Sie sind auf Seite 1von 536

Gebhard Wagener

Editor

Liver Anesthesiology and


Critical Care Medicine

Second Edition

123
Liver Anesthesiology and Critical
Care Medicine
Gebhard Wagener
Editor

Liver Anesthesiology
and Critical Care
Medicine
Second Edition
Editor
Gebhard Wagener
Department of Anesthesiology, Columbia University Medical Center
New York, NY
USA

ISBN 978-3-319-64297-0    ISBN 978-3-319-64298-7 (eBook)


https://doi.org/10.1007/978-3-319-64298-7

Library of Congress Control Number: 2018935600

© Springer International Publishing AG, part of Springer Nature 2012, 2018


This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or
part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way,
and transmission or information storage and retrieval, electronic adaptation, computer software,
or by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in
this book are believed to be true and accurate at the date of publication. Neither the publisher nor
the authors or the editors give a warranty, express or implied, with respect to the material
contained herein or for any errors or omissions that may have been made. The publisher remains
neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Printed on acid-free paper

This Springer imprint is published by the registered company Springer International Publishing
AG part of Springer Nature
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
To Laurie, Ben, and Anna, who allow me to enjoy life and
work, and to my parents, who contributed so much to who I am.
Foreword to the First Edition

Liver transplantation has made remarkable progress in the 48 years since the
first human liver transplant, and especially in the last 30 years, since the intro-
duction of cyclosporine made long-term survival after liver transplantation
feasible.
A procedure that was initially untested and experimental became routine
and is now the accepted treatment for end-stage liver disease in many parts of
the world. About 6000 liver transplants are done in the United States every
year, and graft and patient survival is excellent. We are able to administer
transplants to children, do living related and split liver transplants, and only
the shortage of organs limits the expansion of our field.
This progress is not only due to advances in immunosuppression, surgical
techniques, or organ preservation but also due to improvements in anesthetic
techniques. Anesthesia care initially provided by few experts in a small num-
ber of centers proliferated and is now often standardized and protocolized.
Advances in anesthesiology enabled the development of surgical techniques
such as caval cross-clamp or partial liver transplantation. There are few pro-
cedures in which the close cooperation of surgeon and anesthesiologist is as
essential for the success of the surgery and liver (transplant) surgery would
have never flourished as it did without the teamwork and partnership between
anesthesiologists and surgeons.
Within the last 20 years there has been tremendous progress in clinical
research of liver transplant anesthesia that aims to reduce blood transfusions,
augment organ preservation, and improve overall outcome. Anesthesia for
liver surgery has made a similar astounding progress and now extensive
resections are conceivable that would have been impossible before.
Postoperative critical care medicine as a continuation of the intraoperative
care is now frequently in the hands of anesthesiologists and intensivists spe-
cialized in hepatic intensive care, reflecting the increasing knowledge in this
field.
This book aims to summarize the progress in liver anesthesiology and
critical care medicine of the last 20 years and serves as a guide to those who
care for patients undergoing liver transplantation and liver resections. The
authors are the leaders in the field of liver anesthesiology and critical care in
Europe, Asia, and the United States. The foundation of this book is the
increasing fund of knowledge gained through clinical research as well as
through the extensive clinical experience of the authors that they share with
the readers.

vii
viii Foreword to the First Edition

This textbook provides the necessary background to understand the com-


plexity of the liver and its pathophysiology. It summarizes the elaborate logis-
tics involved in donor and recipient matching in Europe and the United States
and then describes the routine intraoperative management of liver transplant
recipients and patients undergoing hepatic resections. It addresses common
comorbidities and complications and how they may affect the preoperative
work-up and intraoperative management. The postoperative critical care sec-
tion describes the routine care after liver transplantation and resection as well
as diagnosis and management of possible complications including pain
management.
This book aims to summarize our current knowledge of liver anesthesiol-
ogy and critical care. It will serve as a reference for those who routinely care
for patients with liver disease. Those new to our exciting field will gain suf-
ficient knowledge to successfully address many of the complex issues that
may arise during liver anesthesiology and critical care medicine. To those
who have extensive experience in the care of patients undergoing liver (trans-
plant) surgery this book will serve as an authoritative reference and enable an
in-depth immersion into the exciting field of hepatic anesthesiology and criti-
cal care medicine.

Pittsburgh, PA, USA Thomas E. Starzl, MD, PhD (1926–2017)


Preface to the First Edition

Liver transplantation and liver surgery have made enormous strides in the last
20 years. It has been transformed from an often heroic operation requiring
massive amounts of blood transfusions to almost routine surgery with little
blood loss in spite of increasing recipient morbidity. This advancement is
reflected in improved long-term mortality rates in the face of preferentially
allocating more marginal organs to sicker recipients.
Many little steps and advances are responsible for this achievement, not
least improvements of anesthetic techniques and postoperative care. These
little steps may not be immediately obvious but were necessary to accomplish
such a progress. Clinical and preclinical research in liver anesthesiology and
critical care medicine in the last 10 years has thrived, and a new generation of
anesthesiologists and intensive care physicians is willing to scrutinize their
clinical practice using clinical research tools instead of relying only on expe-
rience. This has created a fascinating and productive interaction within the
small group of anesthesiologists and intensivists who care for these severely
sick patients.
This book summarizes their current knowledge by bringing together the
leading experts of our subspecialty. It not only condenses a large amount of
clinical research but also includes opinions and experiences when evidence is
insufficient.
It is an in-depth review of the field and presents the current best knowl-
edge. It aims to be the definitive resource of liver anesthesiology and critical
care medicine. Experienced and busy practitioners will find essential infor-
mation to manage complex conditions of liver disease. The novice anesthesi-
ologist or resident will be able to use this book as a thorough and comprehensive
introduction to our field and rapidly gain extensive knowledge as well as
obtain practical advice for those complex and scary situations that can occur
so frequently during liver transplantation.
This book provides a comprehensive review of the pathophysiology of
liver disease, pharmacology, immunology, and its implications for the anes-
thesiologist and intensivist. Anesthesiologic and postoperative care of liver
transplant recipients requires a thorough appreciation of the intricacies of
liver disease and its complications. Extrahepatic manifestations of liver dis-
ease are addressed in chapters separated by organ systems. Routine manage-
ment as well as common intra- and postoperative complications are described
in detail to provide the knowledge required to care for these patients.

ix
x Preface to the First Edition

Liver transplantation is expanding internationally and a large body of


work and experience originates from centers in Europe and Asia. Experts
from the United States, Europe, and Asia have contributed to this book to give
a global perspective of liver transplant anesthesiology.
A separate section reviews the anesthetic and postoperative management
of patients undergoing liver resection. New surgical approaches have allowed
us to perform more extensive and intricate resections that pose new chal-
lenges to the anesthesiologist and intensivists. Surgical techniques and their
physiologic repercussions are described in detail, and management strategies
for routine as well as complex cases and their possible complications are
offered.
We hope this book will alleviate the apprehension often associated with
caring for these sick patients and encourage many readers to engage in liver
anesthesiology and critical care medicine.

New York, NY, USA Gebhard Wagener, MD


Preface to the Second Edition

The first edition of this book was published six years ago. Since then liver
anesthesiology and critical care medicine has rapidly evolved in pace with
new developments in surgery and transplantation. Laparoscopic and
laparoscopic-­assisted liver surgery that was rarely used before is now routine
in many centers and its use for living donor hepatectomies will greatly
increase acceptance of liver graft donation. Anesthetic management is very
different for this type of surgery, and anesthesiologists need to understand the
risks and benefits of these new technologies. Left lobe living liver donation
for adult recipients is now frequently used and will expand the potential
donor pool and reduce the risk for morbidities for the donor. This would not
have been possible without a better understanding of the regulation of liver
blood flow and improved treatment for early graft dysfunction in the ICU. Pain
procedures have evolved and the use of novel, ultrasound-guided regional
analgesic techniques improved patient comfort and recovery.
The advent of highly successful treatment of hepatitis C with new antiviral
drugs may one day reduce the number of liver transplants. However in the last
six years the need for organs kept rising, resulting in lower quality grafts
assigned to sicker recipients. This greatly complicates the anesthetic and
critical care management of these patients.
Liver anesthesiology and critical care medicine has matured into a sub-
specialty in its own right with national and international societies and meet-
ings. The anesthesiology section of the International Liver Transplant
Society continues to thrive with an annual educational meeting and an
extraordinarily instructive and useful educational website (https://ilts.org/
education/). Independent subspecialty societies such at the Liver Intensive
Care Group of Europe (LICAGE) and the newer Society for the Advancement
of Transplant Anesthesia (SATA) in the United States meet regularly to
share advances in the field, develop guidelines, and facilitate scientific prog-
ress. Many centers now offer fellowships in liver transplant anesthesiology
and societies are currently developing fellowship guidelines to potentially
gain approval by the Accreditation Council for Graduate Medical Education
(ACGME) in the United States.
To reflect these remarkable changes in our field, all chapters in this book
have been revised for this edition. We also added multiple new chapters, for
example, about chronic liver disease, regulation of liver blood flow, evalua-
tion of liver function, and evidence in liver anesthesiology. Among others the

xi
xii Preface to the Second Edition

chapter on pain underwent a major revision and now includes detailed


description of regional analgesic techniques.
We hope that this book remains a useful companion for those who start in
this exciting field as well for the experienced liver anesthesiologist and
intensivist.

New York, NY, USA Gebhard Wagener, MD


Acknowledgments

I sincerely thank the authors of this book for their excellent contributions.
They have spent many hours of diligent and hard work creating delightful,
intelligent, and insightful chapters that were a pleasure to read and edit.
I would also like to thank their families for the time the authors missed with
them while writing these chapters. Dr. Jean Mantz, one of the authors and a
true leader in our field, died last year; I feel privileged to have known such an
outstanding doctor.
This book would not have been possible without the encouragement, sup-
port, and advice of Dr. Margaret Wood who has unwaveringly supported me
throughout my career and all my colleagues and friends at Columbia
University Medical Center. I am immensely grateful to all of you.
I would further like to thank my editors from Springer Science + Business
Media, Asja Parrish, Rebekah Collins, and Saanthi Shankhararaman, who
have been indefatigable and immensely patient with me. Thank you.
Thank you, Taryn Lai and Nina Yoh for your help, insights and advice
with so many aspects of this book (and life!). Also thank you to Tara Richter-
Smith and Erin Hittesdorf who have reviewed and corrected syntax, style and
references of many of these chapters and were essential in finishing this book.
I am sincerely grateful to my colleagues, residents, students, and nurses
that I have had the pleasure to work with for many years and, most ­importantly,
to my patients, who taught me so much about disease, life, and death.

xiii
Contents

Part I Physiology, Pathophysiology and Pharmacology


of Liver Disease

1 Physiology and Anatomy of the Liver. . . . . . . . . . . . . . . . . . . . .    3


Teresa Anita Mulaikal and Jean C. Emond
2 Chronic Liver Failure and Hepatic Cirrhosis . . . . . . . . . . . . . .   21
Lauren Tal Grinspan and Elizabeth C. Verna
3 Acute Hepatic Failure  41
Andrew Slack, Brian J. Hogan, and Julia Wendon
4 The Splanchnic and Systemic Circulation in Liver Disease. . .   63
Nina T. Yoh and Gebhard Wagener
5 Drug Metabolism in Liver Failure. . . . . . . . . . . . . . . . . . . . . . . .   69
Simon W. Lam
6 Evaluation of Liver Function. . . . . . . . . . . . . . . . . . . . . . . . . . . .   79
Vanessa Cowan

Part II Anesthesiology for Liver Transplantation

7 History of Liver Transplantation. . . . . . . . . . . . . . . . . . . . . . . . .   89


John R. Klinck and Ernesto A. Pretto
8 Recipient and Donor Selection and Transplant Logistics:
The European Perspective. . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   101
Gabriela A. Berlakovich and Gerd R. Silberhumer
9 Recipient and Donor Selection and Transplant
Logistics: The US Perspective. . . . . . . . . . . . . . . . . . . . . . . . . . .   109
Ingo Klein, Johanna Wagner, and Claus U. Niemann
10 Surgical Techniques in Liver Transplantation. . . . . . . . . . . . . .   121
Holden Groves and Juan V. del Rio Martin
11 Intraoperative Monitoring. . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   135
Claus G. Krenn and Marko Nicolic

xv
xvi Contents

12 Evidence for Anesthetic Practice in Liver


Transplant Anesthesiology. . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   149
Ryan M. Chadha
13 Caval Cross-Clamping, Piggyback and 
Veno-Venous Bypass. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   155
David Hovord, Ruairi Moulding, and Paul Picton
14 Hemodynamic Changes, Cardiac Output Monitoring
and Inotropic Support . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   163
Anand D. Padmakumar and Mark C. Bellamy
15 Coagulopathy: Pathophysiology, Evaluation,
and Treatment. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   173
Bubu A. Banini and Arun J. Sanyal
16 Physiology, Prevention, and Treatment
of Blood Loss During Liver Transplantation. . . . . . . . . . . . . . .   195
Simone F. Kleiss, Ton Lisman, and Robert J. Porte
17 The Marginal Liver Donor and Organ
Preservation Strategies. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   207
Abdulrhman S. Elnaggar and James V. Guarrera
18 Pediatric Liver Transplantation . . . . . . . . . . . . . . . . . . . . . . . . .   221
Philipp J. Houck
19 Combined Solid Organ Transplantation
Involving the Liver. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   233
Geraldine C. Diaz, Jarva Chow, and John F. Renz
20 Liver Transplantation for the Patient
with High MELD. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .  247
Cynthia Wang and Randolph Steadman
21 Perioperative Considerations for Transplantation
in Acute Liver Failure. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   257
C. P. Snowden, D. M. Cressey, and J. Prentis
22 The Patient with Severe Co-morbidities: Renal Failure. . . . . .   269
Andrew Disque and Joseph Meltzer
23 The Patient with Severe Co-morbidities:
Cardiac Disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   281
Shahriar Shayan and Andre M. De Wolf
24 Pulmonary Complications of Liver Disease. . . . . . . . . . . . . . . .   293
Mercedes Susan Mandell and Masahiko Taniguchi
25 The Patient with Severe Co-morbidities: CNS Disease
and Increased Intracranial Pressure. . . . . . . . . . . . . . . . . . . . . .   307
Prashanth Nandhabalan, Chris Willars, and Georg Auzinger
Contents xvii

Part III Anesthesiology for Liver Surgery

26 Hepatobiliary Surgery: Indications, Evaluation


and Outcomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   333
Jay A. Graham and Milan Kinkhabwala
27 Liver Resection Surgery: Anesthetic Management, Monitoring,
Fluids and Electrolytes. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   349
Emmanuel Weiss, Jean Mantz, and Catherine Paugam-Burtz
28 Anesthetic Aspects of Living Donor Hepatectomy . . . . . . . . . .   367
Paul D. Weyker and Tricia E. Brentjens
29 Complications of Liver Surgery. . . . . . . . . . . . . . . . . . . . . . . . . .   377
Oliver P. F. Panzer
30 The Patient with Liver Disease Undergoing
Non-hepatic Surgery. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   389
Katherine Palmieri and Robert N. Sladen

Part IV Critical Care Medicine for Liver Transplantation

31 Routine Postoperative Care After Liver Transplantation. . . . .   415


Jonathan Hastie and Vivek K. Moitra
32 Immunosuppression. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   431
Enoka Gonsalkorala, Daphne Hotho, and Kosh Agarwal
33 Acute Kidney Injury After Liver Transplantation . . . . . . . . . .   445
Raymond M. Planinsic, Tetsuro Sakai, and Ibtesam A. Hilmi
34 Early Graft Failure. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   451
Avery L. Smith, Srinath Chinnakotla, and James F. Trotter
35 Sepsis and  Infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   455
Fuat Hakan Saner
36 Respiratory Failure and ARDS. . . . . . . . . . . . . . . . . . . . . . . . . .   469
James Y. Findlay and Mark T. Keegan

Part V Critical Care Medicine for Liver Surgery

37 Postoperative Care of Living Donor for Liver Transplant. . . .   485


Sean Ewing, Tadahiro Uemura, and Sathish Kumar
38 Liver Surgery: Early Complications—Liver Failure,
Bile Leak and Sepsis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   497
Albert C. Y. Chan and Sheung Tat Fan
39 Pain Management in Liver Transplantation . . . . . . . . . . . . . . .   507
Paul Weyker, Christopher Webb, and Leena Mathew
Index. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   525
Contributors

Kosh Agarwal, MD Institute of Liver Studies, Kings College Hospital,


London, UK
Georg Auzinger, EDIC, AFICM  Department of Critical Care/Institute of
Liver Studies, King’s College Hospital, London, UK
Bubu A. Banini, MD, PhD  Division of Gastroenterology and Hepatology,
Department of Internal Medicine, Virginia Commonwealth University,
Richmond, VA, USA
Mark C. Bellamy, MA, MB, BS, FRCP(Edin), FRCA, FFICM  St. James’s
University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, West
Yorkshire, UK
Gabriela A. Berlakovich, MD, FEBS Division of Transplantation,
Department of Surgery, Medical University of Vienna, Vienna, Austria
Tricia E. Brentjens, MD Department of Anesthesiology, Columbia
University Medical Center, New York, NY, USA
Catherine Paugam-Burtz, MD, PhD  Department of Anesthesiology and
Critical Care Medecine, Beaujon, HUPNVS, Assistance Publique-Hôpitaux
de Paris (AP-HP), Paris, France
University Paris VII, Paris Diderot, Paris, France
Ryan M. Chadha, MD  Anesthesiology and Perioperative Medicine, Mayo
Clinic Florida, Jacksonville, FL, USA
Albert C. Y. Chan, MBBS, FRCS  Department of Surgery, The University
of Hong Kong, Queen Mary Hospital, Hong Kong, People’s Republic of
China
Srinath Chinnakotla, MD  Department of Surgery, University of Minnesota,
Minneapolis, MN, USA
Jarva Chow, MD Department of Anesthesiology, Loyola University,
Chicago, IL, USA
Vanessa Cowan, MD  Transplant Institute, Beth Israel Deaconess Medical
Center, Boston, MA, USA

xix
xx Contributors

D. M. Cressey, BSc (Hons), MBBS, FRCA  Department of Perioperative


Medicine and Critical Care, Freeman Hospital, Newcastle Upon Tyne, UK
Geraldine C. Diaz, DO Department of Anesthesiology, University of
Illinois at Chicago, Chicago, IL, USA
Andrew Disque, MD Department of Anesthesiology and Perioperative
Medicine, David Geffen School of Medicine, University of California at Los
Angeles, Los Angeles, CA, USA
Abdulrhman S. Elnaggar, MD Department of Surgery, Columbia
University Medical Center, New York, NY, USA
Emmanuel Weiss, MD, PhD  Department of Anesthesiology and Critical
Care Medecine, Beaujon, HUPNVS, Assistance Publique-Hôpitaux de Paris
(AP-HP), Paris, France
University Paris VII, Paris Diderot, Paris, France
Jean C. Emond, MD  Department of Surgery, Columbia University Medical
Center, New York, NY, USA
Sean Ewing, MD  Department of Anesthesiology, University of Michigan
Health System, Ann Arbor, MI, USA
Sheung Tat Fan, MD, PhD, DSc Liver Surgery Centre, Hong Kong
Sanatorium and Hospital, Hong Kong, People’s Republic of China
James Y. Findlay, MB, ChB, FRCA  Department of Anesthesiology and
Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
Enoka Gonsalkorala, MD Institute of Liver Studies, Kings College
Hospital, London, UK
Jay A. Graham, MD  Department of Surgery, Albert Einstein College of
Medicine, Bronx, NY, USA
Lauren Tal Grinspan, MD, PhD Department of Medicine, Columbia
University Medical Center, New York, NY, USA
Holden Groves, MD, MS  Columbia University Medical Center, New York,
NY, USA
Department of Anesthesiology, Columbia University Medical Center, New
York, NY, USA
James V. Guarrera, MD, FACS Division of Liver Transplant and
Hepatobiliary Surgery, Rutgers New Jersey Medical School, University
Hospital, New York, NJ, USA
Jonathan Hastie, MD Department of Anesthesiology, Cardiothoracic
Intensive Care Unit, Columbia University Medical Center, New York, NY,
USA
Contributors xxi

Ibtesam A. Hilmi, MB, ChB  Department of Anesthesiology, University of


Pittsburgh Medical Center, Pittsburgh, PA, USA
Brian J. Hogan, BSc, MBBS, MRCP, FEBTM, FFICM  Institute of Liver
Studies, Kings College London, Kings College Hospital, London, UK
Daphne Hotho, MD Institute of Liver Studies, Kings College Hospital,
London, UK
Philipp J. Houck, MD  Department of Anesthesiology, Columbia University
Medical Center, New York, NY, USA
David Hovord, BA, MB, BChir, FRCA Department of Anesthesiology,
University of Michigan Medical School, Ann Arbor, MI, USA
Jean Mantz, MD, PhD Department of Anesthesiology and Critical Care
Medicine, HEGP, APHP, Paris, France
Mark T. Keegan, MB, MRCPI, MSc, D ABA  Department of Anesthesiology
and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
Milan Kinkhabwala, MD  Department of Surgery, Albert Einstein College
of Medicine, Bronx, NY, USA
Ingo Klein Department of General- and Visceral-, Vascular and Pediatric
Surgery, University of Wuerzburg, Medical Center, Wuerzburg, Germany
Simone F. Kleiss, MD Section of Hepato-Pancreato-Biliary Surgery and
Liver Transplantation, Department of Surgery, University of Groningen,
University Medical Center Groningen, Groningen, The Netherlands
John R. Klinck, MD, FRCA, FRCPC Division of Perioperative Care,
Cambridge University Hospitals, Cambridge, UK
Claus G. Krenn, MD  Department of Anaesthesia, General Intensive Care
and Pain Medicine, Medical University of Vienna, General Hospital Vienna,
Vienna, Austria
Simon W. Lam, PharmD, FCCM  Cleveland Clinic, Cleveland, OH, USA
Ton Lisman, PhD  Section of Hepato-Pancreato-Biliary Surgery and Liver
Transplantation, Department of Surgery, University of Groningen, University
Medical Center Groningen, Groningen, The Netherlands
Mercedes Susan Mandell, MD, PhD Department of Anesthesiology,
University of Colorado, Aurora, CO, USA
Leena Mathew, MD  Department of Anesthesiology, Columbia University
Medical Center, New York, NY, USA
Joseph Meltzer, MD  Division of Critical Care, Department of Anesthesiology
and Perioperative Medicine, David Geffen School of Medicine, University of
California at Los Angeles, Los Angeles, CA, USA
xxii Contributors

Vivek K. Moitra, MD Department of Anesthesiology, Surgical Intensive


Care Unit and Cardiothoracic Intensive Care Unit, Columbia University,
College of Physicians and Surgeons, New York, NY, USA
Ruairi Moulding, BSc, MBBS, FRCA Department of Anaesthesia,
Musgrove Park Hospital, Taunton, UK
Teresa Anita Mulaikal, MD  Division of Cardiothoracic and Critical Care
Medicine, Columbia University Medical Center, New York, NY, USA
Prashanth Nandhabalan, MRCP, FRCA, FFICM, EDIC  Department of
Critical Care/Institute of Liver Studies, King’s College Hospital, London, UK
Marko Nicolic, MD Department of Anaesthesia, General Intensive Care
and Pain Medicine, Medical University of Vienna, General Hospital Vienna,
Vienna, Austria
Claus U. Niemann, MD  Anesthesia and Perioperative Care, University of
California San Francisco, San Francisco, CA, USA
Anand D. Padmakumar, MBBS, FRCA, EDIC, FFICM St. James’s
University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, West
Yorkshire, UK
Katherine Palmieri, MD, MBA Department of Anesthesiology, The
University of Kansas Health System, Kansas City, KS, USA
Oliver P. F. Panzer, MD Department of Anesthesiology, Columbia
University Medical Center, New York, NY, USA
Paul Picton, MB, ChB, MRCP, FRCA Department of Anesthesiology,
University of Michigan Medical School, Ann Arbor, MI, USA
Raymond M. Planinsic, MD  Department of Anesthesiology, University of
Pittsburgh Medical Center, Pittsburgh, PA, USA
Robert J. Porte, MD, PhD  Section of Hepato-Pancreato-Biliary Surgery
and Liver Transplantation, Department of Surgery, University of Groningen,
University Medical Center Groningen, Groningen, The Netherlands
J. Prentis, MBBS, FRCA Department of Perioperative Medicine and
Critical Care, Freeman Hospital, Newcastle Upon Tyne, UK
Ernesto A. Pretto, MD, MPH Division of Transplant and Vascular
Anesthesia, University of Miami Leonard M Miller School of Medicine,
Jackson Memorial Hospital, Miami, FL, USA
John F. Renz, MD, PhD  Section of Transplantation, Department of Surgery,
University of Chicago, Chicago, IL, USA
Juan V. del Rio Martin, MD, FASTS  Hospital Auxilio Mutuo, San Juan,
PR, USA
Tetsuro Sakai, MD, PhD, MHA  Department of Anesthesiology, University
of Pittsburgh Medical Center, Pittsburgh, PA, USA
Contributors xxiii

Fuat Hakan Saner, MD  Department of General-, Visceral- and Transplant


Surgery, Medical Center University Essen, Essen, Germany
Arun J. Sanyal, MD, MBBS  Division of Gastroenterology and Hepatology,
Department of Internal Medicine, Virginia Commonwealth University,
Richmond, VA, USA
Sathish Kumar, MBBS Department of Anesthesiology, University of
Michigan, Ann Arbor, MI, USA
Shahriar Shayan, MD Department of Anesthesiology, Northwestern
Memorial Hospital, Chicago, IL, USA
Gerd R. Silberhumer, MD Division of Transplantation, Department of
Surgery, Medical University of Vienna, Vienna, Austria
Andrew Slack, MBBS, MRCP, EDIC, MD(Res)  Department of Critical
Care, Guy’s and St Thomas’s NHS Foundation Trust, London, UK
Robert N. Sladen, MBChB, MRCP(UK), FRCPC, FCCM  Allen Hyman
Professor Emeritus of Critical Care Anesthesiology at Columbia University
Medical Center, College of Physicians and Surgeons of Columbia University,
New York, NY, USA
Avery L. Smith, MD  Baylor University Medical Center, Dallas, TX, USA
C. P. Snowden, B Med Sci (Hons), FRCA, MD  Department of Perioperative
Medicine and Critical Care, Freeman Hospital, Newcastle Upon Tyne, UK
Randolph Steadman, MD, MS Department of Anesthesiology and
Perioperative Medicine, UCLA Health, Los Angeles, CA, USA
Masahiko Taniguchi, MD, FACS Department of Surgery, Hokkaido
University, Sappora, Japan
James F. Trotter, MD  Baylor University Medical Center, Dallas, TX, USA
Tadahiro Uemura, MD, PhD  Abdominal Transplantation and Hepatobiliary
Surgery, Allegheny General Hospital, Pittsburgh, PA, USA
Elizabeth C. Verna, MD, MS  Transplant Initiative, Division of Digestive
and Liver Diseases, Center for Liver Disease and Transplantation, Columbia
University Medical Center, New York, NY, USA
Gebhard Wagener, MD Department of Anesthesiology, Columbia
University Medical Center, New York, NY, USA
Johanna Wagner Department of General- and Visceral-, Vascular and
Pediatric Surgery, University of Wuerzburg, Medical Center, Wuerzburg,
Germany
Cynthia Wang, MD  Department of Anesthesiology and Pain Management,
VA North Texas Healthcare System, Dallas, TX, USA
Christopher Webb, MD Department of Anesthesiology, Columbia
University Medical Center, New York, NY, USA
xxiv Contributors

Julia Wendon, MbChB, FRCP  Institute of Liver Studies, Kings College


London, Kings College Hospital, London, UK
Paul D. Weyker, MD  Department of Anesthesiology, Columbia University
Medical Center, New York, NY, USA
Chris Willars, MBBS, BSc, FRCA, FFICM  Department of Critical Care/
Institute of Liver Studies, King’s College Hospital, London, UK
Andre M. De Wolf, MD Department of Anesthesiology, Northwestern
Memorial Hospital, Chicago, IL, USA
Nina T. Yoh  Columbia University Medical Center, New York, NY, USA
Part I
Physiology, Pathophysiology and
Pharmacology of Liver Disease
Physiology and Anatomy
of the Liver 1
Teresa Anita Mulaikal and Jean C. Emond

Keywords systemic alterations. Thus, an understanding of


Liver anatomy · Liver segments · External the structure and function of the liver, is critical
anatomy · Embryology · Hepatocytes · Liver for managing the changes in the liver induced
function during surgery. This knowledge, applied through-
out the peri-operative period by anesthesiologists
with interest and training in liver disease, has
Introduction been a major factor in the markedly improved
outcomes of liver surgery during the past
This chapter will review the anatomy and physi- 50 years, and especially since the era of liver
ology of the liver as it pertains to the anesthetic transplantation.
management during liver surgery and transplan- The liver is the largest gland in the human
tation. Anesthetic management of the patient body and the only organ capable of regeneration
with chronic liver disease requires a thorough [1]. This unique ability has been both the subject
understanding of the alterations induced in cir- of ancient Greek mythology and modern medi-
rhosis that affect many organ systems. For exam- cine best illustrated by the myth in which the
ple liver surgery for ablation of tumors may injured liver is restored daily as Zeus’ eternal
reduce the functional mass of the liver resulting punishment to Prometheus. While advances in
in systemic changes that alter hemodynamics and science allow for the temporary support of renal
renal function. In liver transplantation, the body function in the form of dialysis, and of cardiovas-
is deprived of all liver function during the implan- cular and pulmonary function in the form of
tation phase and may receive a new liver with extracorporeal membrane oxygenation (ECMO),
impaired initial function. All types of liver sur- there is currently no effective substitute for the
gery may cause hepatic ischemia and reperfusion immune, metabolic, and synthetic functions of
injury that may induce both acute and chronic the liver other than transplantation (Table 1.1).
The absence of artificial liver support makes a
T. A. Mulaikal, MD (*) thorough understanding of hepatic physiology
Division of Cardiothoracic and Critical Care and pathophysiology imperative to the care of
Medicine, Columbia University Medical Center, critically ill patients with liver injury as their
New York, NY, USA
management requires careful protection of rem-
e-mail: tam36@cumc.columbia.edu
nant function while regeneration occurs.
J. C. Emond, MD
This chapter will review normal liver anatomy,
Department of Surgery, Columbia University Medical
Center, New York, NY, USA histology, and physiology. The first section

© Springer International Publishing AG, part of Springer Nature 2018 3


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_1
4 T. A. Mulaikal and J. C. Emond

covers basic liver anatomy and describes and ventral pancreas while the dorsal foregut
Couinaud’s classification, which divides the liver gives rise to the dorsal pancreas, stomach, and
into eight segments as a function of its portal intestines [5]. The ventral endoderm responds
venous and hepatic arterial supply. These seg- to signals from the cardiac mesoderm to gener-
ments serve as boundaries for the modern day ate the hepatic diverticulum that transforms
hepatectomy. The knowledge of each segment’s into the liver bud and hepatic vasculature [6].
vascular supply, proximity to the vena cava, and The portal vein derives from the vitelline veins
spatial orientation is useful in judging the diffi- [4]. The ductus venosus shunts blood from the
culty of resection and use of surgical techniques umbilical vein, which carries oxygenated blood
such as total vascular isolation to minimize blood from the placenta to the fetus, to the vena cava
loss. For example lesions located posteriorly and thereby supplying oxygenated blood to the
adjacent to the vena cava may necessitate total brain. The ligamentum venosum is the remnant
vascular isolation with broad implications for the of the ductus venosus, and the ligamentum teres
anesthetic management. is the remnant of the umbilical vein.
The next section will comprise basic liver his- The extrahepatic and intrahepatic biliary tracts
tology, including a discussion of microanatomy have different origins. The extrahepatic biliary
and cellular function, which have implications tract, which includes the hepatic ducts, cystic
for the regulation of portal blood flow and the duct, common bile duct, and gallbladder, devel-
pathophysiology of cirrhosis and portal hyperten- ops from the endoderm. The intrahepatic biliary
sion. The last section focuses on basic liver phys- tract, however, develops from hepatoblasts [2].
iology, including the immunological role of the
liver, and its metabolic and synthetic functions.
 acroscopic Anatomy of the Liver
M
and the Visceral Circulation
Embryology
Anatomy relevant to surgical management and
The liver derives from the ventral foregut endo- liver anesthesia includes the blood supply and
derm during the fourth week of gestation, the intrahepatic architecture of the liver. A much
responding to signals from the cardiac meso- more specific knowledge of liver anatomy is
derm for hepatic differentiation [2–4]. The ven- required to plan and execute surgical resections
tral foregut also gives rise to the lung, thyroid, and is beyond the scope of this chapter. The

Table 1.1  Functions of the liver


Metabolic Synthetic Immunologic Regenerative Homeostasis
Xenobiotic metabolism Coagulation factor Innate Restoration after Regulation of
Protein metabolism synthesis immunity hepatectomy or intravascular volume
 Ammonia  Procoagulants Adaptive trauma  Renin-angiogensin-
 Detoxification  Anticoagulants immunity aldosterone
Lipid metabolism  Fibrinolytics Oral and Glucose homeostasis
 B-oxidation F.A.  Antifibrinolytics allograft Regulation of portal
 Triglyceride Plasma protein synthesis tolerance inflow
Glucose metabolism  Albumin  Hepatic arterial
 Gluconeogenesis Steroid hormone synthesis buffer hypothesis
 Glycogenolysis  Cholesterol
 Glygogenesis Thrombopoietin
Angiotensionogen
IGF-1
F.A. Fatty acids
1  Physiology and Anatomy of the Liver 5

afferent blood to the liver is composed of both that the splenic vein contribution to the portal
arterial and portal blood and accounts for 20–25% blood is rich in pancreatic hormones and cyto-
of the cardiac output, and all the blood exits the kines while the superior mesenteric vein brings
liver through the hepatic veins (Fig. 1.1). The nutrients, toxins, and bacteria that are absorbed
hepatic artery is derived from the celiac artery by the GI tract. In situations of increased portal
in most cases but may receive some or all of its vein pressure such as cirrhosis and portal vein
supply from the superior mesenteric artery. The thrombosis, collateral veins known as varices can
artery divides in order to supply the right and left develop. These connections between the portal
lobes and the intraheaptic segments, and the anat- vein and the systemic circulation become
omy includes several variants that are relevant in enlarged and shunt blood away from the liver
hepatic resections and biliary surgery. These vari- (Fig.  1.2). Shunting results in impaired liver
ants do not affect anesthetic management other function, most pronounced in alteration of brain
than the recognition that surgical errors may function discussed later in the chapter. Clinically
result in ischemic injury to segments of the liver. significant varices may result in GI bleeding in
Furthermore, since the biliary tree is primarily the esophagus, stomach and duodenum, as well
supplied by the arterial system, bile duct isch- as the rectum. Other collateral shunts occur in the
emia may result in postoperative complications. retroperitoneum and the abdominal wall, and
The portal blood accounts for the majority of may result in a large amount of porto-systemic
the hepatic blood flow and unites the venous shunting without bleeding but other conse-
return from the entire gastrointestinal (GI) tract quences of impaired portal blood flow. In addi-
with the exception of the rectum that drains into tion to the loss of metabolic ­transformation, the
the iliac vessels. The foregut, including the stom- reticulo-endothelial protective function of the
ach, spleen, pancreas and duodenum drain liver is also bypassed when large shunts are pres-
directly into the portal vein and the splenic vein, ent and may result in bacteremia and sepsis and
while the small intestine and the right colon drain contribute to the hemodynamic alterations of cir-
into the superior mesenteric vein. This means rhosis discussed below.

Inferior vena cava Middle hepatic vein


Right hepatic vein Left hepatic vein
Coronary ligament (cut) Ligamentum venosum
Diaphragm (cut) Falciform ligmament

Branches to caudate lobe Lateral segmental branches


Right lobe of left lobe
segmental branches:
Posterior Medial segmental branches
Anterior of left lobe
Cystic duct and artery
Left hepatic duct and artery
Round ligament of liver
(ligamentum teres hepatis)
Common hepatic duct
Bile duct, hepatic portal vein, and proper hepatic artery
Right hepatic
artery and duct

Fig. 1.1  Arterial and venous circulation of the liver


6 T. A. Mulaikal and J. C. Emond

Fig. 1.2  Sites of Diaphragm Veins of Sappey


collaterals in portal
Splenophreric
hypertension. (With collateral veins
Paraumbilical
permission: Greenfield Esophageal varices
vein of Sappey
textbook of surgery, 5th Spleen
edition, Wolters Kluwer Coronary vein
Health 2016) Umbilicus Omentum
Umbilical vein Abdominal wall
Collateral veins
Gonadal vein
Retroperitoneal
Interior mesenteric vein
veins of Retzius

Epigastric vein
Rectum
Inferior rectal vein

The hepatic veins join at the level of the dia- the liver is defined by the vena cava and the gall-
phragm and enter the right chest and are therefore bladder fossa. The right lobe (segments V–VIII)
exposed to alterations of intrathroacic pressure typically comprises 55–70% of the hepatic tissue
unlike the remainder of the abdominal circula- and is supplied by the right hepatic artery and the
tion. The liver is very sensitive to increases of right portal vein, and is drained by the right
outflow pressure and obstruction of the hepatic hepatic vein. The central plane between the right
veins, for example with Budd-Chiari syndrome and left lobes of the liver is defined by the middle
or right heart failure. Increased hepatic venous hepatic vein. The anatomy of the left lobe is more
pressure can cause hepatic engorgement and complex. An external left lobe is defined by the
severe functional impairment of the liver. During falciform ligament (and is termed by some sur-
liver surgery or transplantation obstruction of the geons as the “left lateral segment”, but consists
hepatic outflow for example by clamping or anatomically of two segments, II and III). The
twisting of the vena cava may result in acute medial portion of the left lobe is morphologically
hemodynamic instability. To avoid hemodynamic described as the quadrate lobe and is actually
collapse as the liver is being manipulated, close segment IV. The left lobe segments are supplied
communication between the surgeon and anes- by the left hepatic artery and portal vein, and
thesiologist is critical. drained by the left and middle hepatic veins. The
Although the external anatomy of the liver has caudate lobe (segment I) is central and supply
been long recognized (Fig. 1.3) [7], the study of and drainage of segment I is fully independent of
corrosion casts of the intrahepatic vessels and the right and left liver lobes.
biliary tree has permitted our current understand-
ing of the intrahepatic anatomy (Fig. 1.4). The
external anatomy is described from gross land- Histology
marks including the gallbladder, the vena cava,
and the hepatic ligaments. The internal anatomy Cellular Classification
is defined by the vascular structures and eight
functionally independent segments. Each seg- The liver is composed of a rich population of
ment has an afferent pedicle that includes artery, specialized cells that allow it to carry out com-
portal vein and bile duct, and efferent hepatic plex functions. They can be grossly character-
vein. From the exterior, the apparent right lobe of ized as “parenchymal” cells (hepatocytes) and
1  Physiology and Anatomy of the Liver 7

Fig. 1.3 External
anatomy of the liver.
Bismuth H. Surgical
anatomy and anatomical
surgery of the liver.
World J Surg. Jan 1982;
6 (1): 3–9
left lobe
right lobe

falciform ligament

teres ligamentum

gallbladder fossa quadrate lobe umbilical fissure

hilus

caudate lobe

“nonparenchymal cells”. The nonparenchymal


cells include stellate cells, sinusoidal endothelial
cells, Kupffer cells, dendritic cells, and lympho-
II
VIII cytes (Fig. 1.5 and Table 1.2). The hepatocytes
or parenchymal cells, make up 60–80% of liver
I
III cells [8] and carry out the metabolic, detoxifi-
cation, and synthetic functions of the liver. The
VII
hepatocytes have a unique relationship with the
IV sinusoidal endothelium that carefully regulates
V
the exposure of hepatocytes to the metabolic
VI substrate that arrives in the portal blood through
fenestrations. The baso-lateral membrane of the
hepatocyte absorbs nutrients from the sinusoids,
which are then processed with excretion of the
Fig. 1.4  Internal anatomy of the liver. Bismuth H. 
Surgical anatomy and anatomical surgery of the liver. metabolic products through the apical cell mem-
World J Surg. Jan 1982; 6 (1): 3–9 brane into the bile duct. Hepatocytes divide under
8 T. A. Mulaikal and J. C. Emond

a b

Fig. 1.5  Hepatic microanatomy. Transmission electron endothelial lining and is seen to contact microvilli of the
micrographs of (a) sinusoidal endothelium (Ec) with parenchymal cells (arrow-heads). Ec endothelial cell, f
attached Kupffer cell (KC) encasing the sinusoid lumen fenestrae, L sinusoidal lumen, N nucleus, SD space of
(L), and perisinusoidal stel-late cell (SC) containing fat Disse (with kind permission from McCuskey [20],
droplets in space of Disse (SD); and (b) Pit cell with typi- Fig. 1.5a, Fig. 6b)
cal dense granules. This Pit cell is in close contact with the

stress and cytokine stimulation and are the princi- underlying mechanism of fibrosis and cirrhosis
pal components of mass restoration during regen- [13]. Hepatic stellate cell secretion of collagen
eration (Table 1.1) [1]. In vitro, hepatic mitotic into the perisinusoidal space of Disse narrows the
activity is stimulated by hepatocyte growth factor sinusoidal lumen, thereby increasing hepatic vas-
(HGF), cytokines, and tumor necrosis factor alpha cular resistance and contributing to portal hyper-
(TNF) and can be clinically seen after hepatec- tension [10]. The impact of this disturbance on
tomy, toxic cell necrosis or trauma [9]. sinusoidal perfusion creates a secondary isch-
Hepatic stellate or Ito cells are vitamin A and emic injury, potentially accelerating the destruc-
fat storing cells located in the perisinusoidal tive impact of an initially limited injury [12].
space of Disse, described by Toshio Ito in 1951 Stellate cells also have intrinsic contractile
[10, 11]. These cells are of tremendous impor- ­function important in the regulation of blood flow
tance and scientific interest as critical regulators and the pathogenesis of portal hypertension.
of hepatic function and prime suspects in the Vasopressin, endothelin-1, and angiotensin II
pathogenesis of cirrhosis. In the normal liver, bind to receptors on stellate cells, activating a rho
stellate cells are quiescent but can become acti- mediated signal transduction pathway and myo-
vated by injury and then transform into collagen sin II contraction [10, 13]. Endothelin-1, angio-
secreting myofibroblasts with contractile proper- tensin II, vasopressin, and their receptors have
ties. This fibroblast-like cellular activity of been studied as therapeutic targets for the treat-
hepatic stellate cells has a protective function in ment of portal hypertension and the management
the generation of scar tissue, promotion of wound of variceal bleeding [14–18].
healing, and remodeling of the extracellular Hepatic endothelial cells are fenestrated cells
matrix [12]. Excessive collagen deposition is the that line the sinusoids and also play an important
1  Physiology and Anatomy of the Liver 9

Table 1.2  Cellular microanatomy


Percentage of
Function Derivation liver cells
Hepatocytes Hepatic regeneration Anterior portion of definitive 60–80%
Xenobiotic metabolism endoderm
Protein synthesis and metabolism
Lipid synthesis and metabolism
APCs
  Innate immunity
Stellate/Ito Vitamin A and fat storage Endoderm 5–15%
cellsv Collagen secreting myofibroblasts or
  Scar tissue and wound healing Septum transversum
  Fibrosis and cirrhosis mesenchyme
Contractile cells
  Regulate vascular resistance
APCs
 Innate immunity
Liver Fenestrated endothelial cells Angiogenesis of existing vessels 15–20%
Sinusoidal Release of nitric oxide (NO) from septum transversum
Endothelial  Regulate vascular resistance mesenchyme
cells APCs
 Innate immunity
Kupffer cells Macrophages Bone marrow 15%
APCs
 Innate immunity
NO, TNF alpha, cytokines
 Ischemia reperfusion injury
Downregulation of APC and T cell activation
mediating tolerance
Dendritic cells APCs Bone marrow <1%
 Innate immunity
Lymphocytes Nonspecific targeting of tumor and viruses Bone marrow 5–10%
 NK  Innate immunity Thymus
 NKT Target lipid antigens Thymus
 T cells  Innate and adaptive immunity Bone marrow
 B cells Cell mediated
 Adaptive immunity
Humoral mediated
 Adaptive immunity
Cholangiocyte Bile duct cells Hepatoblasts → intrahepatic <1%
biliary tree
Ventral endoderm → extrahepatic
biliary tree
Table created from the following publications [8, 10, 12]
APCs (antigen presenting cells), NO (nitric oxide)

role in the regulation of intrahepatic resistance to production in portal hypertension [21]. NO medi-
blood flow through expression endothelial nitric ated increase of splanchnic flow is consistent with
oxide synthase (eNOS) and release of nitric oxide the forward flow theory of portal hypertension that
(NO), a potent vasodilator [19] (Fig. 1.6) [20]. states that portal hypertension is not only due to an
Disruption of sinusoidal endothelial cells in cir- increase in hepatic vascular resistance but also due
rhosis results in a concomitant decrease in the pro- to splanchnic hyperemia [22]. Neoangiogenesis
duction of NO [21]. This is in contrast to the mediated by vascular endothelial derived growth
mesenteric vascular bed that has an increased NO factor (VEGF) also contributes to splanchnic
10 T. A. Mulaikal and J. C. Emond

a b

Fig. 1.6  Electron micrographs of sinusoidal endothelial space of Disse, H hepatic parenchymal cell. (b) Kupffer
cell, hepatic stellate cell, and Kupffer cell. (a) Scanning cell (KC) attached to luminal surface of sinusoidal endo-
electron micrographs of sinusoid illustrating fenestrae thelium by processes that penetrate fenestrae (with kind
organized in clusters as “sieve” plates (arrowheads). SD permission from McCuskey [20], Fig. 5a, Fig. 6b)

hyperemia and the hyperdynamic state of end cells become resident in the liver and function in
stage liver disease [23, 24]. this unique environment as key initiators of
The Kupffer cells are macrophages that reside innate immunity modulating, or in other cases,
in the hepatic sinusoids and constitute 80–90% of activating acute inflammatory responses [28].
the macrophages in the human body [25]. These Though small in number relative to other cell
cells are specialized due to their exposure to high populations in the liver, hepatic lymphocytes
concentrations of endotoxin and oxidative stress play in important role in regulating immune
in the sinusoids and are critical protectors of the defenses within the liver and include natural
systemic circulation from exposure to toxins. killer cells, NKT cells, T lymphocytes, and B
They are part of the innate immune system, lymphocytes. Natural killer (NK) cells are part of
which is the intrinsic host defense system that the innate immune system and are known for
allows nonspecific targeting of foreign antigens, their nonspecific targeting of tumor cells and
in contrast to the adaptive immune system that viruses. NKT cells link the innate and adaptive
allows specific targeting of foreign antigens. immune systems. They are a subpopulation of
There is a close relationship between the regula- lymphocytes with T cell markers and NK cell
tion of blood flow and Kupffer cell macrophage surface receptors. Conventional T and B lympho-
function based on the NO pathway [26] resulting cytes are part of the adaptive immune system and
in a consistent overlap between ischemic and play a role in epitope specific cell and antibody
inflammatory injury to the liver. mediated destruction of foreign antigens.
Hepatic dendritic cells are antigen presenting
cells synthesized in the bone marrow that can
migrate from the liver to lymphoid tissue, though  natomic Lobules and Metabolic
A
they are often localized near the central vein [27]. Zones
They serve a critical role in antigen presentation
and activation of T lymphocytes when encounter- The microscopic anatomy of the liver can be con-
ing an antigen. A sub-population of dendritic ceptualized either as morphologically anatomic
1  Physiology and Anatomy of the Liver 11

hepatic lobules, or functionally, as precise meta- III is also the site of drug detoxification, or phase
bolic zones. The hexagonal hepatic lobule is cen- I and II metabolism [32].
tered around the central vein with the portal triad
(hepatic artery, portal vein, and common bile
duct) at each corner of the hexagon. The central Immunological Function
vein is the terminal branch of the hepatic vein of the Liver
[29]. These microscopic ordered aggregrations of
liver cells are complete and independent units of Innate and Adaptive Immunity
metabolic capacity that recapitulate on a tiny
scale the entire liver. The hepatic artery and por- The liver is an integral part of both the innate and
tal vein travel together and transport blood con- adaptive immune systems. The innate immune
taining oxygen and splanchnic metabolites to the system is the intrinsic host defense system that
liver that the functional hepatocytes in the hepatic allows nonspecific targeting of foreign antigens.
lobule then process and drain into a common cen- Of the nonparenchymal cells in the liver, there
tral vein. Bile from each hepatocyte drains into are four types of antigen presenting cells (APCs)
canaliculi. These canaliculi join to form the duct- that function as immunologic gatekeepers,
ules that aggregate to form the inter-lobular bile engulfing bacteria that enter the portal system
ducts and eventually the macroscopic segmental from the splanchnic circulation, presenting anti-
ducts. Segmental ducts bring bile to the common genic epitopes to effector T and B lymphocytes
bile duct that drains into the gallbladder and duo- and preventing bacterial entry into the systemic
denum. A more functional histologic classifica- circulation. These four APCs are Kupffer cells,
tion of the liver defines metabolic zones that form dendritic cells, stellate cells, and sinusoidal endo-
the hepatic acinus [30, 31]. Zone I is known as thelial cells and are all part of the innate immune
the periportal zone and is centered around the system.
portal triad, making it oxygen rich given its prox- The innate immune system also includes natu-
imity to the hepatic artery. This periportal zone is ral killer (NK) cells and natural killer T (NKT)
the most resilient to hemodynamic stressors, least cells. NK cells are considered lymphocytes
susceptible to necrosis, and the first to regenerate. because they derive from the bone marrow. NK
The cells in zone I also have distinct metabolic cells play a role in the destruction of tumors, bac-
capacity and focus on aerobic functions of the teriae, viruses and parasites by killing cells that
liver such as gluconeogenesis and glycogenoly- lack ‘self’ major histocompatibility complex I
sis, generating a fuel source for the body’s extra- (MHC I) markers [25]. They secrete cytokines
hepatic work [31–33]. Zone I also is the site of that inhibit viral replication and do not require
cholesterol synthesis and beta oxidation of fatty antigen presenting cells to identify their targets
acids. It is active in the degradation of amino [34]. They release granules with perforin that
acids in the urea cycle, which is responsible for puncture cell membranes and granzymes that
the majority of ammonia metabolism in the body lyse internal cellular contents, thereby inducing
[31, 32]. While enzymes involved in this peripor- apoptosis of the infected cell. NK cells typically
tal zone are expressed throughout the acinus, constitute up to 30–50% of liver lymphocytes,
they are metabolically most active in zone I. Zone but may comprise up to 90% of total lympho-
II is the intermediate zone between zones I and cytes in patients with hepatocellular carcinoma.
III. Zone III is the pericentral or perivenous zone Diminished function of NK cells has been associ-
and is in close proximity to the central vein. This ated with increased tumor burden [25, 34]. NKT
zone has the lowest oxygen tension (PaO2), is cells link the innate and adaptive immune sys-
most susceptible to hemodynmamic stressors, tems. They are a subpopulation of lymphocytes
and the last to regenerate. Zone III is involved in with NK cell surface receptors and T cell markers
ketogenesis, which generates ketone bodies for [25]. NKT cells target lipid antigens such as gly-
extrahepatic tissues during fasting states. Zone colipids of mycobacterial cell walls [35, 36].
12 T. A. Mulaikal and J. C. Emond

The adaptive immune system is the acquired This concept of tolerance describes the liver’s
host defense system that allows epitope specific ability to downregulate T cell activation or ‘toler-
cell and antibody mediated destruction of foreign ate’ antigens that present no harm. Tolerance is
antigens, utilizing memory for fighting subse- mediated by cytokines such as TNF alpha and
quent infections. Adaptive immunity comprises interleukin 10 (IL-10). Kupffer cells release these
both cellular and humoral immunity. Members of cytokines, which in turn downregulate the activ-
the liver’s adaptive immune system include con- ity of antigen presenting dendritic and sinusoidal
ventional T and B lymphocytes involved in cell epithelial cells, thereby decreasing T cell activa-
mediated and antibody mediated immunity tion [8]. Tolerogenicity is important in liver
respectively. T lymphoctyes such as CD8 T cells transplantation and may explain why donor leu-
can recognize tumor-associated antigens (TAA) kocytes can improve hepatic allograft survival
and eradicate cells of hepatocellular carcinomas [44].
(HCC) [37]. The liver is exposed to antigens from The mechanism underlying enteric tolerance
the enteric system that enter the portal circulation associated with the liver may be mediated by
and its adaptive immune system is critical in pro- lipopolysaccharide (LPS) endotoxin, a cell wall
tecting the body from exposure of these antigens component of gram negative bacteria [45]. The
to the systemic circulation. In contrast to the cel- portal vein delivers antigens to the liver often in
lular composition in the peripheral circulation, the form of lipopolysaccharide (LPS), which
the hepatic circulation has a predominance of complexes with toll-like receptor 4 (TLR 4) and
nonspecific innate immune cells I as it functions its coreceptors MD 2 and CD 14 on antigen pre-
as an immunologic gatekeeper, regulating the senting cells. The constituitive exposure of LPS
passage of antigens from the splanchnic to the to these antigen presenting cells is thought to
portal and finally to the systemic circulation [38]. result in a dampening of the immune response or
tolerance [45, 46].

Oral and Allograft Tolerance


Hepatic Drug Metabolism
The liver strikes a balance between immunity to
infection and tolerance of commensal bacteria First Pass Metabolism
and orally consumed antigens, a concept known
as oral or systemic tolerance [39]. This immuno- Drugs administered intravenously have 100%
logic adaptation may underlie the physiologic bioavailability because the original form of the
mechanism of allograft tolerance, the transplan- drug reaches the systemic circulation unchanged.
tation of organs between the same species of Drugs ingested orally, however, undergo first
varying genotypes. In 1960 Peter Medawar won pass metabolism. The intestines and liver absorb
the Nobel Prize in Physiology or Medicine for and process drugs thereby decreasing the effec-
describing the tolerance of skin grafts between tive dose that enters systemic circulation. Drugs
dizygotic twin cattle [40, 41]. This observation with a high bio-availability are minimally metab-
was thought to be due to the in utero exposure of olized by enzymes of the entero-hepatic system.
each twin to erythrocytes of the other [42]. In contrast, drugs with a low bioavailability are
Animal models of porcine allogenic transplanta- extensively metabolized by entero-hepatic
tion illustrate the ability to transplant livers enzymes. Drugs that undergo extensive first pass
though not kidneys, between unrelated pigs with- metabolism are particularly susceptible to fluc-
out rejection [43]. Pigs, mice, and rats will accept tuations in blood levels if their enzymatic metab-
unrelated livers without immunosuppressive olism is altered by co-ingestants [47]. Age and
therapy and some human liver transplant recipi- sex can affect the metabolism and bioavailability
ents can wean their immunosuppressive regimen of drugs as well. Alcohol is metabolized by both
over time [28]. alcohol dehydrogenase (ADH) in the liver and
1  Physiology and Anatomy of the Liver 13

gastrointestinal tract, and cytochrome P450 2E1 injury. Acetaminophen toxicity for example
enzymes. Women have higher blood ethanol con- occurs because of the relative depletion of gluta-
centrations than men who ingest equal amounts thione and the accumulation of N-acetyl-p-­
due to decreased gastric ADH that reduces first- benzoquinone-imine (NAPQI), the unconjugated
pass metabolism and increases bioavailability toxic acetaminophen byproduct. The accumula-
[48, 49]. Increased age decreases overall cyto- tion of NAPQI leads to zone III or centrilobular
chrome P450 activity, increasing the risk of older necrosis. Chronic alcohol use can increase the
individuals for drug induced liver injury (DILI), risk of acetaminophen toxicity due to induction
with particular susceptibility to amoxicillin-cla- of cytochrome P450 2E1 (CYP2E1), which
vulanate, isoniazid, and nitrofurantoin. This sus- increases the conversion of alcohol to its toxic
ceptibility has not resulted in an increased rate of metabolite NAPQI [48]. N-acetylcysteine, a pre-
transplantation or death [50]. cursor to glutathione and a free radical scavenger,
may be beneficial in the treatment of acetamino-
phen toxicity [53]. Some studies have also sug-
Phase II and III Metabolism, Phase 0 gested its use to ameliorate ischemia reperfusion
and III Transport injury, primary graft dysfunction, and acute kid-
ney injury in liver transplantation [54, 55]. These
The enzymes involved in drug metabolism in the findings, however, are controversial and not all
liver are part of the P450 cytochrome family studies have proven definitive benefit of
located in the metabolic zone III. Cytochrome N-acetylcysteine in the perioperative transplant
P450s catalyze phase I reactions. Phase I reac- setting [56].
tions are oxidation, reduction, and hydrolysis Phase 0 and III transport involve carrier medi-
reactions that increase the polarity of substances ated uptake and elimination of drugs by trans-
for excretion or for further metabolism by phase porters via the basolateral and canalicular
II enzymes [51]. Phase II enzymes, such as uri- membranes respectively. In phase 0 transport,
dine diphosphate glucuronosyl transferases drugs are absorbed from the blood into the hepa-
(UGTs), sulfotransferases, and glutathione-­ S-­ tocytes by solute carrier (SLC) transporters [57].
transferases, conjugate phase I metabolites to In phase III transport, the hydrophilic substances
substances such as glucuronate, sulfate, and glu- derived from Phase II metabolism must travel via
tathione [51]. These conjugation reactions trans- the lipid soluble canalicular membranes using
form drugs into hydrophilic substances, thereby ATP binding cassette carriers (ABC) [58]. ATP
increasing their solublility in bile and blood for splitting is required to transport these hydrophilic
excretion. Absence or dysfunction of these phase substances through the lipophilic canalicular
I or II enzymes can result in hyperbilirubinemia membrane into the bile [59]. No metabolism or
and encephalopathy. drug alteration is involved in this transport mech-
Gene mutations may affect metabolism and anism and therefore the term “Phase III metabo-
result in specific syndromes. For example in lism” is a misnomer.
Gilbert’s syndrome a mutation in the promoter
region of bilirubin-UGT leads to decreased levels
of normally functioning enzyme, thereby reduced Substrates, Inducers and Inhibitors
conjugation of bilirubin with glucoronide, and an of P450 System: Implications
unconjugated hyperbilirubinemia. In Crigler-­ for Toxicity and Therapeutic Failure
Najjar syndrome a mutation of the coding region
of bilirubin-UGT results in absent or defective Many commonly used drugs in the clinical set-
bilirubin-UGT, unconjugated hyperbilirubine- ting interact with P450 enzyme substrates either
mia, and in some cases kernicterus [52]. as inhibitors or inducers. Inhibitors slow down
Similarly, depletion of molecules involved in P450 enzyme activity, thereby increasing the
these conjugation reactions can result in liver substrate bioavailability. This can result in drug
14 T. A. Mulaikal and J. C. Emond

toxicity, which has profound implications for states by placing them in close proximity to the
medications with a narrow therapeutic index. For oxygen and nutrient rich environment of the por-
example warfarin is a P450 substrate and initiat- tal triad [31]. The precise regulation of glucose
ing treatment with inhibitors such as azoles, mac- homeostasis is clinically relevant in that hypogly-
rolides, beta blockers, calcium channel blockers, cemia is the most dramatic manifestation of liver
amiodarone, and proton pump inhibitors may failure and generally implies a terminal state of
lead to a supra-therapeutic INR and clinically hepatic failure.
significant bleeding. Inherited disorders of glucose and glycogen
Conversely, initiating treatment with a P450 metabolism are known as glycogen storage dis-
inducer such as phenobarbital, phenytoin, fos- eases (GSDs). These are enzymatic defects
phenytoin, carbamazepine, or rifampin, may affecting the liver and muscles, the two main sites
cause therapeutic failure. Women taking oral for glycogen storage [62]. Hepatic manifesta-
contraceptives and anti-epileptic drugs (AEDs) tions of GSDs are characterized by fasting hypo-
that are P450 inducers must be cognizant of glycemia, ketosis, and hepatomegaly [63, 64].
the estrogen and progesterone composition of
their oral contraceptives to avoid therapeutic
failure [60].  rotein Metabolism and Hepatic
P
Encephalopathy

 epatic Glucose, Amino Acid,


H When the body has sufficient protein stores, the
and Lipid Metabolism liver transforms additional amino acids to ammo-
nia in the urea cycle. Ammonia detoxification
Glucose Homeostasis involves the degradation of proteins to their
amino acid components, the breakdown of amino
The liver has the ability to produce glucose dur- acids to alpha ketoacids and ammonia, and the
ing fasting states to maintain euglycemia, and generation of urea. This process occurs in the
provide energy for brain, muscle and red blood oxygen rich periportal zone I. The enzyme gluta-
cells. Initial fasting conditions trigger the release mine synthetase located in the perivenous zone
of glucagon from the pancreas, thereby promot- III, then transforms ammonia and glutamate to
ing glycogenolysis, the release of glucose from glutamine. Liver dysfunction of any etiology
stored glycogen [61]. Epinephrine stimulates results in hyperammonemia from both a
glycogenolysis during states of stress. Prolonged decreased ability to produce urea and glutamine,
fasting or starvation prompts the de novo synthe- and diminished first pass metabolism from porto-
sis of glucose by gluconeogenesis. The liver is systemic shunts [65]. Ammonia is neurotoxic, as
the main site of gluconeogenesis, the synthesis of is the excitatory neurotransmitter glutamate when
glucose from pyruvate, lactate, glycerol, and present in excess [65]. Cerebral astrocytes can
amino acids (non-carbohydrate precursors). Both convert some ammonia to glutamine but supra-
gluconeogenesis and glycogenolysis take part in physiologic levels of glutamine result in an
the periportal metabolic zone I of the liver, the osmotic intracellular gradient and subsequent
zone closest to the portal triad. edema, elevated intracranial pressure, and at its
During nonfasting states the liver is able to worst herniation [65]. This is the basis of the
store glucose by glycogenesis or convert glucose ammonia-glutamine hypothesis of intracranial
to pyruvic acid and ATP by glycolysis. These hypertension in fulminant hepatic failure [66].
processes take place in metabolic zone III, or the Alternatively, some scientists have advocated the
pericentral zone. This zonal heterogeneity or dif- “Trojan horse” hypothesis to explain the mecha-
ferential expression of metabolic enzymes priori- nism of cellular edema. Glutamine acts as a car-
tizes crucial metabolic functions that provide rier or Trojan horse for the uptake of ammonia
energy or glucose to the body during fasting from the astrocyte cytoplasm to the
1  Physiology and Anatomy of the Liver 15

mitochondria. The glutamine-derived ammonia (NASH) [72]. Close to half of patients with NASH
within the mitochondria of astrocytes then gener- develop fibrosis and one sixth develop cirrhosis
ates free radicals and causes cellular edema [67]. that may eventually lead to liver failure requiring
There are two types of cerebral edema: cyto- transplantation [75].
toxic edema that results from cellular swelling
due to an increase in osmotic load and intracellu-
lar water absorption, and vasogenic edema from Liver Coagulation and Fibrinolysis
the increased permeability of solutes and solvents
through a disrupted blood brain barrier [68]. The The liver is a major organ involved in hemostasis
cerebral edema of fulminant hepatic failure is pre- since it is the primary synthetic site of pro-­
dominantly cytotoxic with a preserved blood coagulants, anticoagulants, fibrinolytics, and
brain barrier, and a therapeutic response to antifibrinolytics [76]. While extra-hepatic sites
osmotic diuretics such as mannitol and hypertonic such as the endothelium contribute to synthesis
saline [68, 69]. Intracranial hypertension is rare in of some coagulation factors such as factor VIII
chronic liver failure due to a compensatory intra- and von Willebrand factor (vWF), the liver
cellular increase in solute load. remains the principal synthetic site of coagula-
tion cascade components. Primary and secondary
hemostasis requires the formation of a platelet
 ipid Metabolism and Nonalcoholic
L plug and fibrin clot, triggered by tissue trauma or
Fatty Liver Disease endothelial damage [77]. While platelets are
made in the bone marrow, they are often seques-
The liver is the principal site of lipid metabolism, tered in the spleen of patients with portal hyper-
both in absorption of dietary fats and their de novo tension and splenomegaly [78]. This platelet
synthesis. Dietary fats are emulsified by bile salts sequestration contributes to thrombocytopenia in
and absorbed in the form of micelles by the intes- patients with end stage liver disease. Impaired
tine and delivered to the liver via enterohepatic hepatic synthesis of thrombopoietin, the hormone
circulation. Fatty acids can be hydrolyzed by that stimulates megakaryocyte production, also
beta-oxidation to generate energy or ATP for the contributes to thrombocytopenia in liver disease.
body’s extra-hepatic metabolism. During fasting Bone marrow suppression secondary to alcohol,
states, starvation, or diabetic keto-­acidosis (DKA) viruses, and medications is also a factor [79].
when glucose is not available to the body, the liver The liver synthesizes fibrinogen (factor I),
can generate ketone bodies (acetoacetic acid, beta prothrombin (factor II), factor V, and factors VII–
hydroxybutyric acid, and acetone) from fatty XIII. It also synthesizes anticoagulants such as
acids that can be used by organs such as the brain antithrombin III, protein C, protein S, selected
[70]. Conversely, in non-alcoholic fatty liver dis- fibrinolytics such as plasminogen, and antifibri-
ease (NAFLD) when hepatic lipid content or ste- nolytics such as alpha 2-anitplasmin and throm-
atosis constitutes 5% of liver weight, there is an bin activatable fibrinolysis inhibitor (TAFI) [76].
increase in triglyceride synthesis and defective The balance between pro-coagulants and antico-
insulin mediated inhibition of lipolysis [71, 72]. agulants in liver failure determines the risk of
Metabolic syndrome, defined as visceral obesity bleeding or thrombosis. In end stage liver dis-
associated with hypertension, dyslipidemia, and ease, the balance may be tipped towards antico-
hyperglycemia may also be associated with non- agulant and fibrinolytic factors predisposing
alcoholic fatty liver disease by a similarly patients to bleeding, though cases of venous
impaired insulin mediated inhibition of lipolysis thrombosis can occur secondary to venous stasis
[73, 74] This metabolic derangement of lipid or hepatocellular carcinoma [80]. Traditional
metabolism has striking clinical implications laboratory makers of coagulopathy such as pro-
since NAFLD is the most prevalent liver disease thrombin time (PT) and partial thromboplastin
and can progress to non-­alcoholic steatohepatitis time (PTT) do not accurately portray the balance
16 T. A. Mulaikal and J. C. Emond

between procoagulant and anticoagulant factors Thrombopoietin is a peptide hormone pro-


in liver disease. PT and PTT reflect the degree to duced in the liver that stimulates megakaryocytes
which procoagulants factors are depressed but and platelet production. Low levels of thrombo-
not whether anticoagulants such as protein C can poietin in liver failure may contribute to throm-
offset this deficiency since reagents used in these bocytopenia since these levels as well as platelet
laboratory assays do not contain enough throm- counts are restored with orthotopic liver trans-
bomodulin to activate protein C [81]. plantation [87, 88].
Hyperfibrinolysis has traditionally been asso- Angiotensinogen, the precursor of angioten-
ciated with chronic liver disease as demonstrated sin, is produced in the liver as well. This precur-
by elevated levels of tissue plasminogen activator sor peptide hormone is activated by renin in the
(tPA) and plasmin, both involved in the degrada- renin-angiotensin-aldosterone pathway. This
tion of fibrin clots, as well as decreased levels of pathway is the target of anti-hypertensives such
alpha 2 plasminogen inhibitor and thrombin acti- as ACE inhibitors and angiotensin receptor
vatable fibrinolysis inhibitor (TAFI) [77, 82]. blockers (ARBs). The diuretic spironolactone,
Thirty to forty percent of patients with liver dis- which antagonizes the pathway’s endpoint aldo-
ease have laboratory evidence of hyperfibrinoly- sterone, is used to manage ascites in liver
sis. Whether or not these markers of fibrinolysis disease.
correlate with a clinical bleeding risk remains Lastly, the liver is the site of cholesterol syn-
less clear [83, 84]. thesis and therefore crucial in the genesis of
Other factors that can contribute to clinically endogenous steroid hormones such as cortisol,
significant bleeding include renal failure with aldosterone, and testosterone. While these hor-
platelet dysfunction, portal hypertension, endo- mones are synthesized in the adrenal gland, their
toxemia with fibrinolysis, and disseminated intra- precursors are hepatic in origin. Estrogens and
vascular coagulation [83, 84]. Patients with androgens have receptors in hepatocytes that reg-
isolated hepatic coagulopathy usually have nor- ulate lipid and glucose homeostasis [88].
mal to elevated levels of factor VIII and von
Willebrand factor in contrast to patients with
DIC, though both conditions may coexist [77].
Endotoxemia is associated with both fibrinolysis References
and a procoagulant state. Sepsis induced hyper-
1. Ankoma-Sey V. Hepatic regeneration-­ revisiting
coagulability occurs by the inhibition of activated the myth of prometheus. News Physiol Sci.
protein C and S, as well as by increased tissue 1999;14:149–55.
factor expression [85]. 2. Lemaigre FP. Mechanisms of liver development: con-
cepts for understanding liver disorders and design
of novel therapies. Gastroenterology. 2009;137(1):
62–79.
Hepatic Endocrine Function 3. Kaestner KH. The making of the liver: developmental
competence in foregut endoderm and induction of the
hepatogenic program. Cell Cycle. 2005;4(9):1146–8.
The liver acts as an endocrine organ, producing
4. Collardeau-Frachon S, Scoazec JY. Vascular develop-
hormones such as insulin like growth factor ment and differentiation during human liver organo-
(IGF-1), thrombopoietin, angiotensinogen, and genesis. Anat Rec (Hoboken). 2008;291(6):614–27.
steroid hormones. The liver produces 75% of 5. Tanimizu N, Miyajima A. Molecular mechanism of
liver development and regeneration. Int Rev Cytol.
IGF-1, which is a peptide hormone, mediating
2007;259:1–48.
the effects of human growth hormone (GH). 6. Zhao R, Duncan SA. Embryonic development of the
Growth hormone activates the release of IGF-1, liver. Hepatology. 2005;41(5):956–67.
which stimulates tissue growth. Levels rise dur- 7. Bismuth H. Surgical anatomy and anatomical surgery
of the liver. World J Surg. 1982;6(1):3–9.
ing puberty, are abnormally high in conditions
8. Racanelli V, Rehermann B. The liver as an immu-
such as acromegaly, and may be low in patients nological organ. Hepatology. 2006;43(2 Suppl 1):
with short stature. S54–62.
1  Physiology and Anatomy of the Liver 17

9. Michalopoulos GK. Liver regeneration after partial 26. Teoh NC, Farrell GC. Hepatic ischemia reperfu-

hepatectomy: critical analysis of mechanistic dilem- sion injury: pathogenic mechanisms and basis
mas. Am J Pathol. 2010;176(1):2–13. for hepatoprotection. J Gastroenterol Hepatol.
10. Reynaert H, Thompson MG, Thomas T, Geerts
2003;18(8):891–902.
A. Hepatic stellate cells: role in microcirculation 27. Cataldegirmen G, Zeng S, Feirt N, et al. RAGE limits
and pathophysiology of portal hypertension. Gut. regeneration after massive liver injury by coordinated
2002;50(4):571–81. suppression of TNF-alpha and NF-kappaB. J Exp
11. Ito T. Cytological studies on stellate cells of Kupffer Med. 2005;201(3):473–84.
and fat-storing cells in the capillary wall of human 28. Lau AH, Thomson AW. Dendritic cells and immune
liver. Acta Anat Jpn. 1951;26(42):42–74. regulation in the liver. Gut. 2003;52(2):307–14.
12. Friedman SL. Hepatic stellate cells: protean, multi- 29. Gordillo M, Evans T, Gouon-Evans V. Orchestrating
functional, and enigmatic cells of the liver. Physiol liver development. Development. 2015;142(12):
Rev. 2008;88(1):125–72. 2094–108.
13. Soon RK Jr, Yee HF Jr. Stellate cell contraction: role, 30. Katz NR. Methods for the study of liver cell heteroge-
regulation, and potential therapeutic target. Clin Liver neity. Histochem J. 1989;21(9–10):517–29.
Dis. 2008;12(4):791–803. viii 31. Gebhardt R. Metabolic zonation of the liver: regula-
14. De Gottardi A, Shaw S, Sagesser H, Reichen J. Type tion and implications for liver function. Pharmacol
A, but not type B, endothelin receptor antagonists sig- Ther. 1992;53(3):275–354.
nificantly decrease portal pressure in portal hyperten- 32.
Katz NR. Metabolic heterogeneity of hepato-
sive rats. J Hepatol. 2000;33(5):733–7. cytes across the liver acinus. J Nutr. 1992;122(3
15. Merigan TC Jr, Plotkin GR, Davidson CS. Effect of Suppl):843–9.
intravenously administered posterior pituitary extract 33. Hepatic zonation of carbohydrate metabolism. Nutr
on hemorrhage from bleeding esophageal varices. A Rev. 1989;47(7):219–21.
controlled evaluation. N Engl J Med. 1962;266:134–5. 34. Yoon JC, Yang CM, Song Y, Lee JM. Natural

16. Chojkier M, Groszmann RJ, Atterbury CE, et al. A con- killer cells in hepatitis C: current progress. World J
trolled comparison of continuous intraarterial and intra- Gastroenterol. 2016;22(4):1449–60.
venous infusions of vasopressin in hemorrhage from 35. Weiler-Normann C, Rehermann B. The liver as

esophageal varices. Gastroenterology. 1979;77(3):540–6. an immunological organ. J Gastroenterol Hepatol.
17. Schneider AW, Kalk JF, Klein CP. Effect of losartan, 2004;19(7):279–83.
an angiotensin II receptor antagonist, on portal pres- 36. Bandyopadhyay K, Marrero I, Kumar V. NKT cell
sure in cirrhosis. Hepatology. 1999;29(2):334–9. subsets as key participants in liver physiology and
18. Gonzalez-Abraldes J, Albillos A, Banares R, et al. pathology. Cell Mol Immunol. 2016;13(3):337–46.
Randomized comparison of long-term losartan versus 37. Shuai Z, Leung MW, He X, et al. Adaptive immunity
propranolol in lowering portal pressure in cirrhosis. in the liver. Cell Mol Immunol. 2016;13(3):354–68.
Gastroenterology. 2001;121(2):382–8. 38. Sheth K, Bankey P. The liver as an immune organ.
19. Shah V, Haddad FG, Garcia-Cardena G, et al. Liver Curr Opin Crit Care. 2001;7(2):99–104.
sinusoidal endothelial cells are responsible for nitric 39. Crispe IN. The liver as a lymphoid organ. Annu Rev
oxide modulation of resistance in the hepatic sinu- Immunol. 2009;27:147–63.
soids. J Clin Invest. 1997;100(11):2923–30. 40. Anderson D, Billingham RE, Lampkin GH, Medawar
20. McCuskey RS. The hepatic microvascular system
PB. The use of skin grafting to distinguish between
in health and its response to toxicants. Anat Rec monozygotic and dizygotic twins in cattle. Heredity.
(Hoboken). 2008;291(6):661–71. 1951;5(3):379–97.
21. Wiest R, Groszmann RJ. The paradox of nitric oxide 41.
Billingham RE, Lampkin GH, Medawar PB,
in cirrhosis and portal hypertension: too much, not Williams HL. Tolerance to homografts, twin diagno-
enough. Hepatology. 2002;35(2):478–91. sis, and the freemartin condition in cattle. Heredity.
22.
Groszmann RJ, Abraldes JG. Portal hyperten- 1952;6(2):201–12.
sion: from bedside to bench. J Clin Gastroenterol. 42. Owen RD. Immunogenetic consequences of vas-

2005;39(4 Suppl 2):S125–30. cular anastomoses between bovine twins. Science.
23. Fernandez M, Mejias M, Angermayr B, Garcia-
1945;102:400–1.
Pagan JC, Rodes J, Bosch J. Inhibition of VEGF 43. Calne RY, Sells RA, Pena JR, et al. Induction of
receptor-­2 decreases the development of hyperdy- immunological tolerance by porcine liver allografts.
namic splanchnic circulation and portal-systemic col- Nature. 1969;223(5205):472–6.
lateral vessels in portal hypertensive rats. J Hepatol. 44. Thomson AW, Lu L. Are dendritic cells the key to liver
2005;43(1):98–103. transplant tolerance? Immunol Today. 1999;20(1):
24. Moreau R. VEGF-induced angiogenesis drives col- 27–32.
lateral circulation in portal hypertension. J Hepatol. 45. Crispe IN, Giannandrea M, Klein I, John B, Sampson
2005;43(1):6–8. B, Wuensch S. Cellular and molecular mechanisms of
25. Gao B, Jeong WI, Tian Z. Liver: an organ with pre- liver tolerance. Immunol Rev. 2006;213:101–18.
dominant innate immunity. Hepatology. 2008;47(2): 46. Grakoui A, Crispe IN. Presentation of hepatocellular
729–36. antigens. Cell Mol Immunol. 2016;13(3):293–300.
18 T. A. Mulaikal and J. C. Emond

47. Wilkinson GR. Drug metabolism and variability


64. Roscher A, Patel J, Hewson S, et al. The natural his-
among patients in drug response. N Engl J Med. tory of glycogen storage disease types VI and IX:
2005;352(21):2211–21. long-term outcome from the largest metabolic center
48. Chan LN, Anderson GD. Pharmacokinetic and phar- in Canada. Mol Genet Metab. 2014;113(3):171–6.
macodynamic drug interactions with ethanol (alco- 65. Lemberg A, Fernandez MA. Hepatic encephalopathy,
hol). Clin Pharmacokinet. 2014;53(12):1115–36. ammonia, glutamate, glutamine and oxidative stress.
49. Frezza M, di Padova C, Pozzato G, Terpin M, Baraona Ann Hepatol. 2009;8(2):95–102.
E, Lieber CS. High blood alcohol levels in women. 66. Scott TR, Kronsten VT, Hughes RD, Shawcross

The role of decreased gastric alcohol dehydrogenase DL. Pathophysiology of cerebral oedema in acute liver
activity and first-pass metabolism. N Engl J Med. failure. World J Gastroenterol. 2013;19(48):9240–55.
1990;322(2):95–9. 67. Albrecht J, Norenberg MD. Glutamine: a Trojan horse
50. Chalasani N, Bonkovsky HL, Fontana R, et al.
in ammonia neurotoxicity. Hepatology. 2006;44(4):
Features and outcomes of 899 patients with drug-­ 788–94.
induced liver injury: the DILIN prospective study. 68. Detry O, De Roover A, Honore P, Meurisse M. Brain
Gastroenterology. 2015;148(7):1340–1352.e7. edema and intracranial hypertension in fulminant
51. Glue P, Clement RP. Cytochrome P450 enzymes and hepatic failure: pathophysiology and management.
drug metabolism—basic concepts and methods of World J Gastroenterol. 2006;12(46):7405–12.
assessment. Cell Mol Neurobiol. 1999;19(3):309–23. 69. Ranjan P, Mishra AM, Kale R, Saraswat VA, Gupta
52. Goodman LS, Gilman A, Brunton LL, Lazo JS, Parker RK. Cytotoxic edema is responsible for raised intra-
KL. Goodman and Gilman’s the pharmacological cranial pressure in fulminant hepatic failure: in vivo
basis of therapeutics. 11th ed. New York: McGraw-­ demonstration using diffusion-weighted MRI in
Hill; 2006. human subjects. Metab Brain Dis. 2005;20(3):181–92.
53. Lee WM. Drug-induced hepatotoxicity. N Engl J
70. Masoro EJ. Lipids and lipid metabolism. Annu Rev
Med. 2003;349(5):474–85. Physiol. 1977;39:301–21.
54. Bromley PN, Cottam SJ, Hilmi I, et al. Effects of 71. Kleiner DE, Brunt EM, Van Natta M, et al. Design
intraoperative N-acetylcysteine in orthotopic liver and validation of a histological scoring system
transplantation. Br J Anaesth. 1995;75(3):352–4. for nonalcoholic fatty liver disease. Hepatology.
55. Thies JC, Teklote J, Clauer U, et al. The efficacy 2005;41(6):1313–21.
of N-acetylcysteine as a hepatoprotective agent in 72. Tessari P, Coracina A, Cosma A, Tiengo A. Hepatic
liver transplantation. Transpl Int. 1998;11(Suppl lipid metabolism and non-alcoholic fatty liver disease.
1):S390–2. Nutr Metab Cardiovasc Dis. 2009;19(4):291–302.
56. Hilmi IA, Peng Z, Planinsic RM, et al. N-acetylcysteine 73. Alberti KG, Zimmet P, Shaw J. The metabolic

does not prevent hepatorenal ischaemia-reperfusion syndrome—a new worldwide definition. Lancet.
injury in patients undergoing orthotopic liver trans- 2005;366(9491):1059–62.
plantation. Nephrol Dial Transplant. 2010;25(7): 74. Marchesini G, Brizi M, Bianchi G, et al. Nonalcoholic
2328–33. fatty liver disease: a feature of the metabolic syn-
57. Hediger MA, Clemencon B, Burrier RE, Bruford
drome. Diabetes. 2001;50(8):1844–50.
EA. The ABCs of membrane transporters in health 75. Sheth SG, Gordon FD, Chopra S. Nonalcoholic ste-
and disease (SLC series): introduction. Mol Asp Med. atohepatitis. Ann Intern Med. 1997;126(2):137–45.
2013;34(2–3):95–107. 76. Amitrano L, Guardascione MA, Brancaccio V, Balzano
58.
Ishikawa T. The ATP-dependent glutathione A. Coagulation disorders in liver disease. Semin
S-conjugate export pump. Trends Biochem Sci. Liver Dis. 2002;22(1):83–96.
1992;17(11):463–8. 77. Caldwell SH, Hoffman M, Lisman T, et al. Coagulation
59. Doring B, Petzinger E. Phase 0 and phase III trans- disorders and hemostasis in liver disease: pathophysi-
port in various organs: combined concept of phases ology and critical assessment of current management.
in xenobiotic transport and metabolism. Drug Metab Hepatology. 2006;44(4):1039–46.
Rev. 2014;46(3):261–82. 78. Kaul VV, Munoz SJ. Coagulopathy of liver disease.
60. Harden CL, Leppik I. Optimizing therapy of seizures Curr Treat Options Gastroenterol. 2000;3(6):433–8.
in women who use oral contraceptives. Neurology. 79. Mitchell O, Feldman DM, Diakow M, Sigal SH. The
2006;67(12 Suppl 4):S56–8. pathophysiology of thrombocytopenia in chronic liver
61.
Han HS, Kang G, Kim JS, Choi BH, Koo disease. Hepat Med. 2016;8:39–50.
SH. Regulation of glucose metabolism from a liver-­ 80. Sogaard KK, Horvath-Puho E, Gronbaek H, Jepsen
centric perspective. Exp Mol Med. 2016;48:e218. P, Vilstrup H, Sorensen HT. Risk of venous throm-
62. Adeva-Andany MM, Gonzalez-Lucan M, Donapetry-­ boembolism in patients with liver disease: a nation-
Garcia C, Fernandez-Fernandez C, Ameneiros-­ wide population-based case-control study. Am J
Rodriguez E. Glycogen metabolism in humans. BBA Gastroenterol. 2009;104(1):96–101.
Clin. 2016;5:85–100. 81. Tripodi A, Chantarangkul V, Mannucci PM. Acquired
63. Bhattacharya K. Investigation and management of
coagulation disorders: revisited using global coagu-
the hepatic glycogen storage diseases. Transl Pediatr. lation/anticoagulation testing. Br J Haematol. 2009;
2015;4(3):240–8. 147(1):77–82.
1  Physiology and Anatomy of the Liver 19

82. Leebeek FW, Rijken DC. The fibrinolytic status in liver 86. Peck-Radosavljevic M, Zacherl J, Meng YG, et al.
diseases. Semin Thromb Hemost. 2015;41(5):474–80. Is inadequate thrombopoietin production a major
83. Tripodi A. The coagulopathy of chronic liver disease: cause of thrombocytopenia in cirrhosis of the liver? J
is there a causal relationship with bleeding? No. Eur J Hepatol. 1997;27(1):127–31.
Intern Med. 2010;21(2):65–9. 87. Peck-Radosavljevic M, Wichlas M, Zacherl J, et al.
84. Basili S, Raparelli V, Violi F. The coagulopathy of Thrombopoietin induces rapid resolution of throm-
chronic liver disease: is there a causal relationship with bocytopenia after orthotopic liver transplanta-
bleeding? Yes. Eur J Intern Med. 2010;21(2):62–4. tion through increased platelet production. Blood.
85. Dhainaut JF, Marin N, Mignon A, Vinsonneau
2000;95(3):795–801.
C. Hepatic response to sepsis: interaction between 88. Shen M, Shi H. Sex hormones and their receptors
coagulation and inflammatory processes. Crit Care regulate liver energy homeostasis. Int J Endocrinol.
Med. 2001;29(7 Suppl):S42–7. 2015;2015:294278.
Chronic Liver Failure and 
Hepatic Cirrhosis 2
Lauren Tal Grinspan and Elizabeth C. Verna

Keywords MELD Model for end-stage liver disease


Hepatitis · Chronic liver disease · Hepatic fibrosis NAFLD Nonalcoholic fatty liver disease
· Liver failure · Cirrhosis · Portal hypertension NASH Nonalcoholic steatohepatitis
POPH Portopulmonary hypertension
PSE Portosystemic encephalopathy
Abbreviations PVT Portal venous thrombosis
SAAG Serum-ascites albumin gradient
AASLD American Association for the Study of SBP Spontaneous bacterial peritonitis
Liver Diseases TGF Transforming growth factor
AH Alcoholic hepatitis TIPS Transjugular intrahepatic portosys-
ATP Adenosine triphosphate temic shunt
CDC Center for disease control TLRs Toll-like receptors
CTP Child-Turcotte-Pugh TNF Tumor necrosis factor
ECM Extracellular matrix USPSTF United States preventative services
HBV Hepatitis B virus task force
HCC Hepatocellular carcinoma
HCV Hepatitis C virus
HPS Hepatopulmonary syndrome
HRS Hepatorenal syndrome Introduction
HSC Hepatic stellate cell
HVPG Hepatic venous pressure gradient Chronic liver disease is a condition that is charac-
IL Interleukin terized by persistent liver injury for more than
MDF Maddrey discriminant function 6 months after initial exposure or diagnosis of
liver disease. Causes of chronic liver disease
L. T. Grinspan, MD, PhD include infectious, inflammatory, toxic, vascular
Department of Medicine, Columbia University and congenital/genetic etiologies. The presence
Medical Center, New York, NY, USA of persistent liver injury induces a healing
E. C. Verna, MD, MS (*) response that results in hepatic fibrosis. Cirrhosis
Transplant Initiative, Division of Digestive and Liver is a late stage of hepatic fibrosis characterized by
Diseases, Center for Liver Disease and Transplantation, distortion of normal liver architecture that leads
Columbia University Medical Center,
New York, NY, USA to hepatocellular dysfunction, increased intra-
e-mail: ev77@cumc.columbia.edu hepatic resistance and ultimately to hepatic

© Springer International Publishing AG, part of Springer Nature 2018 21


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_2
22 L. T. Grinspan and E. C. Verna

insufficiency and the development of portal to 102,155 admissions in 2009 based on the
hypertension. Complications of cirrhosis include U.S. Nationwide Inpatient Sample, with overall
impaired synthetic function (including jaundice inpatient mortality of 6.6% [8]. Among individu-
and coagulopathy), portal hypertension (leading als with chronic liver disease, there has been an
to ascites, spontaneous bacterial peritonitis, vari- increase in the prevalence of nonalcoholic fatty
ceal hemorrhage, portosystemic encephalopathy) liver disease (NAFLD) in the US from 46.8% to
and increased risk of hepatocellular carcinoma. 75.1% while hepatitis B (HBV), hepatitis C
Prognosis is dependent on the presence of the (HCV) and alcohol remained relatively stable
previously mentioned complications and can from 1988 to 2008 based on National Health and
be estimated using the Child-Turcotte-Pugh Nutrition Examination Surveys [9]. NAFLD thus
Classification or the Model for End-stage Liver has become an increasingly common cause of cir-
Disease (MELD) scoring systems. Management rhosis. The number of adults with NAFLD await-
of cirrhosis is based on treatment of the underly- ing liver transplants has tripled from 2004 to
ing etiology of chronic liver disease, preventing 2013, making NAFLD the second leading disease
and managing complications, liver transplanta- of patients listed for transplant after HCV [10].
tion, and may soon include approaches at revers-
ing cirrhosis with antifibrotic agents. This chapter
will review the epidemiology, pathophysiology, Etiologies
clinical presentation, complications and treat-
ment of cirrhosis. There are numerous causes of chronic liver disease
and cirrhosis that include infectious, inflammatory,
genetic/congenital and toxic etiologies (Table 2.1).
Epidemiology
Table 2.1  Etiologies of cirrhosis
Liver cirrhosis is a significant source of mor-
bidity and mortality in the United States. Viral Chronic HCV
Chronic liver disease and cirrhosis were the Chronic HBV
sixth leading cause of death in people aged Fatty liver diseases NAFLD
25–64 and the tenth leading cause of death in Alcoholic liver disease
men of all ages in the United States in 2014 [1]. Storage diseases Hemochromatosis
The prevalence of cirrhosis was 0.27% in the Wilson disease
United States in 2010 census data, accounting Alpha-1 antitrypsin deficiency
Autoimmunie Autoimmune hepatitis
for 633,323 adults [2], though this may account
Primary and secondary biliary
for as little as one-third of actual cases as cir-
cirrhosis
rhosis is often identified post-mortem [3, 4]. Primary sclerosing cholangitis
Prevalence of cirrhosis increases with age with IgG4 cholangiopathy
a bimodal age distribution, peaking at age Chronic biliary Recurrent bacterial cholangitis
45–54 and again after age 75. There are differ- disease Bile duct stenosis
ences based on race/ethnicity with highest Cardiovascular Budd-Chiari syndrome
prevalence in non-Hispanic blacks. Higher Right-sided heart failure
prevalence is also associated with patients with Hereditary hemorrhagic
lower education level and socioeconomic status telangiectasia
attributed to higher rates of obesity and diabe- Infiltrative Granulomatous liver disease
tes, intravenous drug use, hepatitis C infection (sarcoid)
and alcohol use [2, 5–7]. Congenital/genetic Polycystic liver disease
The number of hospital admissions for cirrho- Rare Medications
sis has increased from 74,417 admissions in 2003 Porphyria
2  Chronic Liver Failure and Hepatic Cirrhosis 23

The most common causes of end-stage liver dis- fibrosis in an average of 6 years [14].
ease in adults in the United States include NAFLD, Hepatocellular carcinoma (HCC) is an uncom-
HBV, HCV, alcohol, and hemochromatosis. Less mon complication of NAFLD and occurs in the
common causes include autoimmune hepatitis, pri- presence of cirrhosis [14]. Patients with NAFLD
mary biliary cirrhosis, primary sclerosis cholangi- who develop HCC have a worse prognosis than
tis, Wilson’s disease, alpha-1 antitrypsin deficiency, those with HCV-related HCC [14]. According to
vascular and granulomatous etiologies. Here we one study, over 7.6 years NAFLD (even without
will discuss the most common causes in greater fibrosis) increases risk of mortality by 34%
detail. Treatment of these disorders is beyond the compared to the general population, most com-
scope of this chapter. monly due to malignancy, ischemic heart dis-
ease and liver disease [15]. Management of
NAFLD is summarized in the American
 onalcoholic Fatty Liver Disease/
N Association for the Study of Liver Diseases
Nonalcoholic Steatohepatitis (AASLD) practice guidelines [11].

Nonalcoholic fatty liver disease (NAFLD) is the


hepatic manifestation of the metabolic syn- Alcohol-Related Liver Disease
drome and is associated with obesity, diabetes
and dyslipidemia. It encompasses a spectrum of Alcoholic liver disease is characterized by ste-
disease including simple steatosis, steatosis atosis, hepatocyte apoptosis and acute inflam-
associated with inflammation (NASH) and mation and is histologically indistinguishable
NAFLD-­ associated fibrosis and cirrhosis. from NASH. Fatty liver develops in 90% of
Diagnosis of NAFLD requires evidence of individuals with alcohol intake exceeding 60 g/
hepatic steatosis by imaging or biopsy and the day though can occur in individuals who drink
absence of other causes of hepatic fat accumula- less [16]. In the setting of fatty liver disease due
tion such as alcohol, medications, hepatitis C or to alcohol, there is 30% risk of progression to
hereditary disorders [11]. As mentioned above, cirrhosis with continued alcohol intake [17].
NAFLD has become the most common liver dis- Simple fatty liver secondary to alcohol may be
order in the United States and has a prevalence completely reversed with abstinence after
of 46–75% [9, 12]. The prevalence of NASH is 4–6 weeks, however, studies have even sug-
lower ranging from 3 to 5% though is likely ris- gested that even with abstinence, there may be
ing as well [13]. The natural history and progno- progression to fibrosis and cirrhosis in 5–15%
sis of NAFLD is highly variable and depends on of individuals [16, 18, 19]. A subset of patients
the presence of fibrosis [14, 15]. Some studies with alcoholic liver disease will develop acute
suggest that one third of patients with NAFLD/ alcoholic hepatitis (AH) a separate clinical
NASH have progressive fibrosis [14]. Risk of diagnosis based on acute liver dysfunction in
fibrosis in NAFLD is impacted by multiple envi- the setting of excessive alcohol consumption
ronmental, genetic, lifestyle factors, as well as that ranges from mild injury to severe, life-
histological subtype (with NASH leading to the threatening liver injury [16]. Acute AH has a
highest risk of fibrosis and cirrhosis) [14, 15]. very poor short-term prognosis. The Maddrey
Factors that predict progressive fibrosis include discriminant function (MDF) that includes total
Hispanic ethnicity, certain genetic polymor- bilirubin and prothrombin time can help iden-
phisms (PNPLA3 and TM6SF2), increasing age, tify patients with AH who are at high risk of
diabetes and obesity [14]. The rate of fibrosis is mortality. Patients with an elevated MDF ≥ 32
generally slow at an average rate of one stage have a 1 month mortality as high as 30–50%.
per 7.7 years, however, there are rapid progress- Even patients with mild AH are at a high risk of
ers that advance from no fibrosis to late-stage developing progressive liver injury [16].
24 L. T. Grinspan and E. C. Verna

Hepatitis B Virus patients on long-term hemodialysis and individu-


als who have received blood transfusions or
Hepatitis B virus (HBV) is a DNA virus acquired organ transplants before 1992, though the biggest
by exposure to infected blood and bodily fluids, risk factor remains intravenous drug use [35, 38–
most often through perinatal transmission, sexual 40]. Recent breakthroughs in antiviral therapy
contact or injected drug use [20]. HBV is a sig- are likely to dramatically alter the natural history,
nificant global health concern with a prevalence prevalence and significance of HCV in the com-
of 240 million individuals chronically infected as ing years.
of 2005, and represents a leading cause of HCC
worldwide [21, 22]. There has been a decrease in
prevalence from 1990 to 2005 related in part to Pathophysiology
population-wide vaccination against HBV in
newborns, young children and adolescents [21, Liver Injury
23–25]. The risk of developing chronic HBV
depends on age and immune function at the time The various chronic liver diseases cause liver
of infection. Chronic HBV affects 90% of infants injury and cell death via apoptosis, necrosis or
infected during first year of life, more than 50% more often a combination of the two, termed
of immune compromised adults, 30% of children necrapoptosis or nectroptosis [41–44]. Apoptosis,
under age 6 and only 5% of healthy adults [26– programmed cell death, is an active, ATP-­
28]. The natural history of chronic HBV infec- dependent process that is prominent in liver injury,
tion includes a 13–18% 5-year incidence of often induced by specific stimuli secondary to
cirrhosis with 1–17% 5-year risk of hepatocellu- drug-induced liver diseases, viral hepatitis, alco-
lar carcinoma [29]. Incidence of hepatocellular holic liver disease, nonalcoholic fatty liver disease,
carcinoma increases with HBV DNA viral load cholestasis and vascular liver diseases [44].
and levels of HBV replication, and unlike most Although apoptosis occurs at baseline as a method
other chronic liver diseases, HCC may occur of hepatocyte removal, it becomes pathogenic in
even in patients without significant fibrosis [30– the presence of inadequate regeneration and induc-
33]. AASLD guidelines for treatment of HBV tion of an inflammatory response [45]. Necrosis is
were recently published [34]. an energy-independent process activated in the
absence of cellular ATP such as during ischemia or
nitrative/oxidative stress [46]. Without ATP, cells
Hepatitis C Virus lose the ability to maintain ion gradients, swell and
lyse, releasing cellular debris [46]. Due to the
Hepatitis C virus (HCV) is the most common more vigorous release of cellular contents in
indication for liver transplantation worldwide necrosis, inflammation is a more prevalent feature
[35–37] and the most common chronic infection of necrosis compared to apoptosis. Apoptosis and
in the United States. Of those acutely infected necrosis can both be initiated by the same signals
with HCV, 65–75% of patients will develop in which apoptotic cascades are initiated, and
chronic HCV infection. After infection, patients depending on the resulting milieu the path may
remain largely asymptomatic for decades before diverge to apoptosis or necrosis [41, 47, 48].
developing clinical symptoms. Therefore, many
go untreated until cirrhosis and clinically signifi-
cant disease develops. For this reason, the US Fibrosis
Preventive Services Task Force (USPS) and the
Centers for Disease Control and Prevention In chronic liver disease, the liver is subjected to
(CDC) recommend testing individuals at highest repetitive tissue damage resulting in alterations
risk for HCV [38, 39]. High-risk individuals in regenerative capacity, inflammatory response
include those born between 1945 and 1964, and eventually fibrosis (Fig. 2.1) [49–51].
2  Chronic Liver Failure and Hepatic Cirrhosis 25

Initiation Perpetuation

Injury
Oxidative stress Contractility
Proliferation
Apoptotic bodies ↑ET1
LPS ↓NO
↑PDGF
Paracrine stimuli Fibrogenesis
↑VEGF ↑TGFβ1/
↑FGF ↑CTGF

↑NOX
Reversion ↑Leptin ↑MMP2 & MMP9
↑MT1-MMP
↑TIMP1 & TIMP2
Resolution Altered matrix
↑p53 ↑PDGF degradation
↑Chemokines ↑Chemokines
Senescence ↑TLR ligands
↓NFκB ↑Adenosine
↓TIMP1 & TIMP2
T cells
↑TRAIL
B cells
↑Fas
NK cells
Apoptosis HSC
NKT cells
chemotaxis
Dendritic cells
Mast cells
Inflammatory signaling

Fig. 2.1  The pathways of HSC activation include those fibroblast growth factor, HSC hepatic stellate cell, LPS
involved with initiation and those that contribute to per- lipopolysaccharide, MMP matrix metalloproteinase,
petuation. Initiation is stimulated by reactive oxygen MT1-MMP membrane type matrix metalloproteinase,
intermediates, apoptotic bodies, LPS and paracrine stim- NFκB nuclear factor κB, NK natural killer, NKT natural
uli from neighboring cell types, including Kupffer cells, killer T cell, NO nitric oxide, NOX NAPDH oxidase,
sinusoidal endothelial cells and hepatocytes. Perpetuation PDGF platelet-derived growth factor, TGF-β1 transform-
is comprised of proliferation, contractility, fibrogenesis, ing growth factor β1, TIMP tissue inhibitor of metallopro-
altered matrix degradation, chemotaxis and inflammatory teinase, TLR toll-like receptor, TRAIL
signaling. Resolution of hepatic fibrosis occurs with tumor-necrosis-factor-related apoptosis-inducing ligand,
removal of the primary insult to the liver and leads to loss VEGF vascular endothelial growth factor. Figure and leg-
of activated HSCs through apoptosis, senescence or rever- end adapted from Nature Publishing Company ©
sion to a quiescent phenotype. Abbreviations: CTGF con- Friedman, S.L. Nature Reviews Gastroenterology and
nective tissue growth factor, ET1 endothelin 1, FGF Hepatology, 7, 425–436 (2010)

Hepatic fibrosis is a wound-healing response of injury, changes in extracellular matrix composi-


the liver to chronic injury in which extracellular tion and intestinal dysbiosis [51, 55]. In the set-
matrix accumulates around damaged areas and ting of chronic liver disease, hepatocytes undergo
forms a scar that replaces hepatocytes. Central to apoptosis or necrosis, releasing their contents
the development of hepatic fibrosis is the “acti- including DNA, damage-associated molecular
vation” or “transdifferentiation” of hepatic stel- patterns (DAMPs), and reactive oxygen species
late cells (HSCs), the primary sources of that induce release of chemokines, pro-­
extracellular matrix, from quiescent vitamin-A inflammatory (TNF-alpha, IL-1beta and IL-6)
storing cells to proliferative myofibroblasts [51– and pro-fibrotic (TGF-beta) factors from
54]. Additionally, endothelial cells and Kupffer neighboring Kupffer cells. As opposed to the
­
cells are essential modulators of fibrosis progres- regenerative response induced by Kupffer cells in
sion [55]. acute liver injury, there is a fibrotic response in
Activation of the HSC occurs in stages known chronic liver injury. Kupffer cells activate HSCs
as initiation and perpetuation. Initiation involves and summon immune cells through the release of
early events and stimuli, including epithelial chemokines, CCL2 and CCL5.
26 L. T. Grinspan and E. C. Verna

Early in fibrosis, injury of the sinusoidal endo- resistance. The increase in intra-hepatic resis-
thelial cells induces production of altered cellular tance is not only a mechanical consequence of
fibronectin, which leads to creation of increas- abnormal architecture; but is additionally due to
ingly dense extracellular matrix (ECM) in the endothelial dysfunction, reduced hepatic nitric
subendothelial space of Disse [56]. The accumu- oxide and the presence of contractile elements
lation of ECM in the subendothelial space of within the hepatic vasculature that respond to
Disse can alter cell signaling via integrins, cad- the influence of altered vasoactive mediators
herins and selectins through release of soluble [58–62]. Splanchnic vasodilation that is seen in
growth factors, which are also activating of the cirrhosis due to an increase in release of local
HSCs. Once activated, HSCs transform from qui- endothelial factors and humoral vasodilators
escent cells to highly fibrogenic cells called myo- such as carbon monoxide and endocannabinoids
fibroblasts. This transformation leads to leads to increase in portal inflow [63].
acceleration of fibrosis and the accumulation of
scar, loss of hepatocyte microvilli and sinusoidal
endothelial fenestrae and results in deterioration Clinical Presentation
of hepatic function. and Complications
In addition to the intrahepatic injury signaling,
extrahepatic signals contribute to liver fibrosis. Patients with cirrhosis may be asymptomatic
Intestinal dysbiosis and bacterial overgrowth in the compensated stage. With progression of
leading to a “fibrogenic microbiome” and the disease, patients may develop signs and symp-
release of pathogen associated molecular patterns toms of portal hypertension and hepatocellular
(PAMPs) and activation of toll-like receptors dysfunction.
(TLRs) [51]. This contributes to hepatic inflam-
mation and thus to fibrogenesis.
Perpetuation involves response of HSCs to Synthetic Dysfunction
cytokines and growth factors that lead to prolif-
eration, contractility, fibrogenesis, matrix degra- Hepatic dysfunction leads to impaired protein
dation and pro-inflammatory signaling. Unlike biosynthesis, lipid metabolism and excretion
other organs, the liver has regenerative properties of bilirubin. Abnormal metabolism and excre-
such that the liver can sustain injury and chronic tion of bilirubin leads to direct hyperbilirubi-
inflammation over a long period of time before nemia and is manifested as jaundice, scleral
developing end-stage fibrosis and cirrhosis [57]. icterus, dark urine, acholic stool and pruritis.
The presence of coagulopathy indicates severe
hepatic synthetic dysfunction and is due to
Hepatocellular Dysfunction the impaired hepatic synthesis of antithrom-
and Portal Hypertension bin and protein C among others. This coagu-
lopathy leads to abnormal prothrombin time
Cirrhosis represents a late stage of liver fibro- and activated partial thromboplastin time in
sis that is characterized by regenerative nodule laboratory testing and manifests clinically as
formation and organ contraction leading to dis- increased risk of bleeding or thrombosis as
torted architecture of the lobules and vasculature. both pro- and anti-coagulants are decreased.
These changes lead to hepatocellular dysfunc- Notably, thrombocytopenia also contributes
tion including abnormal metabolic and synthetic to abnormal coagulation in ­cirrhosis and is
function of liver. It also causes increased intra- multifactorial secondary to congestive hyper-
hepatic resistance to blood flow that contributes splenism (portal hypertension), relatively
to the development of portal hypertension. Portal decreased thrombopoietin synthesis, immune
hypertension results from an increase in portal complex-associated platelet clearance and
venous inflow and an increase in portal outflow reticuloendothelial destruction [64].
2  Chronic Liver Failure and Hepatic Cirrhosis 27

Portal Hypertension Spontaneous Bacterial Peritonitis

Portal hypertension is responsible for many of Spontaneous bacterial peritonitis (SBP) is defined
the clinical complications of cirrhosis, includ- as a bacterial infection of the ascitic fluid with a
ing gastroesophageal varices, ascites, portosys- positive bacterial culture and/or polymorphonu-
temic encephalopathy, renal dysfunction and clear cell count of greater than 250 cells per mm2
hepatopulmonary syndrome. As stated above, without an identifiable surgically treatable intra-­
portal hypertension results from an increase in abdominal source [76]. SBP should be suspected
portal venous inflow (splanchnic vasodilation) in patients with cirrhosis who develop fever,
and an increase in portal outflow resistance. The abdominal pain or tenderness, altered mental sta-
hepatic venous pressure gradient (HVPG) can tus, hypotension, renal failure or metabolic disar-
be measured to approximate the difference in ray. Most patients with ascites who are admitted
pressure between the portal vein and the hepatic to the hospital for other reasons should undergo
vein and can be used to determine the severity of diagnostic paracentesis to look for evidence of
portal hypertension. The presence of portal infection. Most cases of SBP are due to gut bac-
hypertension is defined by a HVPG ≥5–6 mmHg. teria such as Escherichia coli and Klebsiella and
Clinically significant portal hypertension usu- can be treated with third generation cephalospo-
ally occurs at HVPG ≥10 mmHg. At HVPG rins or fluoroquinolones (unless the patient has
≥10–12 patients are at risk for variceal bleed been receiving a fluoroquinolone for SBP pro-
[65–67]. phylaxis). SBP is associated with a 24.2% 30-day
mortality and a 66.5% 3-year mortality [77]. The
most severe complication of SBP is hepatorenal
Ascites syndrome (up to 30% of cases), therefore, intra-
venous albumin is used as a preventative measure
Ascites refers to the abnormal accumulation of to improve mortality [75, 78]. There is a high rate
fluid within the peritoneal cavity and is related to of recurrence of SBP estimated to be as high as
portal hypertension. Patients with ascites pres- 70% at one year [79]. Antibiotic prophylaxis is
ent with a full, bulging abdomen with dullness to used to decrease the risk of bacterial infection
percussion in the flanks. Testing albumin levels and mortality in patients if they have the follow-
in serum and ascitic fluid can be used to calcu- ing risk factors for SBP: ascitic fluid protein con-
late the serum-ascites albumin gradient (SAAG), centration <1.5 g/dL, cirrhosis and acute
which should be performed for all patients with gastrointestinal bleeding or for secondary pre-
new onset ascites. A SAAG level ≥1.1 can be vention in patients with prior history of SBP.
used to categorize with 97% accuracy that asci-
tes is secondary to portal hypertension [68–70].
Eighty-five percent of patients with ascites have Hepatic Hydrothorax
underlying cirrhosis [68]. Ascites is a poor prog-
nostic indicator with a 15–20% 1-year mortal- Hepatic hydrothorax is a pleural effusion that is
ity and 44% 5-year mortality [71–73]. Initial usually found on the right and occurs in around
management consists of sodium restriction and 5% of patients with cirrhosis and ascites [80]. It
diuretics, usually furosemide and spironolac- is caused by accumulation of ascitic fluid in the
tone [68–70, 74]. Tense ascites can be treated pleural space due to small diaphragmatic defects.
with large volume paracenteses with albumin This fluid, similar to ascites can become infected
infusion. In the case of refractory ascites, serial with bacteria leading to spontaneous bacterial
paracenteses or transjugular intrahepatic porto- empyema [81]. Like ascites it is treated initially
systemic shunt (TIPS) may be considered and with dietary sodium restriction and diuretics and
the patient should be referred for liver transplan- if refractory, TIPS can be considered as second-­
tation [75]. line treatment [68].
28 L. T. Grinspan and E. C. Verna

Esophageal Varices the time of diagnosis ranges from 10 to 21% with


increased prevalence in decompensated cirrhosis
Cirrhotic patients with significant portal hyper- [84–88]. The risk of developing PSE is 5–25%
tension commonly develop portosystemic collat- within the first 5 years after diagnosis with cir-
erals due to increased portal venous inflow and rhosis depending on the presence of other risk
insufficient portal decompression. The most clin- factors. PSE occurs late in cirrhosis and is associ-
ically significant collaterals include esophageal ated with decreased survival with a survival prob-
varices as they may result in variceal hemorrhage ability of 10–73% at 1 year and 3–38% at 3 years
[65]. Patients with cirrhosis and varices usually depending on prognostic index [89]. Diagnosis of
have an HVPG of at least 10–12 mmHg and they PSE is by exclusion of other etiologies of altered
are found in around 50% of patients with cirrho- mental status such as toxic ingestion, electrolyte
sis [65, 66]. The diagnosis of these complications disorders, infections, intracranial bleeds, strokes
is made endoscopically. Endoscopic findings of or lesions. Increased blood ammonia alone has
large varices alone or small varices in the pres- not been shown to add diagnostic, staging or
ence of red wale signs or in a person with Child prognostic value, though if found to be normal
B/C cirrhosis increase variceal bleeding risk and then other diagnoses should be considered [84,
warrant primary prophylaxis with non-selective 90]. PSE is graded from grade I to IV based on
beta-­blockers or variceal ligation. Therefore, symptoms and exam findings. Treatment consists
patients should undergo endoscopic screening for of non-absorbable disaccharides such as lactu-
varices to determine necessity of primary pro- lose for episodic PSE with the addition of rifaxi-
phylaxis. In the setting of acute variceal hemor- min to prevent of recurrence [91]. For refractory
rhage, treatment includes conservative blood PSE, liver transplantation remains the only treat-
transfusion strategy, antibiotic prophylaxis for ment option.
SBP, splanchnic vasoconstriction with octreo-
tide/terlipressin for 3–5 days, and endoscopic
band ligation (treatment of choice). If the patient Hepatopulmonary Syndrome
fails medical/endoscopic management, they can
undergo TIPS versus surgical portosystemic Hepatopulmonary syndrome (HPS) is defined as
shunt with the goal of reducing HVPG by 20% or a widened alveolar-arterial oxygen gradient and
to an absolute value of <12 mmHg [65, 82, 83]. intrapulmonary vasodilation in the setting of
Alternative temporizing treatments include bal- hepatic dysfunction or portal hypertension and in
loon tamponade, and all patients with variceal the absence of intrinsic lung disease [92].
hemorrhage should be considered for liver trans- Intrapulmonary vasodilation is present in 50% of
plantation evaluation. patients with cirrhosis, but only 15–30% have
hepatopulmonary syndrome. Pathogenesis of
HPS is due to increased pulmonary production of
Portosystemic Encephalopathy vasodilators such as NO, endothelin 1 and carbon
monoxide and angiogenesis [65]. Presentation
Portosystemic encephalopathy (PSE) constitutes includes platypnea (increased dyspnea with
a range of neurologic dysfunction that ranges standing), orthodeoxia (worsening hypoxemia
from sleep disturbances (early symptom) to coma with upright position) due to dominance of vaso-
and occurs secondary to hepatic insufficiency or dilation in lung bases [93]. Diagnosis is made by
portosystemic shunting [84]. Asterixis or “flap- echocardiogram that demonstrates delayed
ping tremor” is often present on physical exam shunting of intravenous agitated saline bubbles
and signifies a loss in postural tone. The inci- [92, 94]. HPS is associated with an increase in
dence and prevalence of PSE is correlated to the mortality and can negatively impact the outcome
severity of liver dysfunction and the presence of of liver transplantation. Treatment includes sup-
TIPS. The prevalence of overt encephalopathy at plemental oxygen and liver transplantation. TIPS
2  Chronic Liver Failure and Hepatic Cirrhosis 29

procedure has been attempted in these patients, treatment with nephrotoxic drugs, and (6)
but there is no evidence to support its use [95– absence of parenchymal kidney disease [68,
100]. Liver Transplant is currently the only effec- 109]. Type I HRS is characterized by a rapidly
tive treatment for HPS [101]. Progressive reversal progressive reduction in renal function defined
of HPS can be seen over 12 months post-­ by doubling of creatinine to a value >2.5 mg/dL
transplant with normalization of oxygenation in or a 50% reduction of the initial 24-h creatinine
many cases [102]. clearance to a level <20 mL per minute in less
than 2 weeks. Type II does not have a rapidly pro-
gressive course. Renal dysfunction in a patient
Portopulmonary Hypertension with cirrhosis is associated with a significant
increase in mortality, with a sevenfold increase in
Portopulmonary hypertension (POPH) is defined risk of death and 50% of patients dying within a
as a mean pulmonary artery pressure of month of onset of renal dysfunction [110].
>25 mmHg and pulmonary capillary wedge pres- Measures taken to prevent the onset of HRS,
sure <15 mmHg in the setting of portal hyperten- include infusion of albumin in the setting of SBP
sion and without secondary causes of pulmonary and administration of pentoxyphylline in the case
hypertension [103]. This condition results from of acute alcoholic hepatitis or in the setting of cir-
remodeling of lung vasculature due to prolonged rhosis, ascites and creatinine clearance between
portal hypertension that leads to a hyperdynamic 41 and 80 mL/min. Currently, the medical treat-
state [104]. Histologically, it is similar to primary ment for HRS I consists of albumin, octreotide
pulmonary hypertension. It occurs in 4–8% of and midodrine in the United States [111, 112].
patients and increases mortality after liver trans- Terlipressin, vasopressin and norepinephrine are
plantation [103–105]. POPH is classified based effective for treatment of HRS, and are more
on the mean pulmonary arterial pressures as mild widely used outside of the United States [113–
(25–35 mmHg), moderate (35–59 mmHg) and 116]. For persistent renal failure refractory to
severe (>50 mmHg) [106, 107]. Transthoracic medical management, the treatment is liver trans-
echocardiogram and can be used as an initial plantation. Hemodialysis can be used as a bridge,
screening test. Gold standard for diagnosis is a however without liver transplantation, median
right heart catheterization. Treatments are similar survival in patients with type I HRS is approxi-
to those used in pulmonary arterial hypertension. mately one month [68, 110, 117].
Goal of treatment is to improve hemodynamics
by reducing the pulmonary pressure and pulmo-
nary vascular resistance. Mean pulmonary arte- Cirrhotic Cardiomyopathy
rial pressures >50 mmHg is associated with
100% mortality after liver transplant [108] and is Cirrhotic cardiomyopathy is cardiac dysfunc-
generally considered a contraindication for tion in patients with cirrhosis in the absence of
transplant. known cardiac disease. It is characterized by
electrophysiological abnormalities such as QT
prolongation, diastolic dysfunction and a
Hepatorenal Syndrome blunted contractile response to stress. Diastolic
dysfunction occurs in around 40% of patients
Diagnostic criteria for hepatorenal syndrome with cirrhosis and is associated with a worse
(HRS) include (1) cirrhosis with ascites, (2) prognosis and increased mortality especially
serum creatinine greater than 1.5 mg/dL, (3) no after TIPS and liver transplantation [118–122].
improvement in serum creatinine after at least 2 Cardiac dysfunction is frequently clinically
days with diuretic withdrawal and volume expan- silent as systemic vascular resistance is low in
sion with albumin (1 g/kg body weight per day), cirrhosis, which reduces cardiac afterload.
(4) absence of shock, (5) no current or recent Patients may remain asymptomatic for months
30 L. T. Grinspan and E. C. Verna

to years until systolic heart failure is unmasked blood flow and hypercoagulability [130, 131].
in the setting of physiologic, pharmacologic or Treatment of PVT is controversial and requires
surgical stress. Standard treatments for heart considerations of risks and benefits for individ-
failure have not proven to be effective and cir- ual patients. Historically, chronic PVT in
rhotic cardiomyopathy can be reversed with patients with established cirrhosis is not con-
liver transplantation [123–125]. sidered an indication for anticoagulation.

Portal Vein Thrombosis Hepatocellular Carcinoma

Portal vein thrombosis (PVT) is a frequent Hepatocellular carcinoma (HCC) is the primary
complication of cirrhosis. It is usually diag- tumor of the liver that often develops in the set-
nosed via screening ultrasound and can be con- ting of chronic liver disease, especially hepatitis
firmed with cross-sectional venous phase B and C, or cirrhosis. HCC is one of the few
imaging with CT or MRI. The gold standard of cancers with rising incidence in the United
angiography is rarely necessary. Prevalence of States and is the fastest rising cause of cancer-
PVT in cirrhosis ranges from 10 to 25% [126– related deaths (Fig. 2.2) [22, 132, 133]. It is the
129] with an incidence of PVT of 5–16% [126, third leading cause of cancer-related death
130, 131]. Risk factors for PVT include slow worldwide with an annual incidence of 5% and

Liver and intrahepatic Bile Duct Cancer:


Number of New Cases and Deaths of Per 100,000 People, Age-Adjisted
16
Deaths - US
New Cases - SEER 9
14
Number of Cases Per 100,000 People

12

10

0
1975
1976
1977
1978
1979
1980
1981
1982
1983
1984
1985
1986
1987
1988
1989
1990
1991
1992
1993
1994
1995
1996
1997
1998
1999
2000
2001
2002
2003
2004
2005
2006
2007
2008
2009
2010
2011
2012

Fig. 2.2  Age-adjusted rates of Liver and Intrahepatic Bile Duct Cancer 1975–2013. Mortality source: US Mortality
Files, National Center for Health Statistics, CDC. Incidence source: SEER 9
2  Chronic Liver Failure and Hepatic Cirrhosis 31

is often diagnosed in a later stage due to the Child-Turcotte-Pugh Scoring System


absence of specific symptoms [71, 134]. The
median survival is 20 months in limited disease The Child-Turcotte-Pugh (CTP) score is a prog-
and 6 months in advanced HCC [135, 136]. nostic model originally developed to predict
Thus, HCC screening is currently recommended survival after portosystemic surgery that incor-
for all patients with cirrhosis, and subsets of porates serum albumin, bilirubin, ascites,
non-cirrhotic patients such as some with HBV, encephalopathy and nutritional status [143].
with ultrasound every 6 months [137]. HCC Significant differences in survival (1 year, 2
should be suspected in a previously compen- year) are predicted based on CTP classification:
sated cirrhotic who develops complications Class A (95%, 90%), Class B (80%, 70%) and
such as ascites, encephalopathy, jaundice or Class C (45%, 38%) [73]. While this score has
variceal bleeding. HCC may be associated with been validated in many clinical settings, it is
upper abdominal pain, weight loss, early satiety limited by the subjective nature of assessing
or palpable mass. Less commonly, patients can ascites and encephalopathy and the absence of a
present with obstructive jaundice, diarrhea, measure of renal function [144]. To overcome
bone pain, intraperitoneal bleeding due to tumor these problems with the CTP scoring system,
rupture, fever associated with tumor necrosis, additional prognostic models have been devel-
paraneoplastic syndromes or liver abscess. oped and validated.
Staging is based on Barcelona Clinic Liver
Cancer staging system [138–140]. Treatment is
beyond the scope of this chapter but has been Model for End-Stage Liver Disease
recently reviewed [141], and requires a multi-
disciplinary team approach including surgical The model for end-stage liver disease (MELD) is
(resection or liver transplant), locoregional an assessment tool that predicts liver disease
(such as transarterial chemoembolization) or severity based on serum creatinine, total bilirubin
systemic therapy. and INR. The inclusion of renal function to this
score increases its value as creatinine has been
found to be an independent predictor of progno-
Prognosis and Risk Stratification sis. The MELD score ranges from 6 to ≥40, which
corresponds to a range in 3-month survival of
Prognosis 90% to 7%, respectively [145]. The MELD was
originally developed to risk stratify patients for
Prognosis of cirrhosis depends on etiology, sever- TIPS procedure [146] and has since been demon-
ity, presence of complications and comorbid dis- strated to predict mortality over defined period of
eases. In compensated cirrhosis, in the absence of time [147, 148], and was therefore adapted in the
any of the major complications, mean survival is United States and many other countries for priori-
more than 12 years [73]. Decompensated cirrho- tizing liver graft allocation [149].
sis is marked by the development of any major
complication and is thus defined by the presence
of ascites, variceal bleeding, encephalopathy and Treatment of Cirrhosis
jaundice and associated with a mean survival of
around 2 years [73, 85]. Transition from compen- General Management Strategies
sated to decompensated cirrhosis occurs at a rate
of 5–7% per year. Measurement of HVPG can be Recommendations for treatment of cirrhosis
used to predict clinical decompensation in were recently reviewed in the NEJM [71].
patients with compensated cirrhosis [142]. General principles of cirrhosis management
Different prognostic models have been used to include protecting liver from harm, monitoring
predict mortality in cirrhosis. for development of complications, treating the
32 L. T. Grinspan and E. C. Verna

underlying cause of cirrhosis, and evaluating for cirrhosis was liver transplantation. However,
transplant when appropriate. studies that have shown regression of fibrosis
The liver can be protected by discontinuing with treatment of HBV [151–153] and HCV
hepatotoxic medications, avoiding the use of [154, 155] have sparked interest in the develop-
alcohol, NSAIDs and herbal supplements, moni- ment of antifibrotic agents as an exciting poten-
toring blood pressure and discontinuing antihy- tial therapy. Regression of fibrosis has also been
pertensive agents as needed. Patients with observed in patients with hemochromatosis,
cirrhosis should receive HAV and HBV vaccina- autoimmune hepatitis and biliary cirrhosis
tions if they are not immune. [156–159].
Patients should be followed closely by a gas- With the discovery of the endogenous mecha-
troenterologist or hepatologist (if available) and nisms of fibrosis reversal came the opportunity to
monitored for development of complications. design and develop antifibrotic therapies, though
Screening for HCC with ultrasonography or none are approved yet for clinical use [49, 51, 57,
computed tomography is recommended every 6 160]. The greatest limitation to advances in clini-
months [71, 137]. Endoscopy is recommended to cal use includes that lack of non-invasive bio-
screen for esophageal varices with subsequent markers of fibrosis. Liver biopsies that are being
surveillance per established guidelines [71, 150]. used now are invasive and can sometimes miss
Management of the various complications of cir- the affected area of the liver. Development of
rhosis has been discussed in the previous noninvasive markers of fibrosis would allow for
sections. monitoring of antifibrotic therapy in a less inva-
sive manner.

Indications for Liver Transplantation


Targets for Antifibrotic Agents
Cirrhotic patients should be considered for liver
transplant evaluation once they have an index There are many targets for antifibrotic agents that
complication such as ascites, hepatic encepha- can be categorized as (1) controlling or curing the
lopathy, variceal hemorrhage, or with hepatocel- primary disease process and reducing liver injury;
lular dysfunction resulting in a MELD score of (2) inhibiting myofibroblast activation (3) inhib-
≥15 [145]. Other indications for transplantation iting fibrogenesis and (4) promoting resolution of
include early graft failure (secondary to primary fibrosis (Fig. 2.3). Category 1 focuses on the pri-
nonfunction and hepatic artery thrombosis), mary disease process, while categories 2–4 focus
recurrent disease or chronic graft rejection [145]. on the HSCs and the pro-fibrotic liver microenvi-
Patients with HCC are considered for transplan- ronment, targets that are independent of the
tation if their tumor is within the Milan criteria underlying etiology. These targets and some of
(one lesion <5 cm, up to three lesions <3 cm, no the clinical trials involving the antifibrotic agents
hepatic manifestations and no vascular invasion). were recently reviewed [160].
In primary sclerosing cholangitis, recurrent bac- Treatments aimed at the underlying etiology
terial cholangitis and some selected cases of of liver disease are the most effective antifibrotic
cholangiocarcinoma are indications for liver therapies. For example, treatment of viral hepati-
transplantation. tis has resulted in regression of fibrosis in up to
75% of patients [151, 161]. In addition, control-
ling inflammation and fibrosis with compounds
Antifibrotic Therapies termed “hepatoprotectants” may reduce initial
liver injury [51]. These compounds range from
Historically, fibrosis was thought to be irrevers- inhibitors of apoptosis, inhibitors of lipogenic
ible and other than therapies aimed at the under- pathways, antioxidants and chemokine receptor
lying etiology, the only proven treatment for antagonists [51, 162–167]. In addition, interfer-
2  Chronic Liver Failure and Hepatic Cirrhosis 33

1. Control or cure primary disease 2. Target receptor-ligand interactions

Adiponectin PPAR-α,δ,γ agonist


NASH Other* CB1R antagonist ET-1 antagonist
ACE-I or ARB Tyrosine kinase antagonists
FXR agonist Immunosuppression Ghrelin FXR agonist
Vitamin E UDCA
PPAR-γ agonist Remove iron or copper
Lipogenesis inhibitor Alcholo abstinence
Viral suppression SVR Quiescent Activated
HSC HSC
HBV Liver Injury HCV
*e.g. Wilson disease
Autoimmune
liver disease Hereditary
hemochromatosis
Alcoholic liver disease

Normal Cirrhotic
liver liver

Liver Injury

Latent TGFβ TGFβ CTGF mAb


↓ NF-κB TIMP antagonist
ACE-I LOXL2 mAb
Block CB1R antagonist
Activation ↑Macrophage fibrolytic activity
↑NK cell activity
↑ Collagen ↑ Proliferation ↑ Matrix Degradation
↓ Matrix Degradation ↑ Apoptosis Prevent Cross-Linking
Stellate cell

3. Inhibit fibrogenesis 4. Promote resolution of fibrosis

Fig. 2.3  Mechanisms by which antifibrotic therapies may and by increasing the degradation of extracellular matrix,
lead to fibrosis regression. (1) Disease-specific therapies by fibrolytic macrophages or preventing its cross-linking
that control or cure the underlying disease are still the with antagonists to LOXL2. FXR farnesoid-X-receptor,
most effective antifibrotic approach. (2) Targeting recep- PPAR peroxisome proliferator-activated receptor, UDCA
tor–ligand interactions with either established or experi- ursodeoxycholic acid, SVR sustained virological response,
mental drugs to reduce hepatic stellate cell activation will CB1 cannabinoid receptor type 1, ARB angiotensin II
attenuate fibrosis development, with multiple potential receptor blocker, ET-1 endothelin 1, TGFβ transforming
strategies under development. (3) Inhibition of the most growth factor β, CTGF connective tissue growth factor,
potent of the profibrogenic pathways, for example, pre- mAb monoclonal antibody, NF-κB nuclear factor kappa-­
venting activation of latent TGFβ, or blocking the activity light-­
chain-enhancer of activated B cells, NK natural
of CTGF, are among the more promising antifibrotic strat- killer, TIMP tissue inhibitor of metalloproteinase, LOXL2
egies. (4) The resolution of fibrosis can be promoted by lysyl oxidase 2. Figure and legend reprinted from BMJ ©
enhancing the apoptosis of activated hepatic stellate cells Lee YA, Wallace MC, Friedman SL. Gut, 64, 830–841
either with drugs or through the activity of either NK cells (2015)

ing with pathways of growth factors and cyto- Finally, resolution of fibrosis can be brought
kines can inhibit myofibroblast activation. These about by reducing number of myofibroblasts or by
agents including tyrosine kinase ligands are cur- stimulating matrix degradation, mimicking the
rently being used in treatment of cancer and are endogenous mechanisms of fibrosis resolution.
thus promising as new agents to treat liver fibro- During initiation of fibrosis, the quiescent stellate
sis. Agents that inhibit fibrogenesis work by cell is activated to respond to many growth fac-
blocking the activation of pro-fibrogenic signal- tors and therefore, reduction in number of acti-
ing molecules such as TGF-beta, thus decreasing vated stellate cells is critical to resolution of
fibrosis even in the presence of liver injury. fibrosis. In regression of fibrosis, myofibroblasts
34 L. T. Grinspan and E. C. Verna

are transformed to an inactive phenotype [168]. agnosed cases and 149 diagnosed cases of cirrhosis,
detected in 4929 consecutive autopsies. J Intern
This transition occurs through apoptosis, senes-
Med. 1991;230(2):165–71.
cence or inactivation of the stellate cell [51]. 4. Giarelli L, et al. Occurrence of liver cirrhosis among
Given the possibility that inducing apoptosis may autopsies in Trieste. IARC Sci Publ. 1991;112:37–43.
affect hepatocytes and other untargeted cells, this 5. Mokdad AH, et al. The continuing epidemics of
obesity and diabetes in the United States. JAMA.
is not the most appealing therapy to pursue.
2001;286(10):1195–200.
Interestingly, inactivated HSCs are more prone to 6. Hsu CC, et al. Poverty increases type 2 diabetes inci-
transdifferentiation to myofibroblasts in the set- dence and inequality of care despite universal health
ting of subsequent insults suggesting that a previ- coverage. Diabetes Care. 2012;35(11):2286–92.
7. Denniston MM, et al. Chronic hepatitis C virus
ously injured liver is more susceptible to
infection in the United States, National Health and
fibrogenesis during subsequent insults. In regres- Nutrition Examination Survey 2003 to 2010. Ann
sion of fibrosis, there is also an increase in the Intern Med. 2014;160(5):293–300.
Ly6clo population of macrophages that secrete 8. Mellinger JL, et al. Variation among United States
hospitals in inpatient mortality for cirrhosis. Clin
larger quantities of fibrolytic matrix metallopro-
Gastroenterol Hepatol. 2015;13(3):577–84. quiz e30
teinases and anti-inflammatory cytokines such as 9. Younossi ZM, et al. Changes in the prevalence of the
IL-10 and promote resolution of fibrosis [49, 51]. most common causes of chronic liver diseases in the
This approach targets pro-­fibrolytic macrophages United States from 1988 to 2008. Clin Gastroenterol
Hepatol. 2011;9(6):524–530.e1. quiz e60
such as the Ly6clo cells or other enzymes involved
10. Wong RJ, et al. Nonalcoholic steatohepatitis is the
in extracellular matrix deposition. second leading etiology of liver disease among
adults awaiting liver transplantation in the United
Conclusions States. Gastroenterology. 2015;148(3):547–55.
11. Chalasani N, et al. The diagnosis and management of
Cirrhosis remains a significant cause of morbid-
non-alcoholic fatty liver disease: practice Guideline
ity and mortality in the United States despite by the American Association for the Study of Liver
recent advances in our understanding of its Diseases, American College of Gastroenterology,
pathophysiology and in treatment of chronic and the American Gastroenterological Association.
Hepatology. 2012;55(6):2005–23.
liver disease. The most common etiologies
12. Williams CD, et al. Prevalence of nonalcoholic
remain the same, however, incidence of NAFLD fatty liver disease and nonalcoholic steatohepatitis
has steadily increased in concert with the rise in among a largely middle-aged population utilizing
obesity and metabolic syndrome, making it the ultrasound and liver biopsy: a prospective study.
Gastroenterology. 2011;140(1):124–31.
most common cause of chronic liver disease and
13. Bellentani S, et al. Epidemiology of non-alcoholic
the second leading cause for transplant listing. fatty liver disease. Dig Dis. 2010;28(1):155–61.
Aggressive screening for mostly asymptomatic 14. Calzadilla Bertot L, Adams LA. The natural
liver disease to intervene and prevent the devel- course of non-alcoholic fatty liver disease. Int
J Mol Sci. 2016;17(5). https://doi.org/10.3390/
opment of end-­stage liver disease will be key to
ijms17050774.
combatting this liver disease epidemic. Once 15. Goh GB, McCullough AJ. Natural history of non-
cirrhosis occurs, novel antifibrotic therapies are alcoholic fatty liver disease. Dig Dis Sci. 2016;
promising and may soon revolutionize our abil- 61(5):1226–33.
16. O’Shea RS, Dasarathy S, McCullough AJ. Alcoholic
ity to treat these patients.
liver disease. Hepatology. 2010;51(1):307–28.
17. Teli MR, et al. Determinants of progression to cir-
rhosis or fibrosis in pure alcoholic fatty liver. Lancet.
1995;346(8981):987–90.
References 18. Leevy CM. Fatty liver: a study of 270 patients with
biopsy proven fatty liver and review of the literature.
1. Heron M. Deaths: Leading Causes for 2014. Natl Medicine. 1962;41:249–76.
Vital Stat Rep. 2016;65:1–96. 19. Sorensen TI, et al. Prospective evaluation of alco-
2. Scaglione S, et al. The epidemiology of cirrhosis in hol abuse and alcoholic liver injury in men as pre-
the United States: a population-based study. J Clin dictors of development of cirrhosis. Lancet. 1984;
Gastroenterol. 2015;49(8):690–6. 2(8397):241–4.
3. Graudal N, et al. Characteristics of cirrhosis undiag- 20. Lok AS, McMahon BJ. Chronic hepatitis B: update
nosed during life: a comparative analysis of 73 undi- 2009. Hepatology. 2009;50(3):661–2.
2  Chronic Liver Failure and Hepatic Cirrhosis 35

21. Blum HE. History and global burden of viral hepati- 41. Malhi H, Gores GJ. Mechanisms of liver injury.
tis. Dig Dis. 2016;34(4):293–302. In: Schiff ER, Maddrey WC, Sorrell MF, editors.
22. Chan SL, et al. Infection and cancer: the case of Schiff’s diseases of the liver. 11th ed. Chichester:
hepatitis B. J Clin Oncol. 2016;34(1):83–90. John Wiley and Sons; 2012. p. 216–31.
23. Lavanchy D, Mark K. Global epidemiology of hepa- 42. Lemasters JJ. V. Necrapoptosis and the mitochon-
titis B virus infection. In: Liaw Y-F, Zoulim F, edi- drial permeability transition: shared pathways to
tors. Hepatitis B virus in human diseases. Cham: necrosis and apoptosis. Am J Phys. 1999;276(1 Pt 1):
Springer International Publishing; 2016. G1–6.
24. Ott JJ, et al. Global epidemiology of hepatitis 43. Lemasters JJ. Dying a thousand deaths: redundant
B virus infection: new estimates of age-specific pathways from different organelles to apoptosis and
HBsAg seroprevalence and endemicity. Vaccine. necrosis. Gastroenterology. 2005;129(1):351–60.
2012;30(12):2212–9. 44. Malhi H, Gores GJ, Lemasters JJ. Apoptosis and
25. Wasley A, et al. The prevalence of hepatitis B virus necrosis in the liver: a tale of two deaths? Hepatology.
infection in the United States in the era of vaccina- 2006;43(2 Suppl 1):S31–44.
tion. J Infect Dis. 2010;202(2):192–201. 45. Faouzi S, et al. Anti-Fas induces hepatic chemo-
26. McMahon BJ. Chronic hepatitis B virus infection. kines and promotes inflammation by an NF-kappa
Med Clin North Am. 2014;98(1):39–54. B-independent, caspase-3-dependent pathway. J
27. Hyams KC. Risks of chronicity following acute Biol Chem. 2001;276(52):49077–82.
hepatitis B virus infection: a review. Clin Infect Dis. 46. Jaeschke H, Lemasters JJ. Apoptosis versus oncotic
1995;20(4):992–1000. necrosis in hepatic ischemia/reperfusion injury.
28. Benlloch S, Berenguer M. Chronic viral hepati- Gastroenterology. 2003;125(4):1246–57.
tis. Friedman SL, Keeffe EB, Dienstag JL, editors. 47. Ogasawara J, et al. Lethal effect of the anti-Fas anti-
Philadelphia: Elselvier Saunders; 2012. body in mice. Nature. 1993;364(6440):806–9.
29. Fattovich G, Bortolotti F, Donato F. Natural history 48. Matsumura H, et al. Necrotic death pathway in Fas
of chronic hepatitis B: special emphasis on dis- receptor signaling. J Cell Biol. 2000;151(6):1247–56.
ease progression and prognostic factors. J Hepatol. 49. Trautwein C, et al. Hepatic fibrosis: concept to treat-
2008;48(2):335–52. ment. J Hepatol. 2015;62(1 Suppl):S15–24.
30. Yang HI, et al. Hepatitis B e antigen and the risk 50. Bataller R, Brenner DA. Liver fibrosis. J Clin Invest.
of hepatocellular carcinoma. N Engl J Med. 2005;115(2):209–18.
2002;347(3):168–74. 51. Lee YA, Wallace MC, Friedman SL. Pathobiology
31. Chen CJ, Yang HI, Iloeje UH. Hepatitis B virus of liver fibrosis: a translational success story. Gut.
DNA levels and outcomes in chronic hepatitis 2015;64(5):830–41.
B. Hepatology. 2009;49(5 Suppl):S72–84. 52. Iwaisako K, et al. Origin of myofibroblasts in the
32. Chen JD, et al. Carriers of inactive hepatitis B virus fibrotic liver in mice. Proc Natl Acad Sci U S A.
are still at risk for hepatocellular carcinoma and liver- 2014;111(32):E3297–305.
related death. Gastroenterology. 2010;138(5):1747–54. 53. Lemoinne S, et al. Origins and functions of liver myo-
33. El-Serag HB. Epidemiology of viral hepatitis fibroblasts. Biochim Biophys Acta. 2013;1832(7):|
and hepatocellular carcinoma. Gastroenterology. 948–54.
2012;142(6):1264–1273.e1. 54. Mederacke I, et al. Fate tracing reveals hepatic stel-
34. Terrault NA, et al. AASLD guidelines for treatment of late cells as dominant contributors to liver fibro-
chronic hepatitis B. Hepatology. 2016;63(1):261–83. sis independent of its aetiology. Nat Commun.
35. Ditah I, et al. The changing epidemiology of hepa- 2013;4:2823.
titis C virus infection in the United States: National 55. Friedman SL. Mechanisms of hepatic fibrogenesis.
Health and Nutrition Examination Survey 2001 Gastroenterology. 2008;134(6):1655–69.
through 2010. J Hepatol. 2014;60(4):691–8. 56. Jarnagin WR, et al. Expression of variant fibronec-
36. Alter MJ. Epidemiology of hepatitis C virus infec- tins in wound healing: cellular source and biologi-
tion. World J Gastroenterol. 2007;13(17):2436–41. cal activity of the EIIIA segment in rat hepatic
37. Seeff LB. Natural history of chronic hepatitis fibrogenesis. J Cell Biol. 1994;127(6 Pt 2):
C. Hepatology. 2002;36(5 Suppl 1):S35–46. 2037–48.
38. Centers for Disease Control and Prevention. Recom- 57. Friedman SL. Hepatic fibrosis: emerging therapies.
mendations for prevention and control of hepatitis C Dig Dis. 2015;33(4):504–7.
virus (HCV) infection and HCV-­related chronic disease. 58. Bosch J, et al. Hepatic endothelial dysfunc-
MMWR Recomm Rep. 1998;47(RR-19):1–39. tion and abnormal angiogenesis: new targets in
39. Moyer VA. Screening for hepatitis C virus infec- the treatment of portal hypertension. J Hepatol.
tion in adults: U.S. Preventive Services Task Force 2010;53(3):558–67.
recommendation statement. Ann Intern Med. 2013; 59. Kawada N, et al. The contraction of hepatic stellate
159(5):349–57. (Ito) cells stimulated with vasoactive substances.
40. Surveillance for Viral Hepatitis – United States, 2014. Possible involvement of endothelin 1 and nitric
http://www.cdc.gov/hepatitis/statistics/2014surveillance/ oxide in the regulation of the sinusoidal tonus. Eur J
index.htm. Accessed 18 Aug 2016 Biochem. 1993;213(2):815–23.
36 L. T. Grinspan and E. C. Verna

60. Kaneda K, et al. Endothelin-1-induced vasoconstric- course, predictive factors and prognosis. Hepatology.
tion causes a significant increase in portal pressure 1994;20(6):1495–501.
of rat liver: localized constrictive effect on the distal 79. Tito L, et al. Recurrence of spontaneous bacterial
segment of preterminal portal venules as revealed by peritonitis in cirrhosis: frequency and predictive fac-
light and electron microscopy and serial reconstruc- tors. Hepatology. 1988;8(1):27–31.
tion. Hepatology. 1998;27(3):735–47. 80. Strauss RM, Boyer TD. Hepatic hydrothorax. Semin
61. Zhang JX, Pegoli W Jr, Clemens MG. Endothelin-1 Liver Dis. 1997;17(3):227–32.
induces direct constriction of hepatic sinusoids. Am 81. Xiol X, et al. Spontaneous bacterial empyema in
J Phys. 1994;266(4 Pt 1):G624–32. cirrhotic patients: a prospective study. Hepatology.
62. Gupta TK, et al. Endothelial dysfunction and 1996;23(4):719–23.
decreased production of nitric oxide in the intrahe- 82. D’Amico G, et al. Hepatic vein pressure gradient
patic microcirculation of cirrhotic rats. Hepatology. reduction and prevention of variceal bleeding in
1998;28(4):926–31. cirrhosis: a systematic review. Gastroenterology.
63. Tsochatzis EA, Bosch J, Burroughs AK. Liver cir- 2006;131(5):1611–24.
rhosis. Lancet. 2014;383(9930):1749–61. 83. Bosch J, Garcia-Pagan JC. Prevention of variceal
64. Giannini EG, Savarino V. Thrombocytopenia in liver rebleeding. Lancet. 2003;361(9361):952–4.
disease. Curr Opin Hematol. 2008;15(5):473–80. 84. Vilstrup H, et al. Hepatic encephalopathy in
65. Garcia-Tsao G, et al. Prevention and management of chronic liver disease: 2014 Practice Guideline by
gastroesophageal varices and variceal hemorrhage in the American Association for the Study of Liver
cirrhosis. Hepatology. 2007;46(3):922–38. Diseases and the European Association for the Study
66. Garcia-Tsao G, et al. Portal pressure, presence of of the Liver. Hepatology. 2014;60(2):715–35.
gastroesophageal varices and variceal bleeding. 85. Saunders JB, et al. A 20-year prospective study of
Hepatology. 1985;5(3):419–24. cirrhosis. Br Med J. 1981;282(6260):263–6.
67. Abraldes JG, et al. Hemodynamic response to 86. Romero-Gomez M, et al. Subclinical hepatic
pharmacological treatment of portal hypertension encephalopathy predicts the development of overt
and long-term prognosis of cirrhosis. Hepatology. hepatic encephalopathy. Am J Gastroenterol.
2003;37(4):902–8. 2001;96(9):2718–23.
68. Runyon BA. Introduction to the revised American 87. Jepsen P, et al. Clinical course of alcoholic liver
Association for the Study of Liver Diseases cirrhosis: a Danish population-based cohort study.
Practice Guideline management of adult patients Hepatology. 2010;51(5):1675–82.
with ascites due to cirrhosis 2012. Hepatology. 88. Coltorti M, et al. Liver cirrhosis in Italy. A multi-
2013;57(4):1651–3. centre study on presenting modalities and the impact
69. Runyon BA. Ascites and spontaneous bacterial perito- on health care resources. National Project on Liver
nitis. In: Feldman M, Friedman LS, Brandt LJ, editors. Cirrhosis Group. Ital J Gastroenterol. 1991;23(1):
Sleisenger and Fordtran’s gastrointestinal and liver 42–8.
disease. 9th ed. Philadelphia: Saunder Elsevier; 2010. 89. Bustamante J, et al. Prognostic significance of
70. Runyon BA. Management of adult patients with hepatic encephalopathy in patients with cirrhosis. J
ascites due to cirrhosis: an update. Hepatology. Hepatol. 1999;30(5):890–5.
2009;49(6):2087–107. 90. Lockwood AH. Blood ammonia levels and hepatic
71. Ge PS, Runyon BA. Treatment of patients with cir- encephalopathy. Metab Brain Dis. 2004;19(3–4):345–9.
rhosis. N Engl J Med. 2016;375(8):767–77. 91. Bass NM, et al. Rifaximin treatment in hepatic enceph-
72. Planas R, et al. Natural history of patients hospi- alopathy. N Engl J Med. 2010;362(12):1071–81.
talized for management of cirrhotic ascites. Clin 92. Rodriguez-Roisin R, Krowka MJ. Hepatopulmonary
Gastroenterol Hepatol. 2006;4(11):1385–94. syndrome—a liver-induced lung vascular disorder.
73. D’Amico G, Garcia-Tsao G, Pagliaro L. Natural his- N Engl J Med. 2008;358(22):2378–87.
tory and prognostic indicators of survival in cirrho- 93. Zhang XJ, et al. Intrapulmonary vascular dilatation
sis: a systematic review of 118 studies. J Hepatol. and nitric oxide in hypoxemic rats with chronic bile
2006;44(1):217–31. duct ligation. J Hepatol. 2003;39(5):724–30.
74. Runyon BA. Care of patients with ascites. N Engl J 94. Rodriguez-Roisin R, et al. Pulmonary-hepatic vas-
Med. 1994;330(5):337–42. cular disorders (PHD). Eur Respir J. 2004;24(5):
75. Gines P, et al. Management of cirrhosis and ascites. 861–80.
N Engl J Med. 2004;350(16):1646–54. 95. Corley DA, et al. Lack of efficacy of TIPS for hepatopul-
76. Such J, Runyon BA. Spontaneous bacterial peritoni- monary syndrome. Gastroenterology. 1997;113(2):
tis. Clin Infect Dis. 1998;27(4):669–74. quiz 675–6 728–30.
77. Hung TH, Tsai CC, Hsieh YH. The long-term mor- 96. Allgaier HP, et al. Hepato-pulmonary syndrome:
tality of spontaneous bacterial peritonitis in cirrhotic successful treatment by transjugular intrahepatic
patients: a 3-year nationwide cohort study. Turk J portosystemic stent-shunt (TIPS). J Hepatol.
Gastroenterol. 2015;26(2):159–62. 1995;23(1):102.
78. Follo A, et al. Renal impairment after spontaneous 97. Lasch HM, et al. Use of transjugular intrahepatic
bacterial peritonitis in cirrhosis: incidence, clinical portosystemic shunt as a bridge to liver transplanta-
2  Chronic Liver Failure and Hepatic Cirrhosis 37

tion in a patient with severe hepatopulmonary syn- 115. Nassar Junior AP, et al. Terlipressin versus norepi-
drome. Liver Transpl. 2001;7(2):147–9. nephrine in the treatment of hepatorenal syndrome:
98. Riegler JL, et al. Transjugular intrahepatic porto- a systematic review and meta-analysis. PLoS One.
systemic shunt improves oxygenation in hepatopul- 2014;9(9):e107466.
monary syndrome. Gastroenterology. 1995;109(3): 116. Singh V, et al. Noradrenaline vs. terlipressin in the
978–83. treatment of hepatorenal syndrome: a randomized
99. Selim KM, et al. Transjugular intrahepatic porto- study. J Hepatol. 2012;56(6):1293–8.
systemic shunt: a successful treatment for hepato- 117. Eason JD, et al. Proceedings of consensus confer-
pulmonary syndrome. Am J Gastroenterol. 1998; ence on simultaneous liver kidney transplantation
93(3):455–8. (SLK). Am J Transplant. 2008;8(11):2243–51.
100. Siramolpiwat S. Transjugular intrahepatic porto- 118. Nazar A, et al. LEFT ventricular function assessed
systemic shunts and portal hypertension-­related by echocardiography in cirrhosis: relationship to
complications. World J Gastroenterol. 2014;20(45): systemic hemodynamics and renal dysfunction. J
16996–7010. Hepatol. 2013;58(1):51–7.
101. Krowka MJ. Hepatopulmonary syndrome: recent lit- 119. Karagiannakis DS, et al. Diastolic cardiac dysfunc-
erature (1997 to 1999) and implications for liver trans- tion is a predictor of dismal prognosis in patients
plantation. Liver Transpl. 2000;6(4 Suppl 1):S31–5. with liver cirrhosis. Hepatol Int. 2014;8(4):588–94.
102. Pascasio JM, et al. Prevalence and severity of hepa- 120. Ruiz-del-Arbol L, et al. Diastolic dysfunction is a
topulmonary syndrome and its influence on survival predictor of poor outcomes in patients with cirrho-
in cirrhotic patients evaluated for liver transplanta- sis, portal hypertension, and a normal creatinine.
tion. Am J Transplant. 2014;14(6):1391–9. Hepatology. 2013;58(5):1732–41.
103. Hoeper MM, Krowka MJ, Strassburg CP. Portopul- 121. Cazzaniga M, et al. Diastolic dysfunction is asso-
monary hypertension and hepatopulmonary syn- ciated with poor survival in patients with cirrhosis
drome. Lancet. 2004;363(9419):1461–8. with transjugular intrahepatic portosystemic shunt.
104. Surani SR, et al. Pulmonary complications of hepatic Gut. 2007;56(6):869–75.
diseases. World J Gastroenterol. 2016;22(26): 122. Rabie RN, et al. The use of E/A ratio as a predic-
6008–15. tor of outcome in cirrhotic patients treated with
105. Kochar R, Nevah Rubin MI, Fallon MB. Pulmonary transjugular intrahepatic portosystemic shunt. Am J
complications of cirrhosis. Curr Gastroenterol Rep. Gastroenterol. 2009;104(10):2458–66.
2011;13(1):34–9. 123. Chen Y, et al. A detailed evaluation of cardiac
106. Budhiraja R, Hassoun PM. Portopulmonary hyper- function in cirrhotic patients and its alteration
tension: a tale of two circulations. Chest. 2003; with or without liver transplantation. J Cardiol.
123(2):562–76. 2016;67(2):140–6.
107. Halank M, et al. Portopulmonary hypertension. J 124. Mohamed R, et al. Effect of liver transplantation
Gastroenterol. 2006;41(9):837–47. on QT interval prolongation and autonomic dys-
108. Swanson KL, et al. Survival in portopulmonary function in end-stage liver disease. Hepatology.
hypertension: Mayo Clinic experience categorized by 1996;23(5):1128–34.
treatment subgroups. Am J Transplant. 2008;8(11): 125. Torregrosa M, et al. Cardiac alterations in cirrhosis:
2445–53. reversibility after liver transplantation. J Hepatol.
109. Salerno F, et al. Diagnosis, prevention and treat- 2005;42(1):68–74.
ment of hepatorenal syndrome in cirrhosis. Gut. 126. Tsochatzis EA, et al. Systematic review: portal vein
2007;56(9):1310–8. thrombosis in cirrhosis. Aliment Pharmacol Ther.
110. Fede G, et al. Renal failure and cirrhosis: a system- 2010;31(3):366–74.
atic review of mortality and prognosis. J Hepatol. 127. Francoz C, et al. Splanchnic vein thrombosis in can-
2012;56(4):810–8. didates for liver transplantation: usefulness of screen-
111. Angeli P, et al. Reversal of type 1 hepatorenal syn- ing and anticoagulation. Gut. 2005;54(5):691–7.
drome with the administration of midodrine and 128. Amitrano L, et al. Risk factors and clinical presenta-
octreotide. Hepatology. 1999;29(6):1690–7. tion of portal vein thrombosis in patients with liver
112. Esrailian E, et al. Octreotide/Midodrine therapy sig- cirrhosis. J Hepatol. 2004;40(5):736–41.
nificantly improves renal function and 30-day sur- 129. Amitrano L, et al. Inherited coagulation disorders
vival in patients with type 1 hepatorenal syndrome. in cirrhotic patients with portal vein thrombosis.
Dig Dis Sci. 2007;52(3):742–8. Hepatology. 2000;31(2):345–8.
113. Alessandria C, et al. Noradrenalin vs terlipressin 130. Nery F, et al. Causes and consequences of por-
in patients with hepatorenal syndrome: a prospec- tal vein thrombosis in 1,243 patients with cirrho-
tive, randomized, unblinded, pilot study. J Hepatol. sis: results of a longitudinal study. Hepatology.
2007;47(4):499–505. 2015;61(2):660–7.
114.
Ghosh S, et al. Noradrenaline vs terlipres- 131. Zocco MA, et al. Thrombotic risk factors in patients
sin in the treatment of type 2 hepatorenal syn- with liver cirrhosis: correlation with MELD scoring
drome: a randomized pilot study. Liver Int. system and portal vein thrombosis development. J
2013;33(8):1187–93. Hepatol. 2009;51(4):682–9.
38 L. T. Grinspan and E. C. Verna

132. American Cancer Society. Cancer facts and figures 150. Hwang JH, et al. The role of endoscopy in the
2016. Atlanta, GA: American Cancer Society; 2016. management of variceal hemorrhage. Gastrointest
133. El-Serag HB, Kanwal F. Epidemiology of hepatocel- Endosc. 2014;80(2):221–7.
lular carcinoma in the United States: where are we? 151. Marcellin P, et al. Regression of cirrhosis during
Where do we go? Hepatology. 2014;60(5):1767–75. treatment with tenofovir disoproxil fumarate for
134. Ferlay J, et al. Cancer incidence and mortality chronic hepatitis B: a 5-year open-label follow-up
worldwide: sources, methods and major patterns in study. Lancet. 2013;381(9865):468–75.
GLOBOCAN 2012. Int J Cancer. 2015;​ 152. Xu B, et al. Long-term lamivudine treatment
136(5):E359–86. achieves regression of advanced liver fibrosis/
135. N.A. A new prognostic system for hepatocellular cirrhosis in patients with chronic hepatitis B. J
carcinoma: a retrospective study of 435 patients: the Gastroenterol Hepatol. 2015;30(2):372–8.
Cancer of the Liver Italian Program (CLIP) investi- 153. Buti M, et al. Long-term clinical outcomes in cir-
gators. Hepatology 1998;28(3):751–5. rhotic chronic hepatitis B patients treated with teno-
136. Andersson KL, et al. Cost effectiveness of alterna- fovir disoproxil fumarate for up to 5 years. Hepatol
tive surveillance strategies for hepatocellular carci- Int. 2015;9(2):243–50.
noma in patients with cirrhosis. Clin Gastroenterol 154. D’Ambrosio R, et al. A morphometric and immuno-
Hepatol. 2008;6(12):1418–24. histochemical study to assess the benefit of a sustained
137. Bruix J, Sherman M. Management of hepatocellu- virological response in ­ hepatitis C virus patients
lar carcinoma: an update. Hepatology. 2011;53(3): with cirrhosis. Hepatology. 2012;56(2):532–43.
1020–2. 155. Hezode C, et al. Liver stiffness diminishes with
138. Trojan J, Zangos S, Schnitzbauer AA. Diagnostics antiviral response in chronic hepatitis C. Aliment
and treatment of hepatocellular carcinoma in Pharmacol Ther. 2011;34(6):656–63.
2016: standards and developments. Visc Med. 156. Falize L, et al. Reversibility of hepatic fibrosis in
2016;32(2):116–20. treated genetic hemochromatosis: a study of 36
139. Adhoute X, et al. Usefulness of staging systems and cases. Hepatology. 2006;44(2):472–7.
prognostic scores for hepatocellular carcinoma treat- 157. Czaja AJ, Carpenter HA. Decreased fibrosis during
ments. World J Hepatol. 2016;8(17):703–15. corticosteroid therapy of autoimmune hepatitis. J
140. Bruix J, Reig M, Sherman M. Evidence-based diagno- Hepatol. 2004;40(4):646–52.
sis, staging, and treatment of patients with hepatocellu- 158. Kaplan MM, DeLellis RA, Wolfe HJ. Sustained bio-
lar carcinoma. Gastroenterology. 2016;150(4):835–53. chemical and histologic remission of primary bili-
141. Bhardwaj N, Perera MT, Silva MA. Current treatment ary cirrhosis in response to medical treatment. Ann
approaches to HCC with a special consideration to Intern Med. 1997;126(9):682–8.
transplantation. J Transplant. 2016;2016:7926264. 159. Deniz K, et al. Regression of steatohepatitis-related
142. Ripoll C, et al. Hepatic venous pressure gradient cirrhosis. Semin Liver Dis. 2015;35(2):199–202.
predicts clinical decompensation in patients with 160. Yoon YJ, Friedman SL, Lee YA. Antifibrotic thera-
compensated cirrhosis. Gastroenterology. 2007; pies: where are we now? Semin Liver Dis. 2016;
133(2):481–8. 36(1):87–98.
143. Child CG, Turcotte JG. Surgery and portal hyperten- 161. Chang TT, et al. Long-term entecavir therapy results
sion. Major Probl Clin Surg. 1964;1:1–85. in the reversal of fibrosis/cirrhosis and continued
144. Cholongitas E, et al. Systematic review: the model for histological improvement in patients with chronic
end-stage liver disease—should it replace Child-Pugh’s hepatitis B. Hepatology. 2010;52(3):886–93.
classification for assessing prognosis in cirrhosis? 162. Dixon LJ, et al. Caspase-1-mediated regulation of
Aliment Pharmacol Ther. 2005;22(11–12):1079–89. fibrogenesis in diet-induced steatohepatitis. Lab
145. Martin P, et al. Evaluation for liver transplantation Invest. 2012;92(5):713–23.
in adults: 2013 practice guideline by the American 163. Pockros PJ, et al. Oral IDN-6556, an antiapoptotic
Association for the Study of Liver Diseases and the caspase inhibitor, may lower aminotransferase activ-
American Society of Transplantation. Hepatology. ity in patients with chronic hepatitis C. Hepatology.
2014;59(3):1144–65. 2007;46(2):324–9.
146. Malinchoc M, et al. A model to predict poor survival 164. Baskin-Bey ES, et al. Cathepsin B inactivation
in patients undergoing transjugular intrahepatic por- attenuates hepatocyte apoptosis and liver damage
tosystemic shunts. Hepatology. 2000;31(4):864–71. in steatotic livers after cold ischemia-warm reperfu-
147. Kamath PS, et al. A model to predict survival in sion injury. Am J Physiol Gastrointest Liver Physiol.
patients with end-stage liver disease. Hepatology. 2005;288(2):G396–402.
2001;33(2):464–70. 165. Abu-Elheiga L, et al. Acetyl-CoA carboxylase 2−/−
148. Wiesner RH, et al. MELD and PELD: application mutant mice are protected against fatty liver under high-
of survival models to liver allocation. Liver Transpl. fat, high-carbohydrate dietary and de novo lipogenic
2001;7(7):567–80. conditions. J Biol Chem. 2012;287(15):12578–88.
149. Wiesner R, et al. Model for end-stage liver dis- 166. Baeck C, et al. Pharmacological inhibition of the
ease (MELD) and allocation of donor livers. chemokine C-C motif chemokine ligand 2 (mono-
Gastroenterology. 2003;124(1):91–6. cyte chemoattractant protein 1) accelerates liver
2  Chronic Liver Failure and Hepatic Cirrhosis 39

fibrosis regression by suppressing Ly-6C(+) mac- 168. Kisseleva T, et al. Myofibroblasts revert to an inac-
rophage infiltration in mice. Hepatology. 2014; tive phenotype during regression of liver fibrosis.
59(3):1060–72. Proc Natl Acad Sci U S A. 2012;109(24):9448–53.
167. Sanyal AJ, et al. Pioglitazone, vitamin E, or pla-
cebo for nonalcoholic steatohepatitis. N Engl J Med.
2010;362(18):1675–85.
Acute Hepatic Failure
3
Andrew Slack, Brian J. Hogan, and Julia Wendon

Keywords with expertise in the management of ALF and


Acute hepatic failure · Intracranial hyperten- liver transplantation (LT) is crucial.
sion · Epidemiology · Paracetamol · King’s ALF is rare with around 2800 and 400 cases of
College Criteria · Plasma exchange ALF per year in the United Stated (US) and the
United Kingdom (UK), respectively [1]. There
are multiple etiologies of ALF that vary with
Introduction worldwide geographical location, clinical pre-
sentation, time course, and prognosis. In the
Acute liver failure (ALF) is an unpredictable and developing world the leading cause of ALF are
rapidly progressive, life-threatening multisystem the viral hepatitides, particularly Hepatitis B. In
condition that ensues when an insult causes dif- the US and the UK, drug induced liver injury,
fuse necrosis of liver parenchyma disrupting particularly paracetamol (acetominophen) over-
hepatocyte function in patients who have no pre- dose and sero-negative hepatitis have emerged as
existing liver injury. The subsequent develop- the leading causes (Fig. 3.1) [1, 2].
ment of encephalopathy and coagulopathy within The prognosis of ALF depends on age, etiol-
days or weeks represents the key features of ALF, ogy, and the time course over which the disease
but critically often culminates with multi-organ evolves. Survival rates vary significantly by etiol-
failure (MOF), which impacts significantly on ogy and have improved to around 60% overall
mortality. Timely referral to specialist centers without LT and over 85% with LT [3].
Improvement of survival rates over recent
decades is related to improved critical care man-
agement, better prognostic assessment, and the
timely prioritization of patients for LT. The man-
agement of ALF is focused on support of all
A. Slack, MBBS, MRCP, EDIC, MD(Res) organ systems and the prevention and treatment
Department of Critical Care, Guy’s and St Thomas’s of complications, particularly sepsis. Liver necro-
NHS Foundation Trust, London, UK
sis acts as a focus of inflammation, driving vaso-
B. J. Hogan, BSc, MBBS, MRCP, FEBTM, FFICM plegia and leading to cardiovascular collapse,
J. Wendon, MbChB, FRCP (*)
which exacerbates dysfunction of other vital
Institute of Liver Studies, Kings College London,
Kings College Hospital, London, UK organs, particularly the kidney and brain. The
e-mail: julia.wendon@kcl.ac.uk identification and treatment of the cause of the

© Springer International Publishing AG, part of Springer Nature 2018 41


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_3
42 A. Slack et al.

underlying liver injury should be the primary [5], the decision of who and when to transplant
goal, with a concurrent focus on the optimization is complex. There is emerging support for
of the circulation to promote hepatocellular delaying transplant if the clinical situation is
regeneration and to prevent further insult due to improving in patients with a favorable etiology
ischemic injury. However, despite such endeav- [6]. The option of an auxiliary transplant graft
ors timely recognition that hepatic regeneration is sometimes considered as it allows native
will ultimately not be sufficient is crucial. Under regeneration and withdrawal of immunosup-
these circumstances, Liver transplantation with pression, but due to the increased risk of early
removal of the necrotic liver mass offers the best postoperative complications it necessitates
chance of survival. The decision to prioritize for careful scrutiny of appropriate potential
transplantation requires a multidisciplinary team candidates.
approach incorporating specialist liver transplant
surgeons, hepatologists, and intensivists who can
utilize established prognostic criteria along with
the daily assessment of the levels of organ sup- Classification of ALF
port to best determine which patients are likely to
benefit from being listed for transplant with high The classifications for ALF have evolved since
priority and indeed proceeding to OLT if levels of the initial definition by Trey and Davidson in
organ support permit (Fig. 3.2) [4]. 1970 in an attempt to reflect the impact that both
The availability of donor organs is under etiology and the existence of chronic liver dis-
continued pressure in the UK and worldwide. ease have on prognosis. The two most common
Patients with ALF must fulfill a strict set of definitions concentrate on the time period from
selection criteria based on published risk fac- jaundice to the onset of encephalopathy [1]. This
tors for prioritization before being listed on the classification is important, because the hyper-
national super-urgent transplantation waiting acute forms of ALF including acetominophen
list (Table 3.1). These patients are then strati- overdose and Hepatitis A are associated with
fied by blood group and time while on the mortality due to cerebral edema and kidney
super-urgent waiting list. In most cases a donor injury. However, survival without transplanta-
organ should be available within 48–72 h. tion for this group is superior compared to more
Occasionally an ABO incompatible donor indolent subacute causes, including sero-nega-
organ needs to be considered in light of the tive and idiosyncratic drug reactions (Fig. 3.2)
unavailability of an ABO compatible organ [4, 3]. These etiologies are not as frequently
weighed against the projected deterioration of complicated by the cerebral and renal insults, but
the clinical condition. The currently available carry a higher mortality burden compared to
selection criteria are imperfect and when cou- hyperacute causes (Table 3.1) [2] (Fig. 1.3).
pled with improving transplant free survival
rates, particularly for acetominophen overdose

Table 3.1  Classifications of ALF (time from jaundice to onset of encephalopathy)


Definition Time (days) Most common etiologies Definition Time (weeks)
Hyperacute <7 days POD, hepatitis A and B Fulminant <2
Acute 8–28 days Hepatitis A, B, E, idiosyncratic drug reactions
Subacute 29 days to 8 Idiosyncratic drug reactions, sero-negative Subfulminant >2
weeks hepatitis
3  Acute Hepatic Failure 43

Fig. 3.1 Overall 475


n 480
comparison of etiologies 450
observed among 1033 420
patients with acute liver 390
failure (ALF) in the 360
ALF study Group 330
registry, 1990–2004. A 300
270
preponderance of 240
acetaminophen cases is 210
observed approaching 180 151
50% 150 119
120
90 75
60 55 43 50
31
30 17 9 8
0
M

ug

ri
B

y
A

mm

’s

r
he

ete
mi

nc
hia
on
AC

p
Dr

Ot
He

He

he

na
toi

Ind
ils

-C
Isc

eg
Au

dd

Pr
Bu
Fig. 3.2  Survival for
different etiologies of Survival for different etiologies of acute liver failure
ALF. Organ support to
best determine which
patients are likely to
benefit from being listed Pregnancy related 98%
for transplant with high
priority and indeed Hepatitis A 67%
proceeding to OLT if
levels of organ support Acetaminophen 56%
permit [4]
Hepatitis B 38%

Seronegative 20%
hepatitis
Idiosyncratic drug
2.5%
reactions

Wilson disease <1%

50% 100%
Survival from grade 3–4 encephalopathy

Etiologies of ALF in the UK sought to limit the availability of


paracetamol. Legislation was changed in line
Paracetamol (Acetominophen) with World Health Organization recommenda-
Overdose tions and data from other countries with similar
restrictive policies that had lower rates of
Paracetamol overdose (POD) in the UK had been paracetamol-induced hepatoxicity. Suicidal or
increasing steadily likely due to its easy avail- para-suicidal actions are usually impulsive acts in
ability [7]. In 1998 the Medicine Control Agency reaction to crises; therefore, it was postulated that
44 A. Slack et al.

limiting supply would result in reduced availabil- unstaggered ingestion of paracetamol, NAC can
ity of paracetamol, thus reducing the quantity prevent hepatocellular damage.
ingested and lowering rates of hepatoxicity. The A accurate and precise history regarding the
general sale of paracetamol was restricted to six- timing and quantity of paracetamol ingested is
teen 500 mg tablets, a total of 8 g per packet. important, as is establishing whether the inges-
Reports suggested that this was a successful pol- tion was staggered. However, the circumstances
icy that resulted in a reduction of intensive care that surround any parasuicidal event can make
admissions and of deaths from POD by 43% in this information difficult to establish, especially
the years following the l­egislation [8], despite if patients have ingested opiate-based medication
some debate on the true mortality benefit of pack in addition to paracetamol or are intoxicated with
size reduction alone [9]. alcohol. Additionally, an assessment of potentiat-
In the UK POD comprises up to 50% of all ing factors that lower hepatic glutathione levels
poisoning admissions, compared to only 10% in or increase cytochrome P450 enzyme activity
the US [10]. Due to a combination of the small and increase hepatoxicity should be undertaken.
doses absorbed and the efficacy of early antidote These factors include anorexia nervosa, malnutri-
therapy, only 0.6% of these cases result in hepa- tion, chronic alcohol consumption, and enzyme
toxicity in the UK. Studies assessing the rate of inducing drugs such as phenytoin and
deliberate versus accidental POD display geo- carbamazepine.
graphic variation. In Europe, studies have In an unstaggered overdose presenting within
reported around 86% of POD cases were delib- 24 h a paracetamol level should be measured and
erate and 14% were accidental [11], while US applied to the revised paracetamol poisoning
poisons center data have reported rates of 35% treatment graph. A paracetamol level of more
and 65%, respectively [12]. Paracetamol medi- than 150 mg/kg is generally considered to be
cations combined with narcotics pose a poten- hepatotoxic, though strong evidence ratifying
tial for unintentional hepatoxicity when this is lacking. In a staggered overdose the
addiction to the narcotic within such combined paracetamol level cannot be interpreted and one
analgesics leads to a gradual increase of the must assess the risk of hepatoxicity based on total
ingested dose [13]. This may be a significant dose alone. If there is any doubt about timing or
reason for the discrepancy between the US and if there was a delay in presentation, treatment
the UK with regard to deliberate and uninten- should be commenced until it becomes clear that
tional overdose. The assessment of the risk of hepatotoxicity is unlikely. Patients presenting
developing ALF from POD, whether accidental within 24 h of ingestion without signs of hepato-
or deliberate, is closely related to the total dose toxicity can be managed on the wards, while
ingested, as well as the time from ingestion to those with features of paracetamol-induced hepa-
presentation and treatment with N-acetylcysteine toxicity should be managed in a critical care
(NAC). environment.
Higher doses and prolonged time to NAC
result in increased length of time exposed to the
active unstable paracetamol metabolite, N-acetyl Viral Hepatitis
p-benzoquinone imine (NAPQI). NAPQI
depletes hepatic glutathione levels, with ensuing All hepatitides except for Hepatitis C have been
hepatocellular damage, unless the antidote, the implicated in cases of ALF [1]. Viral hepatitis A
glutathione precursor NAC or methionine is and B are the most common causes of ALF
given in a timely fashion. NAC acts to augment worldwide including France and Japan; Hepatitis
the glutathione reserves in the body, which E is predominant in India.
directly bind to toxic metabolites and protect The risk of ALF is lowest with Hepatitis A at
hepatocytes in the liver from NAPQI toxicity. less than 0.35%, but this risk increases with age
When administered within 12 h of an at the time of exposure. In the western world, it
3  Acute Hepatic Failure 45

appears that native immunity to Hepatitis A is Idiosyncratic Drug Reaction


decreasing. In the US the incidence of ALF due
to Hepatitis A is around 3.1% with around 0.12% The administration of drugs directly affects the
of all cases listed for liver transplantation. In the liver and may cause toxicity as the liver is the
developed world the incidence of Hepatitis A has primary site of metabolism and elimination. In
been decreasing since 1995 likely due to vaccina- the US, hepatoxicity is the main cause for halt-
tion of high risk patients, improved sanitation, ing drug development and withdrawal from the
and improved food preparation techniques [14]. market. Drug-induced liver injury (DILI)
The treatment of Hepatitis A is largely including cases of paracetamol toxicity, is the
supportive. leading cause of ALF and indication for liver
Hepatitis B infection is the cause of ALF in transplantation. The majority of non-
around 1% of all cases with over 50% associated paracetamol DILI cases are idiosyncratic reac-
with hepatitis D co-infection. The mortality of tions that occur in around 1 in 10,000 of exposed
patients developing ALF ranges from 70 to 80% patients. More than 1000 drugs and herbal rem-
[15]. Hepatitis B has eight genotypes A–H and edies have been implicated and idiosyncratic
all have been associated with different clinical reactions comprise 10% of ALF cases [19].
presentations. Antiviral therapy with nucleos(t) Idiosyncratic DILI is a complex phenomenon
ide analogues can alter the outcome in ALF and that appears to be closely related to how cell
is recommended in potential transplant candi- mitochondria balance cellular injury and regen-
dates [16]. eration. The reactions are idiosyncratic as liver
Hepatitis E is common in Asia and Africa injury is unpredictable and not dose-dependant.
with the risk of ALF greatest during pregnancy There are non-allergic and allergic idiosyncratic
(greater than 20%), particularly during the third DILI, the latter characterized by fever, skin
trimester. In the general population, Hepatitis E reactions, eosinophilia with the formation of
carries a low mortality of 0.5–4%, but it can autoantibodies (for example drug-­related eosin-
exceed 75% in developing countries especially ophilic syndrome—DRESS). Several risk fac-
during the second and third trimester of preg- tors for DILI have been identified and include
nancy. It is transmitted by the fecal-oral route, age, female gender, concomitant diseases, and
often through contaminated water supply. specific drugs. DILI algorithms and clinical
Consequently, it has been the cause of epidemics scales may improve consistency and aid the cli-
in Asia, China, and Eastern Europe especially nicians to determine the causality of adverse
after heavy rainfall. The first such epidemics to drug reactions [20].
be documented occurred in New Delhi, India in Genetic polymorphisms have been associ-
1955 and affected 29,000 people [17]. ated with increased risk of DILI, for example,
Viruses including cytomegalovirus (CMV), cytokine polymorphism causing diclofenac
Epstein Barr virus, herpes viruses type 1, 2 and 6, hepatoxicity. Genetic variations are also
and varicella zoster have all been implicated in involved in genetic deficiency of mitochondrial
cases of ALF, frequently in profoundly immuno- long-chain 3-hydroxyacyl-CoA dehydrogenase
compromised patients. Falciparum malaria has that is associated with acute fatty liver of preg-
also been reported as a cause of ALF, primarily in nancy, presumably related to increased levels of
India. The mortality associated with atypical female sex hormones. DILI is commonly diag-
viral hepatitis is around 76% and for falciparum nosed primarily by increased levels of alanine
malaria 24% [1]. Antiviral therapies may be ben- transferase (ALT) and gamma-glutamyl trans-
eficial in some cases of ALF such as nucleos(t) ferase (GGT). Metabolomic studies are cur-
ide analogues for hepatitis B, ribavirin for hepati- rently conducted to identify biomarkers of DILI
tis E [18] and acyclovir and valganciclovir for that will detect injury prior to elevations in
herpes and CMV disease. ALT.
46 A. Slack et al.

Seronegative (Indeterminate) metastatic and lymphomatous infiltration of the


liver. There are no specific biomarkers for tumour
Seronegative ALF is the second most frequent eti- infiltration; elevations of ALT and AST with
ology of ALF (after DILI) with 10–20% of all tumors are usually lower that than with ischemic
cases. With seronegative ALF no definite causes hepatitis. Both appear to have greater sensitivity
of ALF can be found, but a predictable clinical in the presence of hyperbilirubinemia. However,
course is observed characterized in sub-acute liver jaundice does not always manifest in the setting
failure due to hepatic necrosis with loss of liver of tumor infiltration and cases with over 90%
volume and progressive coagulopathy. Patients liver infiltration without jaundice have been
with seronegative ALF often fulfill standard crite- reported. A transjugular liver, bone marrow aspi-
ria late in the course of their illness, as overt ration, and trephine (bone marrow) or lymph
hepatic encephalopathy is a late feature that may node biopsy can be invaluable tools for establish-
occur after many weeks of gradual clinical deterio- ing a diagnosis. Confirmed malignancy is a abso-
ration. In the future assessment using MELD score lute contraindication for liver transplantation,
or liver volumes (as described below) may allow a establishing a diagnosis is therefore crucial.
better prediction of poor prognosis in this group
and facilitate more timely access to LT.
 ascular Insults and Ischemic
V
Hepatitis
Malignancy
ALF following vascular insults are uncommon.
There are numerous case reports of a wide range These include ischemic hepatitis often associated
of solid and hematological tumors that can cause with low cardiac output due to left and right ven-
ALF. A literature review in 2005 cited 34 cases of tricular cardiac dysfunction. Veno-occlusive dis-
primary and metastatic neoplastic infiltration of orders, such as Budd-Chiari syndrome (BCS) are
the liver resulting in ALF [21]. The pathophysiol- also a rare cause of ALF with an incidence of 1 in
ogy of ALF in neoplastic infiltration is multifac- 2.5 million [24]. BCS is characterized by hepatic
torial. Parenchymal ischemia and infarction can venous outflow obstruction and presents with
be caused by diffuse tumor cell infiltration or vas- ALF in around 20% of cases. In the western
cular occlusion from tumor thrombi. It has also world occlusion of the hepatic veins is commonly
been postulated that diffuse tumor cell infiltration due to thrombosis whereas in Asia a membra-
renders the remaining liver parenchyma highly nous web is the most frequent cause. Both inher-
susceptible to ischemic injury. A case series of ited and acquired procoagulant conditions have
three patients with metastatic disease demon- been implicated in Budd-Chiari and often both
strated biopsy-proven hepatic ischemia in the conditions coexist. Veno-occlusive disorders
absence of any discernible episodes of systemic have been associated with inherited conditions
hypotension [21, 22]. Additionally, cytokine-­ such as Factor V Leiden, Protein C, S and anti-
mediated liver injury has been implicated in lym- thrombin deficiency; acquired conditions include
phomatous infiltration [23]. Clinical suspicion paroxysmal nocturnal hemoglobinuria and anti-­
and features suggestive of malignancy such as phospholipid syndrome. The recently discovered
hepatomegaly, enlarged lymph nodes on physical Janus Kinase 2 (JAK-2) mutation has also been
examination along with computer tomography detected in around 40–59% of cases with BCS
(CT) findings suggestive of an infiltrative process [25]. Myeloproliferative disorders also need to be
should prompt an attempt to obtain a biopsy for a ruled out as a cause with an examination of the
definitive histological diagnosis. Radiological bone marrow function using a trephine (bone
imaging including both ultrasonography and tri- marrow) biopsy and aspiration as these disorders
ple phase computer tomography should not solely are most commonly associated with both BCS
be relied on due to their poor sensitivity for and portal vein thrombosis [24].
3  Acute Hepatic Failure 47

Metabolic corticosteroid therapy can be challenging and


MELD-Na score and the UK end stage liver dis-
ALF secondary to inherited and acquired meta- ease score (UKELD) on day 7 after onset may the
bolic disorders is uncommon, these include acute best predictors of failure of standard medical
fatty liver of pregnancy, fructose intolerance, therapy [28]. Patients with hepatic encephalopa-
galactosemia, lecithin-cholesterol acyltransfer- thy or who do not respond rapidly to corticoste-
ase deficiency, Reye’s syndrome, tyrosinemia, roid therapy should be assessed early for LT [28].
and Wilson’s disease (WD).
WD is a rare autosomal recessive condition
caused by a mutation to the WD gene ATP7B that Miscellaneous
encodes a copper transporting P-type ATPase
leading to insufficient copper excretion into bile Other rare causes of ALF include HELLP
and subsequent copper accumulation in brain, (Hemolysis, Elevated Liver enzymes, and Low
liver, and corneas. The incidence of WD is Platelets) syndrome of pregnancy. Amphetamine
approximately 1 in 30,000 and can present derivatives such as 3,4-­methylenedioxymethamp
acutely, usually in pediatric or young female hetamine (MDMA or “ecstasy”) have caused a
patients, or chronically in adults sometimes into number of cases of ALF requiring OLT. Toxins of
their eighth decade of life. ALF in WD is unique mushrooms such as Amanita phalloides or food-
as there is usually some preexisting liver disease borne illnesses by Bacillus cereus are also poten-
at the time when ALF ensues. WD is diagnosed tial causes of ALF.
by measuring indices of copper metabolism,
although in ALF these investigations can be mis-
leadingly normal. Serum copper and caeruloplas-  linical Features and General
C
min, as an acute phase protein, can both be Management
normal or elevated in other causes of
ALF. Elevated levels of urinary copper are a good The identification of the underlying insult is cru-
indicator of WD, but the high incidence of anuric cial in determining potential therapies that could
acute kidney injury in ALF may eliminate this halt the injurious process and potentially reverse
diagnostic tool. Ophthalmic exam of corneas can liver failure. Laboratory investigations should
be useful to detect the presence of Kayser- include hepatitis and atypical viral serology;
Fleischer rings, which in combination with liver autoantibodies (antinuclear, anti-smooth muscle,
disease and copper metabolism abnormalities anti-liver kidney microsomal, anti-soluble liver
strongly supports the diagnosis. Additionally, antigen and anti-mitochondrial antibodies),
Coombs negative hemolytic anemia and low paracetamol levels and urine and serum copper
serum cholinesterase levels can be a feature of levels. A negative paracetamol level does not rule
WD [26]. The ALP/bilirubin and AST/bilirubin out paracetamol as a cause of ALF. Additionally
ratios are often significantly lower in fulminant ultrasonography of the liver and its vasculature
Wilson’s disease than in other categories of ful- should be performed as we as axial imaging with
minant liver failure but this is not necessarily computer tomography if the history and labora-
diagnostic [27]. tory investigations do not confirm viral or drug-­
induced insults. The outcome is largely
determined by the severity of the underlying liver
Autoimmune Hepatitis insult and the development of organ failure; any
episodes of sepsis have a further strong impact on
Twenty percent of patients with autoimmune mortality. (Table 3.1) Early recognition and treat-
hepatitis present with acute liver injury and a pro- ment of sepsis and the prevention and support of
portion of these will go on to develop sub-acute organ dysfunction is therefore key to gain time
ALF. The decision on whether to initiate for hepatic regeneration. Finally, a timely
48 A. Slack et al.

decision for super-­urgent liver transplantation is substantially elevated central venous pressures
required when it becomes sufficiently clear that should prompt further evaluation of myocardial
hepatic regeneration will not occur in time. This function with echocardiography to evaluate left
decision carries particular importance given that and right ventricular filling and function.
the median time from listing to transplantation is Early admission to critical care environment is
around 48 h in the UK. 24% of patients who are recommended to detect and treat rapidly occur-
listed will never receive a transplant; the majority ring deteriorations of clinical condition; patients
(92%) of these patients will die while waiting for with ALF and organ dysfunction should be cared
an organ and the remainder will become “too for in a critical care unit. Some commonly used
sick” for transplantation [29]. Several pre-trans- resuscitation parameters may be problematic in
plant factors have been associated with poor out- ALF; for example ScvO2 is often significantly
comes after LT for ALF such as age >45–50 years, elevated reflecting the hyperdynamic circulation
escalating vasopressor requirements, the use of and microvascular shunting. Lactate concentra-
high-risk organs and ABO-incompatibility [29, tions in ALF may reflect sole circulatory disarray
30]. Other factors should also prompt a discus- but also impaired hepatic clearance of lactate
sion about the suitability for transplantation especially if adequate volume resuscitation has
include fixed dilated pupils for greater than 2 h, been implemented. Hyperlactatemia reflects
necrotizing pancreatitis, severe adult respiratory liver, circulatory and cellular dysfunction; how-
distress syndrome (ARDS), moderate to severe ever the liver has a large reserve for lactate
pulmonary hypertension, culture proven bacterial metabolism. Even after hepatectomy with resec-
or fungal sepsis requiring more than 24 h of anti- tion of more than 50% of the liver lactate levels
microbial therapy before transplantation. All may remain normal [32]. High lactate levels are
these conditions need to be evaluated in relation therefore frequently encountered in ALF where
to age and the degree of associated organ failures. inadequate fluid resuscitation has lead to circula-
The complex nature of ALF requires the involve- tory and cellular metabolism dysfunction. In gen-
ment of wide spectrum of expertise to form a eral hyperlactatemia and the speed of its
cohesive multidisciplinary team. Critical care resolution acts as an important predictor of out-
nurses, physiotherapists, pharmacists, transplant come in both critical illness and ALF [33].
surgeons, hepatologists and liver intensivists Lactate is now recognized as an important prog-
should all be included in this team. nostic variable. Persistently elevated lactate lev-
els >3.0 mmol/L despite aggressive fluid
resuscitation [34] have been incorporated into the
Cardiovascular Kings College Criteria (KCC) adding statistical
strength to the original O’Grady criteria [35].
The circulatory symptoms of established ALF In ALF relative corticosteroid insufficiency,
mirror the hemodynamic changes of sepsis with defined by an abnormal response to adrenocorti-
an elevated cardiac output and vasoplegia. The cotrophic hormone, has a prevalence of 62% and
main vasoactive mediator, nitric oxide, causes steroid replacement therapy is associated with
regional vasodilatation primarily in the splanch- reductions in vasopressor requirements, albeit
nic bed. The management goals for the circula- without any mortality benefit [36, 37]. The diag-
tion in established ALF are similar to the nosis and treatment of critical illness related cor-
recommendations for initial resuscitation in sep- ticosteroid insufficiency (CIRCI) was first
tic shock [31]. The early use of invasive hemody- encountered in sepsis with the demonstration that
namic monitoring is recommended as it may low dose hydrocortisone could accelerate the
provide important additional clinical indices reversal of shock, but no significant effect on
about central circulating volumes and cardiac mortality [38]. The high prevalence of CIRCI in
output. Furthermore, a normal cardiac output (i.e. ALF can be explained by factors that affect corti-
abnormally low for the vasodilatory state) or sol production and metabolism. Firstly, both ALF
3  Acute Hepatic Failure 49

and sepsis often coexist and ALF represents an hypertension (ICH). The mechanisms of lung
additional stress that can lead to RAI. Secondly, injury in ALF include the directly toxic effects of
patients with ALF have low circulating cortisol acetominophen and the release of vasoactive
levels for several reasons: the effects of low ­levels mediators that affect not only the brain and circu-
of HDL cholesterol that is central to cortisol pro- lation but also the lungs causing an increased vas-
duction, increased conversion of cortisol to the cular permeability and capillary leak. This is
inactive form cortisone and the negative effect of further exacerbated by fluid accumulation within
cytokines such as tumor necrosis factor alpha the extravascular compartments as a result of large
(TNF-α) on hypothalamic function all contribute volumes of fluid administration to support a vaso-
to the low circulating TC levels [39]. plegic circulation. Additionally, there is a high
The diagnosis of adrenal insufficiency is often incidence (around 51%) of gram-negative organ-
established by performing the short synacthen isms isolated from tracheal aspirates in intubated
test; however, during critical illness and ALF this ALF patients [44], that directly impact the devel-
is fraught with problems of interpretation as high- opment of ventilator-associated pneumonia (VAP).
lighted by the CORTICUS study and subsequent The management of ICH increases the risk of pul-
investigations [38, 40]. This is mostly related to monary and extrapulmonary sepsis and ARDS as
the decrease of both albumin and cortisol-­binding adequate sedation and specific measures to avoid
globulin (CBG), that leads to an increase of free hyperthermia contribute to limited tracheobron-
cortisol (FC) levels, despite low measured total chial toilet and retention of secretions.
cortisol (TC) levels. Various alternative measures Endotracheal tubes with a large volume, low pres-
or calculations have been explored to better assess sure cuffs and a subglottic suction port may help to
FC levels. Salivary cortisol levels correlate well mitigate some of the VAP risks. Other respiratory
with FC although in ventilated patients this may complications associated with both mechanical
be difficult to obtain. Alternatively, the free corti- ventilation and critical illness have been described
sol index (see equation below) that be calculated in patients with ALF. These include pleural effu-
by measuring both CBG and TC levels correlates sions, atelectasis, and poor compliance due to
well with FC levels [41]. These alternative mea- raised intra-abdominal pressure (IAP) or reduced
sures of FC may prove to be better methods of thoracic compliance due to chest wall edema.
assessing RAI rather than relying on TC levels Conventional lung-protective ventilation
alone. However, hydrocortisone therapy is fre- employed for ARDS may impact on cerebral per-
quently initiated empirically to impact on escalat- fusion and exacerbate ICH. A balanced approach
ing vasopressor levels. is often required, though low tidal volumes
The free cortisol index: (Unbound cortisol (6–8 mL/kg) can achieve normal partial pressures
(mmol/L) = (0.0167 + 0.182 (CBG−TC)) of CO2 (pCO2) in most cases. Increased IAP and
[2] + (0.01 22 × TC) 0.5 − (0.0167 + 0.182 decreased lung compliance due to chest wall
(CBG − TC)) [42]. edema lead to increases in pleural pressure, ren-
dering the plateau pressure a poor measure of
transpulmonary pressure. Therefore, attempts to
Respiratory limit plateau pressure below 30 cm water can be
difficult to attain and indeed are often unneces-
Hepatic encephalopathy in ALF is one of the pri- sary. The combination of ARDS with severely
mary indications for intubation and ventilation in elevated intracranial pressure (ICP) with intact
order to protect the airway from aspiration. A sig- physiological autoregulation requires tight con-
nificant number of patients will also develop respi- trol of PaCO2. When all conventional measures to
ratory complications. ARDS complicates up to optimize ventilation have been exhausted, extra-
30% of paracetamol-induced ALF cases [43]. It corporeal devices can be considered but should
affects primarily those with significant vasopres- be a strategy of last resort due the significant
sor requirements and evidence of intracranial potential for bleeding complications associated
50 A. Slack et al.

with cannulae insertion and limb ischemia. Such steroids, high protein catabolism and multi-organ
devices have been used successfully in traumatic failure [49]. A (percutaneous) tracheostomy is
brain injury and ARDS [45] and have also been often necessary to facilitate weaning from the
employed on few occasions in ALF patients asso- ventilator and sedating medication. Percutaneous
ciated with ARDS, when management of ICH tracheostomy can be performed safely in patients
has remained problematic [46]. with ALF despite coagulopathy and thrombocy-
Patients with fulminant ALF should be posi- topenia [50].
tioned with the head elevated at 30° and attention
to avoiding unnecessary turning and other inter-
ventions that will exacerbate ICH. Consequently, Neurological
high positive end expiratory pressure (PEEP) is
necessary to optimize recruitment and prevent Traditionally intracranial hypertension was a
atelectasis of basal lung segments. The adverse major cause of mortality in ALF; fortunately
effect of high PEEP on ICH may be outweighed more recent reports demonstrated that with mod-
by the improvement of oxygenation and conse- ern critical care management the incidence is
quent improvement of cerebral blood flow. 20%, and mortality rates of affected patients have
Recruitment maneuvers and prone positioning decreased [3]. Hepatic encephalopathy in ALF is
are usually contraindicated due to the impact on multifactorial, however the principal mechanisms
ICH. Refractory hypoxemia may be a reason to involve an accumulation of ammonia that can
consider removing patients with ALF from the cross the blood brain barrier and results in a
transplant waiting list. However, hypoxemia build-up of glutamine in astrocytes. Glutamine,
alone appears to be a nonspecific variable in the ammonia and the systemic inflammatory process
diagnosis of ARDS. Furthermore, a low partial have direct toxic effects on astrocyte function,
pressure of oxygen (PaO2) to FiO2 ratio is com- mitochondrial activity and contribute to astrocyte
mon, often transient and not necessarily associ- swelling resulting in cerebral oedema and intra-
ated with poor outcomes [47]. Transpulmonary cranial hypertension [51].
thermodilution cardiac output monitors can cal- Standard management should include early
culate an estimated measure of lung permeability, intubation and ventilation for airway protection in
the extravascular lung water index, which can be those with >grade 2 hepatic encephalopathy and
a useful variable in guiding management [48]. standard “neuroprotective measures” including
Weaning patients from the ventilator occurs adequate sedation, nursing in the 30° head up posi-
either once the acute phase of the liver injury has tion, avoidance of hyperglycaemia, normocap-
subsided or in the post-transplant period when noea, adequate oxygen delivery and adequate
ICH has settled. An assessment of the recovery of cerebral perfusion. Renal replacement therapy
ICP auto regulatory mechanisms can be achieved should be initiated once the arterial ammonia level
by evaluating ICP responses to enforced eleva- is >150 μmol/L and should be continued with the
tions in PaCO2, mean arterial pressure and reduc- aim of keeping ammonia levels <100 μmol/L [52].
tions in sedation. The return of ICP autoregulation Maintenance of adequate serum sodium levels
permits a more sustained withdrawal of sedation (145–150 mmol/L, best achieved with an infusion
and weaning from mandatory modes of ventila- of hypertonic (30%) saline) reduces the incidence
tion. However, once sedation is decreased or of intracranial hypertension [53]. Rescue therapies
stopped neurological problems may arise such as for acute rises in ICP include bolus doses of
slow emergence from sedation or critical care Mannitol, an osmotic diuretic, and short-term
delirium. Critical illness acquired weakness is hypoventilation to reduce the PaCO2.
highly prevalent, due to the significant number of Invasive ICP monitoring has not demonstrated
risk factors for this condition encountered in any short term mortality benefit and its routine use
ALF, including sepsis, profound systemic inflam- is not recommended. Surrogates of ICP, such as
matory response syndrome (SIRS), exposure to arterial flow on transcranial dopplers and jugular
3  Acute Hepatic Failure 51

venous oxygen saturations can be considered, but  etabolic, Gastroenterology and


M
their accuracy has not been proven in ALF [54]. Nutrition
Therapeutic hypothermia (32–35 °C) has been
used in the treatment of brain injury with the Numerous metabolic abnormalities and their
intention of reducing the cerebral metabolic oxy- associated complications are encountered in ALF
gen demand, reducing cytokine activity and but only a few studies have been undertaken to
improving cerebral blood flow. Hypothermia to assess and identify best practice.
33 °C in patients following cardiac arrest is no Hypoglycemia is a frequent metabolic abnor-
longer recommended after a large randomised mality encountered in ALF due to the loss of
trial (TTM) showed no benefit over avoidance of hepatic glycogen stores, impaired gluconeogen-
hyperthermia (with a target of 36 °C) [55]. A esis and hyperinsulinemia. Hypoglycemia during
similar RCT assessing therapeutic hypothermia the initial presentation is considered a poor prog-
in AHF patients with high-grade hepatic enceph- nostic predictor and hypoglycemia along with
alopathy showed similar results. Currently it is other parameters of hepatic necrosis may help
best to avoid hyperthermia and aim for a target determine which patients require referral to spe-
temperature of 36 °C [56]. Active cooling devices cialist centers (Table 3.2). ALF is also associated
are rarely required to achieve this. with impaired peripheral uptake of glucose and

Table 3.2  Criteria for referral/discussion with specialist center [3]


Non-paracetamol overdose (ALF
classification, time from jaundice to
Paracetamol overdose (time from ingestion, days) encephalopathy)
Organ Day 2 Day 3 Day 4 Hyperacute Acute
system Subacute
Liver INR > 3.0 INR > 4.5 INR > 6 INR > 2.0 INR > 2.0 INR > 1.5
or or or or or or
PT > 50 s PT > 75 s PT > 100 s PT > 30 s PT > 30 s PT > 20 s
or
Shrinking
liver volume
Metabolic pH < 7.3 or pH < 7.3 or pH < 7.3 or Hypoglycemia Hypoglycemia Hypoglycemia
HCO3 < 18 HCO3 < 18 HCO3 < 18
or or or or or or
Lactate > 3.0 Lactate > 3.0 Lactate > 3.0 Hyperpyrexia Hyponatremia Hyponatremia
or or or or <130 μmol/L <130 μmol/L
Hypoglycemia Hypoglycemia Hypoglycemia Hyponatremia
< 130 μmol/L
Kidney Oliguria (<0.5 Oliguria Oliguria AKI Stage AKI Stage AKI Stage
mL/kg/h for (<0.5 mL/kg/h (<0.3 mL/kg/h 1–3 1–3 1–3
>12 h) for >12 h) for >24 h or
anuria for 12 h)
or or or
SCr > SCr > SCr >
200 μmol/L 200 μmol/L 300 μmol/L
Brain HE HE HE Any degree of Any degree of Any degree of
HE HE HE
Hematology Severe Severe Pancytopenia Pancytopenia Pancytopenia
thrombocytopenia thrombocytopenia
HE hepatic encephalopathy, AKI acute kidney injury, SCr serum creatinine, INR international normalised ratio, PT
prothrombin time
52 A. Slack et al.

decreased peripheral insulin sensitivity; this is vitamins and trace elements in patients with
usually restored within 2 weeks in those patients ALF is necessary, especially in patients requir-
that survive [57]. ing continuous renal replacement therapy
It is important to establish and maintain nor- (CRRT) regimen [60, 61]. Hypophosphatemia is
moglycaemia early for example with infusions frequently encountered with CRRT, especially
of 20–50% dextrose until enteral nutrition is high volume dialysis and requires prompt
commenced. Tight blood glucose control has replacement. However, hypophosphatemia may
been controversial since the landmark study by also herald liver regeneration with increased
Van Den Berghe in 2001 and subsequent studies hepatic ATP production and is considered as a
demonstrated more adverse effects and poorer good prognostic marker [62].
outcome with hyperglycemia in critically ill
patients. This was also demonstrated in patients
with neurovascular brain injury and in ALF Immunity and Bacteremia
where hyperglycemia can contribute to poor ICH
control [58]. However, meta-analyses assessing In ALF the incidence of clinical bacteremia is
tight glycemic control studies since 2001 have high (approximately 35%) [44] and there is evi-
not confirmed the mortality benefit demonstrated dence that the complex changes in the innate
in the original study population but instead an immunity are predominantly balanced toward an
increased rate of hypoglycemic episodes with anti-inflammatory environment. The deactivation
intensive insulin regimens. Ultimately, a bal- of monocytes is thought to be the leading cause
anced approach is required with the goal of of increased susceptibility to infection.
achieving blood glucose levels closer to the Approximately 30% of cases of bacteremias
lower limit of 6–8 mmol/L (108–145 mg/dL) manifest without pyrexia and elevation of white
avoiding hypoglycemia and elevated levels cell count reflecting a hypo-responsiveness to
greater than 12 mmol/L (216 mg/dL). infection that may be associated with a mortality
Early enteral nutrition within 24 h of admis- benefit over patients exhibiting classic SIRS cri-
sion aiming to achieve 25–30 kcal/kg/day is rec- teria [63].
ommended. The use of opioid-based sedation, The use of empirical broad-spectrum antibiot-
aggressive fluid regimens causing bowel wall ics, attention to appropriate nutrition, consider-
edema, raised intra-abdominal pressure and ation of gut decontamination, oral hygiene,
constipation all contribute to abnormalities of ventilator care bundles, intense daily scrutiny of
gut motility resulting in decreased absorption. If the indwelling intravenous catheters and vigilant
gut failure and poor absorption persist despite infection control measures are important in limit-
attention to constipation therapy and the use of ing the occurrence of bacteremia. Such interven-
prokinetics early intervention with total paren- tions have affected the epidemiology of
teral nutrition (TPN) may be a last resort. bacteremia in ALF with longer median times to
Previous concerns about TPN-induced liver tox- evolution of bacteremia and a shift toward greater
icity are not observed with newer hypocaloric incidence of gram-negative organisms [44].
regimens [59]. Normal protein intake of approx- Bacteremia and SIRS both appear to influence
imately 1 g/kg/day does not seem to worsen the degree of hepatic encephalopathy (HE) [64].
hyperammonemia and hepatic encephalopathy. ALF is associated with a significant incidence of
This is important, because ALF patients are fungal sepsis (approximately 32%) predomi-
often catabolic with supra-normal energy expen- nantly due to Candida species; early empirical
diture despite significant hepatocyte loss. use of antifungal therapy, preferably with an
Furthermore, there is significant protein catabo- echinocandin antifungal such as anidulafungin is
lism with muscle wasting, amino acid losses, recommended due to the relatively high rates of
and vitamin deficiency, that can affect immune fluconazole resistant Candida and a lower rate of
function. Therefore supplementation of multiple invasive aspergillosis [65, 66].
3  Acute Hepatic Failure 53

ALF generates marked changes of pharmaco- receive LT [68]. In this trial HVP was associated
kinetics and pharmacodynamics that requires with significant reduction in circulating damage
close drug monitoring. If drug level monitoring is associated molecular pattern molecules and
not possible, antibiotic dosing should aim for TNF-alpha, suggesting that HVP acts in part by
higher drug levels because of the immuneparesis controlling the innate immune response.
associated with ALF.
The innate immune system undergoes signifi-
cant changes in response to acute liver injury and Acute Kidney Injury
has a central role in the subsequent development
of the clinical manifestations of ALF. Many of The incidence of AKI in ALF is significantly
these changes closely resemble the features of higher than that of the general critically ill popu-
systemic sepsis that results in vasoplegic circula- lation ranging from 40 to 85% and approximately
tion and MOF. The complex immune responses 75% for POD (AKI defined by Kidney Disease
in ALF are closely related to some of the clinical Improving Global Outcomes (KDIGO) guide-
complications of ALF, particularly, particularly lines) [69]. Stage 3 (increase of serum creatinine
bacteremia and encephalopathy. greater than 300% from baseline) in patients with
The innate immune system is initially acti- previously normal kidney function (SCr 80–120
vated with the mobilization of immune cellular μmol/L) is associated with poor prognosis in
components, including neutrophils, monocytes, ALF and an important clinical criteria for refer-
and macrophages. These cells are involved in the ring to a specialist center and listing patients for
profound release of cytokines as part of the pro- LT (Fig. 3.4 and Tables 3.2 and 3.3).
and anti-inflammatory responses to sustained The mechanisms involved in the development
liver injury together with significant decrease of of AKI in ALF are similar to the pathophysiologi-
complement factors impairing opsonisation of cal models of hepatorenal syndrome and septic
bacteria [67]. Neutrophil function is impaired AKI. The release of vasoactive mediators, like
with reduced chemotaxis, bacteriocidal activity, nitric oxide and other free radicals, leads to a
and decreased production of superoxide and hyperdynamic circulation with circulatory
hydrogen peroxide. Both monocytes and macro- splanchnic vasoplegia “cardiovascular failure”
phages are involved in initiation, propagation, and relative hypovolemia. These vasoactive medi-
and resolution of acute liver injury. Shortly after ator-induced changes to the splanchnic circula-
acute liver injury macrophages release chemo- tion activate responses involving the ­sympathetic
kines and pro-inflammatory cytokines. This pro-­ nervous system and renin angiotensin system
inflammatory state is balanced by (RAS) causing renal arterial vasoconstriction.
anti-inflammatory responses that accompany the Intraglomerular arteriolar vasoconstriction in
recruitment of monocytes to the site of the liver addition to endothelial dysfunction, leukocyte
injury to initiate repair processes. Activated mac- activation and release of cytokines results in pro-
rophages release TNF-a, interleukin (IL)-1, IL-6, found intracellular oxidative stress and ischemic
proteolytic enzymes, reactive oxygen intermedi- acute tubular necrosis. Furthermore microcircula-
ates, and lysosomal enzymes. These elevations of tory changes and renal venous congestion can
TNF correlate with the development of sepsis impede cellular energy mechanisms independent
and increased IL-6 levels are associated with of tissue oxygen availability [70].
MOF and mortality (Fig. 3.3). High-volume Additional renal insults can be caused by
plasma exchange (HVP) was recently studied in a drugs that are either directly nephrotoxic or
randomized controlled trial. Patients with acute cause tubulointerstitial nephritis. Specific glo-
liver failure were randomized to either three merular pathologies, that result in rapidly pro-
cycles of HVP or standard medical therapy. HVP gressive glomerulonephritides, should be
increased overall transplant-free survival with the excluded for example by urine dipstick and
largest effect on survival in patients who did not microscopy for red cell casts in conjunction with
54 A. Slack et al.

