW, Basara N, et al. Toxoplasmosis, a severe complication 315. Jamjoom AB. Short course antimicrobial therapy in intracranial abscess.
in allogeneic hematopoietic stem cell transplantation: successful treat- Acta Neurochir (Wien). 1996;138:835839.
ment strategies during a 5-year single-center experience. Clin Infect Dis. 316. Hirschberg H, Bosnes V. C-reactive protein levels in the differential diag-
2001;32:E1E8. nosis of brain abscesses. J Neurosurg. 1987;67:358360.

Scheld_Ch31.indd 549 2/21/14 5:46 PM


CHAPTER 32 EPIDURAL ABSCESS
HANS-WALTER PFISTER, MATTHIAS KLEIN, ALLAN R. TUNKEL, AND W. MICHAEL SCHELD

less often than in the thoracic or lumbar regions (Table 32.1).


SPINAL EPIDURAL ABSCESS In recent years, the increasing use of spinal interventions for
pain management has caused a relative increase of lumbar
First described by the Italian anatomist Morgagni in the epidural abscesses (2,10). A recent series of 40 cases reported
eighteenth century, a spinal epidural abscess is a suppurative that 11 of 16 patients who were injection drug users had epi-
infection in the space around the spinal cord located between dural abscesses in the cervical region (6). It was postulated
the dura mater and the vertebral periosteum (1). It is both a that this may reflect the venous and lymphatic drainage of the
medical and a surgical emergency, requiring prompt and ac- upper extremity. Whereas the rich circulation ensures ample
curate diagnosis and treatment to prevent irreversible spinal blood supply to the spinal cord, it may also act as a conduit
cord dysfunction, paralysis, and death. along which infection may spread. Infection tracking along
these pathways is the likely explanation for the longitudinal
extension of spinal epidural abscesses that usually affect mul-
Epidemiology tiple adjacent spinal segments. On average, 3.3 2.7 (mean
standard deviation [SD]) spinal cord segments were re-
Although spinal epidural infections are uncommon, recent ported to be involved in five clinical studies with 133 patients
studies suggest that the incidence of this infection is increasing (11,12,1416); however, infections of the entire length of the
(2). Although incidence rates of spinal epidural abscesses have spinal epidural space have been reported (panspinal infection)
not been calculated from population-based data, estimates (3,7,17). Of note, spinal epidural abscesses can also be found
from recent case series noted an increase from 0.2 to 1.2 cases at noncontiguous sites, such as the cervical and lumbar spinal
per 10,000 admissions in the mid-1970s (3) to significantly cord (18).
higher incidence rates of up to 1.96 (4), 2.8 (5), or 11.31 cases
(6) per 10,000 admissions to large tertiary care centers in
the 1980s and early 1990s. The reasons for the increasing
incidence of epidural spinal infections seem to be an aging
population, increased use of spinal procedures, and a rising
rate of injection drug use (47). However, a change in refer-
ral patterns or a greater recognition (more widespread use of
magnetic resonance imaging [MRI]) must also be considered.
Approximately 70% of patients with spinal epidural
abscess are between 31 and 70 years of age, with no obvious
predilection for any given decade (1). The youngest patient in
the literature was 10 days of age and the oldest was 87 years
of age (1). Almost all large case series observed a preference
for the male gender, with an average male-to-female ratio of
1.0:0.56 (1). The reasons for this male predominance are not
apparent. However, some risk factors associated with spinal
epidural abscesses are more common in males, such as alcohol
abuse, injection drug usage, and trauma (1).

Pathogenesis and Pathophysiology


Below the foramen magnum, the epidural space extends the
length of the spine. It is composed of two compartments:
(a) a true space posterior and lateral to the spinal cord con-
taining a cushioning layer of fat embedded with penetrating
arteries and an extensive venous plexus, and (b) a potential an-
terior space where the dura adheres to the posterior surface of
the vertebral body (8,9) (Fig. 32.1). The epidural space is cir-
cumferential around the spinal cord distal to the second sacral
segment, the terminal point of its anterior attachment. Given
these anatomic considerations, it is not surprising that spinal
epidural abscesses are located posteriorly in most cases (3,5). FIGURE 32.1 Vertebral body and sub-/epidural space. Transverse
In addition to the boundary imposed by the anterior at- section of a vertebral body showing the location of the epidural and
tachment to the vertebral canal, the dimensions of the epidural subdural spaces. (a) vertebral body, posterior arch; (b) vertebral body,
space vary from segment to segment. In the cervical region anterior arch; (c) dura mater; (d) arachnoid; (e) pia mater; (f, beige) epi-
where the epidural space is smallest, epidural abscesses occur dural space; (g, yellow) subdural space; (h, blue) subarachnoid space.

550

Scheld_Ch32.indd 550 2/21/14 8:29 PM


Chapter 32: Epidural Abscess 551

TA B L E 3 2 . 1 TA B L E 3 2 . 2
LOCATION OF SPINAL EPIDURAL ABSCESSES SELECTED IMPORTANT RISK FACTORS AND
IN 889 PATIENTS SOURCES OF INFECTION IN 1,005 PATIENTS WITH
SPINAL EPIDURAL ABSCESS
Location No. of Patients (%)
Risk Factors and/or Sources of Infection No. of Patientsa (%)
Cervical 172 (19.4)
Cervicothoracic 60 (6.8) Immunocompromising conditions
Thoracic 294 (33.0) Diabetes mellitus 178 (15.4)
Thoracolumbar 59 (6.6) Alcoholism 52 (4.5)
Lumbar 150 (16.9) Malignancy 26 (2.3)
Lumbosacral 140 (15.7) Chronic kidney disease 24 (2.1)
Sacral 3 (0.3) Corticosteroid therapy 20 (1.7)
Cervicothoracolumbar 8 (0.9) Trauma and invasive procedures
Thoracolumbosacral 3 (0.3) Spinal trauma 55 (4.8)
TOTAL 889 (100) Extraspinal trauma 41 (3.5)
Epidural anesthesia 43 (3.7)
Data from references 1, 1113, 35, 36. Extraspinal operations 42 (3.6)
Spinal operations 39 (3.4)
Paravertebral injections 12 (1.0)
Lumbar puncture 1 (0.1)
Source of Infection Other sources of infection
Infection may be introduced into the epidural space by direct Skin infections 133 (11.5)
extension from a contiguous infection (about 1/3 of cases) or Injection drug use 98 (8.5)
by hematogenous seeding from a remote site (about half of Vertebral osteomyelitis/discitis 59 (5.1)
cases); in the remaining cases, the source of infection is not
Pulmonary/mediastinal infections 45 (3.9)
identified (7) (Table 32.2). Contiguous infections include (1)
vertebral osteomyelitis/discitis; retropharyngeal, perinephric, Sepsis 39 (3.4)
paraspinal, or psoas abscesses; decubitus ulcers; and persistent Urinary tract infection 23 (2.0)
dermal sinus tracts (Fig. 32.2). Local invasion from superficial Vascular access 17 (1.5)
infections can also occur following penetrating injuries, in-
Paraspinal abscess 14 (1.2)
cluding prior surgery or spinal procedures including placement
of catheters or stimulators; epidural drug injections; paraver- Degenerative spinal disorders 50 (4.3)
tebral injections; computed tomography (CT)guided needle Other risk factors or sources of 149 (12.9)
biopsy; and, very rarely, lumbar puncture (1). Metastatic seed- infectionb
ing may result from any bacteremic infection. Skin and soft TOTAL 1,155
tissue infections are typically found, but sources have also in-
cluded endocarditis and infected intravascular catheters, respi- a
Multiple risk factors were present in many patients.
ratory tract infection, urinary tract infection, dental abscesses, b
For example, pharyngitis, wound infection, endocarditis, sinusitis
abdominal infections, and complications from gastrointes- or upper respiratory tract infection, HIV infection, dental abscess,
tinal surgery (1). In contrast to spinal epidural abscesses in retropharyngeal abscess, psoas abscess, liver disease, adrenal insuffi-
adults (Fig. 32.3), adjacent vertebral osteomyelitis is distinctly ciency, ulcerative colitis, Crohn disease, systemic lupus erythematosus,
uncommon in children with this infection; thus, most spinal dermal sinus, herpes zoster neuralgia, decubitus ulcer, pregnancy, and
delivery.
epidural abscesses in children have been attributed to hema- Data from references 1, 12, 16, 19, 20, 35.
togenous seeding from a distant focus (21).
One possible explanation for the development of an epi-
dural abscess after a recent or remote trauma including prior
spinal surgery is that a small hematoma or area of damaged and 16% for cesarean sections) reported only a single case
tissue may provide a fertile area for subsequent hematogenous of an epidural abscess (23). Another mostly retrospective
seeding (22). Whether antecedent trauma is indeed a true risk study estimated the risk at approximately 1 per 5,000 cath-
factor for the subsequent development of a spinal epidural eters (24). A recent prospective study identified nine patients
abscess or whether patient recall bias has influenced this his- with epidural abscess after 17,372 epidural catheters (1 per
tory has not been systematically examined. Of course, spinal 1,930 catheters), with a mean catheterization time of 11 days
trauma may also contribute to the development of an infection (25). Immunocompromise and longer duration of catheter-
by creating a site of entry for microorganisms into the epidural ization were associated with an increased risk of an epidural
space (disruption of anatomic barriers). abscess in that study: only one of the nine patients had no
The reported risk of an epidural abscess after epidural complicating disease (four had malignancies, two had diabe-
catheterization varies widely in the literature. Prolonged use tes mellitus, one had multiple trauma, and one had chronic
of epidural catheters, significant comorbidity, older age, injec- obstructive airway disease); furthermore, all patients with
tion drug use, and diabetes mellitus contribute to the rate of abscess had epidural catheters in situ for 3 days or longer. This
infection (2). A retrospective analysis of 505,000 extradural is best illustrated by the relatively high risk of long-term epi-
blocks in obstetric practice (84% for relief of pain in labor dural treatment of chronic pain (mostly patients with cancer),

Scheld_Ch32.indd 551 2/21/14 8:29 PM


552 Part III: Bacterial and Mycoplasmal Infections

FIGURE 32.2 Congenital dermal sinus as a risk factor for intraspinal infection. T2-weighted MRI
(A, sagittal; B, transversal) of this 26-year-old patient shows a dermal sinus (arrows, incidental finding)
extending to the intraspinal region (C6C7). The patient had a previous surgical decompression of an
Arnold-Chiari malformation.

which was complicated by an epidural abscess in 3%, 4%, treatment and enteroepidural fistulas may be relevant), systemic
or even 12% of patients in three recent studies (13,26,27). lupus erythematosus, chronic granulomatous disease, cortico-
In another study, none of 1,062 patients who had epidural steroid therapy, and acquired immunodeficiency syndrome (1).
anesthesia for a period of less than 14 days developed a serious Compared with the damage produced by spinal tumors
infectious spinal complication (28). and cysts, the damage produced by bacterial spinal epi-
Underlying medical conditions that have been associated dural abscesses is often out of proportion to the size of the
with spinal epidural abscesses include degenerative joint disease, inflammatory mass, that is, the lesion of the spinal cord can
diabetes mellitus, injection drug use, alcoholism and cirrhosis, be more extensive than can be accounted for by mechani-
malignancy, renal failure and hemodialysis (2931), pregnancy, cal effects of compression alone (3). This important feature
ulcerative colitis and Crohn disease (both immunosuppressive may be attributed to many factors including thrombosis and

FIGURE 32.3 Pyogenic osteomyelitis and epidural abscess. The 51-year-old patient, with 1 month of back
pain, presented with fever and cauda equina syndrome. A: On T1-weighted MRI, ventral presacral (arrow)
and epidural (arrowheads) contrast enhancement correspond with abscess formation. B, C: In addition to
the presacral abscess (arrow), T2-weighted sequences reveal hyperintensities of the os sacrum (asterisk) and
the paraspinal muscle (section sign), indicating osteomyelitis and infection of the muscle. Staphylococcus
aureus was identified as the causative pathogen from blood culture.

Scheld_Ch32.indd 552 2/21/14 8:29 PM


Chapter 32: Epidural Abscess 553

thrombophlebitis of veins draining the spinal cord with resul-


tant edema and venous infarction (3); compression of the arte- Microbiology
rial supply to adjacent cord segments with local ischemia and
infarction; focal areas of vasculitis induced by the adjacent in- A microbiologic diagnosis is usually made from blood and/
flammatory mass; or bacterial exotoxin production, especially or intraoperative cultures. Polymicrobial spinal epidural
by Staphylococcus aureus. abscesses are a rarity, blood cultures are positive in about
Still, a rabbit model of spinal epidural abscess has demon- 60% of the patients (7), and pathogens identified by blood
strated that the damage to the spinal cord is more consistent culture are almost always identical with the infectious agents
with mechanical compression than with ischemia or infarction cultured from abscess pus. This situation is, therefore, com-
(32,33). This model, using a clinical isolate of Staph. aureus, pletely different from that in patients with brain abscesses.
also demonstrated many of the pathologic features seen clini- In the latter, blood cultures are positive in only about 10%
cally in spinal epidural abscesses: preservation of gray mat- of patients, and even if the blood cultures are positive, they
ter, white matter edema, vacuolization, liquefaction, myelin do not allow for firm conclusions on the microbial spectrum
degeneration, and axonal swelling. This pattern is consistent of the brain abscess because brain abscess pus often grows
with compressive damage, as spinal cord ischemia is typically mixed cultures (see Chapter 31). The microbial spectrum
characterized by necrosis of the gray matter and relative pres- of spinal epidural abscesses also differs greatly from that of
ervation of the white matter (33). Furthermore, the posterior brain abscesses, as Staph. aureus (Fig. 32.4) is the most com-
spinal arteries and the anterior spinal artery were patent mi- mon etiologic agent in children and adults (responsible for
croangiographically in paraplegic animals with minimal and about two thirds of spinal epidural abscesses). Coagulase-
moderate cord compression and even in some cases of large positive and negative (mostly Staph. epidermidis) staphylo-
epidural abscesses with severe degrees of compression (32). cocci cause approximately 70% of spinal epidural abscesses
Dorsal and anterior spinal veins and the anterior epidural (note that in some studies, up to 15% of the isolates were
venous plexus were also patent in animals with minimal or methicillin-resistant Staphylococcus species [34] with a fre-
moderate compression. Only in rabbits with severe cord com- quency up to 40% in some studies [7,35]). The remaining
pression, the dorsal spinal vein and the perforating arterioles 30% of cases are caused by a wide spectrum of infectious
were occluded ipsilaterally to the abscess. With profound agents (Table 32.3). Most streptococcal spinal epidural ab-
compression, all blood flow was noted to cease. The authors, scesses are due to viridans streptococci and Streptococcus
therefore, concluded that the initial neurologic deficit results pneumoniae (1,45). Escherichia coli is responsible for most
from compression and that vascular compromise (as a result infections due to Enterobacteriaceae. Much less frequently
of compression and probably not due to vasculitis or throm- Proteus, Enterobacter, Salmonella, Serratia, Citrobacter,
bosis) is an important factor in the final pathogenesis of spinal and Klebsiella species have been reported (1). Spinal epidural
epidural abscess. Although these rabbit experiments (32,33) infections due to Pseudomonas aeruginosa are relatively
suggested a primary role for mechanical compression, other uncommon but require particular attention because of this
animal models favored an additive adverse effect of compres-
sion and ischemia on neurologic function (7).
TA B L E 3 2 . 3
MICROBIOLOGY OF SPINAL EPIDURAL ABSCESS:
CULTURE RESULTS (ABSCESS PUS, BLOOD,
AND/OR CSF) FROM 934 PATIENTS WITH
SPINAL EPIDURAL ABSCESSES

Infectious Agent No. of Patients (%)

Staphylococcus aureus 611 (65.5)


Coagulase-negative staphylococci 40 (4.3)
Streptococci 71 (7.6)
Enterobacteriaceae 48 (5.1)
Pseudomonas speciesa 18 (1.9)
Mycobacterium tuberculosis 17 (1.8)
Other bacteriab 15 (1.6)
Mixed bacterial infections 33 (3.5)
Fungic 13 (1.4)
Parasitesd 3 (0.3)
No growth or agent not specified 65 (6.9)

a
Mostly Pseudomonas aeruginosa.
b
For example, Neisseria, Acinetobacter, Actinomyces (37), Actinobacillus,
Brucella (3840), Listeria (41), Clostridium, Propionibacterium, Nocardia
(42), Bacteroides, Morganella, Eikenella, and Haemophilus (43,44) species.
c
Mostly Aspergillus fumigatus, single cases due to Sporothrix schenckii
or Candida species.
d
FIGURE 32.4 Staphylococcus aureus. Gram stain reveals violet (gram- Two cases due to Dracunculus medinensis, one due to Echinococcus
positive) bacteria, which typically present in clusters (arrows). (Courtesy granulosus.
of Dr. Andreas Wieser, Max von Pettenkofer-Institute for Hygiene and Data from references 1, 11, 12, 16, 19, 20.
Microbiology, Ludwig Maximilians University of Munich.)

Scheld_Ch32.indd 553 2/21/14 8:29 PM


554 Part III: Bacterial and Mycoplasmal Infections

pathogens resistance to many antibiotics. Anaerobes, such (4 of whom had documented epidural abscess) with or with-
as Bacteroides, Peptostreptococcus, or Fusobacterium spe- out meningitis, and 8 (12%) had posterior circulation stroke
cies are also an uncommon cause of spinal epidural abscesses. (with or without meningitis). The median time from the
Spinal epidural abscess is rarely caused by agents of actino- last epidural glucocorticoid injection to symptom onset was
mycosis or nocardiosis (1,46). The incidence of mycobacterial 18 days. Eight patients (12%) died, and 7 of them had stroke.
epidural infections (Fig. 32.5) varies greatly geographically. A total of 22 patients had laboratory confirmation of fungal
Although they are very rare, for example, in studies from infection, either Exserohilum rostratum infection (21 patients)
North America or Western Europe, mycobacteria were re- or Aspergillus fumigatus infection (1 patient) (49).
sponsible for 8 (28%) of 29 cases of spinal epidural abscesses Parasitic causes of epidural abscesses have also been reported,
in a series from Taiwan (19) and 19 (53%) of 36 cases in a including Echinococcus (56), guinea worm (Dracunculus medi-
Turkish investigation (47). nensis) (57), and Schistosoma mansoni (58).
Fungal etiologies of spinal epidural abscesses include The microbiology of spinal epidural abscesses in children
Aspergillus species (48,49), Candida species (50), Blastomyces does not differ substantially from that in adults. In 2001, a
dermatitidis (51), Coccidioides immitis (52), Cryptococcus review of the literature summarized 34 cases of childhood
neoformans (53), mucormycosis (54), Sporothrix schenckii spinal epidural abscesses (21): Staph. aureus was cultured in
(14), and Pseudallescheria boydii (55). Recently, an outbreak 21 patients (62%), streptococci in 2 (6%, S. agalactiae in one
of fungal meningitis after epidural or paraspinal glucocorti- and viridans streptococci in another), Salmonella enteritidis
coid injection with contaminated methylprednisolone has in 2 (6%), E. coli in 1 (3%), Pseudomonas aeruginosa in 1
been described (48,49). The outbreak is ongoing and involves (3%), Mycobacterium bovis in 1 (3%), Candida tropicalis in
multiple states. Clinical data from 66 patients showed that 47 1 (3%), and Aspergillus flavus in 1 (3%). Multiple organisms
(71%) had meningitis alone, 11 (17%) suffered from cauda were isolated in one patient and no organisms were isolated in
equina syndrome or focal infection nearby the injection site three children (9%).

FIGURE 32.5 Tuberculous discitis, osteomyelitis, and epidural abscess. A: Plain film in the lateral projec-
tion of the thoracolumbar junction demonstrates narrowing of the disc space, rarefaction and erosion
of the cortical margins of the subjacent vertebrae (arrows), and early gibbus formation. CT scans at the
level of (B) T12 and (C) L4 after intravenous contrast demonstrate osseous destruction of T12 with an
associated epidural abscess (arrows) causing compression of the thecal sac. Note the left-sided psoas
abscess in part C (asterisk). Sagittal (D) and axial (E) T1-weighted MRI scans 3 days later demonstrate
near-complete dissolution of the disc space between T11 (number sign) and T12 (section sign), extensive
osseous destruction, and paraspinal and epidural (arrows) inflammatory disease. F: Sagittal and (G) axial
T1-weighted MRI scans after intravenous administration of gadoliniumdiethylenetriamine pentaacetic
acid demonstrate abnormal enhancement in the T11T12 disc space (F, arrowheads), in the subjacent
vertebrae (F, arrows), and in the paraspinal and epidural compartment (G, arrowheads), indicative of
active inflammatory disease. H: CT-guided percutaneous needle (arrows) aspiration of the psoas abscess
with the patient in the prone position. Cultures yielded Mycobacterium tuberculosis.

Scheld_Ch32.indd 554 2/21/14 8:29 PM


Chapter 32: Epidural Abscess 555

Although the prognostic significance of distinguishing


Clinical Manifestations acute from chronic spinal epidural abscesses has recently been
questioned, patients with acute presentations may have higher
The initial clinical signs and symptoms of a spinal epidural ab- peripheral leukocyte concentrations (mean values ranging
scess, characteristically including fever, back pain, and malaise, from 12,300 to 16,700 cells/L [35]) and higher tempera-
may be subtle. A spinal epidural abscess is often not suspected tures when compared to those of patients with long-standing
on admission. The most common initial misdiagnoses are menin- symptoms who may have only mild elevations in these two
gitis, intervertebral disc prolapse, vertebral osteomyelitis, lumbar parameters (mean peripheral leukocyte concentrations rang-
degenerative joint disease, spinal neoplasm, transverse myelitis, ing from 7,100 to 11,900 cells/L). Other authors, however,
spinal ischemia, and urinary tract infection (1). The duration of reported similar peripheral leukocyte concentrations in acute
symptoms is variable; they may be present for just a few days or up and chronic cases (6). In both presentations, the erythrocyte
to several months before the patient presents for evaluation (4,59). sedimentation rate (ESR) is generally elevated (25 to 30 mm
The four-stage clinical progression of a spinal epidural per hour); however, the magnitude of the ESR is not a reliable
abscess was first described by Rankin and Flothow (60) more clinical clue as to the chronicity of the infection (5,6). Serial
than 50 years ago. In addition to fever and malaise, the first ESR measurements showed good correlation between resolu-
localizing symptoms are backache and focal vertebral pain and tion of clinical signs and decreases in ESR (34). Regardless of
tenderness on examination. This is followed by root pain the clinical course, the average peripheral leukocyte concen-
manifested by radiculopathy and/or paresthesias, which often trations were 15,700 cells/L in 218 patients from the litera-
may be described as electric shocks. Spinal cord dysfunc- ture (range, 1,500 to 42,000 cells/L) and the average ESR
tion, the third stage, is characterized by motor and sensory was 77 mm (range, 2 to 150 mm) in 117 patients from the
deficits or by bladder or bowel dysfunction. This is followed literature (1). Recent studies have also reported elevated serum
by the final stage of complete paralysis (Table 32.4). C-reactive protein (CRP) levels in patients with spinal epidural
Although early symptoms of backache may be indolent and abscesses, 15.0 9.5 mg/dl in one study (20). Serum CRP
persist for weeks or months, back pain usually progresses to levels were also shown to respond more quickly to therapeu-
root pain within 3 to 4 days followed by early signs of spinal tic interventions than, for example, the ESR (61). Similarly, a
cord dysfunction within the subsequent 4 to 5 days. The neuro- persistent strong elevation of the CRP value after 8 days post-
logic deficits at this stage are usually reversible; however, rapid surgery was shown to be associated with a poor outcome (20).
surgical intervention at this point may be crucial because pro- Blood cultures will be positive on average in 72% of patients
gression to complete paralysis may occur within a few hours (mean, range 64% to 82% in three recent studies [4,16,59]).
regardless of the chronicity of the process up to this point. Cerebrospinal fluid (CSF) examination is typically that of a
Specific neurologic signs depend on the level of spinal cord parameningeal focus of infection with elevated protein levels
involvement and is another factor that influences the differen- and increased leukocytes. The CSF leukocytes may be a mix-
tial diagnosis at the time of presentation. ture of polymorphonuclear leukocytes and mononuclear cells or
The presentation of a spinal epidural abscess has historically predominantly polymorphonuclear leukocytes in most patients
been described as acute (symptoms persisting for 2 weeks at (3,59). The CSF leukocyte count was 40 cells/L (median, range
the time of presentation) or chronic (symptoms for 2 weeks at 0 to 27,000 cells/L) in one study (59) and 60 cells/L (mean,
the time of presentation) (3,5). The rate of progression of the ini- range 0 to 820 cells/L) in another report (3). Extremely elevated
tial stages may suggest the route of infection: acute presentations leukocyte counts in the CSF may indicate that the spinal needle
often represent hematogenous seeding and rapid expansion of has entered a lumbar epidural abscess (3,59). CSF protein values
the inflammatory mass, whereas chronic presentations are more are elevated in approximately 90% of patients (59). Markedly
likely the result of a gradually expanding contiguous infection. elevated CSF protein levels (350 mg/dL) are often predictive
of a complete block of the spinal canal; however, an elevated
protein level of lesser magnitude is found in patients with spinal
TA B L E 3 2 . 4 block in nearly two thirds of cases (5). CSF glucose concentra-
PRESENTING SIGNS AND SYMPTOMS IN tions were reported to be decreased (50 mg/dL) in more than
942 PATIENTS WITH SPINAL EPIDURAL ABSCESS half of the cases (59). Except when there is concomitant menin-
gitis, CSF Gram stains rarely demonstrate organisms, and spinal
Signs and Symptoms No. of Patients (%) fluid cultures are positive in only 15% of patients (mean, range
7% to 25% [4,5,59,62]), also similar to other parameningeal
Fever 617 (66) infections. Most authors recommend against performing a lum-
Initial pain symptom: bar puncture in patients with an adjacent spinal epidural abscess
because of the possibility of inducing meningitis or subdural in-
Back pain 679 (72) fection if the needle traverses the epidural space or the potential
Local tenderness 190 (20) risk of neurologic deterioration if performed below the spinal
Radicular pain 209 (22) block (59). Perhaps the principal reason not to perform lumbar
Beginning neurologic deficit: puncture is the meager information that this test provides. The
CSF alterations are nonspecific and when CSF cultures are posi-
Muscle weakness 226 (24) tive, so usually are the blood cultures (59).
Incontinence 232 (25) At the time of surgery, findings may range from frank pus
Sensory deficit 128 (14) to granulomatous tissue. Whereas acute cases are more likely
Advanced neurologic deficit: to have purulent material, this correlation was not found con-
sistently in other case series (4,63). Overall, intraoperative
Paraparesis/paraplegia 313 (33) cultures have the greatest chance of yielding a microbiologic
Tetraparesis/tetraplegia 34 (4) diagnosis; on average 86% of all cases have positive cultures
(mean, range 82% to 90% [35,16,19,59]). However, when a
Data from references 1, 16, 20. patient has been receiving antibiotics for more than 1 week be-
fore culture, diagnostic cultures are unlikely to be positive (5).

Scheld_Ch32.indd 555 2/21/14 8:29 PM


556 Part III: Bacterial and Mycoplasmal Infections

most commonly occurs in the lumbar spine, and the bodies of


Differential Diagnosis the vertebrae are affected more often than the posterior elements.
Helpful ancillary plain film findings include a mass effect or dis-
Signs of fever and back pain should raise suspicion of a spinal placement of the larynx in a retropharyngeal abscess or scoliosis
epidural abscess, especially when spinal tenderness or radicu- and displacement of bowel loops in a lumbar paraspinal abscess.
lar signs are demonstrated on physical examination. However, Normal plain films do not exclude the presence of a spinal epi-
these signs and symptoms may not always be present and lack dural abscess, particularly in acute presentations. In this setting,
specificity for this diagnosis. About half of the patients with additional radiographic examinations are almost always needed
spinal epidural abscess are initially misdiagnosed (7), particu- for further evaluation. Similar to plain films, bone scans often
larly when patients present in early clinical stages (35,59,62). provide clinically not useful information that may be misleading
One review documented that spinal epidural abscesses took in this setting.
significantly longer to diagnose in patients when it was not in- Myelography, which will not directly visualize a spinal epi-
cluded among the differential diagnoses at admission: 6.6 days dural abscess, will demonstrate the associated mass effect on
versus 1.9 days (59). The main differential diagnoses include the spinal cord, thecal sac, or nerve roots. A spinal epidural
intervertebral disc prolapse, spinal tumors, transverse myelitis, abscess can result in a complete extradural obstructive block to
epidural hematoma, vertebral osteomyelitis, spinal subdural the flow of water-soluble contrast. However, in patients with a
or intramedullary abscess, spinal arteriovenous malformation, prominent ventral epidural space in the lower lumbar spine due
and spinal arterial ischemia. The clinical picture of spinal epi- to abundant epidural fat, an epidural abscess may not be de-
dural abscess can also be mimicked by acute spinal cord dys- tected on myelography if it causes indentation or obliteration
function due to myelitis during bacterial meningitis (64). of the epidural fat without indenting the opacified thecal sac.
Both CT and MRI can directly visualize an epidural in-
flammatory mass (6568,70). As is true in the neuroimag-
Approach to Diagnosis ing of many diseases of the brain and spinal canal, MRI has
proven superior to CT for evaluation of patients with epi-
The imaging method of choice for diagnosis of spinal epidural dural abscesses. MRI obviates the need for myelography and
abscess is contrast-enhanced MRI (6570). However, availabil- CT in most cases. The advantages of MRI include its ability
ity, contraindications (e.g., heart pacemaker), or insufficient co- to image long segments of the spinal canal in multiple planes
operation of the patient sometimes requires the use of the other (Figs. 32.6 and 32.7), thereby enabling precise delineation of
methods, such as (postmyelography) CT. Plain films of the spine all loculations of inflammatory tissue; its ability to image all
will not directly visualize the spinal epidural abscess but may compartments around the spinal canal into which an inflam-
demonstrate certain findings that can be an indication of the matory process might extend; its ability to directly visualize the
presence of infection in the spinal canal (66,67). These findings neural elements with high-contrast resolution without the need
include intervertebral disc space narrowing in discitis and loss for intrathecal contrast; its noninvasiveness, which is a particu-
of definition or destruction of both the inferior cortical margin lar advantage over myelography, because of the desire to avoid
of one vertebral body and the superior cortical margin of the inadvertent puncture of the abscess or iatrogenic spread of in-
contiguous vertebral body in osteomyelitis. Rarefaction and loss fection from the epidural to the subarachnoid space following
of bony trabeculae can also be seen. In advanced or rapidly pro- introduction of a needle into the spinal canal. Furthermore,
gressive cases, vertebral body collapse (or dissolution) and gibbus with MRI, patient follow-up is facilitated and assessment of
formation may be seen on plain films. In general, osteomyelitis response to therapy is readily performed. The ability of MRI

FIGURE 32.6 Spinal epidural abscess in a


73-year-old patient. T1-weighted MRI shows
epidural lesions (arrow) with surrounding
contrast enhancement located (A) at C3/C4
and (B) T7T9. Insets show corresponding
axial sections. Staphylococcus aureus was
identified in blood and cerebrospinal fluid.

Scheld_Ch32.indd 556 2/21/14 8:29 PM


Chapter 32: Epidural Abscess 557

FIGURE 32.7 Epidural spinal abscess. The


82-year-old patient, with 4 weeks of back
pain, presented with fever and progres-
sive paraparesis. Sagittal T1-weighted MRI
shows a contrast-enhancing space-occupying
lesion (arrowheads) ranging from (A) T4
to (B) L3 and contrast enhancement in the
paraspinal muscles (asterisk). Insets show
corresponding axial sections. Staphylococcus
aureus was isolated from blood culture and
surgical material.

to distinguish active inflammatory tissue from chronic granu- In the presence of discitis, a characteristic finding on MRI is
lation tissue is further enhanced by intravenous administra- abnormal high signal intensity in the disc on T2-weighted im-
tion of gadolinium. MRI demonstrates epidural inflammatory ages (Fig. 32.9). This is in contradistinction to degenerative
disease to be a soft tissue mass, which compared to the spinal disc disease, which demonstrates low signal intensity. Vertebral
cord is isointense on T1-weighted images and hyperintense on osteomyelitis is seen as low signal intensity on T1-weighted im-
T2-weighted images. Diffusion-weighted imaging may show a ages in the involved marrow cavity, with high signal intensity
signal abnormality within the spinal abscess (70,71). The extra- on T2-weighted images. These osseous changes are usually ob-
dural location of disease and the associated mass effect on the served in conjunction with inflammatory disease of the subja-
thecal sac, spinal cord, or cauda equina are generally readily cent intervertebral disc as described.
appreciated on MRI, as is the extradural location of disease
and the associated mass effect on the thecal sac. The presence of
concomitant paraspinal abscesses is easily identified (Fig. 32.8). Treatment
A spinal epidural abscess is a neurosurgical emergency. The
neurologic deterioration to severe spinal cord dysfunction can
occur in just a few hours, making the diagnosis and treatment
imperative (Fig. 32.10). The initial treatment usually consists
of decompressive surgery and drainage of the abscess, eradica-
tion of the primary underlying infectious focus (if detectable),
and parenteral antibiotic therapy.
Posterior epidural abscesses are usually treated by decom-
pressive laminectomy, removal of pus or granulation tissue, and
postoperative irrigation with sterile saline through extradural
drains for several days. Percutaneous, CT-guided needle aspira-
tion might be a rational alternative to surgical decompression
in selected patients with posteriorly located abscesses, which
extend across multiple spinal segments, exhibit minimal cord
compression, and demonstrate stable neurologic examination
results with minimal or no neurologic deficits (72). For the
FIGURE 32.8 Epidural spinal abscess. Transverse T1-weighted MRI in treatment of anterior epidural abscesses, an anterior approach
this 82-year-old patient shows a contrast-enhancing space-occupying epi- may be necessary. Adjacent sources of infection should also be
dural lesion (arrow) and contrast enhancement (asterisk) in the paraspinal cured surgically (e.g., vertebrectomy for severe osteomyelitis
muscles. Enterococcus faecium was identified from the paraspinal muscles. with vertebral body destruction).

Scheld_Ch32.indd 557 2/21/14 8:29 PM


558 Part III: Bacterial and Mycoplasmal Infections

FIGURE 32.9 Spondylodiscitis. The 65-year-


old patient presented with fever and a history
of back pain for 4 weeks. Clinical exami-
nation revealed dysaesthesia of the left leg,
corresponding to dermatome L5. In compari-
son to (A) plain T1-weighted sagittal MRI,
(B) contrast enhancement of the vertebra L4
and L5 and the intervertebral space L4/L5
was noted. C: On T2-weighted MRI, a hyper-
intense signal of the intervertebral space was
found. CT-guided puncture (L4/L5) revealed
granulocytic inflammatory changes. Isolation
of bacteria was not successful. For A, B, and
C: L4 (asterisk), L5 (number sign), and L4/L5
intervertebral disk space (arrow).

Parenteral antibiotic therapy should be directed against epidural spinal abscess; however, clinical experience with these
the etiologic agents most likely to be involved: Staph. aureus, agents in this disease is limited. A nosocomial spinal epidural
coagulase-negative staphylococci, streptococci, and gram- abscess may be initially treated with meropenem or cefepime
negative rods. A combination of a third-generation cephalo- plus vancomycin. When the etiologic agent has been identi-
sporin (e.g., ceftriaxone) with another antimicrobial showing fied in cultures, the antibiotic regimen should be modified ac-
antistaphylococcal activity (e.g., nafcillin) is recommended (for cording to the antibiogram. Minimum treatment duration of
dosages, see Table 32.5). If methicillin-resistant staphylococci 4 to 6 weeks is usually recommended; this period should be
are suspected or isolated, vancomycin should be used. Linezolid extended to 6 to 8 weeks or more if osteomyelitis is present.
or daptomycin may become alternative antibiotic agents to The administration of corticosteroids (e.g., intravenous dexa-
cover methicillin-resistant staphylococci in the therapy of an methasone) is controversial. Their benefit has not been proven

FIGURE 32.10 Early epidural and paravertebral abscess. The 74-year-old patient presented with subacute
paraparesis and high-grade fever. A: T2-weighted MRI showed hyperintensities of the L3/L4, L4/L5, and L5/S1
intervertebral disc spaces (arrows) and the L3 and L4 vertebral bodies. B: T1-weighted images showed subtle
contrast enhancement of the L3 and L4 vertebral bodies (arrowheads) and (C) epidural material with contrast
enhancement (arrows) as well as contrast enhancement of the paravertebral muscles (asterisk). D: Material ob-
tained during surgery revealed few gram-positive cocci (Gram stain, arrowheads). Microbiologic investigations
identified Staphylococcus aureus, which was also found in blood cultures. (D, Courtesy of Prof. Dr. Armin
Giese, Center for Neuropathology and Prion Research, Ludwig Maximilians University of Munich.)

Scheld_Ch32.indd 558 2/21/14 8:29 PM


Chapter 32: Epidural Abscess 559

TA B L E 3 2 . 5 TA B L E 3 2 . 6
RECOMMENDED DOSAGES FOR ANTIMICROBIAL OUTCOME OF 244 PATIENTS WITH SPINAL EPIDURAL
THERAPY OF EPIDURAL ABSCESSES IN ADULTS ABSCESSES TREATED BETWEEN 1991 AND 2000
(DOSING MAY NEED ADJUSTMENT IN PATIENTS
WITH UNDERLYING RENAL OR LIVER DISEASE) Outcome No. of Patients (%)

Antimicrobial Agent Dosage Per Day Complete recovery 100 (41)


Minor neurologic deficits 68 (28)
Cefotaxime 34 g i.v. q8h
Paresis/paralysis 39 (16)
Ceftriaxone 2 g i.v. q12h
Death 37 (15)
Fosfomycin 5 g i.v. q8h
TOTAL 244 (100)
Linezolid 600 mg i.v. q12h
Meropenem 2 g i.v. q8h Data from references 1, 20.
Metronidazole 500 mg i.v. q68h
Nafcillin 2 g i.v. q4h
Rifampin 10 mg/kg (up to 600 mg)
i.v. q24h Prognosis
Vancomycin 15 mg/kg i.v. q812ha
The outcome of a spinal epidural abscess seems to be directly
a
Need to monitor serum concentrations and maintain at 15 to 20 g/mL. related to several independent variables (Table 32.6). When the
diagnosis is made early in the course of their illness and before
the development of significant neurologic deficit, patients fare
better than when a delay in diagnosis has occurred (1). Complete
and controlled studies do not exist. They may be beneficial in recovery with the return of full neurologic function is most likely
reducing local edema in patients with progressive neurologic to occur in patients without a neurologic deficit at the time of
compromise who are awaiting surgical decompression (7). diagnosis and treatment or when neurologic signs are present
Nonsurgical management using antibiotic therapy alone for less than 24 hours (3). All large case series of the literature
may be considered in selected patients without significant have shown that more severe preoperative neurologic deficits are
neurologic deficit, with complete paralysis for more than generally associated with a worse prognosis (1,19,20). If weak-
3 days, and in patients refusing surgery, with panspinal infec- ness or paralysis exists for more than 36 to 48 hours, complete
tion or in poor medical condition with a high surgical risk. recovery is less likely (1). In some series, patients with cervical or
Identification of the causative pathogen may be attempted by cervicothoracic epidural abscesses had more profound sequelae
CT-guided needle aspiration in conservatively treated patients than those with lumbar, lumbosacral, or sacral abscesses (16,74).
with negative blood cultures and with posteriorly located Other parameters such as older age, underlying disease, septice-
abscesses. Conservatively treated patients without signifi- mia, and higher degree of thecal sac compression were also asso-
cant neurologic deficit need close neurologic monitoring in a ciated with an adverse outcome in several studies (1,5,12,16,74).
hospital with facilities for emergency decompressive surgery.
Retrospective analyses suggest that the outcome is compa-
rable to that of surgically treated patients, if these guidelines INTRACRANIAL EPIDURAL
are followed (11,16,73,74). In the future, MRI criteria may
also be helpful when selecting patients for conservative treat-
ABSCESS
ment. A recent study (75) suggested that a good outcome was
associated with abscesses shorter than 3 cm cephalocaudally, Epidemiology
less than 50% narrowing of the sagittal diameter of the spi-
nal canal, and homogenous contrast enhancement (suggestive The true incidence of intracranial epidural abscesses is not
of phlegmonous or granulomatous tissue, opposed to the known. In one hospital-based series, it was the third most com-
peripheral enhancement of a frank abscess containing pus). mon localized intracranial infection, following brain abscesses
The authors, therefore, suggested that only patients fulfilling and subdural empyemas (77), and in a recent South African
at least two of these criteria should be treated conservatively. series of 82 patients, epidural abscess accounted for 1.8% of all
However, definite conclusions can be drawn only from con- intracranial infections (78). Intracranial epidural abscesses have
trolled studies, which are still lacking and are unlikely to be been reported in a wide age range, from 1 month to 71 years
performed. of age (7779). In contrast to recent studies from develop-
In patients with tuberculous spinal epidural abscess ing countries and earlier studies from the developed world in
(Fig. 32.5), antituberculous chemotherapy (usually with four which intracranial epidural abscesses were principally sequelae
agents pending susceptibility testing) has diminished the need of sinusitis, mastoiditis, and otitis media, recent case series
for surgical intervention except when stabilization of the spine from developed countries document that they are more likely
is required. Because bony changes may progress radiographi- to complicate neurosurgical procedures because up to 2% of
cally for an average of 4 months following the initiation of craniotomies may be complicated by this infection (78,80).
chemotherapy, it has been emphasized that clinical parameters
rather than radiographic changes should be followed to moni-
tor therapy during the early stages of treatment. However, if Pathogenesis and Pathophysiology
neurologic signs and symptoms develop, decompression of a
tuberculous spinal epidural abscess may be required to prevent Above the foramen magnum the dura is essentially the adher-
permanent neurologic deficits (76). (For the recommended ent inner lining of the skull. Here the epidural space represents
antituberculous chemotherapy, see Chapter 29.) only a potential space, which is created when it is violated by

Scheld_Ch32.indd 559 2/21/14 8:29 PM


560 Part III: Bacterial and Mycoplasmal Infections

FIGURE 32.11 Chronic intracranial epidural abscess. The 19-year-old patient had a history of acute sinusitis
followed by recurrent episodes of mild headache and intermittent fevers responsive to oral antibiotics over
a 4-year period. A: Noncontrast CT scan demonstrates left maxillary sinus inflammatory disease (asterisk).
B: Noncontrast CT section of the brain obtained at the same time as part A demonstrates a hypodense mass
(number sign) overlying the left frontal lobe associated with edema and mass effect. The hyperdense rim
(arrows) represents calcification, indicative of the long-standing nature of the inflammatory process. C: T1-
weighted MRI scan obtained after the intravenous administration of gadoliniumdiethylenetriamine penta-
acetic acid demonstrates a hypointense mass (number sign) with a thick enhancing rim (arrows). Left frontal
craniotomy revealed a purulent collection that was negative by Gram stain and culture.

an encroaching mass (tumor, adjacent infection, or hematoma) cranial fossa that facilitate communication between the epidural
or as a result of trauma. As a consequence, infections of the in- space and the paranasal sinuses; and, rarely, dental infection
tracranial epidural space primarily result from the extension of (78,84,85). The abscess expands as the pressure generated by
contiguous infections, in particular, sinusitis (Fig. 32.11) and the growing inflammatory mass dissects the dura away from the
otitis/mastoiditis (8184). Other common risk factors for intra- skull. As a result, an intracranial epidural abscess is a slowly
cranial epidural abscesses are recent transnasal, transmastoid, or growing mass, the property that accounts for its insidious clini-
intracranial surgical procedures (Figs. 32.12 and 32.13); post- cal presentation. Intracranial abscesses rarely dissect beyond the
traumatic infection; congenital osseous defects of the anterior base of the skull because there the dura is even more tightly fixed.