LIVER Hepatic apoptosis

Hepatocellular
damage Hepatic macrophage
population expands

KNUPFFER
CELL Anti-inflamatory cytokines Pro-inflammatory cytokines
IL-10,IL-6 TNF-α, IFN-γ, MIF

Circulatory CARS SIRS


distribution and
differentiation

SEPSIS MOD
Chemokines ACTIVATED
MONOCYTE

Functional
dectivation of
SYSTEMIC
monocytes
COMPARTMENT

Monocyte maturation BONE MARROW


and expansion

Fig. 3.3  A schematic of the inflammatory responses to hepatocellular damage. Adapted from [64]

testing for a­ utoantibodies to exclude small ves- NAC does not protect tubular cells [72].
sel vasculitides and serological testing for lepto- Paracetamol is a phenacetin metabolite that has
spirosis (Weil’s disease), if the history and been implicated in proximal tubule cell apoptosis
examination suggest such diagnoses (See in AKI and chronic kidney disease (CKD).
Fig. 3.4) [71]. Consequently, cellular mechanisms and the
The precise mechanism of renal cell death in induction of apoptosis in renal tubular cells has
paracetamol nephrotoxicity remains unknown been the focus of studies of paracetamol-induced
and yet it is clear that it differs from the mecha- nephrotoxicity. It is likely that the mechanism for
nisms involved in hepatotoxicity as in rat models nephrotoxicity lies with endoplasmic reticulum
3  Acute Hepatic Failure 55

AKI in Acute Liver failure


Glomerular disease Factors associated with greater AKI
susceptibility
Rapidly progressive glomerulonephritis
(A pathological classification based on immumofluorescence patterns [SS]) Reactive increases in afferent arteriolar tone
Typr I (3%) - anti glomerular basement membrane disease ‘Vascular failure’ – acute liver failure
Good pasture’s Sepsis including rarely leptosporosis
Hepatorenal syndrome – Type 1
Type II (45%) - Immune complex mediated
Contrast
Postinfectious (staphylococci/streptococci) Structural failure to decrease afferent arteriolar resistance
Collagen-vascular disease
Lupus nephritis Age
Henoch-Schonlein purpura (immunoglobulin A and systemic vasculitis) Atheroscleosis – includes micro and macro vascular renovascular
Immunoglobulin A nephropathy (no vasculitis) disease
Mixed cryoglobulinemia Cheonic kidney disease
Primary renal disease Chronic hypertension
Membranoproliferative glomerulonephrities Malignant hypertension
Idiopathic Severe pre-eclampsia
Type III (50%) Pauci immune - Antinuclear cytoplasmic antibody mediated Nephroroxix Drugs
Wegener granulomatosis (WG) (Direct toxicity or tubulo-interstitial nephritis)
Microscopic polyangiitis (MPA)
Renal-limited necrotizing crescentic glomerulonephritis (NCGN) Paracetamol
Churg-Strauss syndrome Aminoglycosides
Contrast
Other
Penicillin
Glomeruloendotheliosis - pre-eciampsis Non-Steroidal anti-inflammatory drugs
Thrombotic microangiopathy – TTP, HUS Herbal rememdies

Fig. 3.4  Acute kidney injury (AKI) in acute liver failure

Table 3.3  Criteria for super-urgent listing for liver transplantation [3]
Organ Sera-negative hepatitis (SNH), hepatitis A, hepatitis B, or
system Paracetamol overdose an idiosyncratic drug reaction (IDR)
Liver INR > 6.5 or PT > 100 s with both AKI INR > 6.5 or PT > 100 s or pH < 7.3 with any grade of
Stage 3 and Grade 3/4 HEa HE or three of the following: (INR > 3.5 or PT > 50 s,
bilirubin > 300 μmol/L, jaundice to HE > 7 days,
unfavourable etiology SNH or IDR, age > 40)
Metabolic pH < 7.25 or lactate > 3.0 mmol/La
Kidney AKI Stage 3 (SCr > 300 μmol/L or anuria)
with both (INR > 6.5 or PT > 100 s and
Grade 3/4 HE)a
Brain Grade 3/4 HE with both (INR > 6.5 or Any grade of HE with INR > 6.5 or PT > 100 s
PT > 100 s and AKI Stage 3)a
Cardiac In the UK increased inotrope or
vasopressor requirement in the absence of
sepsis with 2 out of 3 (INR > 6.5 or
PT > 100 s, AKI Stage 3, Grade 3/4 HE)a
HE hepatic encephalopathy, AKI acute kidney injury, SCr serum creatinine, INR international normalised ratio, PT
prothrombin time
a
Assessment at > 24 h post-ingestion and should occur within a 24 h window, despite aggressive fluid resuscitation

stress and caspase-mediated apoptosis [73]. of glutathione conjugates that are considered
Other possible mechanisms include induction of nephrotoxic.
oxidative enzymes such as cytochrome P-450 AKI in patients with ALF frequently requires
mixed function oxidase isoenzymes in the proxi- the use of CRRT for renal-specific and non-­renal-­
mal tubule of the kidney [74]. Furthermore gluta- related reasons. ALF complicates the use of
thione, an important element in the detoxification CRRT specifically when anticoagulation is
of acetaminophen and its metabolites has para- required to extend filter life span. Despite the
doxically also been implicated in the formation coagulopathy and thrombocytopenia associated
56 A. Slack et al.

with ALF, CRRT circuits can clot as a result of the classic Y-shaped model of coagulation and
losses of both pro- and anticoagulation factors reflects activity of clotting factors V, VII, and
[75]. Good vascular access, use of pre-dilution X. Factor VII with the shortest half-life of
fluid replacement, high blood flows to reduce the approximately 2 h, is a good marker of synthetic
ultrafiltration fraction, prompt attention to alarms liver function and the extent of hepatic necrosis.
and the use of prostacyclin anticoagulation may Factor V is a good prognostic indicator in
extend filter life. Prostacyclin has a half-life of Hepatitis B induced ALF [81]. However as the
seconds and therefore represents a safe anticoag- measurement of individual clotting factor levels
ulant in ALF in the absence of hemorrhage. is not routinely available, PT is commonly used
Routine use of heparin for filter anticoagulation for prognostic assessment. In POD a PT greater
is not recommended and citrate anticoagulation than 36 s 36 h after ingestion predicts that 50% of
is complicated by the risk of citrate toxicity, due patients will proceed to develop ALF. A PT
to the integral role of the liver in citrate metabo- increasing on day 4 after ingestion with a peak
lism. However, the use of citrate-based intra-­ PT of greater than 180 s is predictive of a 65%
operative dialysis during a liver transplantation mortality [82]. Prolonged PT however does not
for a paracetamol-induced ALF patient and AKI predict bleeding risk in ALF and thrombin gen-
with no signs of citrate toxicity has been reported eration tests may be a better reflection of coagu-
[76]. The lack of toxicity was likely due to a low lation status [83]. ALF impairs synthesis of
dose of citrate (0.8 mmol/L; only about one-fifth pro- and anticoagulant factors and therefore ALF
of the concentration necessary to achieve antico- patients can develop hypercoagulable states as
agulation) and the predominant role of muscle well as bleeding diathesis [84]. The use of blood
metabolizing citrate. Citrate dialysate for RRT in products containing clotting factors will affect
ALF should if at all only used for short periods the utility of PT as a predictive marker. Blood
and is not a common practice. products to correct coagulopathy should only be
Indications and timing of initiation, dose, anti- used when there is active bleeding or an invasive
coagulation and continuous versus intermittent procedure such as ICP bolt insertion is to be
CRRT remain controversial. The Randomized undertaken.
Evaluation of Normal versus Augmented Level
(RENAL) study showed no mortality benefit of
high ultrafiltration doses of 35–40 mL/kg/h com- Prognosis of ALF
pared to low rates of 20–25 mL/kg/h of CRRT in
critically ill patients [77]. This has been con- Recovery in ALF is largely determined by the
firmed by the IVOIRE (hIgh VOlume in Intensive underlying pathology; therefore, establishing a
Care) study in patients with septic shock (com- diagnosis is important not only to prognosticate
paring 70 mL/kg/h with 35 mL/kg/h) and a sub- but even more importantly to aid the decision if a
sequent meta-analyses [78–80]. RRT needs to be patient should be listed for transplantation.
tailored to address the clinical fluctuations affect-
ing fluid management and the profound meta-
bolic disarray encountered in ALF. King’s College Criteria

Clinical criteria predicting prognosis in patients


Coagulation with ALF were first described at King’s College
Hospital, London. A retrospective analysis of
As synthesis of procoagulant factors is impaired patients with ALF who were medically man-
with acute hepatocyte necrosis coagulation tests aged between 1973 and 1985 was performed
may allow determination of prognosis but not with the aim of identifying significant clinical
necessarily bleeding risk in ALF. Prothrombin prognostic parameters. The King’s College cri-
time (PT) is a measure of the extrinsic pathway of teria (KCC: INR, hepatic encephalopathy,
3  Acute Hepatic Failure 57

acidosis, serum creatinine and lactate) have patients with a liver volume of <1000 mL sur-
become the most widely used criteria for assess- vived without LT [87].
ing prognosis in ALF. The KCC have a high
specificity for mortality without transplantation
but a low sensitivity and negative predictive  ontraindications to Liver
C
value (NPV). Quality of life is significantly Transplantation
affected by transplantation and should be
included as an aspect of the decision-­making Prognostic criteria are inherently biased and
process especially for those patients with POD, often perform best in the study center where they
who may also have chronic psychiatric condi- were originally validated. All currently used cri-
tions. The need to avoid unnecessary transplan- teria are associated with problems of accurate
tations and the scarcity of donor organs have selection of patients for transplantation. The
mandated an ongoing search for additional decision to proceed with transplantation greatly
parameters that can predict prognosis earlier. affects patient survival, graft use from a limited
Persistently elevated blood lactate has been donor pool and physical and psychological
closely associated with mortality and conse- ­consequences associated with long-term immu-
quently incorporated into the KCC for nosuppression. An early initial assessment of
paracetamol-related ALF [34]. The KCC have prognosis must be individualized in the context
been developed for both paracetamol- and of existing validated criteria and continuously
nonparacetamol-­related ALF to assist decisions reviewed during the hospitalization. The decision
regarding referral to specialist centers that per- to list for transplantation needs to re-assessed in
form LT and to decide whom to priority list for case of clinical deterioration that may nullify any
transplantation as outlined in Tables 3.2 and 3.3. mortality benefit from transplantation. The devel-
opment of ongoing specific organ failure, despite
maximal supportive therapies should prompt re-­
Clichy Criteria evaluation of any listing decision by the multidis-
ciplinary team.
The Clichy criteria, developed from a group of Age is an important prognostic factors and it
115 patients with ALF due to acute hepatitis has been incorporated into the non-paracetamol
includes two variables, hepatic encephalopathy classification of ALF transplantation criteria and
and factor V levels. Factor V levels less than 20% confirmed as a poor prognostic variable in a num-
for patients under 20 years and less than 30% for ber of studies. The cut-off age associated with
those older than 30 years were prognostically poor prognosis ranges from as low as 40 to as
important. The Clichy criteria had a positive pre- high as 60 years. Interestingly, while older age
dictive value (PPV) of 75% and a NPV of 58% correlates with overall poor survival, however,
compared to a PPV 80% and NPV 77% for KCC there is no statistical difference between young
in patients with ALF due to hepatitis B [85]. and older patients in spontaneous recovery and
survival (Fig. 3.5).
In our experience transplantation is unlikely to
MELD and Liver Volumes alter outcome if there is circulatory failure with
any of the following: a low cardiac index, right
With changing etiologies of ALF sub-acute ALF heart failure, or pulmonary hypertension with a
is becoming more frequent in these patients the pulmonary artery pressure > 50 mmHg associ-
model for end-stage liver disease (MELD) may ated with escalating vasopressor requirements in
perform better than the KCC [86]. Assessment association with ischemic extremities. In addi-
of liver volumes using CT is an additionally use- tion, severe lung injury requiring high PEEP
ful prognostic tool and is able to identify patients (10–15 cmH2O) and fractional inspired oxygen
with a poor prognosis; in one study only 11% of >0.8 with oxygen saturations <92% are
58 A. Slack et al.

a b
1.0 1.0

0.8 0.8
Cum survival

Cum survival
0.6 0.6

0.4 0.4
<45 Years
>45 Years 1994-99
0.2 0.2 2000-04

0.0 0.0

0 20 40 60 80 0 20 40 60 80
Days after Transplantation Days after Transplantation
c d
1.0 1.0

0.8 0.8
Cum survival

Cum survival

0.6 0.6

0.4 0.4

Low risk graft


0.2 No Vasopressors 0.2 High risk graft
Vasopressors
0.0 0.0

0 20 40 60 80 0 20 40 60 80
Days after Transplantation Days after Transplantation

Fig. 3.5  Survival of patients transplanted (a) aged >45 ing transplantation and (d) with liver grafts with a calcu-
and <45 years, (b) between 1994 and 1999 and 2000 and lated donor risk score either high or low
2004, (c) requiring vasopressor or no vasopressor follow-

associated with poor outcome in ALF and should physiological variables such as a low cardiac
possibly preclude transplantation. However toxic index and hypoxemia are associated with a very
liver syndrome as a cause of lung injury, needs to poor prognosis.
be considered as transplantation may offer a ben-
efit in this setting.
Bacteremia is also an important potential Summary
contraindication for transplantation that should
delay the listing for transplantation until expo- ALF is a multisystem disorder requiring both
sure to targeted antibiotics for a minimum of predictive and reactive management strategies to
24 h has elapsed. Both fungal sepsis and nec- support and protect organs from both the initial
rotizing pancreatitis are similarly associated and subsequent insults. Early referral to a spe-
with an extremely poor outcome in transplan- cialist liver center with the option of liver trans-
tation for ALF. Fixed dilated pupils for greater plantation and an experienced multidisciplinary
than 2 h and a prolonged cerebral perfusion team is recommended. (Table 3.1) Such teams
pressure <45 mmHg in combination with other include liver intensivists, transplant surgeons
3  Acute Hepatic Failure 59

and anesthesiologists, hepatologists, nurses and References


physiotherapists all working to ensure a high
standard of care is delivered. Furthermore, a 1. Khashab M, Tector AJ, Kwo PY. Epidemiology
good understanding of the poor prognostic vari- of acute liver failure. Curr Gastroenterol Rep.
2007;9(1):66–73.
ables is necessary to determine those most at 2. Reuben A, Tillman H, Fontana RJ, et al. Outcomes
risk of developing ALF to facilitate timely and in adults with acute liver failure between 1998 and
safe transfer. 2013: an observational cohort study. Ann Intern Med.
The initial primary goal of management is to 2016;164:724–32.
3. Bernal W, Hyyrylainen A, Gera A, et al. Lessons from
establish a diagnosis to facilitate the initiation look-back in acute liver failure? A single centre expe-
of therapies that may prevent further liver injury. rience of 3300 patients. J Hepatol. 2013;59(1):74–80.
Additionally, optimization of the circulation 4. Bacon BR, et al. Comprehensive clinical hepatology.
with both appropriate early invasive monitoring St. Louis, MO; London: Elsevier Mosby; 2006.
5. Fontana RJ, Ellerbe C, Durkalski VE, et al. Two-year
directing aggressive fluid resuscitation, vaso- outcomes in initial survivors with acute liver failure:
pressor support and assessment for high-volume results from a prospective, multicentre study. Liver
plasma exchange is the key. The early use of Int. 2015;35(2):370–80.
6. O’Grady J. Timing and benefit of liver transplantation
empirical antibiotics and antifungal agents
in acute liver failure. J Hepatol. 2014;60(3):663–70.
along with strict infection control measures are 7. Gunnell D, Murray V, Hawton K. Use of paracetamol
necessary. Due to the high frequency of sepsis (acetaminophen) for suicide and nonfatal poisoning:
without SIRS symptoms a low threshold for worldwide patterns of use and misuse. Suicide Life
Threat Behav. 2000;30(4):313–26.
obtaining cultures and broadening antibiotic
8. Hawton K, Bergen H, Simkin S, et al. Long term
coverage is required when the clinical condition effect of reduced pack sizes of paracetamol on poi-
deteriorates. A keen awareness of the potential soning deaths and liver transplant activity in England
for raised ICH, particularly in the young, neces- and Wales: interrupted time series analyses. BMJ.
2013;346:f403.
sitates appropriate monitoring and management,
9. Bateman DN. Pack size and paracetamol overdose: 16
which will be discussed in detail in a separate years later. Clin Toxicol (Phila). 2014;52(8):821–3.
chapter. In parallel with supportive measures an 10. Dargan PI, Jones AL. Acetaminophen poisoning: an
assessment of the clinical history and prognostic update for the intensivist. Crit Care. 2002;6(2):108–10.
11. Gyamlani GG, Parikh CR. Acetaminophen toxicity:
variables must be undertaken to determine,
suicidal vs. accidental. Crit Care. 2002;6(2):155–9.
which patient fulfills national transplantation 12. Litovitz TL, Klein-Schwartz W, White S, et al. 2000
criteria. The decision to list a patient for super- Annual report of the American Association of Poison
urgent liver transplantation is often difficult and Control Centers Toxic Exposure Surveillance System.
Am J Emerg Med. 2001;19(5):337–95.
can be affected by age, co-morbidities, the
13. Lee WM. Acetaminophen-related acute liver failure
dynamics of the clinical condition and psycho- in the United States. Hepatol Res. 2008;38(Suppl
social factors. [6] The clinical course for those 1):S3–8.
not transplanted is often precarious and associ- 14. Taylor RM, Davern T, Munoz S, et al. Fulminant
hepatitis A virus infection in the United States: inci-
ated with a high mortality [5]. Outcome is
dence, prognosis, and outcomes. Hepatology. 2006;
affected by the speed and degree of hepatic 44(6):1589–97.
regeneration and the impact of the cumulative 15. Miyake Y, Iwasaki Y, Takaki A, et al. Lamivudine
insults such as sepsis, AKI requiring prolonged treatment improves the prognosis of fulminant hepati-
tis B. Intern Med. 2008;47(14):1293–9.
RRT, and critical illness associated weakness
16. Dao DY, Seremba E, Ajmera V, et al. Use of nucleoside
that may result in extended periods of rehabili- (tide) analogues in patients with hepatitis B-related
tation in those that survive. Patients who pro- acute liver failure. Dig Dis Sci. 2012;57(5):1349–57.
ceed to transplantation and receive a good 17. Mahtab MA, Rahman S, Khan M, Mamun AA, Afroz
S. Etiology of fulminant hepatic failure: experience
functioning graft often experience swift resolu-
from a tertiary hospital in Bangladesh. Hepatobiliary
tion of the circulatory and neurovascular disar- Pancreat Dis Int. 2008;7(2):161–4.
ray and have significantly improved outcomes 18. Gerolami R, Borentain P, Raissouni F, Motte A, Solas
albeit offset by the long-term impact of lifelong C, Colson P. Treatment of severe acute hepatitis E by
ribavirin. J Clin Virol. 2011;52(1):60–2.
immunosuppression.
60 A. Slack et al.

19. Russmann S, Kullak-Ublick GA, Grattagliano I. 


paracetamol-induced acute liver failure: a cohort
Current concepts of mechanisms in drug-­induced study. Lancet. 2002;359(9306):558–63.
hepatotoxicity. Curr Med Chem. 2009;16(23): 35. O’Grady JG, Alexander GJ, Hayllar KM, Williams
3041–53. R. Early indicators of prognosis in fulminant hepatic
20. Chalasani NP, Hayashi PH, Bonkovsky HL, et al. failure. Gastroenterology. 1989;97(2):439–45.
ACG Clinical Guideline: the diagnosis and manage- 36. Harry R, Auzinger G, Wendon J. The clinical impor-
ment of idiosyncratic drug-induced liver injury. Am J tance of adrenal insufficiency in acute hepatic dys-
Gastroenterol. 2014;109(7):950–66. quiz 967 function. Hepatology. 2002;36(2):395–402.
21. Harrison HB, Middleton HM 3rd, Crosby JH, Dasher 37. Harry R, Auzinger G, Wendon J. The effects of supra-
MN Jr. Fulminant hepatic failure: an unusual presen- physiological doses of corticosteroids in hypotensive
tation of metastatic liver disease. Gastroenterology. liver failure. Liver Int. 2003;23(2):71–7.
1981;80(4):820–5. 38. Sprung CL, Annane D, Keh D, et al. Hydrocortisone
22. Rajvanshi P, Kowdley KV, Hirota WK, Meyers JB, therapy for patients with septic shock. N Engl J Med.
Keeffe EB. Fulminant hepatic failure secondary to 2008;358(2):111–24.
neoplastic infiltration of the liver. J Clin Gastroenterol. 39. Mookerjee RP, Sen S, Davies NA, Hodges SJ,

2005;39(4):339–43. Williams R, Jalan R. Tumour necrosis factor alpha
23.
Nakagawa K, Miller FN, Sims DE, Lentsch is an important mediator of portal and systemic hae-
AB, Miyazaki M, Edwards MJ. Mechanisms of modynamic derangements in alcoholic hepatitis. Gut.
interleukin-­2-induced hepatic toxicity. Cancer Res. 2003;52(8):1182–7.
1996;56(3):507–10. 40. Boonen E, Vervenne H, Meersseman P, et al. Reduced
24. Darwish Murad S, Plessier A, Hernandez-Guerra
cortisol metabolism during critical illness. N Engl J
M, et al. Etiology, management, and outcome Med. 2013;368(16):1477–88.
of the Budd-Chiari syndrome. Ann Intern Med. 41. Moisey R, Wright D, Aye M, Murphy E, Peacey

2009;151(3):167–75. SR. Interpretation of the short Synacthen test in the
25. Kralovics R, Passamonti F, Buser AS, et al. A gain-­ presence of low cortisol-binding globulin: two case
of-­function mutation of JAK2 in myeloproliferative reports. Ann Clin Biochem. 2006;43(Pt 5):416–9.
disorders. N Engl J Med. 2005;352(17):1779–90. 42. Coolens JL, Van Baelen H, Heyns W. Clinical use
26. Eisenbach C, Sieg O, Stremmel W, Encke J,
of unbound plasma cortisol as calculated from total
Merle U. Diagnostic criteria for acute liver fail- cortisol and corticosteroid-binding globulin. J Steroid
ure due to Wilson disease. World J Gastroenterol. Biochem. 1987;26(2):197–202.
2007;13(11):1711–4. 43. Baudouin SV, Howdle P, O’Grady JG, Webster

27. Sallie R, Katsiyiannakis L, Baldwin D, et al. Failure NR. Acute lung injury in fulminant hepatic fail-
of simple biochemical indexes to reliably differentiate ure following paracetamol poisoning. Thorax.
fulminant Wilson’s disease from other causes of fulmi- 1995;50(4):399–402.
nant liver failure. Hepatology. 1992;16(5):1206–11. 44. Karvellas CJ, Pink F, McPhail M, et al. Predictors of
28. Yeoman AD, Westbrook RH, Zen Y, et al. Prognosis bacteraemia and mortality in patients with acute liver
of acute severe autoimmune hepatitis (AS-AIH): failure. Intensive Care Med. 2009;35(8):1390–6.
the role of corticosteroids in modifying outcome. J 45. McKinlay J, Chapman G, Elliot S, Mallick A. Pre-­
Hepatol. 2014;61(4):876–82. emptive Novalung-assisted carbon dioxide removal
29. Bernal W, Cross TJ, Auzinger G, et al. Outcome
in a patient with chest, head and abdominal injury.
after wait-listing for emergency liver transplantation Anaesthesia. 2008;63(7):767–70.
in acute liver failure: a single centre experience. J 46. Auzinger G, Willars C, Loveridge R, Vercueil A, Best
Hepatol. 2009;50(2):306–13. T, Wendon J. Extracorporeal membrane oxygenation
30. Germani G, Theocharidou E, Adam R, et al. Liver before and after adult liver transplantation: worth the
transplantation for acute liver failure in Europe: effort? Crit Care. 2014;18:P203.
outcomes over 20 years from the ELTR database. J 47. Auzinger G, Sizer E, Bernal W, Wendon J. Incidence
Hepatol. 2012;57(2):288–96. of lung injury in acute liver failure: diagnostic
31. Dellinger RP, Levy MM, Rhodes A, et al. Surviving role of extravascular lung water index. Crit Care.
sepsis campaign: international guidelines for manage- 2004;8(Suppl 1):40.
ment of severe sepsis and septic shock: 2012. Crit 48. Costa EL, Vidal Melo MF. Lung water: what you see
Care Med. 2013;41(2):580–637. (with computed tomography) and what you get (with a
32. Chiolero R, Tappy L, Gillet M, et al. Effect of major bedside device). Anesthesiology. 2009;111(5):933–5.
hepatectomy on glucose and lactate metabolism. Ann 49. Hermans G, De Jonghe B, Bruyninckx F, Van den
Surg. 1999;229(4):505–13. Berghe G. Clinical review: critical illness polyneu-
33. Khosravani H, Shahpori R, Stelfox HT, Kirkpatrick ropathy and myopathy. Crit Care. 2008;12(6):238.
AW, Laupland KB. Occurrence and adverse effect on 50. Auzinger G, O’Callaghan GP, Bernal W, Sizer E,

outcome of hyperlactatemia in the critically ill. Crit Wendon JA. Percutaneous tracheostomy in patients
Care. 2009;13(3):R90. with severe liver disease and a high incidence of
34. Bernal W, Donaldson N, Wyncoll D, Wendon
refractory coagulopathy: a prospective trial. Crit Care.
J. Blood lactate as an early predictor of outcome in 2007;11(5):R110.
3  Acute Hepatic Failure 61

51. Ott P, Vilstrup H. Cerebral effects of ammonia in tation recipients: a propensity score analysis. Liver
liver disease: current hypotheses. Metab Brain Dis. Transpl. 2016;22(4):427–35.
2014;29(4):901–11. 67. Wyke RJ, Rajkovic IA, Eddleston AL, Williams

52. Slack AJ, Auzinger G, Willars C, et al. Ammonia R. Defective opsonisation and complement deficiency
clearance with haemofiltration in adults with liver dis- in serum from patients with fulminant hepatic failure.
ease. Liver Int. 2014;34(1):42–8. Gut. 1980;21(8):643–9.
53. Murphy N, Auzinger G, Bernel W, Wendon J. The 68. Larsen FS, Schmidt LE, Bernsmeier C, et al. High-­
effect of hypertonic sodium chloride on intracra- volume plasma exchange in patients with acute
nial pressure in patients with acute liver failure. liver failure: an open randomised controlled trial. J
Hepatology. 2004;39(2):464–70. Hepatol. 2016;64(1):69–78.
54. Karvellas CJ, Fix OK, Battenhouse H, et al. Outcomes 69. Betrosian AP, Agarwal B, Douzinas EE. Acute renal
and complications of intracranial pressure monitoring dysfunction in liver diseases. World J Gastroenterol.
in acute liver failure: a retrospective cohort study. Crit 2007;13(42):5552–9.
Care Med. 2014;42(5):1157–67. 70. Langenberg C, Wan L, Egi M, May CN, Bellomo
55. Nielsen N, Wetterslev J, Cronberg T, et al. Targeted R. Renal blood flow in experimental septic acute renal
temperature management at 33 degrees C versus failure. Kidney Int. 2006;69(11):1996–2002.
36 degrees C after cardiac arrest. N Engl J Med. 71. Ghosh AK. Mayo Clinic internal medicine review. 8th
2013;369(23):2197–206. ed. New York: Informa Healthcare; 2008.
56. Bernal W, Murphy N, Brown S, et al. A multicentre 72.
Eguia L, Materson BJ. Acetaminophen-related
randomized controlled trial of moderate hypothermia acute renal failure without fulminant liver failure.
to prevent intracranial hypertension in acute liver fail- Pharmacotherapy. 1997;17(2):363–70.
ure. J Hepatol. 2016;65:273–9. 73. Lorz C, Justo P, Sanz A, Subira D, Egido J, Ortiz
57. Clark SJ, Shojaee-Moradie F, Croos P, et al. Temporal A. Paracetamol-induced renal tubular injury: a role
changes in insulin sensitivity following the develop- for ER stress. J Am Soc Nephrol. 2004;15(2):380–9.
ment of acute liver failure secondary to acetamino- 74. Mazer M, Perrone J. Acetaminophen-induced neph-
phen. Hepatology. 2001;34(1):109–15. rotoxicity: pathophysiology, clinical manifestations,
58. Kodakat S, Gopal P, Wendon J. Hyperglycaemia
and management. J Med Toxicol. 2008;4(1):2–6.
is associated with intracranial hypertension in 75. Agarwal B, Shaw S, Shankar Hari M, Burroughs AK,
patients with acute liver failure. Liver Transpl. Davenport A. Continuous renal replacement therapy
2001;7(Suppl):C21. (CRRT) in patients with liver disease: is circuit life
59. Salvino R, Ghanta R, Seidner DL, Mascha E, Xu Y, different? J Hepatol. 2009;51(3):504–9.
Steiger E. Liver failure is uncommon in adults receiv- 76. Winrow RM, Davis C, Halldorson JB, Ahmad

ing long-term parenteral nutrition. JPEN J Parenter S. Intraoperative dialysis during liver trans-
Enteral Nutr. 2006;30(3):202–8. plantation with citrate dialysate. Hemodial Int.
60. Fortin MC, Amyot SL, Geadah D, Leblanc M. Serum 2009;13(3):257–60.
concentrations and clearances of folic acid and 77. Bellomo R, Harris C, Kang Y, Daniel E, Fung JJ,
pyridoxal-5-phosphate during venovenous continu- Bronsther O. Combined veno-venous bypass and high
ous renal replacement therapy. Intensive Care Med. volume hemofiltration during orthotopic liver trans-
1999;25(6):594–8. plantation. ASAIO J. 1993;39(4):954–6.
61. Berger MM, Shenkin A, Revelly JP, et al. Copper, 78. Joannes-Boyau O, Honore PM, Perez P, et al. High-­
selenium, zinc, and thiamine balances during con- volume versus standard-volume haemofiltration
tinuous venovenous hemodiafiltration in critically ill for septic shock patients with acute kidney injury
patients. Am J Clin Nutr. 2004;80(2):410–6. (IVOIRE study): a multicentre randomized controlled
62. Schmidt LE, Dalhoff K. Serum phosphate is an early trial. Intensive Care Med. 2013;39(9):1535–46.
predictor of outcome in severe acetaminophen-induced 79. Jun M, Heerspink HJ, Ninomiya T, et al. Intensities
hepatotoxicity. Hepatology. 2002;36(3):659–65. of renal replacement therapy in acute kidney injury: a
63. Rolando N, Wade J, Davalos M, Wendon J, Philpott-­ systematic review and meta-analysis. Clin J Am Soc
Howard J, Williams R. The systemic inflammatory Nephrol. 2010;5(6):956–63.
response syndrome in acute liver failure. Hepatology. 80. Van Wert R, Friedrich JO, Scales DC, Wald R,

2000;32(4 Pt 1):734–9. Adhikari NK. University of Toronto Acute Kidney
64. Possamai LA, Antoniades CG, Anstee QM, et al.
Injury Research G. High-dose renal replacement ther-
Role of monocytes and macrophages in experimental apy for acute kidney injury: systematic review and
and human acute liver failure. World J Gastroenterol. meta-analysis. Crit Care Med. 2010;38(5):1360–9.
2010;16(15):1811–9. 81. Bernuau J, Goudeau A, Poynard T, et al. Multivariate
65. Rolando N, Philpott-Howard J, Williams R. Bacterial analysis of prognostic factors in fulminant hepatitis
and fungal infection in acute liver failure. Semin Liver B. Hepatology. 1986;6(4):648–51.
Dis. 1996;16(4):389–402. 82. Harrison PM, O’Grady JG, Keays RT, Alexander GJ,
66.
Fortun J, Muriel A, Martin-Davila P, et al. Williams R. Serial prothrombin time as prognostic
Caspofungin versus fluconazole as prophylaxis of indicator in paracetamol induced fulminant hepatic
invasive fungal infection in high-risk liver transplan- failure. BMJ. 1990;301(6758):964–6.
62 A. Slack et al.

83. Habib M, Roberts LN, Patel RK, Wendon J, Bernal 86. McPhail MJ, Farne H, Senvar N, Wendon JA, Bernal
W, Arya R. Evidence of rebalanced coagulation in W. Ability of King’s College Criteria and Model
acute liver injury and acute liver failure as measured for End-Stage Liver Disease Scores to Predict
by thrombin generation. Liver Int. 2014;34(5):672–8. Mortality of Patients With Acute Liver Failure:
84. Agarwal B, Wright G, Gatt A, et al. Evaluation of A Meta-analysis. Clin Gastroenterol Hepatol.
coagulation abnormalities in acute liver failure. J 2016;14(4):516–25.e5
Hepatol. 2012;57(4):780–6. 87. Zabron, A. Quaglia, A. Fatourou, E. Peddu, P. et
85. Yantorno SE, Kremers WK, Ruf AE, Trentadue
al. Clinical and Prognostic Associations of Liver
JJ, Podesta LG, Villamil FG. MELD is superior to Volume determined by Computed Tomography in
King’s college and Clichy’s criteria to assess prog- Acute Liver Failure. Liver Int. 2018 Feb 20. doi:
nosis in fulminant hepatic failure. Liver Transpl. 10.1111/liv.13725.
2007;13(6):822–8.
The Splanchnic and Systemic
Circulation in Liver Disease 4
Nina T. Yoh and Gebhard Wagener

Keywords Physiology
Liver blood flow · Portal hypertension · Portal
flow · Hepatic artery buffer response · Splanchnic The splanchnic vascular bed consists of the celiac
steal · Vasoplegia artery and superior and inferior mesenteric arter-
ies supplying oxygenated blood to abdominal
organs. The liver obtains oxygenated blood
Introduction directly from the hepatic artery that derives from
the celiac artery. The other branches of the celiac
Alterations of the splanchnic circulation are a artery and of the superior and inferior mesenteric
central component of the pathophysiology of artery supply the intestines and the spleen with
liver disease and result in profound changes of oxygenated blood (Fig. 4.1). Venous drainage of
the systemic circulation and ensuing extrahepatic the intestines and the spleen will flow through the
organ damage. An understanding of the changes portal vein to the liver. The liver is therefore the
of the splanchnic and systemic circulation in liver only organ with a dual blood supply through the
disease is important for the anesthesiologist and hepatic artery and the portal vein. In normal
critical care physician as anesthetic agents and physiologic states this blood supply is tightly
surgery or other insults such as infections can regulated through the hepatic artery buffer
further exacerbate these changes and cause dete- response. The aim of the hepatic artery buffer
rioration of the clinical status of patients. response is to maintain steady total hepatic blood
flow. A reduction of portal blood flow will lead to
an increase of hepatic artery flow and vice versa.
This physiologic adaption is not mediated by sys-
temic vasoactive substances or innervation but is
thought to depend on regional release of adenos-
ine; increased portal flow washes vasodilatory
N. T. Yoh adenosin away that is released in the space
Columbia University Medical Center, between artery and portal vein. As a conse-
New York, NY, USA
quence, hepatic artery blood flow decreases.
G. Wagener, MD (*) Total hepatic blood flow is approximately 800–
Department of Anesthesiology, Columbia University
Medical Center, New York, NY, USA 1200 mL/min [1] and the ratio of portal vein to
e-mail: gw72@cumc.columbia.edu hepatic artery blood flow is about 2.5:1.

© Springer International Publishing AG, part of Springer Nature 2018 63


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_4
64 N. T. Yoh and G. Wagener

LIVER

Hepatic
veins oids
us

Sin
Stomach
tery
Ar
c
lia

l
Po

Ce
rta
l
Inferior Caval Vein

vei
Spleen
n

Aorta
Pancreas
r
rio
u pe teric
S en
s
Me
y
ter
Small Ar
intestines

ic

r
te
en
es
M y
ri o r ter
Colon Infe Ar

Fig. 4.1  Anatomy of the splanchnic, portal and hepatic venous circulation. (With permission: Gelman S, Mushlin
PS. Catecholamine-induced changes in the splanchnic circulation affecting systemic hemodynamics. Anesthesiology
2004, 100: 434–439)

Portal Pressure and Hypertension tion is most commonly observed with cirrhosis


and is due in part to activation of hepatic stel-
It is technically difficult to directly measure por- late (Ito) cells in the space of Disse that sur-
tal pressure. Instead, portal venous pressure can rounds the sinusoids. Activated hepatic stellate
be estimated by measuring hepatic vein wedge cells convert into a myofibroblast phenotype
pressure (HVWP) and calculating hepatic thereby becoming contractile and increasing
venous pressure gradient (HVPG = HVWP− resistance to blood flow within the sinusoidal
IVC pressure). To measure HVWP a catheter veins. Furthermore resistance to portal flow
is inserted into the hepatic vein and a balloon is increased when fenestrated hepatic endo-
is inflated. HVPG is slightly lower than portal thelial cells that regulate resistance within the
pressure in normal subjects since some of the sinusoids release less nitric oxide and thereby
portal pressure is decreased due to flow into increase portal pressure [3].
sinusoidal veins. In sinusoidal portal hyperten- Normal HVPG is under 5 mmHg; HVPG
sion HVPG correlates well with portal venous >10 mmHg is usually associated with formation
pressure [2] as the resistance of sinusoidal of varices and HVPG >16 mmHg is a sign of
veins increases (Fig. 4.2). Sinusoidal constric- decompensated cirrhosis.
4  The Splanchnic and Systemic Circulation in Liver Disease 65

a Inferior Sinusoidal b Inferior Sinusoidal


Vena Cava veins Vena Cava veins

Hepatic Hepatic
vein vein
Free Free
IVC IVC
Pressure Portal vein Pressure Portal vein

FHVP Hepatic PVP FHVP Hepatic PVP


WHVP Sinusoids WHVP Sinusoids

Fig. 4.2  A depiction of the measurement of HVPG. (a) In hepatic venous pressure equates to the portal pressure.
healthy individuals the wedged hepatic venous pressure is IVC inferior vena cava, FHVP free hepatic venous pres-
slightly lower than the portal venous pressure to allow for sure, WHVP wedged hepatic venous pressure, PVP portal
blood flow through the sinusoidal veins. (b) In patients venous pressure. Hepatic venous pressure
with portal hypertension due to sinusoidal causes, the gradient = WHVP−FHVP
sinusoidal veins are damaged, and, hence, the wedged

HVPG is a good predictor of survival for bed increases by almost 20% in patients with cir-
example in alcoholic hepatitis [4], cirrhosis rhosis (Fig. 4.3) [9]. Consequently pooling of
[5] and after liver transplantation Measurement blood into the splanchnic area results in volume
of HVPG is further useful to assess response depletion and hypoperfusion in other vascular
to interventions to reduce portal venous pres- beds. Cirrhosis is therefore not associated with
sure [6]. generalized systemic vasodilation but with
Portal hypertension in cirrhosis however is not splanchnic vasodilation that results in hypoperfu-
only due to increased intrahepatic resistance to sion and vasoconstriction in other vascular beds
portal blood secondary of constriction of sinusoi- (“splanchnic steal”). Hypoperfusion of the kid-
dal veins. Systemic circulatory changes in cirrho- neys can be particularly detrimental; renal arte-
sis have been described already more than rial constriction will lead to a decrease of
60 years ago. In 1953 Kowalski and Abelmann glomerular blood flow by activation of the mac-
showed that patients with Laennec’s cirrhosis are ula densa and a reduction of glomerular filtration
in a vasodilatory and hyperdynamic state by rate via the renin-angiotenisn system. Renal arte-
observing a decreased transit time when injecting rial constriction is further aggravated by systemic
Evans blue dye into a vein and sampling blood activation of the sympathetic nervous system to
samples from the brachial artery [7]. Almost maintain systemic vascular tone.
20 years later Kotelanski et al. showed that mes-
enteric blood flow is increased in patients with
hepatic cirrhosis by injecting radioactive iodine-  he Role of Vasopressin in Liver
T
albumine into the superior mesenteric artery and Disease
sampling of blood in the hepatic vein [8]. This
led to the development of the “forward flow” One of the main reasons of this splanchnic vaso-
theory: splanchnic vasodilation results in an dilation is thought to be a low-grade septoid state.
increased splanchnic and therefore portal blood Because of translocation of bacteria from the
flow that increases portal venous pressure. intestines to the portal circulation and impair-
Whole-body scintigraphy using 99mTc-labelled ment of hepatic Kupfer cells to remove bacteriae,
human albumin can measure regional blood vol- the systemic circulation is constantly exposed to
ume distribution. In cirrhotic patients blood vol- bacterial fragments in patients with cirrhosis
ume is shifted from other vascular beds to the [10]. Additionally enterobacteriaceae and other
splanchnic area. Blood volume in the splanchnic enteric microorganisms are found frequently in
66 N. T. Yoh and G. Wagener

similar vasopressin deficiency has been observed


in patients with cirrhosis [13]. Patients undergo-
ing liver transplantation had significantly lower
baseline endogenous vasopressin levels and if
vasodilated, responded to exogenous with an in
increase of blood pressure through splanchnic
vasoconstriction. This effect of vasopressin in
cirrhosis is favorable as it decreases portal vein
flow and pressure and therefore alleviates portal
hypertension [14]. It also explains why vasopres-
sin analogues such as terlipressin and ornipressin
(in addition to fluid administration) are effective
treatments for hepatorenal syndrome [15, 16].
Reducing portal flow is not only important in
patients with cirrhosis but also after partial liver
transplantation or liver resection when the remain-
ing liver may be small relative to portal flow.
Excessive portal flow and pressure not only leads
to congestion of a small liver remnant. It also
increases metabolic work load of the liver while
limiting oxygen delivery by reducing hepatic artery
flow via the hepatic artery buffer response. The
consequence of this disproportional high portal
flow in a small liver remnant may be a deterioration
of liver function called “small for size syndrome”.
Usually liver resection of up to 70–75% of the liver
is considered safe if there is no ischemic injury and
liver and portal flow are normal [17]. The presence
Fig. 4.3  Blood volume distribution in a healthy subject of portal hypertension substantially limits the abil-
(left) and in a patient with cirrhosis (right), as illustrated ity to perform a liver resection and is usually con-
by whole-body scintigraphy with 99mTc-labelled human sidered a relative contraindication for any type of
albumin/red blood cells. Absolute regional blood volumes resection as it substantially increases the risk for
can be determined from regional radioactivity relative to
total radioactivity and total blood volume as determined mortality [18]. In selected cases resections can be
by an indicator dilution technique. With permssion: performed even in the presence of mild portal
Kiszka-Kanowitz M, Henriksen JH, Møller S, Bendtsen hypertension in experienced centers. Resection of
F. Blood volume distribution in patients with cirrhosis: hepatocellular carcinoma (HCC) is possible cura-
aspects of the dual-head gamma-camera technique. J
Hepatol 2001; 35: 605–12 tive and prevents the need for a transplantation [19,
20]. The decision to proceed with resection or
potentially with transplant in patients with resect-
mesenteric lymph nodes. As a consequence able HCC and portal hypertension should be made
­endotoxemia ensues and its severity correlates on an individual basis.
with the severity of cirrhosis [11]. Patients with Partial or living donor liver transplantation is
cirrhosis exhibit higher levels of serum cytokines considered safe if the transplanted graft is larger
such as interleukin 6 and 10 [12]. These systemic than 0.8% of (ideal) body weight [21–23].
responses (even if less severe) are similar to what However even if the graft is larger than this post
has been described for sepsis and septic shock. transplant graft failure may occur with pre-­
Relative vasopressin deficiency is considered existing portal hypertension; decreasing portal
one cause of the vasodilatory state in sepsis. A flow in this situation may then improve graft
4  The Splanchnic and Systemic Circulation in Liver Disease 67

function and avoid graft failure. This can be resistance in the hepatic sinusoids. J Clin Invest.
1997;100:2923–30.
achieved either pharmacologically using vaso-
4. Rincon D, et al. Prognostic value of hepatic venous
pressin or it analogues or mechanically for exam- pressure gradient for in-hospital mortality of patients
ple by ligating the splenic artery. It is now with severe acute alcoholic hepatitis. Aliment
possible to use left lobe liver grafts for adult-to Pharmacol Ther. 2007;25:841–8.
5. Wang YW, et al. Correlation and comparison of the
adult living donor liver transplantation if a suit-
model for end-stage liver disease, portal pressure, and
able donor is available. The use of left lobe grafts serum sodium for outcome prediction in patients with
substantially reduces the risk for morbidity and liver cirrhosis. J Clin Gastroenterol. 2007;41:706–12.
mortality in the donor but will frequently require 6. Garcia-Tsao G, et al. Short-term effects of propranolol
on portal venous pressure. Hepatology. 1986;6:101–6.
portal flow modification [24, 25]. Vasopressin is
7. Kowalski HJ, Abelmann WH. The cardiac out-
routinely used in liver transplant recipients along put at rest in Laennec’s cirrhosis. J Clin Invest.
with measurement of portal flow after reperfu- 1953;32:1025–33.
sion of the graft. If portal flow remains high 8. Lebrec D, Kotelanski B, Cohn JN. Splanchnic hemo-
dynamics in cirrhotic patients with esophageal vari-
despite vasopressin, surgical reduction of portal
ces and gastrointestinal bleeding. Gastroenterology.
flow should be considered. 1976;70:1108–11.
9. Henriksen JH, Kiszka-Kanowitz M, Bendtsen
F. Review article: volume expansion in patients with
cirrhosis. Aliment Pharmacol Ther. 2002;16(Suppl
Summary 5):12–23.
10. Frances R, et al. A sequential study of serum bacterial
Portal hypertension is one of the key symptoms DNA in patients with advanced cirrhosis and ascites.
of hepatic cirrhosis and chronic liver failure. It is Hepatology. 2004;39:484–91.
11. Lin CY, et al. Endotoxemia contributes to the immune
due to increased resistance to flow in the sinusoi-
paralysis in patients with cirrhosis. J Hepatol.
dal spaces and because of splanchnic vasodila- 2007;46:816–26.
tion and hyperemia. Splanchnic vasodilation is 12. Wasmuth HE, et al. Patients with acute on chronic
likely caused by a low-grade septoid state and liver failure display “sepsis-like” immune paralysis. J
Hepatol. 2005;42:195–201.
results in hypoperfusion of other vascular beds
13. Wagener G, et al. Vasopressin deficiency and vasodi-
such as the kidneys. Vasopressin deficiency con- latory state in end-stage liver disease. J Cardiothorac
tributes to splanchnic vasodilation and may be Vasc Anesth. 2011;25:665–70.
ameliorated by administration of exogenous 14. Wagener G, et al. Vasopressin decreases portal vein
pressure and flow in the native liver during liver trans-
vasopressin. Increased portal flow can cause liver
plantation. Liver Transpl. 2008;14:1664–70.
failure after partial or living donor liver trans- 15. Dobre M, Demirjian S, Sehgal AR, Navaneethan

plantation and extensive resections (small for SD. Terlipressin in hepatorenal syndrome: a sys-
size syndrome). Reducing portal flow with vaso- tematic review and meta-analysis. Int Urol Nephrol.
2011;43:175–84.
pressin or splenic artery ligation may allow the
16. Guevara M, et al. Reversibility of hepatorenal syn-
use of smaller, left lobe graft even with pre-exist- drome by prolonged administration of ornipres-
ing portal hypertension. sin and plasma volume expansion. Hepatology.
1998;27:35–41.
17. Guglielmi A, Ruzzenente A, Conci S, Valdegamberi
A, Iacono C. How much remnant is enough in liver
References resection? Dig Surg. 2012;29:6–17.
18. Berzigotti A, Reig M, Abraldes JG, Bosch J, Bruix
J. Portal hypertension and the outcome of surgery for
1. Brown HS, et al. Measurement of normal portal
hepatocellular carcinoma in compensated cirrhosis:
venous blood flow by Doppler ultrasound. Gut.
a systematic review and meta-analysis. Hepatology.
1989;30:503–9.
2015;61:526–36.
2. Zardi EM, Di Matteo FM, Pacella CM, Sanyal
19. Zhong JH, Li LQ. Portal hypertension should not be
AJ. Invasive and non-invasive techniques for detect-
a contraindication of hepatic resection to treat hepa-
ing portal hypertension and predicting variceal bleed-
tocellular carcinoma with compensated cirrhosis.
ing in cirrhosis: a review. Ann Med. 2014;46:8–17.
Hepatology. 2015;62:977–8.
3. Shah V, et al. Liver sinusoidal endothelial cells
20. Capussotti L, Ferrero A, Vigano L, Polastri R, Tabone
are responsible for nitric oxide modulation of
M. Liver resection for HCC with cirrhosis: surgical
68 N. T. Yoh and G. Wagener

perspectives out of EASL/AASLD guidelines. Eur J 23. Ben-Haim M, et al. Critical graft size in adult-to-adult
Surg Oncol. 2009;35:11–5. living donor liver transplantation: impact of the recip-
21. Olthoff KM, et al. Liver regeneration after living
ient’s disease. Liver Transpl. 2001;7:948–53.
donor transplantation: adult-to-adult living donor 24. Soejima Y, et al. Left lobe living donor liver transplan-
liver transplantation cohort study. Liver Transpl. tation in adults. Am J Transplant. 2012;12:1877–85.
2015;21:79–88. 25. Soejima Y, et al. Feasibility of left lobe living donor
22. Moon JI, et al. Safety of small-for-size grafts in adult-­ liver transplantation between adults: an 8-year, single-­
to-­adult living donor liver transplantation using the center experience of 107 cases. Am J Transplant.
right lobe. Liver Transpl. 2010;16:864–9. 2006;6:1004–11.
Drug Metabolism in Liver Failure
5
Simon W. Lam

Keywords Introduction
Cytochrome P450 · Glucuronidation ·
Oxidation · Half-life · Metabolism · The liver is involved in the metabolism and elimi-
Pharmacokinetics · Pharmacodynamics · Liver nation of many medications entering the body and
failure · Sedatives · Opioids · Neuromuscular liver disease leads to widespread alterations in
blockade drug pharmacokinetics and pharmacodynamics.
An understanding of physiologic changes during
liver disease and their corresponding effects on
Abbreviations drug disposition will be useful for clinicians to
optimize therapy and avoid adverse reactions. This
PK Pharmacokinetics chapter describes the complex relationship
PD Pharmacodynamics between drug properties and drug metabolism in
Clh Hepatic clearance liver failure. In addition, it will illustrate the effects
Eh Hepatic extraction of liver failure on the metabolism of specific
Clint Intrinsic clearance classes of medications that are frequently used in
Fu Fraction unbound anesthesiology and critical care such as sedatives,
Q Hepatic blood flow opioids, and neuromuscular blocking agents.
Vd Volume of distribution
CYP Cytochrome
GABA Gamma-aminobutyric acid Pharmacokinetics (PK)
t1/2 Half-life and Pharmacodynamics (PD)
AUC Area-under-the-curve
Cmax Maximum concentration Any medication entering the body follows a
unique process: absorption, distribution, metabo-
lism, and elimination. This process ultimately
determines how much drug is available at the tar-
geted site of action. Pharmacokinetics (PK) refers
to the summation of the processes of what the
body is doing to the drug. In contrast, pharmaco-
dynamics (PD) refers to the physiologic and bio-
S. W. Lam, PharmD, FCCM chemical effects of the drug on the body. The
Cleveland Clinic, Cleveland, OH, USA intended effects of the drug, at a concentration that
e-mail: lams@ccf.org

© Springer International Publishing AG, part of Springer Nature 2018 69


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_5
70 S. W. Lam

minimizes the potential adverse effects, are deter- Both drug and physiologic properties deter-
mined by an intricate balance between PK and PD. mine the extraction efficiency by the liver.
The liver is an organ positioned between the Specifically, the extraction efficiency of a partic-
upper gastrointestinal tract and the general circu- ular drug is dependent on liver blood flow, intrin-
lation and has complex metabolic and synthetic sic clearance of unbound drug (Clint), and the
functions. It participates in drug elimination via fraction of unbound (fu) drug in the blood [2] and
hepatocellular uptake, metabolism, and biliary can be represented by the formula:
excretion. As blood travels through the liver,
low-­molecular weight substances can enter the Eh = ( f u ×Clint ) / (Q + f u ×Clint )

hepatocytes by passive diffusion or active trans-
port. Clearance of drugs is then facilitated by Taken together the equation for hepatic clear-
metabolizing enzymes and transport proteins [1]. ance is:
Metabolism in the liver is a major determinant
Clh = (Q × f u ×Clint ) / (Q + f u ×Clint )
of elimination for a wide variety of drugs and the
hepatic clearance of medications can be affected This equation contains the three primary com-
by patient factors and drug properties. Intrinsic ponents of hepatic drug elimination: blood flow,
patient factors include volume status, perfusion, drug protein binding, and intrinsic clearance.
gut motility (specifically for orally administered Intrinsic clearance can be defined as the sum of
medication), and organ function. The major drug all enzyme and transport activity involved in
properties that affect the quantity of drug elimi- hepatic metabolism. The chemical makeup of a
nation by the liver include hydrophilicity/lipo- drug will determine its susceptibility to hepatic
philicity, extraction ratio, and protein binding [1]. enzymatic metabolism.
To fully understand the impact of hepatic dys- Medications can be categorized according to
function on PK and PD properties of a medication, the extraction efficiency (Eh): high (Eh < 0.7), low
an appreciation of the underlying determinants of (Eh < 0.3) or intermediate (0.3 < Eh < 0.7). Drugs
normal hepatic clearance is necessary. Hepatic with a high extraction ratio are dependent on
clearance (Clh) of a medication is a function of the blood flow and usually relatively insensitive to
hepatic blood flow (Q) and the extraction effi- changes in protein binding or enzyme activity
ciency of the liver for the particular drug (Eh) [2] (Clh  ≈ Q). On the other hand, drugs with low
and it can be represented by the formula: extraction efficiency are affected by changes in
protein binding and intrinsic hepatic clearance
Clh = Q × Eh
(Clh  ≈ fu × Clint) [2]. See Table 5.1 for a list of

Table 5.1  Classifications of relevant medication PK characteristics [4]


Effect of
Hepatic portosystemic
PK profile extraction shunts Examples
Low extraction/low <0.3 None Alprazolam, amoxicillin, doxycycline, fluconazole, isoniazid
protein binding lamivudine, methylprednisolone metronidazole,
(<90%) phenobarbital, prednisone, primidone, theophylline
Low extraction/ <0.3 None Ceftriaxone, chlordiazepoxide, clarithromycin, clindamycin,
high protein diazepam, lansoprazole, lorazepam, oxazepam, methadone,
binding (>90%) mycophenolate, phenytoin, prednisolone, rifampin, valproic
acid
Intermediate 0.3–0.6 Usually not Alfentanil, amiodarone, azathioprine, atorvastatin,
extraction clinically carvedilol, codeine, diltiazem, erythromycin, itraconazole,
relevant lidocaine, meperidine, nifedipine, omeprazole, ranitidine
High extraction >0.6 Clinically Fentanyl, isosorbide dinitrate, morphine, nitroglycerin,
significant sufentanil
5  Drug Metabolism in Liver Failure 71

relevant medications and their corresponding PK portosystemic shunt), which may significantly
profiles and expected effect of liver dysfunction. decrease liver blood flow [3]. Since high extrac-
tion drugs are mostly affected by hepatic blood
flow, cirrhosis may lead to a considerable decrease
 ffect of Liver Faiure on Medication
E in extraction of these medications and therefore
PK and PD an increase in bioavailability. In fact, studies that
evaluate medications with intermediate to high
Hepatic disease may result in many physiologic extraction ratios have found an increase (ranging
changes in the liver leading to alterations in med- from 2 to 12 fold) in bioavailability after enteral
ication PK and PD. administration in cirrhotic patients (Fig. 5.1) [4].
Aside from first-pass effect, cirrhosis may lead to
additional changes to the gastrointestinal tract.
Absorption These include increased plasma gastrin and
delayed gastric emptying and small bowel transit,
Absorption refers to the ability of a drug to which may also lead to erratic gastric absorption
migrate from the site of administration into the [5]. For high extraction drugs that are adminis-
bloodstream. The extent of absorption is typi- tered intravenously, a normal initial dose can be
cally measured in terms of bioavailability, defined administered and the maintenance dose should be
as the fraction of an administered dose that reduced according to hepatic blood flow.
reaches the systemic circulation. All drugs Theoretically, assessment of hepatic blood flow
administered outside the intravenous route are with sonography might be helpful to guide drug
affected by absorption. dosing for high extraction drugs in patients with
Drugs administered orally with a high extrac- significant shunt fraction; however, there is little
tion ratio would normally have a low bioavailabil- clinical evidence to support this approach [4]. The
ity given the significant first pass effect. However, serum bile acid level has shown good c­ orrelation
cirrhosis may lead to endogenous or therapeutic with the shunt index (r = 0.82) and may serve as a
porto-systemic shunts (transjugular intrahepatic surrogate for hepatic blood flow [6].

Effect of liver cirrhosis on pharmacokinetics


High hepatic extraction Low hepatic extraction
In drugs with a high hepatic
extraction, elimination is
retarded and maximal plasma Healthy individuals Healthy individuals
concentration and bioavailability
are increased. In drugs with a low Liver cirrhosis Liver cirrhosis
hepatic extraction, elimination is
retarded and maximal plasma
concentration and bioavailability
are unchanged. Thus, both initial
Plasma concentration

Plasma concentration

and maintenance doses must be


reduced when administering drugs
with a high hepatic extraction to
cirrhotic patients, whereas only the
maintenance dose must be
changed for drugs with low
hepatic extraction.

Time Time

Fig. 5.1  Effect of liver cirrhosis on concentrations of low and high extraction medications
72 S. W. Lam

Protein Binding and Distribution Hydrophilic drugs (e.g., β-lactams, aminoglyco-


sides, vancomycin, linezolid, colistin, morphine,
The distribution of a drug depends largely on the hydromorphone) tend to distribute within the
drug’s hydrophilicity and its acid dissociation plasma volume. In contrast, lipophilic drugs (e.g.,
constant, which affects its binding to proteins and azithromycin, fluoroquinolones, tetracyclines,
other macromolecules. Only a free drug, that is clindamycin, fentanyl, midazolam, propofol)
unbound from protein, can diffuse across tissue have sufficient Vd to penetrate tissue and cells
and have physiologic effect. Many drugs are outside of plasma volume. Serum concentrations
highly bound to either albumin or α1-acid glyco- of lipophilic drugs are only minimally affected by
protein. Decreased protein binding would result fluid shifts and third-spacing. However, water-
in increased free fraction of a drug and decreased soluble drugs may have a significant increase in
total plasma concentration (Fig. 5.2). Hepatic Vd because of the presence of peripheral edema
disease may decrease protein binding via reduced and ascites. As a result, the initial dose of a hydro-
synthetic protein production, accumulation of philic medication should be increased in order to
endogenous compounds that inhibit plasma pro- obtain a similar anticipated effect. Since many
tein binding, and conformational changes in pro- hydrophilic medications are excreted by the kid-
teins that may qualitatively alter binding. neys, renal function should also be considered
Decreased protein binding leads to an increased when choosing an appropriate dose [1].
free fraction and overall volume of distribution
(Vd). Although decreased binding would lower
the total serum concentration, increased free frac- Metabolism
tion may lead to a more pronounced therapeutic
effect. Numerous pathophysiologic changes during
Decreased protein binding is particularly chronic liver failure may affect drug metabolism
important for drugs with a low extraction ratio, and reduce intrinsic hepatic clearance (Clint). A
where hepatic clearance is largely dependent on reduction in liver cell mass may lead to decrease
fraction unbound and intrinsic clearance in enzymatic activity. Furthermore, sinusoidal
(Clh ≈ fu × Clint). Medications with a low extrac- capillarization may impair oxygen and com-
tion ratio can be further broken down to those pound uptake, which further limits drug metabo-
with high protein binding (≥90%) and those with lism. Two different types of reactions are
low protein binding (<90%) (Table 5.1). The primarily responsible for the liver’s metabolizing
drugs with low extraction ratio and low protein capabilities: phase I oxidative metabolism and
binding are most affected by hepatic enzymatic phase II glycosylation and glucuronidation.
activity or intrinsic clearance (Clint). Please refer Phase I reactions, which are usually mediated by
to the metabolism section below for a further the cytochrome P450 isoenzymes (CYP), require
review on the effects of hepatic disease on intrin- the presence of oxygen molecules and therefore
sic hepatic clearance activity. Drugs with low are more susceptible to functional deficiencies
extraction and high protein binding are equally due to lack of oxygenation from decreased
affected by Clint and fraction unbound. An impor- hepatic perfusion. Conjugation phase II reactions
tant distinction to realize in these drugs is that the such as glucuronidation are less susceptible to
total plasma concentration may be decreased the effects of liver cirrhosis [8].
while their free concentrations are either normal
or even increased [7].
Aside from protein binding, physiologic Elimination
changes seen in end-stage hepatic disease may
also lead to changes in Vd. Usually hydrophilic Medications are primarily removed by the kid-
drugs (high water solubility) have lower Vd than neys, although some medications can be excreted
lipophilic drugs (high lipid solubility). via the biliary tract, feces, and respiration.
5  Drug Metabolism in Liver Failure 73

Fig. 5.2  Influence of protein- Influence of protein-binding capacity on the total


binding capacity on the total plasma concentration of highly protein-bound drugs
plasma concentration of highly 0 20 40 60 80 100
protein-bound drugs

Under normal
conditions, 90% of the
drug is albumin-bound
and 10% is free
The total plasma
concentration is100.

Plasma albumin
concentration
decreased by one-third
after reaching the
steady state: the free
concentration is 10, the
free fraction is 14% and
the albumin-bound
fraction is 86%.
The total plasma
concentration is 70.

Plasma albumin
concentration
decreased by two-thirds
after reaching the
steady state: the free
concentration is 10,
the free fraction is 25%
and the albumin-bound
fraction is 75%. The total
plasma concentration is 40.

0 20 40 60 80 100
Plasma concentration
Free drug concentration
Albumin-bound drug concentration
Total drug concentration
74 S. W. Lam

Pathophysiologic processes, such as primary A decreased PD effect has been observed with
sclerosing cholangitis, cholangiocarcinoma, and various diuretic therapies, including furosemide,
primary biliary cirrhosis, may lead to both liver triamterene, torsemide, and bumetanide [12–15].
failure and extrahepatic cholestasis. Furthermore, In general, all of those studies found a decreased
hepatocyte dysfunction and slowing of bile flow PD response to diuretics in cirrhotic patients and
from the liver may lead to intrahepatic cholesta- a higher tubular concentration required to pro-
sis. Reduced formation and secretion of bile into duce the desired sodium excretion effect. One
the duodenum may lead to decreased clearance author suggested that the decreased PD response
of both endogenous and exogenous substances may be due to reductions in number of nephrons
that are primarily eliminated via biliary excre- or due to decreased maximum response per
tion. Drugs and metabolites that are normally ­nephron [15].
excreted by the bile may accumulate in liver fail- An increased PD effect of opioids and ben-
ure patients with biliary obstruction and cho- zodiazepines may be observed in cirrhotics.
lestasis [2]. These medications may cause disproportional
Advanced liver disease is frequently compli- sedation effects beyond PK changes [16–18].
cated by impaired kidney function due to hepa- Hypotheses for the physiological explanation
torenal syndrome. To further complicate for this phenomenon include increase blood
matters, patients with liver failure often have brain barrier permeability, increase in gamma-
reduced muscle mass and impaired metabolism aminobutyric acid (GABA) receptors, and
of creatine to creatinine. Therefore, equations increase GABA baseline activity via accumula-
such as the Cockroft-Gault method may overes- tion of endogenous non-­benzodiazepine recep-
timate true glomerular function. Hence clini- tor compounds.
cians must be cautious even when prescribing a
renaly eliminated medication in patients with
liver disease [2]. Liver Function Assessment

There are no physiologic or laboratory measure-


 harmacodynamic (PD) Changes
P ments to adequately estimate the hepatic clear-
in Liver Failure ance of medications unlike the assessment of
renal function by creatinine clearance that allows
Many studies have alluded to PD changes in a precise estimation of organ performance.
patients with liver disease. However, it should be Furthermore, given the complex interaction
pointed out that few of these studies have taken between drug properties and both physiologic
into account the PK alterations of hepatic dys- changes and altered intrinsic clearance activity in
function as discussed above. It is inherently dif- liver failure, it is unlikely that a single dynamic
ficult to demonstrate an altered therapeutic marker of liver function would accurately predict
response that is independent of the PK effects. PK changes for the majority of medications. The
The discussion on PD changes will focus on Child-Pugh classification of severity of liver dis-
instances where changes in drug receptor binding ease has been used extensively to categorize
or intrinsic activity of the receptor has been patients according to the severity of liver func-
demonstrated tion impairment (Table 5.2) [19]. Although the
Studies have indicated a decrease of the num- Child-­Pugh score is widely used for the assess-
ber of beta adrenoreceptor sites in patients that ment of prognosis in patients with liver cirrhosis,
may correspond with the degree of liver abnor- it does not reflect the hepatic clearance or PD of
mality [9–11]. This translates to both a decrease medications in those patients. As previously
in isoproterenol chronotopic effects [10] and a described, reduced liver function is the result of a
decrease therapeutic effect with B-adrenoreceptor combination of hepatocellular dysfunction and
antagonists [9, 11]. decreased blood supply with portal-systemic
5  Drug Metabolism in Liver Failure 75

Table 5.2  Child-pugh classification of liver disease [19]


Clinical criteria 1 point 2 points 3 points
Serum bilirubin (mg/dL) <2 2–3 >3
Serum albumin (g/dL) >3.5 2.8–3.5 <2.8
Prothrombin time (s > control) <4 4–6 >6
Encephalopathy (grade) None 1 or 2 3 or 4
Ascites Absent Slight Moderate
Points are aggregated and the total score is classified according to severity as follows: 5–6 points: group A (mild), 7–9
points: group B (moderate), 10–15 points: group C (severe)

shunts. The Child-Pugh Score does not provide patients with liver disease and an empiric dose
objective data for either of those functions. reduction of 50% should be employed. Clinicians
Furthermore, two of the five components of the should also be cognizant of possible prolonged
Child-Pugh Score (encephalopathy and ascites) sedative effects. The PK discoveries of mid-
are subjective and may alter with treatment. azolam and diazepam in patients with liver dis-
Despite these deficiencies, the Child-Pugh score ease are in contrast to the findings involving
is endorsed by both the Food and Drug lorazepam. Studies of lorazepam (which is
Administration and the European Medicines metabolized by glucuronidation) in liver disease
Agency to categorize patients according to their have demonstrated little to no PK effects [21].
degree of hepatic impairment for pharmacoki- Propofol is a rapidly acting anesthetic agent
netic studies. with multi-compartmental kinetics. It has an
extremely large Vd and an elimination half-life of
13–44 h [22]. In a controlled study of ten patients
Specific Classes of Medications with cirrhosis, anesthesia was induced with a
propofol infusion and PK parameters were mea-
Sedatives sured and compared with ten control patients
[23]. The investigators found that the termination
Patients with liver failure usually have more t1/2 and total body clearance of propofol were
exaggerated effects to sedatives, which may par- similar between the two groups. Although the
tially be explained by PD alterations, as discussed mean recovery time was significantly longer in
above. However, many of the sedatives com- the cirrhotic group, it did not translate to a clini-
monly used in the management of critically ill cally relevant difference. The authors concluded
liver failure patients also have significant PK that the PK parameters of propofol were not sig-
changes. nificantly affected by cirrhosis.
Midazolam is almost solely transformed by Dexmedetomidine is an α2-adrenergic agonist
CYP 3A4. Patients with moderate liver impair- with sedative properties, which is primarily
ment will experience changes in midazolam metabolized in the liver. It is mainly metabolized
PK. After a single intravenous dose of 0.2 mg/kg in the liver through direct glucuronidation (34%)
of midazolam in ten patients with moderate alco- [24], although it also undergoes hydroxylation by
holic liver disease, the area-under-curve (AUC) CYP enzymes. Dexmedetomidine also has a high
increased by 57% and the half life2 (t1/2) was pro- extraction ratio [24], suggesting that its pharma-
longed by 25% when compared to controls [20]. cokinetics is most likely to be affect by changes
In patients with severe liver cirrhosis, the AUC in blood flow. The effect of hepatic impairment
and t1/2 could potentially double [18]. Similarly, on dexmedetomidine PK was assessed in a phase
investigations of diazepam, which is metabolized I, single dose study with healthy subjects and
CYP, have also demonstrated doubling of elimi- subjects with varying degrees of hepatic insuffi-
nation t1/2 in cirrhotic patients. Diazepam and ciency [25]. A number of findings were observed,
midazolam should be used with caution in including significant decreases in protein binding
76 S. W. Lam

(from 94% in normal patients to 82% in severe vecuronium is primarily dependent on redistribu-
hepatic disease); and decreased clearance. tion termination; however, larger doses depend
Clearances were 74%, 64% and 53% of those in on hepatic function. A dose of <0.1 mg/kg has a
normal patients when compared, respectively, to slower onset and shorter duration of action in cir-
patients with mild, moderate, and severe liver rhotics, which is most likely attributable to
disease. The duration of effect of dexmedetomi- increase Vd. A dose of >0.2 mg/kg has a similar
dine is expected to increase in patients with liver onset time in cirrhotic patients, but a significant
disease due to an increase of the unbound frac- increase in duration of action (91 vs. 65 min)
tion and a decrease of hepatic enzymatic metabo- [30]. Rocuronium elimination is dependent on
lism. Per package insert recommendations, dose biliary excretion as an unchanged drug. A small
adjustments should be made in patients with proportion of rocuronium is also renaly excreted.
severe hepatic dysfunction, although no specific Studies in liver failure patients have demon-
recommendations are provided. Given the strated a larger Vd (longer onset of action) and a
increase of the unbound fraction and clearance prolonged duration of action. Atracurium and
rates, it would be reasonable to start at half the cisatracurium both undergo Hoffmann degrada-
normal dose in patients with severe liver disease. tion and ester hydrolysis. Studies have demon-
strated that the presence of end-stage liver disease
does not alter the elimination t1/2 of either medi-
Neuromuscular Blocking Agents cations [31]. In patients with liver disease where
a prolonged action of neuromuscular blockade
Hepatic failure may contribute to alterations of may not be desirable, preference should be given
PK and PD of neuromuscular blocking medica- to these two agents. However, both of these
tions. Factors leading to these alterations include agents also exhibit increased volume of distribu-
decrease elimination, altered Vd, acid base distur- tion in hepatic disease, and may have a longer
bances, and reduced plasma cholinesterase activ- onset of action.
ity. Prolonged neuromuscular blockade following Recently suggamadex was approved by the
succinylcholine administration has been reported US Food and Drug Adminstration to reverse the
in patients with liver dysfunction [26]. effects of neuromuscular blockade induced by
Furthermore, a delayed onset of action has been rocuronium. Summagadex chelates rocuronium in
observed possibly due to an increased Vd in cir- the plasma, leading to a decrease in concentration
rhotics [27, 28]. of free rocuonium. The ­suggamadex-­rocuronium
Of the neuromuscular blocking agents, pan- complex is rapidly excreted by the kidneys. As
curonium, vecuronium, and rocuronium are the discussed above, patients with liver dysfunction
most likely to be affected by end-stage liver dis- are at risk for prolonged neuromuscular block-
ease. Pancuronium is primarily renally elimi- ade. In an observational study of patients with
nated; however, 35% of it undergoes hepatic liver dysfunction undergoing hepatic surgery,
metabolism with biliary excretion. The Vd of suggamadex led to rapid reversal of neuromus-
pancuronium is increased by 50% in cirrhotics cular blockade [32]. When compared to patient
and its clearance is reduced resulting in a pro- without liver dysfunction, reversal of neuromus-
longed t1/2 (114–208 min). Patients may require a cular blockade was only prolonged for 0.2 min.
larger initial dose for desired effect, but slower Pharmacokinetically, suggamadex is primarily
elimination may lead to prolonged neuromuscu- renally eliminated, with negligible protein bind-
lar blockade [29]. Vecuronium is predominantly ing, and has a low volume of distribution [33].
eliminated via biliary excretion, and only a small As such, in those with hepatic dysfunction,
portion undergoes hepatic metabolism to an there are few anticipated effects aside from the
active metabolite. The effect of liver dysfunction possibility of third-spacing leading to a higher
on the PK of vecuronium depends on the dose volume of distribution and, hence, lower serum
administered. Elimination of smaller doses of concentration.
5  Drug Metabolism in Liver Failure 77

Opioids All of them are metabolized by CYP 3A4; how-


ever, the redistribution from the peripheral to cen-
Morphine is an opioid with partial μ receptor tral compartment is usually the rate-­limiting step.
agonist activity. It is metabolized to intermediate In cirrhosis, hepatic elimination becomes slower
metabolites, including morphine-6-glucuronide than redistribution and turns into the rate-limiting
and morphine-3-glucuronide via phase II reac- step. In general, their PK parameters are spared in
tions that are mostly spared in liver disease. It is mild liver disease, but in severe disease the free
usually 30–40% protein bound to albumin and fractions are higher given decreased protein bind-
extrahepatic clearance accounts for about 40% of ing and the t1/2β is prolonged. The PK of sufentanil
its elimination [34]. For the most part, morphine and fentanyl are more likely to be significantly
PK is unaltered in early liver disease. However, altered by liver disease since their extraction
in severe liver disease, the t1/2 is doubled, and cor- ratios are higher than that of alfentanil (0.8 vs.
relates with prolonged prothrombin time, hypoal- 0.4) [34, 35]. Remifentanil is rapidly acting and
buminemia, encephalopathy, ascites and jaudice metabolized by plasma esterases. Studies have
[17]. The intermediate metabolites of morphine shown that remifentanil PK are not affected by
are renally eliminated and the presence of hepa- liver dysfunction.
torenal syndrome may drastically prolong their Two recent peripherally acting μ-opioid recep-
elimination. In general, the initial intravenous tor antagonists, alvimopan and methylnaltrexone,
dose of morphine does not need to be adjusted to have been evaluated for the treatment of post-­
obtain the desired effect. However, clinicians operative ileus. Alvimopan is orally administered,
should be cognizant of the potential for pro- metabolized to an active metabolite by gut-flora,
longed duration of action and possible increases and eliminated primarily by biliary secretion [36].
in neuroexcitation toxicity, particularly in the In patients with severe hepatic dysfunction, the
presence of hepatorenal syndrome. maximum serum concentration is tenfold higher
Hydromorphone is metabolized via gluc- when compared to healthy volunteers. The pack-
uronidation to hydromorphone-3-glucuronide age insert of alvimopan recommends against its
that is an inactive metabolite but may be neu- use in patients with severe hepatic insufficiency,
rotoxic. Little is known about the PK of intra- and to exercise caution when administering in
venous hydromorphone in patients with liver patients with mild to moderate hepatic disease
dysfunction. However, the PK of orally admin- (Child-Pugh Class A or B). Although alvimopan
istered hydromorphone is moderately affected does not readily crosses the blood-brain barrier,
by liver disease. In patients with advanced cir- there are concerns that increased serum concen-
rhosis, the max concentration (Cmax) and AUC of tration may lead to opioid withdrawal symptoms.
hydromorphone was fourfold higher than nor- In contrast, methylnaltrexone (when administered
mal and the t1/2 remains unchanged. It is unclear subcutaneously) is primarily excreted unchanged
whether these PK changes are solely due to by the kidneys and studies in patients with mild to
increases in the bioavailability from oral admin- moderate hepatic disease did not demonstrate a
istration as a result of decreased extraction and significant increase in either peak concentrations
are not applicable when hydromorphone is or duration of action [37].
given parenterally.
The piperidine opioids (remifentanil, alfent-
anil, sufentanil, fentanyl) exhibit multiple-­ References
compartment PK, where the onset and magnitude
of action is dependent on distribution half-life 1. Rodighiero V. Effects of liver disease on phar-
macokinetics. An update. Clin Pharmacokinet.
(t1/2α), while the duration of action is dependent on 1999;37(5):399–431.
both t1/2α and elimination half-life (t1/2β). Alfentanil, 2. Verbeeck RK. Pharmacokinetics and dosage adjust-
sufentanil, and fentanyl are all highly protein ment in patients with hepatic dysfunction. Eur J Clin
bound (85–96%) and rapidly distribute to tissues. Pharmacol. 2008;64(12):1147–61.
78 S. W. Lam

3. Morgan DJ, McLean AJ. Clinical pharmacokinetic 20. Trouvin JH, Farinotti R, Haberer JP, Servin F, Chauvin
and pharmacodynamic considerations in patients M, Duvaldestin P. Pharmacokinetics of midazolam
with liver disease. An update. Clin Pharmacokinet. in anaesthetized cirrhotic patients. Br J Anaesth.
1995;29(5):370–91. 1988;60(7):762–7.
4. Delco F, Tchambaz L, Schlienger R, Drewe J, 21. Kraus JW, Desmond PV, Marshall JP, Johnson RF,
Krahenbuhl S. Dose adjustment in patients with liver Schenker S, Wilkinson GR. Effects of aging and liver
disease. Drug Saf. 2005;28(6):529–45. disease on disposition of lorazepam. Clin Pharmacol
5. Kalaitzakis E. Gastrointestinal dysfunction in liver cir- Ther. 1978;24(4):411–9.
rhosis. World J Gastroenterol. 2014;20(40):14686–95.
22. Morgan DJ, Campbell GA, Crankshaw
6. Ohkubo H, Okuda K, Iida S, Ohnishi K, Ikawa S, Makino DP. Pharmacokinetics of propofol when given by intrave-
I. Role of portal and splenic vein shunts and impaired nous infusion. Br J Clin Pharmacol. 1990;30(1):144–8.
hepatic extraction in the elevated serum bile acids in 23. Servin F, Cockshott ID, Farinotti R, Haberer JP,

liver cirrhosis. Gastroenterology. 1984;86(3):514–20. Winckler C, Desmonts JM. Pharmacokinetics of
7. Verbeeck RK, Horsmans Y. Effect of hepatic insuf- propofol infusions in patients with cirrhosis. Br J
ficiency on pharmacokinetics and drug dosing. Pharm Anaesth. 1990;65(2):177–83.
World Sci. 1998;20(5):183–92. 24. Valitalo PA, Ahtola-Satila T, Wighton A, Sarapohja T,
8. Westphal JF, Brogard JM. Drug administration in Pohjanjousi P, Garratt C. Population pharmacokinet-
chronic liver disease. Drug Saf. 1997;17(1):47–73. ics of dexmedetomidine in critically ill patients. Clin
9. Caujolle B, Ballet F, Poupon R. Relationship among Drug Investig. 2013;33(8):579–87.
beta-adrenergic blockade, propranolol concentration, 25. Karol MD, Maze M. Pharmacokinetics and interaction
and liver function in patients with cirrhosis. Scand J pharmacodynamics of dexmedetomidine in humans.
Gastroenterol. 1988;23(8):925–30. Best Pract Res Clin Anaesthesiol. 2000;14:261–9.
10. Gerbes AL, Remien J, Jungst D, Sauerbruch T,
26. Viby-Mogensen J, Hanel HK. Prolonged apnoea after
Paumgartner G. Evidence for down-regulation of suxamethonium: an analysis of the first 225 cases
beta-2-adrenoceptors in cirrhotic patients with severe reported to the Danish Cholinesterase Research Unit.
ascites. Lancet. 1986;1(8495):1409–11. Acta Anaesthesiol Scand. 1978;22(4):371–80.
11. Janku I, Perlik F, Tkaczykova M, Brodanova
27. Donati F. Onset of action of relaxants. Can J Anaesth.
M. Disposition kinetics and concentration-effect 1988;35(3 Pt 2):S52–8.
relationship of metipranolol in patients with cir- 28. Khalil M, D’Honneur G, Duvaldestin P, Slavov

rhosis and healthy subjects. Eur J Clin Pharmacol. V, De Hys C, Gomeni R. Pharmacokinetics and
1992;42(3):337–40. ­pharmacodynamics of rocuronium in patients with
12. Dao MT, Villeneuve JP. Kinetics and dynamics of cirrhosis. Anesthesiology. 1994;80(6):1241–7.
triamterene at steady-state in patients with cirrhosis. 29. Ward ME, Adu-Gyamfi Y, Strunin L. Althesin and
Clin Invest Med. 1988;11(1):6–9. pancuronium in chronic liver disease. Br J Anaesth.
13. Marcantonio LA, Auld WH, Murdoch WR, Purohit 1975;47(11):1199–204.
R, Skellern GG, Howes CA. The pharmacokinetics 30. Hunter JM, Parker CJ, Bell CF, Jones RS, Utting
and pharmacodynamics of the diuretic bumetanide JE. The use of different doses of vecuronium in
in hepatic and renal disease. Br J Clin Pharmacol. patients with liver dysfunction. Br J Anaesth.
1983;15(2):245–52. 1985;57(8):758–64.
14. Schwartz S, Brater DC, Pound D, Green PK, Kramer 31. Craig RG, Hunter JM. Neuromuscular blocking drugs
WG, Rudy D. Bioavailability, pharmacokinetics, and and their antagonists in patients with organ disease.
pharmacodynamics of torsemide in patients with cir- Anaesthesia. 2009;64(Suppl 1):55–65.
rhosis. Clin Pharmacol Ther. 1993;54(1):90–7. 32. Fujita A, Ishibe N, Yoshihara T, Ohashi J, Makino
15. Villeneuve JP, Verbeeck RK, Wilkinson GR, Branch H, Ikeda M, et al. Rapid reversal of neuromuscular
RA. Furosemide kinetics and dynamics in patients with blockade by sugammadex after continuous infusion of
cirrhosis. Clin Pharmacol Ther. 1986;40(1):14–20. rocuronium in patients with liver dysfunction under-
16. Bakti G, Fisch HU, Karlaganis G, Minder C, Bircher going hepatic surgery. Acta Anaesthesiol Taiwanica.
J. Mechanism of the excessive sedative response of 2014;52(2):54–8.
cirrhotics to benzodiazepines: model experiments 33. Keating GM. Sugammadex: a review of neuromuscu-
with triazolam. Hepatology. 1987;7(4):629–38. lar blockade reversal. Drugs. 2016;76(10):1041–52.
17. Davis M. Cholestasis and endogenous opioids: liver 34. Tegeder I, Lotsch J, Geisslinger G. Pharmacokinetics
disease and exogenous opioid pharmacokinetics. Clin of opioids in liver disease. Clin Pharmacokinet.
Pharmacokinet. 2007;46(10):825–50. 1999;37(1):17–40.
18. MacGilchrist AJ, Birnie GG, Cook A, Scobie G,
35. Ferrier C, Marty J, Bouffard Y, Haberer JP, Levron JC,
Murray T, Watkinson G, et al. Pharmacokinetics Duvaldestin P. Alfentanil pharmacokinetics in patients
and pharmacodynamics of intravenous midazolam with cirrhosis. Anesthesiology. 1985;62(4):480–4.
in patients with severe alcoholic cirrhosis. Gut. 36.
Curran MP, Robins GW, Scott LJ, Perry
1986;27(2):190–5. CM. Alvimopan. Drugs. 2008;68(14):2011–9.
19. Pugh RN, Murray-Lyon IM, Dawson JL, Pietroni MC, 37. Garnock-Jones KP, McKeage K. Methylnaltrexone.
Williams R. Transection of the oesophagus for bleed- Drugs. 2010;70(7):919–28.
ing oesophageal varices. Br J Surg. 1973;60(8):646–9.
Evaluation of Liver Function
6
Vanessa Cowan

Keywords (ALT) are intracellular enzymes that are


Liver function test · Bilirubin · Indocyanain released during hepatocyte injury and reflect
green · Dynamic liver function tests · the degree of liver injury; for example after
Monoethylgycinexylidide test · Sorbitol ischemia-reperfusion injury. Metabolic func-
clearance tion is frequently assessed by measuring albu-
min and protein levels. Biliary obstruction or
injury causes a release of the enzyme alkaline
Introduction phosphatase that lines the epithelium of the
bile ducts. Metabolic function is assessed by
Conventional liver function tests are insensitive measuring total (unconjugated and conjugated)
and not able to detect discreet injury that affect bilirubin and direct (conjugated) bilirubin.
mostly oxidative pathway of liver metabolism. Unconjugated (lipophilic) bilirubin, as a prod-
Assesment of liver function is important for risk uct of heme (but also myoglobin and cyto-
estimation prior to liver resections and transplants. chrome) metabolism, binds to albumin and is
This chapter will review limitations of conven- transported to the smooth endoplasmic reticu-
tional liver function tests and discuss the use of lum of hepatocytes where it undergoes gluc-
more sensitive, dynamic liver functions tests. uronidation by UDP-glucuronyl transferase 1.
The hydrophilic conjugated bilirubin is then
transported through caniculi to the bile ducts
 ynamic (Quantitative) Liver
D and excreted. This phase II glucuronidation is
Function Tests quite resistant to injury and therefore bilirubin
levels are usually not affected unless a substan-
Conventional liver function test measure mul- tial amount of liver function is lost.
tiple aspects of liver function and injury. Dynamic liver function tests directly measure
Transaminases such as aspartate aminotrans- hepatic metabolism, including phase I oxidative
ferase (AST) and alanine aminotransferase pathways, that are more sensitive to ischemic
injury and thus may be abnormal even with mild
liver dysfunction (Fig. 6.1) [1]. The chemicals
used for dynamic liver function tests are metabo-
V. Cowan, MD lized almost solely by the liver, commonly using
Transplant Institute, Beth Israel Deaconess Medical the cytochrome P-450 pathway, and may repre-
Center, Boston, MA, USA sent a more accurate measurement of liver func-
e-mail: vcowan@bidmc.harvard.edu

© Springer International Publishing AG, part of Springer Nature 2018 79


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_6
80 V. Cowan

Assessing liver function

Static tests Dynamic tests

Hepatocellular Clearance Elimination Metabolite


Excretion Cholestasis Synthesis
integrity half-life capacity formation

Bilirubin AP ASAT Albumin Galatose


γ-GT ALAT Coagulation
(factors V and VII) Indocyanine green (ICG)
GLDH
Caffeine 14 Metabolite
CO2
Bromosulfophthalein exhalation in serum

14
[C] aminopyrine Lidocaine
14
[C] erythromycin Midazolam
14
[C] methacetin

Fig. 6.1  Dynamic and static liver function tests (with Opin Crit Care. 2007 Apr;13(2):207–14. Wolters Kluwer
permission: Sakka SG: Assessing liver function; Curr Health, Inc.)

tion at a given point in time. Common dynamic ICG levels follow a typical pattern after IV
liver function tests include indocyanine green bolus. There is an initial peak that can be used to
(ICG) clearance, monoethylglycinexylidide determine cardiac output, a redistribution phase
(MEGX) test and sorbitol clearance. representing ICG distribution in the body and
finally a hepatic elimination phase that lasts
10–20 min. In normal patients, 97% of the dye
Indocyanine Green Clearance will have been excreted into bile within 20 min of
injection; the half-life of ICG is 3–4 min in nor-
Indocyanine green (ICG) clearance was one of mal subject but prolonged in patients with liver
the first dynamic tests of liver function and is one disease. ICG is generally well tolerated by
of the most widely used in the clinical setting. patients with low incidence of adverse reactions.
ICG is a water-soluble non-toxic tricarbocyanine Most reactions are mild allergic reactions mani-
dye. A principle feature of ICG metabolism is its fested by urticaria and headaches. Severe reac-
exclusive extraction by hepatic parenchyma and tions such as anaphylaxis are rare (1:40,000).
near complete elimination into the bile without Because it contains iodine, ICG should be
entering the enterohepatic circulation. Within avoided in patients with iodine allergy or thyro-
seconds of intravenous injection of ICG, it is toxicosis [2, 3].
bound to plasma proteins. ICG volume of distri- Clinically, several ICG measurement parame-
bution is roughly plasma volume. Protein bound ters are used such as Indocyanine green clearance
ICG is then taken up by parenchymal cells of the (CL-ICG; normal range 500–750 mL/min),
liver and excreted via the canalicular membrane plasma disappearance rate which is the percent-
into bile in an unchanged form. ICG metabolism age of ICG eliminated in 1 min after a ICG bolus
is therefore dependent on blood flow to the liver, (ICG-PDR; normal range 18–25%/min) and
hepatocellular uptake and biliary excretion. retention rate at 15 min (ICGR15; normal range
6  Evaluation of Liver Function 81

0–10%). ICG-PDR and ICGR15 are the most  otential Areas for Clinical
P
widely used parameters. Application
The gold standard for measuring ICG clear-
ance is spectrophotometric concentration analy- Critically Ill Patients
sis on serial blood samples over set time Traditional tools for predicting ICU survival will
intervals however this method is cumbersome, consist of organ injury or physiologic scores such
invasive, costly and requires a significant time as APACHE II, MOD score, SOFA, SAPS II. The
investment. Easy to use, non-invasive bedside prognostic value of ICG-PDR in ICU patients has
devices for the measurement of ICG elimination been investigated in small prospective studies.
are now available for clinical use (LiMON ICG-PDR is significantly lower in non-survivors
(Pulsion Medical Systems, Germany) or DDG than survivors across different subset of ICU
2001 (Nihon Kohden, Japan)). To measure ICG patients. An ICG-PDR < 8% predicted death with
elimination patients wear a finger clip, similar sensitivity and specificity of 81% and 70%
to a conventional pulse oximeter. The transcuta- respectively, with mortality as high as 80%.
neous monitor uses pulse dye densitometry to Additionally, ICG-PDR was able to show signifi-
measure PDR-ICG after bolus injection of indo- cant liver dysfunction in patients sooner and
cyanine green by detecting changes in optical more often than traditional laboratory parameters
absorption (peak infrared absorption range of generally indicate [5, 6]. Patients admitted with
805–890 nm). Results are obtained within min- sepsis had significantly lower ICG-PDR than
utes and the test can be performed at the bedside non-septic patients. ICG-PDR within the first
in hemodynamically stable or unstable patients. 24 h of admission to ICU is as accurate as com-
A dose between 0.25 and 0.5 mg/kg is adequate plex scoring tools such as APACHE II and SAPS
for transcutaneous measurement. The invasive II in predicting ICU survival [6]. In evaluating
and non-invasive methods of detecting ICG- liver transplant patients, a high pre-operative
PDR are highly correlated, making the transcu- MELD score plus a low initial ICG-PDR pre-
taneous method of measurement a good, less dicted significantly longer ICU stay (9 vs. 4
invasive alternative [1, 2, 4]. days), longer hospital stay (42 vs. 22 days) and
As ICG metabolism depends on hepatic blood significantly higher mortality (40 vs. 0%) [7].
flow, hepatocellular uptake and biliary excretion ICG-PDR has also been studied as a tool to
any processes that disrupt any of these three ele- predict survival and need for transplantation in
ments will affect ICG elimination. Local or global acute liver failure. Across all etiologies of
factors that influence hepato-splanchinic blood acute liver failure, ICG-PDR on presentation
flow (i.e. arterial thrombosis, portal HTN, intrahe- was significantly lower in those patients who
patic shunting, low cardiac output) will affect did not spontaneously recover compared to
ICG elimination by the liver. There are circadian those who did. An ICG-PDR on day one of
variations in hepatic blood flow and therefore <6.3%/min predicted death or need for trans-
time of day can influence ICG elimination with plant with a sensitivity and specificity of 85.7
the lowest elimination observed in the afternoon and 88.9%. Additionally, an ICG-PDR of
and the highest elimination at night. Any modifi- <5.3%/min at any point in time predicted death
cation to liver blood flow, for example with pos- or need for transplantation with a sensitivity
tural changes, exercise or medications (ACE and specificity of 85.7% and 66.7% respec-
inhibitors or N-acetylcysteine) can impact ICG tively (compared to Kings College criteria with
elimination. Lastly, cholestasis interferes with a sensitivity and specificity of 69 and 92%) [8].
ICG elimination. Bilirubin is competitive inhibi- Similar results have been found in studies of
tor of indocyanine green because they bind to the pediatric patients, with ICG-PDR < 5%/min
same carrier protein within hepatocytes. ICG val- predicting need for transplantation while those
ues are 10–20% lower when serum bilirubin level with ICG-PDR >6%/min recovered with
is greater than 3 mg/mL [2]. ­medical therapy [4].
82 V. Cowan

 ajor Liver Resections


M Table 6.1  Indocyanine green retention rate at 15 min
(ICGR15; normal range 0–10%) and permissible extent of
Estimation of residual liver function prior to
liver resection [12]
major hepatic resections is important to avoid
postoperative liver failure and potentially death. ICGR15 value
(%) Extent of resection
This is particularly relevant (and difficult) in
<10 Right hepatectomy
patients with pre-existing liver disease. Liver
10–19 Left hepatectomy (up to 1/3 of liver
reserve is usually evaluated using scores such as parenchyma)
Child-Pugh and MELD score, static liver func- 20–29 Segmentectomy (up to 1/6 of liver
tion tests or imaging to determine remnant liver parenchyma)
volume in addition to the clinical judgment of the >30 Limited resection/enucleation
surgeon. A future liver remnant (FLR) of at least
20% is desired with a normal liver. However, a
greater FLR is needed if there is any liver injury adjunct to existing pre-operative assessment of
(30%) or fibrosis/cirrhosis (40%) present [9]. safe limits for hepatic resections.
Currently, post hepatectomy mortality rates are
low (between 0 and 5%). However, post hepatec- Liver Transplantation
tomy liver dysfunction or failure occurs in up to ICG clearance has been investigated as a predic-
30% of cases [10]. tive tool in all aspects of liver transplantation
In patients with pre-existing crisshosis, including pre-transplant mortality, donor selec-
hepatic resection is contraindicated in patients tion, post-operative complications and early graft
with Child C cirrhosis or a MELD score above function in both deceased and living donor liver
14. Surgical resection can be considered in transplants. MELD score is used to predict mor-
patients with Child’s A and selected patients tality in end stage liver disease patients; however
with Child’s B, or those with MELD scores it may not accurately reflect the degree of decom-
between 9 and 14. In many Asian centers pensation experienced by patients, especially for
ICGR15 is used to estimate the acceptable those with MELD scores in the middle ranges.
extent of hepatic resection and its use in preop- MELD-Na is one attempt at more accurately
erative assessment is part of the official guide- assessing patients with mid level MELD scores
lines for treatment of hepatocellular carcinoma to allow better prognostication. ICG half-life has
in Japan. Multiple studies have concluded that a linear relationship to mortality with risk of
liver resections can safely be performed with death rising by 2–4% for every additional minute
ICGR15 < 15% [10]. Imamura et al. developed above the normal range of 3–4 min. ICG half-life
a decision tree for determining extent of permis- is an independent predictor of outcome and can
sible hepatic resection using presence of ascites, be used in a complementary manner with MELD
serum bilirubin level and ICGR15 in patients score. When combined with MELD score, ICG
with impaired liver function. Patients with half-life increased the score by 5–10 points for
decompensated cirrhosis (bilirubin >2 and/or patients with a MELD score between 10 and 30.
uncontrolled ascites) were not candidates for This remains accurate even after interventions
hepatic resection (Table 6.1) [11]. such as TIPS that may change parameters such as
These guidelines were used in operative plan- creatinine and sodium. Validation of MELD-ICG
ning for 915 patients undergoing 1056 hepatecto- score is pending further large-scale prospective
mies, just over half of which were for HCC. There trials [13]. Small, older and retrospective studies
were no mortalities within 30 days of surgery, suggest that ICG-­PDR measurement may be able
overall morbidity was 39% but major morbidity to assist in the assessment of donor graft quality.
was only 5.6%. Within the HCC group, there was One of these studies showed ICG-PDR values of
only one instance of postoperative hepatic failure <15% per minute in the donor were associated
and a 3% major complication rate [12]. These with poor graft outcomes. However, none of
results suggest that ICGR15 may be a useful these small studies have been validated by larger
6  Evaluation of Liver Function 83

prospective studies [3]. With increasing use of exclude HAT thereby potentially avoiding inva-
extended criteria donors, this area deserves fur- sive angiography or contrasted CT scan [17].
ther investigation. Furthermore ICG-PDR can be In the living donor liver transplant population,
used as a marker for early postoperative compli- the ICG elimination rate constant (KICG) has been
cations. ICG-PDR and INR obtained within the studied. KICG less than 0.100/min is a strong pre-
first 24 h were independently associated with dictor of early graft loss [18]. Immediately after
mortality, need for retranplant, duration of LDLT, KICG is excellent, however those who
mechanical ventilation and length of ICU stay develop poor graft function have a significant
after liver transplantation. The authors developed decrease in ICG elimination constant over the
a scoring system that includes 2 or 0 points for first 48 h post transplant and low KICG was signifi-
ICG-PDR less than or greater than 10%/mL and cantly correlated with evidence of graft injury on
0 or 1 point for INR less than or greater than 2.2. biopsy [19].
As the score increased, patients were at higher It is important to note that a change in ICG
risk of death or need for retransplantation and elimination is not indicative of the cause of dys-
required longer time on mechanical ventilation function and results need to be interpreted within
and longer ICU stay [14]. Patients who did not the clinical context for individual patients. Use of
develop complications had stable and high ICG- ICG elimination may allow for earlier detection
PDR during the first 5 days after transplant (aver- of graft dysfunction and therefore earlier inter-
age 24.4%/min) however patients who developed ventions with the hope of improving outcomes.
early complications, including primary nonfunc-
tion, hepatic artery thrombosis, septic shock and
hemorrhage, had a low ICG-PDR (average 8.8%/ Other Dynamic Liver Function Tests
min). ICG-­PDR, in the correct clinical setting,
may further be able to predict rejection. A signifi- Lidocaine Metabolism
cant decrease in ICG-PDR was seen in patients and Monoethylgycinexylidide (MEGX)
who developed biopsy proven rejection, and this Test
preceded any laboratory change in liver enzymes The MEGX test is a dynamic liver function test
[15]. When evaluating the relationship between that measures the ability of the liver to convert
ICG-PDR and early graft function, those with lidocaine to monoethylgycinexylidide (MEGX).
severe graft dysfunction had significantly lower This allows for real-time measurement of hepatic
ICG-PDR post reperfusion and throughout the metabolic function. Lidocaine is primarily
early post operative period. ICG-PDR < 10.8%/ metabolized in the liver by the cytochrome P450
min, indicating severe graft dysfunction, had sig- system, specifically the CYP 3A4 and CYP 1A2
nificantly lower survival [16]. isoenzymes. Lidocaine has a relatively high
Hepatic artery thrombosis (HAT) is a poten- extraction ratio by the liver; therefore lidocaine
tially life threatening complication after liver metabolism depends on both hepatocyte meta-
transplantation with an incidence from 2 to 12%. bolic capacity and liver blood flow. Blood sam-
In up to 75% of cases retransplant is required and ples must be obtained prior to administration and
mortality approaches 50%. Early diagnosis is then 15 and/or 30 min after IV injection of a lido-
required in order for revascularization to work. caine bolus. MEGX concentrations are deter-
ICG-PDR is significantly lower in patients with mined using fluorescence polarization
HAT than in patients without HAT (5.8 vs. immunoassay, high performance liquid chroma-
23.8%/min) and ICG-PDR normalized in the tography or gas liquid chromatography. MEGX
post-operative setting if revascularization was testing is contraindicated in patients with cardiac
successful. With further validation, ICG-PDR disease, arrhythmias or lidocaine allergy but seri-
could be incorporated into standard evaluation ous adverse effects are rare when 1 mg/kg lido-
for HAT. In case of absent arterial flow on ultra- caine is used. Transient mild side effects include
sound but normal ICG-PDR, we may be able to tinnitus, vertigo, drowsiness and light-­
84 V. Cowan

headedness. Decreased hepatic blood flow and as a source of energy, thus increasing galactose
drugs that interfere with the cytochrome P-450 clearance from the blood. Despite its prognostic
system can affect MEGX test results. ability, this test is time consuming and too cum-
The MEGX test has been used in critical care bersome for routine clinical use [9, 21].
medicine and liver transplantations. A wide
inter-­individual variability of MEGX formation Sorbitol Clearance
makes it difficult to use the MEGX test during D-sorbitol is a nontoxic sugar that has a high rate
the initial stages of chronic hepatitis. However in of hepatic extraction and is rapidly metabolized
patients with advanced liver disease, MEGX lev- by the liver. D-sorbitol elimination from the
els decrease with worsening child-Pugh score. In blood correlates well with hepatic blood flow. In
general a MEGX concentration of <20 μg/L the setting of cirrhosis, total hepatic blood flow
15 min after injection indicates poor liver func- does not decrease but the effective blood flow
tion and low MEGX values at 15 and 30 min can decreases due to transhepatic and extrahepatic
be predictive of post transplant complications shunts that allow bypassing of the hepatic sinu-
and post transplant mortality. Some studies sug- soids. Therefore D-sorbitol elimination is more
gest that higher MEGX values are associated likely to be a reflection of functional hepatic
with good early outcomes after living donor liver blood flow. There are very few human studies of
transplantation. MEGX values that decrease sorbitol elimination and its usefulness in clinical
over time in the initial post op setting can be practice remains uncertain [1].
interpreted as a marker of major change in the
graft and possible complications such as HAT,
rejection, or sepsis. In the ICU setting, a sharp References
decrease in MEGX values from admission to
several days post admission to ICU corresponded 1. Wagener G. Assessment of hepatic function, opera-
with patients who developed multiorgan system tive candidacy, and medical management after liver
failure and/or death [1, 20]. Despite these find- resection in the patient with underlying liver disease.
Semin Liver Dis. 2013;33(3):204–12.
ings, MEGX testing is not widely used in the 2. Levesque E, Martine E, Dudau D, Lim C, et al.
clinical setting. Current use and perspective of indocyanine green
clearance in liver diseases. Anaesth Crit Care Pain
 alactose Elimination Capacity Test
G Med. 2016;35:49–57.
3. Vos JJ, Wietash JKG, Absalom AR, et al. Green
Galactose is a monosaccharide sugar that under- light for liver function monitoring using indocyanine
goes conversion to glucose in the liver. Galactose green? An overview of current clinical applications.
elimination can be used to determine the meta- Anaesthesia. 2014;69:1364–76.
bolic capacity of the liver. Galactose is adminis- 4. Halle BM, Poulsen TD, Pedersen HP. Indocyanine
green plasma disappearance rate as dynamic liver
tered intravenously and serial blood samples are function test in critically ill patients. Acta Anaesthesiol
obtained from 20 to 50 min post injection. Scand. 2014;58:1214–9.
Galactose elimination capacity has prognostic 5. Kortgen A, Paxian M, Werth M, Rechnagel P, et al.
significance in chronic liver disease and fulmi- Prospective assessment of hepatic function and
mechanisms of dysfunction in the critically ill. Shock.
nant hepatic failure and low galactose elimina- 2009;32(4):358–65.
tion capacity can predict postoperative 6. Sakka S, Reinhart K, Meier-Hellmann A. Prognostic
complications and death. Two clinical situations value of the indocyanine green plasma disap-
may cause false positive test results: during liver pearance rate in critically ill patients. Chest.
2002;122(5):1715–20.
regeneration the demand for galactose increases 7. Klinzing S, Brandi G, Stehberger P, et al. The com-
to support membrane synthesis because galactose bination of MELD score and ICG liver testing pre-
is an essential component of membrane glyco- dicts length of stay in the ICU and hospital ­mortality
proteins and glycolipids. During states of anaero- in liver transplant recipients. BMC Anesthesiol.
2014;14:103–9.
bic respiration, especially during fasting galactose 8. Merle U, Sieg O, Stremmel W, et al. Sensitivity and
is more rapidly converted into glucose and used specificity of plasma disappearance rate of indocya-
6  Evaluation of Liver Function 85

nine green as a prognostic indicator in acute liver fail- tool to evaluate early graft outcome after liver trans-
ure. BMC Gastroenterol. 2009;9:91–7. plantation. Liver Transpl. 2009;15:1358–64.
9. Mizuguchi T, Kawamoto M, Meguro M, et al. 16. Olmedilla L, Perez-Pena JM, Ripoll C, Garutti I,

Preoperative liver function assessments to estimate et al. Early noninvasive measurement of the indo-
the prognosis and safety of liver resections. Surg cyanine green plasma disappearance rate accurately
Today. 2014;44:1–10. predicts early graft dysfunction and mortality after
10. De Gasperi A, Mazza E, Prosperi M. Indocyanine deceased donor liver transplantation. Liver Transpl.
green kinetics to assess liver function: ready for a 2009;15:1247–53.
clinical dynamic assessment in major liver surgery? 17. Levesque E, Hoti E, Azoulay D, Adam R, et al. Non-­
World J Hepatol. 2016;8(7):355–67. invasive ICG-clearance: a useful tool for the man-
11. Imamura H, Sano K, Sugawara Y, et al. Assessment agement of hepatic artery thrombosis following liver
of hepatic reserve for indication of hepatic resection: transplantation. Clin Transpl. 2011;25:297–301.
decision tree incorporating indocyanine green test. J 18. Du A, Wei Y, Chen K, Chen X, et al. Risk factor
Hepato-Biliary-Pancreat Surg. 2005;12:16–22. and criteria predicting early graft loss after adult-to-­
12. Imamura H, Seyama Y, Kokudo N, Maema A, et al. adult living donor liver transplantation. J Surg Res.
One thousand fifty-six hepatectomies without mortal- 2014;187:673–82.
ity in 8 years. Arch Surg. 2003;138:1198–206. 19. Hori T, Lida T, Yagi S, Taniguchi K, et al. KICG value,
13. Zipprich A, Kuss O, Rogowski S, Kleber G, et al. a reliable real-time estimator of graft function, accu-
Incorporating indocyanin green clearance into the rately predicts outcomes in adult living-donor liver
model for end stage liver disease (MELD-ICG) transplantation. Liver Transpl. 2006;12:605–13.
improves prognostic accuracy in intermediate to 20. Oellerich M, Armstrong VW. The MEGX test: a tool
advanced cirrhosis. Gut. 2010;59:963–8. for the real-time assessment of hepatic function. Ther
14. Olmedilla L, Lisbona CJ, Perez-Pena JM, Lopez-­
Drug Monit. 2001;23:81–92.
Baena JA, et al. Early measurement of indocyanine 21. Hoekstra LT, de Graaf W, Nibourg GA, Heger

green clearance accurately predicts short-term out- M, et al. Physiological and biochemical basis of
comes after liver transplantation. Transplantation. clinical liver function tests: a review. Ann Surg.
2016;100(3):613–20. 2013;257(1):27–36.
15. Levesque E, Saliba F, Benhamida S, Ichai P, et al.
Plasma disappearance rate of indocyanine green: a
Part II
Anesthesiology for Liver Transplantation
History of Liver Transplantation
7
John R. Klinck and Ernesto A. Pretto

Keywords occasional serendipity, but was also built on


Rejection · Immunosuppression · Surgical decades of work by others. This chapter will out-
technique · Organ preservation · Brain death · line the early, hard-won advances in vascular sur-
Caval anastomosis gery, immunology, organ preservation, and
transplantation of the kidney that preceded suc-
cessful liver transplantation, and will describe the
Introduction further evolution of liver transplantation in the
past three decades.
Thomas Starzl (1926–2017) began clinical liver
transplantation in Denver, CO USA in 1963, soon
followed by Roy Calne (1930–) in Cambridge The Early Modern Era
UK. These two surgical pioneers built multidisci-
plinary teams and collaborated closely over the The technical and conceptual foundations of
following two decades, overcoming enormous transplantation were laid by Alexis Carrel (1873–
technical, biological and societal obstacles to 1944), who is widely regarded as the founding
establish transplantation as the only curative father of organ transplantation. He pursued an
treatment for most forms of end-stage liver dis- early interest in the techniques of vascular anas-
ease. The procedure was widely accepted by the tomoses, stimulated by the inability of surgeons
mid-1980s, and today more than 25,000 liver to save the life of the French Prime Minister Sadi
transplants are performed each year worldwide. Carnot, who was stabbed in the street in 1894.
This achievement involved tireless effort, Working with Mathieu Jaboulay (1860–1913) in
intelligent use of advances in other fields, and Lyon, he developed effective techniques of vas-
cular anastomosis and described their use in kid-
ney and heart transplantation in animals in a
celebrated paper in 1902. In 1903 Carrel emi-
J. R. Klinck, MD, FRCA, FRCPC (*) grated to Montreal and soon after to Chicago and
Division of Perioperative Care, Cambridge University
Hospitals, Cambridge, UK New York, where he continued work on surgical
e-mail: john.klinck@addenbrookes.nhs.uk technique and preservation of tissues and organs,
E. A. Pretto, MD, MPH for which he was awarded a Nobel Prize in 1912.
Division of Transplant and Vascular Anesthesia, Carrel later worked at the Rockefeller Institute in
University of Miami Leonard M Miller School of New York and collaborated with the famous avia-
Medicine, Jackson Memorial Hospital, tor and inventor Charles Lindbergh to develop an
Miami, FL, USA

© Springer International Publishing AG, part of Springer Nature 2018 89


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_7
90 J. R. Klinck and E. A. Pretto

apparatus for the extracorporeal perfusion of attempted the first human kidney allografts in
organs, a forerunner of modern perfusion pumps patients who had ingested mercury [3]. He used
and heart-lung bypass. Their book, The Culture Carrel’s techniques to graft kidneys from cadav-
of Organs, was published in 1938 and publicized eric donors to the femoral vessels of recipients
on the cover of Time magazine. under local anesthetic, hoping to obtain enough
Although his own research focused on surgi- function to sustain the recipients until their own
cal technique and tissue viability, Carrel was well kidneys recovered. However, since he accepted
aware of pre-1914 advances in the understanding the contemporary dogma that mismatching of
of host responses to foreign tissue, including the blood groups was unimportant, and was unaware
recognition of the morphology and unique behav- of the harmful effect of prolonged warm isch-
ior of the lymphocyte as distinct from other leu- emia, the grafts still invariably failed. Nonetheless,
kocytes, and of its sensitivity to chemicals and the technical feasibility of kidney implantation in
radiation. Speaking to the press on the occasion humans was demonstrated. Further progress in
of the 1914 meeting of the International Society clinical transplantation awaited advances in labo-
of Surgery in New York, he presciently summa- ratory immunology and chemical immunosup-
rized these developments and the aims of future pression, fields that finally regained the scientific
research in a ‘road map to organ transplantation’. spotlight in the 1950s.
However he did not pursue this emerging field
himself and its scientific momentum was lost
with the outbreak of the First World War.  iology of Rejection and Discovery
B
Important pre-war papers published in German of Immunosuppressive Agents
and French fell into obscurity in the post-war
years when leadership in medical research passed Naturally, Carrel’s laboratory transplants and
to the more affluent United States of America. Voronoy’s attempts to transplant human kidneys
Carrel’s ‘road map’ and progress in immunology in the late 1930s failed consistently due to isch-
faltered, and no further international conference emic injury or the abrupt onset of rejection.
addressing the challenges of transplantation Rejection was thought to be a non-specific
would take place until 1948. Historians refer to inflammatory reaction to any foreign tissue until
this early period as the ‘Lost Era’ of organ trans- Peter Medawar’s (1915–1987) groundbreaking
plantation [1]. work with skin grafts in the 1940s that revealed
Further progress in transplantation then that rejection was both acquired and donor-­
awaited the efforts of the Ukrainian surgeon Yuri specific. Medawar also confirmed that immune
Voronoy (1895–1961) in the 1930s and 1940s rejection was predominantly lymphocyte-­
[2]. Since renal failure had always been far more mediated, as ‘Lost Era’ investigators had sug-
common than liver failure and was usually fatal, gested. In the early 1950s this led to experimental
the potential benefits of kidney transplantation kidney transplants in which whole-body radia-
seemed enormous. After much experimentation tion and donor bone marrow infusion preceded
in animals, he saw an opportunity to attempt clin- implantation. This technique had safely sup-
ical kidney transplantation in the setting of acute pressed rejection in animals but was poorly toler-
renal failure from ingestion of mercury chloride, ated in humans, who often died of sepsis.
a cleaning product at that time increasingly taken However, the simultaneous discovery that corti-
to commit suicide. Experiments had demon- costeroids had immunosuppressive effects miti-
strated the possible role of lymphocyte-­containing gated this setback and proved to be a landmark in
tissues in transplant rejection, and mercury intox- transplant medicine. Endogenous steroids had
ication was known to be associated with atrophy been isolated in the 1930s and were synthesized
of lymphoid tissues such as spleen and lymph at huge expense in the late 1940s after rumors
nodes. Voronoy speculated that immune rejection that Luftwaffe pilots in World War II had used
might therefore be inhibited in this setting, and them to improve performance. Treatment of a
7  History of Liver Transplantation 91

range of inflammatory and autoimmune diseases established, aided by advances in microsurgery.


was transformed by this new ‘wonder drug’, and From then on the pace of experimental work in
transplant pioneers held similar hopes for the pre- the field quickened [4].
vention of rejection. Although it was soon evi-
dent that rejection was not always prevented by
hydrocortisone, it was clearly attenuated and  volution of Organ Preservation
E
risks to the patient were lower than with radia- Techniques and Solutions
tion. It later found a pivotal place in combination
with another breakthrough agent. The development of organ preservation tech-
The suppressive effects of nitrogen mustard niques has been vital to progress in solid organ
(“mustard gas”) on the bone marrow and immu- transplantation. The earliest attempts evolved
nity was known from military use of this agent in from an interest in evaluating physiological func-
both World Wars, and analogs were synthesized tion. Le Gallois [5] predicted that organ function
for treatment of lymphoid cancers in the mid-­ could be restored by perfusion with arterial
1940s. In the 1950s, the capacity of purine ana- blood, and Von Cyon, Ringer and Langendorff
logs to suppress immunity was recognized, and later studied the effects of ex-vivo normothermia,
Roy Calne (1930–) demonstrated that mostly with isolated mammalian hearts [6, 7].
6-­mercaptopurine yielded much better results Carrel described the benefits of cold storage of
than radiation in animals. He speculated correctly explanted vessels, and, with Lindbergh, devel-
that that a dominant effect on cell-mediated as oped a pumped perfusion apparatus able to main-
opposed to humoral immunity might account for tain organs for 20–40 days with normothermic
this, contrary to the belief that all immunosup- serum. This laid the groundwork for later
pression affected T and B-cell function equally, improvements in organ preservation by continu-
thereby inevitably exposing the recipient to lethal ous perfusion [8].
infection. Working with Joseph Murray (1919– Cooling of perfusate was explored in the
2012) in Boston, Calne showed even better 1950s, and its effectiveness later enhanced by
experimental results with another purine analog, altering its composition. In 1956 a canine kidney
azathioprine. However, clinical outcomes with autograft functioned after 24 h of hypothermic
sole use of this agent, as with radiation and with perfusion [9], and the use of cold perfusate was
combinations of the two, remained poor. widely adopted by the early 1960s. Belzer [10]
The next major advance awaited an empiric extended experimental preservation times,
discovery in the early 1960s, by Willard Goodwin obtaining consistent function after 72 h through
(1915–1998) and Thomas Starzl (1926–2017), the use of continuous pulsatile perfusion at 10 °C
that high-dose steroid treatment reversed acute with a cryoprecipitate plasma preparation.
rejection in kidney recipients receiving azathio- Toledo-Pereyra [11] later added silica gel to
prine. Starzl extended this important observation, plasma protein fraction, obtaining even better
suggesting that combining prophylactic low-dose results.
steroids and azathioprine might be beneficial. However, early perfusion preservation
This proved to be the case and rapidly became the required the use of complex, non-portable
standard immunosuppressive regime. Aided by devices, hampering its widespread application.
advances in HLA tissue matching and wider This difficulty was compounded by the emer-
availability of dialysis, kidney transplant became gence of HLA matching for clinical kidney trans-
an established, life-saving treatment. Technical plants in the late 1960s, which increased distances
success and better immunosuppression in kidney between matched donor-recipient pairs. The
transplantation inspired a new generation of sur- impracticality of moving donors or perfusion
geons and immunologists. Small animal models apparatus, and the need for longer preservation
of kidney, heart and liver transplants, designed to times, stimulated further research into simple
study immunosuppression, were by then well cold storage. Although well described in the
92 J. R. Klinck and E. A. Pretto

1950s, and widely adopted in experimental liver sue donation on death, or for relatives to consent
and heart transplantation in the 1960s, simple in the absence of known wishes. However, as
cold storage with solutions then available could described below, these legal initiatives and an
not sustain viability long enough for clinical kid- entirely co-incidental redefinition of death put
ney transplants. G.M. Collins and Paul Terasaki forward by a Harvard committee, were to encoun-
(1929–2016) introduced a superior preservation ter a major setback by high-profile failures fol-
solution and a practical alternative to hypother- lowing the first human heart transplants in 1968,
mic perfusion in 1969 [12]. This was a crystalloid the ‘Year of the Heart’. A series of heart trans-
constituted to resemble intracellular fluid, thereby plant deaths as a consequence of an unseemly
reducing sodium influx and osmotic cell swelling surgical rush for the media spotlight undermined
allowing unperfused cold storage of the kidney public trust and support for transplantation for
for up to 30 h. Although machine perfusion more than 10 years.
remained in use in some centers, and portable During the same decade, advances in anesthe-
pumps were soon developed, Collins’ solution sia had an important impact on the progress of
and its simpler derivative, Euro Collins, rapidly organ transplantation. Muscle relaxants and
became the mainstay of solid organ preservation mechanical ventilators, introduced during the
worldwide. 1950s, improved patients’ tolerance for lengthy
The next significant advance in organ preser- operations and allowed them to be ventilated
vation was reported in 1992 by Folkert Belzer postoperatively, thereby preventing the immedi-
(1931–1995) at the University of Wisconsin with ate death of patients with severe head injuries.
the introduction of the “UW” solution [13]. This The advent of clinical blood gas measurements
solution contained impermeants, shown to fur- facilitated this, and specialist respiratory care
ther reduce cell swelling, as well as anti-oxidants wards were created. Peter Safar (1924–2003), a
and other agents thought to preserve ATP produc- pioneer of cardiopulmonary resuscitation, set up
tion and attenuate ischemia-reperfusion injury. It such a unit at Baltimore City Hospital in 1958,
was very effective in the preservation of kidney coining the term ‘Intensive Care’. This was soon
and liver grafts, though less effective in pancreas replicated in Boston, where admissions increased
and heart. In the 1990s H.J. Bretschneider sixfold between 1961 and 1966.
(1922–1993) introduced the histidine-trypto- As the management of critically ill patients
phan-glutarate (HTK) solution [14]. The UW and became more skillful, it was recognized that
HTK solutions safely extended kidney preserva- some patients who were admitted to Intensive
tion times to 24 h or longer, thereby facilitating Care were kept alive without hope of neurologi-
organ sharing across large geographic regions. cal recovery, causing distress to staff and fami-
lies. Robert Schwab (1903–1972), a neurologist
at the Massachusetts General Hospital, addressed
 rain Death and Heart-Beating
B this in an analysis of prognostic signs associated
Donation with consistent post-mortem findings of exten-
sive, irreversible brain injury. His confidence in
Advances in clinical transplantation in the 1960s these signs led to determination of ‘brain death’
focused attention on legal issues related to organ and consensual termination of artificial ventila-
donation. In many countries no legal framework tion in an increasing number of patients in the
existed and removal of organs from cadavers mid-1960s. The definition of death was debated
depended only on approval by authorities in local in medical, religious and legal circles but not
hospitals. Where laws existed, they addressed resolved. At a meeting of leading transplant sur-
donation for anatomical teaching and corneal geons in London in 1966, the removal of organs
grafting, not rapid removal for transplant. Most from a ‘brain-dead’ donor was reported but
developed countries created a legal framework to essentially rejected by Calne, Starzl and others.
allow individuals to register their consent to tis- Their views were echoed by Norman Shumway
7  History of Liver Transplantation 93

(1923–2006) from UCLA the following year, to-­son living donation. Grafts were now blood
who acknowledged the great advantage this group compatible and some recipients were
would offer, particularly in cardiac transplanta- treated with a costly new immunosuppressant,
tion, but also realized that attitudes in both the hydrocortisone.
medical profession and society would need to In Boston, a newly developed hemodialysis
change before this would be possible. machine, pioneered by Willem Kolff (1911–
Nonetheless, transplant surgeons began to con- 2009) in the Netherlands in 1945, was introduced
sider the possibility of heart-beating donation by John Merrill (1917–1984). Many senior phy-
and to seek the opinions of colleagues in critical sicians, known colloquially as the ‘salt and water
care and neurology. club’ because of their belief that skillful fluid and
Henry Beecher (1904–1976), Professor of dietary management was the key to treatment of
Anesthesia Research at Harvard, pursued this renal failure, initially viewed this device with
matter as chair of the medical faculty’s Standing skepticism. However, innovative US Army doc-
Committee on Human Studies. Motivated by tors working in the MASH (mobile army surgical
what he regarded as the indignity of prolonged, hospital) system in Korea rapidly proved its
futile artificial ventilation, he established a sub-­ value. In Boston, dialysis was used to optimize
committee, the Harvard Ad Hoc Committee on patients before transplant and to sustain them
Brain Death. The committee’s groundbreaking while ischemic grafts recovered, making more
report was published in the Journal of the transplants possible and improving early out-
American Medical Association in August 1968, comes. Wartime advances in vascular repair and
amid growing public disquiet at poor results of the use of homograft blood vessels also contrib-
human heart transplants around the world. An uted to surgeons’ confidence in pursuing experi-
association was inevitably inferred between the mental and clinical transplantation.
redefinition of death to prevent fruitless prolon- Although the problem of rejection remained, in
gation of intensive care and the needs of organ 1954 Joseph Murray (1919–2012) achieved long
transplantation, undermining trust in the medical term function by transplanting a kidney between
profession and deterring legislators from chang- identical twins. This was done without the use of
ing the law. Although critical care physicians immunosuppressive agents and confirmed immu-
readily adopted the Harvard committee’s recom- nologists’ predictions that immune reactivity in
mendations from the outset, this was without this setting would be minimal. Success in twins
legal support. As a result, brain death was not was replicated in other centers, and proved beyond
legally recognized in the United States or United doubt that technical problems had been mastered
Kingdom until the early 1980s [1]. and that a transplanted kidney could sustain good
health for decades. Murray became a leading fig-
ure in the experimental and clinical development
 reakthroughs in Kidney
B of renal transplantation, and was awarded the
Transplantation Nobel Prize in 1990. His work established that
clinical transplantation could be successful, and
Through most of the first half of the twentieth encouraged others to explore the technical chal-
century a tension existed between a dominant tra- lenges presented by the liver and other organs.
dition of holistic bedside medicine and an emerg-
ing culture favoring laboratory assays, technology
and experimentation. Change began to accelerate Overview of Liver Transplantation
in the early years after World War II, exemplified
by the management of renal failure. Several sur- C. Stuart Welch (1909–1980) performed the first
gical teams in France and North America under- experimental liver transplant in 1955, placing a
took experimental kidney transplantation in canine liver graft in the abdomen heterotopically
humans in the early 1950s, including one mother-­ (without removal of the native organ) [15]. The
94 J. R. Klinck and E. A. Pretto

liver was found to be less vulnerable to rejection dures, and demonstrated much better outcomes
than the kidney, but without portal inflow it rap- compared with matched controls with end-stage
idly atrophied and could not be used to study liver disease (ESLD) who were given conven-
rejection. This problem, combined with a belief at tional treatment. In recipients receiving cyclo-
the time that the liver mediated rejection and sporine 1-year survival was 60%, versus 25–35%
therefore a grafted organ might be tolerated if the in the pre-cyclosporine era. Organ donation legis-
native liver was removed, soon prompted the lation, using the Harvard Criteria to define brain
development of the orthotopic procedure. death, and other important advances in liver pro-
Rejection still occurred, but the orthotopic tech- curement and preservation facilitated the use of
nique created an excellent model for experimental liver grafts from brain dead donors, further con-
immunosuppression and a method of implanta- tributing to this success.
tion that remains the standard today. With confi- From 1983 to the 1990s, a positive cycle was
dence in the surgical technique and useful created that produced rapid growth in liver trans-
experimental data on azathioprine and steroid- plant procedures with long-term survival, Better
based immunosuppression, Thomas Starzl per- results brought more referrals, and more experi-
formed the first human liver transplant in Denver ence yielded even better results. Specialists in a
Colorado in 1963 on a 3-year old child with bili- range of supporting disciplines were attracted to
ary atresia [16]. Four more liver transplants were the challenges presented by transplant patients
attempted soon afterward but all died within 23 and brought wider expertise to liver transplant
days, most from primary ischemic injury to the teams, further enhancing care. Today, according
graft but one intraoperatively due to hemorrhage. to the World Health Organization (WHO), more
A self-imposed moratorium followed while Starzl than 25,000 patients receive liver transplants
considered technical refinements. each year. One-year survival is >90%, while 5-
Calne established a pig model in Cambridge and 10-year survival and quality of life for the
in 1965 and began clinical liver transplantation in majority of recipients are excellent. Advances in
1968 [17]. Starzl resumed clinical transplants in liver transplantation have also facilitated the
Denver and both continued experimental work on development of intestinal and multi-visceral
surgical technique, preservation and immunosup- transplantation.
pression. Despite a public and academic climate
unfavorable to clinical transplantation following
the disastrous ‘Year of the Heart’, they perse-  volution of Surgical Technique:
E
vered with human liver transplants during the late Caval Replacement Versus
1960s and throughout the 1970s, making incre- Piggyback, Use of Venovenous
mental progress. However, survival at 1 year Bypass
remained less than 25%, and it was not until the
discovery of cyclosporine and its introduction Although both main techniques of whole-liver
into clinical practice in the late 1970s that rejec- grafting, the classical caval replacement and
tion could be controlled. This provided the break- caval preservation (piggyback) techniques, date
through needed to move liver transplantation and from the first clinical descriptions in the 1960s,
the entire field of organ replacement into main- the relative simplicity and greater laboratory
stream medical care. experience of caval replacement led to its rapid
The National Institutes of Health (NIH) adoption as the standard method. While in animal
Consensus Conference on Liver Transplantation models full caval and portal clamping caused
in 1983 signaled recognition of the operation as fatal splanchnic stasis and hypotension unless an
worthy of broader introduction [18]. At that time extracorporeal portosystemic shunt was used, it
four pioneering liver transplant centers (Denver, was tolerated in humans without shunting, fur-
Cambridge, Hannover and Groningen) presented ther reducing the incentive to apply the more
results of 540 orthotopic liver transplant proce- demanding piggyback technique.
7  History of Liver Transplantation 95

However, most of the early recipients were chil- However, the routine use of venovenous
dren or relatively fit adults with tumors, and with bypass in adult recipients has declined progres-
more experience it became clear that some recipi- sively over the past 15 years, for several reasons.
ents tolerated clamping poorly. Moreover, the dete- First, many long-established programs have used
riorating state of the patient during the anhepatic it only occasionally, including Cambridge UK,
phase meant that implantation needed to be per- London Ontario, University of Minnesota and
formed quickly, by a very experienced surgeon, University of California San Francisco, and it has
which made teaching difficult. Passive shunts were never been used it routinely in children. A num-
tried but some clotted or caused fatal thromboem- ber of fatalities have been associated with its use,
bolism. In Cambridge, Calne developed a tech- mainly due to perforation of central veins when
nique of venoarterial (femoral vein to femoral large-bore percutaneous access is used, and
artery) pumped perfusion with heparinization and observational studies have not shown any clear
an oxygenator, which was implemented in five benefit. Probably most significant is that the pig-
patients intolerant of a trial clamping of the gyback technique has become more widely prac-
IVC. This was reported to restore arterial blood ticed, providing better hemodynamic stability by
pressure, clearly by increasing and redistributing preserving some caval flow during the implanta-
arterial blood volume rather than supporting tion phase.
venous return. All survived the transplant but four
of the five patients died within a few weeks of sur-
gery. An intra-operative death in Pittsburgh in 1982  volution of Anesthesia
E
partly attributed to severe splanchnic stasis led to a and Perioperative Care
trial of a roller-pump driven venovenous bypass
circuit with systemic heparinization. Although this Early descriptions of anesthesia for clinical liver
was successful in several patients, deaths from transplantation come from J. Antonio Aldrete in
uncontrolled bleeding soon followed. Late in 1982, Denver (1969), and John Farman and Michael
a newly developed centrifugal blood pump, caus- Lindop in Cambridge [17]. Most of the key prob-
ing less turbulence than conventional roller pumps lems were identified, including hemodynamic
and already in use without heparin in patients on instability, hemorrhage, hypocalcemia, hypother-
membrane oxygenators, was tested successfully in mia and acidosis. Changes in cardiac output, vas-
animal transplant models. From January 1983 on cular resistances and pulmonary artery occlusion
this pump was used in human liver recipients and pressure were reported by F.J. Carmichael [20],
with the addition of heparin-bonded tubing, became who placed pulmonary artery catheters in a series
standard care in adult liver transplants in Pittsburgh of patients in Cambridge. Similar observations
for the next 20 years [16, 19]. were reported by Marquez and colleagues in
The adoption of venovenous bypass was wide- Pittsburgh [21]. Transient but occasionally severe
spread thereafter, due to the pre-eminence of reperfusion hyperkalemia was also described and
Pittsburgh as a training center for liver transplan- this remains a frequent cause of intraoperative
tation and the facilitating role of bypass in the cardiac arrest and death to this day. Use of the
surgical teaching of liver implantation. In pulmonary artery catheter in the critical care set-
Cambridge, although the venoarterial technique ting declined sharply after a randomized trial in
was abandoned after 1983, use of venovenous 2005 demonstrated no benefit. However it is still
bypass as developed by Shaw and Starzl remained widely used in cardiac surgery and liver trans-
occasional rather than routine. A percutaneous plantation, where the diagnosis and management
technique for outflow and/or return was devel- of pulmonary hypertension and reliable measure-
oped independently in several centers in the mid-­ ment of cardiac index still provide compelling
1980s and is still used, reducing the incidence of reasons for its use [22, 23]. Rapid point-of-care
wound infection and lymphocele associated with measurement of blood gases, available only from
surgical cut-downs for cannula positioning. the late 1970s, was gradually extended to include
96 J. R. Klinck and E. A. Pretto

sodium, potassium, ionized calcium, hemoglobin both therapeutically and prophylactically, in


and lactate over the next 20 years and has been a many transplant programs. Although the pro-
standard of care for many years. thrombotic state found in many liver recipients
Anesthetic agents used in the earliest descrip- has caused concern, the risks associated with its
tions included fluoroxene, trichloroethylene and use are still confined to anecdotes, while large tri-
nitrous oxide. Halothane was widely used in the als in trauma, orthopedics and other settings sug-
1970s but avoided in liver surgery because of rare gest an acceptable safety profile.
but severe hepatotoxicity. Enflurane (from 1975), Further early improvements in anesthesia care
isoflurane (from 1982 and still widely used) and included adequate fluid warming, warm water
later desflurane and sevoflurane became the mattresses and forced-air warming used starting
agents of choice, influenced by the work of in the mid-1980s. Commercial cell salvage sys-
Gelman and others on the effects of anesthetic tems were developed in the early 1980s, coincid-
agents on splanchnic blood flow [24]. High-dose ing with the rapid growth of cardiac and major
fentanyl (50–100 μg/kg) as a sole anesthetic vascular surgery as well as liver transplantation.
agent, then popular in cardiac surgery but associ- Concerns about the safety of donated blood,
ated with reports of awareness, was used in some given the epidemic of HIV at that time, and the
centers in the 1980s, but was replaced by the rising costs of transfusion were major stimuli to
ultra-short-acting opiate remifentanil plus desflu- the introduction of this technology. Rapid infu-
rane from the late 1990s in some centers. sion systems, such as that developed by John
Changes in coagulation, and the use of coagu- J. Sassano in Pittsburgh in 1982 using a fluid res-
lation tests including factor assays and serial ervoir, mechanical pump, countercurrent fluid
thromboelastograms, were well described by warming and air detector, became commercially
Carl Gustav Groth [25]. He reported both hyper- available in the mid-1980s and are now used in
fibrinolysis and unexpected venous thrombosis most liver transplant centers.
and pulmonary emboli, along with treatments
including epsilon-aminocaproic acid, fibrinogen,
heparin and protamine. He also observed that a  ast-Tracking and Early
F
functioning graft was critical to normalization of Postoperative Care
clotting. The use of fresh whole blood was
described by Aldrete [26], and also advocated by Early reports of clinical liver transplantation
Farman. Yoo Goo Kang reported in detail on the describe elective postoperative ventilation for up
use of thromboelastography in the diagnosis and to 24 h [17, 26]. The rapid growth in surgical and
management of hyperfibrinolysis in liver recipi- anesthetic experience through the 1980s and
ents in 1985, establishing it as a valuable point-­ 1990s, the introduction of shorter-acting anes-
of-­care modality [27]. It is now widely used and thetic agents, muscle relaxants and analgesics,
refinements continue to be developed. and better prevention of hypothermia and bleed-
The use of targeted antifibrinolytic therapy as ing led to efforts to wean patients from mechani-
demonstrated by Kang was extended to prophy- cal ventilation earlier. Improved patient selection,
lactic use in many liver transplant units following cost considerations and limited availability of
the publication of a randomized trial of aprotinin critical care beds also contributed to the develop-
in cardiac surgery by Royston in 1987. Significant ment of “fast tracking”. Several centers reported
reduction in blood loss during liver transplants safe extubation of selected patients in the operat-
was later demonstrated in double-blind, random- ing room from the mid-1990s on, and a multi-
ized trials of tranexamic acid and aprotinin. center trial published in 2007 demonstrated its
However, aprotinin was removed from the mar- cost-effectiveness [28]. Fast-tracking, or extuba-
ket in 2008 when studies in cardiac surgery sug- tion in the operating room with subsequent
gested an increased risk of multi-organ failure admission to a high-dependency area, is now well
and death. Tranexamic acid continues to be used, established, although in most units a policy of
7  History of Liver Transplantation 97

ICU admission and extubation within a few hours oping rapidly. These include in situ machine
is common. Early extubation after liver transplant perfusion at retrieval, normothermic and hypo-
depends on good graft function, minimal co-­ thermic perfusion throughout storage, and ex-situ
morbidity and low operative blood loss. normothermic perfusion after cold storage. Some
The use of epidural analgesia was described in of these also allow pre-transplant evaluation of
the early Cambridge series by Lindop [17], graft function as manifest by lactate and enzyme
although it was stopped owing to concerns about levels, bile production and bile pH. This has
the perioperative evolution of coagulation. increased the use of marginal livers that may oth-
Although this and other series have been erwise have been discarded. Although outcomes,
described, including one from King’s College especially in terms of biliary complications,
Hospital in London of over 140 patients, the remain poorer than those seen in standard cadav-
rapid onset of coagulopathy from poor graft func- eric donors, research into improved preservation
tion cannot be predicted and careful assessment techniques has recently gained unprecedented
of risk-benefit has been advocated. momentum.
Living donor liver transplantation (LDLT) has
also developed to meet this need and especially
Trends in Liver Disease, Donation allows treatment of patients in countries where
and Organ Allocation the use of heart-beating donors is outside cultural
or legal norms. LDLT programs have grown rap-
Over the past 30 years, the success of liver trans- idly since the first successful adult-to-child living
plantation has led to a huge increase in referrals donor procedure by Strong and Lynch in Brisbane
for treatment. Epidemics of hepatitis C, alcohol-­ in 1989 [29]. Living donation peaked in the
related disease, non-alcoholic fatty liver disease United States in 2001 at over 500 transplants and
(NAFLD) and hepatocellular carcinoma in aging decreased since then in North America and
populations have compounded this effect. Europe due to donor deaths. Nonetheless, it is the
However, the supply of suitable cadaveric, heart-­ main source of organs in Japan, Korea, Hong
beating donors has been declining since the early Kong, Taiwan, Turkey, India and the Middle East.
1990s, and falls far short of demand. The reasons Recipient survival is now as good as that obtained
for this included an aging population with a high in cadaveric donation but significant donor mor-
prevalence of subclinical liver disease, and bidity and mortality remain a major concern.
improvements in traffic safety and critical care. Innovation to enhance donor safety continues,
New treatments for Hepatitis C and earlier diag- recently reflected in the increasing use of fully
nosis and prevention of NAFLD should reduce laparoscopic techniques for both left and right
the requirement for transplant in these groups in donor hepatectomy.
the long term, but will not benefit the current The management of waiting lists and organ
population with end-stage disease. allocation has evolved significantly in the past
The recent marked increases in the number of 30 years. The choice of recipients from among
potential recipients and their waiting list mortal- size and blood group-matched peers was typi-
ity has stimulated the development of alternative cally carried out by transplant center physicians,
sources of organs for transplant. Technical inno- based on geography, subjective judgments of
vations such as split-liver donation to two recipi- need or assumed benefit, poorly validated prog-
ents have helped, but few donor livers are suitable nostic scoring, or even length of time on the
for this. Livers from marginal donors are increas- waiting list. A move to a “sickest first” model
ingly used, and in some regions donation after based on MELD (Model for End-stage Liver
cardiac death (DCD) protocols have increased Disease) was implemented in the United States
donation rates as much as 30%. Procurement and in 2002, and has now been adopted in varying
perfusion techniques that allow better preserva- forms in most other countries. The MELD score
tion of DCD and marginal grafts are also devel- is derived from three simple laboratory assays
98 J. R. Klinck and E. A. Pretto

(International Normalized Ratio of the pro- National and international academic societies
thrombin time, creatinine and bilirubin) and was contribute enormously to progress in the field by
developed at the Mayo Clinic to predict survival supporting education, mentorship and research,
in end-stage liver patients after transjugular and by advising on standards. These include the
intrahepatic portosystemic shunting (TIPS). It is following:
able to predict transplant waiting list mortality
and improve overall survival when used to pri- • International Liver Transplantation Society
oritize listed patients, although exception rules (ILTS)
are needed for conditions such as hepatocellular • The Transplantation Society (TTS)
carcinoma. This allocation system has been crit- • American Association for the Study of Liver
icized, however, since it may not maximize Diseases (AASLD)
“transplant benefit”, or life-­years gained after • European Association for the Study of the
transplantation. Liver (EASL)
• American Society of Transplantation (AST)
• European Society of Organ Transplantation
 orldwide Growth, Regulation
W (ESOT)
and Academic Organizations • American Society of Transplant Surgeons
(ASTS)
The number of liver transplant programs in North • Liver Intensive Care Group of Europe
America and Europe increased rapidly after NIH (LICAGE)
endorsement in 1983, slowing only in the mid- • Society for the Advancement of Transplant
1990s when the donor supply reached a plateau. Anesthesia (SATA)
From about 2000 on, economic development ini- • European Liver and Intestinal Transplant
tiated a second phase of rapid expansion, mainly Association (ELITA)
in China, Eurasia, the Middle East, India and
South America. Living donation has accounted Conclusion
for much of this growth. Established in Japan, The history of liver transplantation began
Korea and China since the mid-1990s, living with the inspired efforts of a few dedicated
donor programs have grown rapidly in Turkey, physicians and scientists who grasped the
Egypt and India in the past 15 years and contin- potential of the new science of immunology.
ued expansion is likely. There are now more than They built multidisciplinary teams and over-
500 liver transplant centers in 81 countries across came what their peers considered insur-
the world [30]. mountable biological, technical and societal
Organizations to promote and co-ordinate obstacles. Their commitment to continuous
organ procurement and distribution and to mon- refinement of their experimental techniques
itor and maintain standards in liver transplanta- and to integrating key advances in other
tion have been created in all countries in which fields was finally rewarded by a breakthrough
national legislation addressing transplantation in immunosuppression in 1980, when
has been passed. The best-known is the United decades of groundwork came to fruition.
Network for Organ Sharing (UNOS), which Colleagues in anesthesia and critical care
funds the Scientific Registry of Transplant played a vital role in this story, and today’s
Recipients (SRTR) in the United States. clinicians have much to gain from knowledge
There are comparable bodies in European, of how this singular goal was achieved. It
Australasian, Asian and South American coun- highlights the value of empirical research
tries, although data quality, transparency of and multidisciplinary collaboration, and
outcomes and overall effectiveness is reported strengthens mutual respect in a complex
to vary between organizations. endeavor that saves the lives of tens of thou-
sands of patients each year.
7  History of Liver Transplantation 99

References 15. Welch CS. A note on transplantation of the whole


liver in dogs. Transplant Bull. 1955;2:54–5.
16. Starzl TE, Demetris AJ. Liver transplantation: a

1. Hamilton D. A history of organ transplantation. 31-year perspective. Chicago: Year Book Medical
Pittsburgh: University of Pittsburgh Press; 2012. Publishers; 1990.
p. 105–25. 17.
Calne RY, editor. Liver transplantation: the
2. Matevossian E, Kern H, Hüser N, Doll D, Snopok Y, Cambridge-King’s college hospital experience.
Nährig J, Altomonte J, Sinicina I, Friess H, Thorban London: Grune and Stratton; 1983.
S. Surgeon Yurii Voronoy (1895–1961) – a pioneer in 18. National Institutes of Health consensus development
the history of clinical transplantation: in memoriam at conference statement: liver transplantation – June
the 75th anniversary of the first human kidney trans- 20–23, 1983. Hepatology. 1984;4:107S–10S.
plantation. Transpl Int. 2009;22(12):1132–9. 19. Shaw BW Jr, Martin DJ, Marquez JM, Kang YG,
3. Hamilton DN, Reid WA. Yuri Voronoy and the Bugbee AC Jr, Iwatsuki S, Griffith BP, Hardesty RL,
first human kidney allograft. Surg Gynecol Obstet. Bahnson HT, Starzl TE. Venous bypass in clinical
1984;159(3):289–94. liver transplantation. Ann Surg. 1984;200(4):524–34.
4. Bradley JA, Hamilton DN. Organ transplantation: an 20. Carmichael FJ, Lindop MJ, Farman JV. Anaesthesia
historical perspective. In: Hakim NS, Danovitch GM, for hepatic transplantation: cardiovascular and met-
editors. Transplantation surgery. London: Springer abolic alterations and their management. Anesth
Verlag; 2001. Analg. 1985;64:108–16.
5. Le Gallois, M (Julien Jean Cesar), 1770–1814; trans- 21. Marquez J, Martin D, Virji MA, Kang YG, Warty
lated by N.C. and J.G. Nancrede. Experiments on the VS, Shaw B Jr, Sassano JJ, Waterman P, Winter PM,
principle of life, and particularly on the principle of Pinsky MR. Cardiovascular depression secondary to
the motions of the heart, and on the seat of this prin- ionic hypocalcemia during hepatic transplantation in
ciple: including the report made to the first class of the humans. Anesthesiology. 1986;65(5):457–61.
institute, upon the experiments relative to the motions 22. Krowka MJ, Mandell MS, et al. Hepatopulmonary
of the heart. Book, Microform in English; 1813. syndrome and portopulmonary hypertension: a report
6. Miller DJ. Sydney ringer; physiological saline, cal- of the multicenter liver transplant database. Liver
cium and the contraction of the heart. J Physiol. Transpl. 2004;10(2):174–82.
2004;555(Pt 3):585–7. 23. Walia A, Schumann R. The evolution of liver

7. Toledo-Pereyra LH. A study of the historical ori- transplant practices. Curr Opin Organ Transplant.
gins of cardioplegia. Dissertation/Thesis in English; 2008;13(3):275–83.
1984. 24. Gelman S, Dillard E, Bradley EL Jr. Hepatic cir-
8. Malinin TI. Remembering Alexis Carrel and Charles culation during surgical stress and anesthesia with
Lindbergh. Heart Inst J. 1996;23(1):28–35. halothane, isoflurane, or fentanyl. Anesth Analg.
9. Murray JE, Lang S, Miller BJ, et al. Prolonged func- 1987;66(10):936–43.
tional survival of renal autografts in the dog. Surg 25. Groth CG. Changes in coagulation. In: Starzl TE,
Gynecol Obstet. 1956;103:15. Putnam CW, editors. Experience in hepatic transplan-
10. Belzer FO, Boston MD, Sterry Ashby B, Englebert tation. Philadelphia: W B Saunders; 1969. p. 159–75.
Dunphy J. 24-hour and 72-hour preservation of canine 26. Aldrete JA. Anesthesia and intraoperative care. In:
kidneys. Lancet. 1967;290(7515):536–9. Starzl TE, Putnam CW, editors. Experience in hepatic
11. Toledo-Pereyra LH, Simmons RL, Najarian JS,
transplantation. Philadelphia: W B Saunders; 1969.
Condie RM. Effective use of silica gel fraction for p. 83–111.
kidney preservation after 2-yr room storage. Surg 27. Kang YG, Martin DJ, Marquez J, Lewis JH,

Forum. 1976;27(62):311–2. Bontempo FA, Shaw BW Jr, Starzl TE, Winter
12.
Collins GM, Bravo-Shugarman M, Terasaki PM. Intraoperative changes in blood coagulation and
PI. Kidney preservation for transportation. Initial thrombelastographic monitoring in liver transplanta-
perfusion and 30 hours’ ice storage. Lancet. tion. Anesth Analg. 1985;64(9):888–96.
1969;2(7632):1219–22. 28. Mandell MS, Stoner TJ, Barnett R, Shaked A,

13. Belzer FO, D’Alessandro AM, Hoffmann RM,
Bellamy M, Biancofiore G, Niemann C, Walia A,
Knechtle SJ, Reed A, Pirsch JD, Kalayoglu M, Vater Y, Tran ZV, Kam I. A multicenter evaluation
Sollinger HW. The use of UW solution in clini- of safety of early extubation in liver transplant recipi-
cal transplantation: a 4-year experience. Ann Surg. ents. Liver Transpl. 2007;13(11):1557–63.
1992;215(6):579–85. 29. Garcea G, Nabi H, Maddern GJ. Russell strong and
14. Gubernatis G, Pichlmayr R, Lamesch P, Grosse
the history of reduced-size liver transplantation.
H, Bornscheue A, Meyer HJ, Ringe B, Farle M, World J Surg. 2009;33(8):1575–80.
Bretschneider HJ. HTK-solution (Bretschneider) for 30. Busuttil RW. International liver transplantation society
human liver transplantation. Langenbecks Arch Chir. 2009 presidential address: the internationalization of
1990;375(2):66–70. liver transplantation. Liver Transpl. 2010;16:558–66.
Recipient and Donor Selection
and Transplant Logistics: 8
The European Perspective

Gabriela A. Berlakovich and Gerd R. Silberhumer

Keywords Recipient Prioritizing


MELD score · Child score · Organ allocation ·
Graft procurement · Donor selection · In most transplant centers all over the world liver
Donation after cardiac death allocation is based on MELD score [1], which
predicts wait list survival for the upcoming 3
months. For some underlying diseases the sever-
Introduction ity of chronic liver failure is not reflected by labo-
ratory MELD (lab MELD) score, such as
The intermediate and long-term outcomes fol- hepatocellular carcinoma in mild cirrhosis and
lowing orthotopic liver transplantation (OLT) metabolic and biliary diseases among others.
have improved significantly over the years, Therefore, standard exceptions from laboratory
with 1- and 5-year patient survival rates of 90% MELD allocation were defined by Eurotransplant
and 75%, respectively. This success resulted in that usually result in 22 MELD points, equivalent
growing numbers of potential transplant recipi- to a 15% 3-month mortality. Standard exception
ents on waiting lists. The number of transplant- (SE) can be requested for patients at any time
able organs remained stable over the last after registration in the Eurotransplant area but
decades and the increased demand could not be recipients must fulfill country- and disease-­
met. In recent years this discrepancy between specific criteria before the exceptional MELD
patients listed for liver transplantation and (match MELD) can be approved. If the excep-
available organs decreased as less candidates tional MELD was approved, this status is granted
were entered on the waiting lists due to changes for the duration of 90 days and the SE status must
in listing policies (Fig. 8.1). Nevertheless wait be reconfirmed before the expiration of this
list mortality remains a major problem regard- 90-day.
less of various organ allocation policies adopted Eurotransplant introduced MELD allocation
by transplant programs. This chapter will in 2006, but, typically for the heterogeneity in
describe the current situation in Europe with Europe, modalities are somewhat different
special emphasis on efforts to increase the between the countries: Germany, Belgium,
availability of liver grafts. Luxembourg, Hungary and the Netherlands use
a patient-based allocation system in which each
G. A. Berlakovich, MD, FEBS (*) patient is matched with a donor according to
G. R. Silberhumer, MD match MELD. However Austria, Croatia, and
Division of Transplantation, Department of Surgery, Slovenia use a center-oriented allocation system
Medical University of Vienna, Vienna, Austria
e-mail: gabriela.berlakovich@meduniwien.ac.at in which the center is allocated a graft and can
© Springer International Publishing AG, part of Springer Nature 2018 101
G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_8
102 G. A. Berlakovich and G. R. Silberhumer

3000

2500

2000

1500

1000

500

0
1991 1992 1993 1994 1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 2005 2006 2007 2008 2009 2010 2011 2012 2013 2014
Liver waiting list 229 253 203 212 263 327 374 492 593 803 1093 1366 1714 2035 2134 2319 2429 2442 2641 2695 2614 2406 2111 1918
Deceased donor transplants 710 765 878 892 944 1032 1097 1071 1132 1168 1112 1136 1264 1262 1364 1436 1625 1606 1692 1793 1770 1689 1562 1646
Living donor transplants* 5 15 14 24 25 22 41 38 64 116 124 129 133 106 121 116 101 82 99 138 135 121 133 112

Fig. 8.1  Dynamics of the Eurotransplant liver waiting list and liver transplants between 1991 and 2009 [10]

choose an appropriate recipient from the wait- available donor organs and assure a transparent
list. The advantages of MELD score based allo- and objective allocation system. Furthermore,
cation are transparency and objectivity however they assess the importance of factors that have
medical urgency is not always adequately the greatest influence on wait list mortality and
reflected by the MELD score and for several dis- transplant results. OPOs also promote, support,
ease patterns standard exceptions have been and coordinate organ donation and transplanta-
defined as above. The impact of MELD score on tion. In Europe many different OPOs exist:
post-transplant survival is still an issue of debate nationally structured agencies for example in
[2, 3] and recently dynamic changes of MELD Spain, France or Italy as well as multinationally
scores during waiting time (Delta-MELD) have structured agencies. Within a multinational OPO
been found to be a risk factor for death after the national legislation is prioritized over interna-
transplantation independent of donor-derived tional interests of the organization, for example,
variables [4]. when it comes to issues such as presumed or
Another significant disadvantage of a strict informed consent for organ donation. The most
patient-oriented allocation system based on important multinational OPOs in Europe are the
MELD is the inability to directly match donor and following:
recipient based on criteria other than MELD
score. For example extended criteria donor (ECD) –– Scandiatransplant [8] is the Scandinavian
organs may have a higher risk for initial dysfunc- organ exchange organization and covers a
tion and fair even worse with prolonged cold isch- population of 25 million people in five coun-
emia time and may therefore not be suitable for tries (Denmark, Finland, Iceland, Norway,
every candidate. Despite a number of models pre- and Sweden). The most frequently exchanged
dicting post-transplant outcome based on donor organs between centers within
and recipient factors [4–7] the final decision for Scandiatransplant are livers followed by
the suitability of an organ is made by the trans- hearts. The overall exchange rate of kidneys
plant team based on clinical judgment. has stabilized around 12% during the last
years. One third of kidney transplants are per-
formed from living donors.
Organ Distribution –– NHS Blood and Transplant [9] combines the
United Kingdom and the Republic of Ireland
The objectives of Organ Procurement with a total population of 70 millions. Donor
Organizations (OPOs) are similar all over the livers are not allocated to patients but center-­
world, their aim is to achieve an optimal use of specific according to the “Donor Organ
8  Recipient and Donor Selection and Transplant Logistics: The European Perspective 103

Sharing Scheme” prepared by the Liver change strategies. The number of available
Advisory Group. Following these general donors remained stable and does not match the
principles donor to recipient matching should demand even if listing criteria were made stricter
be provided, especially for livers derived from [10, 12, 13]. Therefore, several strategies have
donors with extended criteria. been developed to increase the donor pool
–– Eurotransplant [10] is the central European (Fig.  8.2a). The most popular strategies are the
OPO and covers a population of 135 million use of extended criteria donors (ECDs), donation
people in eight countries (Austria, Belgium, after cardiac death (DCD), and living donation
Croatia, Germany, Hungary, Luxembourg, the (LD) (Fig. 8.2b).
Netherlands, Slovenia). The most frequently
exchanged organs between centers are kid-
neys. In the setting of acute liver failure the Extended Criteria Donor
next available appropriate organ within the ET
area is offered to the requesting transplant Several publications demonstrated that donor
center. Liver graft exchange thereafter follows factors such as age, gender, race, graft type, and
a payback system, which means that the recip- cold ischemic time affect post-transplant survival
ient center has to offer the next available donor [5]. Despite the exact definition of risk factors,
liver of equal blood group to the previously their relative risk for post-transplant primary
donating center. Allocation priority is ranked non-function or poor function is weighted differ-
from “high urgency” to “accepted combined entially [6, 14] and there are no generally
transplantation” to “center” to “ET pool.” accepted criteria for ECD organs yet. Age is one
–– The Spanish transplant system [11] is well of the best-described extended donor factors and
known all over the world as one of the most several studies found that a donor age older than
successful systems with an average of 35 55 was a significant factor resulting in poorer
donors per one million inhabitants. The main graft survival [5, 6, 15]. Nevertheless due to
principles of the Spanish Model of Organ changes of donor demographics in the last
Donation are an unrivaled transplant coordi- decades, donor age and age-related comorbidities
nation network. In-house coordinators per- have increased significantly. Donor death from
form a continuous audit of brain deaths and cardiovascular reason is now more common than
outcome after donation at intensive care units trauma [16] and more than 60% of organs are
in transplant procurement hospitals. These procured from donors who died due to cardiovas-
coordinators are specially trained in commu- cular disease.
nication with hospital staff as well as relatives Cold ischemic time is another very well-­
of potential donors. A central organ donation documented donor risk factor and an imprecise
agency has great influence on medical training cutoff between 10 and 13 h has been investigated
and maintains close relationships with the [5, 6, 17]. In the era of MELD-based allocation
media and intensive care units in order to pro- this is a very importat aspect. Increased local
mote organ donation. donor utilization would result in shorter transpor-
tation times and consecutively reduced cold isch-
emic times.
Donor Selection Donor graft quality is one of the main determi-
nants of post-transplant outcome in liver trans-
The disparity between organ demand and availal- plantation. It is difficult to classify the quality of
ity of grafts represents the most limiting factor in organs based solely on laboratory values. Some
liver transplantation. Since outcome of liver consider donor transaminases levels >150 U/L as
transplantation has improved and the number of a risk factor for graft dysfunction [18, 19].
patients listed for liver transplantation steadily Increased donor gamma-glutamyl transpeptidase
increased, transplant centers were forced to has also been identified as risk factor for increased
104 G. A. Berlakovich and G. R. Silberhumer

a
(%)
100

90

80

70

60

50

40

30

20

10

0
68-82
1983
1984
1985
1986
1987
1988
1989
1980
1991
1992
1993
1994
1995
1996
1997
1998
1999
2000
2001
2002
2003
2004
2005
2006
2007
2008
2009
2010
2011
2012
2013
2014
2015
b Cadaveric Full Size: 86928 Other Typw of Graft: 15830
(%)
100

90

80

70

60

50

40

30

20

10

0
1998
68-82
1987
1988

1990
1991
1992
1993
1994
1995
1996
1997

1999
2000
2001
2002
2003
2004
2005
2006
2007
2008
2009
2010
2011
2012
2013
2014
2015
1989

Domino : 965 Livivng Donor : 7678 Non heart beating: 1789 Reduced : 1696 Splt liver : 5455

Fig. 8.2 (a) Cadaveric versus other types of graft in Europe according to the date of transplantation. (b) Alternatives to
the use of full-size cadaveric liver grafts in Europe [13]

3 months graft failure but not 1-year survival [20]. cellular swelling and dysfunction. Totsuka et al.
Biopsy-proven steatosis can contribute to primary [22] reported comparable outcomes between nor-
non-function rates of up to 25% and is highly cor- monatremic and hypernatremic donors after cor-
related with increased donor age and obesity [21]. rection of sodium levels below 155 mEq/
Direct osmolar damage caused by increased mL. However in our experience peak sodium val-
plasma sodium levels is responsible for hepato- ues during the intensive care unit stay were a sig-
8  Recipient and Donor Selection and Transplant Logistics: The European Perspective 105

nificant factor for post-transplant outcome [20]. to overcome the risk for ischemic cholangiopa-
This finding supports the theory that a short dura- thy. Various techniques have been investigated
tion of deviation of plasma sodium levels may including normothermic or hypothermic perfu-
cause long-lasting damage in hepatocytes due to sion [27, 28]. Extracorporeal perfusion may have
changes of intracellular osmolarity even when the ability to “recondition” the damaged liver
sodium levels are rapidly and aggressively graft that has undergone warm ischemic injury
corrected. during DCD procurement [29, 30]. Despite the
risk of biliary strictures patient and graft survival
rates similar to those of DBD grafts can be
Donation After Cardiac Death achieved with controlled DCD donation when
very restrictive criteria are used [31]. The
Donation After Cardiac Death (DCD) is defined American Society of Transplant Surgeons
as procurement of organs shortly after cardiore- (ASTS) recently published practice guidelines
spiratory support has been withdrawn and cardiac [32] that are similar to the selection criteria rec-
death ensued. Most DCD donors are patients who ommended by European centers [26, 31, 33].
suffered from severe irreversible cerebral injury Considering organ shortage and death on the
but are not dead by neurological criteria and the waiting list DCD grafts remain a small but valu-
family or health care proxies wish to withdraw able resource for scarce organs.
support. Minutes after death has occurred, organs
are harvested for transplantation.
The recent increase of DCD donation in some Living Donor Liver Transplantation
European countries has contributed to an increase
in the number of transplants with outcomes com- Unlike for kidney transplantation, there is no
parable to grafts from brain death donors (DBD). clear evidence for a significant advantage in post-­
However DCD donation may not be necessarily a transplant survival after living donation liver
new and additional source of grafts; data from the transplantation (yet?). The overall results with
Netherlands [23] indicate that the use of DCD good patient and graft survival combined with
organs may have caused a shift from potential acceptable donor morbidity and mortality has led
heart-beating donors to DCD donation. Intensive to the acceptance of LDLT in the transplant
care providers may encourage DCD donation community.
rather than awaiting brain death and subsequent Left-lateral LDLT in children has become a
heart-beating donation. This development could standard procedure with excellent results,
be reversed during the last years, resulting in an whereas LDLT in adults has still some con-
effective increase in organ availability [24]. flicting issues. The number of LDLT proce-
For DCD grafts the effect of cold ischemia is dures peaked in 2001 in Europe and the US,
superimposed by the injury occurring during thereafter showing a significant decrease of
warm ischemia. Biliary epithelium is particularly cases in the US and no further increase in
vulnerable to ischemia – reperfusion injury and a Europe. This development was in part due
high incidence of biliary strictures and/or bile higher failure rate of LDLT due to graft-related
cast syndrome [25, 26] has become of concern. issues and substantial donor morbidity [13].
Ischemic cholangiopathy has been reported in Recipients have a higher risk for primary non-
9–50% of DCD recipients. This complication function or dysfunction due to small for size
tends to present within the first few months after syndrome and a significantly higher risk for
OLT and may resolve with biliary drainage, often technical failures, especially biliary and vas-
requiring repeated interventions and may even cular complications. Additionally the mortal-
lead to graft loss and retransplantation. ity risk of approximately 0.2% and morbidity
In the future extracorporeal machine perfu- risk of 11–28% for donors represent non-­
sion of liver grafts may be a potential technology negligible limitations for the use of LDLT
106 G. A. Berlakovich and G. R. Silberhumer

grafts. LDLT accounts for less than 5% of all References


liver transplants in Europe and US [12, 13]. 1. Wiesner RH, McDiarmid SV, Kamath PS, et al.
The number of LDLT in Asia has continued to MELD and PELD: application of survival models to
increase due to the limitations in DBD caused liver allocation. Liver Transpl. 2001;7:567–80.
by legal and cultural restrictions on deceased 2. Desai NM, Mange KC, Crawford MD, et al.
Predicting outcome after liver transplantation: utility
organ donation. Ninety-five percent of all of the model for end-stage liver disease and a newly
OLTs in Asia excluding mainland China are derived discrimination function. Transplantation.
LDLT [34]. 2004;77:99–106.
One of the main advantages of LDLT is the 3. Onaca NN, Levy MF, Sanchez EQ, et al. A correlation
between the pretransplantation MELD score and mor-
precise scheduling of the procedure and therefore tality in the first two years after liver transplantation.
independence of waiting time and available liver Liver Transpl. 2003;9:117–23.
grafts. OLT can take place according to disease 4. Györi GP, Silberhumer GR, Rahmel A, et al. Impact
severity and recipient conditions. Especially for of dynamic changes in MELD score on survival after
liver transplantation – a Eurotransplant registry analy-
patients suffering from hepatocellular carcinoma, sis. Liver Int. 2016;36:1011–7.
LDLT represents a useful treatment option to 5. Burroughs A, Sabin CA, Rolles K, et al. 3-month and
reduce waiting time and consecutive disease 12-month mortality after first liver transplant in adults
progression. in Europe: predictive models for outcome. Lancet.
2006;367:225–32.
A potential survival benefit due to decreased 6. Feng S, Goodrich NP, Bragg-Grasham JL, et al.
death on the waiting list and reduced disease pro- Characteristics associated with liver graft failure:
gression has to be balanced with higher morbid- the concept of a donor risk index. Am J Transplant.
ity and mortality following transplantation. 2006;6:783–90.
7. Silberhumer GR, Pokorny H, Hetz H, et al.
Future application of LDLT will be based on the Combination of extended donor criteria and changes
accurate definition of risks imposed on donors in the model for end-stage liver disease score predict
compared with potential benefits realized by patient survival and primary dysfunction in liver trans-
recipients. New technologies such as laparo- plantation: a retrospective analysis. Transplantation.
2007;83:588–92.
scopic or adult-to-adult left lobe donation may 8. Scandiatransplant. http://www.scandiatransplant.org.
increase the acceptability of LDLT. Accessed June 2016.
9. NHS Blood and Transplant. http://www.organdona-
Conclusion tion.nhs.uk. Accessed June 2016.
10. Eurotransplant International Foundation. http://www.
Currently the progress of liver transplantation eurotransplant.org. Accessed June 2016.
is limited by organ shortage. Several strategies 11. Matesanz R, Dominguez-Gil B, Coll E, de la Rosa
have been developed to overcome this problem G, Marazuela R. Spanish experience as a leading
during the last few decades. Most important for ­country: what kind of measures were taken? Transpl
Int. 2011;24:333–43.
increasing the pool of deceased donors seems 12. Organ procurement and transplantation network.

to be education of the public and physicians in http://optn.transplant.hrsa.gov. Accessed June 2016.
order to increase awareness for organ donation 13. European liver transplant registry. http://www.eltr.

and transplantation. ICU staff must be continu- org. Accessed June 2016.
14. Dawwas MF, David C, Barber KM, et al. Developing
ously contacted and informed about the bene- a liver transplantation donor risk index in a national
fits of transplantation, and guidelines should be registry. Hepatology. 2007;46:235A.
established to support them with donor man- 15. Busuttil RW, Tanaka K. The utility of marginal donors
agement. Due to the current organ shortage a in liver transplantation. Liver Transpl. 2003;9:651–63.
16. Punch JD, Hayes DH, LaPorte FB, et al. Organ dona-
number of patients will be rejected as recipients tion and utilization in the United States, 1996–2005.
although they would derive a significant sur- Am J Transplant. 2007;7:1327–38.
vival and quality of life benefit from this OLT. It 17. Cameron AM, Ghobrial RM, Yersiz H, et al. Optimal
remains a formidable challenge to balance the utilization of donor grafts with extended criteria: a
single-­ center experience in over 1000 liver trans-
demands of individual autonomy of the recipi- plants. Ann Surg. 2006;243:748–53. discussion
ent and the utility of the donor organ on a back- 753–755
ground of justice and equity.
8  Recipient and Donor Selection and Transplant Logistics: The European Perspective 107

18. Mirza DF, Gunson BK, Da Silva RF, et al. Policies 27. Weeder PD, van Rijn R, Porte RJ. Machine perfusion in
in Europe on “marginal quality” donor livers. Lancet. liver transplantation as a tool to prevent non-­anastomotic
1994;344:1480–3. biliary strictures: rationale, current evidence and future
19. Cuende N, Miranda B, Canon JF, et al. Donor
directions. J Hepatol. 2015;63:265–75.
characteristics associated with liver graft survival. 28. Vairetti M, Ferrigno A, Rizzo V, et al. Correlation
Transplantation. 2005;79:1445–52. between the liver temperature employed during
20. Silberhumer GR, de Vries E, Adam R, et al. Are there machine perfusion and reperfusion damage: role of
new definitions of extended liver donor criteria at the Ca2+. Liver Transpl. 2008;14:494–503.
beginning of the 3rd millennium? Experiences from 29. Westerkamp AC, Karimian N, Matton APM, et al.
6133 donors. Liver Transpl. 2010;16:S102. Oxygenated hypothermic machine perfusion after
21. McCormack L, Petrowsky H, Jochum W, et al.
static cold storage improves hepatobiliary function
Use of severely steatotic grafts in liver transplan- of extended criteria donor livers. Transplantation.
tation: a matched case–control study. Ann Surg. 2016;100:825–35.
2007;246:940–6. discussion 946–948 30. Verhoeven CJ, Farid WRR, de Jonge J, et al.

22. Totsuka E, Dodson F, Urakami A, et al. Influence Biomarkers to assess graft quality during conven-
of high donor serum sodium levels on early postop- tional and machine preservation in liver transplanta-
erative graft function in human liver transplantation: tion. J Hepatol. 2014;61:672–84.
effect of correction of donor hypernatremia. Liver 31. Dubbeld J, Hoekstra H, Farid W, et al. Similar

Transpl Surg. 1999;5:421–8. liver transplantation survival with selected cardiac
23. Monbaliu D, Pirenne J, Talbot D. Liver transplan- death donors and brain death donors. Br J Surg.
tation using donation after cardiac death donors. J 2010;97:744–53.
Hepatol. 2012;56:474–85. 32. Reich DJ, Mulligan DC, Abt PL, et al. ASTS

24. Domínguez-Gil B, Haase-Kromwijk B, Van Leiden H, Recommended Practrice Guidelines for controlled
et al. Current situation of donation after circulatory death donation after cardiac death organ procurement and
in European countries. Transpl Int. 2011;24:676–86. transplantation. Am J Transplant. 2009;9:2004–11.
25. de Vera ME, Lopez-Solis R, Dvorchik I, et al. Liver 33. Andrews PA, Burnapp L, Manas D. Summary of the
transplantation using donation after cardiac death British Transplantation Society guidelines for trans-
donors: long-term follow-up from a single center. Am plantation from donors after deceased circulatory
J Transplant. 2009;9:773–81. death. Transplantation. 2014;97:265–70.
26. Detry O, Donckier V, Lucidi V, et al. Liver trans- 34. Lee SG. Asian contribution to living donor liver trans-
plantation from donation after cardiac death donors: plantation. J Gastroenterol Hepatol. 2006;21:572–4.
initial Belgian experience 2003–2007. Transpl Int.
2010;23:611–8.
Recipient and Donor Selection
and Transplant Logistics: The US 9
Perspective

Ingo Klein, Johanna Wagner,
and Claus U. Niemann

Keywords distribution, resulting in significant geographi-


MELD score · Child score · Share 35 · Waiting cal discrepancies in the US with regard to wait-
time · Liver allocation · Donor management ing time and severity of disease at the time of
transplant. Changes of the current allocation
and distribution policies are evaluated but have
Introduction to balance increased cold ischemia times and
cost efficiency with equity. While the number
Organ allocation for donor livers in the US fol- of ‘standard criteria’ deceased organ donors
lows an algorithm of medical urgency deter- has recently increased, there was also an
mined by 3-month mortality risk based on increase of primarily older donors, donors with
objective laboratory values that are included in co-­morbidities, and donations after cardiac
the MELD score. However, in a significant pro- death. These so called expanded criteria donors
portion of waitlist patients (patients with HCC together with the use of donors with steatotic
and non-cirrhotic patients) the 3-month mortal- livers have broadened the donor pool, but bear
ity risk may not be reflected by standard labora- an increased risk of organ dysfunction to the
tory tests accurately. Therefore, so called liver recipient. Currently clinical outcome
exceptional MELD points based on statistical research focuses on which patient population
mortality risk can be granted upon standard or will benefit the most from which type of
non-standard request. Additionally, based on expanded criteria organ. In the United States,
urgency, the prioritization of organ recipients is living donor liver transplantation has not
embedded in a framework of local and regional evolved into a significant proportion of the total
number of liver transplantations and remains an
option predominantly for pediatric recipients
I. Klein • J. Wagner
Department of General- and Visceral-, Vascular and and recipients who are not prioritized in the cur-
Pediatric Surgery, University of Wuerzburg, Medical rent allocation system.
Center, Wuerzburg, Germany Liver transplantation is the therapy of choice
C. U. Niemann, MD (*) for acute liver failure and many forms of chronic
Anesthesia and Perioperative Care, Department of liver disease including hepatocellular carcinoma
Surgery, University of California San Francisco,
in cirrhotic patients. Significant advances in sur-
San Francisco, CA, USA
e-mail: niemannc@anesthesia.ucsf.edu, gical technique as well as the perioperative
Claus.Niemann@ucsf.edu management have decreased the perioperative

© Springer International Publishing AG, part of Springer Nature 2018 109


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_9
110 I. Klein et al.