FIGURE 32.12 Intracranial epidural abscess. Epidural frontal


abscess in a 41-year-old patient 5 months after meningioma
operation. The patient suffered from headaches, fever, and
(A) a supraorbital swelling (arrow). B: Using computed tomog-
raphy (bone window), the swelling (arrow) could be allocated
in close proximity to the clips (arrowhead) that were inserted
to hold the reinserted bone after trepanation. C: Although
native CT only revealed the extracranial swelling, (D) extra-
(arrow) and also intracranial (black arrowhead) inflammation
were observed after contrast enhancement was administered.
Histologic assessment of material gained from surgery revealed
granulocytic inflammation; a causative pathogen could not be
identified. (A, Courtesy of Dr. Nicole Terpolilli, Department of
Neurosurgery, Ludwig Maximilians University of Munich.)

Scheld_Ch32.indd 560 2/21/14 8:29 PM


Chapter 32: Epidural Abscess 561

FIGURE 32.13 Intracranial epidural abscess.


The 61-year-old patient had a history of previ-
ous meningioma operation and postoperative
bleeding within the operation site. A: T1-
weighted imaging shows a space-occupying
lesion (asterisk) with surrounding contrast
enhancement (arrows) and adjacent posterior
edema (number sign). B: Diffusion-weighted
imaging was hyperintense at the margin of
the lesion (asterisk). Enterobacter aerogenes
was isolated from surgical material.

Depending on the location of the abscess, focal neurologic


Microbiology signs may develop as a result of continued expansion of the
inflammatory mass. However, in the aforementioned series of
The etiology of intracranial epidural abscesses is frequently 82 patients, focal neurologic signs were present in only 4.9%
polymicrobial with the most common organisms includ- of patients (78). Involvement of the apex of the petrous tem-
ing anaerobic gram-positive cocci, Staphylococcus species, poral bone and cranial nerves V and VI may lead to unilateral
Streptococcus species (in particular Streptococcus angino- facial pain and lateral rectus weakness (Gradenigo syndrome)
sus), and anaerobic gram-negative bacilli (84). Anaerobic (89). Although the gradually expanding intracranial abscess
bacteria are cultured from many sinus-associated intra- may remain localized, more often the dura cannot contain
cranial epidural abscesses. Streptococci and staphylococci the expanding epidural abscess. This may result from the
(including Staphylococcus epidermidis and methicillin-resistant development of thrombosis of the valveless emissary veins that
Staphylococcus aureus) are the leading pathogens in postop- run between the skull and meninges or it may result from direct
erative epidural abscess (78,84). penetration through the necrotic dura. When this occurs, the
Other organisms isolated from localized intracranial epi- intracranial epidural abscess may be complicated by subdu-
dural abscesses include Peptostreptococcus species, Salmonella ral empyema, brain abscess, or meningitis. The process may
species, Klebsiella species, Proteus species, E. coli, Providencia be further complicated by venous sinus thrombosis, which,
species, Citrobacter species, Serratia marcescens, Proteus though uncommon, is a serious complication (90). Because the
mirabilis, Haemophilus influenzae, Eikenella corrodens, uncomplicated epidural abscess grows slowly, it is often the
Bacteroides species, Propionibacterium acnes, Enterobacter striking manifestations of the complicationsmeningismus,
cloacae, Mycobacterium tuberculosis, Aspergillus fumigatus, seizures, changing mental status, or comathat may be the
Pseudallescheria boydii, and Rhizopus species (mucormycosis) first indication of an intracranial process.
(55,80,84,8688).

Clinical Manifestations Approach to Diagnosis


The slow-growing intracranial epidural abscess may cause few In contrast to spinal epidural abscess, in an intracranial
symptoms other than fever, localized skull tenderness, dull epidural abscess, a microbiologic etiology is established far
headache, nausea, vomiting, and lethargy (79). Papilledema more reliably from intraoperative cultures than from blood
may develop with increasing intracranial pressure. When cra- cultures (80). Diagnosis is made by contrast-enhanced CT
nial osteomyelitis is present, edema and cellulitis of the face and or MRI. CT can detect a low-attenuation extraaxial mass
scalp also may develop. Thus, although attention is focused on (84). Contrast CT in epidural abscesses reveals a thick me-
the primary process such as sinusitis, cellulitis, skull fracture, dial rim enhancement, which represents inflamed displaced
or a recent neurosurgical or otorhinologic procedure, a devel- dura (Fig. 32.12). The degree of rim enhancement is usually
oping intracranial abscess may remain undetected. A recent thicker and more irregular in an epidural abscess than in sub-
paper on 82 patients with cranial epidural abscess reported dural empyema. Calvarial osteomyelitis, subgaleal or sub-
that the prominent clinical features were fever (57%), frontal periosteal abscesses, and frontal sinus inflammatory disease
subgaleal abscess (Pott puffy tumor, 46%), periorbital edema are typically associated with intracranial epidural abscesses.
(40%), headache (37%), meningismus (35%), and seizures Large epidural abscesses can cause mass effect on the under-
(11%) (78). In 23 cases of postoperative epidural abscess, the lying brain (Fig. 32.14); however, in contradistinction to the
most common sign was wound infection (95.7%), followed clinically more significant subdural empyema, an intracranial
by encephalopathy (44.7%), fever (34.8%), and headache epidural abscess rarely demonstrates parenchymal abnor-
(17.4%) (80). Symptoms may be present for several weeks or malities on CT (Fig. 32.12C). As noted, it is important to
months before the diagnosis is made (78). appreciate that an intracranial epidural abscess often coexists

Scheld_Ch32.indd 561 2/21/14 8:29 PM


562 Part III: Bacterial and Mycoplasmal Infections

FIGURE 32.14 Intracranial epidural abscess.


Intracranial epidural abscess in a 55-year-
old patient 6 weeks after surgical interven-
tion necessary because of chronic subdural
hematoma. A: T1-weighted MRI showed a
contrast-enhancing (arrows) space-occupying
lesion (number sign) with (B) an increased
signal intensity on diffusion-weighted imag-
ing (number sign). C: Histology of surgical
material revealed mainly polymorphonuclear
granulocytes (arrows) and few lymphocytes
(arrowhead) (hematoxylin and eosin [H&E]
staining). D: On Gram stain, gram-positive
bacilli were detected that were later identified
as Propionibacterium species (arrow). E: In
Propionibacterium species cultures, Gram
stain typically reveals violet (gram-positive)
bacteria which can occur singularly (arrow-
head), in pairs (arrow), or in groups (dotted
arrow). (C, D, Courtesy of Prof. Dr. Armin
Giese, Center for Neuropathology and Prion
Research, Ludwig Maximilians University of
Munich; E, Courtesy of Dr. Andreas Wieser,
Max von Pettenkofer-Institute for Hygiene and
Microbiology, Ludwig Maximilians University
of Munich.)

with a subdural empyema, so presence of the former should convexity and/or in the interhemispheric fissure (Fig. 32.15).
prompt a careful search for the latter. MRI offers several advantages over CT in the evaluation of
Patients with postoperative and posttraumatic extraaxial patients with intracranial epidural abscesses. Specifically,
abscesses often present months to years following surgery or in otorhinologically induced abscesses, MRI can accurately
head trauma, respectively (80). These patients may have signs identify and delineate the collections (including small locu-
of local wound infection, but they generally show neither lations) early in the stage of disease, when the findings on
systemic signs of infection nor neurologic changes. Patients CT can be subtle (92). This ability of MRI is attributable to
with postoperative and posttraumatic abscesses usually have the inherent high degree of contrast between the purulent
underlying structural brain lesions that may lead to the persis- collections and the subjacent calvaria, brain, and CSF, com-
tence or the reaccumulation of these collections. Postoperative bined with the absence of streak artifacts from bone. The di-
abscesses occupy the cavity created by the craniotomy defect, agnosis of an intracranial epidural abscess can be confirmed
whereas posttraumatic abscesses are often an iatrogenic com- by diffusion-weighted imaging restriction (Figs. 32.13 and
plication of evacuation of a preexisting subdural or epidural 32.15) (84,93). In postoperative and posttraumatic abscesses,
hematoma. Both postoperative and posttraumatic abscesses MRI can readily differentiate these collections from sterile
are visualized as hypodense extraaxial collections with me- effusions and chronic extraaxial hematomas based on signal-
dial rim enhancement on CT. Edema and mass effect on intensity differences, a distinction that is usually subtle or
the underlying brain are usually minimal and parenchymal undetectable on CT. Intracranial abscesses are usually mildly
abnormalities are rare (91). The indolent clinical course and hyperintense to CSF on T1-weighted images and more mark-
the relatively benign radiographic findings are due to the pres- edly hyperintense to CSF on T2-weighted images (Figs. 32.5C
ence of a discrete limiting membrane from prior surgery or and 32.6). In contrast, sterile effusions are isointense to CSF
trauma, which serves to shield the underlying cerebral cortex. (92,94). Posttraumatic abscesses are hypointense on both T1-
Abscesses in these two clinical circumstances can be very dif- weighted and T2-weighted images in contrast to most chronic
ficult, if not impossible, to differentiate from noninfected ster- hematomas (94).
ile effusions or chronic extraaxial hematomas. A change in Lumbar punctures contribute little to the diagnosis of epi-
density of these collections on serial CT scans, associated with dural abscess, because the CSF findings are nonspecific, cul-
subtle mass effect on adjacent brain, may be the first indica- tures are usually negative, and there is a considerable risk of
tion of an intracranial infection. neurologic deterioration following lumbar puncture because
The morphology of posttraumatic intracranial epidural of transtentorial or foraminal herniation (78). A lumbar punc-
abscesses on MRI corresponds to that seen on CTthat ture should, therefore, not be performed in patients with sus-
is, lentiform or crescentic collections overlying a cerebral pected or proven epidural abscess.

Scheld_Ch32.indd 562 2/21/14 8:29 PM


Chapter 32: Epidural Abscess 563

FIGURE 32.15 Intracranial epidural abscess. The 59-year-old patient developed brain abscess 5 weeks
after surgery for meningioma. A: Transverse T1-weighted MRI showed a contrast-enhancing lesion
(asterisk) with (B) perifocal edema (number sign) on T2-weighted sequences. C: Diffusion-weighted
imaging revealed an increased signal intensity (asterisk).

the purulent material in their series of 82 patients using burr


Treatment holes (21 of 70 patients treated surgically) or limited craniecto-
mies (39 of 70 patients), because the extradural pus collections
Therapy consists of surgical drainage of the epidural abscess were usually liquid and never loculated.
(formal craniotomy or drainage through burr holes or an ex-
tended craniectomy), surgical therapy of underlying infections,
and medical therapy. The medical therapy is that of brain
abscess (see Chapter 31). In brief, empirical antibiotic ther- Prognosis
apy should usually include a third-generation cephalosporin
(e.g., ceftriaxone) and metronidazole in cases of community- The combination of medical and surgical management of
acquired epidural abscesses. Postoperative or posttraumatic uncomplicated intracranial epidural abscesses has generally
epidural cranial abscesses can be treated empirically with a resulted in favorable outcomes. In a recent series of postopera-
combination of vancomycin and meropenem. After culture tive infections, the 18% 5-year case-fatality rate was attrib-
results, the therapy should be modified according to the uted primarily to comorbidities rather than the intracranial
antibiogram. infectious process (80). In the aforementioned South African
The surgical therapy of an intracranial epidural abscess is series of 82 patients, 78 patients recovered completely, 3 had
primarily aimed at drainage of the collection to prevent further a moderate disability, and 1 died (mortality rate, 1.2%) (78).
accumulation and neurologic damage while obtaining material However, similar to spinal epidural abscesses, delays in diag-
for culture. Burr holes may suffice, but craniotomy or craniec- nosis may result in irreversible neurologic deficits, as prog-
tomy may be required, particularly if overlying bone is involved. nosis is inversely related to the degree of encephalopathy at
Nathoo et al. (78) reported that they could adequately drain initial presentation.

References
1. Reihsaus E, Waldbaur H, Seeling W. Spinal epidural abscess: a meta- 11. Grieve JP, Ashwood N, ONeill KS, et al. A retrospective study of surgi-
analysis of 915 patients. Neurosurg Rev. 2000;23:175204. cal and conservative treatment for spinal extradural abscess. Eur Spine J.
2. Riddell J IV, Shuman EK. Epidemiology of central nervous system infec- 2000;9:6771.
tion. Neuroimaging Clin N Am. 2012;22:543556. 12. Klekamp J, Samii M. Extradural infections of the spine. Spinal Cord.
3. Baker AS, Ojemann RG, Swartz MN, et al. Spinal epidural abscess. N Engl 1999;37:103109.
J Med. 1975;293:463468. 13. Strong WE. Epidural abscess associated with epidural catheterization:
4. Hlavin ML, Kaminski HJ, Ross JS, et al. Spinal epidural abscess: a ten-year a rare event? Report of two cases with markedly delayed presentation.
perspective. Neurosurgery. 1990;27:177184. Anesthesiology. 1991;74:943946.
5. Danner RL, Hartman BJ. Update on spinal epidural abscess: 35 cases and 14. Del Curling OJ, Gower DJ, McWhorter JM. Changing concepts in
review of the literature. Rev Infect Dis. 1987;9:265274. spinal epidural abscess: a report of 29 cases. Neurosurgery. 1990;27:
6. Nussbaum ES, Rigamonti D, Standiford H, et al. Spinal epidural abscess: a 185192.
report of 40 cases and review. Surg Neurol. 1992;38:225231. 15. Lange M, Tiecks F, Schielke E, et al. Diagnosis and results of different treat-
7. Darouiche RO. Spinal epidural abscess. N Engl J Med. 2006;355(19): ment regimens in patients with spinal abscesses. Acta Neurochir (Wien).
20122020. 1993;125:105114.
8. Batson OV. The function of vertebral veins and their role in the spread of 16. Tang H, Lin H, Liu Y, et al. Spinal epidural abscessexperience with
metastases. Ann Surg. 1940;112:138149. 46 patients and evaluation of prognostic factors. J Infect. 2002;45:76.
9. Gillilan LA. Veins of the spinal cord. Anatomic details; suggested clinical 17. Solomou E, Maragkos M, Kotsarini C, et al. Multiple spinal epidural
applications. Neurology. 1970;20:860868. abscesses extending to the whole spinal canal. Magn Reson Imaging.
10. Khan SH, Hussain MS, Griebel RW, et al. Comparison of primary and sec- 2004;22:747750.
ondary spinal epidural abscesses: a retrospective analysis of 29 cases. Surg 18. Pfister HW, von Rosen F, Yousry T. MRI detection of epidural spinal
Neurol. 2003;59:2833. abscesses at noncontiguous sites. J Neurol. 1996;243:315317.

Scheld_Ch32.indd 563 2/21/14 8:29 PM


564 Part III: Bacterial and Mycoplasmal Infections

19. Lu CH, Chang WN, Lui CC, et al. Adult spinal epidural abscess: clini- 50. Chia SL, Tan BH, Tan CT, et al. Candida spondylodiscitis and
cal features and prognostic factors. Clin Neurol Neurosurg. 2002;104: epidural abscess: management with shorter courses of anti-fungal
306310. therapy in combination with surgical debridement. J Infect. 2005;51:
20. Soehle M, Wallenfang T. Spinal epidural abscesses: clinical manifestations, 1723.
prognostic factors, and outcomes. Neurosurgery. 2002;51:7985. 51. Bariola JR, Perry P, Pappas PG, et al. Blastomycosis of the central nervous
21. Auletta JJ, John CC. Spinal epidural abscesses in children: a 15-year experi- system: a multicenter review of diagnosis and treatment in the modern era.
ence and review of the literature. Clin Infect Dis. 2001;32:916. Clin Infect Dis. 2010;50:797804.
22. Heusner AP. Nontuberculous spinal epidural infections. N Engl J Med. 52. Wrobel CJ, Chappell ET, Taylor W. Clinical presentation, radiological
1948;239:845854. findings, and treatment results of coccidioidomycosis involving the spine:
23. Scott DB, Hibbard BM. Serious non-fatal complications associated report on 23 cases. J Neurosurg. 2001;95:3339.
with extradural block in obstetric practice. Br J Anaesth. 1990;64: 53. Gupta SK, Chhabra R, Sharma BS, et al. Vertebral cryptococcosis simulat-
537541. ing tuberculosis. Br J Neurosurg. 2003;17:556559.
24. Kindler C, Seeberger M, Siegemund M, et al. Extradural abscess complicat- 54. Buruma OJ, Craane H, Kunst MW. Vertebral osteomyelitis and epidural
ing lumbar extradural anaesthesia and analgesia in an obstetric patient. abscess due to mucormycosis, a case report. Clin Neurol Neurosurg.
Acta Anaesthesiol Scand. 1996;40:858861. 1979;81:3944.
25. Wang LP, Hauerberg J, Schmidt JF. Incidence of spinal epidural abscess 55. Berenguer J, Diaz-Mediavilla J, Urra D, et al. Central nervous system
after epidural analgesia: a national 1-year survey. Anesthesiology. 1999;91: infection caused by Pseudallescheria boydii: case report and review. Rev
19281936. Infect Dis. 1989;11:890896.
26. Du Pen SL, Peterson DG, Williams A, et al. Infection during chronic epi- 56. Kotil K, Tatar Z, Bilge T. Spinal hydatidosis accompanied by a secondary
dural catheterization: diagnosis and treatment. Anesthesiology. 1990;73: infection. Case report. J Neurosurg Spine. 2007;6:585590.
905909. 57. Mathur PP, Dharker SR, Hiran S, et al. Lumbar extradural compression by
27. Sillevis SP, Tsafka A, van den Bent M, et al. Spinal epidural abscess compli- guinea worm infestation. Surg Neurol. 1982;17:127129.
cating chronic epidural analgesia in 11 cancer patients: clinical findings and 58. Ruberti RF, Saio M. Epidural Bilharzioma mansoni compressing the spinal
magnetic resonance imaging. J Neurol. 1999;246:815820. cord: case report. East Afr Med J. 1999;76:414416.
28. Burstal R, Wegener F, Hayes C, et al. Epidural analgesia: prospective audit 59. Darouiche RO, Hamill RJ, Greenberg SB, et al. Bacterial spinal epidural
of 1062 patients. Anaesth Intensive Care. 1998;26:165172. abscess: review of 43 cases and literature survey. Medicine (Baltimore).
29. Kovalik EC, Raymond JR, Albers FJ, et al. A clustering of epidural abscesses 1992;71:369385.
in chronic hemodialysis patients: risks of salvaging access catheters in cases 60. Rankin RM, Flothow PG. Pyogenic infection of the spinal epidural space.
of infection. J Am Soc Nephrol. 1996;7:22642267. West J Surg Obstet Gynecol. 1946;54:320323.
30. Obrador GT, Levenson DJ. Spinal epidural abscess in hemodialysis 61. Brown EM. Infections in neurosurgery: using laboratory data to plan opti-
patients: report of three cases and review of the literature. Am J Kidney mal treatment strategies. Drugs. 2002;62:909913.
Dis. 1996;27:7583. 62. Maslen DR, Jones SR, Crislip MA, et al. Spinal epidural abscess. Optimizing
31. Korzets A, Weinstein T, Ori Y, et al. Back pain and staphylococcal bac- patient care. Arch Intern Med. 1993;153:17131721.
teraemia in haemodialysed patientsbeware! Nephrol Dial Transplant. 63. Redekop GJ, Del Maestro RF. Diagnosis and management of spinal epi-
1999;14:483486. dural abscess. Can J Neurol Sci. 1992;19:180187.
32. Feldenzer JA, McKeever PE, Schaberg DR, et al. The pathogenesis of spi- 64. Kastenbauer S, Winkler F, Fesl G, et al. Acute severe spinal cord dysfunc-
nal epidural abscess: microangiographic studies in an experimental model. tion in bacterial meningitis in adults: MRI findings suggest extensive myeli-
J Neurosurg. 1988;69:110114. tis. Arch Neurol. 2001;58:806810.
33. Feldenzer JA, McKeever PE, Schaberg DR, et al. Experimental spinal epi- 65. Ruiz A, Post MJ, Sklar EM, et al. MR imaging of infections of the cervical
dural abscess: a pathophysiological model in the rabbit. Neurosurgery. spine. Magn Reson Imaging Clin N Am. 2000;8:561580.
1987;20:859867. 66. Stabler A, Reiser MF. Imaging of spinal infection. Radiol Clin North Am.
34. Rigamonti D, Liem L, Sampath P, et al. Spinal epidural abscess: contem- 2001;39:115135.
porary trends in etiology, evaluation, and management. Surg Neurol. 67. Tyrrell PN, Cassar-Pullicino VN, McCall IW. Spinal infection. Eur Radiol.
1999;52:189196. 1999;9:10661077.
35. Huang PY, Chen SF, Chang WN, et al. Spinal epidural abscess in adults 68. Varma R, Lander P, Assaf A. Imaging of pyogenic infectious spondylodiski-
caused by Staphylococcus aureus: clinical characteristics and prognostic tis. Radiol Clin North Am. 2001;39:203213.
factors. Clin Neurol Neurosurg. 2012;114:572576. 69. Dagirmanjian A, Schils J, McHenry MC. MR imaging of spinal infections.
36. Bostrm A, Oertel M, Ryang Y, et al. Treatment strategies and outcome Magn Reson Imaging Clin N Am. 1999;7:525538.
in patients with non-tuberculous spinal epidural abscessa review of 70. Diehn FE. Imaging of spine infection. Radiol Clin North Am. 2012;50:
46 cases. Minim Invasive Neurosurg. 2008;51:3642. 777798.
37. Yung BC, Cheng JC, Chan TT, et al. Aggressive thoracic actinomycosis 71. Eastwood JD, Vollmer RT, Provenzale JM. Diffusion-weighted imaging in
complicated by vertebral osteomyelitis and epidural abscess leading to spi- a patient with vertebral and epidural abscesses. AJNR Am J Neuroradiol.
nal cord compression. Spine. 2000;25:745748. 2002;23:496498.
38. Solera J, Lozano E, Martinez-Alfaro E, et al. Brucellar spondylitis: review 72. Lyu RK, Chen CJ, Tang LM, et al. Spinal epidural abscess successfully
of 35 cases and literature survey. Clin Infect Dis. 1999;29:14401449. treated with percutaneous, computed tomography-guided, needle aspira-
39. Nas K, Gur A, Kemaloglu MS, et al. Management of spinal brucellosis and tion and parenteral antibiotic therapy: case report and review of the litera-
outcome of rehabilitation. Spinal Cord. 2001;39:223227. ture. Neurosurgery. 2002;51:509512.
40. Ekici MA, Ozbek Z, Gkoglu A, et al. Surgical management of cervi- 73. Wheeler D, Keiser P, Rigamonti D, et al. Medical management of spi-
cal spinal epidural abscess caused by Brucella melitensis: report of two nal epidural abscesses: case report and review. Clin Infect Dis. 1992;15:
cases and review of the literature. J Korean Neurosurg Soc. 2012;51: 2227.
383387. 74. Khanna RK, Malik GM, Rock JP, et al. Spinal epidural abscess: evaluation
41. Khan KM, Pao W, Kendler J. Epidural abscess and vertebral osteomyelitis of factors influencing outcome. Neurosurgery. 1996;39:958964.
caused by Listeria monocytogenes: case report and literature review. Scand 75. Tung GA, Yim JW, Mermel LA, et al. Spinal epidural abscess: correla-
J Infect Dis. 2001;33:714716. tion between MRI findings and outcome. Neuroradiology. 1999;41:
42. Graat HC, Van Ooij A, Day GA, et al. Nocardia farcinica spinal osteomy- 904909.
elitis. Spine. 2002;27:E253E257. 76. Jain AK. Treatment of tuberculosis of the spine with neurologic complica-
43. Hung CC, Hsueh PR, Chen YC, et al. Haemophilus aphrophilus bacterae- tions. Clin Orthop. 2002;398:7584.
mia complicated with vertebral osteomyelitis and spinal epidural abscess in 77. Harris LF, Haws FP, Triplett JN Jr, et al. Subdural empyema and epidural
a patient with liver cirrhosis. J Infect. 1997;35:304308. abscess: recent experience in a community hospital. South Med J. 1987;80:
44. Samuel W, Dryden M, Sampson M, et al. Spinal abscess of Haemophilus 12541258.
paraphrophilus: a case report. Spine. 1997;22:27632765. 78. Nathoo N, Nadvi SS, Van DJ. Cranial extradural empyema in the era
45. Turner DPJ, Weston VC, Ispahani P. Streptococcus pneumoniae spinal of computed tomography: a review of 82 cases. Neurosurgery. 1999;44:
infection in Nottingham, United Kingdom: not a rare event. Clin Infect 748753.
Dis. 1999;28:873881. 79. Smith HP, Hendrick EB. Subdural empyema and epidural abscess in chil-
46. West KR, Mason RC, Sun M. Nocardia spinal epidural abscess: 14-year dren. J Neurosurg. 1983;58:392397.
follow-up. Orthopedics. 2012;35:128131. 80. Hlavin ML, Kaminski HJ, Fenstermaker RA, et al. Intracranial suppura-
47. Akalan N, Ozgen T. Infection as a cause of spinal cord compression: a review tion: a modern decade of postoperative subdural empyema and epidural
of 36 spinal epidural abscess cases. Acta Neurochir (Wien). 2000;142: abscess. Neurosurgery. 1994;34:974980.
1723. 81. Hicks CW, Weber JG, Reid JR, et al. Identifying and managing intracranial
48. Pettit A, Kopski J, Castilho J, et al. The index case for the fungal meningitis complications of sinusitis in children: a retrospective series. Pediatr Infect
outbreak in the United States. N Engl J Med. 2012;367:21192125. Dis J. 2011;30:222226.
49. Kainer MA, Reagan DR, Nguyen DB, et al. Fungal infections associ- 82. Mallur PS, Harirchian S, Lalwani AK. Preoperative and postoperative
ated with contaminated methylprednisolone in Tennessee. N Engl J Med. intracranial complications of acute mastoiditis. Ann Otol Rhinol Laryngol.
2012;367:21942203. 2009;118:118123.

Scheld_Ch32.indd 564 2/21/14 8:29 PM


Chapter 32: Epidural Abscess 565

83. Luntz M, Bartal K, Brodsky A, et al. Acute mastoiditis: the role of imag- 88. Shahat AH, Rahman NU, Obaideen AM, et al. Cranial-epidural tuberculo-
ing for identifying intracranial complications. Laryngoscope. 2012;122: sis presenting as a scalp swelling. Surg Neurol. 2004;61:464466.
28132817. 89. Lott T, el Gammal T, Dasilva R, et al. Evaluation of brain and epidural
84. Pradilla G, Ardila GP, Hsu W, et al. Epidural abscesses of the CNS. Lancet abscesses by computed tomography. Radiology. 1977;122:371376.
Neurol. 2009;8:292300. 90. Silverberg AL, DiNubile MJ. Subdural empyema and cranial epidural
85. Kanu OO, Ukponmwan E, Bankole O, et al. Intracranial epidural abscess abscess. Med Clin North Am. 1985;69:361374.
of odontogenic origin. J Neurosurg Pediatr. 2011;7:311315. 91. Zimmerman RD, Leeds NE, Danziger A. Subdural empyema: CT findings.
86. Huang CR, Lu CH, Chien CC, et al. Protean infectious types and frequent Radiology. 1984;150:417422.
association with neurosurgical procedures in adult Serratia marcescens 92. Weingarten K, Zimmerman RD, Becker RD, et al. Subdural and epidural
CNS infections: report of two cases and review of the literature. Clin empyemas: MR imaging. AJR Am J Roentgenol. 1989;152:615621.
Neurol Neurosurg. 2001;103:171174. 93. Tsuchiya K, Osawa A, Katase S, et al. Diffusion-weighted MRI of subdural
87. Diebold P, Humbert J, Djientcheu Vde P, et al. Salmonella epidural abscess and epidural empyemas. Neuroradiology. 2003;45:220223.
in sickle cell disease: failure of the nonsurgical treatment. J Natl Med Assoc. 94. Campbell BG, Zimmerman RD. Emergency magnetic resonance of the
2003;95:10951098. brain. Top Magn Reson Imaging. 1998;9:208227.

Scheld_Ch32.indd 565 2/21/14 8:29 PM


CHAPTER 33 SUBDURAL EMPYEMA AND
SUPPURATIVE INTRACRANIAL PHLEBITIS
BARRY J. HARTMAN AND DAVID C. HELFGOTT

diagnosis, and newer surgical techniques have combined to lower


CRANIAL SUBDURAL EMPYEMA the mortality rate to 10% to 40% (1,4,5,715,21,22,24,28,29).
Physicians depended on their clinical skills and plain roentgeno-
Empyema is an important form of intracranial suppura- grams of the sinuses and skull to direct their attention to the
tion, accounting for 15% to 25% of pyogenic intracranial possibility of intracranial suppuration, until the development
infections (13,97). It represents an infectious process that of cerebral angiography in the early 1960s, which proved an
occupies the space between the dura mater and arachnoid extremely sensitive method of detecting a subdural collection
surrounding the brain. Left undiagnosed and untreated, sub- (13,34). The emergence of CT in the 1970s provided a noninva-
dural empyema is rapidly fatal, so early recognition is criti- sive rapid means of visualizing the cranial contents; its reliability,
cal. In older children and adults, subdural empyema is most safety, and ease of operation made it the first choice for diagnosis
often a complication of otorhinologic infection (1,419,98). of suspected subdural empyema (2,10,26,35). Recently, MRI has
Subdural empyema may also occur as a result of head trauma proven even more sensitive than CT (36).
or surgery, osteomyelitis of the skull, or bacteremic spread
from a distant focus of infection. In infants, leptomeningi-
tis is the most common predisposing cause of subdural em-
pyema (11,20,21,97). Males predominate over females, and Pathogenesis and Anatomic Considerations
about 70% of patients are in their second or third decade of
life (35,710,12,13,15,2226). Patients will present with The clinical features of subdural empyema are easily understood
fever and headache in more than 90% of cases and many if one considers the anatomy of the subdural space with respect
will have associated neurologic abnormalities (1,39,12,13, to its surrounding structures (Fig. 33.1). The subdural space is
1519,21,23,24,2630,99,100,101). The diagnosis of subdu- normally a potential space rather than an actual space because
ral empyema is confirmed using computed tomography (CT) the dura mater follows the contours of the skull and lies adjacent
or magnetic resonance imaging (MRI) scanning techniques. to the arachnoid and pia mater (37). The ability to form an actual
However, a strong clinical suspicion despite the lack of evidence space with fluid collection is greater around the convexities of the
for subdural empyema on scans warrants more invasive inves- cerebral hemispheres where the brain does not approximate the
tigation. Definitive therapy consists of surgical drainage and skull as closely as it does around the basal areas (15,22,33).
systemic antibiotics, yet mortality remains as high as 40% in Posteriorly, the tentorium cerebelli is a reflection of dura
some series (14,21). Spinal subdural empyema also has been separating the cerebellum from the cerebral cortex (38). It is an
described; however, it is quite rare, with fewer than 150 cases effective barrier to the passage of subdural collection infraten-
reported in the literature (31,102). This condition is addressed torially, except at its free anterior margin where fluid may seep
briefly later in the chapter. Suppurative intracranial phlebitis into the subdural space of the posterior fossa. Only about
is a serious complication of cranial and facial infections and 10% of subdural empyemas are infratentorial (15,39,40).
results in thrombosis of the major dural venous sinuses. This is Medially, the falx cerebri is a reflection of dura extending the
discussed at the end of this chapter. length of the cerebrum that separates the cerebral hemispheres
(38). Subdural fluid that accumulates between the falx and
the arachnoid are known as parasagittal, interhemispheric,
Historical Perspective or parafalcine subdural empyemas and are usually second-
ary to surface subdural collections but rarely may be primary
The first comprehensive clinicopathologic descriptions of subdu- (4143). A subdural empyema may communicate with the
ral empyema as a distinct entity were published in the 1940s, contralateral side via the inferior free margin of the falx.
although the first definitive report of subdural empyema dates It therefore follows that infections or trauma of the head are
back to 1861 (32,33). This initial report was followed by sev- the usual causes of subdural empyema. Table 33.1 reviews the
eral more around the turn of the century, with a compilation of conditions predisposing to subdural empyema in series reported
44 cases by Blegvad (32) in 1910. Early names for this disease in- during the past two decades (excluding series of only infants).
cluded pachymeningitis interna, purulent pachymeningitis, Paranasal sinusitis overwhelmingly predominates as the precipi-
pia-arachnoid abscess, phlegmonous meningitis, and sub- tating factor for the development of subdural empyema (101).
dural abscess, but these were rejected by Kubik and Adams in The sinusitis almost always involves a frontal sinus, often with
favor of subdural empyema (8,32). Interestingly, early publica- other sinuses affected as well. The incidence of subdural empy-
tions reported a preponderance of subdural empyema secondary ema following frontal sinusitis is 1% to 2% (45). The frontal
to otogenic infections (32). However, because of the compila- and sphenoid sinuses are intimately associated with the dura
tion of 42 confirmed cases resulting from frontal sinusitis by mater, separated from the dura only by a thin plate of bone.
Courville (33), it has become clear that paranasal sinusitis is the These sinuses communicate with the maxillary and ethmoid
most important causative factor in the development of subdural sinuses, which are more anteriorly placed. Because of its posi-
empyema in older children and adults. During the past 50 years, tion, the frontal sinus is almost always involved in paranasal
much has changed in the areas of therapy and diagnosis of sub- sinus infection that spreads to the subdural space. In addition,
dural empyema. Before the development of antibiotics, subdural the growing posterior wall of the frontal sinus during puberty
empyema was almost always fatal (32,33). Antibiotics, improved has been offered as a possible explanation for the striking

566

Scheld_Ch33.indd 566 2/21/14 5:47 PM


Chapter 33: Subdural Empyema and Suppurative Intracranial Phlebitis 567

Subarachnoid Superior
space sagittal sinus Arachnoid granulations
Calvaria Diploic vein
Epidural space (potential)
Skin
Dura mater (gray)
Galea aponeurotica
Subdural space
Pericardium Arachnoid
Superior cerebral vein mater (purple)
Cerebral artery Pia mater
(yellow)
Cerebral hemisphere
Falx cerebri

FIGURE 33.1 Diagram of the cranial cavity showing the location of the subdural and epidural spaces.
(From Moore KL, Agur AMR, Dalley AF. Clinically Oriented Anatomy. 7th ed. Baltimore: Lippincott
Williams & Wilkins; 2014.)

age susceptibility for the development of subdural empyema the sinus to the emissary veins that link the facial and dural ve-
(7,26,46). nous systems (15,33,48). From the dural sinuses, the infection
Two modes of extension have been proposed for the spread establishes itself in the subdural space at the frontal pole and
of infection from a frontal sinus to the subdural space: direct and may spread posteriorly over the convexity, medially into the in-
indirect (33). Direct extension involves erosion of the posterior terhemispheric region, and contralaterally. This extension may
bony wall of the frontal sinus by infection, with further erosion create significant pressure on a large area of underlying brain
of the underlying dura mater (33). In teenagers primarily, fron- tissue (15,22,49). As stated previously, it is unusual but possible
tal bone osteomyelitis with subperiosteal abscesses (a Pott puffy for the empyema to spread infratentorially (15). Further retro-
tumor) can extend directly into the subdural space (47,103). grade thrombophlebitis often occurs, involving the valveless,
The more likely route is indirect, with extension of infection deeper veins of the cerebrum, which in turn may lead to necrosis
and associated thrombophlebitis through the mucosal veins of and infection of brain tissue (10,13,32,33). Subdural empyema

TA B L E 3 3 . 1
PREDISPOSING CAUSES OF SUBDURAL EMPYEMA

Reference No. No. of Patients Sinusitis Otogenic Head Trauma/Surgery Hematogenous Othera

Bradley and Shaw (1) 47 55% 13% 4% 4% 23%b


Mauser et al. (4) 37 59 12 15 0 15
Miller et al. (5) 23 35 30 26 4 4
Weisberg (6) 8 88 0 0 0 12
Hodges et al. (7) 4 100 0 0 0 0
Khan and Griebel (8) 15 40 7 27 13 13
Hockley and Williams (9) 42 74 12 0 2 12
Zimmerman et al. (10) 49 41 2 31 2 23
Smith and Hendrick (11) 25 32 12 28 4 24b
Bannister et al. (12) 66 68 21 3 3 5
Kaufman et al. (13) 17 41 6 24 6 24b
Le Beau et al. (14) 37 43 0 16 0 41b
Bhandari and Sarkari (15) 37 54 32 5 3 5
McIntyre et al. (16) 14 57 21 14 0 7b
Hoyt and Fisher (17) 17 53 12 0 0 35b
Dill et al. (18) 32 56 0 28 3 13b
Nathoo et al. (44) 699 67 9 8 0 16b
Cole et al. (100) 25 52 0 12 26b
total 1192 62 10 11 1 16
range 32100 032 031 013 041

a
Includes facial infections, local osteomyelitis, unknown.
b
Includes infants with meningitis.

Scheld_Ch33.indd 567 2/21/14 5:47 PM


568 Part III: Bacterial and Mycoplasmal Infections

secondary to an otogenic source of infection differs only in the 50


site at which pus enters the subdural space. The tympanic cavity
is bounded superiorly by the tegmen tympani, a thin plate of

Percent of Patients
40
bone forming part of the temporal bone of the skull separating
the tympanic cavity from the brain (37). Perforating veins pass
through this plate of bone to communicate with the superior 30
petrosal venous sinus of the dura (37). In addition, mastoid air
cells within the temporal bone surrounding the middle ear com-
municate with the tympanic cavity and may lie very close to 20
the posterior cranial fossa separated from the dura by slivers of
bone (37). Otitis or mastoiditis may, therefore, extend directly 10
into the subdural space via erosion of the tegmen tympani or
bone adjacent to the air cells and dura mater or spread infection
indirectly by way of a progressive thrombophlebitis of the per- 0
1 2 3 4 5 6 6
forating veins (27,32). Because the venous sinuses into which
the veins from the middle ear and mastoid bone drain are within Decade of Life
or beneath the tentorium (38), otogenic infection may result in FIGURE 33.2 Age distribution of patients reported with subdural
posterior fossa subdural empyema (27). As opposed to subdural empyema.
empyema secondary to frontal sinusitis, otitis-induced subdural
empyema is initially localized posteriorly or on the tentorium
(50). Hematogenous spread of bacteria to the subdural space
Figure 33.2 displays the age distribution of patients reported in
from a distant site of infection is an uncommon cause of subdu-
series of consecutive patients with subdural empyema. It is clear
ral empyema, accounting for fewer than 5% of all cases in most
that most cases occur during the second and third decades of
series (Table 33.1). Several reports describe the development
life. As stated earlier, the significant growth of the frontal sinus
of subdural empyema via hematogenous spread to a preexist-
during puberty has been proposed as an explanation for the un-
ing subdural hematoma (51,52). Subdural empyema is a rare
even age distribution (7,26,46). However, confirmatory analyses
complication following cranial surgery; in one series, subdural
comparing patients sex, age, and source of infection have not
empyema occurred in slightly more than 1 in 1,000 cranioto-
been reported.
mies (53). In a large series of 16,540 craniotomy procedures
The clinical features of adults with subdural empyema are
from 1997 to 2007, only 0.5% had intracranial infection (only
presented in Table 33.2. Generally, patients have a nonspecific
7 cases were pure subdural infections) (104).
illness for a few days to a few weeks before presentation to the
In adults, the extension of a subdural empyema from acute
hospital acutely ill (1,5,12,13). However, if the infection is a re-
purulent meningitis is very unusual. Although there is a sub-
sult of head trauma or surgery, the symptoms may be milder and
arachnoid inflammatory exudate, the arachnoid is fairly im-
present more subacutely (10,23,55,56,104). The most common
permeable to the bacterial process occurring adjacent to it
symptoms and signs are headache, fever, neurologic deficit, and
(22). Bacterial meningitis in adults is a very unusual cause
stiff neck. Vomiting and malaise are often reported as well (8,15).
of subdural empyema, occurring in less than 1% of cases
Seizures, papilledema, and altered level of consciousness ranging
(21,105). However, in infants, meningitis is an important pre-
from drowsiness and disorientation to coma also occur frequently.
disposing condition for the development of a subdural empy-
These neurologic changes may be presenting signs or, as is often
ema (11,20,21,97,100). Subdural empyema occurs in about
the case, may develop during the course of the illness (6,7,15,28).
2% of infants with bacterial meningitis (54). The pathogenesis
Diffuse neurologic signs such as altered level of conscious-
is presumably infection of an initially sterile subdural effu-
ness, papilledema, and generalized seizures are a result of
sion (13,20,21). Such sterile effusions are variably reported as
increased intracranial pressure (ICP) (33,57). Focal neurologic
occurring in up to 60% of infants with meningitis (13,20,97).
abnormalities such as hemiparesis, jacksonian seizures, dys-
phasia, and cranial neuropathies may be secondary to local
Clinical Features pressure on the underlying cortex by the subdural process
(21,27,32,33,41,49,58) and may be precipitated by cortical
A high clinical suspicion and rapid diagnosis of subdural venous thrombosis with accompanying brain inflammation
empyema are critical for a successful outcome. Certainly, an adult and infarction (21,22,57). Such focal neurologic signs may
with a recent history of sinusitis and a new presentation sugges- help to localize the empyema. This is particularly true in cases
tive of central nervous system (CNS) infection warrants an inves- of infratentorial subdural empyema that occurs infrequently,
tigation to exclude subdural empyema. However, in some cases, but that is easily suspected if cerebellar signs such as ataxia and
the antecedent infection is subtle enough to be unrecognized. In nystagmus are present (27). Interhemispheric (parasagittal,
others, the concurrent complication of sinusitis with a subdural parafalcine) subdural empyema, usually associated with dis-
empyema delays the diagnosis of the latter because symptoms are ease over the convexities but uncommonly occurring alone
attributed to the sinusitis. In other cases, the subdural empyema (43), characteristically produces contralateral leg symptoms,
is not suspected because the precipitating cause for the subdural including weakness and focal seizures (41,43,49,58). As the
empyema is unknown or arises from a distant focus of infection. interhemispheric suppuration extends backward over the ten-
Although the clinical presentation may vary, there are key clinical torium and below the occipital lobes, homonymous hemiano-
features of subdural empyema that if present should result in its pia may result (49,58). Subdural empyema overlying one or
inclusion in ones initial differential diagnosis. The sex and age dis- both convexities yields the most nonspecific neurologic signs.
tributions of patients with subdural empyema are striking. There Clues to the involved areas can be (a) contralateral paresis or
is an overrepresentation of men reported in series of patients seizures, (b) aphasia or dysphasia associated with left-sided
(mostly adults) with subdural empyema published during the last infection, or (c) cranial neuropathies (32,33,49). The clinical
two decades. In those that report only children, males also pre- signs of subdural empyema in infants are similar to those in
dominate. However, in infants, this sex discrepancy may not be adults. In addition, a bulging anterior fontanelle is a common
so marked (35,710,12,13,15,16,18,19,2226,54,99,100,106). finding in infants (11,20,54).