Fig. 9.1  Number of Number of liver transplants in the United States


liver transplants in the 9000
United States from 1990 8000
to 2016. DDLT deceased
donor liver transplant; 7000
LDLT living donor liver
6000
transplant
5000 Total

4000 DDLT

3000 LDLT

2000

1000
0
1990 1995 2000 2005 2010 2015

(3-month) mortality to about 5% in the US and removed from the waiting list for example sec-
Canada. This caused a wide acceptance of the ondary to tumor progression (1,473 in 2015)
procedure and many liver transplant programs [1]. This number has continued to increase for
have emerged throughout the country. As a the last several years, also shown in mortality
result, the number of liver transplants in North rates, that have increased each year since 2009,
America has increased more than threefold from from 11.1 per 100 waitlist years 12.3 per 100
about 2,000 transplants per year in 1989 to waitlist years in 2014. Liver cirrhosis secondary
8,082 liver transplants in 2017 [1]. Since 2004, to Hepatitis C virus infection has remained the
the number of transplanted livers has remained main indication for liver transplantation in the
stable above 6,000 transplants per year with a United States with 1588 patients (25.6%) in
slight increase, among other reasons, since the 2014. The number of transplant recipients with
implementation of the Share 35 rule in 2013 malignant neoplasms has increased from 100
(Fig. 9.1). In addition to liver transplantation for cases in 1999 to 1,365 cases (19.2%) in 2015,
established indications, new developments in which is the largest increase for any indication
the (pre- and post-­transplant) medical manage- over the past decade. The absolute numbers for
ment of the underlying liver diseases and the cholestatic liver disease/cirrhosis and acute
careful evaluation of preexisting conditions and hepatocellular necrosis have remained relatively
co-morbidities (i.e. coronary artery disease, stable since 1999 with a slight decrease since
HIV, and others) of liver transplant candidates 2014; however, the relative percentage of these
have broadened the indications for liver trans- two indications has decreased with the overall
plantation even further. This resulted in a tre- increase in transplant numbers (Table 9.1). It is
mendous discrepancy of patients listed for liver expected that non-alcoholic steatotic hepatitis
transplantation and available organs. In 2015, (NASH) will become one of the most frequent
more than 14,000 patients were actively listed indications for liver transplantation in the
on the United Network for Organ Sharing United States within the next decade.
(UNOS) waiting list for liver transplantation While the absolute number of deceased liver
compared to 7,127 liver transplants performed donors in the United States has increased from
in 2015. The number of patients who died wait- 1,833 in 1988 to 7416 donors in 2015, the pro-
ing for a liver transplant remained relatively portion of young donors aged 11–34 years has
stable over the last several years (1,423 in 2015), decreased by half from 61% in 1988 to 37.6%
however, this number does not account for the in 2015. Especially the proportion of older
substantial number of patients who were donors has increased substantially following
9  Recipient and Donor Selection and Transplant Logistics: The US Perspective 111

critical evaluation of older donors for certain  iver Allocation in the US:


L
transplant indications [2, 3]. In 2015, donors 50 From Waiting Time to Medical
years and older comprised more than one-third Criteria
(32.3%) of the entire deceased donor popula-
tion (Fig. 9.2). During the same period, the pro- The allocation of deceased donor livers in the
portion of donors dying from anoxia has almost United States before 1996 prioritized the patient’s
tripled from about 10% in 1988 to 36.2% in level of care with the first priority given to
2015, while the absolute number of younger patients continuously requiring treatment in
donors aged 11–34 years has increased recently, Intensive Care Units for medical complications
the proportion of head trauma decreased from such as exacerbation of hepatic encephalopathy,
43 to 31.0%. variceal bleeding not manageable by endoscopic
therapy or hepatorenal syndrome. The second
priority was given to patients who required con-
tinuous hospitalization and the third priority was
Table 9.1  Evolution of indications for liver transplanta- given to patients with compensated end stage
tion over time (1999–2015)
liver disease who were managed on an outpatient
1999 2008 2015 basis. With an increasing number of patients
Non-Cholestatic 64.4% 55.9% 67.5% awaiting liver transplantation, waiting time was
cirrhosis (2895) (3391) (4567)
used to prioritize within these groups. Since
Cholestatic liver 11.1% 7.8% 7.9%
disease/cirrhosis (498) (475) (537) ranking into one of the three categories was pri-
Acute hepatic 9% 5.3% 3.6% marily based on center-specific criteria and sub-
necrosis (405) (324) (246) jective interpretation of the patients’ condition,
Biliary atresia 4.2% 3% 2.6% the Child-Turcotte-Pugh scoring system was
(188) (180) (177) introduced in 1996 to categorize patients for
Metabolic diseases 3.3% 3% 3.5% chronic liver failure in the spirit of the existing
(150) (180) (234)
categories: Status 2a with a CTP score ≥ 10 or
Malignant 2.2% 17.5% 15.5%
neoplasms (97) (1061) (1052)
admission to the ICU; status 2b with a CTP ≥ 10
Other 5.9% 7.5 7.4% or CTP ≥ 7 in conjunction with at least one major
(265) (458) (498) complication of portal hypertension or stage 1
hepatocellular carcinoma. Status 3 was attributed
to patients with CTP ≤ 7. The highest priority
(status 1) was reserved for patients with fulmi-
nant hepatic failure, primary graft non-function,

[%]
Proportion of liver donor populations by age over time
60

50
Age group
40 0-10
11-34
30
35-49

20 50-65
>65
Fig. 9.2  Proportion of 10
donor population by age
over time from 1990 to 0
2016 1990 1995 2000 2005 2010 2015
112 I. Klein et al.

hepatic artery thrombosis within 7days after capped at a maximum of 40 points, ranging from
transplantation and decompensated Wilson’s 6 to 40. The serum creatinine value is capped at
Disease (which remained essentially unchanged 4 mg/dL and is set to its maximum if the patient
in the present allocation system). This allocation underwent hemodialysis twice or had continuous
system created three large categories for chronic renal replacement therapy for more than 24 h
liver patients and the amount of waiting time on within the last 7days. Any value less than one for
the list was used to prioritize liver allocation creatinine, bilirubin or INR was fixed at one to
within these groups. Subjectivity in grading prevent negative scores.
hepatic encephalopathy and the amount of asci- For pediatric use, creatinine was removed
tes, important components of the score, posed since it was not found to predict short-term mor-
another inherent problem of the Child-Turcotte-­ tality. Serum albumin, growth failure (yes/no),
Pugh score prioritisation. Furthermore, patients and age (<1 year/>1 year) proved to be important
were listed long before their actual need for liver prognostic factors in infants and children and
transplantation in order to be at the top of the were included in the pediatric end-stage liver dis-
waiting list by the time they finally needed a liver ease model (PELD) [9] (Table 9.2).
transplant. This allocation system resulted in a The MELD score system was validated to
dramatic increase of patients listed for transplan- accurately estimate disease severity and predict
tation and on the other hand a large number of 3-month survival of patients with chronic liver
patients who died waiting for a liver transplant disease at the time of listing and was therefore
because they were not listed early enough to considered suitable to allocate liver grafts on the
accumulate enough waiting time. Evaluation of basis of disease severity and medical urgency
the allocation system revealed that apart from its [10]. However, the overall prognosis of several
subjective components, time spent on the waiting patient populations was not well characterized by
list was not associated with an increased death the mortality risk of intrinsic liver disease [11]. In
rate (higher mortality risk) [4] and did not reflect liver transplant candidates, serum sodium is asso-
any medical need for liver transplantation [5, 6]. ciated with mortality independent of the MELD
In 1998 the Department of Health and Human score, particularly for those with low sodium lev-
Services’ Final Rule determined that objective els. In the range of 125–140 mmol/L serum
medical criteria should determine the priority for sodium there is a mortality increase by 5% for
liver allocation [7]. A report of the Institute of each millimole decrease. This resulted in a modi-
Medicine [8] recommended that short term mor- fication of the original MELD-score, the
tality risk would be a more appropriate measure “Sodium-MELD score” [12]. For candidates
to prioritize liver transplant candidates and the with an initial MELD score greater than 11, the
model for end stage liver disease (MELD score) score is then re-­calculated as follows:
was chosen to rank chronic end-stage liver dis- Table 9.2  Formulas for MELD, Na-MELD and PELD
ease patients for liver transplantation with wait- and associated 90-day-mortality risk for MELD/
ing time being only a subordinate component of Na-MELD
liver transplant allocation. The MELD score was MELD score 3-month mortality (%)
originally developed to assess the short term 6–9 1.9
prognosis of patients evaluated for transjugular 10–19 6.0
intrahepatic porto-systemic shunt (TIPS) proce- 21–29 19.6
dures and utilized three objective, reproducible 30–39 52.6
and patient-specific standard laboratory values ≥40 71.3
(INR, bilirubin, and creatinine) to calculate a MELD = 3.78 × ln[serum bilirubin (mg/dL)] + 11.2 × ln[I
score that can be used to predict 3-month mortal- NR] + 9.57 × ln[serum creatinine (mg/dL)] + 6.43
Na-MELD = [0.025 × MELD × (140 − Na)] + 140
ity (Table 9.2). Several modifications were made PELD = 4.80[Ln serum bilirubin (mg/dL)] + 18.57[Ln
by UNOS before utilizing it as part of the liver INR]−6.87[Ln albumin (g/dL)] + 4.36(<1 year
transplant allocation algorithm: The score was old) + 6.67(growth failure)
9  Recipient and Donor Selection and Transplant Logistics: The US Perspective 113

MELD = MELD(i) + 1.32*(137 − Na) − [0.0 known to affect waiting time significantly. The


33*MELD(i)*(137 − Na)]. three most important additional factors are serum
Sodium values less than 125 mmol/L are set to albumin, ascites, and encephalopathy [15].
125 and values greater than 137 mmol/L are set
to 137.
Furthermore, patients with stage II hepatocel-  urrent Donor Liver Distribution
C
lular carcinoma (HCC) face a much greater risk Resulting in Regional Disparities
of tumor progression beyond the accepted Milan in Liver Allocation
criteria which would result in their removal from
the waiting list and essentially exempt these As a result of the introduction of the MELD score
patients from a curative therapy. To compensate to allocate donor livers the mean MELD score at
for this mortality risk that is unappreciated by the the time of transplantation increased from 18.5 to
laboratory MELD score, stage II HCC patients 24.1 and the mean PELD score from 10.7 to 17.7.
are assigned an arbitrarily higher MELD score The number of patients that had to be removed
starting at 22 irrespective of their ‘true MELD from the waiting list because they were too sick
sore’ (“laboratory-MELD”). Since the risk of to be transplanted or had died on the waiting list
tumor progression increases over time, the decreased for the first time since introduction of
MELD score of these patients can be increased the UNOS waiting list, indicating that the goal to
by 10% mortality risk (i.e. 1–3 exceptional reduce death on the waiting list and to prioritize
MELD points) every 3 months. Regional review the sickest patients for liver transplantation had
boards in each of the 11 UNOS regions were been achieved [16].
appointed to oversee the HCC MELD and other The US distribution system is traditionally
standard exceptional point processes and also based on local, regional and national distribution
decide about individual cases of transplant candi- units, with 63 Donor Service Areas (DSA) for the
dates who may be disadvantaged by the current local Organ Procurement Organizations (OPO,
MELD system. Common examples for the stan- Fig.  9.3) and 11 Organ Procurement Transplant
dard exceptional MELD system are patients with Network (OPTN) regions within the United
severe hepatopulmonary syndrome, familial States (Fig. 9.4). Since its commission in 1986,
amyloidosis, polycystic liver disease, metabolic the OPTN is operated by the United Network for
disorders and other liver tumors. Organ Sharing (UNOS), a private non-profit
As mentioned earlier, exception points for organization under federal contract. The OPTN
HCC has led to a significant increase in the num- regions are therefore usually referred to as OPTN/
ber of liver transplants for HCC (Table 9.1) as UNOS or UNOS regions. For non-emergent adult
well as a significant reduction in ‘time on the patients listed for liver transplantation, deceased
waiting list’ for HCC patients. In addition, new donor livers were first offered to candidates
therapeutic options, such as highly selective trans within the local Donor Service Area (DSA) in
arterial chemoembolization (TACE), transcuta- which the organ was procured, then within the
neous and laparoscopic thermal ablation methods UNOS region, and last to the national list. This
such as radio frequency ablation (RFA) or micro- system resulted in a significant difference in the
wave ablation (MWA) have decreased the mortal- mean MELD score at the time of transplantation
ity of patients with HCC awaiting liver between the 11 UNOS regions, which lead to the
transplantation. This has resulted in a decrease of implementation of Share-15 rule in January 2005.
the initial standard exceptional MELD points for This rule required donor offers to be made first to
HCC patients, their activation status, and the patients with a MELD score ≥ 15 within a UNOS
extent of standard exceptional MELD progres- region, before the organ could be allocated to a
sion for UNOS stage 2 HCC patients. The MELD local recipient with a MELD score < 15.
allocation system is under constant review as fac- Nonetheless, a significant difference in MELD
tors other than the four laboratory values are score at the time of transplantation in the UNOS
114 I. Klein et al.

WA

MT ND
MN ME

OR NY
ID WY
SD WI
NV IA
NE
IN PA
OH
MO IL
CA UT WV VA
CO KS KY
AZ NM
TX TN NC
OK
AR

MS AL GA
LA FL

AK

Fig. 9.3  Map of the current 63 Donor Service Areas (DSA) for the local Organ Procurement Organizations (OPO) in
the United States

6
1
7
9
10 2
5
8
11

Fig. 9.4  Map of the 3


current 11 regions of the
4
Organ Procurement
Transplant Network
(OPTN) in the United
States

regions remained, resulting in the initiation of 1B candidates. The allocation sequence was
Share-35 rule in June 2013. Livers from adult recently changed to provide broader access to
donors are allocated first to the most urgent can- those most in need of a liver (those with scores
didates located in the same UNOS region as the higher than 35) and those who would receive the
donor; Status 1A candidates, followed by Status most benefit (patients with scores higher than
9  Recipient and Donor Selection and Transplant Logistics: The US Perspective 115

15). Therefore, after regional Status 1A and 1B lives saved in the eight-district plan [14]
candidates, liver offers are then made to candi- (Fig. 9.6). Even though restructuring the distri-
dates with MELD/PELD scores 35 and higher bution districts into larger geographic areas
within the donor’s UNOS region, with offers first would result in higher transportation and thus
made locally, then regionally, followed by local, transplantation costs, the reduction of costs dur-
then regional candidates with scores greater than ing pre-transplant, transplant and post-­transplant
15. If the organ cannot be allocated after this care were expected to be significantly reduced
algorithm, it is offered to national candidates, in the proposed four- and eight-district plans. In
first with a Status 1A or 1B, then with a MELD the eight-district plan, the savings even out-
score greater than 15. Lastly, the organ is offered weighed the higher spending. Particularly in
to patients with a MELD score lower than 15, regions currently transplanting lower MELD
locally, regionally, then nationally. patients, transplant centers will face increased
The implementation of Share-35 has led to costs [14]. Thus, further adjustments to mini-
an increased percentage of recipients trans- mize the persisting geographic disparities
planted with higher MELD scores as well as remain under continuous discussion.
shorter waiting time (10 days vs. 15 days) and
higher graft and patient survivals (1-year graft
survival increased from 77 to 80% and 1-year  xpanding the Donor Pool/Amount
E
patient survival from 79 to 82%) [13]. Despite of Transplantable Organs
the Share-35 rule regional differences in the
mean-MELD-score at time of transplantation The shortage of donor organs has been the princi-
remain (Fig. 9.5). Thus, mathematical modeling pal limitation of liver transplantation since the
has been used to assess the effects of redistrict- mid-1980s and resulted in the re-evaluation of
ing into eight or four districts, rather than 11 donor-selection criteria and donors that were pre-
UNOS regions. The model predicted a reduction viously considered unsuitable for transplantation.
in overall mortality, including listed and post- These so called expanded criteria organs that bear
transplant patients with an estimate of 676 saved a higher risk for the recipient can be categorized as
lives in 5 years in the four-district plan and 362 increasing either the disease transmission risk

Fig. 9.5  Disparities in


liver allocation indicated
by MELD-at transplant No program in DSA < 23
in the current 63 Donor 23-24 25-28
Service Areas (DSA) 29+
116 I. Klein et al.

Proposed 4 district distribution model

Predicted median-MELD at transplant in the proposed


4 district distribution model

No liver program
MELD>29
MELD 27-29
MELD 24-26
MELD<24

Fig. 9.6  Proposed geographic four district distribution model and anticipated MELD at transplant in this model

(infection, neoplasm, etc.) or the risk of organ dys- zation of grafts from infectious donors should be
function (primary non function, delayed graft evaluated carefully in light of the recipient’s
function, chronic transplant failure, etc). The utili- immunosuppression. However, there is increas-
9  Recipient and Donor Selection and Transplant Logistics: The US Perspective 117

ingly data that the transmission risk is lower than short cold ischemia time the split organ should be
expected. The risk of transmitting a bacterial considered an expanded criteria organ. In pediat-
infection in case of donor bacteremia is low and ric recipients however, the utilization of a split
can be reduced even further by prophylactic use of liver transplant yielded significantly better results
antibiotics in the recipient. Donors with docu- [22]. During donation after cardiac death (DCD),
mented bacterial meningitis can be safely utilized the donor liver is subjected to a variable period of
using prophylactic antibiotics in the post-trans- warm ischemia and hypo-perfusion. The number
plant period [17]. Donors with unspecified poten- of DCD liver procurements was about 450 organs
tial central nervous infections should not be per year or 6.4% of all recovered livers in 2007
considered without extensive virological workup [23]. Livers from DCD procurements have an
using nuclear acid testing. The general risk of increased rate of primary non function, delayed
transmitting infections like rabies, West Nile fever graft function and a well described increased rate
and others is considered low, however possibly of late ischemic-type biliary complications,
fatal in case of transmission [18]. With the advent resulting in a significant reduction of quality of
of highly effective direct-­ acting antiviral drugs life and graft survival [24, 25]. African-American
(DAAD) for Hepatitis C and B, the risk of trans- race versus white race in the donor, reduced
mitting an untreatable disease seems less likely height (in 10 cm decrements from 170 cm) and
and raises the question if organs with active viral cerebrovascular accident or ‘other” cause of
hepatitis should be allocated to all consented death (not trauma, anoxia, or stroke) as well as
patients on the wait list comparable to CMV- cold ischemia time (indicated by regional or
positive donor organs. The incidence of malig- national share) are other general factors associ-
nancy in organ donors is estimated to be 3% and ated with liver graft failure [20].
the risk of transmitting malignancy by transplanta- Steatosis of the donor liver, especially in the
tion of a solid organ is approximately 0.01% [19]. form of large droplet fat, called macrosteatosis
In terms of decreased organ function, Cox (fat vacuoles >50% of the hepatocyte size)
regression studies identified seven donor charac- potentiates ischemia reperfusion injury and
teristics that independently predicted an increased increases complications after liver transplanta-
risk of graft failure: Donor age > 40 years, dona- tion. The rate of primary graft dysfunction cor-
tion after cardiac death (DCD) and split/partial relates with the extent of steatosis with particular
liver graft were strongly associated with graft poor results if the large droplet steatosis is
failure and cerebrovascular accident and ‘other greater than 60% of the liver parenchyma result-
causes’ of brain death, reduced height and ing in a high rate of primary non function, pro-
African-American race were modestly associated longed ICU stay and hospitalization [26].
with graft failure. All seven factors were quanti- In most cases a frozen section liver biopsy can
tatively combined to the donor risk index (DRI) clarify the suitability of a donor organ by
to objectively assess the risk of post-transplant ­determining the amount and type of steatosis,
graft dysfunction [20]. Older donors were found potential additional fibrosis, inflammatory infil-
to have an increased risk of graft failure starting tration, or hepatocyte necrosis. These criteria
at the age of 40, but particularly >60 years. cannot be quantified macroscopically and may
Advanced age also significantly increased the present a contraindication for organ donation and
severity of hepatitis C viral recurrence in the pre-­ transplantation. However, processing and evalu-
DAAD eara [21]. In adult transplant recipients, ating of a biopsy can prolong cold ischemia time,
the rate of graft failure and post-transplant mor- which should be kept as short as possible when
bidity is significantly higher in split-liver recipi- using expanded criteria organs. A pre-­
ents with a reduced graft volume compared to the procurement biopsy is preferable, but if not fea-
recipient’s standard liver volume and secondary sible, the gain of additional information obtained
to technical challenges. Even when the organ through back-table biopsy has to be weighed
donor is young, with healthy parenchyma and a against the risk of prolonging cold ischemic time.
118 I. Klein et al.

 onor Management Prior


D and long waiting times. The first recipients were
to Procurement primarily children and in 1989 almost 65% of the
donors were their parents. However, right lobe
Brain death is associated with multiple patho- and later left lobe living donor liver transplanta-
physiologic changes that may progress to hemo- tion for adult recipients outnumbered the pediatric
dynamic instability, hypo-perfusion, metabolic liver transplants very quickly and now represent
and endocrine decompensation and may ulti- the majority of living donor liver transplantation
mately result in multi-organ system failure and (67 vs. 23% in 2007). A total of more than 3000
pre-procurement demise [27, 28]. Impaired oxy- living donor liver transplants in more than 100
gen use and a subsequent shift from aerobic to centers have been performed in the US between
anaerobic metabolism with consecutive lactic aci- 1998 and 2007 with about one third in pediatric
dosis has been observed following brain death recipients. Overall the enthusiasm of the proce-
and was associated with decreased levels of triio- dure has declined steadily from a peak of 522 liv-
dthyronin (T3), thyroxin (T4), cortisol, and insu- ing donor liver transplants in 2001 (111 children,
lin. The administration of T4 in donors awaiting 411 adult recipients) to 219 cases in 2009. Six
organ procurement almost completely reverses fatal outcomes in living liver donors have been
the anaerobic metabolism, restores cardiovascular reported in the US, two of them in 2010. Apart
function, and is associated with a significantly from the calculated mortality risk of 0.2–0.5%
higher number of procured organs per donor when there is a significant incidence of postoperative
compared to donors managed without thyroxin morbidity (up to 30%) which emphasizes the risk
[29]. Standard donor specific therapy therefore of this major operation for the organ donor [33].
includes the administration of T4, methylpred- This associated risk has affected the wide applica-
nisolone and insulin as soon as the potential donor tion of this procedure so far in western countries.
requires extensive fluid resuscitation and vaso- However, living donor liver transplantation has
constrictors. Early identification and management emerged as the primary source of organ donation
of disseminated intravascular coagulation (DIC), in East Asia with centers performing over 300 liv-
diabetes insipitus (DI), and neurogenic pulmo- ing donor liver transplants per year. So far, living
nary edema, hypothermia and cardiac arrhythmias donor liver transplantation has not significantly
is essential [30]. A mean arterial blood pressure increased the number of available organs for
between 65 and 100 mmHg, urine output of transplantation in the US. However, for children
1–2 cc/kg/h, hemoglobin of 7–9 g/dL, arterial under 2 years living donor liver transplant is pref-
oxygen partial pressure of ≥80 mmHg, and a core erable over split liver or pediatric whole organ
body temperature of 35.5–38 °C are ideal for transplantation [34]. In adult recipients, the proce-
hemodynamic and metabolic stability prior to dure has evolved into a treatment option for
organ recovery and should be the clinical goal patients with a significant reduction in quality of
parameters. Donor management goals (DMGs) life and relatively preserved hepatic function who
are increasingly used to standardize donor man- are unable to receive MELD exception points in
agement and optimize end-organ function [31]. the current allocation system such as patients with
Clinical studies which focus on pre-conditioning cholestatic cirrhosis.
before organ procurement demonstrated a signifi-
cant effect for mild hypothermia in kidney trans-
plantation [32]. References
1. Based on OPTN data as of April 8, 2017. 2004
Annual Report of the U.S. Organ Procurement and
Living Donor Liver Transplantation Transplantation Network and the Scientific Registry
of Transplant Recipients: Transplant Data 1994–
Living donor liver transplantation in the US has 2003. Rockville, MD: Department of Health and
emerged as a consequence of the organ shortage Human Services, Health Resources and Services
9  Recipient and Donor Selection and Transplant Logistics: The US Perspective 119

Administration, Healthcare Systems Bureau, Division 18. Fischer SA. Emerging viruses in transplantation:

of Transplantation; Richmond, VA: United Network there is more to infection after transplant than CMV
for Organ Sharing; Ann Arbor, MI: University Renal and EBV. Transplantation. 2008;86(10):1327–39.
Research and Education Association. 19. Kauffman HM, McBride MA, Delmonico FL. First
2. Busuttil RW, Tanaka K. The utility of marginal report of the united network for organ sharing trans-
donors in liver transplantation. Liver Transpl. plant tumor registry: donors with a history of cancer.
2003;9(7):651–63. Transplantation. 2000;70(12):1747–51.
3. Alonso O, et al. Advanced donor age increases the 20. Feng S, et al. Characteristics associated with liver
risk of severe recurrent hepatitis C after liver trans- graft failure: the concept of a donor risk index. Am J
plantation. Transpl Int. 2005;18(8):902–7. Transplant. 2006;6(4):783–90.
4. Freeman RB Jr, Edwards EB. Liver transplant wait- 21. Berenguer M, et al. Contribution of donor age to
ing time does not correlate with waiting list mortality: the recent decrease in patient survival among HCV-­
implications for liver allocation policy. Liver Transpl. infected liver transplant recipients. Hepatology.
2000;6(5):543–52. 2002;36(1):202–10.
5. Kamath PS, et al. A model to predict survival in 22. Burdelski MM, Rogiers X. What lessons have we
patients with end-stage liver disease. Hepatology. learned in pediatric liver transplantation? J Hepatol.
2001;33(2):464–70. 2005;42(1):28–33.
6. Wiesner RH, et al. MELD and PELD: application 23. Tuttle-Newhall JE, et al. Organ donation and uti-

of survival models to liver allocation. Liver Transpl. lization in the United States: 1998–2007. Am J
2001;7(7):567–80. Transplant. 2009;9(4 Pt 2):879–93.
7. Network-HRSA, O.P.a.T. Final rule with comment 24. Chan EY, et al. Ischemic cholangiopathy following
period. Federal Register. 1989;63:16296–338. liver transplantation from donation after cardiac death
8. Transplantation., C. o.O.P.a. Assessing current poli- donors. Liver Transpl. 2008;14(5):604–10.
cies and the potential impact of the DHHS final rule. 25. Abt PL, et al. Survival following liver transplan-

Washington, DC: National Academy Press; 1999. tation from non-heart-beating donors. Ann Surg.
9. McDiarmid SV, Anand R, Lindblad AS. Development 2004;239(1):87–92.
of a pediatric end-stage liver disease score to predict 26. McCormack L, Dutkowski P, El-Badry AM, Clavien
poor outcome in children awaiting liver transplanta- PA. Liver transplantation using fatty livers: always
tion. Transplantation. 2002;74(2):173–81. feasible? J Hepatol. 2011;54(5):1055–62. https://doi.
10. Wiesner R, et al. Model for end-stage liver dis-
org/10.1016/j.jehp.2010.11.004.
ease (MELD) and allocation of donor livers. 27. Novitzky D, et al. Change from aerobic to anaero-
Gastroenterology. 2003;124(1):91–6. bic metabolism after brain death, and reversal fol-
11. Sharma P, et al. Liver transplantation for hepatocel- lowing triiodothyronine therapy. Transplantation.
lular carcinoma: the MELD impact. Liver Transpl. 1988;45(1):32–6.
2004;10(1):36–41. 28. Cooper DK, Novitzky D, Wicomb WN. The patho-
12. Biggins SW, Kim WR, Terrault NA, Saab S, Balan V, physiological effects of brain death on potential donor
Schiano T, et al. Evidence-based incorporation of serum organs, with particular reference to the heart. Ann R
sodium concentration into MELD. Gastroenterology Coll Surg Engl. 1989;71(4):261–6.
2006;130:1652–60 29.
DuBose J, Salim A. Aggressive organ donor
13. Nekrasov V, Matsuoka L, Rauf M, Kaur N, Cao S, management protocol. J Intensive Care Med.
Groshen S, Alexopoulos SP. National Outcomes of 2008;23(6):367–75.
Liver Transplantation for Model for End-Stage Liver 30. Roth BJ, et al. Cadaveric organ donor recruitment at
Disease Score ≥40: The Impact of Share 35. Am J Los Angeles County Hospital: improvement after for-
Transplant. 2016 Oct;16(10):2912–24. doi: 10.1111/ mation of a structured clinical, educational and admin-
ajt.13823. Epub 2016 May 13. istrative service. Clin Transpl. 2003;17(Suppl 9):52–7.
14. Gentry SE, Chow EK, Massie A, Luo X, Shteyn E, 31. Franklin GA, et al. Optimization of donor manage-
Pyke J, Zaun D, Snyder JJ, Israni AK, Kasiske B, ment goals yields increased organ use. Am Surg.
Segev DL. Liver sharing and organ procurement orga- 2010;76(6):587–94.
nization performance under redistricted allocation. 32. Niemann CU, Feiner J, Swain S, et al. Therapeutic
Liver Transpl. 2015;21(8):1031–9. hypothermia in deceased organ donors and kidney-
15. Schaubel DE, et al. Survival benefit-based deceased-­ graft function. N Engl J Med 2015;373:405-414.
donor liver allocation. Am J Transplant. 2009;9(4 Pt 33. Ghobrial RM, et al. Donor morbidity after living

2):970–81. donation for liver transplantation. Gastroenterology.
16. Wiesner RH. Patient selection in an era of donor
2008;135(2):468–76.
liver shortage: current US policy. Nat Clin Pract 34. Roberts JP, et al. Influence of graft type on outcomes
Gastroenterol Hepatol. 2005;2(1):24–30. after pediatric liver transplantation. Am J Transplant.
17. Lopez-Navidad A, et al. Successful transplantation of 2004;4(3):373–7.
organs retrieved from donors with bacterial meningi-
tis. Transplantation. 1997;64(2):365–8.
Surgical Techniques in Liver
Transplantation 10
Holden Groves and Juan V. del Rio Martin

Keywords siologist and surgeons and the liver anesthesiologists


Incision · Portocaval shunt · Anhepatic phase · needs to have a good understanding of different
Portal anastomosis · Arterial anastomosis · surgical techniques of this complex surgery. The
Hepatectomy success of the liver transplantation will depend on
the creation of adequate hepatic inflow, both portal
and arterial, and outflow into the inferior vena cava.
Introduction

Liver transplantation is the standard of care for end History of Surgical Technique
stage liver disease, fulminant liver failure, unre-
sectable primary tumors of the liver and metabolic The evolution of liver transplantation has involved
diseases whose associated enzymatic defect are improvements of surgical and anesthesial
located primarily in the liver. The evolution of liver techiques, the development of more effective and
transplantation has involved improvements of sur- less toxic immunosuppression and improved
gical and anesthesiological techniques, the devel- patient selection to achieve outcomes unthikable
opment of more effective and less toxic only a few years ago [1, 2]. The surgical technique
immunosuppression and improved patient selec- of liver transplantation has evolved since the pio-
tion to achieve outcomes unthinkable only few neering times largely due to an increased knowl-
years ago. The surgical technique of liver trans- edge of the liver hemodynamic physiology [3]. It
plantation has evolved since the pioneering times was initially considered too complex to perform
largely due to an increased knowledge of liver the hepatectomy with preservation of the vena
hemodynamic physiology. Intraoperative manage- cava without the expertise to dissect the liver off
ment requires close cooperation between anesthe- the vena cava and therefore the most widely used
technique was the orthotopic liver transplantation
H. Groves, MD, MS without preservation of the recipient vena cava as
Columbia University Medical Center, described by Starzl et al. [4] (Fig. 10.1a). This
New York, NY, USA technique consisted of the removal of the diseased
Department of Anesthesiology, liver during temporary, complete cross-clamping
Columbia University Medical Center, of the vena cava above and below the liver and the
New York, NY, USA portal vein. Clamping of the vena cava and portal
J. V. del Rio Martin, MD, FASTS (*) vein affected the hemodynamic equilibrium of the
Hospital Auxilio Mutuo, San Juan, PR, USA recipient with a drastic reduction of the blood
e-mail: jdelrio@auxiliomutuo.com

© Springer International Publishing AG, part of Springer Nature 2018 121


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_10
122 H. Groves and J. V. del Rio Martin

a (that is inserted using cut-down technique) into


the systemic venous circulation. The benefits of
the bypass were an improvement of hemody-
namic instability during the anhepatic phase, bet-
ter preservation of the renal function, reduction of
blood loss and prevention of portal and systemic
b congestion. However the overall incidence of
complications of VVB was between 10 and 30%
including fatal complications, such as air or
thrombotic pulmonary emboli during cannulation
or accidental decannulation of the bypass circuit.
Other reported complications include hypother-
mia, blood clotting in the bypass system and ves-
sel thrombosis, lymphocele formation, hematoma,
vascular and nerve injury as a complication of
c
catheter placement, wound infection or dehis-
cence, infected vascular suture lines, hemothorax
after insertion of a large bore cannula percutane-
ously, and prolonged operative and warm isch-
emia time [5, 6]. The high incidence of
complications and higher cost, stimulated the
investigation of new surgical techniques.
d Sir Roy Calne described the caval preserva-
tion technique with the use of a pediatric donor
liver into an adult recipient in his initial paper in
1968 [7] (Fig. 10.1b). Others also published the
use of the caval preservation technique [8]. In
1989 Tzakis published the first detailed descrip-
tion of liver transplantation with vena cava pres-
ervation or “piggyback” technique [9]. The first
published large series of liver transplantation
Fig. 10.1  Vena Cava management. (a) Caval removal using “piggyback” technique demonstrated bet-
with end to end anastomosis. (Standard Technique). (b)
Vena Cava preservation and end to side anastomosis ter hemodynamic stability, lower blood transfu-
(Piggyback technique). (c) Modification of the end to side sion requirements and shorter operative time.
anastomosis with extension of the anastomosis into a side The initial description of the piggyback tech-
to side caval anastomosis. (d) Vena Cava preservation nique used the junction of the middle and left
with cavocavostomy and closure of both ends of the donor
Vena Cava. (Belghiti modification) hepatic veins as outflow which could produce
the unintended consequence of outflow dys-
function [10]. Belghiti et al. approached the out-
return and caval and splanchnic bed congestion. flow problem by developing a new technique of
To overcome the potentially deleterious effects of side to side caval anastomosis [11] (Fig. 10.1d).
preload reduction Shaw et al [5] described the use Although the piggyback technique made the
of the ­venovenous bypass (VVB) as follows. The venovenous bypass obsolete, it did not solve the
saphenous or femoral veins are cannulated usu- problem of splanchnic congestion that may
ally by cut-­down or transcutaneously and another complicate the dissection. Tzakis, in 1993 pub-
cannula is inserted into the portal vein. Blood is lished a description of adding a temporary por-
then pumped from the splanchnic bed and lower tocaval shunt (TPCS) that resulted in the
body through a cannula placed in the axillary vein hepatectomy to be performed without portal
10  Surgical Techniques in Liver Transplantation 123

The Liver Transplant Procedure

Before starting a liver transplant it is convenient


to review the unique characteristics of the patient
such as anatomical variations, presence of arte-
rial variants, portal vein thrombosis and inflam-
matory reactions secondary to trans-arterial
chemo-embolization (TACE) or radio-frequency
ablation (RFA) of tumors, especially if those are
close to vascular structures. Indentifying these
variations may help determine the most benefi-
cial technique and is also relevant for the anesthe-
siologist who can prepare for potential blood loss
and instability during the dissection. It is impor-
Fig. 10.2  Creation of a temporary portocaval shunt dur- tant for the surgeon and anesthesiologist to dis-
ing the hilar dissection cuss any variants and the surgical strategy prior
to incision.
hypertension which reduced blood loss and For example a hypertrophied caudate lobe that
achieved outstanding hemodynamic stability wraps around the vena cava could make the use
[12] (Fig. 10.2). Cherqui and Belghiti from of the piggyback technique extremely difficult
France published the first large series of TPCS and the caval removal technique might be the
during liver transplantation in 1994 and 1995 adequate choice. If a hypertrophied left lateral
[13, 14] but it was the Figueras et al. from segment is intimately attached to an enlarged and
Barcelona that proved in a randomized con- congested spleen it may make sense to create a
trolled trial the benefits of this technique. The portocaval shunt to decompress the splanchnic
use of the TPCS allowed better hemodynamic area and then perform a hepatectomy using pig-
stability during the anhepatic phase with lower gyback technique from the right to the left leav-
transfusion requirements and better renal func- ing the attachments to the spleen until the
tion in patients with high portal flow or portoca- hepatectomy is finalized and splenic congestion
val gradient [15]. More recently the Mount Sinai has subsided. These decisions will also affect the
Group demonstrated the benefit of the TPCS in anesthetic management during dissection and
high risk donors [16]. This technique is particu- should be communicated.
larly useful in cases of fulminant hepatic failure It is further critical to maintain in close com-
when there was no time to develop hepatofugal munication with the procurement team in order
(collateral) circulation and cerebral edema lim- to time the initiation of the recipient operation
its the amount fluid that can be given during the and reduce cold ischemic time as well as be
anhepatic phase [17]. The piggyback technique aware of any abnormalities of the donor. It is
demands finesse to dissect the liver from the important to make sure that general conditions of
vena cava by suture ligating of all retro hepatic the recipient have not deteriorated since last time
short vessels and finally dissecting the right, examined by a thorough physical exam and
middle and left hepatic veins. review of laboratory and radiology results.
The clear anatomical definition of vascular All techniques (piggyback with and without
and biliary segmentation of the liver by Couinaud temporary porto-caval shunt, caval cross
allowed techniques of partial hepatectomies as a clamp and veno-venous bypass) are still in use
treatment of benign and malignant diseases of the in centers world-wide. We will restrict the fol-
liver. These surgical techniques are also used to lowing description and illustration on the pig-
split two grafts from a single deceased or living gyback technique with temporary porto-caval
donor [18, 19]. shunt:
124 H. Groves and J. V. del Rio Martin

Back Table Preparation Diaphragm


a
Organ preservation and cold ischemia time (CIT)
are very important factors that affect outcome
after liver transplantation and a short CIT corre-
lates with improved function of the allograft [20].
Close coordination between the surgical
(Donor—Recipient) and the anaesthesiology
team are essential to minimize cold ischemic
time and ideally finish the back table surgery by
the time the recipient team is ready for the anhe-
patic phase. b
Back table preparation involves dissection of Suprahepatic
the diaphragm off the donor liver (Fig. 10.3a). vena cava
Phrenic vein
Next, both ends of the inferior vena cava (IVC) stump
are prepared for anastomosis (Fig. 10.3b, c),
avoiding leaks at reperfusion by stitching up all
branches off the vena cava. The focus is then
shifted towards cleaning the portal vein by dis-
secting it from its origin at the junction of
splenic and superior mesenteric veins to its
bifurcation. Finally, the hepatic arterial supply c
is delineated and dissected all the way up to the
celiac trunk origin along with an aortic patch.
Care should be taken not to extend the dissec-
tion proximally beyond the gastro-duodenal
artery (GDA). At this stage arterial reconstruc-
tion is also performed in case of an aberrant
arterial anatomy. Surgeon’s preference will
Infrahepatic
decide about leaving a short right hepatic artery vena cava
by connecting it to the stump of the gastroduo-
denal artery (Fig. 10.4), or a long right hepatic Right adrenal
vein
artery, while bending the aortic flap and leaving
the superior mesenteric artery or the splenic
artery to anastomose to the native arterial inflow
(Fig. 10.5a, b).
Fig. 10.3  Back table preparation. (a) Dissection of the
donor liver of the diaphragm. Preparation of the inferior
Abdominal Incision and Exposure vena cava: (b) Suprahepatic ligation of the phrenic vein
stump. (c) Infrahepatic ligation of the right adrenal vein
Adequate exposure is fundamental to allow the
appropriate dissection and access for the native hernia due to the poor vascularity at the junction.
liver hepatectomy. The incision most commonly Another commonly used incision is the “J”
used consists of a bilateral sub costal incision shaped (also known as Hockey Stick or
with subxiphoid extension (Fig. 10.6a). This inci- Makuuchi’s incision) which has been used alter-
sion allows excellent exposure of the suprahe- natively as it allows adequate blood supply to the
patic IVC but increases the risk of incisional wound edges [21] (Fig. 10.6b).
10  Surgical Techniques in Liver Transplantation 125

Native Liver Hepatectomy

The hepatectomy can be divided into three gen-


eral steps: (1) mobilization of the liver (2) dissec-
tion and division of the hilar structures (hepatic
artery, portal vein, and common bile duct). (3)
management of the inferior vena cava (IVC). The
order of the steps may change according to sur-
geon preference.
Once the abdomen has been opened, the sur-
Right replaced geon should explore the peritoneal cavity for
artery
possible signs of contraindication for transplanta-
tion such as active infection or advanced tumour
disease; any ascites will be suctioned out and a
sample should be sent for cell count and culture.
Hemodynamic instability can occur with the
drainage of a large amount of ascites.
Gastroduodenal Next the falciform ligament is divided cepha-
artery
lad using electrocautery until the anterior surface
of the suprahepatic IVC is identified. A self-­
retaining retractor will be used based on the sur-
geon’s preference. We prefer to use the Thompson
Fig. 10.4  Short right hepatic artery anastomosis with the retractor. Mobilization of the left lobe of the liver
stump of gastro-duodenal artery is begun by dividing the left triangular ligament
and coronary ligament.

a Common
Commo
hepatic artery
Right replaced
eplaced
artery Left ga
gastric artery

Superior
uperior Splenic artery
mesenteric
senteric
artery

Aorta
Common
Commo hepatic
artery
b
Left ga
gastric artery
eplaced
Right replaced
artery

Fig. 10.5 Preparation Splenic artery


of the arterial
reconstruction with Superior
uperior
aortic flap. (a) With mesenteric
senteric
native aorta. (b) With artery
bending the aortic flap
126 H. Groves and J. V. del Rio Martin

a Thrombus

Portal vein

Coronary vein

Splenic vein

Superior
mesentric
vein

Fig. 10.6  Abdominal incision and exposure with either


subxiphoid extension of the bilateral subcostal incision
(a) or right “J” shaped “Hockey Stick” incision

The hilar dissection starts by dividing the


hepato-­duodenal ligament and encircling the
porta hepatis searching for arterial variants. The
most common variants are the replaced right
hepatic artery from the superior mesenteric artery c
behind the portal vein and left hepatic artery from
the left gastric artery in the gastrohepatic liga-
ment [22]. The porta hepatis is put under tension
by retracting the liver cephalad, the stomach,
duodenum, and transverse colon caudad. Both Portal vein
the left and right hepatic arteries are identified Left renal vein
and divided close to the liver. The common
hepatic artery is dissected proximally towards the Splenorenal
shunt
GDA junction. The common bile duct is isolated
and divided close to the liver. Care must be taken Left renal vein
stump
to preserve the blood supply of the common bile
duct by preserving as much periductal tissue as
Fig. 10.7  Treatment of portal vein thrombus. (a) Eversion
possible. Next the portal vein is dissected all the thrombendovenectomy. (b) Jump graft from patent supe-
way up to the bifurcation proximally and to the rior mesenteric vein to donor portal vein using a donor
superior aspect of pancreas distally. In cases of iliac vein segment. (c) Reno-portal anastomosis
portal vein thrombosis the dissection will con-
tinue up to the junction of the splenic vein and the Management of the portal vein at this point
superior mesenteric vein and the thrombus is will depend on whether the chosen technique will
removed by thrombectomy while the surgeon preserve the vena cava or not. If preservation of
controls the flow proximally [23] (Fig. 10.7a). the vena cava technique is chosen without tempo-
Sudden blood loss at the time of thrombectomy rary portocaval shunt (TPCS) the portal vein will
should be anticipated by the anesthesiologist. be left untouched until the final steps of the hepa-
10  Surgical Techniques in Liver Transplantation 127

tectomy in order to preserve some portal decom- connectors prompted the search for simplified
pression and it will be clamped right before systems and the majority of cases are now done
explantation. using the standard technique without VVB [24].
If a TPCS is performed it is constructed by When portal inflow and supra- and infra-­hepatic
exposing the infra hepatic IVC and performing vena cava are clamped the surgeon should wait
an end-to-side porto caval anastomosis. The rest for the anesthesiologist to confirm that accept-
of the hepatectomy will be facilitated by the com- able hemodynamics can be achieved and main-
plete devascularisation and reduction of the por- tained for the duration of the anhepatic time. If
tal pressure. The caudate lobe is dissected off the the patient is not hemodynamically stable during
anterior portion of the vena cava by suture liga- the test clamp the vessels should be unclamped
ture of the short retrohepatic veins. When the again and the anesthesiologist should attempt to
portal vein is not sectioned this dissection is more improve the hemodynamic situation (for example
difficult. Occasionally it may be necessary to with more vasopressors or fluids) before another
clamp the portal vein if dissection of the retrohe- test clamp is applied. If the patient remains unsta-
patic veins results in a large amount of blood loss ble VVB can be considered [25]. If VVB is used,
and hemodynamic instability. Care should be minimal dissection of the infrahepatic vena cava
taken due to anatomical variations of the retrohe- is needed. It is encircled cephalad to the left renal
patic veins and we recommend using small vein with a vessel loop and the right adrenal vein
clamps and suture ligature when those veins are is suture ligated. After mobilizing the liver bilat-
larger than 3 mms. Once the hepatocaval liga- erally the suprahepatic vena cava is also encircled
ment is divided and suture ligated we recommend with blunt digital dissection and prepared for
to clamp and section the right hepatic vein sepa- clamping.
rately from the middle and left hepatic veins Once supra and infra hepatic vena cava clamps
which can be clamped together. We usually are applied the vena cava is sectioned leaving
extend the dissection of the vena cava above the adequate cuff at both ends for anastomosis and
hepatic veins, severing the attachments on the left the native liver is removed from the field. Before
side and dissecting, ligating and sectioning the initiating the vascular anastomosis adequate
phrenic veins. This way the shape of the vena hemostasis is mandatory, especially in the retro-
cava will round up, improving adequate flow hepatic retroperitoneal area.
return while side clamped.
When using the standard technique, the vena
porta is dissected in the same way and prepared Implantation of the Donor Liver
for clamping and section as proximal to the liver
as possible. When a veno-venous bypass is used The donor liver has been prepared on the back
the portal vein is cannulated to decompress the table for implantation as described above. The
splanchnic territory and connected to a Y shaped liver can be flushed either on the backtable or in
tubing system that also decompresses the subhe- situ. For example prior to moving the graft to the
patic caval system through a cannula placed in operating room the liver can be flushed using
the left femoral or saphenous vein. Both portal either cold albumin or a cristalloid solution to
and systemic flow will then be directed towards remove excess potassium. Alterantively once the
the superior vena cava through a cannula placed upper cava anastomosis has been performed (for
in the left axilary vein either through cut down or example with a 3/0 Prolene running suture),
using a percutaneous (left) internal jugular can- hypothermic or normothermic [26] solution can
nula. The extracorporeal circuit is completed be flushed through the donor portal vein to clear
with the use of a centripetal force pump. The the high potassium content of the UW preserva-
complexity of this process with multiple cannula tion solution. The solutions used differ according
placements, collapsing vessels and the risk of to centers and may include albumin or lactated
thrombosis or gas emboli through the multiple Ringer solution. Most centers now avoid teh sue
128 H. Groves and J. V. del Rio Martin

of starch solutions. The lower caval anastomosis must take place between the surgical and anaes-
is left open for drainage. Once the effluent is thesiology teams to assure that the patient’s
clear the lower cava anastomosis is finalized. At hemodynamics are optimal at this critical stage
this time the anesthesiologist should notify the [28]. If the patient is hyperkalemic prior to reper-
surgeon if the patient is hyperkalemic as the sur- fusion the portal vein anastomosis can be
geon can leave the caval anastomosis slightly unclamped while some of the caval anastomosis
open during reperfusion to flush out high potas- is still open and the caval clamp is still on. Some
sium preservation fluid. Flushing can be per- of the perfusing blood with potentially high
formed antegrade after completion of the portal potassium and acid will then drain out of the
vein anastomosis and brief removal of its clamp caval anastomosis and not reach the patient. This
or retrograde by removing the caval clamps and can however be associated with substantial blood
draining the blood through a loose portal vein loss. Once some of the blood is flushed out, the
anastomosis before tightening it up. portal vein is clamped again, the caval anstomisis
The piggy-back technique entails preserving is completed and then the cava unclamped. It is
the recipient’s entire IVC along with the orifices critical to notify the surgeon early if the patient is
of the hepatic veins that are joined to create one hyperkalemic so he/she can prepare for this.
common orifice (Fig. 10.8). The lower cava is After assuring an uneventful reperfusion, the
closed once the flushing is finalized by a silk tie hepatic arterial anastomosis follows. This anasto-
or vascular stapling [27]. In the cases with TPCS, mosis is performed in an end-to-end fashion
this is now taken down for example by using a between the donor and recipient common hepatic
vascular stapling device. The native portal vein artery. The goal is to make the anastomosis as
will then be anastomosed to the donor portal vein wide and as straight as possible to avoid hepatic
in an end to end fashion. In the modified piggy- artery stenosis or kinking. Dissecting the
back technique by Belghiti (Cavocavoplasty), Common Hepatic Artery off lymphatic tissue and
both the supra- and infrahepatic IVC of the donor ligating and excising the gastroduodenal artery
are oversewn, and the cavocaval anastomosis is will facilitate this. Some surgeons prefer to use
created between the donor and recipient IVCs in the aortic patch for anastomosis to the recipient
a side-to-side fashion [11]. Once the caval anas- arterial inflow. In cases of inadequate arterial
tomosis is finished, the portal vein anastomosis is inflow due to trauma or during retransplantation,
created in an end-to-end fashion. Figure 10.9 one may be forced to use a donor arterial conduit
illustrates each step of the procedure. either from the infrarrenal or supraceliac aorta.
Reperfusion is done after the portal vein anas- The liver transplantation is completed by
tomosis is finished. Considerable coordination performing the biliary reconstruction. A
cholecystectomy is performed on the donor
­
liver. This is a modification from the early days
in which the donor gallbladder was used as part
of the biliary reconstruction. The high risk for
ischemic cholecystitis in the post transplant
period led to this modification. After the chole-
cystectomy we should avoid redundancy of the
donor bile duct by excising the distal end and
limiting ischemic cholangiopathy at the bile
duct anastomosis. The donor’s bile duct is
divided proximal to the cystic duct to assure
adequate blood supply. The bile duct anastomo-
sis is constructed in an end-to-end fashion. The
use of T-tube drain to protect the biliary anasto-
Fig. 10.8  Caval anastomosis mosis is seldomly used anymore after several
10  Surgical Techniques in Liver Transplantation 129

Fig. 10.9  Sequence of steps from top to bottom (photos Flushing of the donor liver to remove high potassium con-
on the left by Juan Del Rio Martin; illustrations on the tent University of Wisconsin solution. [Illustration on
right by Holden Groves) (a) Anhepatic phase with side right: Donor liver (brown), Donor vena cava (orange),
clamp of the hepatic veins and the temporary portocaval flush (grey), recipient hepatic artery (red)]. (d) Portocaval
shunt. [Illustration on right: Side clamp of the recipient shunt takedown with the use of the vascular stapler.
hepatic veins (light blue) and vena cava (dark blue) and (Illustration on right: Portocaval shunt (green), donor liver
the temporary portocaval shunt (green) and recipient (brown), recipient vena cava (dark blue)). (e) Stapled
hepatic artery (red)]. (b) End to Side cavocaval anastomo- stumps of the caval end and main portal vein preparing for
sis. With the inclusion of the Right Hepatic Vein of the anastomosis. (Illustration on right: Donor caval end
recipient the diameter of both ends is similar reducing the (orange) and recipient main portal vein (purple)). (f) End
outflow dysfunction risk. [Illustration on right: Donor to end portal vein anastomosis. (Illustration on right:
liver (brown), donor vena cava (orange), Recipient vena donor vena cava (orange), recipient portal vein (portal
cava (dark blue), recipient hepatic veins (light blue)]. (c) vein))
130 H. Groves and J. V. del Rio Martin

Fig. 10.9 (continued)

studies demonstrated it to be the cause of severe split livers, the biliary reconstruction is done by a
complications [29]. Roux-en-Y hepatico-jejuonostomy.
In certain situations where end-to-end biliary Biliary reconstruction with the technique of
anastomosis is contraindicated either because of hepatico-jejunostomy is done by first dividing
disease of the native bile duct for example with the donor’s bile duct proximal to the cystic duct
primary sclerosing cholangitis (PSC), or techni- junction to guarantee a well vascularised end for
cal reasons, e.g., living related liver transplants or anastomosis. A Roux-en-Y jejunal limb is then
10  Surgical Techniques in Liver Transplantation 131

created by mobilizing a suitable loop of proximal portal vein to the splenic artery, common hepatic
jejunum of approximately 50 cm in length. The artery, or directly to the aorta using a jump graft
anastomosis is then constructed with a standard [34, 35]. Long term patient survival posttrans-
end-to-side Roux-en-Y hepatico-jejuonostomy. plantation with normal liver function and lack of
We strongly recommend the retrocolic and retro- portal hypertension with the use of a calibrated
gastric technique in order to avoid tension on the portal vein arterialization has been recently
anastomosis created by gastric or colonic reported [36]. When a pretransplant portosys-
distension. temic shunt is created distal splenorenal and
mesocaval shunts are safer shunts if subsequent
transplantation is planned [37].
Portal Vein Thrombosis Cavoportal Hemitransposition (CPHT)
involves using the IVC as a source of portal vein
The incidence of portal vein thrombosis (PVT) at inflow. There are a variety of ways to performing
the time of liver transplantation varies from 2.1 CPHT: an end-to-end anastomosis between the
to 26% [30, 31]. PVT used to be an absolute con- native IVC and the portal vein of the liver graft
traindication to liver transplantation however (Fig. 10.1a), side-to-end fashion with deliberate
recently PVT has become only a challenge but luminal constriction (Fig. 10.1b) or calibration of
not a contraindication mainly because of techni- the vascular diameter by placing clips (Fig. 10.1c)
cal advances of the vascular anastomoses, on the retro-hepatic IVC [38, 39].
According to the extent of the thrombosis, PVT A new variant of portal inflow in PVT Grade 4
can be classified into four grades: Grade 1: is the creation of a reno-portal anastomosis
Partially thrombosed PV, in which the thrombus (RPA), an end-to-end anastomosis that is created
is confined to <50% of the vessel lumen with or between the native left renal vein and the donor
without minimal extension to the SMV. Grade 2: portal vein in those cases in which a large sponta-
>50% occlusion of the PV, including total occlu- neous or constructed splenorenal shunt is present
sions, with or without minimal extension to the that will derive most of the splanchnic flow into
SMV. Grade 3: Complete thrombosis of both PV the left renal vein [40, 41] (Fig. 10.7c).
and proximal SMV with patent distal SMV. Grade
4: complete thrombosis of both PV and proximal
as well as distal SMV [30]. Domino Liver Transplantation
For grades 1, 2, and 3 PVT, eversion thrombo-
endovenectomy has been suggested as the surgi- Some rare systemic diseases based on a single
cal technique of choice by many authors enzymatic dysfunction located in the liver paren-
(Fig. 10.7a) [23, 30, 32]. In grade 4 PVT, where chyma result in liver function that is otherwise
the thrombus extends beyond the junction of normal. Familial amyloidotic polyneuropathy
superior mesenteric and splenic veins, eversion (FAP) is a genetic condition residing in the hepa-
thromboendovenectomy is often not feasible tocyte that produces a mutation of transthyretin;
[33]. A good option in this situation is a jump this abnormal protein is deposited in peripheral
graft from a patent segment of the proximal SMV nerves, gastrointestinal tract, heart, and kidneys.
to the donor portal vein using an iliac vein seg- The liver of these patients is otherwise normal
ment of the donor. This graft is tunnelled through apart from producing this abnormal protein, and
the transverse mesocolon (Fig. 10.7b). If the por- has been used as an organ for recipients with des-
tal flow continues to be suboptimal, some authors perate need of a liver transplant [42]. No added
suggested options such as arterialization of the risk to either the FAP patient or their recipients
portal vein, Cavoportal hemitransposition has been found [43].
(CPHT) or renoportal anastomoses (RPA). In these cases the hepatectomy demands the
Arterialization of the portal vein involves aug- removal of a long portion of the retrohepatic vena
menting the portal inflow by anastomosing the cava as a standard technique, with or without the
132 H. Groves and J. V. del Rio Martin

need for VVB. Long arterial and portal segments inferior vena caval occlusion. Surg Gynecol Obstet.
1992;175(3):270–2.
are necessary for the graft and the patient. The
12. Tzakis AG, Reyes J, Nour B, Marino IR, Todo S,
hepatic artery is clamped and divided proximal to Starzl TE. Temporary end to side portacaval shunt in
the take-off of the gastroduodenal artery. The orthotopic hepatic transplantation in humans. Surg
portal vein should be clamped and divided just Gynecol Obstet. 1993;176(2):180–2.
13. Belghiti J, Noun R, Sauvanet A. Temporary porto-
1 cm below portal bifurcation. Finally the vena
caval anastomosis with preservation of caval flow
cava is divided above and below the liver. On the during orthotopic liver transplantation. Am J Surg.
back table the liver is perfused through the portal 1995;169(2):277–9.
vein and the hepatic artery and the biliary tree is 14. Cherqui D, Lauzet JY, Rotman N, et al. Orthotopic
liver transplantation with preservation of the caval
washed. The recipient of the domino liver will be
and portal flows. Technique and results in 62 cases.
transplanted using the piggyback technique, Transplantation. 1994;58(7):793–6.
while the patient with FAP will have a standard 15. Figueras J, Llado L, Ramos E, et al. Temporary por-
procedure as described above [26, 44]. tocaval shunt during liver transplantation with vena
cava preservation. Results of a prospective random-
ized study. Liver Transpl. 2001;7(10):904–11.
16. Ghinolfi D, Marti J, Rodriguez-Laiz G, et al. The
beneficial impact of temporary porto-caval shunt in
References orthotopic liver transplantation: a single center analy-
sis. Transpl Int. 2011;24(3):243–50.
1. Starzl TE, Groth CG, Brettschneider L, et al. 17. Belghiti J, Noun R, Sauvanet A, et al. Transplantation
Orthotopic homotransplantation of the human liver. for fulminant and subfulminant hepatic failure with
Ann Surg. 1968;168(3):392–415. preservation of portal and caval flow. Br J Surg.
2. Jain A, Marcos A, Reyes J, et al. Tacrolimus for 1995;82(7):986–9.
primary liver transplantation: 12 to 15 years actual 18. Starzl TE, Koep LJ, Weil R 3rd, Lilly JR, Putnam CW,
follow-up with safety profile. Transplant Proc. Aldrete JA. Right trisegmentectomy for hepatic neo-
2005;37(2):1207–10. plasms. Surg Gynecol Obstet. 1980;150(2):208–14.
3. Lautt WW. The 1995 Ciba-Geigy Award Lecture. 19. Couinaud C. Contribution of anatomical research to
Intrinsic regulation of hepatic blood flow. Can J liver surgery. Fr Med. 1956;19(5):5–12.
Physiol Pharmacol. 1996;74(3):223–33. 20. Furukawa H, Todo S, Imventarza O, et al. Effect of
4. Starzl TE, Marchioro TL, Vonkaulla KN, Hermann cold ischemia time on the early outcome of human
G, Brittain RS, Waddell WR. Homotransplantation hepatic allografts preserved with UW solution.
of the liver in humans. Surg Gynecol Obstet. Transplantation. 1991;51(5):1000–4.
1963;117:659–76. 21. Mehrabi A, Fonouni H, Muller SA, Schmidt J. Current
5. Shaw BW Jr, Martin DJ, Marquez JM, et al. Venous concepts in transplant surgery: liver transplantation
bypass in clinical liver transplantation. Ann Surg. today. Langenbeck's Arch Surg. 2008;393(3):245–60.
1984;200(4):524–34. 22. Deshpande RR, Heaton ND, Rela M. Surgical anat-
6. Khoury GF, Mann ME, Porot MJ, Abdul-Rasool IH, omy of segmental liver transplantation. Br J Surg.
Busuttil RW. Air embolism associated with veno-­ 2002;89(9):1078–88.
venous bypass during orthotopic liver transplantation. 23. Robles R, Fernandez JA, Hernandez Q, et al. Eversion
Anesthesiology. 1987;67(5):848–51. thromboendovenectomy in organized portal vein
7. Calne RY, Williams R. Liver transplantation in Man-­ thrombosis during liver transplantation. Clin Transpl.
I, observations on technique and organization in five 2004;18(1):79–84.
cases. Br Med J. 1968;4:535–40. 24. Chari RS, Gan TJ, Robertson KM, et al. Venovenous
8. Starzl TE, Brettschneider L, Martin AJ Jr, et al. Organ bypass in adult orthotopic liver transplanta-
transplantation, past and present. Surg Clin North tion: routine or selective use? J Am Coll Surg.
Am. 1968;48(4):817–38. 1998;186(6):683–90.
9. Tzakis A, Todo S, Starzl TE. Orthotopic liver trans- 25. Johnson MW, Powelson JA, Auchincloss H Jr,
plantation with preservation of the inferior vena cava. Delmonico FL, Cosimi AB. Selective use of veno-­
Ann Surg. 1989;210(5):649–52. venous bypass in orthotopic liver transplantation. Clin
10. Parrilla P, Sanchez-Bueno F, Figueras J, et al.
Transpl. 1996;10(2):181–5.
Analysis of the complications of the piggy-back 26. Llado L, Figueras J. Techniques of orthotopic liver
technique in 1112 liver transplants. Transplantation. transplantation. HPB (Oxford). 2004;6(2):69–75.
1999;67(9):1214–7. 27. Eghtesad B, Kadry Z, Fung J. Technical consider-
11. Belghiti J, Panis Y, Sauvanet A, Gayet B, Fekete ations in liver transplantation: what a hepatologist
F. A new technique of side to side caval anastomo- needs to know (and every surgeon should practice).
sis during orthotopic hepatic transplantation without Liver Transpl. 2005;11(8):861–71.
10  Surgical Techniques in Liver Transplantation 133

28. Paugam-Burtz C, Kavafyan J, Merckx P, et al.


37. Mazzaferro V, Todo S, Tzakis AG, Stieber AC,

Postreperfusion syndrome during liver transplantation Makowka L, Starzl TE. Liver transplantation in
for cirrhosis: outcome and predictors. Liver Transpl. patients with previous portasystemic shunt. Am J
2009;15(5):522–9. Surg. 1990;160(1):111–6.
29. Bawa SM, Mathew A, Krishnan H, et al. Biliary
38. Pinna AD, Nery J, Kato T, Levi D, Nishida S, Tzakis
reconstruction with or without an internal biliary stent AG. Liver transplant with portocaval hemitrans-
in orthotopic liver transplantation: a prospective ran- position: experience at the University of Miami.
domised trial. Transpl Int. 1998;11(Suppl 1):S245–7. Transplant Proc. 2001;33(1–2):1329–30.
30. Yerdel MA, Gunson B, Mirza D, et al. Portal vein 39. Olausson M, Norrby J, Mjornstedt L, Liden H, Friman
thrombosis in adults undergoing liver transplantation: S. Liver transplantation using cavoportal hemitrans-
risk factors, screening, management, and outcome. position- a life-saving procedure in the presence of
Transplantation. 2000;69(9):1873–81. extensive portal vein thrombosis. Transplant Proc.
31. Lerut JP, Mazza D, van Leeuw V, et al. Adult liver 2001;33(1–2):1327–8.
transplantation and abnormalities of splanch- 40. Kato T, Levi DM, DeFaria W, Nishida S, Tzakis

nic veins: experience in 53 patients. Transpl Int. AG. Liver transplantation with renoportal anasto-
1997;10(2):125–32. mosis after distal splenorenal shunt. Arch Surg.
32. Robles R, Fernandez JA, Hernandez Q, et al. Eversion 2000;135(12):1401–4.
thromboendovenectomy for organized portal vein 41. Azoulay D, Adam R, Castaing D, et al. Liver trans-
thrombosis encountered during liver transplantation. plantation with cavoportal or renoportal anastomo-
Transplant Proc. 2003;35(5):1915–7. sis: a solution in cases of diffuse portal thrombosis.
33. Seu P, Shackleton CR, Shaked A, et al. Improved Gastroenterol Clin Biol. 2002;26(4):325–30.
results of liver transplantation in patients with portal 42. Azoulay D, Samuel D, Castaing D, et al. Domino liver
vein thrombosis. Arch Surg. 1996;131(8):840–4. dis- transplants for metabolic disorders: experience with
cussion 844–45 familial amyloidotic polyneuropathy. J Am Coll Surg.
34. Erhard J, Lange R, Giebler R, Rauen U, de Groot H, 1999;189(6):584–93.
Eigler FW. Arterialization of the portal vein in ortho- 43. Tincani G, Hoti E, Andreani P, et al. Operative risks of
topic and auxiliary liver transplantation. A report of domino liver transplantation for the familial amyloid
three cases. Transplantation. 1995;60(8):877–9. polyneuropathy liver donor and recipient: a double
35. Stieber AC, Zetti G, Todo S, et al. The spectrum of analysis. Am J Transplant. 2011;11(4):759–66.
portal vein thrombosis in liver transplantation. Ann 44. Figueras J, Pares D, Munar-Ques M, et al. Experience
Surg. 1991;213(3):199–206. with domino or sequential liver transplantation in
36. Bonnet S, Sauvanet A, Bruno O, et al. Long-term familial patients with amyloid polyneuropathy.
survival after portal vein arterialization for portal Transplant Proc. 2002;34(1):307–8.
vein thrombosis in orthotopic liver transplantation.
Gastroenterol Clin Biol. 2010;34(1):23–8.
Intraoperative Monitoring
11
Claus G. Krenn and Marko Nicolic

Keywords Liver failure affects all organ systems and


Pulmonary artery catheter · Pulse pressure induces hemodynamic, hematological, metabolic
variation · Neurological monitoring · and other homeostatic abnormalities; successful
Hemodynamic monitoring · Cardiac output · management of patients undergoing liver trans-
Transesophageal echocardiography plantation requires comprehensive monitoring of
all these systems [6].
Only few recommendations on best monitor-
Introduction ing practices are found in the current literature,
and monitoring during liver transplantation may
Liver transplantation (LTx) has made great vary depending on technique, center and country
strides over the last decades and evolved towards of transplant [5, 7, 8]; specific practice patterns
a well-established procedure offered in hundreds often depend on personal experience and prefer-
of transplant programs in more than 80 countries ences [6, 8–10]. Furthermore management often
worldwide. Despite this impressive progress, needs to be modified for specific patients popula-
challenges in liver transplantation have only tions [8, 11].
shifted and transplantation still remain costly and The aim of this chapter is to provide a discus-
resource-intensive [1–5]. sion of a step-wise approach to intraoperative
The decreasing number of contraindications to monitoring based on recent developments in clin-
liver transplantation (resulting in more co-mor- ical and translational research.
bidities) and use of marginal grafts require opti-
mal monitoring of perioperative therapy [3–5].
Early recognition of homeostatic disturbances
and their timely treatment may improve outcome Conventional Laboratory Tests
and decrease perioperative mortality.
Profound disturbances of homeostasis are fre-
quently seen in patients with liver dysfunction
and failure ­during LTx. These abnormalities also
C. G. Krenn, MD (*) • M. Nicolic, MD compromise other organ functions and overall
Department of Anaesthesia, General Intensive Care
and Pain Medicine, Medical University of Vienna, metabolic function. Detection and monitoring of
General Hospital Vienna, Vienna, Austria rapidly changing metabolic disturbances are
e-mail: claus.krenn@meduniwien.ac.at inevitably part of the intraoperative, anesthetic

© Springer International Publishing AG, part of Springer Nature 2018 135


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_11
136 C. G. Krenn and M. Nicolic

management in LTx [8, 12]. Although monitor- monitoring tools such as microdialysis may
ing of these parameters is common knowledge, eventually emerge as sensitive monitors of early
their specific relevance in the context of recent graft viability in the near future.
publications in the field of LTx is evaluated to Lactate concentrations are a similarly easy to
emphasize the importance of regular intraopera- monitor substrate [27, 28]. Two major mecha-
tive assessment. Furthermore changes of labora- nisms are held responsible for hyperlactatemia in
tory parameters can be used as an estimate of the LTx patients. The shift to anaerobic glycolysis
commencing functioning of the transplanted occurs when oxygen demand exceeds supply in
graft. an attempt to maintain cellular function. Increased
In the absence of firm recommendations mon- lactate production and decreased clearance due to
itoring of conventional parameters should be liver dysfunction leads to accumulation of lactate
adapted to the patient’s actual condition; the [21, 29]. Routine lactate monitoring might not be
choice of monitor as well as intervals of measure- able to discriminate between increased produc-
ment should remain with the discretion of the tion versus impaired clearance but persistent or
anesthesiologist as long as it is in accordance worsening hyperlactatemia will require further
with evidence based practice. investigations and decreasing lactate concentra-
tions are an excellent indicator of stable graft
function. Interstitial lactic acidosis in the donor
Assessment of metablic parameters allograft has been associated with the occurrence
of reperfusion injury; however severe acid base
End stage liver dysfunction is associated with disturbances were solely attributed to cardiovas-
various alterations in metabolism, many of which cular collapse after reperfusion only associated
are corrected after LTx. Some alterations of with higher ASA status but not with prolomged
metabolisms may however not correct rapidly ischemia times in another investigation [23, 30,
and/or are poorly understood [13–15]. Glucose 31].
metabolism is frequently affected and the periph- Another substrate of utmost importance in
eral insulin resistance in patients with end-stage liver disease is serum ammonia resulting from
liver disease is characterized by a decrease in urea cycle and inter-organ trafficking [32].
non-oxidative glucose disposal, which improves Ammonia is taken up by cortical astrocytes and
but does not normalize right after LTx. Metabolic then converted to osmotically active glutamine.
syndrome has a higher prevalence in liver trans- This results in passive influx of water and osmotic
plant recipients than in the general population cerebral edema and subsequent intracranial
and is associated with an increased risk of vascu- hypertension of varying severity [33–35]. The
lar events [16]. association between ammonia neurotoxicity and
Intraoperative blood glucose control hepatic encephalopathy (HE)—although criti-
decreases the incidence of hyperglcymia and cized for the lack of a good correlation between
improves outcome [17–19], although routine use blood levels and the severity of HE—has been
of continuous infusion of insulin for correction the basis for designing treatments to decrease
remains controversial [20]. In a porcine animal plasma ammonia or modulating its intestinal gen-
model of liver failure prolonged cold ischemic eration [36, 37]. The combination of neurotoxic
time was associated with altered glucose metab- ammonia and activation of inflammatory
olism [21]. Correspondingly, intrahepatic glu- ­cytokines in acute and acute-on chronic liver fail-
cose levels may result from glycogen degradation ure may further worsen brain dysfunction [38].
in hepatocytes injured by ischemia [22, 23]. The In the near future we may be able to detect spe-
most precise descriptions of metabolic changes cific metabolic changes and their interdependen-
derive from microdialysis studies in rodents and cies and use distinct specific metabolic pattern or
pigs that have recently been confirmed in adult profiles for monitoring. First results of microdialy-
and pediatric settings [22, 24–26]. Graft- sis retrieved metabolomics are encouraging and
11  Intraoperative Monitoring 137

the transfer of these technologies into clinical ment except postoperative hypophosphatemia in
practice and the standardization into routine appli- living liver donors [55, 56].
cation are tasks for the oncoming decade [39, 40]. If conventional management is limited, the
potential correction of electrolyte and acid-­base
imbalances by the use of intraoperative renal
Electrolytes replacement therapy has recently been shown in a
group of patients with high MELD Scores [57].
Electrolyte imbalances pose serious hazards to
patients undergoing LTx [41, 42] and detection
and correction of electrolyte abnormalities is of Temperature
utmost importance. Hyperkalemia is the most
dreaded electrolyte abnormality. A retrospective Hypothermia is one of the key symptoms of acute
study of 1124 patients undergoing Ltx found that liver failure however it may also occur intraopera-
hyperkalemia was associated with the number of tively inadvertently after reperfusion of a cooled
red blood cell transfusion and higher baseline graft [58, 59]. The negative effects of hypothermia
potassium values during the prereperfusion on wound healing and the coagulation system are
period and warm ischemia time, donor hospital well known but temperature regulation during liver
stay and the use of veno-venous bypass during transplantation remains poorly studied. Active
postreperfusion [43]. warming to avoid hypothermia is recommended
Several recent publication emphasized the [60, 61] but mild hypothermia may be advanta-
effect of hyponatremia on outcome [44]. Low geous as adjunct therapy for increased intracranial
serum sodium was found to be an independent pressure and decrease brain metabolism and oxida-
predictor for waitlist mortality and its inclusion tive stress in acute liver failure [62, 63]. Continual
into the MELD (Model for End-stage Liver (“repeated regularly and frequently in steady rapid
Disease) score improved the predictive accuracy succession”) temperature measurement during sur-
in American and European investigations [6, 45– gery is mandated for any anesthetic by Standards
47]. Hyponatremia is also a risk factor for poor for Basic Anesthetic Monitoring of the American
outcome after LTx, possibly due to heart failure, Society of Anesthesiology.
infectious complications, renal failure and neuro-
logical complications [42, 45, 48]. Osmotic demy-
elinisation and central pontine myelinolysis is Hemostasis and Coagulation
more frequent in hyponatremic patients but can
develop in patients with low, normal or even ele- Even with a reduction in blood transfusion
vated sodium plasma levels when sodium levels requirements during liver transplantation over the
increase too rapidly. Frequent assessment and last decade coagulopathy and hemorrhage are
slow correction of sodium plasma levels may be still relevant features of [64–68]. Coagulopathy
critical in prevention of this devastating complica- in LTx patients results from qualitative and quan-
tion [49–53]. titative deficiencies of pro- and anticoagulant fac-
Hypocalcemia is common with transfusion tors, diminished clearance of activated factors,
blood that contain citrate and need to be cor- hyperfibrinolysis and disturbances in platelet
rected rapidly. Many centers routinely administer function and count [69, 70]. The coagulation sys-
magnesium during the anhepatic phase to ame- tem is often considered rebalanced, as bleeding
liorate the arrhythmogenic effect of reperfusion. and thrombotic complications under unstressed
Preoperative administration of magnesium may conditions are less commonly observed despite
also improve coagulopathy assessed by thromb- severe abnormalities and exhausted compensa-
elastography [54]. tory mechanisms.
Other electrolyte abnormalities are less com- Preoperative conventional laboratory coagula-
mon and less relevant for the anesthetic manage- tion tests are of little value in the prediction of
138 C. G. Krenn and M. Nicolic

intraoperative transfusion requirements [70–73] Near-infrared spectroscopy (NIRS) aims to


and transfusion practice in LTx often depend on asses changes in brain capillary saturation and
the monitoring method used [72–74]. mitochondrial oxygen tension of the frontal lobe
Treatment of coagulopathy should be guided by but has rarely been used during LTx [92, 93] as
point-of-care test such as thrombelastography or hyperbilrubinemia may interfere with NIRS mea-
ROTEM. These tests deliver rapid results and surement [86, 93].
reflect the interaction of coagulation proteins and Transcranial Doppler sonography allows a
cells. The maximum clot firmness as well as the reliable and repeatable—but unfortunately not
form of the curve—to be assessed within minutes— continuous—non-invasive assessment of cere-
provides information about clot formation, fibrino- bral blood flow at the bedside [94, 95]. By calcu-
gen levels, as well as platelet count and function lating resistance, pulsatility indices and
[70–73]. This may aid in the decision-­making about assessment of specific wave forms not only
the use of plasma, factor concentrates and platelet blood flow velocity but also impaired cerebral
transfusions, minimize potential side effects of auto regulation, a result of cerebral edema and
unnecessary transfusions and costly medications intracranial hypertension can be assessed [96].
such as recombinant factor VIIa [75, 76] and reduce Xenon clearance for the determination of cere-
the administration of packed red blood cells and bral blood flow is probably the most precise
other blood products [66]. method but clinically difficult to use and still
only of scientific interest [97].
The use of invasive intracranial probes to mea-
Neurological Monitoring sure intracranial pressure (ICP) in patients with ALF
remains controversial [98, 99]. Epidural devices
In acute liver failure (ALF) hepatic dysfunction have the lowest rate of complications but are less
causes deteriorating neurological function due to reliable [100]. Subdural devices have acceptable
the effects of hyperammonemia, proinflamma- precision and are most commonly used whereas
tory cytokines and oxidative stress among others intraparenchymal are seldom used [101–103].
[77] potentially leading to cerebral edema, intra- Cranial computed tomography is insensitive to
cranial hypertension (ICH) or fatal herniation. detect intracranial hypertension but can be used to
Neurological complications remain one of the rule out other intracranial pathology seen in acute
leading causes of morbidity and mortality in ALF liver failure Head CT scans are recommended in
[35, 78–82]. cases of severe prolonged coma before transplant
The best monitoring modality for the progression to rule out intracranial hemorrhage and hernia-
of hepatic encephalopathy is the physical exam tion. Alternatively magnetic resonance imaging
unless coma supervenes. At this point intracranial (MRI) can be used [104]. The benefit of newer
hypertension needs to be ruled out for example using and less invasive monitoring devices such as auto-
EEG, cerebral blood flow measurement, ultrasound mated pupillometer that measure the papillary
assessment of optic nerve sheath or direct intracra- light response and its recovery remains to be
nial pressure (ICP) measurement [79, 80]. determined [105]. We recommend a step-wise
Quantitative EEG analysis and somatosensory approach using more extensive neurological mon-
evoked and acoustic potentials are sensitive and itoring as the neurological condition worsens.
well established in liver transplant candidates
[83–85]. The use of the bispectral (BIS) index
has been advocated by some as a peri-transplant Hemodynamic Monitoring
monitor of hepatic encephalopathy [86–89] as
well as to guide the anesthetic administration in A large number of patients with liver failure
patients undergoing LTx [90]. The addition of develop abnormal cardiac function that may com-
EEG parameters to the MELD score improved its plicate highly stressful LTx [106, 107]. Liver fail-
prognostic value [91]. ure is marked by circulatory abnormalities such as
11  Intraoperative Monitoring 139

low peripheral resistance and a concomitant catheters can be placed and may also provide a
hyperdynamic state often in combination with more accurate measurement of central blood
altered intravascular volume status and abnormal pressure than peripheral radial catheters [119,
ventricular response to stress [107–111]. This car- 121].
diovascular abnormalities that are specific to liver Pulse oximetry is required for any anesthetic
disease can be combined with primary cardiac and may help detect hypoxia due to hepatopul-
disease such as coronary artery disease and car- monary syndrome prior to induction [122–124].
diomyopathy [106, 107, 110, 111]. Other variables derived from pulse oximetry such
The choice of invasive intraoperative monitor- as hemoglobin concentration and pulse pressure
ing has thus to meet these concerns and may need variation are still under investigation for addi-
to include assessment of cardiac output, pre- and tional benefit [125]. Many less invasive devices
afterload as well as oxygen supply and demand. are not reliable with severe hemodynamic altera-
Monitoring modalities should be adapted accord- tion during LTx.
ing to actual needs and familiarity of the anes-
thetic team with the devices [112–114].
New techniques have been developed that Cardiac Output
allow less invasive and continuous monitoring.
Invasiveness must be matched with the ability to Maintaining adequate tissue perfusion and car-
obtain the necessary information and manage diac output is essential during LTx. Measuring
patients optimally even under extreme situations. cardiac output using a pulmonary artery cath-
All monitoring devices have limitations, [115– eter (PAC) remains the gold-standard however
118] and thorough and cautious interpretation is other technologies such as transpulmonary dye
recommended. and lithium dilution, Doppler echocardiogra-
phy and pulse contour analysis have been used
as well [126–131]. The risk of ventricular
Standard Hemodynamic Monitoring arrhythmias during catheterization with the
PAC must be weighed against the benefit of
Standard hemodynamic monitoring as required direct measurement of pulmonary artery pres-
for example by the American Society of sure to rule out portopulmonary syndrome
Anesthesiology comprises pulse oxymetry, [132]. Continuous cardiac output measurement
electrocardiography and blood pressure and with the PAC, based on short burst of heat dis-
temperature monitoring [112–114]. Continuous sipation, becomes inaccurate when central
arterial blood pressure monitoring for example blood temperature is unstable for example dur-
of the radial artery is required and some cen- ing graft reperfusion. Furthermore sudden
ters routinely use either bilateral radial arterial changes in cardiac output, for example when
catheters or radial and femoral arterial cathe- the inferior vena cava is clamped, are not
ters because of the need for blood sampling at immediately recognized [133].
times of extreme hemodynamic instability and Alternative technologies such as continuous
as a backup should one arterial catheter fail tracking of changes in left ventricular stroke
[119, 120]. ­volume by arterial pulse contour method (con-
Inaccurate pressure measurements may occur tinuously integrating the systolic portion of the
for example due to compression of the subclavian arterial wave tracing) have been studied during
artery during rib cage retraction or with excessive LTx [134]; these algorithms assume constant
vasodilation, when radial arterial pressure under- characteristic impedance, vascular resistance and
estimates central aortic blood pressure. It may arterial compliance [126, 128]. Most have been
therefore be helpful to measure more proximal validated during stable conditions but not during
arterial pressure with a femoral arterial catheter. extreme changes in cardiac function that occur
Alternatives such as brachial or axillary artery during LTx [135, 136].
140 C. G. Krenn and M. Nicolic

Other Hemodynamic Variables initially limited its intraoperative use however


rupture and significant bleeding is rather uncom-
Central venous pressure is easily measured as mon [154–156]. While financial investment into
central venous access is almost always placed echocardiography devices has become reason-
during LTx. It has been suggested that peripheral able and TEE has become more prevalent, exten-
venous pressure reflects central venous pressure sive experience and operating/interpreting skill
however this is seldom used [137]. In general with TEE is still required [152, 153]. The use of
pressure derived variables are not very accurate TEE during LTx not only improved diagnostic
in determining preload and diastolic filling as this abilities but also allowed to significantly change
varies with ventricular compliance [138]. Recent therapeutic algorithms [157].
studies confirmed that preload estimates obtained In addition to the direct visualization of the
with PAC such as central venous or capillary heart to monitor volume status and contractility,
wedge pressure were less reliable than volumet- TEE provides valuable information in case of
ric preload parameters such as global enddia- complications such as large pleural effusion,
stolic (GEDV) and intrathoracic blood volume tension pneumothorax or pulmonary thrombo-
­
(ITBV) [139–141]. However only the PAC allows embolis. TEE is further helpful to detect pulmo-
directs measurement of pulmonary artery pres- nary hypertension, intracardiac clot formation,
sures and therefore rule out portopulmonary and hypertrophic cardiomyopathy [157–160].
hypertension; echocardiographic estimation of Visualization of the large vessels permits the
pulmonary artery pressures is potentially an via- diagnosis for example of incomplete obstruction
ble alternative [142–144]. of the inferior vena cava as a result of an inade-
Adequate oxygen delivery can be assessed by quate venous reconstruction [161].
measuring mixed venous saturation in the pulmo- Transoesophageal echocardiographic assess-
nary artery using a PAC; some catheters allow ment of cardiac output can be done either by
continuous measurements. Central venous satu- pulse waved Doppler analysis of flow accross the
ration measured in the vena cava or the right ven- aortic valve or by planimetry or volumetry of the
tricle is a poor substitute for mixed venous ventricles with good correlation to thermodilu-
saturation and does not correlate well. During tion cardiac output [159].
LTx changes in mixed venous saturation may
reflect not only changes of cardiac output [145]
but also of oxygen carrying capacity and demand. Monitoring of Graft Function
Positive pressure ventilation induces cyclic
changes in left ventricular stroke volume by Early assessment of graft function is crucial for
altering right ventricular filling and ejection dur- successful LTx. Assessment of graft function
ing the ventilatory cycle. Stroke volume variation includes evaluation of hepatic artery, portal vein
or pulse pressure variation [146] reflect fluid and caval blood flow to prevent or d­ iagnose vascu-
responsiveness in LTx patients [147, 148] and lar thrombosis or kinking [162, 163]. Delayed
fluid administration algorithms using stroke vol- detection of complications of the vascular anasto-
ume variation or pulse pressure variation are mosis can result in graft loss and possibly mortal-
promising [149, 150]. ity. Hemodynamic instability may affect
splanchnic and graft perfusion as well and needs to
addressed before concluding that a vascular anas-
Transesophageal Echocardiography tomosis is at fault for impaired graft perfusion.
Conventional liver function testing are not
Increasingly transesophageal echocardiography useful in the immediate assessment of liver graft
(TEE) is used as a monitoring tool in patients viability as derangement will be delayed [164].
undergoing LTx [151–153]. The concern of rup- Assessment of graft function should include
turing esophageal varices with the TEE probe either measurement of homeostatic alterations or
11  Intraoperative Monitoring 141

of metabolism [165] and can affect long term understood [176, 177]. These biomarkers will be
outcome [166–168]. even more important to detect adequate graft
function with the use of extended criteria donors
and/or machine perfusion preservation
Blood Flow Assessment [178–180].

Increased portal blood flow typically leads to a


reduced hepatic artery perfusion via the hepatic Dynamic Liver Function Tests
artery buffer response. After ischemia-­reperfusion
injury hepatic blood flow may be further impeded Dynamic liver function test may be better suited
by tissue swelling. In general the hepatic artery to predict graft survival and overall outcome than
buffer response is preserved after LTx but its conventional liver function tests [165, 181, 182].
capability to compensate varies from graft to Most of these tests assess functional reserve of
graft [169, 170]. the liver by measuring metabolism of adminis-
Ultrasound Doppler allows direct measure- tered substances such as lidocaone or midazolam
ment of hepatic artery and portal vein graft blood by the hepatic microsomal cytochrome P450 sys-
flow and can be done intra- and postoperatively tem. These dynamic liver function tests are dis-
[171, 172] and the use of ultrasound contrast cussed elsewhere in this book [176, 183].
agents can improve signal quality [171, 172].
Temporary implantable flow probes have
been used for research and in selected clinical
situations to detect graft congestion from out- References
flow obstruction in living-donor-liver transplan-
1. Perera MT, Mirza DF, Elias E. Liver transplantation:
tation or to observe critical anastomoses e.g. in issues for the next 20 years. J Gastroenterol Hepatol.
pediatric LTx [173, 174]. Experimental use of 2009;24(Suppl 3):S124–31.
transesophageal echocardiography to monitor 2. Walia A, Schumann R. The evolution of liver trans-
hepatic blood flow has unfortunately never been plantation practices. Curr Opin Organ Transplant.
2008;13(3):275–9.
validated in humans [175]. 3. Zarrinpar A, Busuttil RW. Liver transplantation:
past, present and future. Nat Rev Gastroenterol
Hepatol. 2013;10:434–40.
Conventional Liver Function Tests 4. Jones PD, Hayashi PH, Sidney Barrit A IV. Liver
transplantation in 2013: challenges and controversies.
Minerva Gastroenterol Dietol. 2013;59(2):117–31.
In most cases conventional liver function tests 5. Shukla A, et al. Liver transplantation: east versus
fail to detect acute changes in graft function west. J Clin Exp Hepatol. 2013;3:243–53.
quickly enough. Postoperative transaminases 6. Stravitz RT, et al. Intensive care of patients with
acute liver failure: recommendations of the
reflect the extent of injury caused procurement, U.S. Acute Liver Failure Study Group. Crit Care
cold ischemic time and reperfusion and initial Med. 2007;35(11):2498–508.
graft perfusion but not necessarily graft function. 7. Schumann R. Intraoperative resource utilization in
Total bilirubin is a rather insensitive and slow anesthesia for liver transplantation in the United
States: a survey. Anesth Analg. 2003;97(1):21–8;
marker but levels that don’t decrease within days table of contents
after transplant should be a cause for concern and 8. Manley JL, et al. Controversies in anesthetic man-
further investigation. agement of liver transplantation. HPB (Oxford).
Although recent developments have contrib- 2005;7(3):183–5.
9. Morris-Stiff G, Gomez D, Prasad R. Quantitative
uted to a better understanding of the mechanisms assessment of hepatic function and its rel-
of hepatic regeneration, the role of biochemical evance to the liver surgeon. J Gastrointest Surg.
markers such as cytokines, complement factors 2009;13(2):374–85.
and metabolomic or genomic patterns, in deter- 10. Rando K, et al. Optimizing cost-effectiveness in
perioperative care for liver transplantation: a model
mining graft function is not yet completely
142 C. G. Krenn and M. Nicolic

for low- to medium-income countries. Liver Transpl. tion, and complement activation. Transplantation.
2011;17(11):1247–78. 2008;86(8):1096–103.
11. Ozhathil DK, et al. Impact on center volume on 29. Shah AD, Wood DM, Dargan PI. Understanding lac-
outcomes of increased-risk liver transplants. Liver tic acidosis in paracetamol (acetaminophen) poison-
Transpl. 2011;17(10):1191–9. ing. Br J Clin Pharmacol. 2011;71(1):20–8.
12. Hall TH, Dhir A. Anesthesia for liver transplantation. 30. Murphy ND, et al. Liver and intestinal lactate
Semin Cardiothorac Vasc Anesth. 2013;17(3):180–94. metabolism in patients with acute hepatic failure
13. Giusto M, et al. Changes in nutritional status undergoing liver transplantation. Crit Care Med.
after liver transplantation. World J Gastroenterol. 2001;29(11):2111–8.
2014;20(31):10682–90. 31. Silva MA, et al. Interstitial lactic acidosis in the
14. Kosola S, et al. Cholesterol metabolism altered and graft during organ harvest, cold storage, and reper-
FGF21 levels high after pediatric liver transplanta- fusion of human liver allografts predicts subse-
tion despite normal serum lipids. Am J Transplant. quent ischemia reperfusion injury. Transplantation.
2012;12(10):2815–24. 2006;82(2):227–33.
15. Doycheva I, Leise MD, Watt KD. The intestinal 32. Walker V. Ammonia toxicity and its prevention in
microbiome and the liver transplant recipient: what inherited defects of the urea cycle. Diabetes Obes
we know and what we need to know. Transplantation. Metab. 2009;11(9):823–35.
2016;100(1):61–8. 33. Cordoba J, Minguez B. Hepatic encephalopathy.
16. Laryea M, et al. Metabolic syndrome in liver trans- Semin Liver Dis. 2008;28(1):70–80.
plant recipients: prevalence and association with major 34. Belanger M, et al. Mild hypothermia prevents brain
vascular events. Liver Transpl. 2007;13(8):1109–14. edema and attenuates up-regulation of the astrocytic
17. Ammori JB, et al. Effect of intraoperative hyper- benzodiazepine receptor in experimental acute liver
glycemia during liver transplantation. J Surg Res. failure. J Hepatol. 2005;42(5):694–9.
2007;140(2):227–33. 35. Raghavan M, Marik PE. Therapy of intracranial
18. Park C, et al. Severe intraoperative hyperglycemia hypertension in patients with fulminant hepatic fail-
is independently associated with surgical site infec- ure. Neurocrit Care. 2006;4(2):179–89.
tion after liver transplantation. Transplantation. 36. Ong JP, et al. Correlation between ammonia levels
2009;87(7):1031–6. and the severity of hepatic encephalopathy. Am J
19. Park CS. Predictive roles of intraoperative blood glu- Med. 2003;114(3):188–93.
cose for post-transplant outcomes in liver transplan- 37. Butterworth RF. Pathophysiology of hepatic enceph-
tation. World J Gastroenterol. 2015;21(22):6835–41. alopathy: the concept of synergism. Hepatol Res.
20. Gedik E, et al. Blood glucose regulation during 2008;38(s1The 6 Japan Society of Hepatology
living-donor liver transplant surgery. Exp Clin Single Topic Conference: Liver Failure: Recent
Transplant. 2015;13(Suppl 1):294–300. Progress and Pathogenesis to Management. 28-29
21. Gillispie A, et al. Effect of extended cold ischemia September 2007, Iwate, Japan):S116–21.
time on glucose metabolism in liver grafts: experi- 38. Sawhney R, et al. Role of ammonia, inflammation,
mental study in pigs. J Hepato-Biliary-Pancreat and cerebral oxygenation in brain dysfunction of
Surg. 2007;14(2):183–8. acute-on-chronic liver failure patients. Liver Transpl.
22. Nowak G, et al. Metabolic changes in the liver graft 2016;22(6):732–42.
monitored continuously with microdialysis during 39. Serkova NJ, et al. Early detection of graft failure
liver transplantation in a pig model. Liver Transpl. using the blood metabolic profile of a liver recipient.
2002;8(5):424–32. Transplantation. 2007;83(4):517–21.
23. Tsinari KK, et al. Factors affecting metabolic and 40. Hrydziuszko O, et al. Application of metabolomics
electrolyte changes after reperfusion in liver trans- to investigate the process of human orthotopic liver
plantation. Transplant Proc. 2004;36(10):3051–6. transplantation: a proof-of-principle study. OMICS.
24. Haugaa H, et al. Early bedside detection of ischemia 2010;14(2):143–50.
and rejection in liver transplants by microdialysis. 41. Cimen S, et al. Implications of hyponatremia in liver
Liver Transpl. 2012;18(7):839–49. transplantation. J Clin Med. 2015;4(1):66–74.
25. Pischke SE, et al. Hepatic and abdominal carbon 42. Mandell MS, et al. Risk factors associated with acute
dioxide measurements detect and distinguish hepatic heart failure during liver transplant surgery: a case
artery occlusion and portal vein occlusion in pigs. control study. Transplantation. 2015;99(4):873–8.
Liver Transpl. 2012;18(12):1485–94. 43. Xia VW, et al. Predictors of hyperkalemia in the pre-
26. Haugaa H, et al. Clinical experience with microdi- reperfusion, early postreperfusion, and late postre-
alysis catheters in pediatric liver transplants. Liver perfusion periods during adult liver transplantation.
Transpl. 2013;19(3):305–14. Anesth Analg. 2007;105(3):780–5.
27. Bernal W, et al. Blood lactate as an early predictor of 44. Yun BC, Kim WR. Hyponatremia in hepatic enceph-
outcome in paracetamol-induced acute liver failure: alopathy: an accomplice or innocent bystander? Am
a cohort study. Lancet. 2002;359(9306):558–63. J Gastroenterol. 2009;104(6):1390–1.
28. Waelgaard L, et al. Microdialysis monitoring of liver
grafts by metabolic parameters, cytokine produc-
11  Intraoperative Monitoring 143

45. de Morais BS, et al. Central pontine myelinolysis 64. Hartmann M, Szalai C, Saner FH. Hemostasis
after liver transplantation: is sodium the only villain? in liver transplantation: pathophysiology, moni-
Case report. Rev Bras Anestesiol. 2009;59(3):344–9. toring, and treatment. World J Gastroenterol.
46. Biggins SW, et al. Evidence-based incorpo- 2016;22(4):1541–50.
ration of serum sodium concentration into 65. Nacoti M, et al. Coagulopathy and transfusion
MELD. Gastroenterology. 2006;130(6):1652–60. therapy in pediatric liver transplantation. World J
47. Luca A, et al. An integrated MELD model includ- Gastroenterol. 2016;22(6):2005–23.
ing serum sodium and age improves the prediction 66. Trzebicki J, et al. The use of thromboelastometry in
of early mortality in patients with cirrhosis. Liver the assessment of hemostasis during orthotopic liver
Transpl. 2007;13(8):1174–80. transplantation reduces the demand for blood prod-
48. Nadeem A, et al. Chloride-liberal fluids are associ- ucts. Ann Transplant. 2010;15(3):19–24.
ated with acute kidney injury after liver transplanta- 67. Detry O, et al. Avoiding blood products dur-
tion. Crit Care. 2014;18(6):625–34. ing liver transplantation. Transplant Proc.
49. Boon AP, et al. Central pontine myelinolysis in liver 2005;37(6):2869–70.
transplantation. J Clin Pathol. 1991;44(11):909–14. 68. Dalmau A, Sabate A, Aparicio I. Hemostasis and
50. Lee EM, et al. Risk factors for central pontine and coagulation monitoring and management during
extrapontine myelinolysis following orthotopic liver liver transplantation. Curr Opin Organ Transplant.
transplantation. Eur Neurol. 2009;62(6):362–8. 2009;14(3):286–90.
51. Singh N, Yu VL, Gayowski T. Central nervous 69. Lisman T, Ariens RA. Alterations in fibrine struc-
system lesions in adult liver transplant recipients: ture in patients with liver diseases. Semin Thromb
clinical review with implications for management. Hemost. 2016;42(4):389–96.
Medicine (Baltimore). 1994;73(2):110–8. 70. Roullet S, et al. Rotation thromboelastometry
52. Wszolek ZK, et al. Pontine and extrapontine detects thrombocytopenia and hypofibrinogenaemia
myelinolysis following liver transplantation. during orthotopic liver transplantation. Br J Anaesth.
Relationship to serum sodium. Transplantation. 2010;104(4):422–8.
1989;48(6):1006–12. 71. Coakley M, et al. Transfusion triggers in orthotopic
53. Kumar S, et al. Central pontine myelinolysis, an liver transplantation: a comparison of the thrombo-
update. Neurol Res. 2006;28(3):360–6. elastometry analyzer, the thromboelastogram, and
54. Choi JH, Lee J, Park CM. Magnesium therapy conventional coagulation tests. J Cardiothorac Vasc
improves thromboelastographic findings before liver Anesth. 2006;20(4):548–53.
transplantation: a preliminary study. Can J Anaesth. 72. Herbstreit F, et al. Monitoring of haemostasis in liver
2005;52(2):156–9. transplantation: comparison of laboratory based and
55. Yuan D, et al. Hepatectomy-related hypophos- point of care tests. Anaesthesia. 2010;65(1):44–9.
phatemia may predict donor liver dysfunction in 73. Leon-Justel A, et al. Point-of-care haemostasis mon-
live-donor liver transplantation. Transplant Proc. itoring during liver transplantation reduces transfu-
2010;42(10):4548–51. sion requirements and improves patient outcome.
56. Filik L, et al. Hypophosphatemia in living liver Clin Chim Acta. 2015;446:277–83.
donors. Transplant Proc. 2006;38(2):559–61. 74. Huang PH, et al. Accuracy and trending of continu-
57. Nadim MK. Intraoperative hemodialysis during ous noninvasive hemoglobin monitoring in patients
liver transplantation: a decade of experience. Liver undergoing liver transplantation. Transplant Proc.
Transpl. 2014;20(7):756–64. 2016;48(4):1067–70.
58. Han SB, et al. Risk factors for inadvertent hypother- 75. Lisman T, et al. Recombinant factor VIIa improves
mia during adult living-donor liver transplantation. clot formation but not fibrolytic potential in patients
Transplant Proc. 2014;46(3):705–8. with cirrhosis and during liver transplantation.
59. Leake I. Out in the cold: new supercooling technique Hepatology. 2002;35(3):616–21.
extends liver storage time. Nat Rev Gastroenterol 76. Lodge JP, et al. Efficacy and safety of repeated peri-
Hepatol. 2014;11(9):517. operative doses of recombinant factor VIIa in liver
60. D’Amico DF, et al. Thermal homeostasis and liver transplantation. Liver Transpl. 2005;11(8):973–9.
transplantation. Acta Biomed. 2003;74(Suppl 77. Jalan R, et al. Pathogenesis of intracranial hyper-
2):30–3. tension in acute liver failure: inflammation,
61. Madrid E, et al. Active body surface warming ammonia and cerebral blood flow. J Hepatol.
systems for preventing complications caused by 2004;41(4):613–20.
inadvertent perioperative hypothermia in adults. 78. Clemmesen JO, et al. Cerebral herniation in
Cochrane Database Syst Rev. 2016;4:CD009016. patients with acute liver failure is correlated with
62. Vaquero J, Rose C, Butterworth RF. Keeping cool arterial ammonia concentration. Hepatology.
in acute liver failure: rationale for the use of mild 1999;29(3):648–53.
hypothermia. J Hepatol. 2005;43(6):1067–77. 79. Philips BJ, et al. Cerebral blood flow and metabo-
63. Olthof PB, et al. Protective mechanisms of hypother- lism in patients with chronic liver disease undergo-
mia in liver surgery and transplantation. Mol Med. ing orthotopic liver transplantation. Hepatology.
2015;21:833–46. 1998;27(2):369–76.
144 C. G. Krenn and M. Nicolic

80. Steadman RH, Van Rensburg A, Kramer 96. Zheng Y, et al. Continuous cerebral blood flow auto-
DJ. Transplantation for acute liver failure: periop- regulation monitoring in patients undergoing liver
erative management. Curr Opin Organ Transplant. transplantation. Neurocrit Care. 2012;17(1):77–84.
2010;15(3):368–73. 97. Aggarwal S, et al. Cerebral hemodynamic and meta-
81. Mohsenin V. Assessment and management of cere- bolic profiles in fulminant hepatic failure: relationship
bral edema and intracranial hypertension in acute to outcome. Liver Transpl. 2005;11(11):1353–60.
liver failure. J Crit Care. 2013;28(5):783–91. 98. Blei AT, et al. Complications of intracranial pres-
82. Ferro JM, Viana P, Santos P. Management of neu- sure monitoring in fulminant hepatic failure. Lancet.
rologic manifestations in patients with liver disease. 1993;341(8838):157–8.
Curr Treat Options Neurol. 2016;18:37–53. 99. Le TV, et al. Insertion of intracranial pressure moni-
83. Reeves RR, Struve FA, Burke RS. Quantitative EEG tors in fulminant hepatic failure patients: early expe-
analysis before and after liver transplantation. Clin rience using recombinant factor VII. Neurosurgery.
EEG Neurosci. 2006;37(1):34–40. 2010;66(3):455–8. discussion 458
84. Reeves RR, et al. P300 cognitive evoked potentials 100. Bacani CJ, et al. Emergent, controlled lumbar
before and after liver transplantation. Metab Brain drainage for intracranial pressure monitoring dur-
Dis. 2007;22(2):139–44. ing orthotopic liver transplantation. Neurocrit Care.
85. Hussain E, et al. EEG abnormalities are associ- 2011;14(3):447–52.
ated with increased risk of transplant or poor out- 101. Martinez-Manas RM, et al. Camino intracranial
come in children with acute liver failure. J Pediatr pressure monitor: prospective study of accuracy
Gastroenterol Nutr. 2014;58(4):449–56. and complications. J Neurol Neurosurg Psychiatry.
86. Hwang S. Continuous peritransplant assessment of 2000;69(1):82–6.
consciousness using bispectral index monitoring 102. Munoz SJ, Rajender Reddy K, Lee W. The coagu-
for patients with fulminant hepatic failure under- lopathy of acute liver failure and implications for
going urgent liver transplantation. Clin Transpl. intracranial pressure monitoring. Neurocrit Care.
2010;24(1):91–7. 2008;9(1):103–7.
87. Dahaba AA, et al. The utility of using bispectral 103. Karvellas CJ, et al. Outcomes and complications of
index monitoring as an early intraoperative indicator intracranial pressure monitoring in acute liver fail-
of initial poor graft function after orthotopic or split-­ ure: a retrospective cohort study. Crit Care Med.
graft liver transplantation. Gut. 2009;58(4):605–6. 2014;42(5):1157–67.
88. Dahaba AA, et al. Sensitivity and specificity of 104. Alonso J, Cordoba J, Rovira A. Brain magnetic reso-
bispectral index for classification of overt hepatic nance in hepatic encephalopathy. Semin Ultrasound
encephalopathy: a multicentre, observer blinded, CT MR. 2014;35(2):136–52.
validation study. Gut. 2008;57(1):77–83. 105. Yan S, et al. Clinical utility of an automated
89. Vivien B, et al. Detection of brain death onset using pupillometer for assessing and monitoring
the bispectral index in severely comatose patients. recipients of liver transplantation. Liver Transpl.
­
Intensive Care Med. 2002;28(4):419–25. 2009;15(12):1718–27.
90. Kang JG, et al. The relationship between inhala- 106. Moller S, Dumcke CW, Krag A. The heart and
tional anesthetic requirements and the severity of the liver. Expert Rev Gastroenterol Hepatol.
liver disease in liver transplant recipients according 2009;3(1):51–64.
to three phases of liver transplantation. Transplant 107. Ripoli C, Yotti R, Banares R. The heart in liver trans-
Proc. 2010;42(3):854–7. plantation. J Hepatol. 2011;54:810–22.
91. Montagnese S, et al. Prognostic benefit of the addi- 108. Raval Z, et al. Cardiac risk assessment of liver trans-
tion of a quantitative index of hepatic encephalopa- plant candidate. J Am Coll Cardiol. 2011;58:223–31.
thy to the MELD score: the MELD-EEG. Liver Int. 109. Della Rocca G, et al. Arterial pulse cardiac output
2015;35(1):58–64. agreement with thermodilution in patients in hyper-
92. Nissen P, et al. Near-infrared spectroscopy for dynamic conditions. J Cardiothorac Vasc Anesth.
evaluation of cerebral autoregulation during 2008;22(5):681–7.
orthotopic liver transplantation. Neurocrit Care. 110. Carey WD, et al. The prevalence of coronary artery
2009;11(2):235–41. disease in liver transplant candidates over age 50.
93. Madsen PL, et al. Interference of cerebral near-­ Transplantation. 1995;59(6):859–64.
infrared oximetry in patients with icterus. Anesth 111. Zaky A, Bendjelid K. Appraising cardiac dysfunc-
Analg. 2000;90(2):489–93. tion in liver transplantation: an ongoing challenge.
94. Sidi A, Mahla ME. Noninvasive monitoring of cere- Liver Int. 2015;35(1):12–29.
bral perfusion by transcranial Doppler during fulmi- 112. Ozier Y, Klinck JR. Anesthetic management of
nant hepatic failure and liver transplantation. Anesth hepatic transplantation. Curr Opin Anaesthesiol.
Analg. 1995;80(1):194–200. 2008;21(3):391–400.
95. Bindi ML, et al. Transcranial doppler sonography is 113. Nissen P, Friederiksen HJ, Secher NH. Intraoperative
useful for the decision-making at the point of care in hemodynamic monitoring during liver transplan-
patients with acute hepatic failure: a single centre’s tation: goals and devices. Minerva Gastroenterol
experience. J Clin Monit Comput. 2008;22(6):449–52. Dietol. 2010;56(3):261–77.
11  Intraoperative Monitoring 145

114.
Rudnick MR, De Marchi L, Plotkin 131. Krejci V, et al. Comparison of calibrated and uncali-
JS. Hemodynamic monitoring during liver trans- brated arterial pressure-based cardiac output moni-
plantation: a state of the art review. World J Hepatol. tors during orthotopic liver transplantation. Liver
2015;7(10):1302–11. Transpl. 2010;16(6):773–82.
115. Ripoll C, et al. Cardiac dysfunction during liver 132. Gwak MS, et al. Incidence of severe ventricular
transplantation: incidence and preoperative predic- arrhythmias during pulmonary artery catheteriza-
tors. Transplantation. 2008;85(12):1766–72. tion in liver allograft recipients. Liver Transpl.
116. Robertson A. Intraoperative management of 2007;13(10):1451–4.
liver transplantation in patients with hypertro- 133. Bao FP, Wu J. Continuous versus bolus cardiac output
phic cardiomyopathy: a review. Transplant Proc. monitoring during orthotopic liver transplantation.
2010;42(5):1721–3. Hepatobiliary Pancreat Dis Int. 2008;7(2):138–44.
117. Aggarwal S, et al. Postreperfusion syndrome: hypo- 134. Kim YK, et al. Comparison of stroke volume
tension after reperfusion of the transplanted liver. J variations derived from radial and femoral arterial
Crit Care. 1993;8(3):154–60. 113 pressure waveforms during liver transplantation.
118. Aufhauser DD, et al. Cardiac arrest associated with Transplant Proc. 2009;41(10):4220–8.
reperfusion of the liver during transplantation: inci- 135. Lee M, et al. Agreement in hemodynamic monitor-
dence and proposal for management algorithm. Clin ing during orthotopic liver transplantation: a com-
Transpl. 2013;27(2):185–92. parison of FloTrac/Vigileo and two monitoring sites
119. Lee M, et al. Agreement between radial to femoral with pulmonary artery catheter thermodilution. J
arterial blood pressure measurements during liver Clin Monit Comput. 2017;31(2):343–51.
transplantation. Crit Care Resusc. 2015;17(2):101–7. 136. Schlögelhofer T, Gilly H, Schima H. Semi-invasive
120. Duran JA, et al. Best blood sample draw site measurement of cardiac output based on pulse
during liver transplantation. Transplant Proc. contour: a review and analysis. Can J Anesth.
2009;41(3):991–3. 2014;61:452–79.
121. Krenn CG, De Wolf AM. Current approach to intra- 137. Hoftman N, et al. Peripheral venous pressure
operative monitoring in liver transplantation. Curr as a predictor of central venous pressure during
Opin Organ Transplant. 2008;13(3):285–90. orthotopic liver transplantation. J Clin Anesth.
122. Arguedas MR, et al. Prospective evaluation of out- 2006;18(4):251–5.
comes and predictors of mortality in patients with 138. De Wolf AM, et al. Right ventricular function dur-
hepatopulmonary syndrome undergoing liver trans- ing orthotopic liver transplantation. Anesth Analg.
plantation. Hepatology. 2003;37(1):192–7. 1993;76(3):562–8.
123. Mazzeo AT, et al. Significance of hypoxemia screen- 139. Siniscalchi A, et al. Right ventricular end-diastolic
ing in candidates for liver transplantation: our expe- volume index as a predictor of preload status in
rience. Transplant Proc. 2006;38(3):793–4. patients with low right ventricular ejection fraction
124. Cosarderelioglu C, et al. Portopulmonary hyperten- during orthotopic liver transplantation. Transplant
sion and liver transplant: recent review of the litera- Proc. 2005;37(6):2541–3.
ture. Exp Clin Transplant. 2016;14(2):113–20. 140. Krenn CG, et al. Intrathoracic fluid volumes and pul-
125. Kim SH, et al. Accuracy of continuous noninvasive monary function during orthotopic liver transplanta-
hemoglobin monitoring: a systematic review and tion. Transplantation. 2000;69(11):2394–400.
meta-analysis. Anesth Analg. 2014;119(2):332–46. 141. Fazakas J, et al. Volumetric hemodynamic changes
126. Della Rocca G, et al. Preload and haemodynamic and postoperative complications in liver transplant
assessment during liver transplantation: a com- patients. Transplant Proc. 2011;43(4):1275–7.
parison between the pulmonary artery catheter and 142. Della Rocca G, Brondani A, Costa MG. Intraoperative
transpulmonary indicator dilution techniques. Eur J hemodynamic monitoring during organ transplanta-
Anaesthesiol. 2002;19(12):868–75. tion: what is new? Curr Opin Organ Transplant.
127. Greim CA, et al. Continuous cardiac output monitor- 2009;14(3):29–38.
ing during adult liver transplantation: thermal fila- 143. Starkel P, et al. Outcome of liver transplantation
ment technique versus bolus thermodilution. Anesth for patients with pulmonary hypertension. Liver
Analg. 1997;85(3):483–8. Transpl. 2002;8(4):382–8.
128. Feltracco P, et al. Limits and pitfalls of hemody- 144. Tam NL, He XS. Clinical management of portopul-
namic monitoring systems in liver transplantation monary hypertension. Hepatobiliary Pancreat Dis
surgery. Minerva Anestesiol. 2012;78(12):1372–84. Int. 2007;6(5):464–9.
129. Costa MG, et al. Continuous and intermittent cardiac 145. Acosta F, et al. Does mixed venous oxygen satura-
output measurement in hyperdynamic conditions: tion reflect the changes in cardiac output during liver
pulmonary artery catheter vs. lithium dilution tech- transplantation? Transplant Proc. 2002;34(1):277.
nique. Intensive Care Med. 2008;34(2):257–63. 146. Jardin F, et al. Reevaluation of hemodynamic

130. Ferreira RM, do Amaral JL, Valiatti JL. Comparison consequences of positive pressure ventilation:
between two methods for hemodynamic measure- emphasis on cyclic right ventricular afterload-
ment: thermodilution and oesophageal doppler. Rev ing by mechanical lung inflation. Anesthesiology.
Assoc Med Bras. 2007;53(4):349–54. 1990;72(6):966–70.
146 C. G. Krenn and M. Nicolic

147. Biais M, et al. Uncalibrated stroke volume varia- 161. Shapiro RS, et al. Use of intraoperative Doppler
tions are able to predict the hemodynamic effects ultrasound to diagnose hepatic venous obstruction
of positive end-expiratory pressure in patients with in a right lobe living donor liver transplant. Liver
acute lung injury or acute respiratory distress syn- Transpl. 2001;7(6):547–50.
drome after liver transplantation. Anesthesiology. 162. Puhl G, et al. Initial hepatic microcirculation
2009;111(4):855–62. correlates with early graft function in human
148. Garutti I, et al. Extravascular lung water and pulmo- orthotopic liver transplantation. Liver Transpl.
nary vascular permeability index measured at the end 2005;11(5):555–63.
of surgery are independent predictors of prolonged 163. Lisik W, et al. Intraoperative blood flow measure-
mechanical ventilation in patients undergoing liver ments in organ allografts can predict postoperative
transplantation. Anesth Analg. 2015;121(3):736–45. function. Transplant Proc. 2007;39(2):371–2.
149. Biais M, et al. Uncalibrated pulse contour-derived 164. Hoekstra LT, et al. Physiological and biochemical
stroke volume variation predicts fluid responsiveness basis of clinical liver function tests: a review. Ann
in mechanically ventilated patients undergoing liver Surg. 2013;257(1):27–36.
transplantation. Br J Anaesth. 2008;101(6):761–8. 165. Nagel RA, et al. Use of quantitative liver function
150. Konur H, et al. Evaluation of pleth variability index tests—caffein clearance and galactose elimination
as a predictor of fluid responsiveness during ortho- capacity—after orthotopic liver transplantation. J
topic liver transplantation. Kaohsiung J Med Sci. Hepatol. 1990;10(2):149–57.
2016;32(7):373–80. 166. Ecochard M, et al. Could metabolic liver function
151. Burtenshaw AJ, Isaac JL. The role of trans-­
tests predict mortality on waiting list for liver trans-
oesophageal echocardiography for periop- plantation? A study on 560 patients. Clin Transpl.
erative cardiovascular monitoring during 2011;25(5):755–65.
orthotopic liver transplantation. Liver Transpl. 167. Dubray BJ, Zarrinpar A. Quantification of hepatic
2006;12(11):1577–83. functional capacity: a call for standardisation. Expert
152. Wax DB, et al. Transesophageal echocardiogra- Rev Gastroenterol Hepatol. 2016;10(1):9–11.
phy utilization in high-volume liver transplantation 168. Bruns H, et al. Early markers of reperfusion injury
centers in the United States. J Cardiothorac Vasc after liver transplantation: association with pri-
Anesth. 2008;22(6):811–3. mary dysfunction. Hepatobiliary Pancreat Dis Int.
153. Soong W, et al. United States practice patterns in 2015;14(3):246–52.
the use of transesophageal echocardiography dur- 169.
Garcia-Criado A, et al. Doppler ultrasound
ing adult liver transplantation. J Cardiothorac Vasc findings in the hepatic artery shortly after
Anesth. 2014;28(3):635–9. liver transplantation. Am J Roentgenol.
154. De Wolf A. Transesophageal echocardiography and 2009;193(1):128–35.
orthotopic liver transplantation: general concepts. 170. Feng AC, et al. Hepatic hemodynamic changes
Liver Transpl Surg. 1999;5(4):339–40. during liver transplantation: a review. World J
155. Burger-Klepp U, et al. Transesophageal echocar- Gastroenterol. 2014;20(32):11131–41.
diography during orthotopic liver transplantation in 171. Bueno J, et al. Intraoperative flow measurement
patients with esophageal varices. Transplantation. of native liver and allograft during orthotopic
2012;94(2):192–6. liver transplantation in children. Transplant Proc.
156. Markin NW, et al. The safety of transesophageal 2007;39(7):2278–9.
echocardiography in patients undergoing orthotopic 172. Jamieson LH, et al. Doppler ultrasound velocities
liver transplantation. J Cardiothorac Vasc Anesth. and resistive indexes immediately after pediatric
2015;29(3):588–93. transplantation: normal ranges and predictors of fail-
157. Feierman D. Case presentation: transesophageal ure. Am J Roentgenol. 2014;203(1):W110–6.
echocardiography during orthotopic liver transplan- 173. Kaneko J, et al. Implantable Doppler probe
tation—not only a different diagnosis, but different for continuous monitoring of blood flow after
management. Liver Transpl Surg. 1999;5(4):340–1. liver transplantation. Hepato-Gastroenterology.
158. Ellenberger C, et al. Cardiovascular collapse due 2005;52(61):194–6.
to massive pulmonary thromboembolism dur- 174. Aki TJ, et al. Wireless monitor of liver hemodynam-
ing orthotopic liver transplantation. J Clin Anesth. ics in vivo. PLoS One. 2014;9(7):e102396.
2006;18(5):367–71. 175. Schutz W, et al. Is it feasible to monitor total hepatic
159. Rosendal C, et al. Right ventricular function during blood flow by use of transesophageal echography?
orthotopic liver transplantation: three-dimensional An experimental study in pigs. Intensive Care Med.
transesophageal echocardiography and thermodilu- 2001;27(3):580–5.
tion. Ann Transplant. 2012;17(1):21–30. 176. Kortgen A, et al. Prospective assessment of hepatic
160. Planinsic RM, et al. Diagnosis and treatment of function and mechanisms of dysfunction in the criti-
intracardiac thrombosis during orthotopic liver cally ill. Shock. 2009;32(4):358–65.
transplantation. Anesth Analg. 2004;99(2):353–6; 177. Chan RW, et al. The potential clinical utility of serial
table of contents plasma albumin mRNA monitoring for the post-­
11  Intraoperative Monitoring 147

liver transplantation management. Clin Biochem. 181. Oellerich M, Armstrong VW. The MEGX test: a
2013;46(15):1313–9. tool for the real-time assessment of hepatic function.
178. Schlegel A, Kron P, Dutkowski P. Hypothermic Ther Drug Monit. 2001;23(2):81–92.
machine perfusion in liver transplantation. Curr 182. Levesque E, et al. Plasma disappearance rate of
Opin Organ Transplant. 2016;21(3):308–14. indocyanine green: a tool to evaluate early graft
179. Karangwa SA, et al. Machine perfusion of donor outcome after liver transplantation. Liver Transpl.
livers for transplantation: a proposal for standard- 2009;15(10):1358–64.
ized nomenclature and reporting guidelines. Am J 183. Kortgen A, et al. Vasoactive mediators in patients
Transplant. 2016;16(10):2932–42. with acute liver failure treated with albumin dialysis.
180. Verhoeven CJ, et al. Biomarkers to assess graft qual- Liver Int. 2010;30(4):634–6.
ity during conventional and machine preservation in
liver transplantation. J Hepatol. 2014;61:672–84.
Evidence for Anesthetic Practice
in Liver Transplant Anesthesiology 12
Ryan M. Chadha

Introduction Keywords
Liver transplantation remains one of the most Randomized clinical trial · Pulmonary artery
intricate surgical procedures. Due to the com- catheters · Transesophageal echocardiography
plex involvement of all organ systems in end- · Viscoelastic testing · Coagulation manage-
stage liver disease, it requires a plethora of ment · Renal replacement therapy
invasive monitors, the full complement of car-
diac and pharmacological agents, and constant
communication with the surgeon. Since the
first successful liver transplants in the late Pulmonary Artery Catheters
1960s there has been significant advancement
in surgical technique, donor selection and Since the development of the Swan-Ganz catheter
anesthetic management. In the current era, in 1970, it has been considered to be the standard
good outcomes with long-term graft function for assessing cardiac hemodynamics and function
are standard and to be expected in most cases. in critically ill patients. Advances in the technol-
With an increasing number of liver trans- ogy of the pulmonary artery catheter allows for
plant centers in the United States, identifying continuous measurement of cardiac output and
interventions that improve outcome are criti- mixed venous oxygen saturation. In recent years,
cal. This chapter aims to examine the evi- there has been significant controversy over the
dence, if existent, of common therapies and utility of these catheters, as definite mortality
interventions in liver transplant anesthesiol- benefits have not been shown and some studies
ogy. We will evaluate five intraoperative even suggest increased mortality with its use.
interventions: p­ulmonary artery catheters,
­ Despite this, pulmonary artery catheters remain
transesophageal echocardiography, viscoelas- commonplace in liver transplantation and are
tic testing for coagulation management, intra- used in about 50% of centers internationally.
operative continuous renal replacement There is a scarcity of data regarding the use of
therapy and early extubation. pulmonary artery catheters to assess cardiac func-
tion and manage volume during liver transplan-
tation. Several studies in liver transplant patients
have shown that elevated intrathoracic pressures,
R. M. Chadha, MD
impaired contractility and valvular pathologies
Department of Anesthesiology and Perioperative
Medicine, Mayo Clinic Florida, Jacksonville, grossly affect the pressure measured for a given
FL, USA preload. As a result, static pressure measurements

© Springer International Publishing AG, part of Springer Nature 2018 149


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_12
150 R. M. Chadha

are an unreliable indicator of volume [1, 2]. Costa Cardiovascular Anesthesiologists state that “The
et al. found that stroke volume index correlated consultants agree although the ASA members are
poorly with central venous pressure (CVP) and equivocal regarding the use of TEE during open
pulmonary artery occlusion pressure (PAOP) abdominal aortic procedures and liver transplanta-
[3] and Rocca et al. found that cardiac index did tion” [11]. However, a 2008 survey of high volume
not correlate well with the CVP and PAOP dur- centers in the United States reported that only 13%
ing the dissection, anhepatic and post reperfusion of transplant anesthesiologists routinely use TEE,
phases [4] In addition, some studies suggest that whereas the majority (72%) reserve it for special
pulse wave analysis using the arterial pulse wave situations and rescue settings [12]. Suriani [13]
to estimate cardiac output is more accurate than evaluated the results of the first 100 liver trans-
thermodilution measurements using a pulmonary plants at his institution performed with TEE and
artery. catheter [5]. Greim et al. demonstrated found that intraoperative TEE had an impact on the
that a pulmonary artery catheter with continuous management in 64% patients; in 11% of patients
cardiac output capabilities can accurately mea- TEE had a major impact and in 48% TEE had
sure cardiac output during liver transplantation minor impact in patient management. A major
[6] except during the caval cross-clamping and impact was defined as TEE providing information
reperfusion phases. Beyond its ability to measure that allowed for the treatment of a life-threatening
cardiac output the pulmonary artery catheter is event or if it altered the surgical technique. A minor
required to diagnose portopulmonary hyperten- impact was defined as any finding that changed
sion in patients undergoing liver transplantation. pharmacological management or if the TEE was
Mean Pulmonary artery pressures (PAP) greater used as the primary form of cardiac monitoring
than 50 mmHg in a liver transplant recipient are during the case. Likewise, in a prospective study by
associated with an unacceptable high mortality. Hofer et al. [14], fluid therapy was significantly
Even moderate to severe portopulmonary hyper- influenced by echocardiographic findings in 50%
tension (mean PAP >35-45 mmHg) substantially of patients during liver and lung transplantation.
increases mortality. [7]. However, preoperative Ellis et al. [15] performed TEE to clarify the mech-
treatment of portopulmonary hypertension with anism of myocardial dysfunction that accompanies
a variety of pulmonary vasodilatory medications OLT. Based upon TEE findings, the authors
has led to significant reductions in perioperative reported that isolated right ventricular failure sec-
mortality [8, 9]. It is crucial that severe pulmonary ondary to paradoxical emboli may contribute to the
hypertension is diagnosed prior to or during the hemodynamic instability seen during OLT. Shillcutt
anesthetic induction to allow for cancellation and et al. [16] did a retrospective review of 100 TEE
possible use of a back-up donor [10]. Therefore, performed during liver transplantation and found
while literature indicates that pulmonary artery that intraoperative findings of intracardiac throm-
catheter placement may not be necessary as a bosis or biventricular dysfunction detected by TEE
routine monitor for the anesthesiologist during were predictive of short-term and long-term car-
liver transplantation, it should be considered a diac complications. While routine TEE use may
standard of care in patients with portopulmonary not be warranted yet, evidence suggests to have
hypertension. TEE immediately available should complications
arise during surgery liver transplantation.

Transesophageal Echocardiography
 iscoelastic Testing for Coagulation
V
Transesophageal echocardiography (TEE) can Management
facilitate cardiac and hemodynamic management
during liver transplantation. Practice guidelines The conventional tests of coagulation are platelet
published in 2010 by the American Society of count, prothombin time or international normal-
Anesthesiologists (ASA) and the Society of ized ratio, and fibrinogen levels. While they do
12  Evidence for Anesthetic Practice in Liver Transplant Anesthesiology 151

assess a (limited) part of the coagulation sys- ing placement of a large-bore central venous
tem, they are very poor predictors of bleeding catheter, exposure to an extracorporeal circuit,
tendency, transfusion requirements, and active hypothermia, hypocalcemia, and the possible
in-­vivo clot formation. As a result, anesthesiolo- need for anticoagulation, it has been questioned
gists are attempting to utilize viscoelastic test- whether it is beneficial in the intraoperative
ing, either thromboelastography (TEG) and/or arena. Few studies have examined intraoperative
rotational thromboelastometry (ROTEM), to bet- continuous renal replacement therapy with vary-
ter assess clot formation and strength and guide ing results. A 2011 study reviewed 72 patients
administration of blood products intraoperatively. and found no difference in complications or
Some evidence supports the use of viscoelas- mortality in liver transplant patients chosen
tic testing in liver transplantation. As early as to have intraoperative CRRT [21] and another
1985, Starzl et al. [17] showed that transfusion study showed intraoperative CRRT did not affect
practice guided by TEG resulted in a 33% intraoperative transfusion requirements or inten-
decreased use of blood products; however, this sive care unit (ICU) and hospital lengths of stay.
was during an era with significantly higher intra- In this study, all patients were weaned off renal
operative transfusion requirements. A 2010 study replacement therapy and 1-year patient survival
[18] showed a decreased transfusion rate of fresh was 86% for intraoperative CRRT compared to
frozen plasma in patients monitored by TEG, 71% without, which was not found to be sig-
although differences in overall blood transfused nificant [22]. Townsend et al. retrospectively
and 3-year survival were not significantly differ- reviewed all liver transplants at their institu-
ent. Conversely, a 2014 study [19] using a tion over 10 years and found that intraoperative
ROTEM based algorithm showed no statistically CRRT was used 6.4% of the time, and those
significant differences in the amount of blood cases had a survival of 97.6% at 1 month and
transfused. In addition, viscoelastic testing may 75.6% at 1 year [23]. The scarcity of studies on
have utility by detecting hypercoagulability in this topic do not allow for a definitive recom-
liver disease. Krzanicki et al. showed that throm- mendation on its usage intraoperatively during
boelastography may be useful in detecting hyper- orthotopic liver transplantation. Unfortunateley
coagulability, although his results were not recently a phase II randomized controlled trial of
significant [20]. Further studies will be required Intra-Operative Continuous Renal Replacement
to assess the overall benefit of this testing in liver Therapy in Liver Transplantation (INCEPTION
transplantation. trial) was terminated due to a lack of funding
(clinicaltrials.gov NCT01575015).

I ntraoperative Renal Replacement


Therapy Early Extubation

In the critically ill patient with acute or chronic Liver transplantation outcomes have improved
liver disease, acute kidney injury and hepatore- greatly over the past decade. In the past at the con-
nal syndrome can develop and the number of clusion of the liver transplant procedure, all liver
patients undergoing liver transplants with kidney transplant recipients would be transported to the
disease has increased since the introduction of intensive care unit (ICU) intubated and then post-
the MELD score for graft allocation. Continuous operatively weaned off the ventilator and extu-
renal replacement therapy (CRRT) allows for bated depending on their hemodynamic stability
constant dialysis with improved hemodynamic and metabolic, neurological and respiratory status.
tolerance, reduced risk of exacerbation of cere- However, some institutions have moved towards a
bral edema, and superior metabolic, acid–base, “fast-track” practice with the goal of early extuba-
and azotemic control. However, due to the cost tion in the operating room or immediately on pre-
and risk associated with this intervention includ- sentation to the ICU.
152 R. M. Chadha

An examination of the data has validated the catheter monitoring during anesthesia for liver trans-
plantation. Transplant Proc. 2001;33:1394–6.
safety of early extubation. Extubation in the
3. Costa MG, Chiarandini P, Della Rocca G.
operating room (OR) can be successfully per- Hemodynamics during liver transplantation.
formed in a large fraction of patients without an Transplant Proc. 2007;39:1871–3.
increased risk of subsequent reintubation [24, 4. Rocca GD, Costa MG, Feltracco P, Biancofiore G,
Begliomini B, Taddei S, Coccia C, Pompei L, Di
25]. A 2007 multicenter study showed only a
Marco P, Pietropaoli P. Continuous right ventricular
7.7% complication rate in 361 patients who were end diastolic volume and right ventricular ejection
extubated in the operating room [26]. fraction during liver transplantation: a multicenter
Furthermore, a 2002 study showed that a practice study. Liver Transpl. 2008;14:327–32.
5. Matthieu B, Karine N, Vincent C, Alain V, Francois C,
of intraoperative extubation with transfer directly
Phillipe R, Francois S. Cardiac output measurement
to the surgical ward decreased costs by eliminat- in patients undergoing liver transplantation: pulmo-
ing the ICU stay entirely [27, 28]. Finally, several nary artery catheter versus uncalibrated arterial pres-
studies have established higher survival rates in sure waveform analysis. IARS. 2008;106:1480–6.
6. Greim CA, Roewer N, Thiel H, Laux G, Schulte am Esch
patients who have decreased postoperative
J. Continuous cardiac output monitoring during adult
mechanical ventilation, including one study liver transplantation: thermal filament technique versus
showing a 1% mortality rate in patients that were bolus thermodilution. Anesth Analg. 1997;85:483–8.
extubated immediately after transplantation com- 7. Böttiger BW, Sinner B, Motsch J, Bach A, Bauer H,
Martin E. Continuous versus intermittent thermodilu-
pared to 4% in patients that were extubated
tion cardiac output measurement during orthotopic
within 24 h [29]. Based on the review of the evi- liver transplantation. Anaesthesia. 1997;52:207–14.
dence, early extubation is a safe practice and can 8. Krowka M, Plevak D, Findlay J, Rosen C, Wiesner
be practiced routinely at all institutions. R, Krom R. Pulmonary hemodynamics and periop-
erative cardiopulmonary-related mortality in patients
with portopulmonary hypertenstion undergoing liver
Conclusion transplantation. Liver Transpl. 2000;6:443–50.
There is very little evidence for specific inter- 9. Sussman N, Kaza V, Barshes N, Stribling R, Goss J,
ventions in liver transplant anesthesiology. We O’Mahony C, Zhang E, Vierling J, Frost A. Successful
liver transplantation following medical management
will need additional randomized controlled
of portopulmonary hypertension: a single-center
studies to assess their overall effect and use- series. Am J Transplant. 2006;6:2177–82.
fulness in the perioperative period. Although 10. Ramsay M. Portopulmonary hypertension and right
early extubation and TEE have been shown to heart failure in patients with cirrhosis. Curr Opin
Anaesthesiol. 2010;23:145–50.
be beneficial, their lack of routine application
11. American Society of Anesthesiologists and Society
during liver transplantation is a reminder of the of Cardiovascular Anesthesiologists Task Force
need for further validation by the scientific and on Transesophageal Echocardiography. Practice
clinical community. Therefore, liver transplant guidelines for perioperative transesophageal echo-
cardiography. An updated report by the American
anesthesiologists will need to continue to rely
Society of Anesthesiologists and the Society of
on their clinical judgement and practice with Cardiovascular Anesthesiologists Task Force on
the understanding that evidence is needed Transesophageal Echocardiography. Anesthesiology.
to prove the benefit of their intraoperative 2010;112(5):1084–96.
12.
Wax D, Torres A, Scher D, Leibowitz A.
intervention.
Transesophageal echocardiography utilization in
high-volume liver transplantation centers in the United
States. J Cardiothorac Vasc Anesth. 2008;22:811–3.
13.
Suriani RJ, Cutrone A, Feierman D, et al.
References Intraoperative transesophageal echocardiography dur-
ing liver transplantation. J Cardiothorac Vasc Anesth.
1. Feltracco P, Biancofiore G, Ori C, Saner FH, Della 1996;10:699–707.
Rocca G. Limits and pitfalls of haemodynamic 14. Hofer CK, Zollinger A, Rak M, et al. Therapeutic
monitoring systems in liver transplantation surgery. impact of intra-operative transoesophageal echocar-
Minerva Anestesiol. 2012;78:1372–84. diography during noncardiac surgery. Anaesthesia.
2. Della Rocca G, Pompei L, Costa MG, Coccia C, 2004;59:3–9.
Rossi M, Berloco PM, Pietropaoli P, Cortesini R. 15. Ellis JE, Lichtor JL, Feinstein SB, et al. Right heart
Hemodynamic-volumetric versus pulmonary artery dysfunction, pulmonary embolism, and paradoxical
12  Evidence for Anesthetic Practice in Liver Transplant Anesthesiology 153

embolization during liver transplantation. A trans- 22. LaMattina JC, Kelly PJ, Hanish SI, Ottmann SE,

esophageal two-dimensional echocardiographic Powell JM, Hutson WR, Sivaraman V, Udekwu O,
study. Anesth Analg. 1989;68:777–82. Barth RN. Interoperative continuous veno-venous
16. Shillcutt S, Ringenberg K, Chacon M, Brakke T,
hemofiltration facilitates surgery in liver transplant
Montzingo C, Lyden E, Schulte T, Porter T, Lisco patients with acute renal failure. Transp Proced.
S. Liver transplantation: intraoperative transesopha- 2015;47:1901–4.
geal echocardiography findings and relationship 23. Townsend D, Bagshaw S, Jacka M, Bigam D, Cave D,
to major postoperative adverse cardiac events. J Gibney RT. Intraoperative renal support during liver
Cardiothorac Vasc Anesth. 2016;30:107–14. transplantation. Liver Transpl. 2009;15:73–9.
17. Kang Y, Martin D, Marquez J, Lewis J, Bontempo F, 24. Ulukaya S, Ayanoglu HO, Acar L, Tokat Y, Kilic
Shaw B, Starzl T, Winter P. Intraoperative changes M. Immediate tracheal extubation of the liver trans-
in blood coagulation and thrombelastographic plant recipients in the operating room. Transp Proced.
monitoring in liver transplantation. Anesth Analg. 2002;34:3334–5.
1985;64:888–96. 25. Biancofiore G, Bindi ML, Romanelli AM, et al. Very
18. Trzebicki J, Flakiewicz E, Kosieradzki M, et al. The early tracheal extubation without predetermined cri-
use of thromboelastometry in the assessment of hemo- teria in a liver transplant recipient population. Liver
stasis during orthotopic liver transplantation reduces Transpl. 2001;7(9):777–82.
the demand for blood products. Ann Transplant. 26. Mandell S, Stoner T, Barnett R, et al. A multicenter
2010;15:19–24. evaluation of safety of early extubation in liver trans-
19. Roullet S, Freyburger G, Cruc M, et al. Management plant recipients. Liver Transpl. 2007;13:1557–63.
of bleeding and transfusion during liver transplanta- 27. Mandell S, Lezotte D, Kam I, Zamudio S. Reduced use
tion before and after the introduction of a ROTEM of intensive care after liver transplantation: influence
-based algorithm. Liver Transpl. 2015;21:169–79. of early extubation. Liver Transpl. 2002;8:676–81.
20. Krzanicki D, Sugavanam A, Mallett S. Intraoperative 28. Mandell S, Lockrem J, Kelley S. Immediate tracheal
hyperrcoagulability during liver transplantation as extubation after liver transplantation: experience
demonstrated by thromboelastography. Liver Transpl. of two transplant centers. Anesth Anal. 1997;84:
2013;19:852–61. 249–53.
21. Parmar A, Bigam A, Meeberg G, Cave D, Townsend 29. Biancofiore G, Bindi ML, Romanelli AM, et al. Fast
D, Gibney RT, Bagshaw S. An evaluation of intraop- track in liver transplantation: 5 years’ experience. Eur
erative renal support during liver transplantation: a J Anaesthesiol. 2005;22:584–90.
matched cohort study. Blood Purif. 2011;32:238–48.
Caval Cross-Clamping, Piggyback
and Veno-Venous Bypass 13
David Hovord, Ruairi Moulding, and Paul Picton

Keywords clamps are placed with one below the diaphragm


Caval clamp · Bypass circuit · Piggyback · and one above the renal veins. Resection of the
Blood flow · Anhepatic phase · Reperfusion recipient’s vena cava is achieved by dividing both
the infra- and supra-hepatic vena cava.
Transplantation of the donor organ therefore
Introduction requires both supra and infra-hepatic caval anas-
tomoses and complete caval occlusion occurs
Liver transplantation has historically been associated during the vast majority of the anhepatic phase.
with massive blood transfusion and major hemor-
rhage was considered routine. Surgical techniques
have evolved in an attempt to reduce blood loss and  hysiologic Effects of the Caval
P
reduce transfusion requirements. In this chapter we Cross-Clamp
review the physiologic effects of caval cross-clamp-
ing and explore surgical options to safely establish The physiologic effects of caval cross-clamping
hepatectomy and transplantation including piggy- can be described by considering the effect on
back technique and venovenous bypass (VVB). each major organ system (Table 13.1).

Cardiovascular System
Caval Cross-Clamping Patients with end-stage liver failure typically
have a hyperdynamic circulation, demonstrated
The aim of caval cross-clamping is to elimi- by high cardiac output and low systemic vascular
nate hepatic outflow prior to hepatectomy. resistance (SVR). This may co-exist with a low
Traditionally two inferior vena cava (IVC) cross central blood volume [1, 2]. Application of the
IVC cross-clamp results in a large decrease in
venous return and a reduction of pulmonary
D. Hovord, BA, MB, BChir, FRCA
P. Picton, MB, ChB, MRCP, FRCA (*) wedge pressure. Cardiac output is reduced by up
Department of Anesthesiology, University of to 50% [3]. The reduction in venous return
Michigan Medical School, Ann Arbor, MI, USA depends on the pre-clamp volemic status of the
e-mail: davidhov@med.umich.edu
patient, and the extent to which collateral circula-
ppicton@med.umich.edu
tion has developed. Despite a large decrease in
R. Moulding, BSc, MBBS, FRCA
cardiac output, the effect on systemic blood pres-
Department of Anaesthesia, Musgrove Park Hospital,
Taunton, UK sure (BP) is variable. It is usually possible to
e-mail: ruari.moulding@tst.nhs.uk maintain BP at acceptable levels by increasing

© Springer International Publishing AG, part of Springer Nature 2018 155


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_13
156 D. Hovord et al.

Table 13.1  Physiological effects of caval most centers would consider this adequate to
cross-clamping
continue without the use of VVB.
Cardiac ↓↓Venous return, ↓↓cardiac output,
↓mean blood pressure, ↑heart rate Pulmonary System
↑systemic vascular resistance,
The pulmonary effects of caval clamping are also
Renal ↓Perfusion pressure, ↓↓venous renal
outflow dependent on the acuity of the patients’ disease
GI ↑Venous congestion, ↓↓portal venous [7]. Caval clamping in patients with acute liver
flow disease results in a more profound deterioration
Respiratory ↓Pulmonary capillary wedge pressure, in mixed venous oxygen saturations than in
↑pulmonary vascular resistance, chronic liver disease and a more persistent eleva-
↓mixed venous oxygen tension,
↓pulmonary venous oxygen tension
tion in pulmonary vascular resistance. The rea-
Neurologic ↓Cerebral perfusion pressure
sons for this are unclear; however, patients with
acute liver failure lack the porto-systemic shunts
GI Gastro-intestinal
↓—decreased seen in chronic liver disease. The pulmonary
↓↓—severely decreased effects are transient and normally reverse with
↑—increased the release of the caval clamp.

SVR and to a lesser extent heart rate. The ability Renal System
to compensate is reduced with prolonged clamp As with the wider surgical population, the devel-
time and pre-existing cardiac dysfunction. End-­ opment of postoperative renal failure requiring
stage liver disease may cause depressed barore- renal replacement therapy (RRT) results in a sig-
ceptor sensitivity, and therefore normal nificant increase in mortality following liver
physiologic responses to change in BP and pulse transplantation [8, 9]. Pre-existing renal dysfunc-
may be blunted, similar to patients with dysauto- tion increases the risk of post-operative dysfunc-
nomia. Such patients are unable to intrinsically tion to the degree that a combined liver-kidney
compensate for hemodynamic changes caused by transplant procedure may be mandated [10].
caval cross-clamping or major hemorrhage [4]. Caval clamping reduces overall blood flow to the
Patients with chronic end stage liver failure kidney initially by reducing inflow via its effects
usually tolerate caval clamping with less hemody- on MAP and cardiac output, but also obstructs
namic disturbance than patients with acute hepatic renal venous outflow. Severe renal injury may be
failure. This is due to the development over time precipitated even if renal artery perfusion is
of collateral circulation, mainly via the azygous maintained [11].
system. The presence of Caput Medusa—tortuous Optimal surgical conditions are frequently
venous collaterals radiating from the umbilicus— obtained by reducing CVP in order to reduce
and esophageal varices are indicators that an bleeding and as a consequence transfusion
extensive collateral circulation has developed. requirement during hepatic dissection. This is in
The use of a caval clamp trial has been conflict to some extent to the optimum CVP and
employed to predict the need for VVB. If cardiac cardica output required to optimize renal perfu-
output fell by more than 50%, then VVB would sion. An approach that targets a low CVP through
be necessary. However, this approach has not fluid restriction, vasopressors and/or phlebotomy
been shown to reduce morbidity or mortality [5] has in some studies improved long term survival
and not better than clinical judgement. The only rates and increased the rate of transfusion free
hemodynamic parameters shown to be indepen- liver transplant without increasing the incidence
dently associated with poor surgical outcome are of post-operative renal failure [12]. However,
severe hypotension mean arterial pressure (MAP) when two approaches—low CVP (<5 mmHg) and
<40 or pulmonary artery pressure >40 mmHg [6]. normal CVP (5–10 mmHg)—were directly com-
Therefore, if systemic blood pressure can be pared [8], the low CVP group had reduced blood
maintained after caval clamp placement then product use, but peak creatinine, 30 day mortality
13  Caval Cross-Clamping, Piggyback and Veno-Venous Bypass 157

and the incidence or RRT were all increased com- cerebral edema and raised intracranial pressure
pared to normal CVP. The correct CVP is likely to [17]. The use of vasoconstrictors during caval
be low, but not too low! The precise volume status clamping to maintain MAP is usually adequate to
and MAP at which the kidneys become compro- maintain cerebral perfusion pressure in these
mised to negatively affect survival are yet to be patients [14].
determined and likely to depend on a number of
individual patient and surgical factors.
The Piggyback Technique
Gastrointestinal System
Application of a caval clamp leads to venous con- Initially described by Calne in 1968, piggyback
gestion of the gastrointestinal (GI) tract. The liver transplantation was not widely used until
engorgement of the splanchnic beds causes bleed- Tzakis described a case series in 1988 [18]. The
ing during hepatic dissection and may also lead to piggyback technique aims to preserve the native
bacterial translocation and endotoxin release [13, retrohepatic IVC and avoids complete caval
14]. Venous congestion also leads to bowel edema, clamping (Fig. 13.1).
postoperative ileus, bile leak and cholestasis [15]. This is achieved by identifying and occluding
The physiologic response to hypovolemia may the hepatic veins with a partially occlusive caval
also reduce blood flow to GI tract, resulting in clamp. The piggyback technique helps to main-
intestinal hypoperfusion. Goal-directed therapy tain IVC blood flow, and reduce the decrease in
has been advocated to help improve postoperative cardiac output seen with a complete IVC clamp.
morbidity in a general surgical population. The portal veins are still occluded with this tech-
However the applicability of goal-directed therapy nique, and therefore splanchnic venous conges-
to a liver transplant population is not clear [16]. tion still occurs. A temporary portocaval shunt
can be placed that has the dual effect of reducing
Neurologic System intestinal congestion and increasing venous
A caval clamp that reduces MAP will also return to the heart. Depending on the patients’
decrease cerebral perfusion pressure. This is of anatomy and surgical placement of the cross
critical importance to the subgroup of liver trans- clamp, the partial caval clamp can still occlude
plant patients with fulminant disease who have most if not all of the IVC and flow can completely

Fig. 13.1 Conventional a
caval clamp vs. Piggyback
technique. (a) The Diaphragm Donor Liver
conventional technique of
orthotopic liver
transplantation requires
complete caval cross- Head
clamp and two cavo-caval
anastomoses. (b) The
piggyback technique
preserves the recipient
retrohepatic IVC and b
avoids the caval cross-
clamp and anastomoses.
The inferior portion of the
donor IVC is sutured
closed and the upper
portion is anastomosed to
the native IVC via the
recipient hepatic vein
stump Recipient IVC
158 D. Hovord et al.

cease. Therefore it is important to understand that and a wealth of retrospective data available it is
although less deleterious than a full clamp, the unlikely that a sufficiently powered randomized
partial caval clamp is a not a benign intervention controlled trial will be performed in the future.
and must be carefully handled (Fig. 13.2). Hepatic outflow obstruction has been the
The piggyback technique has been extensively major flaw in the piggyback technique. A modifi-
studied since its widespread uptake and can be cation by Belghiti in 1992 [27] introduced a side-­
used in nearly all liver transplantations and nearly to-­side cavocavostomy and exclusion of the right
all re-transplantations [19–24]. It is considered to hepatic vein has reduced the incidence of this
be associated with less bleeding, lower transfu- complication (Table 13.2).
sion requirements, reduced warm ischemia time
and reduced hospital and ICU length of stay.
Table 13.2  The potential advantages of the piggyback
During piggyback liver transplantation renal per-
technique for orthotopic liver transplantation
fusion pressure can more easily be maintained
Piggyback advantages
towards normal and renal injury is decreased
• Avoids caval reconstruction
[11]. It also reduces the need for VVB, which
• Avoids caval cross-clamping and maintains venous
reduces staffing and equipment costs [25]. return
Most of the evidence supporting the piggy- • Avoids venous renal outflow obstruction
back technique comes from retrospective analy- • Less post-operative renal dysfunction
sis. A Cochrane review reported just three small • Reduced warm ischemic time
randomized controlled trials, all at high risk of • Reduced staffing and equipment costs
bias [26]. These trials showed a much smaller dif- • Reduced blood product use
ference in outcome, with only a reduction in • Reduced bleeding
warm ischemic time with piggyback technique • No complement activation via VVB
when compared to the bi-caval approach. Since • Shorter hospital stay
the piggyback technique is now so widely used • Shorter ICU stay

Satinsky clamp
Recipient hepatic
Piggyback vein stump
anastomosis

Donor
vena cava

Recipient
vena cava

FA
er, M
ch
Fis
HR

Normal flow Reduced flow

Fig. 13.2  Figure illustrating Piggyback clamp placement with normal flow and with reduced flow as may occur espe-
cially during periods of manipulation
13  Caval Cross-Clamping, Piggyback and Veno-Venous Bypass 159

Venovenous Bypass of circuit led to a high incidence of embolic


events. A pump was added to the circuit by Calne
The goal of VVB is to return venous blood from in 1979. However this system required systemic
below the diaphragm to the heart via an extra-­ heparinization, resulting in the potential for mas-
corporeal circuit. This allows improved hemody- sive hemorrhage. The addition of heparin-coated
namic stability during caval cross-clamping. tubing [28] reduced the incidence of hemorrhagic
They key to understanding whether this is benefi- complications (Fig. 13.3). Early studies showed
cial is whether the risk of introducing an extra-­ that adding VVB to a complete caval clamp
corporeal circuit is lower than the complications reduced the post-operative requirement for post-
it is intended to overcome. Initially VVB was operative dialysis [29]. A further randomized
achieved with a passive connection from femoral controlled trail of 77 patients failed to show any
or portal veins to one of the major supra-dia- difference [30].
phragmatic vessels (axillary/subclavian/internal VVB was also thought to reduce blood loss,
jugular) but the low flow rates through this type probably by reducing venous congestion.

Axillary Vein

SVC

Suprahepatic
IVC Inflow

Portal
Infrahepatic Vein
IVC

Venous
Inflow

Femoral
Vein
Pump

Fig. 13.3  Veno-venous bypass circuit


160 D. Hovord et al.

However the addition of an extracorporeal circuit References


leads to platelet activation and hemolysis as the
blood travels through the pump and circuit. 1. Costa MG, Chiarandini P, Della Rocca G.
Hemodynamics during liver transplantation.
Further studies have shown that this effect may Transplant Proc. 2007;39(6):1871–3.
outweigh the benefits of VVB and lead to an 2. Henriksen JH, et al. Reduced central blood volume in
increase in transfusion requirements [14, 19]. cirrhosis. Gastroenterology. 1989;97(6):1506–13.
There is also conflicting evidence on the effect of 3. Carmichael FJ, Lindop MJ, Farman JV. Anesthesia
for hepatic transplantation: cardiovascular and met-
VVB on ICP. Theoretically the increased venous abolic alterations and their management. Anesth
return from the circuit should make it easier to Analg. 1985;64(2):108–16.
maintain cerebral perfusion pressure (CPP), 4. Lhuillier F, et al. Spontaneous baroreflex cardiac sen-
which is especially important in patients with ful- sitivity in end-stage liver disease: effect of liver trans-
plantation. Eur J Anaesthesiol. 2006;23(5):426–32.
minant hepatic failure. However VVB may cause 5. Schwarz B, et al. Liver transplantation without veno-
an increase in PaCO2, therefore increasing ICP venous bypass: morbidity and mortality in patients
via cerebral vasodilatation [24]. VVB may cause with greater than 50% reduction in cardiac output
hypothermia [31] or may correct it if a heat after vena cava clamping. J Cardiothorac Vasc Anesth.
2001;15(4):460–2.
exchanger is added to the system [32]. 6. Reich DL, et al. Association of intraoperative hypo-
The incidence of complications attributable tension and pulmonary hypertension with adverse
to VVB ranges from 10 to 30%. When placing outcomes after orthotopic liver transplantation. J
the cannulae, complications include hemor- Cardiothorac Vasc Anesth. 2003;17(6):699–702.
7. Valta P, et al. Pulmonary effects of caval clamp-
rhage, hematoma formation, lymphocele and ing during liver transplantation without venovenous
brachial plexus injury (axillary vein). bypass in acute or chronic liver failure. Transplant
Inadvertent decannulation may occur, as well Proc. 2001;33(4):2521–2.
as thrombus formation resulting in pulmonary 8. Schroeder RA, et al. Intraoperative fluid management
during orthotopic liver transplantation. J Cardiothorac
embolus, air embolus and complement activa- Vasc Anesth. 2004;18(4):438–41.
tion that can lead to tissue injury and increasing 9. O'Riordan A, et al. Acute renal disease, as defined by
risk of post-operative multi-organ failure [33]. the RIFLE criteria, post-liver transplantation. Am J
The perception that VVB is advantageous Transplant. 2007;7(1):168–76.
10. Pham PTT, Pharn PCT, Wilkinson AH. Renal func-
have waned with the widespread uptake of the tion outcomes following liver transplantation and
piggyback technique as complications associated combined liver-kidney transplantation. Nat Clin Pract
with its use are frequent and severe. Therefore, Nephrol. 2007;3(9):507–14.
there are currently no absolute indications for the 11. Niemann CU, et al. Acute kidney injury dur-

ing liver transplantation as determined by neutro-
routine use of VVB. There are some circum- phil gelatinase-associated lipocalin. Liver Transpl.
stances where its use should be considered. 2009;15(12):1852–60.
Patients with pulmonary hypertension or poor 12. Massicotte L, et al. Effect of low central venous pres-
left ventricular function may benefit, as they may sure and phlebotomy on blood product transfusion
requirements during liver transplantations. Liver
not tolerate even a partial caval clamp. Patients Transpl. 2006;12(1):117–23.
with severe portal hypertension and with anatom- 13. Abdala E, et al. Bacterial translocation during liver
ical reasons for difficult dissection may also ben- transplantation: a randomized trial comparing con-
efit, as may those with fulminant hepatic failure ventional with venovenous bypass vs. piggyback
methods. Liver Transpl. 2007;13(4):488–96.
due to the lack of pre-existing collateral venous 14. Reddy K, Mallett S, Peachey T. Venovenous bypass
return and the need to maintain CPP. VVB may in orthotopic liver transplantation: time for a rethink?
also simplify the hemodynamic management for Liver Transpl. 2005;11(7):741–9.
a less experienced anesthesiology team. The 15. Marcos A, et al. Functional venous anatomy for right-­
lobe grafting and techniques to optimize outflow.
majority of transplant centers no longer use VVB Liver Transpl. 2001;7(10):845–52.
routinely and reserve its use only for a small 16. Giglio MT, et al. Goal-directed haemodynamic ther-
number of selected cases. apy and gastrointestinal complications in major sur-
13  Caval Cross-Clamping, Piggyback and Veno-Venous Bypass 161

gery: a meta-analysis of randomized controlled trials. 25. Polak WG, Peeters PMJG, Slooff MJH. The evolution
Br J Anaesth. 2009;103(5):637–46. of surgical techniques in clinical liver transplantation.
17. Karvellas CJ, et al. Outcomes and complications of A review. Clin Transplant. 2009;23(4):546–64.
intracranial pressure monitoring in acute liver fail- 26. Gurusamy KS, Pamecha V, Davidson BR. Piggy-back
ure: a retrospective cohort study. Crit Care Med. graft for liver transplantation. Cochrane Database
2014;42(5):1157–67. Syst Rev. 2011;1:CD008258.
18. Tzakis A, Todo S, Starzl TE. Orthotopic liver trans- 27. Belghiti J, et al. A new technique of side to side caval
plantation with preservation of the inferior vena cava. anastomosis during orthotopic hepatic transplantation
Ann Surg. 1989;210(5):649–52. without inferior vena caval occlusion. Surg Gynecol
19. Lerut J, et al. Cavocaval adult liver transplantation Obstet. 1992;175(3):270–2.
and retransplantation without venovenous bypass and 28. Shaw BW, et al. Venous bypass in clinical liver-­

without portocaval shunting: a prospective feasibility transplantation. Ann Surg. 1984;200(4):524–34.
study in adult liver transplantation. Transplantation. 29. Shaw BW, et al. Advantages of venous bypass during
2003;75(10):1740–5. orthotopic transplantation of the liver. Semin Liver
20. Mehrabi A, et al. A single-center experience of 500 Dis. 1985;5(4):344–8.
liver transplants using the modified piggyback tech- 30. Grande L, et al. Effect of venovenous bypass on

nique by Belghiti. Liver Transpl. 2009;15(5):466–74. perioperative renal function in liver transplantation:
21. Belghiti J, et al. Feasibility and limits of caval-flow results of a randomized, controlled trial. Hepatology.
preservation during liver transplantation. Liver 1996;23(6):1418–28.
Transpl. 2001;7(11):983–7. 31. Hilmi IA, Planinsic RM. Con: venovenous bypass
22. Khan S, et al. Conventional versus piggyback tech- should not be used in orthotopic liver transplantation.
nique of caval implantation; without extra-corporeal J Cardiothorac Vasc Anesth. 2006;20(5):744–7.
veno-venous bypass. A comparative study. Transpl 32. Barnett R. Pro: veno-veno bypass should routinely be
Int. 2006;19(10):795–801. used during liver transplantation. J Cardiothorac Vasc
23. Nishida S, et al. Piggyback technique in adult ortho- Anesth. 2006;20(5):742–3.
topic liver transplantation: an analysis of 1067 33. Hoffmann K, et al. Is veno-venous bypass still needed
liver transplants at a single center. HPB (Oxford). during liver transplantation? A review of the litera-
2006;8(3):182–8. ture. Clin Transplant. 2009;23(1):1–8.
24. Fonouni H, et al. The need for venovenous bypass in
liver transplantation. Hpb. 2008;10(3):196–203.
Hemodynamic Changes, Cardiac
Output Monitoring and Inotropic 14
Support

Anand D. Padmakumar and Mark C. Bellamy

Keywords
diovascular changes at various stages of LT, modes
Hypotension · Vasopressors · Hemodynamic of hemodynamic monitoring, and use of inotropes
changes · Circulation · Vasodilation · and vasopressors.
Catecholamines

Cardiovascular Changes During LTx


Introduction
Physiological Considerations
Liver transplantation (LTx) poses distinct chal-
lenges to the anesthesiologist. Patients presenting To understand fully the hemodynamic changes
for LTx constitute a high-risk surgical group with during LTx, it is worth reviewing the physiological
unique problems and require meticulous attention principles of liver blood flow. Some of these topics
to their perioperative management. End-stage liver are discussed in more detail elsewhere in the book.
disease (ESLD) is the most common indication for In health, autoregulation smoothes out poten-
LTx and presents complex pathophysiological tially major changes in hepatic blood flow (HBF)
changes involving all organ systems. The severity and protects normal hepatic physiology and func-
of such changes varies enormously between cases. tion. The precise mechanisms that regulate HBF
A further level of complexity is seen in patients pre- are poorly understood. However, there are several
senting with decompensated ESLD and in those hypotheses to explaining intrinsic and extrinsic
presenting with acute hepatic failure. Cardiovascular, factors affecting hepatic flow [1]. The liver has lim-
respiratory, renal, neurological, gastrointestinal and ited inherent ability to control portal venous blood
inflammatory changes all interact to produce a com- flow (PBF); however multiple integrated processes
plex clinical picture. Portopulmonary hypertension, determine PBF, including anatomical and patho-
ascites, varices and dyselectrolytemia are some of logical changes altering portal vascular resistance.
the myriad problems associated with liver disease
that require special consideration before anesthetiz-
ing patients for LTx. In this chapter, we discuss car- Intrinsic Factors

Portal blood flow acts as a main intrinsic factor


A. D. Padmakumar, MBBS, FRCA, EDIC, FFICM
M. C. Bellamy, MA, MB, BS, FRCP(Edin), FRCA, regulating HBF. The hepatic arterial blood flow
FFICM (*) buffers any changes in PBF through the “hepatic
St. James’s University Hospital, Leeds Teaching artery buffer response” to maintain a constant total
Hospitals NHS Trust, Leeds, West Yorkshire, UK HBF. This buffer response seems to be indepen-
e-mail: m.c.bellamy@leeds.ac.uk

© Springer International Publishing AG, part of Springer Nature 2018 163


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_14
164 A. D. Padmakumar and M. C. Bellamy

dent of the metabolic demands of the liver [2]. gery, sepsis, etc. may induce or aggravate such
Myogenic and chemical mechanisms have been complications resulting in hepatorenal syndrome,
postulated to explain this mechanism. As in most variceal bleeding and circulatory failure [7].
other organs, the vascular resistance of the HA is
inversely proportional to blood flow and adenos-
ine plays a key role in the chemical autoregulation Pathogenic Mechanisms
of HBF. Sinusoidal adenosine concentrations,
determined largely by portal venous washout, are Liu et al. have reviewed the pathophysiological
inversely proportional to hepatic artery (HA) tone. processes contributing to the CVS changes in
Thus a reduction in PBF causes accumulation of liver disease [7]. The salient features are summa-
adenosine and ensuing local vasodilation of the rized in Table 14.1.
HA [3]. The liver also has a unique property of
matching its mass to the blood supply it receives
by either proliferation or apoptosis of hepatic cells Table 14.1  Proposed pathogenic mechanisms that con-
tribute to hemodynamic changes in liver disease
possibly mediated via portal flow dependent
growth factors. Adenosine furthermore activates Central Plays a vital role in development of
neural CVS changes in portal hypertension.
the hepatorenal reflex causing fluid retention [4]. activation Exact route of signaling from
periphery to central nervous system
remains unclear
Extrinsic Factors Endogenous Lipid-like substances, acting on G
cannabinoids protein-coupled receptors CB1 &
CB2, show negative inotropic effect
Animal experiments have revealed multiple (for example, Anandamide levels
extrinsic factors that regulate HBF including: increased in cirrhosis) and induce
apoptosis in hepatocytes. This could
• Sympathetic nervous system alter microcirculation and lead to
portal hypertension and hyperdynamic
• Catecholamines state
• Gastrointestinal hormones (secretin, gluca- Nitric oxide Changes in NO activity affect CVS in
gon, cholecystokinin, etc.) (NO) different ways. Increased systemic NO
• Autacoids (histamine, serotonin, bradykinin, production causes peripheral arterial
prostaglandins, etc.) vasodilation and negative inotropic
effect [8]. Cirrhotic rat models show
• Vasoconstrictor peptides (angiotensin-2 and reduced local expression of liver NO
vasopressin) synthase and a corresponding drop in
portal venous pressure [9]
Carbon Mainly produced by the action of heme
monoxide oxygenase (HO): activates soluble
Hemodynamic Changes
guanylate cyclase resulting in increased
levels of cGMP. There is association
Patients with ESLD demonstrate characteristic between elevated cGMP levels and
cardiovascular system (CVS) changes such as a heart failure in animal models of
hyperdynamic or hyperkinetic state secondary to a cirrhotic cardiomyopathy [10]
Beta-­ Expression and responsiveness of
reduction in systemic vascular resistance (SVR)
adrenergic beta-adrenergic receptors and
and a compensatory increase in cardiac output signaling post-receptor signaling pathways are
(CO) [5]. There may be a coexisting cirrhotic car- impaired at various levels in cirrhotic
diomyopathy particularly in alcoholic liver disease cardiomyopathy
(ALD), chronic portal and or pulmonary hyperten- Autacoids Various potent autacoids (bradykinin,
serotonin, histamine & prostaglandins)
sion, ascites, hypoproteinemia and dyselectrolyte- are less likely to play a significant role
mia. These CVS changes worsen as disease in systemic CVS changes due to their
progresses [6] and conditions inducing a neurohu- short half-life
moral stress response, for example trauma, sur- cGMP 3′, 5′ cyclic guanosine monophosphate
14  Liver Transplantation: Hemodynamic Changes, Cardiac Output Monitoring and Inotropic Support 165

The exact pathogenic mechanisms causing pulmonary artery catheter (PAC). For routine use
significant hemodynamic changes in the periop- in patients with previously good cardiac function
erative period of LTx, however, remain unclear. and no structural abnormality, pulse pressure or
pulse power analysis may be sufficient, for exam-
ple using pulse contour cardiac output (PiCCO™)
Measurement of Cardiac Output (CO) or lithium dilution cardiac output (LiDCO™)
systems [12]. In the authors’ institution use of
Although a full discussion of CO monitoring tech- LiDCO™ is standard, with PAC and TEE when
niques is discussed elsewhere in this book, it is indicated. The choice of montoring device should
important to understand their importance and limi- also consider the familiarity of the anesthesiol-
tations. Assessment of CO is important as it helps ogy tream with a specific technique.
guide fluid and inotrope management. Hypotension
may result from low vascular resistance, poor car-
diac contractility, reduced stroke volume or a com-  lassification of Inotropes
C
bination of these factors. (Relative) bradycardia and Vasopressors
may also contribute to low CO, and hence hypo-
tension even in the presence of adequate filling. An understanding of the specific pharmacology
This is particularly important in LTx as brady- of inotropes and vasopressors, and the (some-
cardic hypotension is frequently associated with times subtle) differences between them increases
high central venous pressure, which may compro- their utility during liver transplantation in situa-
mise the pressure gradient between the portal and tions of varying physiological patterns and
central venous systems, compromise graft blood derangements at various stages of the transplant
flow in the immediate post-reperfusion phase and procedure. The key attributes of commonly used
result in primary non-function. agents are summarised in Fig. 14.1.
Cardiac function may be further compromised Other agents with hemodynamic effect include
by pleural or pericardial effusions or pre-existing vasopressin and vasopressin analogues such as
pulmonary hypertension with right ventricular terlipressin and octreotide. These agents have
dysfunction. Furthermore, cardiac filling may be important effects on reducing portal pressure and
impaired by diastolic dysfunction, either irrevers- potentially limiting portal venous bleeding [13,
ible (for example as a result of an established 14] that can be of great value during the dissection
infarct with a fibrotic area), mechanically revers- phase of surgery. In addition, terlipressin and
ible (for example due to pericardial effusions), or vasopressin have a direct vasopressor effect
physiologically reversible (lusitropic and pseudo-­ through its action on vasopressin receptors [15],
lusitropic effects; for example secondary to the enhancing the effects of alpha-adrenergic agents.
effects of transfusion on anaemia-induced myo- This may be particularly valuable in patients with
cardial ischemia or due to ventricular septal shifts low vascular resistance, who may have exhausted
following “venodilatation”). pituitary stores of vasopressin and consequently
The method for CO monitoring selected show a reduced responsiveness to alpha-adrener-
should take account of the patient’s needs, the gic stimulation. This effect has been observed in
expected severity and nature of cardiovascular prolonged septic shock [16] and is also hypothi-
derangement and familiarity of the team. For sized as one cause of the vasodilatory state in liver
example patients at risk of microembolic phe- failure [17]. Vasopressin or its analogues can be
nomena at reperfusion, or patients thought to usefully during liver transplantation to maintain
have an inducible regional wall-motion abnor- vascular resistance and is commonly used in the
mality, or pericardial effusion, may be best moni- ­perioperative management of patients with hepa-
tored using trans-esophageal echocardiography toenal syndrome.
(TEE) [11] but the patient with pulmonary hyper- Calcium supplementation is also frequently
tension however may benefit from the use of a required during liver transplantation because the
166 A. D. Padmakumar and M. C. Bellamy

Fig. 14.1 Classification
of vasopressors and
inotropes. MAO • Direct acting – Selective & Non-selective
Monoamine oxidase, • Mixed acting
COMT Catechol-o-­ Adrenergic agonists
• Indirect acting – Releasing agents, uptake
methyl transferase, NO inhibitors, MAO/COMT inhibitors
Nitric oxide, LNMMA
l-NG-mono-methyl
arginine citrate

• Vasopressin analogs
• Nitric oxide antagonists
• LNMMA
Non-adrenergic agents
• Stromafree hemoglobin
• Methylene blue
• Calcium

concentration of ionized calcium in the circula- nary vasculature and should be used with caution
tion falls rapidly, particularly during the anhepatic in patients with pulmonary hypertension.
phase. This is due to chelation by citrate added to
blood products at a time when there is no meta-
bolic route for the elimination of citrate [18].  linical Features of Hemodynamic
C
Administration of calcium at this time, to main- Disturbance and Their Management
tain an ionized calcium value above 0.9 mmol per
liter, can have both a dramatic positive inotropic Pre-existing CVS changes in liver disease are
effect, a vasopressor effect, and is of value in further affected during induction and mainte-
maintaining normal perfusion pressure [19]. nance of anesthesia as intravenous and volatile
Free radical scavengers such as mannitol and agents frequently reduce CO and SVR. ESLD is
N-acetylcysteine have also been described as associated with low SVR that may decrease even
helping improve hemodynamic stability during further with induction of anesthesia. This is in
liver transplantation, particularly in the period part offset by an increase of CO that contributes
following graft reperfusion. Similar claims have to the “hyperdynamic state”. Increases of CO are
been made for aprotinin, a broad-spectrum serine a response to low SVR but the extent to which the
protease inhibitor that had been used for preven- CO can compensate for a low vascular resistance
tion of fibrinolysis and maintenance of clotting is further dependent on adequate ventricular fill-
[20]. Aprotinine is now not available anymore ing, venous return (dependent in part on vascular
because of increased mortality associated with its tone in capacitance vessels) and ventricular dia-
use in cardiac surgery. stolic function. In ESLD diastolic function may
Methylene blue has been used as an inhibitor of be abnormal due to alcoholic cardiomyopathy
the nitric oxide (NO) pathway and acts by inhibi- [22], pleural or pericardial effusions, or myocar-
tion of guanylate cyclase. Used as a bolus at the dial ischemia. Consequently, close physiological
time of reperfusion, it may increase blood pressure monitoring and an intelligent approach to multi-
but its overall effect on outcome is unclear [21]. modal cardiovascular manipulation are required.
The biological role of NO inhibition in sepsis is The nature and magnitude of these CVS changes
controversial as NO also appears to exert a protec- may necessitate intervention with fluids, inotro-
tive effect. Methylene blue can cause increased pes or vasopressor agents. The hemodynamic
pulmonary artery pressures due to inhibition of the changes during the various phases of LTx and
vasodilatory effect of intrinsic NO in the pulmo- their causes are summarized in Table 14.2.
14  Liver Transplantation: Hemodynamic Changes, Cardiac Output Monitoring and Inotropic Support 167

Table 14.2 Cardiovascular changes during various ably between patients. The principles of manage-
phases of liver transplantation (LT)
ment are maintenance of an adequate perfusion
Phase of LT CO Causes for change in CO pressure and hemodynamic optimization.
Dissection/ ↓ Hypovolemia, transient IVC Significant volume loading may be necessary to
pre-anhepatic compression, fluid shift with
achieve an optimal stroke volume. However, it is
ascitic decompression
↓ Reduced venous return due to
important also to pay attention to filling pressures
Anhepatic
clamping of PV & IVC, and electrolyte changes; excessive elevation of
acidosis filling pressure or PAP may both lead to reduced
Reperfusion/ ↑ Hyperkalemia, release of right ventricular performance and increased
neohepatic vasoactive substances, diuresis bleeding. For this reason, cardiovascular moni-
CO cardiac output toring is important at this stage, and the use of
inotropes or vasopressors may help mitigate
excessive fluid administration. Agents commonly
employed at this stage, both to help optimie
Intraoperative Hemodynamic stroke volume and to fine tune fluid administra-
Changes and Interventions tion, include norepinephrine, phenylephrine or
less commonly dopamine. The problems of the
 emodynamic Changes During
H dissection phase may be further exacerbated by
Dissection Phase portal hypertension and variceal bleeding. A log-
ical combined approach to the hyperdynamic
During the course of the surgical dissection state similar to sepsis and bleeding secondary to
(pre-­
anhepatic) phase further hemodynamic portal hypertension is the use of vasopressin or a
compromise may occur due to decompression suitable analogue. Vasopressin by infusion, terli-
of ascites, hemorrhage and gut translocation. pressin and octreotide have all been used in these
These issues are further exacerbated by lifting situations and they have the advantage of enhanc-
and rotation of the liver causing transient caval ing catecholamine sensitivity while at the same
compression. To allow removal of the native time promoting splanchnic vasoconstriction and
liver portal bypass as part of the veno-venous reducing portal hypertension. There may be an
bypass technique, complete cross-clamping of additional theoretical advantage in the reduction
portal vein and vena cave or piggyback tech- in portal flow around the time of graft reperfusion
nique with or without creation of a portocaval that may help minimize the potential for the
shunt are used. The choice of technique and “small for size” syndrome [23].
therefore its hemodynamic consequences, will
vary according to patient anatomy, surgeon
preference and local protocol as discussed else-  emodynamic Changes During
H
where in this book. Drainage of potential mas- Anhepatic Phase
sive ascites at the beginning of surgery is often
accompanied by a reduction in aorto-caval com- During the anhepatic phase, there is a progressive
promise and possibly even an improvement in reduction in body temperature and worsening of
overall systemic hemodynamics. This may fur- coagulopathy and fibrinolysis. These effects
ther be enhanced by a reduction in pulmonary interact with the hemodynamic situation. In those
artery pressure (PAP) however it is not uncom- techniques involving partial caval clamping or
mon to observe substantial hypovolemia at this complete cross-clamping in the absence of veno-
time as well. venous bypass, there is additionally the effect of
reduced venous return. While this can to some
I nterventions During Dissection Phase extent be offset by fluid administration, any
Prior to the anhepatic phase of the procedure, improvement seen is generally transient, and may
fluid and inotrope requirements vary consider- overall contribute to a worsening of the clinical
168 A. D. Padmakumar and M. C. Bellamy

situation because gut edema and fluid overload resulting in transient bradycardia, dysrhyth-
may ensue that becomes manifest after clamp mias and myocardial depression. Cardiac arrest
removal and graft reperfusion. due to hypokalemia and right ventricular fail-
ure is not uncommon (over 3% incidence in
I nterventions During Anhepatic Phase one series) and associated with significantly
The extra fluid volume required to maintain worse outcome even if spontaneous circulation
hemodynamic stability has been estimated as can be restored [25].
around 4 L or more [24]. Vasopressors can be As liver cell membranes become more func-
used to reduce fluid requirement while maintain- tional, there is rapid sequestration of potas-
ing hemodynamic stability during the anhepatic sium into intracellular locations. Cardiac
phase, especially in the presence of complete output rises, but the effects of complement
caval occlusion. Norepinephrine, vasopressin or activation and release of inflammatory media-
phenylephrine by infusion are generally the tors, together with generation of oxygen-
drugs of choice; they help maintain blood pres- derived free radicals, result in the “post
sure both by raising SVR, and importantly, reperfusion syndrome” [26, 27]. This is char-
through action on venous capacitance vessels acterized by hypotension and low SVR occur-
resulting in modestly improved venous return ring 5 min or more after reperfusion and lasting
and cardiac filling. This is particularly important at least 1 h [28].
in the presence of partial caval clamping.
Hemodynamic consequences of IVC occlusion, I nterventions During Graft Reperfusion
and therefore the effectiveness of alpha-agonists, In general, management of the immediate reper-
are dependent on the extent to which the variceal fusion phase consists of both preemptive and
circulation has resulted in collateralization, reactive elements. The preemptive element
facilitating venous return in the absence of vena includes administration of a bolus of calcium,
cava flow. Therefore hemodynamic changes dur- either as calcium chloride or gluconate, immedi-
ing complete or partial caval clamp in patients ately prior to graft reperfusion. This has com-
long standing chronic liver disease and extensive bined effects on protecting the myocardium
collateralization are often less severe than in against a potassium surge, while at the same time
patients with acute liver failure. replenishing or restoring deficient calcium ion
concentration to a physiological level.
Hypocalcemia during the late anhepatic phase is
 emodynamic Changes During Graft
H common, as a consequence of citrate accumula-
Reperfusion tion, and this may be clinically significant [22]. A
bolus of ionized calcium at this stage can be
At the time of graft reperfusion, caval blood highly effective. In some cases, a short infusion
flow is restored resulting initially in an of sodium bicarbonate may also be of value to
improvement of hemodynamics unless there is control peri-reperfusion hyperkalemia, and helps
substantial bleeding from the caval anastomo- maintain pH above 7.2. This is important to
sis. This is followed shortly afterwards by maintain vasopressor receptor responsiveness.
unclamping of the portal vein and reperfusion Appropriately judging the use of these agents
of the graft. The initial stages are affected by mandates blood gas analysis immediately prior to
the washout of cold fluid from the graft, poten- graft reperfusion. Some pracitioners prefer a
tially containing high concentrations of potas- small amount of epinephrine during reperfusion
sium, acid and traces of preservation fluids that as prophylaxis.
includes adenosine in the case of University of The reactive components of management of
Wisconsin solution. Therefore, the immediate the reperfusion process depend on the extent to
effect is due to acute myocardial cooling, which hypotension occurs. Small, incremental
exposure to potassium and adenosine, possibly boluses of epinephrine may be required.
14  Liver Transplantation: Hemodynamic Changes, Cardiac Output Monitoring and Inotropic Support 169

Depending on the specific clinical situation, of PAP. This is probably a feature of a fixed or


fluids may also be needed, for example where moderately elevated pulmonary vascular resis-
the patient is relatively hypovolemic or if there tance in the presence of a rising CO [30]. An
is unexpected bleeding at reperfusion. increase in left ventricular stroke volume is also
Cases who have been managed without frequently seen at this stage. Patients with pre-­
veno-­venous bypass may have received signifi- existing pulmonary hypertension or right ven-
cant fluid loading during the anhepatic phase, tricular dysfunction are at particular risk of
depending on the degree of vena caval occlu- decompensation secondary to elevation of PAP
sion and whether or not a temporary porto-sys- with a subsequent shift of the right ventricular
temic shunt had been created. As a result, there pressure flow-volume loop to the right. In these
may be an increased venous return as the vena situations there is a substantial risk of right heart
caval clamps are removed; such patients may failure resulting in very high venous pressures
show elevated right heart pressures within the and graft failure as a result of the loss of a pres-
seconds and minutes following liver reperfu- sure gradient between the portal and central cir-
sion and therefore, fluid administration is inap- culations. Graft blood flow is further compromised
propriate in this group. Epinephrine is generally by the potential low CO state and hypotension
a good choice of agent rather than phenyleph- that can result from inadequate left ventricular
rine in this situation. Constriction of venous filling secondary to right heart failure.
capacitance vessels can further contribute to
fluid overload. Occasionally, it is necessary to I nterventions During the Neo-Hepatic
combine epinephrine with a nitrate to achieve Phase
simultaneous improvement in cardiac function Standard management of persistent hypotension
and venous offloading. This, however, is a following liver graft reperfusion consists of the
strategy that requires considerable experience use of an alpha-agonist, commonly norepineph-
and very close monitoring. Injudicious use of rine by infusion. Epinephrine may be a suitable
nitrates at this stage can result in catastrophic alternative where a reduced or inappropriately
hypotension. Other agents that have been used low CO is also a feature. Patients who exhibit
experimentally to offset the hypotension and right heart failure at this time may benefit from
graft reperfusion include methylene blue, administration of epinephrine and/or a nitrate.
though there is very limited evidence to sup- There may also, in such situations, be a role for
port the use of this agent and therefore, its use dobutamine for inotropic support (with caution
cannot be advocated in routine clinical because of the vasodilatory properties of dobuta-
practice. mine). Dobutamine can be unpredictable in this
Cardiovascular collapse and arrest during this situation, as it is a racemic mixture, whose iso-
time needs aggressive treatment including treat- mers exhibit a differential alpha-agonist effect.
ment of hyperkalemia and other reversible causes An important and often overlooked contribu-
in addition to cardiopulmonary resuscitation. If tion to maintain hemodynamic stability during
everything else fails extracorporeal membrane vasodilation and major hemorrhage is plasma
oxygenation (ECMO) has been used in rare cases viscosity as a function of hematocrit among other
with success [29]. factors. Although conventional teaching has been
that a lower hematocrit is associated with reduced
plasma viscosity and hence better tissue perfu-
 emodynamic Changes During
H sion, current evidence questions this. At low
the Neo-Hepatic Phase plasma viscosity, reduced vascular sheer results
in altered signaling, probably via a NO pathway
Following reperfusion, reduction in SVR results among others, that can in turn result in vasocon-
in an elevation in CO. This, in turn, is accompa- striction and reduced tissue perfusion [31].
nied by (and is related to) progressive elevation Maintaining an adequate hematocrit is also ben-
170 A. D. Padmakumar and M. C. Bellamy

eficial for preserving diastolic function and hence load are taken into account [33]. Therefore,
helping to avoid the catastrophic rise in right judicious use of inotropes and vasopressors at
heart pressure, that would compromise hepatic this stage of the procedure directly influence the
perfusion at a stage when the liver is entirely need for postoperative ventilation and the time
dependent on portal venous flow. course of critical care unit discharge.
Classically, diuresis is described during the
neohepatic phase however, this depends on the
quality of the liver graft function, adequate perfu- References
sion pressure and the absence of preoperative 1. Richardson PD. Physiological regulation of the
renal impairment. Additionally, perioperative hepatic circulation. Fed Proc. 1982;41(6):2111–6.
factors such as massive hemorrhage during the 2. Lautt WW. Relationship between hepatic blood flow
dissection phase may compromise renal function and overall metabolism: the hepatic arterial buffer
response. Fed Proc. 1983;42(6):1662–6.
and limit the potential for a diuresis. To some 3. Lautt WW. Mechanism and role of intrinsic regulation
extent, decisions on volume replacement and of hepatic arterial blood flow: hepatic arterial buffer
potassium supplementation depend on observa- response. Am J Phys. 1985;249(5 Pt 1):G549–56.
tion of an adequate urine output and decreasing 4. Lautt WW. Regulatory processes interacting to main-
tain hepatic blood flow constancy: vascular compli-
serum potassium at this stage of the procedure. ance, hepatic arterial buffer response, hepatorenal
Clearly, the inotrope and vasopressor require- reflex, liver regeneration, escape from vasoconstric-
ments at the time of graft reperfusion differ from tion. Hepatol Res. 2007;37(11):891–903.
those required for support in the ensuing time 5. Kontos HA, Shapiro W, Mauck HP, Patterson JL Jr.
General and regional circulatory alterations in cirrho-
period. sis of the liver. Am J Med. 1964;37:526–35.
Cardiovascular changes persist well into the 6. Baik SK, Jee MG, Jeong PH, Kim JW, Ji SW, Kim
postoperative period. Systemic vascular resis- HS, Lee DK, Kwon SO, Kim YJ, Park JW, Chang
tance remains low for up to 24 h after surgery, but SJ. Relationship of hemodynamic indices and prog-
nosis in patients with liver cirrhosis. Korean J Intern
they will gradually normalize over the next Med. 2004;19(3):165–70.
24–48 h if graft function is good. Improvements 7. Liu H, Gaskari SA, Lee SS. Cardiac and vascular
of SVR are often already seen intraoperatively changes in cirrhosis: pathogenic mechanisms. World
with a well working graft. The normalization of J Gastroenterol. 2006;12(6):837–42.
8. Smith TW, Balligand JL, Kaye DM, Wiviott SD,
SVR seems to be independent of the reduction in Simmons WW, Han X, Michel T, Singh K, Kelly
CO, that also self-­corrects over a slightly greater RA. The role of the NO pathway in the control of car-
time course. It is therefore not entirely clear diac function. J Card Fail. 1996;2(4 Suppl):S141–7.
whether the reduction of CO is compensatory or 9. Van de Casteele M, Omasta A, Janssens S, Roskams
T, Desmet V, Nevens F, Fevery J. In vivo gene transfer
a consequence of separate neurohumoral regula- of endothelial nitric oxide synthase decreases portal
tion [32]. In patients with increased postoperative pressure in anaesthetised carbon tetrachloride cir-
PAP and wedge pressure, these usually remain rhotic rats. Gut. 2002;51(3):440–5.
high for at least a few days after surgery. 10. Liu H, Song D, Lee SS. Role of heme oxygenase-­
carbon monoxide pathway in pathogenesis of cirrhotic
Therefore, there is frequently an ongoing require- cardiomyopathy in the rat. Am J Physiol Gastrointest
ment for vasopressor support, although these can Liver Physiol. 2001;280(1):G68–74.
usually be decreased in the hours following repr- 11. Burtenshaw AJ, Isaac JL. The role of trans-­

fusion and surgery. Spontaneous improvement in oesophageal echocardiography for perioperative
cardiovascular monitoring during orthotopic liver
mean arterial pressure and organ perfusion is transplantation. Liver Transpl. 2006;12(11):1577–83.
associated with significant diuresis during the 12. Benington S, Ferris P, Nirmalan M. Emerging trends
process of weaning from artificial ventilation. In in minimally invasive haemodynamic monitoring and
most units, this is feasible within few hours after optimization of fluid therapy. Eur J Anaesthesiol.
2009;26(11):893–905.
surgery. In units with fast track protocols, venti- 13. Escorsell A, Bandi JC, Moitinho E, Feu F, García-­
lation and extubation at the end of surgery are Pagan JC, Bosch J, Rodés J. Time profile of the
feasible when by intraoperative fluid require- haemodynamic effects of terlipressin in portal hyper-
ments and the absence of pulmonary fluid over- tension. J Hepatol. 1997;26(3):621–7.
14  Liver Transplantation: Hemodynamic Changes, Cardiac Output Monitoring and Inotropic Support 171

14. Baik SK, Jeong PH, Ji SW, Yoo BS, Kim HS, Lee DK, plantation with venovenous bypass. Anaesthesia.
Kwon SO, Kim YJ, Park JW, Chang SJ, Lee SS. Acute 1995;50(9):776–8.
hemodynamic effects of octreotide and terlipressin in 25. Matsusaki T, Hilmi IA, Planinsic RM, Humar A,

patients with cirrhosis: a randomized comparison. Am Sakai T. Cardiac arrest during adult liver trans-
J Gastroenterol. 2005;100(3):631–5. plantation: a single institution's experience with
15. Ryckwaert F, Virsolvy A, Fort A, Murat B, Richard S, 1238 deceased donor transplants. Liver Transpl.
Guillon G, Colson PH. Terlipressin, a provasopressin 2013;19(11):1262–71.
drug exhibits direct vasoconstrictor properties: con- 26. Bellamy MC, Gedney JA, Buglass H, Gooi JH. Leeds
sequences on heart perfusion and performance. Crit Liver Group. Complement membrane attack com-
Care Med. 2009;37(3):876–81. plex and hemodynamic changes during human
16. Landry DW, Oliver JA. The pathogenesis of vasodila- orthotopic liver transplantation. Liver Transpl.
tory shock. N Engl J Med. 2001;345(8):588–95. 2004;10(2):273–8.
17. Wagener G, Kovalevskaya G, Minhaz M, Mattis
27. Bellamy MC, Galley HF, Webster NR. Changes in
F, Emond JC, Landry DW. Vasopressin deficiency inflammatory mediators during orthotopic liver trans-
and vasodilatory state in end-stage liver disease. J plantation. Br J Anaesth. 1997;79(3):338–41.
Cardiothorac Vasc Anesth. 2011;25(4):665–70. 28. Aggarwal S, Kang Y, Freeman JA, Fortunato FL,

18. Gray TA, Buckley BM, Sealey MM, Smith SC,
Pinsky MR. Postreperfusion syndrome: cardiovascu-
Tomlin P, McMaster P. Plasma ionized calcium moni- lar collapse following hepatic reperfusion during liver
toring during liver transplantation. Transplantation. transplantation. Transplant Proc. 1987;19(4 Suppl
1986;41(3):335–9. 3):54–5.
19. Díaz J, Acosta F, Parrilla P, Sansano T, Contreras RF, 29. Tejani M, Yi SY, Eudailey KW, George I, Guarrera
Bueno FS, Martínez P. Correlation among ionized cal- JV, Wagener G. Extracorporeal membrane oxygen-
cium, citrate, and total calcium levels during hepatic ation as a rescue device for postreperfusion cardiac
transplantation. Clin Biochem. 1995;28(3):315–7. arrest during liver transplantation. Liver Transpl.
20. Molenaar IQ, Begliomini B, Martinelli G, Putter H, 2015;21(3):410–4.
Terpstra OT, Porte RJ. Reduced need for vasopressors 30.
Webster NR, Bellamy MC, Lodge JP, Sadek
in patients receiving aprotinin during orthotopic liver SA. Haemodynamics of liver reperfusion: compari-
transplantation. Anesthesiology. 2001;94(3):433–8. son of two anaesthetic techniques. Br J Anaesth.
21. Koelzow H, Gedney JA, Baumann J, Snook NJ,
1994;72(4):418–21.
Bellamy MC. The effect of methylene blue on the 31. Salazar Vázquez BY, Martini J, Chávez Negrete A,
hemodynamic changes during ischemia reperfusion Cabrales P, Tsai AG, Intaglietta M. Microvascular
injury in orthotopic liver transplantation. Anesth benefits of increasing plasma viscosity and maintain-
Analg. 2002;94(4):824–9. ing blood viscosity: counterintuitive experimental
22. Møller S, Henriksen JH. Cirrhotic cardiomyopathy. J findings. Biorheology. 2009;46(3):167–79.
Hepatol. 2010;53(1):179–90. 32. Glauser FL. Systemic hemodynamic and cardiac

23. Busani S, Marconi G, Schiavon L, Rinaldi L, Del function changes in patients undergoing orthotopic
Buono M, Masetti M, Girardis M. Living donor liver liver transplantation. Chest. 1990;98(5):1210–5.
transplantation and management of portal venous 33. Glanemann M, Busch T, Neuhaus P, Kaisers U. Fast
pressure. Transplant Proc. 2006;38(4):1074–5. tracking in liver transplantation. Immediate postop-
24. Cheema SP, Hughes A, Webster NR, Bellamy
erative tracheal extubation: feasibility and clinical
MC. Cardiac function during orthotopic liver trans- impact. Swiss Med Wkly. 2007;137(13–14):187–91.
Coagulopathy: Pathophysiology,
Evaluation, and Treatment 15
Bubu A. Banini and Arun J. Sanyal

Keywords TFPI Tissue factor pathway inhibitor


Bleeding · Rotational thrombelastogram · TM Thrombomodulin
Thrombelastography · Clotting · tPA Tissue plasminogen activator
Hypercoaguability · Thrombosis TRALI Transfusion related acute lung injury
TxA Tranexamic acid
VWF von Willebrand factor
Abbreviations

AT Antithrombin Introduction
CVP Central venous pressure
DDAVP Desmopressin Hemostasis consists of processes that promote
EACA Epsilon-aminocaproic acid coagulation and those that favor fibrinolysis. Both
EPCR Endothelial protein C receptor processes are essential to creating clot while local-
FFP Fresh frozen plasma izing thrombosis to the site of injury and prevent-
INR International normalized ratio ing uncontrolled thrombotic extension. In liver
PAI-1 Plasminogen activator inhibitor disease, changes occur in both anti- and pro-hemo-
PT Prothrombin time static mechanisms, leading to a rebalanced coagu-
PTT Partial thromboplastin time lation system. In cirrhosis, the rebalanced state can
RBC Red blood cell be disrupted by several factors including stasis,
TACO Transfusion associated circulatory portal hypertension, dysfibrinogenemia, produc-
overload tion of endogenous heparinoids, platelet and endo-
TAFI Thrombin activatable fibrinolysis thelial dysfunction, renal failure and infection.
factor Whether planning an invasive procedure, major
TEG Thromboelastography surgery or transplantation, there is much dilemma
TF Tissue factor in how to properly treat these patients and their
coagulopathic status. This chapter will explore the
balance of hemostatic pathways, and review the
B. A. Banini, MD, PhD
A. J. Sanyal, MD, MBBS (*) defects that occur in progressive liver disease. We
Division of Gastroenterology and Hepatology, will also discuss how to evaluate and treat coagu-
Department of Internal Medicine, Virginia lopathy in this patient population, with specific
Commonwealth University, Richmond, VA, USA attention given to application towards liver
­
e-mail: bubu.banini@vcuhealth.org; arun.sanyal@
vcuhealth.org transplantation.