Scheld_Ch33.indd 568 2/21/14 5:47 PM


Chapter 33: Subdural Empyema and Suppurative Intracranial Phlebitis 569

TA B L E 3 3 . 2
CLINICAL SIGNS OF SUBDURAL EMPYEMA

Signs/Symptoms % No. of Patientsa References

Fever 78 451 3, 4, 69, 13, 1519, 21, 23, 26, 27, 30, 99
Headache 74 467 39, 13, 1519, 21, 23, 24, 26, 27, 30
Hemiparesis 71 389 35, 79, 13, 17, 18, 21, 23, 24, 26, 28, 30
Altered consciousness 69 544 1, 39, 12, 13, 15, 1719, 21, 23, 27, 30
Nuchal rigidity 60 416 4, 68, 13, 1519, 21, 23, 24, 26, 27, 29, 30
Seizures 49 607 49, 13, 1519, 21, 23, 24, 26, 28, 29, 30,99
Papilledema 33 238 69, 13, 15, 17, 18, 21, 24, 26, 27, 30
Altered speech 22 364 36, 8, 9, 15, 18, 24, 26, 28
b
Other focal deficits 46 296 59, 13, 15, 18, 24, 26, 28, 30, 99

a
Total number of patients in whom finding was assessed.
b
Includes cranial neuropathies, hemianopia, other.

characterization of these extraaxial (extraparenchymal) inflam-


Differential Diagnosis matory lesions and their associated intraaxial (parenchymal)
sequelae when compared to carotid arteriography (10). In ad-
The cardinal features of headache, fever, stiff neck, and neuro- dition, CT provides an important adjunct to standard clinical
logic signs are not specific for subdural empyema. The differ- parameters in the assessment of the adequacy of patient re-
ential diagnosis also includes brain abscess, epidural abscess, sponse to therapy. The CT, findings during the early stages of
meningitis, meningoencephalitis, subdural hematoma, and in- development of a subdural empyema may be subtle and easily
tracerebral thrombophlebitis (21,48). Of these, the presence of overlooked (10). Noncontrast CT scans typically demonstrate a
focal neurologic signs makes meningitis much less likely. The crescentic hypodense collection over one or both cerebral con-
presence of nuchal rigidity is unusual in brain abscess and sub- vexities and/or around the interhemispheric tissue (Fig. 33.3A).
dural hematoma. Unfortunately, clinical grounds alone do not Contrast-enhanced CT increases the conspicuity of the collec-
allow the exclusion of most of these possibilities. Therefore, tions that represent active inflammatory disease either in the
more specific testing should be undertaken as soon as the diag- leptomeninges or in the subjacent cerebral cortex (Fig. 33.3B).
nosis of subdural empyema is suspected. Thick, irregular enhancement of the falx in association with
a spindle-shaped collection is seen in interhemispheric subdu-
ral empyemas (43) (Fig. 33.4). Parenchymal changes at this
Diagnostic Studies early stage include thickening and hyperdensity of the underly-
Routine studies such as blood tests and plain roentgenograms ing cortical gray matter and hypodensity of the white matter
are of very little value in patients with suspected subdural em- on noncontrast CT images; these changes indicate the presence
pyema. Most patients are found to have a peripheral blood of edema, hyperemia, and ischemia (10). Additionally, gyral
leukocytosis (3,11,13,27,30,33). Plain films of the skull are enhancement subjacent to an extraaxial empyema on con-
not useful except to demonstrate a sinusitis or mastoiditis or trast CT scans is a common finding, indicative of meningitis,
to show widened sutures in infants (21). In infants, cranial cerebritis, and/or venous thrombosis (Fig. 33.3B). Extensive
ultrasonography can detect a subdural collection and may mass effect on the ipsilateral cerebral hemisphere that is out
differentiate a reactive effusion from a subdural empyema of proportion to the small size of the extraaxial collection is
(107). Before the development of CT, cerebral arteriography, invariably present, and it is manifested as ventricular compres-
with a diagnostic accuracy of 80% to 90%, was the proce- sion, sulcal effacement, and midline shift. It is important to
dure of choice to diagnose subdural empyema (13,15,21,27). examine the paranasal sinuses, middle ear cavity, and orbits
Although nearly perfect for the detection of hemispheric and for the presence of inflammation, which may reflect the origin
parafalcine subdural collections, the sensitivity of carotid and extent of the intracranial abnormalities (26,29) (Figs. 33.5
angiograms for posterior fossa subdural empyema was not and 33.6A).
as great (27). Presently, the safety, ease of application, and Unrecognized and untreated, the subdural empyema rap-
reliability of CT and MRI make them the modalities of choice idly grows and develops loculations and the parenchymal
to diagnose subdural empyema. abnormalities progress to cortical infarction and abscess
formation. MRI is proving to have a greater sensitivity and
Computed Tomography and Magnetic specificity in the workup of patients with an extraaxial
empyema; this is attributed to several inherent advantages of
Resonance Imaging MRI over CT (36,59,60). MRI uses several standard pulse
The radiologic evaluation of patients with subdural and epi- sequences referred to as T1 weighted, proton-density
dural empyemas has been revolutionized by the advent of CT weighted, and T2 weighted (61). T1-weighted images
in 1972 and MRI in 1984. The introduction of CT has had a emphasize contrast between the brain and cerebrospinal fluid
major impact on the management and prognosis of subdural (CSF), and proton-densityweighted and T2-weighted images
and epidural empyemas because CT allows (in a noninvasive emphasize contrast between brain and pathologic processes
manner) earlier and more accurate detection, delineation, and (61) (Fig. 33.6B and C).

Scheld_Ch33.indd 569 2/21/14 5:47 PM


570 Part III: Bacterial and Mycoplasmal Infections

A B

FIGURE 33.3 Subdural and epidural empyemas due to


paranasal sinus inflammatory disease. A: Noncontrast
computed tomographic (CT) scan of the brain demonstrat-
ing right hemispheric edema with compression of the right
lateral ventricle. No definite empyema is seen. B: Contrast-
enhanced CT scan demonstrating a right convexity sub-
dural empyema with a thin rim of marginal enhancement
(large arrows) and gyral enhancement (small arrows).
Note the disproportionate degree of mass effect on the
underlying brain with ventricular compression when com-
pared to the size of the empyema.

MRI has been found to have six inherent advantages over occur secondary to a subdural empyema. Fifth, MRI is more
CT. First, MRI permits excellent visualization of superficial specific than CT in differentiating a subdural from an epidural
brain anatomy, precise localization of extraaxial empyemas empyema (36,63). A hypodense medial rim, representing in-
(Fig. 33.4C), and more definitive separation of extraaxial col- flamed displaced dura, is seen on MRI of an epidural empyema
lections from their associated intraaxial complications such but not on that of a subdural empyema. Finally, the delineation
as edema, cerebritis, and venous thrombosis, which are more of extraaxial inflammatory disease, leptomeningeal disease,
readily visualized on MRI than on CT (36,62). Second, streak and parenchymal abnormalities is improved with the use of
artifacts from the bony calvaria, which are particularly prob- the MRI contrast agent, gadolinium diethylenetriamine pen-
lematic on CT, are not limitations on MRI. Third, MRI is su- taacetic acid (64), and diffusion-weighted imaging (108,109).
perior to CT in differentiating noninfected subdural effusions CT and MRI play a role in the follow-up of patients
and hygromas from infected empyemas. As with other protein- with extraaxial empyemas (10,36,59). Residual or recur-
aceous fluids, the T1- and T2-weighted values of purulent col- rent collections that may necessitate reexploration are par-
lections are smaller than those of CSF. These collections are, ticularly prone to occur in the parafalcine or subtemporal
therefore, mildly hyperintense to CSF on T1-weighted images regions. These locations are well imaged on MRI because
and markedly hyperintense to CSF on T2-weighted images of its ability to obtain direct coronal sections of the brain.
(36). Fourth, the unprecedented sensitivity of MRI to subtle Long-term follow-up CT or MRI examinations frequently
changes in tissue water content is uniquely suited to the goal demonstrate cortical atrophy adjacent to a previous extra-
of early detection of the parenchymal abnormalities that can axial empyema.

A B C

FIGURE 33.4 Otorhinologically induced subdural empyema. A: Contrast-enhanced computed tomo-


graphic scan demonstrating subtle right hemispheric empyema. B: Interhemispheric subdural empyema
(arrows) delineated by the falx medially and with early membrane formation laterally. C: T2-weighted
MRI scan taken the same day. Note the improved visualization of the right convexity compared to that of
A (long arrows); also note the interhemispheric subdural empyema (short arrows) and the inflammatory
disease in the frontal sinus (arrowheads).

Scheld_Ch33.indd 570 2/21/14 5:47 PM


Chapter 33: Subdural Empyema and Suppurative Intracranial Phlebitis 571

In addition to providing no valuable diagnostic informa-


tion, lumbar puncture is a potentially dangerous procedure
in patients with signs of increased ICP (66). Several deaths
from cerebral herniation have been reported in patients with
subdural empyema shortly after undergoing lumbar puncture
(13,25,26,40,44). Certainly, patients with papilledema or a
focal neurologic abnormality or patients with suspected in-
creased ICP should not undergo a lumbar puncture.

Bacteriology
The microbiologic etiology of subdural empyema is established
by Gram stain and culture of evacuated pus from the subdural
FIGURE 33.5 Contrast-enhanced computed tomographic scan of the
space. Unfortunately, cultures of subdural pus are sterile in
paranasal sinuses demonstrating extensive maxillary, ethmoid, and
sphenoid sinus inflammation (arrows). about one third of patients (25) because patients are almost
always receiving antibiotics preoperatively. It has been sug-
gested that the high number of negative cultures is also related
to the lack of proper handling and culture for anaerobes (25).
Lumbar Puncture In one study in which paranasal sinus cultures and subdural
Lumbar puncture is often performed in patients who are sub- cultures were compared, three of the four sinus isolates did
sequently diagnosed with subdural empyema, but it is neither not correlate with the subdural isolates (26). Blood cultures
sensitive nor specific for this disease. Recovery of a causative may provide additional diagnostic information in about 10%
organism in the CSF is rare, except in infants in whom men- of cases in which the subdural fluid is sterile (11,21,26). The
ingitis preceded the development of the subdural empyema organisms cultured most often from subdural infections are
(11,20,21,54). The CSF formula in children and adults with aerobic and anaerobic streptococci. Staphylococci are cultured
subdural empyema is unpredictable, as shown in Table 33.3. less often, followed by aerobic gram-negative bacilli and non-
Typically, the white blood cell (WBC) count is elevated; how- streptococcal anaerobes (Table 33.4). In most patients, a single
ever, many series report patients with zero to five CSF leu- organism is responsible for subdural empyema. However, sev-
kocytes per cubic milliliter (13,25,26,29,30). The differential eral series have included cases in which multiple organisms
cell count on the CSF is highly variable: although a polymor- have been cultured (3,5,7,16,17,19,21,22,25,27).
phonuclear pleocytosis is more common, the mononuclear cell Generally, the causative organism is predictable based
predominates in close to 40% of patients. A normal protein on the anatomic focus from which the infection originated
concentration suggests the absence of a subdural empyema be- (3,5,8,13,45,55,56,100). Otorhinogenic subdural empyemas
cause there is an inflammatory response by the arachnoid to are most often due to aerobic and anaerobic streptococci and
the overlying subdural process. However, because the arach- are less often due to coagulase-positive staphylococci and
noid is generally impermeable to the infectious agent, CSF other anaerobes. Infections secondary to head trauma, sur-
Gram stain and culture almost never demonstrate the bacterial gery, or an indwelling foreign device are most often caused
cause of the subdural empyema and are, therefore, not helpful by coagulase-positive and coagulase-negative staphylococci
in choosing antibiotic therapy. and gram-negative bacilli. Four cases of postneurosurgical

A B C

FIGURE 33.6 Subdural empyema in the setting of orbital and paranasal sinus inflammatory disease. A,
left: Contrast-enhanced computed tomographic (CT) scan demonstrating a right orbital abscess (long
arrows) and ethmoid sinusitis (short arrows). Proton-density MRI scan (B, center) and T2-weighted
MRI (C, right) taken the same day as for A, demonstrating a small right convexity subdural collection
(arrows), which is brighter than CSF in B and which is, therefore, not a sterile effusion or hygroma. Note
that the conspicuity of the empyema on MRI is greater than that on CT.

Scheld_Ch33.indd 571 2/21/14 5:47 PM


572 Part III: Bacterial and Mycoplasmal Infections

TA B L E 3 3 . 3
CEREBROSPINAL FLUID FINDINGS IN SUBDURAL EMPYEMA

CSF Parametera No. of LPs with Parameter Reported Number (%) References

5 WBCs 108 11 (10%) 68, 13, 21, 23, 2527, 29, 30, 57, 65
6499 WBCs 94 63 (67) 8, 13, 21, 23, 2527, 29, 30, 57, 65
500999 WBCs 94 8 (9) 8, 13, 21, 23, 2527, 29, 30, 57, 65
1,000 WBCs 94 15b (16) 8, 13, 21, 23, 2527, 29, 30, 57, 65
50% polys 64 43 (67) 8, 13, 21, 23, 2527, 29, 30, 57, 65
Elevated proteinc 93 76 (82) 7, 8, 13, 23, 2527, 29, 30, 57, 65
Normal glucosed 72 66e (92) 7, 8, 13, 21, 23, 25, 26, 29, 57, 65
Positive Gram stain 34 2 (6) 68, 25, 27, 65
Positive CSF culture 156 14b (9) 68, 13, 16, 19, 21, 23, 2527, 29, 30, 65, 100

LP, lumbar puncture.


a
From series with more than three lumbar punctures reported.
b
Two with preceding bacterial meningitis.
c
Reported as elevated or 50 mg/100 mL.
d
Reported as normal or 50 mg/100 mL.
e
Four of six with decreased glucose with bacterial meningitis or preceding neurosurgery.

subdural empyema caused by Propionibacterium acnes, a Treatment and Outcome


gram-positive anaerobic bacillus, were reported often occur-
ring several weeks after the surgical procedure (67,68). The clinical suspicion of subdural empyema requires the im-
Subdural empyemas originating from distant foci of infection mediate institution of parenteral antibiotic therapy. Antibiotics
are caused by a variety of organisms. In infants with leptomen- should be chosen based on the suspected source of the infec-
ingitis, subdural empyema is caused by the same organism re- tion and on the organisms known to commonly cause subdural
sponsible for the meningitis, usually Streptococcus pneumoniae empyema. Although no prospective comparisons of antibiotic
or Haemophilus influenzae (21,54). Many organisms other than regimens for subdural empyema have been conducted, an
those mentioned have been reported to cause subdural empy- acceptable empirical therapy includes a -lactamasestable
ema. These include Salmonella species (69,70), Campylobacter penicillin, a third-generation cephalosporin, and metroni-
fetus (52), Serratia marcescens (71), Neisseria meningitidis dazole. Depending on the prevalence of methicillin-resistant
(72,73), Pasteurella multocida (52,74), Actinomyces israelii, Staphylococcus aureus or the likelihood of coagulase-negative
and Actinobacillus actinomycetemcomitans (new genus name staphylococci, vancomycin may be used in place of the
Aggregatibacter) (75). In the Far East, a greater percentage of bac- -lactamasestable penicillin. Although there is no consensus,
terial pathogens are gram-negative aerobes (Enterobacteriaceae), some advocate irrigation of the subdural space with antibiotics
particularly Klebsiella pneumoniae (110). (2,8,9,12,14,49). No current data support a specific duration
of antibiotic therapy; however, most patients are treated for 3
to 4 weeks after drainage (21). Empirical therapy for seizure
TA B L E 3 3 . 4 prophylaxis has been advocated (8,13,28,45), and steroids
and mannitol have been used successfully to decrease ICP in
MICROBIOLOGY OF ADULT SUBDURAL EMPYEMA
individual cases (5,13,24).
Organism Incidencea Although anecdotal cases have been successfully treated with
antibiotics alone (76,77), surgical drainage of a subdural empy-
Streptococci ema is imperative. Disagreement exists, however, over the opti-
mal mode of surgery. The comparative efficacy of multiple burr
Aerobicb 36%
holes versus craniotomy is complicated by clinical factors that
Anaerobic 10% may contribute to outcome. Several parameters have been sug-
Staphylococci gested to be important in predicting patient mortality, includ-
Coagulase positive 9% ing age of patient (12), source of infection (12), microbiology
(12), time from presentation to surgery (13), management of
Coagulase negative 3% the primary source of infection (1), extent of spread of empy-
Aerobic gram-negative bacillic 10% ema (4), level of consciousness at presentation (1,4,79,12,27),
Other anaerobes 6% and surgical technique (4,5,79,12,14,22,24,27). Analysis of
the few reviews that correlate the primary source of infection
Sterile 29%
with ultimate outcome suggest that subdural empyema second-
a
ary to paranasal sinusitis is associated with less overall mortality
Over 200 evaluable cases from references 35, 7, 8, 12, 13, 1619, compared to other primary sources of infection (1,3,1214,18).
2123, 2527, 30; total greater than 100% because of multiple
isolates from single cases. However, level of consciousness at presentation and surgi-
b
Includes -, -, nonhemolytic. cal technique have correlated better with outcome in several
c
Mostly enteric bacilli. studies. Table 33.5 compares patient mortality with level of
consciousness at presentation. Those patients presenting awake

Scheld_Ch33.indd 572 2/21/14 5:47 PM


Chapter 33: Subdural Empyema and Suppurative Intracranial Phlebitis 573

TA B L E 3 3 . 5
ASSOCIATION OF LEVEL OF CONSCIOUSNESS WITH MORTALITY IN SUBDURAL EMPYEMA

Grade

I II III IV
Reference No. Deaths No. Deaths No. Deaths No. Deaths

Bradley and Shaw (1) 20 0 18 9 0 0 5 5


Mauser et al. (4) 16 1 36 5 22 6 24 10
Hodges et al. (7) 0 0 5 0 7 0 2 2
Khan and Griebel (8) 2 0 8 0 4 1 1 1
Hockley and Williams (9) 7 1 22 1 0 0 13 5
Bannister et al. (12) 22 2 30 10 6 1 8 6
Dill et al. (18) 13 0 12 0 5 2 2 1
Morgan and Williams (27) 1 1 4 1 0 0 2 2
total 81 5 135 26 44 10 57 32
% Deaths 6% 19% 23% 56%

I, awake and alert; II, drowsy and disoriented; III, responsive to painful stimuli; IV, unresponsive to pain.

and alert (grade I) have the greatest chance of survival and those Several groups have advocated craniotomy over burr-hole
presenting unresponsive to pain (grade IV) are least likely to drainage, citing increased survival in the group treated by
survive. Patients who are drowsy and disoriented (grade II) or craniotomy (4,5,79,12,14,22,24,27,44) (Table 33.6). The
responsive only to painful stimuli (grade III) have intermediate advantage of craniotomy is considered to be related to the
survival statistics. Of the survivors, decreased level of conscious- greater ease of evacuating pus from a larger area. Few inves-
ness at presentation correlates with more severe neurologic tigators, however, have considered the level of patients con-
sequelae (4,7,8). sciousness when evaluating mortality of the surgical groups.