© Springer International Publishing AG, part of Springer Nature 2018 173


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_15
174 B. A. Banini and A. J. Sanyal

Primary Hemostasis transmembrane signal, allowing activation and


release of ADP, thromboxane A2, and alpha and
The initial step in the hemostatic pathway occurs dense granules by the platelet (Fig. 15.1c).
by formation of the platelet plug. Platelet aggrega- Platelet–platelet interaction via integrin μIIbβ3
tion creates the scaffolding on which thrombosis can then occur, leading to further platelet activa-
can then occur. When the vessel wall is damaged, tion and aggregation. In the meantime, the coagu-
subendothelial collagen is exposed to von lation cascade initiates, leading to platelet
Willebrand factor (VWF) in the serum. VWF stabilization.
binds to the site of injury, momentarily interacting
with platelets expressing glycoprotein GPIb
(Fig. 15.1a). This slows the flow of platelets until a Secondary Hemostasis
more lasting attachment is made between the
exposed collagen and platelet-expressed receptor Many interactions occur simultaneously at the
μ2β1 and glycoprotein VI, or platelet integrin site of endothelial damage. As platelets aggre-
μIIbβ3 and fibronectin with collagen (Fig. 15.1b). gate, the coagulation cascade initiates at the
Glycoprotein VI on the platelet surface initiates a platelet surface, forming a fibrin clot and rein-

Slo
win
a g b
GPIb

Platelet

GPVI
GPIb GPIb
α2β1 αIIbβ3
VWF
VWF

GPIb

fibro
VWF
GPIb

coll

VWF

nec
age

VWF

tin
n

VWF VWF VWF VWF VWF VWF VWF VWF VWF VWF
Endothelium Endothelium

Injury Site
-Subendothelial
collagen/matrix

Platelet adhesion
and aggregation
A2
TX
nd
Pa
AD
form mbin
n
atio

Platelet interaction via


o
Thr

VWF/fibronectin and
αIIbβ3 αIIbβ3

VWF VWF VWF VWF VWF

Fig. 15.1 (a) Subendothelial collagen binds to von μIIbβ3 and fibronectin with collagen. (c) Glycoprotein VI
Willebrand factor (VWF), which momentarily interacts on the platelet surface initiates a transmembrane signal,
with platelets expressing glycoprotein GPIb. (b) This pro- allowing for the release of ADP, thromboxane A2, and
cess slows the flow of platelets to create a more lasing alpha and dense granules by the platelet. Platelet aggrega-
attachment between the collagen- and platelet-expressed tion can then occur
receptor μ2β1 and glycoprotein VI or platelet integrin
15  Coagulopathy: Pathophysiology, Evaluation, and Treatment 175

forcing platelet aggregation. When the endothe- on the platelet surface [2–4]. Factor XI activates
lium is damaged, tissue factor (TF) is released Factors IXa and VIIIA, which then come together
into the bloodstream, binding to Factor VII, initi- to form the FIXa/FVIIIA, tenase complex.
ating the thrombin burst. This initial generation The tenase complex cleaves factor X into an
of thrombin promotes maximal platelet activa- activated form (Xa). Factor V is activated by
tion [1], as well as activation of additional coagu- FXa. This creates the first sufficient amount of
lation cofactors (Fig. 15.2). While this is not thrombin (IIa) to generate fibrin and stabilize
enough to generate a fibrin clot on its own, it the platelet plug. This is known as the “propa-
primes the clotting system for a burst of platelet gation” phase of thrombin generation
aggregation by activating Factors V, VIII, and XI (Figs. 15.3 and 15.4). FII is converted into FIIa

Fig. 15.2 Endothelial Secondary Hemostasis: The Initiation Phase


damage—tissue factor
(TF) interaction with
factor VII, initiating the
thrombin burst, VII
promoting platelet
activation, and Intraluminal blood flow
activation of additional
coagulation cofactors endothelium Prothrombin (II)

IX X

Xa Thrombin (IIa)
IXa VIIa
Va

T T T
F F F

Adventitial cell

Secondary Hemostasis: The Amplification Phase

Intraluminal blood flow

XI VIII V
Fig. 15.3 Amplification
phase—the tenase
complex, FIXa/FVIIIA,
cleaves factor X into an thrombin
activated form (Xa).
FXa activates Factor V,
Plt Plt
which creates the first
sufficient amount of ADP
thrombin (IIa) to Plt TXA2
endothelium VWF
generate fibrin and
stabilize the platelet
plug collagen
176 B. A. Banini and A. J. Sanyal

(thrombin), in turn cleaving fibrinogen into I nhibition and Fibrinolysis


fibrin, which forms a strong meshwork to pro- in Coagulation
mote clot stability and thrombosis. The coagu-
lation cascade only emphasizes the procoagulant Hemostasis is composed of “forward” driving
factors of the hemostasis. Equally important to forces that promote coagulation, and those that
understand are those steps which provide bal- “reverse” the process to favor fibrinolysis. Both
ance and inhibit the prothrombotic steps of the forces maintain a balance in order to localize
coagulation cascade. thrombosis to the site of injury and prevent
uncontrolled thrombotic extension. Thrombin
(factor IIa) generation is directly inhibited by tis-
sue factor pathway inhibitor (TFPI) and anti-
thrombin (AT) (Fig. 15.5). TFPI inactivates

Fig. 15.4 Propagation Secondary Hemostasis: The Propagation Phase


phase—the activity of
the coagulation cascade
continues to generate
thrombin, creating a Intraluminal blood flow
stable thrombin clot Xa Xa
thrombin

Xa Xa
Xa
Xa IX VIII V Xa
thrombin thrombin
thrombin

endothelium V Plt V Plt endothelium


W W
F F
collagen

AT &TFPI Limit Thrombosis to Endothelial Surface

VII

Serum
VIIa TFPI

Fig. 15.5  Tissue factor Serum


X

pathway inhibitor
(TFPI) inactivates
Xa

factors VIIa and Xa,


IIa
inactivating factor IIa
V Va
(thrombin) generation.
Also, antithrombin Thrombin (IIa) Antithrombin
inactivates thrombin.
TFPI is active only in
serum, unable to inhibit
at the cellular surface.
This localizes thrombin Xa Xa
generation to the
surfaces of platelets and endothelium endothelium
endothelium where
damage is present collagen
15  Coagulopathy: Pathophysiology, Evaluation, and Treatment 177

factors VIIa and Xa, and AT inactivates throm- bin generation to the site of damage. If thrombin
bin. However, TFPI is active only in serum, escapes the site of injury onto intact endothelial
unable to inhibit at the cellular surface. This cells, it will be bound to the endothelial surface
localizes thrombin generation to the surfaces of receptor thrombomudulin (TM), forming a
platelets and endothelium where damage is pres- thrombin/TM complex. This complex can no lon-
ent [5, 6]. ger carry out normal coagulant functions [9] and
The vitamin K-dependent factors Protein C activates protein C to bind protein S, inhibiting
and S further regulate the coagulation cascade. clot formation (Fig. 15.6). Other mechanisms
Protein C is a protease [7] whose activity is further reverse fibrin production through
enhanced by protein S, and together, they inhibit fibrinolysis.
factors Va and VIIIa. Protein C is localized to the These latter mechanisms (Fig. 15.7) utilize
endothelial cell surface by the endothelial protein factors which include tissue plasminogen acti-
C receptor (EPCR) [8], further localizing throm- vator (tPA), plasminogen activator inhibitor

Protein C and S: Antithrombotic Activity

c Va
X
Va
thrombomodulin thrombin S Protein C receptor
c c

endothelium

Fig. 15.6  Protein C is a protease, enhanced by protein S, injury is bound to the endothelial surface receptor throm-
which together inhibit factors Va and VIIIa. Protein C is bomudulin (TM), forming a thrombin/TM complex, and
localized to the endothelial cell surface by the endothelial losing the ability to carry out normal coagulant functions
protein C receptor (EPCR). Thrombin escaping the site of

Fibrinolysis: Clot Reversal


Thrombin

+ PAI-1

TPA
uPA
Fibrinogen --- Fibrin TAFI
FXIII
+

Plasmin Plasminogen

Fibrin Degradation
Products (FDP)
Plasmin inhibitor

Fig. 15.7  Fibrinolysis—tPA released from endothelial able fibrinolysis factor (TAFI) inactivates the conversion
cells, macrophages, and renal epithelial cells, activates of plasminogen into plasmin, by cleaving the C-terminal
plasminogen to plasmin. FXIII inhibits the degradation of lysine and arginine residues on tPA and plasminogen pre-
fibrin into fibrin degradation products. Thrombin activat- venting their binding to one another
178 B. A. Banini and A. J. Sanyal

(PAI-­1), plasminogen, alpha2-antiplasmin, his-  oagulation Cascade and Liver


C
tidine-rich glycoprotein, and factor XIII, all of Disease
which except tPA and PAI-1, are synthesized by
the liver [10]. tPA is released from endothelial The coagulation cascade is comprised of redun-
cells, macrophages, and renal epithelial cells, dant steps that keep the entire system in balance.
and activates plasminogen to plasmin. Plasmin In a healthy individual, only 20–50% of normal
is an enzyme capable of degrading fibrin into levels of procoagulants are needed to achieve
soluble fibrin degradation products. This pro- hemostasis [13]. Thus, there is a fair amount of
cess is regulated by inhibitory factors, including overlap between pro- and anticoagulant factors
FXIII, which inhibit the degradation of fibrin that provide a buffering system for hemostasis in
and stabilize the fibrin meshwork. Thrombin healthy individuals. That buffering system is ten-
activatable fibrinolysis factor (TAFI) inactivates uous in the liver disease patient and becomes
the conversion of plasminogen into plasmin by increasingly difficult to balance as small changes
cleaving the C-terminal lysine and arginine resi- in factor levels can lead to significant changes in
dues on tPA and plasminogen and preventing the entire system. PT and INR have been heavily
their binding to one another. relied upon to assess the degree of coagulopathy
When evaluating the effects of liver disease on in liver patients. This reflects a shortcoming in
the coagulation cascade, these inhibitory factors our understanding of hemostasis and barriers in
are often not considered. Unfortunately there is our testing strategies. As PT and INR are a mere
no simple way to test for their activity at the bed- measure of “procoagulant” factors, they disre-
side. They are a major contributor to the overall gard the effect of liver disease on “anticoagulant”
coagulation status of the liver disease patients factors. In liver disease, all procoagulant factors
and need to be considered before treating except FVIII are reduced. In addition, the antico-
coagulopathy. agulant factors such as protein C and S are also
reduced [14, 15]. The reduction of anticoagulant
factors seen in liver disease is not reflected in PT
Hemostasis in Liver Disease and INR measurements. Thus, in this rebalanced
state of hemostasis, thrombin generation may
After reviewing normal hemostasis, we can explore actually be normal in the setting of increased PT,
how advanced liver disease affects this process. PTT, and INR [16], emphasizing their inade-
Because hepatic parenchymal cells synthesize quacy in evaluation coagulation status in liver
many of the pro- and anticoagulant proteins disease. In cirrhosis, FVIII and other procoagu-
involved in coagulation, it is easy to understand lants including vWF can be increased [17].
how liver disease disrupts hemostasis. Recent stud- PT and INR only assess one aspect of the coag-
ies have led to a greater understanding of the ‘rebal- ulation cascade—namely vitamin K dependent
ance’ of the coagulation system in liver disease, factors FII, VII, IX, and X—as these tests were
involving increased levels of circulating von originally developed to measure the therapeutic
Willebrand factor and factor VIII, and reduced syn- effects of drugs like warfarin that affects vitamin
thesis of anticoagulant factors [11, 12]. However, K dependent factors. In vivo, hemostasis in liver
in cirrhosis, the rebalance is unstable and can be disease involves reduced levels of fibrinogen, pro-
disrupted by several factors including stasis and thrombin, the vitamin K dependent factors, as
portal hypertension, dysfibrinogenemia, produc- well as protein C and S and other anticoagulants.
tion of endogenous heparinoids, platelet and endo- Moreover, factors including changes in platelet
thelial dysfunction, renal failure, and increased function, fibrinolysis and endothelial function all
susceptibility to infection. impact hemostasis in liver disease patients.
15  Coagulopathy: Pathophysiology, Evaluation, and Treatment 179

Platelet Function in Liver Disease collagen and fibrinogen as long as platelet count
and hematocrit are adjusted to levels found in
In primary hemostasis, the initial platelet plug healthy patients [36, 37].
provides the scaffolding for the coagulation cas-
cade and thrombin generation. Impaired primary
hemostasis has traditionally been linked to abnor- Hyperfibrinolysis in Liver Disease
malities of platelet number and function in liver
disease [15]. Platelet numbers are decreased due The role of hyperfibrinolysis in patients with cir-
to the effects of portal hypertension and increased rhosis is controversial and widely debated in the
sequestration in the spleen, and with worsening current literature [38]. Whether hyper- or hypofi-
liver failure, thrombopoietin levels decrease [18, brinolysis is occurring, it is agreed that it compli-
19]. Hepatitis C, alcohol toxicity, and nutritional cates the picture [39–42]. Hyperfibrinolysis
folic acid deficiency compound this problem by seems to be more problematic as liver disease
depressing megakaryocytopoiesis [20–22]. The progresses [43–46], and low-grade fibrinolysis
role disseminated intravascular coagulation has been shown to occur in 30–46% of patients
(DIC) as a cause of thrombocytopenia in these with end-stage disease [47].
patients is contentious [23]; however, low-level Plasminogen, alpha2-antiplasmin, histidine-­
consumption associated with DIC possibly rich-­glycoprotein, factor XIII, and TAFI [48–57]
decreases platelet life span as well [24]. are all produced in the liver, and therefore their
Reduced platelet function further complicates levels are reduced in liver disease. However,
impaired primary hemostasis, and there is strong increased levels of tissue plasminogen activator
evidence that platelet aggregation is reduced [25– (tPA) and plasminogen activator inhibitor-1 (PAI-­
27]. Platelet activation is affected by both intrin- 1) are present as these are not synthesized in the
sic and extrinsic stresses. Intrinsically, decreased liver [10]. tPA is elevated most likely due to
thromboxane A2 synthesis, altered transmem- reduced hepatic clearance [58, 59]; PAI-1 levels
brane signaling and a reduction in glycoprotein Ib seem to be correlated with the clinical stage of
and platelet integrin μIIbβ3 reduce platelet activa- the liver disease. PAI-1 is elevated in patients
tion [27–32]. Extrinsically, elevated levels of with chronic, smoldering liver disease [44, 60],
nitric oxide and prostacyclin inhibit platelet func- and decreased in patients with severe liver failure
tion [33] as a result of endothelial dysfunction. [44, 61]. Patients with acute liver failure have a
The platelet phospholipid membrane may also be higher circulating amount of acute phase reactant
affected by abnormal high density lipoprotein PAI-1, and more of a shift towards hypofibrinoly-
particles in plasma [34]. Moreover, in liver dis- sis [41]. Therefore, theoretically, increased fibri-
ease, blood flow defects occur, and these may be nolysis should occur in patients with severe liver
compounded by reduced hemoglobin [35]. failure who have an increased pool of tPA, and
It is still not clear how these abnormalities depressed levels of PAI-1 and alpha2-antiplasmin
contribute to bleeding time as elevated levels of to balance it.
VWF (and decreased levels of ADAMTS-13, a Decreased levels of TAFI have also been linked
metalloproteinase that cleaves VWF) may com- to hyperfibrinolysis in cirrhosis [62]. Colluci and
pensate for the impairment [12]. In fact, the ele- colleagues [63] demonstrated that TAFIa genera-
vation of VWF may lead to a higher rate of tion was low in cirrhosis due to decreased levels of
thrombin generation and clot formation [12]. It TAFI, suggesting that depleted TAFIa was a sig-
appears that in liver disease, thrombin generation nificant contributor to hyperfibrinolysis. However,
is not necessarily negatively affected and that Lisman et al. [64] came to the conclusion that
under physiologic conditions of flow, platelets TAFI deficiency was not a significant contributor
from a patient with cirrhosis can interact with to hyperfibrinolysis in cirrhosis. This disparity
180 B. A. Banini and A. J. Sanyal

may be due to the inability to test and measure Evaluation of Coagulation


global fibrinolysis, and newer methods of testing
global fibrinolysis confirmed the presence of Bleeding Time
hyperfibrinolysis in chronic liver disease [65].
Fibrinolysis is important not necessarily because Bleeding time is performed by inflicting a stan-
of its potential to initiate bleeding in a liver disease dardized cut on the volar aspect of the forearm
patient but because of the important role it plays in while applying a blood pressure cuff to the upper
delaying primary and secondary hemostasis, con- arm. There are several limitations to the repro-
tributing to the severity or recurrence of bleeding ducibility to the test including the skill of the
events. In liver transplantation, many studies report technician, skin thickness, ambient temperature,
enhanced fibrinolytic activity during the anhepatic and endothelial dysfunction [80–82]. Bleeding
stage [66]. The lack of tPA clearance and the reduc- time is not well validated [81], and studies using
tion of alpha2-­antiplasmin may be responsible for desmopressin as treatment showed improved
this enhanced fibrinolysis [67]. After liver trans- bleeding time but no effect on risk of variceal
plantation, fibrinolysis may persist for a prolonged bleeding in liver disease patients [82–84]. The
time especially in case of early allograft dysfunc- lack of correlation between bleeding time and
tion [67–69]. An initial rise in tPA during the anhe- risk of bleeding in liver disease patients makes
patic stage is followed by further increases after the test not very helpful in assessing
reperfusion in 75% of patients [68, 70]. The recog- coagulation.
nition, monitoring and treatment of fibrinolysis in
moderate to severe liver disease are important and
will be addressed later in this chapter.  latelet Function Analyzer: 100
P
(PFA-100)

 ndothelial Dysfunction and Liver


E The platelet function analyzer-100 (PFA-100) is
Disease an vitro test that provides a quick way to quanti-
tatively evaluate primary hemostasis under shear
Sinusoidal endothelial cells produce and release stress. The test measures platelet adhesion as
vasoactive substances that regulate intrahepatic blood flows through a collagen membrane under
vascular resistance [71]. Endothelial dysfunction is the draw of a vacuum. The time it takes to occlude
thought to be due to a defective vasodilatory the channel in the collagen membrane is the mea-
response to acetylcholine and insufficient endothe- surement of platelet adhesion [80, 85]. One study
lial NO synthase to produce NO [72–74]. Increased demonstrated that closure time was decreased if
production of Thromboxane A2 also leads to hematocrit was normalized in the blood of liver
increased intrahepatic resistance in advanced liver disease patients [26] however in general the PFA-­
disease [75, 76]. Portal hypertension is the liver’s 100 has not been considered overwhelmingly
response to its inability to accommodate fluctua- helpful and is rarely used to platelet function.
tions in increased portal circulation.
As portal hypertension develops, deleterious
processes such as splanchnic vasodilatation occur. Prothrombin Time
The endothelium in this system responds by pro-
ducing more NO resulting in arterial dilatation The prothrombin time evaluates the extrinsic path-
and a hyperdynamic circulation which is related way of the coagulation cascade and is responsive
to complications such as variceal bleeding, asci- to deficiencies in factors X, VII, V, II and fibrino-
tes, hepatorenal syndrome, and hepatopulmonary gen. The test was developed by Armand Quick
syndrome [77–79]. There are several proposed and measures the time it takes a blood sample to
ways to monitor the response of the endothelium clot once thromboplastin and calcium chloride are
in liver disease, and these will be addressed later. added [86]. Results are measured in seconds and
15  Coagulopathy: Pathophysiology, Evaluation, and Treatment 181

commonly standardized using the International Thrombin generation test


Normalized Ratio (INR). The INR was developed Time to peak peak
as a way to account for differences in reagents
(thromboplastin) across different laboratories, and
was originally used to standardize treatment of

thrombin
patients using vitamin K antagonists like warfarin. Inactivation
phase
The use of INR is an imperfect system and in cou- slope
madinized patients, a variability of 13% has been
observed depending on where lab samples are
obtained [87]. This variability is accentuated in lag AUC
liver disease patients and is accentuated in liver
disease patients and mean INR variations increase time
further with advanced liver disease [88–91]. Fig. 15.8  Thrombin generation test—tissue factor (TF)
Not only are PT and INR variable but they and phospholipids are added to plasma to generate throm-
also reflect an incomplete picture of coagulopa- bin and trigger coagulation. AUC (area under curve) = ETP
thy in liver disease and are unreliable in this (Endogenous thrombin potential), or the amount of work
that can potentially be done by thrombin. Proposed poten-
patient population [92–96]. This test for example tial of this test is to quantitate how much and how long
does not reflect the parallel depletion of protein C thrombin is active
and S in vivo [16], and is without sufficient levels
of thrombomodulin for thrombin-mediated acti-
vation of protein C [97]. PT is also limited by its Thrombin, a potent platelet activator and phos-
inability assess the role of platelet and endothe- pholipids, representing the platelet surface, feed
lial dysfunction. forward to allow for explosive thrombin genera-
tion. In the thrombin generation test (Fig. 15.8),
tissue factor and phospholipids are added to trig-
Activated Partial Thromboplastin ger coagulation, and thrombin generation is plot-
Time ted over time. The initial part of the curve is the
lag time, while the time from tissue factor addi-
The activated partial thromboplastin time (aPTT) tion to peak of thrombin is referred to as time to
assesses the intrinsic pathway of the coagulation peak. The area under the curve (AUC) measures
cascade and is increased with deficiencies of all the effectiveness of thrombin generation in a sys-
coagulation factors except for factors VII and tem where both pro- and anti-­ coagulants are
XIII. It represents the time (in seconds) for phos- examined as they operate in plasma. Reliability
pholipids, representing the platelet membrane, to of the thrombin generation test is yet to be deter-
generate a thrombus by activating factors like mined but may have acceptable levels of varia-
factor XII. It is often used clinically to monitor tion. Further clinical studies are needed to
the anticoagulant effects of heparin in patients, evaluate the usefulness of the thrombin genera-
though there is no standardization between labo- tion test as it applies to liver disease and
ratories. As previously mentioned, relying solely transplantation.
on this test to evaluate coagulation in liver dis-
ease patients is fraught with difficulties.
Thromboelastography

Thrombin Generation Test Thromboelastography (TEG) provides a graph-


ical representation of the viscoelastic changes
This test utilizes tissue factor and phospholipids that occur during coagulation in vitro
to trigger thrombin generation and is arguably the (Fig. 15.9). A stationary pin is introduced into a
closest representation of what occurs in vivo. sample of whole blood that oscillates back and
182 B. A. Banini and A. J. Sanyal

Fig. 15.9  Thromboelastography (TEG)


Thromboelastography Coagulation Fibrinolysis
(TEG)—R reflects
coagulation factor and
platelet activities; K
reflects activity of
fibrinogen, Factor II,
and effects of
hematocrit; α represents α
MA
clotting factor
A30
deficiency; MA
R
indicates platelet, fibrin,
and factor XIII function; 30 min
Ly 30 reflects
fibrinolysis. A30 further
represents fibrinolysis
and is the amplitude K Ly 30
30 min after MA

forth six times per minute. Kaolin is added, ini-  onitoring Fibrinolysis During Liver
M
tiating thrombin generation [98, 99] and subse- Transplant
quently fibrinogen is converted to fibrin. As
fibrin is stabilized by platelets [100] and the Several studies have reported hyperfibrinolysis
clot is strengthened, the pin detects viscoelastic during the anhepatic stage when venous return is
changes and records these dynamic changes in maintained via a venovenous shunt [66], as well
a graph. The technique can provide continuous as during graft reperfusion [67] Some attribute
observation and quantitative measurement of this to decreased hepatic clearance of tPA, and
different stages of hemostasis, including clot Porte et al. demonstrated that tPA levels are
formation, strength, platelet function, and fibri- increased during reperfusion in 75% of patients
nolysis. Technical difficulties have limited the [70]. If a transplanted liver has sustained
use of TEG in the past, however improved tech- increased damage during transport due to isch-
nology have led to standardization of the tech- emia, it may take longer for the hyperfibrinolysis
nique and improved reproducibility. A to resolve. TEG or ROTEM remain key instru-
modification of the TEG, the rotational throm- ments in monitoring every step of hemostasis
boelastometry or ROTEM, uses a rotating sen- during liver transplant including fibrinolysis;
sor shaft rather than a rotating cup and is less however, the measures discussed below are also
sensitive and provides a simpler and more stan- helpful.
dardized user interface. Liver transplantation
was one of the first procedures to utilize TEG
[101], and TEG and ROTEM are now increas- Prevention and  Treatment
ingly used as standard tests to evaluate coagula- Guidelines for Bleeding During
tion intraoperatively [98, 102]. As the tests Liver Surgery
monitor different phases of hemostasis, intra-
operative therapy can be individualized and While some bleeding is inevitable during liver
reevaluated in a short period of time. In a recent resection and transplantation, blood loss rates have
study by Stravitz et al. [103], clinically evident decreased substantially as surgical technique and
thrombosis correlated with prolonged R-time preventive measures through volume management
on TEG, a value that indicated hypocoagulabil- have become more sophisticated. Particularly per-
ity. More studies are needed to assess the abil- tinent in the coagulopathic liver disease patient,
ity of TEG to determine bleeding versus extensive bleeding may require transfusion of
thrombotic risk. blood or blood products, which is associated with
15  Coagulopathy: Pathophysiology, Evaluation, and Treatment 183

increased rates of morbidity and mortality [104– (33.5 mL/kg) [114]. Currently the recommended
109]. Recommendations regarding the administra- therapeutic dosage of FFP is 10–20 mL/kg, until
tion of blood products have improved the better defined with future studies.
transfusion requirements and should help guide While dosage remains an important issue,
the decision to treat coagulopathy during liver another difficulty is determining when to trans-
transplantation. We will present these recommen- fuse plasma and which diagnostic markers to use
dations here. to guide transfusion. Massicotte et al. demon-
strated that preoperative plasma transfusion did
not decrease the need for intraoperative red blood
Fresh Frozen Plasma cell (RBC) transfusion and found no difference in
the number of plasma or RBC transfusion units
Fresh frozen plasma may be obtained from whole between patients with an INR >1.5 and those
blood or via plasmapharesis and is frozen within with an INR <1.5 during 200 liver transplants
8 h at −30° C. FFP contains both pro- and antico- [115]. They further demonstrated that transplant
agulation factors, acute phase proteins, immuno- patients with an INR >1.5 who did not receive
globulins and albumin [110], and it is often used plasma subsequently did not incur any more RBC
to prevent or stop bleeding. Factor VIII is typi- transfusion when compared to patients with an
cally the only plasma protein whose level is qual- INR <1.5.
ity controlled, and while coagulation factor FFP transfusion is certainly not without risk,
content can be maintained for up to 5 days at and may have the highest incidence of complica-
1–6° C, there is evidence that the levels of FV tions of all blood products in liver transplantation
and FVIII fall over time. There is variability in [116, 117]. The most important complications
factors between units, with heterogeneity reflect- relevant to liver disease patients are transfusion
ing genetic differences between donors and/or related acute lung injury (TRALI), an inflamma-
adverse effects of pathogen killing techniques tory reaction producing non-cardiogenic pulmo-
[111]. One unit of FFP measures about 300 mL, nary edema acutely within 6 h of transfusion
and appropriate dosing is loosely agreed upon. [118], and transfusion associated circulatory
Most of the recommendations for dosing are overload (TACO). TACO is an acute syndrome
based upon mathematical extrapolation of factor producing elevated blood pressure and dyspnea
content and physiologic response to the effects of associated with large volume transfusions and
plasma infusion [112]. Data has been variable as resulting in longer hospital stay and increased
to the efficacy of FFP as a therapeutic agent. A mortality compared to TRALI. Variable report-
prospective evaluation of 324 units of FFP on ing of both complications have made it difficult
120 patients demonstrated only 15% of patients to define the incidence. TRALI has been related
corrected halfway to normal PT and INR and 1% to female donors through England’s Serious
completely corrected with FFP transfusion [106]. Hazards of Transfusion hemovigilance program
Even more troubling in this retrospective study is [119], and as a consequence male donors are pre-
that there was no correlation between clinical ferred. Allergic reactions to FFP occur at a rare
bleeding and diagnostic test results. There was rate of 1–3% of all FFP transfusions [116], and
also no evidence of a dose-dependent response of there is a minor risk of infectious complications
plasma transfusion. Another study specific to related to FFP.
liver disease patients found that the median Despite the risks associated with FFP infu-
reduction of INR attained after FFP transfusion sion, plasma is no longer directly linked to
was 0.2 (range 0–0.7) [113]. Similar results have decreased survival rate. Rather, there is a known
been attained with studies that also evaluated a association between increased 1-year mortality,
volume-related benefit: lower doses (12.2 mL/ RBC transfusion and Child Pugh Score and
kg) had less therapeutic benefit and increased plasma transfusion has been the variable with the
harm compared to higher doses FFP transfusion strongest association with blood transfusion
184 B. A. Banini and A. J. Sanyal

[120]. In another retrospective study done by nolytic state after transplant, increased levels of
Massicotte et al. [121], the proposed sequence of tissue plasminogen activator released from the
events was that the patient with coagulation graft, and increased platelet activation after trans-
defects receives plasma (10–15 mL/kg), the plant [123, 129–131]. Factors indicating platelet
expanded volume would increase CVP causing activation and degranulation have been detected
more bleeding prior to the anhepatic phase. The in serum and graft samples at elevated levels
resultant increased rate of bleeding associated [123, 125, 127, 132]. Due to altered platelet func-
with plasma infusion and subsequent blood trans- tion, a simple serum platelet count is inadequate
fusion is what is thought to be responsible for in guiding transfusion thresholds.
increased 1-year mortality in liver transplant Platelet transfusion should considered a means
patients. to decrease bleeding and minimize RBC transfu-
While the American Society of Anesthesiology sions, as there is demonstrated association
recommends transfusion of FFP for an interna- between transplant complications and increased
tional normalized ratio greater than 2.0 in patients RBC transfusion [105, 109, 133–135]. The risks
with excessive microvascular bleeding [122], of TRALI and TACO with plasma transfusion are
better control of volume status and lower central clearly recognized, however the risks of platelet
venous pressure may be preferential over attempt- transfusion during liver transplantation are less
ing to correct coagulopathy strictly defined by an well described. With improved surgical technique
INR of 2.0. For now, it is advised that INR of 2.0 and perioperative strategies to minimize bleed-
with clinical bleeding may be an indication for ing, older studies associating large volume plate-
FFP transfusion; however, more work is required let transfusions and poor survival after surgery
in this area. are less helpful [109]. A retrospective study by de
Boer et al. identified RBC and platelet transfu-
sions as risk factors affecting 1-year survival in
Platelet Transfusion first-time liver transplant patients, independent of
markers for worse disease, such as model for end
Apart from the function of platelets in primary stage liver disease score [133]. This study evalu-
hemostasis, platelets may actually play an impor- ated 433 liver transplant patients, and analyzed
tant role in regulating inflammation, angiogene- 26 variables and found that 1-year survival risk
sis, tissue repair/regeneration and ischemia and decreased dose-related with platelet transfusion
reperfusion injury [123–126], all of which are with a hazard ratio of 1.377 per unit of platelets
relevant during liver transplantation. It is there- (P = 0.01). Platelet transfusion also appeared to
fore important to know which platelet levels to have a negative impact on graft survival, but this
strive for pre-, intra-, and post-operatively inde- association was not present on multivariate anal-
pendent of their role in hemostasis. Since there is yses. It is difficult to determine causality for poor
currently no optimal way to monitor the extent of outcomes of transplantation on platelet transfu-
primary hemostasis, it is difficult to predict opti- sion alone using retrospective studies. However
mal platelet thresholds for surgery. in the absence of randomized trials these results
What we do know about the thrombocytope- should be considered when administering plate-
nic liver patient is that primary hemostasis may lets during liver transplantation. We recommend
not be as compromised as previously thought that platelet transfusion should be reserved for
[16, 37]. Platelet levels are in a constant state of active bleeding and low platelet count and not be
flux during and after liver transplantation due to used prophylactically.
hemodilution, immunologic reactions, and Patients with platelet counts greater than
30–55% reduction in levels due to entrapment in 75 × 109/L and an INR <1.5 are at no increased
the liver during liver reperfusion [127, 128]. In risk for bleeding during invasive procedures.
addition to alterations in platelet numbers, plate- Patients with a platelet count >50 × 109/L without
let function is also altered due to the hyperfibri- evidence of bleeding should also not be trans-
15  Coagulopathy: Pathophysiology, Evaluation, and Treatment 185

fused perioperatively, as these platelet levels are tion, and fibrinolysis, and include DDAVP, apro-
not correlated with bleeding risk in invasive pro- tinin, lysine analogues, and recombinant activated
cedures such as paracentesis [136, 137]. Dosing factor VII (rFVIIa).
of platelets should be tailored for each individual
patient and guided by bleeding, TEG, and whether
or not a consumptive platelet process is present. Desmopressin
Endogenously produced platelets typically sur-
vive around 10 days; however, transfused platelets DDAVP or 1-deamino-8-D-arginine is a synthetic
do not last this long, with further reduced life- analogue to vasopressin. It acts by releasing fac-
spans if platelet consumption is present. Patients tor VII and VWF from endothelial storage pools
with platelet levels below 100 × 109/L have shorter into the serum. The effect is rapid and short-lived
platelet life spans when compared to patients with and has a decreased response over repeated use
levels greater than this [138]. of the drug without adequate time for storage
A single dose of random-donor platelets pools to replenish. This agent has been used suc-
contains approximately 3 × 1011 platelets, sus- cessfully in the treatment of minor to moderate
pended in 50–70 mL of plasma. The optimal bleeding during surgical procedures for patients
dose specific to liver patients has not been with von Willebrand deficiency or hemophilia
determined and often has more to do with indi- A. Dosing is usually 0.3 ug/kg intravenously
vidual patient requirements and availability, as infused over 20 min.
platelets are a limited resource [139–143]. The There is very little literature supporting its use
standard dose prescription for platelets is during a major operation such as liver transplan-
10 mL/kg, with a maximum dose of five ran- tation, however, intranasal DDAVP was shown to
dom donor platelet units. Single donor platelet be equally effective as blood product transfusion
apharesis units contain at least 300 × 109 plate- in achieving hemostasis in cirrhotic patients
lets, suspended in 200–400 mL of plasma [144]. undergoing tooth extraction [147]. In this study,
The hematology literature has found little dif- patients were matched evenly and there was no
ference in efficacy between different platelet difference between groups transfused with plate-
preparations prepared from random donors ver- lets versus those given DDAVP, including MELD
sus those collected by apharesis [145, 146]. score, number of tooth extractions, and coagula-
However, platelets obtained by apharesis are tion profile. For minor to intermediate procedures
obtained from one donor and decrease the inci- like tooth extraction, patients receiving DDAVP
dence of immune-mediated refractoriness. received no rescue transfusions after the proce-
They are used preferentially in patients who dure, whereas one platelet-transfused patient
may be refractory to random donor platelet required rescue transfusion [147]. Outcomes
units. Platelet transfusion is recommended were considered the same between the two
when patient platelet levels drop below groups.
20–50 × 109/L or if platelet count cannot be Results with DDAVP have been favorable in
obtained and there is evidence of microvascular cardiac surgery and for patients on cardiopul-
bleeding and coagulopathy unless viscoelastic monary bypass [148, 149]. Patients undergoing
testing can be obtained. complex spinal fusion however did not show
reduced blood loss when treated with DDAVP
versus placebo [150]. Moreover, DDAVP infu-
 rocoagulant Drugs and Liver
P sion did not improve hemostasis in cirrhotic
Transplantation patients [151]. While DDAVP should provide
some theoretical benefit to liver disease patients
A limited amount of information is available on undergoing transplant surgery, we currently
pharmacologic alternatives to blood product use. cannot recommend routine use of DDAVP as a
Agents in this class target hemostasis, coagula- hemostatic agent.
186 B. A. Banini and A. J. Sanyal

Lysine Analogues 158]. The second proposed mechanism involves


rFVII binding directly to the platelet surface and
Epsilon-aminocaproic acid (EACA) and activating factor X [159]. The normal FVII:FVIIa
tranexamic acid (TxA) are synthetic analogues of ratio in the serum is 100:1 (10 and 0.10 nmol/L,
the amino acid lysine, which competitively block respectively), and with rFVII infusion, FVIIa lev-
lysine-binding sites on plasminogen to limit els increase 100-fold to 3–20 nmol/L [160]. The
hyperfibrinolysis. The Cochrane group meta-­ agent seems to be well tolerated and widespread
analysis found aprotinin and lysine analogues to activation of coagulation resulting in DIC has not
have a nearly similar effect on limiting blood been reported. rFVII has proven to be useful in
transfusion [152], and a recent randomized con- other off label uses such as controlling bleeding
trolled trial of liver transplant patients failed to in trauma, obstetrical complications, and surgical
demonstrate a difference between TxA and apro- patients with complex coagulation disorders
tinin with regards to transfusion requirements [161, 162].
[134]. EACA was first shown by Kang et al. to rFVII corrects vitamin-K dependent decreases
decrease the amount of residual bleeding during of coagulation factors [163]. This has led to its
the hyperfibrinolytic state after liver transplanta- use in liver patients with a depletion in vitamin-K
tion in 20 of 97 liver transplant patients [153]. A dependent coagulation factors. rFVII is able to
standard dose of 1 gram of EACA bolus was used. effectively reverse prolonged prothrombin time
A more recent study compared EACA, TXA, and (PT) in cirrhotic patients without active bleeding
placebo used as prophylaxis to decrease transfu- [164], and Lisman et al. concluded that a single
sions during and after transplant and found that dose of rFVIIa could lead to stable clot formation
TXA was beneficial over EACA and placebo, in cirrhotic patients [165].
with no difference in RBC transfusion sparing Results have been mixed when using rFVIIa
effect between EACA and placebo [154]. Dosage in liver transplant patients. There are multiple
for TXA has varied greatly, and different studies case reports and series reporting efficacy of
have shown reduction in transfusion at dosages of rFVIIa at varying doses such as 100 [166], 80
10 and 40 mg/kg−1/h−1 [154, 155]. [167], or 68.4 μg/kg [168]. However, better
Limited research of both of these agents information is derived from several randomized
makes it difficult to recommend specific dosage, control and retrospective case control studies.
and further trials should be conducted. Also, Two retrospective case control studies con-
comparative data is extremely limited between cluded that administration of rFVIIa preopera-
these two agents, and it is difficult to recommend tively decreased blood transfusion requirements
using one over the other. in transplant patients [169, 170]. The dose of
rFVIIa in one of those studies of 22 patients was
58 μg/kg. Two randomized control studies failed
Recombinant Factor VIIa to show efficacy of rFVIIa in reducing transfu-
sion requirements while using 20, 40, 60, 80, or
Recombinant Factor VIIa (rFVIIa) has been 120  μg/kg bolus doses of rFVIIa when com-
approved for use in hemophilia patients with pared to placebo [171, 172]. A more recent ran-
inhibitors in both surgical and non-surgical set- domized, double-blind trial found that patients
tings. The agent is very similar to endogenous undergoing orthotopic liver transplant who
FVIIa, and is thought to enhance thrombin gen- received 40 μg/kg of rFVIIa perioperatively
eration at sites of endothelial injury. Two mecha- required fewer transfused units of blood when
nisms have been suggested in rFVII driven compared to placebo [173]. A recent meta-anal-
thrombin generation: a high concentration to tis- ysis on the use of rFVIIa for bleeding prophy-
sue factor (TF) and rFVIIa accumulate at the site laxis in hepatobiliary surgery was unable to
of vascular damage [156], which in turn activates draw conclusions for clinical application due to
factor Xa and triggers thrombin generation [157, limited data [174].
15  Coagulopathy: Pathophysiology, Evaluation, and Treatment 187

As we previously stated, prothrombin time prior to liver transplant and other invasive proce-
does not necessarily correlate with improved dures. It is essential to optimize platelet count
coagulation profile in liver disease, and correc- prior to surgery, however, unnecessary platelet
tion of prothrombin time does not necessarily and RBC transfusion should be avoided. Further
decrease transfusion rate or improve outcome. studies are needed to improve our understanding
While it seems that more randomized, controlled of the intricate imbalances comprising coagulop-
studies should be implemented, the current con- athy in the liver disease patient, and to more
sensus European Guidelines on the use or rFVIIa effectively perform invasive procedures and care
during liver transplantation or resection recom- for the liver transplant patient.
mends that rFVIIa not be used routinely (grade B
evidence) [175].
References
Topical Agents in Hemostasis 1. Davie EW, Ratnoff OD. Waterfall sequence for intrin-
sic blood clotting. Science. 1964;145(3638):1310–2.
Several types of agents are available for intraop- 2. Hoffman M, Monroe DM, Roberts HR. Cellular inter-
actions in hemostasis. Haemostasis. 1996;26(Suppl
erative topical application to stimulate hemosta- 1):12–6.
sis. Products that provide a matrix for coagulation 3. Oliver JA, Monroe DM, Roberts HR, Hoffman
to occur such as collagen and cellulose, those that M. Thrombin activates factor XI on activated plate-
mimic coagulation like fibrin sealants, and prod- lets in the absence of factor XII. Arterioscler Thromb
Vasc Biol. 1999;19(1):170–7.
ucts that combine the use of endogenous and 4. Baglia FA, Walsh PN. Prothrombin is a cofactor for
exogenous coagulation factors are currently the binding of factor XI to the platelet surface and
employed during liver transplant surgery [131, for platelet-mediated factor XI activation by throm-
176]. Results are mixed showing that while there bin. Biochemistry. 1998;37(8):2271–81.
5. Franssen J, Salemink I, Willems GM, Wun TC,
may be a decrease in blood transfusion intra- and Hemker HC, Lindhout T. Prothrombinase is pro-
peri-operatively, there may actually be no mortal- tected from inactivation by tissue factor pathway
ity benefit [177, 178]. A 2003 Cochrane review inhibitor: competition between prothrombin and
[179] and much of the endoscopic literature sup- inhibitor. Biochem J. 1997;323(Pt 1):33–7.
6. Lu G, Broze GJ Jr, Krishnaswamy S. Formation
ports the utility of fibrin sealants; however, a ran- of factors IXa and Xa by the extrinsic pathway:
domized study of 300 liver resection patients differential regulation by tissue factor ­ pathway
found no difference in total blood loss, transfu- inhibitor and antithrombin III. J Biol Chem.
sion requirement, or morbidity with or without 2004;279(17):17241–9.
7. Esmon CT, Stenflo J, Suttie JWA. new vitamin
the product [180]. K-dependent protein. A phospholipid-­ binding
zymogen of a serine esterase. J Biol Chem.
1976;251(10):3052–6.
Summary 8. Esmon CT. The endothelial protein C receptor. Curr
Opin Hematol. 2006;13(5):382–5.
9. Esmon CT, Esmon NL, Harris KW. Complex forma-
Coagulation management is a fascinating and tion between thrombin and thrombomodulin inhibits
rapidly developing subfield in transplantion. It both thrombin-catalyzed fibrin formation and factor
extends across all fields of medicine presenting V activation. J Biol Chem. 1982;257(14):7944–7.
10. Lisman T, Leebeek FW. Hemostatic alterations
challenges for anesthesiologists, hepatologists, in liver disease: a review on pathophysiology,
and liver transplant surgeons. Patients with liver clinical consequences, and treatment. Dig Surg.
disease are in a state of rebalanced hemostasis 2007;24(4):250–8.
that may be affected by a number of factors. A 11. Tripodi A. The coagulopathy of chronic liver dis-
ease: is there a causal relationship with bleeding?
high INR does not necessarily translate to No. Eur J Intern Med. 2010;21(2):65–9.
increased bleeding risk in this patient population. 12. Lisman T, Bongers TN, Adelmeijer J, Janssen HL,
Viscoelastic testing may facilitate better assess- de Maat MP, de Groot PG, et al. Elevated levels of
ment of bleeding risk and coagulation balance von Willebrand Factor in cirrhosis support plate-
188 B. A. Banini and A. J. Sanyal

let adhesion despite reduced functional capacity. arachidonic acid metabolism in response to agonists.
Hepatology. 2006;44(1):53–61. Hepatology (Baltimore, MD). 1988;8(6):1620–6.
13. Monroe DM, Hoffman M. The coagulation cascade 28. Laffi G, La Villa G, Pinzani M, Ciabattoni G,
in cirrhosis. Clin Liver Dis. 2009;13(1):1–9. Patrignani P, Mannelli M, et al. Altered renal and
14. Raya-Sanchez JM, Gonzalez-Reimers E, Rodriguez-­ platelet arachidonic acid metabolism in cirrhosis.
Martin JM, Santolaria-Fernandez F, Molina-Perez M, Gastroenterology. 1986;90(2):274–82.
Rodriguez-Moreno F, et al. Coagulation inhibitors in 29. Laffi G, Marra F, Failli P, Ruggiero M, Cecchi
alcoholic liver cirrhosis. Alcohol (Fayetteville, NY). E, Carloni V, et al. Defective signal transduc-
1998;15(1):19–23. tion in platelets from cirrhotics is associated with
15. Hedner U, Erhardsten E. Hemostatic disorders in increased cyclic nucleotides. Gastroenterology.
liver diseases. In: Schiff ER, Sorrell MF, Maddrey 1993;105(1):148–56.
WC, editors. Diseases of the liver. Philadelphia: 30. Ordinas A, Maragall S, Castillo R, Nurden AT. A
Lippincott Wiliams & Wilkins; 2003. glycoprotein I defect in the platelets of three patients
16. Tripodi A, Salerno F, Chantarangkul V, Clerici M, with severe cirrhosis of the liver. Thromb Res.
Cazzaniga M, Primignani M, et al. Evidence of nor- 1978;13(2):297–302.
mal thrombin generation in cirrhosis despite abnor- 31. Sanchez-Roig MJ, Rivera J, Moraleda JM, Garcia
mal conventional coagulation tests. Hepatology VV. Quantitative defect of glycoprotein Ib in severe
(Baltimore, Md). 2005;41(3):553–8. cirrhotic patients. Am J Hematol. 1994;45(1):10–5.
17. Jennings I, Calne RY, Baglin TP. Predictive value of 32. Pasche B, Ouimet H, Francis S, Loscalzo J. Structural
von Willebrand factor to ristocetin cofactor ratio and changes in platelet glycoprotein IIb/IIIa by plasmin:
thrombin-antithrombin complex levels for hepatic determinants and functional consequences. Blood.
vessel thrombosis and graft rejection after liver trans- 1994;83(2):404–14.
plantation. Transplantation. 1994;57(7):1046–51. 33. Cahill PA, Redmond EM, Sitzmann JV. Endothelial
18. Aster RH. Pooling of platelets in the spleen: role in dysfunction in cirrhosis and portal hypertension.
the pathogenesis of “hypersplenic” thrombocytope- Pharmacol Ther. 2001;89(3):273–93.
nia. J Clin Invest. 1966;45(5):645–57. 34. Desai K, Mistry P, Bagget C, Burroughs AK,
19. Goulis J, Chau TN, Jordan S, Mehta AB, Watkinson Bellamy MF, Owen JS. Inhibition of platelet aggre-
A, Rolles K, et al. Thrombopoietin concentrations gation by abnormal high density lipoprotein parti-
are low in patients with cirrhosis and thrombocyto- cles in plasma from patients with hepatic cirrhosis.
penia and are restored after orthotopic liver trans- Lancet (London, England). 1989;1(8640):693–5.
plantation. Gut. 1999;44(5):754–8. 35. Turitto VT, Baumgartner HR. Platelet interac-
20. Nagamine T, Ohtuka T, Takehara K, Arai T, Takagi tion with subendothelium in a perfusion system:
H, Mori M. Thrombocytopenia associated with hepa- physical role of red blood cells. Microvasc Res.
titis C viral infection. J Hepatol. 1996;24(2):135–40. 1975;9(3):335–44.
21. Levine RF, Spivak JL, Meagher RC, Sieber F. Effect 36. Tripodi A, Primignani M, Chantarangkul V, Clerici
of ethanol on thrombopoiesis. Br J Haematol. M, Dell'Era A, Fabris F, et al. Thrombin g­ eneration
1986;62(2):345–54. in patients with cirrhosis: the role of platelets.
22. Klipstein FA, Lindenbaum J. Folate deficiency in Hepatology (Baltimore, MD). 2006;44(2):440–5.
chronic liver disease. Blood. 1965;25:443–56. 37. Lisman T, Adelmeijer J, de Groot PG, Janssen HL,
23. Ben-Ari Z, Osman E, Hutton RA, Burroughs Leebeek FW. No evidence for an intrinsic plate-
AK. Disseminated intravascular coagulation in let defect in patients with liver cirrhosis—stud-
liver cirrhosis: fact or fiction? Am J Gastroenterol. ies under flow conditions. J Thromb Haemost.
1999;94(10):2977–82. 2006;4(9):2070–2.
24. Carr JM. Disseminated intravascular coagula- 38. Caldwell SH, Hoffman M, Lisman T, Macik BG,
tion in cirrhosis. Hepatology (Baltimore, Md). Northup PG, Reddy KR, et al. Coagulation disor-
1989;10(1):103–10. ders and hemostasis in liver disease: pathophysiol-
25. Ordinas A, Escolar G, Cirera I, Vinas M, Cobo F, ogy and critical assessment of current management.
Bosch J, et al. Existence of a platelet-adhesion defect Hepatology (Baltimore, MD). 2006;44(4):1039–46.
in patients with cirrhosis independent of hema- 39. Brophy MT, Fiore L, Deykin D. Hemostasis. 3rd ed.
tocrit: studies under flow conditions. Hepatology Philadelphia: W.B. Saunders; 1996.
(Baltimore, Md). 1996;24(5):1137–42. 40. Steib A, Gengenwin N, Freys G, Boudjema K, Levy
26. Escolar G, Cases A, Vinas M, Pino M, Calls J, Cirera S, Otteni JC. Predictive factors of hyperfibrinolytic
I, et al. Evaluation of acquired platelet dysfunctions activity during liver transplantation in cirrhotic
in uremic and cirrhotic patients using the platelet patients. Br J Anaesth. 1994;73(5):645–8.
function analyzer (PFA-100): influence of hemato- 41. Pernambuco JR, Langley PG, Hughes RD, Izumi
crit elevation. Haematologica. 1999;84(7):614–9. S, Williams R. Activation of the fibrinolytic system
27. Laffi G, Cominelli F, Ruggiero M, Fedi S, Chiarugi in patients with fulminant liver failure. Hepatology
VP, La Villa G, et al. Altered platelet function in cir- (Baltimore, MD). 1993;18(6):1350–6.
rhosis of the liver: impairment of inositol lipid and
15  Coagulopathy: Pathophysiology, Evaluation, and Treatment 189

42. Gunawan B, Runyon B. The efficacy and safety of 57. Wang W, Hendriks DF, Scharpe
epsilon-aminocaproic acid treatment in patients with SS. Carboxypeptidase U, a plasma carboxypep-
cirrhosis and hyperfibrinolysis. Aliment Pharmacol tidase with high affinity for plasminogen. J Biol
Ther. 2006;23(1):115–20. Chem. 1994;269(22):15937–44.
43. Hu KQ, Yu AS, Tiyyagura L, Redeker AG, 58. Rijken DC, Emeis JJ. Clearance of the heavy and light
Reynolds TB. Hyperfibrinolytic activity in hospital- polypeptide chains of human tissue-type plasmino-
ized cirrhotic patients in a referral liver unit. Am J gen activator in rats. Biochem J. 1986;238(3):643–6.
Gastroenterol. 2001;96(5):1581–6. 59. Einarsson M, Smedsrod B, Pertoft H. Uptake and
44. Boks AL, Brommer EJ, Schalm SW, Van Vliet degradation of tissue plasminogen activator in rat
HH. Hemostasis and fibrinolysis in severe liver fail- liver. Thromb Haemost. 1988;59(3):474–9.
ure and their relation to hemorrhage. Hepatology 60. Amitrano L, Guardascione MA, Brancaccio V,
(Baltimore, MD). 1986;6(1):79–86. Balzano A. Coagulation disorders in liver disease.
45. Leebeek FW, Kluft C, Knot EA, de Maat MP, Semin Liver Dis. 2002;22(1):83–96.
Wilson JH. A shift in balance between profibrino- 61. Violi F, Ferro D, Basili S, Quintarelli C, Musca
lytic and antifibrinolytic factors causes enhanced A, Cordova C, et al. Hyperfibrinolysis resulting
fibrinolysis in cirrhosis. Gastroenterology. from clotting activation in patients with different
1991;101(5):1382–90. degrees of cirrhosis. The CALC Group. Coagulation
46. Ferro D, Quintarelli C, Saliola M, Alessandri C, Abnormalities in Liver Cirrhosis. Hepatology
Basili S, Bonavita MS, et al. Prevalence of hyperfibri- (Baltimore, MD). 1993;17(1):78–83.
nolysis in patients with liver cirrhosis. Fibrinolysis. 62. Van Thiel DH, George M, Fareed J. Low levels of
1993;7:59–62. thrombin activatable fibrinolysis inhibitor (TAFI)
47. Kujovich JL. Hemostatic defects in end stage liver in patients with chronic liver disease. Thromb
disease. Crit Care Clin. 2005;21(3):563–87. Haemost. 2001;85(4):667–70.
48. Stein SF, Harker LA. Kinetic and functional stud- 63. Colucci M, Binetti BM, Branca MG, Clerici C,
ies of platelets, fibrinogen, and plasminogen in Morelli A, Semeraro N, et al. Deficiency of throm-
patients with hepatic cirrhosis. J Lab Clin Med. bin activatable fibrinolysis inhibitor in cirrhosis
1982;99(2):217–30. is associated with increased plasma fibrinolysis.
49. Aoki N, Yamanaka T. The alpha2-plasmin inhibi- Hepatology (Baltimore, MD). 2003;38(1):230–7.
tor levels in liver diseases. Clin Chim Acta. 64. Lisman T, Leebeek FW, Mosnier LO, Bouma BN,
1978;84(1–2):99–105. Meijers JC, Janssen HL, et al. Thrombin-activatable
50. Knot EA, Drijfhout HR, ten Cate JW, de Jong E, fibrinolysis inhibitor deficiency in cirrhosis is not
Iburg AH, Kahle LH, et al. alpha 2-Plasmin inhibi- associated with increased plasma fibrinolysis.
tor metabolism in patients with liver cirrhosis. J Lab Gastroenterology. 2001;121(1):131–9.
Clin Med. 1985;105(3):353–8. 65. Aytac S, Turkay C, Bavbek N, Kosar A. Hemostasis
51. Marongiu F, Mamusa AM, Mameli G, Mulas G, and global fibrinolytic capacity in chronic liver dis-
Solinas A, Demelia L, et al. alpha 2 Antiplasmin and ease. Blood Coagul Fibrinolysis. 2007;18(7):623–6.
disseminated intravascular coagulation in liver cir- 66. Hambleton J, Leung LL, Levi M. Coagulation:
rhosis. Thromb Res. 1985;37(2):287–94. consultative hemostasis. In: American Society
52. Gram J, Jespersen J, Ingeberg S, Bentsen KD, Bach of Hematology Education Program, editor.
E. Plasma histidine-rich glycoprotein and plasmin- Hematology/the Education Program of the American
ogen in patients with liver disease. Thromb Res. Society of Hematology; 2002. p. 335–52.
1985;39(4):411–7. 67. Senzolo M, Burra P, Cholongitas E, Burroughs
53. Leebeek FW, Kluft C, Knot EA, De Maat AK. New insights into the coagulopathy of liver dis-
MP. Histidine-rich glycoprotein is elevated in mild ease and liver transplantation. World J Gastroenterol.
liver cirrhosis and decreased in moderate and severe 2006;12(48):7725–36.
liver cirrhosis. J Lab Clin Med. 1989;113(4):493–7. 68. Porte RJ. Coagulation and fibrinolysis in orthotopic
54. Biland L, Duckert F, Prisender S, Nyman liver transplantation: current views and insights.
D. Quantitative estimation of coagulation factors in Semin Thromb Hemost. 1993;19(3):191–6.
liver disease. The diagnostic and prognostic value 69. Homatas J, Wasantapruek S, Von Kaulla E, Von
of factor XIII, factor V and plasminogen. Thromb Kaulla KN, Eiseman B. Clotting abnormalities fol-
Haemost. 1978;39(3):646–56. lowing orthotopic and heterotopic transplantation of
55. Bajzar L, Manuel R, Nesheim ME. Purification marginally preserved pig livers. Acta Hepatosplenol.
and characterization of TAFI, a thrombin-­ 1971;18(1):14–26.
activable fibrinolysis inhibitor. J Biol Chem. 70. Porte RJ, Bontempo FA, Knot EA, Lewis JH, Kang
1995;270(24):14477–84. YG, Starzl TE. Systemic effects of tissue plasmino-
56. Hendriks D, Wang W, Scharpe S, Lommaert MP, gen activator-associated fibrinolysis and its relation
van Sande M. Purification and characterization of to thrombin generation in orthotopic liver transplan-
a new arginine carboxypeptidase in human serum. tation. Transplantation. 1989;47(6):978–84.
Biochim Biophys Acta. 1990;1034(1):86–92. 71. Iwakiri Y, Groszmann RJ. Vascular endothelial dys-
function in cirrhosis. J Hepatol. 2007;46(5):927–34.
190 B. A. Banini and A. J. Sanyal

72. Gupta TK, Toruner M, Chung MK, Groszmann reporting of the international normalized ratio:
RJ. Endothelial dysfunction and decreased produc- progress and problems. Arch Pathol Lab Med.
tion of nitric oxide in the intrahepatic microcircula- 2007;131(11):1641–7.
tion of cirrhotic rats. Hepatology (Baltimore, MD). 88. Trotter JF, Brimhall B, Arjal R, Phillips C. Specific
1998;28(4):926–31. laboratory methodologies achieve higher model for
73. Rockey DC, Chung JJ. Reduced nitric oxide pro- endstage liver disease (MELD) scores for patients
duction by endothelial cells in cirrhotic rat liver: listed for liver transplantation. Liver Transpl.
endothelial dysfunction in portal hypertension. 2004;10(8):995–1000.
Gastroenterology. 1998;114(2):344–51. 89. Kovacs MJ, Wong A, MacKinnon K, Weir K,
74. Sarela AI, Mihaimeed FM, Batten JJ, Davidson Keeney M, Boyle E, et al. Assessment of the validity
BR, Mathie RT. Hepatic and splanchnic nitric of the INR system for patients with liver impairment.
oxide activity in patients with cirrhosis. Gut. Thromb Haemost. 1994;71(6):727–30.
1999;44(5):749–53. 90. Denson KW, Reed SV, Haddon ME, Woodhams B,
75. Graupera M, Garcia-Pagan JC, Pares M, Abraldes Brucato C, Ruiz J. Comparative studies of rabbit and
JG, Rosello J, Bosch J, et al. Cyclooxygenase-1 inhi- human recombinant tissue factor reagents. Thromb
bition corrects endothelial dysfunction in cirrhotic Res. 1999;94(4):255–61.
rat livers. J Hepatol. 2003;39(4):515–21. 91. Robert A, Chazouilleres O. Prothrombin time in
76. Graupera M, March S, Engel P, Rodes J, Bosch J, liver failure: time, ratio, activity percentage, or inter-
Garcia-Pagan JC. Sinusoidal endothelial COX-1-­ national normalized ratio? Hepatology (Baltimore,
derived prostanoids modulate the hepatic vascular MD). 1996;24(6):1392–4.
tone of cirrhotic rat livers. Am J Physiol Gastrointest 92. Ewe K. Bleeding after liver biopsy does not correlate
Liver Physiol. 2005;288(4):G763–70. with indices of peripheral coagulation. Dig Dis Sci.
77. Iwakiri Y, Groszmann RJ. The hyperdynamic cir- 1981;26(5):388–93.
culation of chronic liver diseases: from the patient 93. Segal JB, Dzik WH. Paucity of studies to support that
to the molecule. Hepatology (Baltimore, MD). abnormal coagulation test results predict bleeding
2006;43(2 Suppl 1):S121–31. in the setting of invasive procedures: an evidence-­
78. Groszmann RJ, Abraldes JG. Portal hyperten- based review. Transfusion. 2005;45(9):1413–25.
sion: from bedside to bench. J Clin Gastroenterol. 94. Diaz LK, Teruya J. Liver biopsy. N Engl J Med.
2005;39(4 Suppl 2):S125–30. 2001;344(26):2030.
79. Wiest R, Groszmann RJ. The paradox of nitric 95. Terjung B, Lemnitzer I, Dumoulin FL, Effenberger W,
oxide in cirrhosis and portal hypertension: too Brackmann HH, Sauerbruch T, et al. Bleeding com-
much, not enough. Hepatology (Baltimore, MD). plications after percutaneous liver biopsy. An analy-
2002;35(2):478–91. sis of risk factors. Digestion. 2003;67(3):138–45.
80. Rand ML, Leung R, Packham MA. Platelet function 96. Bravo AA, Sheth SG, Chopra S. Liver biopsy. N
assays. Transfus Apher Sci. 2003;28(3):307–17. Engl J Med. 2001;344(7):495–500.
81. Rodgers RP, Levin J. A critical reappraisal of 97. Dahlback B. Progress in the understanding of the
the bleeding time. Semin Thromb Hemost. protein C anticoagulant pathway. Int J Hematol.
1990;16:1):1–20. 2004;79(2):109–16.
82. de Franchis R, Arcidiacono PG, Carpinelli L, 98. Reikvam H, Steien E, Hauge B, Liseth K, Hagen KG,
Andreoni B, Cestari L, Brunati S, et al. Randomized Storkson R, et al. Thrombelastography. Transfus
controlled trial of desmopressin plus terlipressin vs. Apher Sci. 2009;40(2):119–23.
terlipressin alone for the treatment of acute vari- 99. Di Cera E. Thrombin as procoagulant and anticoagu-
ceal hemorrhage in cirrhotic patients: a multicenter, lant. J Thromb Haemost. 2007;5(Suppl 1):196–202.
double-blind study. New Italian Endoscopic Club. 100. Bombeli T, Spahn DR. Updates in periopera-
Hepatology (Baltimore, MD). 1993;18(5):1102–7. tive coagulation: physiology and management of
83. Wong AY, Irwin MG, Hui TW, Fung SK, Fan ST, thromboembolism and haemorrhage. Br J Anaesth.
Ma ES. Desmopressin does not decrease blood loss 2004;93(2):275–87.
and transfusion requirements in patients undergoing 101. Kang YG, Martin DJ, Marquez J, Lewis JH,
hepatectomy. Can J Anaesth. 2003;50(1):14–20. Bontempo FA, Shaw BW Jr, et al. Intraoperative
84. Pivalizza EG, Warters RD, Gebhard R. Desmopressin changes in blood coagulation and thrombelasto-
before liver transplantation. Can J Anaesth. graphic monitoring in liver transplantation. Anesth
2003;50(7):748–9. Analg. 1985;64(9):888–96.
85. Kundu SK, Heilmann EJ, Sio R, Garcia C, Davidson 102. Koh MB, Hunt BJ. The management of periopera-
RM, Ostgaard RA. Description of an in vitro plate- tive bleeding. Blood Rev. 2003;17(3):179–85.
let function analyzer—PFA-100. Semin Thromb 103. Stravitz RT, Lisman T, Luketic VA, Sterling RK,
Hemost. 1995;21(Suppl 2):106–12. Puri P, Fuchs M, et al. Minimal effects of acute liver
86. Quick AJ. The prothrombin in hemophilia and in injury/acute liver failure on hemostasis as assessed by
obstructive jaundice. J Biol Chem. 1935;109:73–4. thromboelastography. J Hepatol. 2012;56(1):129–36.
87. Olson JD, Brandt JT, Chandler WL, Van Cott EM, 104. Hendriks HG, van der Meer J, de Wolf JT, Peeters
Cunningham MT, Hayes TE, et al. Laboratory PM, Porte RJ, de Jong K, et al. Intraoperative blood
15  Coagulopathy: Pathophysiology, Evaluation, and Treatment 191

transfusion requirement is the main determinant of 120. Massicotte L, Lenis S, Thibeault L, Sassine MP, Seal
early surgical re-intervention after orthotopic liver RF, Roy A. Effect of low central venous pressure and
transplantation. Transpl Int. 2005;17(11):673–9. phlebotomy on blood product transfusion require-
105. Cacciarelli TV, Keeffe EB, Moore DH, Burns W, ments during liver transplantations. Liver Transpl.
Busque S, Concepcion W, et al. Effect of intraopera- 2006;12(1):117–23.
tive blood transfusion on patient outcome in hepatic 121. Massicotte L, Sassine MP, Lenis S, Seal RF, Roy
transplantation. Arch Surg (Chicago, Ill: 1960). A. Survival rate changes with transfusion of blood
1999;134(1):25–9. products during liver transplantation. Can J Anaesth.
106. Stainsby D, Williamson L, Jones H, Cohen H. 6 2005;52(2):148–55.
Years of shot reporting—its influence on UK blood 122. American Society of Anesthesiologists Task Force
safety. Transfus Apher Sci. 2004;31(2):123–31. on Perioperative Blood Transfusion and Adjuvant
107. de Boer MT, Molenaar IQ, Hendriks HG, Slooff MJ, Therapies. Practice guidelines for perioperative
Porte RJ. Minimizing blood loss in liver transplan- blood transfusion and adjuvant therapies: an updated
tation: progress through research and evolution of report by the American Society of Anesthesiologists
techniques. Dig Surg. 2005;22(4):265–75. Task Force on Perioperative Blood Transfusion
108. Porte RJ, Hendriks HG, Slooff MJ. Blood con- and Adjuvant Therapies. Anesthesiology.
servation in liver transplantation: the role of apro- 2006;105(1):198–208.
tinin. J Cardiothorac Vasc Anesth. 2004;18(4 123. Sindram D, Porte RJ, Hoffman MR, Bentley RC,
Suppl):31S–7S. Clavien PA. Platelets induce sinusoidal endothelial
109. Ramos E, Dalmau A, Sabate A, Lama C, Llado L, cell apoptosis upon reperfusion of the cold ischemic
Figueras J, et al. Intraoperative red blood cell trans- rat liver. Gastroenterology. 2000;118(1):183–91.
fusion in liver transplantation: influence on patient 124. Cywes R, Packham MA, Tietze L, Sanabria JR,
outcome, prediction of requirements, and measures Harvey PR, Phillips MJ, et al. Role of platelets
to reduce them. Liver Transpl. 2003;9(12):1320–7. in hepatic allograft preservation injury in the rat.
110. O’Shaughnessy DF, Atterbury C, Bolton Maggs P, Hepatology (Baltimore, MD). 1993;18(3):635–47.
Murphy M, Thomas D, Yates S, et al. Guidelines for 125. Porte RJ, Blauw E, Knot EA, de Maat MP, de
the use of fresh-frozen plasma, cryoprecipitate and Ruiter C, Minke Bakker C, et al. Role of the
cryosupernatant. Br J Haematol. 2004;126(1):11–28. donor liver in the origin of platelet disorders and
111. Goodnough LT, Johnston MF, Shah T, Chernosky ­hyperfibrinolysis in liver transplantation. J Hepatol.
A. A two-institution study of transfusion practice in 1994;21(4):592–600.
78 consecutive adult elective open-heart procedures. 126. Lesurtel M, Graf R, Aleil B, Walther DJ, Tian Y,
Am J Clin Pathol. 1989;91(4):468–72. Jochum W, et al. Platelet-derived serotonin mediates
112. Dara SI, Rana R, Afessa B, Moore SB, Gajic liver regeneration. Science. 2006;312(5770):104–7.
O. Fresh frozen plasma transfusion in critically ill 127. Himmelreich G, Hundt K, Neuhaus P, Roissant
medical patients with coagulopathy. Crit Care Med. R, Riess H. Decreased platelet aggregation after
2005;33(11):2667–71. reperfusion in orthotopic liver transplantation.
113. Burns ER, Goldberg SN, Wenz B. Paradoxic effect Transplantation. 1992;53(3):582–6.
of multiple mild coagulation factor deficiencies on 128. Himmelreich G, Hundt K, Isenberg C, Bechstein
the prothrombin time and activated partial thrombo- WO, Neuhaus P, Riess H. Thrombocytopenia and
plastin time. Am J Clin Pathol. 1993;100(2):94–8. platelet dysfunction in orthotopic liver transplanta-
114. Dzik WH. Predicting hemorrhage using preoperative tion. Semin Thromb Hemost. 1993;19(3):209–12.
coagulation screening assays. Curr Hematol Rep. 129. Day JR, Punjabi PP, Randi AM, Haskard DO,
2004;3(5):324–30. Landis RC, Taylor KM. Clinical inhibition of the
115. Massicotte L, Beaulieu D, Thibeault L, Roy seven-transmembrane thrombin receptor (PAR1) by
JD, Marleau D, Lapointe R, et al. Coagulation intravenous aprotinin during cardiothoracic surgery.
defects do not predict blood product require- Circulation. 2004;110(17):2597–600.
ments during liver transplantation. Transplantation. 130. Federici AB, Berkowitz SD, Lattuada A, Mannucci
2008;85(7):956–62. PM. Degradation of von Willebrand factor in patients
116. MacLennan S, Williamson LM. Risks of fresh with acquired clinical conditions in which there is
frozen plasma and platelets. J Trauma. 2006;60(6 heightened proteolysis. Blood. 1993;81(3):720–5.
Suppl):S46–50. 131. Porte RJ, Leebeek FW. Pharmacological strategies
117. Holness L, Knippen MA, Simmons L, Lachenbruch to decrease transfusion requirements in patients
PA. Fatalities caused by TRALI. Transfus Med Rev. undergoing surgery. Drugs. 2002;62(15):2193–211.
2004;18(3):184–8. 132. Schalm SW, Terpstra JL, Achterberg JR, Noordhoek
118. Toy P, Popovsky MA, Abraham E, Ambruso DR, Hegt V, Haverkate F, Popescu DT, et al. Orthotopic
Holness LG, Kopko PM, et al. Transfusion-related liver transplantation: an experimental study on
acute lung injury: definition and review. Crit Care mechanisms of hemorrhagic diathesis and thrombo-
Med. 2005;33(4):721–6. sis. Surgery. 1975;78(4):499–507.
119. Fresh frozen plasma use. [Internet]. Available from: 133. de Boer MT, Christensen MC, Asmussen M, van
http://www.shotuk.org. der Hilst CS, Hendriks HG, Slooff MJ, et al. The
192 B. A. Banini and A. J. Sanyal

impact of intraoperative transfusion of platelets and 149. Levi M, Cromheecke ME, de Jonge E, Prins MH,
red blood cells on survival after liver transplantation. de Mol BJ, Briet E, et al. Pharmacological strategies
Anesth Analg. 2008;106(1):32–44; table of contents. to decrease excessive blood loss in cardiac surgery:
134. Dalmau A, Sabate A, Koo M, Rafecas A, Figueras J, a meta-analysis of clinically relevant endpoints.
Jaurrieta E. Prophylactic use of tranexamic acid and Lancet (London, England). 1999;354(9194):1940–7.
incidence of arterial thrombosis in liver transplanta- 150. Theroux MC, Corddry DH, Tietz AE, Miller F,
tion. Anesth Analg. 2001;93(2):516. Peoples JD, Kettrick RG. A study of desmopressin
135. Mor E, Jennings L, Gonwa TA, Holman MJ, Gibbs and blood loss during spinal fusion for neuromus-
J, Solomon H, et al. The impact of operative bleed- cular scoliosis: a randomized, controlled, double-­
ing on outcome in transplantation of the liver. Surg blinded study. Anesthesiology. 1997;87(2):260–7.
Gynecol Obstet. 1993;176(3):219–27. 151. Arshad F, Stoof SC, Leebeek FW, Ruitenbeek K,
136. Grabau CM, Crago SF, Hoff LK, Simon JA, Melton Adelmeijer J, Blokzijl H, et al. Infusion of DDAVP
CA, Ott BJ, et al. Performance standards for does not improve primary hemostasis in patients
therapeutic abdominal paracentesis. Hepatology with cirrhosis. Liver Int. 2015;35(7):1809–15.
(Baltimore, MD). 2004;40(2):484–8. 152. Henry DA, Moxey AJ, Carless PA, O'Connell
137. Lin CH, Shih FY, Ma MH, Chiang WC, Yang CW, D, McClelland B, Henderson KM, et al. Anti-­
Ko PC. Should bleeding tendency deter abdominal fibrinolytic use for minimising perioperative alloge-
paracentesis? Dig Liver Dis. 2005;37(12):946–51. neic blood transfusion. Cochrane Database Syst Rev.
138. Hanson SR, Slichter SJ. Platelet kinetics in patients 2001;1:CD001886.
with bone marrow hypoplasia: evidence for a fixed 153. Kang Y, Lewis JH, Navalgund A, Russell MW,
platelet requirement. Blood. 1985;66(5):1105–9. Bontempo FA, Niren LS, et al. Epsilon-aminocaproic
139. Greeno E, McCullough J, Weisdorf D. Platelet uti- acid for treatment of fibrinolysis during liver trans-
lization and the transfusion trigger: a prospective plantation. Anesthesiology. 1987;66(6):766–73.
analysis. Transfusion. 2007;47(2):201–5. 154. Dalmau A, Sabate A, Acosta F, Garcia-Huete L,
140. Cameron B, Rock G, Olberg B, Neurath D. Evaluation Koo M, Sansano T, et al. Tranexamic acid reduces
of platelet transfusion triggers in a tertiary-­care hos- red cell transfusion better than epsilon-aminocaproic
pital. Transfusion. 2007;47(2):206–11. acid or placebo in liver transplantation. Anesth
141. Rinder HM, Arbini AA, Snyder EL. Optimal dosing Analg. 2000;91(1):29–34.
and triggers for prophylactic use of platelet transfu- 155. Boylan JF, Klinck JR, Sandler AN, Arellano R,
sions. Curr Opin Hematol. 1999;6(6):437–41. Greig PD, Nierenberg H, et al. Tranexamic acid
142. Tinmouth AT, Freedman J. Prophylactic plate- reduces blood loss, transfusion requirements, and
let transfusions: which dose is the best dose? coagulation factor use in primary orthotopic liver
A review of the literature. Transfus Med Rev. transplantation. Anesthesiology. 1996;85(5):1043–
2003;17(3):181–93. 8. discussion 30A-31A.
143. Schlossberg HR, Herman JH. Platelet dosing. 156. van’t Veer C, Mann KG. The regulation of the fac-
Transfus Apher Sci. 2003;28(3):221–6. tor VII-dependent coagulation pathway: rationale
144. American Association of Blood Banks. Blood for the effectiveness of recombinant factor VIIa
transfusion therapy: a physician’s handbook. 7th in refractory bleeding disorders. Semin Thromb
ed. Bethesda, MD: American Association of Blood Hemost. 2000;26(4):367–72.
Banks; 2002. 157. Butenas S, Brummel KE, Bouchard BA, Mann
145. Rebulla P, Finazzi G, Marangoni F, Avvisati G, KG. How factor VIIa works in hemophilia. J Thromb
Gugliotta L, Tognoni G, et al. The threshold for Haemost. 2003;1(6):1158–60.
prophylactic platelet transfusions in adults with 158. Butenas S, Brummel KE, Paradis SG, Mann
acute myeloid leukemia. Gruppo Italiano Malattie KG. Influence of factor VIIa and phospholipids on
Ematologiche Maligne dell’Adulto. N Engl J Med. coagulation in “acquired” hemophilia. Arterioscler
1997;337(26):1870–5. Thromb Vasc Biol. 2003;23(1):123–9.
146. Rebulla P. In vitro and in vivo properties of various 159. Monroe DM, Hoffman M, Oliver JA, Roberts
types of platelets. Vox Sang. 1998;74(Suppl 2):217–22. HR. Platelet activity of high-dose factor VIIa
147. Stanca CM, Montazem AH, Lawal A, Zhang is independent of tissue factor. Br J Haematol.
JX, Schiano TD. Intranasal desmopressin ver- 1997;99(3):542–7.
sus blood transfusion in cirrhotic patients with 160. Pusateri AE, Park MS. Mechanistic implications
coagulopathy undergoing dental extraction: a ran- for the use and monitoring of recombinant acti-
domized controlled trial. J Oral Maxillofac Surg. vated factor VII in trauma. Crit Care. 2005;9(Suppl
2010;68(1):138–43. 5):S15–24.
148. Laupacis A, Fergusson D. Drugs to minimize 161. Dutton RP, McCunn M, Hyder M, D'Angelo M,
perioperative blood loss in cardiac surgery: meta-­ O'Connor J, Hess JR, et al. Factor VIIa for cor-
analyses using perioperative blood transfusion as the rection of traumatic coagulopathy. J Trauma.
outcome. The International Study of Peri-operative 2004;57(4):709–18. discussion 18-9.
Transfusion (ISPOT) Investigators. Anesth Analg. 162. Boehlen F, Morales MA, Fontana P, Ricou B, Irion
1997;85(6):1258–67. O, de Moerloose P. Prolonged treatment of massive
15  Coagulopathy: Pathophysiology, Evaluation, and Treatment 193

postpartum haemorrhage with recombinant factor 171. Planinsic RM, van der Meer J, Testa G, Grande L,
VIIa: case report and review of the literature. BJOG. Candela A, Porte RJ, et al. Safety and efficacy of
2004;111(3):284–7. a single bolus administration of recombinant factor
163. Deveras RA, Kessler CM. Reversal of warfarin-­ VIIa in liver transplantation due to chronic liver dis-
induced excessive anticoagulation with recombinant ease. Liver Transpl. 2005;11(8):895–900.
human factor VIIa concentrate. Ann Intern Med. 172. Lodge JP, Jonas S, Jones RM, Olausson M, Mir-­
2002;137(11):884–8. Pallardo J, Soefelt S, et al. Efficacy and safety of
164. Bernstein DE, Jeffers L, Erhardtsen E, Reddy repeated perioperative doses of recombinant fac-
KR, Glazer S, Squiban P, et al. Recombinant fac- tor VIIa in liver transplantation. Liver Transpl.
tor VIIa corrects prothrombin time in cirrhotic 2005;11(8):973–9.
patients: a preliminary study. Gastroenterology. 173. Pugliese F, Ruberto F, Summonti D, Perrella S,
1997;113(6):1930–7. Cappannoli A, Tosi A, et al. Activated recombi-
165. Lisman T, Leebeek FW, Meijer K, Van Der Meer nant factor VII in orthotopic liver transplantation.
J, Nieuwenhuis HK, De Groot PG. Recombinant Transplant Proc. 2007;39(6):1883–5.
factor VIIa improves clot formation but not fibro- 174. Chavez-Tapia NC, Alfaro-Lara R, Tellez-Avila F,
lytic potential in patients with cirrhosis and during Barrientos-Gutierrez T, Gonzalez-Chon O, Mendez-­
liver transplantation. Hepatology (Baltimore, MD). Sanchez N, et al. Prophylactic activated recombinant
2002;35(3):616–21. factor VII in liver resection and liver transplantation:
166. Kalicinski P, Kaminski A, Drewniak T, Ismail H, systematic review and meta-analysis. PLoS One.
Szymczak M, Markiewicz M, et al. Quick correc- 2011;6(7):e22581.
tion of hemostasis in two patients with fulminant 175. Vincent JL, Rossaint R, Riou B, Ozier Y, Zideman D,
liver failure undergoing liver transplantation by Spahn DR. Recommendations on the use of recom-
recombinant activated factor VII. Transplant Proc. binant activated factor VII as an adjunctive treatment
1999;31(1–2):378–9. for massive bleeding—a European perspective. Crit
167. Hendriks HG, Meijer K, de Wolf JT, Klompmaker Care (London, England). 2006;10(4):R120.
IJ, Porte RJ, de Kam PJ, et al. Reduced transfu- 176. Berrevoet F, de Hemptinne B. Use of topical hemo-
sion requirements by recombinant factor VIIa static agents during liver resection. Dig Surg.
in orthotopic liver transplantation: a pilot study. 2007;24(4):288–93.
Transplantation. 2001;71(3):402–5. 177. Heaton N. Advances and methods in liver sur-
168. Surudo T, Wojcicki M, Milkiewicz P, Czuprynska gery: haemostasis. Eur J Gastroenterol Hepatol.
M, Lubikowski J, Jarosz K, et al. Rapid correction of 2005;17(Suppl 1):S3–12.
prothrombin time after low-dose recombinant factor 178. Chapman WC, Clavien PA, Fung J, Khanna A,
VIIA in patients undergoing orthotopic liver trans- Bonham A. Effective control of hepatic bleeding
plantation. Transplant Proc. 2003;35(6):2323–5. with a novel collagen-based composite combined
169. Niemann CU, Behrends M, Quan D, Eilers H, with autologous plasma: results of a randomized
Gropper MA, Roberts JP, et al. Recombinant factor controlled trial. Arch Surg (Chicago, Ill: 1960).
VIIa reduces transfusion requirements in liver trans- 2000;135(10):1200–4. discussion 5.
plant patients with high MELD scores. Transfus 179. Carless PA, Henry DA, Anthony DM. Fibrin sealant use
Med (Oxford, England). 2006;16(2):93–100. for minimising peri-operative allogeneic blood transfu-
170. Kalicinski P, Markiewicz M, Kaminski A, Laniewski sion. Cochrane Database Syst Rev 2003(2):CD004171.
P, Ismail H, Drewniak T, et al. Single pretransplant 180. Figueras J, Llado L, Miro M, Ramos E, Torras J,
bolus of recombinant activated factor VII amelio- Fabregat J, et al. Application of fibrin glue sealant
rates influence of risk factors for blood loss during after hepatectomy does not seem justified: results
orthotopic liver transplantation. Pediatr Transplant. of a randomized study in 300 patients. Ann Surg.
2005;9(3):299–304. 2007;245(4):536–42.
Physiology, Prevention,
and Treatment of Blood Loss 16
During Liver Transplantation

Simone F. Kleiss, Ton Lisman, and Robert J. Porte

Keywords patients can nowadays be transplanted without


Transfusion · Plasma · Hemostasis · Bleeding · any requirement for blood transfusion [5, 6].
Antifibrinolytics · Coagulopathy Improvements in surgical technique and anesthe-
siological management have contributed to this
major reduction in blood loss, but in addition a
Introduction better understanding of the nature of the abnor-
malities in the hemostatic system have led to a
Historically, bleeding was one of the major chal- more rational approach to the prevention of
lenges during liver transplantation. The first bleeding. Nevertheless, severe and uncontrolla-
patient receiving a liver transplant in 1963 exsan- ble bleeding still occurs and has to be treated
guinated during the procedure [1], and massive appropriately. This chapter will discuss causes of
perioperative blood loss remained a major clini- bleeding during liver transplantation, strategies
cal challenge until the 1980s. Most, if not all, to prevent blood loss, and treatment possibilities
liver transplant procedures required transfusion in case major bleeding does occur.
of blood products in those days, and transfusion
requirements would often exceed 100 units of red
blood cell concentrates (RBCs), with a mean  emostatic Alterations in Liver
H
transfusion requirement around 20–40 units of Disease and During Liver
blood products (RBC, fresh frozen plasma, plate- Transplantation
let concentrates, cryoprecipitate) [2, 3]. Blood
products were, and still are, expensive and The liver is the site of synthesis of most proteins
accounted for a significant part of the total costs involved in initiation, propagation, and regulation
of liver transplantation [4]. In the last 15–20 years, of both coagulation and fibrinolysis. Consequently,
massive blood loss during liver transplantation major alterations in the levels of hemostatic pro-
has become rare and a significant proportion of teins occur in patients with liver disease
(Table  16.1) [7–9]. In addition, a substantially
decreased platelet count is present in a large pro-
S. F. Kleiss, MD • T. Lisman, PhD portion of patients, which may be accompanied by
R. J. Porte, MD, PhD (*)
Section of Hepato-Pancreato-Biliary Surgery and
platelet function defects [10, 11]. Routine diag-
Liver Transplantation, Department of Surgery, nostic tests of hemostasis such as platelet count,
University of Groningen, University Medical Center prothrombin time (PT), and activated partial
Groningen, Groningen, The Netherlands thromboplastin time (APTT) are c­ onsequently fre-
e-mail: r.j.porte@chir.umcg.nl

© Springer International Publishing AG, part of Springer Nature 2018 195


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_16
196 S. F. Kleiss et al.

Table 16.1 Alterations in the hemostatic system in quently abnormal in a patient with liver disease.
patients with liver disease that contribute to bleeding (left)
Abnormal test results have long been interpreted
or counteract bleeding (right)
as suggestive of a bleeding tendency [12]. Recent
Changes that impair Changes that promote advances in the understanding of both clinical and
hemostasis hemostasis
laboratory aspects of hemostasis in liver disease
Thrombocytopenia Elevated levels of von
Willebrand Factor (VWF) have led to an alternate view of the status of the
Platelet function defects Decreased levels of hemostatic system in these patients [13]. We have
ADAMTS-13 coined this alternate view the ‘concept of rebal-
Enhanced production of Elevated levels of factor anced hemostasis’ in patients with liver disease.
nitric oxide and VIII The rebalanced hemostasis theory states that with
prostacyclin
liver disease the hemostatic system is in a rebal-
Low levels of factors II, Decreased levels of protein
V, VII, IX, X, and XI C, protein S, antithrombin,
anced status due to concomitant alterations in both
α2-macroglobulin, and pro- and anticoagulant pathways (Fig. 16.1). This
heparin cofactor II balance is present in patients who may have severe
Vitamin K deficiency Low levels of plasminogen abnormalities in routine hemostasis tests such as
Dysfibrinogenemia the PT (either expressed in seconds or as interna-
Low levels of α2-­ tional normalized ration [INR]), APTT, and plate-
antiplasmin, factor XIII, let count indicating that these tests do not reflect
and TAFI
the true hemostatic status of patients with complex
Elevated t-PA levels
alterations of hemostasis, for example seen with
Source: Reproduced from J Hepatol. 2010;53(2):362–
371, with permission
liver disease [12]. Patients with liver disease thus
do not necessarily have a hemostasis-­ related
bleeding tendency that is suggested by the low

Fig. 16.1  The concept


of rebalanced
hemostasis in patients
with liver disease. In
healthy individuals
(left), hemostasis is in a
solid balance. In patients
with liver disease (right),
concomitant changes in
pro- and antihemostatic
pathways result in a
‘rebalance’ in the
hemostatic system. This Pro- anti- Pro- anti-
new balance, however, hemo- hemo- hemo- hemo-
presumably is less stable static static static static
than the balance in factors factors factors factors
healthy volunteers and
may thus more easily tip
towards either bleeding
or thrombosis Normal liver Cirrhotic liver
16  Physiology, Prevention, and Treatment of Blood Loss During Liver Transplantation 197

platelet count and/or prolonged PT. Many bleed- The hemostatic balance is clinically evident by a
ing complications that occur are unrelated to substantial proportion of patients that can be
deranged hemostasis, but rather related to compli- transplanted without any blood transfusion [5,
cations of portal hypertension, such as esophageal 26–29]. Moreover, recent laboratory data indi-
varices [14, 15]. However, in patients with liver cate a rebalanced platelet-mediated hemostasis as
disease, the hemostatic balance is more easily dis- a result of a hyperreactive von Willebrand factor
turbed as compared to healthy individuals, which system, that is responsible for attachment of
may lead to bleeding but also to thrombotic com- platelets to damaged vasculature [30, 31]. In
plications (summarized in Table 16.1) [13, 16, 17]. addition, despite profoundly prolonged routine
Importantly, current laboratory tests, including laboratory tests of coagulation (PT, APTT), the
many newly developed point-of-care tests fail to coagulation potential appears preserved or even
predict which patients are at risk for either bleed- hyperreactive throughout the transplant proce-
ing or thrombosis. dure when tested with modern thrombin genera-
A thorough review of the pathophysiology of tion tests or thromboelastography [24, 32]. With
coagulation in liver disease and during liver improvements in graft preservation and shorter
transplantation is found elsewhere in this book. cold ischemia times, hyperfibrinolysis is nowa-
Throughout liver transplantation specific days less common.
changes in the hemostatic system occur. During Despite the observation that the hemostatic
the anhepatic phase hemostatic proteins are not balance is frequently relatively well preserved
synthesized, but more importantly, activated during liver transplantation, there are individual
hemostatic proteins and protein-inhibitor com- patients with severe and uncontrollable bleeding
plexes accumulate in the circulation due to a lack that require large amounts of blood products.
of clearance by the liver. As a result, dissemi- Causes of these bleeding complications and treat-
nated intravascular coagulation can develop, ment possibilities will be discussed in this chap-
resulting in consumption of platelets and coagu- ter. In addition, there is increasing recognition of
lation factors and accompanied by secondary the potential for perioperative thrombotic com-
hyperfibrinolysis. Hyperfibrinolysis during the plications. A discussion on diagnosis and treat-
anhepatic phase has been attributed to stable or ment of thromboembolic complications during
increased tissue plasminogen activator (tPA) and and after liver transplantation is discussed else-
decreased plasminogen activator inhibitor-1 where in this book.
(PAI-1) levels [18]. During reperfusion, hyperfi-
brinolysis may further develop as a consequence
of release of tPA from the graft [19–21]. The  auses of Bleeding During Liver
C
degree of hyperfibrinolysis correlates with the Transplantation
severity of ischemia/reperfusion injury of the
hepatic endothelium, the source of tPA upon graft Liver transplantation is a potentially lengthy pro-
reperfusion [19, 22]. Moreover, the liver graft cedure with extensive surgical wound surfaces and
may release heparin-like substances that can fur- transection of collateral veins. Bleeding complica-
ther inhibit coagulation [23]. Additionally, hypo- tions that may occur during the procedure are
thermia, metabolic acidosis, and hemodilution often due to surgical causes, and meticulous surgi-
may adversely affect the hemostatic status during cal hemostasis is important to limit blood loss. In
liver transplantation [5]. Although the additional addition, the presence of portal hypertension may
changes in the hemostatic system during liver contribute to bleeding, as will be discussed below.
transplantation have long been held directly Decreasing portal hypertension by fluid restriction
responsible for the bleeding seen in these patients, and maintenance of a low central venous pressure
accumulating evidence suggest that many liver may be usefull to reduce pressure-associated
transplant recipients may remain in hemostatic bleeding c­omplications [26] and a liberal fluid
balance throughout the procedure [12, 24, 25]. management (including the liberal use of blood
198 S. F. Kleiss et al.

products such as fresh frozen plasma) may aggra- Blood Products


vate the bleeding tendency during liver surgery by
increasing central venous pressure (CVP) and In the early days of liver transplantation, prophy-
splanchnic venous pressure [5, 26, 33–36]. lactic administration of blood products prior to
Strategies to avoid this will be discussed below. the procedure was standard of care. It was believed
Dysfunctional hemostasis may contribute to bleed- that (partial) correction of abnormal hemostasis
ing in some patients, and multiple potential causes tests prior to surgery improves the overall hemo-
may be present. Firstly, hypothermia, metabolic static status of the patients, resulting in a reduced
acidosis, and low ionized calcium levels directly bleeding risk. Consequently, liver transplant pro-
affect the hemostatic system, and prevention and cedures routinely started with administration of
treatment of these complications is important to fresh frozen plasma (FFP) to correct the pro-
prevent bleeding [5]. Secondly, although the role longed PT/APTT, platelet concentrates to reverse
of thrombocytopenia and coagulation factor thrombocytopenia, and cryoprecipitate to increase
defects as a cause of bleeding during liver trans- the circulating level of fibrinogen [5].
plantation has never been convincingly shown, it Administration of RBCs to reverse anemia is
has been established that hyperfibrinolysis is asso- believed to improve hemostasis by virtue of the
ciated with an increased bleeding risk [19, 37, 38]. pivotal role of red blood cells in platelet attach-
Earlier studies suggested that patients with ment to the damaged vasculature in flowing blood
more severe liver disease are at an increased [46]. During the procedure, frequent assessment
bleeding risk, however more recent studies found of the hemostatic status using PT, APTT, platelet
no correlation between disease severity and blood count, and fibrinogen measurements is performed
loss [39]. Furthermore, a steadily decrease in to guide additional administration of blood prod-
transfusion requirements is seen in the last years ucts. Alternatively, thromboelastography may be
despite a progressive increase in MELD score of used to guide transfusion [47–49].
the recipients [29]. The most important predictor Although prophylactic administration of
of blood product use is likely the center (or better blood products prior to and during liver trans-
the surgical or anesthesiology team), an indicator plantation is still common practice in many cen-
that surgical and anesthesiological factors rather ters, little evidence for the efficacy of such a
than a defective hemostatic system are the pri- strategy exists, and there may be valid arguments
mary cause for perioperative bleeding [40, 41]. against prophylactic administration of blood
products [5, 14, 36, 50]. Administration of blood
products is associated with the potential for vol-
 rophylactic Strategies to Prevent
P ume overload and results in elevation of CVP and
Blood Loss splanchnic venous pressure. This is particularly
true in critically ill transplant recipients with a
There are multiple reasons to support a proactive hyperdynamic circulation with increased cardiac
attitude towards prevention of bleeding during liver output and active shunts between the systemic
transplantation. Firstly, a dry surgical field is benefi- and portal venous circulation. In a patient with
cial for the surgeon and lack of bleeding complica- portal hypertension, elevation of CVP by admin-
tions will shorten the procedure. Secondly, excessive istration of blood products may thus paradoxi-
blood loss is associated with a worse outcome also cally induce bleeding by pressure effects rather
due to the direct detrimental effects of blood product than decreasing bleeding risk by improving the
transfusion [42–45]. Finally, reduction of transfu- hemostatic status [34, 35]. Furthermore, adminis-
sion requirements as well as reduction of the dura- tration of blood products is associated with
tion of surgery will save costs. Multiple prophylactic adverse effects and affects morbidity and mortal-
strategies to reduce or avoid bleeding exist, and the ity [26, 27, 51–54]. Normalization of routine
advantages and disadvantages of these strategies will laboratory tests is hardly ever achieved with
be discussed in the following paragraphs. blood products [55]. Recently is has also been
16  Physiology, Prevention, and Treatment of Blood Loss During Liver Transplantation 199

suggested that administration of FFP or cryopre- Pharmacological Agents


cipitate increases the risk for postoperative
venous thrombosis [56]. A major advance in management of bleeding
Rather than administering blood products pro- complications in liver transplantation has been the
phylactically in a patient that is not (yet) bleed- use of antifibrinolytic agents. The serine protease
ing, a wait-and-see policy is increasingly used. In inhibitor aprotinin has been shown to reduce
this scenario, the anesthesiologist and surgeon transfusion requirements during liver transplanta-
accept (profoundly) abnormal PT, APTT, platelet tion by around 30–50% [37, 38, 62], indicating
and fibrinogen levels, as they do not accurately the clinical relevance of the hyperfibrinolytic sta-
reflect the hemostatic status, and commence with tus during liver transplantation. Aprotinin not
the procedure only to initiate blood product trans- only inhibits the fibrinolytic protease plasmin, but
fusion in case of active bleeding complications also has anti-inflammatory properties by virtue of
[5, 36, 57]. Since abnormal preoperative hemo- inhibition of kallikrein and by prevention of the
stasis tests do not appear to predict perioperative activation of the protease activated receptor type 1
bleeding risk and many centers can nowadays (PAR-1) [63]. Administration of aprotinin in liver
transplant a large proportion of patients without transplant patients does not appear to be associ-
any blood transfusions, this wait-and-see policy ated with adverse effects such as thrombosis or
appears justified [5, 26–29]. When active bleed- renal failure [64–66], that have been reported to
ing does occur, administration of blood products occur in cardiac surgery. Despite the apparent
may be guided by conventional laboratory tests excellent risk/benefit profile of aprotinin in liver
or viscoelastic tests such as thromboelastography transplantation, safety concerns in cardiac surgery
[47, 48]. Hypofibrinogenemia may prompt trans- have led to the withdrawal of aprotinin from the
fusion of fibrinogen concentrate or cryoprecipi- market both in the US and Europe. After the with-
tate, whereas evidence of hyperfibrinolysis on the drawal of aprotinin the prevalence of fibrinolysis
thromboelastograph may be a reason to start anti- increased during liver transplantation. This how-
fibrinolytic therapy. ever did not result in an increase in transfusion
There is no consensus about on-demand requirements in centers that switched to different
transfusion strategies, and there is a large antifibrinolytics [67]. The lysine analogues
amount of variability in strategies between cen- tranexamic acid and ε-aminocaproic acid are
ters [40, 43]. The ratio of blood products potentially suitable alternatives for aprotinin [5].
administered in bleeding patients is likely Although both drugs are widely used, only
important, and some authors suggested that tranexamic acid has been shown to reduce trans-
whole blood transfusion may be more appropri- fusion requirements in randomized studies [68–
ate than transfusion of individual blood compo- 70]. When comparing the administration of
nents [58]. In vitro studies have demonstrated aprotinin to tranexamic acid in liver transplanta-
little effect of FFP or platelet transfusion on the tion, no difference has been shown with regards to
hemostatic status in non-bleeding cirrhotics blood loss, transfusion requirements, renal func-
[59, 60] therefore studying alternative strate- tion and 1-year mortality rate [71]. Other pro-
gies would be of great interest. Maintenance of coagulant drugs may also be beneficial in reducing
a specific hematocrit may be effective in main- bleeding. An initial non-controlled trial suggested
taining adequate platelet function, and pro- recombinant factor VIIa (rFVIIa) to reduce trans-
thrombin complex concentrates may be a fusion requirements during liver transplantation
superior alternative to FFP (see next para- [72, 73]. However, two subsequent randomized
graph). Conversely, there is in vitro evidence controlled trials did not show any benefit from
for efficacy of fibrinogen concentrate [61] and routine rFVIIa administration, despite a profound
in vivo proof for efficacy of antifibrinolytic correction of the PT [74–76]. Intraoperative
therapy during liver transplantation (see next administration of factor VIIa during liver trans-
paragraph) [37]. plantation can be associated with higher blood
200 S. F. Kleiss et al.

product use and higher costs [77]. Although pro- 85]. Liver disease and portal hypertension are asso-
phylactic administration of rFVIIa does not ciated with increased plasma volume and disturbed
reduce perioperative blood loss and showed an cardiac function and the administration of fluids
increase in arterial thrombotic events [78], rFVIIa results in a further increase in portal and central
may be an option as ‘rescue agent’ in patients venous pressure, promoting rather than preventing
with intractable bleeding [79]. bleeding tendencies when surgical damage is
Improvement of platelet function parameters, inflicted [14, 33–36, 50, 84]. Avoidance of fluid
in particular shortening of the bleeding time by overload by a very conservative transfusion policy
administration of 1-deamino-8-D-arginine vaso- and by restriction of colloids and/or crystalloids
pressin (DDAVP), does not translate into clinical thus likely reduces bleeding risk. A RCT compar-
improvement of hemostasis. Several studies ing a restrictive transfusion policy and low central
showed no effect of DDVAP on variceal bleeding venous pressure with a liberal transfusion policy
or on blood loss in patients undergoing partial found that the former policy leads to a significant
hepatectomy or liver transplantation [80], indicat- reduction in intraoperative blood loss and transfu-
ing that correction of the bleeding time may not sion requirement, especially during the preanhe-
necessarily result in improvement of hemostasis. patic phase [86]. Liberal use of blood products,
Indeed, an ex vivo study showed no relevant including preoperative correction of abnormal lab-
effects of DDAVP on laboratory indices of pri- oratory values may thus even be counterproduc-
mary hemostasis in patients with cirrhosis, tive, as these blood products increase venous
whereas a clear improvement of these parameters pressure and thereby ‘fuel the fire’. Some groups
were observed in patients with hemophilia A [81]. have taken more drastic steps to maintain a low
A pharmacological pro-hemostatic strategy perioperative CVP by combining fluid restriction
that may have potential, but has not yet been tested protocols with preoperative phlebotomy [26, 85,
in adequately powered clinical studies is the use of 87]. One center has reported that ~80% of patients
prothrombin complex concentrates (PCCs). The could be transplanted without the requirement for
theoretical advantage of PCCs over FFP is the low any transfusion when using fluid restriction in
volume of PCCs that prevents the inevitable rise in combination with preoperative phlebotomy [85]. It
CVP and splanchnic venous pressure associated is important to realize that substantial center and
with FFP infusion. However PCCs only contain a individual experience and acceptance of a low
selection of coagulation factors and its use may be hematocrit (20–25%) is essential for such a strat-
associated with a thrombotic risk. One retrospec- egy. A major concern regarding the use of fluid
tive observational study suggests that administra- restriction protocols is the risk of complications
tion of fibrinogen and/or PCCs does not increase such as air embolism, systemic tissue hypoperfu-
the risk of thrombotic, thromboembolic and isch- sion, and renal failure [27, 85, 88, 89]. Although
emic events in liver transplant patients [82]. An one non-­controlled study showed an increase in
ongoing randomized clinical trials (RCT) will renal failure using a low CVP strategy [89], a num-
assess safety and efficacy of PCCs in reducing ber of other studies, including one RCT, have con-
bleeding during liver transplantation [83]. cluded that fluid restriction during liver
transplantation is safe and does not lead to an
increased incidence of post-operative renal failure
Fluid Restriction [26, 85, 86].

Emerging evidence indicates that the hemostatic


balance during liver transplantation is relatively  urgical and Anesthesiological
S
well maintained [12], and that portal hypertension, Techniques
fluid overload, and the hyperdynamic circulation
are more important determinants of perioperative Surgical experience is an important determinant
bleeding than possible coagulation defects [26, 84, of perioperative bleeding, and specific improve-
16  Physiology, Prevention, and Treatment of Blood Loss During Liver Transplantation 201

ments in surgical technique have therefore been of using these classic laboratory values, the use
instrumental in reducing blood loss [5, 36]. The of viscoelastic tests such as thromboelastography
introduction of venovenous bypass in the 1980s and thromboelastometry may result in a more
has possibly contributed to a reduction of blood rational use of blood products however definitve
loss by avoiding major hemodynamic changes evidence is for this lacking. Viscoelastic testing
during the anhepatic phase [90]. Subsequent can distinguish between a specific platelet or
introduction of the piggyback technique has lead coagulation defect and is the only clinically avail-
to a further significant decrease in transfusion able rapid test that can indicate hyperfibrinolysis.
requirements [91, 92]. A major advantage of the Viscoelastic tests can be used to determine func-
piggyback technique with respect to blood loss is tional platelet count or functional fibrinogen con-
the avoidance of dissection of the retroperito- centration and can guide administration of
neum, that includes multiple collateral veins. But fibrinogen [93, 94], FFP, and fibrinogen and pos-
more importantly, the piggyback technique has sibly antifibrinolytic therapy [47, 49]. Some cen-
enabled reduction of intraoperative fluid load ters use viscoelastic tests for prophylactic
[91, 92]. transfusion of blood products, whereas other cen-
Other anesthesiological interventions that pre- ters only transfuse blood products in case of
vent excessive bleeding include maintenance of active bleeding. There are an increasing number
body temperature, normal pH and ionized cal- of variations of viscoelastic tests on the market
cium levels. Normothermia can be achieved by that differ in the use of coagulation amplifiers/
heating blankets and administration of warm flu- triggers (none, tissue factor, kaolin) or additives
ids [5, 89]. Frequent measurements of serum ion- that specifically neutralize specific components
ized calcium levels and aggressive replacement is of coagulation (heparins, platelets, fibrinolysis).
key especially when large amounts of RBCs or
FFPs are transfused. Citrate that is used as an
anticoagulant in blood products by chelating cal- Adverse Effects of Blood Products
cium. Citrate is metabolized by the liver and
plasma citrate levels may be high with liver dis- Use of blood products varies greatly between
ease, especially during the anhepatic phase centers [40] partly due to differences in the expe-
rience of the teams, but also because of a lack of
uniformity of transfusion protocols. An impor-
 aboratory Monitoring of Bleeding
L tant difference between centers is the choice
and Transfusion between prophylactic administration of blood
products based on pre- and perioperative labora-
Traditionally, laboratory tests such as the PT, tory parameters and an on-demand approach in
APTT, platelet count, fibrinogen level, and hema- which blood products are only transfused when
tocrit were used to guide transfusion. Cut-offs for active bleeding occurs. When deciding to trans-
transfusion differ substantially from center to fuse blood products, one has to weigh the possi-
center and there is little evidence to support blood ble (and in liver transplantation often uncertain)
product transfusion at certain laboratory thresh- benefits against potential adverse events.
olds (e.g., a platelet count below 50 × 109/L or a A number of adverse effects associated with
PT >1.5–2 times the upper limit of normal) in the blood product use are well recognized [52, 95].
absence of active bleeding [36, 37, 43, 50]. Even Although the risk of viral transmission has not
in the presence of active bleeding, target labora- yet been fully eliminated, the chance of contract-
tory values have never been clearly established. ing a virus through blood product transfusion is
However, aiming for a hemostatic profile of extremely low in high-income countries [42, 96].
platelet count >50 × 109/L, PT <1.5–2 times the Transmission of bacteria can still occur, in par-
upper limit of normal and a fibrinogen level of ticular with transfusion of platelet concentrates
1–2 g/L is considered by most reasonable. Instead that are stored at room temperature facilitating
202 S. F. Kleiss et al.

bacterial growth [53, 97, 98]. Hemolytic and out any blood transfusion, suggest that dysfunc-
allergic transfusion reactions have been well tional hemostasis is not necessarily the prime
described but are fortunately relatively rare [95]. cause for perioperative bleeding. Nevertheless,
A recently recognized risk of blood product occasionally patients with dysfunctional hemo-
transfusion is transfusion-related acute lung stasis as the primary cause for excessive blood
injury (TRALI), an antibody mediated transfu- loss are encountered and hyperfibrinolysis is
sion reaction that is rare, but may be fatal [99]. often observed in these patients.
The risk of TRALI appears highest with the use Although clearly defined transfusion thresholds
of FFP, in particular FFP from female donors have not been established, it appears reasonable
[100–102]. Liver surgery (particularly transplan- that in case of larger transfusion requirements,
tation) has been identified as a risk factor for RBC, FFP and platelets should be concomitantly
TRALI [103], emphasizing the need for cautious administered in physiological ratios. On-demand
transfusion of blood products. Blood product use of antifibrinolytics may be considered, espe-
administration results in depression of the cially with evidence of hyperfibrinolysis on throm-
immune response, which—in theory—may be boelastography. Additional studies on the optimal
beneficial to prevent rejections after liver trans- management of intractable bleeding during liver
plantation. However, transfusion-related immune transplantation are required even if it will be diffi-
modulation also increases the incidence of post- cult to achieve adequate power for these studies.
operative infections. Additionally, fluid overload,
also called transfusion associated circulatory
overload (TACO) is a clinically relevant compli- References
cation of transfusion of blood products [54].
Several studies have demonstrated that blood 1. Starzl TE, Marchioro TL, Vonkaulla KN, Hermann
product transfusion is associated with increased G, Brittain RS, Waddell WR. Homotransplantation
morbidity and mortality even after thorough of the Liver in Humans. Surg Gynecol Obstet.
1963;117:659–76.
adjustment for potential confounders [45, 104, 2. Lewis JH, Bontempo FA, Cornell F, Kiss JE, Larson
105]. A dose-effect has been demonstrated, indi- P, Ragni MV, et al. Blood use in liver transplantation.
cating that even in patients who already received Transfusion. 1987;27(3):222–5.
some blood products during the transplantation, 3. Farrar RP, Hanto DW, Flye MW, Chaplin H. Blood
component use in orthotopic liver transplantation.
further minimization of transfusion may be Transfusion. 1988;28(5):474–8.
important [5]. 4. Showstack J, Katz PP, Lake JR, Brown RS Jr,
Dudley RA, Belle S, et al. Resource utilization in
liver transplantation: effects of patient characteristics
and clinical practice. NIDDK Liver Transplantation
 onclusion: A Rational Approach
C Database Group. JAMA. 1999;281(15):1381–6.
to Prevention or Treatment 5. de Boer MT, Molenaar IQ, Hendriks HG, Slooff MJ,
of Bleeding Porte RJ. Minimizing blood loss in liver transplan-
tation: progress through research and evolution of
techniques. Dig Surg. 2005;22(4):265–75.
Increasing laboratory evidence suggests that the 6. Massicotte L, Thibeault L, Roy A. Classical notions
hemostatic system in a patient with liver disease of coagulation revisited in relation with blood losses,
is in a rebalanced situation, and consequently transfusion rate for 700 consecutive liver transplan-
much more competent than suggested by routine tations. Semin Thromb Hemost. 2015;41(5):538–46.
7. Hugenholtz GC, Adelmeijer J, Meijers JC, Porte
laboratory tests such as the PT and platelet count. RJ, Stravitz RT, Lisman T. An unbalance between
During liver transplantation, the hemostatic sta- von Willebrand factor and ADAMTS13 in acute
tus appears to remain in balance, when tested by liver failure: implications for hemostasis and
more sophisticated laboratory tests. These find- clinical outcome. Hepatology (Baltimore, MD).
2013;58(2):752–61.
ings, combined with the clinical observations that 8. van der Werf J, Porte RJ, Lisman T. Hemostasis in
an increasing number of centers report that many patients with liver disease. Acta Gastroenterol Belg.
patients can undergo a liver transplantation with- 2009;72(4):433–40.
16  Physiology, Prevention, and Treatment of Blood Loss During Liver Transplantation 203

9. Lisman T, Leebeek FW, de Groot PG. Haemostatic tion despite prolonged conventional coagulation
abnormalities in patients with liver disease. J tests. J Hepatol. 2010;52(3):355–61.
Hepatol. 2002;37(2):280–7. 25. Warnaar N, Lisman T, Porte RJ. The two tales of
10. Hugenholtz GG, Porte RJ, Lisman T. The platelet coagulation in liver transplantation. Curr Opin
and platelet function testing in liver disease. Clin Organ Transplant. 2008;13(3):298–303.
Liver Dis. 2009;13(1):11–20. 26. Massicotte L, Lenis S, Thibeault L, Sassine MP, Seal
11. Lisman T, Porte RJ. The role of platelets in liver RF, Roy A. Effect of low central venous pressure and
inflammation and regeneration. Semin Thromb phlebotomy on blood product transfusion require-
Hemost. 2010;36(2):170–4. ments during liver transplantations. Liver Transpl.
12. Lisman T, Porte RJ. Rebalanced hemostasis in 2006;12(1):117–23.
patients with liver disease: evidence and clinical 27. Cacciarelli TV, Keeffe EB, Moore DH, Burns W, Busque
consequences. Blood. 2010;116(6):878–85. S, Concepcion W, et al. Effect of intraoperative blood
13. Lisman T, Caldwell SH, Burroughs AK, Northup transfusion on patient outcome in hepatic transplanta-
PG, Senzolo M, Stravitz RT, et al. Hemostasis and tion. Arch Surg (Chicago, Ill: 1960). 1999;134(1):25–9.
thrombosis in patients with liver disease: the ups and 28. Ramos HC, Todo S, Kang Y, Felekouras E, Doyle
downs. J Hepatol. 2010;53(2):362–71. HR, Starzl TE. Liver transplantation without the use
14. Westerkamp AC, Lisman T, Porte RJ. How to mini- of blood products. Arch Surg (Chicago, Ill: 1960).
mize blood loss during liver surgery in patients with 1994;129(5):528–32; discussion 32–3.
cirrhosis. HPB. 2009;11(6):453–8. 29. Jabbour N, Gagandeep S, Shah H, Mateo R, Stapfer
15. Garcia-Tsao G, Bosch J. Management of varices M, Genyk Y, et al. Impact of a transfusion-free pro-
and variceal hemorrhage in cirrhosis. N Engl J Med. gram on non-Jehovah’s Witness patients undergoing
2010;362(9):823–32. liver transplantation. Arch Surg (Chicago, Ill: 1960).
16. Sogaard KK, Horvath-Puho E, Gronbaek H, Jepsen 2006;141(9):913–7.
P, Vilstrup H, Sorensen HT. Risk of venous throm- 30. Lisman T, Bongers TN, Adelmeijer J, Janssen HL,
boembolism in patients with liver disease: a nation- de Maat MP, de Groot PG, et al. Elevated levels of
wide population-based case-control study. Am J von Willebrand Factor in cirrhosis support plate-
Gastroenterol. 2009;104(1):96–101. let adhesion despite reduced functional capacity.
17. Senzolo M, Sartori MT, Lisman T. Should we give Hepatology (Baltimore, MD). 2006;44(1):53–61.
thromboprophylaxis to patients with liver cirrhosis 31. Pereboom IT, Adelmeijer J, van Leeuwen Y,
and coagulopathy? HPB. 2009;11(6):459–64. Hendriks HG, Porte RJ, Lisman T. Development of
18. Roullet S, Freyburger G, Labrouche S, Morisse E, a severe von Willebrand factor/ADAMTS13 dysbal-
Stecken L, Quinart A, et al. Hyperfibrinolysis dur- ance during orthotopic liver transplantation. Am J
ing liver transplantation is associated with bleeding. Transplant. 2009;9(5):1189–96.
Thromb Haemost. 2015;113(5):1145–8. 32. Krzanicki D, Sugavanam A, Mallett S. Intraoperative
19. Porte RJ, Bontempo FA, Knot EA, Lewis JH, Kang hypercoagulability during liver transplantation
YG, Starzl TE. Systemic effects of tissue plasmino- as demonstrated by thromboelastography. Liver
gen activator-associated fibrinolysis and its relation Transpl. 2013;19(8):852–61.
to thrombin generation in orthotopic liver transplan- 33. Jarnagin WR, Gonen M, Maithel SK, Fong Y,
tation. Transplantation. 1989;47(6):978–84. D’Angelica MI, Dematteo RP, et al. A prospective
20. Kang YG, Martin DJ, Marquez J, Lewis JH, randomized trial of acute normovolemic hemodilu-
Bontempo FA, Shaw BW Jr, et al. Intraoperative tion compared to standard intraoperative manage-
changes in blood coagulation and thrombelasto- ment in patients undergoing major hepatic resection.
graphic monitoring in liver transplantation. Anesth Ann Surg. 2008;248(3):360–9.
Analg. 1985;64(9):888–96. 34. Smyrniotis V, Kostopanagiotou G, Theodoraki
21. Lewis JH, Bontempo FA, Awad SA, Kang YG, K, Tsantoulas D, Contis JC. The role of central
Kiss JE, Ragni MV, et al. Liver transplantation: venous pressure and type of vascular control in
intraoperative changes in coagulation factors in blood loss during major liver resections. Am J Surg.
100 first transplants. Hepatology (Baltimore, MD). 2004;187(3):398–402.
1989;9(5):710–4. 35. Jones RM, Moulton CE, Hardy KJ. Central venous
22. Dzik WH, Arkin CF, Jenkins RL, Stump pressure and its effect on blood loss during liver
DC. Fibrinolysis during liver transplantation in resection. Br J Surg. 1998;85(8):1058–60.
humans: role of tissue-type plasminogen activator. 36. Alkozai EM, Lisman T, Porte RJ. Bleeding in liver
Blood. 1988;71(4):1090–5. surgery: prevention and treatment. Clin Liver Dis.
23. Senzolo M, Cholongitas E, Thalheimer U, Riddell 2009;13(1):145–54.
A, Agarwal S, Mallett S, et al. Heparin-like effect 37. Porte RJ, Molenaar IQ, Begliomini B, Groenland
in liver disease and liver transplantation. Clin Liver TH, Januszkiewicz A, Lindgren L, et al. Aprotinin
Dis. 2009;13(1):43–53. and transfusion requirements in orthotopic liver
24. Lisman T, Bakhtiari K, Pereboom IT, Hendriks HG, transplantation: a multicentre randomised double-­
Meijers JC, Porte RJ. Normal to increased thrombin blind study. EMSALT Study Group. Lancet
generation in patients undergoing liver transplanta- (London, England). 2000;355(9212):1303–9.
204 S. F. Kleiss et al.

38. Findlay JY, Rettke SR, Ereth MH, Plevak DJ, Krom plasma and red blood cell transfusions on acute lung
RA, Kufner RP. Aprotinin reduces red blood cell injury and infection risk following liver transplanta-
transfusion in orthotopic liver transplantation: a tion. Liver Transpl. 2011;17(2):149–58.
prospective, randomized, double-blind study. Liver 53. Juffermans NP, Prins DJ, Vlaar AP, Nieuwland R,
Transpl. 2001;7(9):802–7. Binnekade JM. Transfusion-related risk of sec-
39. Massicotte L, Beaulieu D, Roy JD, Marleau D, ondary bacterial infections in sepsis patients: a
Vandenbroucke F, Dagenais M, et al. MELD retrospective cohort study. Shock (Augusta, GA).
score and blood product requirements during 2011;35(4):355–9.
liver transplantation: no link. Transplantation. 54. Li G, Rachmale S, Kojicic M, Shahjehan K,
2009;87(11):1689–94. Malinchoc M, Kor DJ, et al. Incidence and transfu-
40. Ozier Y, Pessione F, Samain E, Courtois F, French sion risk factors for transfusion-associated circula-
Study Group on Blood Transfusion in Liver tory overload among medical intensive care unit
Transplantation. Institutional variability in transfu- patients. Transfusion. 2011;51(2):338–43.
sion practice for liver transplantation. Anesth Analg. 55. Youssef WI, Salazar F, Dasarathy S, Beddow T,
2003;97(3):671–9. Mullen KD. Role of fresh frozen plasma infu-
41. Massicotte L, Sassine MP, Lenis S, Roy sion in correction of coagulopathy of chronic liver
A. Transfusion predictors in liver transplant. Anesth disease: a dual phase study. Am J Gastroenterol.
Analg. 2004;98(5):1245–51; table of contents. 2003;98(6):1391–4.
42. Dodd RY. Current risk for transfusion transmitted 56. Emuakhagbon V, Philips P, Agopian V, Kaldas FM,
infections. Curr Opin Hematol. 2007;14(6):671–6. Jones CM. Incidence and risk factors for deep venous
43. Lopez-Plaza I. Transfusion guidelines and liver thrombosis and pulmonary embolus after liver trans-
transplantation: time for consensus. Liver Transpl. plantation. Am J Surg. 2016;211(4):768–71.
2007;13(12):1630–2. 57. Massicotte L, Beaulieu D, Thibeault L, Roy
44. Spiess BD. Transfusion of blood products affects JD, Marleau D, Lapointe R, et al. Coagulation
outcome in cardiac surgery. Semin Cardiothorac defects do not predict blood product require-
Vasc Anesth. 2004;8(4):267–81. ments during liver transplantation. Transplantation.
45. de Boer MT, Christensen MC, Asmussen M, van 2008;85(7):956–62.
der Hilst CS, Hendriks HG, Slooff MJ, et al. The 58. Laine E, Steadman R, Calhoun L, Blackall D, Levin
impact of intraoperative transfusion of platelets and P, Braunfeld M, et al. Comparison of RBCs and FFP
red blood cells on survival after liver transplantation. with whole blood during liver transplant surgery.
Anesth Analg. 2008;106(1):32–44; table of contents. Transfusion. 2003;43(3):322–7.
46. Lisman T, Caldwell SH, Porte RJ. Anemia as a 59. Tripodi A, Chantarangkul V, Primignani M, Clerici
potential contributor to bleeding in patients with M, Dell’era A, Aghemo A, Mannucci PM. Thrombin
liver disease—neglected but not forgotten. J Hepatol. generation in plasma from patients with cirrhosis
2011;54(3):2. supplemented with normal plasma: considerations
47. Wang SC, Shieh JF, Chang KY, Chu YC, Liu CS, on the efficacy of treatment with fresh-frozen plasma.
Loong CC, et al. Thromboelastography-guided trans- Intern Emerg Med. 2012;7(2):139–44.
fusion decreases intraoperative blood transfusion 60. Tripodi A, Primignani M, Chantarangkul V, Lemma
during orthotopic liver transplantation: randomized L, Jovani M, Rebulla P, Mannucci PM. Global
clinical trial. Transplant Proc. 2010;42(7):2590–3. hemostasis tests in patients with cirrhosis before
48. Tripodi A, Primignani M, Chantarangkul V, Viscardi and after prophylactic platelet transfusion. Liver Int.
Y, Dell’Era A, Fabris FM, et al. The coagulopathy 2013;33(3):362–7.
of cirrhosis assessed by thromboelastometry and its 61. Groeneveld DJ, Adelmeijer J, Hugenholtz GC, Ariëns
correlation with conventional coagulation param- RA, Porte RJ, Lisman T. Ex vivo addition of fibrino-
eters. Thromb Res. 2009;124(1):132–6. gen concentrate improves the fibrin network structure
49. Salooja N, Perry DJ. Thrombelastography. Blood in plasma samples taken during liver transplantation.
Coagul Fibrinolysis. 2001;12(5):327–37. J Thromb Haemost. 2015;13(12):2192–201.
50. Steib A, Freys G, Lehmann C, Meyer C, Mahoudeau 62. Pereboom IT, de Boer MT, Porte RJ, Molenaar
G. Intraoperative blood losses and transfusion IQ. Aprotinin and nafamostat mesilate in liver surgery:
requirements during adult liver transplanta- effect on blood loss. Dig Surg. 2007;24(4):282–7.
tion remain difficult to predict. Can J Anaesth. 63. Poullis M, Manning R, Laffan M, Haskard DO,
2001;48(11):1075–9. Taylor KM, Landis RC. The antithrombotic effect
51. Brems JJ, Hiatt JR, Colonna JO 2nd, el-Khoury of aprotinin: actions mediated via the protease-
G, Quinones WJ, Ramming KP, et al. Variables activated receptor 1. J Thorac Cardiovasc Surg.
influencing the outcome following orthotopic liver 2000;120(2):370–8.
transplantation. Arch Surg (Chicago, Ill: 1960). 64. Molenaar IQ, Porte RJ. Aprotinin and thrombo-
1987;122(10):1109–11. embolism in liver transplantation: is there really a
52. Benson AB, Burton JR Jr, Austin GL, Biggins SW, causal effect? Anesth Analg. 2002;94(5):1367–8;
Zimmerman MA, Kam I, et al. Differential effects of author reply 8.
16  Physiology, Prevention, and Treatment of Blood Loss During Liver Transplantation 205

65. Molenaar IQ, Legnani C, Groenland TH, Palareti G, 77. Scheffert JL, Taber DJ, Pilch NA, McGillicuddy
Begliomini B, Terpstra OT, et al. Aprotinin in ortho- JW, Baliga PK, Chavin KD. Timing of factor VIIa
topic liver transplantation: evidence for a prohemo- in liver transplantation impacts cost and clinical out-
static, but not a prothrombotic, effect. Liver Transpl. comes. Pharmacotherapy. 2013;33(5):483–8.
2001;7(10):896–903. 78. Levi M, Levy JH, Andersen HF, Truloff D. Safety of
66. Molenaar IQ, Begliomini B, Grazi GL, Ringers J, recombinant activated factor VII in randomized clin-
Terpstra OT, Porte RJ, et al. The effect of aprotinin ical trials. N Engl J Med. 2010;363(19):1791–800.
on renal function in orthotopic liver transplantation. 79. Kozek-Langenecker SA, Afshari A, Albaladejo P,
Transplantation. 2001;71(2):247–52. Santullano CA, De Robertis E, Filipescu DC, et al.
67. Schofield N, Sugavanam A, Thompson K, Mallett Management of severe perioperative bleeding: guide-
SV. No increase in blood transfusions during liver lines from the European Society of Anaesthesiology.
transplantation since the withdrawal of aprotinin. Eur J Anaesthesiol. 2013;30(6):270–382.
Liver Transpl. 2014;20(5):584–90. 80. Wong AY, Irwin MG, Hui TW, Fung SK, Fan ST,
68. Dalmau A, Sabate A, Acosta F, Garcia-Huete L, Ma ES. Desmopressin does not decrease blood loss
Koo M, Sansano T, et al. Tranexamic acid reduces and transfusion requirements in patients undergoing
red cell transfusion better than epsilon-aminocaproic hepatectomy. Can J Anaesth. 2003;50(1):14–20.
acid or placebo in liver transplantation. Anesth 81. Arshad Freeha F. Infusion of DDAVP does not
Analg. 2000;91(1):29–34. improve primary hemostasis in patients with cirrho-
69. Boylan JF, Klinck JR, Sandler AN, Arellano R, sis. Liver Int. 2015;35(7):1809–15.
Greig PD, Nierenberg H, et al. Tranexamic acid 82. Kirchner C, Dirkmann D, Treckmann JW, Paul A,
reduces blood loss, transfusion requirements, and Hartmann M, Saner FH, et al. Coagulation man-
coagulation factor use in primary orthotopic liver agement with factor concentrates in liver trans-
transplantation. Anesthesiology. 1996;85(5):1043– plantation: a single-center experience. Transfusion.
8; discussion 30A–31A. 2014;54(10 Pt 2):2760–8.
70. Kaspar M, Ramsay MA, Nguyen AT, Cogswell 83. Arshad Freeha F. Prothrombin complex concentrate in
M, Hurst G, Ramsay KJ. Continuous small-dose the reduction of blood loss during orthotopic liver trans-
tranexamic acid reduces fibrinolysis but not transfu- plantation: PROTON-trial. BMC Surg. 2013;13:22.
sion requirements during orthotopic liver transplan- 84. Balci ST, Pirat A, Torgay A, Cinar O, Sevmis S,
tation. Anesth Analg. 1997;85(2):281–5. Arslan G. Effect of restrictive fluid management and
71. Massicotte L, Denault AY, Beaulieu D, Thibeault acute normovolemic intraoperative hemodilution on
L, Hevesi Z, Roy A. Aprotinin versus tranexamic transfusion requirements during living donor hepa-
acid during liver transplantation: impact on blood tectomy. Transplant Proc. 2008;40(1):224–7.
product requirements and survival. Transplantation. 85. Massicotte L, Perrault MA, Denault AY, Klinck
2011;91(11):1273–8. JR, Beaulieu D, Roy JD, et al. Effects of phlebot-
72. Hendriks HG, Meijer K, de Wolf JT, Klompmaker omy and phenylephrine infusion on portal venous
IJ, Porte RJ, de Kam PJ, et al. Reduced transfu- ­pressure and systemic hemodynamics during liver
sion requirements by recombinant factor VIIa transplantation. Transplantation. 2010;89(8):920–7.
in orthotopic liver transplantation: a pilot study. 86. Feng ZY, Xu X, Zhu SM, Bein B, Zheng SS. Effects
Transplantation. 2001;71(3):402–5. of low central venous pressure during preanhepatic
73. Hendriks HG, Meijer K, de Wolf JT, Porte RJ, phase on blood loss and liver and renal function in liver
Klompmaker IJ, Lip H, et al. Effects of recombinant transplantation. World J Surg. 2010;34(8):1864–73.
activated factor VII on coagulation measured by 87. Hashimoto T, Kokudo N, Orii R, Seyama Y, Sano
thromboelastography in liver transplantation. Blood K, Imamura H, et al. Intraoperative blood salvage
Coagul Fibrinolysis. 2002;13(4):309–13. during liver resection: a randomized controlled trial.
74. Lodge JP, Jonas S, Jones RM, Olausson M, Mir-­ Ann Surg. 2007;245(5):686–91.
Pallardo J, Soefelt S, et al. Efficacy and safety of 88. Melendez JA, Arslan V, Fischer ME, Wuest D,
repeated perioperative doses of recombinant fac- Jarnagin WR, Fong Y, et al. Perioperative outcomes
tor VIIa in liver transplantation. Liver Transpl. of major hepatic resections under low central venous
2005;11(8):973–9. pressure anesthesia: blood loss, blood transfusion,
75. Planinsic RM, van der Meer J, Testa G, Grande L, and the risk of postoperative renal dysfunction. J Am
Candela A, Porte RJ, et al. Safety and efficacy of a Coll Surg. 1998;187(6):620–5.
single bolus administration of recombinant factor 89. Schroeder RA, Collins BH, Tuttle-Newhall
VIIa in liver transplantation due to chronic liver dis- E, Robertson K, Plotkin J, Johnson LB, et al.
ease. Liver Transpl. 2005;11(8):895–900. Intraoperative fluid management during orthotopic
76. Chavez-Tapia NC, Alfaro-Lara R, Tellez-Avila F, liver transplantation. J Cardiothorac Vasc Anesth.
Barrientos-Gutierrez T, Gonzalez-Chon O, Mendez-­ 2004;18(4):438–41.
Sanchez N, et al. Prophylactic activated recombinant 90. Shaw BW Jr, Martin DJ, Marquez JM, Kang
factor VII in liver resection and liver transplantation: YG, Bugbee AC Jr, Iwatsuki S, et al. Venous
systematic review and meta-analysis. PLoS One. bypass in clinical liver transplantation. Ann Surg.
2011;6(7):e22581. 1984;200(4):524–34.
206 S. F. Kleiss et al.

91. Tzakis A, Todo S, Starzl TE. Orthotopic liver trans- 100. Eder AF, Herron RM Jr, Strupp A, Dy B, White J,
plantation with preservation of the inferior vena Notari EP, et al. Effective reduction of transfusion-­
cava. Ann Surg. 1989;210(5):649–52. related acute lung injury risk with male-predominant
92. Jovine E, Mazziotti A, Grazi GL, Ercolani G, plasma strategy in the American Red Cross (2006–
Masetti M, Morganti M, et al. Piggy-back ver- 2008). Transfusion. 2010;50(8):1732–42.
sus conventional technique in liver transplanta- 101. Popovsky MA, Moore SB. Diagnostic and pathoge-
tion: report of a randomized trial. Transpl Int. netic considerations in transfusion-related acute lung
1997;10(2):109–12. injury. Transfusion. 1985;25(6):573–7.
93. Herbstreit F, Winter EM, Peters J, Hartmann 102. Densmore TL, Goodnough LT, Ali S, Dynis M,
M. Monitoring of haemostasis in liver transplanta- Chaplin H. Prevalence of HLA sensitization in female
tion: comparison of laboratory based and point of apheresis donors. Transfusion. 1999;39(1):103–6.
care tests. Anaesthesia. 2010;65(1):44–9. 103. Toy P, Gajic O, Bacchetti P, Looney MR, Gropper
94. Sabate A, Dalmau A. Fibrinogen: a clinical MA, Hubmayr R, Lowell CA, Norris PJ, Murphy
update on liver transplantation. Transplant Proc. EL, Weiskopf RB, Wilson G, Koenigsberg M, Lee
2015;47(10):2925–8. D, Schuller R, Wu P, Grimes B, Gandhi MJ, Winters
95. Alter HJ, Klein HG. The hazards of blood JL, Mair D, Hirschler N, Sanchez Rosen R, Matthay
transfusion in historical perspective. Blood. MA. TRALI Study Group. Transfusion-related
2008;112(7):2617–26. acute lung injury: incidence and risk factors. Blood.
96. Pomper GJ, Wu Y, Snyder EL. Risks of transfusion-­ 2012;119(7):1757–67.
transmitted infections: 2003. Curr Opin Hematol. 104. Pereboom IT, de Boer MT, Haagsma EB, Hendriks
2003;10(6):412–8. HG, Lisman T, Porte RJ. Platelet transfusion dur-
97. Stroncek DF, Rebulla P. Platelet transfusions. Lancet ing liver transplantation is associated with increased
(London, England). 2007;370(9585):427–38. postoperative mortality due to acute lung injury.
98. Koch CG, Li L, Sessler DI, Figueroa P, Hoeltge GA, Anesth Analg. 2009;108(4):1083–91.
Mihaljevic T, et al. Duration of red-cell storage and 105. Hendriks HG, van der Meer J, de Wolf JT, Peeters
complications after cardiac surgery. N Engl J Med. PM, Porte RJ, de Jong K, et al. Intraoperative blood
2008;358(12):1229–39. transfusion requirement is the main determinant of
99. Lee JS, Gladwin MT. Bad blood: the risks of red cell early surgical re-intervention after orthotopic liver
storage. Nat Med. 2010;16(4):381–2. transplantation. Transpl Int. 2005;17(11):673–9.
The Marginal Liver Donor
and Organ Preservation Strategies 17
Abdulrhman S. Elnaggar and James V. Guarrera

Keywords demand for organs. Though there is no consensus,


MELD score · Steatosis · Donation after car- donors with advanced age, hepatic steatosis, sero-
diac death · Donor risk index · Organ preserva- positivity for hepatitis B virus (HBV) and hepati-
tion · Ischemia/reperfusion injury tis C virus (HCV), donation after cardiac death
(DCD) as well as occult malignancy are com-
monly considered extended criteria donors (ECD)
and were until recently considered a contraindica-
Introductions tions for donation (Table 17.1). Many single-cen-
ter experiences have illustrated that utilization of
According to the United Network for Organ such livers allows an expansion of the donor pool
Sharing (UNOS) data, 8,082 liver transplants and reduction of the wait-list mortality at the pro-
have been performed in 2017 with 14,177 patients jected cost of inferior outcomes. In this chapter,
on the waiting list as of March 2018 [1]. The we will highlight these “marginal” donor factors
advances in patient and donor selection, surgical and strategies that have enabled the use of these
techniques, immunosuppression, organ preserva- organs for select recipients.
tion and critical care management have made
lifesaving liver transplantation possible to those
with irrevocable liver damage and acute liver fail-  onor Demographics and Graft
D
ure. Yet the scarcity of organs continues to be a Outcome
major obstacle to the greater application of liver
transplantation (Fig. 17.1). Donor Age
This disequilibrium between supply and
demand has forced transplant programs to use Advanced donor age was previously thought to be
more marginal donors to fulfill the ever-­increasing a relative contraindication to transplantation due to
increased risk of poor graft function. Evidence
A. S. Elnaggar, MD indicates that liver grafts from donors 70 years or
Department of Surgery, Columbia University Medical older have similar outcomes to that of younger
Center, New York, NY, USA donors [2]. Accordingly, UNOS data has shown a
J. V. Guarrera, MD, FACS (*) steady increase in the upper age limit for livers
Division of Liver Transplant and Hepatobiliary used in transplantation. In 1995, 4.9% (n = 216) of
Surgery, Rutgers New Jersey Medical School,
University Hospital, New York, NJ, USA the transplanted livers were above the age of 65,
e-mail: james.guarrera@njms.rutgers.edu which increased to 7.7% (n = 570) in 2015 [3].

© Springer International Publishing AG, part of Springer Nature 2018 207


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_17
208 A. S. Elnaggar and J. V. Guarrera

Fig. 17.1  Number of Deceased Donor Tx Living Donor Tx Liver Waiting List
transplants and size of 15,000
active waiting list
(Source: 2009 OPTN/
SRTR Annual Report)
10,000

5,000

0
1999 2000 2001 2002 2003 2004 2005 2006 2007 2008
SRTR The number of patients awaiting a liver transplant at year-end peaked in 2011:
this is clearly related to the introduction of the MELD/PELD allocation system in 2002.
The number who received a deceased donor liver transplant has gradually increased,
reaching a peak in 2006. The gap between the numbers of candidates and recipients
has been slowly shrinking since 2002.

compliant although these morphologic changes


Table 17.1  Extended criteria donor characteristics that
can affect severity of preservation injury in liver do not necessarily equate to impaired functional
transplantation
capacity. The liver is resilient to the forces of
Elderly donors More susceptible to ischemic aging and has great functional reserve and regen-
(>65 years) endothelial injury erative capacity that allows it to function effec-
Decreased ATP availability on
reperfusion tively after donation. Cumulative experiences
Less tolerant of prolonged cold with advanced age donors report excellent out-
ischemia comes especially with minimal cold ischemic
May have decreased synthetic time (CIT) justifying the use of such organs in
function and regenerative capacity
this era of organ shortage and aging donor and
Underlying liver Macrosteatosis → predisposes to
histopathology early allograft dysfunction and recipient population [4–7]. Careful attention by
primary nonfunction the donor surgeon is paramount in selecting
Ischemic changes/necrosis appropriate elderly donor organs that may have
Significant alcohol features of fibrosis or steatosis [8]. Transmission
abuse → steatohepatitis
Hepatitis B and C activity/portal of occult malignancy is another consideration
inflammation due to the higher incidence of unrecognized
Fibrosis → may be associated with malignancies in the elderly.
hepatitis C or alcohol abuse and
Caution must be exercised with the use of
may affect long-term outcomes
elderly donors in HCV-positive recipients.
Ischemia Donation after cardiac
associated with death → frequently profound Livers with advanced donor age (>60 years)
donor injury ischemia injury increase the risk for deleterious histologic out-
High-dose vasopressors comes and graft failure due to disease recur-
Prolonged or uncorrected
rence [9–11]. However, hepatitis C treatment
hypoxemia or acidosis
has dramatically evolved in the past 5 years
Biochemical Hypernatremia
changes Rising transaminases or bilirubin with the availability of new HCV antivirals that
result in viral clearance in over 95% of patients
[12–14]. As such most transplant centers are
Elderly donors need to be assessed on a case-­ now treating patients on the waiting list prior to
by-­case basis. Liver grafts from donors with transplant. For untreated patients, transplanta-
advanced age tend to be smaller, fibrotic and less tion with an elderly donor or a histologically
17  The Marginal Liver Donor and Organ Preservation Strategies 209

HCV seropositive organ with post transplant with 14–16 or more hours of cold ischemia [21,
HCV antiviral therapy is an excellent strategy to 22]. Beyond the short-term consequences, pro-
gain access to transplantation. longed cold ischemia is also associated with
long-term biliary complications [23]. Extensive
ischemic injury may also induce immunogenicity
Steatosis of the grafts and contribute to acute and chronic
rejection of the grafts [24]. In marginal grafts
About 20–25% of deceased donor liver allografts with risk factors such as steatosis, donation after
are steatotic. The mechanism of hepatic dysfunc- cardiac death (DCD), and donors with advanced
tion in fatty livers is multifaceted. The fatty vacu- age, the CIT should be further minimized ideally
oles in the hepatocytes increase the cell volume to under 6–8 h [25].
and compromise the sinusoidal space [15]. This
alters the microcirculation of the liver. Moreover,
fatty livers are less tolerant of ischemia/reperfu-  epatitis B Virus and Hepatitis C
H
sion injuries associated with cold preservation Virus: Seropositive Donors
[16]. Kupffer cell dysfunction, endothelial cell
necrosis, and intensified leukocyte adhesion and The fear of transmission of HBV and HCV made
lipid peroxidation are also characteristic of stea- using organs from seropositive donors controver-
totic graft dysfunction [17]. sial in the past. The reported risk of HBV trans-
Two histologic patterns of fatty infiltration mission in the recipient is very variable and
can be observed in donor liver biopsies: microve- ranges widely between 15% and 95% [26, 27];
sicular steatosis, in which the cytoplasm con- however transplantation of HBV core antibody
tains diffuse small droplet of fat vacuoles, and seropositive (HBcAb+) grafts is considered safe.
macrovesicular steatosis, in which large vacuole Recipients positive for antibodies against both
deposits displace the nuclei [18, 19]. The pres- hepatitis surface antigen (HBsAb) and core anti-
ence of macrosteatosis adversely affects the gen (HBcAb) have been most resistant to HBV
function of the graft [18, 20], while the presence reactivation with HBcAb+ grafts, whereas those
and extent of the microsteatosis does not appear with no serologic indications for HBV immunity
to impact graft function. Grafts with less than or infection have a theoretical risk of viral activa-
30% of macrosteatosis can be safely used for tion [28–32]. Fortunately, prophylactic antiviral
transplantation, whereas grafts with macroste- agents and hepatitis B immunoglobulin (HBIg)
atosis of 30–60% are at high risk for graft dys- have minimized the risk of HBV transmission by
function and should be used only after careful HBcAb+ grafts in both HBV-naïve and HBV-­
evaluation by an experienced surgeon. Grafts positive recipients [33–35]. Most centers will uti-
with over 60% macrosteatosis are at very high lize single agent antiviral prophylaxis in naive
risk for primary nonfunction (PNF) and should patients receiving HBcAb+ livers. Investigations
be discarded. on HCV-positive vs. HCV-negative grafts dem-
onstrate equivalent graft function and short-term
patient survival when used in HCV-positive
Prolonged Cold Ischemic Time recipients [36–38]. There is also no evidence that
using grafts from donors with dual seropositivity
Even in the era of modern preservation tech- for both HBV and HCV has an effect on graft
niques and solutions and modulation of the function and 5-year survival outcome [39].
hepatic microenvironment, it is of paramount HBcAb+ and HCV-positive allografts will con-
importance to minimize cold ischemic time tinue to be utilized in patients undergoing
(CIT). Ample evidence points to the increasing ­transplantation for HBV and HCV, respectively,
incidents of PNF, early allograft dysfunction and HBcAb+ grafts may be used HBV-naïve
(EAD), and declining graft viability associated patients with appropriate prophylaxis.
210 A. S. Elnaggar and J. V. Guarrera

Historically, Hepatitis C recurrence in HCV-­ and observation of the patient for any possibility
positive recipient was inevitable with comparable of auto-resuscitation.
patient outcomes between HCV-positive and A retrospective analysis of 1567 patients who
-naïve grafts [36–38]. Hepatitis C viremia per- received DCD livers has identified the recipient and
sists in 95% of patients receiving transplant due donor characteristics that affect morbidity and mor-
to HCV cirrhosis [40], and graft damage by HCV tality [52]. Male gender, recipient age over 55, hep-
is expedited, particularly with HCV-1b, com- atitis C seropositivity, African-­American race, the
pared to the indolent course of de novo hepatitis need for hospitalization and life support at the time
C infection in immunocompetent patients [41]. of transplant, and MELD score greater than 35 in
Approximately 25% of HCV transplants experi- recipients were all attributed to graft failure. Donor
ence recurrence of cirrhosis within a median time age greater than 55 or donor weight greater than
interval of 5 years and subsequent decompensa- 100 kg, increasing cold and WIT also correlated
tion 1 year later [9]. Retransplantation for hepati- with morbidity. Several factors were correlated with
tis C is controversial and has suboptimal post-transplant mortality, namely, recipient factors
outcomes. The most recent advances in HCV of age greater than 55, hospitalization at the time of
treatment with newer protease inhibitor such as transplant as well as donor factors of weight greater
Telaprevir and Boceprevir led to the development than 100 kg, and prolonged CIT.
of pre-transplantation treatment protocols achiev- However carefully selected DCD livers
ing seronegativity at the time of transplant and from younger donors with short cold ischemia
preventing HCV recurrence [42]. Moreover, pro- time have superior outcomes compared to
tease inhibitor-based triple therapy in transplant older donors following brain death (or death by
recipients achieved sustained viral response neurological criteria) [53]. A few groups have
ranging from 40% to 60% including patients with used extracorporeal membrane oxygenation
advanced fibroses and previous non-responders (ECMO) to maintain organ perfusion and mini-
[43, 44]. Long-term effects on graft and patient mize WIT during DCD donationwith promis-
survival is yet to be determined. At our center we ing results [54].
treat all HCV positive patients postoperatively Use of fibrinolytic agents such as tPA in the
who were not already cleared pretransplant. The preservation flush solution and instilled in the
current armamentarium of antivirals has an an arterial tree prior to arterial reperfusion has been
outstanding success rate with a sustained viral advocated by small case series. The anesthesiolo-
response (SVR) in greater than 95% of cases. gist of the recipient needs to know if tPA was
used in the DCD donor in case there is hyperfibri-
nolysis after reperfusion. Further study is needed
Donation After Cardiac Death to develop DCD protocols that have reproducible
benefits on outcomes.
The utilization of donation after cardiac death
(DCD) livers grafts has increased steadily, now
comprising approximately 6% of all liver trans- Donor Risk Index
plants [45]. A wider application of DCD has been
hindered by inferior outcomes when compared to In 2006, Feng et al. described a scoring system
standard criteria donor (SCD) [46–49], mainly as that identified donor-specific risk factors and
a sequela of ischemic cholangiopathy and diffuse quantified their effect on outcome [55]. The donor
intrahepatic biliary strictures [49–51]. Unlike the risk index (DRI) was developed by retrospectively
neurologic death where a controlled withdrawal analyzing data over 20,000 liver transplants from
of cardiopulmonary support and aortic cross-­ the Scientific Registry of Transplant Recipients
clamping minimizes warm ischemia time (WIT), (SRTR) from1998 to 2002. The authors identified
DCD is associated with prolonged warm isch- seven risk factors that had an independent associ-
emia time by virtue of the necessary intervention ation with increased graft loss: age, African–
17  The Marginal Liver Donor and Organ Preservation Strategies 211

Table 17.2  Donor risk index = exponent of the sum below


0.154 0.274 0.424
If age >/= 40 and If age >/= 50 and If age >/= 60 and 0.501
Age <50 <60 <70 If age >70
Cause of death (COD) 0.079 0.145 0.184
If COD = anoxia If COD = CVA If COD = other
Donor race 0.176 0.126
If race = African-American If race = other
Donation after cardiac 0.411
death If DCD
Partial/split transplant 0.422
If partial/split
Donor height 0.066170–height/10
Area of organ sharing 0.105 0.244
If regional share If national share
Cold ischemic time 0.010 × cold time

American race of the donor, donor height, cause Alternative Procurement


of death of the donor, and partial or split liver Techniques: Split, Reduced,
transplant (SLT). Two non-­donor factors, CIT and and Adult Living Donor Liver
sharing of grafts outside the local organ sharing Transplant
area, were also significantly and independently
associated with increased graft failure and there- The scarcity of organ donors has expedited the
fore included in the calculation of the DRI. The technical advances in split liver transplant
DRI is calculated according to Table 17.2. It (SLT). SLT can expand the donor pool as each
should not be the only criterion to accept or donor liver can benefit two patients, most com-
decline an organ but may help decisions by quan- monly one pediatric recipient using the left lobe
tifying the donor quality and allows comparison or segments and one adult using the right lobe.
between centers and regions. Results from SLT technique unfortunately have
been accompanied by its own set of morbidi-
ties, including parenchymal leakage of bile,
Malignancy thrombosis of hepatic artery, infection second-
ary to the necrotic tissue remnant, and poor
The incidence of untoward transmission of donor graft function due to insufficient hepatic vol-
malignancy is extremely low. Though the num- ume [60]. Early reports on the outcomes from
ber of reports have been increasing since the cre- SLT from optimal allografts paralleled that of
ation of Disease Transmission Advisory the whole organ transplant of ECD in terms of
Committee in 2005, only eight probable or graft failure and mortality [60, 61]. More recent
proven donor-transmitted malignancies in all investigations found that SLT renders long-
solid organ transplants were recorded in 2013 term outcomes comparable to standard criteria
[56]. CNS tumors, such as medulloblastoma and donor (SCD) and holds promise for another
glioblastoma multiforme [57], along with non- potentially underutilized organ resource [62].
CNS tumors including high-grade melanoma and In the future advances in surgical and anesthetic
choriocarcinoma, were associated with highest techniques may allow the use of full left lobe
transmission risk [58]. Allografts with lymphoma and right lobe split liver transplantation for two
often culminate in dire outcomes, and vigilance appropriate-sized adult recipients. This will
should be exercised to diagnose occult lymphoma require an increased understanding of small for
in donors and to avoid the use of such grafts [59]. size syndrome, portal hyperperfusion, and flow
212 A. S. Elnaggar and J. V. Guarrera

modulation combined with improvements in  echanism of Ischemia


M
liver preservation. and Reperfusion Injury
Nowadays adult-to-adult living donor liver
transplant has excellent outcomes. While donor Understanding organ preservation warrants the
morbidity remains a concern with few well-­ appreciation of the complexity of ischemia/
publicized donor mortalities, a sub-analysis of reperfusion injury (IRI) (Fig. 17.2). As the met-
the adult-to-adult living donor liver transplant abolic rate and ATP requirements for cell sus-
(A2ALL) trial illustrated that donor serologic tainment drop precipitously with decreasing
markers of liver function and transaminases, temperature, most organ preservation tech-
with the exception of platelet count, returned to niques routinely incorporate hypothermia.
baseline within a year post-donation, suggestive However, this artificial cellular ambience of
of good hepatic functional recovery [63]. As the hypothermia and anoxia results in the disrup-
deceased donor pool remains limited, living tion of chemiosmotic gradients and structural
donor transplantation remains an excellent tech- integrity of the membrane phospholipid
nique to extend patient access to transplanta- bilayer. Hypothermia alters the polarity and
tion. Center volume and anesthetic and surgical permeability of plasma membrane as well as
experience are important factors that ensure the the activity of membrane-bound enzymes cul-
success of an living donor liver transplant minating in cell swelling. Ischemia necessi-
program. tates the transition from aerobic to anaerobic
metabolism, creating acidosis and reducing
ATP production. Consequently, the activity of
Organ Preservation enzymes such as Na+–K+-ATPase and Ca2+-
ATPase that maintain the chemiosmotic gradi-
Since the beginning of orthotopic liver trans- ent diminishes, exacerbating the disrupted
plantation, the optimization of the graft has ionic traffic. Calcium influx in particular
been a collaborative effort between donor and induces calmodulin and phospholipase activa-
recipient sites. Interventions to optimize the tion, alteration in mitochondrial activity and
graft condition can take place as precondition- vasospasm by its action on myofibrils, pro-
ing, organ preservation, and post-conditioning longing and exacerbating ischemia. Hence, the
in the donor site, as well as en route to the preservation solution is hypertonic and con-
recipient site and during the process of trans- tains impermeants in order to minimize cellu-
plantation. Injuries to the allograft can occur in lar edema.
at least three phases: warm ischemia, cold On a cellular level, ischemia and reperfusion
ischemia, and reperfusion. Warm ischemia activates Kupffer cells, which release chemokines
starts with aortic cross-clamping in brain dead/ like tumor necrosis factor-a (TNF-a). Subsequent
dead by neurological criteria donors or with- release of interleukin-8 (IL-8) recruits neutrophils
drawal of cardiac support or impaired hemody- to the ischemic area. The interaction between neu-
namic status with donation after cardiac death trophils and sinusoidal endothelium occurs via
(DCD). Cold ischemia is iatrogenic, initiated intercellular adhesion molecule-1 (ICAM-1),
by the flushing of the liver with cold preserva- selectin and integrin, that result in extravasation
tion solution. Reperfusion injury is incurred of inflammatory cells. Lysosomal enzymes,
when the allograft is reperfused after compet- hydrogen peroxide, nitric oxide, and endothelin
ing the anastomosis to the recipient. The scope perpetuate further structural destruction [64].
of this chapter will be to describe the patho- Sinusoidal endothelial cells (SECs) are known to
physiology of organ preservation injury and be more susceptible to cold ischemia than warm
techniques to minimize such insults, which is ischemia [65, 66]. There is growing evidence that
of particular interest in optimizing the use of both cold and warm ischemia also damage biliary
ECD livers. epithelial cells [67].
17  The Marginal Liver Donor and Organ Preservation Strategies 213

Fig. 17.2 (a) Simplified a Change in


diagram of preservation
polarity,
injury due to
Permeability of
hypothermia and
membrane
ischemia. (b) Simplified
Hypothermia Cellular Der anged chemiosmotic
schematic of ischemia/
edema gradient
reperfusion injury.
Ischemia-reperfusion Change in activity
activates Kupffer cells of membrane-bound
which release TNF-a, enzymes
IL-8, and other
chemokines. Neutrophil
is activated, recruited,
Acidosis
and infiltrated the
sinusoidal endothelial Aerobic → Anaerobic Disrupted
layer to damage Ischemia chemical
metabolism
hepatocytes. ROS from gradient
Kupffer cell and Reduced ATP
neutrophil incur tissue production
injury. Endothelin (ET-1) b
activates Kupffer cells as
well as stellate cell (Ito
cell) resulting in Neutrophil extravasation
Space of Disse
vasoconstriction and via ICAM-1, selectin, integrin
further ischemia. SEC
Lysosomal enzymes, Sinusoid
nitric oxide (not shown),
and complement TNF-α, IL-1, IL-8
activation also contribute Tissue Injury
Chemokines
to further injury (SEC:
sinusoidal endothelial
cell)
Kupffer cell Neutrophil
Ischemia
ET-1
ROS

SEC apoptosis

Stellate Cell

Hepatocyte Bile cannaliculus

Interestingly, more damage is incurred during ferent levels. ROS destroys the microvasculature in
reperfusion than ischemia. For instance, though liver, perpetuating local anoxia after reperfusion
cold ischemia damages sinusoids, mounting evi- [72], impairs mitochondrial function, and induces
dence points to the notion that reperfusion results lipid peroxidation [73]. Reperfusion injury is also
in apoptosis of sinusoidal endothelial cells [68]. mediated by cytokines and nitric oxide [69].
Such fatal injuries occur mainly via reactive oxy-
gen species (ROS) such as superoxide radical,
hydroxyl, and hydrogen peroxide [69], synthesized  rgan Preservation and Modalities
O
from Kupffer cells, neutrophils via cytosolic xan- to Attenuate Ischemia/Reperfusion
thine oxidase [70]. Such a sudden and tremendous Injury
oxidative stress eclipses the compensatory abilities
of endogenous antioxidants such as superoxide dis- As previously mentioned, IRI is associated with
mutase, glutathione, catalase, and beta-carotene PNF, EAD, acute as well as chronic rejection and
[71]. The consequent damages are observed on dif- intrahepatic biliary stricture. Hypothermia
214 A. S. Elnaggar and J. V. Guarrera

reduces the metabolic rate at the cost of chemios- (kidney) by Dr. Belzer in 1967 utilized the
motic derangements. Hence, successful organ machine perfusion technology, the convenience
preservation with organ preservation solutions of standard cold storage had largely replaced
entails a balance of attenuating metabolic strain HMP until its recent revival [79, 80]. The advan-
by hypothermia and ameliorating IRI. One of the tages of HMP are (1) continuous supply of meta-
earliest used solutions was Euro-Collins that sim- bolic substrates for ATP production, (2) washout/
ulates the intracellular chemiosmotic composi- dilution of waste products such as lactic acid and
tion in order to mitigate cell swelling. However, ROS, (3) assessment of organ viability and
the maximal possible CIT in an ex vivo rat model ­functionality prior to transplant, and (4) intraop-
of liver preservation with Euro-Collins was only erative therapeutic interventions including down-
8 h [74]. This was also the perceived CIT regulation of the mRNA from precursors of IRI
limit when used in clinical settings in the early such as TNF-a, IL-8, and ICAM-1 (Fig. 17.3).
days of liver transplantation. Other preservation Interest in HMP techniques for the liver has
solutions, such as histidine-tryptophan-a-­recently returned not only due to its superiority to
­
ketoglutarate solution [74, 75], University of SCS in preserving SCD liver but also because of
Wisconsin solution (UW) [76], Celsior [77], and improved outcomes including reducing IRI in
Polysol [78], further ameliorated IRI in standard rodent [78, 81] and swine models [82] of ECD
cold storage (SCS) and hypothermic machine graft liver transplantation.
perfusion (HMP) for different organ grafts Increasingly clinical experience in HMP has
including the liver. A novel solution called been gained. Guarrera et al. demonstrated in a
Vasosol is considered to have enhanced vasodila- phase I clinical trial that HMP of SCD human
tory and antioxidant capacity with clinical evi- liver grafts results in shorter postoperative
dence of improved early graft function and recovery time, diminished biliary complication,
survival benefits [79]. Despite the variable com- and attenuated serum markers of IRI [79, 83]
ponents in preservation solutions, the essential (Fig. 17.4). Over the past 5 years, there has been
components include buffers and impermeants to excellent clinical experience with HMP in small
diminish cellular edema, enriched with metabolic series including those recovered from extended
substrates, amino acids, free radical scavengers, criteria donors [84] and donation after cardiac
as well as vasodilators. death (DCD) donors [85, 86]. While some vari-
A notable advance in liver preservation is ability exists in technique, all clinical HMP
hypothermic machine perfusion (HMP). series to date have reported improved outcomes
Although the inception of organ preservation with reductions in early allograft dysfunction,

Washout of Toxics and


Ex Vivo Modeling
Anaerobic End Products

Maintenance of
Economic Benefit
Vasoconstriction

Increases Donor Pool: Machine Administration of


ECD, DCD Perfusion Cryoprotective Agents

Fig. 17.3 Advantages
of hypothermic machine On Pump Assessment Provide Nutrients:
of Organs ATP, Oxygen, Glucose
perfusion. The left
column lists outcome
benefits, while the right Endothelial Protection:
Pharmacological Sustains Expression of
column outlines Interventions Flow Related Genes
mechanistic advantages
17  The Marginal Liver Donor and Organ Preservation Strategies 215

Fig. 17.4 (a) Schematic


a
diagram of hypothermic
machine perfusion. (b)
Liver graft undergoing
hypothermic machine
perfusion (Ao Aorta,
CHA Common hepatic Heat 40 µm filter
artery, IVC Inferior vena Bubble exchanger
(disposable)
cava, PV Portal vein) chamber

Closed circuit
tubing

Pump head

b IVC effluent cannula


Temperature probe

Ao IVC
CHA

PV

Pressure
monitor
Cannulae

biliary complications and reduced hospital of “ischemic time” are less relevant using
length of stay. These benefits, together with the NMP. U.S. IDE trials are scheduled by two
development of innovative portable HMP companies that produce normothermic perfu-
devices and further adoption by more centers sion devices. The European COPE (Consortium
worldwide, have “broken the ice” for more for Organ Preservation in Europe) trial of NMP
widespread use of HMP and subsequent has enrolled over 200 patients up to now and the
expanded studies of the benefits of MP in liver results are pending.
transplantation. While comparing HMP and NMP is beyond
Despite the success of HMP, optimal tem- the scope of this chapter, both HMP and NMP
peratures and protocols remain controversial have shown reductions in clinically relevant
[87]. Normothermic MP (NMP) is currently markers of Ischemia/Reperfusion Injury and in
undergoing clinical evaluation in Europe with some trials improved clinical endpoints and with
promising early clinical reports [88–90]. It the ability to use of organs that had been declined
appears that previously considered limitations by other transplant centers [84, 89].
216 A. S. Elnaggar and J. V. Guarrera

 uture Directions in Liver


F 6. Hoofnagle JH, Lombardero M, Zetterman RK, Lake
J, Porayko M, Everhart J, Belle SH, Detre KM. Donor
Preservation age and outcome of liver transplantation. Hepatology.
1996;24(1):89–96.
The study of ex vivo machine perfusion of the 7. Paterno F, Wima K, Hoehn RS, Cuffy MC, Diwan
liver has thrived and the optimal MP techniques TS, Woodle SE, Abbott DE, Shah SA. Use of elderly
allografts in liver transplantation. Transplantation.
and protocols are under intense investigations.
2016;100(1):153–8.
Strategies under investigation include enhancing 8. Rull R, Vidal O, Momblan D, González FX, López-­
perfusate, optimizing temperature, perfusion Boado MA, Fuster J, Grande L, Bruguera M, Cabrer
pressure, and flow. The performance of perfusate K, García-Valdecasas JC. Evaluation of potential liver
donors: limits imposed by donor variables in liver
may further improve with agents such as anti-­
transplantation. Liver Transpl. 2003;9(4):389–93.
apoptotic drugs, vasodilators, inhibitors of 9. Rayhill SC, Wu YM, Katz DA, Voigt MD, Labrecque
inflammatory cytokines, antioxidants, and matrix DR, Kirby PA, Mitros FA, Kalil RS, Miller RA,
metalloproteinase inhibitors [91]. Stolpen AH, Schmidt WN. Older donor livers
show early severe histological activity, fibrosis, and
Other technical advances such as periopera-
graft failure after liver transplantation for hepatitis
tive carbon monoxide inhalation [91, 92], phar- C. Transplantation. 2007;84(3):331–9.
macological or ischemic preconditioning [93] of 10. Mutimer DJ, Gunson B, Chen J, Berenguer J, Neuhaus
the liver graft or direct therapeutic interventions P, Castaing D, Garcia-Valdecasas JC, Salizzoni
M, Moreno GE, Mirza D. Impact of donor age and
may be incorporated into HMP protocol in the
year of transplantation on graft and patient survival
future. IRI may be further mitigated by introduc- following liver transplantation for hepatitis C virus.
ing superoxide dismutase gene by adenovirus Transplantation. 2006;81(1):7–14.
[94]. Novel therapeutic interventions to “defat” 11. Condron SL, Heneghan MA, Patel K, Dev A,
McHutchison JG, Muir AJ. Effect of donor age on
steatotic allografts, leading to improved graft
survival of liver transplant recipients with hepatitis C
function are also being explored in a small ani- virus infection. Transplantation. 2005;80(1):145–8.
mal model. 12. Pungpapong S, Aqel BA, Koning L, Murphy JL,

Henry TM, Ryland KL, Yataco ML, Satyanarayana
R, Rosser BG, Vargas HE, Charlton MR, Keaveny
AP. Multicenter experience using telaprevir or
Summary boceprevir with peginterferon and ribavirin to treat
hepatitis C genotype 1 after liver transplantation.
Improvements in donor management, organ pres- Liver Transpl. 2013;19(7):690–700.
13. Coilly A, Roche B, Dumortier J, Leroy V, Botta-­
ervation and attenuation of IRI hold promise in
Fridlund D, Radenne S, Pageaux GP, Si-Ahmed SN,
allowing safe expansion of the donor pool and Guillaud O, Antonini TM, Haïm-Boukobza S, Roque-­
improvement of outcomes in Liver Transplantation. Afonso AM, Samuel D, Duclos-Vallée JC. Safety
and efficacy of protease inhibitors to treat hepatitis C
after liver transplantation: a multicenter experience. J
Hepatol. 2014;60(1):78–86.
References 14. Coilly A, Dumortier J, Botta-Fridlund D, Latournerie
M, Leroy V, Pageaux GP, Agostini H, Giostra E,
1. https://www.unos.org/. Moreno C, Roche B, Antonini TM, Guillaud O,
2. Emre S, Schwartz ME, Altaca G, Sethi P, Fiel MI, Guy Lebray P, Radenne S, Saouli AC, Calmus Y, Alric
SR, Kelly DM, Sebastian A, Fisher A, Eickmeyer D, L, Debette-Gratien M, De Ledinghen V, Durand F,
Sheiner PA, Miller CM. Safe use of hepatic allografts Duvoux C, Samuel D, Duclos-Vallée JC. Multicenter
from donors older than 70 years. Transplantation. experience with boceprevir or telaprevir to treat
1996;62(1):62–5. ­hepatitis c recurrence after liver transplantation: when
3. https://optn.transplant.hrsa.gov/data/view-data-reports/ present becomes past, what lessons for future? PLoS
national-data/. One. 2015;10(9):e0138091.
4. Oh CK, Sanfey HA, Pelletier SJ, Sawyer RG, 15. Takeda Y, Arii S, Kaido T, Niwano M, Moriga T,
McCullough CS, Pruett TL. Implication of advanced Mori A, Hanaki K, Gorrin-Rivas MJ, Ishii T, Sato
donor age on the outcome of liver transplantation. M, Imamura M. Morphologic alteration of hepa-
Clin Transplant. 2000;14(4 Pt 2):386–90. tocytes and sinusoidal endothelial cells in rat fatty
5. Wall WJ, Mimeault R, Grant DR, Bloch M. The liver during cold preservation and the protective
use of older donor livers for hepatic transplantation. effect of hepatocyte growth factor. Transplantation.
Transplantation. 1990;49(2):377–81. 1999;67(6):820–8.
17  The Marginal Liver Donor and Organ Preservation Strategies 217

16. Fukumori T, Ohkohchi N, Tsukamoto S, Satomi


29. Dickson RC, Everhart JE, Lake JR, Wei Y, Seaberg
S. Why is fatty liver unsuitable for transplantation? EC, Wiesner RH, Zetterman RK, Pruett TL, Ishitani
Deterioration of mitochondrial ATP synthesis and MB, Hoofnagle JH. Transmission of hepatitis B by
sinusoidal structure during cold preservation of a liver transplantation of livers from donors positive for anti-
with steatosis. Transplant Proc. 1997;29(1–2):412–5. body to hepatitis B core antigen. The National Institute
17. Lemasters JJ, Peng XX, Bachmann S, Currin RT, of Diabetes and Digestive and Kidney Diseases
Gao W, Thurman RG. Dual role of Kupffer cell acti- Liver Transplantation Database. Gastroenterology.
vation and endothelial cell damage in reperfusion 1997;113(5):1668–74.
injury to livers stored for transplantation surgery. J 30. Roche B, Samuel D, Gigou M, Feray C, Virot V,
Gastroenterol Hepatol. 1995;10(Suppl 1):S84–7. Schmets L, David MF, Arulnaden JL, Bismuth A,
18. Markin RS, Wisecarver JL, Radio SJ, Stratta RJ,
Reynes M, Bismuth H. De novo and apparent de novo
Langnas AN, Hirst K, Shaw BW. Frozen section hepatitis B virus infection after liver transplantation. J
evaluation of donor livers before transplantation. Hepatol. 1997;26(3):517–26.
Transplantation. 1993;56(6):1403–9. 31. Dodson SF, Issa S, Araya V, Gayowski T, Pinna

19. Fishbein TM, Fiel MI, Emre S, Cubukcu O, Guy SR, A, Eghtesad B, Iwatsuki S, Montalvo E, Rakela
Schwartz ME, Miller CM, Sheiner PA. Use of livers J, Fung JJ. Infectivity of hepatic allografts with
with microvesicular fat safely expands the donor pool. antibodies to hepatitis B virus. Transplantation.
Transplantation. 1997;64(2):248–51. 1997;64(11):1582–4.
20. Trevisani F, Colantoni A, Caraceni P, Van Thiel
32. Uemoto S, Sugiyama K, Marusawa H, Inomata Y,
DH. The use of donor fatty liver for liver trans- Asonuma K, Egawa H, Kiuchi T, Miyake Y, Tanaka K,
plantation: a challenge or a quagmire? J Hepatol. Chiba T. Transmission of hepatitis B virus from hepa-
1996;24(1):114–21. titis B core antibody-positive donors in living related
21. Porte RJ, Ploeg RJ, Hansen B, van Bockel JH,
liver transplants. Transplantation. 1998;65(4):494–9.
Thorogood J, Persijn GG, Hermans J, Terpstra 33. Nery JR, Gedaly R, Vianna R, Berho M, Weppler D,
OT. Long-term graft survival after liver transplanta- Levi D, Kato T, Khan F, Pinna A, Schiff E, Tzakis
tion in the UW era: late effects of cold ischemia and A. Are liver grafts from hepatitis B surface anti-
primary dysfunction. European Multicentre Study gen negative/anti-hepatitis B core antibody positive
Group. Transpl Int. 1998;11(Suppl 1):S164–7. donors suitable for transplantation? Transplant Proc.
22. Ploeg RJ, D'Alessandro AM, Knechtle SJ, Stegall
2001;33(1–2):1521–2.
MD, Pirsch JD, Hoffmann RM, Sasaki T, Sollinger 34. Joya-Vazquez PP, Dodson FS, Dvorchik I, Gray E,
HW, Belzer FO, Kalayoglu M. Risk factors for pri- Chesky A, Demetris AJ, Shakil O, Fung JJ, Vargas
mary dysfunction after liver transplantation—a multi- HE. Impact of anti-hepatitis Bc-positive grafts on the
variate analysis. Transplantation. 1993;55(4):807–13. outcome of liver transplantation for HBV-related cir-
23. Scotté M, Dousset B, Calmus Y, Conti F, Houssin D, rhosis. Transplantation. 2002;73(10):1598–602.
Chapuis Y. The influence of cold ischemia time on 35. Loss GE, Mason AL, Blazek J, Dick D, Lipscomb J,
biliary complications following liver transplantation. Guo L, Perrillo RP, Eason JD. Transplantation of liv-
J Hepatol. 1994;21(3):340–6. ers from hbc Ab positive donors into HBc Ab nega-
24. Hariharan S, Johnson CP, Bresnahan BA, Taranto SE, tive recipients: a strategy and preliminary results. Clin
McIntosh MJ, Stablein D. Improved graft survival Transplant. 2001;15(Suppl 6):55–8.
after renal transplantation in the United States, 1988 36. Salizzoni M, Lupo F, Zamboni F, Franchello A,

to 1996. N Engl J Med. 2000;342(9):605–12. Lavezzo B, Cerutti E, Strignano P. Outcome of
25. Yersiz H, Shaked A, Olthoff K, Imagawa D, Shackleton patients transplanted with liver from hepatitis C posi-
C, Martin P, Busuttil RW. Correlation between donor tive donors. Transplant Proc. 2001;33(1–2):1507–8.
age and the pattern of liver graft recovery after trans- 37. Velidedeoglu E, Desai NM, Campos L, Olthoff KM,
plantation. Transplantation. 1995;60(8):790–4. Shaked A, Nunes F, Zeldin G, Stewart C, Blumberg
26. Castells L, Vargas V, Rodríguez-Frías F, Allende
E, Abrams J, Markmann JF. Effect of donor
H, Jardí R, Margarit C, Esteban R, Guardia ­hepatitis C on liver graft survival. Transplant Proc.
J. Transmission of hepatitis B virus by transplantation 2001;33(7–8):3795–6.
of livers from donors positive for antibody to hepatitis 38. Ghobrial RM, Steadman R, Gornbein J, Lassman C,
B core antigen. Transplant Proc. 1999;31(6):2464–5. Holt CD, Chen P, Farmer DG, Yersiz H, Danino N,
27. Uemoto S, Inomata Y, Sannomiya A, Koshiba T,
Collisson E, Baquarizo A, Han SS, Saab S, Goldstein
Kurokawa T, Takatsuki M, Hino H, Yokoi A, Tanaka LI, Donovan JA, Esrason K, Busuttil RW. A 10-year
K. Posttransplant hepatitis B infection in liver trans- experience of liver transplantation for hepatitis C:
plantation with hepatitis B core antibody-positive analysis of factors determining outcome in over 500
donors. Transplant Proc. 1998;30(1):134–5. patients. Ann Surg. 2001;234(3):384–93. discussion
28. Wachs ME, Amend WJ, Ascher NL, Bretan PN,
393-4.
Emond J, Lake JR, Melzer JS, Roberts JP, Tomlanovich 39. Saab S, Chang AJ, Comulada S, Geevarghese SK,
SJ, Vincenti F. The risk of transmission of hepatitis Anselmo RD, Durazo F, Han S, Farmer DG, Yersiz H,
B from HBsAg(−), HBcAb(+), HBIgM(−) organ Goldstein LI, Ghobrial RM, Busuttil RW. Outcomes
donors. Transplantation. 1995;59(2):230–4. of hepatitis C- and hepatitis B core antibody-­positive
218 A. S. Elnaggar and J. V. Guarrera

grafts in orthotopic liver transplantation. Liver donors after cardiac death: the untold story. Surgery.
Transpl. 2003;9(10):1053–61. 2009;146(4):543–52. discussion 552-3.
40. Wright TL, Donegan E, Hsu HH, Ferrell L, Lake JR, 51. Abt P, Crawford M, Desai N, Markmann J, Olthoff
Kim M, Combs C, Fennessy S, Roberts JP, Ascher K, Shaked A. Liver transplantation from con-
NL. Recurrent and acquired hepatitis C viral infec- trolled non-heart-beating donors: an increased inci-
tion in liver transplant recipients. Gastroenterology. dence of biliary complications. Transplantation.
1992;103(1):317–22. 2003;75(10):1659–63.
41. Gane EJ, Portmann BC, Naoumov NV, Smith
52. Mathur AK, Heimbach J, Steffick DE, Sonnenday
HM, Underhill JA, Donaldson PT, Maertens G, CJ, Goodrich NP, Merion RM. Donation after cardiac
Williams R. Long-term outcome of hepatitis C death liver transplantation: predictors of outcome. Am
infection after liver transplantation. N Engl J Med. J Transplant. 2010;10(11):2512–9.
1996;334(13):815–20. 53. Scalea JR, Redfield RR, Foley DP. Liver trans-

42. Verna EC, O'Leary JG. Hepatitis C treatment in
plant outcomes using ideal donation after cir-
patients on the liver transplant waiting list. Curr Opin culatory death livers are superior to using older
Organ Transplant. 2015;20(3):242–50. donation after brain death donor livers. Liver Transpl.
43. Burton JR, O'Leary JG, Verna EC, Saxena V, Dodge 2016;22(9):1197–204.
JL, Stravitz RT, Levitsky J, Trotter JF, Everson GT, 54. Lazzeri C, Bonizzoli M, Valente S, Cianchi G,

Brown RS, Terrault NA. A US multicenter study of Migliaccio ML, Gensini GF, Peris A. The role of
hepatitis C treatment of liver transplant recipients extracorporeal membrane oxygenation in dona-
with protease-inhibitor triple therapy. J Hepatol. tion after circulatory death. Minerva Anestesiol.
2014;61(3):508–14. 2014;80(11):1217–27.
44. Verna EC, Saxena V, Burton JR, O'Leary JG, Dodge 55. Feng S, Goodrich NP, Bragg-Gresham JL, Dykstra
JL, Stravitz RT, Levitsky J, Trotter JF, Everson GT, DM, Punch JD, DebRoy MA, Greenstein SM, Merion
Brown RS, Terrault NA, Consortium C-C. Telaprevir- RM. Characteristics associated with liver graft failure:
and boceprevir-based triple therapy for hepatitis C the concept of a donor risk index. Am J Transplant.
in liver transplant recipients with advanced recur- 2006;6(4):783–90.
rent disease: a multicenter study. Transplantation. 56. Green M, Covington S, Taranto S, Wolfe C, Bell W,
2015;99(8):1644–51. Biggins SW, Conti D, DeStefano GD, Dominguez
45. Singhal A, Wima K, Hoehn RS, Quillin RC, Woodle E, Ennis D, Gross T, Klassen-Fischer M, Kotton C,
ES, Paquette IM, Paterno F, Abbott DE, Shah LaPointe-Rudow D, Law Y, Ludrosky K, Menegus
SA. Hospital resource use with donation after cardiac M, Morris MI, Nalesnik MA, Pavlakis M, Pruett T,
death allografts in liver transplantation: a matched Sifri C, Kaul D. Donor-derived transmission events in
controlled analysis from 2007 to 2011. J Am Coll 2013: a report of the Organ Procurement Transplant
Surg. 2015;220(5):951–8. Network Ad Hoc Disease Transmission Advisory
46.
Selck FW, Grossman EB, Ratner LE, Renz Committee. Transplantation. 2015;99(2):282–7.
JF. Utilization, outcomes, and retransplantation of 57. Kauffman HM, McBride MA, Cherikh WS, Spain
liver allografts from donation after cardiac death: PC, Delmonico FL. Transplant tumor registry: donors
implications for further expansion of the deceased-­ with central nervous system tumors1. Transplantation.
donor pool. Ann Surg. 2008;248(4):599–607. 2002;73(4):579–82.
47. Yamamoto S, Wilczek HE, Duraj FF, Groth CG,
58. Myron Kauffman H, McBride MA, Cherikh WS,

Ericzon BG. Liver transplantation with grafts from Spain PC, Marks WH, Roza AM. Transplant tumor
controlled donors after cardiac death: a 20-year registry: donor related malignancies. Transplantation.
follow-up at a single center. Am J Transplant. 2002;74(3):358–62.
2010;10(3):602–11. 59. Nalesnik MA, Woodle ES, Dimaio JM, Vasudev B,
48. Pine JK, Aldouri A, Young AL, Davies MH, Attia M, Teperman LW, Covington S, Taranto S, Gockerman
Toogood GJ, Pollard SG, Lodge JP, Prasad KR. Liver JP, Shapiro R, Sharma V, Swinnen LJ, Yoshida A,
transplantation following donation after cardiac Ison MG. Donor-transmitted malignancies in organ
death: an analysis using matched pairs. Liver Transpl. transplantation: assessment of clinical risk. Am J
2009;15(9):1072–82. Transplant. 2011;11(6):1140–7.
49. Grewal HP, Willingham DL, Nguyen J, Hewitt
60. Alkofer B, Samstein B, Guarrera JV, Kin C, Jan D,
WR, Taner BC, Cornell D, Rosser BG, Keaveny Bellemare S, Kinkhabwala M, Brown R, Emond JC,
AP, Aranda-Michel J, Satyanarayana R, Harnois D, Renz JF. Extended-donor criteria liver allografts.
Dickson RC, Kramer DJ, Hughes CB. Liver trans- Semin Liver Dis. 2006;26(3):221–33.
plantation using controlled donation after cardiac 61. Olthoff KM, Merion RM, Ghobrial RM, Abecassis
death donors: an analysis of a large single-center MM, Fair JH, Fisher RA, Freise CE, Kam I, Pruett
experience. Liver Transpl. 2009;15(9):1028–35. TL, Everhart JE, Hulbert-Shearon TE, Gillespie BW,
50. Skaro AI, Jay CL, Baker TB, Wang E, Pasricha
Emond JC, Group AAS. Outcomes of 385 adult-­
S, Lyuksemburg V, Martin JA, Feinglass JM, to-­
adult living donor liver transplant recipients: a
Preczewski LB, Abecassis MM. The impact of isch- report from the A2ALL Consortium. Ann Surg.
emic cholangiopathy in liver transplantation using 2005;242(3):314–23. discussion 323-5.
17  The Marginal Liver Donor and Organ Preservation Strategies 219

62. Hong JC, Yersiz H, Busuttil RW. Where are we


78. Bessems M, Doorschodt BM, van Vliet AK, van Gulik
today in split liver transplantation? Curr Opin Organ TM. Machine perfusion preservation of the non-heart-­
Transplant. 2011;16(3):269–73. beating donor rat livers using polysol, a new preserva-
63. Trotter JF, Gillespie BW, Terrault NA, Abecassis
tion solution. Transplant Proc. 2005;37(1):326–8.
MM, Merion RM, Brown RS, Olthoff KM, Hayashi 79. Guarrera JV, Henry SD, Samstein B, Odeh-Ramadan
PH, Berg CL, Fisher RA, Everhart JE, Adult-to-Adult R, Kinkhabwala M, Goldstein MJ, Ratner LE, Renz
Living Donor Liver Transplantation Cohort Study JF, Lee HT, Brown RS, Emond JC. Hypothermic
Group. Laboratory test results after living liver dona- machine preservation in human liver transplan-
tion in the adult-to-adult living donor liver transplanta- tation: the first clinical series. Am J Transplant.
tion cohort study. Liver Transpl. 2011;17(4):409–17. 2010;10(2):372–81.
64. Jaeschke H. Molecular mechanisms of hepatic ischemia- 80. Moers C, Smits JM, Maathuis MH, Treckmann J, van
reperfusion injury and preconditioning. Am J Physiol Gelder F, Napieralski BP, van Kasterop-Kutz M, van
Gastrointest Liver Physiol. 2003;284(1):G15–26. der Heide JJ, Squifflet JP, van Heurn E, Kirste GR,
65.
Gao W, Bentley RC, Madden JF, Clavien Rahmel A, Leuvenink HG, Paul A, Pirenne J, Ploeg
PA. Apoptosis of sinusoidal endothelial cells is a RJ. Machine perfusion or cold storage in deceased-­
critical mechanism of preservation injury in rat liver donor kidney transplantation. N Engl J Med.
transplantation. Hepatology. 1998;27(6):1652–60. 2009;360(1):7–19.
66. Caldwell-Kenkel JC, Currin RT, Tanaka Y, Thurman 81. Lee CY, Zhang JX, Jones JW, Southard JH, Clemens
RG, Lemasters JJ. Kupffer cell activation and endo- MG. Functional recovery of preserved livers following
thelial cell damage after storage of rat livers: effects warm ischemia: improvement by machine perfusion
of reperfusion. Hepatology. 1991;13(1):83–95. preservation. Transplantation. 2002;74(7):944–51.
67. Vajdová K, Smreková R, Kukan M, Lutterová
82. Iwamoto H, Matsuno N, Narumi Y, Uchiyama M,
M, Wsólová L. Bile analysis as a tool for assess- Kozaki K, Degawa H, Hama K, Kikuchi K, Takeuchi
ing integrity of biliary epithelial cells after cold H, Kozaki M, Nagao T. Beneficial effect of machine
ischemia—reperfusion of rat livers. Cryobiology. perfusion preservation on liver transplantation
2000;41(2):145–52. from non-heart-beating donors. Transplant Proc.
68. Clavien PA, Harvey PR, Strasberg SM. Preservation 2000;32(7):1645–6.
and reperfusion injuries in liver allografts. An over- 83. Guarrera JV, Henry SD, Chen SW, Brown T, Nachber
view and synthesis of current studies. Transplantation. E, Arrington B, Boykin J, Samstein B, Brown RS,
1992;53(5):957–78. Emond JC, Lee HT. Hypothermic machine preser-
69. Kupiec-Weglinski JW, Busuttil RW. Ischemia and
vation attenuates ischemia/reperfusion markers after
reperfusion injury in liver transplantation. Transplant liver transplantation: preliminary results. J Surg Res.
Proc. 2005;37(4):1653–6. 2011;167(2):e365–73.
70. Jaeschke H. Reactive oxygen and ischemia/reper-
84. Guarrera JV, Henry SD, Samstein B, Reznik E, Musat
fusion injury of the liver. Chem Biol Interact. C, Lukose TI, Ratner LE, Brown RS, Kato T, Emond
1991;79(2):115–36. JC. Hypothermic machine preservation facilitates
71. Atalla SL, Toledo-Pereyra LH, MacKenzie GH,
successful transplantation of “orphan” extended crite-
Cederna JP. Influence of oxygen-derived free radi- ria donor livers. Am J Transplant. 2015;15(1):161–9.
cal scavengers on ischemic livers. Transplantation. 85. Dutkowski P, Schlegel A, de Oliveira M, Müllhaupt
1985;40(6):584–90. B, Neff F, Clavien PA. HOPE for human liver grafts
72. Menger MD. Microcirculatory disturbances sec-
obtained from donors after cardiac death. J Hepatol.
ondary to ischemia-reperfusion. Transplant Proc. 2014;60(4):765–72.
1995;27(5):2863–5. 86. Dutkowski P, Polak WG, Muiesan P, Schlegel A,

73. McCord JM. Oxygen-derived free radicals in postisch- Verhoeven CJ, Scalera I, DeOliveira ML, Kron P,
emic tissue injury. N Engl J Med. 1985;312(3):159–63. Clavien PA. First comparison of hypothermic oxy-
74.
Momii S, Koga A. Time-related morphologi- genated perfusion versus static cold storage of
cal changes in cold-stored rat livers. A compari- human donation after cardiac death liver transplants:
son of Euro-Collins solution with UW solution. an international-matched case analysis. Ann Surg.
Transplantation. 1990;50(5):745–50. 2015;262(5):764–70. discussion 770-1.
75. Gebhard MM, Bretschneider HJ, Gersing E, Preusse 87. Karangwa SA, Dutkowski P, Fontes P, Friend PJ,

CJ, Schnabel PA, Ulbricht LJ. Calcium-free cardio- Guarrera JV, Markmann JF, Mergental H, Minor T,
plegia—pro. Eur Heart J. 1983;4(Suppl H):151–60. Quintini C, Selzner M, Uygun K, Watson CJ, Porte
76. Belzer FO, Southard JH. Principles of solid-organ RJ. Machine perfusion of donor livers for trans-
preservation by cold storage. Transplantation. plantation: a proposal for standardized nomencla-
1988;45(4):673–6. ture and reporting guidelines. Am J Transplant.
77. Menasché P, Termignon JL, Pradier F, Grousset C, 2016;16:2932–42.
Mouas C, Alberici G, Weiss M, Piwnica A, Bloch 88. Ravikumar R, Jassem W, Mergental H, Heaton N,
G. Experimental evaluation of Celsior, a new heart Mirza D, Perera MT, Quaglia A, Holroyd D, Vogel
preservation solution. Eur J Cardiothorac Surg. T, Coussios CC, Friend PJ. Liver transplantation
1994;8(4):207–13. after ex vivo normothermic machine preservation: a
220 A. S. Elnaggar and J. V. Guarrera

phase 1 (first-in-man) clinical trial. Am J Transplant. reperfusion injury through p38 MAP kinase pathway.
2016;16(6):1779–87. Hepatology. 2002;35(4):815–23.
89. Mergental H, Perera MT, Laing RW, Muiesan P, Isaac 92. Kaizu T, Nakao A, Tsung A, Toyokawa H, Sahai R,
JR, Smith A, Stephenson BT, Cilliers H, Neil DA, Geller DA, Murase N. Carbon monoxide inhalation
Hübscher SG, Afford SC, Mirza DF. Transplantation ameliorates cold ischemia/reperfusion injury after rat
of declined liver allografts following normothermic ex-­ liver transplantation. Surgery. 2005;138(2):229–35.
situ evaluation. Am J Transplant. 2016;16:3235–45. 93.
de Rougemont O, Lehmann K, Clavien
90. Watson CJ, Kosmoliaptsis V, Randle LV, Russell
PA. Preconditioning, organ preservation, and postcon-
NK, Griffiths WJ, Davies S, Mergental H, Butler ditioning to prevent ischemia-reperfusion injury to the
AJ. Preimplant normothermic liver perfusion of a liver. Liver Transpl. 2009;15(10):1172–82.
suboptimal liver donated after circulatory death. Am J 94. Lehmann TG, Wheeler MD, Froh M, Schwabe RF,
Transplant. 2016;16(1):353–7. Bunzendahl H, Samulski RJ, Lemasters JJ, Brenner
91. Amersi F, Shen XD, Anselmo D, Melinek J, Iyer DA, Thurman RG. Effects of three superoxide dis-
S, Southard DJ, Katori M, Volk HD, Busuttil RW, mutase genes delivered with an adenovirus on graft
Buelow R, Kupiec-Weglinski JW. Ex vivo expo- function after transplantation of fatty livers in the rat.
sure to carbon monoxide prevents hepatic ischemia/ Transplantation. 2003;76(1):28–37.
Pediatric Liver Transplantation
18
Philipp J. Houck

Keywords the late 1960’s eight pediatric patients survived


Biliary atresia · Metabolic liver disease · the initial transplantation, only to face difficulties
Fulminant hepatic failure · Hepatic encepha- with immunosupression. The introduction of
lopathy · Infant · Children cyclosporine A in 1978 made acceptable long-­
term survival rates possible [2] and liver trans-
plantation became standard of care in the 1980’s
Introduction for liver failure and end-stage liver disease. The
resulting shortage of organs for small children
Anesthetic management of pediatric liver trans- triggered further surgical innovations in the late
plants is in many ways very different from taking 80’s and early 90’s such as living-donor liver and
care of adult liver transplant recipients. Different split liver donations. The introduction of the
indications combined with the distinct physiology PELD score in 2002 shifted waitlist priority for
of infants and children result in a pathophysiology organ allocation from time on the waiting list to
that requires specific skill sets and techniques. the severity of the disease. This evolution over
With meticulous attention to details results in almost 50 years lead to today’s excellent long
pediatric liver transplantation are excellent and term outcome after pediatric liver transplantation
very gratifying. This chapter will review basic with one and five year survival rates of 90% and
indications and management for infants, toddlers, 85%, respectively [3]. Problems related to life-­
pre-teenagers and teenagers undergoing cadaveric long immunosupression and donor scarcity how-
or living donor liver transplants. ever remain challenges.

 istory of Pediatric Liver


H  llocation of Organs for Pediatric
A
Transplantation Liver Transplantation.

The history of pediatric liver transplantation In the United States the allocation of organs is
started with the first unsuccessful liver transplan- overseen since 1986 by the Organ Procurement
tation in 1963 by Thomas Starzl in Denver [1]. In Transplantation Network (OPTN). Organ alloca-
tion was initially based on time on the waiting list
P. J. Houck, MD and home, hospital or ICU location as a surrogate
Department of Anesthesiology, Columbia University for the severity of illness. However studies found
Medical Center, New York, NY, USA that waitlist time had no correlation with death,
e-mail: pjh2001@cumc.columbia.edu

© Springer International Publishing AG, part of Springer Nature 2018 221


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_18
222 P. J. Houck

except for status 1 patients. Until 2002, patients continue to include the value assigned for age
needing liver transplants were grouped into four (<1 year) until the patient reached the age of
medical urgency categories and this system did 24 months) The score is multiplied by 10 and
not take the urgency or the actual severity of the rounded to the nearest whole number. Additional
illness in consideration. In 1995 a group of trans- points are given for hepatopulmonary syndrome,
plant physicians from the United States and urea cycle defects and hepatic neoplasms [6]. In
Canada formed a collaborative research group, general, pediatric organs are given to pediatric
the Studies of Pediatric Liver Transplantation patients. Patients with acute liver failure with an
(SPLIT) to share data and create a national data- expected life expectancy of 7 days or less catego-
base of pediatric liver transplants. Based on the rized as status one (usually less then 1% of all
data from the SPLIT group the Pediatric Endstage listed patients) and these patients have the highest
Liver Disease (PELD) score was established in priority independent of PELD score. Most patients
2002 [4]. The PELD score was developed to pre- who received allocated organs achieved high
dict the mortality or ICU admission of a patient PELD scores through special exceptions points or
within the next 3 months without a liver trans- receive transplants as status one patients.
plantation using growth failure, albumin, biliru- The introduction of the PELD score led to fewer
bin, INR and age at the time of listing and is valid healthy patients on the waiting list as there was no
for patients younger than 12 years [4, 5]. The benefit in listing patients early during their disease
PELD score is calculated using the formula in process. Initially there was concern that this system
Table  18.1. Scores for patients listed for liver would lead to worse outcomes, since organs are allo-
cated to sicker patients, however the SPLIT research
Table 18.1  Metabolic diseases of the liver group demonstrated that post-transplant survival
C. was similar with either allocation system [7].
A. Causes hepatic
Structural adenomas or
damage to B. hepatocellular
liver Extrahepatic damage carcinoma
Age Distribution
Alpha-1-­ Urea cycle Glycogen
antitrypsin disorders storage disease Between 1988 and 2015 less than one third of all
deficiency type I and III transplanted pediatric patients were younger than
Cystic Hyperoxaluria Hereditary 12 months, 20% were older than 10 years
fibrosis tyrosinemia (Fig.  18.1). The highest proportion of living
Wilson Tyrosenemia type I Galactosemia donor transplantation was used for recipients
disease
under 1 year (Fig. 18.2).
Familial Alpha-1-­
hypercholesteremia antitrypsin
Sixty-five percent of patients under 1 were
deficiency transplanted for biliary atresia. The most com-
Organic acidemias PFIC type II mon indication for patients older than 13 years
was unspecific type of cirrhosis; fulminant liver
transplantation before the patient’s first birthday

Age distribution of pediatric liver transplants in the US


< 1 Year
200 1-5 Years
Number/year

6-10 Years
11-17 Years
Fig. 18.1 Age
100
distribution of pediatric
liver transplantations in
the US. (2016 data;
Organ Procurement and 0
Transplantation 07 08 09 10 11 12 13 14 15 16
Network) 20 20 20 20 20 20 20 20 20 20
18  Pediatric Liver Transplantation 223

Fig. 18.2 Distribution
of deceased and living 250
donor liver
transplantation in 200
different age groups.
(2016 data; Organ
150
Procurement and Living Donor
Transplantation Deceased Donor
100
Network)

50

0
< 1 Year 1-5 Year 6-10 Year 11-17 Year

Fig. 18.3  Indication for liver transplantation by age groups in the US (2016 data; Organ Procurement and Transplantation
Network)

failure as an indication for liver transplantation base of pediatric liver transplants and collects
was highest in this age group [8] (Fig. 18.3). data on more than 80% of all pediatric liver trans-
plants in the US and Canada; the following epi-
demiological data is extracted from the SPLIT
I ndications for Pediatric Liver database [8] (Fig. 18.2).
Transplantation and Their
Implications
Cholestatic Liver Disease
Indications for pediatric liver transplantation can
be divided into four general categories: choles- Biliary atresia is the indication for almost half of
tatic liver disease, metabolic liver disease, fulmi- all pediatric liver transplants, other cholestatic
nant hepatic failure and liver tumors. The SPLIT liver diseases such as Alagille syndrome, scleros-
group that organizes the US and Canadian data- ing cholangitis, progressive familial intrahepatic
224 P. J. Houck

cholestasis account for 15% of all pediatric liver signs and symptoms are persistent jaundice, pale
transplantations. Liver transplantation is consid- stools, dark urine, failure to thrive and coagulop-
ered curative for patients with cholestatic liver athy unresponsive to vitamin K. Late signs are
disease, however some patients may develop a hepatosplenomegaly and ascites, suggestive of
recurrence of cholestatic disease due to autoanti- progressing cirrhosis. The diagnosis is usually
bodies that interfere with the canalicular function made in early infancy with a percutaneous liver
in the graft [9]. biopsy.
Biliary atresia is the most common indication
for pediatric liver transplantation. Left untreated
Biliary Atresia it is lethal within 3 years in most cases. Up to
20% of patients have other congenital abnormali-
Biliary atresia is an inflammatory destruction of ties, including splenic malformation, situs inver-
both intra- and extrahepatic bile ducts in neo- sus or absence of an inferior vena cava. Standard
nates. The obliterative process is thought to begin of care in industrialized countries is a Kasai por-
in the neonatal period in patients with isolated toenterostomy (Fig. 18.4), which is a palliative
biliary atresia, whereas syndromic biliary atresia procedure in which a Roux-en-Y loop is anasto-
is thought to begin at an earlier stage of the mosed to the exposed ductules at the surface of
embryonic development [10]. The presenting the porta hepatis. Long term outcome is thought

Small intestine inserted over wedge

Liver

Pancreas Stomach

Small intestine pulled


up and used to create
bile duct
Small
intestine
Fig. 18.4 Kasai Small intestine
portoenterostomy: a
Roux-en-Y loop is
anastomosed to the
exposed ductules at the Rest of intestine stitched to
surface of the porta side of small intestine
hepatis. (Illustration by (Roux-en-Y connection)
Holden Groves, with
special thanks)
18  Pediatric Liver Transplantation 225

to be better the earlier the Kasai procedure is per- organ systems and (C) metabolic defects that can
formed. Although controversial, studies show cause hepatic neoplasms [12, 13]. Patients with
that procedures done after day 90 of life have primary hepatic metabolic disease such as Wilson
shorter native liver survival and worse bile drain- disease, α-1-antitrypsin deficiency, tyrosinemia
age and best results are achieved if the procedure and cystic fibrosis present with endstage liver
is done within the first 30 days of life [11]. disease or liver failure at the time of transplanta-
Successful portoenterostomies drain bile and will tion. Extrahepatic injury can be significant as for
normalize plasma bilirubin level within 6 months example in cystic fibrosis, where pulmonary dis-
of the procedure. Possible postoperative compli- ease is the leading manifestation in many patients.
cations include bile leaks, ascending cholangitis Only a subgroup of patients with cystic fibrosis
and later fat malabsorption and malnutrition. Up have hepatic disease, and of those only few prog-
to 80% of patients who underwent a successful ress to liver failure (Table 18.1).
Kasai procedure survive with a native liver for Wilson disease is an autosomal recessive dis-
longer than 10 years. Despite adequate bile drain- order of the copper metabolism with an incidence
age the disease will progress with worsening por- of 1:30,000; it is the cause of 5% of all patients
tal fibrosis, cirrhosis and portal hypertension to a with acute liver failure. Patients who are diag-
point at which a liver transplantation is indicated. nosed prior to fulminant liver failure and receive
Patients who are diagnosed late with biliary atre- pharmacological treatment have an excellent
sia and already have cirrhosis may undergo a prognosis. Copper-induced injury leads to liver
liver transplantation without prior Kasai proce- failure, neuropsychiatric decline, hemolysis,
dure. Patients with biliary atresia splenic malfor- proximal renal tubular dysfunction and other sys-
mation syndrome will require preoperative temic manifestations. Patients with Wilson dis-
imaging study to evaluate the anatomy of the por- ease may only have mild hepatic disease with
tal vein. progressive neuropsychiatric deterioration and
The anesthesiologist will encounter patients have chronic active hepatitis. They may present
with biliary atresia prior to the transplantation for in their teens with an acute deterioration that
liver biopsies and for esophago-gastro-­leads to fulminant hepatic failure. Severe hemo-
duodenoscopies to rule out or treat esophageal lysis at this time can be treated by removal of
varices. Blind esophageal instrumentation should copper from the circulation. The mortality of
be avoided for 7 days after banding of esophageal acute liver failure with Wilson disease without a
varices, because of the increased risk of esopha- liver transplantation is almost 100% [14] and
geal perforation or variceal bleed. Patients under- longstanding neurological deficits may to persist
going a liver transplantation after a Kasai despite liver transplantation.
procedure will have greater blood loss during the In patients with primary nonhepatic disease
pre-anhepatic phase because of adhesions. After (ornithine transcarbamylase deficiency, familial
reperfusion there is no need to create a new hypercholesteremia, primary hyperoxaluria type
Roux-en-Y limb in most cases as the existing 1 or organic academia) the indication for trans-
limb from the Kasai procedure can be used. plantation is not liver failure or end-stage liver
disease; the liver is structurally normal in these
patients but lacks a specific metabolic or syn-
Metabolic Disease thetic function. The purpose of the liver trans-
plant is to prevent further extrahepatic damage.
Metabolic disease is the indication for 13% of all Transplantation is curative for extrahepatic com-
pediatric liver transplants. The metabolic dis- plications of these patients and outcome is usu-
eases can be divided into (A) diseases that lead to ally excellent. Liver transplantation in these cases
structural liver damage with or without extrahe- can be considered as a crude form of gene ther-
patic injury, (B) metabolic defects that are apy to prevent the accumulation of toxic metabo-
expressed in the liver but cause injury to other lites [13]. Timing of the transplantation is difficult
226 P. J. Houck

in this setting. In patients with familial hypercho- mitochondrial hepatopathy or diseases that lead
lesteremia, the rapid progression of coronary to replacement of hepatic parenchyma such as
artery disease and aortic stenosis from lipid congenital leukemia, neuroblastoma and nephro-
deposits can make transplants necessary in pre-­ blastoma [17].
schoolers, before children reach the minimum Neonatal hypoxia does not cause isolated liver
weight for plasmapheresis. Outcomes are mixed failure in the neonate as the neonatal liver is rela-
with intraoperative and post-operative mortality tively refractory to hypoxia. Gestational alloimmune
due to coronary and vascular disease, however liver disease (GALD) is causing the majority of neo-
there are few alternative options [15]. natal acute liver failures. Recent studies found that
Metabolic diseases that cause hepatic ade- most cases of neonatal hemochromatosis were actu-
noma or hepatocellular carcinoma include, ally caused by GALD [18]. Neonatal hemosiderosis
among others, glycogen storage disorders, hered- (iron overload and tissue siderosis) is a phenotype of
itary tyrosinemia, galactosemia and alpha-1-­ liver disease, not a diagnosis. GALD can present
antitrypsin deficiency. In patients with adenomas with or without neonatal hemochromatosis. The
auxiliary transplants are avoided due to the risk alloimmune injury to the liver usually happens in
of progression to hepatocellular carcinomas. midgestation. GALD is associated with a significant
Glycogen storage disorders pose a special chal- coagulopathy with INR between 4 and 10, prematu-
lenge for the anesthesiologist: glycogen deposits rity, a patent ductus venosus, ascites and hypoglyce-
can cause myocardial hypertrophy, subaortic ste- mia [19]. Identifying the cause of neonatal acute
nosis and macroglossia, that can make airway liver failure is paramount to avoid transplanting
management extremely difficult. Patients are at patients with mitochondrial disease associated with
risk for hypoglycemia and lactic acidosis and a poor outcome or patients with hemophagocytic
usually receive a glucose containing solution dur- lymphohistiocytosis (HLH), which is treated medi-
ing the preoperative fastening period. Indications cally using chemotherapy. Biopsies of the salivary
for transplantation are large or multiple adeno- glands are useful to find evidence of sidorosis.
mas and poor metabolic control. Anesthetic care of neonates for liver trans-
plantation can be challenging because of their
size and the neonatal physiology, even though the
Fulminant Hepatic Failure outcomes of neonatal liver transplantation are
comparable to the other pediatric liver transplant.
Fulminant hepatic failure accounts for 11% of Hepatic encephalopathy is difficult to detect in
pediatric liver transplantations. Often no diagno- the pediatric population, and almost impossible
sis can be found and an unspecified viral etiology to appreciate in newborns. Extremes in intracra-
is assumed. Sometimes, due to the time con- nial pressure can be approximated by palpating
straints and urgency of transplantation, metabolic the open fontanel. ABO incompatible grafts can
diseases or autoimmune hepatitis cannot be ruled be used in neonates because neonates are not sen-
prior to transplantation [16]. Patients with fulmi- sitized to the major blood group antigens.
nant hepatic failure are generally older and have
worse long term outcome.
Liver Tumors

Neonatal Acute Liver Failure Liver tumors account for 4% of all pediatric liver
transplantations, with hepatoblastoma as the
All neonatal liver failures are acute by definition, most common pediatric liver tumor. If the tumor
meaning liver failure within 8 weeks of onset of is unresectable after appropriate systemic chemo-
symptoms. Neonatal acute liver failure is caused therapy, liver transplantation can be offered; even
by gestational alloimmune liver disease, viral metastatic disease unresponsive to chemotherapy
infections, hemophagocytic lymphohistiocytosis, is not a contraindication to transplantation.
18  Pediatric Liver Transplantation 227

 otal Parental Nutrition (TPN)


T and to prioritize the repair of the cardiac lesion
Induced Liver Failure versus addressing the liver disease with a liver
transplantation. Patients with congenital right to
Intestinal failure from either congenital abnormal- left shunts are at risk for paradoxical emboli dur-
ities or after bowel resections may require chronic ing reperfusion of the graft and throughout the
TPN administration that may lead to TPN-induced procedure and meticulous de-airing of the caval
liver disease. TPN-induced liver disease is seen in anastomoses as well as all intravenous fluid lines
40–60% of pediatric patients receiving chronic is paramount.
TPN. The TPN-induced liver dysfunction in pedi- Heterotaxy syndrome is an abnormal arrange-
atric patients differs significantly from adults: in ment of thoracic and or abdominal viscera with a
adults steatosis is more common whereas infants wide array of anatomical abnormalities. Patients
often present with cholestasis. Biliary sludge for- with heterotaxy syndrome may have, in addition
mation and cholelithiasis are seen in both popula- to congenital heart defects an abnormal vascular
tions [20]. Early small-bowel transplantation in a anatomy. The hepatic segment of inferior vena
TPN dependent infant may avoid the need for a cava (IVC) can be present or absent (so-called
combined liver and small-bowel transplantation “interrupted IVC”). The hepatic veins can be nor-
since early TPN cholestasis is reversible after ces- mal (joining the IVC just proximal to the IVC-­
saton of TPN [20]. atrial junction) or can connect independently to
atria. An interrupted IVC with hemiazygos con-
tinuation might be advantageous for the intraop-
Hepatic Encephalopathy erative management since crossclamping the
suprahepatic IVC may not lead to a decrease of
Hepatic encephalopathy has a different pathogen- venous return. Extrahepatic portosystemic shunts
esis in children compared to adults. In adults it is should be ruled out in all patients with heterotaxy
usually seen in the setting of chronic liver failure syndrome. Congenital extrahepatic shunts
and cirrhosis, whereas in children hepatic enceph- decrease the metabolism of galactose and ammo-
alopathy is usually due to acute on chronic liver nia by bypassing mesenteric circulation through
failure. Cerebral edema is seen at earlier stages in the liver and can cause encephalopathy. Newborn
children and frequently not recognized in a timely screening for elevated galactose levels can be
manner. Supportive care for patients with hepatic positive because of congenital extrahepatic por-
encephalopathy should address fluid manage- tosystemic shunts [21].
ment, potassium, sodium and glucose control. To
achieve normovolemia it is usually necessary to
restrict fluid intake. Hypokalemia and alkalosis I ntra-Operative Anesthetic Care
impair ammonia detoxification and increase renal for Pediatric Liver Transplantation
ammonia production, which may worsen the
hepatic encephalopathy. Artificial hepatic support Anesthetic management for liver transplantation
has been used in children however there is no evi- varies depending of the age group. For the ease of
dence that it improves outcome. discussion patients can be divided in three
groups: Infants & Toddlers, Preteens and Teens.

 ongenital Heart Disease


C
and Vascular Anomalies I nfants (0–1 year) and Toddlers
(1–3 years)
Patients with complex congenital heart disease
pose a special challenge. A multidisciplinary Infants typically present with biliary atresia and a
effort is necessary to ensure that all team mem- history of a “failed” Kasai procedure. Inhalational
bers clearly understand the cardiac physiology induction of anesthesia can be used, if there are no
228 P. J. Houck

contraindications such as a full stomach, massive clamp. Veno-venous bypass is not routinely used
ascites or actively bleeding gastro-­esophageal var- in this age group because of the risk of thrombo-
ices. The airway is secured with a conventional embolic complications due to low flow in the
uncuffed endotracheal tube or an endotracheal extracorporeal circuit; if crossclamping of the
tube with a high volume / low pressure cuff. The vena cava is not tolerated the piggyback technique
leak around the cuff should be maintained around as described earlier in this book can be used [22].
25 cm H20 and may need to be checked frequently Infants are at higher risk for hypothermia
as the airway may become more edematous. due to the larger skin-surface to body mass
Surgical exposure, ascites, pleural effusions and ratio and the inefficient shivering thermogene-
organomegaly causes a reduction of functional sis. Infants have to rely on non-shivering ther-
residual capacity (FRC); postive end-expiratory mogenesis, which may persist up to the age of
pressure (PEEP) and higher airway pressures need 2 years. Placing the cold donor organ in the
to be applied to ensure adequate oxygenation. abdominal cavity of an already hypothermic
Gastric emptying can be delayed, the presence infant will result in an even lower core temper-
of massive ascites with an associated increase in ature that may be difficult to correct. Warming
intra-abdominal pressure further increases the the operating room, use of radiant heat lamps,
risk of pulmonary aspiration; a rapid sequence convective forced-air warmers and airway
induction of anesthesia is commonly performed humidifiers can prevent hypothermia.
in this scenario. Placement of temperature probes in the rectum
Central vascular access is usually obtained in the addition to the esophagus prevents erro-
after the airway is secured, either with a Broviac neous temperature readings when the cold
catheter (a soft, tunneled central venous catheter) organ is placed in the immediate vicinity of the
or with a conventional central line. Broviac cath- esophageal temperature probe.
eters are long and have a small lumen, which pre- Arterial blood gases with a hemoglobin level
cludes rapid infusion of fluids or blood products. should be sampled hourly, because bleeding is
Therefore adequate peripheral access is required frequently unrecognized and difficult to estimate
in addition to the Broviac catheter. Peripheral in this age group. Transfusion of FFP and plate-
intravenous lines should be placed in the upper lets is done very restricted because of the con-
extremity, because there might be inadequate stant threat of hepatic artery thrombosis. It is not
drainage of the infrahepatic IVC to the central uncommon to start a heparin infusion if the
circulation during caval crossclamping. Obtaining hepatic artery anastomosis is considered to be at
intravenous access can be a challenge in these risk for thrombosis. Arterial blood gases analysis
patients who may have had multiple central line should also include glucose and electrolyte deter-
placements in the past. At times a venous mag- minations: decreased glycogen storage capacities
netic resonance study or venous Doppler study in infants and prior infusion of glucose contain-
can be obtained preoperatively to verify patency ing fluids such as TPN predisposes these patients
of veins. The use of an ultrasound machine in the to hypoglycemia and may make a glucose infu-
OR can be helpful to establish not only central sion and monitoring necessary. Hyperkalemia is
access but also peripheral access in the antecubi- of similar concern as in adults especially during
tal veins. An arterial line can be established in reperfusion and needs to be treated aggressively.
either extremity; the upper extremity is preferred Although some infants can safely be endotra-
because of the possibility of intraoperative partial cheally extubated at the conclusion of the proce-
aortic occlusion from aortic sideclamping. dure, postoperative ventilation provides time to
Infants typically tolerate cross-clamp of the ensure adequate diuresis, reduction of possible
inferior vena cava with only minimal hemody- airway swelling and stable hemodynamics and
namic support and most infants only require for hemostasis and facilitates optimal imaging stud-
example a dopamine infusion with optimization ies. In one case series safe immediate extubation
of their intravascular volume to tolerate the cross- was achieved in two thirds of all patients.
18  Pediatric Liver Transplantation 229

Immediate extubation was not considered in thetic setup and management is similar to the
pediatric patients with: management of adults. Teenagers usually have
good cardiac reserves and a very compliant ven-
• open abdomen/temporary closure, tricle with the exception of patients with familial
• anticipation of a return to the operating room hypercholesterolemia, who may have significant
within the next 24 h coronary artery disease and myocardium at risk.
• significant volume overload with symptom- Large bore central venous access, with the pos-
atic of impaired pulmonary gas exchange or sibility to float a pulmonary artery catheter is
swelling compromising the airway required. Pulmonary artery catheters or other
• severe hemodynamic instability with escalat- measures of ventricular filling can be used in
ing vasopressor requirements teenager with significant cardiac disease or pul-
• encephalopathy with preoperative ventilation monary hypertension as described for adults else-
or compromised airway protection where in this book. Transesophageal
• pretransplant ventilatory dependence [23]. echocardiography may be a good monitoring
option in teenagers of appropriate size.
Patients as small as 1.7 kg have successfully
undergone a liver transplantation, however metic-
ulous surgical technique and anesthetic care are Post Operative Care
necessary to ensure the success of the operation
in this extreme patient group. Anesthetic issues Concerns in the immediate post-operative period
that are similar to those encountered with prema- care are similar to patients who underwent major
ture neonates such as glycemic management, abdominal procedures and remained intubated.
avoidance of hyperosmolar medications, ventila- Additional considerations specific to liver
tory management and the physiology of neonatal ­transplantation are the detection of graft related
circulation need to be considered. A Broviac complication such as hepatic artery or portal vein
catheter should be placed preoperatively in very thrombosis, rejection or infectious complications.
small or and premature neonates to allow ade- If the abdominal wall cannot be closed ini-
quate central administration of fluids and vasoac- tially, tracheal extubation is deferred until after
tive medications. closure of the abdominal wall and other related
procedures. The higher rate of re-explorations
in infants, facilitation of pain management and
Pre-Teenager (4–9 years) imaging of intubated patients make it prudent
not to extubate the patient too prematurely. In
Preteens can receive large bore central venous patients with relative large grafts special care
lines and usually it is not necessary or feasible to must be taken to rule out abdominal compart-
place a pulmonary artery catheter. Preteens have ment syndrome. Increasing airway pressure,
normal shivering thermogenesis but still have a respiratory insufficiency from worsening
larger skin-surface to body mass ratio and require ventilation-­perfusion mismatch, hemodynamic
meticulous temperature management. Unlike compromise from compression of the vena cava
adults, pre-teenager do not commonly have cir- and worsening abdominal distention are signs of
rhotic cardiomyopathy. abdominal compartment syndrome and should
prompt urgent evaluation and possible re-explo-
ration. If not addressed rapidly, the high airway
Teenager pressures may cause further hemodynamic com-
promise and increase intracranial pressure.
Hemodynamic prertubations seen with teens dur- Renal function may deteriorate due to compres-
ing liver transplantation are comparable, but still sion of the renal veins. With a normal postoper-
less grave than with adults, however the anes- ative course renal dysfunction is not as frequent
230 P. J. Houck

as in the adult population, however reduced per- treated with revision of the anastomosis or using
fusion pressure, impaired venous return and percutaneous interventions such as angioplasty
renal vasoconstriction from calcineurin inhibi- or stent placement. Portal thrombosis in the late
tors may precipitate renal injury and lead to post-operative period becomes clinically appar-
renal dysfunction. ent by splenomegaly, thrombocytopenia and gas-
Due to large volume intraoperative fluid trointestinal hemorrhage. If there is still an open
administration fluid overload is common and lumen percutaneous techniques may be used, but
fluid shift in the early postoperative period may with complete thrombosis portal venous shunt
require aggressive diuresis. Extensive use of loop placement may be needed.
diuretic however may lead to (contraction) meta-
bolic alkalosis and may cause hypoventilation in
the extubated child. Biliary Complications
There are three categories of graft-related
problems in the early post-operative period: vas- The incidence of biliary complications is up to
cular complications, biliary complications and 30% in pediatric liver transplant recipients and it
allograft rejection. is the most common surgical complication in
Patient with any of the complications may patients receiving reduced sized organs [25].
present with cholestasis, elevation of hepatocel- Most biliary complications are bile leaks and
lular enzymes, lethargy and fever. Urgent diag- biliary strictures are less common [26]. In the
nosis of the specific cause is required to initiate early postoperative period bile can be found in
timely treatment. Doppler ultrasound may help the abdominal drains, if a bile leak is present.
exclude vascular complications and identify The vascular supply of the extrahepatic bile
fluid collections from bile leaks. Vascular and ducts is quite precarious and bile leaks are at
biliary complications frequently require re- times caused by hepatic artery thrombosis, lead-
explorations. Rejections are not common very ing to necrosis of the bile duct and leakage of
early after surgery however treatment should be bile into the abdominal cavity. Biliary strictures
commenced rapidly either when there is a high present with recurrent cholangitis, elevate alka-
level of suspicion or after a biopsy confirmed line phosphatase and GGT and dilated intrahe-
the rejection [24]. patic biliary ducts on ultrasound examination.
Biliary complications almost always require sur-
gical re-­exploration if endoscopic or percutane-
Vascular Complications ous interventions are not feasible.

In pediatric patients hepatic artery thrombosis is


the most common serious postoperative compli- Rejection
cation and up to four times more frequent than in
adult patients due to the smaller size of the ves- Hyperacute rejection is rare, usually occurs
sel. Early occlusion of the hepatic artery leads to very early and is caused by antibodies that bind
graft necrosis and may cause graft loss if not to the endovascular epithelium of the graft. It
addressed immediately. Daily routine doppler can lead to intraparenchymal vascular thrombo-
ultrasound examinations are recommended to sis and rapid graft loss. Acute rejection presents
verify a patent vessel. with irritability, fever, increased bilirubin,
Patients with biliary atresia typically have a transaminases and leukocytes occurs later and
hypoplastic portal vein and may need a replace- usually requires confirmation with a liver
ment of the portal vein up to the superior mesen- biopsy. Treatment consists for example of a
teric vein and the splenic vein. These patients are course of steroids over 3–6 days followed by a
at a higher risk for portal vein thrombosis with an steroid taper. Acute rejection seldom leads to
incidence of up to 10%. Portal thrombosis is graft loss.
18  Pediatric Liver Transplantation 231

Primary Nonfunction syndrome is a major contributor to the long-term


morbidity after adult liver transplantation and it
25% of postoperative graft loss is due to primary is suspected to be as prevalent in pediatric patients
nonfunction requiring re-transplantation and is [30, 31].
associated with a 67% mortality [27]. Patients
with primary non-function present with worsen-
ing coagulopathy, acidemia, rising liver enzymes Summary
and cholestasis without a clear etiology. Early
diagnosis and relisting for transplant is crucial. If The perioperative care for patients who are
not re-transplanted in time the disease may prog- undergoing a liver transplantation is one of the
ress to fulminant liver failure and death. most satisfying challenges of a pediatric anesthe-
siologist. Anesthetizing young, critically ill
patients, who are undergoing urgent and major
Infectious Complications surgery, is even more rewarding in view the
excellent long-term outcomes compared with
Induction immunosuppressive therapy in the other solid organ transplants.
early post-operative period renders patients at
high risk for gram-negative enteric bacteria,
enterococci and staphylococci. Indwelling cath- References
eters should be removed as soon as possible.
Epstein-Barr virus, cytomegaly virus and herpes 1. Starzl TE, et al. Homotransplantation of the Liver in
Humans. Surg Gynecol Obstet. 1963;117:659–76.
simplex virus are the most common causes of a 2. Kamath BM, Olthoff KM. Liver transplanta-
viral infection post-operatively. An antifungal tion in children: update 2010. Pediatr Clin N Am.
prophylaxis may be given even before the trans- 2010;57(2):401–14; table of contents.
plantation, whereas most center reserve prophy- 3. Rook M, Rand E. Predictors of long-term outcome
after liver transplant. Curr Opin Organ Transplant.
laxis only for high-risk patients. Diagnosis and 2011;16(5):499–504.
treatment of infectious complications is similar 4. Freeman RB Jr, et al. The new liver allocation system:
to adults described elsewhere in this book. moving toward evidence-based transplantation policy.
Liver Transpl. 2002;8(9):851–8.
5. McDiarmid SV, Anand R, Lindblad AS. Development
of a pediatric end-stage liver disease score to predict
Outcome poor outcome in children awaiting liver transplanta-
tion. Transplantation. 2002;74(2):173–81.
In the SPLIT database the overall one and five 6. McDiarmid SV, et al. Selection of pediatric candidates
under the PELD system. Liver Transpl. 2004;10(10
year patient survival was 89.8% and 84.8% and Suppl 2):S23–30.
graft survival among the 5-year survivors was 7. Bourdeaux C, et al. PELD score and posttransplant
93% and 88% respectively. Twelve percent of outcome in pediatric liver transplantation: a retro-
5-year-survivors needed a second liver transplan- spective study of 100 recipients. Transplantation.
2005;79(9):1273–6.
tation and 2% needed a third transplantation [28]. 8. McDiarmid SV, Anand R, Lindblad AS. Studies
Post-transplant lymphoproliferative disease was of pediatric liver transplantation: 2002 update. An
seen in 6% of patients and 60% of patients expe- overview of demographics, indications, timing, and
rienced an episode of acute cellular rejection immunosuppressive practices in pediatric liver trans-
plantation in the United States and Canada. Pediatr
within the first 5 years [28]. The reported inci- Transplant. 2004;8(3):284–94.
dence of renal insufficiency in long-term survi- 9. Santos JL, Choquette M, Bezerra JA. Cholestatic
vors varies between 13% and 32% [29]. The use liver disease in children. Curr Gastroenterol Rep.
of calcineurin-inhibitors is associated with post-­ 2010;12(1):30–9.
10. Kelly DA, Davenport M. Current management of bili-
transplant metabolic syndrome (obesity, hyper- ary atresia. Arch Dis Child. 2007;92(12):1132–5.
tension, elevated triglycerides, low HDL and 11. Nio M, et al. Five- and 10-year survival rates
glucose intolerance). Post-transplant metabolic after surgery for biliary atresia: a report from the
232 P. J. Houck

Japanese Biliary Atresia Registry. J Pediatr Surg. 22. Nery J, et al. Routine use of the piggyback technique
2003;38(7):997–1000. in pediatric orthotopic liver transplantation. J Pediatr
12. Hansen K, Horslen S. Metabolic liver disease in chil- Surg. 1996;31(12):1644–7.
dren. Liver Transpl. 2008;14(4):391–411. 23. Fullington NM, et al. Immediate extubation after

13. Treem WR. Liver transplantation for non-hepatotoxic pediatric liver transplantation: a single-center experi-
inborn errors of metabolism. Curr Gastroenterol Rep. ence. Liver Transpl. 2015;21(1):57–62.
2006;8(3):215–23. 24. Spada M, et al. Pediatric liver transplantation. World J
14. Rosencrantz R, Schilsky M. Wilson disease: patho- Gastroenterol. 2009;15(6):648–74.
genesis and clinical considerations in diagnosis and 25. Broering DC, et al. Is there still a need for living-­
treatment. Semin Liver Dis. 2011;31(3):245–59. related liver transplantation in children? Ann Surg.
15. Mansoorian M, et al. Liver transplantation as a
2001;234(6):713–21. discussion 721–2.
definitive treatment for familial hypercholester- 26. Heffron TG, et al. Biliary complications in pediatric
olemia: a series of 36 cases. Pediatr Transplant. liver transplantation. A comparison of reduced-size
2015;19(6):605–11. and whole grafts. Transplantation. 1992;53(2):391–5.
16. Narkewicz MR, et al. Pattern of diagnostic evalu- 27. Sieders E, et al. Graft loss after pediatric liver trans-
ation for the causes of pediatric acute liver failure: plantation. Ann Surg. 2002;235(1):125–32.
an opportunity for quality improvement. J Pediatr. 28. Ng VL, et al. Outcomes of 5-year survivors of pediat-
2009;155(6):801–6. e1 ric liver transplantation: report on 461 children from
17. Taylor SA, Whitington PF. Neonatal acute liver fail- a north american multicenter registry. Pediatrics.
ure. Liver Transpl. 2016;22(5):677–85. 2008;122(6):e1128–35.
18. Whitington PF. Gestational alloimmune liver disease 29. Bucuvalas J. Long-term outcomes in pediatric

and neonatal hemochromatosis. Semin Liver Dis. liver transplantation. Liver Transpl. 2009;15 Suppl
2012;32(4):325–32. 2:S6–11.
19. Sharma R, et al. Neonatal acute liver failure compli- 30. Rothbaum Perito E, et al. Posttransplant metabolic
cated by patent ductus venosus: diagnosis and man- syndrome in children and adolescents after liver
agement. Liver Transpl. 2013;19(9):1049–52. transplantation: a systematic review. Liver Transpl.
20. Kelly DA. Liver complications of pediatric par-
2012;18(9):1009–28.
enteral nutrition—epidemiology. Nutrition. 31. Perito ER, et al. Posttransplant metabolic syndrome
1998;14(1):153–7. in the withdrawal of immunosuppression in Pediatric
21. Murray CP, Yoo SJ, Babyn PS. Congenital extra-
Liver Transplant Recipients (WISP-R) pilot trial. Am
hepatic portosystemic shunts. Pediatr Radiol. J Transplant. 2015;15(3):779–85.
2003;33(9):614–20.
Combined Solid Organ
Transplantation Involving 19
the Liver

Geraldine C. Diaz, Jarva Chow, and John F. Renz

Keywords availability of TIPS (transjugular intrahepatic por-


Liver–kidney transplantation · Renal trans- tosytemic shunt) procedures and effective medica-
plantation · Heart–liver transplantation · tions to control hepatitis B and hepatitis C viral
Lung–liver transplantation · Intraoperative replication have significantly increased the life
management · Renal failure expectancy of patients with ESLD [2]. Improved
life expectancy results in additional organ-system
failures in some that are potentially amenable to
Introduction transplantation. Furthermore, improved outcomes
of isolated LTX and thoracic organ transplant
Transplantation of the liver (LTX) in combination recipients have led to a natural extension of these
with other solid organs, both abdominal and tho- techniques to dual organ transplantation in select
racic, has significantly increased over the past transplant centers.
decade (Fig. 19.1) [1]. This is the result of many Combined solid organ transplantations intro-
factors including improved medical management duce entirely new dimensions to the practicing
of patients with end-­stage liver disease (ESLD), anesthesiologist. These procedures are a signifi-
advances in thoracic organ transplantation and a cant clinical challenge requiring unique physio-
Model for End-stage Liver Disease (MELD)- logic considerations. Perioperative management
based liver allocation policy [2–4]. The introduc- must be adjusted to the underlying etiology of
tion of prophylactic beta-blockers to decrease organ failure, severity of illness, and the patient’s
portal hypertension, widespread application of estimated physiologic reserve. This chapter dis-
endoscopic modalities to treat esophageal varices, cusses the three most common combined solid
organ transplant procedures involving the liver
and their anesthetic implications.
G. C. Diaz, DO (*)
Department of Anesthesiology, University of Illinois
at Chicago, Chicago, IL, USA
e-mail: gdiaz@uic.edu
Simultaneous Liver–Kidney
J. Chow, MD
Department of Anesthesiology, Loyola University,
Transplantation
Chicago, IL, USA
The implementation of a MELD-based liver allo-
J. F. Renz, MD, PhD
Section of Transplantation, Department of Surgery, cation policy in February 2002 [4] has dramati-
University of Chicago, Chicago, IL, USA cally increased the number of simultaneous

© Springer International Publishing AG, part of Springer Nature 2018 233


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_19
234 G. C. Diaz et al.

Fig. 19.1 Heart–liver Heart/Liver and Lung/Liver Transplants 1995-2015


and lung–liver 60
transplants from 1995 to Heart/Liver
2015 in the US Lung/Liver
50

40

30

20

10

0
95
96
97
98
99
00
01
02
03
04
05
06
07
08
09
10
11
12
13
14
15
19
19
19
19
19
20
20
20
20
20
20
20
20
20
20
20
20
20
20
20
20
Fig. 19.2 Kidney–liver Kidney/Liver Transplants 1995-2015
from 1995 to 2015 in the 1400
US
1200

1000

800

600

400

200

0
1995 1997 1999 2001 2003 2005 2007 2009 2011 2013 2015

liver–kidney transplantats (SLKT) (Fig. 19.2, post-LTX on patient survival have further accel-
Table  19.1) [5]. The MELD score predicts the erated demand. Organ Procurement and
90-day mortality from liver failure based upon Transplant Network (OPTN) data (reported as
serum total bilirubin, the international normalized part of an allocation policy proposal for SLKT in
ratio (INR) of prothrombin time, and serum cre- 2016) demonstrated that patients with renal fail-
atinine [4]. The MELD is overweighted by serum ure (defined as ≥2 mo of dialysis or estimated
creatinine and the requirement for renal replace- glomerular filtration rate (GFR) <25 mL/min
ment therapy because historically mortality was prior to LTX,) had a significant survival advan-
high with pre-transplant renal failure. This has tage when receiving received SLKT compared to
resulted in a >400% increase in the performance LTX alone (1-year patient survival: 86.2% versus
of SLKT since MELD-based liver allocation. 81.1%, 5-year patient survival: 70.1% versus
The first SLKT was reported in 1984 [6] and 65.9%) [9]. This confirms previous data demon-
since then outcomes have continuously improved strating a clear survival advantage for patients
[7, 8]. MELD-based allocation has contributed to with renal failure undergoing SLKT [10].
an increased number of SLKT. Furthermore, rec- Advances in critical care, adoption of a defini-
ognition of the deleterious effects of renal failure tion of acute kidney injury (AKI) [11], and the
19  Combined Solid Organ Transplantation Involving the Liver 235

Table 19.1  Eligibility criteria for simultaneous liver kid- criteria (risk, injury, failure, loss, end-stage) strat-
ney transplantation [9]
ified acute renal dysfunction based upon changes
Chronic in serum creatinine or urine output. These criteria
kidney disease were later modified by the Acute Kidney Injury
End stage renal disease with chronic
Network that expanded the definition of AKI to
dialysis
encompass a wider range of renal failure [19].
GFR <60 mL/min for >90 days prior
to listing and a GFR <35 mL/min at Increased mortality with worsening RIFLE class
listing has been validated in multiple patient populations
Sustained AKI including cirrhotics and post-LTX [20–22]. In
GFR <25 mL/min for ≥6 consecutive 2010, a working party that included several mem-
weeks bers from the ADQI and the International Ascites
Combination of dialysis and GFR Club, set forth a proposal to apply the RIFLE cri-
<25 mL/min for ≥6 consecutive
weeks teria to define AKI in patients with cirrhosis, irre-
Metabolic spective of the cause [23].
diseases While the presence of end stage renal disease
AKI Acute kidney injury, GFR Glomerular filtration rate (ESRD) and ESLD makes the decision for SLKT
straightforward, differentiating HRS from acute
widespread availability of continuous renal replace- kidney injury (AKI) and CKD in the setting of
ment therapy (CRRT) [12], have dramatically ESLD and determining its reversibility may be
increased access of critically ill patients to LTX and difficult. Thus, selection criteria of appropriate
improved post-LTX outcomes. Historically, hepa- candidates for SLKT remains controversial
torenal syndrome (HRS) was a leading contributor despite multiple consensus conferences [7, 24,
to waitlist morbidity and mortality [13]. HRS is a 25]. To date, there are no reliable instruments to
serious complication of advanced liver disease, determine the etiology, severity, and reversibility
with an annual incidence of up to 15% in patients of renal failure in the setting of liver disease.
with liver disease who develop ascites [14]. HRS Thus, duration of dialysis has been a surrogate to
results from activation of the sympathetic nervous determine SLKT candidacy in several published
system and renin angiotensin aldosterone system guidelines [7, 9, 24]. A new SLKT policy has
causing renal vasoconstriction, a compensatory been proposed in the Spring of 2016 [9]
response to the profound systemic arterial vasodila- (Table 19.1). This classifies candidates into three
tation and decrease in effective central blood vol- groups: CKD, sustained AKI, and metabolic dis-
ume seen in patients with portal hypertension ease. For the first time, specific criteria are pro-
[14–16]. The result is decreased renal perfusion and posed to fulfill eligibility for SLKT. CKD
glomerular filtration rate with preserved renal tubu- requiring SLKT was defined as GFR <60 mL/min
lar function [14, 16]. HRS is classically thought to for >90 days prior to listing and a GFR of <35 mL/
be reversible following LTX and is not a recognized min at the time of listing. Sustained AKI fulfilling
indication for SLKT [17]. However, improvements criteria for SLKT was defined as a combination of
in medical management with the use of vasocon- dialysis and GFR <25 mL/min for six consecutive
strictors and albumin infusion, critical care, and weeks. The need for SLKT among candidates
CRRT have dramatically prolonged the period with metabolic disease such as citrullinemia has
whereby patients can wait for an allograft [13, 18]. been widely recognized and was not changed [9].
This has created a new clinical dilemma of when
should consideration for SLKT be entertained
among LTX candidates with HRS who require pro- Pre-Operative Evaluation
longed hemodialysis.
The Acute Dialysis Quality Initiative (ADQI) Preoperative evaluation includes a thorough under-
Workgroup developed a consensus definition and standing of the etiology of both liver and kidney
classification for AKI in 2004 [11]. The RIFLE disease, as their pathology is typically intertwined.
236 G. C. Diaz et al.

Associated complications such as uremic or hepatic cal team where the kidney will be implanted (left
encephalopathy, pericardial effusion, cirrhotic car- lower quadrant, right lower quadrant, or intra-
diomyopathy, hepatopulmonary syndrome, and abdominal) so that area can be avoided for vascular
coagulopathy must be identified and medical ther- access. The typical LTX incision may also be mod-
apy optimized prior to SLKT. The anesthesiologist ified to accommodate SLKT. Venous and arterial
should query the duration of dialysis and the last catheter placement should be optimized to prevent
hemodialysis session prior to surgery. Serum elec- unnecessary complications such as discovering a
trolytes must be obtained and reviewed. catheter in the vascular clamp. Such issues are eas-
Central venous access can be difficult in this ily avoided with effective communication. Two
patient population. Venous access for SLKT arterial catheters, radial and femoral, are used in
includes a dialysis catheter and central access for most centers. Radial arterial monitoring alone
volume and pharmacologic resuscitation. Because should be interpreted with caution as aortic pres-
of the nature of renal and hepatic failure, these sure can be underestimated in hypotensive states,
patients often have a history of multiple prior cen- especially during hepatic allograft reperfusion [28,
tral venous access and may demonstrate vascular 29]. Pulmonary artery catheter (PAC) with contin-
pathology. Pre-operative magnetic resonance or uous cardiac output monitoring, traditionally used
ultrasound venous mapping during evaluation and in many liver transplant centers is increasingly
utilization of real-time Doppler ultrasound during supplemented and even replaced by continuous
catheter insertion may be essential. transesophageal echocardiography (TEE) to assess
volume status, ventricular function, and detect air
or thrombotic emboli [30, 31].
Intra-Operative Management

The allograft with the least cold-ischemic toler-  iver Transplantation in the Presence
L
ance must be transplanted first and therefore SLKT of Renal Failure
usually begins with the liver transplantation fol-
lowed by renal transplantation. The surgeries are Renal failure complicates LTX via impaired acid-­
typically sequential; however, when appropriate, base physiology, coagulopathy, and the ability to
LTX can be completed and the patient stabilized in compensate for acute volume/electrolyte shifts
the intensive care unit prior to performing the renal secondary to blood transfusion and reperfusion
transplant. We have found this a very effective [32]. Anesthetic management often requires the
strategy when LTX is complicated by coagulopa- utilization of intra-operative continuous renal
thy, hypothermia, hemodynamic instability, high replacement therapy, aggressive red cell washing
vasopressor requirements, or early hepatic prior to transfusion, frequent laboratory analysis,
allograft dysfunction. Resuscitation in the inten- venting of the hepatic allograft prior to reperfu-
sive care unit for several hours may optimize the sion, and volume resuscitation guided by TEE
patient prior to returning to the operating room for and PAC pressures [33]. Our group has also
SLKT completion through a separate skin inci- found flushing the hepatic allograft with low-­
sion. During this period, the renal allograft is potassium histidine-tryptophan-ketoglutarate
maintained on a cold-perfusion, machine preser- preservation solution prior to implantation to be
vation system [26]. It is our practice to place all particularly effective at reducing hemodynamic
renal allografts destined for SLKT on cold-perfu- instability associated with allograft reperfusion.
sion, machine preservation as soon as practical fol- Frequent coagulation studies and assessment of
lowing procurement. coagulation using clinical judgement and visco-
Rapid sequence induction is frequently indi- elastic tests guide transfusion therapy [34, 35].
cated in SLKT secondary to delayed gastric empty- Desmopressin can be used to increase factor VIII
ing in patients with renal and hepatic failure [27]. levels and von Willebrand antigen in the presence
The anesthesiologist should clarify with the surgi- of uremic coagulopathy [32].
19  Combined Solid Organ Transplantation Involving the Liver 237

 enal Transplantation in the Presence


R imaging may demonstrate abnormalities in blood
of a Newly Transplanted Liver flow and determine the necessity for re-­
exploration. Delayed renal allograft function is
Similar concerns arise for renal transplantation in common among SLKT recipients who often
the presence of a newly transplanted liver. require a period of renal replacement therapy.
Aggressive fluid resuscitation during kidney Persistent encephalopathy may contribute to dif-
allograft implantation may result in congestion of ficulty liberating from mechanical ventilation
the hepatic allograft. Heparin is frequently and increases the risk of aspiration [27]. A multi-
administered during renal transplantation to disciplinary team including hepatologists, sur-
ensure graft vessel patency but may worsen coag- geons, nephrologists, and intensivists should
ulopathy in the setting of hepatic allograft review immunosuppressive therapy, particularly
dysfunction. the use of nephrotoxic calcineurin inhibitors.
Anesthetic considerations include judicious
fluid resuscitation guided by TEE, PAC pres-
sures, and urine output. The use of diuretics Combined Heart–Liver
should be discussed with the surgical team and Transplantation
often depends on the function of the newly trans-
planted liver. Heparin administration should also First described by Starzl in 1984, combined heart
be reviewed on an individual basis. Vasopressors and liver transplantation (CHLT) has been widely
should be avoided if possible due to deleterious applied to adults and children [39–42]. While
vasoconstrictive effects on the newly transplanted CHLT remains a procedure performed by a only
renal allografts [36]. a small number of US transplant centers, the inci-
dence reported by the Scientific Registry of
Transplant Recipients (SRTR) database has
Post-Operative Management increased annually (Fig. 19.1) [1]. The success of
cardiac mechanical assist devices, coupled with
The complexity of the postoperative period for improved medical management, is increasing the
SLKT patients depends on the duration of sur- longevity of cardiac failure patients and the inci-
gery and the functional recovery of both allografts dence of cardiac hepatopathy [43]. This has
[37]. Hepatic allograft dysfunction manifests as become a principal driver of CHLT demand.
persistent acidemia, coagulopathy, hypoglyce- Definitive CHLT indications have yet to be
mia, and encephalopathy [38]. Renal allograft established but can be broadly categorized as a
dysfunction is associated with oliguria, acidemia, procedure to optimize the performance of a sin-
and electrolyte abnormalities. Hypotension is gle organ, characterized by single organ failure
common in the postoperative period and may with minimal portal hypertension, or true dual
result from hypovolemia, hemorrhage, myocar- organ failure, characterized by concomitant car-
dial ischemia, arrhythmia from electrolyte/acid-­ diac and liver failure, with portal hypertension
base abnormalities, and vasodilatory shock. An and its complications (Table 19.2) [44].
echocardiogram to supplement PAC data may be CHLT outcomes have been excellent and as
helpful in the diagnosis and treatment of hypo- improvements in surgical technique and immu-
tension. Judicious vasopressor administration is nosuppression continue to advance, sicker
paramount to optimize perfusion to both patients are increasingly eligible for this complex
allografts without causing excessive renal vaso- procedure. Cannon et al. performed a review of
constriction. Assessment of abdominal drains, SRTR data that included 97 CHLT performed
measurement of serum and abdominal drain between 1987 and 2010 with 1-year, 3-year, and
hemoglobin can diagnose ongoing hemorrhage. 5-year patient survival of 84%, 74%, and 72%,
Viscoelastic and conventional coagulation tests respectively [45]. US allocation policy under-
should guide transfusion therapy [35]. Ultrasound serves the CHLT population by not uniformly
238 G. C. Diaz et al.

Table 19.2 Indications for combined heart–liver tively in these patients. The decision for combined
transplantation
transplantation should include consensus between
A.  Procedure to “optimize” performance of a single the heart transplant team, liver transplant team,
organ: anesthesiologists, cardiac surgeons, cardiologists,
Heart failure secondary to a metabolic defect in the
liver hepatobiliary surgeons, hepatologists, as well as
Familial hypercholesterolemia social work.
Homozygous beta-thalassemia
Familial amyloidotic polyneuropathy
Familial hypertrophic restrictive cardiomyopathy Intra-Operative Management
B.  True dual organ failure
Heart and liver failure The successful performance of CHLT mandates
Hemochromatosis unique anesthetic considerations. Two extensive
Cryptogenic cirrhosis/cardiomyopathy operative procedures must be performed on a
Alcoholic cardiomyopathy/cirrhosis patient with limited physiologic reserve as a
result of cardiac and hepatic failure. The cardiac
transplant is performed first to minimize cardiac
permitting the cardiac and liver allografts to be ischemia and because the failing heart will poorly
allocated as a single unit. Thus, less than 30% of tolerate the fluid shifts and hemodynamic insta-
patients listed nationally for CHLT receive trans- bility associated with hepatic reperfusion.
plantation, and the overall mortality in this popu- There is no consensus regarding the ideal sur-
lation is greater than predicted by the sum of gical technique for CHLT. Operative strategies
MELD and cardiac status scores [46]. range from complete cardiac transplantation with
sternal closure prior to proceeding with the
abdominal dissection, to maximal abdominal dis-
Pre-Operative Evaluation section before initiating cardiopulmonary bypass
(CPB) [44, 48–50]. Shaw described the first three
Understanding the etiology of cardiac and hepatic cases of CHLT in 1985 using CPB during cardiac
failure is essential to the successful performance transplant and venovenous bypass (VVB) includ-
of CHLT. The indication for CHLT affects the ing portal vein decompression during LTX [51].
complexity of the planned surgery. If the CHLT is The authors postulated VVB augmented cardiac
to optimize the performance of a single organ, as support and enhanced hemodynamic stability
in amyloidosis and familial hypercholesterol- during LTX; however, coagulopathy, hypother-
emia, the operative course may be much less dif- mia, acidosis, and platelet dysfunction associated
ficult due to absence of portal hypertension. with CPB must be corrected [38, 51]. Barbara
However, when CHLT is mandated by true dual reported avoiding CPB to avert coagulopathy and
organ failure, as in hemochromatosis or alcoholic inflammation [52]. Proponents of CPB assert the
cardiomyopathy, the surgery will be much more risks of CPB during LTX are outweighed by the
challenging secondary to portal hypertension and advantages of decreased hemodynamic and met-
cirrhotic physiology [47]. abolic disturbances to the newly transplanted
Pretransplant evaluation for CHLT includes heart during hepatic reperfusion [53].
clinical and laboratory examinations, echocar- Subsequent strategies to reduce hemorrhage
diography, right and left heart catheterization, advocated separate thoracic and abdominal trans-
chest radiography, carotid and peripheral artery plant operations with interruption of extracorpo-
Doppler ultrasound, as well as total body com- real circulation and heparin neutralization in
puted tomography [44, 48]. Preoperative evalua- between [50]. Although this technique reduced
tion of pulmonary hypertension is essential as the duration of anticoagulation, it significantly
acute right ventricular failure is a dreadful compli- increased hepatic allograft cold ischemia.
cation that can occur both intra- and post-opera- Conversely, Offstad advocated complete abdomi-
19  Combined Solid Organ Transplantation Involving the Liver 239

nal dissection prior to sternotomy [49]. This tech- Vaso-mediated pulmonary hypertension or
nique facilitates abdominal dissection without portopulmonary hypertension (PPHTN) can be
the presence of anticoagulation but significantly exacerbated during hepatic allograft reperfusion
adds to the length of the total operative procedure and precipitate right ventricular dysfunction.
as well as the cold ischemia time of both Pulmonary arterial catheterization permits imme-
allografts. Preservation of the IVC during LTX, diate recognition of increasing pulmonary vascu-
(“piggyback” technique), improves hemody- lature pressures, as well as guides the use of
namic stability during the anhepatic phase, allow- pulmonary vasodilators. In addition, TEE is ben-
ing cirrhotic patients to better tolerate portal eficial in evaluating right ventricular function.
clamping without necessitating anticoagulation Patients with liver failure also suffer from
[44] especially when combined with a temporary impaired acid–base regulation, hypothermia,
porto-caval shunt. All of the above strategies thrombocytopenia, and clotting factor deficien-
have demonstrated acceptable outcomes. cies further complicating the cardiac transplant
While no superior approach has emerged, it is procedure [27]. Averting disseminated intravas-
crucial that coordination between the cardiotho- cular coagulation (DIC), metabolic acidosis,
racic anesthesiologist, liver anesthesiologist, arrhythmias, and pulmonary hypertension
intensivists, cardiothoracic surgeon, liver trans- through vigilant management of acid–base,
plant surgeon, and perfusionists occur prior to hemostasis, and volume status, are crucial to the
surgery. Discussions should include surgical success of the procedure [47].
sequence, use of CPB and possibly VVB, place- If the patient develops cardiac graft dysfunc-
ment of bypass cannulas and central venous cath- tion ECMO may be required and the liver trans-
eters, PAC, arterial lines, heparin utilization and plant may need to be done while the patient is
reversal. supported on ECMO.

Cardiac Transplantation  iver Transplantation in the Presence


L
in the Presence of Liver Failure of a Newly Transplanted Heart

The physiology of portal hypertension compli- LTX incurs unique demands upon the newly
cates the anesthetic management of cardiac trans- transplanted heart. The cardiac allograft demon-
plantation. Gastric and intestinal motility are strates a normal Starling relationship between
impaired in cirrhotics secondary to electrolyte end-diastolic pressure and cardiac output [55].
disturbances and ascites [27]. While rapid The cardiac allograft is preload dependent and
sequence induction is ideal to prevent aspiration limited in its tolerance of sudden declines in total
in the presence of a full stomach, it is associated venous return, as could occur with hemorrhage or
with hemodynamic instability among patients clamping of the inferior vena cava [47]. Large
with cardiac failure. Balanced anesthesia using transfusion requirements associated with LTX
opioids, benzodiazepines, and muscle relaxants and ischemia reperfusion injury increase the risk
may be supplemented with low-dose volatile of elevated pulmonary vascular resistance, right
anesthetics to minimize vasopressor require- ventricular systolic dysfunction, and increased
ments and avoid hypotension associated with myocardial demand. Satisfactory right ventricu-
higher concentrations of volatile anesthetics [54]. lar function is necessary to maintain adequate
Cirrhotic patients demonstrate hyperdynamic cardiac output, hemodynamic stability, and end-­
physiology, characterized by low systemic vascu- organ perfusion [48]. PAC monitoring and TEE
lar resistance and high cardiac output [54] and are essential to intra-operative hemodynamic
will almost always require vasopressors to main- management.
tain blood pressure. Reperfusion of the hepatic allograft can be com-
plicated by acidosis, electrolyte abnormalities,
240 G. C. Diaz et al.

hypothermia, and ischemia/reperfusion injury [56]. Early cardiac function dramatically affects the
The “cytokine storm” triggered by ischemia/reper- newly transplanted hepatic graft. Right ventricu-
fusion increases cardiac demand and may precipi- lar failure secondary to prolonged CPB, isch-
tate arrhythmias in the newly transplanted heart. emia/reperfusion injury, or increased pulmonary
VVB offers the theoretical advantage of attenuat- vascular resistance precipitates hepatic conges-
ing sudden declines in venous return and hemody- tion and allograft dysfunction. In addition, biven-
namic instability secondary to allograft reperfusion tricular failure causing systemic hypotension
[57]. Judicious fluid management combined with with increasing vasopressor requirements is del-
immediate correction of electrolyte and acid–base eterious to the hepatic allograft. Successful
abnormalities are imperative for the optimization CHLT while the recipient has required intra-­
of cardiac and hepatic performance. Utilization of operative or immediate post-operative extracor-
the piggyback technique with caval preservation poreal membrane oxygenation and/or ventricular
often avoids the need for VVB [52]. assist devices for poor initial cardiac function has
been reported [43, 59, 60]. The utility of an intra-­
aortic balloon pump has yet to be defined due to
Post-Operative Management concerns regarding interrupted aortic flow to the
celiac trunk, the potential for celiac trunk obstruc-
The postoperative course of the CHLT recipient tion, and a perceived increased risk of hepatic
depends upon the patient’s functional status prior artery thrombosis secondary to disrupted hepatic
to transplantation, intra-operative complications, arterial supply in diastole. While there are many
and the immediate function of both grafts. acceptable ways of performing this complex pro-
Successful recovery requires meticulous, coordi- cedure, an individualized strategy to optimize
nated care balancing the interests of cardiac and operative efficiency and reperfusion time in addi-
hepatic transplant multidisciplinary teams. tion to weighing the risks associated with pro-
Integration, communication, and a precise treat- longed CPB is the goal.
ment plan for nurses and intensivists is essential.
Immunosuppression is typically similar to that
of an isolated heart transplant. Hemodynamics Combined Lung–Liver
should be monitored closely with both a PAC and Transplantation
arterial catheter. Echocardiograms supplement
these data and should be obtained as necessary. Combined lung–liver transplantation (CLLT) is
PAC pressures, mixed venous oxygen saturation, rare with less than 100 total procedures reported
arterial pressures, liver function tests, and urine in the United States, and less than ten performed
output are principal determinants for discontinu- annually [1]. In May 2005, the time-accrual sys-
ation of inotropic and vasopressor support. Chest tem for lung allocation was replaced with the
tube output should be monitored closely and fre- Lung Allocation Score (LAS) to address issues
quent laboratory tests obtained within the initial relating to resource utilization and optimization
24 h including arterial blood gas, lactate, liver of need and benefit [61]. The LAS is a more
function tests, complete blood count, and coagu- objective method to determine allocation of lung
lation panel. A hepatic ultrasound is frequently grafts based upon disease severity, comparable to
obtained to evaluate vascular flow and patency the MELD system for LTX. The LAS score has
within the hepatic allograft [48, 51]. Persistent significantly improved access for sicker patients
coagulopathy from CPB and hepatic dysfunction to lung transplantation but not significantly
may manifest as continued abdominal and tho- improved post-transplant outcomes [61].
racic hemorrhage. Cardiac tamponade must be Given the limited number of CLLT procedures
suspected in the setting of acute hypotension, performed, true performance benchmarks have
elevation with equalization of diastolic pressures, yet to be established. Barshes et al. have reported
or decreased chest tube output [58]. 1- and 5-year patient survival from the SRTR of
19  Combined Solid Organ Transplantation Involving the Liver 241

79% and 63% that is comparable to outcomes of portal hypertension, hypersplenism, variceal
isolated liver or isolated bilateral lung transplan- bleeding, and ultimately ESLD occurs.
tation [62, 63]. In this cohort, the majority of A less frequent indication for CLLT is PPHTN,
patients were children or young adults under the with a 3–8% prevalence among LTX candidates
age of 30. As found in CHLT, there is increased [68, 69]. Mild PPHTN, defined as mean pulmo-
wait-list mortality and no prioritization under nary artery pressures (mPAP) of 25–35 mmHg is
current UNOS allocation policy [64]. Yi et al. amenable to LTX. Patients with moderate
published the largest single-center report of eight PPHTN, defined as mPAP 35–45 mmHg, and
CLLT procedures in 2013 [65]. The authors severe PPHTN of mPAP >45 mmHg who respond
reported a one-year patient survival of 71% and to medical therapy and preserve right heart func-
advocated the procedure be limited to patients tion may be eligible for LTX. Severe PPHTN
with an LAS < 50. refractory to medical therapy, but with preserved
Indications for CLLT are shown in Table 19.3 right heart function indicate CLLT while right
and can be broadly categorized as: end stage lung ventricular dysfunction is a contraindication to
disease (obstructive or restrictive) with advanced CLLT given the high risk of intra-operative mor-
liver disease (for example cystic fibrosis, idio- tality secondary to cardiac failure [70]. These
pathic pulmonary fibrosis or alpha 1 antitrypsin patients require evaluation for combined heart–
deficiency) or end stage liver disease with sec- lung–liver transplantation (CHLLT) [70, 71].
ondarily compromised lung function as found in
portopulmonary hypertension (PPHTN) and
cirrhosis-­related hypoxemia with intrapulmonary Pre-Operative Evaluation
shunting [3, 62, 65].
The most common indication for CLLT is cys- As with other simultaneous transplants, it is
tic fibrosis. Hepatic multilobar cirrhosis is imperative to determine the etiology and severity
observed in 20–30% of cystic fibrosis patients, of end organ disease. A CLLT preoperative proto-
typically in the first decade of life [66]. Focal col proposed by Yi et al. based liver transplant
biliary fibrosis that represents chronic, intrahe- candidacy upon the presence of biopsy proven
patic, biliary obstruction from inspissated, tena- cirrhosis with a portal gradient greater than
cious bile is the characteristic early hepatic 10 mmHg [65]. Lung transplant candidacy was
lesion. Chronic biliary obstruction with resultant based on LAS that was the principal determinant
cholangitis leads to biliary cirrhosis [64]. Serum of allocation.
albumin, prothrombin time, and transaminase Evaluation of cardiac function, particularly
may be normal or only mildly impaired in spite right heart function, is paramount in triaging the
of the presence of advanced multilobar cirrhosis patient to liver–lung versus liver–heart–lung [3,
[67]. Despite these normal values, progression to 65, 71]. Acute right heart failure is the leading
cause of early mortality following CLLT [65].
Table 19.3 Indications for combined liver–lung The ventilation/perfusion scan, mean pulmonary
transplantation artery pressures, and arterial blood gas assist in
End stage lung disease with advanced liver disease determining ventilation strategies. Adequate pre-­
 – Cystic fibrosis operative nutrition is paramount and may not be
 – Alpha-1-antitrypsin deficiency reflected by the body mass index. Coordination
 – Sarcoidosis between the thoracic anesthesiologist, liver trans-
 – Alagille syndrome plant anesthesiologist, thoracic surgeon, liver
End stage liver disease with secondarily compromised surgeon, and perfusionist is essential during the
lung function preoperative period with discussions focused
 – Cirrhosis-associated hypoxemia with upon surgical sequence, catheter placement, car-
intrapulmonary shunting diopulmonary bypass, venovenous bypass, and
 – PPHTN incision location.
242 G. C. Diaz et al.

Intra-Operative Management Intra-operative monitoring should include


radial and femoral arterial catheters, PAC, and
Similar to CHLT, there is variation in CLLT sur- TEE. The PAC is positioned only to the central
gical technique. Lung transplantation is gener- venous position during the initial placement, and
ally performed first followed by LTX. Various relocated more distally into the pulmonary artery
surgical sequences have been reported. The ini- after unclamping of the pulmonary arteries. TEE
tial technique involved integrated, concomitant allows for diagnosis of the etiology of hemody-
dissection of the chest and abdomen prior to namic instability, assessment of RV function
CPB, followed by initiation of CPB and com- after clamping of the pulmonary artery during the
bined en bloc thoracic and liver transplantation lung transplant procedure and during hepatic
[72]. A technique of complete thoracic organ allograft reperfusion, detection of air and throm-
implantation and discontinuation of CPB prior to boembolus, as well as assessment of the surgical
laparotomy, abdominal dissection, and liver anastomosis [3, 73]. Balanced anesthesia with
transplantation has also been reported [64]. The opioids and volatile agents is helpful in providing
latter technique decreases hepatic warm isch- hemodynamic stability.
emia by permitting liver allograft preparation The most common intra-operative complica-
during the thoracic dissection. In this sequence, tion reported is pulmonary hypertension during
abdominal dissection occurs after the reversal of reperfusion of the liver allograft. Zimmerman
heparin. A third technique advocated by Yi et al. et al. reported successful management of severe
begins with complete abdominal dissection prior pulmonary hypertension in a 14-year-old girl
to both the lung and liver implantation. The aim with cystic fibrosis utilizing prostaglandin E 1
is to avoid pulmonary edema within the newly and dobutamine administered via a PAC [74].
transplanted lung(s) from fluid resuscitation and Pirenne reported two cases of CLLT for cirrhosis
blood transfusion associated with the and severe refractory PPHTN. The first case
­hepatectomy [65]. resulted in fatal heart failure after liver reperfu-
There is wide variation in CPB utilization dur- sion despite the use of portal and systemic
ing lung transplantation. Use of CPB has been VVB. The second patient successfully received
advocated by several centers while others utilize en bloc heart–lung transplant followed by liver
one-lung ventilation (OLV) to avoid inflamma- transplant, which had been planned preopera-
tion, dilutional coagulopathy and thrombocyto- tively due to anticipated risk of intra-operative
penia associated with CPB. Avoiding additional heart failure after liver reperfusion [50]. More
causes of coagulopathy is particularly beneficial recently inhaled nitric oxide is advocated as a
in the setting of advanced liver disease [50, 63– selective pulmonary vasodilator without systemic
65, 72]. Prolonged OLV is challenging as hypoxia vasodilation, as well as an important mediator of
and hypercapnia may increase pulmonary vascu- ischemia-reperfusion injury [3, 75].
lar resistance and precipitate right ventricular
failure. Hoechter et al. advocates for pressure
controlled ventilation because of improved Postoperative Management
effects on respiratory function [3]. Pressure-­
controlled volume-guaranteed modes may avoid Coordination between the pulmonary and hepatic
alternating tidal volumes from varying compli- transplant teams is essential as their clinical goals
ance [3]. Goals of ventilation include: minimal are frequently contradictory. Immunosuppression
inspired oxygen fraction to maintain SpO2 is generally dictated by the pulmonary transplant
92–96%, Vt 4–6 mL/kg ideal body weight, PEEP protocol since the risk of rejection is lower for
3–10 cmH2O [3]. Following the transplant, lung liver grafts [65, 71]. While the pulmonary trans-
protective strategies should be employed but it is plant team frequently advocates for early fluid
under study whether the settings should be donor restriction to avoid complications of pulmonary
or recipient based. edema and facilitate early extubation, the liver
19  Combined Solid Organ Transplantation Involving the Liver 243

transplant team is more concerned about main- understanding of the etiology and pathophysi-
taining adequate hydration to avoid hypoperfu- ology resulting in end organ failure as well as
sion of the new liver allograft. Yi and colleagues effective communication between all clinical
reported early successful extubation of CLLT parties to avert morbidity through minimizing
recipients with lower LAS scores while higher ischemia/reperfusion injury and facilitating
LAS scores generally required tracheostomy optimal allograft function.
[65]. Vasopressor use has been implicated in the
high incidence of biliary complications.
References
Open discussion and data-driven management
are critical to a successful outcome. Complications 1. SRTR. United Network for Organ Sharing. In:
to monitor for include sepsis, ischemia of the Scientific Registry of Transplant Recipients; 2016.
bronchial anastomosis, bile duct leak, and bile 2. Ge P, Runyon B. Treatment of patients with cirrhosis.
duct ischemia. Cognitive impairment is a recog- NEJM. 2016;375:767–77.
3. Hoechter D, von Dossow V. Lung transplantation:
nized complication in as many as 67% of lung from the procedure to managing patients with lung
transplant recipients [3]. Prolonged graft isch- transplant. Curr Opin Anaesthesiol. 2016;29:8–13.
emia, use of CPB, OLV and double lung trans- 4. Wiesner R, McDiarmid S, Kamath P, et al. MELD and
plantation correlate with the incidence of PELD: application of survival models to liver alloca-
tion. Liver Transpl. 2001;7:567–80.
cognitive impairment that can be particularly dif- 5. Gonwa T, McBride M, Anderson K, Mai M, Wadei H,
ficult to differentiate from hepatic encephalopa- Ahsan N. Continued influence of preoperative renal
thy or drug toxicity in the CLLT recipient. function on outcome of orthotopic liver transplant in
Laboratory values, such as arterial blood gas, lac- the US: where will MELD lead us? Am J Transplant.
2006;6:2651–9.
tate, ammonia, and liver function tests, are essen- 6. Margreiter R, Kramar R, Huber C, et al. Combined liver
tial in assessing lung and liver allograft function. and kidney transplantation. Lancet. 1984;1:1077–8.
Bronchoscopic examination may be performed 7. Eason J, Gonwa T, Davis C, Sung R, Gerber D,
routinely or when clinically indicated. Doppler Bloom R. Proceedings of consensus conference on
simultaneous liver kidney transplantation (SLK). Am
ultrasonography of the hepatic artery and portal J Transplant. 2008;8:2243–51.
vein is useful in the postoperative period to sup- 8. Puri V, Eason J. Simultaneous liver–kidney transplan-
plement laboratory data and diagnose technical tation. Curr Transpl Rep. 2015;2:297–302.
complications of the liver graft. 9. Formica R, Aeder M, Boyle G, et al. Simultaneous
liver–kidney allocation policy: a proposal to ­optimize
The most common cause of mortality fol- appropriate utilization of scarce resources. Am J
lowing CLLT is sepsis. Clinical experience, Transplant. 2016;16:758–66.
effective communication, and judicious immu- 10. Fong T, Khemichiam S, Shah T, Hutchinson I, Cho
nosuppression are essential to minimize mor- Y. Combined liver–kidney transplantation is prefer-
able to liver transplant alone for cirrhotic patients with
bidity. LAS scores correlate with post-CLLT renal failure. Transplantation. 2012;94:411–6.
morbidity and a ceiling LAS score of 50 has 11. Bellomo R, Ronco C, Kellum J, Mehta R, Palevsy
been proposed where CLLT may be considered P, ADQI Workgroup. Acute renal failure—defini-
too high risk [65]. tion, outcome measures, animal models, fluid ther-
apy and information technology needs: the Second
International Consensus Conference of the Acute
Conclusion Dialysis Quality Initiative (ADQI) Group. Crit Care.
Notable achievements in the performance of 2004;8:R204–R12.
isolated solid organ transplantation have 12. Kellum J, Mehta R, Angus D, Palevsky P, Ronco
C. The first international censensus conference of
broadened the indications for combined solid continuous renal replacement therapy. Kidney Int.
organ procedures and advanced the field. 2002;62:1855–63.
Transplantation of the liver with additional 13. Modi R, Patel N, Metwally S, Mumtaz K. Outcomes
thoracic or abdominal organs is increasing in of liver transplantation in patients with hepatorenal
syndrome. World J Hepatol. 2016;8:999–1011.
frequency. Fundamental to the successful per- 14. Gines A, Escorsell A, Gines P, et al. Incidence, predic-
formance of these procedures from an anes- tive factors, and prognosis of hepatorenal syndrome in
thesia/critical care perspective is an intricate cirrhosis. Gastroenterology. 1993;105:229–36.
244 G. C. Diaz et al.

15. Arroyo V, Gines P, Gerbes A, et al. Definition and diag- 32. Sprung J, Kapural L, Bourke D, O'Hara J. Anesthesia
nostic criteria of refractory ascites and hepatorenal for kidney transplant surgery. Anesthesiol Clin North
syndrome in cirrhosis. Hepatology. 1996;23:164–76. America. 2000;18:919–51.
16. Salerno F, Gerbes A, Gines P, Wong F, Arroyo
33. Agopian V, Dhillon A, Baber J, et al. Liver transplan-
V. Diagnosis, prevention, and treatment of hepatore- tation in recipients receiving renal replacement ther-
nal syndrome in cirrhosis. Gut. 2007;56:1310–8. apy: outcomes analysis and the role of intra-operative
17. Papafragkakis H, Martin P, Akalin E. Combined
hemodialysis. Am J Transplant. 2014;14:1638–47.
liver and kidney transplantation. Curr Opin Organ 34. Kang Y. Transfusion based on clinical coagulation
Transplant. 2010;15:263–8. monitoring does reduce hemorrhage during liver
18. Angeli P, Gines P, Wong F, et al. Diagosis and man- transplantation. Liver Transpl Surg. 1997;3:655–9.
agement of acute kidney injury in patients with cir- 35. Mallet S. Clinical utility of viscoelastic tests of

rhosis: revised consensus recommendations of the coagulation (TEG/ROTEM) in patients with liver dis-
International Club of Ascites. Gut. 2015;64:531–7. ease and during liver transplantation. Semin Thromb
19. Mehta R, Kellum J, Shah S, et al. Acute kidney injury Hemost. 2015;41:527–37.
network: report of an initiative to improve outcomes 36. Lemmens H. Kidney transplantation: recent develop-
in acute kidney injury. Crit Care. 2007;11:R31. ments and recommendations for anesthetic management.
20. Cholongitas E, Calvaruso V, Senzolo M, et al. RIFLE Anesthesiol Clin North America. 2004;22:651–62.
classification as predictive factor of mortality in 37. Faenza S, Arpesella G, Bernardi E, et al. Combined
patients with cirrhosis admitted to intensive care unit. liver transplants: main characteristics from the stand-
J Gastroenterol Hepatol. 2009;24:1639–47. point of anesthesia and support in intensive care.
21. du Cheyron D, Bouchet B, Parienti J, Ramakers M, Transplant Proc. 2006;38:1114–7.
Charbonneau P. The attributable mortality of acute 38. Shaw B, Stratta R, Donovan J, Langnas A, Wood R,
renal failure in critically ill patients with liver cirrho- Markin R. Postoperative care after liver transplanta-
sis. Intensive Care Med. 2005;31:1693–9. tion. Semin Liver Dis. 1989;9:202–30.
22. O'Riordan A, Wong V, McQuillan R, McCormick P, 39. Starzl T, Bahnson H, Hardesty R, et al. Heart–liver
Hegarty J, Watson A. Acute renal disease, as defined transplantation in a patient with familial hypercholes-
by the RIFLE criteria, post-liver transplantation. Am terolaemia. The Lancet. 1984;323:1382–3.
J Transplant. 2007;7:168–76. 40. Olivieri N, Liu P, Sher G, et al. Brief report: combined
23. Wong F, Nadim M, Kellum J, et al. Working party liver and heart transplantation for end-stage iron-­
proposal for a revised classification system of induced organ failure in an adult with homozygous
renal dysfunction in patients with cirrhosis. Gut. beta-thalassemia. N Engl J Med. 1994;330:1125–7.
2011;60:702–9. 41. Surakomol S, Olson L, Rastogi A, et al. Combined
24. Davis C, Feng S, Sung R, et al. Simultaneous liver-­ orthotopic heart and liver transplantation for
kidney transplantation: evaluation to decision making. genetic hemochromatosis. J Heart Lung Transplant.
Am J Transplant. 2007;7:1702–9. 1997;16:573–5.
25. Nadim M, Sung R, Davis C, et al. Simultaneous
42.
Pilato E, Dell’Amore A, Botta L, Arpesella
liver–kidney transplantation summit: current state and G. Combined heart and liver transplantation for famil-
future directions. Am J Transplant. 2012;12:2901–8. ial amyloidotic neuropathy. Eur J Cardiothorac Surg.
26. De Deken J, Kocabayoglu P, Moers C. Hypothermic 2007;32:180–2.
machine preservation in kidney transplantation. Curr 43. DiStefano Y, Cvetkovic D, Malekan R, McGoldrick
Opin Organ Transplant. 2016;21:294–300. K. Anesthetic management of combined heart-liver
27. Sladen R. Anesthetic concerns for the patient with transplantation in a patient with ischemic cardio-
renal or hepatic disease. ASA Refresher Courses in myopathy and cardiac cirrhosis: lessons learned. J
Anesthesiology. 2001;29:213–28. Cardiothorac Vasc Anesth. 2016;31:646–52.
28. Arnal D, Garutti I, Perez-Pena J, Olmedilla L,
44. Careddu L, Zanfi C, Pantaleo A, et al. Combined
Tzenkiv I. Radial to femoral arterial blood pressure heart-liver transplantation: a single-center experience.
differences during liver transplantation. Anaesthesia. Transpl Int. 2015;28:828–34.
2005;60:766–71. 45. Cannon R, Hughes M, Jones C, Eng M, Marvin

29. Krenn C, De Wolf A. Current approach to intraop- M. A review of the United States experience with
erative monitoring in liver transplantation. Curr Opin combined heart-liver transplantation. Transpl Int.
Organ Transplant. 2008;13:285–90. 2012;25:1223–8.
30. Soong W, Sherwani S, Ault M, Baudo A, Herborn 46. Porrett P, Desai S, Timmins K, Twomey C, Sonnad S,
J, De Wolf A. United States practice patterns in the Olthoff K. Combined orthotopic heart and liver trans-
use of transesophageal echocardiography during adult plantation: the need for exception status listing. Liver
liver transplantation. J Cardiothorac Vasc Anesth. Transpl. 2004;10:1539–44.
2014;28:635–9. 47. Diaz G, Renz J, Nishanian E, Kinkhabwala M, Emond
31. Rudnick M, Marchi L, Plotkin J. Hemodynamic mon- J, Wagener G. Anesthetic management of combined
itoring during liver transplantation: a state of the art heart-liver translantation. J Cardiothorac Vasc Anesth.
review. World J Hepatol. 2015;7:1302–11. 2007;21:253–6.
19  Combined Solid Organ Transplantation Involving the Liver 245

48. Nagpal A, Chamogeorgakis T, Shafii A, et al.


62. Barshes N, DiBardino D, McKenzie E, et al.

Combined heart and liver transplantation: the Combined lung and liver transplantation: the United
Cleveland Clinic experience. Ann Thorac Surg. States experience. Clin Transplant. 2005;80:1161–7.
2013;95:179–82. 63. Barshes N, Udell I, Joyce D, Southard R, O'Mahony
49. Offstad J, Schrumpf E, Geiran O, Soreide O, Simonsen C, Goss J. A pooled analysis of posttransplant sur-
S. Plasma exchange and heart-liver transplantation in vival following combined heart-liver transplantation.
a patient with homozygous familial hypercholesterol- Transplantation. 2007;83:95–8.
emia. Clin Transplant. 2001;15(6):432. 64. Couetil J, Houssin D, Soubrane O, et al. Combined
50. Pirenne J, Verleden G, Nevens F, et al. Combined liver lung and liver transplantation in patients with cystic
and (heart-) lung transplantation in liver transplant fibrosis: a 4 1/2-year experience. J Thorac Cardiovasc
candidates with refractory portopulmonary hyperten- Surg. 1995;110:1415–23.
sion. Transplantation. 2002;73:140–56. 65. Yi S, Burroughs S, Loebe M, et al. Combined lung
51. Shaw B, Bahnson H, Hardesty R, Griffith B, Starzl and liver transplantation: analysis of a single-center
T. Combined transplantation of the heart and liver. experience. Liver Transpl. 2013;20:46–53.
Ann Surg. 1985;202:667–72. 66. Lenaerts C, Lapierre C, Patriquin H, et al. Surveillance
52. Barbara D, Rehfeldt K, Heimbach J, Rosen C, Daly R, for cystic fibrosis-associated hepatobiliary disease:
Findlay J. Combined heart-liver transplantation expe- early ultrasound changes and predisposing factors. J
rience and improved organ utilization. Ann Thorac Pediatr. 2003;143:343–50.
Surg. 2015;99:1488–9. 67. Feigelson J, Anagnostopoulos C, Poquet M, Pecau Y,
53. Hennessey T, Backman S, Cecere R, et al. Combined Munck A, Navarro J. Liver cirrhosis in cystic fibrosis-­
heart and liver transplantation on cardiopulmo- therapeutic implications and long term follow up.
nary bypass: report of four cases. Can J Anesth. Arch Dis Child. 1993;68:653–7.
2010;57:355–60. 68. Castro M, Krowka M, Schroeder D, et al. Frequency
54. Baker J, Yost C, Niemann C. Organ transplantation. and clinical implications of increased pulmonary
In: Miller R, editor. Miller’s anesthesia. Philadelphia: artery pressures in liver transplant patients. Mayo Clin
Elsevier; 2005. p. 2231–83. Proc. 1996;71:543–51.
55. Cheng D, Ong D. Anaesthesia for non-cardiac sur- 69. Ramsay M, Simpson B, Nguyen A, Ramsay K, East
gery in heart-transplanted patients. Can J Anesth. C, Klintmalm G. Severe pulmonary hypertension
1993;40:981–6. in liver transplant candidates. Liver Transpl Surg.
56. Steadman R. Anesthesia for liver transplant surgery. 1997;3:494–500.
Anesthesiol Clin North America. 2004;22:687–711. 70.
Krowka M, Mandell M, Ramsay M, et al.
57. Rossi G. Veno-venous bypass versus no bypass
Hepatopulmonary syndrome and portopulmonary
in orthotopic liver transplantation: hemody- hypertension: a report of the multicenter liver trans-
namic, metabolic, and renal data. Transplant Proc. plant database. Liver Transpl. 2004;10:174–82.
1998;30:1871–3. 71.
Scouras N, Matsusaki T, Boucek C, et al.
58. Chuttani K, Tischler M, Pandian N, Lee R, Mohanty Portopulmonary hypertension as an indication for
P. Diagnosis of cardiac tamponade after cardiac sur- combined heart, lung, and liver or lung and liver
gery: relative value of clinical, echocardiographic, and transplantation: literature review and case presenta-
hemodynamic signs. Am Heart J. 1994;127:913–8. tion. Liver Transpl. 2011;17:137–43.
59. Eyraud D, Vaillant J, Ionescu C, et al. Early primary 72. Wallwork J, Williams R, Calne R. Transplantation of
cardiac graft fialure and combined heart-liver trans- liver, heart, and lungs for primary biliary c­ irrhosis and pri-
plantation: need for an uncommon doule bypass. Br J mary pulmonary hypertension. Lancet. 1987;25:182–5.
Anaesth. 2011;107:280–1. 73. Slinger P. Anesthetic management for lung transplan-
60. Fitzsimmons M, Ichinose F, Vagefi P, et al. Successful tation. Tx Med. 2012;24:27–34.
right ventricular mechanical support after combined 74. Zimmerman A, Howard T, Huddleston C. Combined
heart-liver transplantation. J Cardiothorac Vasc lung and liver transplantation in a girl with cystic
Anesth. 2014;28:1583–5. fibrosis. Can J Anesth. 1999;46:571–5.
61. Merlo C, Weiss E, Orens J, et al. Impact of U.S. lung 75. Benedetto M, Romano R, Bacha G. Inhaled nitric
alocation score on survival after lung transplantation. oxide in cardiac surgery: evidence or tradition? Nitric
J Heart Lung Transplant. 2009;28:769–75. Oxide. 2015;49:67–79.
Liver Transplantation
for the Patient with High MELD 20
Cynthia Wang and Randolph Steadman

Keywords  he Model for End-Stage Liver


T
MELD score · Renal insufficiency · Outcome · Disease (MELD) Score
Transfusion · Organ allocation · Futility
MELD is a system for scoring the severity of
liver disease. The model was developed in 2000
Introduction to predict survival in patients undergoing tran-
sjugular intrahepatic portosystemic shunt place-
The model for end-stage liver disease (MELD) ment. In 2002, the Organ Procurement and
score has been used for more than 15 years for the Transplantation Network adopted the MELD
allocation of liver grafts in the U.S. and many score as the standard for prioritization of graft
other countries. As a results liver transplant recipi- allocation for liver transplantation [1–3]. With
ents have been sicker and presented with more few exceptions (hepatocellular carcinoma and
severe liver disease. Patients with severe, decom- acute liver failure for example), those patients
pensated liver disease pose a challenge for the with highest MELD scores have the highest pri-
anesthesiologist as most organ systems are usually ority for organ allocation for liver transplantation
affected. This chapter will review management (LTX) in many countries, including the United
strategies and common complications of liver States. Since the implementation of the MELD
transplant recipients with high MELD scores. system, wait-list mortality has significantly
decreased, waiting time to liver transplantation
has been reduced by over 100 days and the
MELD score has proven to be a good marker for
1-year posttransplantation survival [4–7]. The
MELD score is a composite of three laboratory
values: international normalized ratio (INR) [8],
serum creatinine, and serum bilirubin [9].
MELD = 9.6 × loge(creatinine) + 3.78 ×
C. Wang, MD
Department of Anesthesiology and Pain loge(bilirubin) + 11.2 × loge (INR) + 6.43 9.
Management, VA North Texas Healthcare System, Any laboratory value less than one is set at
Dallas, TX, USA one for the purpose of MELD calculation to pre-
R. Steadman, MD, MS (*) vent negative MELD scores. For serum creati-
Department of Anesthesiology and Perioperative nine levels above 4 mg/dL or for patients
Medicine, UCLA Health, Los Angeles, CA, USA requiring dialysis twice or more per week, a
e-mail: rsteadman@mednet.ucla.edu

© Springer International Publishing AG, part of Springer Nature 2018 247


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_20
248 C. Wang and R. Steadman

c­ reatinine value of 4.0 is entered into the formula plantation is either euvolemic or hypovolemic at
[10]. Patients with high MELD scores (MELD the time of surgery. Regardless of the patient’s
>30) who present to the operating room for LTX initial volume status, fluid management is very
have characteristics that are associated with challenging in this patient population. The poten-
greater perioperative challenges and risks as tial for massive blood loss and high transfusion
compared to patients with lower MELD scores requirements during surgery in an oliguric or
[11, 12]. Although these characteristics are often anuric patient dictates close monitoring of intra-
directly associated with the MELD score (i.e., vascular volume status. While there is no evi-
renal insufficiency and coagulopathy), there are dence that the use of an intraoperative pulmonary
also MELD-unrelated factors in this patient pop- artery catheter improves outcome, many clini-
ulation that contribute significantly to periopera- cians consider it a helpful guide to fluid manage-
tive risk. ment during surgery. Intraoperative use of
transesophageal echocardiography as a monitor
to assess volume status is increasingly common
Renal Insufficiency during liver transplantation and may be espe-
cially useful in patients with high MELD scores
The etiology of preoperative renal insufficiency and/or significant co-morbidities. Central venous
in patients awaiting liver transplantation is often pressure (CVP), though frequently monitored
multifactorial and presents unique perioperative and recorded during liver transplant procedures,
considerations. Patients with end-stage liver dis- is not an accurate reflection of intravascular vol-
ease (ESLD) and coexisting renal failure are at ume status. Multiple studies have shown that
higher risk of death while awaiting transplanta- there is no correlation between CVP and effective
tion when compared to patients with ESLD and circulating blood volume [17]. Vigilant monitor-
preserved renal function [13, 14]. It is estimated ing of fluid administration is crucial, especially
that survival, in the absence of transplantation, in during periods of sudden fluctuation in volume
patients with cirrhosis and renal failure is approx- status, for example during clamping of the vena
imately 50% at 1 month and 20% at 6 months cava and portal vein prior to hepatectomy, vent-
[15]. Post-transplantation, these patients are at ing of the liver prior to reperfusion, and during
higher risk for complications, prolonged hospi- brisk blood loss in the dissection phase.
talization, and decreased survival. Even in patients with preexisting nonoliguric
LTX is a complex and lengthy procedure asso- renal insufficiency the circulatory and hemody-
ciated with major hemodynamic alterations, fluid namic disturbances associated with the transplant
shifts, and metabolic derangements. These procedure may worsen renal function. These
changes are less well tolerated in patients pre- patients may become oliguric intraoperatively,
senting to the operating suite with preexisting most commonly during the anhepatic and neohe-
renal dysfunction whether or not they have been patic phases. There is no evidence that renal pro-
receiving renal replacement therapy (RRT) pre- tective measures such as mannitol, furosemide,
operatively [16]. and dopamine have any benefit in preventing fur-
The intravascular volume status of cirrhotic ther deteriorations of renal [18, 19].
patients with renal dysfunction is difficult to Metabolic abnormalities during liver trans-
assess. Cirrhotic patients are prone to systemic plantation are more frequent and challenging in
vasodilatation, extravasation of intravascular vol- patients with preexisting renal dysfunction.
ume due to low oncotic pressure, and decreased Reperfusion of the newly transplanted graft is
effective circulating blood volume. Volume over- associated with an influx of potassium, lactic
load can occur in patients with renal insuffi- acid, and inflammatory mediators into the circu-
ciency, particularly prior to the institution of lation. Hyperkalemia and acidemia encountered
RRT. More commonly, however, the patient in upon reperfusion can be fatal, especially in
renal failure on RRT who presents for liver trans- patients with compromised renal function who
20  Liver Transplantation for the Patient with High MELD 249

are unable to compensate for these intraoperative Table 20.1  Indications for combined liver and kidney
transplantation
physiologic changes. Furthermore, if the patient
has received large volumes of banked blood, I. Advanced liver disease with chronic kidney
disease
potassium can be dangerously high by the time of
(a) Coincidental
reperfusion, further increasing the risk for life-­
– Glomerulonephritis/glomerulopathy
threatening arrhythmias [20]. Patients with sig- (membranous, membranoproliferative, IgA
nificant acidemia or electrolyte disturbances may nephropathy, focal glomerulosclerosis,
not be able to tolerate reperfusion. These situa- Anti-GbM disease, scleroderma, SLE,
tions must be anticipated, and intraoperative RRT diabetes mellitus)
should be considered prior to arrival to the oper- – Interstitial renal disease (chronic
pyelonephritis, analgaesic nephropathy,
ating room so that sufficient time (at least 1–2 h) sickle cell anaemia, renal transplant failure,
is allotted to correct the acidosis and/or hyperka- sarcoidosis)
lemia prior to reperfusion. Intraoperative contin- – Structural (obstructive uropathy, medullary
uous venovenous hemodialysis (CVVHD) is cystic disease, nephrolithiasis, malignant
hypertension, renal artery thrombosis)
frequently used in many centers; however, it may
(b) Associated
be less effective than single-pass conventional
– Polycystic disease
hemodialysis in correcting acidemia and electro-
– Glomerulonephritis/glomerulopathy
lyte disturbances over a limited time period [21]. associated with viral hepatitis (HBV, HCV)
Large bore venous access is required for CVVHD – HCV chronic liver disease in chronic renal
or single-pass hemodialysis and should be placed failure patients on hemodialysis (HD)
prior to surgery. The decision of whether to initi- (c) Calcineurin inhibitors (CNI) toxicity
ate RRT intraoperatively during LTX is not well II. Advanced liver disease with acute renal failure/
studied. In a retrospective analysis of 500 patients acute on chronic
receiving preoperative RRT, emergency intraop- – Hepatorenal Syndrome (HRS)
erative RRT, when compared to planned or no – Acute Tubular Necrosis (ATN)
III. Metabolic
intraoperative RRT, was associated with more
(a) Affecting both organs
complications. Independent predictors of the
– Sickle cell disease
need of intraoperative RRT included DCD (dona-
– Alpha 1 antitrypsin deficiency
tion after cardiac death) donors, retransplanta-
– Glycogen Storage Disease type I
tion, and preoperative vasopressor use, but not (b) Affecting mainly kidney, liver serving as a
acidosis [22]. Potential benefits of intraoperative gene therapy for correcting the metabolic
RRT must be weighed against risks such as disorder
include emboli and hypothermia, which may be – Primary hyperoxaluria I
more likely intraoperatively when longer circuits – Amyloidosis
with connections are used that are often obscured – Haemolytic uraemic syndrome
by operative drapes. – Methylmalonic acidaemia
Combined liver–kidney transplantation (dis- IV. Miscellaneous
cussed in more detail elsewhere in this book) is – Immunoprotection of kidney in positive
cross-match
indicated in cirrhotic patients with preexisting
– Abdominal fibromatosis
chronic renal disease whose renal failure is not
– COACH syndrome
expected to improve after successful transplanta- – Acute intoxication of chromium-copper
tion of a new liver. Indications are listed in
Chava, S.P., et al., Current indications for combined liver
Table 20.1 [23]. and kidney transplantation in adults. Transplant Rev.
In patients who have developed renal disease (Orlando), 2009; 23(2): p. 111–9
as a result of liver failure, that is, in patients with
hepatorenal syndrome, the guidelines for com- ney transplant in these patients is generally based
bined transplantation are less well defined. The on the length of time the patient has been on RRT
determination to perform a combined liver–kid- prior to surgery. The length of time on RRT, and
250 C. Wang and R. Steadman

hence the time at which renal failure is consid- Massive bleeding and transfusion require-
ered irreversible, has been described from 1 to ments during LTX exacerbate the complex circu-
12 weeks. Patients who required dialysis longer latory and metabolic derangements already
than 3 months prior to liver transplantation have present in patients with end-stage liver disease
an increased survival with combined liver–kid- and are associated with reduced graft and patient
ney transplant compared to isolated liver trans- survival [33]. Despite the fact that many cirrhotic
plantation (87% vs. 75%, P = 0.02) [15]. It has patients have a prolonged INR due to the inabil-
been recommended that patients with severe ity of the liver to synthesize coagulation factors,
renal dysfunction defined by a glomerular filtra- patients with severe liver disease are also at
tion rate <25–35 mL/min, and those with rapidly increased risk of hypercoagulability secondary to
progressing renal disease, should be considered abnormal polymerization of clot and accelerated
candidates for combined liver–kidney transplan- intravascular coagulation. These disturbances in
tation [24]. Other aspects of combined liver–kid- coagulation are often exacerbated by sepsis, cir-
ney transplantation are discussed in more detail culatory failure, or blood loss necessitating mas-
elsewhere in this book. sive transfusion [26, 34]. Inherited thrombophilias
such as protein C and S deficiencies, antithrom-
bin deficiency, factor V Leiden, and lupus antico-
Coagulopathy and  Transfusion agulant may also increase the risk of perioperative
thrombotic events, increasing the morbidity of
Liver transplant surgery is often associated with liver transplant recipients [35, 36].
massive blood loss and transfusion, factors that The data on the use of recombinant factor
are linked to poor postoperative outcomes. VIIa in patients with high MELD scores is lim-
These include a higher incidence of postopera- ited and its indication for these patients contro-
tive infections, hemolysis, allergic reactions, versial. Though recombinant factor VIIa may
and death [25–27]. Patients with high MELD reduce transfusion requirements in selected
scores are at greater risk of requiring large vol- cases, several randomized trials have failed to
umes of intraoperative transfusions than those show a benefit [30–32]. The administration of
with lower MELD scores [28]. Elevated INR factor VIIa may put the patient at risk for poten-
and creatinine levels, both components of the tially fatal thromboembolic complications as
MELD score, have been found to be associated liver failure results not only in coagulopathy, but
with elevated intraoperative blood loss and also hypercoagulability due to decreased levels
transfusion requirements [29]. of anticoagulant factors and elevated levels of
In addition to more severe coagulopathy, von Willebrand factor and factor
patients with high MELD scores often have lower VIII. Administration of factor concentrates such
preoperative hematocrit and fibrinogen levels as prothrombin complex concentrate (PCC) and
[11]. There is a positive correlation between fibrinogen concentrate has become increasingly
MELD score and transfusion requirements dur- common for liver transplantation in the high
ing LTX. Single-center studies have demon- MELD patient. The successful use of factor con-
strated that patients with MELD scores greater centrates has been described in the literature for
than 30 require on average five more units of cardiac surgery and trauma surgery. PCC, as a
packed red blood cells and seven more units of balanced 4-factor concentrate containing factors
fresh frozen plasma when compared to patients II, VII, IX, and X, as well as proteins C and S, has
with lower MELD scores [12]. Transfusion been shown to significantly decrease require-
requirements for cryoprecipitate and platelets ments for allogeneic blood products with no sig-
were also doubled in this patient population. nificant increase in thromboembolic
Furthermore, patients with high MELD scores complications when used with the guidance of
also received rescue anti fibrinolytic agents more viscoelastic testing [37, 38]. When deciding if a
frequently than those with lower MELD scores. patient would benefit from factor concentrate
20  Liver Transplantation for the Patient with High MELD 251

administration, clinical factors and co-morbidi- anti-coagulants in end-stage liver disease [41].
ties in addition to laboratory values must be con- Using viscoelastic testing during the intraopera-
sidered. Those with a history of thrombosis, tive period may help guide transfusion and
small hepatic arteries, or history of hypercoagu- decrease overall requirements for blood products.
lability potentially should be excluded from As aforementioned, many of the algorithms for
receiving factor concentrates. Though no firm the administration of factor concentrates rely on
inclusion/exclusion criteria exist for the adminis- viscoelastic testing values [42, 43].
tration of factor concentrates, these elements in In the high MELD patient, volume replace-
the patient’s medical history should be taken ment therapy is best managed with a combination
under consideration. Trials on the use of fibrino- of packed red blood cells and fresh frozen plasma,
gen concentrate in cardiac and trauma surgical factor concentrates, or a combination of both. In
patients have yielded similar results. Although the setting of poor hemostasis and ongoing coag-
randomized studies on the use of PCC in the liver ulopathy due to hypofibrinogenemia and throm-
transplantation population have yet to be pub- bocytopenia, cryoprecipitate (or fibrinogen
lished, results in other surgical populations are concentrates if available) and platelets may also
encouraging. Overall, the trend toward targeted, be administered. Viscoelastic testing guided
focused repletion of factors and components is management in cardiac surgery is associated with
strong [39, 40]. increased transfusion of fibrinogen [44].
It is prudent to ensure adequate venous access However, there is no absolute transfusion thresh-
and a sufficient supply and easy access to banked old for these products, and transfusion practices
blood products for patients with high MELD may vary by center. The excessive use of crystal-
scores. Particular attention must be paid to the loid and colloid solutions may lead to worsening
presence of other factors that may exacerbate of preexisting coagulopathy by hemodilution and
intraoperative bleeding, such as a history of prior bleeding from overfilled varices. Intraoperative
abdominal surgeries and/or significant portal blood salvage is available at some centers during
hypertension. Monitoring coagulation status by LTX but should not be used in patients with hepa-
following fibrinogen, PT, PTT, INR, and platelet tocellular carcinoma or in patients with bacterial
levels at frequent intervals during the operation peritonitis due to the possibility of bacterial con-
may help guide transfusion therapy. However, it tamination. The use of leukocyte depletion filters
is increasingly apparent that conventional labora- with intraoperative blood salvage devices may
tory tests may not present an accurate, compre- reduce the complications associated with alloge-
hensive picture of the state of coagulation. neic transfusion [45].
Although thromboelastography (TEG®) or rota-
tional thromboelastometry (ROTEM®) is not
used routinely at all institutions, it may provide Severity of Disease
better insight into the complex milieu of coagula-
tion in vivo in the patient with end-stage liver dis- In addition to MELD-related indicators of liver
ease. TEG® or ROTEM® are viscoelastic tests disease (INR, creatinine, and bilirubin), the
that present a dynamic, composite picture of the MELD score has been shown to correlate with
interaction between plasma, blood cells, and MELD-unrelated markers that reflect severity of
platelets. When comparing TEG® parameters to liver disease. Patients with high MELD scores
INR values, TEG® was more predictive of clini- have a higher incidence of ascites and more fre-
cal pathology such as bleeding in patients with quently require preoperative ventilator and vaso-
end-stage liver disease than INR. In fact, in stable pressor support, all markers for advanced disease.
cirrhotics, viscoelastic testing parameters are These patients also have longer preoperative hos-
often within normal limits. This may be a reflec- pital stays, predisposing them to additional co-­
tion of the fact that overall hemostasis may be morbidities prior to liver transplantation [11, 12].
preserved due to a rebalanced state of pro- and Intraoperatively, patients with high MELD scores
252 C. Wang and R. Steadman

have demonstrated a greater need for fluid tion does not take into account the quality of the
boluses and vasopressor infusions. The need for donor organ. Centers may avoid accepting
vasopressors in this patient population may be extended criteria donor organs for the sickest
exacerbated by the increased incidence of patients with the highest MELD scores. Extended
­intraoperative blood loss. Excessive vasopressor criteria organs include those that come from
use may be problematic, causing decreased older donors, donors who have undergone a pro-
hepatic perfusion and potentially worsening out- longed period of mechanical ventilation and/or
come [12]; although in populations with lower hospitalization in an intensive care unit prior to
MELD scores, vasopressor use when combined procurement, organs with evidence of high-grade
with lowered central blood volumes is associated steatosis, or grafts with exceedingly long warm
with reduced intraoperative blood loss [46]. and/or cold ischemia times. The defining features
Ascites alone is associated with increased of the extended criteria organ are not standard-
requirements for intraoperative vasopressors. ized between centers [49–51].
High MELD scores are also associated with In light of the scarcity of organs, the use of
excessive changes in cerebral blood flow during grafts donated after cardiac death (DCD) has
transplantation and that may affect the ability to become increasingly common. For DCD grafts,
assess the etiology of mental status changes. death is declared on the basis of cardiopulmonary
Both a high MELD score and pretransplantation criteria rather than the cessation of brain func-
mechanical ventilation are predictive of postop- tion. This subjects the graft to additional warm
erative altered mental status [47]. Brain perfusion ischemia time due to an often unspecified period
scans during LTX have suggested that patients of hypotension prior to death. Higher incidences
with high MELD scores experience cerebral of non-anastomotic biliary stricture, hepatic
hyperperfusion intraoperatively that may cause artery thrombosis, hepatic abcesses, and primary
neurological damage due to cerebral hyperten- graft nonfunction have been in described in
sive episodes. These neurological complications patients who received DCD organs [52].
can be devastating and a major source of postop- Traditionally, DCD organs have been avoided in
erative morbidity and mortality [47, 48]. the sickest patients with the highest MELD
Other MELD-unrelated factors such as hyper- scores and matching DCD organs with patients
tension, diabetes mellitus, and coronary artery with lower MELD scores may be the best way to
disease exhibit little variation between patients utilize this resource effectively [53–57].
with high vs. low MELD scores. Nevertheless, Donor and graft characteristics have been
understanding both MELD-related and MELD used to create a mathematical model known as
unrelated factors that contribute to increased the donor risk index (DRI) to predict graft sur-
perioperative risk and postoperative morbidity vival. Organs with a high DRI are associated with
and mortality can help guide management, higher rates of graft failure. Recent evidence has
resource utilization, and steps to improve patient also suggests that patients with high MELD
outcomes. scores experience a greater survival benefit when
transplanted low DRI grafts. The survival benefit
of transplantation remains, but is less, when high
Organ Allocation MELD recipients are transplanted with high DRI
grafts [58–61].
Despite the reduction in wait-list mortality with Living-donor liver transplantation is another
the introduction of the MELD system, the scar- source of organs, though living donation presents
city and quality of donor organs remains a major a risk to both the donor and the recipient. Recent
concern when allocating organs to patients await- studies that have compared the differences in sur-
ing liver transplantation. The disproportion vival between patients with high and low MELD
between organ demand and supply continues to scores receiving adult-to-adult living-donor liver
increase and the current system for organ alloca- transplants have suggested that there is no differ-
20  Liver Transplantation for the Patient with High MELD 253

ence in survival between the low MELD group country, thereby increasing waitlist deaths.
and the high MELD group [62, 63]. However, Despite these criticisms, simulations suggested
these findings are based on studies with limited that this model may lead to an overall decrease in
sample sizes and potentially select patient groups, waitlist ­mortality [67].
and further studies of the use of live donors for A national study in 2015 on the effects of this
patients with high MELD scores are needed policy change analyzed data from the SRTR that
before any definitive conclusions can be drawn includes information on all donors, waitlisted
[64]. However, living-donor liver transplantation candidates and transplant recipients in the United
may be a viable option when considering the high States. Liver distribution and mortality within the
wait-list mortality of patients with MELD scores first 12 months following implementation of
above 30 [65]. Share 35 was compared to an equivalent time
period before. Under Share 35, new listings with
MELD ≥35 increased slightly from 9.2% to 9.7%
Share 35 of listings. However, the proportion of deceased
donor liver grafts allocated to recipients with
Although candidates with acute liver failure (sta- MELD ≥35 increased from 23.1% to 30.1%. The
tus 1A) receive the highest priority for liver trans- proportion of regional sharing increased from
plantation, patients with the highest MELD scores 18.9% to 30.4%. The adjusted discard rates
may suffer from equal or even greater mortality decreased by 14%. The waitlist mortality
rates. A review of data from the Scientific Registry decreased by 30% among patients with a baseline
of Transplant Recipients (SRTR) suggests that MELD >30 but patients with lower MELD scores
candidates with MELD >40 have nearly two times experienced no changes in waitlist mortality. Of
the wait-list mortality than status 1A candidates. note, although transport distances and times
For patients with MELD of 36–40, wait-list mor- increased, cold ischemia time was unaffected.
tality rates are no different than status 1A candi- Liver graft quality, as assessed by the DRI, also
dates. This, in addition to the ongoing debate of remained unchanged. Although the authors
how to equitably allocate organs to those who are acknowledge that the observed changes could not
most medically urgent, led to a re-evaluation of be definitively attributed to Share 35, there were
allocation policies in the US [66]. On June 18, no other major shifts in liver transplantation or
2013, the Organ Procurement and Transplantation organ allocation that might result in the substan-
Network (OPTN) in the United States imple- tial decrease in discard rates and waitlist ­mortality
mented a program known as “Share 35”. This [68, 69].
changed the way deceased donor organs were A separate analysis of patients with MELD
allocated. In the pre-Share 35 era, organs were ≥40 revealed similar improvements follow-
offered first to waitlist candidates in the local ing implementation of Share 35. Patients with
Donation Service Area (DSA). Only after these MELD ≥40 are markedly different from other
organs were refused for candidates with MELD liver transplantation patients, as they represent
≥15 in the DSA were they offered to other DSAs the sickest of the cohort. These patients suffer
within the OPTN Region. In the post-Share 35 from significantly higher rates of complications
era, deceased donor livers are offered to candi- such as renal failure and infection in the periop-
dates in the Region with MELD ≥35, regardless erative period. Nationally, the 1-year post-trans-
of DSA. Although this policy would theoretically plantation patient survival for recipients with a
increase the number of regionally shared livers for MELD ≥40 was 10% lower than 1-year survival
patients with higher MELD scores, the concern rates in recipients with MELD <40. Share 35
was that travel distances and times—and there- has effectively decreased the time on the wait-
fore cold ischemia time—could potentially ing list, decreased pre-transplantation hospital-
increase. Furthermore, Share 35 could potentially ization time, and improved national graft and
decrease organ availability in certain parts of the patient survival [70]. These findings are only the
254 C. Wang and R. Steadman

very beginning of the outcomes from Share 35. complicated surgical procedure that is physi-
As more time passes, further analyses on utility, ologically taxing but lifesaving for the patient.
efficiency and cost are necessary to determine the
true sacrifices and benefits of broader sharing for
high-MELD patients. References
1. Avolio AW, Nardo B, Agnes S, et al. The mismatch
Futility choice in liver transplantation: a suggestion for the
selection of the recipient in relation to the characteris-
tics of the donor. Transplant Proc. 2005;37(6):2584–6.
A single center retrospective study of 169 (13%) 2. Malinchoc M, Kamath PS, Gordon FD, et al. A
of 1522 liver transplant recipients who had a model to predict poor survival in patients undergo-
MELD score of 40 or more attempted to deter- ing transjugular intrahepatic portosystemic shunts.
mine independent factors associated with the Hepatology. 2000;31(4):864–71.
3. Wiesner RH, McDiarmid SV, Kamath PS, et al.
poorest post-transplant outcomes [71]. Recipients MELD and PELD: application of survival models to
with futile outcomes, defined as post-transplant liver allocation. Liver Transpl. 2001;7(7):567–80.
mortality within three-months or prior to hospital 4. Freeman RB, Harper A, Edwards EB. Excellent liver
discharge, included a higher proportion with transplant survival rates under the MELD/PELD sys-
tem. Transplant Proc. 2005;37(2):585–8.
increased cardiac risk, age-adjusted Charlson 5. Saab S, Wang V, Ibrahim AB, et al. MELD score
Co-morbidity Index ≥6, life support (mechanical predicts 1-year patient survival post-orthotopic liver
ventilation or RRT) and septic shock prior to transplantation. Liver Transpl. 2003;9(5):473–6.
transplant. Cardiac risk included severe valvular 6. Huo TI, Lee SD, Lin HC. Selecting an optimal
prognostic system for liver cirrhosis: the model
disease (aortic stenosis <1 cm2, severe tricuspid for endstage liver disease and beyond. Liver Int.
regurgitation), coronary artery disease with 2008;28(5):606–13.
>70% stenosis or revascularization, history of 7. Kamath PS, Wiesner RH, Malinchoc M, et al. A
ventricular tachycardia or atrial fibrillation, ele- model to predict survival in patients with end-stage
liver disease. Hepatology. 2001;33(2):464–70.
vated pre-transplant troponin (>0.2 ng/mL) and/ 8. Weinrieb RM, Lucey MR. Treatment of addictive
or new wall motion abnormalities. Cardiac and behaviors in liver transplant patients. Liver Transpl.
infectious causes of death were more frequent in 2007;13(11 Suppl 2):S79–82.
the futile than in the non-futile groups. In a study 9. Kamath PS, Kim WR. The model for end-stage liver
disease (MELD). Hepatology. 2007;45(3):797–805.
that analyzed United Network of Organ Sharing 10. Huo TI, Lin HC, Lee SD. Model for end-stage liver
database recipients with MELD scores >40 (8% disease and organ allocation in liver transplantation:
of 33,400 recipients between 2002 and 2011), where are we and where should we go? J Chin Med
predictors of futility included age >60 years, obe- Assoc. 2006;69(5):193–8.
11. Xia VW, Taniguchi M, Steadman RH. The changing
sity, intensive care unit admission with mechani- face of patients presenting for liver transplantation.
cal ventilation and multiple co-morbidities [72]. Curr Opin Organ Transplant. 2008;13(3):280–4.
Despite this, patients with MELD scores >40 12. Xia VW, Du B, Braunfeld M, et al. Preoperative char-
demonstrate the largest increase in post-­transplant acteristics and intraoperative transfusion and vaso-
pressor requirements in patients with low vs. high
survival, so the benefit of transplantation must be MELD scores. Liver Transpl. 2006;12(4):614–20.
weighed against the perioperative risk [60, 61]. 13. Wong LP, Blackley MP, Andreoni KA, et al. Survival
of liver transplant candidates with acute renal fail-
Conclusions ure receiving renal replacement therapy. Kidney Int.
2005;68(1):362–70.
The patient with a high MELD score who 14. Biancofiore G, Davis CL. Renal dysfunction in the
comes to the operating suite for an LTX pres- perioperative liver transplant period. Curr Opin Organ
ents a unique set of challenges in the periop- Transplant. 2008;13(3):291–7.
erative period. A thorough understanding of 15. Gines P, Schrier RW. Renal failure in cirrhosis. N
Engl J Med. 2009;361(13):1279–90.
the impact of the MELD score on the manage- 16. Townsend DR, Bagshaw SM, Jacka MJ, et al.

ment of these patients is crucial to success- Intraoperative renal support during liver transplanta-
fully navigate through a technically tion. Liver Transpl. 2009;15(1):73–8.
20  Liver Transplantation for the Patient with High MELD 255

17. Marik PE, Baram M, Vahid B. Does central venous 35. Spaggiari M, Di Benedetto F, Masetti M, et al. The
pressure predict fluid responsiveness? A systematic impact of inherited thrombophilia on liver transplan-
review of the literature and the tale of seven mares. tation. Transplantation. 2009;87(7):1103–4.
Chest. 2008;134(1):172–8. 36. Bustelos R, Ayala R, Martinez J, et al. Living donor
18. Planinsic RM, Lebowitz JJ. Renal failure in end-stage liver transplantation: usefulness of hemostatic
liver disease and liver transplantation. Int Anesthesiol and prothrombotic screening in potential donors.
Clin. 2006;44(3):35–49. Transplant Proc. 2009;41(9):3791–5.
19. Ozier Y, Klinck JR. Anesthetic management of
37. Gorlinger K, et al. Reduction of fresh frozen plasma
hepatic transplantation. Curr Opin Anaesthesiol. requirements by perioperative point-of-care coagula-
2008;21(3):391–400. tion management with early calculated goal-directed
20. Xia VW, Ghobrial RM, Du B, et al. Predictors of therapy. Transfus Med Hemother. 2012;39(2):104–13.
hyperkalemia in the prereperfusion, early postre- 38. Schochl H e a. Trauma bleeding management: the
perfusion, and late postreperfusion periods dur- concept of goal-directed primary care. Anesth Analg.
ing adult liver transplantation. Anesth Analg. 2014;119(5):1064–73.
2007;105(3):780–5. 39. Grottke O. Coagulation management. Curr Opin Crit
21. Davies HT, Leslie GD. Intermittent versus continuous Care. 2012;18(6):641–6.
renal replacement therapy: a matter of controversy. 40. Kircher C, et al. Coagulation management with factor
Intensive Crit Care Nurs. 2008;24(5):269–85. concentrates in liver transplantation: a single-center
22. Agopian VG, et al. Liver transplantation in recipients experience. Transfusion. 2014;54(10):2760–8.
receiving renal replacement therapy: outcomes analy- 41. Lisman T, et al. Hemostasis and thrombosis in patients
sis and the role of intraoperative hemodialysis. Am J with liver disease: the ups and downs. J Hepatol.
Transplant. 2014;14(7):1638–47. 2010;53:362–71.
23. Chava SP, Singh B, Zaman MB, et al. Current indica- 42. Mallett SV. Clinical utility of viscoelastic tests of
tions for combined liver and kidney transplantation in coagulation (TEG/ROTEM) in patients with liver dis-
adults. Transplant Rev (Orlando). 2009;23(2):111–9. ease and during liver transplantation. Semin Thromb
24. Jeyarajah DR, McBride M, Klintmalm GB, et al.
Hemost. 2015;41(5):527–37.
Combined liver-kidney transplantation: what are the 43. Stravitz RT. Potential applications of thromboelastog-
indications? Transplantation. 1997;64(8):1091–6. raphy in patients with acute and chronic liver disease.
25. Donaldson MD, Seaman MJ, Park GR. Massive blood Gastroenterol Hepatol. 2012;8(8):513–20.
transfusion. Br J Anaesth. 1992;69(6):621–30. 44. Spalding GJ, et al. Cost reduction of perioperative
26. Ozier Y, Albi A. Liver transplant surgery and transfu- coagulation management in cardiac surgery: value
sion. Int Anesthesiol Clin. 2004;42(3):147–62. of ‘bedside’ thrombelastography (ROTEM). Eur J
27. Wojciechowski PJ, Samol N, Walker J. Coagulopathy Cardiothorac Surg. 2007;31:1052–7.
in massive transfusion. Int Anesthesiol Clin. 45. Liang TB, Li JJ, Li DL, et al. Intraoperative blood
2005;43(4):1–20. salvage and leukocyte depletion during liver trans-
28. Massicotte L, Sassine MP, Lenis S, et al. Transfusion plantation with bacterial contamination. Clin Transpl.
predictors in liver transplant. Anesth Analg. 2010;24(2):265–72.
2004;98(5):1245–51; table of contents. 46. Massicotte L, et al. Effects of phlebotomy and phen-
29. McCluskey SA, Karkouti K, Wijeysundera DN, et al. ylephrine infusion on portal venous pressure and
Derivation of a risk index for the prediction of mas- systemic hemodynamics during liver transplantation.
sive blood transfusion in liver transplantation. Liver Transplantation. 2010;89(8):920–7.
Transpl. 2006;12(11):1584–93. 47. Panzera P, Cicco G, Memeo R, et al. MELD predic-
30. Planinsic RM, van der Meer J, Testa G, et al. Safety and tive value of alterations of brain perfusion during liver
efficacy of a single bolus administration of recombi- transplantation. Transplant Proc. 2005;37(6):2622–5.
nant factor VIIa in liver transplantation due to chronic 48. Kanwal F, Chen D, Ting L, et al. A model to predict
liver disease. Liver Transpl. 2005;11(8):895–900. the development of mental status changes of unclear
31. Lodge JP, Jonas S, Jones RM, et al. Efficacy and cause after liver transplantation. Liver Transpl.
safety of repeated perioperative doses of recombinant 2003;9(12):1312–9.
factor VIIa in liver transplantation. Liver Transpl. 49. Cameron AM, Ghobrial RM, Yersiz H, et al. Optimal
2005;11(8):973–9. utilization of donor grafts with extended criteria: a
32.
Niemann CU, Behrends M, Quan D, et al. single-center experience in over 1000 liver trans-
Recombinant factor VIIa reduces transfusion require- plants. Ann Surg. 2006;243(6):748–53. discussion
ments in liver transplant patients with high MELD 753–5.
scores. Transfus Med. 2006;16(2):93–100. 50. Gastaca M. Extended criteria donors in liver trans-
33. Boin IF, Leonardi MI, Luzo AC, et al. Intraoperative plantation: adapting donor quality and recipient.
massive transfusion decreases survival after liver Transplant Proc. 2009;41(3):975–9.
transplantation. Transplant Proc. 2008;40(3):789–91. 51. Renz JF, Kin C, Kinkhabwala M, et al. Utilization
34. Warnaar N, Lisman T, Porte RJ. The two tales of of extended donor criteria liver allografts maximizes
coagulation in liver transplantation. Curr Opin Organ donor use and patient access to liver transplantation.
Transplant. 2008;13(3):298–303. Ann Surg. 2005;242(4):556–63. discussion 563–5.
256 C. Wang and R. Steadman

52. Pine JK, Aldouri A, Young AL, et al. Liver trans- 62. Hayashi PH, Forman L, Steinberg T, et al. Model for
plantation following donation after cardiac death: end-stage liver disease score does not predict patient
an analysis using matched pairs. Liver Transpl. or graft survival in living donor liver transplant recipi-
2009;15(9):1072–82. ents. Liver Transpl. 2003;9(7):737–40.
53. de Vera ME, Lopez-Solis R, Dvorchik I, et al. Liver 63. Akyildiz M, Karasu Z, Arikan C, et al. Impact of pre-
transplantation using donation after cardiac death transplant MELD score on posttransplant outcome in
donors: long-term follow-up from a single center. Am living donor liver transplantation. Transplant Proc.
J Transplant. 2009;9(4):773–81. 2004;36(5):1442–4.
54. Mateo R, Cho Y, Singh G, et al. Risk factors for graft 64. Marubashi S, Dono K, Asaoka T, et al. Risk fac-
survival after liver transplantation from donation after tors for graft dysfunction after adult-to-adult liv-
cardiac death donors: an analysis of OPTN/UNOS ing donor liver transplantation. Transplant Proc.
data. Am J Transplant. 2006;6(4):791–6. 2006;38(5):1407–10.
55. Reich DJ, Mulligan DC, Abt PL, et al. ASTS recom- 65. Selzner M, Kash fi A, Cattral MS, et al. Live donor
mended practice guidelines for controlled donation liver transplantation in high MELD score recipients.
after cardiac death organ procurement and transplan- Ann Surg. 2010;251(1):153–7.
tation. Am J Transplant. 2009;9(9):2004–11. 66. Sharma P, et al. End-stage liver disease candidates at
56. Grewal HP, Willingham DL, Nguyen J, et al. Liver the highest MELD scores have higher wait-list mor-
transplantation using controlled donation after car- tality than status-1A candidates. Hepatology. 2012
diac death donors: an analysis of a large single-center Jan;55(1):192–8.
experience. Liver Transpl. 2009;15(9):1028–35. 67. Washburn K, et al. Liver allocation and distribution:
57. Foley DP, Fernandez LA, Leverson G, et al. Donation possible next steps. Liver Transpl. 2011;17:1005–12.
after cardiac death: the University of Wisconsin 68. Massie AB, et al. Early changes in liver distribu-
experience with liver transplantation. Ann Surg. tion following implementation of Share 35. Am J
2005;242(5):724–31. Transplant. 2015;15:659–67.
58. Avolio AW, Siciliano M, Barbarino R, et al. Donor 69. Feng S, al e. Share 35: a liver in time saves lives? Am
risk index and organ patient index as predictors of J Transplant. 2015;15:581–2.
graft survival after liver transplantation. Transplant 70. Nekrasov V, et al. National outcomes of liver trans-
Proc. 2008;40(6):1899–902. plantation for Model for End-stage Liver Disease
59. Feng S, Goodrich NP, Bragg-Gresham JL, et al.
score ≥40: the impact of Share 35. Am J Transplant.
Characteristics associated with liver graft failure: 2016;16:2912–24.
the concept of a donor risk index. Am J Transplant. 71. Petrowsky H, et al. Liver transplantation in highest
2006;6(4):783–90. acuity recipients: identifying factors to avoid futility.
60. Merion RM, Schaubel DE, Dykstra DM, et al.
Ann Surg. 2014;259:1186–94.
The survival benefit of liver transplantation. Am J 72. Panchal HJ, et al. Survival outcomes in liver trans-
Transplant. 2005;5(2):307–13. plant recipients with Model for End-stage Liver
61. Schaubel DE, Sima CS, Goodrich NP, et al. The sur- Disease scores of 40 or higher: a decade-long experi-
vival benefit of deceased donor liver transplantation ence. HPB. 2015;17:1074–84.
as a function of candidate disease severity and donor
quality. Am J Transplant. 2008;8(2):419–25.
Perioperative Considerations
for Transplantation in Acute Liver 21
Failure

C. P. Snowden, D. M. Cressey, and J. Prentis

Keywords Patient Population


Fulminant hepatic failure · Intracranial pres-
sure · Coagulopathy · Fluid management · The paucity of literature concerning the anes-
Recovery · Intracranial hypertension thetic management of ALF patients may in some
part be explained by the fact that they comprise
only 7% [1] to 10% [2] of all liver transplantation
Introduction recipients. Paracetamol-induced liver injury
remains the major etiological factor for ALF in
In contrast to the intensive care management of adult patients in North America [3]. Similarly in
the patient with acute liver failure (ALF), the the UK, paracetamol remains the predominant
perioperative anesthetic management of emer- cause of ALF, but following the restrictions
gency liver transplantation for acute liver failure imposed on sales of paracetamol in 1998, the
has received limited attention in the medical lit- incidence has decreased over the last two decades
erature. Although many of the principles regard- and is now estimated at 39% [4, 5]. In contrast, in
ing recipient management can be transferred South Asia and Hong Kong, the most likely cause
from the intensive care unit (ICU) setting into the of ALF is viral hepatitis with drug-induced liver
intraoperative period, the liver transplantation injury being less commonly observed. The per-
procedure (including transfer to the operating sistence of paracetamol-induced injury as the
room) creates specific concerns that require par- major etiological factor for ALF in Western
ticular anesthetic attention. countries is still reflected in a predominantly
younger age group presenting for emergency
liver transplantation, with less co-morbid disease
burden to increase perioperative risk. However,
the rapid onset of preoperative multiorgan failure
(MOF), especially involving cardiovascular,
renal and cerebrovascular dysfunction, creates
specific practical and physiological challenges
for the transplant anesthesiologist.
C. P. Snowden, B Med Sci (Hons), FRCA, MD (*)
D. M. Cressey, BSc (Hons), MBBS, FRCA Even with the development of MOF, the cur-
J. Prentis, MBBS, FRCA rent outcome of patients with ALF who undergo
Department of Perioperative Medicine and Critical transplantation is excellent and in some series
Care, Freeman Hospital, Newcastle Upon Tyne, UK (especially following paracetamol poisoning)
e-mail: Chris.Snowden@nuth.nhs.uk

© Springer International Publishing AG, part of Springer Nature 2018 257


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_21
258 C. P. Snowden et al.

rivals that of elective liver transplantation for Table 21.1  Key discussion points prior to transfer of
chronic disease. In 2012 the European Liver patients to the operating room
Transplant Registry (ELTR) reported 1, 5 and Factor to
10 year survival of 74%, 68% and 63% with graft consider Discussion points
Invasive • Vascular access and line position
survival of 63%, 57% and 50%, respectively [6, 7].
access/ related to operating room
A newer study from the UK suggests even better monitoring requirements
1 year survival rates of 86% [8]. The improve- • Available method of cardiac
ment in outcome is due to appropriate early iden- output assessment
• Presence of ICP measurement
tification of patients who may benefit from
device
transplantation, expedient transfer to an appro- • Access for established continuous
priate center for specialist management, the rec- veno-venous haemofiltration
ognition of encephalopathy as an important (CVVH)
indicator of clinical progression and aggressive Ventilation • Modes and pressure settings
required to maintain adequate
intensive care therapy from the outset. Distinction parameters oxygenation and PaCO2 levels
of patients too sick to survive the transplantation • Availability of these same
procedure or who are likely to have poor post-­ ventilation modes in operating
operative survival (i.e. recidivist heavy intake room
• Presence/absence of permeability
alcoholics, active repeat suicide risk) may have pulmonary oedema.
also improved outcome figures. There is also Stability issues • Cardiovascular and ICP stability
value in distinguishing patients who show early prior to operating room transfer
signs of recovery [8, 9]. In addition, where time • Specific vasopressor doses
from listing to organ procurement is extended, a • Response to therapy.
degree of self selection will occur, whereby exist- Renal support • Overall fluid balance
• Details of CVVH flow rates and
ing supportive measures are not able to maintain dialysate composition
rapidly deteriorating patients and the decision is Sedation and • Regimes and responses to
taken to remove the patient from the waitlist. paralysis sedation regime
This often leaves the most physiologically adapt- • Recent administration of paralysis
able patients to receive the available organs. If agent
Coagulation • Adequacy of preoperative
artificial liver support and bridging therapies
issues correction
improve it may be possible to support even sicker • Availability of pre-ordered blood
patients until transplantation. However, at present products
there is no evidence to suggest existing bridging
therapies alter outcome in ALF [10].
ventilatory strategies and renal support, is impor-
tant to maintain stability for the forthcoming opera-
Preoperative Considerations tive period. Relevant practical considerations prior
to transfer of the patient to the operating operating
Preoperative management of the ALF patient in the room are shown in Table 21.1. All infusions should
ICU is particularly relevant to the transplant anes- be continued to ensure stability and nearly com-
thesiologist and early communication with the pleted drug infusions changed prior to transfer.
intensivist is important prior to and following Ventilation is optimally provided by portable
transplantation listing. Patient transfer is poten- mechanical ventilation given the inherent variation
tially destabilising and must be performed care- in manual “bag” ventilation with the risk of hyper-
fully. The specific supportive measures for MOF capnia and intracranial hypertension. Patients with
will already have been established in the ICU as established lung injury require maintenance of
part of pre-operative optimization. Continuation of appropriate positive end-expiratory pressure levels.
these modalities into the operating room, including Head positioning including 15-degree head raise in
inotropic and vasopressor infusions, protective the neutral position must also be ensured to avoid
21  Perioperative Considerations for Transplantation in Acute Liver Failure 259

intracranial pressure (ICP) elevations. Many gical experience and expertise. No published
­centers prefer not to use continuous muscle relax- study has demonstrated an advantage of any one
ation in the ICU to reduce the risk of critical illness surgical technique for ALF. However conven-
neuro-myopathy. However, muscle paralysis prior tional caval clamping technique without veno-­
to operating room transfer reduces the risk of venous bypass is—in the authors’ opinion—likely
surges in ICP associated with valsalva manoeuvres to result in more cardiovascular and neurological
caused by coughing and allow more consistent challenges due to the dramatically decreased
ventilation during transfer. If continuous veno- venous return and the potential need for increased
venous hemofiltration (CVVH), has been used in fluid administration [17]. However, use of veno-­
the ICU and is to be recommenced in the operating venous bypass is associated with the risks of
room, it is advisable to electively “wash back” the complex line insertion even though it may result
circuit prior to transfer, as mechanical cranking of in more cardiovascular stability. The “Piggy-
circuits during transfer is impractical. back” technique, especially when using a tempo-
rary porto-caval shunt to maintain splanchnic
venous drainage during crossclamp, may be pref-
Pre-emptive Total Hepatectomy erential for patients with ALF.

When total cardiovascular or neurological col-


lapse secondary to liver failure seems imminent Anesthetic Considerations
and a donor organ is not yet available, the possi-
bility of elective total hepatectomy with portoca- The primary considerations, for the anesthesiolo-
val shunting prior to liver transplantation should gist involved in transplantation for patients with
be considered as “toxic liver syndrome” may be ALF (in addition to those for non-emergency
treated with total hepatectomy [11, 12]. In most transplantation) are:
reports, this procedure has demonstrated a stabi-
lizing effect on the neurological status in patients (a) Cerebrovascular stability (closely linked to
with ALF [13, 14]. In contrast, the effect on car- cardiovascular stability)
diovascular stability has been variable [15, 16]. (b) Avoidance of severe coagulopathy
This procedure provokes some difficult ethical (c) Perioperative fluid balance (including the use
issues as once the liver is removed, the patient of intraoperative continuous veno-venous
clearly has no hope of survival beyond a limited hemofiltration (CVVH))
time without a donor organ. If there was any (d) Potential for use of marginal donor organs
doubt that the patient might have a chance to sur- and ABO incompatible donor organs
vive without a transplant, then the ethic of “do no
(e) Acceptance of requirement for extended
harm” may be evoked. In our institution, hepatec- postoperative recovery
tomy has only been used in extreme cases if a
suitable donor organ is known to be available and Cerebrovascular Stability
harvest is imminent, or more commonly if the Patients with fulminant hepatic failure (FHF) and
donor organ has been already viewed and deemed encephalopathy may have impaired cerebral
macroscopically usable. autoregulation and variations in mean arterial
pressure will tend to result in marked changes in
cerebral blood flow. It follows that cardiovascular
Liver Transplantation Procedure stability during transplantation is of paramount
importance to the maintenance of cerebral perfu-
Surgical Considerations sion pressure (CPP). A decrease in cerebral com-
pliance leading to an increase in ICP may occur
The type of surgical procedure for emergency through either excess cerebral blood flow or an
liver transplantation will depend on regional sur- increase in interstitial fluid secondary to
260 C. P. Snowden et al.

e­ ndothelial leak. Alternatively, ICP rises may be early report [24] patients always had higher ICP
secondary to ischemia. Gaining a balance during surgery compared to preoperative ICU
between the two distinct entities is critical to values. Detry et al. [21] suggested that those
cerebral protection during transplantation. patients who developed preoperative rises in ICP
Many patients with acute liver failure have evi- may be at greater risk of intraoperative changes
dence of cerebral “luxury” perfusion and cerebral in intracranial pressure presumably representing
hyperaemia secondary to reduced cerebrovascular a reduced brain compliance. However, this find-
resistance. It has been demonstrated that ICP surges ing has not been universally accepted. Individual
in FHF are likely due to an increase in cerebral variation may be explained by the complex rela-
blood flow (CBF) [18]. Intraoperative measure- tionship between CBF, CPP and cerebrovascular
ments of the ICP, CMRO2 and CBF during trans- resistance in patients with abnormal autoregula-
plantation in patients with FHF have also tion, that in itself is not an “all or nothing”
demonstrated that ICP increases are frequently phenomenon.
more related to rises in CBF than ischemia induced Given this degree of variation of ICP response
by reduction in CPP secondary to systemic hypo- during transplantation, the management of phasic
tension [19]. Therefore, although a threshold CPP ICP changes during the procedure is complex.
must be maintained, relative hypertension during The use of moderate hypothermia to control
and after reperfusion, may be more detrimental in changes in ICP has been applied to patients with
terms of increasing the risk of increased microvas- FHF [25]. Jalan et al. [22] have shown that mod-
cular pressure, cerebral hyperperfusion and ulti- erate hypothermia abolished ICP variability
mately cerebral edema. Unfortunately, intraoperative throughout the transplantation procedure even in
changes of ICP are often hemodynamically silent. patients with difficult to control ICP prior to
Direct measurement of the ICP may be advanta- transplantation. Other evidence, suggests that
geous before and during liver transplantation in hypothermia may reinstate cerebral autoregula-
order to rapidly identify and treat increases of ICP; tion and reduce cerebral hyperperfusion [26, 27].
there is however a definite risk of intracerebral Even though the significance of these changes
haemorrhage with instrumentation in the presence has not been demonstrated in an outcome study,
of coagulopathy. hypothermia and especially mild hypothermia
Changes in ICP are often temporally predict- can be considered a reasonable therapeutic strat-
able during different stages of emergency trans- egy where ICP control is troublesome.
plantation. Lidofsky [20], demonstrated that Whilst hypothermia can be used as an impor-
peaks of ICP occurred during the dissection, tant baseline strategy for reducing surges in ICP,
anhepatic and early reperfusion phases. other interventions may be required. Variations in
However, in more recent reports [21, 22], the ICP during the early dissection phase can be
changes in ICP seem to occur more consistently reduced by expeditious hepatic artery/portal vein
during the reperfusion and dissection phase only clamping. The development of an anhepatic state
while ICP remains stable or may even decrease often promotes a reduction in the requirement for
during the anhepatic phase. The temporal vasoconstrictors and inotropes [28], enabling bet-
changes of ICP during the transplantation pro- ter cardiovascular stability. Furthermore, rapid
cedure have been attributed to various mecha- fluid removal via CVVH and mild hyperventila-
nisms including release of inflammatory tion in anticipation of increased CO2 production,
substances from the failing liver, de novo cyto- may attenuate increases in intracranial pressures
kine production from the newly perfused liver at reperfusion. Where acute rises in ICP occur,
and cerebral hyperperfusion secondary to an standard active measures including the use of
increase in venous return [23]. mannitol and single dose indomethacin (25 mg)
Although, temporal ICP rises during the pro- remain the emergency measures of choice.
cedure are somewhat predictable, the cerebral Filho and colleagues [29] provide preliminary
response of an individual patient is variable. In an evidence to promote the use of hypertonic saline
21  Perioperative Considerations for Transplantation in Acute Liver Failure 261

in patients undergoing transplantation for controlled hypovolemia [31].Whilst cell salvage


FHF. Their study compared ten patients receiving is ideally suited to transplantation for ALF, rela-
hypertonic saline, to historical controls and dem- tive hypotension and controlled hypovolemia
onstrated a stabilisation of cerebral perfusion conflict with the need to maintain optimal cardio-
pressure during the anhepatic phase and an vascular stability for neurological protection. In
increase following reperfusion. This was associ- the pre-reperfusion phase of transplantation, the
ated with a higher level of sodium at the end of emphasis is on maintaining adequate coagulation
the anhepatic pause and 3 hours post reperfusion. control secondary to the complete loss of liver
However use of hypertonic saline can rapidly function while maintaining a neutral circulating
increase plasma sodium levels and may expose volume if blood loss becomes prominent.
patients with preoperative hyponatremia to the The use of factor concentrates instead of
risk of central pontine myelinolysis. whole blood components is now standard. These
include;
Coagulopathy
Given that the vast majority of procoagulant and • Fibrinogen concentrates
anticoagulant factors are either synthesized or • Low fibrinogen levels have been associated with
metabolised in the liver (von Willebrand factor, increased blood loss and requirement of blood
tissue plasminogen activator (t-PA) and thrombo- products. A fibrinogen level of >2 g/L seems to be
modulin being amongst the exceptions), the the optimal level to prevent clot instability [32]. A
development of ALF can lead to complex multi- fibrinogen concentration <1.5 or signs of fibrino-
factorial coagulopathy. Although, spontaneous gen deficiency on functional testing (TEG/
hemorrhage is relatively uncommon in ALF in ROTEM) may be used as a trigger for consider-
the ICU, early correction of coagulopathy is ation of use. Fibrinogen concentrates have the
important when operative intervention is immi- advantage of providing a standard dose with
nent. Standard methods of correction utilize FFP, reduced risk of pathogen transmission.
cryoprecipitate and platelets guided by labora- Nevertheless, there is limited evidence to demon-
tory studies. However, this may lead to excess strate that fibrinogen concentrates actually reduce
blood product transfusion. Indeed the balance blood transfusion requirements [33]. A placebo-
between blood product transfusion to control controlled study of the pre-emptive use of fibrino-
coagulopathy and replace blood loss, in the set- gen concentrates showed no effect on transfusion
ting of cerebral dysfunction remains a constant requirements [34] in patients undergoing trans-
concern throughout the transplantation proce- plantation, although there were less thrombotic
dure. Blood conservation techniques to avoid complications in the fibrinogen concentrate group.
large-volume transfusion is recommended, whilst • Prothrombin complex concentrates
maintaining coagulation stability with alternative • Prothrombin Complex Concentrates (PCC)
coagulation strategies and the minimal use of are purified coagulation concentrates from
selective blood products. pooled plasma, containing the vitamin K
Visco-elastic coagulation testing such as dependent factors, with approximately 25
thromboelastography (TEG) or rotational throm- times higher concentrations of clotting factors
belastometry (ROTEM) can provide a measure of [35]. Like FC, they allow the correction of
overall clot formation and function, including the coagulation using small volumes with lower
presence of hyperfibrinolysis, within 20–30 min. risk of viral transmission. Current evidence
TEG guided therapy as a whole (including deter- suggests that even in high-risk patients for
mining the need for platelets, FFP and cryopre- thrombosis, PCCs are safe and that thrombo-
cipitate) may reduce transfusion requirement by embolic events are rare [36]. A randomised
up to 33% [30]. Other methods to avoid excessive controlled trial (the PROTON trial) studying
transfusion in acute liver transplantation include PCCs effect on RBC transfusion requirements
the use of cell salvage, relative hypotension and in LTx is currently in progress [37].
262 C. P. Snowden et al.

• Recombinant factor VIIa and standard laboratory coagulation studies


• Recombinant factor VIIa (rFVIIa) has been will not identify fibrinolysis. The diagnosis
extensively studied in liver transplantation and can be made using viscoelastic tests such as
several studies have supported the use in trans- TEG or ROTEM (Fig. 21.1). Spontaneous
plantation for FHF [38, 39]. Within 15 min of recovery from hyperfibrinolysis during post-­
an intravenous dose of rFVIIa, almost complete reperfusion commences after 30–60 min but
correction of prothrombin time may be does not return to normal before 2 h [30]. If
achieved. This effect seems to persist until brisk hemorrhage ensues possibly related to
reperfusion [40] and the judicious use of rFVIIa hyperfibrinolysis, waiting for spontaneous
has demonstrated a reduction in transfusion recovery will allow consumptive and dilu-
requirement in some studies [41]. The main tional coagulopathies to supervene. Therefore
concern in the use of rFVIIa (and indeed any early therapy based on regular assessment of
procoagulant) is an increase in thromboem- the TEG or ROTEM trace is appropriate.
bolic events especially hepatic artery thrombo- • Prior to the withdrawl of the market of apro-
sis (HAT). HAT post-transplant is associated tinin (a serine protease inhibitor which prevents
with a high risk of graft loss and greatly plasminogen splitting to form plasmin) in 2008
increased mortality. Therefore, any actions to after a randomized trial in cardiac surgery
correct coagulopathy must be balanced against (BART trial) [52] aprotinin was commonly
the risk of excessive procoagulation. Several used in liver transplantation as the first line
studies report no increased incidence of throm- anti-fibrinolytic agent, with reductions of trans-
boembolic complications with rFVIIa. fusion requirements when used prophylacti-
However, other authors suggest caution in its cally [53]. Now the two available lysine
use [42]. Meta-analysis and systematic reviews analogues that inhibit conversion of plasmino-
of the use of rFVIIa in hepatic surgery (includ- gen—transexamic acid and epsilon amino-
ing transplantation) failed to show a benefit in caproic acid (EACA, unlicensed in
the number of blood transfusions, yet showed a Europe)—are the agents of choice. An intrave-
significant increase in the incidence of arterial nous dose of either given either prophylacti-
thrombotic events [43–45]. Consequently, the cally prior to reperfusion or, preferably, after
prophylactic use of rFVIIa is therefore not rec- establishing the presence of hyperfibrinolysis
ommended, reserving its use only as rescue using TEG or ROTEM, will provide rapid
therapy for uncontrolled bleeding [46]. reversal of that aspect of coagulopathy.
• Antifibrinolytics
• Post-reperfusion, primary hyperfibrinolysis is Perioperative Fluid Balance
a common cause of coagulation dysfunction Optimal fluid management is particularly diffi-
with an 80% incidence, 40% being severe cult to maintain in the operative phase of trans-
[30]. This is due mainly to an imbalance plant for fulminant failure. The need for exacting
between hepatic metabolism of t-PA and syn- control to prevent ICP surges whilst maintaining
thesis of plasminogen activator inhibitor-1 adequate intravascular filling to maximise car-
(PAI-1). Hyperfibrinolysis has also been diac function in the presence of often significant
related to the quality of the graft [47]. During and rapid blood loss is a major challenge. The use
the anhepatic phase, t-PA breakdown in the of some form of cardiac output monitoring is the
liver ceases [48]. Immediately post-­norm. Many methods are available, each with
reperfusion, a large amount of t-PA is released their own advantages and disadvantages.
into the circulation from the donor liver endo- Although the use of pulmonary artery catheters is
thelium, accumulated during the cold much less common in the ICU there may be some
ischeemic period, and accentuates [49] t-PA benefit of its use in these highly complex cases.
excess and overwhelms activity of PAI-1 [50]. Renal replacement therapy is frequently used
rFVIIa has no effect on hyperfibrinolysis [51] in patients with fulminant liver failure due to the
21  Perioperative Considerations for Transplantation in Acute Liver Failure 263

3 kaolin
a anhepatic Sample: 15/09/2008 13: 11-14:24

Hb 10.7

Plt 49
PT 16

R K Angle MA G EPL LY30 LY60 A CI


min min deg mm d/sc % % % mm
8.6 4.4 38.3 47.4 4.5K 1.3 1.3 *0.4* 44.4 -6.3
4–8 0–4 47–74 54–72 60.K – 13.2K 0 – 15 0–8 0 – 15 -3 – 3

4 kaolin
b post-reperfusion Sample: 15/09/2008 14: 10-15:05

Hb 9.4

Plt 49
PT 14

R K Angle MA G EPL LY30 LY60 A CI


min min deg mm d/sc % % % mm
7.5 3.9 45.1 23.5 1.5K 74.8 74.8 *81.6* 0.2 -7.9
4–8 0–4 47–74 54–72 60.K – 13.2K 0 – 15 0–8 0 – 15 -3 – 3

Fig. 21.1  TEG traces. (a) Anhepatic phase represents a the two lines to a single straight line indicating clot lysis.
relatively normal clotting profile with minor abnormali- The standard laboratory results for a blood sample taken
ties associated with a corrected coagulation state in a simultaneously with the TEG sample are displayed. Note
patient with ALF and (b) after reperfusion in the same the minimal change of laboratory results despite a radical
patient, demonstrates marked hyperfibrinolysis associated change in coagulation status
with reperfusion of the donor liver. The initial formation
of clot with line divergence is followed by rapid return of

high incidence of acute kidney injury. CVVH in the complexity of the transplant procedure with
the ICU in patients with renal impairment has more staff and operating room equipment
become a standard procedure in patients with required. However, in the presence of ALF with
ALF to allow fluid management, acid base main- MOF and established acute renal failure and the
tenance, reducing hyperammonaemia, ICP con- prospect of needing to use marginal donor livers,
trol and enable coagulation control without our institution would regard continuing use of
volume overload. Continuation of CVVH into CVVH in operating room an important compo-
the operating room for the same goals is an nent in the success of present and future trans-
accepted practice. Intraoperative CVVH adds to plantation in the ALF patient. More recent
264 C. P. Snowden et al.

evidence has also supported this viewpoint in prior to transplant to improve function. This may
high MELD score patients undergoing liver enable greater use of these organs in the ALF
transplantation [54] although the reported studies patient group. It is hoped that these techniques
are limited in patient numbers and do not include may ameliorate the adverse effects seen at reper-
patients with fulminant hepatic failure [55, 56]. fusion as well as improve longer-term graft
Of particular importance in addition to maintain- function.
ing fluid balance is the amelioration of acidemia If size matching has not been perfect either
and hyperkalemia that may accompany re-­ “small for size” syndrome or difficulties closing
perfusion. Some centres described the use of the abdomen may be encountered. Indeed, split-
hemofiltration filters in parallel to the VVB cir- ting the donor liver is sometimes necessary to
cuit in a modified CVVH system [57]. Advances obtain a size match. In the unstable ALF trans-
in CVVH technology and improved accuracy in plant recipient with difficult ICP control and
fluid exchange rates have made it more attractive impaired respiratory function, attempting to close
to continue standard CVVH during liver trans- an abdomen over an over-sized liver may create
plant, although optimal intraoperative exchange major physiological difficulties. The resultant
regimes have not been defined [58]. Usually the increase in abdominal pressure may impair dia-
CVVH treatment regime that was started in the phragmatic excursions and potentially reduce
ICU pre-operatively will be continued into the perfusion pressures to the new liver and other
operating room. We commonly used a dialysate intra-abdominal organs (i.e. gut and kidneys).
flow of 35 mL/kg/h using lactate and potassium Furthermore, the increase in intra-thoracic pres-
free solutions as standard. Anticoagulation for sure necessary to maintain ventilation may have
the circuit is usually not needed given the under- an adverse effect on ICP control. It may be pru-
lying deranged coagulopathy in ALF. The ability dent to opt for a delay in total abdominal closure
for fluid removal with CVVH allows the transfu- for 24–48 h with surgical packs or vacuum-type
sion of significant volumes of blood products and dressings in place to minimise this deleterious
better control of ICP pressure spikes, anhepatic impact.
acidosis and pre-reperfusion hyperkalemia [59]. In the UK, major blood group (ABO) incom-
patibility (ie. “A” donor with “O” recipient) liver
transplantation is not considered an appropriate
 arginal Donors and ABO
M use of a limited resource, as survival results are
Incompatibility inferior to group matched transplants, (although
success with one such UK liver transplant has
Rapid deterioration and profound MOF in ALF been reported, in a patient with ALF [60]. Minor
may restrict the choice of donor organ within the ABO incompatibility (i.e. “A” recipient receiving
period where successful transplant for these an “O” liver) is accepted (particularly the A2-to-O
patients can be achieved. As a result, clinicians subtype where results are superior to other mis-
may decide that use of marginal donor organs or matches [61] and may be used for ALF treatment
ABO incompatible organs is necessary for sur- with time restraints and limitation on donor sup-
vival. For the transplant anesthesiologist this may ply. The main concern with this practice is a
provide greater challenges. Marginal donor graft-versus-host reaction caused by passenger
organs may produce more unstable reperfusion lymphocytes released from the donor liver pro-
related physiological effects. Recovery of func- ducing anti-A antibodies resulting in the poten-
tion with its improvement of coagulopathy and tial for recipient red cell haemolysis. If marked
other physiological parameters may also be this is treated by transfusing donor compatible
greatly delayed with a suboptimal donor liver. red cells (to which the A recipient will not pro-
There is a strong focus of current research into duce antibody), B-cell suppression, intravenous
the use of machine perfusion of marginal donor immunoglobulin (IVIG) and plasmapheresis. In
organs (either normothermic or hypothermic) other countries, (e.g. Japan) major ABO incom-
21  Perioperative Considerations for Transplantation in Acute Liver Failure 265

patible transplantation is an accepted method tion, the early initiation of the anhepatic state will
using a live related donor, where ABO compati- improve the patients’ condition both in terms of
ble donors are not available. With enhanced cerebral and cardiovascular stability. Surges in
immunosupression using mycophenolate mofetil cerebral blood flow are often more relevant to
(MMF), aggressive use of B lymphocyte suppres- increases in ICP, whereas CPP is usually main-
sion (i.e. with rituximab) and removal of pre- tained even during periods of systemic hypoten-
formed immunoglobulins with plasmaphoresis, sion due to the reduction in cerebrovascular
immunoadsorption and IgG, it is possible to pro- resistance seen in acute liver failure. Management
vide conditions for successful transplant. In ALF of the complex coagulopathies seen in ALF is a
there may be no other choice and the relative challenge in itself and can be enhanced by the use
increased risk is outweighed by benefit of expedi- of TEG monitoring. Close control of intraopera-
ent transplantation. There has been a series of tive fluid balance and early post-reperfusion
recently published successes in treating ALF hepatic function are important to intraoperative
using major ABO-mismatched donor organs success. The pressure to use marginal or not per-
internationally [62]. In these cases, particular fectly matched donor organs can provide addi-
attention to the use of appropriately matched tional intra-operative challenges. Delayed
blood products is vital. postoperative recovery needs to be anticipated
and managed appropriately.

 ealistic Expectations of Delayed


R
Recovery References
It is important to be realistic about the recovery 1. Hoofnagle JH, Carithers RL, Shapiro C, Ascher
of the patient with established ALF even after N. Fulminant hepatic failure: summary of a work-
seemingly successful liver transplantation. shop. Hepatology. 1995;21:240–52.
2. Bernal W, Wendon J. Liver transplantation in adults
Preoperative severe organ dysfunction, delayed with acute liver failure. J Hepatol. 2004;40(2):192–7.
new liver function, continuing raised intracranial 3. Stravitz RT. Critical management decisions in patients
pressure, pulmonary oedema and persistent renal with acute liver failure. Chest. 2008;134(5):1092–102.
failure are all reasons why recovery is likely 4. Marudanayagam R, Shanmugam V, Gunson B,
Mirza DF, Mayer D, Buckels J, et al. Aetiology
going to be delayed requiring prolonged support- and outcome of acute liver failure. HPB (Oxford).
ive ICU care. 2009;11(5):429–34.
5. Bernal W, Wendon J. Acute liver failure. N Engl J
Med. 2014;370(12):1170–1.
6. Farmer DG, Anselmo DM, Ghobrial RM, Yersiz H,
Summary McDiarmid SV, Cao C, et al. Liver transplantation
for fulminant hepatic failure: experience with more
Patients with ALF make up only 7–10% of liver than 200 patients over a 17-year period. Ann Surg.
transplant recipients but create unique challenges 2003;237(5):666–75. discussion 675–6.
7. Germani G, Theocharidou E, Adam R, Karam
for the anesthesiologist. Advances in supportive V, Wendon J, O’Grady J, et al. Liver transplanta-
ICU care have improved the likelihood of patients tion for acute liver failure in Europe: outcomes
surviving until a donor organ is procured, but over 20 years from the ELTR database. J Hepatol.
where premorbid organ failure and imminent 2012;57(2):288–96.
8. Bernal W, Lee WM, Wendon J, Larsen FS, Williams
demise seems likely, “bridging” therapies includ- R. Acute liver failure: a curable disease by 2024? J
ing total hepatectomy should be considered. If Hepatol. 2015;62(1 Suppl):S112–20.
emergency liver transplantation is performed, 9. O’Grady J. Timing and benefit of liver transplantation
cerebrovascular stability remains a priority. The in acute liver failure. J Hepatol. 2014;60(3):663–70.
10.
Kjaergard LL, Liu J, Als-Nielsen B, Gluud
use of moderate hypothermia seems to abolish C. Artificial and bioartificial support systems for
ICP variability and may constitute an important acute and acute-on-chronic liver failure: a systematic
therapeutic strategy in high-risk patients. In addi- review. JAMA. 2003;289(2):217–22.
266 C. P. Snowden et al.

11. Husberg BS, Goldstein RM, Klintmalm GB, Gonwa 25. Dmello D, Cruz-Flores S, Matuschak GM. Moderate
T, Ramsay M, Cofer J, et al. A totally failing liver hypothermia with intracranial pressure monitoring as
may be more harmful than no liver at all: three cases a therapeutic paradigm for the management of acute
of total hepatic devascularization in preparation for liver failure: a systematic review. Intensive Care Med.
emergency liver transplantation. Transplant Proc. 2010;36(2):210–3.
1991;23(1 Pt 2):1533–5. 26. Detry O, De Roover A, Honore P, Meurisse M. Brain
12. Ringe B, Pichlmayr R. Total hepatectomy and liver edema and intracranial hypertension in fulminant
transplantation: a life-saving procedure in patients with hepatic failure: pathophysiology and management.
severe hepatic trauma. Br J Surg. 1995;82(6):837–9. World J Gastroenterol. 2006;12(46):7405–12.
13. Ejlersen E, Larsen FS, Pott F, Gyrtrup HJ, Kirkegaard 27. Blei AT. Monitoring cerebral blood flow: a useful
P, Secher NH. Hepatectomy corrects cerebral hyper- clinical tool in acute liver failure? Liver Transpl.
perfusion in fulminant hepatic failure. Transplant 2005;11(11):1320–2.
Proc. 1994;26(3):1794–5. 28. Noun R, Zante E, Sauvanet A, Durand F, Bernuau
14. Rozga J, Podesta L, LePage E, Hoffman A, Morsiani J, Belghiti J. Liver devascularisation improves the
E, Sher L, et al. Control of cerebral oedema by total hyperkinetic syndrome in patients with fulminant
hepatectomy and extracorporeal liver support in fulmi- and subfulminant hepatic failure. Transplant Proc.
nant hepatic failure. Lancet. 1993;342(8876):898–9. 1995;27(1):1256–7.
15. Bustamante M, Castroagudín JF, Gonzalez-Quintela 29. Filho JAR, Machado MAC, Nani RS, Rocha JPS,
A, Martinez J, Segade FR, Fernández A, et al. Intensive Figueira ERR, Bacchella T, et al. Hypertonic saline
care during prolonged anhepatic state after total hepa- solution increases cerebral perfusion pressure during
tectomy and porto-caval shunt (two-stage procedure) clinical orthotopic liver transplantation for fulmi-
in surgical complications of liver transplantation. nant hepatic failure: preliminary results. Clinics (Sao
Hepatogastroenterology. 2000;47(35):1343–6. Paulo). 2006;61(3):231–8.
16. Domínguez Fernández E, Lange K, Lange R, Eigler 30. Kang YG, Martin DJ, Marquez J, Lewis JH,

FW. Relevance of two-stage total hepatectomy and Bontempo FA, Shaw BW, et al. Intraoperative
liver transplantation in acute liver failure and severe changes in blood coagulation and thrombelasto-
liver trauma. Transpl Int. 2001;14(3):184–90. graphic monitoring in liver transplantation. Anesth
17. Cheema SP, Hughes A, Webster NR, Bellamy
Analg. 1985;64(9):888–96.
MC. Cardiac function during orthotopic liver trans- 31. Massicotte L, Lenis S, Thibeault L, Sassine M-P, Seal
plantation with venovenous bypass. Anaesthesia. RF, Roy A. Effect of low central venous pressure and
1995;50(9):776–8. phlebotomy on blood product transfusion require-
18. Jalan R. Pathophysiological basis of therapy of raised ments during liver transplantations. Liver Transpl.
intracranial pressure in acute liver failure. Neurochem 2005;12(1):117–23.
Int. 2005;47(1–2):78–83. 32. Bolliger D, Szlam F, Molinaro RJ, Rahe-Meyer N,
19. Ardizzone G, Arrigo A, Panaro F, Ornis S, Colombi Levy JH, Tanaka KA. Finding the optimal concen-
R, Distefano S, et al. Cerebral hemodynamic and tration range for fibrinogen replacement after severe
metabolic changes in patients with fulminant haemodilution: an in vitro model. Br J Anaesth.
hepatic failure during liver transplantation. TPS. 2009;102(6):793–9.
2004;36(10):3060–4. 33. Wikkelsø A, Lunde J, Johansen M, Stensballe J,

20. Lidofsky SD. Liver transplantation for fulminant
Wetterslev J, Møller AM, et al. Fibrinogen concen-
hepatic failure. Gastroenterol Clin North Am. trate in bleeding patients. Cochrane Database Syst
1993;22(2):257–69. Rev. 2013;8:CD008864.
21. Detry O, Arkadopoulos N, Ting P, Kahaku E,
34. Sabate A, Gutierrez R, Beltran J, Mellado P, Blasi
Margulies J, Arnaout W, et al. Intracranial pressure A, Acosta F, et al. Impact of pre-emptive fibrino-
during liver transplantation for fulminant hepatic fail- gen concentrate on transfusion requirements in liver
ure. Transplantation. 1999;67(5):767–70. transplantation: a multicenter, randomized, double-­
22. Jalan R, Olde Damink SWM, Deutz NEP, Davies NA, blind, placebo-controlled trial. Am J Transplant.
Garden OJ, Madhavan KK, et al. Moderate hypo- 2016;16:2421–9.
thermia prevents cerebral hyperemia and increase 35. Clevenger B, Mallett SV. Transfusion and coagula-
in intracranial pressure in patients undergoing liver tion management in liver transplantation. World J
transplantation for acute liver failure. Transplantation. Gastroenterol. 2014;20(20):6146–58.
2003;75(12):2034–9. 36. Franchini M, Lippi G. Prothrombin complex concen-
23. Jalan R, Pollok A, Shah SHA, Madhavan K, Simpson trates: an update. Blood Transfus. 2010;8(3):149–54.
KJ. Liver derived pro-inflammatory cytokines may be 37. Arshad F, Ickx B, van Beem RT, Polak W, Grüne F,
important in producing intracranial hypertension in Nevens F, et al. Prothrombin complex concentrate
acute liver failure. J Hepatol. 2002;37(4):536–8. in the reduction of blood loss during orthotopic
24. Lidofsky SD, Bass NM, Prager MC, Washington DE, liver transplantation: PROTON-trial. BMC Surg.
Read AE, Wright TL, et al. Intracranial pressure mon- 2013;13:22.
itoring and liver transplantation for fulminant hepatic 38. Shami VM, Caldwell SH, Hespenheide EE, Arseneau
failure. Hepatology. 1992;16(1):1–7. KO, Bickston SJ, Macik BG. Recombinant activated
21  Perioperative Considerations for Transplantation in Acute Liver Failure 267

factor VII for coagulopathy in fulminant hepatic 50. Porte RJ, Knot EA, Bontempo FA. Hemostasis

failure compared with conventional therapy. Liver in liver transplantation. Gastroenterology.
Transpl. 2003;9(2):138–43. 1989;97(2):488–501.
39. Lodge JPA, Jonas S, Jones RM, Olausson M, Mir-­ 51. Lisman T, Leebeek FWG, Meijer K, Van Der Meer J,
Pallardo J, Soefelt S, et al. Efficacy and safety of Nieuwenhuis HK, De Groot PG. Recombinant factor
repeated perioperative doses of recombinant fac- VIIa improves clot formation but not fibrolytic poten-
tor VIIa in liver transplantation. Liver Transpl. tial in patients with cirrhosis and during liver trans-
2005;11(8):973–9. plantation. Hepatology. 2002;35(3):616–21.
40. Surudo T, Wojcicki M, Milkiewicz P, Czuprynska
52. Fergusson DA, Hébert PC, Mazer CD, Fremes S,

M, Lubikowski J, Jarosz K, et al. Rapid correction of MacAdams C, Murkin JM, et al. A comparison of
prothrombin time after low-dose recombinant factor aprotinin and lysine analogues in high-risk cardiac
VIIA in patients undergoing orthotopic liver trans- surgery. N Engl J Med. 2008;358(22):2319–31.
plantation. Transplant Proc. 2003;35(6):2323–5. 53. Porte RJ, Molenaar IQ, Begliomini B, Groenland

41. Hendriks HG, Meijer K, de Wolf JT, Klompmaker TH, Januszkiewicz A, Lindgren L, et al. Aprotinin
IJ, Porte RJ, de Kam PJ, et al. Reduced transfusion and transfusion requirements in orthotopic liver
requirements by recombinant factor VIIa in orthotopic transplantation: a multicentre randomised double-­
liver transplantation: a pilot study. Transplantation. blind study. EMSALT Study Group. The Lancet.
2001;71(3):402–5. 2000;355(9212):1303–9.
42. Pavese P, Bonadona A, Beaubien J, Labrecque P,
54. LaMattina JC, Kelly PJ, Hanish SI, Ottmann SE,

Pernod G, Letoublon C, et al. FVIIa corrects the Powell JM, Hutson WR, et al. Intraoperative con-
coagulopathy of fulminant hepatic failure but may tinuous veno-venous hemofiltration facilitates surgery
be associated with thrombosis: a report of four cases. in liver transplant patients with acute renal failure.
Can J Anaesth. 2005;52(1):26–9. Transplant Proc. 2015;47(6):1901–4.
43. Chavez-Tapia NC, Alfaro-Lara R, Tellez-Avila F,
55. Douthitt L, Bezinover D, Uemura T, Kadry Z, Shah
Barrientos-Gutiérrez T, González-Chon O, Mendez-­ RA, Ghahramani N, et al. Perioperative use of con-
Sanchez N, et al. Prophylactic activated recombinant tinuous renal replacement therapy for orthotopic liver
factor VII in liver resection and liver transplantation: transplantation. Transplant Proc. 2012;44(5):1314–7.
systematic review and meta-analysis. PLoS One. 56. Sedra AH, Strum E. The role of intraoperative hemo-
2011;6(7):e22581. dialysis in liver transplant patients. Curr Opin Organ
44. Yank V, Tuohy CV, Logan AC, Bravata DM,
Transplant. 2011;16(3):323–5.
Staudenmayer K, Eisenhut R, et al. Systematic review: 57. Tobias MD, Jobes CS, Aukburg SJ. Hemofiltration
benefits and harms of in-hospital use of recombinant in parallel to the venovenous bypass circuit for oli-
factor VIIa for off-label indications. Ann Intern Med. guric hypervolemia during liver transplantation.
2011;154(8):529–40. Anesthesiology. 1999;90(3):909–11.
45. Levi M, Levy JH, Andersen HF, Truloff D. Safety of 58. Blackwell MM, Chavin KD, Sistino JJ. Perioperative
recombinant activated factor VII in randomized clini- perfusion strategies for optimal fluid management in
cal trials. N Engl J Med. 2010;363(19):1791–800. liver transplant recipients with renal insufficiency.
46. Kozek-Langenecker SA, Afshari A, Albaladejo P,
Perfusion. 2003;18(1):55–60.
Santullano CAA, De Robertis E, Filipescu DC, et al. 59. Davenport A. Continuous renal replacement thera-
Management of severe perioperative bleeding: guide- pies in patients with liver disease. Semin Dial.
lines from the European Society of Anaesthesiology. 2009;22(2):169–72.
Eur J Anaesthesiol. 2013;30(6):270–382. 60. Srinivas Reddy M, Wilson C, Torpey N, Manas

47. Blasi A, Hessheimer AJ, Beltrán J, Pereira A,
DM. ABO incompatible liver transplantation: a case
Fernández J, Balust J, et al. Liver transplant from of immediate need. Transpl Int. 2007;20(10):904–5.
unexpected donation after circulatory determination 61. Kluger MD, Guarrera JV, Olsen SK, Brown RS,

of death donors: a challenge in perioperative manage- Emond JC, Cherqui D. Safety of blood group A2-to-O
ment. Am J Transplant. 2016;16(6):1901–8. liver transplantation: an analysis of the United
48.
Dzik WH, Arkin CF, Jenkins RL, Stump Network of Organ Sharing database. Transplantation.
DC. Fibrinolysis during liver transplantation in 2012;94(5):526–31.
humans: role of tissue-type plasminogen activator. 62. Shen T, Lin B-Y, Jia J-J, Wang Z-Y, Wang L, Ling Q,
Blood. 1988;71(4):1090–5. et al. A modified protocol with rituximab and intrave-
49. Porte RJ, Bontempo FA, Knot EA, Lewis JH, Kang nous immunoglobulin in emergent ABO-incompatible
YG, Starzl TE. Tissue-type-plasminogen-activator- liver transplantation for acute liver failure.
associated fibrinolysis in orthotopic liver transplanta- Hepatobiliary Pancreat Dis Int. 2014;13(4):395–401.
tion. TPS. 1989;21(3):3542.
The Patient with Severe
Co-morbidities: Renal Failure 22
Andrew Disque and Joseph Meltzer

Keywords
Liver Disease (MELD) scoring system was
Renal insufficiency · Chronic kidney disease ·
implemented for prioritizing patients on the liver
Acute kidney injury · Hepatorenal syndrome ·
transplant waiting list due to its ability to predict
Renal replacement therapy · Dialysis
survival for patients with end-stage liver disease
[4]. It replaced the Child-Pugh scoring system.
Both UNOS and Eurotransplant now use the
Introduction MELD score for allocating organs to patients
awaiting liver transplantation. Serum creatinine,
Renal injury and failure is a frequent and poten- a marker of renal function, is one of only three
tially devastating complication of liver cirrhosis variables used in the MELD score, highlighting
and patients undergoing liver transplantation [1]. the importance of renal function for survival in
When renal injury progresses to failure the prog- the face of liver disease. A patient with renal fail-
nosis for patients with concomitant cirrhosis is ure requiring dialysis or with a serum creatinine
poor [2]. Preoperative renal dysfunction is also over 4 mg/dL already has a MELD score of 20
associated with significantly worsened outcomes even with normal liver function. As a result the
in patients who undergo liver transplantation [3]. use of the MELD scoring system has given pref-
In 2002, the United Network for Organ Sharing erence to patients with impaired renal function
(UNOS) modification to the Model for End Stage and increased the number of those patients who
undergo liver transplantation [5]. Up to 25% of
liver transplant candidates have pre-­ operative
A. Disque, MD AKI; more than 10% of patients who undergo
Department of Anesthesiology and Perioperative
Medicine, David Geffen School of Medicine, liver transplantation have a serum creatinine of
University of California at Los Angeles, greater than 2 mg/dL and more than 5% undergo
Los Angeles, CA, USA transplantation while receiving renal replacement
J. Meltzer, MD (*) therapy (RRT) [3]. Preoperative renal function is
Division of Critical Care, Department of one of the most important predictors of post-
Anesthesiology and Perioperative Medicine, David transplant survival. These facts reinforce the
Geffen School of Medicine, University of California
at Los Angeles, Los Angeles, CA, USA importance of renal function and dysfunction in
e-mail: jmeltzer@mednet.ucla.edu patients with advanced liver disease.

© Springer International Publishing AG, part of Springer Nature 2018 269


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_22
270 A. Disque and J. Meltzer

Defining Renal Failure teria [9]. The five categories of RIFLE criteria
represent three grades of increasing severity of
Arriving at a standardized definition of renal AKI (Risk, Injury, and Failure) and two out-
failure has been surprisingly difficult. Renal fail- come classes (Loss, and End-Stage Kidney
ure is commonly divided into either acute renal Disease). Absolute increase of serum creatinine,
failure (now termed acute kidney injury, or AKI) percentage increase in creatinine, percentage
or chronic kidney disease (CKD) (previously reduction in glomerular filtration rate (GFR) and
termed chronic renal insufficiency or chronic decrement in urine output over time define the
renal failure) [6]. More than 35 definitions had categories. Figure 22.1 summarizes the RIFLE
existed for renal failure [7]. The absence of a criteria. Rather than reductively equating renal
consensus definition has had a negative impact function and serum creatinine, the RIFLE crite-
on basic science as well as clinical research in ria attempted to standardize the definition and
the field of acute kidney injury. In the last decade severity of renal injury and facilitate evaluation,
there have been attempts to unify the definition treatment and communication amongst health-
for classifying and diagnosing AKI. The diagno- care providers. A recent study demonstrated the
sis of AKI requires both a patient’s clinical his- utility of the RIFLE criteria as a predictor of
tory and relevant laboratory data. The Acute mortality in patients with cirrhosis admitted to
Kidney Injury Network (AKIN) introduced spe- the intensive care unit (ICU) [10].
cific criteria for the diagnosis of AKI including a
rapid time course (less than 48 h) and a decre-
ment of kidney function [8]. A reduction of kid-  cute Kidney Injury in Cirrhosis
A
ney function was defined as either an absolute and Hepatorenal Syndrome
increase in serum creatinine of >0.3 mg/dL, a
percentage increase in serum creatinine of AKI is a rapid loss of kidney function and is
>50%, or a reduction in urine output to a level of commonly categorized into three broad catego-
<0.5 mL/kg/h for more than 6 h. Prior to the ries: pre-renal, intrinsic-renal, and post-renal
introduction of AKIN criteria, the Acute Dialysis kidney injury. Acutely, renal function can dete-
Quality Initiative (ADQI) uniformly defined and riorate over a period of hours to days, most
staged acute kidney injury using the RIFLE cri- often as a result of multiple insults. Pre-renal

GFR Criteria* Urine Output Criteria

Increased SCreat x1.5 or UO< .5ml/kg/h


Risk GFR decreses > 25% x 6 hr
High
Sensitivity
Increased SCreat x2 UO< .5ml/kg/h
or GFR decrese > 50% x 12 hr
Injury

Increased SCreat x3 UO< .3ml/kg/h


ria

or GFR decrese 75% x 12 hr or


Oligu

Failure OR SCreat ≥4mg/dl Anuria x 12 hrs


Acute rise ≥0.5mg/dl

High
Fig. 22.1 RIFLE
Sensitivity
criteria (risk, injury, Persistent ARF**=complete loss
failure, loss of function, Loss
of kidney function >4 weeks
endstage kidney disease
(with permission: End Stage Kidney Disease
Bellomo et al. Critical ESKD (>3 months)
Care 2004 8:R204)
22  The Patient with Severe Co-morbidities: Renal Failure 271

causes of kidney injury include any mechanism portal hypertension and advanced cirrhosis
that decreases the effective blood flow to the [13–15]. Vasodilatation thus triggers the activa-
kidney. Common causes of pre-renal kidney tion of the renin-angiotensin system and along
injury include dehydration, hypovolemia, hem- with sympathetic stimulation, results in intense
orrhage, hypotension, and heart failure. Pre- renal vasoconstriction. In compensated cirrho-
renal injury is often rapidly reversible when the sis, cardiac output and plasma volume both
underlying mechanism is corrected, thus glo- increase to restore effective arterial volume and
merular or tubular injury can be avoided. thereby renal perfusion and function is pre-
However, prolonged pre-renal azotemia may served. However, in decompensated cirrhosis,
progress to intrinsic acute kidney injury. cardiac output and heart rate are maximized
Intrinsic causes of kidney injury can result from and cannot increase further to augment blood
direct injury to the glomeruli, tubules, or inter- pressure, resulting in a further increase in circu-
stitium of the kidney. Common causes of intrin- lating vasoconstrictors and renal vasoconstric-
sic kidney injury include glomerulonephritis tion, sodium and water retention and ascites
(GN), acute interstitial nephritis (AIN), and formation [1]. This results in decreased renal
acute tubular necrosis (ATN) [6]. Infection and perfusion pressure and reduced glomerular fil-
sepsis are common causes of acute kidney tration rate (GFR). Previously the International
injury in patients with cirrhosis who present for Club of Ascites (ICA) defined two types of
liver transplantation. Cirrhotic patients are at hepatorenal syndrome exist: Type 1 HRS is
high risk for sepsis from a multitude of causes characterized by a rapid decline in renal func-
including, but not limited to, spontaneous bac- tion, while Type 2 HRS entails a more chronic
terial peritonitis, pneumonia, or central-line deterioration in renal function that is associated
associated blood stream infection [11]. with ascites formation. Differentiating HRS
Additionally, these patients are often chroni- from ATN can be difficult because diagnosing
cally ill and at risk for toxin-mediated ATN the former involves excluding other causes of
from aminoglycoside antibiotics, intravenous AKI and there is no single test that confirms
contrast agents or non-steroidal anti-inflamma- HRS [16]. Although mortality is very high
tory medications. Immunosuppressants and among patients with cirrhosis and renal failure,
chemotherapeutic agents are also implicated if patients with Type 1 HRS have the worst prog-
administered. Post-renal kidney injury involves nosis—without liver transplant, patients have a
obstruction at any point along the urinary out- 50% survival rate at 1 month and a 20% sur-
flow tract by, for example, malignancies, stones, vival rate at 6 months [17]. Therapeutic options
or a hypertrophied prostate. Treatment of any are limited for patients with HRS. While albu-
type of kidney injury is centered on treating the min combined with vasopressin (or its ana-
underlying etiology while providing supportive logues) is of some benefit, optimal medical
care and avoiding nephrotoxic substances and management should include the evaluation for
further renal insult. Patients with liver cirrhosis liver transplantation [18].
are at risk for all three types of acute kidney In 2015 the ICA revised their definition of
injury, but they can also develop a unique entity HRS (now called HRS-AKI) [19]. The new defi-
known as hepatorenal syndrome (HRS) [12]. nition of HRS-AKI is similar to definitions of
HRS is a form of renal injury caused by circula- AKI in other clinical scenarios and includes a
tory dysfunction secondary to an imbalance of recent increase of serum creatinine (>0.3 mg/dL
circulating vasodilatory and vasoconstrictive within 48 h or >50% within 7 days). HRS-AKI
substances. This dysfunction is the result of a has now three stages depending on the severity of
decrease in systemic vascular resistance result- the increase of serum creatinine. The new defini-
ing primarily from splanchnic vasodilatation tion removed urine output criteria (that previ-
due to nitric oxide, prostaglandins, and other ously have seldom been used clinically or in
vasoactive substances released in patients with research) and some of the criteria that were
272 A. Disque and J. Meltzer

s­ pecific for AKI in liver failure. The new defini-  ssessment and Management
A
tion reflects recent studies that HRS is clinically of Acute Kidney Injury
very difficult to discern from conventional in Cirrhosis—Preoperative
AKI. The old and new definitions of HRS are Approach
described in Tables 22.1 and 22.2.
Managing AKI in patients with cirrhosis depends
Table 22.1  Major diagnostic criteria of hepato-renal not only on the cause, but also the severity of
syndrome (HRS) -1996 defintion injury. The most practical way to assess renal
Major diagnostic criteria of HRS [14] function is by measurement of factors included
Hepatic failure and ascites in the RIFLE criteria: serum creatinine, GFR and
Creatinine >1.5 mg/dL urine output. Although commonly used and
No shock, ongoing bacterial infection, nephrotoxic widely accepted, serum creatinine is unfortu-
agents or fluid losses nately insensitive and not linearly related to GFR
No improvement after diuretic withdrawal and fluid [20]. Moreover, in patients with advanced liver
resuscitation
disease, serum creatinine is often an unreliable
Proteinuria <500 mg/day, normal renal sonography
indicator of renal function due to a decreased
HRS Type I Type II
amount of creatinine production with reduced
Serum >2 × baseline or >1.5 mg/dL
creatinine >2.5 mg/dL (133 μmol/L) muscle mass [21]. Therefore, a normal or low
(221 μmol/L) serum creatinine is likely to overestimate
Creatinine <20 mL/min <40 mL/min GFR. Urine output may not be a reliable marker
clearance of renal function or injury as many patients
Onset <2 weeks >2 weeks receive chronic diuretic therapy. Recently, there
Medians 1 month 6 months has been a promising search for biomarkers of
survival
renal function and injury that can potentially

Table 22.2  New (2015) definition of hepatorenal syndrome-acute kidney injury [19]
Subject Definition
Baseline sCr A value of sCr obtained in the previous 3 months, when available, can be used as baseline sCr. In
patients with more than one value within the previous 3 months, the value closest to the admission
time to the hospital should be used.In patients without a previous sCr value, the sCr on admission
should be used as baseline.
Definition of • Increase in sCr ≥0.3 mg/dL (≥26.5 μmol/L) within 48 h; or,
AKI • A percentage increase sCr ≥50% from baseline which is known, or presumed, to have occurred
within the prior 7 days
Staging of • Stage 1: increase in sCr ≥0.3 mg/dL (26.5 μmol/L) or an increase in sCr ≥1.5- to 2-fold from
AKI baseline
• Stage 2: increase in sCr >2- to 3-fold from baseline
• Stage 3: increase of sCr >3-fold from baseline or sCr ≥4.0 mg/dL (353.6 μmol/L) with an acute
increase ≥0.3 mg/dL (26.5 μmol/L) or initiation of renal replacement therapy
Progression Progression Regression
of AKI Progression of AKI to a higher stage and/or Regression of AKI to a lower stage for RRT
need for RRT
Response to No response Partial response Full response
treatment No regression of AKI Regression of AKI stage Return of sCr to a
with a reduction of sCr to value within 0.3 mg/
≥0.3 mg/dL dL (26.5 μmol/L) of
(26.5 μmol/L) above the the baseline value
baseline value
With permission: Gut. 2015 Apr;64(4):531–7:Diagnosis and management of acute kidney injury in patients with cir-
rhosis: revised consensus recommendations of the International Club of Ascites
22  The Patient with Severe Co-morbidities: Renal Failure 273

detect AKI much earlier than changes in creati- Discontinuation of diuretics and optimization of
nine. Serum cystatin C, a protein produced by all fluid status and renal blood flow with the admin-
nucleated cells at a constant rate independent of istration of isotonic crystalloid or colloid solu-
age, sex, race, or muscle mass is a more accurate tions may be necessary to prevent progression of
marker of GFR than creatinine [22]. The pres- the injury. There is little convincing evidence
ence of cystatin C in the urine can indicate injury favoring colloids or crystalloids, however 6%
of the proximal convoluted tubule, where the hydroxyethyl starch (and probably all types of
injured cells no longer uptake the biomarker starches) should be avoided in the setting of AKI
[53]. Further studies are needed to test its clini- due to the increased risk of AKI in clinical stud-
cal utility. Neutrophil gelatinase-associated lipo- ies [26, 27]. In more acute situations of hypovo-
calin (NGAL) is a protein that is produced by lemia, for example due to gastrointestinal
renal tubular cells in response to renal injury bleeding, rapid administration of plasma expand-
[23]. It can be detected easily in the urine within ers and/or blood products may be needed to
minutes of injury and is highly sensitive and spe- reverse hemodynamic instability. Sepsis should
cific to acute kidney injury—levels are much always be considered as a cause of renal injury in
less increased in chronic kidney disease. It may cirrhotic patients [28]. Early and aggressive treat-
further allow a differentiation between conven- ment should be initiated if sepsis is suspected,
tionally defined HRS and AKI [24]. Other bio- including source control, appropriate antibiotics,
markers that may be useful in the future include intravenous fluid administration [29], lung pro-
N-acetyl-b-d-glucosaminidase (a urine marker tective ventilation in the setting of acute respira-
indicating proximal convoluted tubule lysosomal tory distress syndrome (ARDS) [30], the
damage), kidney injury molecule 1, microalbu- avoidance of severe hyperglycemia [31], early
min, interleukin 18, and fatty liver acid binding enteral nutritional, and potentially steroid ther-
protein [25]. apy for adrenal insufficiency or refractory vaso-
Patients with advanced liver disease and those plegia [32]. Bacterial infections should be treated
presenting for liver transplantation may have kid- rapidly and appropriately [33]—initial empiric
ney injury with a wide variety of causes and therapy is often dictated by local and hospital
severity. Unfortunately, despite countless studies, antibiograms. “Renal-dose” dopamine remains in
there is no proven preventive measure or treat- use as it often increases urine output and may
ment for AKI [20]. Therefore, the management increase cardiac output and therefore renal perfu-
of AKI centers on identifying and treating the sion in patients with low cardiac output and/or
underlying etiology, providing renal support bradycardia. However, multiple large random-
including maintaining renal blood flow and oxy- ized controlled trials demonstrated that there is
gen delivery and avoiding nephrotoxic agents. no role for dopamine in prophylaxis or treatment
Most commonly, pre-renal causes of AKI in cir- of AKI [34–36]. Loop diuretics can be used in the
rhotic patients include hypovolemia secondary to setting of AKI as long as euvolemia is restored
bleeding, fluid losses, reduced oral intake or prior to their administration to avoid further renal
diuretic administration. Gastrointestinal bleed- hypoperfusion and exacerbation of AKI. Loop
ing, including esophageal variceal bleeding, can diuretics have multiple effects on the injured kid-
occur as a consequence of portal hypertension. ney. They may relieve obstructed tubules by
Excessive fluid losses from the gastrointestinal clearing necrotic cell debris. They increase pros-
tract (for example due to diarrhea of an infectious taglandin synthesis which, in turn, can increase
etiology or from excessive lactulose administra- renal blood flow while decreasing active tubular
tion) or renal fluid loss secondary to excessive sodium reabsorption, thus decreasing metabolic
diuresis can cause pre-renal injury [1]. Treatment demand [20]. However, most large studies have
of pre-renal injury can be simple, but requires shown no direct effect of loop diuretics on pre-
quick recognition of the cause and appropriate vention or treatment of AKI [37]. Many vasoac-
treatment to avoid a more permanent renal injury. tive drugs have been studied as possible
274 A. Disque and J. Meltzer

prevention or treatment of renal injury. Studies of abnormalities, and/or volume overload. Once
renal vasodilators such as dopamine, prostaglan- renal function has reached this level of severity,
dins, and fenoldopam have been either too small the patient should be treated with renal replace-
or discouraging [38]. Vasopressors can be effec- ment therapy (RRT). Although there are several
tive in AKI, primarily in the setting of HRS Type renal replacement modalities, three major types
1 [1], by reversing splanchnic vasodilatation and exist—intermittent hemodialysis (iHD), perito-
restoring central blood volume and renal perfu- neal dialysis (PD), and continuous renal replace-
sion. Several different vasoconstrictors such as ment therapies (CRRT). iHD, the standard
terlipressin (a vasopressin analogue), octreotide, treatment for severe acute renal failure for more
norepinephrine, and midodrine have been stud- than four decades, is most often used in patients
ied. Results from recent randomized control trials without acute hemodynamic abnormalities.
were especially promising for the use of vaso- Peritoneal dialysis is an alternative for iHD that
pressin analogues, with possibly added benefit allows a more independent life style but is contra-
with co-administration of intravenous albumin indicated in patients with ascites. During the
[18, 39]. Patients with cirrhosis are deficient of perioperative period different modalities of
endogenous vasopressin and administration of CRRT are commonly used to maintain hemody-
vasopressin can restore blood pressure and main- namic stability during dialysis. There is little data
tain renal perfusion pressure [40]. Overall, vaso- validating one method over another and there is
pressin analogues can be effective in 40–50% of debate over the timing and dosage of RRT in the
patients with HRS but in these studies there was perioperative period [6]. The CRRT modality
no 3- and 6-month mortality benefit [1]. used is often determined by institutional experi-
The International Club of Ascites recom- ence and can be quite variable. Continuous veno-
mends a step-wise approach to diagnosis and venous hemodialysis (CVVHD) is probably the
treatment of HRS-AKI as part of their new defi- most commonly used and safest modality for
nition of AKI (Fig. 22.2). Despite maximum patients in the perioperative period with a tenu-
pharmacologic therapy, AKI and/or HRS-AKI ous hemodynamic status. Regardless of the
can cause renal function to decline to a point of method of RRT, complications such as bleeding,
metabolic disarray, acidosis, severe electrolyte infection, and hypotension should be recognized.

Stage 1 AK1# Stage 2 and 3 AKI#

Close monitoring Withdrawal of diuretics


Remove risk factors (withdrawal of (if not withdrawn
nephrotoxic drugs, vasodilators already) and volume
Fig. 22.2 Hepatorenal and NSAIDs, decrease/withdrawal expansion with albumin
syndrome-acute kidney of diuretics, treatment of infections* (1 g/kg) for 2 days
injury (HRS-AKI) when diagnosed), plasma volume
treatment algorithm expansion in case of hypovolemia
proposed by the Response
International Club of
Ascites. With YES NO
permission: Gut. 2015 Resolution Stable Progression
Apr;64(4):531–
Meets criteria of HRS
7:Diagnosis and
management of acute Close follow up
kidney injury in patients NO YES
with cirrhosis: revised
consensus
Futher treatment of AKI
recommendations of the Specific treatment for Vasocontrictors
decided on a
International Club of case-by-case basis§ other AKI phenotypes and albumin
Ascites
22  The Patient with Severe Co-morbidities: Renal Failure 275

In addition to RRT, there are other nonpharmaco- tion. As with any surgery, maintaining a normal
logic therapies used in patients with combined blood pressure and euvolemia to ensure adequate
kidney and liver dysfunction. Placement of a perfusion and oxygen delivery to all tissues is
transjugular intrahepatic portosystemic shunt paramount. Volatile anesthetics as maintenance
(TIPS) can improve renal perfusion and GFR of anesthesia can decrease GFR primarily as a
[41, 42]. Unproven and experimental artificial result of decreased systemic vascular resistance
liver support systems currently under clinical [20]. This may be exacerbated by hypovolemia
investigation include the Molecular Adsorbent and antidiuretic hormone (ADH) secretion as a
Recirculating System (MARS), single pass albu- response to surgical stress [45]. There is no evi-
min dialysis (SPAD), and the Prometheus System dence that sevoflurane causes clinically relevant
and their effect on renal function remains the be renal injury despite the theoretical possibility that
seen [43]. release of fluoride induces renal injury [46].
Intraoperative positive pressure ventilation
reduces cardiac output, renal blood flow, and thus
Liver Transplantation: GFR through activation of the sympatho-adrenal
Intraoperative Management system. Although not specific to liver transplan-
of Renal Function tation, anesthesiologists should be aware of any
medications that may accumulate or have adverse
Liver transplant remains the preferred treatment effects in patients who have renal dysfunction—
of advanced cirrhosis. Patients with combined barbiturates, benzodiazepines, succinylcholine,
renal and liver failure should be considered for morphine, meperidine, and most nondepolarizing
combined liver kidney transplant (CLKT) [44]. It neuromuscular blockers should be used with
is not clear which patients benefit from CLKT, caution.
but consideration of the type of renal failure, par- Liver transplantation is a lengthy procedure
ticularly the presence of HRS, along with the and is associated with hemodynamic instability,
severity and duration should be made. Without bleeding, coagulopathy, transfusion and meta-
CLKT, renal function often improves after liver bolic disarray—all of which can cause and exac-
transplantation if renal failure has not been pro- erbate kidney injury. The role of the
longed [1]. However, those patients with severe, anesthesiologist, among other things, is to main-
longstanding renal failure requiring RRT do not tain adequate intravascular volume and hemody-
typically improve after liver transplant to accept- namic stability, ensure renal perfusion, and
able levels of renal function. Given organ scarcity minimize further renal injury. Vascular occlu-
and the need for rationing, CLKT possibilities sion of the portal triad and interruption of the
are limited. Newer approaches have used com- inferior vena cava is often part of the surgical
bined liver and dual-kidney transplants, which is procedure and, in the absence of veno-venous
the transplantation of two marginal kidneys that bypass, results in a significant decrease in car-
would otherwise be discarded. The outcomes diac preload and cardiac output and, therefore,
using this approach remain to be seen [54]. More renal perfusion [47]. Fluid management strate-
details about CKLT are discussed elsewhere in gies such as “low-­CVP” techniques and conser-
this book. vative fluid management to prevent liver
As stated before, patients present for liver congestion, bleeding, and transfusion require-
transplant with varying types and severities of ments may have harmful effects on renal perfu-
kidney dysfunction and may only have mild and sion and predispose patients to perioperative
short-lived elevations of creatinine or they may kidney injury [48, 49]. Significant alterations in
present with severe AKI requiring CRRT. The the acid–base balance occur intraoperatively and
intraoperative management of these patients is two of the most critical phases of liver transplan-
complex, and conventional anesthetic goals can tation, the anhepatic and neohepatic phases are
often have detrimental effects on kidney func- associated with significant and serious lactic aci-
276 A. Disque and J. Meltzer

dosis, often demonstrated by a base deficit of Vasopressors are often required during liver
less than −10 to −12 mmol/L [50]. Correcting transplantation to treat hypotension and vasodila-
acidemia and base deficit may help prevent the tion; norepinephrine and arginine vasopressin are
many serious manifestations of re-­perfusion of the two most commonly used agents. Glomerular
the donor liver, such as severe acidosis, hypoten- filtration is determined by the net difference in
sion, hyperkalemia, myocardial depression, arterial pressure between the afferent and efferent
arrhythmias, and cardiovascular collapse. arterioles across the glomerular capillary bed
Sodium bicarbonate can be administered during known as the transcapillary filtration pressure.
the anhepatic phase to prevent a further deterio- Norepinephrine can constrict the glomerular
ration of a severe metabolic acidosis during afferent arteriole, decrease the filtration pressure
reperfusion. The anesthesiologist must be cogni- and therefore contribute to and prolong the course
zant of potential adverse effects such as hyper- of acute renal failure. However, in a vasodilatory
carbia, hypernatremia, rebound alkalosis and state norepinephrine may actually increase filtra-
worsening intracellular acidosis [50] and sodium tion pressure. Arginine vasopressin constricts the
bicarbonate should be administered slowly while glomerular efferent arteriole and therefore
hperventilating the patient to allow removal of increases filtration pressure and consequently the
excess carbondioxide. Tris-­hydroxymethyl ami- glomerular filtration rate. Administration of low
nomethane (THAM) is a buffer that appears to dose vasopressin also compensates for endoge-
safely control acidosis during the reperfusion nous vasopressin in liver failure [40] and works
phase of liver transplantation and is considered synergistic with sympathomimetics such as
by some an alternative to sodium bicarbonate. norepenpehrine.
However, THAM accumulates in patients with Invasive monitors such as arterial, central
renal dysfunction and its ubiquitous use cannot venous and pulmonary arterial catheters and pos-
be recommended. As of 2017 it is not available sibly a transesophageal echocardiography can be
anymore in the US. Severe bleeding is frequently used to guide hemodynamic management as dis-
encountered during liver transplantation requir- cussed elsewhere in this book. Additionally, since
ing massive transfusion of blood products. Blood patients are ventilated and paralyzed, pulse pres-
transfusion in patients with renal failure may sure variation and stroke volume variation can
cause hyperkalemia and this may further be further help guide management [51, 52].
exacerbated by reperfusion of the graft. It should Obviously, urine output must be closely moni-
be aggressively treated using insulin-­glucose to tored with a Foley catheter. Frequent point-of-­
drive extracellular potassium into cells, calcium care assessment of the acid–base status and
to ameliorate the effect of potassium on the myo- electrolytes balance aid in determining if renal
cardium, and loop diuretics to increase renal replacement is needed.
potassium secretion. However, preexisting kid-
ney dysfunction or acute kidney injury might
make the loop of Henle resistant or unresponsive  enal Replacement Therapy (RRT)
R
to loop diuretics. Several strategies can be used During and After Liver
to increase the effectiveness of loop diuretics, if Transplantation
resistance is suspected, including using an intra-
venous infusion over intermittent bolus dosing Given significant acidosis and, frequently,
or the concomitant administration of a thiazide hyperkalemia, RRT, either as iHD or as CRRT,
(hydrochlorothiazide) or thiazide-­like (metola- may be required in the perioperative period
zone) diuretic. It appears that these strategies [53]. Sustained Low Efficiency Dialysis
increase urine output and natiuresis without (SLED) is a hybrid form of RRT that is essen-
increased side effects. tially a slower version of iHD using the same
22  The Patient with Severe Co-morbidities: Renal Failure 277

machinery with lower blood flow, longer dialy- solute and fluid removal. CVVHDF is the pre-
sis sessions (8–10 h vs. 3–4 h) and possibly less ferred method during liver transplantation
hypotension [54]. As stated before, CRRT is because of its ability to control both fluid and
preferred in the perioperative setting due to its solute clearance. It is safe and allows close con-
hemodynamic stability. It has yet to be deter- trol of fluid balance [55]. For the patient under-
mined (but is an active area of investigation) going liver transplantation, CVVHDF requires
which patients will benefit most from intraop- a large bore double lumen catheter that allows
erative CRRT. Some centers have developed blood flows of 150–300 mL/min and counter-
protocols to identify patients who may benefit current dialysate flows of 2–6 L/h without the
from CRRT during surgery [53]. In addition to need for anticoagulation [20, 55]. Alternatively,
renal failure CRRT may be used for other indi- the CVVHDF machines can be linked into a
cations during liver transplantation. ESLD with veno-venous bypass circuit if dialysis vascular
ascites is associated with refractory hyponatre- access is not available; the 1–4 L/min flows
mia and massive fluid shifts and transfusion through the bypass circuit provide excellent
during surgery can result in too rapid correction flows for continuous RRT. Most commonly
and central pontine myelinolysis; RRT can be CRRT is continued through the postoperative
used to maintain a more stable sodium level if period until renal function has recovered or the
sodium levels of the dialysate fluid is adjusted patient can be transitioned back to
to the patient’s sodium levels, for example by iHD. Although RRT is generally very safe, the
addition of D5W. This needs to be done with complications related to its use are: vascular
close collaboration with the nephrology ser- access complications from catheter placements,
vice. Use of standard dialysate fluid for exam- air embolus, circuit and catheter clotting, and
ple with a sodium concentration of 140 mEqu/L significant hypothermia due to long circuit tub-
will result in too fast correction of sodium lev- ing and poor heat exchanging [52].
els in hyponatremic patients and increase the
risk of central pontine myelinolysis.
Intraoperative CRRT may also be beneficial Summary
in patients with fulminant hepatic failure. The
cerebral swelling due to large amounts of Renal dysfunction in the setting of liver dys-
retained ammonia places these patients at risk function is an important cause of morbidity
for perioperative brainstem herniation; brain- and mortality in the perioperative period of
stem herniation is the most common cause of liver transplantation. All types of renal injury
death in these patients. CRRT may reduce the can coexist with advanced cirrhosis and recog-
large alterations of intracranial pressure associ- nizing and treating the underlying etiology is
ated with IVC cross-clamping and large-volume of paramount importance. In addition, hepa-
transfusion [52]. torenal syndrome should be treated appropri-
There are several forms of CRRT including, ately, although liver transplantation is the only
but not limited to, slow continuous ultrafiltra- long-term treatment. Unfortunately, there is no
tion (SCUF), continuous venovenous hemofil- therapy that prevents or treats acute kidney
tration (CVVH), continuous venovenous injury. As a result, perioperative management
hemodialysis (CVVHD), and continuous veno- of acute kidney injury should include main-
venous hemodiafiltration (CVVHDF). Basic taining renal blood flow, renal perfusion, nor-
circuits for different modalities of renal replace- movolemia, and preventing further injury. Use
ment therapies are depicted in Fig. 22.3. These of intraoperative CRRT can be used in patients
forms of RRT use the principles of ultrafiltra- with severe renal dysfunction or renal failure
tion, hemofiltration, and/or hemodialysis for to manage severe volume overload, hyperkale-
278 A. Disque and J. Meltzer

Access
Access
SCUF
Return
CVVH
Return

P
R
I P
S R
M I
A S
M
A

Effluent
Effluent

CVVHD
Access CVVHDF

Dialysate Access
Return Dialysate
Return

P
R
I
S P
M R
A I
S Replacement
M
A

Effluent

Effluent

Fig. 22.3  Modalities of renal replacement therapy: SCUF Slow continuous ultrafiltration, CVVH Continuous veno-venous
hemofiltration, CVVHD Continuous veno-­venous hemodialysis, CHHDF Continuous veno-venous hemodiafiltration
22  The Patient with Severe Co-morbidities: Renal Failure 279

mia and metabolic abnormalities, but may be sodium and water retention in cirrhosis. Hepatology.
1988;8(5):1151–7.
logistically challenging.
16. Salerno F, Gerbes A, Gines P, Wong F, Arroyo

V. Diagnosis, prevention and treatment of hepatorenal
syndrome in cirrhosis. Gut. 2007;56(9):1310–8.
References 17. Alessandria C, Ozdogan O, Guevara M, et al. MELD
score and clinical type predict prognosis in hepa-
torenal syndrome: relevance to liver transplantation.
1. Gines P, Schrier RW. Renal failure in cirrhosis. N
Hepatology. 2005;41(6):1282–9.
Engl J Med. 2009;361(13):1279–90.
18. Martin-Llahi M, Pepin MN, Guevara M, et al.