TA B L E 3 3 . 6
COMPARATIVE SURVIVAL RATES FOR SURGICAL APPROACHES TO SUBDURAL EMPYEMAS

Burr Holes Craniotomy

Reference No. Deaths % No. Deaths %

Mauser et al. (4) 46 11 24 47 10 21


Miller et al. (5) 18 4 22 4 1 25
Hodges et al. (7) 11 1 9 2 0 0
Khan and Griebel (8) 9 1 11 5 1 20
Hockley and Williams (9) 22 6 27 20 1 5
Bannister et al. (12) 15 7 47 24 2 8
Le Beau et al. (14) 11 8 73 26 6 23
Dill et al. (18) 8 1 12 22 2 10
Bok and Peter (19) 44 3 7 45 4 9
Feuerman et al. (22) 11 2 18 7 0 0
Borzone et al. (24) 2 0 0 12 2 17
Morgan and Williams (27) 3 3 100 4 1 25
Skelton et al. (30) 3 0 0 7 0 0
Nathoo et al. (44) 90 21 23 322 26 8
Matt Nayan et al. (111) 50 2 4 40 4 10
Liu et al. (97) 18 15
total 361 70 19 602 60 10
range 0100 025

Scheld_Ch33.indd 573 2/21/14 5:47 PM


574 Part III: Bacterial and Mycoplasmal Infections

In several studies in which both the level of consciousness and compared to craniotomy, with one study having a reoperation
the mode of surgery are established, it is notable that patients rate of 50% with burr holes compared to only 20% in the
with grades III and IV coma were more likely to undergo burr- craniotomy group (4,98).
hole drainage (5,7,22,27). In fact, Mauser et al. (4) report a The overall mortality of subdural empyema and the extent
higher death rate in the craniotomy group when patients pre- of neurologic sequelae reported in survivors are summarized in
senting with grades III and IV consciousness are considered Table 33.7. In the past, mortality rates averaged 11% to 12% and
but lower mortality in the craniotomy group among patients morbidity rates were 17% to 45% (98). Some recent studies with
with grades I and II consciousness. Hence, the increased sur- aggressive antibiotic and surgical approaches have reduced the
vival with craniotomy may be related, in part, to its more mortality to less than 5% and morbidity to less than 13% (98).
frequent use in a patient population starting with a better The potential extent of neuroanatomic sequelae is illustrated by
prognosis. A study by Nathoo et al. (44) of 699 patients with necropsy studies (33). Venous sinus thrombosis is a common
subdural empyema reported improved mortality rates using finding in patients with subdural empyema because the route
craniotomy rather than burr holes or limited craniectomy re- of infection to the subdural space is generally via these venous
gardless of severity of disease. However, a good outcome was sinuses. As a result of the absence of valves in the venous system
achieved in 71% of those undergoing burr-hole drainage and of the brain, thrombophlebitis may extend to the cortical and
86% undergoing craniotomy. Nathoo et al. (44) suggest that subcortical veins of the cerebrum. Thrombosis of these vessels
craniotomy provides the best decompression of the brain and results in venous stasis and subsequent congestion and softening
the most complete evacuation of pus. The optimal treatment of adjacent brain tissue. Brain infarction and necrosis (Fig. 33.7),
for all patients remains unclear; however, regardless of the with or without abscess formation, may ensue. Therefore, clini-
initial surgical approach (multiple burr holes vs. craniotomy), cal neurologic sequelae may be a consequence of brain abscess or
several studies report a number of patients requiring reopera- brain infarction secondary to increased ICP or venous thrombo-
tion (4,7,9,98). Several studies have documented increased sis. More than 10% of patients with subdural empyema develop
reoperation rates in those patients treated with burr holes venous sinus thrombosis or brain abscess (13,33,45).

TA B L E 3 3 . 7
MORTALITY AND NEUROLOGIC SEQUELAE OF SUBDURAL EMPYEMA

% Sequelae in Survivors

References Total Cases % Mortality Severea Mild/Moderateb Seizures

Bradley and Shaw (1) 47 32


Mauser et al. (4) 102 26 18 18 29
Miller et al. (5) 23 17 5 11 37
Hodges et al. (7) 14 14
Khan and Griebel (8) 15 15 15 31 46
Hockley and Williams (9) 42 17
Zimmerman et al. (10) 49 12
Smith and Hendrick (11) 22 1
Bannister et al. (12) 66 29
Kaufman et al. (13) 17 35
Le Beau et al. (14) 37 40
Bhandari and Sarkari (15) 37 34 16 32
McIntyre et al. (16) 14 21
Hoyt and Fisher (17) 17 12
Dill et al. (18) 32 9 38 10 3
Bok and Peter (19) 90 8 5 10
Farmer and Wise (21) 17 41 0 33 11
Feuerman et al. (22) 22 14 25 48 8
Borzone et al. (24) 14 14 8 42 37
Cowie and Williams (28) 89 27 0 23 34
Renaudin and Frazee (29) 23 22
Skelton et al. (30) 10 0 0 20 20
Matt Nayan et al. (111) 90 7 0 11
Cole et al. (100) 42 5 20
Legrand et al. (98) 23 4 13

a
Disabling hemiparesis or aphasia.
b
Sequelae not inhibiting activity.

Scheld_Ch33.indd 574 2/21/14 5:47 PM


Chapter 33: Subdural Empyema and Suppurative Intracranial Phlebitis 575

It can be distinguished from spinal epidural abscess by the


absence of tenderness to palpation in most cases of spinal
subdural empyema (78,79,81,102). In children, however,
spinal tenderness is more common (112). Therefore, in chil-
dren, the clinical presentation may be difficult to differenti-
ate from acute transverse myelitis. Spinal subdural empyema
most commonly occurs in the thoracolumbar spine, although
subdural infection of the cervical spine has been reported
(31,80,8285,112).
Spinal subdural empyema most often arises as a result
of hematogenous spread of infection to the spinal subdural
space, with S. aureus being the most common etiologic agent
(78,102). Other reported pathogens include streptococci
(31,86), coagulase-negative staphylococci (79,87), and gram-
negative bacilli (78,80). Occasionally, this may arise from local
trauma or surgical procedures including incidental dural tears
(113,114) and even acupuncture (115). In a series of child-
hood spinal subdural empyema, the most common patho-
A gens were Mycobacterium tuberculosis and Echinococcus
granulosis (112).
Diagnosis is best accomplished by MRI with gadolinium
with decreased signal on T1 and increased signal on T2. Fat-
suppressive techniques with MRI help define the subdural
space (112). Metrizamide-enhanced spinal CT can be used
in areas where MRI is not available or is contraindicated.
Treatment consists of empirical antibiotic therapy initially
directed against S. aureus, streptococci, and gram-negative
enteric bacilli, in association with laminectomy for drainage
of the empyema. Antibiotics can then be adjusted based on
specific culture results for a duration of 2 to 4 weeks.

SUPPURATIVE INTRACRANIAL
PHLEBITIS
Infections within the sinuses or facial structures can lead to
intracranial complications that include subdural empyema,
cerebral abscesses, epidural abscesses, meningitis, and less
B commonly suppurative intracranial phlebitis of the dural
veins. Suppurative phlebitis can occur as a primary complica-
FIGURE 33.7 Two-year-old child with Haemophilus influenzae men- tion or as a secondary complication to other intracranial infec-
ingitis. A, top: Contrast-enhanced computed tomographic (CT) scan
demonstrating extraaxial collections over both frontal convexities and
tions such as subdural empyema (Table 33.8).
in the anterior aspect of the interhemispheric fissure (arrows). Note
the similarity of these collections to the CSF in the ventricular sys-
tem, making it difficult to distinguish them from sterile effusions and Pathogenesis
prominent subarachnoid spaces. Spinal tap revealed purulent men-
ingitis. B, bottom: Follow-up CT scan 2 months later demonstrates Infection usually spreads to the dura via venous drainage
extensive zones of infarction and hydrocephalus. from the sinuses (91), middle ear, face, or scalp. Within the
dura mater are seven paired venous sinuses and five unpaired
sinuses, all of which are spaces between two layers of dura.
They collect blood from the veins of the brain, skull, and
SPINAL SUBDURAL EMPYEMA face, and they empty into the internal jugular veins (38).
Emissary (perforating) veins allow the passage of blood
Spinal subdural empyema is a rare condition, with fewer from the larger veins of the face and scalp into the dural
than 80 cases previously reported in the literature, with most venous sinuses, serving as a potential communication for
reports from the Western Hemisphere in patients in their more superficial infection of the head with the venous system
sixth and seventh decade of life. In 2013, Sandler et al. (112) of the brain (37). The venous system of the head and brain
reported 11 cases of spinal subdural empyema in children is valveless, allowing retrograde spread of thrombophlebitis
throughout the world and reviewed 73 additional cases. The from infected venous sinuses into dural and cortical venous
median age was 6.5 years with 38% of cases younger than channels (21,37,116).
3 years of age and 15% younger than 1 year of age. Most Suppurative intracranial phlebitis can lead to infarction or
of the children were male (2:1 ratio) and 53% were associ- brain inflammation (21,22,57) with subsequent neurologic
ated with spinal congenital abnormalities. Fifty-six percent sequelae such as seizures, hemiparesis, or cranial neuropathies.
of cases lived in the Eastern Hemisphere with only 17% from Cavernous sinus thrombosis is more commonly associated
the United States. with ethmoid and sphenoid disease, whereas occasionally,
In adults, signs and symptoms include fever, back pain, ophthalmic vein thrombophlebitis may extend posteriorly to
and subsequent signs of spinal cord compression (7880). cause cavernous sinus thrombosis (91).

Scheld_Ch33.indd 575 2/21/14 5:47 PM


576 Part III: Bacterial and Mycoplasmal Infections

TA B L E 3 3 . 8
MAJOR SITES OF SUPPURATIVE INTRACRANIAL PHLEBITIS

Dural Venous Sinus Source of Blood Flow Predisposing Site of Infection Clinical Presentation Miscellaneous

Cavernous sinus Superior or inferior Sphenoid and ethmoid Headache Acute onset
ophthalmic vein sinusitis Fever Most common site of
Facial infections Proptosis suppurative phlebitis
Otitis media Ptosis 30% mortality
Dental infection Chemosis 39% complete recovery
Cranial nerve dysfunction
(III, IV, VI; rarely V1, V2)
Decreased visual acuity
Lethargy/coma
Superior sagittal Ethmoid veins Bacterial meningitis Hemiparesis Largest venous sinus
sinus Frontal parietal veins Frontal and maxillary Intracranial hypertension 78% mortality
Occipital superior sinusitis (rarely ethmoid
cerebral vein sinusitis)
Diploic veins Subdural or epidural Headache Rare patient with
empyema Nausea and vomiting complete recovery
Seizures
Confusion/coma
Lateral sinuses Superior sagittal sinus Acute or chronic otitis media Unilateral headache Subacute presentation
Straight sinus Mastoiditis Earache 012% mortality
Pharyngitis Papilledema 75% complete
Facial pain recovery
Diplopia
Vertigo
Cranial nerve dysfunction (V, VI)

Data from references 8890.

to the pathogens involved in chronic otitis media, which is


Clinical Features often its predisposing condition (88). Immunosuppression and
hematologic malignancies may be associated with fungal infec-
Most patients present with headache and fever associated with tions such as mucormycosis (92). A study from Pakistan and
a toxic appearance and leukocytosis. Increased ICP and pap- the Middle East reviewed 109 cases of cerebral venous throm-
illedema are present in 53% to 65% of cases of cavernous bosis of which 20 (18%) had infectious causes ranging from
and lateral sinus thrombosis but less commonly with superior tuberculous meningitis, rhino-orbital fungal infection, bacte-
sagittal thrombosis (88,89). Focal neurologic findings involve rial meningitis, mastoiditis, malignant otitis, and others (117).
cranial nerves primarily based on the location of some cra-
nial nerves to the venous sinuses. Cranial nerves III, IV, and
VI controlling the extraocular muscles are primarily involved
with cavernous sinus thrombosis as they pass through or near
Diagnosis
the inflamed cavernous sinuses (89). Lateral sinus thrombosis High-resolution contrast-enhanced CT scans with fine cuts
is most often associated with unilateral sixth nerve palsy. On through the suspected areas have been used most extensively
rare occasions, the ophthalmic and maxillary branches of the over the years. However, MRI has now become the modality of
trigeminal nerve (V1 and V2, respectively) are involved with choice for diagnosing suppurative intracranial phlebitis, par-
cavernous sinus thrombosis and less commonly with lateral ticularly cavernous sinus thrombosis, using thin-section coro-
sinus thrombosis. Hemiparesis may occur in up to 61% of pa- nal images as well as magnetic resonance angiography (MRA)
tients with superior sagittal sinus thrombosis due to cerebral and magnetic resonance venography (MRV) (91,93,117120).
infarcts (88). In a recent review of cerebral venous thromboses (118), the ad-
vantages and disadvantages of CT and MRI modalities are dis-
cussed. Although the exact sensitivity and specificity for MRI
Microbiology and Pathogenesis techniques are unknown, it is estimated that CT/computed to-
mography venography (CTV) shows 95% sensitivity and 91%
S. aureus is the most common bacterial pathogen, occurring specificity and that MRI/MRV is equal or better than CT/CTV
in more than 60% to 70% of cases of cavernous sinus throm- (118). In cases where MRI is not available or contraindicated,
bosis (8890). Other gram-positive bacteria such as strepto- CT techniques are fairly accurate.
coccal species and pneumococci also can be associated with Cerebral angiography and venography are used only rarely
acute suppurative intracranial phlebitis. In septic lateral sinus (121) since the advent of CT and MRI. Lumbar puncture with
thrombosis, gram-negative bacteria such as Proteus mirabilis CSF evaluation always is abnormal with neutrophilic pleocy-
and Escherichia coli as well as anaerobes such as Bacteroides tosis and elevated pressure but is generally not diagnostic and
fragilis and anaerobic streptococci may play a larger role due may pose some risk in patients with severely elevated ICP.

Scheld_Ch33.indd 576 2/21/14 5:47 PM


Chapter 33: Subdural Empyema and Suppurative Intracranial Phlebitis 577

thrombosis, but its use in septic sinus thrombosis is not clear


Treatment and may be hazardous (95).
All intracranial suppurative infections require the use of
appropriate high-dose antibiotics with good penetration of
the bloodbrain barrier. Appropriate surgical intervention to Prognosis
drain the sinuses or subdural collections is usually manda-
tory. In addition, anticoagulants may be added to the regi- Antibiotics have markedly reduced the morbidity and mortal-
men for cavernous and sinus thrombosis using heparin or ity of suppurative intracranial phlebitis. However, mortality
low-molecular-weight heparin followed by warfarin for a remains as high as 78% for superior sagittal thrombosis, 30%
6-week course or until the thrombus resolves radiographi- for cavernous sinus thrombosis, and only 0% to 12% for lat-
cally (119). The use of anticoagulants, however, is contro- eral sinus thrombosis (88,96). Permanent neurologic sequelae
versial because of the risk of bleeding from carotid artery result in 25% to 100% of those patients who survive and are
rupture or cerebral venous infarction (94). Direct throm- most likely to occur in those with the most severe disease and
bolytic therapy has been used for refractory cerebral sinus those with superior sagittal sinus thrombosis.

References
1. Bradley PJ, Shaw MDM. Subdural empyema management of the primary 29. Renaudin JW, Frazee J. Subdural empyemaimportance of early diagnosis.
source. Br J Clin Med. 1984;38:8588. Neurosurgery. 1980;7:447479.
2. Joubert MJ, Stephanov S. Computed tomography and surgical treatment in 30. Skelton R, Maixner W, Isaacs D. Sinusitis-induced subdural empyema.
intracranial suppuration. J Neurosurg. 1977;47:7378. Arch Dis Child. 1992;67:14781480.
3. Harris LF, Haws FP, Triplett JN, et al. Subdural empyema and epi- 31. Levy ML, Wieder BH, Schneider J, et al. Subdural empyema of the cer-
dural abscess: recent experience in a community hospital. South Med J. vical spine: clinicopathologic correlates and magnetic resonance imaging.
1987;80:12541258. J Neurosurg. 1993;79:929935.
4. Mauser HW, Van Houwelingen HC, Tulleken CA. Factors affect- 32. Kubik CS, Adams RD. Subdural empyema. Brain. 1943;66:1842.
ing the outcome in subdural empyema. J Neurol Neurosurg Psychiatry. 33. Courville CB. Subdural empyema secondary to purulent frontal sinusitis.
1987;50:11361141. Arch Otolaryngol. 1944;39:211230.
5. Miller ES, Dias PS, Uttley D. Management of subdural empyema: a series 34. Kim KS, Weinberg PE, Magidson M. Angiographic features of subdural
of 24 cases. J Neurol Neurosurg Psychiatry. 1987;50:14151418. empyema. Radiology. 1976;118:621625.
6. Weisberg L. Subdural empyema: clinical and computed tomographic 35. Kaufman DM, Leeds NE. Computed tomography (CT) in the diagnosis of
correlations. Arch Neurol. 1986;43:497500. intracranial abscesses. Neurology. 1977;27:10691073.
7. Hodges J, Anslow P, Gillett G. Subdural empyemacontinuing diagnostic 36. Weingarten K, Zimmerman RD, Becker RD, et al. Subdural and epidural
problem in the CT scan era. Q J Med. 1986;59:387393. empyemas: MR imaging. AJNR Am J Neuroradiol. 1987;10:8187.
8. Khan M, Griebel R. Subdural empyema: a retrospective study of 15 patients. 37. Warwick R, Williams PL, eds. Grays Anatomy. 35th ed. Philadelphia: WB
Can J Surg. 1984;27:283288. Saunders; 1973.
9. Hockley AD, Williams B. Surgical management of subdural empyema. 38. Clemente CD. Anatomy: A Regional Atlas of the Human Body. 2nd ed.
Childs Brain. 1983;10:294300. Baltimore: Urban & Schwarzenberg; 1981.
10. Zimmerman RD, Leeds NE, Danziger A. Subdural empyema: CT findings. 39. Weinman D, Samarasinghe HHR. Subdural empyema. Aust N Z J Surg.
Radiology. 1984;150:417422. 1972;41:324330.
11. Smith HP, Hendrick EB. Subdural empyema and epidural abscess in chil- 40. van Alphen HAM, Dreissen JJR. Brain abscess and subdural empyema:
dren. J Neurosurg. 1983;58:392397. factors influencing mortality and results of various surgical techniques.
12. Bannister G, Williams B, Smith S. Treatment of subdural empyema. J Neurol Neurosurg Psychiatry. 1976;39:481490.
J Neurosurg. 1981;55:8288. 41. Van Dellen JR, Boles DM, Van Der Heever CM. Interhemispheral subdural
13. Kaufman DM, Miller MM, Steigbigel NH. Subdural empyema: analysis of empyema. S Afr Med J. 1977;52:266269.
17 recent cases and review of the literature. Medicine. 1975;54:485498. 42. Stephanov S, Joubert MJ, Welchman JM. Combined convexity and para-
14. Le Beau J, Creissard P, Harispe L, et al. Surgical treatment of brain abscess falx subdural empyema. Surg Neurol. 1979;11:147151.
and subdural empyema. J Neurosurg. 1973;38:198203. 43. Holtzman RNN, Tepperberg J, Schwartz O. Parasagittal subdural empy-
15. Bhandari YS, Sarkari NBS. Subdural empyema: a review of 37 cases. ema: a case report with computerized tomographic scan documentation.
J Neurosurg. 1970;32:3539. Mt Sinai J Med. 1980;47:6267.
16. McIntyre PB, Lavercombe PS, Kemp RJ, et al. Subdural and epidural empy- 44. Nathoo N, Nadvi SS, Gouws E, et al. Craniotomy improves outcomes for
ema: diagnostic and therapeutic problems. Med J Aust. 1991;154:653657. cranial subdural empyemas: computed tomography-era experience with
17. Hoyt DJ, Fisher SR. Otolaryngologic management of patients with subdu- 699 patients. Neurosurgery. 2001;49:872878.
ral empyema. Laryngoscope. 1991;101:2024. 45. Rosenbaum GS, Cunha BA. Subdural empyema complicating frontal and
18. Dill SR, Cobbs CG, McDonald CK. Subdural empyema: analysis of ethmoid sinusitis. Heart Lung. 1989;18:199202.
32 cases and review. Clin Infect Dis. 1995;20:372386. 46. Zizmor J, Noyak AM. Radiology of the nose and paranasal sinuses. In:
19. Bok APL, Peter JC. Subdural empyema: burr holes or craniotomy? Paparella MM, Shumrick DA, eds. Otolaryngology: Basic Science and
J Neurosurg. 1993;78:574578. Related Disciplines. Philadelphia: WB Saunders; 1980:10101066.
20. Curless RG. Subdural empyema in infant meningitis: diagnosis, therapy, 47. Grullen A, Brell M, Cardona E, et al. Potts puffy tumour: still not an eradi-
and prognosis. Childs Nerv Syst. 1985;1:211214. cated entity. Childs Nerv Syst. 2001;17:359362.
21. Farmer TW, Wise GR. Subdural empyema in infants, children and adults. 48. Layon J, McCulley D. Subdural empyema and group C Streptococcus.
Neurology. 1973;23:254261. South Med J. 1985;78:6466.
22. Feuerman T, Wackym PA, Gade GF, et al. Craniotomy improves outcome 49. Osgood CP, Dujovny M, Holm E, et al. Delayed post-traumatic subdural
in subdural empyema. Surg Neurol. 1989;32:105110. empyema. J Trauma. 1975;15:916921.
23. Luken MG, Whelan MA. Recent diagnostic experience with subdural 50. Hadj-Djilani M, Calliauw L. A contribution to the rapid diagnosis of sub-
empyema. J Neurosurg. 1980;52:764771. dural empyema. Acta Neurochir. 1982;61:187199.
24. Borzone M, Capuzzo T, Rivano C, et al. Subdural empyema: fourteen cases 51. Casson IR, Patel P, Blair D, et al. Subdural empyema: caused by infection of
surgically treated. Surg Neurol. 1980;13:449452. preexisting subdural hematoma. N Y State J Med. 1981;81(3):389391.
25. Yoshikawa TT, Chow AW, Guze LB. Role of anaerobic bacteria in subdural 52. Khan MI, Chan R. Pasteurella multocida subdural empyema: a case report.
empyema: report of four cases and review of 327 cases from the English Can J Neurol Sci. 1981;8:163165.
literature. Am J Med. 1975;58:99104. 53. Hlavin ML, Kaminski HJ, Fenstermaker RA, et al. Intracranial suppura-
26. Kaufman DM, Litman N, Miller MM. Sinusitis: induced subdural tion: a modern decade of postoperative subdural empyema and epidural
empyema. Neurology. 1983;33:123132. abscess. Neurosurgery. 1994;34:974981.
27. Morgan DW, Williams B. Posterior fossa subdural empyema. Brain. 54. Jacobson PL, Farmer TW. Subdural empyema complicating meningitis in
1985;108:983992. infants: improved prognosis. Neurology. 1981;31:190193.
28. Cowie R, Williams B. Late seizures and morbidity after subdural empyema. 55. Hardy TL, Minor F, Phinney ES. Chronic subdural empyema. Surg Neurol.
J Neurosurg. 1983;58:569573. 1981;16:154156.