2. Nair S, Verma S, Thuluvath PJ. Pretransplant renal
Terlipressin and albumin vs albumin in patients with
function predicts survival in patients undergo-
cirrhosis and hepatorenal syndrome: a randomized
ing orthotopic liver transplantation. Hepatology.
study. Gastroenterology. 2008;134(5):1352–9.
2002;35(5):1179–85.
19. Angeli P, Gines P, Wong F, et al. Diagnosis and man-
3. Gonwa TA, Klintmalm GB, Levy M, Jennings LS,
agement of acute kidney injury in patients with cir-
Goldstein RM, Husberg BS. Impact of pretransplant
rhosis: revised consensus recommendations of the
renal function on survival after liver transplantation.
International Club of Ascites. Gut. 2015;64(4):531–7.
Transplantation. 1995;59(3):361–5.
20. Wagener G, Brentjens TE. Anesthetic concerns in
4. Wiesner R, Edwards E, Freeman R, et al. Model for
patients presenting with renal failure. Anesthesiol
end-stage liver disease (MELD) and allocation of
Clin. 2010;28(1):39–54.
donor livers. Gastroenterology. 2003;124(1):91–6.
21. Sherman DS, Fish DN, Teitelbaum I. Assessing renal
5. Kamath PS, Kim WR. The model for end-stage liver
function in cirrhotic patients: problems and pitfalls.
disease (MELD). Hepatology. 2007;45(3):797–805.
Am J Kidney Dis. 2003;41(2):269–78.
6. Thadhani R, Pascual M, Bonventre JV. Acute renal
22. Hojs R, Bevc S, Ekart R, Gorenjak M, Puklavec

failure. N Engl J Med. 1996;334(22):1448–60.
L. Serum cystatin C as an endogenous marker of renal
7. Kellum JA, Levin N, Bouman C, Lameire
function in patients with mild to moderate impair-
N. Developing a consensus classification sys-
ment of kidney function. Nephrol Dial Transplant.
tem for acute renal failure. Curr Opin Crit Care.
2006;21(7):1855–62.
2002;8(6):509–14.
23. Wagener G, Minhaz M, Mattis FA, Kim M, Emond
8. Mehta RL, Kellum JA, Shah SV, et al. Acute
JC, Lee HT. Urinary neutrophil gelatinase-­associated
Kidney Injury Network: report of an initiative to
lipocalin as a marker of acute kidney injury after
improve outcomes in acute kidney injury. Crit Care.
orthotopic liver transplantation. Nephrol Dial
2007;11(2):R31.
Transplant. 2011;26(5):1717–23.
9. Bellomo R, Ronco C, Kellum JA, Mehta RL, Palevsky
24. Verna EC, Brown RS, Farrand E, et al. Urinary neu-
P. Acute renal failure—definition, outcome measures,
trophil gelatinase-associated lipocalin predicts mor-
animal models, fluid therapy and information tech-
tality and identifies acute kidney injury in cirrhosis.
nology needs: the Second International Consensus
Dig Dis Sci. 2012;57(9):2362–70.
Conference of the Acute Dialysis Quality Initiative
25. Matuszkiewicz-Rowinska J, Malyszko J, Wieliczko
(ADQI) Group. Crit Care. 2004;8(4):R204–12.
M. Renal support during liver transplantation: when
10. Cholongitas E, Calvaruso V, Senzolo M, et al. RIFLE
to consider it? Transplant Proc. 2013;45(9):3157–62.
classification as predictive factor of mortality in
26. Schortgen F, Lacherade JC, Bruneel F, et al. Effects
patients with cirrhosis admitted to intensive care unit.
of hydroxyethylstarch and gelatin on renal function in
J Gastroenterol Hepatol. 2009;24(10):1639–47.
severe sepsis: a multicentre randomised study. Lancet.
11. Schrier RW, Wang W. Acute renal failure and sepsis.
2001;357(9260):911–6.
N Engl J Med. 2004;351(2):159–69.
27. Finfer S, Bellomo R, Boyce N, French J, Myburgh
12. Meltzer J, Brentjens TE. Renal failure in patients

J, Norton R. A comparison of albumin and saline for
with cirrhosis: hepatorenal syndrome and renal
fluid resuscitation in the intensive care unit. N Engl J
support strategies. Curr Opin Anaesthesiol.
Med. 2004;350(22):2247–56.
2010;23(2):139–44.
28. Tandon P, Garcia-Tsao G. Bacterial infections, sepsis,
13. Martin PY, Gines P, Schrier RW. Nitric oxide as a
and multiorgan failure in cirrhosis. Semin Liver Dis.
mediator of hemodynamic abnormalities and sodium
2008;28(1):26–42.
and water retention in cirrhosis. N Engl J Med.
29. Rivers E, Nguyen B, Havstad S, et al. Early goal-­
1998;339(8):533–41.
directed therapy in the treatment of severe sepsis and
14. Arroyo V, Gines P, Gerbes AL, et al. Definition and
septic shock. N Engl J Med. 2001;345(19):1368–77.
diagnostic criteria of refractory ascites and hepatore-
30. The Acute Respiratory Distress Syndrome Network.
nal syndrome in cirrhosis. International Ascites Club.
Ventilation with lower tidal volumes as compared
Hepatology. 1996;23(1):164–76.
with traditional tidal volumes for acute lung injury
15. Schrier RW, Arroyo V, Bernardi M, Epstein M,

and the acute respiratory distress syndrome. N Engl J
Henriksen JH, Rodes J. Peripheral arterial vasodila-
Med. 2000;342(18):1301–8.
tion hypothesis: a proposal for the initiation of renal
280 A. Disque and J. Meltzer

31. Finfer S, Chittock DR, SY S, et al. Intensive versus systemic stent-shunt in non-transplant cirrhotics
conventional glucose control in critically ill patients. with hepatorenal syndrome: a phase II study. Gut.
N Engl J Med. 2009;360(13):1283–97. 2000;47(2):288–95.
32. Tsai MH, Peng YS, Chen YC, et al. Adrenal insuf- 43. Phua J, Lee KH. Liver support devices. Curr Opin Crit
ficiency in patients with cirrhosis, severe sepsis and Care. 2008;14(2):208–15.
septic shock. Hepatology. 2006;43(4):673–81. 44. Davis CL, Feng S, Sung R, et al. Simultaneous liver-­
33. Dellinger RP, Levy MM, Carlet JM, et al. Surviving kidney transplantation: evaluation to decision making.
Sepsis Campaign: international guidelines for man- Am J Transplant. 2007;7(7):1702–9.
agement of severe sepsis and septic shock: 2008. Crit 45. Kusudo K, Ishii K, Rahman M, et al. Blood flow-­
Care Med. 2008;36(1):296–327. dependent changes in intrarenal nitric oxide levels
34. Bellomo R, Chapman M, Finfer S, Hickling K,
during anesthesia with halothane or sevoflurane. Eur J
Myburgh J. Low-dose dopamine in patients with early Pharmacol. 2004;498(1–3):267–73.
renal dysfunction: a placebo-controlled randomised 46. Gentz BA, Malan TP Jr. Renal toxicity with sevoflu-
trial. Australian and New Zealand Intensive Care rane: a storm in a teacup? Drugs. 2001;61(15):2155–62.
Society (ANZICS) Clinical Trials Group. Lancet. 47. Clavien PA, Petrowsky H, DeOliveira ML, Graf

2000;356(9248):2139–43. R. Strategies for safer liver surgery and partial liver
35. Friedrich JO, Adhikari N, Herridge MS, Beyene
transplantation. N Engl J Med. 2007;356(15):1545–59.
J. Meta-analysis: low-dose dopamine increases urine 48. Schroeder RA, Kuo PC. Pro: low central venous

output but does not prevent renal dysfunction or pressure during liver transplantation—not too low. J
death. Ann Intern Med. 2005;142(7):510–24. Cardiothorac Vasc Anesth. 2008;22(2):311–4.
36. Lassnigg A, Donner E, Grubhofer G, Presterl E,
49. Massicotte L, Beaulieu D, Thibeault L. Con: low

Druml W, Hiesmayr M. Lack of renoprotective effects central venous pressure during liver transplantation. J
of dopamine and furosemide during cardiac surgery. J Cardiothorac Vasc Anesth. 2008;22(2):315–7.
Am Soc Nephrol. 2000;11(1):97–104. 50. Nahas GG, Sutin KM, Fermon C, et al. Guidelines
37. Ho KM, Sheridan DJ. Meta-analysis of frusemide for the treatment of acidaemia with THAM. Drugs.
to prevent or treat acute renal failure. BMJ. 1998;55(2):191–224.
2006;333(7565):420. 51. Solus-Biguenet H, Fleyfel M, Tavernier B, et al.

38. Moreau R, Lebrec D. Acute renal failure in
Non-invasive prediction of fluid responsive-
patients with cirrhosis: perspectives in the age of ness during major hepatic surgery. Br J Anaesth.
MELD. Hepatology. 2003;37(2):233–43. 2006;97(6):808–16.
39. Sanyal AJ, Boyer T, Garcia-Tsao G, et al. A random- 52. Biais M, Nouette-Gaulain K, Roullet S, Quinart A,
ized, prospective, double-blind, placebo-controlled Revel P, Sztark F. A comparison of stroke volume
trial of terlipressin for type 1 hepatorenal syndrome. variation measured by Vigileo/FloTrac system and
Gastroenterology. 2008;134(5):1360–8. aortic Doppler echocardiography. Anesth Analg.
40. Wagener G, Kovalevskaya G, Minhaz M, Mattis
2009;109(2):466–9.
F, Emond JC, Landry DW. Vasopressin deficiency 53. Agopian VG, Dhillon A, Baber J, et al. Liver transplan-
and vasodilatory state in end-stage liver disease. J tation in recipients receiving renal replacement ther-
Cardiothorac Vasc Anesth. 2011;25(4):665–70. apy: outcomes analysis and the role of intraoperative
41. Wong F, Pantea L, Sniderman K. Midodrine, octreo- hemodialysis. Am J Transplant. 2014;14(7):1638–47.
tide, albumin, and TIPS in selected patients with cir- 54. Goren O, Matot I. Perioperative acute kidney injury.
rhosis and type 1 hepatorenal syndrome. Hepatology. Br J Anaesth. 2015;115(Suppl 2):ii3–14.
2004;40(1):55–64. 55. Townsend DR, Bagshaw SM, Jacka MJ, Bigam D,
42. Brensing KA, Textor J, Perz J, et al. Long term
Cave D, Gibney RT. Intraoperative renal support during
outcome after transjugular intrahepatic porto- liver transplantation. Liver Transpl. 2009;15(1):73–8.
The Patient with Severe
Co-morbidities: Cardiac Disease 23
Shahriar Shayan and Andre M. De Wolf

Keywords plications [2]. Poor left ventricular function


Coronary artery disease · Cardiomyopathy · (ejection fraction <35%) or severe cardiac dis-
Preoperative evaluation · Heart failure · ease that cannot be improved or corrected should
Myocardial stress test · Arrhythmias be considered to be contraindications for LTx and
only rarely can a patient with these conditions be
considered for combined heart Tx/LTx [3]. For
Introduction the purpose of this book portopulmonary hyper-
tension is considered a pulmonary morbidty and
In order to properly discuss the anesthetic man- will be discussed elsewhere in this book.
agement of patients with cardiac co-­morbidities
undergoing liver transplantation (LTx), we will
first briefly describe the cardiovascular changes  he Cardiovascular Changes in End-­
T
that occur as a result of liver failure, including Stage Liver Disease (ESLD)
hemodynamic changes and cirrhotic cardiomy-
opathy. We will then concentrate on the following Severe liver disease results in significant changes
co-morbidities: coronary artery disease, valvular in circulatory and cardiac function that can be
heart disease, arrhythmias, and hypertrophic summarized as a hyperdynamic circulation; this
obstructive cardiomyopathy. Preoperative diag- is characterized by increased cardiac output,
nosis of cardiac co-morbidities is essential to heart rate, and blood volume; peripheral vasodi-
ensure preoperative optimization and proper lation; and low systemic blood pressure [4]. With
intraoperative management and helps to deter- mild liver dysfunction the cardiovascular changes
mine the potential need for combined cardiac sur- may be well compensated and nearly impercep-
gery and LTx. Cardiac complications after liver tible clinically, however the circulatory effects
transplantation is an important cause for postop- may already have well progressed. The arterial
erative mortality with previous cardiac disease as compliance increases and the overall systemic
the main risk factor [1]. Early postoperative death vascular resistance (SVR) decreases incremen-
(within 30 days of LTx) is as high as 2.9% and tally, corresponding to the degree of liver failure.
40% of these mortalities are due to cardiac com- As liver dysfunction progresses, the circulatory
burden of biologically active compounds such as
S. Shayan, MD • A. M. De Wolf, MD (*) estrogen, bradykinin, prostacyclin, nitric oxide
Depatment of Anesthesiology, Northwestern (NO) and vasoactive intestinal peptide exert a
Memorial Hospital, Chicago, IL, USA predominantly vasodilator effect on the vascular
e-mail: a-dewolf@northwestern.edu

© Springer International Publishing AG, part of Springer Nature 2018 281


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_23
282 S. Shayan and A. M. De Wolf

smooth muscle. These and other vasodilating lation of the SNS and RAAS results in a large
substances are overproduced or cleared less (as a increase in stroke volume and cardiac output.
result of reduced metabolism in the diseased liver Eventually, with progressive liver failure, the
or due to bypassing the liver), furthermore there SNS and RAAS become maximally stimulated
may be an increased sensitivity to their vasodila- and the increase in cardiac output and vasocon-
tory effects. In addition, peripheral arteriovenous striction in certain vascular beds is insufficient
communications form and the sensitivity to vaso- to maintain an effective circulatory volume
constrictors such as norepinephrine and endothe- and compensate for the massive vasodilation
lin-­
1 decreases due to a reduced number of of the splanchnic system. As a consequence
receptors in combination with post-receptor blood pressure decreases and progressive auto-
defects. nomic dysfunction and baroreceptor insensi-
Although SVR decreases in patients with tivity will further exacerbate this inadequate
severe liver disease, not all vascular beds are compensation.
affected in the same way. As the primary distur- Activation of the SNS and RAAS can be det-
bance in ESLD, portal hypertension develops as rimental to the function of other organs. Indeed,
a result of increased hepatic vascular resistance the persistent sympathetic stimulation results in
at the level of the sinusoids and is a direct con- vasoconstriction of coronary, cerebral, and renal
sequence of local structural changes (fibrosis vessels. This is most apparent in the kidneys,
and regeneration nodules) and sinusoidal vaso- where reduction of blood flow in addition to a
constriction (locally decreased NO production, reduced circulatory volume may result in the pro-
and increased local release of and sensitivity to gression to hepatorenal syndrome with fluid
vasoconstrictors such as endothelin, angioten- retention, hyponatremia, and ascites formation.
sin II, catecholamines, and leukotrienes). The Although activation of the SNS results in a
spanchnic’s circulatory response to portal hyper- persistent state of sympathetic stimulation, it
tension is characterized by a massively increased does not necessarily lead to a better myocardial
local production of NO resulting in severe vaso- performance. To the contrary ESLD may cause
dilation of the splanchnic circulation. In addition, progressive myocardial dysfunction called cir-
splanchnic vessels are less responsive to vaso- rhotic cardiomyopathy. Cardiac dysfunction in
constrictors and release of substances such as liver disease unrelated to alcohol was first
vascular endothelial growth factor results in the described by Ma in 1996, and consists of systolic
creation of portosystemic collaterals. Other vas- dysfunction, diastolic dysfunction and electro-
cular beds however undergo vasoconstriction as a physiologic abnormalities [6]. Despite increased
result of activation of compensatory mechanisms. cardiac output in ESLD, the systolic contractility
The severe splanchnic vasodilatation leads and diastolic relaxation are attenuated.
to intravascular volume redistribution, which Furthermore repolarization changes such as pro-
results in a reduction in central and arterial blood longed QT interval (which may improve after
volume and an increase in non-central blood β-blocker therapy), and reduced inotropic and
volume (mainly splanchnic system) (Fig. 23.1) chronotropic response to β-adrenergic stimula-
[5]. This is detected by central baroreceptors, tion may occur. Although cirrhotic cardiomyopa-
and leads to activation of compensatory mecha- thy is usually not apparent at rest, it becomes
nisms, mainly the sympathetic nervous system noticeable during cardiac stress (increase in pre-
(SNS) and the Renin Angiotensin Aldosterone load or afterload). For example cardiac dysfunc-
System (RAAS) Initially there may also an tion may become clinically relevant for the first
increased release of vasopressin by the pituitary time after transjugular intrahepatic portosystemic
gland, and an increased concentration of circu- shunt (TIPS) placement or in the early postopera-
latory endothelins. However duing later stages tive period after LTx. The cause of cirrhotic car-
vasopressin levels are low in cirrhosis. In com- diomyopathy is multifactorial; this includes
bination with the reduction in SVR, the stimu- circulating myocardial depressant substances
23  The Patient with Severe Co-morbidities: Cardiac Disease 283

Portal hypertension
Portosystemic
shunting

Increased
Arteriolar
Splanchnic blood
vasodilation
flow

Increased
• Cardiac output Low SVR
• Heart rate

Activation of Low
• SNS • Blood pressure
• RAAS • Central blood volume
• ET-1 • Lung blood volume

Fig. 23.1  Pathophysiology of hemodynamic changes in there is activation of sympathetic nervous system (SNS)
cirrhosis: systemic overproduction of vasodilators results and Renin Angiotensin Aldosterone System (RAAS), and
in arteriolar vasodilation and low systemic vascular resis- increased plasma concentrations of endothelin-1 (ET-1).
tance (SVR), resulting in low blood pressure. This leads to increases in cardiac output, heart rate,
Redistribution of blood results in a reduction in central plasma volume (fluid and water retention), and splanchnic
blood volume and lung blood volume. Consequently, blood flow

(tumor necrosis factor-α, bile acids, endotoxins, correlates well with systolic dysfunction; how-
cytokines, carbon monoxide, endogenous canna- ever there is evidence that diastolic dysfunction
binoids, etc.), and down-regulation of β-receptors precedes systolic dysfunction [7]. Diastolic dys-
(reduced β-receptor density, desensitization of function results in a higher incidence of heart
β-receptors, and abnormal excitation-contraction failure after LTx [8].
coupling). Furthermore morphologic changes in
the heart such as cardiac hypertrophy and patchy
areas of fibrosis and subendothelial edema may Coronary Artery Disease (CAD)
occur and further contribute to the systolic and
diastolic dysfunction. One of the early indicators In the 1960s and 1970s it was thought that patients
of cirrhotic cardiomyopathy is diastolic dysfunc- with severe liver disease had a low incidence of
tion, which can be seen in many patients with CAD, based on a lower incidence of hypercholes-
ESLD. Typically there is a decreased E/A ratio terolemia, increased levels of circulating estrogen
on Doppler echocardiographic examination of (resulting in protection against atherosclerosis)
the blood flow through the mitral valve; the E and decreased SVR thereby eliminating, at least
wave represents early passive transmitral flow, in theory, hypertension as a risk factor for CAD
while the A wave represents transmitral flow as a [9]. However there is increasing evidence that the
result of atrial contraction. It is unclear whether prevalence of CAD in patients with ESLD is at
diastolic dysfunction is a good marker for the least the same if not even higher than in the general
degree of cirrhotic cardiomyopathy or whether it population (20% vs. 12%, ­respectively) [10, 11].
284 S. Shayan and A. M. De Wolf

Obesity, nonalcoholic steatohepatitis (NASH) the true incidence of CAD in patients with ESLD
and other inflammatory liver conditions, and remains unknown.
advancing age of the LTx candidate have lead to
an increasing prevalence of atherosclerosis [12,
13]. Interestingly, the prevalence of CAD is much  onsequence of CAD in Patients
C
higher in patients with alcoholic liver disease Undergoing LTx
(31%) and NASH (27%) than in patients with cir-
rhosis due to other causes (2.4%) [14]. This could Why is there so much emphasis on the preopera-
be related to a higher incidence of smoking, diabe- tive diagnosis of CAD? LTx is a procedure that
tes mellitus, older age, and hypertension in patient creates a substantial stress for the heart with virtu-
with alcoholic liver disease and NASH, but it is ally unavoidable episodes of often severe tachy-
unlikely that these risk factors by themselves can cardia and hypotension. Furthermore plaque
account for the higher incidence of CAD. There is rupture resulting in acute coronary artery throm-
also evidence that while light to moderate alcohol bosis and myocardial infarction may be related to
intake reduces the risk for CAD, heavy episodic a chronic inflammatory state. Episodes of hyper-
alcohol drinking may actually increase its risk coagulability further increase the perioperative
[15]. The prevalence of CAD in patients with viral risk through intracoronary thrombus formation
cirrhosis however is lower than in patients with- triggered by an area of coronary atherosclerosis.
out cirrhosis [16, 17]. Although there is limited Therefore, CAD is considered to increase the
comparative data about the prevalence of CAD in peri- and postoperative risk. In 1996 Plotkin et al.
patients with cirrhosis with different etiologies, reported a 50% 3 year mortality rate after LTx in
one must assume that CAD has a higher incidence patients with CAD, irrespective of whether the
in patients with ESLD than in the general popula- management of CAD was medical or surgical
tion, mainly due to the high incidence of CAD in [22]. Management options for CAD have evolved
patients with alcoholic liver disease and NASH. since then and we can now choose among medical
The reported prevalence of significant CAD management, percutaneous transluminal coro-
(defined as at least one coronary artery stenosis nary angioplasty (PCTA), coronary stenting with
≥50%) in patients with ESLD varies widely bare metal or drug eluting stents, coronary artery
from 2.5% to 27%. There are several reasons for bypass surgery (CABG) and off-pump CABG
this variability. First, most studies have looked at (OPCAB), with cardiac surgery being performed
a relatively small number of patients and second before LTx or as a combined procedure. As a
some studies based the diagnosis of significant result, a more recent study demonstrated an
CAD on abnormal screening tests such as posi- improved outcome, although the mortality rates
tive dobutamine stress echocardiography. Third, were still much higher than in the general LTx
the only method to determine the true incidence population: 1 year mortality rate of 11.9% vs.
of CAD is by coronary angiography and in most 2.4%, and 3 year mortality rate of 26.2% vs. 7.1%,
studies coronary angiography was only per- respectively [23]. Also, post-­ operatively, CAD
formed in the subgroup of patients with abnor- continues to be a significant cause of mortality
mal screening tests or with multiple risk factors after otherwise successful LTx [24]. Interestingly,
for CAD [18–20]. Interestingly, Carey found an a recent retrospective multicenter study suggests
incidence of CAD of 27% in 37 LTx candidates that properly managed obstructive CAD (stenting
older than 45 years who underwent coronary or CABG) results in survival rates that are no dif-
angiography without consideration of other risk ferent from those in patients with non-obstructive
factor [21]; these results raise doubt on the CAD, with 3 year mortality rates 25.3 and 22.7%,
appropriateness of risk stratification of patients respectively (not much different from the survival
that were referred to coronary angiography in rates in all patients undergoing LTx according to
other studies however this study was limited due the ­OPTN/SRTR 2010 Annual Report) [25].
to its small sample size (37 patients). Therefore However, it is important to recognize that these
23  The Patient with Severe Co-morbidities: Cardiac Disease 285

patients were considered to be acceptable LTx however is reduced from 86% to 80% when non-­
candidates and therefore may not represent all diagnostic tests (due to inability of up to 50% of
patients with CAD. patients to reach the target heart rate) are
included [33]. Others found an even lower nega-
tive predictive value (75 and 79%) [34, 35].
Preoperative Evaluation Another interesting finding is that patients who
did not reach the target heart rate during DSE
Preoperative risk stratification is guided by tradi- (“chronotropic incompetence”) had a higher
tional CAD risk factors that include age >50 years, incidence of cardiac complications up to
diabetes mellitus, peripheral vascular disease and 4 months after LTx [31]. The positive predictive
history of CAD [26]. Interestingly, acute renal fail- value of DSE is not nearly as good, ranging from
ure also increases cardiovascular risk in LTx 22% to 44% [18, 19, 30, 32, 34, 35]. Therefore,
patients [27]. Patients with no prior screening tests an abnormal DSE is not necessarily caused by
but several risk factors for CAD had a 26% inci- significant CAD. It has been suggested that the
dence of moderate or severe CAD during coronary positive predictive value may be improved by the
angiography, suggesting that CAD is quite com- use of real time contrast myocardial echocar-
mon in patients with ESLD [20]. However not all diography for patients with intermediate risk
LTx candidates can or should undergo coronary factors for CAD [35]. The wide variability
angiography as the procedure is associated with among studies likely arises from differences in
significant risks of complications such as femoral institutional protocols in selecting patients for
artery and renal injury [28, 29]. LTx candidates DSE, coronary angiography, and definitions of
often present with a poor functional status and outcomes. For example CAD can be defined as
hepatic encephalopathy, making the clinical diag- coronary obstruction >50% vs. >70%, perioper-
nosis of significant CAD through eliciting signs ative myocardial infarction can be diagnosed
and symptoms or exercise tolerance challenging based on different troponin cut-offs and endpoint
and nearly impossible. For the same reasons, exer- could be cardiac mortality or any-­cause mortal-
cise testing is often not feasible. Therefore, there is ity. In addition, many patients failed to achieve
a real need for improved understanding who should the predicted maximal heart rate, rendering the
receive what screening test and who should then ability of interpreting the DSE rather marginal
undergo coronary angiography. [31, 34]. This may be the result of the use of
β-blockers as part of medical management of
 obutamine Stress Echocardiography
D portal hypertension, in addition to down-­
(DSE) regulation of β-receptors in ESLD (see above).
DSE is the most frequently used screening test Withholding β-blockers before the test and the
for CAD in LTx candidates. Dobutamine is administration of atropine has been recom-
administered at an increasing dose in an attempt mended to reduce the number of inconclusive
to achieve 85% of the predicted maximal heart tests due to submaximal heart rates [31] but
rate. The associated increase in myocardial oxy- withholding β-blockers may increase the risk of
gen demand attempts to mimic the physiologic variceal bleeding [36]. Because of the relatively
stress that the myocardium undergoes in the poor predictive value of DSE in predicting peri-
perioperative period. Obstructive CAD is operative cardiac events or early mortality, some
detected by regional wall motion abnormalities clinicians use alternative or additional screening
in the myocardial territories at maximal heart tests for CAD in order to avoid unnecessary cor-
rate. Several studies show that a negative DSE is onary angiographies. However, in our opinion it
highly predictive of a myocardial injury-free is still much better to obtain some false positive
perioperative course [18, 19, 30–32], and thus a screening test results (resulting in unnecessary
normal DSE has a good negative predictive value coronary angiographies) than too many false
(range 89–100%). The negative predictive value negative results resulting in patients accepted for
286 S. Shayan and A. M. De Wolf

LTx with unrecognized significant CAD. Also, obstruction. There is a good correlation between
no other screening test has a better positive pre- the CAC score and the presence of risk factors for
dictive value than DSE at this time. CAD [45, 46] but currently no studies compare
the CAC scores to traditional contrast coronary
 yocardial Perfusion Scan
M angiography in the catheterization laboratory, nor
Single photon emission computed tomography are there any outcome studies. However, not all
(SPECT) myocardial perfusion scintigraphy is plaques are calcified and using the same test CT
another screening test for CAD. It uses exercise, coronary angiography theoretically allows the
dobutamine or vasodilators such as adenosine or detection of noncalcified plaques [45]. Again,
dipyridamole to stress the myocardium and there are no studies that compare abnormal CT
determines the relative blood flow to different coronary angiography tests with traditional con-
areas of the myocardium. Defects in perfusion trast coronary angiography and therefore the use-
can be classified as fixed (scar) or reversible fulness of CT coronary angiography in patient
(presumably ischemia) and defects in at least with ESLD remains to be determined.
three segments (out of 17 or 20) are indicative of In conclusion, the currently available screening
at least moderate risk for CAD [37]. Overall, the tests for CAD are not very good. Both DSE and
positive predictive value (range: 15–50%) and myocardial perfusion scan have a good negative
the negative predictive value (range: 77–99%) predictive value, but the positive predictive value
are worse than for DSE [38–41]. These results is not nearly as good, although slightly better for
are worse than those in patients without liver dis- DSE than for myocardial perfusion scan. There is
ease; this can be attributed to the decreased base- little experience with CT coronary angiography
line arterial vascular resistance in patients with and it is therefore difficult to estimate its ability as
ESLD, as the typical response of the coronary a screening test for CAD in LTx candidates. Since
arteries to vasodilators may not be achieved [39]. DSE gives additional information about systolic
In addition, false positive tests could be the result and diastolic cardiac function, valvular disease,
of abnormal coronary microvascular tone [42], hypertrophic cardiomyopathy, peak right ventricu-
which has also been observed in patients without lar pressure, and hepatopulmonary syndrome, it
severe liver disease [43]. This abnormal micro- seems to be the preferred screening test at this time
vascular (coronary) blood flow (in the presence [11]. Our algorithm for preoperative screening and
of normal coronary angiography) may be associ- management of CAD is presented in Fig. 23.2.
ated with a higher perioperative morbidity and
mortality rate, sepsis, and graft failure [44]. I nvasive, Diagnostic Evaluation of CAD
Furthermore, ascites may result in attenuation Coronary angiography using the standard dye
artifacts in the inferior wall that may mimic isch- technique in the catheterization laboratory is
emia or scar tissue [40]. Therefore, a high num- considered the gold standard for detection of
ber of false-positive results makes this test less CAD. A positive screening test for CAD should be
accurate [41] and myocardial perfusion scan ­followed by coronary angiography to confirm the
may be only indicated as a screening test in presence of CAD considering the relatively low
patient with several risk factors for CAD who do positive predictive value of these screening tests.
not tolerate or have an inconclusive DSE. Infrequently coronary angiography is performed
in candidates with several cardiac risk factors
Computerized Tomography (CT) (e.g., diabetes, age >50 years, hypertension, smok-
Coronary Angiography and Coronary ing, family history of CAD, and hypercholester-
Artery Calcification (CAC) olemia) even in the presence of normal screening
Coronary artery calcification (CAC) determined tests. This may be justified in patients with >2 risk
by multisection CT reflects the degree of calcifi- factors for CAD [20], especially in patients with
cation of coronary atherosclerotic lesions and alcoholic liver disease and NASH, as the incidence
may be an indicator of the degree of coronary of CAD is significantly higher in these patients.
23  The Patient with Severe Co-morbidities: Cardiac Disease 287

Risk factors for CAD

CAD or significant PVD Major risk factors Minor risk factors


Diabetes, NASH, EtOH, Age > 50 yr, HTN,
Smoking, Renal failure Hyperlipidemia, Obesity

Multiple risk factors Some risk factors Minimal or no risk factors

DSE

Positive Inconclusive/ Negative


Chronotropicincompetence

Alternative test, e.g.,:


Myoc. perf. scan
CT angiography
Positive Negative
Coronary angiography

Obstructive CAD No sign of obstructive CAD

Reject candidacy
OPCAB/CABG/Stent
Simultaneous CABG/LTx Proceed with LTx

Fig. 23.2  Coronary artery disease in orthotopic liver EtOH Alcohol disease, DSE Dobutamine stress
transplantation: pretransplant assessment and manage- Echocardiogram, HTN Hypertension, OPCABG Off-­pump
ment. CAD Coronary artery disease, PVD Peripheral vas- Coronary artery bypass graft, CABG Coronary artery
cular disease, NASH Non-alcoholic steatotic hepatitis, bypass graft, Myoc. Perf. Scan Myocradial perfusion scan

Cardiac catheterization and coronary angiography optimized prior to LTx, because of excessively
are associated with a higher number of complica- high perioperative mortality if left untreated [47].
tions in patients with ESLD compared to patients With proper management, the outcome is not
without ESLD: patients with ESLD may have much different from that in the general LTx pop-
less renal function reserve, resulting in a higher ulation [25]. The best strategy to accomplish this
incidence of renal dysfunction and there is an has not been determined, since no randomized
increased incidence of bleeding complications at controlled trials have compared percutaneous
the site of vascular access [29]. Using the radial revascularization to surgical techniques in this
artery for vascular access is becoming more com- population. The main therapeutic options besides
mon as it may have a reduced complication rate. medical management are placement of coronary
stents, coronary artery bypass grafting (CABG),
and off-pump coronary artery bypass (OPCAB).
Management of CAD
 oronary Stent Placement
C
If significant CAD is diagnosed preoperatively, Although coronary stent placement is an effec-
the coronary status of these patients should be tive method of revascularization it is not without
288 S. Shayan and A. M. De Wolf

risks in patients with ESLD. Antiplatelet ther-  ff-Pump Coronary Artery Bypass


O
apy is required after stent placement in order to (OPCAB)
maintain patency and this further increases the Off-pump coronary artery bypass (OPCAB)
risk of bleeding complications. However, the offers several theoretical advantages over CABG:
potential for clot formation is not as low in no need for cardiopulmonary bypass and there-
patients with ESLD as previously thought [48], fore less requirement for anticoagulation, and
at least in part due to increased concentration of better pulsatile organ perfusion. If CAD is the
von Willebrand factor [49]. Most commonly only cardiac lesion to be corrected, then OPCAB
bare metal stents are used instead of drug elut- would theoretically offer significant advantages,
ing stents, because bare metal stents are cov- especially in patients with ESLD [51]. While
ered faster by an endothelial layer and therefore some confirmed this hypothesis [52, 57, 58], oth-
do not require prolonged dual antiplatelet ther- ers found no improvement in incidence of hepatic
apy (1–3 months vs. 12 months). The disadvan- dysfunction and overall mortality when OPCAB
tage of bare metal stents is the long-term higher was used [59].
restenosis rate, but this may not result in a
higher incidence of acute myocardial infarc-
tion or death. The risks associated with arterial Valvular Disease
vascular access are similar to coronary
angiography. Mild or moderate valvular disease in patients
with ESLD is usually well tolerated. The inci-
CABG dence of mild or moderate tricuspid and mitral
Coronary artery bypass grafting (CABG) may regurgitation is higher than in the general popula-
be the only option in patients with significant tion [60] possibly due to cirrhotic cardiomyopa-
CAD that cannot be corrected by coronary stent thy and subsequent ventricular remodeling.
placement. However, CABG in patients with These conditions require no special consideration
ESLD and CAD prior to LTx is associated with perioperatively, although patients may require
a high mortality, mainly as the result of postop- more blood transfusions and inotropic support
erative liver failure [50–53]. Other complica- [60]. Also, patients with severe valve disease
tions include renal failure, infections, and with mild liver disease tolerate cardiac surgery
bleeding [50, 51, 53, 54]. Patients with mild better with a somewhat increased complication
cirrhosis (Childs A) have up to 25% morbidity rate similar to patients with mild liver disease
(usually late postoperative liver failure and undergoing CABG [51, 54].
wound infections) but a low incidence of mor- Perioperative management of patients with
tality [55]. Patients with moderated cirrhosis severe valvular disease and severe liver disease is
(Childs B) have a morbidity of almost 100% very complex. If an attempt is made to surgically
and mortality of up to 30%. Non-pulsatile blood correct the valvular disease using cardiopulmo-
flow during cardiopulmonary bypass results in nary bypass prior to LTx the outcome will be as
systemic inflammation further contributing to poor as the results of CABG in patients with
liver dysfunction or liver failure. CABG using ESLD [51, 52, 54]. Few patients underwent such
cardiopulmonary bypass is therefore an unat- an operation successfully [61, 62] and other
tractive option for myocardial revascularization options need to be explored. Percutaneous bal-
in patients with ESLD awaiting LTx. A better loon valvuloplasty or transcatheter aortic valve
alternative may be simultaneous CABG/LTx, implantation (TAVI), avoiding cardiopulmonary
with the cardiac procedure performed first, bypass, could be used to correct severe mitral
resulting in excellent results, although it ­disease or aortic stenosis [63]. Another option is
requires significant multidisciplinary coordina- a simultaneous valve replacement and LTx
tion and cooperation from the cardiac surgical although this requires a thoracoabdominal inci-
team [56]. sion, cardiopulmonary bypass at the time of LTx
23  The Patient with Severe Co-morbidities: Cardiac Disease 289

and initiation of immunosuppression [64]. A a sudden drop in cardiac output. Echocardiography


recent report suggests that tricuspid regurgitation is the most useful method of diagnosing HOCM
is associated with a higher mortality and graft as it allows visualization of the HOCM, diagno-
failure; this may be the result of prolonged sis of SAM and estimation of the degree of
hepatic congestion, or it may indicate poor car- obstruction [68]. Volume status, afterload, and
diovascular reserve [65]. myocardial contractility all affect the degree of
LVOT obstruction and mitral regurgitation.
Specifically, low SVR and a hyperdynamic left
Arrhythmias: Atrial Fibrillation ventricle will worsen LVOT obstruction espe-
cially in hypovolemic patients. The hemody-
Atrial fibrillation, the most common cardiac namic goal is to prevent conditions that would
arrhythmia, can be paroxysmal or chronic-­ result in obliteration of the LV cavity and ulti-
persistent. The incidence in LTx candidates is mately LVOT obstruction. Such treatment modal-
higher than in the general population (4.5% vs. ities are focused on increasing LV cavity size by
0.8–1.5%), and although it results in higher peri- avoiding hypovolemia and reducing contractility
operative cardiac morbidity, overall graft and with β-blockers. Myectomy and alcohol septal
patient survival are unchanged [66]. Nevertheless, ablation are reserved for patients with drug-­
it seems reasonable to attempt to prevent recur- refractory heart failure symptoms [68].
rence of atrial fibrillation by β-blocker therapy HOCM poses a particular difficulty for
before LTx. In a more recent study using a patients with ESLD as some of the circulatory
national database, atrial fibrillation was found to abnormalities in ESLD promote LVOT obstruc-
be associated with a higher incidence of major tion. LVOT obstruction can be diagnosed by
adverse cardiovascular events and a lower 1-year DSE, but the incidence seems to be quite vari-
survival after LTx [67]. able ranging from low (two out of 157 patients
developed high LVOT gradients during DSE)
[31] up to 43% of all patients [69]. It is possible
 ypertrophic Obstructive
H that the diagnosis of LVOT obstruction with
Cardiomyopathy (HOCM) DSE depends on if one is actually looking for
LVOT obstruction. A LVOT gradient of
Hypertrophic obstructive cardiomyopathy >35 mmHg has resulted in denial for transplan-
(HOCM) is characterized by an asymmetrically tation, even though the reported perioperative
hypertrophied non-dilated left ventricle, poten- mortality is not neccessarily increased [69].
tially causing left ventricular outflow tract Options for patients rejected for LTx because of
(LVOT) obstruction. It has a genetic inheritance a high LVOT obstruction include myectomy and
pattern, although it can be the result of de novo alcohol septal ablation. Myectomy in patients
genetic mutation, and has an incidence of about with ESLD may be a poor choice with high mor-
0.2% of the general population [68]. Although tality rate mainly resulting from the need for car-
frequently asymptomatic, some patients develop diopulmonary bypass [51], although a combined
anginal chest pain, dyspnea or syncope, and it myectomy—LTx can be an option. Alcohol sep-
can progress to congestive heart failure or sudden tal ablation is less invasive but may be associated
death as a result of dynamic LVOT obstruction, with several complications as well [70] and cur-
mitral regurgitation, diastolic dysfunction, myo- rently there are only a few case reports of patients
cardial ischemia, or arrhythmias [68]. LVOT with ESLD who received alcohol septal ablation
obstruction caused by septal hypertrophy prior to LTx [71, 72].
becomes hemodynamically more significant in Although ESLD and LTx result in hemody-
the presence of systolic anterior motion (SAM) namic conditions that worsen LVOT obstruction,
of the anterior mitral leaflet, that prevents com- these patients can be transplanted safely when
plete ejection of the stroke volume and results in meticulous hemodynamic management is used,
290 S. Shayan and A. M. De Wolf

such as intraoperative avoidance of inotropic 12. Burke A, Lucey MR. Non-alcoholic fatty liver dis-
ease, non-alcoholic steatohepatitis and orthotopic
agents (epinephrine) and hypovolemia. TEE
liver transplantation. Am J Tranplant. 2004;4:686–93.
monitoring is essential in order to avoid hypovo- 13. Zetterman RK, Belle SH, Hoofnagle JH, Lawlor S,
lemia and to closely follow the degree of LVOT Wei Y, Everhart J, et al. Age and liver transplanta-
obstruction and SAM [73–75]. During the anhe- tion: a report of the Liver Transplantation Database.
Transplantation. 1998;66:500–6.
patic stage veno-venous bypass facilitates the
14. Kalaitzakis E, Björnsson E. Coronary artery disease
avoidance of hypovolemia, while hypotension in liver cirrhosis: does the aetiology of liver disease
should be rapidly and aggressively treated with matter? J Hepatol. 2009;51:962–3.
potent vasoconstrictors such as norepinephrine or 15. Murray RP, Connett JE, Tyas SL, Bond R, Ekuma O,
Silversides CK, et al. Alcohol volume, drinking pat-
vasopressin and volume. Also, calcium should be
tern, and cardiovascular disease morbidity and mor-
administered slowly in order to avoid a hypercon- tality: is there a U-shaped function? Am J Epidemiol.
tractile state [76]. 2002;155:242–8.
16. Berzigotti A, Bonfiglioli A, Muscari A, Bianchi G,
LiBassi S, Bernardi M, et al. Reduced prevalence of
ischemic events and abnormal supraortic flow patterns
References in patients with liver cirrhosis. Liver Int. 2005;25:331–6.
17. Kadayifci A, Tan V, Ursell PC, Merriman RB, Bass
1. Fouad TR, Abdel-Razek WM, Burak KW, Bain VG, NM. Clinical and pathologic risk factors for athero-
Lee SS. Prediction of cardiac complications after liver sclerosis in cirrhosis: a comparison between NASH-­
transplantation. Transplantation. 2009;87:763–70. related cirrhosis and cirrhosis due to other aetiologies.
2. VanWagner LB, Lapin B, Levitsky J, Wilkins JT, J Hepatol. 2008;49:595–9.
Abecassis MM, Skaro AI, et al. High early cardio- 18. Donovan CL, Marcovitz PA, Punch JD, Bach DS,
vascular mortality after liver transplantation. Liver Brown KA, Lucey MR, et al. Two-dimensional and
Transpl. 2014;20:1306–16. dobutamine stress echocardiography in the preop-
3. Shaw BW Jr, Bahnson HT, Hardesty RL, Griffith BP, erative assessment of patients with end-stage liver
Starzl TE. Combined transplantation of the heart and disease prior to orthotopic liver transplantation.
liver. Ann Surg. 1985;202:667–72. Transplantation. 1996;61:1180–8.
4. Møller S, Henriksen JH. Cardiopulmonary complica- 19. Plotkin JS, Benitez M, Kuo PC, Njoku MJ, Ridge LA,
tions in chronic liver disease. World J Gastroenterol. Lim JW, et al. Dobutamine stress echocardiography
2006;12:526–38. for preoperative cardiac risk stratification in patients
5. Schrier RW, Arroyo V, Bernardi M, Henriksen JH, undergoing orthotopic liver transplantation. Liver
Rodes J. Peripheral arterial vasodilation hypothesis: Transpl Surg. 1998;4:253–7.
a proposal for the initiation of renal sodium and water 20. Tiukinhoy-Laing SD, Rossi JS, Bayram M, De Luca
retention in cirrhosis. Hepatology. 1988;8:1151–7. L, Gafoor S, Blei A, et al. Cardiac hemodynamic
6. Ma Z, Lee SS. Cirrhotic cardiomyopathy: getting to and coronary angiographic characteristics of patients
the heart of the matter. Hepatology. 1996;24:451–9. being evaluated for liver transplantation. Am J
7. Henriksen JH, Møller S. Cardiac and systemic hemo- Cardiol. 2006;98:178–81.
dynamic complications of liver cirrhosis. Scand 21. Carey WD, Dumot JA, Pimentel RR, Barnes DS,

Cardiovasc J. 2009;43:218–25. Hobbs RE, Henderson JM, et al. The prevalence of
8. Dowsley TF, Bayne DB, Langnas AN, Dumitru I, coronary artery disease in liver transplant candidates
Windle JR, Porter TR, et al. Diastolic dysfunction over age 50. Transplantation. 1995;27:859–64.
in patients with end-stage liver disease is associated 22. Plotkin JS, Scott VL, Pinna A, Dobsch BP, De Wolf
with development of heart failure early after liver AM, Kang Y. Morbidity and mortality in patients with
transplantation affecting survival. Transplantation. coronary artery disease undergoing orthtotopic liver
2012;94:646–51. transplantation. Liver Transpl Surg. 1996;2:426–30.
9. Howell WL, Manion WC. The low incidence of myo- 23. Diedrich DA, Findlay JY, Harrison BA, Rosen

cardial infarction in patients with portal cirrhosis of CB. Influence of coronary artery disease on out-
the liver: a review of 639 cases of cirrhosis of the liver comes after liver transplantation. Transplant Proc.
from 17,731 autopsies. Am Heart J. 1960;60:341–4. 2008;40:3554–7.
10. Kalaitzakis E, Rosengren A, Skommevik T, Björnsson 24. Johnston SD, Morris JK, Cramb R, Gunson BK,

E. Coronary artery disease in patients with liver cir- Neuberger J. Cardiovascular morbidity and mortality
rhosis. Dig Dis Sci. 2010;55:467–75. after orthotopic liver transplantation. Transplantation.
11. Ehtisham J, Altieri M, Salamé E, Saloux E, Ollivier I, 2002;73:901–6.
Hamon M. Coronary artery disease in orthotopic liver 25. Wray C, Scovotti JC, Tobis J, Niemann CU, Planinsic
transplantation: pretransplant assessment and man- R, Walia A, et al. Liver transplantation outcome in
agement. Liver Transpl. 2010;16:550–7. patients with angiographically proven coronary artery
23  The Patient with Severe Co-morbidities: Cardiac Disease 291

disease: a multi-institutional study. Am J Transplant. 39. Davidson CJ, Gheorghiade M, Flaherty JD, Elliott
2013;13:184–91. MD, Reddy SP, Wang NC, et al. Predictive value of
26. Fleisher LA, Beckman JA, Brown KA, Calkins H, stress myocardial perfusion imaging in liver trans-
Chaikof EL, Fleischmann KE, et al. ACC/AHA 2007 plant candidates. Am J Cardiol. 2002;89:359–60.
guidelines on perioperative cardiovascular evaluation 40. Zoghbi GJ, Patel AD, Ershadi RE, Heo J, Bynon
and care for noncardiac surgery. J Am Coll Cardiol. JS, Iskandrian AE. Usefulness of preoperative stress
2007;50:1707–32. perfusion imaging in predicting prognosis after liver
27. Cholongitas E, Senzolo M, Patch D, Shaw S,
transplantation. Am J Cardiol. 2003;92:1066–71.
O’Beirne J, Burroughs AK. Cirrhotics admitted 41. Aydinalp A, Bal U, Atar I, Ertan C, Aktas A, Yildirir
to intensive care unit: the impact of acute renal A, et al. Value of stress myocardial perfusion scan-
failure on mortality. Eur J Gastroenterol Hepatol. ning in diagnosis of severe coronary artery disease
2009;21:744–50. in liver transplantation candidates. Transplant Proc.
28. Lester SJ, Hurst RT. Liver transplantation: do you 2009;41:3757–60.
have the heart for it? Liver Transpl. 2006;12:520–2. 42. Senzolo M, Bassanello M, Graziotto A, Zucchetta P,
29. Sharma M, Yong C, Majure D, Zellner C, Roberts JP, Cillo U, Maraglino G, et al. Microvascular autonomic
Bass NM, et al. Safety of cardiac catheterization in dysfunction may justify false-positive stress myocar-
patients with end-stage liver disease awaiting liver dial perfusion imaging in patients with liver cirrho-
transplantation. Am J Cardiol. 2009;103:742–6. sis undergoing liver transplantation. Transplant Proc.
30. Findlay JY, Keegan MT, Pellikka PP, Rosen CB,
2008;40:1916–7.
Plevak DJ. Preoperative dobutamine stress echocar- 43. Lanza GA, Crea P. Primary coronary microvascular
diography, intraoperative events, and intraoperative dysfunction. Clinical presentation, pathophysiology,
myocardial injury in liver transplantation. Transplant and management. Circulation. 2010;121:2317–25.
Proc. 2005;37:2209–13. 44. Guckelberger O, Byram A, Klupp J, Neumann UP,
31. Umphrey LG, Hurst RT, Eleid MF, Lee KS, Reuss CS, Glanemann M, Stockmann M, et al. Coronary event
Hentz JG, et al. Preoperative dobutamine stress echo- rates in liver transplant recipients reflect the increased
cardiographic findings and subsequent short-term prevalence of cardiovascular risk factors. Transpl Int.
adverse cardiac events after orthotopic liver transplan- 2005;18:967–74.
tation. Liver Transpl. 2008;14:886–92. 45. Assy N, Djibre A, Farah R, Grosovski M, Marmor
32. Safadi A, Homsi M, Maskoun W, Lane KA, Singh I, A. Presence of coronary plaques in patients with nonalco-
Sawada SG, et al. Perioperative risk predictors of car- holic fatty liver disease. Radiology. 2010;254:393–400.
diac outcomes in patients undergoing liver transplan- 46. McAvoy NC, Kochar N, McKillop G, Newby DE,
tation surgery. Circulation. 2009;120:1189–94. Hayes PC. Prevalence of coronary artery calcification
33.
Williams K, Lewis JF, Davis G, Geiser in patients undergoing assessment for orthotopic liver
EA. Dobutamine stress echocardiography in transplantation. Liver Transpl. 2008;14:1725–31.
patients undergoing liver transplantation evaluation. 47. Plotkin JS, Johnson LB, Rustgi V, Kuo PC. CAD and
Transplantation. 2000;69:2354–6. liver transplantation: the state of the art. Liver Transpl.
34. Harinstein ME, Flaherty JD, Ansari AH, Robin J, 2000;6:S53–6.
Davidson CJ, Rossi JS, et al. Predictive value of dobu- 48. Warnaar N, Lisman T, Porte RJ. The two tales of
tamine stress echocardiography for coronary artery coagulation in liver transplantation. Curr Opin Organ
disease detection in liver transplant candidates. Am J Transplant. 2008;13:298–303.
Transplant. 2008;8:1523–8. 49. Lisman T, Bongers TN, Adelmeijer J, Janssen HL, de
35. Tsutsui J, Mukherjee S, Elhendy A, Xie F, Lyden Maat MP, de Groot PG, et al. Elevated levels of von
ER, O’Leary EO, et al. Value of dobutamine stress Willebrand factor in cirrhosis support platelet adhe-
myocardial contrast perfusion echocardiography in sion despite reduced functional capacity. Hepatology.
patients with advanced liver disease. Liver Transpl. 2006;44:53–61.
2006;12:592–9. 50. Kaplan M, Cimen S, Kut MS, Demirtas MM. Cardiac
36. Abraczinskas DR, Ookubo R, Grace ND, Groszmann operations for patients with chronic liver disease.
RJ, Bosch J, Garcia-Tsao G, et al. Propranolol for Heart Surg Forum. 2002;5:60–5.
the prevention of first esophageal variceal hem- 51. Hayashida N, Shoujima T, Teshima H, Yokokura Y,
orrhage: a lifetime commitment? Hepatology. Takagi K, Tomoeda H, et al. Clinical outcome after
2001;34:1096–102. cardiac operations in patients with cirrhosis. Ann
37. Berman DS, Abidov A, Kang X, Hayes SW, Friedman Thorac Surg. 2004;77:500–5.
JD, Sciammarella MG, et al. Prognostic validation of 52. Filsoufi F, Salzberg SP, Rahmanian PB, Schiano TD,
a 17-segment score derived from a 20-segment score Elsiesy H, Squire A, et al. Early and late outcome of
for myocardial perfusion SPECT interpretation. J cardiac surgery in patients with liver cirrhosis. Liver
Nucl Cardiol. 2004;11:414–23. Transpl. 2007;13:990–5.
38. Kryzhanovski VA, Beller GA. Usefulness of preop- 53. Klemperer JD, Ko W, Krieger KH, Connolly M,

erative noninvasive radionuclide testing for detecting Rosengart TK, Altorki NK, et al. Cardiac opera-
coronary artery disease in candidates for liver trans- tions in patients with cirrhosis. Ann Thorac Surg.
plantation. Am J Cardiol. 1997;79:986–8. 1998;65:85–7.
292 S. Shayan and A. M. De Wolf

54. An Y, Xiao YB, Zhong QJ. Open-heart surgery in 66. Bargehr J, Trejo-Gutierrez JF, Patel T, Rosser B,

patients with liver cirrhosis. Eur J Cardiothorac Surg. Aranda-Michel J, Yataco ML, et al. Preexisting atrial
2007;31:1094–8. fibrillation and cardiac complications after liver trans-
55. Bizouarn P, Ausseur A, Desseigne P, Le Teurnier Y, plantation. Liver Transpl. 2015;21:314–20.
Nougarede B, Train M, et al. Early and late outcome 67. Van Wagner LB, Serper M, Kang R, Levitsky J,

after elective cardiac surgery in patients with cirrho- Hohmann S, Abecassis M, et al. Factors associ-
sis. Ann Thorac Surg. 1999;67:1334–8. ated with major adverse cardiovascular events after
56. Axelrod D, Koffron A, DeWolf A, Baker A, Fryer J, liver transplantation among a national sample. Am J
Baker T, et al. Safety and efficacy of combined ortho- Transplant. 2016;16(9):2684–94.
topic liver transplantation and coronary artery bypass 68. Maron BJ. Hypertrophic cardiomyopathy: a system-
grafting. Liver Transpl. 2004;10:1386–90. atic review [review]. JAMA. 2002;287:1308–20.
57. Carr C, Desai J. OPCAB surgery in a cirrhotic hepa- 69.
Maraj S, Jacobs LE, Maraj R, Contreras R,
tocellular carcinoma patient awaiting liver transplant. Rerkpattanapipat P, Malik TA, et al. Inducible left
Ann Thorac Surg. 2004;78:1460–2. ventricular outflow tract gradient during dobutamine
58. Ben Ari A, Elinav E, Elami A, Matot I. Off-pump cor- stress echocardiography: an association with intraop-
onary artery bypass grafting in a patient with Child erative hypotension but not a contraindication to liver
class C liver cirrhosis awaiting liver transplantation. transplantation. Echocardiography. 2004;21:681–5.
Br J Anaesth. 2006;97:468–72. 70.
Rigopoulos AG, Panou F, Kremastinos DT,
59. Shaheen AA, Kaplan GG, Hubbard JN, Myers
Seggewiss H. Alcohol septal ablation in hypertrophic
RP. Morbidity and mortality following coronary obstructive cardiomyopathy [review]. Hell J Cardiol.
artery bypass graft surgery in patients with cirrhosis: a 2009;50:511–22.
population-based study. Liver Int. 2009;29:1141–51. 71. Paramesh AS, Fairchild RB, Quinn TM, Leya F,

60. Alper I, Ulukaya S, Demir F, Kilic M. Effects of George M, Van Thiel DH. Amelioration of hypertro-
cardiac valve dysfunction on perioperative man- phic cardiomyopathy using nonsurgical septal abla-
agement of liver transplantation. Transplant Proc. tion in a cirrhotic patient prior to liver transplantation.
2009;41:1722–6. Liver Transpl. 2005;11:236–8.
61. Pocar M, Passolunghi D, Moneta A, Donatelli
72. Hage FG, Bravo PE, Zoghbi GJ, Bynon JS, Aqel
F. Primary and redo valve replacement before and RA. Hypertrophic obstructive cardiomyopathy in
after liver transplantation. Eur J Cardiothorac Surg. liver transplant patients. Cardiol J. 2008;15:74–9.
2007;32:674–5. 73. Lim YC, Doblar DD, Frenette L, Fan PH, Poplawski
62.
Iino K, Tomita S, Yamaguchi S, Watanabe S, Nanda NC. Intraoperative transesophageal echo-
G. Successful aortic valve replacement using dilu- cardiography in orthotopic liver transplantation in
tional ultrafiltration during cardiopulmonary bypass a patient with hypertrophic cardiomyopathy. J Clin
in a patient with Child-Pugh class C cirrhosis. Interact Anesth. 1995;7:245–9.
Cardiovasc Thorac Surg. 2008;7:331–2. 74. Harley ID, Jones EF, Liu G, McCall PR, McNicol
63. Kalarickal P, Liu Q, Rathor R, Ishag S, Kerr T,
PL. Orthotopic liver transplantation in two patients
Kangrga I. Balloon aortic valvuloplasty as a bridge to with hypertrophic obstructive cardiomyopathy. Br J
liver transplantation in patients with severe aortic ste- Anaesth. 1996;77:675–7.
nosis: a case series. Transplant Proc. 2014;46:3492–5. 75. Cywinski JB, Argalious M, Marks TN, Parker

64. Eckhoff DE, Frenette L, Sellers MT, McGuire BM, BM. Dynamic left ventricular outflow tract obstruc-
Contreras JL, Bynon JS, et al. Combined cardiac tion in an orthotopic liver transplant recipient. Liver
surgery and liver transplantation. Liver Transpl. Transpl. 2005;11:692–5.
2001;7:60–1. 76. Chin JH, Kim YK, Choi DK, Shin WJ, Hwang

65. Kia L, Shah SJ, Wang E, Sharma D, Selvaraj S, Medina GS. Aggrevation of mitral regurgitation by calcium
C, et al. Role of pretransplant echocardiographic eval- administration in a patient with hypertrophic cardio-
uation in predicting outcomes following liver trans- myopathy during liver transplantation: a case report.
plantation. Am J Transplant. 2013;13:2395–401. Transplant Proc. 2009;41:1979–81.
Pulmonary Complications
of Liver Disease 24
Mercedes Susan Mandell and Masahiko Taniguchi

Keywords commonly in patients with liver disease than in


Respiratory failure · Hepato-pulmonary syn- the general population. These pulmonary
drome · Porto-pulmonary hypertension · defects can affect the perioperative manage-
Restrictive lung disease · Postoperative venti- ment of patients and may influence the decision
lation · Pulmonary function test to proceed with transplantation. This chapter
will present an overview of the changes in lung
mechanics and gas exchange that occur in
Introduction patients with liver disease and explore the more
common causes of pulmonary disease in cir-
Liver disease affects the function of all other rhotic patients.
organ systems and can be thought of as a sys- Liver disease is a systemic disease that
temic disease that results in multisystem organ results in multisystem organ failure as a princi-
failure as a principal cause of death. The lung is pal cause of death. Lung function is highly
particularly sensitive to changes in hepatic influenced by hepatic function and can be a pri-
function and respiratory failure is a common mary cause of early mortality. Previously the
complication of advanced liver disease. association between lung and liver disease was
Historically, physicians considered the associa- considered rare. But more recent studies have
tion between lung and liver disease to be rare. shown that symptoms such as hypoxemia at rest
But more recent studies have shown that symp- occur in at least 27–33% of liver transplant can-
toms such as hypoxemia at rest occur in at least didates. Hypoxemia is the end manifestation of
27–33% of liver transplant candidates [1]. a number of processes, some that occur more
Hypoxemia is caused by a wide variety of dis- commonly in patients with liver disease than in
eases. Some pulmonary diseases occur more the general population. These pulmonary
defects can affect the perioperative manage-
ment of patients and may influence the decision
M. S. Mandell, MD, PhD (*) to proceed with transplantation. This chapter
Department of Anesthesiology, University of
will present an overview of the changes in lung
Colorado, Aurora, CO, USA
e-mail: Susan.Mandell@UCDenver.edu mechanics and gas exchange that occur in
patients with liver disease and cover some of
M. Taniguchi, MD, FACS
Department of Surgery, Hokkaido University, the more common causes of pulmonary disease
Sappora, Japan in cirrhotic patients.

© Springer International Publishing AG, part of Springer Nature 2018 293


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_24
294 M. S. Mandell and M. Taniguchi

 ulmonary Function in Patients


P Table 24.1  Pulmonary function tests in liver disease
with Cirrhosis Pulmonary function Restrictive Obstructive
tests (spirometry) pattern pattern
Patients with liver disease have well-documented FVC Decrease Decrease
defects in respiratory mechanics, alveolar blood FEV1 Normal-­ Decrease
decrease
supply and in gas exchange at the alveolar sur-
FEV1/FVC ratio Normal Decrease
face [1, 2]. One or all of these important func-
TLC Decrease Normal or
tions can be affected. Thus, patients with cirrhosis increased
can suffer from hypoxemia even when there is no
Patients with liver disease usually have mixed obstructive
identifiable pulmonary disease process. and restrictive pattern of pulmonary function tests con-
Respiratory mechanics are commonly compro- ducted by Spirometry. Ascites and pleural effusion cause
mised by portal hypertension, anasarca ascites restrictive changes in pulmonary function tests as the
lungs cannot fully expand. This is reflected in the decrease
and an increase in the size of the abdominal
of total lung capacity and lung volumes and capacities that
organs. These can cause symptoms suggestive of make up the total lung volume. The forced expiratory vol-
restrictive lung disease. There are significant umes are either normal or slightly decreased. In contrast,
reductions in the chest wall motion, the normal some patients may demonstrate a predominant obstructive
pattern due to liver disease. In this case the lung volumes
underlying lung recoil and excursion of the dia-
increase while the expiratory flow rates decrease. The
phragm, caused by an increase in the size of the ratio between the forced expiratory volume in 1 s to the
abdominal organs, ascites and/or increase in the forced vital capacity (FEV1/FVC) helps determine if a
abdominal blood volume. The enlarged abdomi- restrictive or obstructive pattern predominates
nal volume pushes the diaphragm upwards and
holds the ribs in a more horizontal position Ascites and pleural effusion cause restrictive
thereby increasing the resting diameter of the changes in pulmonary function tests where the
chest wall [3]. The consequent reduction in lung lungs cannot fully expand. This is reflected in the
volumes and chest wall excursion limits the decrease of total lung capacity and lung volumes
expansion and elastic recoil of the lung. This in and capacities that make up the total lung volume.
turn reduces respiratory volumes, and especially The forced expiratory volumes are either normal or
the functional residual volume falls, while clos- slightly decreased. In contrast, some patients may
ing capacity increases [4]. demonstrate a predominant obstructive pattern due
All these changes reduce respiratory reserve to liver disease. In this case, the lung volumes
and place patients at risk of developing hypox- increase, while the expiratory flow rates decrease.
emia. Additional complications such as pleural The ratio between the forced expiratory volume in
effusions further impair the normal expansion 1 s and the forced vital capacity helps determine if
and elastic recoil of the lung and chest wall. a restrictive or obstructive pattern predominates.
Compression of the lower airways leads to signs The ratio is normal in a pure restrictive pattern but
and symptoms of small airway obstruction [2]. is reduced in an obstructive pattern.
These changes lead to a reduction of expiratory Defects in gas exchange produce an increase
flow volumes: the ratio between the volume that in the gradient between the alveolar and arterial
can be forcibly expired in 1 s as a percentage of concentration of oxygen (A-a gradient) in a large
the total forced vital capacity (FEV1/FVC) is number of patients with liver disease [3].
commonly reduced. A similar reduction is seen Hypoxemia (PaO2 < 80 mmHg) has been reported
in the forced expiratory flows in the small air- in up to one-third of transplant candidates and
ways (FEF25–50) [5]. The mixed restrictive-­ correlates with the severity of liver disease as
obstructive pattern observed in pulmonary measured by the Child-Pugh and the model for
function tests correlates with the severity of ill- end-stage liver disease (MELD) score [1]. There
ness and patients in child’s class B and those with are three well-recognized mechanisms that
ascites tend to have a greater impairment [5] explain the increase in A-a gradient in patients
(Table 24.1). with liver disease: an imbalance in match between
24  Pulmonary Complications of Liver Disease 295

alveolar ventilation and perfusion, true shunt DLCO in patients with cirrhosis (as the A-a gradi-
where there is perfusion without alveolar ventila- ent increases, the DLCO falls) suggests that mech-
tion and diffusion defects [6, 7]. anisms influencing the DLCO play an important
Patients with liver disease have a mismatch role in oxygen exchange [10]: several different
between alveolar ventilation and capillary perfu- hypotheses try to explain why the end capillary
sion. Compression of the lung tissue by organo- partial pressure of a gas would not be equal to its
megaly, ascites, and pleural effusion explains alveolar value [15]. One possible reason is a “dif-
some of the ventilation-to-perfusion mismatch. A fusion-perfusion defect” where the central stream
decrease in functional residual capacity along of red blood cells in dilated capillaries does not
with an increase in closing volume favors a drop have time to equilibrate with the alveolar oxygen
off in alveolar ventilation due to simple mechani- [16]. Elevated cardiac output associated with the
cal compression of the alveoli. This leads to a hyperdynamic state of hepatic cirrhosis may cause
mismatch between the ventilation and perfusion a rapid transit of blood through dilated alveolar
ratio. Evidence suggests that the ventilation-to-­ vessels. The transit time exceeds the time needed
perfusion imbalance could be due to changes in for the alveolar blood to fully equilibrate with the
the pulmonary microvascular tone [8]. This may alveolar oxygen content. Other theories propose a
occur even though general hypoxia would thickening of the capillary-alveolar interface [17]
increase in pulmonary resistance in cirrhotic or a decrease in capillary blood flow (despite an
patients with well compensated disease [9]. increase in central blood volume [10]. In general
Autonomic dysfunction may play a role in an increase of the A-a gradient and hypoxemia is
ventilation-­to-perfusion mismatch, and hypox- usually caused by multiple mechanisms in patients
emia is more commonly seen in patients with with liver disease (Fig. 24.1).
greater severity of illness [10].
In many patients with liver disease intrapulmo-
nary shunts substantially contribute to an increase  iseases of the Hepatopulmonary
D
in the A-a gradient and hypoxemia [11]. Harmonic Axis
imaging by echocardiography has revealed the
presence of intrapulmonary shunting in up to 80% Certain pulmonary diseases have a higher than
of patients assessed for liver transplantation [12]. expected incidence in patients with liver disease.
Intrapulmonary shunting has also been observed Some of these are acquired while others have a
by the multiple inert gas elimination technique clear genetic pattern of inheritance. The acquired
[13]. Shunting causes blood to completely bypass diseases tend to fall into one of three categories:
ventilating units and empty into the arterial sys- pulmonary vascular diseases, parenchymal dis-
tem causing venous admixture. Shunting that ease, and diseases of the pleural space. Although
results in clinical hypoxemia is considered diag- inherited diseases can be roughly categorized in a
nostic for hepatopulmonary syndrome (HPS). similar manner, their classification is often not as
Liver disease also affects the traversing of gas clear as the principal disease process tends to
across lung tissue and alveolar membrane result- affect multiple aspects of lung function.
ing in a notable decrease in the diffusing capacity
of the lung for carbon monoxide (DLCO) [14].
The DLCO test is a single-breath pulmonary func- Inherited Diseases
tion test that assesses two components: the rate of
gas exchange across the alveolar membrane and Cystic Fibrosis and Alpha-1
the binding of carbon monoxide to the hemoglobin Antitrypsin Deficiency
molecule. The latter is a function of the rate of
binding of carbon monoxide to hemoglobin and The most common inherited diseases that affect
the alveolar capillary hemoglobin volume. The both the lung and liver are α1-antitrypsin and cys-
inverse correlation between the A-a gradient and tic fibrosis [18]. Both are autosomal recessive dis-
296 M. S. Mandell and M. Taniguchi

Fig. 24.1 Three Autonomic Ventilation-Perfusion Lung Compression


principle causes of Dysfunction Defect
hypoxemia and a
widened A-a gradient in
patients with liver
disease. All three
mechanisms may be Intrapulmonary
Shunts
present in the same
patient

Diffusion-
Hypoxemia
Perfusion
Defect

Diffusion
Defect Shunts

Alveolar thickening Pleural, Shunts

orders. Cystic fibrosis occurs in 1 in 3000 births common in cystic fibrosis but often not clinically
while severe α1-antitrypsin deficiency is present in significant [25], and less than one-third of patients
1 in 3500 births [19, 20]. The majority of patients with cystic fibrosis develop detectable hepatobili-
with cystic fibrosis have some degree of hepatobi- ary disease [26, 27]. Focal biliary cirrhosis is the
liary disease during their lifetime. In contrast, most common lesion in these patients [25].
liver disease is rare in α1-antitrypsin deficiency. Severe liver disease is associated with only a few
A mutant allele for the gene of cystic fibrosis of the more than 1500 known mutations in the
membrane conductance regulator (CFTR) causes CFTR gene. Although clinically significant dis-
defects in in the transport of ions including chlo- ease develops in 5–7% of patients with focal bili-
ride and bicarbonate on apical epithelial surfaces ary cirrhosis [27, 28], complications of portal
[21]. Quantiative and/or qualitative reduction of hypertension are rare [27, 28]. Currently the only
CFTR result in diverse presentations and severity available treatment for liver disease due to cystic
of disease and multiple organ systems can be fibrosis is ursodeoxycholic acid [29, 31].
affected. Patients with cystic fibrosis have a pre- Most patients with hepatobiliary disease due
dicted mean survival of 37.4 years [22]. Nearly to cystic fibrosis also have lung disease [30]. The
90% of patients are diagnosed under the age of ten lower airways become obstructed by viscous
years with symptoms of exocrine pancreatic secretions and patients experience multiple epi-
insufficiency and pulmonary disease [23]. Early sodes of infection [32]. This leads to parenchy-
presentation is more common in males than mal destruction, severe obstructive disease with
females [21]. A second group of patients are pri- ­diffuse loss of lung volumes, and finally respira-
marily diagnosed with cystic fibrosis as adults. tory failure. The majority of patients have chronic
The latter patients tend to have milder lung dis- lower airway infections most commonly with
ease and predominant pancreatic insufficiency Staphylococcus aureus, Hemophilus influenzae,
[24]. Patients who are diagnosed in adulthood and Pseudomonas aeruginosa. There are no clear
tend to have long survival and are less likely to pulmonary criteria that predict posttransplant
require lung transplantation. survival in these patients and chronic coloniza-
Elevations in the serum levels of the amino- tion and infection with bacteria does not seem to
transferases and gamma glutamyl transferase are affect transplant outcome.
24  Pulmonary Complications of Liver Disease 297

However in general survival after liver trans- ciency. Liver transplantation is curative since the
plantation is worse in patients with cystic fibrosis new liver synthesizes normal α1-antitrypsin and
according to data from the United Network for patients have an excellent outcome with similar
Organ Sharing [33]. Propensity score matching 1-, 3-, and 5-year survivals as those reported for
of patients (average age 14 ± 3.1 years) with and all liver transplants in the United States [40].
without cystic fibrosis identified a mortality haz-
ard of 3 following liver transplantation. The peri-
operative morbidity and mortality are mainly Autoimmune Diseases
related to lung disease [34, 35]. There is not as
much experience with liver transplantation in Autoimmune diseases often affect both the lung
adult patients with cystic fibrosis; however, and liver [46]. For example, one quarter of
investigators have only reported a 40% survival patients with rheumatoid arthritis have lung dis-
rate at 5 years due to a variety of causes [36]. ease including chronic pleural effusions and
Alpha-1 antitrypsin deficiency is an autosomal interstitial pneumonitis, pulmonary fibrosis, and
recessive disease where each allele contributes pulmonary hypertension. These patients also
50% of the circulating enzyme. The normal gene have an increased incidence of autoimmune hep-
product is designated as PiM [20]. Defects in α1-­ atitis and nodular regenerative hyperplasia. Liver
antitrypsin are the most common metabolic cause and lung disease also occur in patients with der-
of liver disease in neonates and children [37]. matomyositis, scleroderma, and systemic lupus
Adults are usually affected in the fifth decade erythematosus. Furthermore, drugs used to con-
[38]. The genetic variants that are associated with trol symptoms of autoimmune disease can inde-
lung or liver disease are PiS (expressing 50–40% pendently cause liver disease. Cases of acute and
α1-antitrypsin) and PiZ (expressing 10–20% α1-­ chronic hepatic injury have been described with
antitrypsin). The most common deficiency types the use of most anti-inflammatory drugs includ-
that cause disease are PiSS, PiSZ, and PiZZ [39]. ing high dose nonsteroidal anti-inflammatory
Alpha-1 antitrypsin is a serine protease inhibi- drugs, methotrexate [47], and tumor necrosis fac-
tor of neutrophil elastase [40]. Failure to inhibit tors inhibitors [48].
elastase causes early onset pan-lobar emphy- Primary biliary cirrhosis is similar to other
sema. The accumulation of α1-antitrypsin poly- autoimmune diseases in that a sibling has a
mers within the endoplasmic reticulum of 10.5% relative risk of developing the disease
hepatocytes causes liver disease when the S and [49], and it is more common in females than in
Z gene are co-inherited [41, 42]. These two genes males. The disease is characterized by autoanti-
code for errors in the steps that transport α1-­ bodies to mitochondrial antigens [50]. It causes
antitrypsin out of the hepatocyte. Approximately progressive destruction of small and medium
37% of asymptomatic PiZZ patients have cirrho- intrahepatic bile ducts and can lead to cirrhosis.
sis at the time of death [43]. Other factors such as There is an association between primary biliary
male gender and obesity increase the risk of cirrhosis and other autoimmune diseases.
hepatic disease [44]. Thyroiditis, Sjogren’s syndrome, scleroderma,
The common environmental and genetic fac- and rheumatoid arthritis occur more frequently
tors that predispose patients to develop lung and/ in patients with primary biliary cirrhosis [51]. In
or liver disease are unknown. Patients with liver fact, a crossover syndrome between primary
disease due to α1-antitrypsin have an increased biliary cirrhosis and autoimmune hepatitis has
incidence of cholangiocarcinoma and hepato- been reported [52]. The pulmonary manifesta-
cholangiocarcinoma. Patients with emphysema tions can be complex. There is an increased
due to α1-antitrypsin deficiency have been treated incidence of lymphocytic interstitial pneumo-
with intravenous α1-antitrypsin augmentation nia, intrapulmonary granulomas, bronchiolitis
therapy [45]; however, there is no medical treat- obliterans, obstructive airway disease and pul-
ment for liver disease due to α1-antitrypsin defi- monary hypertension [53]. The high incidence
298 M. S. Mandell and M. Taniguchi

and diverse presentation of lung disease in venous malformation [62] include hemoptysis,
patients with autoimmunity indicates that a spontaneous hemothorax and severe pulmonary
careful preoperative evaluation of pulmonary hypertension [63]. Pulmonary hypertension
function is warranted in these patients. occurs in patients with liver involvement second-
Disease recurrence is estimated at 8–12% in ary to high cardiac output. Some patients with
the first year and 36–68% at 5 years following hereditary hemorrhagic telangiectasia develop a
transplanation [54]. Overall 5-year post-­plexogenic pulmonary arteriopathy that is identi-
transplantationsurvival of patients with autoim- cal to portopulmonary hypertension (POPH). An
mune hepatitis is 75%, similar to patients with overall median survival of 87% has been reported
alcoholic liver disease [55]. However, this is sig- at 47 months following transplantation.
nificantly worse than the 5-year survival of 83% Antiangiogenic drugs such as Bevacizumab have
reported in a multicenter study for patients with been successfully used to control disease pro-
primary biliary cirrhosis [55]. Young patients gression [64]. Patient and graft survival follow-
with autoimmune hepatitis had moderately better ing liver transplantation reported in 40 patients
survival than patients over the age of 50. from the European Registry was greater than
Infectious complications were a major cause of 80% [65].
mortality, and pulmonary disease did not have a
significant effect on overall outcome. Even
though patients with primary biliary cirrhosis had Acquired Diseases
better survival, approximately 9–35% of patients
will develop recurrent disease [56]. Diseases of the Pleural Space

Hepatic hydrothorax is the most common


Hereditary Hemorrhagic acquired pulmonary complication of cirrhosis,
Telangiectasia occurring in 4–6% of all patients [66]. The diag-
nosis is made when there is a pleural effusion
Hereditary hemorrhagic telangiectasia (Osler– (>500 mL) with no evidence of primary lung or
Weber–Rendu disease) is a group of autosomal heart disease. In the majority of patients the effu-
dominant disorders characterized by the pres- sion is right-sided and usually occurs in patients
ence of abnormal arteriovenous malformations with ascites. Congenital fenestrations in the ten-
caused by genetic mutations in the transforming dinous part of the diaphragm allow the passage of
growth factors beta signaling pathway [43, 57]. ascites fluid. Elevated intra-abdominal pressure
A number of organs systems can be involved combined with cyclic negative intrathoracic pres-
aside from the lung and liver, such as skin, brain, sure will cause a unidirectional flow of ascites
and the gastrointestinal tract. The liver is affected into the pleural space. Hepatic hydrothorax rarely
in up to 84% of patients [58]; however, only occurs on the left because there are fewer fenes-
5–8% have symptomatic liver disease [59]. trations as the hemidiaphragm is thicker and
High-output cardiac failure is the most common more muscular.
clinical presentation and is caused by significant Patients with hepatic hydrothorax have an
shunting through arteriovenous malformations increased risk of developing spontaneous bacte-
in the liver [60]. rial empyema [67]. The effusion is initially
Approximately one-third of affected patients treated in the same way as ascites, with diuretics
experience hypoxemia due to pulmonary arterio- and by restricting sodium intake. If these treat-
venous malformations [61]. There is an increased ments fail, thoracocentesis is an option but has
incidence of arterial embolic complications been associated with an increase risk of infec-
including stroke and brain abscess due to direct tious complications [66]. The application of bio-
arteriovenous connections. Complications in logic glue or sclerosing agents to seal
15–45% of patients who have pulmonary arterio- diaphragmatic defects using video-assisted tho-
24  Pulmonary Complications of Liver Disease 299

racoscopic surgery has a high rate of success a normal pulmonary capillary wedge pressure
[68]. Transjugular intrahepatic portosystemic (<15 mmHg), a pulmonary vascular resistance
shunting has also been used successfully to con- greater than 240 dynes s cm−5 and the presence of
trol hepatic hydrothorax [69]. A large effusion portal hypertension [77]. Most cases are initially
may impede positive pressure ventilation during found on screening echocardiography. Systolic
liver transplant surgery and may require insertion flow through a regurgitant tricuspid valve corre-
of a chest tube for drainage at the beginning of lates with the systolic pressure gradient between
surgery. However intercostal vessels may be the right ventricle and the right atrium. When
engorged in cirrhosis and increase the risk of right atrial pressure is known (or estimated) right
bleeding if injured during chest tube placement. ventricular systolic pressure (RVSP) can be cal-
culated. RVSP is equal to the systolic pulmonary
pressure in the absence of pulmonary stenosis.
Parenchymal Diseases RVSP great than 50 mmHg is associated with
97% sensitivity but only 77% specificity for the
A large number of infectious and immune-­ diagnosis of POPH by right heart catheterization
mediated diseases affect the lung parenchyma. [78]. It has however been criticized that estimates
Patients with liver disease have changes in their of pulmonary pressures from echocardiography
immune system that makes them susceptible to lack precision and therefore right heart catheter-
pulmonary infections and complications. With ization is needed to confirm the diagnosis and
hepatic cirrhosis, there is an increased incidence assess the severity of disease [79]. All patients
of pulmonary infections with bacterial, fungal, placed on the waiting list for liver transplantation
viral, and mycobacterial species; additionally, should be screened for POPH using a resting
complications such as bronchitis obliterans are echocardiogram and follow-up right heart cathe-
more common than in the general population terization if the estimated RSVP ≥50 mmHg
[70]. because there are no diagnostic clinical symp-
Immune-mediated lung injury has been toms of POPH. Fatigue and dyspnea on exertion
reported in patients with hepatitis C and in are the most common complaints and are not eas-
patients receiving interferon antiviral therapy. ily distinguished from general symptoms of liver
Fibrosing alveolitis has been reported in patients disease. Patients with syncope or chest pain usu-
with hepatitis C [71]. This serious complication ally have severe POPH and right heart failure, but
is probably due to mixed cryoglobulinemia the absence of these symptoms does not preclude
caused by an innate immune response to infec- a diagnosis of POPH.
tion with hepatitis C. Similar lung pathology has The etiology of POPH is still uncertain even
been observed in patients receiving treatment though there has been considerable research in
with sirolimus [72] and pegylated interferon [73]. this area in human populations and animal mod-
els. No specific molecular signallling pathway or
genetic mutation has been confirmed as a cause
 ortopulmonary Hypertension
P of POPH. Investigators think the interaction
between genetic predisposition and hyperdy-
Pulmonary hypertension (POPH) is a rare but namic circulation causes disease [80]. There are
severe and potentially life-threatening disease significant changes of the function of vascular
that affects up to 6% of patients with portal endothelium within the pulmonary circulation,
hypertension waiting for transplantation [74, 75], and these changes lead to progressive vasocon-
markedly higher than the 1.1–2.4 cases per mil- striction, inflammation, angiogenesis and in situ
lion reported by multiple registries form the US, thrombosis. If left untreated, most patients will
France, China and the United Kingdom [76]. progress to right heart failure and death [81].
Diagnostic criteria include a mean pulmonary Three molecular end signals in the lung vascular
artery pressure greater than 25 mmHg at rest with endothelium are affected in POPH patients: the
300 M. S. Mandell and M. Taniguchi

vasodilatory signaling of nitric oxideand prosta- the potentially life-saving benefit of a transplant
cyclin is decreased and the vasoconstrictor sig- for the patients with the possible “waist” of a
naling of endothelin is increased [82]. Medical scarce graft in case of perioperataive death.
therapy is aimed at restoring a normal balance of
these mediators. Treatment is similar to patients
with other causes of pulmonary hypertension and Hepatopulmonary Syndrome
relies on the use of endothelin receptor antago-
nists, prostacyclin derivatives, and drugs that Hypoxemia has been reported 25% in patients
increase the amount of nitric oxide [83]. with portal hypertension [91]. HPS is the most
Without any intervention, half of the patients common form of pulmonary vascular disease in
with POPH die within 1 year of their diagnosis patients with cirrhosis. Diagnostic criteria vary
[84, 85]. With medical treatment, approximately and include a room-air PaO2 between 70 and
45% of patients are alive at 5 years and 67% of 80 mmHg, the presence of intrapulmonary shunt-
patients who received both medical therapy and ing, and a diagnosis of portal hypertension with
liver transplantation were alive at 5 years. This is or without cirrhosis. The effects of patients posi-
in sharp contrast to patients who were trans- tioning during measurement of arterial oxygen-
planted without medical therapy; only 25% of ation affects diagnosis since blood is preferentially
these patients survived 5 years. Overall surgical perfused through larger shunts in the bases of the
survival is affected by the severity of POPH; lung while upright [92]. In contrast lying supine
mean pulmonary artery pressures over 35 mmHg improves the distribution of blood in the pulmo-
are associated with an increased risk of mortality. nary bed and consequently oxygenation [91, 92].
Similarly right ventricular performance also pre- This seemingly paradoxical improvement of oxy-
dicts outcome, and preserved right heart function genation with supine position is called orthede-
correlates with a better outcome [86]. oxia. Some investigators inlcude the
Patients with POPH in the United States alveolar-arterial oxygen gradient as a diagnostic
receive additional MELD points for the allocation criteria for hypoxemia/HPS as it is more sensitive
of organs if their mean pulmonary artery pressure and adjusts for changes in arterial carbon dioxide
is 35 mmHg or less with or without medical treat- (PaCO2). The PaCO2 is often decreased in
ment [87]. The priority given to patients with patients with liver disease due to hyperventila-
POPH is based upon better outcomes in patients tion. Approximately 30% of patients with
with lower pulmonary artery pressures [88] advanced liver disease are diagnosed with HPS
(Goldberg). The long-term outcome of POPH using these criteria.
patients is still confusing. Some patients may HPS can be divided into mild, moderate,
have a complete resolution of the disease, while severe, and very severe based upon the degree of
others experience a worsening of their disease hypoxemia (Table 24.2). This classification is
[89, 90]. Still others remain stable but do not
show any significant improvement, and in rare
cases, there is new onset POPH following trans- Table 24.2  The classification of hepatopulmonary syn-
drome into severity of disease
plantation. To date, there are no patient character-
istics known that predict long-­term outcome. Severity of disease PaO2 mmHg A-a gradient
Severe pulmonary hypertension for example Mild ≥80 ≥15
with mean pulmonary artery pressure over Moderate ≥60 to <80 ≥15
45 mmHg should prompt a multi-disciplinary Severe ≥50 to <60 ≥15
discussion about eligbility for transplant prior to Very severe <50 ≥15
listing, possible medical optimization and peri- There are four classifications of HPS based upon the arte-
rial oxygen partial pressure. All severities of disease
operative management. This discussion should require that the A-a gradient is greater than 15. A-a
include all stakeholders (anesthesiologists, sur- alveolar-­arterial gradient; HPS hepatopulmonary
geons, hepatologist and intensivists) and weigh syndrome
24  Pulmonary Complications of Liver Disease 301

important as it correlates with patient survival brate with the alveolar oxygen [6]. Furthermore,
and transplant outcome [93]. Standard arterial there is direct arteriovenous admixture due to the
oxygen measurements for diagnosis should be presence of anatomic shunts. Ventilation-­
performed with the patient in the upright perfusion mismatch and shunts explain the pres-
position. ence of orthodeoxia [95]. Patients respond to an
Pulmonary function tests are not diagnostic for increase in inspired oxygen concentration when
HPS. The only consistent finding is a low diffus- mismatching predominates as a cause of hypox-
ing capacity but this has low specificity. emia. A diffusion-perfusion defect probably dom-
Intrapulmonary shunts are the hallmark of HPS inates in severe cases of HPS and is made worse
and are identified by one of two methods [93]. by the concomitant increase in cardiac output that
Contrast-enhanced transthoracic echocardiograph further decreases capillary transit time and there-
with agitated saline is the most commonly used fore the time available for oxygen uptake.
method. Microbubbles from the saline appear in A selective increase in the pulmonary produc-
the left heart in approximately 3–6 beats follow- tion of nitric oxide is one of the key pathological
ing the opacification of the right heart. This distin- changes underlying the development of HPS
guishes HPS from right to left intracardiac shunts [96]; however, HPS does not appear to be solely
where bubbles are observed in the left heart within due to nitric oxide overproduction as inhibitors of
1–2 beats following opacification of the right nitric oxide do not entirely reverse hypoxemia
heart. Echocardiography cannot estimate the [97]. Additionally to this and possibly other
severity of disease but is less invasive than the unknown mechanisms, an increase in endothelin
injection of technetium-labeled macroaggregated B type receptors in the pulmonary circulation of
albumin particles. With this technique, the amount patients with HPS further causes pulmonary vas-
of radiolabel technetium-­ labeled albumin that cular vasodilation and hypoxia [98].
accumulates over the brain allows an estimate of Currently, the only definitive treatment for
the severity of disease. However, technetium HPS is liver transplantation. The median survival
scanning cannot determine the site of shunting in patients with HPS without a transplant is only
and therefore cannot distinguish intracardiac 24 months with a 5-year survival of 23% [99].
defects from intrapulmonary shunts. Patients with similar characteristics but without
The principal defects in HPS are an increase in HPS had a median survival of 87 months, and
the number of pulmonary precapillary and capil- 63% were alive at 5 years [100]. The outcome is
lary vessels in combination with vasodilation. worse with a PaO2 less than 50 mmHg at the time
Investigators have also identified anomalous pleu- of diagnosis or a macroaggregated albumin shunt
ral, pulmonary, and portopulmonary arteriovenous fraction greater than 20% [101]. The cause of
connections. In addition, patients with HPS have a death is usually multifactorial with complications
reduction of pulmonary vascular tone and impaired due to liver disease predominating. There are a
pulmonary vasoconstriction in response to hypox- few reports of improved hypoxemia after tran-
emia [94]. These latter findings suggest the pres- sjugular intrahepatic portosystemic shunting
ence of autonomic dysfunction of the pulmonary [102] or after cavoplasty in patients with HPS
circulation in patients with HPS, and it is possible due to Budd-Chiari syndrome [103].
that some of the severity of disease is related to the Patient with HPS have a high risk of periopera-
degree of autonomic dysfunction. tive death [104]. Algorithms for acute periopera-
Patients with HPS are hypoxemic due to three tive hypoxemia have been described [104].
causes: There is a ventilation-to-perfusion mis- Manoevers include, Trendelnberg position, inhaled
match due to a selective increase in pulmonary epoprostenol or nitric oxide, methylene blue and
blood flow in areas of low ventilation [94]. embolization of pulmonary shunts or extracorpo-
Investigators also think that a diffusion-perfusion real oxgyenation [105]. Up to 21% of HPS patients
defect is caused by blood in the center of enlarged can suffer severe intraoperative hypoxemia with a
vessels that does not have adequate time to equili- 45% chance of mortality [104, 106]. A PaO2 of
302 M. S. Mandell and M. Taniguchi

less than 50 mmHg is associated with at least a 3. Mohamed R, Freeman JW, Guest PJ, et al. Pulmonary
gas exchange abnormalities in liver transplant candi-
29% perioperative mortality [105]. Life-
dates. Liver Transpl. 2002;8:802–8.
threatening decrease in oxygenation can occur 4. Yao EH, Kong BC, Hsue GL, et al. Pulmonary
anytime during surgery and in the early postopera- function changes in cirrhosis of the liver. Am J
tive period. Due to the increased risk of early Gastroenterol. 1987;82:352–4.
5. Yigit IP, Hacievliyagil SS, Seckin Y, et al. The rela-
death, patients with HPS receive additional MELD
tionship between severity of liver cirrhosis and pul-
points in the United States when the PaO2 is less monary function tests. Dig Dis Sci. 2008;53:1951–6.
than 50 mmHg. The amount of additional MELD 6. Martínez GP, Barberà JA, Visa J, et al.
points given for additional priority are under Hepatopulmonary syndrome in candidates for liver
transplantation. J Hepatol. 2001;34:651–7.
debate as investigators suggest there is no associa-
7. Martínez-Palli G, Gómez FP, Barberà JA, et al.
tion between oxygenation and waitlist mortality Sustained low diffusing capacity in hepatopulmo-
until the PaO2 reaches 44 mmHg [106]. nary syndrome after liver transplantation. World J
Gastroenterol. 2006;12:5878–83.
8. Castaing Y, Manier G. Hemodynamic disturbances
and VA/Q matching in hypoxemic cirrhotic patients.
Summary Chest. 1989;96:1064–9.
9. Naeije R, Halleman R, Mols P, Melot C. Hypoxic
An intricate link between pulmonary and liver pulmonary vasoconstriction in liver cirrhosis. Chest.
1981;80:570–4.
function exists, and abnormalities of the circula-
10. Degano B, Mittaine M, Guénard H, et al. Nitric
tion and neurohormonal balance of patients with oxide and carbon monoxide lung transfer in patients
liver disease cause lung disease. Some diseases with advanced liver cirrhosis. J Appl Physiol.
such as hydrothorax and pneumonitis are acquired 2009;107:139–43.
11. Andrivet P, Cadranel J, Housset B, et al. Mechanisms
and result from liver dysfunction. Other diseases
of impaired arterial oxygenation in patients with
such as cystic fibrosis and α1-antitrypsin are liver cirrhosis and severe respiratory insufficiency.
genetically based, and the coexisting lung injury Effects of indomethacin. Chest. 1993;103:500–7.
is part of the wider disease process. Diseases such 12. Kim BJ, Lee SC, Park SW, et al. Characteristics
and prevalence of intrapulmonary shunt detected
as POPH and HPS are acquired but probably have
by contrast echocardiography with harmonic imag-
a genetic predisposition. The wide variety of lung ing in liver transplant candidates. Am J Cardiol.
diseases associated with liver dysfunction makes 2004;94:525–8.
the preoperative assessment, selection, and peri- 13. Rodríguez-Roisin R, Krowka MJ, Hervé P, et al.
Pulmonary-hepatic vascular disorders (PHD). Eur
operative management of transplant recipients
Respir J. 2004;24:861–80.
challenging for anesthesiologists and intensivists. 14. Hourani JM, Bellamy PE, Tashkin DP, et al.
As newer medical therapies continue to emerge, Pulmonary dysfunction in advanced liver disease:
they will change the outcomes of patients with frequent occurrence of an abnormal diffusing capac-
ity. Am J Med. 1991;90:693–700.
combined pulmonary and liver disease. Thus, the
15. Wagner PD, Rodriguez-Roisin R. Clinical advances
perioperative care of the liver transplant patient is in pulmonary gas exchange. Am Rev Respir Dis.
a work in progress that will improve through 1991;143:883–8.
advances of the scientific knowledge that under- 16. Rodríguez-Roisin R, Krowka MJ. Hepatopulmonary
syndrome—a liver-induced lung vascular disorder.
lies this unique specialty practice.
N Engl J Med. 2008;358:2378–87.
17. Matsubara O, Nakamura T, Uehara T, Kasuga
T. Histometrical investigation of the pulmo-
nary artery in severe hepatic disease. J Pathol.
References 1984;143:31–7.
18. Moyer K, Balistreri W. Hepatobiliary disease in
1. Przybyłowski T, Krenke R, Fangrat A, et al. Gas patients with cystic fibrosis. Curr Opin Gastroenterol.
exchange abnormalities in patients listed for liver 2009;25:272–8.
transplantation. J Physiol Pharmacol. 2006;57(Suppl 19. Rowe SM, Miller S, Sorscher EJ. Cystic fibrosis. N
4):313–23. Engl J Med. 2005;352:1992–2001.
2. Agustí AG, Roca J, Bosch J, Rodriguez-Roisin 20. Fairbanks KD, Tavill AS. Liver disease in
R. The lung in patients with cirrhosis. J Hepatol. alpha 1-antitrypsin deficiency: a review. Am J
1990;10:251–7. Gastroenterol. 2008;103:2136–41.
24  Pulmonary Complications of Liver Disease 303

21. Elborn JS. Cystic fibrosis. Lancet. 2016;388:2519– 39. Chappell S, Hadzic N, Stockley R, et al. A poly-
31. https://doi.org/10.1016/S0140-6736(16)00576- morphism of the alpha1-antitrypsin gene repre-
6. [Epub ahead of print]. sents a risk factor for liver disease. Hepatology.
22. Nick JA, Chacon CS, Brayshaw SJ, et al. Effects of 2008;47:127–32.
gender and age of diagnosis on disease progression 40. Stoller JA, Aboussouan A. A review of α1 anti-
in long-term cystic fibrosis survivors. Am J Respir trypsin deficiency. Am J Respir Crit Care Med.
Crit Care Med. 2010;182(5):614–26. 2012;185:246–59.
23. Farrell PM, Rosensteinn BJ, White TB, et al. 41. Lindblad D, Blomenkamp K, Teckman J. Alpha-1-­
Guidelines for diagnosis of cystic fibrosis in newborns antitrypsin mutant Z protein content in individual
through older adults: Cystic Fibrosis Foundation hepatocytes correlates with cell death in a mouse
consensus report. J Pediatr. 2008;153:S4–14. model. Hepatology. 2007;46:1228–35.
24. Rodman DM, Polis JM, Heltshe SL, et al. Late diag- 42. Mahadeva R, Chang WS, Dafforn TR, et al.
nosis defines a unique population of long-term sur- Heteropolymerization of S, I, and Z alpha1-­
vivors of cystic fibrosis. Am J Respir Crit Care Med. antitrypsin and liver cirrhosis. J Clin Invest.
2005;171(6):621. 1999;103:999–1006.
25. Davis PB. Cystic fibrosis since 1938. Am J Respir 43. Hinds R, Hadchouel A, Shanmugham NP,
Crit Care Med. 2006;173:475–82. Al-Hussaini A, et al. Variable degree of liver
26. Wilschanski M. Patterns of gastrointestinal dis- involvement in siblings with PiZZ alpha-1-­
ease associated with mutations of CFTR. Curr antitrypsin deficiency-­related liver disease. J Pediatr
Gastroenterol Rep. 2008;10:316–23. Gastroenterol Nutr. 2006;43:136–8.
27. Feranchak AP, Sokol RJ. Cholangiocyte biology 44. Bowlus CL, Willner I, Zern MA, et al. Factors asso-
and cystic fibrosis liver disease. Semin Liver Dis. ciated with advanced liver disease in adults with
2001;21:471–88. alpha1-antitrypsin deficiency. Clin Gastroenterol
28. Lindblad A, Glaumann H, Strandvik B. A two-year Hepatol. 2005;3:390–6.
prospective study of the effect of ursodeoxycholic 45. Tonelli AR, Rouhani F, Li N, et al. Alpha-1-­
acid on urinary bile acid excretion and liver mor- antitrypsin augmentation therapy in deficient indi-
phology in cystic fibrosis-associated liver disease. viduals enrolled in the Alpha-1 Foundation DNA
Hepatology. 1998;27:166–74. and Tissue Bank. Int J Chron Obstruct Pulmon Dis.
29. Desmond CP, Wilson J, Bailey M, et al. The benign 2009;4:443–52.
course of liver disease in adults with cystic fibrosis 46. Meyer CA, White CS, Sherman KE. Diseases of the hep-
and the effect of ursodeoxycholic acid. Liver Int. atopulmonary axis. Radiographics. 2000;20:687–98.
2007;27:1402–8. 47. Curtis JR, Beukelman T, Onofrei A, et al. Elevated
30. Gooding I, Dondos V, Gyi KM, et al. Variceal hemor- liver enzyme tests among patients with rheuma-
rhage and cystic fibrosis: outcomes and implications for toid arthritis or psoriatic arthritis treated with
liver transplantation. Liver Transpl. 2005;11:1522–6. methotrexate and/or leflunomide. Ann Rheum Dis.
31. Siano M, De Gregorio F, Boggia B, et al. 2010;69:43–7.
Ursodeoxycholic acid treatment in patients with cys- 48. Sokolove J, Strand V, Greenberg JD, et al. Risk of
tic fibrosis at risk for liver disease. Dig Liver Dis. elevated liver enzymes associated with TNF inhibi-
2010;42:428–31. tor utilisation in patients with rheumatoid arthritis.
32. Lipuma JJ. The changing microbial epidemiology in Ann Rheum Dis. 2010;69(9):1612–7.
cystic fibrosis. Clin Microbiol Rev. 2010;23:299–323. 49. Jones DE. Pathogenesis of primary biliary cirrhosis.
33. Black SM, Woodley FW, Tumin D, et al. Cystic J Hepatol. 2003;39:639–48.
fibrosis associated with worse survival after liver 50. Kaplan MM. Primary biliary cirrhosis. N Engl J
transplantation. Dig Dis Sci. 2016;61:1178–85. Med. 1996;335:1570–80.
34. Fridell JA, Bond GJ, Mazariegos GV, et al. Liver 51. Liu B, Zhang FC, Zhang ZL, et al. Interstitial lung
transplantation in children with cystic fibrosis: a disease and Sjögren’s syndrome in primary bili-
long-term longitudinal review of a single center’s ary cirrhosis: a causal or casual association? Clin
experience. J Pediatr Surg. 2003;38:1152–6. Rheumatol. 2008;27:1299–306.
35. Molmenti EP, Squires RH, Nagata D, et al. Liver 52. Neuhauser M, Bjornsson E, Treeprasertsuk S, et al.
transplantation for cholestasis associated with Autoimmune hepatitis-PBC overlap syndrome: a
cystic fibrosis in the pediatric population. Pediatr simplified scoring system may assist in the diagno-
Transplant. 2003;7:93–7. sis. Am J Gastroenterol. 2010;105:345–53.
36. Ikegami T, Sanchez EQ, Uemura T, et al. Liver trans- 53. Shen M, Zhang F, Zhang X. Primary biliary cirrho-
plantation for cystic fibrosis in adults. Surg Today. sis complicated with interstitial lung disease: a pro-
2008;38:26–9. spective study in 178 patients. J Clin Gastroenterol.
37. Kelly E, Greene CM, Carroll TP, et al. Alpha-1 anti- 2009;43:676–9.
trypsin deficiency. Respir Med. 2010;104:763–72. 54. Kerkar N, Yanni G. “De Novo” and “recurrent” auto-
38. Topic A, Alempijevic T, Milutinovic AS, Kovacevic immune hepatitis after liver transplantation: a com-
N. Alpha-1-antitrypsin phenotypes in adult liver dis- prehensive review. J Autoimmun. 2016;16:17–24.
ease patients. Ups J Med Sci. 2009;114:228–34. https://doi.org/10.1016/j.jaut.2015.08.017.
304 M. S. Mandell and M. Taniguchi

55. Schramm C, Bubenheim M, Adam R, et al. European 70. Almaslmani M, Derbala MF, Albozom I, et al.
liver intestine transplant association. Primary liver Bronchiolitis obliterans organizing pneumonia
transplantation for autoimmune hepatitis: a com- associated with Pneumocystis jiroveci infection in
parative analysis of the European Liver Transplant orthotopic liver transplantation. Transpl Infect Dis.
Registry. Liver Transpl. 2010;16:461–9. 2008;10:339–42.
56. Silveira MG, Talwalkar JA, Lindor KD, Wiesner 71. Witte L, Rupp J, Heyer P, et al. Fibrosing alveoli-
RH. Recurrent primary biliary cirrhosis after liver tis with hepatitis C-related cryoglobulinemia. Dtsch
transplantation. Am J Transplant. 2010;10:720–6. Med Wochenschr. 2008;133:709–12.
57. Torring PM, Larsen MJ, Kiedsen AD, et al. Global 72. Howard L, Gopalan D, Griffiths M, Mahadeva
gene expression of telangectisial tissue from patients R. Sirolimus-induced pulmonary hypersensitiv-
with hereditary telangiectasia. Microvasc Res. ity associated with a CD4 T-cell infiltrate. Chest.
2015;99:118–26. 2006;129:1718–21.
58. Buonamico P, Suppressa P, Lenato GM, et al. Liver 73. Carrillo-Esper R, González-Avila D, Uribe-Ríos M,
involvement in a large cohort of patients with heredi- Méndez-Sánchez N. Interstitial pneumonitis asso-
tary hemorrhagic telangiectasia: echo-color-Doppler ciated with pegylated interferon alpha-2b therapy
vs multislice computed tomography study. J Hepatol. for chronic hepatitis C: case report. Ann Hepatol.
2008;48:811–20. 2008;7:87–90.
59. Memeo M, Stabile Ianora AA, et al. Hereditary 74. Krowka MJ. Hepatopulmonary syndrome and por-
haemorrhagic telangiectasia: study of hepatic topulmonary hypertension: implications for liver
vascular alterations with multi-detector row heli- transplantation. Clin Chest Med. 2005;26:587–97.
cal CT and reconstruction programs. Radiol Med. 75. Colle IO, Moreau R, Godinho E, et al. Diagnosis of
2005;109:125–38. portopulmonary hypertension in candidates for liver
60. Garcia-Tsao G. Liver involvement in hereditary transplantation: a prospective study. Hepatology.
hemorrhagic telangiectasia (HHT). J Hepatol. 2003;37:401–9.
2007;46:499–507. 76. Thenappan T, Ryan JJ, Archer SL. Evolving epide-
61. Rozenberg D, Leek E, Faughnan ME. Prevalence miology of pulmonary arterial hypertension. Am J
and nature of dyspnea in patients with hereditary Crit Care Med. 2012;186:707–9.
hemorrhagic telangiectasia [HHT]. Respir Med. 77. Galie N, Hoeper MM, Humbert M, et al. Guidelines
2015;109:768–77. for the diagnosis and treatment of pulmonary
62. Cottin V, Plauchu H, Bayle JY, et al. Pulmonary arte- hypertension: the Task Force for the Diagnosis
riovenous malformations in patients with hereditary and Treatment of Pulmonary Hypertension of the
hemorrhagic telangiectasia. Am J Respir Crit Care European Society of Cardiology (ESC) and the
Med. 2004;169:994–1000. European Respiratory Society (ERS), endorsed by
63. Faughnan ME, Granton JT, Young LH. The pulmo- the International Society of Heart and Lung
nary vascular complications of hereditary haemor- Transplantation (ISHLT). Eur Heart J.
rhagic telangiectasia. Eur Respir J. 2009;33:1186–94. 2009;34:2493–537.
64. Epperla N, Hocking W. Blessing for the bleeder bev- 78. Kim WR, Krowka MJ, Plevak DJ, et al. Accuracy of
acizumab in hereditary hemorrhagic telangiectasia. Doppler echocardiography in the assessment of pul-
Clin Med Res. 2015;13:32–5. monary hypertension in liver transplant candidates.
65. Lerut J, Orlando G, Adams R, et al. Liver transplan- Liver Transpl. 2000;6:453–8.
tation for hereditary hemorrhagic telangiectasia: 79. D’Alto M, Romeo E, Argiento P, et al. Accuracy
reprot of the European liver transplant registry. Ann and precision of echocardiography versus right heart
Surg. 2006;244:854–62. catheterization for the assessment of pulmonary
66. Gur C, Ilan Y, Shibolet O. Hepatic hydrothorax– hypertension. Int J Cardiol. 2013;168:4058–62.
pathophysiology, diagnosis and treatment—review 80. Roberts KE, Fallon MB, Krowka MJ, et al.
of the literature. Liver Int. 2004;24(4):281. Pulmonary Vascular Complications of Liver Disease
67. Sese E, Xiol X, Castellote J, et al. Low complement Study Group. Genetic risk factors for portopulmo-
levels and opsonic activity in hepatic hydrothorax: nary hypertension in patients with advanced liver dis-
its relationship with spontaneous bacterial empy- ease. Am J Respir Crit Care Med. 2009;179:835–42.
ema. J Clin Gastroenterol. 2003;36:75–7. 81. Ramsay M. Portopulmonary hypertension and right
68. Ferrante D, Arguedas MR, Cerfolio RJ, et al. Video-­ heart failure in patients with cirrhosis. Curr Opin
assisted thoracoscopic surgery with talc pleurodesis Anaesthesiol. 2010;23:145–50.
in the management of symptomatic hepatic hydro- 82. Porres-Aguilar M, Zuckerman MJ, Figueroa-Casas
thorax. Am J Gastroenterol. 2002;97:3172–5. JB, Krowka MJ. Portopulmonary hypertension: state
69. Ditah IC, Bawardy BF, Saberi B, et al. Transjugular of the art. Ann Hepatol. 2008;7:321–30.
intrahepatic portosystemic stent shunt for medically 83. Ghofrani HA, Humbert M. The role of combination
refractory hepatohydrothorax: a systematic review in managing pulmonary arterial hypertension. Eur
and cumulative meta-analysis. Wrold J Hepatol. Respir Rev. 2014;134:469–75.
2015;13:1797–806.
24  Pulmonary Complications of Liver Disease 305

84. Swanson KL, Wiesner RH, Nyberg SL, et al. Survival 95. Gómez FP, Martínez-Pallí G, Barberà JA, et al. Gas
in portopulmonary hypertension: Mayo Clinic expe- exchange mechanism of orthodeoxia in hepatopul-
rience categorized by treatment subgroups. Am J monary syndrome. Hepatology. 2004;40:660–6.
Transplant. 2008;8:2445–53. 96. Degano B, Mittaine M, Hervé P, et al. Nitric oxide
85. Robalino BD, Moodie DS. Association between pri- production by the alveolar compartment of the lungs
mary pulmonary hypertension and portal hyperten- in cirrhotic patients. Eur Respir J. 2009;34:138–44.
sion: analysis of its pathophysiology and clinical, 97. Gómez FP, Barberà JA, Roca J, et al. Effects of nebu-
laboratory and hemodynamic manifestations. J Am lized N(G)-nitro-L-arginine methyl ester in patients
Coll Cardiol. 1991;17:492–8. with hepatopulmonary syndrome. Hepatology.
86. Le Pavec J, Souza R, Herve P, et al. Portopulmonary 2006;43:1084–91.
hypertension: survival and prognostic factors. Am J 98. Tang L, Luo B, Patel RP, et al. Modulation of pulmo-
Respir Crit Care Med. 2008;178:637–43. nary endothelial endothelin B receptor expression
87. Goldberg DS, Batra S, Sahay S, et al. MELD excep- and signaling: implications for experimental hepato-
tions for portopulmonary hypertension. Current pulmonary syndrome. Am J Physiol Lung Cell Mol
policy and future implications. Am J Transplant. Physiol. 2007;292:L1467–72.
2014;9:2081–7. 99. Krowka MJ, Mandell MS, Ramsay MA, et al.
88. Fallon MB, Mulligan DC, Gish RG, Krowka Hepatopulmonary syndrome and portopulmonary
MJ. Model for end-stage liver disease (MELD) hypertension: a report of the multicenter liver trans-
exception for hepatopulmonary syndrome. Liver plant database. Liver Transpl. 2004;10:174–82.
Transpl. 2006;12(12 Suppl 3):S105–7. 100. Swanson KL, Wiesner RH, Krowka MJ. Natural his-
89. Rafanan AL, Maurer J, Mehta AC, Schilz tory of hepatopulmonary syndrome: impact of liver
R. Progressive portopulmonary hypertension after transplantation. Hepatology. 2005;41:1122–9.
liver transplantation treated with epoprostenol. 101. Arguedas MR, Abrams GA, Krowka MJ, Fallon
Chest. 2000;118:1497–500. MB. Prospective evaluation of outcomes and pre-
90. Koneru B, Ahmed S, Weisse AB, et al. Resolution of dictors of mortality in patients with hepatopulmo-
pulmonary hypertension of cirrhosis after liver trans- nary syndrome undergoing liver transplantation.
plantation. Transplantation. 1994;58:1133–5. Hepatology. 2003;37:192–7.
91. Pascasio JM, Grilo I, Lopez-Pardo FJ, et al. 102. Benítez C, Arrese M, Jorquera J, et al. Successful
Prevalence and severity of Hepatopulmonary treatment of severe hepatopulmonary syndrome with
syndrome and its influence on survival on cir- a sequential use of TIPS placement and liver trans-
rhotic patients evaluated for transplantation. Am J plantation. Ann Hepatol. 2009;8:71–4.
Transplant. 2014;14:1391–9. 103. De BK, Sen S, Biswas PK, et al. Occurrence of
92. Fierro Alanis MP, Bastón Rey I, Nieves-Maldonado hepatopulmonary syndrome in Budd-Chiari syn-
S. Platypnea-orthodeoxia syndrome: Importance drome and the role of venous decompression.
of patient position for correct diagnosis at the time Gastroenterology. 2002;122:897–903.
of (99m)Tc-MAA injection. Rev Esp Med Nucl 104. Nayyar D, Man HS, Granton J, et al. Proposed man-
Imagen Mol. 2015;34:261–3. agement algorithm for severe hypoxemia after liver
93. El-Shabrawi MH, Omran S, Wageeh S, et al. transplantation in the hepatopulmonary syndrome.
99mTechnetium-macroaggregated albumin perfu- Am J Transplant. 2015;15:903–13.
sion lung scan versus contrast enhanced echocar- 105. Schiffer E, Majno P, Mentha G, et al.
diography in the diagnosis of the hepatopulmonary Hepatopulmonary syndrome increases the postop-
syndrome in children with chronic liver disease. Eur erative mortality rate following liver transplantation:
J Gastroenterol Hepatol. 2010;22(8):1006–12. a prospective study in 90 patients. Am J Transplant.
94. Rodriguez-Roisin R, Roca J, Agusti AG, et al. Gas 2006;6:1430–7.
exchange and pulmonary vascular reactivity in 106. Taille C, Cadranel J, Bellocq A, et al. Liver trans-
patients with liver cirrhosis. Am Rev Respir Dis. plantation for hepatopulmonary syndrome: a ten
1987;135:1085–92. year experience in Paris, France. Transplantation.
2003;75:1482–9.
The Patient with Severe
Co-morbidities: CNS Disease 25
and Increased Intracranial Pressure

Prashanth Nandhabalan, Chris Willars,
and Georg Auzinger

Keywords
I ntracranial Hypertension in Acute
Intracranial hypertension · Ammonia · Cerebral
Liver Failure
blood flow · Intracranial pressure · Transcranial
Doppler · Therapeutic hypothermia · Cerebral
 tiology and Pathophysiology
E
edema · Hepatic encephalopathy
of Encephalopathy and Cerebral
Edema in Acute Liver Failure

Introduction Intracranial hypertension (ICH) is a common


cause of death in acute liver failure (ALF) [1].
Intracranial hypertension presents specific chal- The concept of cerebral edema and hyperemia as
lenges for the anesthesiologist during liver a cause of the acute rise in intracranial pressure
transplantation. The goal of reducing and avoid- (ICP) in ALF was first described in the early
ing surges of intracranial pressure is often in 1970s [2]. While the incidence of ICH is decreas-
conflict with general anesthetic goals. It is cru- ing, it is still associated with high mortality [3].
cial to maintain cerebral perfusion and protect In a large retrospective review of 3300 patients
the brain during surgery. Neurological compli- with ALF, the incidence of ICH had decreased to
cations are frequent and can be detrimental. 20% but mortality was still high at 55%. The
This chapter will review the pathophysiology of onset of ICH in ALF is rapid and allows insuffi-
intracranial hypertension in acute liver failure as cient time for adaptive processes. The underlying
well as perioperative strategies to prevent and etiology is likely to be multifactorial and includes
treat it in order to avoid catastrophic neurologi- the development of cerebral edema, disordered
cal outcomes. cerebral blood flow and inflammatory processes.

Etiology: Cerebral Cytotoxic Edema

The failing liver’s inability to metabolize ammo-


nia to urea is associated with the development of
hepatic encephalopathy. In an analysis of 165
P. Nandhabalan, MRCP, FRCA, FFICM, EDIC patients with ALF of varying etiology [4], a high
C. Willars, MBBS, BSc, FRCA, FFICM arterial ammonia concentration was an indepen-
G. Auzinger, EDIC, AFICM (*)
Department of Critical Care/Institute of Liver
dent risk factor for severe encephalopathy and
Studies, King’s College Hospital, London, UK ICH. A level of >100 μmol/L predicts the onset
e-mail: georg.auzinger@nhs.net of severe encephalopathy with 70% accuracy,

© Springer International Publishing AG, part of Springer Nature 2018 307


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_25
308 P. Nandhabalan et al.

and ammonia levels of >200 μmol/L are associ- process potentiated by ammonia) where it is
ated with the development of ICH and the possi- hydrolyzed to produce glutamate and ammonia
bility of herniation. Persistently high ammonia (Fig.  25.1) [12]. This leads to the formation of
levels are commonly found in patients with ICH reactive oxygen species and induction of the mito-
and are associated with a greater incidence of chondrial permeability transition pore leading to
complications and significantly higher mortality astrocyte swelling through up-regulation of aqua-
[5]. Higher MELD (Model for End-Stage Liver porins. Alternative mechanisms attribute the toxic
Disease) scores, younger age and requirement for effects of glutamine to restricted transfer out of
vasopressors or renal replacement therapy are the astrocyte cell rather than increased synthesis
additional independent risk factors for hepatic within it [13].
encephalopathy [6]. Gene expression may also be altered in response
Ammonia plays a crucial role in the patho- to the onset of FHF, particularly those genes cod-
genesis of cytotoxic cerebral edema and has been ing for astrocytic proteins. These proteins have
identified as a potent neurotoxin with a multi- important roles in the regulation of cell volume
tude of molecular effects [7]. Cytotoxic cerebral and in neurotransmission. The expression of the
edema arises as a result of astrocyte swelling [8] astrocytic/endothelial glucose transporter gene, the
and is generally considered the principal cause aquaporin-4 water channel and glutamate trans-
of intracranial hypertension in ALF. Ammonia porter gene have been specifically studied and
produced by bacteria in the bowel is taken up by demonstrated to be altered in FHF; however, the
astrocytes and converted into glutamine through significance of any of these processes in isolation
the actions of glutamine synthetase. Brain glu- remains unknown.
tamine concentrations are increased in animal Evidence of predominant cytotoxic edema for-
models of fulminant hepatic failure (FHF) [9] mation in ALF is based on findings of diffusion-­
and also in samples taken post-mortem from weighted MRI scanning [14]. The diffusion
patients with FHF [10]. Furthermore, cerebral coefficient that quantifies movement of water mol-
microdialysis studies in patients with ALF con- ecules across cell membranes is significantly lower
firm a strong correlation of arterial ammonia in ALF patients with resolution of abnormal find-
concentrations with brain glutamine content and ings following recovery of liver failure. However,
ICP [11]. Persistent elevations of both param- more recent MRI studies have revealed the pres-
eters may identify individuals at risk of ICH. ence of both cytotoxic and interstitial edema,
Having entered the astrocyte, a glutamine car- suggesting that vasogenic edema may also be of
rier transports glutamine to the mitochondria (a relevance [15]. A possible mechanism comprises
dysfunction of the blood-brain barrier (BBB) in
the context of preserved structural integrity as a
result of substances released from the injured liver.
Mitochondria One such substance, matrix metalloproteinase 9
ROS NH4++ GLU (MMP-9), has been heavily implicated in induc-
4
3 GLNasa ing significant alterations in tight junction proteins
GLN-TX
leading to increased BBB permeability [16].
NH4+ 2
In summary, cerebral edema may be vasogenic
GS
Glutamine with inflammatory disruption of the blood–brain
1

GLU barrier, allowing extracellular edema formation, or


cytotoxic with an increase in intracellular water as
a result of accumulation of glutamine and oxidative
Fig. 25.1  The detoxification of ammonia to glutamine stress. These changes occur with varying temporal
(GLU) mediated by glutamine synthetase (GS) and subse- and spatial resolution with recent evidence suggest-
quent creation of reactive oxygen species (ROS) follow-
ing transport into the mitochondria. GLN-Tx glutamine ing an early role for vasogenic edema, particularly
transporter, GLNase glutaminase in the basal ganglia and motor cortex, followed by
25  The Patient with Severe Co-morbidities: CNS Disease and Increased Intracranial Pressure 309

predominantly frontal cortical cytotoxic edema as lation can further lead to increased cerebral blood
severity of encephalopathy progresses [17]. flow and blood volume and therefore ICH. This
concept is supported by findings from animal
models and seen in patients with FHF [24]. The
 tiology: Cerebral Blood Flow
E loss of autoregulation has been attributed to the
and Systemic Inflammation effects of nitric oxide (NO) on the cerebral vas-
culature, but it may be that elevated NO levels
Recent evidence has highlighted the prominent only occur secondary to increase in cerebral
role of systemic inflammation in the pathogenesis blood flow rather than as a primary and causative
of HE [18]. FHF is associated with an accumula- ­phenomenon [25].
tion of toxic metabolites and a massive systemic
inflammatory response with the release of a vast
quantity of pro-inflammatory cytokines causing a Etiology: Inflammation Within
hyperdynamic state. The presence and degree of the Brain
systemic inflammation is associated with progres-
sion to more severe encephalopathy and greater Inflammation within the brain itself has also been
mortality. Alterations of cerebral blood flow are implicated in the development of hepatic enceph-
directly attributable to this inflammatory milieu; alopathy and ICH. Pro-inflammatory cytokines
one study showed a direct correlation between including IL-1β, IL-6 and TNF-α are released into
increased cerebral blood flow and serum levels of the cerebral circulation in patients with ALF and
cytokines, resulting in higher ICP [19]. Blood arterial cytokine levels correlate well with sever-
flow is coupled to cerebral metabolic rate and ity of intracranial hypertension. These cytokines
changes in ventilation and acid–base status. In directly cause astrocyte swelling, and are potenti-
conjunction with blood–brain barrier injury, ated by previous exposure to ammonia [26].
increased cerebral blood flow and hyperemia can Further evidence comes from experimental mod-
potentiate cerebral edema independently of astro- els of ALF where gene deletions for IL-1 and
cyte glutamine concentration, contributing to the TNF-α receptors significantly delayed the onset
rise in ICP. Although cerebral blood flow can vary of cerebral edema [27].
in patients with ALF, higher flow rates are associ- Cellular dysfunction in ALF has been shown to
ated with poorer outcomes. extend beyond astrocyte swelling. Microglial acti-
Cerebral edema is diminished in anhepatic vation occurs early in ALF and increases as
rats compared with those with experimentally encephalopathy and cerebral edema develop.
induced FHF [20]. Intrasplenic transplantation Lactate, ammonia and manganese have all been
of allogeneic hepatocytes prevents development of implicated in stimulating microglial activation and
ICH in pigs with acute ischemic liver failure and subsequent release of pro-inflammatory cytokines.
transient hepatectomy [21]. The creation of a por- Anti-inflammatory interventions that counter these
tacaval shunt has been used successfully in ALF changes can slow the progression of HE and have
patients with intractable ICH as a bridge to trans- been suggested as potential mechanisms for cur-
plantation. The observation that ICH occurs with rent and hypothetical therapeutic strategies.
FHF, but not chronic liver disease, lends further
weight to the ‘toxic liver hypothesis’. ICP mea-
surements during transplant surgery have demon-  athophysiology of Intracranial
P
strated that ICP increases during the manipulation Hypertension
and dissection of the necrotic liver [22], but then
decreases during the anhepatic phase. Evidence Normal ICP is approximately 7–15 mmHg in a
from case reports suggests that the levels of pro-­ supine adult. Definitions of ICH vary, but a
inflammatory cytokines are diminished following pressure of >20 mmHg for a period of 20 min
removal of the toxic liver [23]. Loss of autoregu- or more can be considered as an episode of sig-
310 P. Nandhabalan et al.

nificant ICH. Accurate measurement of ICH FHF. Rather, the radiological appearances are con-
requires the insertion of an ICP monitor. sistent with acute cerebral edema. Brain edema has
Current AASLD guidelines recommend ICP been demonstrated in rabbits with galactosamine-
monitoring for patients with advanced hepatic induced fulminant hepatitis [30] and ammonia-
encephalopathy who are awaiting liver trans- induced cerebral edema in rats. Hyperemia due to
plantation (LTx) in centers with appropriate defective autoregulation or circulating inflamma-
expertise and osmotic therapy with mannitol tory mediators may further compound the rise in
for ICP ≥ 25 mmHg [28]. ICP. The main complication of profound ICH is
According to Monro and Kellie, the cranial diencephalic transtentorial herniation, causing:
compartment is essentially an incompressible
box with a fixed internal volume. Blood, CSF and • Posterior cerebral artery insufficiency with
brain tissue (~90% of the total) exist in a state of temporal, thalamic and occipital infarction
volume equilibrium and are relatively incom- • Compression of the cerebral aqueducts and
pressible, such that any increase in the volume of subarachnoid space with resultant obstructive
one of the cranial constituents must be compen- hydrocephalus
sated for by a decrease in the volume of another. • Brain stem compression, ischemia and death
CT studies in FHF have demonstrated that ven-
tricular spaces are either unchanged or compressed To summarize, two predominant mechanisms
[29], and therefore, the expansion of the CSF com- are thought to cause the rise in ICP seen in FHF
ponent is not responsible for rises in ICP in [31] (Fig. 25.2):

Fulminant hepatic failure

Ammonia-glutamine Toxic liver hypothesis


hypothesis

Toxic products of the failing liver

Inflammatory cytokines

Deficit in liver detoxification of ammonia

Systemic hyperammonemia

Astrocyte accumulation of glutamine

Astrocyte swelling
Increase in cerebral blood flow
Cytotoxic brain edema

Increase in brain volume Increase in intracranial blood volume

Increase in intracranial volume

Increase hypertension
brain stem death
Fig. 25.2  Etiology of ICH
25  The Patient with Severe Co-morbidities: CNS Disease and Increased Intracranial Pressure 311

• The uptake and detoxification of ammonia to with subsequent sedation and mechanical venti-
osmotically active glutamine by astrocytes, lation [28]. Under these circumstances, the only
leading to cytotoxic cerebral edema (the reliable early monitor of raised ICP—the patient’s
ammonia-glutamine hypothesis) own conscious level—has been lost, although
• The loss of cerebral autoregulation leading to clonus, hypertonicity and decerebrate posturing
increased CBF secondary to circulating inflam- may still be detected. Pupillary changes, sys-
matory mediators (the toxic liver hypothesis) temic hypertension and reflex bradycardia are
with disruption of the blood–brain barrier and late changes, and radiographic changes are non-­
vasogenic edema formation. specific. A relatively ‘tight’ brain is often seen on
CT imaging but correlates poorly with severity of
Both of these seemingly contradictory mecha- cerebral edema or the presence of ICH.
nisms are probably responsible for ICH and pres-
ent specific targets for interventions. They are
compounded by the brain’s response to the acutely ICP Monitoring
failing liver in modifying gene expression and
releasing pro-inflammatory mediators which drive Insertion of an ICP monitor (after correction of
both cytotoxic and vasogenic cerebral edema for- coagulopathy) and jugular bulb oximetry readings
mation, and propagate the inflammatory cascade. allow for continuous monitoring of ICP and give
an indication of the cerebral oxygen supply/
demand relationship. ICH may develop rapidly
Cerebral Perfusion Pressure and is subject to flux. Inadequate sedation, seizure
activity and worsening edema/hyperemia can
ICH compromises cerebral perfusion pressure cause sudden and potentially dangerous surges in
(CPP) given their relationship: CPP = MAP−ICP ICP. Continuous monitoring enables rapid detec-
(MAP—mean arterial pressure). A sustained tion of ICH and implementation of target therapy
decrease of CPP to less than 40 mmHg for 2 h or accordingly. ICP monitoring further allows assess-
more is associated with a poor outcome, although ment of prognosis and possible neurological out-
there are reports of complete neurological recov- come. Clinical signs do not adequately allow a
ery despite prolonged periods of perfusion pres- quantification of ICP.
sure below this threshold [32]. There is no However, a lack of consensus over the thera-
consensus as to the optimum CPP in patients with peutic goals has done little to promote the role of
ALF but current guidelines advocate maintaining ICP monitoring in ALF. Trials evaluating the effi-
CPP around 60–80 mmHg [28]. While every cacy of ICP monitoring in Traumatic Brain Injury
attempt should be made to maintain cerebral per- have consistently failed to identify a clear sur-
fusion within well-defined limits, our own expe- vival benefit. A large multicenter randomized
rience indicates that a transient decrement in controlled trial demonstrated no difference in
cerebral perfusion should not be interpreted in outcome between protocol-delivered care accord-
isolation as a marker of poor prognosis. ing to ICP monitoring and imaging/clinical
examination [33]. Similarly, although random-
ized controlled trials are lacking, studies in ALF
 iagnosis and Multimodality
D have shown no benefit of ICP monitoring. A
Monitoring study of 332 patients with ALF reported the
experience with ICP monitoring in 24 centers
ICH should be suspected in any patient who pres- [34]. ICP monitoring was used in only 92 patients
ents with hepatic encephalopathy in the context (28% of the cohort), and the 30-day survival for
of acute or fulminant liver failure and/or signifi- liver transplantation recipients was similar in
cantly elevated arterial ammonia levels. Patients both monitored and unmonitored groups (85%
with ALF and rapidly evolving encephalopathy vs. 85%). Most recently, a retrospective cohort
will need to undergo endotracheal intubation study by the USALF group identified a similar
312 P. Nandhabalan et al.

prevalence of 22% of patients with ALF and High saturations >80% are found during cerebral
advanced HE undergoing ICP monitoring [35]. hyperemia, with inadequate neuronal metabolism or
ICP monitoring was associated with no benefit in with neuronal cell death. Both high and low jugular
the acetaminophen-induced ALF group and an venous saturations are equally associated with poor
increased mortality in those patients with non-­ outcomes [38]. In patients with acute on chronic
acetaminophen induced ALF. It should be noted liver failure, abnormalities in SjvO2 are indepen-
that any observational study is unlikely to be able dently associated with death [39]. A major disadvan-
to identify the benefit of a monitoring device, tage of SjvO2 is that it provides an estimate of global
particularly if the outcome is associated with oxygenation and metabolism, and smaller areas of
many other confounding factors. critical ischemia may not affect overall cerebral
Widespread use of ICP monitoring has been venous oxygen content. However, rises in ICP, effect
curtailed by concerns over safety and increased risk of hyperventilation therapy, hypotension and cere-
of bleeding in potentially coagulopathic critically bral vasospasm may all be detected with SjvO2.
ill patients. The procedure carries an, although in SjvO2 is reduced in the following clinical
expert hands small, yet significant bleeding risk. In scenarios:
the studies above, 5–7% patients experienced clini-
cally significant intracranial hemorrhage. A retro- • Cerebral vasoconstriction (e.g. as a result of
spective analysis of 115 patients in our institution hyperventilation and hypocarbia)
demonstrated much lower rates of associated hem- • Hypoxemia
orrhage of 2.6% [36]. A retrospective study of ICP • Anemia
monitoring in pediatric ALF revealed a small sub- • Diminished CPP
dural haematoma in one of 14 patients; these how- • Inappropriately high CPP and vasoconstric-
ever resulted in no demonstrable neurological tion induced by exogenous vasoconstrictor
deficit [37]. • Seizure activity
Monitoring modalities differ between centers:
Extradural monitoring is less accurate and asso- SjvO2 is elevated in:
ciated with significant baseline drift, but penetra-
tion of the dura is associated with higher rates of • Hyperemia
bleeding. Patients whose ICP is monitored • Vasodilation (e.g. as a result of hypoventila-
undergo more treatment interventions, but it is tion and hypercarbia)
not clear whether these interventions result in • Brain death
better neurological outcomes.

Transcranial Doppler
Jugular Bulb Oximetry
Transcranial Doppler ultrasound (TCD) is a sim-
Blood from the cerebral venous sinuses drains ple and non-invasive method of quantifying
into the internal jugular vein. Monitoring of oxy- blood flow velocities in the basal cerebral arteries
gen saturation in the jugular bulb allows an esti- (most commonly the middle cerebral artery).
mation of the balance of global oxygen supply Cerebral blood flow is calculated from the mean
versus demand ratio and hence of cerebral metab- flow velocity if the cross-sectional area of the tar-
olism. Both intermittent sampling and continu- geted artery is known:
ous monitoring may be used, although the latter CBF = mean flow velocity × area of artery
requires the insertion of a fiberoptic catheter. × cosine angle of insonation
The normal range for jugular venous oxygen
saturations (SjvO2) is 60–75%. Desaturations to less Successive measures of CBF are only compara-
than 55% are indicative of cerebral hypoperfusion ble if the angle of insonation and the diameter of the
due to inadequate CPP or a sign of increased cere- target vessel remain the same. Varying vessel diam-
bral oxygen uptake as seen with seizure activity. eters with vasospasm are a potential source of error.
25  The Patient with Severe Co-morbidities: CNS Disease and Increased Intracranial Pressure 313

An increase in flow velocities is seen with to FHF has not been fully evaluated; in pediat-
hyperemia and increased cerebral blood flow and ric patients with ALF it appears to help iden-
during episodes of cerebral vasospasm. In order tify those patients with a poor prognosis [45]
to differentiate between these two very different and has been used in the perioperative liver
phenomena, the ratio of middle cerebral artery to transplant setting as an indirect surrogate of
extracranial internal carotid artery flow can be ICP [46]. This tool may be a useful adjunct if
determined. The MCA velocity is normally about the indications for ICP monitoring are unclear,
60–70 cm/s with an ICA velocity of 40–50 cm/s. or in quantifying the risk of ICH and identify-
The MCA/ICA ratio is therefore 1.76 ± 0.1. An ing those patients who are most likely to ben-
MCA velocity > 120 cm/s is considered signifi- efit from direct monitoring.
cantly elevated and when accompanied by a high
MCA/ICA ratio likely due to vasospasm. If
MCA/ICA ratios are lower, hyperemia is the Preoperative Management
more likely diagnosis.
The pulsatility index (PI = peak systolic veloc- Recommended strategies for the reduction of
ity−end diastolic velocity/mean flow velocity) is ICH include specific therapies targeting ICP and
less subject to inter-observer variation and corre- the reduction of the volume of brain tissue, as
lates well with ICP in patients with intracranial well as general measures to protect against sec-
pathology [40]. Elevation of the PI indicates ondary brain damage following the primary insult
intracranial hypertension and values >1.6 are (Rosner’s conjecture). This should embrace all
associated with a worse outcome. Measurement the factors responsible for causing secondary
of PI may help to determine prognosis in patients insult via cerebral ischemia.
with FLF [41, 42]. Transcranial Doppler wave- Medical management thus falls under a number
form analysis can also provide a quantitative of broad categories:
assessment of cerebral hemodynamics and may
be an alternative to PI measurements [43]. • General supportive measures
• Prevention and treatment of raised ICP
• Achieving an appropriate CPP
Non-Invasive Monitoring of ICP • Specific medical therapies
• Anticipation and management of complications
Non-invasive monitoring of ICP with computed
tomography, MRI, or PET scanning is inaccurate, An ICP >15 mmHg is considered abnormally
non-continuous and often impractical in advanced high. Various authors have suggested different
stages of ALF. Tympanic tonometry is inaccurate thresholds for treatment under different circum-
compared with direct ICP measurement but may stances. The Brain Trauma Foundation [47] sug-
be useful in detecting changes in ICP. Near infra- gests a treatment threshold of 20 mmHg, whilst
red spectrophotometry (NIRS) shows promise in AASLD guidelines advise maintaining ICP
detecting changes in cerebral blood flow and below 20–25 mmHg. The limits within which
blood volume in patients with ALF [44]. CPP should be maintained are also not clearly
The optic nerve sheath distends when CSF defined.
pressure is elevated. Measurement of optic
nerve sheath diameter may therefore be an
acceptable surrogate for the measurement of ICP-Targeted Therapies
raised ICP. MRI and ocular sonography fol-
lowing TBI have demonstrated a correlation The majority of treatment strategies are similar to
between nerve sheath diameter and presence of those described in the neurosurgical literature,
ICH. This method of assessment is user depen- but many of the pathophysiological mechanisms
dent but non-invasive and can be performed at of cerebral edema in ALF are unique and not
the bedside. At present, its use in ICH related applicable to other patient groups.
314 P. Nandhabalan et al.

Positioning and Environment There is no general consensus regarding the


mode of ventilation to be used. Given that acute
The head of the bed should be elevated at ~30° to respiratory distress syndrome (ARDS) may accom-
facilitate venous and CSF drainage. Further ele- pany the systemic inflammatory response of FHF
vations can cause a paradoxical increase in (particularly with the development of raised ICP),
ICP. The head and neck are kept in a neutral posi- a protective ventilatory strategy should be adopted
tion, approximating the midline. Environmental where possible (limiting tidal volumes to ~6 mL/
stimulation is kept to a minimum. kg and plateau pressure to <30 cmH2O). Permissive
hypercapnia is poorly tolerated as any rise in PaCO2
will be associated with a concomitant rise in ICP.
Ventilation High levels of positive end-expiratory pressure
(PEEP) can diminish venous return and reduce
Encephalopathy is usually graded using the West hepatic blood flow; at the same time, PEEP levels
Haven criteria for encephalopathy (Table 25.1). up to 15 cmH2O have been used safely in patients
Endotracheal intubation is performed for airway with TBI and ARDS. A ‘best PEEP’ strategy
protection in advanced grade III/IV encepha- (choosing PEEP levels that will provide maximal
lopathy, to facilitate the control of ICP (cerebral recruitment whilst avoiding alveolar over-disten-
blood flow is coupled to cerebral metabolic rate sion to optimize oxygen delivery) is advisable.
and to PaO2 and PaCO2) and for the treatment Recruitment maneuvers should be used with cau-
of respiratory failure. Induction of anesthesia tion as experimental studies have indicated that
should aim to attenuate surges in ICP on laryn- such maneuvers may be associated with transient
goscopy and intubation whilst maintaining CPP liver dysfunction [48]. Hypoxia and hypercapnia
within acceptable limits. cause CBF (and therefore ICP) to increase.
Prophylactic hyperventilation may reduce brain
edema and delay the onset of brain herniation [49];
however, it may result in unwanted cerebral vaso-
Table 25.1  West Haven criteria for semiquantative grad-
constriction which could be detrimental for oxygen
ing of mental state (encephalopathy grades)
delivery to marginal/at-risk areas of brain tissue.
Grade 1 Trivial lack of awareness
The Brain Trauma Foundation recommends the
Euphoria or anxiety
use of hyperventilation as a temporizing measure
Shortened attention span
only and suggests that it should be avoided during
Impaired performance of addition
the first 24 h after TBI. In the setting of ALF, con-
Grade 2 Lethargy or apathy
trolled studies have failed to show any benefit, with
Minimal disorientation for time or place
no reduction in the number of episodes of raised
Subtle personality change
ICP [49]. Hyperventilation should be guided by
Inappropriate behavior
jugular bulb oximetry or other forms of monitor-
Impaired performance of subtraction
ing of adequacy of cerebral oxygen supply; as with
Grade 3 Somnolence to semistupor, but responsive
to verbal stimuli TBI, it should only be used for the emergency res-
Confusion cue of imminent diencephalic herniation.
Gross disorientation
Grade 4 Coma (unresponsive to verbal or noxious
stimuli) Temperature
With permission: Ferenci P, Lockwood A, Mullen K, Tarter
R, Weissenborn K, Blei AT. Hepatic encephalopathy—defi- In general, normothermia should be maintained.
nition, nomenclature, diagnosis, and quantification: final
report of the working party at the 11th World Congresses of
Fever needs to be treated aggressively because
Gastroenterology, Vienna, 1998. Hepatology 2002;35: it stimulates cerebral metabolism and conse-
716–21 quently induces vasodilatation. Cooling blankets
25  The Patient with Severe Co-morbidities: CNS Disease and Increased Intracranial Pressure 315

and paracetamol are both suitable for this pur- Arterial ammonia levels should be monitored as
pose. As many patients will require extracorpo- even moderate enteral protein loads can trigger
real renal replacement therapy, low-temperature marked hyperammonemia in patients with major
control can be easily maintained on extracorpo- acute hepatic insufficiency. Parenteral feeding
real circuits. may be indicated in patients whom enteral feed-
ing is not possible [55].

Glycemic Control
Infection Prophylaxis
Hyperglycemia may exacerbate secondary brain
injury (Rosner’s conjecture) and exacerbate Infection is a frequent complication of ALF and
ICH in patients with ALF [50]. Dysglycaemia is associated with progression of hepatic encepha-
has several deleterious effects on the brain lopathy. ALF patients develop a functional immu-
through a variety of mechanisms including noparesis with monocyte dysfunction, defective
upregulation of cellular glucose transporters complement activation and impaired neutrophil
and neuronal glucose overload leading to oxi- phagocytosis [56]. Respiratory tract infections,
dative stress [51]. After conflicting results from including ventilator-associated pneumoniae are
previous trials, a large randomized controlled most prevalent, and likely aggravated by aggres-
trial definitively demonstrated an increased sive measures to reduce ICH such as heavy
mortality with tight glycemic control in criti- sedation, induced-hypothermia and avoidance
cally ill patients [52]. In TBI, tight glycemic of regular bronchial toilette. Central-line associ-
control can lead to critical brain tissue hypoxia ated blood stream infections (CLABSIs), urinary
and in the first week after injury, has been asso- sepsis, abdominal sepsis secondary to bacterial
ciated with poor ICP control, higher incidence translocation and de novo septicemia are also
of bacteremia and worse survival [53]. Given common. Gram-positive cocci (Staphylococci,
that there is no compelling evidence that tight Streptococci) and enteric Gram-­negative bacilli
glycemic control is beneficial in this popula- are the most frequently isolated organisms. Fungal
tion and ALF is associated with a propensity infections are also common and may occur in a
towards hypoglycemia, a common complica- third of ALF patients [57]. It is routine practice
tion of tight glycemic control in the large trial to treat early and aggressively with antifungal
above, it would seem prudent to aim for less therapy. To avoid CLABSIs intravenous catheters
restrictive glucose levels. should be monitored on a regular basis, changed
if suspected as a cause for infection and removed
when not needed anymore.
Nutrition Antibiotic prophylaxis is instituted as a matter
of routine in all patients with advanced encepha-
ALF induces a systemic inflammatory response lopathy and when infection seems likely on the
and hypermetabolic state. Catabolism predomi- basis of clinical and laboratory investigations.
nates with a negative nitrogen balance and immu- Empirical antibiotic therapy has previously been
nodeficiency. The energy expenditure even in the advised for patients listed for LTx as new infec-
resting state is considerable, and early nutritional tive episodes may preclude transplantation and
support is therefore recommended, although imminent immunosuppression. However, current
there is little evidence of benefit in this patient AASLD guidelines [28] state that:
population. Nonetheless, current guidelines rec-
Periodic surveillance cultures are recommended to
ommend enteral feeding with standard dosing
detect bacterial and fungal pathogens as early as
regimes for critically ill patients according to possible. Antibiotic treatment should be initiated
dry/usual weight instead of actual weight [54]. promptly according to surveillance culture results
316 P. Nandhabalan et al.

at the earliest sign of active infection or deteriora- lism coupling. Propofol is a widely used agent
tion (progression to high grade hepatic encepha- in this context and may attenuate CBF more
lopathy or elements of the systemic inflammatory
response syndrome) effectively than benzodiazepines. Cerebral
All ALF patients are at risk for bacterial or fungal metabolic rate (CMRO2) is elevated with inad-
infection or sepsis, which may preclude liver equate anesthesia/sedation and will often be
transplantation or complicate the post-operative reflected by a low SjvO2. Infusion of an opiate
course. Prophylactic antibiotics and antifungals
have not been shown to improve overall outcomes such as fentanyl is commonplace for synergistic
in ALF and therefore cannot be advocated in all sedative effect, to facilitate endotracheal tube
patients, particularly those with mild hepatic tolerance, as an anti-­tussive agent, to attenuate
encephalopathy. surges in ICP. Opiates themselves have little
Further evidence against the indiscriminant use effect on cerebral metabolism and blood flow.
of antibiotic prophylaxis comes from a recent ret- Neuromuscular blockade is rarely required
rospective cohort analysis of over 1500 patients when adequate sedation and analgesia are used.
by the US Acute Liver Failure Study Group Neuromuscular blocking agents mask seizure
(US-ALFG). Here, antibiotic prophylaxis failed to activity and may be associated with the develop-
reduce the incidence of bloodstream infection or ment of critical care polyneuromyopathy. Their
improve survival [58]. However, less than half of routine use cannot be recommended, although
the study group comprised patients with high- practice varies between centers. They are gen-
grade encephalopathy and of these, only 55% erally used to prevent coughing, straining and
received antibiotic prophylaxis. The study also ventilator dys-synchrony and associated surges
confirmed that the presence of blood-­stream infec- in ICP. Lidocaine can be administered intrave-
tions was associated with a worse 21-day mortal- nously or via the tracheal tube prior to the appli-
ity, particularly in non-­acetaminophen ALF. cation of tracheal suction to attenuate coughing
When used, antimicrobial coverage should but is not necessarily common practice.
encompass commonly responsible organisms given
the likely site of infection, the known bacterial flora
of the intensive care unit at the time and the results Seizure Prophylaxis
of blood, urine and sputum cultures, chest radio-
graphs and other surveillance modalities. Further Grade III/IV encephalopathy is associated with a
details about infections and antibiotic treatment in high incidence of non-convulsive seizure activity.
liver disease and transplantation can be found else- Commonly used sedative agents such as propofol
where in this book. and benzodiazepines are well established in the
treatment of epilepsy and provide some degree of
prophylaxis/protection of the sedated and venti-
 edation and Neuromuscular
S lated ALF patient. Studies failed to show a bene-
Blockade fit of phenytoin in reducing seizures, cerebral
edema and improving survival [59], the prophy-
Sedation should be maintained in a continu- lactic use anti-epileptics is not recommended. If
ous manner and be maintained at a depth that BIS monitoring is used to assess the depth of
will prevent straining or coughing against the sedation, then discordant readings may prompt
ventilator. BIS monitoring to evaluate depth further evaluation with continuous EEG monitor-
of sedation is not routinely used and recom- ing. The latter should also be considered for neu-
mended. Intravenous anesthetic agents (with rological deteriorations and to assess burst
the exception of ketamine) decrease cerebral suppression when barbiturate coma is induced to
metabolism and reduce CBF via flow-metabo- treat refractory ICH.
25  The Patient with Severe Co-morbidities: CNS Disease and Increased Intracranial Pressure 317

Ammonia-Reducing Strategies ammonia; part of the effect can be explained by


temperature control and reduction in ammonia
Considering the strong correlation between ele- production. There is no evidence that prophylac-
vated arterial ammonia levels and the development tic use of CRRT in the absence of other indica-
of encephalopathy and ICH, ammonia-reducing tions for renal replacement therapy improves
strategies may be useful; however, there is no level outcome in patients with ALF and encephalopa-
1 evidence to support this practice. Many of the thy. However, a recent study demonstrated effec-
agents that are regarded effective in chronic liver tive reduction in arterial ammonia concentrations
disease have little data to support their use in ALF. within 24 h in patients with ALF and hyperam-
There are no randomized controlled trials of monemia [63]. Higher ultrafiltration rates were
lactulose administration in ALF, and it is often directly associated with greater ammonia clear-
poorly tolerated in critically ill patients receiving ance. The authors concluded that hyperammone-
high-dose sedation and analgesia, as reduced gut mia should be considered as an additional
motility frequently leads to worsening gaseous dis- indication for CRRT in critically ill adult patients
tension that may also cause technical difficulties with liver insufficiency. As well as controlling
during transplantation. The routine use of lactulose ammonia levels, early use of RRT can ameliorate
is therefore not necessarily recommended. biochemical disturbances, facilitate temperature
Neomycin, rifaximin and other non-­absorbable control and regulate fluid balance.
antibiotics, such as metronidazole, oral vanco-
mycin, paromomycin and oral quinolones, are
administered to patients with chronic cirrhosis in Extracorporeal Liver Assist Devices
an effort to decrease the colonic concentration of
ammoniagenic bacteria. While a large random- Several extracorporeal liver assist devices have
ized controlled trial has shown rifaximin to be been produced to support the failing liver until
effective in treating HE in chronic liver disease transplantation or native recovery occurs. These
[60], there is no strong evidence base supporting can broadly be classified into biological or non-­
its use or that of other non-absorbable antibiotics biological. The most extensively studied of these
in ALF. within the critically ill population is the Molecular
L-Ornithine-L-aspartate (LOLA) reduces the Adsorbent Recirculating System (MARS) device,
hyperammonemia of hepatic encephalopathy which uses albumin dialysis to remove both pro-
[61] by increasing ammonia detoxification in the tein and water-based toxins. Early studies dem-
muscle; however, there is no evidence of an out- onstrated an ability to reduce serum bilirubin and
come benefit. A placebo-controlled blinded study ammonia levels [64] however a randomized con-
randomized 201 patients with ALF to either pla- trolled trial of 102 patients with ALF showed no
cebo or LOLA infusions (30 g daily) for 3 days difference in outcome [65]. The high proportion
[62]. Arterial ammonia was measured at baseline of patients who underwent emergency LTx (66%)
and then daily for 6 days. There was no reduction in this trial within 24 h of randomization ensured
in mortality with LOLA treatment and no differ- that any meaningful conclusion with respect to
ence between the two groups in the improvement efficacy or lack thereof is difficult to reach. A
in encephalopathy grade, consciousness recovery trial comparing the use of a bioartificial liver
time, survival time or complications like seizures device using porcine hepatocytes with standard
and renal failure. treatment also failed to show a survival benefit
Continuous renal replacement therapy (CRRT) [66]. At this time most of the clinical evidence
is indicated for acute renal failure, oligo-anuria discourages the use of any type of extracorporeal
and acidemia and reduces circulating levels of liver assist devices in ALF.
318 P. Nandhabalan et al.

Plasma Exchange Hypertonic saline includes any concentra-


tion > 0.9% NaCl, but solutions used for osmoth-
High volume plasma exchange (HVPE) is effec- erapy in ALF are commonly 2.7–30%. The
tive in reducing arterial ammonia in patients with indications for hypertonic saline are similar to
ALF [67]. Recently, a large multicenter random- those of mannitol. It also acts by establishing an
ized controlled trial compared HVPE with stan- osmotic gradient across the blood–brain barrier
dard medical treatment in 182 patients with ALF with a subsequent reduction in brain water as
and at least moderate HE [68]. Plasma exchange water is drawn out of the brain parenchyma down
was associated with an increase in transplant-free its osmotic gradient. There is a biphasic reduc-
survival after 3 months, with a particular benefit tion in ICP, similar to that of mannitol.
seen in those patients that did not undergo LTx. Serum sodium levels of 145–155 mmol/L are
Concerns relating to worsening sepsis, cerebral commonly used as a target and reduce the inci-
edema or coagulopathy through immunomodula- dence of ICP rise above 25 mmHg. In practice,
tion of unidentified signaling pathways proved patients with FHF are often anuric and require
unfounded with no difference in complications continuous renal replacement therapy, so serum
between the two groups. This suggests that HVPE sodium levels rarely exceed these values even
may be a useful rescue therapy in patients in with prolonged infusion.
whom LTx is contraindicated, not readily avail- The osmotic-reflection coefficient across the
able or the subgroup of patients who fail to intact blood–brain barrier is higher (i.e. the
respond to initial management. BBB is less permeable) for hypertonic saline
than for mannitol. It is therefore less likely to
accumulate significantly in the brain paren-
Fluid Management and Osmotherapy chyma and, in theory, should be a more effec-
tive osmotic agent. It has been postulated that
Fluid management should be directed towards rebound ICH may be less severe with hyper-
the provision of adequate hydration and treat- tonic saline than with mannitol. Hypertonic
ment of hypovolemia. The blood–brain barrier saline also causes effective volume expansion
will allow the passage of fluids and electrolytes without a secondary diuresis.
along their osmotic gradients, and hypotonic flu- As mentioned, plasma osmolality should be
ids should therefore not be used as they have a kept below 320 mosmol/L, although this ­threshold
tendency to exacerbate cerebral edema. has recently been questioned and poorer outcomes
Osmotherapy is effective in attenuating cere- have only been associated with very high serum
bral edema. Mannitol and hypertonic saline are sodium levels and corresponding plasma osmolali-
both recommended for this purpose. Mannitol ties of 335–345 mosmol/L. Complications of
elicits a classically described biphasic response hypertonic saline relate to the administration (tis-
[69]. There is an early fall in ICP as blood rheol- sue necrosis, thrombophlebitis) and metabolic side
ogy improves. The improved blood flow enhances effects (hyperchloremic acidosis, hypokalemia,
oxygen delivery and, via flow/metabolism cou- hypocalcemia). Osmotic myelinolysis may be pre-
pling, results in cerebral vasoconstriction. A sub- cipitated if serum sodium concentrations are cor-
sequent decrease in ICP is observed approximately rected too rapidly.
30 min later as mannitol increases plasma osmo- Recent meta-analyses have suggested that
lality and draws brain water across the blood– hypertonic saline is superior to mannitol in reduc-
brain barrier down its osmotic gradient. Mannitol ing ICP in brain-injured patients [70, 71]. Clinical
also acts as an oxygen free-radical scavenger. trials are in progress comparing the two therapies
Plasma osmolality should not exceed 320 mos- in patients with ALF and the results are pending.
mol/kg. There is a risk of fluid overload in oligu- For now, guidelines suggest prophylactic induc-
ric/anuric patients in which case RRT may be tion of hypernatremia to 145–155 mEq/L with
required. hypertonic saline in patients at highest risk for
25  The Patient with Severe Co-morbidities: CNS Disease and Increased Intracranial Pressure 319

cerebral edema and bolus doses of mannitol as lation threshold or if autoregulatory mechanisms
first line therapy for ICP > 25 mmHg [28]. have failed altogether and CBF is proportional to
CPP. This may be desirable, but risks exacerbat-
ing ICH through increased cerebral blood flow
Therapies Targeting Cerebral and blood volume and worsening cerebral edema
Perfusion Pressure (CPP) (increased hydrostatic pressures). Increasing
CPP can also cause cerebral vasoconstriction
Under normal conditions, autoregulatory mecha- (thus lowering the ICP) if autoregulation is intact
nisms ensure that CBF remains constant at (Fig. 25.3).
approximately 50 mL/100 g brain tissue/min The target CPP has been the subject of some
within a CPP range 50–150 mmHg. In the injured controversy. In polytrauma cases at risk of raised
brain, the relationship between CPP and CBF ICP, a MAP of 90 mmHg has traditionally been tar-
changes—the autoregulation curve tends to shift geted in patients without ICP monitoring. In
to the right, so that a CPP > 50 mmHg may be patients with ICP monitoring, a target CPP of
required to maintain flow and normal autoregula- 70 mmHg was initially recommended by the Brain
tion may be disrupted, such that CBF becomes Trauma Foundation in 1995. A contrasting view is
proportional to CPP. that setting a higher CPP target will worsen brain
General principles of fluid management apply, edema by increasing the hydrostatic pressure gradi-
and fluid therapy is perhaps best guided by the ents across tissue beds. There is also some evidence
appropriate use of cardiac output monitoring that targeting higher CPPs may promote the devel-
that can provide dynamic measures of preload opment of ARDS [72], although the underlying
responsiveness and indicate whether or not stroke mechanism is unclear. This led to the Brain Trauma
volume improves in response to increased filling. Foundation to lower the target CPP to 60 mmHg in
Injudicious use of fluids may worsen cerebral TBI. The brain tissue oxygen partial pressure
edema and associated lung injury. If hemody- (PbO2) may plateau at a CPP of 60 mmHg [73].
namic optimization with fluid therapy alone fails The current AASLD guidelines suggest maintain-
to achieve adequate mean arterial pressures in the ing CPP between 60 – 80 mmHg or a mean arterial
face of diminished systemic vascular resistance, pressure ≥ 75 but there is little evidence to support
vasopressors may be required to augment the this. Others advocate accepting a lower target of
CPP. CPP > 50 mmHg [74].
Increasing CPP may increase CBF, particu- Whilst continuing to note the dangers of a CPP
larly in injured regions of the brain, but this will >70 or <50 mmHg, a recent recommendation is to
only occur if CPP has fallen below the autoregu- monitor markers of cerebral oxygenation and

75
Maximum
Cerebral Blood Flow (ml/100 g/min)

Passive Zone of Normal Maximum


Collapse Dilatation Autoregulation Constriction

Fig. 25.3 The 50
relationship between
systolic blood pressure
(SBP) and cerebral
blood flow (CBF) in 25
the uninjured brain.
The ability to
autoregulate blood flow
may be lost in brain
injury, and flow may 0
become pathologically 0 25 50 75 100 125 150
dependent on pressure Systolic Blood Pressure (mmHg)
320 P. Nandhabalan et al.

metabolism to adopt an individualized approach benefit. Other complications of sodium thio-


to therapy within the CPP range of 50–70 mmHg pental therapy include immunosuppression,
[75]. Autoregulation thresholds vary over time; bronchoconstriction, electrolyte disturbances
hence, CPP goals have to be adapted to changing (notably profound hypokalemia), renal impair-
clinical conditions. ment (reduced renal blood flow and increased
The choice of vasopressor has not been sub- ADH secretion) and ileus.
ject of controlled clinical trials. Norepinephrine After prolonged infusion, the metabolism of
is generally considered the first-line agent as it sodium thiopental becomes ‘zero order’—the
may best augment peripheral organ perfusion hepatic enzyme systems responsible for its metabo-
whilst preserving splanchnic blood flow. Low-­ lism become overwhelmed and the lipid-­soluble
dose vasopressin is increasingly used following drug accumulates in tissues such as fat and muscle.
experience in septic shock and TBI patients. The duration of action is therefore greatly prolonged
Early concerns regarding increase in cerebral and ‘washout’ of the drug takes considerable time.
hyperemia with use of vasopressin or vasopressin In addition, sodium thiopental is partly metabolized
analogues are probably unfounded and recent to pentobarbitone that has a longer half-life than
evidence demonstrates the ability of terlipressin sodium thiopental itself.
to increase CPP and cerebral perfusion without
an associated increase in ICP [76]. Epinephrine is
poorly tolerated due to its effect on aerobic gly- Indomethacin
colysis and associated worsening of lactic acido-
sis. Relative adrenal insufficiency is common in Indomethacin, a non-selective cyclo-oxygenase
ALF, patients with ongoing haemodynamic insta- inhibitor, has been used in the treatment of refrac-
bility should undergo short SynACTHen testing tory cerebral hyperemia [77, 78]. Doses of 25 mg
and receive supplemental steroid therapy. iv over 1 min may have a vasoconstrictor effect,
Whether steroid replacement improves outcome although in these circumstances, CBF may actu-
remains unclear. ally increase (as measured by transcranial
Doppler) as ICP is reduced and CPP is restored.
Indomethacin has been used more extensively in
 trategies for Treating Refractory
S traumatic ICH, in patients with space occupying
Increases in ICP lesions and animal models and its use is not
widely reported in ALF. Therapeutic benefit with
Barbiturate Coma indomethacin in ALF has been attributed to its
anti-inflammatory effects and potentially lends
Barbiturates can be titrated to induce burst support to the neuro-inflammatory theory of the
suppression of the EEG and decrease cerebral pathogenesis of HE.
metabolism (CMRO2) and cerebral blood flow
by virtue of flow-metabolism coupling. Sodium
thiopental can be used as ‘rescue therapy’ to Therapeutic Hypothermia (TH)
lower ICP refractory to other measures. A load-
ing dose of 5–10 mg/kg of sodium thiopental Hypothermia antagonizes the pathogenesis of HE
is required, followed by a continuous infusion through multiple mechanisms including reduc-
of 3–5 mg/kg/h. EEG monitoring should be ing intestinal production and cerebral uptake
used to guide further therapy. Increasing doses of ammonia, attenuating extracellular accumu-
above those required for burst suppression lation of glutamate and lactate within the brain
causes unwanted side effects such as arterial and decreasing microglial activation as well as
hypotension through negative inotropy and a improving hemodynamic variations and reducing
lowering of systemic vascular resistance (dose- cerebral hyperemia [79]. Cooling the patient’s
dependent) without conferring any additional core temperature to as low as 32–33 °C reduces
25  The Patient with Severe Co-morbidities: CNS Disease and Increased Intracranial Pressure 321

otherwise refractory elevation in ICP in patients The study was stopped prematurely due to the
with ALF [80]. CPP improves as a result of likely outcome already having been demonstrated.
diminished ICP. However, hypothermia has sev- There was no difference in the incidence of clini-
eral deleterious systemic effects, including coag- cally significant elevations in ICP with an unex-
ulopathy, immune suppression, insulin resistance pected trend towards increased ICP’s in the
and an increased risk in nosocomial infections— intervention group. Mortality was similar between
particularly ventilator-associated pneumonia. the two groups (41% vs. 46% in controls) although
Prolonged hypothermia in patients not progress- the study was not powered to detect a difference
ing to transplantation requires the use of lengthy in survival. Although a temperature of 34 °C was
periods of deep sedation and/or paralysis to targeted in the intervention group, values achieved
attenuate shivering. Mild to moderate hypother- were closer to 33 °C. These results are consistent
mia, targeting temperatures of 35–36 °C, have with those of recent large randomized controlled
been suggested as a reasonable compromise. ICP trials of therapeutic hypothermia that also failed
is reduced, although perhaps not as effectively to show any outcome benefit, both for patients
as with more profound cooling techniques, and with TBI [83] and following out of hospital car-
ammonia production is less affected, but the del- diac arrest [84].
eterious consequences of profound hypothermia In summary, there remains a discrepancy
are minimized. Allowing a passive decline of between the substantial body of experimental
core temperature using an extracorporeal circuit evidence indicating a benefit for the use of thera-
is a simple way of inducing and maintaining mild peutic hypothermia for the treatment of ICH in
hypothermia. patients with ALF and the results of contempo-
Despite the abundance of animal studies and rary clinical studies. Although the latter have
case series in favor of hypothermia, more robust failed to demonstrate its efficacy, evidence relat-
clinical studies have failed to demonstrate ing to the safety of TH has been reassuring. Most
improved outcomes with therapeutic hypother- institutions continue to endorse moderate hypo-
mia in ALF. A retrospective multi-center cohort thermia in ALF patients at risk of cerebral edema
study by the USALF Study Group of over 1200 and as rescue therapy in those with refractory
patients with ALF and Grade 3 or 4 HE compared ICH. Further evidence is required to identify
97 patients who were cooled to 32–35 °C for a which patients may benefit and how this inter-
median of 2 days with the remaining group [81]. vention should be used.
There was no difference in 21-day mortality or If hypothermia is used it is mandatory to con-
transplant-free survival rates although therapeu- tinue to do so in the operating room at least until
tic hypothermia was tolerated well with no excess reperfusion if the patient proceeds for LTx to
in infectious and non-infectious complications. avoid rebound ICH.
Possible explanations include the limitations of
retrospective study design, potential greater ben-
efit for the subgroup of hyperacute presentations Hepatectomy
and whether the intervention was intended as
treatment for ICH or prophylaxis. Refractory increases in ICP have been treated by
The only prospective randomized controlled total hepatectomy with portocaval shunt creation
trial of therapeutic hypothermia in ALF was pub- as a bridge to LTX [85]. Marked reductions of
lished in 2016 [82]. The authors hypothesized that ICP following removal of the toxic liver supports
induction of moderate hypothermia may prevent the postulate that pro-inflammatory cytokines are
or delay the onset of ICH in patients with ALF, involved in the pathogenesis of cerebral edema
high grade HE and ICP monitoring in situ. They and/or hyperemia in ALF. Anhepatic periods of
compared 17 patients who underwent targeted up to 60 h have been described without perma-
temperature management to 34 °C with 26 control nent neurological deficit. The procedure may be
patients treated to a temperature of 36 °C.  lifesaving for extreme cases but requires the
322 P. Nandhabalan et al.

availability of a transplantable organ within a raise ICP. However, others have demonstrated that
short time. Nonetheless, good outcomes are still cerebral perfusion can be preserved without VVB
achievable; a case series recently reported that in such patients [90]. The procedure itself carries
only one of six patients treated with two stage a small but significant risk although the incidence
liver transplantation died [86]. of complications has fallen considerably to around
2% [91]. A recent meta-analysis attempted to
evaluate the efficacy of VVB but was limited by
Intra-Operative Considerations lack of robust clinical outcome data [92]. Overall
the use of piggyback technique for LTx and avoid-
Patients are at risk of brain herniation intra-­ ance of complete caval clamping has reduced the
operatively as well as during the peri-operative need for routine VVB. In most centers VVB is
phases. In an analysis of 116 FHF patients, 13 reserved for selected cases only and not used rou-
(11.2%) developed brain death during or shortly tinely even for patients with ALF.
after LTX [87], and the exact timing of the neuro-
logical insult is unclear.
Detry et al. observed that of 12 patients trans- The Neurology of 
planted for FHF, the four patients with normal Chronic Liver Disease
preoperative ICPs maintained normal pressures
intra-operatively [22]. Of the eight patients with Brain edema and ICH are not common features
preoperative episodes of increased ICP, four of terminal chronic liver failure, although occa-
patients developed six episodes of ICH during sional cases have been reported in the literature.
surgery. The dissection and reperfusion phases Indeed, radiographic evidence of cerebral
were most frequently associated with cerebral edema with CT scanning was seen in only 4% of
insufficiency secondary to surges in ICP and con- patients with acute on chronic liver failure in a
sequent reduction in CPP. The anhepatic phase recent retrospective study at our institution [93].
was associated with a decrease of the ICP. At the There were no cases of cerebral edema in the
end of the anhepatic phase, the ICP was lower chronic liver disease group despite high-grade
preoperatively in all patients and below 15 mmHg encephalopathy. Of note, the onset of hepatic
in all but one patient. encephalopathy in patients with chronic liver
This observation is in concordance with a illness is indicative of severe progressive dis-
small study of six cases from King’s College ease with poor outcomes unless transplantation
Hospital, London that demonstrated higher ICP is offered. In a study by Jepsen et al., 1-year
pre-anhepatic and during graft reperfusion and mortality in patients with cirrhotic liver disease
similarly reduced ICP during the anhepatic phase who developed HE was 64% [94]. Independent
[88]. Lidofsky et al. noted that thiopental treat- risk factors of mortality in patients with HE
ment was most frequently required during liver include age, bilirubin, creatinine and grade of
dissection, but ICP invariably normalized within encephalopathy.
15 min of caval cross-clamping [89]. This group Clinical symptoms and cerebral edema are less
also noted transient rises in ICP at the time of severe with chronic liver disease compared with
graft reperfusion. ALF since encephalopathy in chronic liver disease
The use of veno-venous bypass (VVB) during progresses more slowly and adaptive responses
LTx has been advocated to maintain cerebral per- can develop. The distribution of edema differs in
fusion. It has been suggested that the lack of ade- chronic liver disease; excess brain water is mostly
quate collateral venous circulation leads to intracellular with ALF, whereas with chronic liver
hemodynamic instability and require volume disease, it is mostly extracellular. This may result
replacement that can exacerbate cerebral edema. from the loss of organic solute and water from
Furthermore, the release of CO2 during reperfu- cells with restoration of volume and minimal
sion can exacerbate cerebral vasodilatation and effect on function.
25  The Patient with Severe Co-morbidities: CNS Disease and Increased Intracranial Pressure 323

The pathogenesis of HE in chronic liver dis- and severity of the decrease of serum sodium
ease shares some common features with those levels. A gradual drop, even to very low levels,
in ALF. However, the relative importance of may be tolerated well if it occurs over several
contributing factors may differ. Shawcross et al. days or weeks. However serum sodium levels of
demonstrated that systemic inflammation and <120 mmol/L can significantly lower the seizure
infection are closely associated with the develop- threshold, and serum sodium concentration is an
ment of HE in patients with liver cirrhosis [95]. independent predictor of EEG abnormalities in
More recently, a nested prospective cohort study patients with HE. Lethargy, seizures and coma
of 101 patients with acute on chronic liver fail- may be seen with variable frequency with serum
ure identified a strong correlation between sever- sodium levels <110 mmol/L. In addition to
ity of encephalopathy and increasing ammonia hyponatremia associated osmotic disequilibrium
levels in the face of conflicting evidence from resulting in astrocyte swelling, the generation of
previous studies [39]. The authors reported the action potential and synaptic transmission
an association between persistent hyperam- are also dependent on ionic gradients and the
monaemia and increased mortality as well as movement of sodium down its electrochemical
improvement in HE with a concurrent reduction gradient through Na-specific voltage-gated ion
in ammonia levels. Raised markers of systemic channels.
inflammation were strongly predictive of death The resolution of hyponatremia in cirrhotics
but this was not specific to those patients with leads to improvement in related neurological
HE. Furthermore, disturbances in jugular venous symptoms. To avoid hyponatremia, causes such
bulb oximetry were associated with grade of HE as diuretic use, infusion of hypotonic fluids and
and higher mortality suggesting that abnormal gastrointestinal losses due to diarrhea or medica-
cerebral oxygenation may be a relevant fac- tion (lactulose, enema) should be considered. It is
tor in the pathophysiology of HE in this group important to distinguish between hypovolemic
of patients. and hypervolemic hyponatremia, as this will
determine whether saline infusion or fluid restric-
tion is the appropriate treatment.
 he Patient with Severe
T In ALF, osmotherapy with hypertonic saline
Hyponatremia and CNS Dysfunction infusion increases serum sodium to levels of
145–155 mmol/L and is associated with a reduc-
Hyponatremia is common, both in patients with tion in the incidence and severity of episodes of
cirrhosis and ALF, and occurs, among other rea- ICH. In chronic cirrhosis with hepatic encepha-
sons as a result of hypersecretion of antidiuretic lopathy and hyponatremia, saline infusions may
hormone. Early postoperative morbidity and mor- be administered if signs of hypovolemia or
tality is increased in patients with low serum recent diuretic use are evident. Under these cir-
sodium levels undergoing liver transplantation cumstances, paracentesis may be the preferred
[96]. Hyponatraemia is associated with increased treatment modality for resistant ascites. Sodium
frequency of HE in patients with cirrhosis. A pos- levels should be normalized as much as possible
sible mechanism suggests astrocyte swelling as a prior to liver transplantation in hyponatremic
result of intracellular glutamine accumulation is patients to avoid rapid sodium shifts during sur-
followed by a second osmotic insult with hypona- gery. Sodium levels above 130 mmol/L are usu-
tremia leading to exhaustion of counteractive sys- ally considered safe for LTx but little is known
tems and low grade cerebral edema [97]. about the effect of more severe hyponatremia on
Hyponatremia in combination with hepatic outcome after LTx and if preoperative correction
encephalopathy leads to a clinical picture of improves outcome.
confusional syndrome and is similar to other Intra-operative increases of sodium concen-
metabolic encephalopathies. The severity of trations are common and associated with
neurological symptoms correlates with the speed increased ICU and hospital length of stay [98].
324 P. Nandhabalan et al.

Occasionally pre- and intra-operative CRRT is serum sodium concentration can precipitate osmotic
indicated to prevent postoperative neurological myelinolysis that can cause profound and often per-
complications. manent neurological deficits. Severe damage of the
Vaptans are a class of selective vasopressin-2 myelin sheath of nerve cells in the corticobulbar and
receptor antagonists that induce dose-dependent corticospinal tracts of the brainstem may cause
solute-free water excretion and have been used quadriparesis, dysphagia, dysarthria, diplopia, loss
in the treatment of hypervolemic hyponatrae- of consciousness and locked-in syndrome. Central
mia. A recent meta-analysis demonstrated a sig- Pontine Myelinolysis occurs in 1–2% patients fol-
nificant effect in both improving serum sodium lowing liver transplantation although its true preva-
levels and treatment of refractory ascites in lence may be under-reported [101]. The MRI in
patients with cirrhosis [99]. Although these Fig. 25.4 is of a patient who underwent LTX with
effects are likely to be short-lived, vaptans can serum sodium 128 mmol/L. Subsequent to LTx, the
be considered in patients with refractory hypo- serum sodium rose to 135 mmol/L. The patient was
natremia prior to transplantation or those who extubated successfully but underwent re-laparot-
remain symptomatic despite conventional treat- omy the following day for ongoing blood loss with
ment [100]. consequent infusion of colloid, packed cells and
The rapidity of correction of hyponatremia is blood products. The day following re-laparotomy,
based on the speed of onset. If the speed is not the serum sodium had risen to 142 mmol/L and
known, slow rise in serum sodium concentration at there was an associated deterioration in respiratory
a rate of <0.5 mmol/L/h is advisable. Rapid rises in

Fig. 25.4  There is a large, central area of high-T2 signal abnormality in the pons in keeping with osmotic
myelinolysis
25  The Patient with Severe Co-morbidities: CNS Disease and Increased Intracranial Pressure 325

function and GCS. Pontine myelinolysis is evident tle radiological changes may take months to
on the MRI. become evident. Furthermore, the high signal
intensity changes in the basal ganglia as a result
of possible manganese deposition that are asso-
 eurological Outcomes After
N ciated with HE also appear to diminish follow-
Liver Transplantation ing liver transplantation [107].
A number of studies have documented an
Neurological complications are common fol- improvement in cognitive function following LTx
lowing liver transplantation with an incidence and an improvement in quality of life index mark-
of 13–47% [102, 103]. They occur in part due ers. This is not always the case and for a substan-
to co-­morbidities present at the time of surgery tial number of patients, cognitive deficits persist
(HE, hepatitis C, arterial hypertension, etc.) long into the postoperative period. The etiology
and are more common in alcoholic liver dis- for this is likely to be multifactorial but include
ease and primary biliary cirrhosis. Neurological the presence of hepatic encephalopathy pre-trans-
morbidity post-transplant has traditionally been plant, subsequent neuronal loss, brain atrophy
attributed to opportunistic infections and immu- (commonly seen in cirrhosis), presence of cere-
nosuppressant neurotoxicity with or without bral small vessel disease pre-transplant, perioper-
seizure activity but recent studies suggest these ative vascular complications, immunosuppression
may have been superseded by cerebrovascular (especially calcineurin inhibitors) and the persis-
events [104]. Other important causes include tence of portosystemic collaterals that take time to
seizures, metabolic disturbances, neuromuscular resolve. Indeed, in a prospective study of neuro-
disorders and central pontine myelinolysis. Such psychological function before and after LTx, the
complications increase morbidity, mortality and incidence of post-transplant cognitive dysfunc-
hospital stay. Recommendations have been pub- tion was 13% and was associated with preopera-
lished in order to guide prevention, diagnosis tive HE and post-operative loss of brain volume
and treatment [105]. [108]. Persisting cognitive dysfunction is associ-
Intracranial hypertension usually resolves ated with co-morbidities such as diabetes mel-
within 48 h following liver transplantation litus, arterial hypertension, hyperlipidemia and
assuming that the graft is functioning well. In increasing age (all associated with other causes of
those with marked encephalopathy prior to trans- neuronal loss such as small vessel disease).
plantation, neurological outcome is in general
favorable, with eventual improvement of cogni-
tive function in the majority of patients; however,  ther Hepatic Diseases
O
resolution of neurological symptoms may be slow with Cerebral Manifestation:
in some and persist in a minority. Encephalopathy Wilson’s Disease and Acquired
post-transplantation is not uncommon with one Hepatocerebral Degeneration
study reporting an incidence of 23% within the
first month of LTx [106]. Preoperative infection is Wilson’s disease is an autosomal recessive disor-
an independent risk factor for postoperative neu- der of chromosome 13 that results in defective
rological complications. biliary copper excretion and copper accumula-
The most tangible radiological evidence for tion in the tissues. It was first described by Dr.
the resolution of cerebral edema comes from Samuel Alexander Kinnier Wilson, a professor of
magnetic resonance imaging that demonstrates neurology at King’s College Hospital, London.
an increase in the volume of the ventricles in Most of the symptoms are attributable to the
association with an improvement in neurologi- deposition of copper through the body. Patients
cal and cognitive function after liver transplan- present early with liver disease or late with the
tation (and is therefore unlikely due to an neurological syndrome that consists of neuropsy-
absolute loss of brain parenchyma). These sub- chiatric symptoms and movement disorders.
326 P. Nandhabalan et al.

Acquired (non-Wilsonian) hepatocerebral 7. Rama Rao KV, Jayakumar AR, Norenberg MD. Brain
degen­eration (AHD) is a chronic progressive edema in acute liver failure: mechanisms and con-
cepts. Metab Brain Dis. 2014;29(4):927–36.
neurological syndrome in patients with porto- 8. Kato M, Hughes RD, Keays RT, Williams
systemic shunts characterized by dementia, dys- R. Electron microscopic study of brain capillaries
arthria, ataxia of gait, intention tremor and in cerebral edema from fulminant hepatic failure.
choreoathetosis (i.e. neuropsychiatric and extra- Hepatology. 1992;15(6):1060–6.
9. Swain M, Butterworth RF, Blei AT. Ammonia and
pyramidal symptomatology). AHD and Wil- related amino acids in the pathogenesis of brain edema
son’s disease are often mistaken—the diagnosis in acute ischemic liver failure in rats. Hepatology.
depends on age of onset (Wilson’s usually pres- 1992;15(3):449–53.
ents <30 years), serum caeruloplasmin concen- 10. Record CO, Buxton B, Chase RA, Curzon G,
Murray-Lyon IM, Williams R. Plasma and brain
tration and the presence of Kayser-Fleischer amino acids in fulminant hepatic failure and their
rings in the latter. MRI appearances may help to relationship to hepatic encephalopathy. Eur J Clin
distinguish between the two conditions. The Investig. 1976;6(5):387–94.
disease is associated with multiple metabolic 11. Tofteng F, Hauerberg J, Hansen BA, Pedersen CB,
Jorgensen L, Larsen FS. Persistent arterial hyperam-
insults and has been variously linked to the fail- monemia increases the concentration of glutamine
ure of clearance of toxins such as ammonia and and alanine in the brain and correlates with intra-
manganese. Microscopically, there is patchy cranial pressure in patients with fulminant hepatic
cortical necrosis, diffuse proliferation of failure. J Cereb Blood Flow Metab. 2006;26(1):
21–7.
Alzheimer type II glial cells and neuronal loss. 12. Albrecht J, Norenberg MD. Glutamine: a Trojan horse
Treatment options remain limited, although in ammonia neurotoxicity. Hepatology. 2006;44(4):
rifaximin has been used to successfully amelio- 788–94.
rate symptoms [109]. In addition, a recent case 13. Desjardins P, Du T, Jiang W, Peng L, Butterworth
RF. Pathogenesis of hepatic encephalopathy and
series described both sustained clinical and brain edema in acute liver failure: role of glutamine
radiographic reversal of AHD with living donor redefined. Neurochem Int. 2012;60(7):690–6.
liver transplantation [110]. 14. Ranjan P, Mishra AM, Kale R, Saraswat VA, Gupta
RK. Cytotoxic edema is responsible for raised intra-
cranial pressure in fulminant hepatic failure: in vivo
demonstration using ­diffusion-­weighted MRI in
human subjects. Metab Brain Dis. 2005;20(3):
References 181–92.
15. Rai V, Nath K, Saraswat VA, Purwar A, Rathore RK,
1. O’Grady JG, Alexander GJ, Hayllar KM, Williams Gupta RK. Measurement of cytotoxic and intersti-
R. Early indicators of prognosis in fulminant hepatic tial components of cerebral edema in acute hepatic
failure. Gastroenterology. 1989;97(2):439–45. failure by diffusion tensor imaging. J Magn Reson
2. Ware AJ, D’Agostino AN, Combes B. Cerebral Imaging. 2008;28(2):334–41.
edema: a major complication of massive hepatic 16. Nguyen JH. Blood-brain barrier in acute liver fail-
necrosis. Gastroenterology. 1971;61(6):877–84. ure. Neurochem Int. 2012;60(7):676–83.
3. Bernal W, Hyyrylainen A, Gera A, Audimoolam VK, 17. Cauli O, Lopez-Larrubia P, Rodrigo R, Agusti A,
McPhail MJ, Auzinger G, et al. Lessons from look-­ Boix J, Nieto-Charques L, et al. Brain region-­
back in acute liver failure? A single centre experi- selective mechanisms contribute to the progression
ence of 3300 patients. J Hepatol. 2013;59(1):74–80. of cerebral alterations in acute liver failure in rats.
4. Bernal W, Hall C, Karvellas CJ, Auzinger G, Sizer E, Gastroenterology. 2011;140(2):638–45.
Wendon J. Arterial ammonia and clinical risk factors 18. Coltart I, Tranah TH, Shawcross DL. Inflammation
for encephalopathy and intracranial hypertension in and hepatic encephalopathy. Arch Biochem Biophys.
acute liver failure. Hepatology. 2007;46(6):1844–52. 2013;536(2):189–96.
5. Kumar R, Shalimar Sharma H, Prakash S, Panda 19. Jalan R, Olde Damink SWM, Hayes PC, Deutz NEP,
SK, Khanal S, et al. Persistent hyperammonemia is Lee A. Pathogenesis of intracranial hypertension in
associated with complications and poor outcomes in acute liver failure: inflammation, ammonia and cere-
patients with acute liver failure. Clin Gastroenterol bral blood flow. J Hepatol. 2004;41(4):613–20.
Hepatol. 2012;10(8):925–31. 20. Olafsson S, Gottstein J, Blei AT. Brain edema and
6. Clemmesen JO, Larsen FS, Kondrup J, Hansen BA, intracranial hypertension in rats after total hepatec-
Ott P. Cerebral herniation in patients with acute liver tomy. Gastroenterology. 1995;108(4):1097–103.
failure is correlated with arterial ammonia concen- 21. Arkadopoulos N, Chen SC, Khalili TM, Detry O,
tration. Hepatology. 1999;29(3):648–53. Hewitt WR, Lilja H, et al. Transplantation of hepa-
25  The Patient with Severe Co-morbidities: CNS Disease and Increased Intracranial Pressure 327

tocytes for prevention of intracranial hypertension 36. Peck M, Wendon J, Sizer E, Auzinger G, Bernal
in pigs with ischemic liver failure. Cell Transplant. W. Intracranial pressure monitoring in acute liver
1998;7(4):357–63. failure: a review of 10 years experience. Crit Care.
22. Detry O, Arkadopoulos N, Ting P, Kahaku E, 2010;14(1):1.
Margulies J, Arnaout W, et al. Intracranial pressure 37. Kamat P, Kunde S, Vos M, Vats A, Gupta N, Heffron
during liver transplantation for fulminant hepatic T, et al. Invasive intracranial pressure monitoring is
failure. Transplantation. 1999;67(5):767–70. a useful adjunct in the management of severe hepatic
23. Jalan R, Pollok A, Shah SH, Madhavan K, Simpson encephalopathy associated with pediatric acute liver
KJ. Liver derived pro-inflammatory cytokines may failure. Pediatr Crit Care Med. 2012;13(1):e33–8.
be important in producing intracranial hypertension 38. Sheinberg M, Kanter MJ, Robertson CS, Contant
in acute liver failure. J Hepatol. 2002;37(4):536–8. CF, Narayan RK, Grossman RG. Continuous moni-
24. Larsen FS, Ejlersen E, Hansen BA, Knudsen GM, toring of jugular venous oxygen saturation in head-­
Tygstrup N, Secher NH. Functional loss of cerebral injured patients. J Neurosurg. 1992;76(2):212–7.
blood flow autoregulation in patients with fulminant 39. Sawhney R, Holland-Fischer P, Rosselli M, Mookerjee
hepatic failure. J Hepatol. 1995;23(2):212–7. RP, Agarwal B, Jalan R. Role of ammonia, inflamma-
25. Blei AT. The pathophysiology of brain tion, and cerebral oxygenation in brain dysfunction of
edema in acute liver failure. Neurochem Int. acute-on-chronic liver failure patients. Liver Transpl.
2005;47(1–2):71–7. 2016;22(6):732–42.
26. Rama Rao KV, Jayakumar AR, Tong X, Alvarez VM, 40. Bellner J, Romner B, Reinstrup P, Kristiansson KA,
Norenberg MD. Marked potentiation of cell swelling Ryding E, Brandt L. Transcranial Doppler sonogra-
by cytokines in ammonia-sensitized cultured astro- phy pulsatility index (PI) reflects intracranial pres-
cytes. J Neuroinflammation. 2010;7(1):1–8. sure (ICP). Surg Neurol. 2004;62(1):45–51.
27. Bémeur C, Qu H, Desjardins P, Butterworth RF. IL-1 41. Kawakami M, Koda M, Murawaki Y. Cerebral pul-
or TNF receptor gene deletion delays onset of enceph- satility index by transcranial Doppler sonography
alopathy and attenuates brain edema in experimental predicts the prognosis of patients with fulminant
acute liver failure. Neurochem Int. 2010; 56(2):213–5. hepatic failure. Clin Imaging. 2010;34(5):327–31.
28. Lee WM, Stravitz RT, Larson AM. Introduction to 42. Abdo A, Perez-Bernal J, Hinojosa R, Porras F,
the revised American Association for the Study of Castellanos R, Gomez F, et al. Cerebral hemo-
Liver Diseases Position Paper on acute liver failure dynamics patterns by transcranial Doppler in
2011. Hepatology. 2012;55(3):965–7. patients with acute liver failure. Transplant Proc.
29. Munoz SJ, Robinson M, Northrup B, Bell R, Moritz 2015;47(9):2647–9.
M, Jarrell B, et al. Elevated intracranial pressure and 43. Aggarwal S, Brooks DM, Kang Y, Linden PK,
computed tomography of the brain in fulminant hepa- Patzer JF. Noninvasive monitoring of cerebral per-
tocellular failure. Hepatology. 1991;13(2):209–12. fusion pressure in patients with acute liver failure
30. Traber PG, Ganger DR, Blei AT. Brain edema in rab- using transcranial doppler ultrasonography. Liver
bits with galactosamine-induced fulminant hepatitis. Transpl. 2008;14(7):1048–57.
Regional differences and effects on intracranial pres- 44. Nielsen HB, Tofteng F, Wang LP, Larsen
sure. Gastroenterology. 1986;91(6):1347–56. FS. Cerebral oxygenation determined by near-infra-
31. Detry O, De Roover A, Honore P, Meurisse M. Brain red spectrophotometry in patients with fulminant
edema and intracranial hypertension in fulminant hepatic failure. J Hepatol. 2003;38(2):188–92.
hepatic failure: pathophysiology and management. 45. Helmke K, Burdelski M, Hansen HC. Detection
World J Gastroenterol. 2006;12(46):7405–12. and monitoring of intracranial pressure dysregula-
32. Davies MH, Mutimer D, Lowes J, Elias E, tion in liver failure by ultrasound. Transplantation.
Neuberger J. Recovery despite impaired cerebral 2000;70(2):392–5.
perfusion in fulminant hepatic failure. Lancet. 46. Krishnamoorthy V, Beckmann K, Mueller M,
1994;343(8909):1329–30. Sharma D, Vavilala MS. Perioperative estimation of
33. Chesnut RM, Temkin N, Carney N, Dikmen S, the intracranial pressure using the optic nerve sheath
Rondina C, Videtta W, et al. A trial of intracranial- diameter during liver transplantation. Liver Transpl.
pressure monitoring in traumatic brain injury. N 2013;19(3):246–9.
Engl J Med. 2012;367(26):2471–81. 47. Bratton SL, Chestnut RM, Ghajar J, McConnell
34. Vaquero J, Fontana RJ, Larson AM, Bass NM, Hammond FF, Harris OA, Hartl R, et al. Guidelines
Davern TJ, Shakil AO, et al. Complications and use for the management of severe traumatic brain injury.
of intracranial pressure monitoring in patients with VIII. Intracranial pressure thresholds. J Neurotrauma.
acute liver failure and severe encephalopathy. Liver 2007;24(Suppl 1):S55–8.
Transpl. 2005;11(12):1581–9. 48. Kredel M, Muellenbach RM, Brock RW, Wilckens
35. Karvellas CJ, Fix OK, Battenhouse H, Durkalski V, HH, Brederlau J, Roewer N, et al. Liver dysfunction
Sanders C, Lee WM. Outcomes and complications after lung recruitment manoeuvres during pressure-­
of intracranial pressure monitoring in acute liver controlled ventilation in experimental acute respira-
failure: a retrospective cohort study. Crit Care Med. tory distress. Crit Care. 2007;11(1):R13.
2014;42(5):1157–67.
328 P. Nandhabalan et al.

49. Ede RJ, Gimson AE, Bihari D, Williams R. Controlled 62. Acharya SK, Bhatia V, Sreenivas V, Khanal S,
hyperventilation in the prevention of cerebral oedema in Panda SK. Efficacy of L-ornithine L-aspartate in
fulminant hepatic failure. J Hepatol. 1986;2(1):43–51. acute liver failure: a double-blind, randomized,
50. Kodakat S, Gopal P, Wendon J. Hyperglycaemia is placebo-controlled study. Gastroenterology. 2009;
associated with intracranial hypertension in patients 136(7):2159–68.
with acute liver failure. Liver Transpl. 2001;7:C-21. 63. Slack AJ, Auzinger G, Willars C, Dew T, Musto
51. Sonneville R, Vanhorebeek I, den Hertog HM, R, Corsilli D, et al. Ammonia clearance with hae-
Chrétien F, Annane D, Sharshar T, et al. Critical mofiltration in adults with liver disease. Liver Int.
illness-induced dysglycemia and the brain. Intensive 2014;34(1):42–8.
Care Med. 2015;41(2):192–202. 64. Novelli G, Rossi M, Pretagostini M, Pugliese F,
52. Finfer S, Chittock DR, Su SY, Blair D, Foster D, Ruberto F, Novelli L, et al. One hundred sixteen cases
Dhingra V, et al. Intensive versus conventional glu- of acute liver failure treated with MARS. Transplant
cose control in critically ill patients. N Engl J Med. Proc. 2005;37(6):2557–9.
2009;360(13):1283–97. 65. Saliba F, Camus C, Durand F, Mathurin P, Letierce
53. Meier R, Bechir M, Ludwig S, Sommerfeld J, Keel A, Delafosse B, et al. Albumin dialysis with a non-
M, Steiger P, et al. Differential temporal profile of cell artificial liver support device in patients with
lowered blood glucose levels (3.5 to 6.5 mmol/l ver- acute liver failure: a randomized, controlled trial.
sus 5 to 8 mmol/l) in patients with severe traumatic Ann Intern Med. 2013;159(8):522–31.
brain injury. Crit Care. 2008;12(4):R98. 66. Demetriou AA, Brown RS Jr, Busuttil RW, Fair J,
54. McClave SA, Taylor BE, Martindale RG, Warren McGuire BM, Rosenthal P, et al. Prospective, ran-
MM, Johnson DR, Braunschweig C, et al. Guidelines domized, multicenter, controlled trial of a bioarti-
for the Provision and Assessment of Nutrition ficial liver in treating acute liver failure. Ann Surg.
Support Therapy in the Adult Critically Ill Patient: 2004;239(5):660–7.
Society of Critical Care Medicine (SCCM) and 67. Clemmesen JO, Kondrup J, Nielsen LB, Larsen FS, Ott
American Society for Parenteral and Enteral Nutrition P. Effects of high-volume plasmapheresis on ammo-
(A.S.P.E.N.). JPEN J Parenter Enteral Nutr. 2016; nia, urea, and amino acids in patients with acute liver
40(2):159–211. failure. Am J Gastroenterol. 2001;96(4):1217–23.
55. Plauth M, Cabre E, Campillo B, Kondrup J, 68. Larsen FS, Schmidt LE, Bernsmeier C, Rasmussen A,
Marchesini G, Schutz T, et al. ESPEN guidelines on Isoniemi H, Patel VC, et al. High-volume plasma
parenteral nutrition: hepatology. Clin Nutr. 2009; exchange in patients with acute liver failure: an
28(4):436–44. open randomised controlled trial. J Hepatol. 2016;
56. Antoniades CG, Berry PA, Wendon JA, Vergani D.  64(1):69–78.
The importance of immune dysfunction in determin- 69. Marshall LF, Smith RW, Rauscher LA, Shapiro
ing outcome in acute liver failure. J Hepatol. 2008; HM. Mannitol dose requirements in brain-injured
49(5):845–61. patients. J Neurosurg. 1978;48(2):169–72.
57. Rolando N, Harvey F, Brahm J, Philpott-Howard J, 70. Kamel H, Navi BB, Nakagawa K, Hemphill JC 3rd,
Alexander G, Casewell M, et al. Fungal infection: a Ko NU. Hypertonic saline versus mannitol for the
common, unrecognised complication of acute liver treatment of elevated intracranial pressure: a meta-­
failure. J Hepatol. 1991;12(1):1–9. analysis of randomized clinical trials. Crit Care
58. Karvellas CJ, Cavazos J, Battenhouse H, Durkalski Med. 2011;39(3):554–9.
V, Balko J, Sanders C, et al. Effects of antimicrobial 71. Li M, Chen T, Chen S-d, Cai J, Hu Y-h. Comparison
prophylaxis and blood stream infections in patients of equimolar doses of mannitol and hypertonic
with acute liver failure: a retrospective cohort study. saline for the treatment of elevated intracranial pres-
Clin Gastroenterol Hepatol. 2014;12(11):1942–9.e1. sure after traumatic brain injury: a systematic review
59. Bhatia V, Batra Y, Acharya SK. Prophylactic phe- and meta-analysis. Medicine. 2015;94(17):e736.
nytoin does not improve cerebral edema or survival 72. Robertson CS, Valadka AB, Hannay HJ, Contant CF,
in acute liver failure—a controlled clinical trial. J Gopinath SP, Cormio M, et al. Prevention of second-
Hepatol. 2004;41(1):89–96. ary ischemic insults after severe head injury. Crit
60. Sharma BC, Sharma P, Lunia MK, Srivastava S, Care Med. 1999;27(10):2086–95.
Goyal R, Sarin SK. A randomized, double-blind, 73. Lang EW, Czosnyka M, Mehdorn HM. Tissue
controlled trial comparing rifaximin plus lactulose oxygen reactivity and cerebral autoregulation
with lactulose alone in treatment of overt hepatic after severe traumatic brain injury. Crit Care Med.
encephalopathy. Am J Gastroenterol. 2013;108(9): 2003;31(1):267–71.
1458–63. 74. Shalimar Acharya SK. Management in acute liver
61. Rees CJ, Oppong K, Al Mardini H, Hudson M, failure. J Clin Exp Hepatol. 2015;5(Suppl 1):
Record CO. Effect of L-ornithine-L-aspartate on S104–15.
patients with and without TIPS undergoing gluta- 75. Balestreri M, Czosnyka M, Hutchinson P, Steiner LA,
mine challenge: a double blind, placebo controlled Hiler M, Smielewski P, et al. Impact of intracranial
trial. Gut. 2000;47(4):571–4. pressure and cerebral perfusion pressure on severe
25  The Patient with Severe Co-morbidities: CNS Disease and Increased Intracranial Pressure 329

disability and mortality after head injury. Neurocrit 88. Keays R, Potter D, O’Grady J, Peachey T,
Care. 2006;4(1):8–13. Alexander G, Williams R. Intracranial and cere-
76. Eefsen M, Dethloff T, Frederiksen H-J, Hauerberg J, bral perfusion pressure changes before, during
Hansen BA, Larsen FS. Comparison of terlipressin and immediately after orthotopic liver transplan-
and noradrenalin on cerebral perfusion, intracranial tation for fulminant hepatic failure. Q J Med.
pressure and cerebral extracellular concentrations 1991;79(289):425–33.
of lactate and pyruvate in patients with acute liver 89. Lidofsky SD, Bass NM, Prager MC, Washington
failure in need of inotropic support. J Hepatol. 2007; DE, Read AE, Wright TL, et al. Intracranial pressure
47(3):381–6. monitoring and liver transplantation for fulminant
77. Clemmesen JO, Hansen BA, Larsen FS. Indomethacin hepatic failure. Hepatology. 1992;16(1):1–7.
normalizes intracranial pressure in acute liver failure: 90. Prager MC, Washington DE, Lidofsky SD, Kelley SD,
a twenty-three-year-old woman treated with indo- White JD. Intracranial pressure monitoring during
methacin. Hepatology. 1997;26(6):1423–5. liver transplant without venovenous bypass for ful-
78. Tofteng F, Larsen FS. The effect of indomethacin minant hepatic failure. Transplant Proc. 1993;25(2):
on intracranial pressure, cerebral perfusion and 1841.
extracellular lactate and glutamate concentrations 91. Sakai T, Planinsic RM, Hilmi IA, Marsh JW. 
in patients with fulminant hepatic failure. J Cereb Complications associated with percutaneous place-
Blood Flow Metab. 2004;24(7):798–804. ment of venous return cannula for venovenous
79. Vaquero J. Therapeutic hypothermia in the man- bypass in adult orthotopic liver transplantation.
agement of acute liver failure. Neurochem Int. Liver Transpl. 2007;13(7):961–5.
2012;60(7):723–35. 92. Gurusamy KS, Koti R, Pamecha V, Davidson BR. Veno-
80. Jalan R, Olde Damink SW, Deutz NE, Hayes PC, venous bypass versus none for liver transplantation.
Lee A. Moderate hypothermia in patients with acute Cochrane Database Syst Rev. 2011;(3):Cd007712.
liver failure and uncontrolled intracranial hyperten- 93. Joshi D, O’Grady J, Patel A, Shawcross D, Connor
sion. Gastroenterology. 2004;127(5):1338–46. S, Deasy N, et al. Cerebral oedema is rare in acute-­
81. Karvellas CJ, Todd Stravitz R, Battenhouse H, Lee on-­chronic liver failure patients presenting with
WM, Schilsky ML, USALFSG. Therapeutic hypo- high-grade hepatic encephalopathy. Liver Int. 2014;
thermia in acute liver failure: a multicenter retro- 34(3):362–6.
spective cohort analysis. Liver Transpl. 2015;21(1): 94. Jepsen P, Ott P, Andersen PK, Sorensen HT, Vilstrup
4–12. H. Clinical course of alcoholic liver cirrhosis: a
82. Bernal W, Murphy N, Brown S, Whitehouse T, Danish population-based cohort study. Hepatology.
Bjerring PN, Hauerberg J, et al. A multicentre ran- 2010;51(5):1675–82.
domized controlled trial of moderate hypothermia to 95. Shawcross DL, Sharifi Y, Canavan JB, Yeoman AD,
prevent intracranial hypertension in acute liver fail- Abeles RD, Taylor NJ, et al. Infection and systemic
ure. J Hepatol. 2016;65:273–9. inflammation, not ammonia, are associated with
83. Andrews PJ, Sinclair HL, Rodriguez A, Harris BA, Grade 3/4 hepatic encephalopathy, but not mortality
Battison CG, Rhodes JK, et al. Hypothermia for in cirrhosis. J Hepatol. 2011;54(4):640–9.
intracranial hypertension after traumatic brain injury. 96. Dawwas MF, Lewsey JD, Neuberger JM, Gimson
N Engl J Med. 2015;373(25):2403–12. AE. The impact of serum sodium concentration on
84. Nielsen N, Wetterslev J, Cronberg T, Erlinge D, mortality after liver transplantation: a cohort multi-
Gasche Y, Hassager C, et al. Targeted temperature center study. Liver Transpl. 2007;13(8):1115–24.
management at 33 degrees C versus 36 degrees C 97. Gines P, Guevara M. Hyponatremia in cirrhosis:
after cardiac arrest. N Engl J Med. 2013;369(23): pathogenesis, clinical significance, and manage-
2197–206. ment. Hepatology. 2008;48(3):1002–10.
85. Rozga J, Podesta L, LePage E, Hoffman A, 98. Hudcova J, Ruthazer R, Bonney I, Schumann R. 
Morsiani E, Sher L, et al. Control of cerebral Sodium homeostasis during liver transplantation
oedema by total hepatectomy and extracorporeal and correlation with outcomes. Anesth Analg. 2014;
liver support in fulminant hepatic failure. Lancet. 119(6):1420–8.
1993;342(8876):898–9. 99. Yan L, Xie F, Lu J, Ni Q, Shi C, Tang C, et al. The
86. Sanabria Mateos R, Hogan NM, Dorcaratto D, treatment of vasopressin V2-receptor antagonists in
Heneghan H, Udupa V, Maguire D, et al. Total cirrhosis patients with ascites: a meta-analysis of
hepatectomy and liver transplantation as a two-stage randomized controlled trials. BMC Gastroenterol.
procedure for fulminant hepatic failure: a safe proce- 2015;15:65.
dure in exceptional circumstances. World J Hepatol. 100. Lizaola B, Bonder A, Tapper EB, Mendez-­Bocanegra
2016;8(4):226–30. A, Cardenas A. The changing role of sodium manage-
87. Bismuth H, Samuel D, Castaing D, Adam R, Saliba F, ment in cirrhosis. Curr Treat Options Gastroenterol.
Johann M, et al. Orthotopic liver transplantation 2016;14(2):274–84.
in fulminant and subfulminant hepatitis. The Paul 101. Živković SA. Neurologic complications after liver
Brousse experience. Ann Surg. 1995;222(2):109–19. transplantation. World J Hepatol. 2013;5(8):409–16.
330 P. Nandhabalan et al.

102. Saner FH, Nadalin S, Radtke A, Sotiropoulos GC, logical complications and one-month mortality after
Kaiser GM, Paul A. Liver transplantation and neurolog- liver transplantation. Front Neurol. 2014;5:275.
ical side effects. Metab Brain Dis. 2009;24(1):183–7. 107. Rovira A, Alonso J, Cordoba J. MR imaging find-
103. Chan G, Taqi A, Marotta P, Levstik M, McAlister ings in hepatic encephalopathy. Am J Neuroradiol.
V, Wall W, et al. Long-term outcomes of emergency 2008;29(9):1612–21.
liver transplantation for acute liver failure. Liver 108. Garcia-Martinez R, Rovira A, Alonso J, Jacas C,
Transpl. 2009;15(12):1696–702. Simon-Talero M, Chavarria L, et al. Hepatic enceph-
104. Vizzini G, Asaro M, Miraglia R, Gruttadauria S, alopathy is associated with posttransplant cognitive
Fili D, D’Antoni A, et al. Changing picture of cen- function and brain volume. Liver Transpl. 2011;
tral nervous system complications in liver transplant 17(1):38–46.
recipients. Liver Transpl. 2011;17(11):1279–85. 109. Kok B, Foxton MR, Clough C, Shawcross DL. 
105. Guarino M, Benito-Leon J, Decruyenaere J, Sch- Rifaximin is an efficacious treatment for the
mutzhard E, Weissenborn K, Stracciari A. Neuro- Parkinsonian phenotype of hepatic encephalopathy.
logical problems in liver transplantation. In: Gilhus Hepatology. 2013;58(4):1516–7.
NE, Barnes MR, Brainin M, editors. European hand- 110. Qavi AH, Hammad S, Rana AI, Salih M, Shah NH,
book of neurological management. 2nd ed. Oxford: Dar FS, et al. Reversal of acquired hepatocerebral
Wiley-Blackwell; 2011. p. 491–9. degeneration with living donor liver transplantation.
106. KA F, DiNorcia J, Sher L, Velani SA, Akhtar S, Liver Transpl. 2016;22(1):125–9.
Kalayjian LA, et al. Predictive factors of neuro-
Part III
Anesthesiology for Liver Surgery
Hepatobiliary Surgery: Indications,
Evaluation and Outcomes 26
Jay A. Graham and Milan Kinkhabwala

Keywords
Liver Anatomy
Hepatic resection · Surgical technique · Pringle
maneuver · Laparoscopic liver resection · Liver
It is impossible to begin a discussion of hepatobili-
abscesses · Liver tumor
ary surgery without referencing Claude Couinaud’s
(1922–2008) pioneering work [1]. By wax casting
the vasculature and biliary tree of cadaveric livers
Introduction in his laboratory in Neuilly-sur-Seine, Couinaud
ushered in a new understanding of biliary and
Complex surgical intervention for liver disease is segmental hepatic anatomy. Published in 1957,
a relatively new addition in medical practice and Le Foie: Études Anatomiques et Chirurgicales is
reliable standards of success were only achieved Couinaud’s seminal work detailing hepatobiliary
only in the past three decades. Earlier attempts at anatomy that has paved the way for development
surgical resection were commonly associated of surgical approaches for hepatic resection: an
with high rates of morbidity and mortality. anatomic approach permits precise parenchymal
Advancements in our understanding of liver resection with optimal preservation of the remnant,
physiology and anatomy, technological advance- which is the fundamental goal of the liver surgeon.
ments, dissemination of standardized surgical In standard human anatomy, the liver can be
techniques, and advances in perioperative anes- divided into eight segments, with four segments
thesia management have all contributed to sub- accounting for the right lobe (segments V, VI,
stantial improvements in the success of major VII, and VIII, approximately 55–60% of liver
hepatic surgery. volume), and three segments accounting for the
The aim of this chapter is to review the surgi- left lobe (segments II, III, IV, approximately
cal techniques and indications for major hepatic 30–40% of liver volume) (Fig. 26.1). Segment 1
surgery. is the caudate lobe, located posteriorly surround-
ing partially the inferior vena cava, which has
vascular derivation from both right and left pedi-
cles. Hepatic outflow is dependent on three main
hepatic veins in standard anatomy. Hepatic veins
drain into the suprahepatic vena cava just below
the diaphragm. The middle hepatic vein (MHV)
J. A. Graham, MD • M. Kinkhabwala, MD (*) defines the junction between the right and left
Department of Surgery, Albert Einstein College hepatic lobes. The MHV receives branches from
of Medicine, Bronx, NY, USA both right and left lobes in varying degrees and
e-mail: mkinkhab@montefiore.org

© Springer International Publishing AG, part of Springer Nature 2018 333


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_26
334 J. A. Graham and M. Kinkhabwala

Right posterior Right anterior Left medial Left lateral


section section section section

Right hepatic vein


Middle hepatic vein
Left hepatic vein

7
2
8

1
4

6 5
Hepatic
duct
Inferior
vena cava
Cystic Bile Portal Hepatic
Gall bladder duct duct vein artery

Fig. 26.1  Anatomy of the human liver. The human liver is a medial section (segment 4) separated by the left hepatic
divided by the falciform ligament into an anatomical right vein and the falciform ligament (not shown). Each segment
lobe and left lobe. However, the liver also has a functional can be individually resected. Black dashed lines show the
right and left side, divided by Cantlie’s line: a hypothetical demarcations between sections. Reprinted by permission
line from the gallbladder fossa to the middle hepatic vein. from Nature Publishing Group: Siriwardena AK, Mason JM,
Each functional hemi-liver is composed of two sections: on Mullamitha S, Hancock HC, Jegatheeswaran S. Management
the right, an anterior section (segments 5 and 8) and a poste- of colorectal cancer presenting with synchronous liver
rior section (segments 6 and 7) separated by the right hepatic metastases. Nat Rev Clin Oncol. 2014;11(8):446–459.
vein; and on the left, a lateral section (segments 2 and 3) and Copyright 2-14

patterns. Externally the middle hepatic vein is not result in acute congestion of that segment with
visible, so most hepatobiliary surgeons use intra- eventual segmental atrophy if intrahepatic collat-
operative sonography to define the location of the eral outflow tracts are not present. Consequently if
middle hepatic vein during parenchymal transec- functional extended right hepatectomy is planned,
tion. The location of the MHV can be estimated the surgeon should estimate the remnant volume
with Cantlie’s line, which is a virtual line between based only on segments II, III, and the left side of
the gallbladder fossa inferiorly and the suprahe- segment I. Estimation of remnant volume is essen-
patic vena cava superiorly (Fig. 26.1). tial to success in hepatic surgical resection.
Anatomic right hepatectomy is defined as resec- Trisegmentectomy is a misnomer, though the term
tion of the four right lobe segments to the right of is still widely utilized to describe an extended right
the MHV, with or without resection (though usu- hepatectomy. The actual number of segments being
ally with) of the right-sided portion of the caudate resected in “trisegmentectomy” is five (S4–8); only
lobe. Anatomic right hepatectomy preserves the the lateral segments (S2 and S3) and the left por-
MHV with the remnant left lobe, thus the outflow tion of the caudate remains after “trisegmentec-
of segment IV (S4) is preserved. Functional tomy” (Fig. 26.2).
extended right hepatectomy includes the MHV Non-anatomic resections are those that are
with the right lobe resection, depriving S4 of some not based on particular segmental vascular pedi-
outflow. Outflow obstruction in a segment will cles. “Wedge” resections of surface lesions, for
26  Hepatobiliary Surgery: Indications, Evaluation and Outcomes 335

a Formal right hepatectomy (right bisectionectomy) b Extended right resection (right trisectionectomy)

7 2 7 2
8 8
1 1
4 4
3 3

6 5 6 5

c Left lateral sectionectomy d Left hepatectomy (left bisectionectomy)

7 2 7 2
8 8
1 1
4 4
3 3

6 5 6 5

Fig. 26.2  Types of resections. Current terminology for (c) Left lateral sectionectomy involves removal of the
major liver resections illustrating sections and segments liver segments 2 and 3. (d) Liver segments 2–4 are
removed at each procedure. (a) In formal right hepatec- removed during left hepatectomy. Reprinted by permis-
tomy the right anterior and right posterior sections, com- sion from Nature Publishing Group: Siriwardena AK,
prising segments 5–8, are resected. (b) Liver sections Mason JM, Mullamitha S, Hancock HC, Jegatheeswaran
removed in extended right resection. The right anterior S. Management of colorectal cancer presenting with syn-
and right posterior sections of the liver (segments 5–8) are chronous liver metastases. Nat Rev Clin Oncol.
resected plus left medial section (segment 4); therefore, 2014;11(8):446–459. Copyright 2-14
this procedure is sometimes referred to as right trisectio-
nectomy. Note that the arterial and portal inflow to the
left-­lateral section is preserved in right trisectionectomy.

example, are non-anatomic resections. Surgeons Basic Techniques


usually prefer anatomic resections because they of Hepatic Resection
permit resection based on pedicle ligation that
reduces operative time and blood loss. In addition, While complex hepatic surgery continues to be
for cancer operations, anatomic resection removes mostly performed in larger centers, the number
the entire associated parenchyma based on a ped- of centers with substantial liver experience and
icle that may more fully include satellite disease resources has grown, so that now most large hos-
within the vascular distribution of the pedicle. pitals routinely perform hepatobiliary procedures
Nonanatomic resections are typically performed that would have been inconceivable in the past.
for very small surface lesions or when the configu- Large hepatobiliary centers are characterized by
ration of a lesion precludes an anatomic resection. a high volume of surgical procedures, availability
An example includes gallbladder fossa resection of a broad set of infrastructure resources, true
for gallbladder cancer that corresponds to portions multidisciplinary care and a commitment to dis-
of both S4 and S5. ease specific quality outcomes. Hepatology, criti-
336 J. A. Graham and M. Kinkhabwala

cal care medicine, interventional and diagnostic section line in right or left hepatic lobectomy. In
radiology are core disciplines that must be avail- this modification, an umbilical tape is passed
able, in addition to an experienced operative team around the hepatic vein, between the liver and
that is able to manage high acuity surgical retrohepatic cava, and then around the portal vein
procedures. so that the liver is suspended on the tape. The sur-
Surgical approaches to hepatic parenchymal geon can then use the tape as a target and guide
transection have evolved with time. In general, during the transection [5].
operative times, blood loss, and injury to the rem- During transection, surgeons may divide the
nant liver have all improved in the past two liver using as simple a method as fracture with a
decades [2, 3]. Transfusion requirements with fine clamp or use more technologically advanced
liver resection are significantly less than in previ- tools. The most commonly used device for tran-
ous years. Bleeding risk is minimized by several section other than clamp fracture is the
improvements: (1) hypovolemic anesthesia dur- Cavitron Ultrasonic Dissector (CUSA), origi-
ing the transection, in order to maintain low cen- nally developed for neurosurgery. The CUSA tip
tral venous pressures and reduce back-bleeding vibrates at a very high frequency, which divides
from the hepatic veins, (2) reduction of bleeding parenchyma but leaves intact (in principle) blood
risk from the inflow vessels through pedicle liga- vessels and bile ducts, allowing the surgeon to
tion or Pringle (inflow occlusion, Fig. 26.3) prior ligate those structures separately. Another device
to transection, (3) two surgeon technique, and (4) similar in practice to the CUSA utilizes a high
use of technology to facilitate transection. velocity water jet to divide parenchyma. Other
The goal of hepatic transection is division of devices are designed to precoagulate liver tissue,
the liver parenchyma with identification of blood allowing the surgeon to divide the tissue with a
vessels and bile ducts, so that they may be standard scissors or cautery without blood loss.
secured with ligatures or clips prior to transec- There are many precoagulating devices, all of
tion. There are many different variations and which rely on the transmission of energy (radio-
technologies used for parenchymal transection, frequency or microwave energy, for example) to
though the basic tenet is meticulous, fine tech- a handheld probe that coagulates liver paren-
nique and patience in identification and ligation chyma. There are very few controlled studies that
of each small structure. The surgeon must also compare various techniques of parenchymal divi-
pay close attention to the plane of transection, sion. Recently the LigaSure® device, an electro-
which must be identified and planned prior to thermal bipolar tissue sealing system has also
beginning parenchymal transection using ana- been used for the transection of the liver paren-
tomic landmarks and ultrasonography. A well chyma with good success [6]. Many surgeons
planned transection plane reduces transection minimize transection time and possibly morbid-
time (important when there is inflow occlusion), ity by using two experienced surgeons during
blood loss, risk of involved margins in cancer transection and utilizing a standard technique
surgery [4], and risk of injury to the remnant with only minor modification in every case.
liver. While this may seem self-evident, transec- A number of variations have arisen to the
tion planes can in fact be quite difficult to main- basic technique. For example, when lesions are
tain when the parenchyma is bulky, the field close to major vascular structures like the IVC or
bloody, or there are anatomic distortions (atrophy portal vein, selective application of total vascular
of a lobe, for example). Consequently, hepatobi- isolation (TVI, Fig. 26.3) can be useful to reduce
liary surgeons often “check” their planes con- bleeding risk.
stantly during the transection and make TVI is an extension of Pringle inflow occlu-
adjustments to stay on track. The development of sion directly derived from liver transplantation
the “hanging technique” has been especially use- total hepatectomy: in TVI the IVC above and
ful in maintaining a straight parenchymal tran- below the liver as well as porta hepatis is tempo-
26  Hepatobiliary Surgery: Indications, Evaluation and Outcomes 337

a b

c d

Fig. 26.3 Types of vascular occlusion: (a) Pringle flow blow (with permission from: Lang, H: Technik der
maneuver, (b) “Hemi-Pringle”, (c) Total vascular exclu- Leberresektion; Chirurg 2007;78:761–774)
sion (TVE) and (d) TVE with maintenance of caval blood

rarily occluded, so that the transection can occur advantage of TVI is the need for additional
in an asanguinous environment, reducing the mobilization of the liver and retrocaval IVC that
risk of hemorrhage near large vascular struc- would otherwise not be necessary. TVI also
tures. Most parenchymal transection can be requires more complex fluid management by the
accomplished in 30 min of occlusion time or anesthesiologist because the return of caval
less, which is generally well tolerated by the blood flow to the heart is interrupted, which can
liver. With longer inflow occlusion, more sub- be associated with hemodynamic instability
stantial ischemia-­reperfusion injury to the liver without volume loading and in some cases pres-
can be expected, which can increase morbidity sor support. Potential use of TVI should be dis-
especially if the remnant size is small. One dis- cussed between surgeon and anesthesiologist at
338 J. A. Graham and M. Kinkhabwala

the start of the operation, so that the anesthesi- Minimally Invasive Hepatic Surgery
ologist can ensure proper monitoring and prepa-
ration for the cross clamp. The surgeon must The first use of laparoscopic approaches in
communicate timing of TVI and perform a test hepatic surgery was reported back in 1992 by
clamp to ensure hemodynamic stability. Gagner et al. [9, 10]. These cases consisted
mostly of wedge biopsies of the liver for staging
of lymphoma and various case reports and small
Alternatives to Standard Resections series of laparoscopic resection of peripheral,
mostly benign lesions.
While complete resection of a solid lesion with More recently, reports of anatomic left lobec-
a negative margin (R-0 resection) is considered tomy and right lobectomy have energized the
the goal in cancer surgery for solid tumors, field [11]. Factors contributing to this boost is the
nonresectional cytoablative approaches to better knowledge of liver surgical anatomy, the
tumor control have been increasingly applied development of laparoscopic parenchymal tran-
when resection is not feasible because of section devices (electrosurgical devices/staplers)
underlying liver disease, comorbidities, or and that this type of surgery has become an
anatomic location and pattern of lesions. The exclusive field of experience hepatobiliary sur-
current targeted cytoablative options include geons in high volume centers [12].
localization of a lesion either by direct visu- Laparoscopic and minimally invasive liver
alization or intraoperative sonography, fol- surgery has the potential to change the diagnostic
lowed by insertion of a probe or antenna into and treatment algorithms for the management of
the lesion to accomplish transfer of energy to liver tumors, both benign and malignant. The
destroy the tissue. Radiofrequency ablation is benefits of minimally invasive hepatic surgery
the most common technique for cytoablation, compared with an open approach, can be sum-
though chemical ablation (alcohol, for exam- marized as:
ple), microwave ablation, and other methods
have been developed. Cytoablative techniques • Noninferiority in terms of cancer outcomes
are less invasive and associated with less [13]
morbidity than hepatic resections while still • Reduced morbidity in relation to the subcostal
achieving local tumor control, especially for incision
smaller lesions. For larger lesions and lesions • Better cosmetic outcome
in proximity to vascular structures, complete • Shorter length of stay and recovery to full
durable tumor control is less likely. In addi- function
tion, ablation can be performed sequentially,
in combination with other nonsurgical inter- The incidence of major surgical morbidity
ventional therapies such as transarterial embo- (bleeding, bile leak, liver failure) and mortality are
lization, and for control of multiple lesions. very similar to the open approach. Some authors
Ablation can even be used in combination with described the risk of CO2 embolism that may cause
resection to achieve R-0 control of bilobar dis- hemodynamic instability and morbidity. Animal
ease. The disadvantage of ablation (compared experiments demonstrated that CO2 embolism can
to resection) is that without complete extirpa- be detected by intraoperative transesophageal
tion and pathologic analysis of the lesion, it echocardiography but they do not necessarily cre-
is not certain that all of the tissue has been ate significant hemodynamic instability [14]. CO2
destroyed. With larger lesions, the possibility embolism is considered much safer than air embo-
of local recurrence is increased with ablative lism because of the greater solubility of CO2 com-
treatment compared to resection [7, 8]. pared to nitrogen.
26  Hepatobiliary Surgery: Indications, Evaluation and Outcomes 339

There are different forms of laparoscopic and Table 26.1  Indications for liver resection
laparoscopic assisted liver resections. For example Solid tumors
the right lobe can be mobilized laparoscopically Benign tumors
followed by a midline incision to resect the right Adenoma
lobe. The technique avoids the painful subcostal Hemangioma
incision. Smaller lesion can be removed either Focal nodular hyperplasia
laparoscopic-assisted using a hand port or fully Inflammatory pseudotumor
laparoscopically. Laparoscopic hepatectomy can Malignant tumors
be performed using inflow occlusion (Pringle) and Metastatic disease to the liver
intraoperative sonography analogous to open sur- Primary liver and biliary tract carcinoma
gery. Some locations in the liver are more amena- Hepatocellular carcinoma
ble to a laparoscopic approach, for example Cholangiocarcinoma
peripheral lesions are easier to access, including Gallbladder cancer
the lateral segments and the inferior segment of the Primary liver sarcomas
right lobe (segment 6). Lesions in Segment 7 (pos- Angiosarcoma
terior close to the diaphragm) are more difficult to Cystic lesions
access laparoscopically. Overall laparoscopic liver Cystadenoma/cystadenosarcoma
resection is a good option for the treatment of Simple epithelial cyst
patients with liver lesions, but requires a team that Polycystic liver disease
is experienced in both open and laparoscopic liver Pyogenic liver abscess
surgery for the best outcomes. Amebic abscess
Hydatid (echinococcal) cyst
Biliary tract disorders
Primary sclerosing cholangitis
Indications for Hepatic Surgery
Choledochal cystic disease/Caroli’s disease

A list of common indications for hepatic surgery


is outlined in Table 26.1. Common indications effectiveness and broad availability, and minimal
for hepatic surgery are influenced by geographic risk to the patient. US is therefore recommended
region. For example, hydatid cystic disease is for liver cancer screening in high risk populations,
more common in South/Central America and in like patients with chronic liver disease who are at
Eastern Europe. Gallbladder cancer is more com- risk for development of liver cancer [15]. When
mon in China, North India and South America, liver lesions are identified through screening, addi-
whereas hepatocellular carcinoma is more preva- tional liver specific contrast enhanced cross sec-
lent in Asia and Africa due to childhood exposure tional imaging is indicated to confirm diagnosis
to hepatitis B virus. Consequently, the approach and stage the lesion and to assess size and anatomy
to clinical diagnosis and evaluation of liver of the liver should hepatic resection be required. In
pathology is based partly on geography. some cases, contrast imaging may permit a diag-
Nevertheless, there are common strategies in nosis based on imaging criteria alone without need
evaluating a patient for potential hepatic surgery. for biopsy [15]. In biliary tract disorders, contrast
For example, a standard diagnostic work up will imaging together with cholangiography can be per-
always include a thorough history and physical formed simultaneously with MRI and MRCP, that
exam with a focus on liver disease risk factors, a is critical for preoperative evaluation of high biliary
laboratory evaluation that includes core liver func- tract lesions such as cholangiocarcinomas of hilar
tion tests and hepatitis serologies, and liver imaging. region (Klatskin tumor). Modern imaging software
Ultrasonography (US) is the most widely utilized allows detailed reconstructions of the hepatic vas-
liver imaging modality because of its relative cost culature and size of each segment, which results in
340 J. A. Graham and M. Kinkhabwala

an improved margin of safety when planning and Amebic Abscess


executing complex hepatic resections [16]. Like pyogenic abscess, amebic abscesses are typi-
cally treated with percutaneous drainage, antimi-
crobial therapy with metronidazole and luminal
Liver Abscesses agents such as idoquinanol or paromycin [21].
Amebic abscesses are more common in areas of
Pyogenic Abscess the world with a contaminated water supply.
The classic symptoms of liver abscess are fever Fecal-oral ingestion of entamoeba histolytica
and right upper quadrant pain, sometimes but cysts and trophozoite transformation in the host’s
not always together with jaundice. When jaun- colon, results in invasive disease that seeds the
dice is present, biliary infection (cholangitis) liver. Amebic abscess is the result of liquefactive
should be suspected. Because the liver receives necrosis due to trophozoic invasion that is histori-
the entire portal circulation, pyogenic abscesses cally described as anchovy sauce in appearance
are associated with intraabdominal infections [22]. Notably, the progressive hepatic necrosis is
including complicated appendicitis [17], diver- limited to Glisson’s capsule due to e. histolytica’s
ticulitis, or biliary stone disease. Liver abscess lack of hydrolytic enzymes so that amebic
is also known to occur in the setting of oral and abscesses can be seen abutting the liver capsule
dental infections, with underlying liver malig- without evidence of rupture. Patients usually pres-
nancy, or as a complication of liver directed ther- ent with fever, chills, anorexia, right upper quad-
apy like ablation [18]. Imaging usually reveals a rant tenderness and are typically between the ages
cystic lesion or lesions in the liver with evidence of 20 and 40 who have recently traveled to or are
of surrounding inflammatory change [19]. Liver from an endemic area.
abscesses can be solitary or multifocal, and may
have components of solid and fluid components. Hydatid Cyst
Segmental biliary dilation may be evident proxi- Hydatid disease (Echinococcosis) is endemic in
mal to the lesion, especially if the lesion is large. mostly rural areas of the world and is classically
Central or extrahepatic biliary dilation may be associated with sheep farming as sheep are the
present if the underlying etiology is related to intermediate host. While humans are not the defi-
biliary tract pathology like stone disease. Goal nite host, ingestion of eggs from sheep herding
of treatment is drainage and long-term intrave- dogs in a fecal-oral route leads to the develop-
nous antibiotics that provide coverage of biliary ment of hydatid cyst formation [23]. Present in an
tract organisms (gram negative and enterococ- equal male:female ratio, the average age of diag-
cus). Typically 6 weeks of antibiotic therapy is nosis is at 45 years. Patients usually present with
recommended [20] with serial imaging to verify vague abdominal pain, reflux and vomiting.
resolution of the process. Antibiotic therapy While hepatomegaly is a common sequelae of
can be modified after obtaining microbiology both amebic abscesses and hydatid cysts, jaun-
and sensitivities. Surgery is typically reserved dice and fever are rarely present with hydatid
for refractory sepsis or inability to effectively cysts. Echinococcal cysts can grow indolently
drain the abscess due to location or consistency until they are quite large and multiple. Free rup-
of the infected tissue. When surgery is required, ture of a hydatid cyst is potentially lethal due to
hepatotomy (unroofing of the abscess through anaphylaxis, and for this reason surgery is the
liver incision) can be performed, especially if mainstay of treatment, with antimicrobial treat-
the lesion is superficial. Formal resection may ment (albendazole typically) used as a preopera-
be required if the zone of infection is too large tive and postoperative adjunct to therapy [24].
or multilobulated to successfully drain with Patients with complex cystic disease who trav-
hepatotomy, if there is major biliary or vascu- elled from endemic areas associated with hydatid
lar involvement, or if there is suspicion for an disease should have cross sectional imaging in
underlying malignancy. addition to serologic testing for echinococcus.
26  Hepatobiliary Surgery: Indications, Evaluation and Outcomes 341

Radiographic findings of hydatid cysts include a which in some cases may even require liver
rosette arrangement and calcified walls on cross transplantation.
sectional imaging [25]. Ultrasonography is the gold Adenomas have potential malignant poten-
standard for diagnosis of echinococcal ­ disease. tial and can bleed. Larger adenomas have a
Budding and free-floating cysts can often be seen greater likelihood of hemorrhage or occult
and are classically termed hydatid sand. Larger malignancy, therefore resection is recom-
cysts do not respect tissue planes and can invade mended when the lesion is over 5 cm [28] or
surrounding structures including the diaphragm and when a lesion grows over a period of obser-
IVC. Patients presenting with pulmonary hyperten- vation despite discontinuation of oral contra-
sion and large hydatid cysts in continuity with the ception. Stable smaller lesions less than 3 cm
IVC may have systemic venous erosion of the cyst can be watched with careful surveillance, but
contents into the chest, which required careful oper- the patient will need to adhere to long-term
ative planning for possible IVC reconstruction at serial imaging. After baseline cross sectional
the time of hepatic resection [26]. In these patients, contrast imaging, ultrasonography can be used
a total vascular isolation (TVI) surgical technique for subsequent surveillance to assess changes
may be necessary to improve safety of the resection in size. There is no standard of care for how
(TVI is described later in this chapter). Prior to often or how long to perform surveillance in
instrumenting the liver, the abdominal operative small adenomas. Patients presenting with solid
space is packed with hypertonic soaked lap pads to liver lesions may understandably be alarmed,
prevent the possibility of disseminated echinococ- but the morbidity of surgical intervention is not
cosis. Patients are also treated with albendazole or justified in most cases. However, all patients
mebendazole pre- and post-operatively to help with solid liver lesions should be evaluated
reduce the echinococcal burden and mitigate opera- by a liver disease specialist or hepatobiliary
tive spillage complications. surgeon who can perform a correct diagnostic
Careful preoperative planning and communi- evaluation and propose intervention or surveil-
cation between surgery and anesthesia is impor- lance based on evidence. Cross sectional con-
tant to mitigate adverse outcomes, and the trast enhanced imaging with a liver directed
participation of an experienced infectious disease protocol is essential in the diagnostic work
parasitologist is helpful for diagnosis, pre and up. Hemangiomas and FNH can often be diag-
postoperative care and follow up. The anesthesi- nosed on imaging alone and biopsy is generally
ologist should have steroids and epinephrine not recommended.
available in case of anaphylaxis. Serum sodium Hemangiomas and FNH do not require rou-
needs to be followed closely with frequent ABG’s tine resection or intervention unless they are
during the operation because of the risk of hyper- symptomatic. Symptoms are typically vague
natremia associated with the use of hypertonic right upper quadrant or flank pain, or symptoms
saline in the peritoneal cavity. related to gastric extrinsic compression (satiety,
reflux). These symptoms are not usually dis-
abling but the constant nature of the symptoms
Benign Solid Liver Lesions may be disturbing to patients’ sense of well-­
being. Hepatobiliary surgeons must inform the
The most common benign solid tumors are hem- patient of the risks of resection in relation to their
angiomas, adenomas, and focal nodular hyper- symptoms and the extent of resection. Procedures
plasias (FNH). Adenomas and FNH lesions are with lower expected morbidity, like laparoscopic
associated with oral contraception and may be lateral segmentectomy, may be acceptable risk to
exacerbated by pregnancy. Men who use anabolic patients wishing to alleviate symptoms, whereas
steroids may also be prone to developing this it may be safer to defer any intervention for larger
hepatic lesion [27]. Glycogen storage disorders right sided benign lesions requiring major open
are associated with multifocal adenomatosis, hepatectomy and its associated morbidity.
342 J. A. Graham and M. Kinkhabwala

On contrast imaging, hemangiomas show typ- Emergency surgery may be necessary in rare cases
ical delayed filling. Adenomas show early where the condition of the patient and the rate of
enhancement, that is also a characteristic of hepa- hemorrhage preclude even embolization. In these
tocellular carcinomas; therefore adenomas can be cases the patient is triaged and managed like an
difficult to differentiate from well differentiated abdominal trauma patient.
HCC on imaging. Like HCC, hepatic adenomas
do not excrete gadoxetate disodium (Eovist®, a
gadolinium based contrast agent) on MRI and are Malignant Liver Lesions
“cold” on nuclear sulfur colloid scanning because
ultrastructurally they lack Kupffer cells and bile Secondary malignancies (metastases) to the liver
ductules. Clinical differentiation of adenoma ver- are more common than primary liver cancers in the
sus HCC is based on the statistical likelihood of a United States and colorectal cancer metastases are
typically enhancing lesion representing a dys- the most common type of metastatic lesion.
plastic nodule or frank cancer in patients with Worldwide, however, primary liver cancer (hepato-
underlying liver disease, compared to the very cellular carcinoma) is the second leading cause of
different demographic of patients presenting with cancer deaths and its incidence is increasing in the
adenoma, who are typically younger and without United States. Hepatic resection is the mainstay of
underlying liver disease. curative intervention in both primary and second-
Focal Nodular Hyperplasia is also enhancing, ary liver cancer, though the majority of patients are
but excretes gadoxetate disodium on MRI and is unresectable at presentation [33]. Selected patients
isointense on sulfur colloid scanning. FNH may with primary liver cancer may benefit from initial
also exhibit a pathognomonic central fibrous liver transplantation rather than resection, espe-
scar. Like adenomas, FNH is also influenced by cially if advanced liver disease is present. Metastatic
oral contraception and pregnancy. Unlike adeno- colorectal cancer to the liver is not an accepted
mas, FNH lesions have no increased malignancy indication for transplantation, though some patients
risk and the risk of bleeding is exceedingly rare. with neuroendocrine tumor metastases to the liver
Therefore FNH does not require intervention if could benefit from a transplant [34].
there is a typical appearance on contrast MRI. In Treatment of colorectal liver metastases, like
our practice, we follow FNH lesions with serial other solid tumors, is based on a multidisciplinary
imaging: after an initial confirmation of the diag- approach that may include preoperative or post-
nosis with cross sectional contrast imaging (state operative chemotherapy including the use of tar-
of the art is MRI), ultrasound is used every geted biologic agents such as bevacizumab.
6 months for two years and then surveillance is Systemic therapy has been shown to prolong sur-
discontinued unless the patient wants to be vival in Stage III colon cancer when used in the
become pregnant, in which case the lesion under- adjuvant setting. Systemic therapy is also com-
goes frequent surveillance imaging during monly used for metastatic disease, either in com-
­pregnancy [29, 30]. bination with liver directed therapy or alone. First
Hemorrhage is a known complication of adeno- line systemic therapy is based on a regimen of
mas; some patients with free peritoneal rupture 5FU and Oxaliplatin with leukovorin (Folfox).
can present in extremis [31]. Patients with sus- Second line therapy may replace Oxaliplatin with
pected rupture of an adenoma or HCC should be Irinotecan (Folfiri). These regimens have impli-
adequately resuscitated, moved to a critical care cations for hepatic resection because they have
monitored setting, and evaluated for emergency liver toxicity that can lead to substantial injury
embolization by interventional radiology. Though with prolonged treatment courses. The typical
not a definitive treatment, selective hepatic artery pattern of toxicity is microsteatosis, which can
embolization is usually successful in controlling impair liver regeneration post hepatectomy [35].
acute hemorrhage, which may permit a staged Cytotoxic chemotherapeutic drugs can also cause
resection under more elective circumstances [32]. hepatic sinusoidal fibrosis or veno-occlusive dis-
26  Hepatobiliary Surgery: Indications, Evaluation and Outcomes 343

ease [36]. Drug induced ultrastructural hepato- liver disease), most patients presenting with HCC
cellular injury may have unintended consequences will already have fibrotic change if not frank cir-
during liver resection, including an increased risk rhosis, which limits the application of resection
of posthepatectomy liver failure and portal hyper- as a curative modality in most patients.
tension. A residual volume/standard liver volume Many therapeutic interventions have been
(RV/SLV) ratio of 30% is considered the thresh- introduced for HCC over the past 20 years,
old for safe hepatic resection in these patients including ablation, transcatheter embolization,
However, in patients with liver dysfunction a and most recently stereotactic radiotherapy [38].
higher RV/SLV ratio is a requirement to mini- The Barcelona Liver Clinic Liver Cancer Group
mize potential postoperative hepatic insuffi- (BCLC) proposed an algorithm for HCC treat-
ciency. Preoperative portal vein embolization has ment in 2012 that has been widely adopted as a
been may decrease morbidity of major hepatec- standard of care [39]. The BCLC algorithm iden-
tomy in the setting of underlying liver disease by tifies three potentially curative pathways for early
increasing functional liver remnant volume. stage HCC: resection, liver transplantation, and
ablation. More advanced stages have limited
 ssociating Liver Partition and Portal
A options for cure and are best treated with life pro-
Vein Ligation for Staged Hepatectomy longing embolization, potentially with the addi-
(ALPPS) tion of systemic therapy. The only FDA approved
More recently (ALPPS) has been described as a systemic agent for HCC is the multikinase inhibi-
technique to increase functional liver residual tor Sorafenib [40]. Current areas of investigation
volume (FLRV). ALPPS, is a two stage opera- include the benefits of ablation compared to
tion: During the first stage, the hepatic paren- resection for small HCC, the utility of Stereotactic
chyma is divided leaving the hepatic artery and Body Radiation Therapy (SBRT) in HCC as a
bile duct intact, but the portal vein ipsilateral to curative modality, identification of biologic/
the tumor is ligated. During the second stage, genetic markers of HCC, and new technologies in
which can occur as early as 7–10 days after the embolization.
first operation, a completion hepatectomy is per- Cholangiocarcinoma (CCA) can arise sporad-
formed to remove the tumor bearing liver, by ically without prior illness but is also associated
which time the remnant liver has increased sub- with chronic liver disease, especially Primary
stantially in volume and a formal resection is Sclerosing Cholangitis (PSC). CCA can present
much better tolerated. Larger case series have as an intrahepatic mass lesion (in which case it
demonstrated that the ALPPS procedure is feasi- may appear similar to HCC), as a hilar biliary
ble with acceptable morbidity in patients who lesion causing biliary obstruction (Klatskin
would otherwise not be resectable [37]. tumor) or as a distal lesion at the head of the pan-
creas. The surgical approach and to some extent
 epatocellular Carcinoma (HCC)
H prognosis depends on the location of the lesion.
and Cholangiocarcinoma Mass lesions in the liver are treated with hepatic
Primary liver cancers most often arise in a field of resection. Hilar lesions most often require hepatic
underlying liver disease. The mechanisms for resection and bile duct resection with biliary—
carcinogenesis are different in hepatitis B com- enteric reconstruction (Roux en Y hepatojejunos-
pared to other chronic liver diseases. All chronic tomy, Fig. 26.4), and distal lesions may require a
liver disease is associated with an increase in can- pancreaticoduodenectomy (Whipple procedure)
cer risk, but patients with Hepatitis B infection in order to achieve negative margins. Resections
may present with HCC at an earlier stage of liver for hilar cancers are among the most difficult in
disease than Hepatitis C and other chronic liver all of surgery because of the complex decision
diseases (alcohol, fatty liver, etc.). Because the making when lesions are in proximity to vital
etiology of liver disease in the U.S. is commonly structures like the hepatic artery and portal vein.
HCV and alcohol (and now increasingly fatty In addition, extended hepatic resections may be
344 J. A. Graham and M. Kinkhabwala

Fig. 26.4  The Roux-en-Y


hepatojejunostomy With permission
from: Liu, Y., Yao, X., Li, S., Liu,
W., Liu, L., & Liu, J. (2014).
Comparison of Therapeutic Effects
of Laparoscopic and Open Operation
for Congenital Choledochal Cysts in
Adults. Gastroenterology Research
and Practice, 2014, 670260

required to remove all of the cancer bearing bili- Because the portal circulation is valveless,
ary and peribiliary tissue. changes in pressure are transmitted across the
entire portal circulation. Normally the large sinu-
soidal cross sectional area of the liver results in
 ost Hepatectomy Liver Failure
P very low resistance across the liver to portal flow.
and Liver Resection Morbidity With hepatic resection, a portion of this sinusoi-
dal area is removed, resulting in increased resis-
Current expected total morbidity (mortality + mor- tance to flow until the liver regenerates. Therefore
bidity) associated with hepatic resection overall is after any major hepatic resections portal pressure
approximately 15–20%, which reflects a rate of increases to some degree, though the changes
3–5% perioperative mortality and 10–15% mor- may be subclinical and of no consequence if the
bidity, according to the The American College of size and health of the remnant liver is sufficient to
Surgeons National Surgical Quality Improvement permit regeneration.
Program (ACS-NSQIP) database [2]. Transfusion Surgeons estimate the size of the remnant liver
rates in liver surgery have dropped dramatically after resection using volumetric cross sectional
over the past decade [41, 42]. Total morbidity is imaging either by computer tomography (CT) or
generally influenced by three factors: Magnetic Resonance Imaging (MRI). Volumetric
1.  Size/extent of hepatectomy measurements are labor intensive for the radiolo-
2. Presence and severity of underlying liver gist, though commercially available software
disease exists for calculating volumes and depicting
3.  Other comorbidities resected segments and remnants based on vascular
26  Hepatobiliary Surgery: Indications, Evaluation and Outcomes 345

pedicles. The surgeon should work closely with underlying liver disease is important in planning
the diagnostic radiologist to correctly describe the the maximum allowable resection [46]. Severity
potential resection plan for the radiologist in cal- of illness associated with cirrhosis has been eval-
culating volumes. Guidelines for remnant volumes uated using semiquantitative scoring systems
are based not only based on size but also on the (Child-Turcotte-Pugh—CTP score), and quanti-
presence of underlying parenchymal disease (hep- tative systems (Model For End-Stage Liver
atitis C or steatosis are common conditions). Disease—MELD score). Both CTP and MELD
Suggested size of the remnant liver to avoid small score are designed to predict long-term mortality
for size syndrome are 30% in a healthy liver and at related to liver disease, but not necessarily post-
least 40–50% in the presence of liver disease. operative risk after liver resection. ASA
When the size and health of the remnant (American Society of Anesthesiologists) [47] sta-
is insufficient, post hepatectomy liver failure tus and Charlson Index of Morbidity may have
(PHLF) may occur [43]. PHLF is caused by greater predictive value after hepatic resection
insufficient remnant hepatocellular function and than MELD but a combination of these scoring
is manifested by stigmata of portal hyperten- systems is probably most valuable in clinical
sion (ascites, hypoalbuminemia, intraabdominal practice [48]. Irrespective of the scoring systems
varices, and bowel wall edema) and diminished used, it is clear that with advancing cirrhosis, sur-
synthetic function (jaundice, coagulopathy, gical risks increase dramatically. MELD
encephalopathy). While the exact mechanisms score > 11 was associated with increased risk of
responsible for cell signaling in regeneration is postoperative mortality in a series of patients
poorly understood, portal hypertension itself undergoing resection for hepatocellular carci-
seems to contribute to cellular injury and may noma (HCC) [49]. Mortality in Childs B and C
inhibit regeneration. It has been demonstrated patients undergoing abdominal surgery were
that portal decompression or attenuation of portal 31% and 76%, respectively [50]. These rates
blood flow may facilitate liver regeneration and have improved more recently, but the risk of sub-
lessen the risk of PHLF, which has substantial stantial morbidity remains high. Decompensation
mortality risk in advanced stages. of chronic liver disease to acute-on chronic liver
In patents with underlying liver disease and/or failure is most common in patients with portal
small expected remnant size, preoperative portal hypertension that may be clinically not apparent.
vein embolization (PVE) can reduce morbidity Consequently, irrespective of CTP or MELD
by inducing hyperplasia of the remnant liver [44]. score, patients with preoperative clinical findings
After PVE the interruption of portal blood flow of portal hypertension are considered poor candi-
acts as a signal for the regenerative response. dates for resection. Clinical findings of portal
PVE is performed by interventional radiologists hypertension include thrombocytopenia, spleno-
who typically access the portal vein either tran- megaly, ascites, and/or varices. Subclinical portal
shepatically or through the internal jugular vein hypertension may be present in the absence of
(similar to TIPS). Once accessed, the PV can be these findings and only detectable using hepatic
embolized on the ipsilateral side of the resection, vein wedge pressure (HVWP) measurement.
thus inducing growth of the opposite lobe within HVWP measurement is indicated in patients with
3–4 weeks. One limitation of PVE is the delay in known cirrhosis being evaluated for major hepa-
potentially curative surgery that may occur while tectomy (three segments or greater). In patients
waiting for hyperplasia, though there is emerging with portal hypertension, preoperative portal
evidence that interval “bridge” arterial emboliza- decompression using TIPS has been suggested as
tion is useful in combination with PVE [45]. a means of improving postoperative morbidity
Patients with cirrhosis have increased risk of for non hepatic abdominal surgery, however this
liver decompensation and death after major has not been evaluated in controlled studies and
abdominal surgery and hepatic resection. TIPS itself has associated risks including exacer-
Assessment of the perioperative risk related to bation of hepatic encephalopathy. A decision to
346 J. A. Graham and M. Kinkhabwala

perform a transjugular intrahepatic portosystemic 15. Heimbach J, Kulik LM, Finn R, et al. AASLD guide-
shunt (TIPS) as a means of facilitating general lines for the treatment of hepatocellular carcinoma.
Hepatology. 2018;67:358–80.
abdominal surgery should only be made after 16. Soler L, Nicolau S, Pessaux P, Mutter D, Marescaux
multidisciplinary evaluation including an experi- J. Real-time 3D image reconstruction guidance in
enced hepatologist; TIPS itself does not facilite liver resection surgery. Hepatobiliary Surg Nutr.
hepatic resection and may in fact increase risk of 2014;3(2):73–81.
17. Bidwell LA. Hepatic abscess following acute appen-
acute postoperative liver failure because of depri- dicitis. Br Med J. 1910;2(2591):507–8.
vation of portal flow to the remnant liver. 18. Choi D, Lim HK, Kim MJ, et al. Liver abscess after
percutaneous radiofrequency ablation for hepatocel-
lular carcinomas: frequency and risk factors. Am J
Roentgenol. 2005;184(6):1860–7.
References 19. Hammer MM, Mellnick VM, Raptis CA, Schmidt R,
Fowler KJ. Peripheral restricted diffusion: a common
1. Couinaud CLF. Etudes anatomiques et chirurgicales. appearance of hepatic abscesses. J Comput Assist
Paris: Masson; 1957. Tomogr. 2015;39(6):890–5.
2. Aloia TA, Fahy BN, Fischer CP, et al. Predicting poor 20. Seeto RK, Rockey DC. Pyogenic liver abscess.
outcome following hepatectomy: analysis of 2313 Changes in etiology, management, and outcome. Med-
hepatectomies in the NSQIP database. HPB (Oxford). icine (Baltimore). 1996;75(2):99–113.
2009;11(6):510–5. 21. Pearce NW, Knight R, Irving H, et al. Non-operative
3. Mayo SC, Pawlik TM. Current management of management of pyogenic liver abscess. HPB (Oxford).
colorectal hepatic metastasis. Expert Rev Gastroenterol 2003;5(2):91–5.
Hepatol. 2009;3(2):131–44. 22. Agrawal BV, Somani PN, Khanna MN, Srivastava
4. Are C, Gonen M, Zazzali K, et al. The impact of mar- PK, Jha BN, Verma SP. Amebic pericardial effusion:
gins on outcome after hepatic resection for colorectal a rare complication of amebic liver abscess. Am Surg.
metastasis. Ann Surg. 2007;246(2):295–300. 1975;41(6):373–7.
5. Liddo G, Buc E, Nagarajan G, Hidaka M, Dokmak 23. Mihmanli M, Idiz UO, Kaya C, et al. Current status
S, Belghiti J. The liver hanging manoeuvre. HPB of diagnosis and treatment of hepatic echinococcosis.
(Oxford). 2009;11(4):296–305. World J Hepatol. 2016;8(28):1169–81.
6. Yoshimoto M, Endo K, Hanaki T, et al. Effectiveness 24. Sayek I, Onat D. Diagnosis and treatment of uncom-
of the LigaSure small jaw vessel-sealing system in plicated hydatid cyst of the liver. World J Surg.
hepatic resection. Yonago Acta Med. 2014;57(2):93–8. 2001;25(1):21–7.
7. Jarnagin W, Chapman WC, Curley S, et al. Surgical 25. Czermak BV, Akhan O, Hiemetzberger R, et al.
treatment of hepatocellular carcinoma: expert consen- Echinococcosis of the liver. Abdom Imaging. 2008;
sus statement. HPB (Oxford). 2010;12(5):302–10. 33(2):133–43.
8. Minami Y, Kudo M. Radiofrequency ablation of hepa- 26. Berney T, Mentha G, Morel P. Total vascular exclu-
tocellular carcinoma: current status. World J Radiol. sion of the liver for the resection of lesions in contact
2010;2(11):417–24. with the vena cava or the hepatic veins. Br J Surg.
9. Gagner M, Pomp A, Heniford BT, Pharand D, Lacroix A.  1998;85(4):485–8.
Laparoscopic adrenalectomy: lessons learned from 27. Rebouissou S, Bioulac-Sage P, Zucman-Rossi J. 
100 consecutive procedures. Ann Surg. 1997;226(3): Molecular pathogenesis of focal nodular hyperplasia
238–46. discussion 246–7 and hepatocellular adenoma. J Hepatol. 2008;48(1):
10. Gagner M, Rogula T, Selzer D. Laparoscopic liver 163–70.
resection: benefits and controversies. Surg Clin North 28. Deneve JL, Pawlik TM, Cunningham S, et al. Liver
Am. 2004;84(2):451–62. cell adenoma: a multicenter analysis of risk factors
11. Dagher I, O’Rourke N, Geller DA, et al. Laparoscopic for rupture and malignancy. Ann Surg Oncol. 2009;
major hepatectomy: an evolution in standard of care. 16(3):640–8.
Ann Surg. 2009;250(5):856–60. 29. Weimann A, Mossinger M, Fronhoff K, Nadalin S,

12. Nguyen KT, Gamblin TC, Geller DA. World review Raab R. Pregnancy in women with observed focal nodu-
of laparoscopic liver resection-2,804 patients. Ann lar hyperplasia of the liver. Lancet. 1998;351(9111):
Surg. 2009;250(5):831–41. 1251–2.
13. Kazaryan AM, Marangos IP, Rosok BI, et al. 30. Noels JE, van Aalten SM, van der Windt DJ, et al.

Laparoscopic resection of colorectal liver metastases: Management of hepatocellular adenoma during preg-
surgical and long-term oncologic outcome. Ann Surg. nancy. J Hepatol. 2011;54(3):553–8.
2010;252(6):1005–12. 31. Huurman VA, Schaapherder AF. Management of ruptured
14. Eiriksson K, Fors D, Rubertsson S, Arvidsson D.  hepatocellular adenoma. Dig Surg. 2010;27(1):56–60.
High intra-abdominal pressure during experimental 32. Stoot JH, van der Linden E, Terpstra OT, Schaapherder
laparoscopic liver resection reduces bleeding but AF. Life-saving therapy for haemorrhaging liver ade-
increases the risk of gas embolism. Br J Surg. 2011; nomas using selective arterial embolization. Br J Surg.
98(6):845–52. 2007;94(10):1249–53.
26  Hepatobiliary Surgery: Indications, Evaluation and Outcomes 347

33. Kelly ME, Spolverato G, Le GN, et al. Synchronous 43. Rahbari NN, Garden OJ, Padbury R, et al. Posthepa-
colorectal liver metastasis: a network meta-analysis tectomy liver failure: a definition and grading by the
review comparing classical, combined, and liver- International Study Group of Liver Surgery (ISGLS).
first surgical strategies. J Surg Oncol. 2015;111(3): Surgery. 2011;149(5):713–24.
341–51. 44. Okabe H, Beppu T, Ishiko T, et al. Preoperative por-
34. Gedaly R, Daily MF, Davenport D, et al. Liver trans- tal vein embolization (PVE) for patients with hepa-
plantation for the treatment of liver metastases from tocellular carcinoma can improve resectability and
neuroendocrine tumors: an analysis of the UNOS may improve disease-free survival. J Surg Oncol.
database. Arch Surg. 2011;146(8):953–8. 2011;104(6):641–6.
35. Selzner M, Clavien PA. Fatty liver in liver transplanta- 45. Sotiropoulos GC, Druhe N, Sgourakis G, et al. Liver
tion and surgery. Semin Liver Dis. 2001;21(1):105–13. transplantation, liver resection, and transarterial che-
36. Khanna R, Sarin SK. Non-cirrhotic portal hyper-
moembolization for hepatocellular carcinoma in cir-
tension—diagnosis and management. J Hepatol. rhosis: which is the best oncological approach? Dig
2014;60(2):421–41. Dis Sci. 2009;54(10):2264–73.
37. Alvarez FA, Ardiles V, de Santibanes M, Pekolj J, 46. Wagener G. Assessment of hepatic function, opera-
de Santibanes E. Associating liver partition and por- tive candidacy, and medical management after liver
tal vein ligation for staged hepatectomy offers high resection in the patient with underlying liver disease.
oncological feasibility with adequate patient safety: Semin Liver Dis. 2013;33(3):204–12.
a prospective study at a single center. Ann Surg. 47. Schroeder RA, Marroquin CE, Bute BP, Khuri S,

2015;261(4):723–32. Henderson WG, Kuo PC. Predictive indices of mor-
38. Wulf J, Guckenberger M, Haedinger U, et al. Stereotactic bidity and mortality after liver resection. Ann Surg.
radiotherapy of primary liver cancer and hepatic metas- 2006;243(3):373–9.
tases. Acta Oncol. 2006;45(7):838–47. 48. Simons JP, Ng SC, Hill JS, Shah SA, Zhou Z, Tseng
39. Forner A, Gilabert M, Bruix J, Raoul JL. Treatment of JF. In-hospital mortality from liver resection for hepa-
intermediate-stage hepatocellular carcinoma. Nat Rev tocellular carcinoma: a simple risk score. Cancer.
Clin Oncol. 2014;11(9):525–35. 2010;116(7):1733–8.
40. Llovet JM, Ricci S, Mazzaferro V, et al. Sorafenib in 49. Cucchetti A, Ercolani G, Vivarelli M, et al. Impact
advanced hepatocellular carcinoma. N Engl J Med. of model for end-stage liver disease (MELD) score
2008;359(4):378–90. on prognosis after hepatectomy for hepatocellular
41. Day RW, Brudvik KW, Vauthey JN, et al. Advances carcinoma on cirrhosis. Liver Transpl. 2006;12(6):
in hepatectomy technique: Toward zero transfu- 966–71.
sions in the modern era of liver surgery. Surgery. 50. Hsu KY, Chau GY, Lui WY, Tsay SH, King KL, Wu
2016;159(3):793–801. CW. Predicting morbidity and mortality after hepatic
42. Tzeng CW, Cooper AB, Vauthey JN, Curley SA,
resection in patients with hepatocellular carcinoma:
Aloia TA. Predictors of morbidity and mortality after the role of Model for End-Stage Liver Disease score.
hepatectomy in elderly patients: analysis of 7621 World J Surg. 2009;33(11):2412–9.
NSQIP patients. HPB (Oxford). 2014;16(5):459–68.
Liver Resection Surgery:
Anesthetic Management, 27
Monitoring, Fluids and Electrolytes

Emmanuel Weiss, Jean Mantz,
and Catherine Paugam-Burtz

Keywords
of segment IV. Left lobectomy is a left hepatec-
Risk assessment · Ischemia-reperfusion injury ·
tomy restricted to segments II and III. Liver
Cardiac stresstest · Hemodynamic monitoring ·
resection is considered major when three or more
Intravenous fluids · Transfusion
segments are involved.
Better patient selection, improvement of sur-
gical techniques (e.g. parenchymal sparing resec-
Introduction tions), optimization of anesthetic management
and creation of specialized high-­volume centers
Liver resection surgery has become a cornerstone have decreased hepatic resection-related mortal-
of the therapeutic strategies for primary hepato- ity over the past decade [1–3]. Several studies
cellular carcinoma (HCC) and liver metastases indicate that survival following liver resection is
often in combination with systemic chemother- significantly affected by the volume of liver
apy, embolization or radiotherapy. Other com- resected, preoperative liver function, response to
mon indications are polycystosis, hydatidosis, portal vein embolization, and condition of the
benign tumors, pheochromocytoma, and trauma. remnant liver parenchyma [4, 5]. In particular,
Two major resection subtypes can be distin- metabolic syndrome with non-alcoholic steato-
guished on anatomical bases: right hepatectomy, hepatitis or preoperative chemotherapy with anti-
which includes resection of segments V–VIII, angiogenic factors may worsen postoperative
and left hepatectomy, which consists of resection outcome [6–8]. Postoperative 30-day mortality
of segments II–IV and sometimes I. Right lobec- and liver failure after liver resection is around
tomy consists of right hepatectomy plus resection 2–3% in patients with non-­cirrhotic parenchyma,
while it may reach 8–10% in patients with
E. Weiss, MD, PhD • C. Paugam-Burtz, MD, PhD (*) chronic liver disease such as cirrhosis [1, 2, 4, 5,
Department of Anesthesiology and Critical Care 9–16]. Postoperative morbidity ranges from 15 to
Medecine, Beaujon, HUPNVS, Assistance 50% of patients. Besides the status of the rem-
Publique-Hôpitaux de Paris (AP-HP),
nant liver, age, and comorbidities such as diabe-
Paris F-75018, France
tes and compromised cardiovascular or
University Paris VII, Paris Diderot,
respiratory functions also impact outcome [8,
Paris F-75018, France
e-mail: catherine.paugam@aphp.fr 17]. Increasingly elderly patients with substantial
comorbidities present for liver resection surgery
J. Mantz, MD, PhD
Department of Anesthesiology and Critical Care and require optimization of the preoperative sta-
Medicine, HEGP, APHP, Paris, France tus and complex intraoperative management.

© Springer International Publishing AG, part of Springer Nature 2018 349


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_27
350 E. Weiss et al.

This chapter will review the goals of preopera- Cardiovacular Risk Assessment
tive risk evaluation, the anesthetic agents and
techniques relevant to liver resection, the hemo- Careful evaluation of the risk for cardiovascu-
dynamic consequences of vascular cross clamp- lar adverse events is important in patients with
ing during liver resection, the intraoperative compromised coronary or myocardial function
monitoring with emphasis on recent controver- undergoing liver resection surgery. Especially,
sies on hemodynamic topics, and a discussion of the presence and severity of metabolic syndrome
vascular filling, electrolytes, transfusion, and should be carefully assessed. The overall inci-
blood saving agents and techniques. We will also dence of major cardiovascular adverse events
briefly discuss the benefits of enhanced recovery is approximately 3%. The guidelines for preop-
strategies. erative cardiac risk assessment and periopera-
tive cardiac management in noncardiac surgery
should be applied [18, 19]. Evidence suggests
Preoperative Risk Evaluation that systematic coronary angiography and revas-
cularization (by stenting or bypass) prior to
 valuation of the Liver Function,
E surgery are not beneficial to patients undergo-
Specific Morbidity and Specific Risk ing noncardiac surgery [20]. Exercise tolerance
of patients may be better suited to assess car-
Rigourous preoperative evaluation of liver diac risk; limited function (measured in meta-
function is mandatory to select appropriate bolic equivalents) should be a cause for further
liver resection candidates and avoid postopera- examination. In patients with (or suspected of)
tive liver failure. This assessment allows tai- compromised coronary function, noninvasive
loring of patient’s preoperative management. assessment of coronary reserve can be done by
The specific evaluation of liver function before either stress echocardiography or stress angio-
liver resection and its impact on management scintigraphy. Thalium-persantine angioscintig-
and outcome is developed elsewhere in this raphy may be less useful in this population if
book (Fig. 27.1). there is pre-existing cirrhosis due to pre-existing
Additionally, other non-liver factors, such as vasodilation. Surgery for cancer often needs to
ASA physical status or renal function affect out- proceed without preoperative coronary revascu-
come and should be assessed preoperatively. larization for example by coronary stenting as
cancer may rapidly grow and become unresect-
able if surgery is delayed even with bare-metal
stenting (4–6 weeks). Cardiovascular medica-
tions such as beta-­blockers and statins should be
VIII IV continued throughout the perioperative period
III
[21], but initiation of beta-­blockade in patients
not on chronic beta-blocker therapy before sur-
VII gery is not recommended. In patients with chronic
beta-blockade therapy, particular caution should
Left medial branch be paid to the intraoperative hemodynamic mon-
V
VI Left portal vein itoring and optimization, since the reduction
of postoperative major cardiovascular adverse
Right portal Portal vein
vein
events and mortality attributed to beta-blockers
can be counterbalanced by an increase incidence
Fig. 27.1  Anatomical segmentation of the liver of stroke and noncardiac mortality [22, 23].
27  Liver Resection Surgery: Anesthetic Management, Monitoring, Fluids and Electrolytes 351

In patients with chronic antiplatelet therapy for Nutritional Assessment


secondary prevention of thrombotic events,
a growing body of evidence suggests that the Close attention should be paid to nutritional
risk of discontinuation of antiplatelet therapy status during the pre-anesthetic evaluation.
before surgery exceeds the risk of maintain- Malnutrition is an imbalance between the intake
ing this treatment throughout the perioperative of nutrients and metabolic needs. It can be
period [24, 25]. However at least two studies assessed by loss of weight, hypo-albuminemia
indicated that there is no superiority in main- or low pre-albumine. Malnutrition is common in
taining versus stopping aspirin preoperatively in cirrhotic patients and, a fortiori in the context
noncardiac surgical patients at vascular risk [26, of neoplasia and surgery [30]. Malnutrition has
27]. Therefore, specifically with the increased been shown to increase postoperative morbidity
risk for bleeding during liver surgery preopera- (especially related to infectious and pulmonary
tive cessation of antiplatelet therapy should be complications) and hypoalbuminemia [1, 3] and
considered. low body mass index (BMI < 18.5 kg/m2) have
been described as prognosticators [31]. More
recently, sarcopenia, defined as a progressive
Pulmonary Function Assessment loss of muscular mass occurring even in obese
patients and measured by evaluating muscular
Postoperative pulmonary complications are fre- atrophy by CT scan has been shown to accu-
quent after liver resection. Nobili et al. found in a rately predict short- and long-term outcome
large series of 555 patients who underwent elec- after hepatectomy [32–34]. Patients with nutri-
tive liver resection that almost 45% of patients tional risk patients should benefit from preoper-
experienced pulmonary complications including ative nutritional care using dietary supplements
pleural effusions, pneumoniae and pulmonary that may be hypercaloric and hyperproteinated
embolisms (40.5%, 13% and 2.9% respectively) in case of sarcopenia. Preoperative immunonu-
[28]. Interestingly, aside from diabetes melitus, trition in cancer surgery reduces hospital length
most risk factors were modifiable: prolonged sur- of stay and should be considered before liver
gery, presence of a nasogastric tube, intraopera- tumor resection [35, 36]. A multicenter ran-
tive blood transfusion and a transverse subcostal domized controlled phase IV trial is currently
bilateral muscle cutting incision [28]. evaluating the efficacy of preoperative immu-
nonutrition in reducing postoperative morbidity
after liver resection for cancer [37].
Renal Function Assessment

Postoperative acute kidney injury (AKI) is another Intraoperative Management


frequent complication after liver surgery with an
incidence of approximately 15%. AKI is usually Anesthetic Agents
multifactorial and highly related to mortality [29].
A prediction score including preoperatively ele- The main goals of anesthesia for liver resection
vated alanine aminotransferase (ALT), preexisting surgery are, first, to maintain intraoperative
cardiovascular disease, chronic renal failure, and hemodynamic stability, particularly in case of
diabetes has been described and is potentially use- massive blood loss and in response to vascular
ful to identify patients that may benefit from more clamping and unclamping, and, second to mini-
invasive intraoperative hemodynamic manage- mize blood loss and follow an appropriate
ment [29]. ­transfusion strategy. The risk of aspiration of the
352 E. Weiss et al.

gastric content at induction of anesthesia is high some studies but remains controversial [42, 43].
in cirrhotic patients with voluminous ascites, and Finally, during the last 10 years, some studies
rapid sequence induction should be used as indi- suggested a beneficial effect of preconditioning
cated. Pharmacokinetics of drugs is highly vari- strategies using volatile anesthetics on liver
able in severe cirrhotic patients because of major ischemia-­reperfusion injury. It will be further dis-
changes in distribution volumes and sodium cussed in vascular occlusion section of this
retention, albumin plasma levels, metabolism, chapter.
and elimination processes. The pharmacology of
anesthetic drugs in patients with liver failure is
discussed elsewhere in this book. Hemodynamics

Vascular occlusions, hemorrhage during dissec-


Hypnotics tion and especially transection, liver mobilization
(“liver luxation”), inferior vena cava compression
Anesthetics for which elimination primarily compromising venous return or gas embolism are
depends on renal clearance or redistribution (such all potential causes for hemodynamic instability
as propofol, etomidate, fentanyl, sufentanil) are during liver surgery. Beat-to-beat blood pressure
the first-choice drugs, while those depending on measurement using arterial catheters is manda-
hepatic metabolism, for example, using the P450 tory in almost all cases of liver surgery.
cytochrome system such as thiopental and alfent-
anil should be avoided. Remifentanil and cisatra-
curium may be used, as they do not accumulate Hemodynamic Consequences
even when administered by continuous infu- of Vascular Occlusions
sion. Target-controlled propofol infusion may
be an interesting and worthwhile concept since Occlusion of the portal triad (Pringle’s maneuver)
it may help to blunt intraoperative hemodynamic and total vascular exclusion (simultaneous clamp-
changes [38, 39]. Recently, Wang et al. suggested ing of the infrahepatic and suprahepatic vena cava)
in a randomized double-blind trial including 44 are commonly used to minimize intraoperative
patients undergoing elective hepatectomy with blood loss during transection of the liver paren-
inflow occlusion that perioperative dexmedeto- chyma [44]. The Pringle’s maneuver (vascular
midine administration might reduce intestinal occlusion of the portal triad) is associated with a
and hepatic injury [40]. decrease in blood loss by approximately 800 mL
The risk of hepatotoxicity of volatile anesthet- but also worsens postoperative liver injury and has
ics depends on their degree of hepatic metabo- no demonstrable effect on red cell transfusion,
lism. Halothane use has been shown to lead to mortality or postoperative liver failure [45]. It is
fulminant hepatitis through the production of the associated with a 15% decrease in venous return
hepatotoxic metabolite trifluoroacetic acid but and cardiac output, that is usually well tolerated by
this risk could now be considered as historic. a compensatory increase of sympathetic tone [46–
More recent poorly metabolized volatile anes- 48]. Hepatic inflow occlusion unavoidably causes
thetics such as isoflurane or desflurane can be ischemic injury that may jeopardize liver regener-
safely used for maintenance of anesthesia during ation after hepatic resection surgery.
liver surgery. Isoflurane may improve hepatic Total vascular exclusion of the liver is associ-
blood flow and hepatic oxygen supply [41]. A ated with a substantial decrease of venous return.
beneficial effect of volatile anesthesics (over pro- Cardiac output and mean arterial pressure are
pofol) on postoperative inflammatory response decreased by 40% and 10%, respectively due
and hepatocellular injury was also suggested by to a marked increase in sympathetic tone [49].
27  Liver Resection Surgery: Anesthetic Management, Monitoring, Fluids and Electrolytes 353

Tolerance to this situation depends on the intravas- erties during a brief period before a severe ischemic
cular volume status, the presence of portosystemic insult to minimize ischemic damages. According
shunts and possible impairment of ventricular to several randomized controlled trials [54, 55] and
function. These parameters have to be evaluated meta-­analysis [56], ischemic preconditioning (10-
and optimized before considering to proceed with min long portal triad clamping before the start of
total vascular exclusion. However, reliable predic- transection) is as effective as intermittent clamping.
tors of intolerance to this maneuver remain to be Intermittent portal triad clamping appears superior,
established. If a prolonged total vascular exclusion however, for duration of occlusion >75 min of the
of the liver is necessary and anticipated, the tran- triad [57]. Nonsurgical tools for liver protection
sient use of a veno-venous bypass can be sched- have been reviewed and include pharmacologic
uled in experienced centers. Other intraoperative interventions targeting microcirculation, oxida-
procedures such as the “hanging liver maneuver” tive stress, proteases, and inflammation [44, 53].
in which the liver is suspended by lifting it up Anesthetic volatile agents (especially sevoflurane)
with a tape that is passed behind the liver facilitate may be one of these preconditioning stimuli dur-
resection using the anterior approach for major ing hepatic resection. Beck-Schimmer et al. con-
hepatectomy. Reduction of liver mobility during ducted a randomized controlled trial comparing a
dissection achieved by this maneuver significantly standard care consisting in total intravenous anes-
improves intraoperative hemodynamic stability thesia (TIVA) with a preconditioning strategy con-
[50, 51]. Lateral clamping of the inferior vena cava sisting in the replacement of TIVA by sevoflurane
is also at times used to reduce bleeding during par- 30 min before inflow occlusion [58]. Although the
ticular delicate phases of liver dissection. number of patients was limited, the precondition-
ing strategy reduced hepatic injury (assessed by
postoperative rise in transaminases) and surgical
 anagement of Ischemia-­
M complications [58]. More recently, the same team
Reperfusion Injury described the same beneficial effect for postcondi-
tioning consisting of immediate and short-term use
Pringle’s maneuvers may cause ischemia-­ reper­ of volatile anesthesics after reperfusion that may
fusion syndrome characterized by loss of vascular be more appropriate to emergency situations [59].
tone and hypotension at unclamping. At the molec- Several mechanisms have been proposed for hepa-
ular level, ischemia-reperfusion leads to a release toprotective effect of volatile agents: stimulation
by damaged endothelial cells of Damage-Associ- of antioxydative system through heme-oxygenase
ated-Molecular Patterns (DAMPs) that stimulate 1 upregulation [60], increased production of anti-­
immune cells (such has Kuppfer cells) and trigger inflammatory IL-1Ra and antiapoptotic Bcl2 [61]
hepatocyte exaggerated inflammatory response, or attenuation neutrophil inflammatory response
hepatocyte necrosis and finally liver dysfunction elicited by CXC cytokines through an interfer-
[52]. ence with their cognate receptors [62]. Of note, a
Intermittent portal triad clamping (cycles of protective effect of remifentanyl pretreatment on
15-min inflow occlusion followed by 5-min liver injury was also suggested in a murine model
reperfusion) is thought by some to protect against of ischemia-reperfusion [63]. Inducible nitric oxide
liver injury due to prolonged ischemia compared synthase partly mediated this effect by exhausting
to continuous, uninterrupted Pringle’s maneuver reactive oxygen species and attenuating inflam-
[53, 54]. matory response. Ischemic preconditioning has
Other hepatoprotective methods against ische­ been recently suggested to exhibit multiple ben-
mia-­reperfusion injury rely on preconditioning eficial clinical endpoints, but further RCTs seem
either by exposure to ischemia or pharmacological to be needed to confirm its clinical benefits [64].
agents with antioxidant or anti-inflammatory prop- Finally, omega-3 polyunsaturated fatty acids may
354 E. Weiss et al.

Fig. 27.2 Preload– Fluid Challenge


dependence of cardiac
output as illustrated by Principle of fluid challenge
the hemodynamic Q
response to a fluid
challenge (from Curr Volume unresponsive
Opin Crit Care 2005; 11:
260–70, with
permission)

Volume responsive CVP ↑ 2 mmHg

CO ↑ 300 ml/min
(↑ ScvO2)
Pra Curr op crt care 2005;
11:260-70

exert beneficial effect on hepatic steatosis, regen- Invasive Arterial Pressure


eration and inflammatory insults such as ischemic
injury after surgery [65, 66]. Results from a ran- A large amount of information can be obtained
domized controlled trial hypothesizing that their from monitoring invasive arterial blood pressure.
use as preconditioning strategy may reduce post- Blood pressure does not reflect blood flow and
operative complications are pending [67]. Aside hypovolemia and hypoperfusion may be pres-
from these pathophysiological interesting effects, ent even with normal blood pressure. However,
the real clinical benefit of these strategies remains there are clearly thresholds below which some
to be confirmed. organs, such as the brain, the liver, or the kidney
are not adequately perfused and intraoperative
hypotension is as a predictor of 6-month mor-
Hemodynamic Monitoring Tools tality in cirrhotic patients undergoing surgery
[73]. Therefore, any severe intraoperative hypo-
Perioperative hemodynamic optimization through tensive episodes during liver resection should be
cardiac output (or stroke volume) monitoring has promptly corrected by fluid loading and/or vaso-
been shown to improve outcome in digestive sur- pressors. Monitoring of respiratory variations
gery [68–71] (Fig. 27.2). However, this well docu- of the arterial pulse pressure (pulse pressure
mented and demonstrated concept is considered variation—PPV) has become increasingly popu-
controversial in liver surgery where low CVP, at lar also in major hepatic surgery. This index has
least before and during the transection, is consid- been initially reported as a reliable predictor of
ered as a standard technique to decreased intra- responsiveness to a fluid challenge in mechani-
operative bleeding [72]. Therefore, any device cally ventilated patients with ARDS [74]. It can
that allows direct or indirect evaluation of left be obtained by applying the following formula:
ventricular stroke volume and responsiveness to
fluid loading may be useful for routine use in liver PPV ( % ) = 100 ( PPmax − PPmin ) / [ PPmax + PPmin ]
resection surgery. /2.
27  Liver Resection Surgery: Anesthetic Management, Monitoring, Fluids and Electrolytes 355

Over the last few years, some random- Esophageal Doppler


ized controlled trials have demonstrated that
intraoperative fluid management based on mon- The esophageal Doppler monitor (ODM) aims to
itoring and optimization of dynamic param- estimate stroke volume and cardiac output. A
eters such as PPV or stroke volume variation small number of studies performed outside the
results in a significant reduction of morbidity frame of liver resection on a restricted number of
and hospital length of stay after major surgery patients have shown that intraoperative ODM
[75–77]. Although Solus-Biguenet et al. sug- may improve patient recovery [85–87]; however,
gested that PPV could predict fluid respon- none of them were designed to examine mortality
siveness in a small number of 48 patients (54 or other “hard” outcomes as a primary endpoint.
fluid challenges) undergoing major hepatic More recently, the OPTIMIZE study included
surgery, some limitations of PPV should be 734 high-risk patients undergoing major gastro-
kept in mind. First, recently published data intestinal surgery failed to demonstrate a benefit
showed that decreasing tidal volume to 8 mL/ of a cardiac output-guided hemodynamic strat-
kg enhances patients’ clinical outcomes dur- egy (as compared to standard care) on complica-
ing major abdominal surgery and thus should tions and 30-day mortality [70]. However,
be implemented in the management protocols including their data in an updated meta-analysis
of patients undergoing major surgery [78]. of more than 6500 patients, the authors found
Lower tidal volumes however are limiting the that the intervention reduced complication rate
use of PPV as it increases the number of false- [70]. Although most of the studies included in
negatives [79]. Secondly, PPV value is highly this meta-analysis used ODM or pulse contour
depends on intra-abdominal pressure. An analysis-based methods, current level of evidence
increased intra-abdominal pressure impedes supporting the use of one device over another for
the ability of PPV to predict fluid responsive- hemodynamic monitoring in patients undergoing
ness [80]; this situation may occur during lapa- major abdominal surgery is weak.
roscopic surgery that is increasingly used for
liver resection (see above). PPV may also be
affected by open abdominal surgery and has Pulmonary Artery Catheter
never been validated in this context. Thirdly,
Cannesson et al. identified a “grey zone” of Pulmonary artery (Swan Ganz) catheter represents
PPV values i.e. a range of PPV values (between a potentially useful device to guide optimization
9 and 13%), for which fluid responsiveness of hemodynamics (fluid loading, catecholamines)
cannot be reliably predicted and that accounted in patients undergoing liver transplantation,
for up to 25% of patients. Finally, other limi- although no outcome benefit has been reported.
tations of PPV are spontaneous breathing, It provides accurate, continuous, rapid response
irregular heart rate beats, low heat rate/respira- time, precise, reproducible, operator-­independent,
tory rate ratio and open thoracic cavity [81]. and low-cost information on systemic and pulmo-
The ability of a similar dynamic index derived nary hemodynamics. A major limitation of the
from arterial waveform pulse contour analy- pulmonary artery catheter is the poor performance
sis (Flowtrac®) to predict fluid responsiveness of the pulmonary wedge pressure as a reliable
during abdominal surgery is controversial [82, marker of left ventricular filling [88]. Mixed (or
83]. Other non-invasive volume-clamp (Nex- central) venous oxygen saturation obtained with
fin®) or bioreactance-based (Nicom®) methods a pulmonary artery catheter may provide useful
allowing fluid responsiveness monitoring are information on rapid changes occurring in patients
interesting [84] but their accuracy also remains with compromised left ventricular failure [89].
to be clarified in liver surgery. The routine use of the pulmonary artery catheter
356 E. Weiss et al.

cannot be ­recommended due to its invasiveness the internal jugular vein for venovenous bypass
and potential for complications. [91]. Its perioperative use is generally associ-
ated with a low incidence of complications and
the presence of esophagogastric varices is not
Transoesophageal Echocardiography contraindication to its use (especially grade
(TEE) 1 or 2 esopageal varices) [92, 93] if precau-
tions are taken. Of note, a preliminary study
TEE is an increasingly popular imaging tool performed during liver surgery suggested that
that provides immediate visual information TEE-measured left atrial dimension may change
about the dynamic function of the heart. It deliv- earlier than CVP during acute blood loss [94].
ers accurate, precise, real-time information on TEE use is expensive and dependent on opera-
ventricular filling, stroke volume, and myocar- tor experience and cannot be continuously used
dial dynamics [90]. TEE further visualizes air in the postoperative period. Moreover, no data
embolism in the right heart circulation originat- demonstrated the superiority of this monitor
ing from the hepatic veins or inferior vena cava over any other device in patients undergoing
and can help guide percutaneous cannulation of major liver surgery (Fig. 27.3).

Fig. 27.3  Intraoperative view of the supradiaphragmatic graphic view, Lower panel: Time-motion Doppler
inferior vena cava by transesophageal Echocardiography recording via an axis corresponding to the dotted line,
(TEE) illustrating the respiratory variations of the diam- with (from top to bottom) the right atrium, the superior
eter of the inferior vena cava. Upper panel: 2D echo- vena cava and the left atrium
27  Liver Resection Surgery: Anesthetic Management, Monitoring, Fluids and Electrolytes 357

In summary, the best hemodynamic monitor- hepatic sinusoid pressure, lowering CVP during
ing for liver resection remains to be determined. liver resection can reduce hepatic parenchymal
The preference should be given to a monitoring congestion and subsequent blood loss by helping
device that is able to predict the response to ven- to control hepatic venous hemorrhage [99]. High
tricular filling, such as DPP or SVV or TEE when CVP values (>10 mmHg) may cause uncontrolla-
feasible. Frequent measurements of lactate con- ble retrograde bleeding occurring during clamping
centrations may be useful to ensure adequate of the portal triad, but whether CVP can be rec-
intraoperative oxygen delivery to the tissues. ommended as a monitor or endpoint to guide the
Recently, Vibert et al. showed that lactate con- hemodynamic management of patients undergo-
centration at the end of surgery is an early out- ing major hepatic surgery (liver resection or trans-
come predictor after major hepatectomy [17]. plantation) remains a matter of debate [100–103].
These finding are highlighting the impact of Continuous monitoring of CVP has been justified
intraoperative events and of hemodynamic man- for a long time in liver surgery because of several
agement during liver resection surgery [17]. studies suggested that maintaining a CVP below
5 mmHg was associated with improved outcome
and a decreased transfusion requirements dur-
Hemodynamic Management ing liver resection [99, 104, 105] or transplanta-
tion [106, 107]. Noteworthy, most of the studies
Maintaining effective intravascular volume to that support a low CVP strategy have some major
ensure tissue perfusion and cellular oxygenation methodological flaws as they were either retro-
is the physiological goal independent of the type spective or prospective nonrandomized cohorts
of surgery. This consideration also applies to with a low number of patients or underpowered
major liver surgery (resection, transplantation), randomized controlled trials. Surgical situations
with particular emphasis on liver perfusion and can affect CVP measurement reliability: wrong
oxygenation. Fluid therapy has to be balanced placement of the pressure transducer, liver manipu-
between underuse leading to hypovolemia and lation, occasional clamping of inferior vena cave,
inappropriate tissue perfusion and, administra- hepatic veins or even portal vein by the surgeon,
tion of excessive fluids with subsequent risks of changes in pericardial, intrathoracic (in particular
pulmonary and peripheral edema and hepatic positive and expiratory pressure, PEEP) and intra-
congestion. abdominal pressure and frequent patient position
For a few years, fixed regimens of fluid infu- changes [108]. Measured CVP may therefore not
sion (measured in milliliters per kilogram per always reflect hepatic vein or transection zone
hour) that can be liberal or restrictive were used pressures. Moreover, a meta-­ analysis including
but a growing body of evidence suggests now that all randomized clinical trials comparing various
intraoperative hemodynamic goal-directed ther- cardiopulmonary interventions aimed at decreas-
apy to increase blood flow may best to reduce ing blood loss and transfusion requirements in
postoperative morbidity [68, 70, 71, 95] and mor- patients undergoing liver resection and showed
tality in the higher-risk groups of patients [68, no significant beneficial impact of the “low CVP”
96]. Fluid therapy should therefore be individual- strategy on transfusion, surgical complications or
ized by using specific goals of care (such as mortality [109]. Thus, the benefit of “low CVP”
stroke volume or cardiac output optimization) to technique seems only controversial while its mor-
allow early correction of fluid deficits and avoid bidity remains poorly evaluated. Potential fatal
excessive administration by fluid titration. consequences of the low CVP technique during
The use of low CVP as an index of preload has hepatectomy include air embolism and unneces-
been popular in hepatic surgery [97]. Although sary hypoperfusion [104, 105]. Briefly, reducing
CVP may indirectly reflect volume status, it is not CVP can only be obtained by rendering the patient
a reliable predictor of the response to fluid load- hypovolemic, by hemorrhage, partial clamping of
ing [98] and associated with many limitations in the inferior vena cava, elective addition of diuretics
hepatic surgery. Because CVP is thought to reflect or vasodilators, intraoperative epidural analgesia or
358 E. Weiss et al.

increasing depth of anesthesia. This is feasible in optimization will hopefully provides some possible
minor or intermediate liver resection or in young answers to this question [120]. For the time we must
healthy patient but not in older patients with severe assume that any starches are associated with an
comorbidities undergoing major liver resection. increased risk of renal injury and are to be avoided.
Some studies employed strategies to reduce CVP The administration of (large volume of) 0.9%
included the use of diuretics and even severe hypo- saline may contribute to the development of
volemia requiring high-dose vasopressors to main- hyperchloremic metabolic acidosis and AKI
tain blood pressure. This strategy increases the risk [121–123]. The use of balanced rather than non-­
of postoperative renal failure and is not recom- balanced crystalloid solutions has been pro-
mended for routine use. posed as a pragmatic alternative to 0.9% saline,
In general, minimizing fluid administration but only limited evidence is currently available
until the resection is completed (irregardless of concerning comparable efficacy and safety of
the CVP) is likley useful to decrease bleeding and use [111] without demonstration of a clinical
improve visualization of the surgical field. This is benefit in surgical patients [124, 125].
usually well tolerated as long as the patient is rela- Plasmalyte solutions are preferrable over lacate
tively healthy and fluid deficits are corrected after ringer as the buffers in plasmalyte, gluconate
resection and hemostasis are completed. and acetate do not require hepatic metabolism
(unlike lactate).

Type of Fluids
Transfusion
Numerous studies, reviews and meta-analyses
reported that new-generation and low-molecular-­ Red cell transfusion is a cornerstone of periop-
weight preparations of hydroxyethyl starch (HES) as erative care and determinant of outcome after
compared to cristalloids increase the risk for acute major liver surgery. Five to 20% (up to 50% in
kidney injury, renal replacement therapy and trans- some studies) of elective liver resections require
fusion while providing no short-­term hemodynamic red cell transfusions intraoperatively [126].
resuscitation benefit for critically ill or septic patients Massive red cell transfusion is necessary in 1 out
[110–114]. These data have led the of about 20–30 patients, while the incidence of
Pharmacovigilance Risk Assessment Committee transfusion of plasma and platelets is very low.
(PRAC) of the European Medicines Agency to sus- Risk factors for transfusion are the presence of
pend the authorization to use HES in cases of sepsis, cirrhosis, the extent of liver resection, and portal
burn injury or critically ill patients but have allowed hypertension. Scores with good discriminatory
the continued use of HES in surgical patients. ability to predict the necessity of red cell transfu-
Indeed, the detrimental effects of intraoperative use sion during liver resection have been developed
of colloids are less clear. While some studies also [126–128]. Growing evidence supports that red
reported dose dependent renal toxicity [115] (includ- cell transfusion is associated with a significantly
ing in liver transplantation [116]) and decreased worsened outcome and cost after anesthesia and
coagulation [117] with hydroxyethylstarchs (HES), surgery [129]. Operative blood loss during resec-
majority of meta-analyses including studies con- tion of hepatocellular carcinoma was found to
ducted on heterogeneous patient groups with a low be a predictor of recurrence and survival rates
risk of complications and reported no difference of [130]. Increasing attention has focused on the
death or renal dysfunction between crystalloids and potential risk of cancer progression associated
low molecular weight HES [113, 118, 119]. An with red cell transfusions. In an experimental
ongoing multicenter controlled trial randomizing study of rats, transfusion of autologous or allo-
patients with moderate-to-high risk of postoperative geneic aged red cells was responsible for an
complications to receive 0.9% saline and last gener- increased retention of tumoral cells in the lung,
ation HES during individualized goal-directed fluid and this was clearly related to erythrocytes and
27  Liver Resection Surgery: Anesthetic Management, Monitoring, Fluids and Electrolytes 359

not leukocytes or soluble factors contained in the As described in detail elsewhere in this book,
plasma. A possible explanation would be that the evaluation of the coagulation in liver disease
transfused erythrocytes impair cellular immunity, solely based on conventional coagulation mark-
­particularly natural killer (NK) cells, by decreas- ers is insufficient [140]. Other changes that
ing their efficacy to eliminate tumoral cells [131]. accompany chronic liver disease may restore the
These considerations underscore the importance balance of anticoagulant and procoagulant effects
of minimizing perioperative blood loss in patients [140]. The decision to transfuse blood products
undergoing liver resection. Although some blood should therefore not be based on the biological or
salvage techniques or pharmacologic interven- laboratory abnormalities of coagulation only
tions to reduce blood loss may be safe and effec- (i.e., increased INR or low platelet count), since
tive in patients undergoing liver resection surgery these abnormalities poorly predict intraoperative
[132, 133], none of these interventions targeting bleeding. Mallett et al. recently showed that
reduction of perioperative bleeding have resulted whereas major liver resection is assumed to trig-
in a demonstrable decreased mortality or morbid- ger a potential bleeding risk, the kinetic of pro-
ity rate [109, 134]. Tranexamic acid decreases and anticoagulant factors show an imbalance
fibrinolysis and may allow a reduction in blood suggesting a global prothrombotic state in the
loss and transfusion requirements [132, 135]. early postoperative period [141].
There were no significant differences in throm-
bosis between groups although none of these
studies were designed to evaluate safety and only Mechanical Ventilation
a small overall number of events were recorded.
A randomized trial by Lodge et al. did not find There is currently a trend towards a reduction in
that recombinant coagulation factor VIIa reduced tidal volume, not only in ICU patients with acute
either the number of patients requiring transfu- lung injury, but also in patients with healthy lungs
sion or the amount of red cell units administered undergoing surgery. The IMPROVE study [78]
during liver resection [136]. Without an available showed that in patients at intermediate to high
alternative therapy, red cell transfusion remains risk of pulmonary complications after major
the only way to compensate for severe blood abdominal surgery protective ventilation (tidal
loss or persisting hemorrhage. Delaying transfu- volume (TV) of 6 mL/kg of ideal body weight
sions because of underestimation of the severity (IBW) associated with 6–8 cm H2O PEEP and
of hemorrhage undoubtedly causes a significant recruitment manœuvres) during surgery is asso-
number of deaths within the 24 first postoperative ciated with a better post-surgical outcome than
hours [137, 138]. Jarnagin et al. showed that in non-protective ventilation (TV 10–12 mL/kg
patients undergoing major liver resection, a target IBW, no PEEP and no recruitment manœuvres).
hemoglobin level of 8 g/dL resulted in a signifi- A different study comparing intraoperative
cant reduction in red cell requirements compared mechanical ventilation settings and outcome
to standard transfusion strategy [139]. However done in 29,343 patients who underwent general
there was no effect of acute normovolemic hemo- anesthesia found an increased 30-day mortality
dilution on postoperative complications [139]. It and hospital length of stay in patients with low
should be emphasized that an intraoperative target tidal volume and minimal PEEP (<3 cm H2O)
hemoglobin concentration of 8 g/dL corresponds suggesting that low TD is beneficial only in con-
to the “standard” practice in many institutions, junction with the application of PEEP and recur-
while transfusing patients with hemoglobin lev- rent recruitment manœuvres [142]. During liver
els greater than this was defined as the “standard resection surgery PEEP has traditionally been
practice” in this study. A reasonable threshold thought to worsen blood loss during hepatic tran-
for transfusion in the operating room is within a section by increasing CVP and hepatic venous
hemoglobin level between 7 and 8 g/dL, in the pressures [143, 144]. However, this model has
absence of active coronary disease. recently been challenged by a post-hoc analysis
360 E. Weiss et al.

of the IMPROVE study assessing the impact of 154]. However, some components of ERAS
mechanical ventilation with PEEP on bleeding in programmes remain debated, especially in
the subgroup of patients that underwent hepatec- liver surgery. Regional analgesia techniques
tomy. Intraoperative blood loss and transfusion are effective in decreasing postoperative pain
requirements did not differ between the 41 after major abdominal surgery [155] and sub-
patients in the lung protective (TV: 6 mL/kg of group analysis of randomized controlled trials
IBW, 6–8 cm H2O PEEP and recruitment suggests a benefit from postoperative epidural
manœuvres) and the 38 in the non-protective analgesia with local anesthetics with a reduc-
groups (TV 10–12 mL/kg IBW, no PEEP and no tion of respiratory complications after major
recruitment manœuvres). Whether these results abdominal surgery. However, there are unre-
were also related to a decrease in TV, as previ- solved issues regarding safety and efficacy of
ously suggested by Lasndorp et al. is unknown epidural anesthesia for patients undergoing
[145]. The posthoc analysis of the IMPROVE hepatic resection [156]. Firstly, the difficult
study has some inevitable limitations and its assessment of coagulopathy that may develop
results need to be confirmed but suggest that, as during the postoperative period may increase
in the case of hemodynamic management, intra- the risk of epidural hematoma. Although, no
operative mechanical ventilation optimization is study was, to date, sufficiently powered to
feasible during liver resection surgery. address this question, postoperative coagulop-
athy may result either in delayed removal of
epidural catheter or unnecessary fresh frozen
Postoperative Analgesia plasma transfusion [156]. Other regional anes-
and Rehabilitation thesia techniques such as intrathecal morphine
[157] or continuous local anesthetic infiltration
Postoperative rehabilitation is highly recom- via wound catheter [158] may provide effective
mended and adopted for colorectal and ortho- control of postoperative pain after liver surgery
paedic surgery as it facilitates recovery by and can be an acceptable alternative to thoracic
reducing length of hospital stay and morbidity epidural analgesia.
while improving the value of care and patient Minimally invasive surgical techniques such as
satisfaction after surgery [146–148]. Enhanced laparoscopic liver resection (LLR) should be more
recovery after surgery (ERAS) programms frequently included into ERAS programmes.
focus on patient education, goal-directed fluid Laparoscopic liver resection (LLR) has been pro-
management, decreased use of unnecessary gressively developed along the past two decades
nasogastric tubes and peritoneal drains, mini- yet, it is still only used in 15% of liver surgery in
mal use of opioid analgesia, as well as early countries with long­ standing tradition in surgery
mobilization and resumption of oral intake partly because of careful patient selection (number,
[147, 149–151]. After liver surgery some size and position of tumors). When technically fea-
ERAS components (e.g. immediate postopera- sible LLR should become a standard practice as far
tive removal of the nasogastric tube [152]) are as it does not seem to affect oncological results or
encouraged and we now have some evidence long-term survival and it allows shorter hospital
suggesting that perioperative surgical path- length of stay, earlier return of bowel activity and
ways are also safe and effective in the context lesser requirement of analgesics, as compared to
of open liver resection surgery [149, 153, 154]. open techniques [159, 160].
ERAS programmes are well endorsed by care To conclude, the optimal management of
providers and are associated with reduction patients undergoing liver resection surgery
in opioid use, faster recovery, shorter hospital requires the implementation of a bundle of care
stay and decreased hospital costs [149, 153, rather than one main measure or treatment. The
27  Liver Resection Surgery: Anesthetic Management, Monitoring, Fluids and Electrolytes 361

improvement of liver resection outcome over the 10. Mullen JT, Ribero D, Reddy SK, et al. Hepatic insuf-
past decade is due to a combination of factors ficiency and mortality in 1,059 noncirrhotic patients
undergoing major hepatectomy. J Am Coll Surg.
including better preoperative evaluation, closer 2007;204:854–62; discussion 62–4.
intraoperative management of hemodynam- 11. Schroeder RA, Marroquin CE, Bute BP, Khuri S,
ics, transfusion and mechanical ventilation and Henderson WG, Kuo PC. Predictive indices of mor-
increasing diffusion of enhanced recovery pro- bidity and mortality after liver resection. Ann Surg.
2006;243:373–9.
grammes. New approaches for the management 12. Ercolani G, Ravaioli M, Grazi GL, et al. Use of vascu-
of ischemia-reperfusion injuries or the develop- lar clamping in hepatic surgery: lessons learned from
ment of minimally invasive surgery may repre- 1260 liver resections. Arch Surg. 2008;143:380–7;
sent therapeutic prospects. discussion 8.
13. Imamura H, Seyama Y, Kokudo N, et al. One thousand
fifty-six hepatectomies without mortality in 8 years.
Arch Surg. 2003;138:1198–206; discussion 206.
References 14. Belghiti J, Regimbeau JM, Durand F, et al. Resection
of hepatocellular carcinoma: a European experi-
1. Jarnagin WR, Gonen M, Fong Y, et al. ence on 328 cases. Hepato-Gastroenterology. 2002;
Improvement in perioperative outcome after |49:41–6.
hepatic resection: analysis of 1,803 consecu- 15. Adam R, Chiche L, Aloia T, et al. Hepatic resec-
tive cases over the past decade. Ann Surg. tion for noncolorectal nonendocrine liver metasta-
2002;236:397–406; discussion 7. ses: analysis of 1,452 patients and development of a
2. Farges O, Goutte N, Bendersky N, Falissard B.  prognostic model. Ann Surg. 2006;244:524–35.
Group AC-FHS. Incidence and risks of liver resec- 16. European Association for the Study of the L,
tion: an all-inclusive French nationwide study. Ann European Organisation For R, Treatment of C.
Surg. 2012;256:697–704; discussion 5. EASL-EORTC clinical practice guidelines: man-
3. Kamiyama T, Nakanishi K, Yokoo H, et al. agement of hepatocellular carcinoma. J Hepatol.
Perioperative management of hepatic resection 2012;56:908–43.
toward zero mortality and morbidity: analysis of 793 17. Vibert E, Boleslawski E, Cosse C, et al. Arterial
consecutive cases in a single institution. J Am Coll lactate concentration at the end of an elective hepa-
Surg. 2010;211:443–9. tectomy is an early predictor of the postoperative
4. Konopke R, Kersting S, Bunk A, et al. Colorectal course and a potential surrogate of intraoperative
liver metastasis surgery: analysis of risk factors pre- events. Ann Surg. 2015;262:787–92; discussion
dicting postoperative complications in relation to the 92–3.
extent of resection. Int J Color Dis. 2009;24:687–97. 18. Fleisher LA, Beckman JA, Brown KA, et al. ACC/
5. Citterio D, Facciorusso A, Sposito C, Rota R, AHA 2006 guideline update on perioperative cardio-
Bhoori S, Mazzaferro V. Hierarchic interaction of vascular evaluation for noncardiac surgery: focused
factors associated with liver decompensation after update on perioperative beta-blocker therapy--a report
resection for hepatocellular carcinoma. JAMA Surg. of the American College of Cardiology/American
2016;151(9):846–53. Heart Association task force on practice guidelines
6. Aussilhou B, Dokmak S, Faivre S, Paradis V, Vilgrain (writing committee to update the 2002 guidelines on
V, Belghiti J. Preoperative liver hypertrophy induced perioperative cardiovascular evaluation for noncardiac
by portal flow occlusion before major hepatic resec- surgery). Anesth Analg. 2007;104:15–26.
tion for colorectal metastases can be impaired by 19. Poldermans D, Bax JJ, Boersma E, et al. Guidelines
bevacizumab. Ann Surg Oncol. 2009;16:1553–9. for pre-operative cardiac risk assessment and periop-
7. Scoggins CR, Campbell ML, Landry CS, et al. erative cardiac management in non-cardiac surgery: the
Preoperative chemotherapy does not increase mor- task force for preoperative cardiac risk assessment and
bidity or mortality of hepatic resection for colorectal perioperative cardiac management in non-cardiac sur-
cancer metastases. Ann Surg Oncol. 2009;16:35–41. gery of the European Society of Cardiology (ESC) and
8. Belghiti J, Hiramatsu K, Benoist S, Massault P, endorsed by the European Society of Anaesthesiology
Sauvanet A, Farges O. Seven hundred forty-seven (ESA). Eur J Anaesthesiol. 2010;27:92–137.
hepatectomies in the 1990s: an update to evaluate 20. McFalls EO, Ward HB, Moritz TE, et al. Coronary-­
the actual risk of liver resection. J Am Coll Surg. artery revascularization before elective major vascu-
2000;191:38–46. lar surgery. N Engl J Med. 2004;351:2795–804.
9. Schindl MJ, Redhead DN, Fearon KC, et al. The 21. Dunkelgrun M, Boersma E, Schouten O, et al.
value of residual liver volume as a predictor of Bisoprolol and fluvastatin for the reduction of peri-
hepatic dysfunction and infection after major liver operative cardiac mortality and myocardial infarction
resection. Gut. 2005;54:289–96. in intermediate-risk patients undergoing noncar-
362 E. Weiss et al.

diovascular surgery: a randomized controlled trial 35. Braga M, Gianotti L, Vignali A, Carlo VD. Preoperative
(DECREASE-IV). Ann Surg. 2009;249:921–6. oral arginine and n-3 fatty acid supplementation
22. Group PS, Devereaux PJ, Yang H, et al. Effects of improves the immunometabolic host response and
extended-release metoprolol succinate in patients outcome after colorectal resection for cancer. Surgery.
undergoing non-cardiac surgery (POISE trial): a ran- 2002;132:805–14.
domised controlled trial. Lancet. 2008;371:1839–47. 36. Braga M, Gianotti L, Radaelli G, et al. Perioperative
23. Wijeysundera DN, Duncan D, Nkonde-Price C, immunonutrition in patients undergoing cancer sur-
et al. Perioperative beta blockade in noncardiac sur- gery: results of a randomized double-blind phase 3
gery: a systematic review for the 2014 ACC/AHA trial. Arch Surg. 1999;134:428–33.
guideline on perioperative cardiovascular evalua- 37. Ciacio O, Voron T, Pittau G, et al. Interest of pre-
tion and management of patients undergoing non- operative immunonutrition in liver resection for
cardiac surgery: a report of the American College cancer: study protocol of the PROPILS trial, a mul-
of Cardiology/American Heart Association Task ticenter randomized controlled phase IV trial. BMC
Force on practice guidelines. J Am Coll Cardiol. Cancer. 2014;14:980.
2014;64:2406–25. 38. Servin FS, Bougeois B, Gomeni R, Mentre F,
24. Mantz J, Samama CM, Tubach F, et al. Impact of Farinotti R, Desmonts JM. Pharmacokinetics of
preoperative maintenance or interruption of aspirin propofol administered by target-controlled infusion
on thrombotic and bleeding events after elective to alcoholic patients. Anesthesiology. 2003;99:
non-cardiac surgery: the multicentre, randomized, 576–85.
blinded, placebo-controlled, STRATAGEM trial. Br 39. Passot S, Servin F, Pascal J, Charret F, Auboyer C,
J Anaesth. 2011;107:899–910. Molliex S. A comparison of target- and manually
25. Oscarsson A, Gupta A, Fredrikson M, et al. To controlled infusion propofol and etomidate/desflu-
continue or discontinue aspirin in the perioperative rane anesthesia in elderly patients undergoing hip
period: a randomized, controlled clinical trial. Br J fracture surgery. Anesth Analg. 2005;100:1338–42;
Anaesth. 2010;104:305–12. table of contents.
26. Devereaux PJ, Mrkobrada M, Sessler DI, et al. 40. Wang ZX, Huang CY, Hua YP, Huang WQ, Deng
Aspirin in patients undergoing noncardiac surgery. LH, Liu KX. Dexmedetomidine reduces intestinal
N Engl J Med. 2014;370:1494–503. and hepatic injury after hepatectomy with inflow
27. Myles PS, Smith JA, Forbes A, et al. Stopping vs. occlusion under general anaesthesia: a randomized
continuing aspirin before coronary artery surgery. N controlled trial. Br J Anaesth. 2014;112:1055–64.
Engl J Med. 2016;374:728–37. 41. Gatecel C, Losser MR, Payen D. The postoperative
28. Nobili C, Marzano E, Oussoultzoglou E, et al. effects of halothane versus isoflurane on hepatic
Multivariate analysis of risk factors for pulmonary artery and portal vein blood flow in humans. Anesth
complications after hepatic resection. Ann Surg. Analg. 2003;96:740–5; table of contents.
2012;255:540–50. 42. Yang LQ, Tao KM, Cheung CW, Liu YT, Tao Y,
29. Slankamenac K, Breitenstein S, Held U, Beck-­ Wu FX, Yu WF. The effect of isoflurane or pro-
Schimmer B, Puhan MA, Clavien PA. Development pofol anaesthesia on liver injury after partial
and validation of a prediction score for postopera- hepatectomy in cirrhotic patients. Anaesthesia.
tive acute renal failure following liver resection. Ann 2010;65:1094–100.
Surg. 2009;250:720–8. 43. Song JC, Sun YM, Yang LQ, Zhang MZ, Lu ZJ,
30. Corish CA, Kennedy NP. Protein-energy undernu- Yu WF. A comparison of liver function after hepa-
trition in hospital in-patients. Br J Nutr. 2000;83: tectomy with inflow occlusion between sevoflu-
575–91. rane and propofol anesthesia. Anesth Analg. 2010;
31. Gedaly R, McHugh PP, Johnston TD, Jeon H, 111:1036–41.
Ranjan D, Davenport DL. Obesity, diabetes, and 44. Clavien PA, Petrowsky H, DeOliveira ML, Graf
smoking are important determinants of resource uti- R. Strategies for safer liver surgery and partial liver
lization in liver resection: a multicenter analysis of transplantation. N Engl J Med. 2007;356:1545–59.
1029 patients. Ann Surg. 2009;249:414–9. 45. Rahbari NN, Wente MN, Schemmer P, et al.
32. Harimoto N, Shirabe K, Yamashita YI, et al. Systematic review and meta-analysis of the effect of
Sarcopenia as a predictor of prognosis in patients portal triad clamping on outcome after hepatic resec-
following hepatectomy for hepatocellular carci- tion. Br J Surg. 2008;95:424–32.
noma. Br J Surg. 2013;100:1523–30. 46. Delva E, Camus Y, Paugam C, Parc R, Huguet
33. Otsuji H, Yokoyama Y, Ebata T, et al. Preoperative sar- C, Lienhart A. Hemodynamic effects of por-
copenia negatively impacts postoperative outcomes tal triad clamping in humans. Anesth Analg.
following major hepatectomy with extrahepatic bile 1987;66:864–8.
duct resection. World J Surg. 2015;39:1494–500. 47. Delva E, Nordlinger B, Parc R, Lienhart A, Hannoun
34. Voron T, Tselikas L, Pietrasz D, et al. Sarcopenia L, Huguet C. Hepatic vascular exclusion (HVE) for
impacts on short- and long-term results of hepa- major liver resections. Int Surg. 1987;72:78–81.
tectomy for hepatocellular carcinoma. Ann Surg. 48. Belghiti J, Noun R, Zante E, Ballet T, Sauvanet
2015;261:1173–83. A. Portal triad clamping or hepatic vascular exclu-
27  Liver Resection Surgery: Anesthetic Management, Monitoring, Fluids and Electrolytes 363

sion for major liver resection. A controlled study. response by interfering with CXC receptor-2 signal-
Ann Surg. 1996;224:155–61. ling. Br J Anaesth. 2015;114:143–9.
49. Eyraud D, Richard O, Borie DC, et al. Hemodynamic 63. Yang LQ, Tao KM, Liu YT, et al. Remifentanil pre-
and hormonal responses to the sudden interruption conditioning reduces hepatic ischemia-reperfusion
of caval flow: insights from a prospective study of injury in rats via inducible nitric oxide synthase
hepatic vascular exclusion during major liver resec- expression. Anesthesiology. 2011;114:1036–47.
tions. Anesth Analg. 2002;95:1173–8; table of 64. Simillis C, Robertson FP, Afxentiou T, Davidson
contents. BR, Gurusamy KS. A network meta-analysis
50. Lai EC, Fan ST, Lo CM, Chu KM, Liu CL. Anterior comparing perioperative outcomes of interven-
approach for difficult major right hepatectomy. tions aiming to decrease ischemia reperfusion
World J Surg. 1996;20:314–7; discussion 8. injury during elective liver resection. Surgery.
51. Ogata S, Belghiti J, Varma D, et al. Two hundred 2016;159:1157–69.
liver hanging maneuvers for major hepatectomy: a 65. Marsman HA, de Graaf W, Heger M, et al. Hepatic
single-center experience. Ann Surg. 2007;245:31–5. regeneration and functional recovery following partial
52. Zhai Y, Busuttil RW, Kupiec-Weglinski JW. Liver liver resection in an experimental model of hepatic
ischemia and reperfusion injury: new insights into steatosis treated with omega-3 fatty acids. Br J Surg.
mechanisms of innate-adaptive immune-­ mediated 2013;100:674–83.
tissue inflammation. Am J Transplant. 2011;11: 66. Raptis DA, Limani P, Jang JH, et al. GPR120 on
1563–9. Kupffer cells mediates hepatoprotective effects of
53. Selzner N, Rudiger H, Graf R, Clavien PA. Protective omega3-fatty acids. J Hepatol. 2014;60:625–32.
strategies against ischemic injury of the liver. 67. Linecker M, Limani P, Botea F, et al. A random-
Gastroenterology. 2003;125:917–36. ized, double-blind study of the effects of omega-3
54. Petrowsky H, McCormack L, Trujillo M, Selzner fatty acids (Omegaven) on outcome after major liver
M, Jochum W, Clavien PA. A prospective, random- resection. BMC Gastroenterol. 2015;15:102.
ized, controlled trial comparing intermittent portal 68. Hamilton MA, Cecconi M, Rhodes A. A systematic
triad clamping versus ischemic preconditioning with review and meta-analysis on the use of preemp-
continuous clamping for major liver resection. Ann tive hemodynamic intervention to improve postop-
Surg. 2006;244:921–8; discussion 8–30. erative outcomes in moderate and high-risk surgical
55. Scatton O, Zalinski S, Jegou D, et al. Randomized patients. Anesth Analg. 2011;112:1392–402.
clinical trial of ischaemic preconditioning in major 69. Grocott MP, Dushianthan A, Hamilton MA, et al.
liver resection with intermittent Pringle manoeuvre. Perioperative increase in global blood flow to explicit
Br J Surg. 2011;98:1236–43. defined goals and outcomes following surgery. Coch­
56. O’Neill S, Leuschner S, McNally SJ, Garden OJ, rane Database Syst Rev. 2012;11:CD004082.
Wigmore SJ, Harrison EM. Meta-analysis of isch- 70. Pearse RM, Harrison DA, MacDonald N, et al.
aemic preconditioning for liver resections. Br J Surg. Effect of a perioperative, cardiac output-guided
2013;100:1689–700. hemodynamic therapy algorithm on outcomes fol-
57. Benzoni E, Lorenzin D, Baccarani U, et al. Resective lowing major gastrointestinal surgery: a random-
surgery for liver tumor: a multivariate analysis ized clinical trial and systematic review. JAMA.
of causes and risk factors linked to postopera- 2014;311:2181–90.
tive complications. Hepatobiliary Pancreat Dis Int. 71. Gomez-Izquierdo JC, Feldman LS, Carli F, Baldini
2006;5:526–33. G. Meta-analysis of the effect of goal-directed ther-
58. Beck-Schimmer B, Breitenstein S, Urech S, et al. A apy on bowel function after abdominal surgery. Br J
randomized controlled trial on pharmacological pre- Surg. 2015;102:577–89.
conditioning in liver surgery using a volatile anes- 72. Li Z, Sun YM, Wu FX, Yang LQ, Lu ZJ, Yu
thetic. Ann Surg. 2008;248:909–18. WF. Controlled low central venous pressure reduces
59. Beck-Schimmer B, Breitenstein S, Bonvini JM, blood loss and transfusion requirements in hepatec-
et al. Protection of pharmacological postcondition- tomy. World J Gastroenterol. 2014;20:303–9.
ing in liver surgery: results of a prospective random- 73. Ziser A, Plevak DJ, Wiesner RH, Rakela J, Offord
ized controlled trial. Ann Surg. 2012;256:837–44. KP, Brown DL. Morbidity and mortality in cir-
discission 44–5 rhotic patients undergoing anesthesia and surgery.
60. Ryter SW, Alam J, Choi AM. Heme oxygenase-1/ Anesthesiology. 1999;90:42–53.
carbon monoxide: from basic science to therapeutic 74. Michard F, Chemla D, Richard C, et al. Clinical use
applications. Physiol Rev. 2006;86:583–650. of respiratory changes in arterial pulse pressure to
61. Barrier A, Olaya N, Chiappini F, et al. Ischemic monitor the hemodynamic effects of PEEP. Am J
preconditioning modulates the expression of sev- Respir Crit Care Med. 1999;159:935–9.
eral genes, leading to the overproduction of IL-1Ra, 75. Salzwedel C, Puig J, Carstens A, et al. Perioperative
iNOS, and Bcl-2 in a human model of liver ischemia-­ goal-directed hemodynamic therapy based on radial
reperfusion. FASEB J. 2005;19:1617–26. arterial pulse pressure variation and continuous
62. Muller-Edenborn B, Frick R, Piegeler T, et al. cardiac index trending reduces postoperative com-
Volatile anaesthetics reduce neutrophil inflammatory plications after major abdominal surgery: a multi-­
364 E. Weiss et al.

center, prospective, randomized study. Crit Care. 90. Burtenshaw AJ, Isaac JL. The role of trans-­
2013;17:R191. oesophageal echocardiography for perioperative
76. Goepfert MS, Richter HP, Zu Eulenburg C, et al. cardiovascular monitoring during orthotopic liver
Individually optimized hemodynamic therapy transplantation. Liver Transpl. 2006;12:1577–83.
reduces complications and length of stay in the 91. Planinsic RM, Nicolau-Raducu R, Caldwell JC,
intensive care unit: a prospective, randomized con- Aggarwal S, Hilmi I. Transesophageal echocardio­
trolled trial. Anesthesiology. 2013;119:824–36. graphy-­guided placement of internal jugular percu-
77. Ramsingh DS, Sanghvi C, Gamboa J, Cannesson M, taneous venovenous bypass cannula in orthotopic
Applegate RL 2nd. Outcome impact of goal directed liver transplantation. Anesth Analg. 2003;97:648–9.
fluid therapy during high risk abdominal surgery in 92. Burger-Klepp U, Karatosic R, Thum M, et al.
low to moderate risk patients: a randomized con- Transesophageal echocardiography during orthotopic
trolled trial. J Clin Monit Comput. 2013;27:249–57. liver transplantation in patients with esophagoastric
78. Futier E, Constantin JM, Paugam-Burtz C, et al. A varices. Transplantation. 2012;94:192–6.
trial of intraoperative low-tidal-volume ventilation in 93. Pantham G, Waghray N, Einstadter D, Finkelhor RS,
abdominal surgery. N Engl J Med. 2013;369:428–37. Mullen KD. Bleeding risk in patients with esopha-
79. De Backer D, Heenen S, Piagnerelli M, Koch M, geal varices undergoing transesophageal echocar-
Vincent JL. Pulse pressure variations to predict fluid diography. Echocardiography. 2013;30:1152–5.
responsiveness: influence of tidal volume. Intensive 94. Pissarra F, Oliveira A, Marcelino P. Transoesophageal
Care Med. 2005;31:517–23. echocardiography for monitoring liver sur-
80. Duperret S, Lhuillier F, Piriou V, et al. Increased gery: data from a pilot study. Cardiol Res Pract.
intra-abdominal pressure affects respiratory varia- 2012;2012:723418.
tions in arterial pressure in normovolaemic and 95. Corcoran T, Rhodes JE, Clarke S, Myles PS, Ho
hypovolaemic mechanically ventilated healthy pigs. KM. Perioperative fluid management strategies in
Intensive Care Med. 2007;33:163–71. major surgery: a stratified meta-analysis. Anesth Analg.
81. Michard F, Chemla D, Teboul JL. Applicability of 2012;114:640–51.
pulse pressure variation: how many shades of grey? 96. Cecconi M, Corredor C, Arulkumaran N, et al.
Crit Care. 2015;19:144. Clinical review: goal-directed therapy-what is the
82. Lahner D, Kabon B, Marschalek C, et al. Evaluation evidence in surgical patients? The effect on different
of stroke volume variation obtained by arterial pulse risk groups. Crit Care. 2013;17:209.
contour analysis to predict fluid responsiveness 97. Stephan F, Rezaiguia-Delclaux S. Usefulness of a
intraoperatively. Br J Anaesth. 2009;103:346–51. central venous catheter during hepatic surgery. Acta
83. Derichard A, Robin E, Tavernier B, et al. Automated Anaesthesiol Scand. 2008;52:388–96.
pulse pressure and stroke volume variations from 98. Marik PE, Baram M, Vahid B. Does central venous
radial artery: evaluation during major abdominal sur- pressure predict fluid responsiveness? A systematic
gery. Br J Anaesth. 2009;103:678–84. review of the literature and the tale of seven mares.
84. de Wilde RB, de Wit F, Geerts BF, et al. Non- Chest. 2008;134:172–8.
invasive continuous arterial pressure and pulse pres- 99. Jones RM, Moulton CE, Hardy KJ. Central venous
sure variation measured with Nexfin((R)) in patients pressure and its effect on blood loss during liver
following major upper abdominal surgery: a com- resection. Br J Surg. 1998;85:1058–60.
parative study. Anaesthesia. 2016;71:788–97. 100. Choi SJ, Gwak MS, Ko JS, et al. Can peripheral
85. Abbas SM, Hill AG. Systematic review of the lit- venous pressure be an alternative to central venous
erature for the use of oesophageal Doppler monitor pressure during right hepatectomy in living donors?
for fluid replacement in major abdominal surgery. Liver Transpl. 2007;13:1414–21.
Anaesthesia. 2008;63:44–51. 101. Schroeder RA, Kuo PC. Pro: low central venous
86. Grocott MP, Mythen MG, Gan TJ. Perioperative pressure during liver transplantation--not too low. J
fluid management and clinical outcomes in adults. Cardiothorac Vasc Anesth. 2008;22:311–4.
Anesth Analg. 2005;100:1093–106. 102. Massicotte L, Beaulieu D, Thibeault L. Con: low
87. Wakeling HG, McFall MR, Jenkins CS, et al. central venous pressure during liver transplantation.
Intraoperative oesophageal Doppler guided fluid J Cardiothorac Vasc Anesth. 2008;22:315–7.
management shortens postoperative hospital stay 103. Niemann CU, Feiner J, Behrends M, Eilers H,
after major bowel surgery. Br J Anaesth. 2005;95: Ascher NL, Roberts JP. Central venous pressure
634–42. monitoring during living right donor hepatectomy.
88. Halpern SD, Taichman DB. Misclassification of pul- Liver Transpl. 2007;13:266–71.
monary hypertension due to reliance on pulmonary 104. Melendez JA, Arslan V, Fischer ME, et al.
capillary wedge pressure rather than left ventricular Perioperative outcomes of major hepatic resections
end-diastolic pressure. Chest. 2009;136:37–43. under low central venous pressure anesthesia: blood
89. Dahmani S, Paugam-Burtz C, Gauss T, et al. loss, blood transfusion, and the risk of postoperative
Comparison of central and mixed venous saturation renal dysfunction. J Am Coll Surg. 1998;187:620–5.
during liver transplantation in cirrhotic patients: a 105. Smyrniotis V, Kostopanagiotou G, Theodoraki
pilot study. Eur J Anaesthesiol. 2010;27:714–9. K, Tsantoulas D, Contis JC. The role of central
27  Liver Resection Surgery: Anesthetic Management, Monitoring, Fluids and Electrolytes 365

venous pressure and type of vascular control in 119. Gillies MA, Habicher M, Jhanji S, et al. Incidence
blood loss during major liver resections. Am J Surg. of postoperative death and acute kidney injury asso-
2004;187:398–402. ciated with i.v. 6% hydroxyethyl starch use: sys-
106. Massicotte L, Lenis S, Thibeault L, Sassine MP, Seal tematic review and meta-analysis. Br J Anaesth.
RF, Roy A. Effect of low central venous pressure and 2014;112:25–34.
phlebotomy on blood product transfusion require- 120. Futier E, Biais M, Godet T, et al. Fluid loading in
ments during liver transplantations. Liver Transpl. abdominal surgery—saline versus hydroxyethyl
2006;12:117–23. starch (FLASH Trial): study protocol for a random-
107. Feng ZY, Xu X, Zhu SM, Bein B, Zheng SS. Effects ized controlled trial. Trials. 2015;16:582.
of low central venous pressure during preanhepatic 121. Yunos NM, Bellomo R, Hegarty C, Story D, Ho L,
phase on blood loss and liver and renal function in Bailey M. Association between a chloride-liberal
liver transplantation. World J Surg. 2010;34:1864–73. vs chloride-restrictive intravenous fluid administra-
108. Sondergaard S, Parkin G, Aneman A. Central tion strategy and kidney injury in critically ill adults.
venous pressure: we need to bring clinical use into JAMA. 2012;308:1566–72.
physiological context. Acta Anaesthesiol Scand. 122. Reid F, Lobo DN, Williams RN, Rowlands BJ,
2015;59:552–60. Allison SP. (Ab)normal saline and physiological
109. Gurusamy KS, Li J, Vaughan J, Sharma D, Davidson Hartmann’s solution: a randomized double-blind
BR. Cardiopulmonary interventions to decrease crossover study. Clin Sci (Lond). 2003;104:17–24.
blood loss and blood transfusion requirements 123. McCluskey SA, Karkouti K, Wijeysundera D,
for liver resection. Cochrane Database Syst Rev. Minkovich L, Tait G, Beattie WS. Hyperchloremia
2012;5:CD007338. after noncardiac surgery is independently associ-
110. Zarychanski R, Abou-Setta AM, Turgeon AF, et al. ated with increased morbidity and mortality: a
Association of hydroxyethyl starch administration propensity-matched cohort study. Anesth Analg.
with mortality and acute kidney injury in critically 2013;117:412–21.
ill patients requiring volume resuscitation: a system- 124. Burdett E, Dushianthan A, Bennett-Guerrero E,
atic review and meta-analysis. JAMA. 2013;309: et al. Perioperative buffered versus non-buffered
678–88. fluid administration for surgery in adults. Cochrane
111. Myburgh JA, Finfer S, Bellomo R, et al. Hydroxyethyl Database Syst Rev. 2012;12:CD004089.
starch or saline for fluid resuscitation in intensive 125. Young P, Bailey M, Beasley R, et al. Effect of a
care. N Engl J Med. 2012;367:1901–11. buffered crystalloid solution vs saline on acute
112. Bayer O, Reinhart K, Kohl M, et al. Effects of kidney injury among patients in the intensive care
fluid resuscitation with synthetic colloids or crys- unit: the SPLIT randomized clinical trial. JAMA.
talloids alone on shock reversal, fluid balance, and 2015;314:1701–10.
patient outcomes in patients with severe sepsis: 126. Sima CS, Jarnagin WR, Fong Y, et al. Predicting the
a prospective sequential analysis. Crit Care Med. risk of perioperative transfusion for patients undergo-
2012;40:2543–51. ing elective hepatectomy. Ann Surg. 2009;250:914–21.
113. Perel P, Roberts I, Ker K. Colloids versus crystal- 127. Pulitano C, Arru M, Bellio L, Rossini S, Ferla G,
loids for fluid resuscitation in critically ill patients. Aldrighetti L. A risk score for predicting periopera-
Cochrane Database Syst Rev. 2013;2:CD000567. tive blood transfusion in liver surgery. Br J Surg.
114. Navickis RJ, Haynes GR, Wilkes MM. Effect of 2007;94:860–5.
hydroxyethyl starch on bleeding after cardiopulmo- 128. Janny S, Eurin M, Dokmak S, Toussaint A, Farges
nary bypass: a meta-analysis of randomized trials. J O, Paugam-Burtz C. Assessment of the external
Thorac Cardiovasc Surg. 2012;144:223–30. validity of a predictive score for blood transfusion in
115. Rioux JP, Lessard M, De Bortoli B, et al. Pentastarch liver surgery. HPB (Oxford). 2015;17:357–61.
10% (250 kDa/0.45) is an independent risk factor of 129. Spahn DR, Moch H, Hofmann A, Isbister JP. Patient
acute kidney injury following cardiac surgery. Crit blood management: the pragmatic solution for the
Care Med. 2009;37:1293–8. problems with blood transfusions. Anesthesiology.
116. Hand WR, Whiteley JR, Epperson TI, et al. 2008;109:951–3.
Hydroxyethyl starch and acute kidney injury in 130. Katz SC, Shia J, Liau KH, et al. Operative blood loss
orthotopic liver transplantation: a single-center ret- independently predicts recurrence and survival after
rospective review. Anesth Analg. 2015;120:619–26. resection of hepatocellular carcinoma. Ann Surg.
117. Skhirtladze K, Base EM, Lassnigg A, et al. Comparison 2009;249:617–23.
of the effects of albumin 5%, hydroxyethyl starch 131. Atzil S, Arad M, Glasner A, et al. Blood transfusion
130/0.4 6%, and Ringer’s lactate on blood loss and promotes cancer progression: a critical role for aged
coagulation after cardiac surgery. Br J Anaesth. 2014; erythrocytes. Anesthesiology. 2008;109:989–97.
112:255–64. 132. Wu CC, Ho WM, Cheng SB, et al. Perioperative
118. Martin C, Jacob M, Vicaut E, Guidet B, Van Aken H, parenteral tranexamic acid in liver tumor resec-
Kurz A. Effect of waxy maize-derived hydroxyethyl tion: a prospective randomized trial toward a “blood
starch 130/0.4 on renal function in surgical patients. transfusion”-free hepatectomy. Ann Surg. 2006;243:
Anesthesiology. 2013;118:387–94. 173–80.
366 E. Weiss et al.

133. Schmidt A, Sues HC, Siegel E, Peetz D, Bengtsson Enhanced Recovery After Surgery (ERAS((R)))
A, Gervais HW. Is cell salvage safe in liver resec- Society recommendations. World J Surg. 2013;37:
tion? A pilot study. J Clin Anesth. 2009;21:579–84. 259–84.
134. Gurusamy KS, Li J, Sharma D, Davidson 148. Geltzeiler CB, Rotramel A, Wilson C, Deng L,
BR. Pharmacological interventions to decrease blood Whiteford MH, Frankhouse J. Prospective study of
loss and blood transfusion requirements for liver resec- colorectal enhanced recovery after surgery in a com-
tion. Cochrane Database Syst Rev. 2009;4:CD008085. munity hospital. JAMA Surg. 2014;149:955–61.
135. Dalmau A, Sabate A, Acosta F, et al. Tranexamic 149. Page AJ, Ejaz A, Spolverato G, et al. Enhanced
acid reduces red cell transfusion better than epsilon-­ recovery after surgery protocols for open hepatec-
aminocaproic acid or placebo in liver transplanta- tomy--physiology, immunomodulation, and imple-
tion. Anesth Analg. 2000;91:29–34. mentation. J Gastrointest Surg. 2015;19:387–99.
136. Lodge JP, Jonas S, Oussoultzoglou E, et al. 150. Schultz NA, Larsen PN, Klarskov B, et al. Evaluation
Recombinant coagulation factor VIIa in major liver of a fast-track programme for patients undergoing
resection: a randomized, placebo-controlled, dou- liver resection. Br J Surg. 2013;100:138–43.
ble-blind clinical trial. Anesthesiology. 2005;102: 151. Hughes MJ, McNally S, Wigmore SJ. Enhanced recov-
269–75. ery following liver surgery: a systematic review and
137. de Saint MG, Pequignot F, Auroy Y, et al. Patient meta-analysis. HPB (Oxford). 2014;16:699–706.
blood management and transfusion. Anesthesiology. 152. Pessaux P, Regimbeau JM, Dondero F, Plasse
2009;111:444–5. author reply 5–6 M, Mantz J, Belghiti J. Randomized clinical trial
138. Lienhart A, Auroy Y, Pequignot F, et al. Survey evaluating the need for routine nasogastric decom-
of anesthesia-related mortality in France. pression after elective hepatic resection. Br J Surg.
Anesthesiology. 2006;105:1087–97. 2007;94:297–303.
139. Jarnagin WR, Gonen M, Maithel SK, et al. A pro- 153. Page AJ, Gani F, Crowley KT, et al. Patient outcomes
spective randomized trial of acute normovolemic and provider perceptions following implementation
hemodilution compared to standard intraoperative of a standardized perioperative care pathway for
management in patients undergoing major hepatic open liver resection. Br J Surg. 2016;103:564–71.
resection. Ann Surg. 2008;248:360–9. 154. Jones C, Kelliher L, Dickinson M, et al. Randomized
140. Tripodi A, Mannucci PM. The coagulopathy of clinical trial on enhanced recovery versus standard
chronic liver disease. N Engl J Med. 2011;365: care following open liver resection. Br J Surg.
147–56. 2013;100:1015–24.
141. Mallett SV, Sugavanam A, Krzanicki DA, et al. 155. Bonnet F, Marret E. Influence of anaesthetic and
Alterations in coagulation following major liver analgesic techniques on outcome after surgery. Br J
resection. Anaesthesia. 2016;71:657–68. Anaesth. 2005;95:52–8.
142. Levin MA, McCormick PJ, Lin HM, Hosseinian 156. Tzimas P, Prout J, Papadopoulos G, Mallett SV. Epi-
L, Fischer GW. Low intraoperative tidal volume dural anaesthesia and analgesia for liver resection.
ventilation with minimal PEEP is associated with Anaesthesia. 2013;68:628–35.
increased mortality. Br J Anaesth. 2014;113:97–108. 157. De Pietri L, Siniscalchi A, Reggiani A, et al. The use
143. Johnson M, Mannar R, Wu AV. Correlation between of intrathecal morphine for postoperative pain relief
blood loss and inferior vena caval pressure during after liver resection: a comparison with epidural
liver resection. Br J Surg. 1998;85:188–90. analgesia. Anesth Analg. 2006;102:1157–63.
144. McNally SJ, Revie EJ, Massie LJ, et al. Factors in 158. Hughes MJ, Harrison EM, Peel NJ, et al. Randomized
perioperative care that determine blood loss in liver clinical trial of perioperative nerve block and con-
surgery. HPB (Oxford). 2012;14:236–41. tinuous local anaesthetic infiltration via wound cath-
145. Lansdorp B, Hofhuizen C, van Lavieren M, et al. eter versus epidural analgesia in open liver resection
Mechanical ventilation-induced intrathoracic pres- (LIVER 2 trial). Br J Surg. 2015;102:1619–28.
sure distribution and heart-lung interactions*. Crit 159. Coelho FF, Kruger JA, Fonseca GM, et al. Laparoscopic
Care Med. 2014;42:1983–90. liver resection: experience based guidelines. World J
146. Kehlet H, Dahl JB. Anaesthesia, surgery, and chal- Gastrointest Surg. 2016;8:5–26.
lenges in postoperative recovery. Lancet. 2003;362: 160. Schiffman SC, Kim KH, Tsung A, Marsh JW, Geller
1921–8. DA. Laparoscopic versus open liver resection for
147. Gustafsson UO, Scott MJ, Schwenk W, et al. Guidelines metastatic colorectal cancer: a metaanalysis of 610
for perioperative care in elective colonic surgery: patients. Surgery. 2015;157:211–22.
Anesthetic Aspects of Living
Donor Hepatectomy 28
Paul D. Weyker and Tricia E. Brentjens

Keywords As the wait list for liver transplants far exceeds


Preoperative assessment · Ethics · Bile leak · the availability of cadaveric donors, the use and
Graft weight to recipient body weight ratio · widespread acceptance of LDLT has increased as
Anesthetic management · Surgical technique an alternative to cadaveric transplantation. However,
the need to protect the donor from unacceptable risk
is of paramount concern. In one case series, hospital
mortality from hepatic resection was 3% [4].
Introduction Fortunately, the worldwide experience for LDLT
has demonstrated a much lower mortality rate of
Led by Thomas E. Starzl, the era of liver trans- 0.4–0.6% [5] for living liver donation, yet an order
plantation began in 1963 at the University of of magnitude higher than the risk for renal donation
Colorado and by 1967 the first patient trans- [6]. It is therefore imperative that a potential liver
planted by this group survived more than a year donor is thoroughly investigated and screened to
[1]. However living donor liver transplantation optimize the safety of the procedure.
(LDLT) was only attempted many years later
after further advances of knowledge, experience,
and surgical technique in the field of split liver Preoperative Evaluation
technique allowed the transplantation of one
donor graft into two recipients [2], was first suc- In 2000, The Live Organ Donor Group published
cessfully performed in 1989 by Broelsch et al. [3] a consensus statement, providing a guideline how
at the University of Chicago. A young girl born to screen prospective liver donors [7]. Variations
with biliary atresia was the recipient of her moth- of this guideline exist from center to center as to
er’s left lobe of liver. Since that time, experience which evaluation or procedure is performed dur-
in the field has grown and expanded to include ing which phase of the screening process.
adult-to-adult living liver transplantation.

First Evaluation Phase

The first evaluation phase involves pre-screen-


P. D. Weyker, MD • T. E. Brentjens, MD (*)
Department of Anesthesiology, Columbia University
ing the prospective donor, usually performed
Medical Center, New York, NY, USA by a registered nurse to confirm that a poten-
e-mail: tb164@cumc.columbia.edu tial donor meets the following criteria [8]: The

© Springer International Publishing AG, part of Springer Nature 2018 367


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_28
368 P. D. Weyker and T. E. Brentjens

prospective donor should be of legal age and Table 28.1  Laboratory investigations during first phase
of evaluation
sufficient intellectual ability to understand
the procedure and the associated risks. There Laboratory investigations [11]
should be evidence of an emotional relation- Amylase Serology for HBV
ship between the prospective donor and recipi- Lipase HCV
HIV
ent; and potential donors who are believed or Glucose
CMV
known to have been coerced into the process Protein EBV
must be excluded. It is paramount to safeguard Protein electrophoresis HSV
the donor and ensure that their welfare super- Triglycerides
sedes all other concerns including those of the Cholesterol
recipient. The potential donor must also have TSH
C-reactive protein Protein C
the ability and willingness to comply with long-
Ferritin Protein S
term follow-up. ABO incompatible grafts are Antithrombin III
Transferring saturation
known to have a poorer long-term outcome and Factor V Leiden mutations
Alpha-1-antitrypsin Prothrombin mutations
thus, ABO compatibility is considered a pre-
Ceruloplasmin Homocysteine
requisite for donation [8]. The donor should
Antinuclear antibodies Factor VIII
be negative for hepatitis B surface antigen and Cardiolipin
Coagulation profile
hepatitis C antibody. Some centers may accept Anti-phospholipid
Urinalysis
hepatitis B core antibody positive donors. As antibodies
these donors have been exposed to hepatitis B
at some point in the past, it is prudent to per- Table 28.2  Non-invasive investigations during the sec-
form a liver biopsy if the candidate is to be ond phase of evaluation
further considered. About 18–34% of potential
Non-invasive investigations [11]
candidates are rejected in this first phase with-
Electrocardiography Doppler ultrasound of carotid
out utilizing significant resources or undergo- arteries
ing invasive testing [9, 10]. Chest roentgenogram Abdominal ultrasound
Pulmonary function Echocardiography
test
Second Evaluation Phase
Table 28.3  Tests to determine graft feasibility during the
The second phase requires a thorough medical, third phase of evaluation
laboratory (Table 28.1) and psychological evalu- Volumetric CT or MRI scan of liver
ation. The potential donor is presented to the Splanchnic arteriography
transplant team and a decision is made whether Endoscopic retrograde cholangiopancreatography
to proceed to comprehensive donor evaluation. Liver biopsy
The patient’s overall health status is assessed
and specifically the absence of diabetes, severe
or uncontrolled hypertension, and any hepatic, ensure that the donor is able to consent without
cardiac, renal, or pulmonary disease is confirmed coercion by recipient, recipient’s family, or
(Table  28.2) [11]. A thorough pre-operative transplant team.
anesthetic evaluation should be done at this time
as well.
A transplant psychologist and/or a social  hird Evaluation Phase: Graft
T
worker will conduct the psychosocial evalua- Feasibility Determination
tion. The goal is to educate the potential donor
about the psychosocial impact of donor surgery The tests listed in Table 28.3 will aid in deter-
and recovery, identify potential psychological mining graft suitability however not all of these
or psychiatric issues that preclude donation, and tests are routinely performed in all centers. It is
28  Anesthetic Aspects of Living Donor Hepatectomy 369

important to ascertain hepatic volumetric data, The committee defined essential ethical ele-
delineate hepatic anatomy including hepatic ments that need to be followed by the transplant
artery, portal vein, hepatic veins, and assess the center:
degree of steatosis [7]. The degree of steatosis The responsibility of the transplant team per-
can be assessed using imaging techniques [12]. forming live donation includes:
The percentage of steatosis is subtracted from
the estimated liver volume, thus yielding a cor- • Involvement of healthcare professionals
rected liver volume [13]. If deemed necessary, exclusively responsible to the donor.
percutaneous liver biopsy can also be per- • Repetition of the information.
formed. It is center-specific whether a candidate • Psychosocial evaluation.
with significant steatosis is accepted. • Provide a reflection period after medical
The three phases of the evaluation of the acceptance and decision to donate.
potential liver donor are listed in Table 28.4. • Assess donor retention of information and under-
standing.
• External review committees.
Ethical Considerations
Informed consent needs to include:
In 2006, The Transplantation Society issued an
ethics statement with respect to the living lung, • Cognitive capacity
liver, pancreas and intestinal (extra-renal) donor. • Voluntary decision
(Care of the live kidney donor was addressed 2 • Donor understanding
years earlier at the International Forum on the care • Disclosure, including recipient conditions
of the Live Kidney Donor held in Amsterdam.) which may impact the decision to donate with
The Transplantation Society concluded: recipient’s permission
The Ethics Committee of TTS recommends that live • Expected transplant outcomes (favorable and
lung, liver, pancreas and intestine donation should un-favorable) for the recipient
only be performed when the aggregate benefits to • Information on alternative types of treatments
the donor-recipient pair (survival, quality of life,
psychological, and social well being) outweigh the
for the recipient, including cadaveric organ
risks to the donor- recipient pair (death, medical, transplantation
psychological, and social morbidities). [14] • Donor registries

Table 28.4  Living donor evaluation criteria


Phase I Phase I Phase II Phase II Phase II Phase III
Relationship Psychosocial Medical evaluation Laboratory
Age support evaluation Graft assessment
18–60 Emotionally Adequate Comprehensive Hematologic, Volumetric MRa scan
related to psychosocial history and physical serum chemistry, excludes occult mass
recipient; support systems examination liver, and kidney lesions, documents
ABO as determined negative for acute or function normal; adequate liver volume;
compatible; by pediatric chronic illness normal EKG and graft represents at least
negative transplant team, affecting operative CXRa; negative 50% of expected
serology for psychiatry, and risk serology for recipient liver mass;
hepatitis and social services hepatitis and HIV arteriography documents
HIV viruses viruses arterial supply for
anticipated graft (for
adult LRTa only)
a
EKG electrocardiogram, CXR chest X-ray, MR magnetic resonance, LRT living related donor transplant
Reprinted with permission from Samstein B, Emond J: Liver transplants from living related donors. Annu Rev Med
2001; 52: 147–60
370 P. D. Weyker and T. E. Brentjens

Donor autonomy needs to be assured includ- have hepatic steatosis. Another concern is the
ing the freedom to withdraw from the donation presence of patent foramen ovale (PFO) in the
process at any time, with reasons for not proceed- donor, as the risk of paradoxical air-embolism
ing kept confidential. during the resection is increased [15]. It has even
Donor selection should include: been advocated that the pre-operative evaluation
should include echocardiography to rule-out PFO
• Legally incompetent or those who lack the capac- [16].
ity for autonomous decision-making should be
excluded from donation
• Rarely an independent advocate for the donor Surgical Technique
needs to be appointed
• In the event that non-directed or distant In 1957, Claude Couinaud, a French surgeon,
acquaintance live organ donation is consid- published his seminal work Le Foie: Études
ered, special considerations to prevent donor anatomiques et chirurgicales [17]. By delineat-
exploitation should be made ing the segmental anatomy of the liver (Fig. 28.1),
• Centers should regard long-term access to hepatectomy surgery became possible.
health care after the procedure as a prerequisite Four anatomic allografts are classically described
for donation. for LDLT [18, 19]. The entire right liver lobe
• The donation process and follow-up should be (Couinaud segments V–VIII) is most commonly
cost neutral for the donor. transplanted, comprising more than 60% of the
donor’s total liver mass. Normal liver volume is
1294–1502 mL in women and 1796–1956 mL in
Contraindications to Donations [5] men [20]. The entire left liver lobe (Couinaud seg-
ments II–IV) is approximately 35% of the total liver
A calculated remnant liver less than 30% of origi- volume, yielding 300–500 cc allografts that are ide-
nal liver volume with complete venous drainage ally suited for recipients weighing approximately
puts the donor at risk of too-small-for-size syn- 50 kg or less. The left lateral segment (Couinaud
drome. Pre-operative volumetric imaging may segments II–III) yields 20% of total liver volume, a
actually overestimate actual liver volume by 10%. 200–300 cc allograft, and this is used in large donor-
Similarly, an estimated graft liver volume to recip- to-­recipient size disparity and the recipient weight
ient body weight ratio (GWBWR) of <0.8%– 0.6% for a left lateral segment graft is usually restricted
is a contraindication for donation. Other contrain- to less than 40 kg. Extended right liver (Couin-
dications are: aud segments IV–VIII) hepatectomy is the least
­commonly utilized graft and provides greater than
• ABO incompatibility except in special cir- 70% of standard liver volume and is suitable for a
cumstances such as infants <1 year of age small donor-to-large recipient situation. Risks to the
without presence of isoagglutinins, and in donor by removal of such a large portion of the liver
emergencies where a cadaveric transplanta- make this technique unjustifiable in most situations
tion is not possible (Table 28.5).
• Portal or sinusoidal fibrosis Options for pediatric LDLT include entire left
• Non-alcoholic steatohepatitis (NASH) liver lobe, left lateral segment and left lateral seg-
• Steatosis >20% (only for right liver) ment with a part of segment IV [19]. To assess
• Portal inflammation and necrotic-­inflammatory graft size adequacy, a graft weight to recipient
changes body weight ratio (GWBWR) is calculated [21].
• HIV, HCV, or HBV (HBsAg+) positive Alternately, the percentage of the calculated stan-
dard liver volume (SLV) can be used [22, 23].
A BMI > 30 kg/m2 is a relative contraindica- The graft size is considered adequate if the
tion to donation as these candidates commonly GWBWR is within 1–3% [19]. A ratio of 0.8% is
28  Anesthetic Aspects of Living Donor Hepatectomy 371

Fig. 28.1 Couinaud Right Right


segmental anatomy of Posterior Anterior Left Medial Left Lateral
the liver With Section Section Section Section
permission from: (VI and VII) (V and VIII) (IV) (II and II)
Modern Pathology
(2014) 27, 472–491, Right Middle
Towards an international Hepatic Vein Hepatic Vein
pediatric liver tumor
consensus classification:
VIII II
proceedings of the Los
Angeles COG liver IV
tumors symposium.

VII
I
III

V
Umbilical Fissue/
Ligamentum Teres
VI

For pediatric living donor liver transplanta-


Table 28.5  Definition of the four donor anatomic allografts
tion, laparoscopic left lateral segmentectomy to
Couinaud Percentage liver Volume resect segments II and III and removal through
Allograft segments removed (%) yield (cc) a Pfannenstiel incision has been reported [25].
Entire V–VIII 60 600–900 Laparoscopic right hepatectomy has also been
right lobe
described for adult living donor transplantation,
Entire left II–IV 35 300–500
lobe and is now routinely employed at our center
Left lateral II–III 20 200–300 [26]. Laparoscopic assisted hepatectomy for
segment donation will consist of laparoscopic mobiliza-
Extended IV–VIII 70 800–1000 tion of the right lobe followed by resection
right liver through a midline incision, thereby avoiding
the subcostal incision. However in most cases,
considered the minimum to prevent small-for-­ a right or bilateral subcostal incision with mid-
size syndrome in the recipient, however, experi- line extension is performed for live liver organ
ence at our center has shown successful grafting donation.
with graft ratios of as low as 0.49%. Recipients
with severe portal hypertension or decompen-
sated disease will require a larger graft, irrespec- Anesthetic Management
tive of calculated GWBWR. In general, left lobe
will be used for recipient with a body weight Due to the potential for large volume blood loss
20–40 kg, and left lateral segment or left lateral during the hepatectomy, central venous catheteriza-
segment plus portion of segment IV for recipients tion should be considered if there is any doubt about
with a body weight < 40 kg [19]. In instances large volume intravenous access to allow for rapid
where a graft larger than left lobe is necessary, volume replacement and monitoring of central
left half of caudate lobe can be added. [24]. venous pressure (CVP). A low CVP (2–4 mmHg)
372 P. D. Weyker and T. E. Brentjens

is desirable in order to minimize blood loss [4]. postoperative day 1 and discharged from hospital
Pringle’s maneuver, the surgical technique of (inter- postoperative around days 5–10.
mittently) occluding inflow is routinely used to min- As living donors are generally healthy without
imize blood loss in hepatectomy surgery, however, any chronic disease, post-operative pain is often
the risk of ischemic injury to the graft has in the greater than in patients who underwent hepatic
past precluded its use in living donor hepatectomy. resection for example for cancer [32, 33]. Pre-­
Recent evidence shows that this procedure can be operative epidural catheter placement may be an
safe for the graft while providing a cleaner surgical good option for post-operative analgesia [34]
field and a lower incidence of biliary complications with the additional benefits of a shorter duration
[27, 28]. Techniques utilizing Trendelenburg posi- of post-operative ileus, attenuated stress response,
tion, volume restriction, nitroglycerine infusion, fewer pulmonary complications and early ambu-
and furosemide administration have all been used lation [35]. However some centers avoid epidural
to reduce CVP and potentially bleeding [29]. In analgesia as significant post-operative derange-
addition to reducing CVP, Trendelenburg position ments of the coagulation profile can occur within
of 15° is advocated to reduce the risk of venous air the first postoperative days and these may com-
embolism. plicate the removal of the epidural catheter at a
The anesthesiologist must also be cognizant time when the patient is getting ready for dis-
to minimize possible insult to the resected graft charge home [36].
[30]. Firstly, hepatotoxic drugs should be avoided. In addition to the risk of postoperative coag-
Approximately 20% of halothane is metabolized ulopathy due to lower hepatic volume, heparin
by the liver and the risk of autoimmune hepatitis administration to prevent graft thrombosis at the
is greater than that of any other available inhaled end of liver parenchymal dissection may further
anesthetic. Secondly, perfusion to the liver should prevent anesthesiologists from placing an epi-
be optimized. Hepatic blood flow is decreased by dural catheter [36, 37]. It is recommended that
the nitrous oxide, an elevated CVP, and as a con- heparin administration be delayed 1 h after cath-
sequence of reflex vasoconstriction of the hepatic eter placement, and catheter removal delayed
arterial and portal venous system in response to 2–4 h after the last dose of heparin and not until
elevated pressures in the hepatic sinusoids. Lastly, the aPTT is checked [38] and fortunately, the
graft edema must be minimized to reduce the risk average time of heparin administration from
of graft thrombosis and the administration of man- epidural catheter placement is usually greater
nitol to the living donor may aid in reducing graft than four hours [36].
edema [30]. Ultimately, LDLT has the advantage Patient-controlled analgesia (PCA) is another
of minimizing cold ischemic time to 1 h or less mode of analgesia commonly used in many cen-
as compared to the 4 up to 12 h of cold ischemic ters after donor hepatectomy [30, 37]. Our center
time with deceased donor transplantation. As a uses intrathecal morphine (ITM) placed prior to
consequence inflammatory markers after reperfu- surgery in combination with post-operative PCA,
sion are lower in LDLT and may improve graft a regimen that is superior to PCA use alone [39].
survival [31]. A mild self-limiting pruritus is the most common
adverse effect of ITM [39]. Pre-operative ITM is
not inferior to epidural catheter use as determined
Post-operative Management by the Visual Analog Scale, but intravenous opi-
oid use and incidence of pruritus is greater [40].
At the conclusion of the operation, muscle relax- Regional techniques such as transverse abdomi-
ation is adequately reversed, and the majority of nal plane (TAP) block either as single injection or
patients can be safely extubated in the operating using continuous catheters are other good options
room. At our institution, intensive care admission for postoperative pain while avoiding the limita-
is routine and with an uneventful recovery tions with mobilization and removal when using
patients can be transferred to the surgical floor on epidural catheters.
28  Anesthetic Aspects of Living Donor Hepatectomy 373

Complications An early report of 100 consecutive hepatic


resections reported that 59 of these patients
The altruistic nature of living donor hepatec- received exogenous blood products [4]. A com-
tomy for transplantation necessitates that all mon strategy to minimize exogenous blood
precautions to protect the donor must be taken. product administration is the use of intraopera-
Deep vein thrombosis (DVT) leading to pul- tive blood salvage techniques, for example
monary embolism is a potentially catastrophic using Cell-Saver™ (Haemonetics Laboratories,
post-­operative complication that can result in Boston, MA) and re-transfusion of the red
donor morbidity and/or mortality [41]. The use blood cells at the conclusion of the hepatec-
of graduated compression stockings and inter- tomy [16]. Pre-operative autologous blood
mittent pneumatic compression intra- and post-­ donation, erythropoietin administration, and
operatively has been well validated in reducing isovolumetric hemodilution are other possible
the incidence of DVT [42]. Additionally, the strategies that have been employed.
prophylactic administration of subcutaneous Postoperative recovery and regeneration of the
heparin can reduce the risk of DVT by 50–70% remnant liver begins immediately after resection.
[43]. Transaminase enzymes peak within 48 h and bili-
Blood loss depends on the type of hepatec- rubin usually peaks approximately on day 3 [16].
tomy performed. A right hepatectomy is a more Small-for-size syndrome, usually described as a
lengthy and challenging procedure and associ- transplanted graft that is inadequate in size and
ated with a significantly longer hospital stay, function can occur in the donor if the remaining
anesthesia time, larger blood loss, and greater liver volume is too low. A too small liver remnant
derangement of the coagulation profile occurs as can present with prolonged cholestasis, transami-
compared to left hepatectomy (LH) or left lateral nitis, and synthetic function derangements [16].
hepatectomy (LL) [6] (Table 28.6). The care for small-for-size syndrome is mainly
supportive, however various strategies have been
proposed, including octreotide therapy to reduce
Table 28.6  Clinical and biological outcome of living portal pressure, and intraportal glucose and insu-
liver donation
lin infusions to hasten remnant liver regeneration
RH LH LL [16]. One case of liver failure in a living donor
Mean ± SD Mean ± SD Mean ± SD
requiring liver transplantation has been reported
Clinical
[44].
Hospital— 7 ± 2.5 5.9 ± 1.3 6.66 ± 1.5
Stay (days) Biliary leaks are the most common complica-
Anesthesia 528 ± 108 453 ± 73 340 ± 39 tion after donor hepatectomy [44]. One case
time (min) series reported biliary leaks in 13% of donors.
Estimated 583 ± 277 400 ± 175 294 ± 145 Twenty percent of these cases resolved with
blood (mL) external drainage using the Jackson-Pratt drain
Biological placed during the initial surgery, half required
INR peak 1.75 ± 0.3 1.37 ± 0.2 1.27 ± 0.2 additional percutaneous drainage and 30%
TBili peak 3.05 ± 1.4 2.6 ± 1 1.5 ± 1.3 required endoscopic drainage. The source of the
(mg/dL)
leak is commonly the cut surface, but may also
AST peak 348 ± 260 239 ± 225 289 ± 226
(IU/L) be at the stump of the right hepatic duct [45]. A
RH right hepatectomy, LH left hepatectomy, LL left lateral
lower rate of biliary leaks (5–10%) was observed
hepatectomy, INR Internation Normalzie ration, TBili with left lateral segmentectomy [19]. Biliary
total bilirubin, AST aspartate aminotransferase strictures occur less often; the same case series
Reprinted and adapted with permission from Salame E, reported this complication in 1.5% of all donors
Goldstein MJ, Kinkhabwala M, Kapur S, Finn R, Lobritto
S, Brown R, Jr., Emond JC: Analysis of donor risk in
[45]. Biliary strictures will more frequently
living-­donor hepatectomy: the impact of resection type on require invasive interventions for example using
clinical outcome. Am J Transplant 2002; 2: 780–8 temporary endoscopic retrograde biliary stenting
374 P. D. Weyker and T. E. Brentjens

and one donor required hepaticojejunostomy 13. Marcos A, Fisher RA, Ham JM, et al. Liver regen-
eration and function in donor and recipient after right
20 months after surgery.
lobe adult to adult living donor liver transplantation.
The most common reason for reoperation in Transplantation. 2000;69:1375–9.
the living donor is to repair an incisional hernia 14. Pruett TL, Tibell A, Alabdulkareem A, et al. The eth-
[46]. Herniae are more frequent in the obese pop- ics statement of the Vancouver Forum on the live lung,
liver, pancreas, and intestine donor. Transplantation.
ulation (BMI > 30) [47], however obesity is only
2006;81:1386–7.
a relative contraindication and does not necessar- 15. Hatano Y, Murakawa M, Segawa H, Nishida Y, Mori
ily preclude donation. A bilateral incision with K. Venous air embolism during hepatic resection.
midline extension has a higher risk of incisional Anesthesiology. 1990;73:1282–5.
16. Gruessner R, editor. Living donor organ transplanta-
hernia compared to a right subcostal incision
tion. 1st ed. New York: McGraw-Hill; 2008.
with midline extension [19]. 17. Couinaud C. Le Foie: Etudes Anatomique et Chirurgi-
cales. Paris: Masson; 1957.
18. Everson GT, Trotter JF, editors. Liver transplanta-
References tion: challenging controversies and topics. Totowa:
Humana Press; 2009.
1. Starzl TE, Fung JJ. Themes of liver transplantation. 19. Busuttil R, Goran KK, editors. Transplantation of the
Hepatology. 2010;51:1869–84. Liver. 2nd ed. Philadelphia: Elsevier Saunders; 2005.
2. Otte JB, de Ville de Goyet J, Alberti D, Balladur P, 20. Andersen V, Sonne J, Sletting S, Prip A. The volume
de Hemptinne B. The concept and technique of the of the liver in patients correlates to body weight and
split liver in clinical transplantation. Surgery. 1990; alcohol consumption. Alcohol Alcohol. 2000;35:
107:605–12. 531–2.
3. Broelsch CE, Whitington PF, Emond JC, et al. 21. Kiuchi T, Kasahara M, Uryuhara K, et al. Impact of
Liver transplantation in children from living related graft size mismatching on graft prognosis in liver
donors. Surgical techniques and results. Ann Surg. transplantation from living donors. Transplantation.
1991;214:428–37. discussion 37–9 1999;67:321–7.
4. Cunningham JD, Fong Y, Shriver C, Melendez J, 22. Kawasaki S, Makuuchi M, Matsunami H, et al. Pre-
Marx WL, Blumgart LH. One hundred consecutive operative measurement of segmental liver volume of
hepatic resections. Blood loss, transfusion, and opera- donors for living related liver transplantation. Hepa-
tive technique. Arch Surg. 1994;129:1050–6. tology. 1993;18:1115–20.
5. Barr ML, Belghiti J, Villamil FG, et al. A report of the 23. Heinemann A, Wischhusen F, Puschel K, Rogiers

Vancouver Forum on the care of the live organ donor: X. Standard liver volume in the Caucasian population.
lung, liver, pancreas, and intestine data and medical Liver Transpl Surg. 1999;5:366–8.
guidelines. Transplantation. 2006;81:1373–85. 24. Miyagawa S, Hashikura Y, Miwa S, et al. Concomitant
6. Salame E, Goldstein MJ, Kinkhabwala M, et al. caudate lobe resection as an option for donor hepa-
Analysis of donor risk in living-donor hepatectomy: tectomy in adult living related liver transplantation.
the impact of resection type on clinical outcome. Am Transplantation. 1998;66:661–3.
J Transplant. 2002;2(8):780. 25. Cherqui D, Soubrane O, Husson E, et al. Laparoscopic
7. Abecassis M, Adams M, Adams P, et al. Consensus living donor hepatectomy for liver transplantation in
statement on the live organ donor. JAMA. 2000;284: children. Lancet. 2002;359:392–6.
2919–26. 26. Suh KS, Yi NJ, Kim J, et al. Laparoscopic hepatec-
8. Samstein B, Emond J. Liver transplants from living tomy for a modified right graft in adult-to-adult living
related donors. Annu Rev Med. 2001;52:147–60. donor liver transplantation. Transplant Proc. 2008;40:
9. Renz JF, Mudge CL, Heyman MB, et al. Donor selec- 3529–31.
tion limits use of living-related liver transplantation. 27. Imamura H, Kokudo N, Sugawara Y, et al. Pringle’s
Hepatology. 1995;22:1122–6. maneuver and selective inflow occlusion in living
10. Baker A, Dhawan A, Devlin J, et al. Assessment of donor liver hepatectomy. Liver Transpl. 2004;10:
potential donors for living related liver transplanta- 771–8.
tion. Br J Surg. 1999;86:200–5. 28. Taketomi A, Morita K, Toshima T, et al. Living donor
11. Sauer P, Schemmer P, Uhl W, Encke J. Living-­donor hepatectomies with procedures to prevent biliary
liver transplantation: evaluation of donor and recipi- complications. J Am Coll Surg. 2010;211:456–64.
ent. Nephrol Dial Transplant. 2004;19(Suppl 4): 29. Wang WD, Liang LJ, Huang XQ, Yin XY. Low cen-
iv11–5. tral venous pressure reduces blood loss in hepatec-
12. Kamel IR, Kruskal JB, Raptopoulos V. Imaging for tomy. World J Gastroenterol. 2006;12:935–9.
right lobe living donor liver transplantation. Semin 30. Beebe DS, Carr R, Komanduri V, Humar A, Gruessner
Liver Dis. 2001;21:271–82. R, Belani KG. Living liver donor surgery: report of
28  Anesthetic Aspects of Living Donor Hepatectomy 375

initial anesthesia experience. J Clin Anesth. 2000;12: gesia is an effective and safe method for immediate
157–61. postoperative pain control in live liver donors. Liver
31. Jassem W, Koo DD, Cerundolo L, Rela M, Heaton Transpl. 2009;15:381–9.
ND, Fuggle SV. Cadaveric versus living-donor livers: 40. De Pietri L, Siniscalchi A, Reggiani A, et al. The use
differences in inflammatory markers after transplanta- of intrathecal morphine for postoperative pain relief
tion. Transplantation. 2003;76:1599–603. after liver resection: a comparison with epidural anal-
32. Siniscalchi A, Begliomini B, De Pietri L, et al.
gesia. Anesth Analg. 2006;102:1157–63.
Increased prothrombin time and platelet counts in 41. Wakeno-Takahashi M, Nakao S, Ikeda S, et al. A case
living donor right hepatectomy: implications for epi- of suspected severe pulmonary embolism in a living-­
dural anesthesia. Liver Transpl. 2004;10(9):1144. related liver transplantation donor. J Clin Anesth. 2005;
33. Cywinski JB, Parker BM, Xu M, Irefin SA. A com- 17:218–20.
parison of postoperative pain control in patients after 42. Nicolaides AN, Breddin HK, Fareed J, et al. Preven-
right lobe donor hepatectomy and major hepatic tion of venous thromboembolism. International Con-
resection for tumor. Anesth Analg. 2004;99:1747–52; sensus Statement. Guidelines compiled in accordance
table of contents. with the scientific evidence. Int Angiol. 2001;20:
34. Choudhry DK, Schwartz RE, Stayer SA, Shevchenko 1–37.
Y, Rehman M. Anesthetic management of living liver 43. Collins R, Scrimgeour A, Yusuf S, Peto R. Reduction
donors. Can J Anaesth. 1999;46:788–91. in fatal pulmonary embolism and venous thrombo-
35. Fotiadis RJ, Badvie S, Weston MD, Allen-Mersh
sis by perioperative administration of subcutaneous
TG. Epidural analgesia in gastrointestinal surgery. Br heparin. Overview of results of randomized trials in
J Surg. 2004;91:828–41. general, orthopedic, and urologic surgery. N Engl J
36. Choi SJ, Gwak MS, Ko JS, et al. The changes in coag- Med. 1988;318:1162–73.
ulation profile and epidural catheter safety for living 44. Brown RS Jr, Russo MW, Lai M, et al. A survey of
liver donors: a report on 6 years of our experience. liver transplantation from living adult donors in the
Liver Transpl. 2007;13:62–70. United States. N Engl J Med. 2003;348:818–25.
37. Cammu G, Troisi R, Cuomo O, de Hemptinne B, Di 45. Ito T, Kiuchi T, Egawa H, et al. Surgery-related mor-
Florio E, Mortier E. Anaesthetic management and bidity in living donors of right-lobe liver graft: les-
outcome in right-lobe living liver-donor surgery. Eur J sons from the first 200 cases. Transplantation. 2003;
Anaesthesiol. 2002;19:93–8. 76:158–63.
38. Horlocker TT, Wedel DJ, Rowlingson JC, et al.
46. Rudow DL, Brown RS Jr, Emond JC, Marratta D,
Regional anesthesia in the patient receiving antithrom- Bellemare S, Kinkhabwala M. One-year morbid-
botic or thrombolytic therapy: American Society of ity after donor right hepatectomy. Liver Transpl.
Regional Anesthesia and Pain Medicine Evidence-­ 2004;10:1428–31.
Based Guidelines (Third Edition). Reg Anesth Pain 47. Moss J, Lapointe-Rudow D, Renz JF, et al. Select uti-
Med. 2010;35:64–101. lization of obese donors in living donor liver trans-
39. Ko JS, Choi SJ, Gwak MS, et al. Intrathecal morphine plantation: implications for the donor pool. Am J
combined with intravenous patient-controlled anal- Transplant. 2005;5:2974–81.
Complications of Liver Surgery
29
Oliver P. F. Panzer

Keywords Assessment of the surgical risk of patients


Hemorrhage · Antifibrinolytic · Liver failure · undergoing elective liver surgery must include the
Small-for-size syndrome · ALPPS procedure · preoperative state of the patient, the degree of cir-
Minimal invasive resection rhosis, steatosis, fibrosis (Child-Pugh classifi-
cation or the MELD score), the extent of liver
dysfunction and associated co-­morbidities such as
coagulopathies, renal insufficiency, portopulmo-
Introduction nary hypertension among others, and the extent of
liver resection with remaining hepatic function.
Over the years improvements in the understand- All of the above factors affect periperative out-
ing of the functional anatomy of the liver, patient come and need to be considered carefully prior to
selection and surgical technique have greatly surgery. (See Table 29.1: MELD score.) It is well
reduced the rate of complications of hepatic sur- known that the extent of intraoperative blood loss
gery. Consequently, the number of liver resec- with subsequent transfusions is related to increased
tions has greatly increased over the last decade. morbidity and mortality [1–6]. Patient selection
Nonetheless, intraoperative complications con- and preoperative work-up is discussed in more
tinue to exist. This chapter will discuss the most detail elsewhere in this book.
common complications associated with non-
transplant liver surgery and evaluate the poten-
tial treatment options available. We will focus
on methods to reduce intraoperative blood loss, Bleeding and Excessive Blood Loss
ischemia reperfusion injury after hepatic resec-
tions and lastly, discuss the issue of low residual Next to the amount of liver segments resected dur-
hepatic mass as a consequence of excessive liver ing surgery, perioperative blood loss is one of the
resection. most important predictors for perioperative mor-
bidity and mortality [1–6]. In 1977 Foster et al.
reported a mortality of over 20% in major hepatic
surgery, major hemorrhage causing 20% of the
deaths alone [7]. In a recent analysis of more
O. P. F. Panzer, MD
than 1800 patients undergoing liver resections the
Department of Anesthesiology, Columbia University
Medical Center, New York, NY, USA perioperative mortality was only 5%, with almost
e-mail: op2015@cumc.columbia.edu 0% in the last 200 cases. The authors ascribed the

© Springer International Publishing AG, part of Springer Nature 2018 377


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_29
378 O. P. F. Panzer

improvement in perioperative mortality to sub- Surgical Techniques


stantially improved parenchyma-sparing surgical
techniques and a decrease in estimated blood loss. Improvements in surgical techniques have greatly
The resection of more than three segments or the reduced the extent of intraoperative blood loss. In
performance of complex hepatectomies, however order to control bleeding during transection of the
still caused increased blood loss and transfusion liver the surgeon must manipulate vascular inflow
requirements [8]. Other studies reported the per- and hepatic venous backflow. The Pringle maneu-
formance of en bloc resection, a surgeon with ver aims to reduce vascular inflow by clamping
low case volume, tumor size, tumor proximity to the portal vein and the hepatic artery (Fig. 29.1:
major hepatic vessels [9] and operative time [10] Pringle Maneuver). Total vascular occlusion
to be independent risk factors for perioperative involves the Pringle maneuver and additional
blood loss during hepatectomies [11]. Poor liver clamping of the infrahepatic and suprahepatic
function especially with impaired hemostasis has vena cava [15]. Both techniques have allowed for
long been thought to increase intraoperative blood decreased intraoperative blood loss at the cost of
loss. This has recently been challenged by labo- ischemic liver injury and impairment of liver
ratory studies in patients with cirrhosis [4, 12] regeneration [16]. The quality of the liver tissue
and reports of patients with cirrhosis undergoing and the surgical dissection method utilized will
major hepatic surgery without the need for trans- also affect parenchymal bleeding [17]. Better
fusion of blood products [4]. Evidence shows that understanding of the liver anatomy and major
the deficiencies in procoagulant factors are in advances in hepatic imaging led to the develop-
part compensated by a simultaneous downregu- ment of parenchymal sparing surgical techniques
lation of anti coagulant function [13, 14] main- for liver resection. Advantages of a segmentally
taining hemostasis in patients with liver disease oriented resection particularly in patients with
(Table 29.1). However this rebalanced hemostatic preexisting liver disease result in better conserva-
equilibrium is easily challenged by triggers like tion of functional liver parenchyma and reduced
sepsis or intraoperative hemorrhage.
Table 29.1  Hemostatic changes in patients with liver dis-
ease that either contribute (A) or counteract (B) bleeding Main
scissura
(A) Changes that impair hemostasis
Right scissura

Left scissura

Thrombocytopenia
Reduced hematocrit I

Platelet function defects


Enhanced production of nitric oxide and prostacyclin VIII
VII IV III II
Low levels of coagulation factors II, V, VII, IX, X,
and XI VI V

Vitamin K deficiency
Dysfibrinogenemia
Low levels of plasmin inhibitor, factor XIII, and TAFI
Elevated levels of tPA
(B) Changes that promote hemostasis
Elevated levels of VWF Right Liver Left
Elevated levels of factor VIII
Fig. 29.1  Segmental organization of the liver. The
Decreased levels of protein C, protein S, and
hepatic veins reside in the portal scissurae, which divide
antithrombin
the liver into four sectors. Each sector is composed of one
Low levels of plasminogen or more anatomic segments. The right hemi-liver consists
From: Lisman and Leebeek. Hemostatic alterations in liver of segments V, VI, VII, and VIII. The left hemi-liver con-
disease: a review on pathophysiology, clinical consequences, sists of segments II, III, and IV. (From: Blumgart LH, ed.
and treatment. Dig Surg (2007) vol. 24 (4) pp. 250–8, Surgery of the Liver and Biliary Tract. 2nd ed. London:
Table 1, with permission Churchill-Livingstone; 1994, with permission)
29  Complications of Liver Surgery 379

Fluid Management and Transfusion


VII
VIII The anesthesiologist’s role in reducing intraop-
II erative blood loss and complications cannot be
underestimated and impacts intraoperative fluid
V management, transfusion requirements, and phar-
III macological interventions. Fluid management
IV
strategies during liver surgery have long been
VI debated with the focus on central venous pressure
(CVP) monitoring. While some form of hepatic
A
inflow occlusion is routinely used during liver
surgery, intraoperative bleeding is mostly caused
Fig. 29.2  Exploded views of the liver demonstrating the
liver segments according to Couinaud’s nomenclature, as by backflow from valveless hepatic veins. Thus
seen in the patient. (From: Blumgart LH, ed. Surgery of reducing CVP below 5 mmHg has been advocated
the Liver and Biliary Tract. 2nd ed. London: Churchill-­ and reported to improve the surgical field during
Livingstone; 1994, with permission)
dissection and reduce transfusion requirements
in several studies for liver resection [19–25] or
hemorrhagic complications compared to the clas- transplantation [26, 27]. Methods to achieve a low
sic lobar or wedge resection respectively. The CVP consist of volume contraction by restrictive
anatomical basis for the segmental resection is use of volume replacement, the use of vasodilating
built on the fact that the three hepatic veins divide agents, the use of diuretics or even perioperative
the liver up into four sectors, where each sector is phlebotomy. Opponents of a low intraoperative
fed by a distinct portal vein pedicle (Figs. 29.1 CVP state the higher risk for complications includ-
and 29.2). A similar distribution of the hepatic ing systemic tissue hypoperfusion and postopera-
arteries and bile duct system is true. Identification tive renal failure [19]. It is important to note that
and clamping of one portal pedicle leads to the there are no definitive studies showing evidence
demarcation of the corresponding liver segments for improvement of postoperative outcome in
and allows a resection without damaging the vas- terms of survival or long-term morbidity using
cular supply of the neighboring tissue. a reduced CVP. Furthermore, it has been argued
New dissection devices for the transection of that pulmonary artery occlusion pressure and CVP
the liver parenchyma such as ultrasonic dissec- fail to predict ventricular filling volume, cardiac
tion, hydro-jet dissection, and radiofrequency performance or the response to volume infusion in
ablation-based devices have been introduced normal subjects [28].Other intraoperative methods
with varying degrees of success [17]. Although for monitoring preload (transesophageal echocar-
most of these devices have shown an advantage diography) remain to be tested and are covered in
in decreasing blood loss during transection, some more detail elsewhere in this book.
of them perform slowly and the overall benefits In the setting of major liver resection or trans-
cannot be clearly elucidated at this time [18]. plant, transfusion of Packed Red Blood Cells
Personal preference and experience and the (PRBC), Fresh Frozen Plasma (FFP), Platelets
availability of each device in operating centers (Plts) and Cryoprecipitate may be necessary. The
are the main factors that determine the use of a triggers and goals for blood product transfusion
specific device. Additional prospective studies should be discussed by the anesthesiology and
are required to evaluate the superiority of each the surgical teams individually for each patient
method or device. Vascular control is needed to prior to surgery. This topic is highly controversial
limit the extent of blood loss, but the method of as transfusion does not come without increased
control should be selected based on the location intraoperative and postoperative risks. (Please
and complexity of the resection and the skill level see elsewhere in this book for a more detailed
of the surgical team. discussion on intraoperative transfusion.)
380 O. P. F. Panzer

Pharmacologic Agents ence in blood loss or transfusion requirements


between patients who received Factor VIIa or
Pharmacologic agents, such as topical hemostatic placebo [17]. Tissue Factor VIIa is used most
agents, antifibrinolytic drugs, and procoagulant often in “last resort” situations of massive hemor-
drugs, are available as complementary measures rhage, and must be tested more extensively as a
to reduce intraoperative blood loss. Topical prophylactic drug in liver resections and trans-
hemostatic agents work by mimicking coagula- plant more specifically, before it can be recom-
tion at the transected surface of the parenchyma, mended as a routine pharmacological agent to
for example, fibrin, or by creating a matrix for limit blood loss.
endogenous coagulation, like collagen, gelatin or
cellulose sponges [17, 29]. Fibrin sealants in liver
surgery have been tested in a large, randomized, Perioperative Hepatic Insufficiency
controlled trial in 300 patients undergoing partial
liver resection [30]. The results showed no differ- The incidence of perioperative hepatic insuffi-
ence in total blood loss, transfusion requirement ciency is about 3% in patients undergoing resec-
or postoperative morbidity between the treatment tion for cancer or metastases. Many of these
group (fibrin sealants) and a control group (no patients have evidence of impaired liver function
fibrin sealants) [30]. Future prospective studies preoperatively, reflecting a higher risk of periop-
are required to further test the efficacy of the vari- erative hepatic failure and death [39]. In fact the
ous topical agents in reducing intraoperative preoperative Model for Endstage Liver Disease
blood loss. (MELD) score correlates well with the incidence
Antifibrinolytic drugs are either inhibitors of of post-liver- resection hepatic failure and has
plasminogen, for example tranexamic acid and been suggested to help select patients appropri-
aminocaproic acid, or inhibitors of plasmin, like ately for hepatectomies. In one study patients
aprotinin. Aprotinin is not available anymore in undergoing hepatectomy with a MELD
Europe and the US due to the association between score > 10 developed liver failure in 37.5% of the
aprotinin and serious end-organ damage in car- cases, whereas none did with a MELD score < 9
diac surgery [31] (but not in liver transplant sur- [40]. Ischemia reperfusion injury, small-for-size
gery [32, 33]). Both aprotinin and tranexamic syndrome and major blood loss are the most
acid reduce blood loss and transfusion require- important etiologies involved in postoperative
ments by approximately 30–40% [32]. The use of liver failure after hepatectomy [41].
antifibrinolytics in liver resections has not been
extensively studied and warrants future prospec-
tive analysis. It is important to note that these Ischemia Reperfusion Injury
drugs are used as adjuncts to compliment surgical
technique and proper anesthetic care during liver Ischemia reperfusion (IR) injury can lead to liver
surgery, and cannot be utilized as solitary hemo- failure resulting in coagulopathy, renal failure,
static methods. severe metabolic acidosis, cerebral edema and
Factor VIIa has been studied as a procoagu- hypothermia and finally a higher mortality. The
lant drug in several randomized clinical trials in pathophysiology of IR injury during surgical
patients undergoing liver resection or transplant clamping and liver resection is complex and occurs
[17, 34–38]. Recombinant Factor VIIa generates in two stages. In the initial phase (within approxi-
thrombin through Tissue Factor (TF) or by bind- mately 2 h after reperfusion) reactive oxygen spe-
ing to the platelet surface directly. In all of the cies (ROS) are released and activated Kupffer cells
clinical trials assessing the efficacy and safety of (liver macrophages) may release proinflammatory
Factor VIIa in liver surgery and transplant, there mediators such as tumor necrosis factor-α (TNFα)
were no major safety issues. However, these tri- and nitric oxide (NO), triggering a systemic inflam-
als also failed to demonstrate a significant differ- matory response. The oxidative stress ultimately
29  Complications of Liver Surgery 381

Vascular occlusion techniques

PTC HHVC THVE SHVE

I/R injury
Antioxidants
Ischaemic preconditioning Anti-inflammatory therapy
Remote preconditioning Vasodilatants
Intermittent clamping Apoptosis inhibitors
Pharmacological preconditioning

Surgical strategies Pharmacological strategies

Fig. 29.3 Synopsis of surgical and pharmacological (THVE) or selective hepatic vascular exclusion (SHVE).
strategies for hepatic ischemia–reperfusion (IR) injury (From Bahde and Spiegel. Hepatic ischaemia-reperfusion
induced by portal triad clamping (PTC), hemihepatic vas- injury from bench to bedside. Br J Surg (2010) vol. 97
cular clamping (HHVC), total hepatic vascular exclusion (10) pp. 1461–75, with permission)

may cause necrosis or apoptosis of hepatocytes as aggravate the preexisting supply-demand mismatch
well as endothelial cells [42]. During the late phase and amplify hepatocyte destruction (Fig. 29.3).
(6–48 h after reperfusion) infiltration by activated
neutrophils dominates the process. The inflamma-
tory response is maintained and impairment of the Treatment
microcirculation via the release of Endothelin 1
(vasoconstriction) and platelet aggregation leads to There are currently no proven treatment modali-
further hepatocyte damage [42]. Steatotic as well ties for early reperfusion injury resulting in
as cirrhotic livers are more susceptible to IR injury improved clinical outcome; in animal studies
than a healthy hepatic parenchyma. In fatty liver interventions like ischemic preconditioning
disease mitochondrial changes paired with altered (IPC) and intermittent vascular clamping (IC) or
receptor expression leads to decreased intracellu- pharmacological strategies have shown promis-
lar ATP levels. The altered anatomy and increased ing results for liver resections, however the clini-
concentrations of endothelin-­ 1 (vasoconstrictor) cal translation has been disappointing so far.
in cirrhotic patients may cause a marginal blood During IPC the vascular supply to the liver is
supply, limiting the tolerance of hypoperfusion. interrupted intermittently only for short periods
Subsequent ischemia during surgery will thus of time provoking a conditioning response in
382 O. P. F. Panzer

order to tolerate prolonged periods of ischemia nitric oxide donor), and sirolimus (an immuno-
better. On a molecular basis Adenosine, NO and suppressive agent) have shown hepatoprotective
induction of cytoprotective genes seem to play a properties and demonstrated improved survival
key role [42, 43], however the exact mechanisms in rat models [54–56]. Although these pharmaco-
are not well understood. The potential beneficial logical agents are promising, their effectiveness
effects of IC are probably based on the same has only been successful in animal models, and
mechanisms as IPC. IC describes a vascular warrants future clinical trials in human models.
occlusion during liver surgery with intermittent Additional research highlights the early use of
release intervals. Various clamp-release-time beta-adrenergic drugs such as dobutamine, that
regimens have been published, resulting in atten- may augment hepatic oxygen supply and uptake
uated liver injury but no difference in clinical in cirrhotic livers. Volatile anesthetics, which are
outcomes compared to continuous vascular routinely utilized in the operating room, have
occlusion [44, 45]. Based on current Cochrane shown additional benefits by induction of anti-­
meta-analyses by Gurusamy et al., it seems that inflammatory, anti-apoptotic and anti-oxidative
at present IPC and IC reduce hepatic injury evi- properties. In a small study in patients undergo-
denced by laboratory tests and reduce transfu- ing liver resection with continuous inflow occlu-
sion requirements but so far failed to show any sion, preconditioning with sevoflorane not only
clear clinical benefit [46–48]. reduced hepatic injury parameters but also lead to
In experimental studies numerous pharma- fever complications in the perioperative period.
cological agents have shown some promise in Patients with steatotic livers seemed to benefit
decreasing liver damage caused by the occluded even more [57]. However some authors caution
blood supply [49]. Amrinone, prostaglandin E1, using volatile anesthetics, holding them respon-
pentoxifylline, dopexamine, dopamine, ulina- sible for hepatic injuries ranging from transa-
statin, gantaile, sevoflurane, and propofol have all minitis to fulminant hepatic failure [58]. Larger
been evaluated. Ulinastatin (a neutrophil-­elastase randomized controlled trials are needed before
inhibitor) experimentally used for the treatment any of these pharmacological agents can be rec-
of septic shock, circulatory shock and adult respi- ommended for routine clinical practice [45]. For
ratory distress syndrome [50] had significantly now, adequate perfusion with maintenance of
lower postoperative enzyme markers of liver adequate portal vein to CVP gradient to ensure
injury; however, there was no significant differ- flow remains paramount during the reperfusion
ence in mortality, liver failure, or postoperative phase.
complications [49]. Potential pharmacological
hepatoprotective drugs, such as pentoxifylline,
have shown promise when used as a pretreatment Small-for-Size Syndrome
of a small graft and of the recipient, and it may
reduce the likelihood of inadequate liver function There is no definite test to determine preopera-
in the liver remnant [51]. Pentoxifylline is a TNF-­ tively how much hepatic tissue can be safely
alpha synthesis inhibitor found in Kupffer cells, resected. Should the liver remnant volume
and its usefulness has only been studied in the (LRV) be below a certain threshold it cannot
murine model of partial liver transplant. Another sustain normal physiological functions and will
agent, Acetylcysteine, has also been examined progress into liver failure in the postoperative
for its roll in hepatoprotection [52, 53]. However, period (approximately 3–5 days after surgery).
clinical trials of its use in the perioperative treat- “Small-­for-­size syndrome” (SFSS) is the term
ment of patients undergoing liver transplant have used to describe the postoperative course of
not shown an overt benefit for the patient. Other jaundice, coagulopathy, encephalopathy, cere-
molecules, such as cardiotrophin-1 (an interleu- bral edema, ascites, renal and pulmonary failure
kin-­6 cytokine), somatostatin (a beneficial agent seen with this potentially catastrophic compli-
in reducing portal pressure), FK 409 (a low-dose cation [59, 60].
29  Complications of Liver Surgery 383