Scheld_Ch33.indd 577 2/21/14 5:47 PM


578 Part III: Bacterial and Mycoplasmal Infections

56. Post EM, Modesti LM. Subacute postoperative subdural empyema. 92. Harril WC, Stewart MG, Lee AG, et al. Chronic rhinocerebral mucormy-
J Neurosurg. 1981;55:761765. cosis. Laryngoscope. 1996;106:12921297.
57. Sadhu VK, Handel SF, Pinto RS, et al. Neuroradiologic diagnosis of sub- 93. Eustis HS, Mafee MF, Walton C, et al. MR imaging and CT of orbital
dural empyema and CT limitations. AJNR Am J Neuroradiol. 1980;1: infections and complications in acute rhinosinusitis. Radiol Clin North
3944. Am. 1998;36:11651178.
58. List CF. Diagnosis and treatment of acute subdural empyema. Neurology. 94. Bhatia K, Jones NS. Septic cavernous sinus thrombosis secondary to sinus-
1955;5:663670. itis: are anticoagulants indicated? A review of the literature. J Laryngol
59. Zimmerman RD, Weingarten K. Evaluation of intracranial inflammatory Otol. 2002;116:667676.
disease by CT and MRI. In: Theodore WH, ed. Clinical Neuroimaging. 95. Smith TP, et al. Treatment of docral sinus thrombosis by urokinase infu-
New York: Alan R. Liss; 1988:75100. sion. AJNR Am J Neuroradiol. 1994;15:801807.
60. Sze G, Zimmerman RD. The magnetic resonance imaging of infections and 96. DiNubile MJ. Septic thrombosis of the cavernous sinuses. Arch Neurol.
inflammatory diseases. Radiol Clin North Am. 1988;26:839860. 1988;45:567571.
61. Wehrli FW. Principles of magnetic resonance imaging. In: Stark DD, Bradley 97. Liu ZH, Chen NY, Tu PH, et al. The treatment and outcome of postmen-
WG, eds. Magnetic Resonance Imaging. St. Louis: CV Mosby; 1988:323. ingitic subdural empyema in infants. J Neurosurg Pediatr. 2010;6:3842.
62. Rick PM, Deasy NP, Jarosz JM. Intracranial dural empyema. Br J Radiol. 98. Legrand M, Roujeau T, Meyer P, et al. Paediatric intracranial empyema:
2000;73:13291336. differences according to age. Eur J Pediatr. 2009;168:12351241.
63. Bockova J, Rigamonti D. Intracranial empyema. Pediatr Infect Dis J. 99. Wu TJ, Chen NC, Huang FY. Subdural empyema in children20-year
2000;19:735737. experience in a medical center. J Microbiol Immunol Infect. 2008;41:
64. Bydder GM. Clinical applicability of gadolinium-DTPA. In: Stark DD, 6267.
Bradley WG, eds. Magnetic Resonance Imaging. St. Louis: CV Mosby; 100. Cole TS, Clark ME, Jenkins AF, et al. Pediatric focal intracranial suppuration:
1988:182200. a UK single-center experience. Childs Nerv Syst. 2012;28(12):21092114.
65. Farkas AG, Marks JC. Subdural empyema: an important diagnosis not to 101. Osborne M, Steinberg JP . Subdural empyema and other suppurative com-
miss. Br Med J. 1986;293:118119. plications of paranasal sinusitis. Lancet Infect Dis. 2007;7(1):6267.
66. Duffy GP. Lumbar puncture in the presence of raised intracranial pressure. 102. Darouche RO. Spinal epidural and subdural empyema. Handb Clin
Br Med J. 1969;1:407409. Neurol. 2010;96:9199.
67. Chu RM, Tummala RP, Halb WA. Local intracranial infections due to 103. Tsai B, Lin KL, Lin TY, et al. Potts puffy tumor in children. Childs Nerv
Propionibacterium acnes: report of three cases. Neurosurg. 2001;49: Syst. 2010;26:5360.
717720. 104. Dashti SR, Baharvadat H, Spetzler RF, et al. Operative intracranial infec-
68. Nawashiro H, Uozumi Y. Letter to editor. Neurosurg. 2002;50:1172. tion following craniotomy. Neurosurg Focus. 2008;24(6):E10.
69. Okudera H, Yasutuki T, Kyoshima K. Bilateral subdural empyema due to 105. Van de Beek D, deGans J, Tunkel A, et al. Community-acquired bacterial
Salmonella enteritidis in an infant. Childs Nerv Syst. 1989;5:4546. meningitis. N Engl J Med. 2006;354:4453.
70. Grosinger L, Lauter CB. Salmonella subdural empyema in a patient with 106. Banerjee AD, Pandey P, Devi BI, et al. Pediatric supratentorial subdu-
brain metastasis. Rev Infect Dis. 1986;8:830831. ral empyemas: a retrospective analysis of 65 cases. Pediatr Neurosurg.
71. Safani MM, Ehrensaft DV. Successful treatment of subdural empyema due 2009;45:1118.
to Serratia marcescens. Drug Intell Clin Pharm. 1982;16:777779. 107. Chen CY, Huang CC, Chang YC, et al. Subdural empyema in 10 infants:
72. Edwards MS, Baker CJ. Subdural empyema: an unusual complication of US characteristics and clinical correlates. Radiology. 1998;207:609617.
meningococcal meningitis. South Med J. 1982;75:6869. 108. Fanning NF, Laffan EE, Shroff MM. Serial diffusion-weighted MRI cor-
73. Nithyanandam S, DSouza O, Rao S, et al. Rhinocerebral actinomycosis. relates with clinical cause and treatment response in children with intra-
Ophthal Plast Reconstr Surg. 2001;17:134136. cranial pus collections. Pediatr Radiol. 2006;36:2637.
74. Stern J, Bernstein CA, Whelan MA, et al. Pasteurella multocida subdural 109. Foerster BR, Thurnher MM, Malane PN, et al. Intracranial infections:
empyema. J Neurosurg. 1981;54:550552. clinical and imaging characteristics. Acta Radiol. 2007;8:875893.
75. Louie JA, Kusske JA, Rush JL, et al. Actinomycotic subdural empyema: 110. Tsou TP, Lee PL, Lu CY, et al. Microbiology and epidemiology of brain
case report. J Neurosurg. 1979;51:852855. abscess and subdural empyema in a medical center: a 10-year experience.
76. Mauser HW, Ravijst RAP, Elderson A, et al. Nonsurgical treatment of sub- J Microbiol Immunol Infect. 2009;42:405412.
dural empyema: case report. J Neurosurg. 1985;63:128130. 111. Matt Nayan SA, Mohd Herjani MS, Abd Latif AZ, et al. Two surgical
77. Rosazza A, de Tribolet N, Deonna T. Nonsurgical treatment of interhemi- methods used in 90 patients with intracranial subdural empyema. J Clin
spheric subdural empyemas. Helv Paediatr Acta. 1979;34:577581. Neurosci. 2009;16:15671571.
78. Dacey RG, Winn R, Jane JA, et al. Spinal subdural empyema: report of two 112. Sandler AL, Thompson D, Goodrich JT, et al. Infections of the spinal
cases. Neurosurgery. 1978;3:400403. subdural space in children: a series of 11 contemporary cases and review
79. Theodotou B, Woosley RE, Whaley RA. Spinal subdural empyema: diagno- of all the published reports. A multinational collaborative effort. Childs
sis by spinal computed tomography. Surg Neurol. 1984;21:610612. Nerv Syst. 2013;29:105117.
80. Bartels RH, De Jong R, Grotenhuis JA. Spinal subdural abscess: case 113. Wu AS, Griebel RW, Megurfo K, et al. Spinal subdural empyema after a
report. J Neurosurg. 1992;76:307311. dural tear. Neurosurg Focus. 2004;17:E10.
81. Fraser RAR, Ratzan K, Wolpert SM, et al. Spinal subdural empyema. Arch 114. Vural M, Arslantas A, Adapinar B, et al. Spinal subdural Staphylococcus
Neurol. 1973;28:235238. aureus abscess: case report and review of the literature. Acta Neurol
82. Hirson C. Spinal subdural abscess. Lancet. 1965;2:12151217. Scand. 2005;112:34346.
83. Heindel CS, Ferguson JP, Kumarasamy T. Spinal subdural empyema com- 115. Chen MH, Chen MH, Huang JS. Cervical subdural empyema following
plicating pregnancy: case report. J Neurosurg. 1974;40:654656. acupuncture. J Clin Neurosci. 2004;11:908911.
84. Negrin J Jr, Clark RA Jr. Pyogenic subdural abscess of the spinal meninges: 116. Syed MI, Baring D, Addidle M, et al. Lemmiere syndrome: two cases and
report of two cases. J Neurosurg. 1952;9:95100. a review. Laryngoscope. 2007;117:16051610.
85. Scully RE, Mark EJ, McNeely BU. Case records of the Massachusetts 117. Khealani BA, Wasay M, Saadah M, et al. Cerebral venous thrombosis.
General Hospital: case 471984. N Engl J Med. 1984;311:13651370. A descriptive multicenter study of patients in Pakistan and Middle East.
86. Gelfand MS, Bakhtian BJ, Simmons BP. Spinal sepsis due to Streptococcus Stroke. 2008;39:27072711.
milleri: two cases and review. Rev Infect Dis. 1991;13:559563. 118. Leach JL, Fortuna RB, Jones BV, et al. Imaging of cerebral venous throm-
87. Peacher WG, Robertson RCL. Neurologic complications following the bosis: current techniques, spectrum of findings, and diagnostic pitfalls.
use of continuous caudal anesthesia. Arch Neurol Psychiatry. 1944;52: Radiographics. 2006;26:519543.
531533. 119. Saposnik G, Barinagarrementeria F, Brown RD, et al. Diagnosis and man-
88. Southwick FS, Richardson EP, Swartz MN. Septic thrombosis of the dural agement of cerebral venous thrombosis: a statement for the healthcare
sinuses. Medicine. 1986;65:82106. professionals from the American Heart Association and American Stroke
89. Amran M, Sidek DS, Hamzah M, et al. Cavernous sinus thrombosis Association. Stroke. 2011;42:11581192.
secondary to sinusitis. J Otolaryngol. 2002;31:165169. 120. Hughes DC, Raghawan A, Mordekar SR, et al. Role of imaging and the
90. Ebright JR, Pace MT, Niazi AF. Septic thrombosis of the cavernous sinuses. diagnosis of acute bacterial meningitis and its complications. Postgrad
Arch Intern Med. 2001;161:26712676. Med J. 2010;86:478485.
91. Gallagher RM, Gross CW, Phillips CD. Suppurative intracranial complica- 121. Rodallec MH, Krainek A, Feydy A, et al. Cerebral thrombosis and multide-
tions of sinusitis. Laryngoscope. 1998;108:16351642. tector CT angiography: tips and tricks. Radiographics. 2006;26:S5S18.

Scheld_Ch33.indd 578 2/21/14 5:47 PM


CHAPTER 34 COMPLICATIONS OF INFECTIVE
ENDOCARDITIS
STEFANO GIULIERI, RETO ANTOINE MEULI, AND MATTHIAS CAVASSINI

Central nervous system (CNS) complications of infective endo- incidence and mortality mask the continually evolving spec-
carditis (IE) occur in about 30% of patients, with the highest trum of IE and the contrasts that exist between various types
incidence among patients referred to tertiary care centers and in- of IE. For instance, subacute IE due to viridans streptococci
tensive care units, as well as patients with mitral valve endocar- has an early mortality rate of only 5% to 10%, whereas acute
ditis and IE due to virulent microorganisms (e.g., Staphylococcus prosthetic valve endocarditis (PVE) in an elderly patient with
aureus, gram-negative bacilli, fungi). Ischemic stroke accounts congestive heart failure can be fatal in up to 70% of patients.
for up to two thirds of CNS complications, followed by intra- The overall figures reflect shifting frequency of predisposing
cranial hemorrhage, meningitis, brain abscess, intracranial in- factors. In developed countries, the decreasing prevalence of
fectious aneurysm (ICIA), and encephalopathy. Embolization chronic rheumatic heart disease is counterbalanced by an in-
of infected vegetations is the main pathogenetic mechanism. creased number of patients with prosthetic valves and intra-
Imaging (both computed tomography [CT] and magnetic reso- vascular devices, injection drug use, and elderly patients with
nance imaging [MRI], coupled with noninvasive angiography) degenerative heart disease (10). The mean age of patients pre-
plays a pivotal role in the diagnosis of CNS complications. senting with IE has increased, from 34 to 43 years in the early
Optimal management includes early institution of antimicro- antibiotic period to 52 to 55 years more recently, and has even
bial treatment, which has the highest impact on the risk of fur- passed 60 years in some reports (3,11,12).
ther emboli. Anticoagulation should be cautiously continued
in patients with established cardiac indications (e.g., prosthetic
valve). In the presence of an established indication, patients with
IE and ischemic stroke should undergo cardiac surgery without
PREDISPOSING FACTORS
delay, whereas it should be postponed for at least 4 weeks in Degenerative valve disease has replaced rheumatic heart dis-
case of intracerebral hematoma. Unruptured ICIA may be man- ease as the main underlying condition in patients with IE (10).
aged medically with radiologic follow-up, whereas enlarging or Degenerative valvular disease with calcified atheromatous
ruptured ICIA should be treated surgically or by an endovascu- deposits is associated with IE in a proportion of cases that
lar approach. Despite improvement in the management, patients increases with age and may be a predisposing factor in up to
with CNS complications generally carry a worse prognosis com- 50% of the IE cases in elderly patients (13). Mitral valve pro-
pared to patients without neurologic complications. lapse (MVP) associated with valve dysfunction is another rec-
ognized risk factor for IE, found in 10% to 30% of patients
(14). In a casecontrol study, the risk of IE was eight times
HISTORY higher among patients with MVP (15), but, given the high in-
cidence of MVP (5%) in the general population, the absolute
Infective endocarditis (IE) was probably recognized as early risk per patient is low, estimated to be 0.0175% per year (14).
as 1646 by Rivire, but the full clinical spectrum of malig- Although it remains a common predisposing condition in
nant endocarditis was first comprehensively described by developing countries, rheumatic heart disease is associated
Sir William Osler (1) in his Gulstonian lectures at the Royal with IE in less than 5% of cases in the Western countries (16).
College of Physicians in London in 1885. He not only de- Congenital lesions are found in 6% to 20% of cases (17), mostly
scribed the different modes of onset, and the extraordinary ventricular septal defect, bicuspid aortic valve, patent ductus
diversity of symptoms which may arise, but carefully ana- arteriosus, and tetralogy of Fallot. A bicuspid aortic valve is an
lyzed the pathophysiology of distant complications, particu- important factor in elderly persons in whom it may be present
larly of central nervous system (CNS) manifestations such in up to 20% of IE cases; this is probably because of associated
as meningitis: The meningeal complications of endocarditis valvular sclerosis and abnormalities of blood flow (18).
have not received much attention, considering the frequency Prosthetic valves account for 7% to 25% of the cases of
with which it has occurred . . . somewhat over 12 per cent (1). IE and the annual incidence of IE in patients with prosthetic
He also related IE and mycotic aneurysms to a common infec- valves is approximately 1% (19). In-hospital mortality rate
tious etiology and later emphasized the central importance of ranges from 20% to 30% (20).
blood cultures in diagnosis (2). IE related to hospitalization and ambulatory invasive
treatments (health care associated) now accounts for one
third of cases (21). The proportion of IE associated with
EPIDEMIOLOGY other intracardiac devices such as pacemakers or implant-
able cardioverter-defibrillators was 6.4% in the ICE-PCS (22).
In developed countries, the reported incidence of IE is 3 to 7 per Nosocomial bacteremias (mainly Staphylococcus aureus) rep-
100,000 per year (36). This rate has remained fairly constant resents as many as 25% of the cases in certain series (23,24).
over time (7). Incidence of IE increases sharply with age, with IE is reported in 2% to 6% of patients undergoing long-term
a peak of 15 episodes per 100,000 per year in patients older hemodialysis (25). Intravenous drug users (IVDUs) are at high
than 70 years (4,8). The male:female ratio is 2:1. In-hospital risk for IE. Moreover, 20% to 40% of IVDUs suffering from
and 6-month mortality in recent series are 15% to 20% and IE have preexisting cardiac lesions, often caused by previous
25%, respectively (9). These apparently stable overall rates of infection (26).

579

Scheld_Ch34.indd 579 2/21/14 5:47 PM


580 Part III: Bacterial and Mycoplasmal Infections

countries (6). In the International Collaboration on


PATHOGENESIS Endocarditis-Prospective Cohort Study (ICE-PCS), S. aureus
and streptococci both accounted for 32% of 1,881 cases of
Certain lesions of the endothelium of the valve or heart cavities definite NVE (16). The increase in frequency of S. aureus IE
can easily be infected should a bacteremia occur. This has been is explained by several invasive medical procedures associated
shown in animal models: An intravascular polyethylene cathe- with S. aureus bacteremia (e.g., hemodialysis, long-term intra-
ter placed across the aortic or the tricuspid valve will induce the venous catheters) (24). The clinical course of S. aureus IE is
deposition of platelets and fibrin and lead to the formation of usually acute and complicated. In-hospital mortality rate was
nonbacterial thrombotic endocarditis (NBTE) (27). Many fac- 22% in a recent prospective study (24). Coagulase-negative
tors can induce endothelial lesions, which promote the forma- staphylococci rarely cause NVE but appear to be increasing
tion of NBTE. The most important are valvular organic lesions in frequency and can pursue an aggressive clinical course (34).
with associated perturbations of blood flow and prosthetic Special attention has been drawn to Staphylococcus lugdunen-
valves. Even microscopic lesions are prone to infection by circu- sis, being commonly associated with valve destruction (35).
lating bacteria (28), so not surprisingly, up to 50% of patients In a recent French series of 497 patients with definite IE
with IE have no known predisposing heart condition at the time (79% with NVE), streptococci, although less frequent com-
of diagnosis (29). Transient asymptomatic bacteremias occur pared to a previous study (4), were still the most common
frequently, often following minor mucosal trauma induced by cause of IE and were responsible for 48% of cases (6). The
daily domestic activities such as chewing or tooth brushing. viridans group (e.g., Streptococcus sanguis, Streptococcus ora-
Bacteremia may also be triggered by various iatrogenic proce- lis, Streptococcus mutans, and Streptococcus mitis) accounts
dures. Among the many bacterial species that can be recovered for at least two thirds of these cases. The clinical course is gen-
during transient bacteremias, streptococci account for most IE erally subacute and an underlying cardiac condition is often
cases, probably because of the frequency with which they enter present. Streptococcus milleri, an occasional cause of IE, has
the bloodstream and the adherence properties of their surfaces. a peculiar propensity to cause distant abscesses and local peri-
Circulating bacteria readily attach to NBTE. The attachment valvular invasion (36). Streptococcus bovis group (including S.
process is probably mediated on the host side by receptor-like gallolyticus subsp. gallolyticus, S. gallolyticus subsp. pasteria-
structures such as fibronectin, fibrinogen, laminin, or collagen, nus, and S. infantarius subsp. coli) accounts for up to 20% of
which interact with the surface of bacteria (8). Platelets also play streptococcal IE (37) and is often associated with neoplasms
a role in these initial events (30,31). On the microbial side, only of the gut, of which it may be the first manifestation (38,39).
bacteria with the ability to adhere to valvular lesions can pro- Streptococcus pneumoniae and streptococci of the Lancefield
duce IE. Gram-positive cocci (e.g., staphylococci, streptococci) groups A, B, C, G may also occasionally cause endocarditis
adhere more strongly than enterobacteria to heart valves (32). (40,41). Enterococci account for 10% of all IE (42); these
Once microorganisms have attached to NBTE, they start to strains are particularly resistant to the bactericidal activity of
multiply and stimulate further deposition of fibrin and platelets antibiotics. Older people are usually affected and outcome can
through the activation of tissue factors or procoagulant activ- be fatal in approximately 16% of patients (42).
ity. This thrombotic process buries some bacterial colonies deep Numerous other bacteria are well known but uncom-
within the vegetation where they are protected from circulating mon causes of NVE, for example, Coxiella burnetii, Brucella,
neutrophils. This has led to the concept of the vegetation as an and bacteria of the HACEK group (Haemophilus species,
area of localized agranulocytosis. Bacteria deeply seated in the Actinobacillus actinomycetemcomitans, Cardiobacterium hom-
vegetation are metabolically inactive and thus resistant to the ac- inis, Eikenella corrodens, Kingella species) (43). Actinobacillus
tion of some antibiotics. Furthermore, some antibiotics do not actinomycetemcomitans has now been reclassified as
fully penetrate into the vegetation. This combination of factors Aggregatibacter actinomycetemcomitans (44). Bartonella is an
favors the bacteria and helps explain why cure of endocarditis unusual cause of endocarditis in homeless people (45).
requires the prolonged administration of bactericidal antibiotics.
Because the infected vegetation is located within the blood-
stream, persistent bacteremia is a hallmark of bacterial endocar-
ditis, permitting the diagnosis through blood cultures in most Prosthetic Valve Endocarditis
patients. Moreover, further deposition of platelets, fibrin, and
circulating bacteria will compensate for the fragmentation, em- PVE cases with onset within 2 months after surgery are called
bolism, and/or resorption of the vegetation by the inflammatory early PVE (46). Early PVE cases are presumably related to
response, in a delicate and complex balance. IE also induces im- intraoperative contamination. S. aureus is responsible for
munologic responses that contribute to some clinical manifesta- 35% of early PVE cases; coagulase-negative staphylococci are
tions of IE such as glomerulonephritis or vasculitis (30). found in 17% of cases. The frequency of gram-negative bacilli
(10% to 20%), diphtheroids (0% to 3%), and fungi (5% to
10%) is higher in PVE than in NVE (47), and they are usually
ETIOLOGIC AGENTS isolated in early PVE.
Cases occurring 2 months or more after surgery are called
The list of microorganisms that can cause IE includes most late PVE and are largely community acquired. As compared to
human pathogenic bacteria, as well as rickettsiae, Bartonella, early PVE, streptococci are more common and are responsible
chlamydiae, and fungi (19). However, gram-positive cocci pre- for approximately 20% of late PVE cases (46).
dominate. The pattern of distribution of these etiologic agents
differs profoundly according to underlying risk factors, the
valve type (native vs. prosthetic), and geography (33). Intravenous Drug Users
S. aureus accounts for more than 50% of IVDU cases probably
Native Valve Endocarditis because of its high frequency on the skin in local infections at the
injection site (cellulitis, abscesses, suppurative thrombophlebitis)
S. aureus is now as common as streptococci as a causative and on drug paraphernalia (48). In some regions, methicillin-
pathogen of native valve endocarditis (NVE) in developed resistant S. aureus (MRSA) have been responsible for IE among

Scheld_Ch34.indd 580 2/21/14 5:47 PM


Chapter 34: Complications of Infective Endocarditis 581

IVDUs. Several other bacterial species such as Pseudomonas findings (59). Based on the number of major and minor criteria,
aeruginosa, enterococci, and Serratia species or fungi are all more any case of suspected IE is classified as definite, possible, or
common in addicts than in the general population (49). In 40% rejected IE. These criteria have been primarily developed for
to 70% of the cases, the tricuspid valve is affected (50). Right- research purpose, but they can help integrate diagnostic findings
sided IE among IVDUs has a mortality rate of less than 10% (48). (Table 34.1)

TA B L E 3 4 . 1
Culture-Negative Infective Endocarditis
CRITERIA AND DIAGNOSIS OF INFECTIVE
Patients with a clinical presentation suggestive of IE but with ENDOCARDITIS
negative blood cultures account for 5% to 10% of the cases
(8). The most common cause for culture-negative IE is prior Criteria
administration of antibiotics. In the absence of prior antibiotic
Major
administration and with appropriate blood culture media, most
bacteria will eventually grow but may require repeated blood 1. Blood culture Two separate blood cultures with
culture and prolonged incubation periods (51). Some micro- typical microorganism
organisms, such as Aspergillus species, C. burnetii, Legionella Persistently positive blood cultures
species (usually Legionella pneumophila), or Tropheryma (12 hours apart or three of three
whippelii, cannot be recovered from the blood, so diagnosis or more)
relies on other methods such as tissue culture, in situ hybrid- Single positive blood culture for
ization, polymerase chain reaction (52), or special serologic Coxiella burnetii or antiphase I
tests (53). By applying a standardized diagnostic algorithm, IgG antibody titer 1 : 800
Fournier et al. (33) were able to identify an etiologic agent
2. Endocardial New regurgitant murmur
among 476 out of 759 (63%) patients referred for blood
involvement
culturenegative IE. C. burnetii (229 cases) and Bartonella sp.
(86 cases) were the most common etiologic agents. A noninfec- Positive echocardiogram for IE
tious origin of culture-negative IE (e.g., marantic endocarditis, (oscillating mass, abscess, new
systemic lupus erythematosus) was diagnosed in 19 patients. dehiscence of PV)
Minor
1. Predisposition, predisposing heart condition, or
CLINICAL MANIFESTATIONS injection drug use
2. Fever, temperature 38C
The signs and symptoms of IE are extremely variable because of
the diversity of the etiologic agents and the various organs in- 3. Vascular phenomena: emboli, infectious aneurysm,
volved. Clinical presentation may range from a chronic disease intracranial hemorrhage
with unspecific systemic symptoms to acute life-threatening 4. Immunologic phenomena: glomerulonephritis, Osler
sepsis. Fever with a heart murmur is the most common clinical nodes, Roth spots, RF
presentation; it may be the only clue to the diagnosis, especially 5. Microbiology: positive blood culture but not meeting
early during the course of the disease and with microorgan- major criteria, serological evidence of active infection
isms of low virulence. In a prospective study of 109 episodes with organism consistent with IE
of IE, fever was present in 98% of cases (54). A new murmur Diagnosis
and skin lesions were found in 59% and 32%, respectively.
Splenomegaly and musculoskeletal manifestations were pres- Definite
ent in 16% (54). Congestive heart failure complicates one third Pathology or bacteriology of vegetations
of episodes in recent studies (4,16,54). Distant complications or
such as major emboli, lumbar pain, or rupture of a infectious
Two major criteria
aneurysm may be the first presenting manifestations of IE. All
the various clinical manifestations of IE arise from one or more or
of four main mechanisms: bacteremia, embolization (55), im- One major and three minor criteria
munologic manifestations (56), and local complications (57). or
Five minor criteria
Possible
DIAGNOSIS
One major criterion and one minor criterion
The diagnosis of IE requires the integration of clinical, mi- or
crobiologic, and echocardiographic data. Hematologic ab- Three minor criteria
normalities are frequently observed: Anemia, leukocytosis,
and thrombocytopenia are present in 66%, 50%, and 18% of Rejected
cases, respectively. Urinalysis shows abnormalities in 30% or Firm alternative diagnosis
more of patients, mainly evidence of proteinuria or microscopic or
hematuria, one third of patients has elevated creatinine (54). Resolution on 4 days of antibiotics
Circulating immune complexes may be detected, but they are
nondiagnostic (56). Repeated electrocardiography can reveal
PV, prosthetic valve; RF, rheumatoid factor.
new atrioventricular block, particularly in the setting of aortic From Li JS, Sexton DJ, Mick N, et al. Proposed modifications to the
valve endocarditis, suggesting perivalvular invasion (58). Duke criteria for the diagnosis of infective endocarditis. Clin Infect
The Duke criteria combine predisposing factors to IE, the blood Dis. 2000;30:633638, with permission.
culture isolate or histologic examination, and echocardiographic

Scheld_Ch34.indd 581 2/21/14 5:47 PM


582 Part III: Bacterial and Mycoplasmal Infections

Echocardiography has proven to be a valuable diagnostic during the course of medical treatment in almost 50% of pa-
tool in patients with suspected IE. Transesophageal echocar- tients with IE (63). In the ICE-PCS, the rate of surgical inter-
diography (TEE) has substantially better ability to detect vege- vention (48%) was comparable among patients with NVE and
tations, perivalvular extension, and myocardial abscesses than PVE (16). Nearly all patients with fungal endocarditis require
transthoracic echocardiography (TTE). TTE has a sensitivity combined surgical and medical treatment (64). Hemodynamic
of 40% to 63% for detecting vegetations in NVE (60). TEE deterioration, local complications, and uncontrolled infection
increases the sensitivity for detecting vegetations in NVE to are the major indications for surgery (10,65). Although the
more than 90% (61). TEE is particularly useful in PVE, with need for surgery in patients presenting with hemodynamic
which vegetations are detected in more than 80% of the cases deterioration and uncontrolled infection is widely recognized
compared with less than 30% with TTE. A negative TEE has a (66), it remains controversial whether surgery is indicated for
negative predictive value for IE of 86% to 97%. prevention of systemic embolism. In a randomized study from
Korea, patients at high risk for systemic embolism (i.e., with
severe valvular regurgitation and large vegetations) had signifi-
TREATMENT AND PREVENTION cantly less systemic embolic events if operated early during the
course of disease (67). Figure 34.1 summarizes current indica-
Once the diagnosis of IE has been made, antibiotic treatment tions for valve surgery in patients with IE. Other interventions
should be started promptly to eradicate the infecting mi- that should be considered in patients with IE are the removal
croorganisms as soon as possible. It is well established that of intravascular devices (e.g., pacemakers, intracardiac car-
a prolonged course of antibiotics is needed for cure even if dioverter defibrillators, central venous catheters) (68), and the
the microorganisms are highly sensitive to the drug used. The drainage of extracardiac infections (e.g., septic arthritis).
antibiotic agent must be bactericidal and should be selected Attempted prevention of IE is aimed at patients with
according to in vitro susceptibility test results and the experi- known underlying predisposing factors who undergo a proce-
ence gathered from experimental and clinical studies (10,62). dure that may result in bacteremia. Possible portals of entry,
Surgery has an important role in the management of IE, as an such as oral and dental lesions or urinary or gastrointestinal
optimal therapeutic approach requires operative intervention tract pathology, should be sought and treated if necessary.

HIGH EMBOLIC RISK PERSISTENT SEPSIS


HEART FAILURE (large vegetation >10 mm, (periannular abscess,
previous embolic event) difficult microorganism)

YES NO YES NO YES NO

Severe Controlled Other poor prognostic features


by medical treatment (difficult organism,
severe valve regurgication)

Severe
valve regurgitation
YES NO

YES NO

Emergency/ Urgent/ Medical Urgent Individual Medical Urgent Medical


urgent surgery elective surgery management surgery decision* management surgery management

*based on size and mobility of vegetation,


affected valve, previous embolism, duration
of antibiotic treatment

FIGURE 34.1 Indications and timing of valve surgery in patients with IE. (Adapted from Prendergast
BD, Tornos P. Surgery for infective endocarditis: who and when? Circulation. 2010;121:11411152.)

Scheld_Ch34.indd 582 2/21/14 5:47 PM


Chapter 34: Complications of Infective Endocarditis 583

Recommendations for use of prophylactic antibiotics have IE hospitalized in 33 intensive care units (ICUs) in France,
been updated, and indications for prophylaxis have been 108 (55%) presented with neurologic complications (106).
considerably restricted to four high-risk conditions: prosthetic In contrast, the rate of CNS complications was only 22% in
cardiac valve, history of previous IE, some forms of congenital a series of IE observed in a large community hospital (11).
heart disease (CHD) (e.g., unrepaired CHD), and heart trans- This referral bias would alter not only the number but also
plantation recipients with valvulopathy (69). the type and severity of the complications because referral is
often precipitated by CNS involvement. On the other hand,
in a series gathered from six hospitals of different types (uni-
CENTRAL NERVOUS SYSTEM versity, private, and Veterans), the frequency of CNS compli-
cations was very similar (92).
COMPLICATIONS OF INFECTIVE
ENDOCARDITIS Age and Sex
Among the various manifestations of IE, neurologic complica- In many studies of adults with IE, the age of the patients did not
tions are particularly important for three main reasons: They appear to greatly influence the overall frequency of CNS com-
occur often, they may be the first or the predominant manifes- plications. In a recent series, however, major neurologic events
tation of the disease, and they are a leading cause of death and were recorded in 22%, 28%, and 34% of patients younger
complications due to IE (70). than 40 years, 40 to 60 years, and older than 60 years, re-
spectively. In elderly patients, neurologic manifestations tend
to be more common as a presenting clinical sign of IE (107);
Incidence as many as one fourth to one third of them may present with
neurologic signs (86), compared with 5% to 17% in the gen-
The reported overall incidence of CNS complications of IE eral population. If disorientation is included as a neurologic
varies greatly. In most series, the incidence of CNS involve- finding, the figure can be as high as 45%. Neurologic compli-
ment during the course of IE ranges between 20% and 40%, cations in children reported in five series ranged from 15% to
with an average of 30%. The incidence has not changed much 32% (108). Because major neurologic events are uncommon
over time, despite wide differences in the diagnostic criteria in children or young adults, everyone should recall the dictum,
employed in various studies and many evolutionary changes In hemiplegia in young adults or children, always think of
that have occurred in the natural history of IE over the past infective endocarditis.
60 years. Among 743 patients pooled from seven series pub- Generally, the sex distribution of the patients with or with-
lished before the advent of antibiotics, 176 (24%) were noted out neurologic complications appears to be similar in both
to have neurologic manifestations (1,7176). Among 1,622 sexes (83).
patients from eight studies reported between 1947 and 1978,
457 (28%) had such complications (7785). Among 1,329 Location of Vegetations in the Heart
episodes of IE from seven series published between 1981 and Neurologic complications are a hallmark of left-sided valvular
1993, 437 (33%) were accompanied by neurologic manifesta- abnormalities. This association was noted as early as 1852,
tions (11,8693). And among 2,162 episodes of IE from eight before IE was recognized as a clinical entity (109). The inci-
series published between 1996 and 2007, 512 (24%) were dence of neurologic complications is markedly lower in iso-
complicated by neurologic events (70,94101). Recent studies lated right-sided IE. In a series of 40 IVDUs with tricuspid IE
may indicate a lower rate of neurologic complications: The in- due to S. aureus, none presented with cerebral manifestations,
cidence of stroke in a French cohort of 390 patients and in the whereas 2 of 5 of those with left-sided involvement had cere-
ICE-PCS was 18% and 17%, respectively (4,16). However, bral septic emboli (110). Of 97 episodes of IE in drug addicts,
data on other neurologic complications were not reported. none of the nine major CNS events that were present on initial
Finally, among 513 patients presenting with complicated evaluation occurred in the 32 cases that involved the right side
native-valve IE, focal neurologic findings, and altered mental of the heart (111), although the infection was caused by highly
status were described in 18% and 16%, respectively (9). virulent organisms such as S. aureus. CNS complications were
Incidence of asymptomatic CNS events is much higher. By observed in 54 (41%) of 133 patients with aortic and/or mitral
systematically using resonance imaging of the brain, Snygg- valve involvement and in only 4 (12%) of 33 of those with tri-
Martin et al. (102) identified cerebrovascular complications cuspid IE (92). When neurologic manifestations occur in right-
in 65% of patients; about half of them were asymptomatic. sided IE, they present as meningitis, cerebral abscesses, or
In another study of systematic brain imaging in IE, radiologic encephalopathy, rather than as strokes, and are most often re-
evidence of brain embolism was detected in 80% of patient, lated to virulent pathogens such as S. aureus or S. pneumoniae
whereas 25% had clinical stroke (103). (112). Systemic embolization is rare in right-sided IE but does
occasionally occur either from septic thrombi arising in the
pulmonary veins or via paradoxical embolization through a
Factors Influencing the Incidence of Central patent foramen ovale, which is present in 18% of the normal
Nervous System Complications population.
Mitral valve endocarditis is associated with a higher rate
The reported incidence is understandably higher in stud- of neurologic complications (83,106,113,114). For example,
ies devoted primarily to the neurologic aspects of IE and in a recent French study of 198 patients with IE, mitral valve
in studies based on autopsies, because brain damage often involvement was independently associated with neurologic
causes or contributes to death in IE (11,78,79). In one au- complications (OR 1.54, CI 1.07 to 2.21) (106). A study
topsy series of 69 cases of IE, cerebral emboli were found from Duke University including 707 patients with IE found a
in up to two thirds of the young adults and in 46% of the twofold risk of stroke among patients with mitral valve endo-
total cases (104). Frequency of CNS complications also tends carditis as compared to aortic valve endocarditis (114). This
to be higher in series gathered from referral centers (105) association was not reported by older studies (115), probably
or specialized units. For example, among 198 patients with because of small size (88).

Scheld_Ch34.indd 583 2/21/14 5:47 PM


584 Part III: Bacterial and Mycoplasmal Infections

Neurologic complications in the context of PVE are of spe- 100


cial concern because of the threat of superimposed intrace-
rebral hemorrhage, which might be caused or promoted by
anticoagulation. Most published data are derived from PVE
complicating mechanical prosthetic valves. The overall inci-
dence of focal neurologic events associated with PVE varies
from 11% to 44% (46,116122), which is considerably higher 50
than the overall thromboembolic rate of 1% to 4% per year
observed for anticoagulated patients with uninfected pros-
thetic valves (123).
Studies comparing the incidence of CNS complications
between NVE and PVE have given conflicting results. In
one study, 11 (13%) of 82 patients with NVE, compared to
6 (33%) of 18 with PVE, presented with such complications 0
(117); however, another study reported 40 (35%) of 113 for

3
4

90

08
4

5
16
13

=8

=5
patients with NVE and 24 (39%) of 62 for those with PVE

n=

=1
n=
n=

n'

n'

n'
(88). Other series did not report any statistically significant
difference regarding the number of NVE and PVE CNS com- Pruitt, 1978 Heiro, 2000 Sonneville, 2011
plications (46,98,124).
It is unclear whether the incidence of embolic events is af- Culture negative Other Enterococci
fected by the time of onset of PVE in relation to the placement Viridans Coagulase-negative S. aureus
of the prosthetic valve. In one study, CNS embolic episodes streptococci staphylococci
were reported in 0% to 11% of patients with early onset PVE
and in 23% to 28% of those with late-onset PVE (120). The FIGURE 34.2 Microbiology of infective endocarditis in patients with-
higher rate in late-onset PVE was confirmed in another study out (n) and with (n) CNS complications.
in which peripheral manifestations occurred in 10% of early
onset PVE cases, compared to 34% in late-onset PVE cases
(125). However, in a recent prospective study of 78 cases of
observed with some other bacteria, particularly group D and
PVE, the rate of CNS complications did not differ between
nongroup D streptococci, which ranged from 25% to 47%
early and late PVE (126).
(83,88,92,97,113).
The incidence of CNS complications in PVE is influenced
Some species of bacteria other than S. aureus, such as
by anticoagulant therapy. In patients insufficiently anticoagu-
Enterobacteriaceae or anaerobic bacteria, have been associ-
lated, CNS complication rates of 38% to 71% were noted,
ated with a high rate of neurologic complications (83,128).
compared to only 8% to 10% in patients on appropriate an-
Moreover, certain microorganisms are prone to a high rate
ticoagulant therapy (116,119). In one series of 61 patients,
of certain types of neurologic involvement. For example, al-
there was no difference in the rate of embolism or risk of
though S. pneumoniae has become rare in IE, causing only
bleeding between patients receiving no anticoagulation or sub-
1% to 3% of cases (129), in these few cases, associated pneu-
therapeutic doses and those who were adequately anticoagu-
mococcal meningitis is common, being found in 40% to 60%
lated (121). When anticoagulated patients develop neurologic
(130,131). Purulent complications are also frequently encoun-
complications, they are at high risk for major hemorrhage
tered with S. aureus IE, either as meningitis or as brain abscess.
(83,120,127).
Certain bacteria such as the Haemophilus species, members of
The type of prosthetic valve may influence the rate of CNS
the genus Abiotrophia (formerly nutritionally variant strepto-
manifestations. Of 33 patients with bioprosthetic valve in-
cocci), or fastidious organisms have been associated with large
fection gathered from three studies, 4 (12%) had neurologic
emboli; these emboli may occlude major vessels including
events (117,120,121), a figure lower than that with mechani-
cerebral arteries. In patients with culture-negative IE, major
cal valves. However, in another study of 62 patients with PVE,
embolic phenomena, including those to the brain, were noted
there was no statistical difference when the type of prosthetic
twice as often as in patients with culture-positive IE. Cases due
valve involved was compared among patients with and with-
to Candida spp. are also associated with large vegetations and
out neurologic complications (88).
a high rate of embolic phenomena (132,133). Of 27 patients

Das könnte Ihnen auch gefallen