The pathophysiology of SFSS is not necessar- liver), may benefit from portal vein embolization
ily related only to the size of the remaining graft (PVE) or from Associating Liver Partition and
but also to the hemodynamic alterations within Portal vein ligation for Staged hepatectomy
the remaining liver parenchyma. Increased portal (ALPPS). Selective embolization leads to consid-
vein flow (PVF) can lead to a rise in portal pres- erable shrinking of the diseased areas and simul-
sure and increased shear stress in the hepatic taneous hypertrophy of the LRV. PVE not only
parenchyma that may contribute to sinusoidal increases the volume of the remaining liver, but
endothelial cell injury, vascular occlusion and also improves its function and decreases the inci-
perisinusoidal hemorrhage and hepatocellular dence of postoperative hepatic dysfunction [66].
necrosis [54, 56, 61]. These changes typically If PVE cannot be accomplished ALPPS may be a
manifest as early as minutes after the hepatic suitable alternative in experienced centers.
resection is completed. Furthermore excessive ALPPS is a two stage procedure especially for
PVF leads via the hepatic artery buffer response patients where the future liver remnant (FLR) is
to a compensatory hepatic arterial vasoconstric- considered too small [67]. The first step is the
tion triggering ischemic cholangitis and centrol- partition of the liver with the ligation of the portal
obular necrosis. This complication is seen in the vein, interruption of intrahepatic flow but mainte-
later stages of SFSS [62]. While a certain degree nance of arterial flow, thus avoiding compete loss
of portal hypertension is critical for liver regen- of function [68]. Hence the future liver remnant
eration, a pressure higher than 20 mmHg can lead can regenerate and grow with reduced risk of
to graft failure and decreased survival in patients overt liver failure. Once the FLR has hypertro-
after living-donor liver transplantation [63]. phied sufficiently (usually within a week or two),
Prediction of liver failure is difficult and not the partitioned liver is removed in a second step.
only depends on the extent of hepatic resection, However this approach has a high operative mor-
but also is influenced by the severity of any pre- tality and morbidity, patients need to be carefully
existing liver disease, liver function and patient selected and the procedure is commonly reserved
age. The first step is to assess how much liver tis- for experienced liver surgery centers.
sue needs to be resected in the individual patient.
The consensus is that in patients with normal
hepatic parenchyma up to four segments can Strategies to Prevent SFSS
safely be resected (approximately 50–60%). In
healthy individuals with a normal liver even Increased portal vein (PVF) flow and portal vein
resections up to 80% without complications have pressures (PVP) as well as decreased hepatic
been reported. A recent study questioned that and artery flow through the hepatic arterial buffer
suggested that the total remaining liver remnant response (HABR) are key factors in the develop-
volume (LRV) measured by CT volumetry is a ment of SFSS and therefore most early treat-
better predictor than the actual number of ana- ments have focused on modulating blood flow to
tomical segments resected. An analysis of 126 the remnant liver. Both surgical and pharmaceuti-
patients undergoing liver resection for colorectal cal methods have been described.
metastases showed that 90% of the patients with Splenic artery ligation (SAL) results in
less than 25% remaining volume developed liver decreased splanchnic blood flow and as a result
failure, whereas none did with >25% of liver lower portal flow and increased hepatic artery
remnant volume [64]. Thus the recommended blood flow via the hepatic artery buffer response.
minimal functional LRV in patients with normal In split liver transplant (living and cadaveric)
livers undergoing extended hepatectomies should recipients this method may improve graft function
be >25%, and >40% in a impaired liver such as and patient survival, even in a subset of patients
steatosis, cirrhosis, fibrosis or following chemo- that had persistent portal hypertension. The role
therapy [65]. Patients deemed unresectable based of a splenectomy in these patients remains unclear,
on a too small predicted LRV (<25% in a normal however it may prevent associated complications
384 O. P. F. Panzer

of SAL such as abscess formation after splenic associated with pneumoperitoneum and compli-
infarction. The creation of portocaval shunts cations of the operative procedure necessitating
(PCS) to reduce PVF and PVP may also reduce conversion to an open procedure [75]. Reduced
the incidence of SFSS and patient survival in access to achieve sufficient hemostasis still lim-
small FLR and liver grafts. However the long term its the widespread use of minimally invasive liver
adverse effects of a persistent PCS are not well surgery. However, with the use of newer surgical
known in this patient population. devices and increasing experience of surgeons,
Pharmacological interventions to reduce PVF laparoscopic liver surgery represents an effective
and PVP using vasoactive agents may prove ben- method of surgery even for more complex liver
eficial in avoiding postoperative hepatic dysfunc- resections. Hemodynamic goals that are used in
tion/SFSS. Vasopressin or terlipressin both conventional liver surgery (i.e. severe hypovo-
decreased portal venous blood flow and pressure. lemia and low CVP) may be more difficult to
However a safe reduction of PVP and subsequent achieve during laparoscopic liver surgery as infla-
improvement in survival and regeneration of the tion of the peritoneum already decreases substan-
liver tissue has only been demonstrated in animal tially preload.
models for SFSS [69, 70]. Intraportal infusion of Over the years the number of hepatic trans-
Prostaglandin E1, thromboxane A2 synthase plants and partial resections has increased mostly
inhibitor, nafamostat mesilate, octreotide and due to safer surgical techniques and newer phar-
esmolol all demonstrated beneficial effects in macological therapies. While this may have lead to
various animal studies and clinical case reports, beneficial results in many patients, it has also
however, only future clinical trials will determine allowed for the surgical option in extremely sick
their ultimate use before their routine use can be patients previously deemed inoperable, thus lead-
recommended [71–73]. ing to intraoperative complications. Interestingly
the perioperative morbidity and mortality are still
improving, most likely secondary to development
Minimal Invasive Resection of parenchymal sparing surgical techniques,
reduced blood loss and potential hepatoprotective
Minimally invasive liver resection is gaining strategies. There is and always will be the threat of
popularity, however its limitations continue to be intraoperative complications, and it is only with
intraoperative control of bleeding and hemostasis. the skillfulness of the surgical team and the fore-
Due to concerns about compromising oncologic sight of the anesthesiologist, that we can effec-
resection and technically difficult manipulations tively combat these complications.
in patients with large tumors, and tumors near the
hilum are commonly not considered for laparo-
scopic surgery. Initial attempts at laparoscopic References
resection were reserved for peripheral lesions,
mainly on the left lobe [74]. Recent advances in 1. Hendriks HGD, van der Meer J, de Wolf JTM, et al.
Intraoperative blood transfusion requirement is the
surgical devices to control parenchymal bleed- main determinant of early surgical re-intervention
ing in open hepatectomies, have also lead to the after orthotopic liver transplantation. Transpl Int.
increased use of laparoscopic techniques for 2005;17:673–9.
technically more challenging hepatectomies. 2. Cacciarelli TV, Keeffe EB, Moore DH, et al. Effect of
intraoperative blood transfusion on patient outcome in
The Pringle maneuver can be performed lapa- hepatic transplantation. Arch Surg. 1999;134:25–9.
roscopically to control vascular inflow, and the 3. Stainsby D, Williamson L, Jones H, Cohen H. 6 Years
parenchymal bleeding that is encountered during of shot reporting—its influence on UK blood safety.
transection can be controlled with pressure appli- Transfus Apher Sci. 2004;31:123–31.
4. de Boer MT, Molenaar IQ, Hendriks HGD, Slooff
cation techniques and newer hemostatic devices MJH, Porte RJ. Minimizing blood loss in liver trans-
[18, 74]. The major complications that can occur plantation: progress through research and evolution of
are difficulty with access, physiologic alterations techniques. Dig Surg. 2005;22:265–75.
29  Complications of Liver Surgery 385

5. Ramos E, Dalmau A, Sabate A, et al. Intraoperative 21. Moug SJ, Smith D, Leen E, Angerson WJ, Horgan
red blood cell transfusion in liver transplantation: PG. Selective continuous vascular occlusion and
influence on patient outcome, prediction of require- perioperative fluid restriction in partial hepatectomy.
ments, and measures to reduce them. Liver Transpl. Outcomes in 101 consecutive patients. Eur J Surg
2003;9:1320–7. Oncol. 2007;33:1036–41.
6. Porte RJ, Hendriks HGD, Slooff MJH. Blood conser- 22. Chen H, Merchant NB, Didolkar MS. Hepatic resection
vation in liver transplantation: the role of aprotinin. J using intermittent vascular inflow occlusion and low
Cardiothorac Vasc Anesth. 2004;18:31S–7S. central venous pressure anesthesia improves morbidity
7. Foster JH, Berman MM. Solid liver tumors. Major and mortality. J Gastrointest Surg. 2000;4:162–7.
Probl Clin Surg. 1977;22:1–342. 23. Smyrniotis V, Kostopanagiotou G, Theodoraki K,

8. Jarnagin WR, Gonen M, Fong Y, et al. Improvement Tsantoulas D, Contis JC. The role of central venous
in perioperative outcome after hepatic resection: anal- pressure and type of vascular control in blood loss
ysis of 1,803 consecutive cases over the past decade. during major liver resections. Am J Surg. 2004;187:
Ann Surg. 2002;236:397–406; discussion 406–7. 398–402.
9. Helling TS, Blondeau B, Wittek BJ. Perioperative fac- 24. Wang W-D, Liang L-J, Huang X-Q, Yin X-Y. Low
tors and outcome associated with massive blood loss central venous pressure reduces blood loss in hepa-
during major liver resections. HPB (Oxford). 2004;6: tectomy. World J Gastroenterol. 2006;12:935–9.
181–5. 25. Eid EA, Sheta SA, Mansour E. Low central venous
10. Sirichindakul B, Chanwat R, Nonthasoot B, Suphapol pressure anesthesia in major hepatic resection. Middle
J, Nivatvongs S. Risk factors associated with major East J Anesthesiol. 2005;18:367–77.
intraoperative blood loss in hepatic resection for hepa- 26. Massicotte L, Lenis S, Thibeault L, Sassine M-P, Seal
tobiliary tumor. J Med Assoc Thail. 2005;88(Suppl 4): RF, Roy A. Effect of low central venous pressure and
S54–8. phlebotomy on blood product transfusion requirements
11. Bergenzaun L, Gudmundsson P, Öhlin H, et al.
during liver transplantations. Liver Transpl. 2006;12:
Assessing left ventricular systolic function in shock: 117–23.
evaluation of echocardiographic parameters in inten- 27. Feng Z-Y, Xu X, Zhu S-M, Bein B, Zheng S-S. Effects
sive care. Crit Care. 2011;15:R200. of low central venous pressure during preanhepatic
12. Tripodi A, Salerno F, Chantarangkul V, et al. Evidence phase on blood loss and liver and renal function in
of normal thrombin generation in cirrhosis despite liver transplantation. World J Surg. 2010;34:1864–73.
abnormal conventional coagulation tests. Hepatology. 28. Kumar A, Anel R, Bunnell E, et al. Pulmonary

2005;41:553–8. artery occlusion pressure and central venous pres-
13. Lisman T, Leebeek FWG. Hemostatic alterations
sure fail to predict ventricular filling volume, cardiac
in liver disease: a review on pathophysiology, clini- ­performance, or the response to volume infusion in
cal consequences, and treatment. Dig Surg. 2007;24: normal subjects. Crit Care Med. 2004;32:691–9.
250–8. 29. Berrevoet F, de Hemptinne B. Use of topical hemo-
14. Westerkamp AC, Lisman T, Porte RJ. How to mini- static agents during liver resection. Dig Surg. 2007;24:
mize blood loss during liver surgery in patients with 288–93.
cirrhosis. HPB (Oxford). 2009;11:453–8. 30. Figueras J, Llado L, Miro M, et al. Application of
15. van Gulik TM, de Graaf W, Dinant S, Busch ORC, fibrin glue sealant after hepatectomy does not seem
Gouma DJ. Vascular occlusion techniques during justified: results of a randomized study in 300 patients.
liver resection. Dig Surg. 2007;24:274–81. Ann Surg. 2007;245:536–42.
16. Yi B, Qiu Y-H, Liu C, et al. Hepatic blood inflow 31. Fergusson DA, Hebert PC, Mazer CD, et al. A com-
occlusion with/without hemihepatic artery control parison of aprotinin and lysine analogues in high-risk
versus the Pringle maneuver in resection of hepatocel- cardiac surgery. N Engl J Med. 2008;358:2319–31.
lular carcinoma: a retrospective comparative analysis. 32. Molenaar IQ, Warnaar N, Groen H, Tenvergert EM,
Chin Med J. 2010;123:1413–6. Slooff MJH, Porte RJ. Efficacy and safety of antifi-
17. Alkozai EM, Lisman T, Porte RJ. Bleeding in liver brinolytic drugs in liver transplantation: a systematic
surgery: prevention and treatment. Clin Liver Dis. review and meta-analysis. Am J Transplant. 2007;7:
2009;13:145–54. 185–94.
18. Takayama T, Makuuchi M, Kubota K, et al.
33. Warnaar N, Polak WG, de Jong KP, et al. Long-term
Randomized comparison of ultrasonic vs clamp tran- results of urgent revascularization for hepatic artery
section of the liver. Arch Surg. 2001;136:922–8. thrombosis after pediatric liver transplantation. Liver
19. Melendez JA, Arslan V, Fischer ME, et al. Perioperative Transpl. 2010;16:847–55.
outcomes of major hepatic resections under low central 34. Mangano DT, Tudor IC, Dietzel C. Multicenter Study
venous pressure anesthesia: blood loss, blood transfu- of Perioperative Ischemia Research Group, Ischemia
sion, and the risk of postoperative renal dysfunction. J Research and Education Foundation. The risk associ-
Am Coll Surg. 1998;187:620–5. ated with aprotinin in cardiac surgery. N Engl J Med.
20. Jones RM, Moulton CE, Hardy KJ. Central venous 2006;354:353–65.
pressure and its effect on blood loss during liver resec- 35. Lodge JPA, Jonas S, Oussoultzoglou E, et al. Recombi-
tion. Br J Surg. 1998;85:1058–60. nant coagulation factor VIIa in major liver resection: a
386 O. P. F. Panzer

randomized, placebo-controlled, double-­blind clinical 51. Tian Y, Jochum W, Georgiev P, Moritz W, Graf R,
trial. Anesthesiology. 2005;102:269–75. Clavien P-A. Kupffer cell-dependent TNF-alpha
36. Planinsic RM, van der Meer J, Testa G, et al. Safety and signaling mediates injury in the arterialized small-
efficacy of a single bolus administration of recombi- for-­size liver transplantation in the mouse. Proc Natl
nant factor VIIa in liver transplantation due to chronic Acad Sci U S A. 2006;103:4598–603.
liver disease. Liver Transpl. 2005;11:895–900. 52. Weigand MA, Plachky J, Thies JC, et al. N-acetylcyste-
37. Hendriks HG, Meijer K, de Wolf JT, et al. Reduced ine attenuates the increase in alpha-­glutathione S-trans-
transfusion requirements by recombinant factor VIIa ferase and circulating ICAM-1 and VCAM-1 after
in orthotopic liver transplantation: a pilot study. reperfusion in humans undergoing liver transplantation.
Transplantation. 2001;71:402–5. Transplantation. 2001;72:694–8.
38. de Meijer VE, Kalish BT, Puder M, Ijzermans JNM.  53. Bucuvalas JC, Ryckman FC, Krug S, Alonso MH,
Systematic review and meta-analysis of steatosis as Balistreri WF, Kotagal U. Effect of treatment with
a risk factor in major hepatic resection. Br J Surg. prostaglandin E1 and N-acetylcysteine on pediat-
2010;97:1331–9. ric liver transplant recipients: a single-center study.
39. Simmonds PC, Primrose JN, Colquitt JL, Garden Pediatr Transplant. 2001;5:274–8.
OJ, Poston GJ, Rees M. Surgical resection of 54. Man K, Lee TK, Liang TB, et al. FK 409 ameliorates
hepatic metastases from colorectal cancer: a sys- small-for-size liver graft injury by attenuation of por-
tematic review of published studies. Br J Cancer. tal hypertension and down-regulation of Egr-1 path-
2006;94:982–99. way. Ann Surg. 2004;240:159–68.
40. Cucchetti A, Ercolani G, Vivarelli M, et al. Impact of 55. Bustos M, Beraza N, Lasarte J-J, et al. Protection
model for end-stage liver disease (MELD) score on against liver damage by cardiotrophin-1: a hepatocyte
prognosis after hepatectomy for hepatocellular carci- survival factor up-regulated in the regenerating liver
noma on cirrhosis. Liver Transpl. 2006;12:966–71. in rats. Gastroenterology. 2003;125:192–201.
41. Hartog A, Mills G. Anaesthesia for hepatic resection 56. Xu X, Man K, Zheng S-S, et al. Attenuation of acute
surgery. Continuing Educ Anaesth Critical Care Pain. phase shear stress by somatostatin improves small-
2009;9:1–5. for-size liver graft survival. Liver Transpl. 2006;12:
42. Bahde R, Spiegel H-U. Hepatic ischaemia-­reperfusion 621–7.
injury from bench to bedside. Br J Surg. 2010;97: 57. Beck-Schimmer B, Breitenstein S, Urech S, et al. A
1461–75. randomized controlled trial on pharmacological pre-
43. Gurusamy KS, Gonzalez HD, Davidson BR. Current pro- conditioning in liver surgery using a volatile anes-
tective strategies in liver surgery. World J Gastroenterol. thetic. Ann Surg. 2008;248:909–18.
2010;16:6098–103. 58. Beck C, Schwartges I, Picker O. Perioperative liver
44. Esaki M, Shimada K, Nara S, et al. Left hepatic trisec- protection. Curr Opin Crit Care. 2010;16:142–7.
tionectomy for advanced perihilar cholangiocarcinoma. 59. Dahm F, Georgiev P, Clavien P-A. Small-for-size syn-
Br J Surg 2013;100:801–7. drome after partial liver transplantation: definition,
45. Belghiti J, Marty J, Farges O. Techniques, hemody- mechanisms of disease and clinical implications. Am
namic monitoring, and indications for vascular clamping J Transplant. 2005;5:2605–10.
during liver resections. J Hepatobiliary Pancreat Surg 60. Kam I, Lynch S, Svanas G, et al. Evidence that host size
1998;5:69–76. determines liver size: studies in dogs receiving ortho-
46. Gurusamy KS, Kumar Y, Pamecha V, Sharma D,
topic liver transplants. Hepatology. 1987;7:362–6.
Davidson BR. Ischaemic pre-conditioning for elective 61. Man K, Su M, Ng KT, et al. Rapamycin attenuates
liver resections performed under vascular occlusion. liver graft injury in cirrhotic recipient--the signifi-
Cochrane Database Syst Rev. 2009:CD007629. cance of down-regulation of Rho-ROCK-VEGF path-
47. Gurusamy KS, Kumar Y, Sharma D, Davidson BR.  way. Am J Transplant. 2006;6:697–704.
Methods of vascular occlusion for elective liver 62. Kelly DM, Bennett R, Brown N, et al. Predicting the
resections. Cochrane Database Syst Rev. 2007: discharge status after liver transplantation at a single
CD006409. center: a new approach for a new era. Liver Transpl.
48. Gurusamy KS, Sheth H, Kumar Y, Sharma D,
2012;18:796–802.
Davidson BR. Methods of vascular occlusion for 63. Boillot O, Delafosse B, Méchet I, Boucaud C,

elective liver resections. Cochrane Database Syst Rev. Pouyet M. Small-for-size partial liver graft in an
2009:CD007632. adult recipient; a new transplant technique. Lancet.
49. Abu-Amara M, Gurusamy KS, Hori S, Glantzounis G, 2002;359:406–7.
Fuller B, Davidson BR. Pharmacological interventions 64.
Shoup M, Gonen M, D'Angelica M, et al.
versus no pharmacological intervention for ischaemia Volumetric analysis predicts hepatic dys-
reperfusion injury in liver resection surgery performed function in patients undergoing major liver
under vascular control. Cochrane Database Syst Rev. resection. J Gastrointest Surg. 2003;7:
2009:CD007472. 325–30.
50. Sato N, Murakami K, Ishida K, Ikeda K, Saito K.  65. Tucker ON, Heaton N. The ‘small for size’ liver syn-
Pulmonary hypertension and polymorphonuclear leu- drome. Curr Opin Crit Care. 2005;11:150–5.
kocyte elastase after esophageal cancer operations. 66. Uesaka K, Nimura Y, Nagino M. Changes in hepatic
Ann Thorac Surg. 1991;51:754–8. lobar function after right portal vein embolization. An
29  Complications of Liver Surgery 387

appraisal by biliary indocyanine green excretion. Ann injury in living donor adult liver transplantation.
Surg. 1996;223:77–83. Transpl Int. 2005;18:923–8.
67. Baumgart J, Lang S, Lang H. A new method for induc- 72. Jia C, Dai C, Bu X, et al. Co-administration of pros-
tion of liver hypertrophy prior to right trisectionectomy: taglandin E1 with somatostatin attenuates acute
a report of three cases. HPB (Oxford). 2011;13:71–2. liver damage after massive hepatectomy in rats via
68. de Santibañes E, Clavien PA. Playing Play-Doh to pre- inhibition of inflammatory responses, apoptosis
vent postoperative liver failure: the ‘ALPPS’ approach. and endoplasmic reticulum stress. Int J Mol Med.
Ann Surg. 2012;255:415–7. 2013;31:416–22.
69. Fahrner R, Patsenker E, de Gottardi A, et al. Elevated 73. Busani S, Marconi G, Schiavon L, et al. Living donor
liver regeneration in response to pharmacological liver transplantation and management of portal venous
reduction of elevated portal venous pressure by ter- pressure. Transplant Proc. 2006;38:1074–5.
lipressin after partial hepatectomy. Transplantation. 74. Abu Hilal M, Underwood T, Taylor MG, Hamdan K,
2014;97:892–900. Elberm H, Pearce NW. Bleeding and hemostasis in
70. Ren Z, Xu Y, Zhu S. The effect of terlipressin on
laparoscopic liver surgery. Surg Endosc. 2010;24:
hepatic hemodynamics in small-for-size livers. Hepato- 572–7.
Gastroenterology. 2012;59:208–11. 75. Sandadi S, Johannigman JA, Wong VL, Blebea J,
71. Suehiro T, Shimada M, Kishikawa K, et al. Effect of Altose MD, Hurd WW. Recognition and management
intraportal infusion to improve small for size graft of major vessel injury during laparoscopy. J Minim
Invasive Gynecol. 2010;17:692–702.
The Patient with Liver Disease
Undergoing Non-hepatic Surgery 30
Katherine Palmieri and Robert N. Sladen

Keywords  he Impact of Liver Disease


T
Non-hepatic surgery · Perioperative outcome · on Perioperative Outcome
Preoperative Evaluation · Anesthetic
Management · Abdominal surgery · Events that most commonly contribute to mor-
Cardiothoracic surgery bidity and mortality include bleeding, sterile or
infected ascites, pulmonary or genitourinary sep-
sis, decompensated liver failure and/or severe
acute kidney injury (AKI) [8–11]. In a 2014
Introduction study of 194 patients with cirrhosis undergoing
abdominal surgery, Neeff et al. observed that
It has been estimated that 5–10% of all patients 69% had a postoperative complication. Of these,
with cirrhosis of the liver will undergo surgery about half were related to the surgical procedure
other than liver transplantation during their last 2 and half to medical complications due to the
years of life [1]. Patients with any form of severe underlying liver disease. Postoperative mortality
liver disease who undergo non-­hepatic surgery was forbidding: 20% at 30 days and 30% at
are at markedly increased risk of perioperative 90 days. Median survival after hospital discharge
complications and mortality [2–7] and pose a was 2.83 years, with long-term survival at 1, 3
substantial perioperative challenge to anesthesi- and 5 years rapidly declining from 68.9% to
ologist. The myriad manifestations of liver dis- 50.1% to 32.8% respectively [12].
ease and its high operative risk imply that surgery The major determining factors are the sever-
should never be taken lightly in this group of ity of their liver disease, and the complexity
patients. and urgency of the surgical procedure
(Table  30.1). Del Olmo et al. [2] studied 135
patients with cirrhosis of the liver undergoing a
K. Palmieri, MD, MBA variety of non-­hepatic procedures. This group
Department of Anesthesiology, The University of had an increased blood transfusion require-
Kansas Health System, Kansas City, KS, USA
e-mail: kpalmieri@kumc.edu ment, longer hospital stays, more complica-
tions and a mortality rate of 16.3% compared to
R. N. Sladen, MBChB, MRCP(UK), FRCPC, FCCM (*)
Allen Hyman Professor Emeritus of Critical Care 3.5% in 86 matched controls. Similar differ-
Anesthesiology at Columbia University Medical ences were observed in patients with compen-
Center, College of Physicians and Surgeons of sated cirrhosis undergoing cholecystectomy,
Columbia University, New York, NY, USA colectomy, abdominal aortic aneurysm repair
e-mail: rs543@columbia.edu

© Springer International Publishing AG, part of Springer Nature 2018 389


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_30
390 K. Palmieri and R. N. Sladen

Table 30.1  Mortality rates associated with specific types Table 30.2 Independent predictors of postoperative
of surgery in patients with cirrhosis complications in cirrhotic patients undergoing surgery [3]
Overall Patient factors
Type of procedure mortality rate – Elevated serum creatinine (SCr)
Appendectomy [73] 9% – Preoperative infection
Bariatric surgerya [44] 10% – Chronic obstructive pulmonary disease (COPD)
Cardiac surgery [74, 75] 16–25% – American Society of Anesthesiology (ASA)
Cholecystectomy physical status 4 or 5
   Open, CTP Class C [36] 23–50% Disease factors
   Laparoscopic, CTP Class A and B 0–1% – Etiology of cirrhosis other than primary biliary
only [38, 76] cirrhosis (PBC)
   Endoscopic sphincterotomy for 7% – Child-Turcotte-Pugh (CTP) class B or C
common bile duct stones [77] – Preoperative upper gastrointestinal (UGI) bleeding
Esophageal surgery [14, 78, 79] 17–26% – Ascites
Herniorrhaphy [14] Surgical factors
   Incisional 6% – Invasiveness of procedure
   Umbilical, elective 2% – Intraoperative hypotension
   Umbilical, emergency 11%
Laparotomy for trauma [11] 45%
Table 30.3 Independent predictors of postoperative
Total knee arthroplasty [80] 0% mortality in cirrhotic patients undergoing surgery [3]
CTP = Child-Turcotte-Pugh
Patient factors
a
Includes perioperative and late deaths from liver failure
in 91 patients who continued with bariatric surgery despite – Male gender
the finding of unexpected cirrhosis – Preoperative infection
– American Society of Anesthesiology (ASA)
physical status 4 or 5
or coronary artery bypass grafting. Compared Disease factors
to non-cirrhotic controls, cirrhotic patients had – Etiology of cirrhosis other than primary biliary
an unadjusted mortality three- to eight-fold cirrhosis (PBC)
higher, total hospital charges 20–45% higher – Child-Turcotte-Pugh (CTP) class B or C
and lengths of stay 21–31% longer. Outcomes – Ascites
for patients with cirrhosis complicated by por- Surgical factors
tal hypertension were even worse [7]. – Thoracic surgery
In a retrospective review of 733 patients with
cirrhosis who underwent surgery other than
liver transplantation, Ziser et al. explored the the number of independent predictors and the
relationship between risk factors, morbidity and risk of perioperative complications (Fig. 30.1).
mortality [3]. Postoperative complications asso- The incidence of complications with one risk
ciated with increased mortality included pneu- factor was 9.3%, compared to 63% of patients
monia, ventilator dependence, infection, with 4 or 5 risk factors, and 100% with 7 or 8
new-onset or worsening ascites and cardiac risk factors. In sum, the risks and benefits of any
arrhythmias. Independent predictors of morbid- intervention should be carefully discussed with
ity and mortality that emerged after multivariate the patient or their surrogate, and non-surgical
analysis are listed in Tables 30.2 and 30.3. There therapy should be considered whenever possible
was an almost exponential r­ elationship between and appropriate.
30  The Patient with Liver Disease Undergoing Non-hepatic Surgery 391

Fig. 30.1  Effect of


number of risk factors

% of Patients with Complications


on perioperative 100
complication rate [3]

80

60

40

20

0
0 1 2 3 4 5 6 7-8
# of Risk Factors

 coring Systems to Assess Severity


S operative assessment may have known liver dis-
of Liver Disease ease or it may be revealed by a careful history and
physical exam. When the preoperative evaluation
The assessment of perioperative risk in patients reveals signs or symptoms of significant hepatic
with liver disease is complex, requiring consider- dysfunction in a patient without such a diagnosis,
ation of disease etiology, severity and chronicity, additional workup is appropriate. This may
and unrelated co-morbidity such as cardiac ­disease, include liver function tests (LFTs), hepatitis and
chronic obstructive pulmonary disease (COPD) or drug screening, right upper quadrant (RUQ) ultra-
diabetes mellitus [13]. The urgency and type of sur- sound, complete blood count (CBC) with platelet
gery also have a major impact on outcome. count, electrolytes, blood urea nitrogen (BUN),
Routine screening laboratory testing for serum creatinine, coagulation studies and other
hepatic disease in an otherwise asymptomatic and tests as indicated. Assessing the risk of periopera-
healthy surgical candidate is neither cost effective tive morbidity and mortality is important even in
nor helpful. Not only is the prevalence of liver patients with well-­ compensated liver disease.
disease low in the general population but an iso- Two disparate but well-established risk assess-
lated abnormal laboratory test is of questionable ment systems are helpful in decision-making for
significance and unlikely to change management the patient with liver disease undergoing non-
or outcome. The Mayo Clinic gave up routine pre- hepatic surgery.
operative screening with liver function tests in
1988 after only 0.3% of 3700 healthy, asymptom-
atic patients presenting for elective surgery were Child-Turcotte-Pugh (CTP)
found to have an abnormality in their liver Classification
enzymes. Of those laboratory abnormalities,
almost 20% were predictable based on history and In this well-established system, a score of 1, 2
physical exam and none were associated with or 3 is assigned based on the degree of abnor-
adverse outcome [14]. Patients presenting for pre- mality in each of five parameters: serum biliru-
392 K. Palmieri and R. N. Sladen

Table 30.4  Modified Child-Turcotte-Pugh score for cirrhosis


Point valuea
Variable 1 2 3
Ascites None Slight Moderate
Albumin <2 mg/dL 2–3 mg/dL >3 mg/dL
Prothrombin time (PT)
    Seconds > control <4 4–6 >6
    INR <1.7 1.7–2.3 >2.3
Encephalopathy None Grade 1–2 Grade 3–4
The modified Child-Turcotte-Pugh score utilizes the International normalized ratio (INR) in lieu of the PT. Encephalopathy
grades: 1 = constructional apraxia; 2 = asterexis, confabulation; 3 = stupor; 4 = coma
a
Class A = total score 5–6, Class B = total score 7–9, Class C = total score 10–15

bin, serum albumin, prothrombin time (PT) in Patients with CTP Class C disease represent a
seconds above normal, grade of encephalopa- very high risk of operative mortality and, as such,
thy and severity of ascites (Table 30.4). On this elective surgery is contraindicated in almost all
basis the minimum CTP score is 5 and the max- cases. Patients with CTP Class B disease fall into
imum score is 15. A score of 5–6 is assigned as an intermediate category and should be evaluated
CTP Class A, 7–9 as Class B and 10–15 as on an individual basis; consideration may be
Class C. given to elective surgery if preoperative medical
Observational studies dating back to the 1980s interventions can improve their status or surgery
and 1990s established the usefulness of the CTP provides a substantial benefit.
Class in predicting perioperative morbidity and There are a number of risk factors that are not
mortality. The risk of perioperative mortality was taken into consideration by the CTP Classification.
estimated as approximately 10% for Class A, For example, even in patients with CTP Class A
30% for Class B and as high as 82% for Class C liver disease, preoperative portal hypertension is
patients [15, 16]. an independent predictor of postoperative com-
In 2010 Telem et al. observed substantially plications such as jaundice, encephalopathy and
lower postoperative mortality rates of 2%, 12% ascites [18]. Preoperative placement of a tran-
and 12% respectively [17]. A reasonable sjugular intrahepatic portosystemic shunt (TIPS)
­assumption might be that mortality rates declined may decrease perioperative complications in
because of advances in surgical technique, anes- these patients [19].
thetic management and perioperative care.
However, about 40% of the patients in the Telem
study underwent less-risky laparoscopic proce-  odel for End-Stage Liver Disease
M
dures. In 2014, Neeff et al. described 30-day (MELD)
mortality rates of 6.3%, 12.8% and 53.2% for
CTP Class A, B and C [12]. Given these inconsis- The Model for End-stage Liver Disease (MELD)
tent results, additional studies are warranted to is based on a complex nomogram that incorpo-
better understand the relationship between CTP rates exponentials of three variables: total biliru-
Class and perioperative mortality rates. bin, serum creatinine and INR (international
Nonetheless, it is reasonable to conclude that normalized ratio of the prothrombin time).
patients with CTP Class A disease pose minimal MELD scores start at 6 and are capped at 40, with
to low risk for elective surgery and should pro- higher scoring patients at greatest risk of 3-month
ceed if there are no other contraindications. mortality (Fig. 30.2).
30  The Patient with Liver Disease Undergoing Non-hepatic Surgery 393

MELD = 3.8[Ln SB] + 11.2[Ln INR] + 9.6[Ln SCr] + 6.4

Fig. 30.2  Model for end-stage liver disease (MELD) c­ reatinine in mg/dL, Ln is the natural logarithm of each
score. SB = serum bilirubin in mg/dL, INR = International variable, A number of calculators are available on-line for
Normalized Ratio of prothrombin time, SCr = serum calculating MELD

Fig. 30.3  MELD score 80%


and risk of 30-day
mortality after non-­ 70%
transplant surgery in
patients with cirrhosis 60%
30-day Mortality %

[30]
50%

40%

30%

20%

10%

0%
5 10 15 20 25 30 35 40 45
MELD Score

Although originally developed to estimate 11, 17, 26], but a high score of 45 increased
3-month survival in patients undergoing TIPS, risk exponentially, to 67% [27]. Hanje
the MELD score has become the key indicator and Patel suggest that a MELD score of <10
for priority listing in patients considered for ­presents an acceptable risk for elective surgery,
liver transplantation. It has also been validated whereas the risk posed by a score >15 should
in assessing prognosis across a broad range of preclude elective surgery. Patients with a
liver diseases and severity, as well as in acute MELD score between 10 and 15 should
variceal bleeding and acute alcoholic hepatitis be evaluated individually, with particular
[20–23]. attention paid to the urgency and nature of
­
The MELD score has shown promise in pre- ­surgery [28].
dicting perioperative morbidity and mortality In general, there appears to a good correlation
in patients with liver disease undergoing between CTP Class and MELD score in predict-
abdominal, orthopedic, cardiovascular and ing perioperative morbidity and mortality [12,
other non-­ transplant surgeries [4, 24–26]. 24, 25, 29]. Neeff et al. [12] observed no clear
Northrup et al. retrospectively reviewed 140 advantage of one over the other, but Befeler et al.
non-transplant surgeries performed on patients found the MELD score to be superior to the CTP
with cirrhosis and found the MELD score to be Class in predicting mortality after intra-­
an independent predictor of 30-day mortality abdominal surgery [26]. They inferred this to be
(Fig.  30.3). A linear increase in preoperative because the CTP Class incorporates subjective
MELD score from 5 through 25 was associated criteria such as the degree of ascites and
with an equivalent increase in mortality risk [5, encephalopathy.
394 K. Palmieri and R. N. Sladen

 ther Laboratory Measures


O Table 30.5 Absolute contraindications to elective
­surgery in patients with liver disease [5, 15]
of Hepatic Function
Fulminant hepatic failure
Several quantitative tests of liver function have Acute viral or alcoholic hepatitis
been evaluated as predictors of perioperative Child-Turcotte-Pugh class C cirrhosis
morbidity and mortality in patients with liver dis- Severe coagulopathy
ease. These include the aminopyrine breath test,     PTT > 3 s above control despite treatment
    Platelet count <50,000/mm3
indocyanine green clearance, galactose elimina-
Severe extra-hepatic complications
tion capacity and the rate of metabolism of lido-
    Hypoxemia
caine to monoethylglycinexylidide (MEGX) [5].
    Cardiomyopathy, heart failure
However, their complexity and expense offer no
    Acute kidney injury
superiority to the CTP Class or MELD score in
clinical decision-making.
elective surgery (Table 30.5) [5, 30]. These
include CTP Class C cirrhosis, acute viral or alco-
 actors Affecting Outcome After
F holic hepatitis, symptomatic active hepatitis and
Non-hepatic Surgery fulminant liver failure. If severe coagulopathy
exists (PT > 3 s than control or platelet count
I mpact of the Severity and Nature <50,000/mcL) that is not readily correctable,
of Liver Disease elective surgery should not be performed. Severe
co-existing cardiac, renal or pulmonary failure
Most studies have examined perioperative risk in also contraindicate elective surgery.
the patient with cirrhosis and little information is
available about less severe liver disease or spe-  cute Liver Disease
A
cific liver diseases. However, the etiology of the Over the last four decades the poor outcome after
liver disease appears to have less impact on out- surgery in patients with acute alcoholic and viral
come than the degree of preoperative hepatocel- hepatitis has been well documented [13, 31, 32].
lular dysfunction. Severe liver failure is estimated As most cases of acute hepatitis are self-limited,
to be the proximate cause of as many as 50% of all but the most emergent procedures should be
postoperative deaths in patients with advanced postponed until the patient has made a full recov-
liver disease undergoing emergency gastrointes- ery or underwent transplantation. This implies
tinal procedures [10]. Kim et al. compared peri- resolution of active inflammation as measured by
operative morbidity and mortality in surgical transaminase levels or cellular infiltration on
patients with cirrhosis, non-cirrhotic liver liver biopsy. If the decision is made to proceed
­disease, or without liver disease. Patients with with surgery after recovery, these patients should
non-­cirrhotic liver disease experienced signifi- be carefully managed preoperatively to medi-
cantly more postoperative complications than cally optimize any co-existing systemic
those without liver disease (11.8% vs. 6.1%). derangements.
However, postoperative mortality was rare and
not different between these two groups (0.6% vs.  hronic Liver Disease
C
0.7%). In contrast, patients with cirrhosis had a The prognosis and life expectancy of a patient
significantly higher incidence of complications with chronic liver disease depends on patient
(32.5%) and mortality (10.2%) due to bleeding age, disease etiology and severity, the presence
(48%), sepsis (32%), acute liver failure (12%) of complications and the availability of treat-
and acute renal failure (8%) [11]. ment options [33]. In a study of the natural
Certain conditions pose such an increased risk ­history of compensated cirrhosis, almost half
of postoperative morbidity and mortality that they the patients developed at least one major com-
are generally recognized as contraindications to plication, including ascites, jaundice, hepatic
30  The Patient with Liver Disease Undergoing Non-hepatic Surgery 395

encephalopathy or gastrointestinal hemorrhage. abdominal viscera [39]. Vascular adhesions


In comparison with patients without major com- from prior abdominal surgery exacerbate
plications, their median survival time was 1.6 ­intraoperative blood loss and the potential for
versus 8.9 years [34]. Preoperative elucidation ischemic liver injury [5]. Co-existing portal
of a history of one of these key complications hypertension causes hepatic venous engorge-
provides important insight into the patient’s risk ment that further exacerbates perioperative
of postoperative morbidity and mortality. bleeding. Preoperative decompression of ele-
vated portal vein pressure by a TIPS procedure
may decrease the incidence of postoperative
Impact of the Surgical Procedure complications [26].
There is considerable evidence that laparo-
Emergency Surgery scopic surgery is safer for patients with cirrhosis
The risk of perioperative mortality increases two who require abdominal surgery and should be
to three-fold when cirrhotic patients undergo preferred whenever possible [40]. Potential ben-
emergency abdominal or trauma surgery [4, 16, efits include decreased perioperative loss of
24, 35, 36], and is reliably predicted by the CTP blood and ascites, rate of infection and inflamma-
Child-Turcotte and MELD score [4, 16]. In cir- tory response and a shorter hospital length of stay
rhotic patients emergency abdominal surgery is (LOS) [40, 41].
an independent predictor of postoperative hospi-
tal length of stay (LOS) [4] and is associated Cholecystectomy
with a mortality between 50% and 100%, versus Gallstones are twice as prevalent in patients with
(a still impressive) 18% for non-emergent proce- cirrhosis than in the general population, and it is
dures [16], Demetriades et al. compared not uncommon for these patients to present for
­outcomes in cirrhotic and non-cirrhotic patients cholecystectomy [42]. Open cholecystectomy has
undergoing emergency laparotomy for traumatic been associated with an unacceptable periopera-
injury [36]. Cirrhotic patients had a significantly tive mortality, between 26 and 50% [43, 44]. Even
higher risk of complications, and increased patients with a relatively low MELD (8 or
intensive care unit (ICU) LOS, hospital charges above)—especially when accompanied by throm-
and overall mortality (45% vs. 24%). These dif- bocytopenia—have a substantially increased risk
ferences occurred with even relatively minor of postoperative complications [25].
trauma, and the authors recommended ICU Laparoscopic cholecystectomy, previously
admission for any cirrhotic patient undergoing a rejected because of concerns for uncontrollable
trauma-related laparotomy regardless of injury bleeding and postoperative hepatic failure, is now
severity of injury. considered to be the far safer approach. This is
particularly applicable to patients with CTP Class
Abdominal Surgery A or B disease, whose cirrhosis is well-­
The postoperative mortality of patients with compensated and who do not have portal hyper-
cirrhosis undergoing open, non-hepatic abdom- tension [45–48]. Compared to open
inal surgery is forbidding, ranging from 14% to cholecystectomy, documented benefits include
35% [15, 26, 35, 37, 38]. As one might expect, shorter operative times, decreased intraoperative
the mortality after intra-abdominal surgery is blood loss, fewer postoperative complications,
considerably higher than after procedures con- faster return to a normal diet and shorter hospital
fined to the abdominal wall—35% vs. 8% LOS (Table 30.6) [48, 49]. However, even after
respectively in a retrospective study by Neeff laparoscopic cholecystectomy, patients with a
et al. [35]. MELD >13 have an increased risk of requiring
Open laparotomy considerably impairs conversion to open surgery and complications
hepatic blood flow, in part due to reflex vasodi- such as intraoperative hemorrhage, ascites and
lation and hypotension induced by retraction of infection [8].
396 K. Palmieri and R. N. Sladen

Table 30.6  Laparoscopic versus open cholecystectomy [40]


Laparoscopic cholecystectomy Open cholecystectomy p
Operative times (min) 123.3 150.2 <0.042
Intraoperative blood loss (mL) 113 425.2 <0.015
Hospital length of stay (d) 6 12.2 <0.001
Data compiled from a meta-analysis of four studies comparing laparoscopic with open cholecystectomy in patients with
hepatic cirrhosis [40]. For explanation, see text

Bariatric Surgery and mortality increase with higher CTP Class


Obesity is a worldwide problem affecting more and MELD. A large meta-analysis revealed early-­
than two billion people [50], and predisposes to hospital mortality rates of 8.92%, 31.38% and
hepatic fat deposition (steatosis). In some it pro- 47.62% for patients of CTP Class A, B and C
gresses to non-alcoholic fatty liver disease respectively. Severe complications occur in
(NAFLD). About 30% of patients with NAFLD 10–20% of cases, and include acute kidney
will develop an inflammatory response to fat injury, respiratory failure, cardiovascular decom-
called non-alcoholic steatohepatitis (NASH). In pensation and sepsis. Although acute hepatic fail-
about a quarter of these patients inflammation ure is relatively less common (5–6% of cases) it
results in diffuse fibrosis and end stage liver dis- may be associated with a mortality rate as high as
ease (ESLD) [51, 52]. 75% [59, 60].
As bariatric surgery plays a more prominent Standard recommendations are to avoid car-
role as obesity intervention, so does the finding of diac surgery in patients of CTP Class B and C
NAFLD at the time of surgery without any his- [29, 61, 62], but more recently it is suggested that
tory of overt liver disease. Data from multiple it could be considered in carefully selected CTP
studies in patients undergoing laparoscopic Class B patients [63–65]. It is generally accepted
Roux-Y gastric bypass or sleeve gastrectomy that a MELD >13–15 presents risks that out-
reveal that almost all patients have histological weigh benefits and preclude elective cardiac sur-
findings of steatosis, 24–98% have NASH, and gery [66–68].
1–7% have ESLD [53, 54]. At the same time, Regardless of CTP Class or MELD, perioper-
there is considerable evidence that successful ative risk is further increased in the presence of
bariatric surgery can not only result in weight coagulopathy, immune dysfunction, poor nutri-
loss but also stop and even reverse NASH and tional status, renal and/or pulmonary dysfunction
NAFLD, including a regression of fibrosis and cirrhotic cardiomyopathy. Existing coagu-
[55–58]. lopathy is aggravated by CPB-induced platelet
Nonetheless, patients who present for bariatric dysfunction, fibrinolysis and hypocalcemia. The
surgery with NAFLD or NASH have increased potential for postoperative hepatic decompensa-
perioperative complications and mortality. One tion is increased by prolonged CPB, requirement
review found an overall complication rate of for vasopressor therapy, and non-pulsatile (ver-
21.3%, an early mortality rate of 1.6% and a late sus pulsatile) CPB [5].
mortality rate of 2.45%—rates higher than those
observed in patients without liver disease [55].  ff-Pump Coronary Artery Bypass
O
Grafting (OPCAB)
Cardiothoracic Surgery Avoiding cardiopulmonary bypass may amelio-
rate the inflammatory response during cardiac
Cardiac Surgery with Cardiopulmonary surgery and thus the risk of developing acute on
Bypass chronic liver failure for patients with chronic
It is well recognized that ESLD predicts adverse liver disease. Randomized controlled trials com-
outcomes after cardiac surgery with cardiopul- paring coronary artery bypass grafting (CABG)
monary bypass (CPB). Perioperative morbidity with CPB to off-pump CABG (OPCAB) in
30  The Patient with Liver Disease Undergoing Non-hepatic Surgery 397

patients with liver disease have not been per- B disease had morbidity and mortality rates of
formed. However, several retrospective series 30.8% and 7.6% respectively [77].
and case reports describe successful outcomes
after OPCAB in cirrhotic patients with disease as
severe as CTP class B and C [65, 69–73]. A ret- Preoperative Evaluation
rospective review of U.S. Nationwide Inpatient and Preparation for Surgery
Sample (NIS) data evaluated more than 6000 cir-
rhotic patients who underwent CABG between  scites, Fluid and Electrolyte
A
1998 and 2009 [74]. About one third underwent Imbalance
OPCAB. A subgroup analysis revealed that cir-
rhotic patients assigned to OPCAB were sicker, Ascites elevates the diaphragm and decreases
with a higher incidence of emergency surgery or functional residual capacity (FRC) and is associ-
requirement for an intra-aortic balloon pump ated with basal atelectasis and sympathetic pleu-
(IABP). Despite this, morbidity and mortality ral effusions that further compromise oxygenation
was comparable with patients undergoing CABG and ventilation in the perioperative period. The
with CPB, and the authors concluded that presence of ascites also increases the risk of post-
OPCAB is preferred for cirrhotic patients when- operative wound dehiscence or abdominal wall
ever possible. herniation.
Severe, tense ascites may result in a compart-
Transcatheter Aortic Valve ment syndrome when abdominal pressure is ele-
Replacement vated >20 mmHg. This impairs cardiac output,
Over the last decade, transcatheter aortic valve renal blood flow and renal venous outflow result-
replacement (TAVR) has revolutionized the ing in progressive impairment of renal function.
management of high-risk patients with severe Restoring cardiac output does not reverse the sit-
aortic stenosis. Preliminary retrospective obser- uation and only decompressing the elevated
vations in cirrhotic patients undergoing TAVR intra-abdominal pressure will relieve it. A preop-
suggested a decrease in transfusion requirement, erative TIPS procedure may be very helpful in
perioperative complications and in-hospital preventing recurrence, but at the risk of increas-
mortality compared to open aortic valve replace- ing hepatic encephalopathy. In patients without
ment (AVR) [75]. A subsequent propensity- edema or in those for whom there is not enough
matched study comparing TAVR and open AVR time for a course of diuretics, it may be helpful to
in 60 cirrhotic patients supported these observa- drain tense ascites preoperatively or during lapa-
tions, and demonstrated a halving of the post- rotomy. Paracentesis must be performed cau-
procedural length of stay from about 2 weeks to tiously, however, given the risk of inducing acute
1 week [76]. intravascular hypovolemia and hypotension that
The experience with OPCAB and TAVR sug- may then further exacerbate liver injury and renal
gests that the least invasive procedure possible dysfunction.
should be considered for patients with ESLD Many patients with liver disease are pre-
who require cardiovascular interventions. scribed the aldosterone antagonist, spironolac-
tone, which is very effective in maintaining a
Thoracic Surgery modest potassium-sparing diuresis. However, its
There are few data on perioperative morbidity onset and offset are slow (2–3 days) and its
and mortality in patients with cirrhosis undergo- potassium-­sparing effect in acute renal insuffi-
ing thoracic surgery. In a small retrospective ciency may provoke acute hyperkalemia. If pos-
review of 17 cirrhotic patients who underwent sible, spironolactone therapy should be
surgery for non-small cell lung cancer, patients discontinued 3–4 days before surgery.
with CTP class A disease experienced no mor- Hyponatremia is common in patients with
bidity or mortality, whereas those with CTP class severe liver dysfunction, although usually not
398 K. Palmieri and R. N. Sladen

symptomatic. Treatment including fluid restric- moderate perioperative blood loss can have the
tion may be warranted preoperatively if the serum same result.
sodium concentration is less than 120– Two factors produced in the endothelium, von
125 mmol/L. Excessively rapid serum sodium Willebrand factor (VWF) and Factor VIII, are
elevation can rarely result in a devastating neuro- elevated in patients with cirrhosis, support plate-
logic complication, central pontine myelinolisis. let adhesion and can to a certain extent compen-
There is merit to the adage to correct hyponatre- sate for defects in platelet number and function
mia at a rate similar to that with which it devel- [79]. Even with severe degrees of renal failure,
oped to allow the brain to slowly adjust to the hepatic conversion of ammonia to urea is so
new plasma osmolality. impaired that uremic suppression of vWF-VIII
complex is unlikely.
Dysfibrinogenemia is characteristic of advanced
Coagulopathy liver failure and implies abnormal fibrinogen func-
tion even though plasma levels may be normal or
Liver disease is associated with complex altera- even elevated. Hyperfibrinogenemia is observed
tions of the hemostatic system categorized as a more frequently in patients with chronic hepatitis,
state of “rebalanced hemostasis” that may be hepatocellular carcinoma or cholestasis. About
tipped toward bleeding or thrombosis—or both 60–70% of fibrinogen is non-functional, with
[78]. The coagulopathy associated with liver dis- abnormal alpha chains and a high sialic acid con-
ease is multifactorial, but dominated by impaired tent, similar to that produced by immature
synthesis of the Vitamin K-dependent coagula- hepatocytes.
tion factors, Factors II, VII, IX and X. This is Anemia is common and may occur via several
exacerbated by malnutrition, cholestasis and use mechanisms including acute or chronic blood
of antibiotics such as neomycin that eliminate the loss, malnutrition and bone marrow suppression.
gut bacteria that produce Vitamin K. Inactive Chronic alcoholism may be associated with mac-
forms of the procoagulants are produced, known rocytic anemia.
as proteins induced by Vitamin K absence The liver is the source of the endogenous anti-
(PIVKA). coagulants, Protein C and Protein S that are vita-
Abnormalities in routine laboratory coagula- min K-dependent and produced by hepatocytes
tion tests may not accurately predict bleeding and antithrombin III that is vitamin K-independent
risk in patients with hepatic disease [78]. and produced in the hepatic endothelium.
However, the severity of hepatocellular disease is Impaired synthesis of any of these factors may
directly reflected by the degree of prolongation of create a prothrombotic state. Hypercoagulability
the prothrombin time (PT) or International is characteristic of primary biliary cirrhosis, pri-
Normalized Ratio (INR). The activated partial mary sclerosing cholangitis and hepatocellular
thromboplastin time (aPTT) is usually preserved carcinoma but can occur with liver disease of any
until ESLD is established, unless affected by etiology. Patients with NAFLD are also relatively
acute processes such as disseminated intravascu- prothrombotic [78]. It is not uncommon for
lar coagulation (DIC). hypercoagulability and hypocoagulability to
Moderate thrombocytopenia, i.e. a platelet coexist in patients with severe liver disease.
count 50–75,000/mcL, is common, a conse- Increased coagulability appears to be more prom-
quence of increased splenic sequestration (portal inent in the liver itself, predisposing to portal or
hypertension-induced hypersplenism) and hepatic vein thrombosis, whereas the hypocoagu-
decreased production (impaired hepatic throm- lability manifests extrahepatically with increased
bopoietin synthesis). Acute complications such risk of gastrointestinal or surgical bleeding.
as gastrointestinal bleeding or DIC may pro- Fibrinolysis is also disordered, because of
foundly decrease platelet count to <20,000/mcL. impaired synthesis of some fibrinolysins (notably
Given the already depleted platelet reserve, even plasminogen) and antifibrinolysins (histidine-­
30  The Patient with Liver Disease Undergoing Non-hepatic Surgery 399

rich glycoprotein, thrombin-activatable fibrinoly- patients with cirrhosis [81–83]. However, the
sis inhibitor). However, the most important drug is very costly. Administration of recombi-
fibrinolysin, tissue plasminogen activator (tPA), nant factor VIIa did not show a benefit in decreas-
is synthesized in the endothelium independently ing transfusion requirement in a study of patients
of liver function. undergoing partial hepatectomy [84]. However
some advocate concomitant administration of CP
Perioperative Correction to increase the fibrinogen substrate for clot
of Coagulopathy formation.
Attempts to correct coagulopathy should not be It is important to recognize that transfusion of
based solely on abnormal preoperative laboratory any blood component carries a risk of adverse
values. The PT and aPTT, in particular, do not pre- events, and evidence of the benefit of prophylac-
dict the risk of bleeding in patients with such com- tic correction of laboratory abnormalities is
plex hemostatic alterations as encountered in liver scanty [85, 86]. Rational perioperative transfu-
disease [78]. If available, whole blood thrombo- sion strategies in patients with liver disease
elastography (such as TEG® or ROTEM®) may be should be similar to those for any patient. These
used to identify specific coagulopathies to target include attempts to eliminate sepsis and optimize
the most appropriate intervention. cardiovascular and renal function prior to sur-
An attempt to correct a prolonged PT or INR gery; recognizing that hypothermia, hypocalce-
may be made using parenteral Vitamin K and/or mia and acidosis adversely affect endogenous
fresh frozen plasma (FFP). Vitamin K adminis- coagulation; and taking care to avoid rapid trans-
tration takes 12–18 h to become fully effective in fusion of FFP that predisposes to transfusion
restoring Factor VII levels, and the administra- associated circulatory overload (TACO).
tion of several units of FFP represents a substan- There is increasing evidence that patients with
tial volume load that may induce acute pulmonary chronic liver disease are not protected from peri-
congestion or even right heart failure. Patients operative thrombotic complications by being
with severe liver disease may become refractory “auto-anticoagulated”. Indeed, epidemiological
to Vitamin K or FFP transfusion. studies suggest that liver disease may actually
Prothrombin complex concentrates (PPC) confer an increased risk of deep venous thrombo-
contain Factors II, VII, IX and X as well as small sis (DVT) [87]. Prophylactic anticoagulation in
quantities of protein C and protein S to provide selected patients with cirrhotic liver disease, but
“hemostatic balance”. They are advocated for the without high-risk esophageal varices, appears to
rapid (10 min) reversal of a prolonged PT induced be safe [88], and should not exclude careful post-
by warfarin. Evidence for benefit of PPC admin- operative administration of thrombembolic pro-
istration to correct a prolonged PT secondary to phylaxis either using subcutaneous heparin or
ESLD is at present anecdotal. A small observa- low molecular weight (LMW) heparin if bleed-
tional study demonstrated rapid and “very good” ing risk is not excessive [89].
hemostasis in 16 of 22 patients with severe liver
disease given a virus-inactivated PCC without
adverse effects or evidence of viral disease trans- Pulmonary Disease
mission [80].
Cryoprecipitate (CP), that contains fibrinogen, Careful preoperative assessment of pulmonary
vWF, factor VIII and factor XIII in a relatively function, including arterial blood gas analysis
small volume may be indicated to treat severe and simple spirometry is appropriate in advanced
hypofibrinogenemia (normal levels 100–150 mg/ liver disease, and is essential for in any patient
dL). with a history of dyspnea or other pulmonary
Several studies have demonstrated that admin- symptoms. Compromised ventilatory reserve is a
istration of recombinant factor VIIa is a safe and given with advanced ascites, pleural effusions or
effective method of correcting coagulopathy in hydrothorax. Two conditions associated with
400 K. Palmieri and R. N. Sladen

progressive hypoxemia occur in about 6–10% of arteriovenous malformations, responds poorly to


patients with ESLD who are candidates for liver supplemental oxygen, and represents a very high
transplantation. They appear to be the conse- risk for surgery.
quence of splanchnic metabolites that in one The only recourse for reversal of severe HPS
(hepatopulmonary syndrome) induces pulmo- of either type is liver transplantation. Outcomes
nary vasodilation, and in the other, pulmonary are usually good and if possible, elective surgery
vasoconstriction [90]. should be deferred until after successful
transplantation.
Hepatic Hydrothorax
Hepatic hydrothorax is estimated to occur in  ortopulmonary Hypertension (PPH)
P
5–6% of patients with ESLD, and it is more Portopulmonary hypertension (PPH) is defined
commonly right-sided [91]. The severity of as a mean pulmonary artery pressure (MPAP)
symptoms depends on the rapidity and size of greater than 25 mmHg in a patient with coexist-
the effusion. If hydrothorax becomes refractory ing portal hypertension and no other obvious eti-
to sodium restriction and diuretics, patients may ology for pulmonary hypertension [95]. This is a
require escalating interventions such as thera- potentially lethal condition, particularly when
peutic thoracentesis, pleurodesis, video-assisted MPAP reaches moderately severe (35–50 mmHg)
thorascopic surgery (VATS) closure of dia- or severe (>50 mmHg) levels, with a high risk of
phragmatic defects or a TIPS procedure. acute right heart failure [96]. Workup requires
Preoperative thoracentesis should be reserved right heart catheterization and transesophageal
for patients with large, symptomatic effusions, echocardiography. Patients may be on the entire
because mild to moderate effusions rapidly gamut of pulmonary vasodilator therapy includ-
reaccumulate [92]. Postoperative thoracentesis ing prostacyclins (e.g. epoprostenol), phosphodi-
may be useful when a large pleural effusion esterase V inhibitors (e.g. sildenafil) and/or
impairs weaning from mechanical ventilatory endothelin antagonists (e.g. bosentan). Patients
support. with moderate or severe PPH are candidates for
no other surgery than liver transplantation. Even
Hepatopulmonary Syndrome (HPS) when MELD exceptions are applied to facilitate
Hepatopulmonary Syndrome (HPS) is character- early transplantation, mortality is high, with very
ized by fixed hypoxemia—an increased alveolar-­ unpredictable amelioration of pulmonary hyper-
arterial (A-a) gradient—and widespread tension [97].
intrapulmonary arteriovenous vasodilation and
shunting in the setting of chronic liver disease. Its
incidence is unpredictable and very variable [93, Renal Dysfunction and Injury in Liver
94]. Unique manifestations of HPS include Disease
upright dyspnea (platypnea), and hypoxemia
relieved by lying flat (orthodeoxia). Platypnea Hepatorenal Syndrome (HRS)
appears to be induced by rapid blood flow through Hepatorenal syndrome (HRS) is a poorly under-
the pulmonary circulation as a consequence of stood, complex, multifactorial disorder that is
low pulmonary vascular resistance (PVR) that often precipitated by complications in a patient
results in greater delivery of deoxygenated blood with advanced liver disease. One proposed
to the left atrium. The A-a gradient is worsened ­mechanism is the absorption of endotoxin into
by pooling of deoxygenated in the lung bases in the systemic circulation via porta-systemic
the upright position. shunts, exacerbated by Kupffer cell and hepatic
Type I or minimal pattern HPS has a very reticulo-­ endothelial dysfunction (Fig. 30.4).
finely dispersed radiographic pattern and is gen- Endotoxin is directly nephrotoxic to the tubular
erally responsive to an increase in supplemental epithelium and also induces disordered renal
oxygen. Type II HPS is characterized by discrete perfusion.
30  The Patient with Liver Disease Undergoing Non-hepatic Surgery 401

Whatever the triggering factor, progressive Historically there were two descriptive forms
splanchnic vasodilation occurs [98], ultimately of HRS (Table 30.7). Type 1 HRS is associated
with reflex renal vasoconstriction and sodium with shock or spontaneous bacterial peritonitis
retention so that the patient develops oliguria and is rapidly progressive. The serum creatinine
with a very low urine sodium (characteristically, (SCr) may increase from baseline to >2.5 mg/dL
<10 mEq/L).The oliguria is unresponsive to with a few weeks, and mortality is 50% within a
aggressive hydration, akin to a resistant prerenal month of onset. Acute renal decompensation can
syndrome. Histologically, there is no evidence be temporized by inducing efferent arteriolar
of cellular injury, reinforcing the concept that vasoconstriction with terlipressin (a vasopressin
this is a functional disorder. In fact, it has been analogue), and infusing albumin [99], but the
observed that normal kidney function is restored definitive therapy for Type 1 HRS is liver trans-
in a kidney donated from a patient dying of liver plantation [100].
failure into a recipient with normal liver Type 2 HRS is associated with refractory asci-
function. tes and elevated renal venous pressure, analogous
to a compartment syndrome, the SCr increases
slowly to about 1.5 mg/dL, and the median sur-
Hepatorenal Syndrome vival is about 6 months. These patients respond
very well to the TIPS procedure, which should be
considered as a preoperative intervention for
non-transplant surgery.
More recently the International Club of
Ascites revised the definition of HRS and
renamed it HRS-AKI. This new definition
describes three stages of HRS similar to stages of
Endotoxin AKI in other scenarios graded by severity of SCr
UNa < 10 increase. It was previously difficult to differenti-
ate clinically between AKI from HRS and the
Fig. 30.4  One of the proposed mechanisms of hepatore- new definition therefore does not discern between
nal syndrome (HRS). In advanced liver disease, porta-­ these two diagnosis anymore (Table 30.8).
systemic shunting facilitates the absorption of endotoxin
from the gut. It thereby bypasses the hepatic reticulo-­
endothelial filter and gains entry into the systemic circula-
 valuation of Renal Function
E
tion. Kupffer cell function is impaired so that any In advanced liver disease blood urea nitrogen
endotoxin that does perfuse the hepatic sinusoids is not (BUN) and SCr are unreliable and even mislead-
eliminated. Endotoxin makes its way to the renal circula- ing markers of the severity of renal dysfunction.
tion, where it causes disruption of circulatory homeosta-
sis, as well as direct cellular toxicity. The kidney reacts by
Amino acids are deaminated in the liver, produc-
avidly conserving sodium, so that urine sodium (UNa) is ing ammonia, which is converted to urea in the
typically < 10 mEq/L arginine cycle (Fig. 30.5). In ESLD the arginine

Table 30.7  Previously used definition and grades of hepatorenal syndrome (HRS)
Type HRS 1 HRS 2
Time course Rapidly progressive Slowly progressive
Serum creatinine (SCr) SCr > 2.5 mg/dL in 2 weeks SCr > 1.5 mg/dL
Complicates Shock, SBP Refractory ascites
Outcome Mortality 50% in 1 month Median survival 6 months
Treatment Terlipressin + albumin TIPS
Liver transplantation
SCr serum creatinine, SBP spontaneous bacterial peritonitis, TIPS Transjugular intrahepatic portosystemic shunt
Terlipressin is a vasopressin analog not available in the United States
402 K. Palmieri and R. N. Sladen

Table 30.8  New definitions and stages of hepatorenal syndrome(HRS)


Subject Definition
Baseline sCr •  SCr obtained in the previous 3 months,
•  Without a previous sCr value, use the sCr on admission
Definition of •  Increase in sCr ≥0.3 mg/dL (≥26.5 μmol/L) within 48 h; or,
AKI •  Increase sCr ≥50% from baseline which is known, or presumed, within the prior 7 days
Staging of •  Stage 1: increase in sCr ≥ 0.3 mg/dL (26.5 μmol/L) ≥ 1.5-fold to 2-fold from baseline
AKI •  Stage 2: increase in sCr > 2-fold to 3-fold from baseline
• Stage 3: increase of sCr > 3-fold from baseline to sCr ≥ 4.0 mg/dL (353.6 μmol/L) with an acute
increase ≥ 0.3 mg/dL (26.5 μmol/L) or initiation of renal replacement therapy
Progression Progression Regression
of AKI Progression of AKI to a higher Regression of AKI to a lower stage for RRT
stage and/or need
Response to No response Partial response Full response
treatment No regression of AKI Regression of AKI stage with a Return of sCr to a value
reduction of sCr to ≥0.3 mg/dL within 0.3 mg/dL
(26.5 μmol/L) above the (26.5 μmol/L) of the
baseline value baseline value
SCr serum creatinine, AKI acute kidney injury
With permission: Gut. 2015 Apr;64(4):531–7:Diagnosis and management of acute kidney injury in patients with
­cirrhosis: revised consensus recommendations of the International Club of Ascites

i­ndicate severe renal injury. Patients with


Ammonia and Urea Metabolism
advanced liver disease depleted muscle mass
and/or increased total body water, resulting in
amino acids
low levels of SCr that underestimate any decrease
Liver Kidney
in glomerular filtration rate (GFR).

NH3 Preoperative Preparation


arginine Hemodynamic derangements induced by anes-
Urea
cycle thesia and surgery increase the risk of renal hypo-
perfusion that exacerbates existing renal
dysfunction. Nephrotoxic medications, diuretics,
Fig. 30.5  Schematic of ammonia and urea metabolism. large-volume paracentesis, gastrointestinal
All amino acids undergo deamination in the liver, whereby bleeding or infection further increase the risk of
the -NH2 group is split off and converted into ammonia
(NH3). Ammonia enters the arginine cycle and is con- superimposed acute kidney injury (AKI). There
verted to urea, which is excreted in the urine. In acute kid- is limited evidence that pharmacologic interven-
ney injury (AKI), urea accumulates and the blood urea tions may ameliorate renal dysfunction in certain
nitrogen (BUN) rises. With an acute gastrointestinal bleed complications associated with advanced liver dis-
(e.g. variceal bleed), blood protein is absorbed and urea
production increases. In acute liver injury, the arginine ease. In one moderately large study in cirrhotic
cycle is impaired and ammonia is not converted to urea. patients with spontaneous bacterial peritonitis,
Ammonia accumulates and BUN remains very low, even the addition of large doses of albumin (1–1.5 g/
in the face of AKI or variceal bleeding kg) to antibiotic therapy substantially decreased
the incidence of AKI and mortality. Plasma renin
cycle fails, so ammonia accumulates and urea activity was decreased in patients receiving
production and BUN levels are markedly ­albumin, suggesting a salutary effect on renal
decreased, often to <10 mg/dL even in the face of perfusion [101]. Large volume paracentesis for
severe prerenal states (hypovolemia, gastrointes- severe ascites can induce acute intravascular
tinal bleeding) or frank renal failure. Thus, even hypovolemia and potentially, AKI. Administration
moderate elevations of BUN above normal of midodrine (an oral alpha-adrenergic agonist)
30  The Patient with Liver Disease Undergoing Non-hepatic Surgery 403

appears to be as effective as albumin infusion in Elevated arterial ammonia (NH3) is commonly


preventing paracentesis-induced circulatory dys- associated with abnormal central nervous system
function, with increased urine flow and natriure- (CNS) function, but it is a marker of disordered
sis [102]. When severe obstructive jaundice protein metabolism rather than a primary etio-
develops, excretion of bile salts—natural anti- logic factor. Acute encephalopathy may be pre-
toxin—is impaired and endotoxin may enter the cipitated by any number of additional factors that
portal system, where it is normally cleared by the are likely to occur in the perioperative period,
liver. However, in the presence of porto-systemic including hypovolemia, hypoglycemia, gastroin-
shunting and Kupffer cell dysfunction, endotoxin testinal bleeding, renal failure, active infection
gains access to the systemic circulation where it and administration of sedatives or opioids [30].
induces renal vasomotor dysregulation and direct Non-ionic diffusion trapping of NH3 is an
tubular injury. Preoperative bile salt administra- important CNS regulatory process. In an acidic
tion with sodium deoxycholate has been demon- milieu, NH3 gains a hydrogen ion to become
strated not only to decrease the incidence and ionized ammonium (NH4+) that cannot cross
severity of endotoxemia in obstructive jaundice lipid membranes. In an alkalotic milieu, NH4+
[103], but also to provide perioperative renal pro- loses the hydrogen ion to become NH3, that, as
tection [104]. it is non-ionized, freely crosses lipid blood-
Ultimately, renal function is best preserved in brain barrier and enters the CNS. Whether or
the perioperative period by minimizing nephro- not this is the precise mechanism, encephalopa-
toxic, surgical and circulatory perturbations, and thy worsens in the presence of alkalosis. Many
maintaining hemodynamic stability. patients with advanced liver disease have sec-
Unfortunately, major perturbations may occur ondary hyperaldosteronism, characterized by
despite the best attempts to prevent them, espe- hypokalemic a­lkalosis that can exacerbate
cially in emergency surgery or with complica- hepatic encephalopathy.
tions such as bleeding, major fluid shifts or Patients with acute encephalopathy should
infection. Moreover, given the underlying renal have elective procedures postponed until their
circulatory impairment that exists in advanced mental status returns to baseline. Precipitating
liver disease, acute deterioration in renal function factors of encephalopathy should be identified
may occur even after apparently minor periopera- and corrected. Metabolic alkalosis associated
tive events. with hypokalemia should be treated by careful
correction with potassium chloride. In severe sit-
uations dilute (0.1N) hydrochloric acid has been
Neurologic Dysfunction infused slowly via a central line [105]. Additional
treatment options include efforts to decrease
Hepatic Encephalopathy absorption of gut protein by oral lactulose,
Hepatic encephalopathy is the most important titrated to 3–4 soft stools per day or oral rifaxi-
neurologic complication of severe liver disease. min in patients that are intolerant of lactulose
The earliest stages—Grade 0 (minimal) and [106]. Protein restriction, although used in the
Grade 1 (mild)—of encephalopathy may be very management of encephalopathy, is not supported
difficult to detect without psychometric testing by clinical evidence and may complicate wound
because they manifest with signs as subtle as dif- healing in the already malnourished patient.
ficulty with driving, mood changes or a shortened
attention span. Grade 2 (moderate) encephalopa- Alcohol-Induced Cirrhosis
thy is more overt, with confusion, dysgraphia and and Encephalopathy
the appearance of asterixis (“flapping tremor”). It has been estimated that 20% of heavy drinkers
In Grade 3 (severe) encephalopathy the patient is develop alcoholic hepatitis and 25% develop cir-
stuporose but still arousable, and in Grade 4 the rhosis [107] The National Institute on Alcohol
patient is completely comatose. Abuse and Alcoholism has estimated that 44% of
404 K. Palmieri and R. N. Sladen

all deaths from liver disease in 2003 were attrib- further decreased by liver disease and through its
utable to alcohol [108]. When the patient with interaction with transferrin, contributes to iron
alcoholic liver disease presents for surgery, a his- overload [110].
tory of current or recent alcohol abuse indicates a First line treatment is a copper-chelating agent
very high risk of acute withdrawal and delirium such as D-penicillamine, that however can sup-
tremens in the perioperative period. Patients with press collagen production and impair postopera-
acute alcoholic intoxication should not be sub- tive wound healing [111] Discontinuation of the
jected to elective surgery because of sensitivity to medication is not recommended because this
all sedative agents, risk of aspiration and impaired may result in prompt hepatic decompensation
platelet aggregation [1]. and liver failure. It is suggested that in women
Wernicke’s encephalopathy, characterized by scheduled for Cesarean section, the
dementia, ataxia and ophthalmoplegia, is a com- D-penicillamine dose be decreased by 25–50% in
plication of chronic alcoholism induced by thia- the third trimester to promote postoperative
mine deficiency, and may benefit from its wound healing in [112]. This guideline could rea-
preoperative supplementation [109]. sonably be extended to other types of operative
intervention, or at least 2 weeks before and after
surgery. Neuropsychiatric involvement in
 reoperative Consideration for Rare
P Wilson’s disease may preclude a patient from
Liver Diseases providing informed consent.

Hereditary Hemochromatosis Autoimmune Hepatitis


Human hemochromatosis protein (HFE protein) Patients with chronic autoimmune hepatitis are
regulates circulating iron uptake by transferrin. treated with glucocorticoids, and so should be
Hereditary hemochromatosis is an autosomal considered for perioperative stress dose steroid
recessive disorder in which mutations of the HFE therapy.
gene cause increased intestinal iron absorption
and deposition in the liver, leading to cirrhosis
with an increased risk of hepatocellular carci-  nesthetic Planning in Patients
A
noma [110]. Iron is also deposited in the heart with Liver Disease
(cardiomyopathy, arrhythmias), pancreas (diabe-
tes), adrenals (hypoadrenalism), bones and joints Pharmacologic Considerations
(arthritis). Resistance to siderophilic microrgan-
isms (Vibrio, Listeria, Yersinia, Salmonella, No specific anesthetic drugs or techniques have
Klebsiella and E. coli) is impaired. Patients been shown to be superior in managing the
should be carefully screened for the presence of patient with significant liver disease. Obviously it
these manifestations and medically optimized is prudent to avoid any agent with known hepato-
prior to undergoing elective surgery. toxic or nephrotoxic effects. All anesthetic tech-
niques have the potential to decrease cardiac
Wilson’s Disease output and blood pressure and thereby decrease
Wilson’s disease is an autosomal recessive disor- hepatic blood flow. This may be exacerbated if
der caused by a defect in the gene that codes for splanchnic vasoconstriction is induced by hypo-
copper binding. This defect leads to defective volemia, stress or shock.
biliary excretion of copper and its subsequent Regional anesthesia may be safely used in the
accumulation in multiple organs, potentially absence of thrombocytopenia and if coagulation
leading to hepatic, neuropsychiatric, renal and studies (INR, aPTT) are within normal limits.
other dysfunction. The diagnosis is usually con- Local anesthesia may help to preserve hepatic
firmed by finding low serum levels of the copper-­ blood flow if blood pressure and cardiac out-
containing enzyme, ceruloplasmin, which is put are maintained. The common presence of
30  The Patient with Liver Disease Undergoing Non-hepatic Surgery 405

c­ oagulopathy, ascites and encephalopathy unfor- both drugs, laudanosine, may accumulate after
tunately limits its application in this population. high doses of continuous infusions and is associ-
Pharmacokinetic and pharmacodynamic ated in dogs with neurotoxicity including seizure
abnormalities are prominent in liver disease and activity. This effect has not been encountered in
all sedative and anesthetic medications must be humans or reported in patients [30] and there is
carefully titrated to the desired effect. The liver evidence that laudanosine may actually be neuro-
plays a central role in drug metabolism by con- protective in patients undergoing neurosurgery
verting fat-soluble active moieties to water-­ [113]. Neuromuscular blocking agents that are
soluble metabolites that can be excreted in the dependent on hepatic biotransformation such as
bile or urine. It does so through two distinct path- vecuronium or rocuronium should be used if at
ways: biotransformation via the CP450 enzyme all sparingly and with caution.
system, which is highly susceptible to liver
injury; and simple glucuronide conjugation, Volatile Anesthetics
which is more robust. For example, the elimina- All currently used volatile anesthetic agents
tion of midazolam, a benzodiazepine dependent (sevoflurane, isoflurane, desflurane) decrease
on biotransformation, is more readily affected by hepatic blood flow due to their effects on the cen-
liver dysfunction than its congener, lorazepam, tral circulation, but this can usually be overcome
which is rendered water soluble through simple with appropriate hemodynamic management.
glucuronide conjugation. Halothane should be avoided at all costs in
Liver disease alters drug pharmacokinetics not patients with liver disease. With the advent of
only because of impaired hepatic biotransforma- sevoflurane toward the end of the twentieth
tion or conjugation, but also because of an Century, halothane was largely discontinued in
increased volume of distribution. Thus, loading Western developed countries. However, because
dose requirements for certain drugs may be of its perceived cost-effectiveness, it is still
higher but emergence may be substantially widely used in low income countries [114]. Of all
delayed. This applies particularly to neuromus- the volatile anesthetics, halothane undergoes the
cular blocking agents such as vecuronium and greatest degree of metabolism: about 20%, com-
rocuronium, whose initial dosing is increased and pared with sevoflurane (5%), enflurane (2%), iso-
onset of action is delayed in patients with severe flurane and desflurane (both 0.2%). Postoperative
liver disease due to an increased volume of distri- liver dysfunction (POLD) is most often related to
bution, but whose recovery time may be pro- halothane anesthesia. Mild halothane hepatotox-
longed by impaired hepatic clearance. icity (Type 1 POLD) is very common, and
induced by reductive metabolites. It is of little
 euromuscular Blocking Agents
N clinical consequence and usually missed unless
The effects of succinylcholine may be slightly laboratory evidence of mild transaminitis is
prolonged in the patient with severe liver dys- looked for and found. Type 2 POLD, known as
function because of low levels of plasma cholin- halothane hepatitis or anesthesia-induced hepati-
esterase. However the prolongation is seldom tis (AIH) is a highly fatal condition of immunoal-
clinically important and should not preclude the lergic centrilobular necrosis associated with
use of succinylcholine for rapid sequence induc- re-exposure to halothane. The fulminant hepato-
tion (see below). toxicity appears to be a consequence of sensitiza-
Atracurium and cisatracurium are intermedi- tion of oxidative trifluoroacetate metabolites
ate neuromuscular blocking agents that undergo produced by the CP-450 2E1 system [115].
spontaneous pH-mediated breakdown in the Although there is a genetic predisposition and it
blood (Hoffman elimination) or nonspecific ester is more common in obese middle-aged women,
hydrolysis. Their elimination is independent of the most important precipitating factor is halo-
liver function and they are logical and safe agents thane re-exposure, especially within 1–2 weeks.
in a patient with liver disease. A metabolite of The co-existent use of drugs like acetaminophen
406 K. Palmieri and R. N. Sladen

that stimulate mixed function oxidases increases propofol must be used with caution as it depresses
the risk of AIH by increasing production of oxi- the circulation through inhibition of reflex tachy-
dative metabolites. The incidence has been esti- cardia and vasodilation. These effects may be
mated to be 1:35,000 cases of halothane exposure, particularly detrimental in a patient who is
but there have been very rare reports of hepatitis already hypotensive at baseline.
occurring with re-exposure to the newer volatile
agents. Dexmedetomidine
Dexmedetomidine is a highly selective alpha-2
Opioids agonist administered by continuous infusion. It
All opioids, including fentanyl, morphine, hydro- provides central sedation and sympatholysis, and
morphone and methadone, undergo hepatic bio- has an analgesia-sparing effect without inducing
transformation and/or glucuronide conjugation in respiratory depression. Its sympatholytic action
the liver and may accumulate in patients with may induce bradycardia and hypotension in sus-
significant liver dysfunction. Delayed emergence ceptible patients, which an important consider-
should be anticipated if they are used at standard ation in patients with ESLD and low systemic
doses. Opioids will have an enhanced vascular resistance. There is considerable evi-
­pharmacodynamic effect with even latent hepatic dence that it decreases the incidence of postop-
encephalopathy and should be dosed with great erative delirium. It is a useful preinduction agent
caution, especially in patients with an unpro- in highly agitated patients, or to facilitate awake
tected airway. intubation, and provides excellent conditions for
Remifentanil is a potent opioid that undergoes anesthetic emergence and tracheal extubation. It
rapid hydrolysis by esterases in the blood, and undergoes hepatic glucuronide conjugation to
has an elimination half-life of 8 min. As such its inactive metabolites, and as such has been con-
pharmacokinetic properties are independent of sidered to be relatively contraindicated in patients
liver function and remifentanil is ideally suited with severe liver dysfunction [116]. However, it
for administration by continuous infusion in has been demonstrated to be safe and without
patients with severe liver disease. However, the accumulation even when used at high doses for
same caveat regarding its pharmacodynamic prolonged periods of time in the ICU [117].
effects applies and it should be titrated to effect There is also experimental and clinical evidence
very cautiously. that dexmedetomidine may be protective against
hepatic ischemia-reperfusion injury [118].
Benzodiazepines
Benzodiazepines should be avoided in patients
with liver disease because of delayed elimination Immediate Preoperative Preparation
as well as their potential to unmask or exacerbate
hepatic encephalopathy and promote postopera- Patients with severe liver disease are very sensi-
tive delirium. This applies to both “long-acting” tive to the depressant effects of all sedatives and
(lorazepam) and “short-acting” (midazolam) premedication is best omitted except for aspira-
agents. tion prophylaxis. Small doses of IV sedation can
be given in the induction room or operating room
Propofol under direct observation in patients without sig-
Propofol is a potent induction and maintenance nificant ascites or encephalopathy. Proper moni-
agent that is rapidly cleared from the CNS toring of vital signs and the ability to take over
because of its high lipid solubility. This is the ventilation at any time must be assured. Because
most important determinant of its duration of these patients are so susceptible to infection spe-
action and even if its hepatic metabolism is cial attention must be given to universal precau-
slowed, it remains relatively short-acting in tions and scrupulous aseptic technique. At the
patients with advanced cirrhosis. Nonetheless same time all staff should be aware of the ­possible
30  The Patient with Liver Disease Undergoing Non-hepatic Surgery 407

danger to themselves of viral transmission, and The severity of the patient’s liver disease and
as a precaution uniform staff hepatitis B vaccina- the complexity of the planned surgical procedure
tion is recommended. should dictate the extent of invasive monitoring
and vascular access. Given the constant specter
Anesthetic Induction of coagulopathy and surgical bleeding, it is pru-
Hepatic encephalopathy places patients at risk to dent to place large bore intravenous access for all
hiccoughs, nausea and vomiting. Gastric empty- but the least invasive procedures. Similar consid-
ing is delayed in patients with severe liver disease eration should be given to arterial cannulation
and increases the risk of regurgitation and aspira- and monitoring.
tion during anesthetic induction. This risk is Central venous cannulation with or without
exacerbated by severe ascites with increased pulmonary artery catheterization can be helpful
abdominal pressure. Management of anesthetic when substantial fluid shifts are anticipated in the
induction should incorporate thorough preoxy- patient with cardiovascular and/or pulmonary
genation, generous fluid loading and aspiration morbidity or large volume transfusion is antici-
precautions. If the patient has an easy airway, pated. Transthoracic and transesophageal echo-
rapid sequence induction and intubation is cardiography are excellent tools for monitoring
­recommended. If the airway is difficult, awake cardiac filling and function intraoperatively,
fiberoptic intubation maybe facilitated by low although the former may be limited by the loca-
doses of a drug that does not depress ventilation tion of the surgical field and the latter by the pres-
such as dexmedetomidine should be considered. ence of esophageal varices and coagulopathy.

Intraoperative Monitoring  mergence and Postoperative Care


E
and Management Anesthetic emergence may be delayed and com-
Intraoperative liver injury may occur secondary plicated by vomiting, aspiration, hypotension,
to a diverse number of insults. These include respiratory depression and acute respiratory fail-
hypotension, hypoxemia, medications, blood loss ure. To reduce the risk of aspiration, patients
and transfusion, infection and the stress response should have their trachea extubated only when
to surgery. Delivery of blood and oxygen to the they are fully awake. A short period of postopera-
liver may be impaired by hemodynamic instabil- tive mechanical ventilation should always be con-
ity, surgical retraction of the abdomen, manipula- sidered to allow controlled emergence, avoid
tion of the liver, endogenous and exogenous reversal agents and facilitate the evaluation of
vasoconstrictors and anesthetic agents. neurologic and ventilatory function prior to tra-
Hypocarbia and hypercarbia decrease portal cheal extubation. Potential postoperative compli-
blood flow. The anesthesiologist should antici- cations include bleeding, oliguria, encephalopathy,
pate, attempt to prevent and vigorously treat acute respiratory failure, sepsis, wound dehis-
these factors. Intraoperative complications that cence and acute hepatic failure. The detailed post-
may occur as a consequence of severe liver dis- operative management of patients with liver
ease include hypoxemia (ascites, hepatopulmo- disease is addressed elsewhere in this book.
nary syndrome), bleeding (coagulopathy),
oliguria (hepatorenal syndrome) and hypoglyce-
mia. These complications should be anticipated References
and minimized by careful maintenance of intra-
operative systemic, hepatic and renal hemody-
1. Stoelting RK, Stephen F, editors. Anesthesia and
co-existing disease. 4th ed. Philadelphia: Churchill
namics. Blood glucose levels should be checked Livingstone, Elsevier Inc.; 2002.
frequently throughout the perioperative period,
2. del Olmo JA, Flor-Lorente B, Flor-Civera B,
with intravenous infusion of a glucose-­containing Rodriguez F, Serra MA, Escudero A, et al. Risk fac-
fluid to prevent hypoglycemia and appropriate tors for nonhepatic surgery in patients with cirrhosis.
World J Surg. 2003;27(6):647–52.
insulin dosing as needed.
408 K. Palmieri and R. N. Sladen

3. Ziser A, Plevak DJ, Wiesner RH, Rakela J, Offord lar carcinoma in cirrhotic patients: prognostic value
KP, Brown DL. Morbidity and mortality in cir- of preoperative portal pressure. Gastroenterology.
rhotic patients undergoing anesthesia and surgery. 1996;111(4):1018–22.
Anesthesiology. 1999;90(1):42–53. 19. Azoulay D, Buabse F, Damiano I, Smail A, Ichai P,
4. Teh SH, Nagorney DM, Stevens SR, Offord KP, Dannaoui M, et al. Neoadjuvant transjugular intra-
Therneau TM, Plevak DJ, et al. Risk factors for hepatic portosystemic shunt: a solution for extra-
mortality after surgery in patients with cirrhosis. hepatic abdominal operation in cirrhotic patients
Gastroenterology. 2007;132(4):1261–9. with severe portal hypertension. J Am Coll Surg.
5. Friedman LS. The risk of surgery in patients with 2001;193(1):46–51.
liver disease. Hepatology. 1999;29(6):1617–23. 20. Chalasani N, Kahi C, Francois F, Pinto A,
6. Rice HE, O’Keefe GE, Helton WS, Johansen Marathe A, Bini EJ, et al. Model for end-stage
K. Morbid prognostic features in patients with liver disease (MELD) for predicting mortality in
chronic liver failure undergoing nonhepatic surgery. patients with acute variceal bleeding. Hepatology.
Arch Surg. 1997;132(8):880–4. discussion 4–5 2002;35(5):1282–4.
7. Csikesz NG, Nguyen LN, Tseng JF, Shah 21. Sheth M, Riggs M, Patel T. Utility of the Mayo
SA. Nationwide volume and mortality after elec- End-Stage Liver Disease (MELD) score in assessing
tive surgery in cirrhotic patients. J Am Coll Surg. prognosis of patients with alcoholic hepatitis. BMC
2009;208(1):96–103. Gastroenterol. 2002;2:2.
8. Delis S, Bakoyiannis A, Madariaga J, Bramis J, 22. Kamath PS, Wiesner RH, Malinchoc M, Kremers W,
Tassopoulos N, Dervenis C. Laparoscopic chole- Therneau TM, Kosberg CL, et al. A model to pre-
cystectomy in cirrhotic patients: the value of MELD dict survival in patients with end-stage liver disease.
score and Child-Pugh classification in predicting Hepatology. 2001;33(2):464–70.
outcome. Surg Endosc. 2010;24(2):407–12. 23. Said A, Williams J, Holden J, Remington P, Gangnon
9. Miller RD, editor. Miller’s anesthesia. 7th ed. R, Musat A, et al. Model for end stage liver disease
Philadelphia: Churchill Livingstone, Elsevier Inc.; score predicts mortality across a broad spectrum of
2010. liver disease. J Hepatol. 2004;40(6):897–903.
10. Douard R, Lentschener C, Ozier Y, Dousset B. Operative 24. Farnsworth N, Fagan SP, Berger DH, Awad
risks of digestive surgery in cirrhotic patients. SS. Child-Turcotte-Pugh versus MELD score as
Gastroenterol Clin Biol. 2009;33(6–7):555–64. a predictor of outcome after elective and emer-
11. Kim TH, Um SH, Yim SY, Seo YS, Yim HJ, Jeen gent surgery in cirrhotic patients. Am J Surg.
YT, et al. The risk of perioperative adverse events 2004;188(5):580–3.
in patients with chronic liver disease. Liver Int. 25. Perkins L, Jeffries M, Patel T. Utility of preoperative
2015;35(3):713–23. scores for predicting morbidity after cholecystec-
12. Neeff HP, Streule GC, Drognitz O, Tittelbach-­ tomy in patients with cirrhosis. Clin Gastroenterol
Helmrich D, Spangenberg HC, Hopt UT, et al. Early Hepatol. 2004;2(12):1123–8.
mortality and long-term survival after abdominal 26. Befeler AS, Palmer DE, Hoffman M, Longo W,
surgery in patients with liver cirrhosis. Surgery. Solomon H, Di Bisceglie AM. The safety of intra-­
2014;155(4):623–32. abdominal surgery in patients with cirrhosis: model
13. Martin P. Perioperative considerations for patients for end-stage liver disease score is superior to Child-­
with liver disease. Cleve Clin J Med. 2009;76(Suppl Turcotte-­Pugh classification in predicting outcome.
4):S93–7. Arch Surg. 2005;140(7):650–4. discussion 5
14. Narr BJ, Hansen TR, Warner MA. Preoperative 27. Northup PG, Wanamaker RC, Lee VD, Adams
laboratory screening in healthy Mayo patients: cost-­ RB, Berg CL. Model for End-Stage Liver
effective elimination of tests and unchanged out- Disease (MELD) predicts nontransplant surgi-
comes. Mayo Clin Proc. 1991;66(2):155–9. cal mortality in patients with cirrhosis. Ann Surg.
15. Garrison RN, Cryer HM, Howard DA, Polk HC Jr. 2005;242(2):244–51.
Clarification of risk factors for abdominal opera- 28. Hanje AJ, Patel T. Preoperative evaluation of patients
tions in patients with hepatic cirrhosis. Ann Surg. with liver disease. Nat Clin Pract Gastroenterol
1984;199(6):648–55. Hepatol. 2007;4(5):266–76.
16. Mansour A, Watson W, Shayani V, Pickleman 29. Suman A, Barnes DS, Zein NN, Levinthal GN,
J. Abdominal operations in patients with cir- Connor JT, Carey WD. Predicting outcome after car-
rhosis: still a major surgical challenge. Surgery. diac surgery in patients with cirrhosis: a comparison
1997;122(4):730–5. discussion 5–6 of Child-Pugh and MELD scores. Clin Gastroenterol
17. Telem DA, Schiano T, Goldstone R, Han DK, Buch Hepatol. 2004;2(8):719–23.
KE, Chin EH, et al. Factors that predict outcome of 30. Sladen RN. Anesthetic concerns for the patient with
abdominal operations in patients with advanced cir- liver disease. Anesth Analg. 2008;106(4):1–7.
rhosis. Clin Gastroenterol Hepatol. 2010;8(5):451– 31. Harville DD, Summerskill WH. Surgery in acute hep-
7. quiz e58 atitis. Causes and effects. JAMA. 1963;184:257–61.
18. Bruix J, Castells A, Bosch J, Feu F, Fuster J, Garcia-­ 32. Greenwood SM, Leffler CT, Minkowitz S. The
Pagan JC, et al. Surgical resection of hepatocellu- increased mortality rate of open liver biopsy
30  The Patient with Liver Disease Undergoing Non-hepatic Surgery 409

in alcoholic hepatitis. Surg Gynecol Obstet. 48. de Goede B, Klitsie PJ, Hagen SM, van Kempen BJ,
1972;134(4):600–4. Spronk S, Metselaar HJ, et al. Meta-analysis of lapa-
33. Propst A, Propst T, Zangerl G, Ofner D, Judmaier G, roscopic versus open cholecystectomy for patients
Vogel W. Prognosis and life expectancy in chronic with liver cirrhosis and symptomatic cholecystoli-
liver disease. Dig Dis Sci. 1995;40(8):1805–15. thiasis. Br J Surg. 2013;100(2):209–16.
34. Gines P, Quintero E, Arroyo V, Teres J, Bruguera 49. Puggioni A, Wong LL. A metaanalysis of laparo-
M, Rimola A, et al. Compensated cirrhosis: natu- scopic cholecystectomy in patients with cirrhosis. J
ral history and prognostic factors. Hepatology. Am Coll Surg. 2003;197(6):921–6.
1987;7(1):122–8. 50. Ng M, Fleming T, Robinson M, Thomson B, Graetz
35. Neeff H, Mariaskin D, Spangenberg HC, Hopt UT, N, Margono C, et al. Global, regional, and national
Makowiec F. Perioperative mortality after non-­ prevalence of overweight and obesity in children and
hepatic general surgery in patients with liver cir- adults during 1980–2013: a systematic analysis for
rhosis: an analysis of 138 operations in the 2000s the Global Burden of Disease Study 2013. Lancet.
using child and MELD scores. J Gastrointest Surg. 2014;384(9945):766–81.
2011;15:1–11. 51. Harrison SA, Di Bisceglie AM. Advances in the
36. Demetriades D, Constantinou C, Salim A, Velmahos understanding and treatment of nonalcoholic fatty
G, Rhee P, Chan L. Liver cirrhosis in patients under- liver disease. Drugs. 2003;63(22):2379–94.
going laparotomy for trauma: effect on outcomes. J 52. Ekstedt M, Franzen LE, Mathiesen UL, Thorelius
Am Coll Surg. 2004;199(4):538–42. L, Holmqvist M, Bodemar G, et al. Long-term fol-
37. Nguyen GC, Correia AJ, Thuluvath PJ. The low-­up of patients with NAFLD and elevated liver
impact of cirrhosis and portal hypertension on enzymes. Hepatology. 2006;44(4):865–73.
mortality following colorectal surgery: a nation- 53. Machado M, Marques-Vidal P, Cortez-Pinto
wide, population-based study. Dis Colon Rectum. H. Hepatic histology in obese patients undergoing
2009;52(8):1367–74. bariatric surgery. J Hepatol. 2006;45(4):600–6.
38. Ghaferi AA, Mathur AK, Sonnenday CJ, Dimick 54. Wolter S, Dupree A, Coelius C, El Gammal A,
JB. Adverse outcomes in patients with chronic liver Kluwe J, Sauer N, et al. Influence of liver disease
disease undergoing colorectal surgery. Ann Surg. on perioperative outcome after bariatric surgery
2010;252(2):345–50. in a northern german cohort. Obes Surg. 2017
39. Gelman S. General anesthesia and hepatic circula- Jan;27(1):90–95.
tion. Can J Physiol Pharmacol. 1987;65(8):1762–79. 55. Jan A, Narwaria M, Mahawar KK. A systematic
40. Sabbagh C, Fuks D, Regimbeau JM. Non-hepatic review of bariatric surgery in patients with liver cir-
gastrointestinal surgery in patients with cirrhosis. J rhosis. Obes Surg. 2015;25(8):1518–26.
Visc Surg. 2014;151(3):203–11. 56. Barker KB, Palekar NA, Bowers SP, Goldberg JE,
41. Cobb WS, Heniford BT, Burns JM, Carbonell AM, Pulcini JP, Harrison SA. Non-alcoholic steatohepati-
Matthews BD, Kercher KW. Cirrhosis is not a con- tis: effect of Roux-en-Y gastric bypass surgery. Am J
traindication to laparoscopic surgery. Surg Endosc. Gastroenterol. 2006;101(2):368–73.
2005;19(3):418–23. 57. Furuya CK Jr, de Oliveira CP, de Mello ES, Faintuch
42. Poggio JL, Rowland CM, Gores GJ, Nagorney DM, J, Raskovski A, Matsuda M, et al. Effects of bariatric
Donohue JH. A comparison of laparoscopic and surgery on nonalcoholic fatty liver disease: prelimi-
open cholecystectomy in patients with compen- nary findings after 2 years. J Gastroenterol Hepatol.
sated cirrhosis and symptomatic gallstone disease. 2007;22(4):510–4.
Surgery. 2000;127(4):405–11. 58. Liu X, Lazenby AJ, Clements RH, Jhala N,
43. Aranha GV, Sontag SJ, Greenlee Abrams GA. Resolution of nonalcoholic steato-
HB. Cholecystectomy in cirrhotic patients: a formi- hepatits after gastric bypass surgery. Obes Surg.
dable operation. Am J Surg. 1982;143(1):55–60. 2007;17(4):486–92.
44. Bloch RS, Allaben RD, Walt AJ. Cholecystectomy 59. Hsieh WC, Chen PC, Corciova FC, Tinica G. Liver
in patients with cirrhosis. A surgical challenge. Arch dysfunction as an important predicting risk factor
Surg. 1985;120(6):669–72. in patients undergoing cardiac surgery: a system-
45. Sleeman D, Namias N, Levi D, Ward FC, atic review and meta-analysis. Int J Clin Exp Med.
Vozenilek J, Silva R, et al. Laparoscopic chole- 2015;8(11):20712–21.
cystectomy in cirrhotic patients. J Am Coll Surg. 60. Hessel EA 2nd. Abdominal organ injury after car-
1998;187(4):400–3. diac surgery. Semin Cardiothorac Vasc Anesth.
46. Fernandes NF, Schwesinger WH, Hilsenbeck SG, 2004;8(3):243–63.
Gross GW, Bay MK, Sirinek KR, et al. Laparoscopic 61. Kaplan M, Cimen S, Kut MS, Demirtas MM. Cardiac
cholecystectomy and cirrhosis: a case-control study operations for patients with chronic liver disease.
of outcomes. Liver Transpl. 2000;6(3):340–4. Heart Surg Forum. 2002;5(1):60–5.
47. D’Albuquerque LAC, de Miranda MP, Genzini T, 62. Klemperer JD, Ko W, Krieger KH, Connolly M,
Copstein JL, de Oliveira e Silva A. Laparoscopic Rosengart TK, Altorki NK, et al. Cardiac opera-
cholecystectomy in cirrhotic patients. Surg Laparosc tions in patients with cirrhosis. Ann Thorac Surg.
Endosc. 1995;5(4):272–6. 1998;65(1):85–7.
410 K. Palmieri and R. N. Sladen

63. Vanhuyse F, Maureira P, Portocarrero E, Laurent N, in patients with liver cirrhosis. Ann Thorac Surg.
Lekehal M, Carteaux JP, et al. Cardiac surgery in cir- 2006;82(3):1027–32.
rhotic patients: results and evaluation of risk factors. 78. Weeder PD, Porte RJ, Lisman T. Hemostasis in
Eur J Cardiothorac Surg. 2012;42(2):293–9. liver disease: implications of new concepts for
64. Lopez-Delgado JC, Esteve F, Javierre C, Ventura perioperative management. Transfus Med Rev.
JL, Manez R, Farrero E, et al. Influence of cirrho- 2014;28(3):107–13.
sis in cardiac surgery outcomes. World J Hepatol. 79. Lisman T, Bongers TN, Adelmeijer J, Janssen HL,
2015;7(5):753–60. de Maat MP, de Groot PG, et al. Elevated levels of
65. Filsoufi F, Salzberg SP, Rahmanian PB, Schiano TD, von Willebrand Factor in cirrhosis support plate-
Elsiesy H, Squire A, et al. Early and late outcome of let adhesion despite reduced functional capacity.
cardiac surgery in patients with liver cirrhosis. Liver Hepatology. 2006;44(1):53–61.
Transpl. 2007;13(7):990–5. 80. Lorenz R, Kienast J, Otto U, Egger K, Kiehl M,
66. Ailawadi G, Lapar DJ, Swenson BR, Siefert SA, Lau Schreiter D, et al. Efficacy and safety of a prothrom-
C, Kern JA, et al. Model for end-stage liver disease bin complex concentrate with two virus-inactivation
predicts mortality for tricuspid valve surgery. Ann steps in patients with severe liver damage. Eur J
Thorac Surg. 2009;87(5):1460–7. discussion 7–8 Gastroenterol Hepatol. 2003;15(1):15–20.
67. Modi A, Vohra HA, Barlow CW. Do patients with 81. Bernstein DE, Jeffers L, Erhardtsen E, Reddy
liver cirrhosis undergoing cardiac surgery have KR, Glazer S, Squiban P, et al. Recombinant fac-
acceptable outcomes? Interact Cardiovasc Thorac tor VIIa corrects prothrombin time in cirrhotic
Surg. 2010;11(5):630–4. patients: a preliminary study. Gastroenterology.
68. Thielmann M, Mechmet A, Neuhauser M, Wendt 1997;113(6):1930–7.
D, Tossios P, Canbay A, et al. Risk prediction and 82. Jeffers L, Chalasani N, Balart L, Pyrsopoulos N,
outcomes in patients with liver cirrhosis undergo- Erhardtsen E. Safety and efficacy of recombinant
ing open-heart surgery. Eur J Cardiothorac Surg. factor VIIa in patients with liver disease undergo-
2010;38(5):592–9. ing laparoscopic liver biopsy. Gastroenterology.
69. Hayashida N, Shoujima T, Teshima H, Yokokura Y, 2002;123(1):118–26.
Takagi K, Tomoeda H, et al. Clinical outcome after 83. Levy JH, Fingerhut A, Brott T, Langbakke IH,
cardiac operations in patients with cirrhosis. Ann Erhardtsen E, Porte RJ. Recombinant factor VIIa
Thorac Surg. 2004;77(2):500–5. in patients with coagulopathy secondary to anti-
70. Yamamoto T, Takazawa K, Hariya A, Ishikawa N, coagulant therapy, cirrhosis, or severe traumatic
Dohi S, Matsushita S. Off-pump coronary artery injury: review of safety profile. Transfusion.
bypass grafting in a patient with liver cirrhosis. Jpn J 2006;46(6):919–33.
Thorac Cardiovasc Surg. 2002;50(12):526–9. 84. Shao YF, Yang JM, Chau GY, Sirivatanauksorn
71. Tang AT, Karski J, Cusimano RJ. Successful off-­ Y, Zhong SX, Erhardtsen E, et al. Safety and
pump pericardiectomy and coronary artery bypass in ­hemostatic effect of recombinant activated fac-
liver cirrhosis. J Card Surg. 2005;20(3):284–6. tor VII in cirrhotic patients undergoing partial
72. Carr C, Desai J. OPCAB surgery in a cirrhotic hepa- hepatectomy: a multicenter, randomized, dou-
tocellular carcinoma patient awaiting liver trans- ble-blind, placebo-­ controlled trial. Am J Surg.
plant. Ann Thorac Surg. 2004;78(4):1460–2. 2006;191(2):245–9.
73. Ben Ari A, Elinav E, Elami A, Matot I. Off-pump 85. Tinmouth A. Evidence for a rationale use of frozen
coronary artery bypass grafting in a patient with plasma for the treatment and prevention of bleeding.
child class C liver cirrhosis awaiting liver transplan- Transfus Apher Sci. 2012;46(3):293–8.
tation. Br J Anaesth. 2006;97(4):468–72. 86. Stanworth SJ, Hyde CJ, Murphy MF. Evidence for
74. Gopaldas RR, Chu D, Cornwell LD, Dao TK, indications of fresh frozen plasma. Transfus Clin
Lemaire SA, Coselli JS, et al. Cirrhosis as a modera- Biol. 2007;14(6):551–6.
tor of outcomes in coronary artery bypass grafting 87. Tripodi A, Anstee QM, Sogaard KK, Primignani
and off-pump coronary artery bypass operations: a M, Valla DC. Hypercoagulability in cirrhosis:
12-year population-based study. Ann Thorac Surg. causes and consequences. J Thromb Haemost.
2013;96(4):1310–5. 2011;9(9):1713–23.
75. Greason KL, Mathew V, Wiesner RH, Suri RM, 88. Buresi M, Hull R, Coffin CS. Venous thromboem-
Rihal CS. Transcatheter aortic valve replace- bolism in cirrhosis: a review of the literature. Can J
ment in patients with cirrhosis. J Card Surg. Gastroenterol. 2012;26(12):905–8.
2013;28(5):492–5. 89. Senzolo M, Sartori MT, Lisman T. Should we
76. Thakkar B, Patel A, Mohamad B, Patel NJ, Bhatt P, give thromboprophylaxis to patients with liver
Bhimani R, et al. Transcatheter aortic valve replace- cirrhosis and coagulopathy? HPB (Oxford).
ment versus surgical aortic valve replacement in 2009;11(6):459–64.
patients with cirrhosis. Catheter Cardiovasc Interv. 90. Aldenkortt F, Aldenkortt M, Caviezel L, Waeber
2016;87(5):955–62. JL, Weber A, Schiffer E. Portopulmonary hyper-
77. Iwasaki A, Shirakusa T, Okabayashi K, Inutsuka K, tension and hepatopulmonary syndrome. World J
Yoneda S, Yamamoto S, et al. Lung cancer surgery Gastroenterol. 2014;20(25):8072–81.
30  The Patient with Liver Disease Undergoing Non-hepatic Surgery 411

91. Baikati K, Le DL, Jabbour II, Singhal S, 105. Brimioulle S, Berre J, Dufaye P, Vincent JL, Degaute
Anand S. Hepatic hydrothorax. Am J Ther. JP, Kahn RJ. Hydrochloric acid infusion for treat-
2014;21(1):43–51. ment of metabolic alkalosis associated with respira-
92. Keegan MT, Plevak DJ. Preoperative assessment of tory acidosis. Crit Care Med. 1989;17(3):232–6.
the patient with liver disease. Am J Gastroenterol. 106. Mas A, Rodes J, Sunyer L, Rodrigo L, Planas R,
2005;100(9):2116–27. Vargas V, et al. Comparison of rifaximin and lac-
93. Hopkins WE, Waggoner AD, Barzilai B. Frequency titol in the treatment of acute hepatic encepha-
and significance of intrapulmonary right-to-left lopathy: results of a randomized, double-blind,
shunting in end-stage hepatic disease. Am J Cardiol. double-dummy, controlled clinical trial. J Hepatol.
1992;70(4):516–9. 2003;38(1):51–8.
94. Stoller JK, Lange PA, Westveer MK, Carey WD, 107. Grant BF, Dufour MC, Harford TC. Epidemiology
Vogt D, Henderson JM. Prevalence and reversibil- of alcoholic liver disease. Semin Liver Dis.
ity of the hepatopulmonary syndrome after liver 1988;8(1):12–25.
transplantation. The Cleveland Clinic experience.
­ 108. Yoon YH, Yi HY. (2006) Surveillance report #75:
West J Med. 1995;163(2):133–8. liver cirrhosis mortality in the United States, 1970–
95. Castro M, Krowka MJ. Hepatopulmonary syndrome. 2003. Arlington, VA: National Institute on Alcohol
A pulmonary vascular complication of liver disease. Abuse and Alcoholism (Alcohol Epidemiologic
Clin Chest Med. 1996;17(1):35–48. Data System).
96. Krowka MJ, Cartin-Ceba R. Portopulmonary hyper- 109. Wiklund RA. Preoperative preparation of patients
tension: formidable dual threat versus hopeful dual with advanced liver disease. Crit Care Med.
therapy. Liver Transpl. 2014;20(6):635–6. 2004;32(4 Suppl):S106–15.
97. Goldberg DS, Batra S, Sahay S, Kawut SM, Fallon 110. Heidelbaugh JJ, Bruderly M. Cirrhosis and chronic
MB. MELD exceptions for portopulmonary hyper- liver failure: part I. Diagnosis and evaluation. Am
tension: current policy and future implementation. Fam Physician. 2006;74(5):756–62.
Am J Transplant. 2014;14(9):2081–7. 111. Ansell BM, Moran H, Arden GP. Penicillamine and
98. Kramer L, Horl WH. Hepatorenal syndrome. Semin wound healing in rheumatoid arthritis. Proc R Soc
Nephrol. 2002;22(4):290–301. Med. 1977;70(Suppl 3):75–7.
99. Sagi SV, Mittal S, Kasturi KS, Sood GK. Terlipressin 112. Roberts EA, Schilsky ML. A practice guideline on
therapy for reversal of type 1 hepatorenal syndrome: Wilson disease. Hepatology. 2003;37(6):1475–92.
a meta-analysis of randomized controlled trials. J 113. Fodale V, Santamaria LB. The possible neuropro-
Gastroenterol Hepatol. 2010;25(5):880–5. tective effect of laudanosine, an atracurium and
100. Boyer TD, Sanyal AJ, Garcia-Tsao G, Regenstein F, cisatracurium metabolite. Acta Anaesthesiol Scand.
Rossaro L, Appenrodt B, et al. Impact of liver trans- 2003;47(6):780. 780−1
plantation on survival of patients treated for type 114. Mahboobi N, Esmaeili S, Safari S, Habibollahi P,
1 hepatorenal syndrome (HRS-1). Liver Transpl. Dabbagh A, Alavian SM. Halothane: how should
2011;17:1328–32. it be used in a developing country? East Mediterr
101. Sort P, Navasa M, Arroyo V, Aldeguer X, Planas R, Health J. 2012;18(2):159–64.
Ruiz-del-Arbol L, et al. Effect of intravenous albu- 115. Kenna JG. Immunoallergic drug-induced hepatitis:
min on renal impairment and mortality in patients lessons from halothane. J Hepatol. 1997;26(Suppl
with cirrhosis and spontaneous bacterial peritonitis. 1):5–12.
N Engl J Med. 1999;341(6):403–9. 116. Shehabi Y, Nakae H, Hammond N, Bass F,

102. Singh V, Dheerendra PC, Singh B, Nain CK, Chawla Nicholson L, Chen J. The effect of dexmedeto-
D, Sharma N, et al. Midodrine versus albumin in the midine on agitation during weaning of mechani-
prevention of paracentesis-induced circulatory dys- cal ventilation in critically ill patients. Anaesth
function in cirrhotics: a randomized pilot study. Am Intensive Care. 2010;38(1):82–90.
J Gastroenterol. 2008;103(6):1399–405. 117. Iirola T, Aantaa R, Laitio R, Kentala E, Lahtinen M,
103. Cahill CJ, Pain JA, Bailey ME. Bile salts, endotoxin Wighton A, et al. Pharmacokinetics of prolonged
and renal function in obstructive jaundice. Surg infusion of high-dose dexmedetomidine in critically
Gynecol Obstet. 1987;165(6):519–22. ill patients. Crit Care. 2011;15(5):R257.
104. Pain JA, Cahill CJ, Gilbert JM, Johnson CD, 118. Wang ZX, Huang CY, Hua YP, Huang WQ,

Trapnell JE, Bailey ME. Prevention of postoperative Deng LH, Liu KX. Dexmedetomidine reduces
renal dysfunction in patients with obstructive jaun- intestinal and hepatic injury after hepatectomy
dice: a multicentre study of bile salts and lactulose. with inflow occlusion under general anaesthe-
Br J Surg. 1991;78(4):467–9. sia: a randomized controlled trial. Br J Anaesth.
2014;112(6):1055–64.
Part IV
Critical Care Medicine for Liver
Transplantation
Routine Postoperative Care After
Liver Transplantation 31
Jonathan Hastie and Vivek K. Moitra

Keywords complications affect ICU length of stay


Electrolytes · Hyperglycemia · Coagulation · (Fig. 31.2). Systems processes, such as care bun-
Hemodynamic monitoring · Ventilation · dles, clinical pathways, and enhanced communi-
Sedation cation tools impact patient outcomes and length
of stay [3–5].
A multidisciplinary team of intensivists, hepa-
tologists, and transplant surgeons should manage
Introduction liver transplant patients. Consultations are fre-
quently requested with specialists in cardiology,
Routine care immediately after liver transplanta- pulmonary medicine, infectious diseases and
tion includes in most cases and institutions nephrology for patients with comorbidities or
admission to the intensive care unit (ICU) for when complications arise. This chapter reviews
monitoring of organ systems, i­nitiation of immu- the routine management of the liver transplanta-
nosuppression, evaluation of graft function and tion patient in the ICU.
management of perioperative physiological
changes, including coagulopathy and hemody-
namic effects (Fig. 31.1). Although some institu-
tions may bypass the ICU for select patients, I nitial Organ Response to Liver
most c­ enters admit all liver transplant recipients Transplantation
to the intensive care unit, for even as short a dura-
tion as 24 h [1, 2]. Preexisting conditions, intra- Cardiovascular Response
operative factors and intra- and post-operative
The cardiovascular system of patients with end-­
stage liver disease mimicsthe hyperdynamic cir-
J. Hastie, MD
Department of Anesthesiology, Cardiothoracic
culatory changes of patients with sepsis.
Intensive Care Unit, Columbia University Medical Tachycardia, elevated cardiac output, low arterial
Center, New York, NY, USA blood pressure, and low systemic vascular resis-
V. K. Moitra, MD (*) tance are characteristically observed [6].
Department of Anesthesiology, Surgical Intensive Hypotension after liver transplantation has mul-
Care Unit and Cardiothoracic Intensive Care Unit, tiple etiologies (Fig. 31.3). Although patients
Columbia University, College of Physicians and
Surgeons, New York, NY, USA
with robust cardiovascular reserve are best able
e-mail: vm2161@cumc.columbia.edu to tolerate liver transplantation, this procedure

© Springer International Publishing AG, part of Springer Nature 2018 415


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_31
416 J. Hastie and V. K. Moitra

Fig. 31.1 Postoperative
care of the liver ICU Care
transplant recipient
Hemodynamics
Operating Room Graft & Organ Function Post-ICU Care
Coagulopathy
Immunosuppression

Transplant Surgeon
Hepatologist
Intensivist

Neurological

Hepatic Encephalopathy
Cerebral Edema
Cardiovascular Respiratory

Vasodilated State Hepatic Hydrothorax


Hyperdynamic Cardiac Function Acute Lung Injury
Drug-induced Hypertension Aspiration Risk

Liver Transplant

Metabolic Renal
Hyponatremia Hepatorenal Syndrome
Hypomagnesemia Nephrotoxic Drugs
Hyperglycemia

Massive Transfusion Hematologic


Hypothermia Disseminated Intravascular Coagulation
Lung Injury Decreased Synthesis
Electrolyte Disturbances Platelet Dysfunction

Fig. 31.2  Organ systems and how they are affected by liver disease

Hypotension after Liver Transplant

Hypovolemia Myocardial Dysfunction Vasodilation

Surgical Bleeding
Baseline Infiltrative Disease Infection
Under-resuscitation
Ischemic Cardiomyopathy Inflammatory Response
New Dilated Cardiomyopathy Graft Failure
Postreperfusion Syndrome

Fig. 31.3  Differential diagnosis of hypotension after liver transplantation


31  Routine Postoperative Care After Liver Transplantation 417

has been reported in patients in patients with cor- develop after transplantation [10]. This myocar-
onary artery disease [7] as well as in patients with dial depression may require therapy with inotro-
cardiomyopathies secondary to alcoholic liver pic agents and diuretics, and it is often reversible.
disease, amyloidosis, and hemochromatosis. Inadequate support for impaired contractility will
Vasodilation is common after liver transplan- cause elevations of central venous pressures and
tation. After unclamping of the portal vein desat- hepatic congestion. Hepatic congestion, in turn,
urated blood from the obstructed portal increases portal venous pressures and reduces
circulation is released into the systemic circula- enteral perfusion pressures, causing bacterial
tion, carrying with it potassium, protons, cold translocation, inflammation, and hypotension
components, and inflammatory mediators such as and a vicious cycle may ensue.
interleukin-6 and tumor necrosis factor alpha. Chronic hypertension, inadequate pain con-
These mediators decrease systemic vascular trol, volume overload, hypoglycemia, immuno-
resistance and impair myocardial contractility. A suppression with calcineurin inhibitors and the
30% decrease in mean arterial blood pressure onset of cerebral edema can cause postoperative
sustained for more than 1 min during the first hypertension. Calcium channel blockers such as
5 min after reperfusion is commonly defined as amlodipine are often used to manage cyclospo-
postreperfusion syndrome [8]. This postreperfu- rine- or tacrolimus-induced hypertension.
sion hypotension should resolve by the time the Atrial fibrillation is the most common cardiac
recipient arrives in the ICU unless there is some arrhythmias after liver transplantation [11], and
primary graft dysfunction. Causes of vasodila- predisposing factors include abnormal potassium
tion in the ICU include liver failure, infection, and magnesium levels, malpositioned central
and an inflammatory response to surgery. The venous catheters, and right atrial stretch from
­intraoperative use of steroids is associated with a fluid shifts. Immediate biphasic synchronized
decrease in postoperative vasopressor require- cardioversion is performed in patients with
ments [9]. hemodynamically unstable atrial fibrillation,
Management of hypotension from vasodila- while beta-blockers and calcium channel block-
tion includes administration of vasocontricting ers can be used to manage atrial fibrillation in the
agents such as norepinephrine or vasopressin, stable patient. Administration of non-­
fluid resuscitation to expand the circulating vol- dihydropyridine calcium channel blockers such
ume, and determination of the underlying cause as diltiazem or verapamil should be avoided,
of vasodilation. Aggressive fluid resuscitation because these medications increase levels of cal-
without assessment of fluid responsiveness cineurin inhibitor immunosuppression via inhibi-
should be avoided. Administration of excessive tion of the cytochrome P450 system. Short-term
intravascular fluid to a non-fluid-responsive use of amiodarone is useful in treating atrial
patient increases cardiac filling pressures, which fibrillation, however the potential for hepatic tox-
can impair graft function via hepatic congestion, icity has limited its long-term use in patients after
and can cause pulmonary edema requiring tra- liver transplantation.
cheal reintubation.
Cirrhotic patients may have a supra-normal
cardiac output at baseline from significantly Pulmonary Response
decreased peripheral vascular resistance. When
the postoperative systemic vascular resistance Hepatic hydrothorax, hepatopulmonary syn-
normalizes, the patient’s circulatory system may drome, underlying chronic pulmonary disease
be challenged, and a reduction in ejection f­ raction [12], and occasionally acute respiratory distress
and cardiac output may follow. Echocardiography syndrome (ARDS) [13, 14] can cause postopera-
may show impairment of myocardial func- tive hypoxemia and respiratory failure in patients
tion similar to that found in s­eptic patients. after liver transplantation. Restrictive fluid
Consequently, dilated cardiomyopathy may administration in the operating room decreases
418 J. Hastie and V. K. Moitra

postoperative hypoxemia [15]. Atelectasis from lose administration, drainage of ascites and
immobility, splinting, and the compressive effects diuretic therapy. Additional risk factors include
of ascites is common. Ascitic fluid can enter the female sex, weight > 100 kg, pre-existing diabe-
pleural space through small channels in the dia- tes [16], and severity of preoperative liver dis-
phragm to cause a hepatic hydrothorax that usu- ease. As cirrhosis progresses, a reduction in
ally predominates on the right side. Muscle systemic vascular resistance causes compensa-
wasting and intra-abdominal hypertension from tory activation of the renin-angiotensin and sym-
ascites increase the work of breathing. Resolution pathetic nervous systems, which leads to ascites,
of hepatopulmonary syndrome after transplant is edema, intra-renal vasoconstriction and renal
possible but may take months. Postoperative hypoperfusion. Hepatorenal syndrome (HRS) is
ARDS may be a result of direct surgical insult, caused by functional renal vasoconstriction in
generalized inflammatory processes, intraopera- response to splanchnic arterial vasodilation [19].
tive aspiration, or transfusion of blood products. Postoperative renal function generally correlates
These conditions cause noncardiogenic pulmo- with preoperative glomerular filtration rates [19];
nary edema by increasing the permeability of the however in patients with cirrhosis and HRS resis-
alveolar capillary membrane, creating a capillary tant to diuretics, renal function may improve
leak syndrome with exudation of fluid and pro- after transplantation.
tein into the alveolar space. In its most dramatic Following liver transplant, the incidence of
form, ARDS presents as the acute onset of a mas- stage II to III acute kidney injury by Acute
sive outpouring of proteinaceous fluid from the Kidney Injury Network (AKIN) definitions
endotracheal tube. Noncardiogenic pulmonary ranges from 9 to 48% [17, 20]. Intraoperative fac-
edema is characterized by normal or low left tors such as hypovolemia, hypotension, and
atrial or pulmonary artery wedge pressures and a increased renal venous pressure during the anhe-
high protein concentration in the edema fluid patic phase may all impact renal function. Initial
(albumin concentration 90% or greater than that serum creatinine level on arrival to the ICU may
of serum albumin); these distinguish it from pul- be “falsely low”, reflecting dilution from blood
monary edema from heart failure. loss and transfusion rather than steady state clear-
ance of creatinine Postoperative reductions in
renal function are commonly multifactorial and
Renal Response attributed to sepsis, renal ischemia, and nephro-
toxic medications. Calcineurin inhibitors such as
Acute kidney injury (AKI) is a common compli- cyclosporin A and FK506 (tacrolimus) contribute
cation following liver transplantation, occurring to renal injury by decreasing renal blood flow via
in as many as half of all recipients [16]. Patients afferent arterial vasoconstriction. Chronic
with preoperative acute kidney injury (AKI) and nephropathy commonly occurs in patients who
cirrhosis have a higher incidence of short- and have received liver, heart, lung, or kidney trans-
long-term complications and increased risk of plants. Managing liver transplantation patients
mortality after liver transplantation than those with low urine output, renal dysfunction, or
without renal failure [17, 18]. Patients with cir- ­postoperative hyperkalemia is challenging, and
rhosis have a lower creatinine production rate continuous renal replacement therapy may be
from muscle wasting and malnutrition, and serum needed to manage volume shifts, acid-base bal-
creatinine may not be reflective of the glomerular ance, and electrolyte disturbances.
filtration rate (GFR). Even small decreases in kid-
ney function are associated with poorer outcomes
and long-term renal complications [17, 18]. Neurological Response
Patients at risk for kidney injury include those
with preoperative hypovolemia from gastrointes- Hepatic encephalopathy, a neuropsychiatric
tinal bleeding, diarrhea from infection or lactu- complication of acute and chronic liver disease,
31  Routine Postoperative Care After Liver Transplantation 419

ranges from mild confusion to cerebral edema 24]. Both states are associated with a decrease in
with intracranial hypertension. Patients with effective circulating volume.
encephalopathy have disturbances in conscious- Hyponatremia is associated with HRS, ascites,
ness, cognitive abilities, behavior, neuromuscu- increased risk of death from liver disease, and
lar function, concentration, reaction time, postoperative mortality after transplantation.
memory, and/or electroencephalogram readings. Patients who undergo liver transplantation are at
The pathogenesis of hepatic encephalopathy is risk for perioperative central pontine myelinolysis
poorly understood, but most theories implicate (CPM), a neurological condition characterized
elevated levels of ammonia, a gut-derived neuro- by symmetric non-inflammatory demyelinating
toxin, that is shunted to the systemic circulation lesions in the basis pontis. The etiology of CPM is
from the portal system. Bacteria in the intestinal uncertain, but some theories implicate osmotic
tract produce ammonia, which crosses the blood- stress on central nervous system cells, and CPM
brain barrier into astrocytes that detoxify it to correlates with rapid correction of hyponatremia
glutamine. An increased concentration of intra- [24]. Sodium levels will almost always increase
cellular glutamine causes swelling of astrocytes, during liver transplantation and sodium needs to
that reduces their ability to regulate neurotrans- be frequently measured during and after surgery
mission. Patients with hepatic encephalopathy in the patient with hyponatremia to avoid too
have high serum ammonia levels, but ammonia rapid correction.
levels do not correlate with the severity of neuro- Hypomagnesemia may be worsened by calci-
logical symptoms. This observation suggests neurin inhibitors, loop diuretics, and significant
that other factors, such as hyponatremia, gastro- blood loss. Total serum calcium is often low
intestinal bleeding, infection and inflammation because liver transplant patients commonly have
contribute to the development of hepatic enceph- low serum albumin with reduced calcium bind-
alopathy [21–23]. In acute liver failure cerebral ing. Therefore, ionized calcium levels should be
edema can progress to intracranial hypertension followed routinely and corrected as indicated.
and herniation of the brain. In chronic liver dis- The administration of packed red blood cells also
ease the brain has time to adjust and cerebral contributes to hypocalcemia by citrate chelation.
edema is extremely rare. Hepatic encephalopa- Malnutrition and vitamin D deficiency may also
thy typically improves in patients who receive a cause perioperative hypocalcemia.
well-functioning graft. By contrast, graft dys-
function may lead to persistence or new onset of
altered mental status, a state that should prompt Glycemic Response
further investigations.
Elevated blood glucose is often a result of the
stress response and its presence may correlate
Sodium and Electrolyte Response with illness severity. After liver surgery, the meta-
bolic and endocrine function of transplant patients
Preoperative hyponatremia is commonly observed can range from mild hyperglycemia with no clini-
in patients with advanced cirrhosis. Hypervolemic cal consequence to the severely altered neuroen-
hyponatremia is due to impaired excretion of sol- docrine responses associated with chronic critical
ute-free water resulting in expanded extracellular illness. Acute hyperglycemia has emerged as a
volume, ascites, and edema. By contrast, patients marker of outcome after liver transplantation sur-
with hypovolemic hyponatremia, due to renal loss gery and acute hypoglycemia is associated with
of extracellular fluid (overdiuresis) or loss from poor graft function and sepsis [25, 26]. Immediate
the gastrointestinal tract, rarely have ascites or postoperative glucose control can be challenging
edema but present with pre-renal azotemia from because multiple factors affect glucose levels,
low circulating volume or hepatic encephalopathy including inflammatory response to transplanta-
from a rapid reduction in serum osmolality [19, tion surgery, steroid administration, hepatic dys-
420 J. Hastie and V. K. Moitra

function, altered glycogen stores and insulin Uncontrolled hemorrhage and massive trans-
resistance of liver failure. fusion in the operating room or the ICU may
cause the lethal triad of acidosis, coagulopathy,
and hypothermia. Left uncorrected, each of these
Coagulation Response abnormalities can exacerbate the others, creating
a vicious cycle. Early complications of massive
Patients with liver disease have hemostatic transfusion that may become apparent in the
changes that promote both bleeding and throm- intensive care unit include acute hemolytic trans-
bosis. Tendency to bleeding is caused by inade- fusion reactions, febrile nonhemolytic transfu-
quate synthesis of all coagulation factors (except sion reactions, transfusion-related acute lung
for von Willebrand factor), thrombocytopenia, injury, transfusion-associated circulatory over-
platelet function defects, dysfibrinogenemia, and load, allergic reactions, bacterial sepsis, hypocal-
elevated tissue plasminogen activator levels. In cemia, and hyperkalemia [32].
contrast, elevations of von Willebrand factor and
factor VIII, and decreased levels of ADAMTS-13,
protein C, protein S, antithrombin, alpha I nitial Care After Liver
2-­macroglobulin, plasminogen, and heparin Transplantation
cofactor II, favor thrombosis. Thrombin genera-
tion is normal in cirrhosis, and in coagulation Hemodynamic Monitoring
tests (prothrombin time and activated partial
thromboplastin time), thrombin generation is Liver transplant recipients will arrive in the ICU
considered a function of procoagulant factors, with several invasive hemodynamic monitors.
and anticoagulant factors that inhibit thrombin Intensivists use monitors to diagnose and manage
are not considered [27–30]. hemodynamic disturbances from fluid shifts, hem-
Levels of fibrinogen, an acute phase reactant, orrhage, myocardial dysfunction, or vasodilation.
are normal or increased in liver disease. Patients The early detection and treatment of sepsis can
with severe hepatic dysfunction, however, may have a significant impact on morbidity and mortal-
synthesize fibrinogen poorly, which increases the ity [33]. This of particular importance since liver
risk for bleeding. Despite high concentrations of transplant recipients are at risk for wound infec-
fibrinogen found in patients with chronic hepati- tion, urinary tract infection, catheter-­ associated
tis, cholestatic jaundice, and hepatocellular carci- blood stream infection, and pneumonia.
noma, clot formation is not enhanced because Immediately after arrival, the intensivist will
fibrinogen is dysfunctional. Patients may have an assess fluid responsiveness, vascular tone, myo-
abnormal thrombin time with normal values for cardial function, and the adequacy of tissue
prothrombin time [31] and activated partial ­oxygen delivery and organ perfusion (Fig. 31.4).
thromboplastin time. An electrocardiogram should be obtained on
Several perioperative factors that promote arrival to detect myocardial ischemia and electro-
bleeding, such as heparin effects, disseminated lyte abnormalities. A radial arterial catheter is
intravascular coagulation or hyperfibrinolysis, are used for beat-to-beat blood pressure monitoring
typically resolved before leaving the operating and frequent blood sampling. Pulse pressure vari-
room. However, the early postoperative phase ation, calculated from the invasive arterial tracing
may be characterized by clinical bleeding from in the ventilated patient, aids in the assessment of
dilutional coagulopathy, ongoing platelet and fac- fluid responsiveness. Peripheral arterial pressure
tor consumption, and poor graft function. Even a measurements may not be reliable in patients with
well functioning transplanted liver may not pro- severe vasoconstriction or vasodilation and a sec-
duce sufficient levels of coagulation factors for ond arterial catheter is often placed in the femoral
several days; consequently plasma, cryoprecipi- artery to measure central blood pressure. Central
tate, or platelet transfusions may be required. venous access, established in the operating room,
31  Routine Postoperative Care After Liver Transplantation 421

Initial Hemodynamic and Perfusion Assessment

Questions
Is the patient fluid responsive?
What is the cardiac function?
Is there adequate tissue perfusion?

Examination Clinical Status Laboratory Assessment Hemodynamic Monitors


Vital Signs Urine Output Mixed Venous Saturation Cardiac Output by Pulmonary Artery Catheter
Extremity Temperature & Pulses Mental Status Arterial Lactate Arterial Waveform Analysis
Capillary Refill Acid-Base Status Filling Pressures
Echocardiography

Fig. 31.4  Algorithm to assess the hemodynamic and perfusion state of the liver transplant recipient

is used to measure right atrial pressure and to room or within 6 h after the operation, mechani-
administer vasoactive drugs and blood products. cal ventilation is sometimes required for 24–48 h
Central venous pressure monitoring does not reli- after transplantation. Prolonged intubation and
ably measure blood volume or change in blood mechanical ventilation are associated with
volume [34]. The use of a pulmonary artery cath- ventilator-­associated pneumonia and muscle
eter depends on local institutional practice and wasting.
the physiological state of the patient. Mixed Management of mechanical ventilation is
venous oxygen saturation values indirectly mea- guided by the physical examination, assessment
sure cardiac output by the Fick principle and of arterial blood gas values, ventilator mechanics
adequacy of global oxygen delivery, but these and chest radiography. Initial ventilation settings
measurements are also affected by changes in include a mode of assisted control volume con-
oxygen-carrying capacity and oxygen consump- trol ventilation with a tidal volume of 6–8 mL/kg
tion. Echocardiography assesses ventricular fill- ideal body weight (determined by the patient’s
ing, contractility, and function. Diagnoses such height); a respiratory rate of 10–15 breaths per
as myocardial ischemia, pulmonary embolism, minute, and an FIO2 titrated to maintain an oxy-
pleural effusions, and technical complications of gen saturation > 95%. Respiratory alkalosis may
the inferior vena cava anastomosis can be be observed in patients with a high respiratory
detected with transesophageal echocardiography drive from liver dysfunction. Alternatively,
(TEE) and the risk of injury or rupture of esopha- mechanical ventilation may be used to compen-
geal varices with TEE is rare. Bladder pressure sate for a lactic acidosis associated with liver dys-
should be measured to evaluate intra-abdominal function or incomplete fluid resuscitation.
hypertension, especially in patients with ascites The use of positive-pressure mechanical ven-
or bleeding complications when elevated intra- tilation and positive end-expiratory pressure
abdominal pressure is more common. (PEEP) is of concern in the liver transplant
patients as both increase intrathoracic pressure
thereby decreasing venous return from the infe-
Mechanical Ventilation rior vena cava and hepatic veins. This may lead to
congestion of the graft. However, the clinical rel-
Tracheal extubation should be performed when evance remains controversial, as studies with
there is resolution of respiratory failure and evi- PEEP of up to 10 cmH2O have shown no adverse
dence of satisfactory graft function. Although effects on hepatic arterial, portal venous, or
many patients can be extubated in the operating hepatic venous flow [7]. Ventilator management
422 J. Hastie and V. K. Moitra

General Extubation Guidelines logical parameters (Table 31.1). The Riker


Sedation-Agitation Scale (SAS), the Motor
Resolution of Respiratory Failure
Awake, Cooperative
Activity Assessment Scale (MAAS), the
Pain Controlled Adaptation to the Intensive Care Environment
Manageable Secretions (ATICE), and the AVRIPAS scale, which incorpo-
Stable Hemodynamics
rates heart rate and respiration have all been uti-
lized to guide sedation [36–40]. It is important to
Fig. 31.5  Extubation guidelines
note that these scales assess the level of sedation
only and not pain, anxiety, or level of cognition;
in ARDS should focus on limiting lung stretch they cannot be used in the presence of neuromus-
via low tidal volumes (4–6 mL/kg) and permit- cular blockade and none of them have been exclu-
ting hypercapnia to minimize barotrauma and sively validated in patients with neurological
volutrauma. injury such as cerebral edema.
A protocolized approach to awakening and Sedation can be considered as a combination
weaning of mechanical ventilation can shorten of three components: anxiolysis (that is indicated
time of mechanical ventilation and ICU length of for almost every ICU patient), hypnosis (i.e., the
stay. Accordingly, a daily spontaneous awaken- induction of sleep, which may be indicated in
ing trial (accomplished by reducing doses of acutely ill and/or ventilated patients), and amne-
sedative infusions) in liver transplantation sia (loss or lack of recall). Sedation is distinct
patients permits ventilator weaning by allowing from analgesia (the relief of pain), and sedative
an assessment of hemodynamic stability, respira- agents such as propofol or benzodiazepines
tory strength, and oxygenation. Mechanical ven- (lorazepam and midazolam) have no analgesic
tilation should be discontinued when the effects. Administration of sedation as treatment
following conditions are met: the underlying rea- for pain-induced agitation may disinhibit control
son for respiratory failure has resolved, the graft functions and lead to a seemingly paradoxical
is functioning, the patient is awake and coopera- increase in agitation. Also, although amnesia is
tive, the patient’s pain is well controlled, tracheo- essential during general anesthesia in the operat-
bronchial secretions are manageable, and ing room, the potent anterograde amnesia induced
vasopressor requirements are stable (Fig. 31.5). by benzodiazepines, even at subhypnotic doses,
results in confusion and disorientation on awak-
ening, and therefore may predispose toward ICU
Sedation and Analgesia delirium. In contrast, propofol provides amnesia
only during sleep and emergence is likely
Sedation in the ICU is used to promote comfort, smoother.
alleviate anxiety, and to facilitate mechanical ven- The intensivist should consider an “analgesia
tilation. The use of a sedation scale such as the first” or “A-1” approach to relieve the patient’s
Ramsay scale [35] can facilitate the appropriate pain before administration of sedation [41]. This
level of sedation, improve communication approach will avoid disinhibiting a patient whose
between care providers and prevent overdose and agitation is due to pain as discussed above. There
hypotension. The Richmond Agitation-Sedation is evidence that an A-1 approach decreases both
Scale (RASS) has achieved prominence because sedation requirements and mechanical ventila-
its continuum represents a balance between tion time [42–46]. ICU patients experience pain
degrees of agitation and sedation. As such, it cor- both from surgical causes, such as incisions and
relates better with electroencephalographic drains, as well as routine ICU care, including
assessment and has been integrated with an ­tracheal intubation, endotracheal tube suction-
assessment of delirium called the Confusion ing, and repositioning. Failure to treat pain exac-
Assessment Method for the ICU (CAM-ICU) erbates endogenous catecholamine activity,
(Fig. 31.6). Several other instruments for assess- which predisposes to myocardial ischemia,
ment of sedation focus on a­ gitation and physio-
31  Routine Postoperative Care After Liver Transplantation 423

Confusion Assessment Method for the ICU (CAM-ICU) Flowsheet

1. Acute Change or Fluctuating Course of Mental Status:


CAM-ICU negative
• Is there an acute change from mental status baseline? OR NO
NO DELIRIUM
• Has the patient’s mental status fluctuated during the past 24 hours?

YES
2. Inattention:
• “Squeeze my hand when I say the letter ‘A’.” 0-2 CAM-ICU negative
Read the following sequence of letters: S A V E A H A A R T Errors NO DELIRIUM
ERRORS: No squeeze with ‘A’ & Squeeze on letter other than ‘A’
• If unable to complete Letters → Pictures
> 2 Errors

3. Altered Level of Consciousness: RASS other CAM-ICU positive


Current RASS level than zero DELIRIUM Present

RASS = zero

4. Disorganized Thinking:
> 1 Error
1. Will a stone float on water?
2. Are there fish in the sea?
3. Does one pound weight more than two?
4. Can you use a hammer to pound a nail? 0-1
Error CAM-ICU negative
Command: “Hold up this many fingers” (Hold up 2 fingers) NO DELIRIUM
“Now do the same thing with the other hand” (Do not demonstrate)
OR “Add one more finger” (If patient unable to move both arms)

Fig. 31.6  The Confusion Assessment Method for the ICU (CAM-ICU). Copyright © 2002, E. Wesley Ely, MD, MPH
and Vanderbilt University, all rights reserved

Table 31.1  Sedation scales


Modified Ramsay Sedation Scale [35]
1 Awake Anxious, agitated, restless
2 Awake Cooperative, orientated, serene
3 Awake Responding only to commands
4 Asleep Brisk response to stimulationa
5 Asleep Sluggish response to stimulationa
6 Asleep No response to stimulationa
Richmond Agitation-Sedation Scale (RASS)
Score Assessment Description
+4 Combative Overtly combative, violent, danger to staff with observation
+3 Very agitated Pulls or removes tube(s) or catheters, aggressive with observation
+2 Agitated Frequent nonpurposeful movement with observation or
dyssynchrony with ventilator
+1 Restless Anxious, apprehensive, but not aggressive
0 Alert and calm upon observation
−1 Drowsy With loud speaking voice awakens >10 s, not fully alert
−2 Light sedation With loud speaking voice briefly awakens to voice <10 s
−3 Moderate sedation With loud speaking voice has movement or eye opening without
eye contact
−4 Deep sedation Movement to physical stimulation
−5 Unarousable No response to physical stimulation
Stimulation = glabellar tap or loud noise
a
424 J. Hastie and V. K. Moitra

hypercoagulability, hypermetabolic states, sleep In contrast to gamma-aminobutyric acid ago-


deprivation, and delirium [47, 48]. nists, dexmedetomidine sedates without changes
Opioids are the mainstay of pain manage- in respiratory rate, oxygen saturation, or arterial
ment in the ICU (Table 31.2) and synthetic anal- carbon dioxide tension [60]. Unlike benzodiaz-
gesics such as fentanyl and remifentanil are epines, clinical doses of dexmedetomidine are
commonly used. These agents are administered not associated with anterograde amnesia.
as a bolus or as an infusion to manage pain and Patients are easily aroused from light levels of
facilitate patient synchrony with mechanical sedation and emerge without confusion or dis-
ventilation. Fentanyl has a high hepatic extrac- orientation. When left undisturbed, they go back
tion ratio and its metabolism is slowed in to their previous level of sedation. Thus, dexme-
patients with liver disease (e.g., cirrhosis) or detomidine produces interactive, or coopera-
hepatic dysfunction (e.g., congestive heart fail- tive, sedation and facilitates neurological
ure and shock) [49]. examination [61–63]. Although dexmedetomi-
Benzodiazepines such as midazolam and dine may decrease the incidence of delirium in
lorazepam are lipid-soluble, and because they the ICU, this effect has not been extensively
accumulate in fat stores, prolonged infusions studied in liver transplantation patients who
result in markedly delayed emergence. Patients may have a pretransplant encephalopathy.
with hepatic dysfunction may be sensitive to ben- Dexmedetomidine is dependent on hepatic
zodiazepines. Conversely, patients who have a elimination.
history of alcohol abuse may require increased In the postoperative period, early weaning of
doses of benzodiazepines. Lorazepam is diluted sedation allows for assessment of mental status.
in propylene glycol, which has been associated The preoperative mental function, as determined
with AKI and metabolic acidosis; accordingly the by medical history, chart review, and discussion
osmolar anion gap should be calculated in with family and other medical providers, allows a
patients receiving lorazepam doses greater than point of comparison. Unless contraindicated by
1 mg/kg/day. medical conditions, sedation should be inter-
Propofol is a potent sedative that decreases rupted daily to facilitate weaning of mechanical
catecholamine levels, induces vasodilation, and ventilation and assessment of mental status.
limits baroreflex cardiovascular responses. Altered mental status should prompt thorough
Although propofol sedation may promptly lower evaluation, excluding common causes such as
intracranial pressure (ICP) in the liver transplant residual anesthetic agents, electrolyte and glu-
patient with cerebral edema [50–52], it may also cose abnormalities, infection, inadequate gas
induce hypotension, especially in hypovolemic exchange, and intracranial pathology. The failure
patients, and thus decrease cerebral perfusion to find a specific cause may suggest graft
pressure [51, 53]. Because propofol is highly dysfunction.
lipid soluble, it is suspended in a 20% fat emul-
sion that may predispose to infection, hypertri-
glyceridemia, and pancreatitis [54–58]. Prolonged Immunosuppression
high-dose propofol infusion in the setting of
shock, high endogenous or exogenous catechol- Immunosuppression is usually started by postop-
amines, and corticosteroids is associated with the erative day 1. Intraoperative renal injury, how-
very rare but potentially fatal propofol infusion ever, is frequently not yet apparent postoperative
syndrome that appears to result from an intracel- day 1, and aggressive immunosuppression may
lular block in fat oxidation, resulting in intracta- compound renal injury, even precipitating acute
ble lactate acidosis, myocardial depression, and kidney injury. The decisions of when and how to
death [59]. start immunosuppression in the liver transplant
recipient should therefore involve the hepatolo-
Table 31.2  Intravenous analgesics and sedatives
Medication Intermittent (Bolus) dose Infusion dose Onset of action Half-life Active metabolites Unique adverse effects
Fentanyl 25–50 mcg 10–400 mcg/h 2 min 1.5–6 h N Accumulation of parent compound
Remifentanil Not recommended 0.05–0.2 mcg/kg/min 3–10 min N Hyperalgesia
Hydromorphone 0.25–0.5 mg 0.5–1 mg/h 15 min 2–3 h N
Morphine 0.5–10 mg 1–10 mg/h 15 min 3–7 h Y Histamine release
Accumulation of metabolite in renal failure
Midazolam 1–2 mg 1–10 mg/h 2–5 min 3–11 h Y Accumulation of parent compound
31  Routine Postoperative Care After Liver Transplantation

Accumulation of metabolite in renal failure


Lorazepam 0.5–1 mg 1–10 mg/h 5–20 min 8–15 h N High-dose PG-related acidosis or renal failure
Propofol Not recommended 5–70 mcg/kg/min Immediate 26–32 h N PRIS
Infection risk
Elevated triglycerides
Dexmedetomidine Not recommended 0.2–1.5 mcg/kg/h 30 min 2–5 h N Bradycardia
PG propylene glycol, mcg micrograms, mg milligrams, PRIS propofol infusion syndrome
425
426 J. Hastie and V. K. Moitra

gists, transplant surgeons, and intensivists. measurements with ultrasound may offer a non-
Immunosuppressive regimens are discussed in invasive means of monitoring elevated ICP in
detail elsewhere in this book. these patients. Drugs or conditions that exacer-
bate elevations in ICP should be avoided. Other
neurological complications include stroke, sei-
Hepatic Encephalopathy zures, and coma. A focal ­deficit diagnosed in the
ICU should prompt c­ onsideration for stroke or
Initial management of hepatic encephalopathy bleed and trigger immediate head CT.
should include the identification and treatment
of reversible triggers of this neuropsychiatric
syndrome, such as gastrointestinal bleeding
and infection. Non-absorbable disaccharides Sodium and Electrolyte Management
such as lactulose decrease absorption of ammo-
nia from the intestinal tract via catharsis. Distinguishing between hypervolemic and hypo-
Excessive dosing of lactulose causes dehydra- volemic hyponatremia is essential to guide treat-
tion. Oral antibiotics (rifaximin, neomycin, ment. Hypervolemic hyponatremia is managed
vancomycin, paromomycin, or metronidazole) with fluid restriction (1–1.5 L/day) and withhold-
reduce ammonia-­ producing enteric bacteria. ing of diuretics. Vaptans, medications that block
Rifaximin, in combination with lactulose, may the vasopressin-2 receptor, increase solute-free
prevent episodes of hepatic encephalopathy water excretion by blocking renal vasopressin 2
[21, 23, 64]. receptors and may preclude water restriction so
Patients who develop fulminant hepatic fail- that diuretics can be continued. Patients with
ure are at risk for hepatic encephalopathy, cere- hypovolemic hyponatremia are treated with
bral edema with increased ICP, and the saline administration to increase plasma volume
possibility of herniation. ICP monitoring should and sodium [24].
be strongly considered for patients with fulmi- Rapid changes in the concentration of serum
nant hepatic failure and encephalopathy and sodium cannot be predicted, and a safe rate of
continued postoperatively until intracranial correction of hyponatremia has not been defin-
hypertension resolved and/or the patient recov- itively established; sodium correction at a rate
ered sufficiently to assess neurological status of less than 12 mEq/L/day is usually consid-
clinically. Historically, many clinicians avoided ered safe. Preoperative correction of hypona-
ICP monitoring as it carries the risk of intracra- tremia may prevent a rapid rise in serum
nial bleeding. More details of ICP monitoring sodium intraoperatively and postoperatively.
are discussed elsewhere in this book. Persistent Intraoperative resuscitation with sodium bicar-
ICP elevations to greater than 25 mmHg should bonate may be associated with large sodium
be treated with mannitol to increase serum loads, leading to inadvertent rapid correction
osmolarity and reduce cerebral edema. of serum sodium or even hypernatremia at the
Preoperative hyperventilation has not been end of surgery.
shown to improve outcome, and corticosteroids Potassium, magnesium, and calcium levels
are not indicated. Pentobarbital coma may be should be monitored frequently and abnormal
indicated for patients who are unresponsive to levels corrected. Metabolic acidosis is managed
mannitol, but coma may worsen cerebral perfu- by improving hemodynamic parameters to ensure
sion by causing systemic hypotension. Many adequate tissue perfusion; until acidosis resolves,
centers consider sustained cerebral hypoperfu- the pH may be buffered by increasing the minute
sion (cerebral perfusion pressure < 40 mmHg) ventilation. Severe metabolic acidosis may
as a contraindication to transplant because of require slow administration sodium bicarbonate
the high risk for brain death. Transcranial or tromethamine (THAM) in combination with
Doppler ultrasonography or optic nerve sheath hyperventilation.
31  Routine Postoperative Care After Liver Transplantation 427

Glycemic Control Table 31.3  Signs of adequate graft function


• Clearance of lactic acid (conversion to pyruvate)
Although the role of “tight glycemic” control has • Restoration of vascular tone and decreasing
not been adequately studied immediately after vasopressor requirements
liver transplantation, the results of the • Production of glucose (gluconeogenesis and
glycogenolysis)
Normoglycemia in Intensive Care Evaluation and
• Resolution of encephalopathy
Survival Using Glucose Algorithm Regulation
• Emergence from anesthesia (biotransformation of
(NICE-SUGAR) investigation, a large multi- anesthetic agents)
center, multinational, randomized, and non-­ • Normothermia (metabolic activity)
blinded trial of medical and surgical patients, • Normalization of coagulopathy
suggest that intensive insulin therapy does not • Decreasing total bilirubin level
improve outcomes [65]. Maintaining glucose lev- • Production of bile (visible if a biliary tube was
els between 140 and 180 mg/dL is a reasonable placed)
goal in most situations. Insulin should preferably • Resolution of HRS (resolving endotoxemia)
be administered by intravenous infusion, and glu- • Adequate urine output
cose should be monitored continually.
Metabolic alkalemia can develop as a result of
citrate metabolism. Potassium levels tend to nor-
 anagement of Coagulopathy
M malize with the onset of hepatocyte function.
and Bleeding A number of recipients exhibit hyperglycemia
resistant to insulin.
Warming the room, using active warming systems An important task in the ICU is the identifi-
administering blood products and fluids through a cation and management of poorly functioning
fluid warmer, and heating and humidifying liver grafts. Graft dysfunction ranges from
inspired gases reduce the risk of hypothermia. mildly impaired function to complete failure of
Dilutional coagulopathy and thrombocytopenia graft synthetic and metabolic function. In the
may be prevented by transfusing packed red blood ICU several signs suggest adequate graft func-
cells, fresh frozen plasma, and platelets in a 1:1:1 tion (Table 31.3). Preservation and reperfusion
ratio [32]. The risk of transfusion-related acute injury causes transaminases to rise immedi-
lung injury may be reduced by avoiding unneces- ately after surgery, but enzyme levels usually
sary transfusions. When transfusions are required, fall within 24–48 h. Even high levels of trans-
packed red blood cells with a short storage time aminases are not necessarily a reason for major
and fresh frozen plasma from men or nulliparous concern as they are an indicator of past injury
women may also decrease this risk. Overaggressive and not present function. New synthesis of
transfusion can elevate central venous pressures coagulation factors will not correct fac-
and cause acute congestion and ischemia of the tor depletion for hours to days after the trans-
transplanted liver. plant. As the transplanted liver begins to
function, other organ systems will generally
improve.
Evaluating Graft Function Extrahepatic organ dysfunction may be an
indirect sign of postoperative graft dysfunction or
Assessment of graft function begins in the oper- failure. Graft dysfunction is characterized by lac-
ating room. Good texture and color of the graft, tic acidosis, hypoglycemia, and altered mental
evidence of bile production, improving hemody- status or persistent encephalopathy. Severe dys-
namic stability and decreasing lactate levels are function should prompt the clinician to exclude
signs of adequate graft function. Hypocalcemia surgical complications such as anastomotic prob-
often resolves quickly as the graft metabolizes lems that may require re-exploration. In addition,
citrate during the final phases of the procedure. a general and gradual worsening of the patient’s
428 J. Hastie and V. K. Moitra

clinical status days after transplant may be due to and inflammation impair protective mechanisms.
allograft rejection. This clinical setting may Coagulopathy and mechanical ventilation are
prompt a diagnostic liver biopsy. In contrast to independent risk factors for stress ulceration, and
past practices, routine biopsies are seldom pharmacological ulcer prophylaxis with
performed. H2-receptor blockers, proton-pump inhibitors or
sucralfate should be initiated after transplantation
[66]. Enteral nutrition alone is not sufficient to
 ssessment of Vascular and Biliary
A prevent stress ulceration. Theoretically, an
Complications increase in gastric pH via gastric acid suppres-
sion promotes bacterial colonization of the gas-
Vascular complications are occasionally seen trointestinal tract and may increase the risk for
early after liver transplant, and they include anas- nosocomial pneumonia and Clostridium difficile
tomotic bleeding, luminal stenosis, or occlusion infections [31, 67–69].
from thrombosis or kinking of vessels.
Thrombosis of the portal vein or hepatic artery
are of particular concern as they compromise Venous Thromboembolism
viability of the graft. Abdominal Doppler ultra-
sound is used to assess the hepatic vessels and Contrary to conventional belief, patients who
routinely done within the first few hours in all undergo liver transplantation may be normo- or
patients at our institution. Hepatic artery throm- even hypercoagulable and are not protected
bosis, which is more common than portal vein against venous thromboembolism. Liver trans-
thrombosis, may cause hepatic necrosis leading plant patients are not “autoanticoagulated.”
to liver failure. Portal vein thrombosis may cause Imbalances in the clotting cascade towards hyper-
liver dysfunction, tense ascites and variceal coagulability, as well as immobility, surgery, and
bleeding. Patients with bile leaks experience system inflammation, increase the risk of venous
fever, abdominal pain, and peritoneal irritation. thromboembolism and pulmonary embolism [28–
Bile leaks develop early in the postoperative 30, 70, 71]. Thromboembolic prophylaxis includes
course and may be identified by bilious fluid in graduated compression stockings and/or intermit-
drains. Ultrasonography that shows abdominal tent pneumatic compression and low-dose unfrac-
fluid collections or cholangiography can confirm tionated heparin or low-­molecular-­weight heparin.
the diagnosis. Ductal structures may be compro- If there is no evidence of active bleeding, pharma-
mised by poor vascular flow as described above; cological thromboembolic prophylaxis should be
they are associated with an elevation in alkaline initiated on postoperative day one.
phosphatase and gamma-glutamyl transpepti-
dase. Bile leaks are managed with endoscopic
placement of a biliary stent or surgical explora- References
tion and repair. These problems may be amenable
to radiological intervention, or require 1. Ramsay M. Justification for routine intensive care after
re-exploration. liver transplantation. Liver Transpl. 2013;19(Suppl
2):S1–5.
2. Mandell MS, et al. Reduced use of intensive care after
liver transplantation: influence of early extubation.
Prevention of ICU Complications Liver Transpl. 2002;8(8):676–81.
3. Toledo AH, et al. Reducing liver transplant length of
Gastrointestinal Stress Ulcers stay: a Lean Six Sigma approach. Prog Transplant.
2013;23(4):350–64.
4. Ely EW, et al. Large scale implementation of a respira-
ICU patients are at risk for developing gastroin- tory therapist-driven protocol for ventilator weaning.
testinal stress ulcers because hypovolemia, hypo- Am J Respir Crit Care Med. 1999;159(2):439–46.
perfusion, sympathetic nervous system activation
31  Routine Postoperative Care After Liver Transplantation 429

5. McElroy LM, et al. Operating room to intensive care 24. Gines P, Guevara M. Hyponatremia in cirrhosis:

unit handoffs and the risks of patient harm. Surgery. pathogenesis, clinical significance, and management.
2015;158(3):588–94. Hepatology. 2008;48(3):1002–10.
6. Iwakiri Y, Groszmann RJ. The hyperdynamic circula- 25. Ammori JB, et al. Effect of intraoperative hyper-

tion of chronic liver diseases: from the patient to the glycemia during liver transplantation. J Surg Res.
molecule. Hepatology. 2006;43(2 Suppl 1):S121–31. 2007;140(2):227–33.
7. Srivastava P, et al. Perioperative management of 26. Park C, et al. Severe intraoperative hyperglycemia
liver transplantation with concurrent coronary artery is independently associated with surgical site infec-
disease: report of two cases. Indian J Anaesth. tion after liver transplantation. Transplantation.
2013;57(6):599–602. 2009;87(7):1031–6.
8. Paugam-Burtz C, et al. Postreperfusion syndrome 27. Lisman T, Porte RJ. Rebalanced hemostasis in

during liver transplantation for cirrhosis: outcome and patients with liver disease: evidence and clinical con-
predictors. Liver Transpl. 2009;15(5):522–9. sequences. Blood. 2010;116(6):878–85.
9. Daker C, et al. Beneficial effect of intra-­operative 28. Senzolo M, et al. New insights into the coagulopa-
methylprednisolone on immediate post liver trans- thy of liver disease and liver transplantation. World J
plant intensive care course. Ann Transplant. Gastroenterol. 2006;12(48):7725–36.
2015;20:76–84. 29. Dabbagh O, et al. Coagulopathy does not pro-

10. Zardi EM, et al. Cirrhotic cardiomyopathy. J Am Coll tect against venous thromboembolism in hospi-
Cardiol. 2010;56(7):539–49. talized patients with chronic liver disease. Chest.
11. Xia VW, et al. Postoperative atrial fibrillation in liver 2010;137(5):1145–9.
transplantation. Am J Transplant. 2015;15(3):687–94. 30. Dalmau A, Sabate A, Aparicio I. Hemostasis and
12. Kia L, et al. The utility of pulmonary function testing coagulation monitoring and management during
in predicting outcomes following liver transplanta- liver transplantation. Curr Opin Organ Transplant.
tion. Liver Transpl. 2016;22(6):805–11. 2009;14(3):286–90.
13. Kim WR, et al. Hyponatremia and mortality among 31. Apte NM, et al. Gastric colonization and pneumonia
patients on the liver-transplant waiting list. N Engl J in intubated critically ill patients receiving stress ulcer
Med. 2008;359(10):1018–26. prophylaxis: a randomized, controlled trial. Crit Care
14. Hemprich U, Papadakos PJ, Lachmann B. Respiratory Med. 1992;20(5):590–3.
failure and hypoxemia in the cirrhotic patient 32. Sihler KC, Napolitano LM. Complications of massive
including hepatopulmonary syndrome. Curr Opin transfusion. Chest. 2010;137(1):209–20.
Anaesthesiol. 2010;23(2):133–8. 33.
Razonable RR, et al. Critical care issues in
15. Sahmeddini MA, et al. Restricted crystalloid fluid patients after liver transplantation. Liver Transpl.
therapy during orthotopic liver transplant surgery 2011;17(5):511–27.
and its effect on respiratory and renal insufficiency 34. Marik PE, Baram M, Vahid B. Does central venous
in the early post-operative period: a random- pressure predict fluid responsiveness? A systematic
ized clinical trial. Int J Organ Transplant Med. review of the literature and the tale of seven mares.
2014;5(3):113–9. Chest. 2008;134(1):172–8.
16. Hilmi IA, et al. Acute kidney injury following ortho-
35. Ramsay MA, et al. Controlled sedation
topic liver transplantation: incidence, risk factors, and with alphaxalone-­ alphadolone. Br Med J.
effects on patient and graft outcomes. Br J Anaesth. 1974;2(5920):656–9.
2015;114(6):919–26. 36. Sessler CN, et al. The Richmond Agitation-Sedation
17. Barri YM, et al. Acute kidney injury following
Scale: validity and reliability in adult intensive
liver transplantation: definition and outcome. Liver care unit patients. Am J Respir Crit Care Med.
Transpl. 2009;15(5):475–83. 2002;166(10):1338–44.
18. Zand MS, et al. High mortality in orthotopic liver 37. Riker RR, Picard JT, Fraser GL. Prospective evalu-
transplant recipients who require hemodialysis. Clin ation of the Sedation-Agitation Scale for adult criti-
Transpl. 2011;25(2):213–21. cally ill patients. Crit Care Med. 1999;27(7):1325–9.
19. Gines P, Schrier RW. Renal failure in cirrhosis. N 38. Hogg LH, et al. Interrater reliability of 2 sedation
Engl J Med. 2009;361(13):1279–90. scales in a medical intensive care unit: a preliminary
20. O’Riordan A, et al. Acute renal disease, as defined report. Am J Crit Care. 2001;10(2):79–83.
by the RIFLE criteria, post-liver transplantation. Am 39. De Jonghe B, et al. Adaptation to the Intensive Care
J Transplant. 2007;7(1):168–76. Environment (ATICE): development and validation of
21. Bass NM, et al. Rifaximin treatment in hepatic enceph- a new sedation assessment instrument. Crit Care Med.
alopathy. N Engl J Med. 2010;362(12):1071–81. 2003;31(9):2344–54.
22.
Bernal W, et al. Acute liver failure. Lancet. 40. Avripas MB, et al. Development of an intensive

2010;376(9736):190–201. care unit bedside sedation scale. Ann Pharmacother.
23.
Teperman LW, Peyregne VP. Considerations 2001;35(2):262–3.
on the impact of hepatic encephalopathy treat- 41. Riker RR, Fraser GL. Altering intensive care sedation
ments in the pretransplant setting. Transplantation. paradigms to improve patient outcomes. Crit Care
2010;89(7):771–8. Clin. 2009;25(3):527–38. viii–ix
430 J. Hastie and V. K. Moitra

42. Breen D, et al. Decreased duration of mechanical 56. Eddleston JM, Shelly MP. The effect on serum

ventilation when comparing analgesia-based sedation lipid concentrations of a prolonged infusion of
using remifentanil with standard hypnotic-based seda- propofol—hypertriglyceridaemia associated with
tion for up to 10 days in intensive care unit patients: propofol administration. Intensive Care Med.
a randomised trial [ISRCTN47583497]. Crit Care. 1991;17(7):424–6.
2005;9(3):R200–10. 57. Leisure GS, et al. Propofol and postoperative pancre-
43. Dahaba AA, et al. Remifentanil versus morphine
atitis. Anesthesiology. 1996;84(1):224–7.
analgesia and sedation for mechanically ventilated 58. Casserly B, et al. Propofol infusion syndrome: an
critically ill patients: a randomized double blind unusual cause of renal failure. Am J Kidney Dis.
study. Anesthesiology. 2004;101(3):640–6. 2004;44(6):e98–101.
44. Muellejans B, et al. Remifentanil versus fentanyl for 59. Vasile B, et al. The pathophysiology of propofol infu-
analgesia based sedation to provide patient comfort sion syndrome: a simple name for a complex syn-
in the intensive care unit: a randomized, double-­ drome. Intensive Care Med. 2003;29(9):1417–25.
blind controlled trial [ISRCTN43755713]. Crit Care. 60. Venn RM, Hell J, Grounds RM. Respiratory effects
2004;8(1):R1–R11. of dexmedetomidine in the surgical patient requiring
45. Muellejans B, et al. Sedation in the intensive care unit intensive care. Crit Care. 2000;4(5):302–8.
with remifentanil/propofol versus midazolam/fen- 61. Nelson LE, et al. The alpha2-adrenoceptor ago-

tanyl: a randomised, open-label, pharmacoeconomic nist dexmedetomidine converges on an endogenous
trial. Crit Care. 2006;10(3):R91. sleep-promoting pathway to exert its sedative effects.
46. Rozendaal FW, et al. Remifentanil-propofol analgo-­ Anesthesiology. 2003;98(2):428–36.
sedation shortens duration of ventilation and length 62. Shelly MP. Dexmedetomidine: a real innovation or
of ICU stay compared to a conventional regimen: a more of the same? Br J Anaesth. 2001;87(5):677–8.
centre randomised, cross-over, open-label study in the 63. Venn RM, Grounds RM. Comparison between dex-
Netherlands. Intensive Care Med. 2009;35(2):291–8. medetomidine and propofol for sedation in the inten-
47. Hall LG, Oyen LJ, Murray MJ. Analgesic agents. sive care unit: patient and clinician perceptions. Br J
Pharmacology and application in critical care. Crit Anaesth. 2001;87(5):684–90.
Care Clin. 2001;17(4):899–923. viii 64. Zafirova Z, O’Connor M. Hepatic encephalopathy:
48. Lewis KS, et al. Effect of analgesic treatment on the current management strategies and treatment, includ-
physiological consequences of acute pain. Am J Hosp ing management and monitoring of cerebral edema
Pharm. 1994;51(12):1539–54. and intracranial hypertension in fulminant hepatic
49. Devlin JW, Roberts RJ. Pharmacology of commonly failure. Curr Opin Anaesthesiol. 2010;23(2):121–7.
used analgesics and sedatives in the ICU: benzo- 65. Finfer S, et al. Intensive versus conventional glu-

diazepines, propofol, and opioids. Crit Care Clin. cose control in critically ill patients. N Engl J Med.
2009;25(3):431–49. vii 2009;360(13):1283–97.
50. Kelly DF, et al. Propofol in the treatment of mod- 66. Cook DJ, et al. Risk factors for gastrointestinal bleed-
erate and severe head injury: a randomized, pro- ing in critically ill patients. Canadian Critical Care
spective double-blinded pilot trial. J Neurosurg. Trials Group. N Engl J Med. 1994;330(6):377–81.
1999;90(6):1042–52. 67. Vincent JL, et al. The prevalence of nosocomial

51. Pinaud M, et al. Effects of propofol on cerebral
infection in intensive care units in Europe. Results
hemodynamics and metabolism in patients with brain of the European Prevalence of Infection in Intensive
trauma. Anesthesiology. 1990;73(3):404–9. Care (EPIC) Study. EPIC International Advisory
52. Hartung HJ. Intracranial pressure in patients with cra- Committee. JAMA. 1995;274(8):639–44.
niocerebral trauma after administration of propofol 68. Dubberke ER, et al. Clostridium difficile-­associated
and thiopental. Anaesthesist. 1987;36(6):285–7. disease in a setting of endemicity: identification of
53.
Hutchens MP, Memtsoudis S, Sadovnikoff novel risk factors. Clin Infect Dis. 2007;45(12):1543–9.
N. Propofol for sedation in neuro-intensive care. 69. Herzig SJ, et al. Acid-suppressive medication use

Neurocrit Care. 2006;4(1):54–62. and the risk for hospital-acquired pneumonia. JAMA.
54. Bennett SN, et al. Postoperative infections traced to 2009;301(20):2120–8.
contamination of an intravenous anesthetic, propofol. 70. Lisman T, et al. Hemostasis and thrombosis in patients
N Engl J Med. 1995;333(3):147–54. with liver disease: the ups and downs. J Hepatol.
55. Henry B, Plante-Jenkins C, Ostrowska K. An out- 2010;53(2):362–71.
break of Serratia marcescens associated with the 71. Warnaar N, Lisman T, Porte RJ. The two tales of
anesthetic agent propofol. Am J Infect Control. coagulation in liver transplantation. Curr Opin Organ
2001;29(5):312–5. Transplant. 2008;13(3):298–303.
Immunosuppression
32
Enoka Gonsalkorala, Daphne Hotho,
and Kosh Agarwal

Keywords Acute cellular rejection (ACR) is graft


Rejection · Calcineurin inhibitors · Steroids · d­ysfunction due to inflammation affecting
Antimetabolites · Polyclonal antibodies · interlobular bile ducts and vascular endothelia
Induction therapy (Fig.  32.1) and, grading of severity is per the
Banff Schema [1]. ACR does not usually affect
long-term graft survival, except in hepatitis C
Introduction infected recipients, and has conversely been
associated with increased patient and graft sur-
The aims of this chapter are to provide an over- vival. One study found that patients who had at
view of the processes involved in immunological least one episode of acute rejection had
rejection after liver transplantation, outline improved 4-year patient (82.8% vs. 75.9%) and
immunosuppression strategies post liver trans- graft survival (76.5% vs. 71.7%) [2]. Ongoing
plantation, review the pharmacology of immuno- hepatitis C infection post liver transplantation
suppressive agents and provide an overview for is associated with approximately 10% risk of
treatment of liver graft rejection. graft loss. Rejection episodes requiring bolus
corticosteroid therapy however are associated
with worse outcomes in patients with hepatitis
Immunological Rejection C recurrence [3].
The incidence of acute cellular rejection was
Liver transplantation has a lower incidence of 60% in the 1990s [4]. Since 2000 it has improved
rejection compared to other organs and does not to 15% due to the introduction of new therapeutic
require HLA matching of donor and recipient options and improved management of immuno-
prior to transplantation. However, a substantial suppression [5]. Most cases occur within 90 days
number of recipients still develop graft rejection. of surgery and respond to high dose corticoste-
Table 32.1 is a summary of patterns of rejection roids [6].
seen in liver transplant recipients. Chronic cellular rejection is mediated by both
immunological and non-immunological factors.
The greatest risk factor for chronic cellular rejec-
E. Gonsalkorala, MD • D. Hotho, MD
K. Agarwal, MD (*) tion is frequency and severity of acute cellular
Institute of Liver Studies, Kings College Hospital, rejection. Age of donor and quality of liver graft
London, UK are non-immunological risk factors for chronic
e-mail: enoka.gonsalkorala@health.qld.gov.au; rejection.
daphne.hotho@gmail.com; kosh.agarwal@nhs.net

© Springer International Publishing AG, part of Springer Nature 2018 431


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_32
432

Table 32.1  Patterns of liver transplant rejection


Type Timing Incidence Pathogenesis Histology Treatment Outcome
Hyperacute Minutes– Rare Pre-sensitization to donor Endothelial injury, fibrin Urgent retransplantation
hours antigen deposition
Acute cellular 0–90 days 15–25% Inflammation induced by T Portal inflammation, bile duct High-dose corticosteroids No adverse effects on
rejection cell activation by antigen inflammation, venous endothelial ATG graft or patient
presenting cell inflammation. Fig. 32.1 Alemtuzumab outcomes, except in
HCV, who have worse
outcomes
Chronic >90 days <4% Inadequate Ductopaenia and obliterative Augmentation of Reduced graft survival
cellular immunosuppression vasculopathy affecting large and immunosuppression
rejection Donor age medium sized arteries and portal Re-transplantation in
microcirculation Fig. 32.2 [7] severe cases
Acute antibody 0–90 days 2–6%, 10% in Activation of complement Microvasculitis with diffuse Multimodal including Limited data, can be
mediated patients with pathway by donor specific portal and sinusoidal C4d plasmapheresis and B-cell associated with chronic
rejection idiopathic graft antigens staining Fig. 32.3 targeting agents rejection and graft
loss failure
Chronic >90 days Unknown Yet to be defined Yet to be defined Unknown Unknown
antibody
mediated
rejection
E. Gonsalkorala et al.
32 Immunosuppression 433

The steps involved in the development of ing cells (APC) (dendritic cells, macrophages,
acute cellular rejection are as follows (Fig. 32.2): B lymphocytes) in lymphoid organs, e.g.
spleen, regional lymph nodes. These are
• Allograft recognition—Foreign antigens are loaded onto the major histocompatibility com-
presented to lymphocytes by antigen present- plex (MHC) by the APC that are recognized
by CD3 (as well as CD4/CD8). The T-cell
receptor on CD3 interacts with the MHC of
the APC, this is stabilized by CD4/CD8,
resulting in “SIGNAL 1”, a calcium-­dependent
pathway.
• T-cell activation—This is achieved by binding
of co-stimulatory molecules (CD-28, CD-40,
PD1) on T-cells with ligands on the antigen
presenting cell—“SIGNAL 2”, a Ca2+-
independent process. Both signals are required
for naïve T-cell activation and are mediated by
calcineurin and Protein Kinase C activation of
nuclear factor of activated T cells (NF-AT),
nuclear factor kB (NF-KB) and AP-1. These
bind to gene promoters associated with T-cell
Fig. 32.1  H&E section shows a portal tract completely activation and proliferation, i.e. promotes IL2
occupied by a florid mixed inflammatory cell infiltrate, production which initiates G0 to G1 transition
including eosinophils, and involving bile ducts. of the cell cycle [8]. Inhibition of this pathway
Endotheliitis is present in the lower half of the field. H&E
x 20. (Courtesy of Dr Alberto Quaglia, Institute of Liver
has been the predominant site of action in
Studies, King’s College Hospital) immunosuppression therapies utilizing calci-

T CELL
OKT3

SIGNAL 1
CD3
Fig. 32.2 Mechanism MHC
of allograft rejection and
of immunosuppressive
drugs. APC antigen
Tacrolimus NFAT
presenting cell, MHC Calcineurin
Cyclosporine
major histocompatibility Protein kinase C NFKB
complex, IL2R AP1
interleukin-20 receptor,
IL-2 interleukin 2, NFAT SIGNAL 2
nuclear factor of CD80 CD28 Azathioprine
APC
activated T-cells, NFKB MMF
nuclear factor kappa-­ IL-2
light-­chain-enhancer of
Cell
activated B cells, AP1
cycle
activator protein 1,
mTOR mammalian
target of rapamycin, MAP
SIGNAL 3 STAT5
MAP mitogen-activated IL2R
protein, STAT5 signal mTOR
transducer and activator
of transcription 5, OKT
orthoclone, MMF Daclizumab Sirolimus
mycophenylate Basiliximab Everolimus
434 E. Gonsalkorala et al.

Anti-HLA class I DSA form complexes with


soluble class I-MHC antigens released by the
donor liver and are removed from circulation
from liver Kupffer cells [11], providing further
protection against AMR. Additionally, AMR
can occur in the presence of acute cellular rejec-
tion. AMR is suspected in setting of unexplained
graft dysfunction with thrombocytopenia, hypo-
complementemia and microvasculitis with dif-
fuse C4d staining on liver biopsy (Fig. 32.3)
[12]. Whilst AMR is well recognized after renal
and cardiac transplantation, its role in graft
rejection post liver transplantation has only
Fig. 32.3  Immunohistochemistry for C4d show stromal been recognized recently. The revised Banff cri-
and vascular endothelial stain in a portal tract. (Courtesy
of Dr Lara Neves-Souza, Institute of Liver Studies, King’s teria for AMR diagnosis requires histopatholog-
College Hospital) ical pattern of injury consistent with AMR,
positive serum DSA, diffuse microvascular C4d
deposition and exclusion of other causes that
neurin inhibitors (CNIs) such as tacrolimus may cause graft dysfunction [13].
and cyclosporine. Steps in the development of antibody medi-
• Clonal expansion—“SIGNAL3”: auto/para- ated rejection [14] [15].
crine activation of T-cells. Receptor of the IL2
family activates JAK 1/3 in T-cells [9] which • Donor specific antigens can be present at time of
activates mammalian target of rapamycin transplantation or develop post-­transplantation
(mTOR), STAT5 and Ras-Raf MAP kinase (de novo DSA). These a­ ntigens bind to HLA on
resulting in cell proliferation, DNA synthesis graft endothelium. DSA can be directly mea-
and cell division. Sirolimus and everolimus sured in the serum.
inhibit SIGNAL 3. Other molecules are pro- • Classical complement pathway activation
duced that inhibit SIGNAL 2 (e.g. CD152) occurs when plasma C1q attaches to Fc seg-
and decrease T-cell receptor signaling [10]. ment on DSA
Azathioprine and mycophenolate mofetil • A series of enzymatic reactions involving deg-
inhibit purine and DNA synthesis. radation of C2 and C4 follows. One of the
• Inflammation—Activated T cells result in byproducts of C4 degradation, C4d, is depos-
release of cytokines that recruit cytotoxic ited on the allograft and can be detected
T-cells, B-cells, activated macrophages and through immunohistochemistry staining of
adhesion molecules. Further activated T-cells liver biopsy specimen.
are attracted by these leading to release of • Degradation products of C2 and C4 ultimately
TNF α/β perforin, granzymes. Corticosteroids leads to formation of C3b which then activates
and antilymphocyte antibody act via inhibi- C5 and allows formation of membrane attack
tion of this route. complexes. The end result is endothelial dam-
age and inflammation.
Antibody mediated rejection (AMR) is due
to de-novo or pre-existing anti-HLA donor-spe-
cific antibodies (DSA) and is seen in acute Immunosuppressive Agents
rejection with reported association with chronic
rejection. The liver has been considered to be Most immunosuppressive regimens use a combina-
resistant to AMR due to large organ size and tion of drugs with different sites of action in the
dual blood supply (arterial and portal) [11]. T-cell response pathway. This enables variable dos-
32 Immunosuppression 435

Table 32.2  Side effects of common immunosuppression dependent phosphatase. This prevents the
medication
dephosphorylation of activated T-cells that inhib-
Drug Common adverse effects its their nuclear entry and thus upregulation of
Tacrolimus Nephrotoxicity, neurotoxicity, proinflammatory cytokines including IL-2
hypertension, diabetes, metabolic (Signal 2 pathway) [19].
acidosis
Tacrolimus inhibits calcineurin by binding to
Cyclosporine Nephrotoxicity, neurotoxicity,
hypertension, diabetes, metabolic FK-binding protein-12. This in turn binds to a
acidosis, hyperlipidemia, gingival separate site to cyclosporine/cyclophilin on calci-
hyperplasia, hypertrichosis neurin resulting in a similar inhibitory pathway
Corticosteroids Hypertension, diabetes, for IL-2 production. These two drugs cannot be
osteoporosis, obesity, cataracts,
used simultaneously as they compete with other
poor wound healing
Mycophenolate Myelosuppression, diarrhea, viral
for immunosuppressive action.
mofetil infections
Sirolimus Hepatic artery thrombus, poor  harmacokinetics and Metabolism
P
wound healing, hyperlipidemia, The original formulation of cyclosporine was as
myelosuppression, pneumonitis, Sandimmune. It is a corn oil based agent with a
rash
highly variable absorption and an average bio-
availability of 10%. Absorption was dependent
age and treatment adjustment according to response on the presence of bile salt availability and the
and adverse effects. The current mainstay of treat- use of T-tubes that interrupted enterohepatic cir-
ment involves the use of calcineurin inhibitors culation after transplantation necessitated intra-
(CNI) in combination with steroids. There is an venous administration. A microemulsion form,
increasing use of tailored protocols individualized Neoral, was subsequently developed and adopted
to the patient and etiology to stratify risk of rejec- into regular practice. This formulation is less
tion and protect long-­ term graft function while dependent on bile acids for absorption resulting
minimizing adverse effects. See Table 32.2 for an in improved overall bioavailability. Distribution
overview of currently used immunosuppressive is concentration dependent and is predominantly
agents and their adverse effects. in adipose, adrenal, hepatic, pancreatic and renal
tissue. In blood it is primarily bound to lipopro-
teins. The half-life is 18 h and it is mainly
Calcineurin Inhibitors: Cyclosporine excreted into bile [20].
and Tacrolimus Tacrolimus is well absorbed from the gastro-
intestinal tract with a bioavailability in liver
Cyclosporine was the first CNI to be routinely transplant patients of approximately 22%. The
used post transplantation. It was derived from rate of absorption is best under fasting condi-
the fungus Tolyplocladium inflatum in 1972 tions. It is 95% bound to erythrocytes, with 99%
and was evaluated for use as an immunosup- of the remaining 5% bound to plasma proteins.
pressive agent in 1976 [16]. Its use has now Less than 0.1% is unbound, and it is this fraction
often been superceded by Tacrolimus (FK506) that exerts the pharmacological activity [21]. The
which is approximately 100 times more potent half-life varies from 31 to 48 h.
on a molar level [17]. Tacrolimus is a macrolide CNIs are metabolized by the cytochrome
antibiotic similar to erythromycin that was P450 3A4 (CYP3A4) enzyme in the gastrointes-
derived from the fungus Streptomyces tsuku- tinal epithelium (approximately 50%) and the
baensis in 1984 [18]. liver where first pass hepatic metabolism accounts
for a further 10%. The metabolites have minimal
Method of Action immunosuppressive effects. Drugs that interact
Cyclosporine binds to cyclophilin that causes with CYP3A4 will affect the concentration of
inhibition of calcineurin, a calcium/calmodulin-­ CNIs (Table 32.3).
436 E. Gonsalkorala et al.

Table 32.3  Drugs that increase and decrease CNI and mon are convulsions, confusion, psychosis and
sirolimus levels
reduced consciousness.
Increase levels Decrease levels Metabolic effects: Diabetes, hyperlipidaemia,
Calcium antagonists Anticonvulsants hyperkalaemia and metabolic acidosis are fre-
    Verapamil, nifedipine, Phenytoin, quently observed. Gingival hyperplasia and
diltiazem carbamazepine,
hypertrichosis are specific to cyclosporine [27].
henobarbital
Antifungals Antibiotics
    Fluconazole, itraconazole, Rifampicin, rifabutin
Clinical Use
etoconazole, voriconazole, Tacrolimus (Prograf™) has mostly superseded
clotrimazole cyclosporine as the first line drug in liver trans-
Macrolides St. John’s wort plantation. Several studies have demonstrated a
    Azithromycin, lower incidence of acute cellular rejection with
erythromycin, tacrolimus compared to cyclosporine with simi-
clarithromycin
lar patient and graft survival, and tacrolimus is
Protease inhibitors
usually the first choice CNI in de novo trans-
    E.g. ritonavir, darunavir,
saquinavir plants [28–30].
Metoclopramide In the immediate post-operative period tacro-
Amiodarone limus can be administered orally or via an oro- or
nasogastric tube if the patient remains intubated,
usually at a starting dose of 1–2 mg twice daily. It
Adverse Effects is given in combination with intravenous steroid.
Major long-term adverse effects are related to Levels are checked and the dose is adjusted
nephrotoxicity. CNIs cause a reduction in renal accordingly.
blood flow and GFR by vasoconstriction of the
afferent renal arteriole [22]. Longitudinal studies  herapeutic Drug Monitoring
T
of liver transplant patients with chronic renal The immunosuppressive effects of CNIs are
insufficiency demonstrate that CNI toxicity is the related to the total drug exposure that is repre-
most common clinical and histologic diagnosis in sented by the area under the drug-concentration-­
patients who progress to end stage renal failure time curve (AUC). Both drugs have a narrow
after transplant [23]. Both cumulative dose and therapeutic window. For tacrolimus, the 12-h
duration of CNI exposure are related to the degree trough concentration is a good estimation of the
of renal injury. These changes are reversible in AUC: and blood samples taken 10–14 h after
the short term however nearly 20% of liver trans- dosage are predictive of exposure [31]. There is
plant recipients go on to develop renal failure no clear consensus as to the optimal dosing regi-
within 5 years [24]. This is a major clinical issue men in transplantation. In the past levels as high
in post transplant care and the concern about as 10–20 ng/mL in the first post transplant month
renal toxicity has led to CNI sparing regimes in have been recommended. However, there is now
patients with pre-existing renal dysfunction. increasing evidence for lower tacrolimus levels
Hypertension is commonly seen, often due to post liver transplantation. Trough concentration
the renal changes [25] and amlodipine is the drug between 6 and 10 ng/mL in the first 4–6 weeks
of choice used to treat CNI-induced hyperten- post transplantation with a reduction to 4–8 ng/
sion. Neurotoxicity is potentiated by low magne- mL long term has been recommended [32].
sium levels and often improves with magnesium Levels are adjusted according to renal function
supplementation [26]. Tremor, headache and and the presence or absence of rejection. A new
insomnia are the other adverse effects. Less com- once daily formulation of tacrolimus
32 Immunosuppression 437

(Advagraf™) has recently been introduced. operative period after a bolus of 500 mg methyl-
Once-daily dosing may improve compliance prednisolone in the operating room.
while allowing the same total daily dose and Methylprednisolone is continued until enteral
monitoring strategies [33]. administration is possible and the dose is then
converted to prednisolone. The aim is to taper the
dose gradually depending on the overall response
Corticosteroids to immunosuppression and etiology of the under-
lying liver disease. Withdrawal within 3–6 months
Corticosteroids are the most frequently used non-­ in those with no evidence of rejection or autoim-
CNI drug immunosuppressants in liver transplan- mune disease is often successful [34]. High dose
tation and pulse dose methylprednisolone pulsed steroids are used to treat acute cellular
remains the first line treatment for acute cellular rejection. Typically hydrocortisone 100 mg daily
rejection. Corticosteroids were initially used in for 3 days or methylprednisolone 500 mg daily
high doses in the early era of transplantation and for 2 days is administered in conjunction with an
resulted in inevitable high morbidity. The current increased dose of tacrolimus.
practice is based upon their use as induction ther-
apy with early dose reduction and possible with-
drawal due to the myriad adverse effects. Antimetabolites: Azathioprine
and Mycophenolate Mofetil (MMF)

Method of Action Antimetabolites were not initially used in liver


transplantation. They were used as part of strate-
Corticosteroids have a wide variety of immuno- gies to reduce the frequency of CNI related renal
modulatory and anti-inflammatory actions. They failure and to treat refractory rejection.
bind to glucocorticoid receptors resulting in inhibi- Azathioprine is the pro-drug form of
tion of gene transcription of pro-inflammatory 6-­mercaptopurine (6-MP) that is then converted to
cytokines including IL-2, IL-6, TNF-α and IFN-­γ. 6-thioguanine, 6-methyl-MP and 6-thiouric acid.
These cytokines are required for the macrophage These active compounds interfere with DNA repli-
and lymphocyte response to allograft antigens. In cation. Thiopurine methyltransferase (TPMT) is the
addition, there is direct suppression of complement enzyme required for the conversion of azathioprine
and antibody binding, stabilization of lysosomal to 6-MP. Polymorphisms of TPMT exist that cause
enzymes, suppression of prostaglandin synthesis decreased activity and allow toxic level of azathio-
and reduction of histamine and bradykinin release. prine to build up resulting in acute myelosuppres-
sion [35]. It is therefore essential to check TPMT
activity prior to commencing therapy. Further
Adverse Effects metabolism is via xanthine oxidase and therefore it
must not be used with allopurinol, a xanthine oxi-
These are well known and summarized in dase inhibitor, as toxicity will be potentiated.
Table 32.2. Use in liver transplantation has been limited
due to adverse effects including liver toxicity,
cholestatic jaundice, hepatic veno-occlusive dis-
Clinical Use ease, hypersensitivity, pancreatitis and bone mar-
row suppression, particularly in patients with
Typical regimens use methylprednisolone portal hypertension. It is currently used primarily
10–50 mg intravenously in the immediate post-­ as adjunctive therapy.
438 E. Gonsalkorala et al.

Mycophenolate Mofetil is derived from pain [39]. Bone marrow suppression can also
Penicillium and was first discovered in 1893 occur. If these adverse effect do not improve
however its evaluation as an immunosuppressant with dose reduction, MMF should be stopped.
was not until the 1990s [36]. Two forms are avail- There is also an increased incidence of viral
able: MMF (CellCept, Roche) and enteric coated and fungal infections including cytomegaly
mycophenolate sodium (Myfortic, Novartis). virus (CMV), herpes simplex virus (HSV) and
candida with the use of MMF. Its use is not rec-
ommended in pregnancy due to the increased
Method of Action risk of congenital malformation and spontane-
ous abortion.
The active compound is mycophenolate acid
(MPA). MPA inhibits the action of inosine mono-
phosphate dehydrogenase (IMDPH), the rate-­ Clinical Use
limiting enzyme in the synthesis of guanosine
nucleotides which are essential for DNA synthe- Predominant use is as a CNI-sparing agent as
sis. Most cell types have a second pathway for MMF is not nephrotoxic. It is more frequently
nucleotide synthesis, however lymphocytes do used in patients requiring additional long-term
not possess such activity. There are also two iso- immunosuppression e.g. following documented
forms of the IMDPH enzyme. The second previous rejection [40]. MMF has replaced aza-
­isoform is more prominent in lymphocytes, and thioprine as it is associated with a lower inci-
has preferential selectivity for MMF [37]. dence of biopsy proven rejection when combined
with CNI [41]. There is no role of MMF as mono-
therapy due to the high incidence of ACR, steroid
Pharmacokinetics and Metabolism resistant rejection and chronic rejection requiring
re-transplantation [42].
MMF is well absorbed from the gastrointestinal
tract and undergoes immediate hepatic first-pass Therapeutic Drug Monitoring
metabolism to MPA. The half-life is approxi-
mately 18 h with bioavailability estimated at The data to support monitoring is of limited qual-
90%. Food decreases MPA concentration so ity as drug levels and effects are affected by a vari-
MMF should be administered at least 1 h before ety of factors including serum protein levels, other
or 2 h after eating. MPA is 97% protein bound, immunosuppressive agents and renal function
with free MPA as the active fraction. MPA is fur- leading to significant inter-patient variability [43].
ther metabolized by the liver to mycophenolic
acid glucuronide (MPAG) that has 93% urinary
elimination. Liver disease impairs MPA conjuga-
tion, thus increasing its half-life. MPAG is also  TOR Inhibitors: Sirolimus
m
excreted into bile. Further hydrolysis back to and Everolimus
MPA by gut organisms leads to enterohepatic
recirculation of MPA and a second peak concen- The two mammalian target of rapamycin (mTOR)
tration 6–12 h post ingestion [38]. inhibitors licensed for use in transplantation are
sirolimus and everolimus. Sirolimus was discov-
ered in soil samples from Easter Island (Rapa Nui)
Adverse Effects in 1964 and initially developed as an anti-­fungal
[44]. It is structurally similar to tacrolimus and is a
The most common dose related adverse effect naturally occurring product of streptomyces
is diarrhea. Other gastrointestinal adverse hygroscopicus. Everolimus is a chemically modi-
effects include nausea, vomiting and abdominal fied form of sirolimus to improve absorption.
32 Immunosuppression 439

Method of Action introduction of sirolimus may be most benefi-


cial to prevent progression of renal complica-
Sirolimus and everolimus bind to the FK-binding tions of CNI.
protein-12 but do not inhibit calcineurin. Instead Sirolimus has a potential anti-tumor effect:
they inhibit mTOR that is required for mRNA patients transplanted with HCC have been found
translation necessary for cell cycle progression, to have a prolonged survival with sirolimus com-
(which is halted in the G1 phase), IL-2 production pared to CNI [47] but further confirmatory stud-
and cellular proliferation. T-cell activation occurs, ies are required.
but IL-2 induced proliferation does not occur.

Therapeutic Drug Monitoring


Pharmacokinetics and Metabolism
Sirolimus levels are measured by either immu-
Sirolimus is a highly lipophilic compound that is noassay or chromatography. It is essential that
readily absorbed when in oily solution or the same method is consistently used. Trough
­microemulsion (bioavailability 14–18%). It has a levels <6 ng/mL are associated with an
half-­
life of 62 h and reaches steady state in increased incidence of rejection; levels >15 ng/
5–7 days. The long half-life necessitates regular mL have an increased risk of hyperlipidemia
drug monitoring. It is extensively bound to plasma and thrombocytopenia [48]. Trough levels
proteins and metabolized by CYP3A4 (Table 32.3) obtained 5–7 days after dose adjustment are
in the intestine and liver. Most of the metabolites sufficient due to the long half-life of sirolimus.
are excreted in feces via a P-glycoprotein pump.

Antibody-Based Therapies
Adverse Effects
These are generally utilized as induction of
Hyperlipidaemia, thrombocytopenia, anemia immunosuppression or as salvage for steroid
and leucopenia are commonly seen. Less fre- refractory rejection.
quent adverse effects include aphthous ulcer-
ation, acne, arthralgia and intersitital pneumonitis
(that resolves on withdrawal) [45]. Specifically  olyclonal Antibodies: Anti-­
P
in liver transplantation, an increased incidence Thymocyte and Anti-Lymphocyte
of hepatic artery thrombosis and wound dehis- Globulin
cence in the first month post transplant has been
reported [46]. These agents are prepared by inoculation of rab-
bits with human lymphocytes or thymocytes.
A purified gamma globulin fraction of anti-sera is
Clinical Use used to prevent serum sickness. They were first
used in the early era of transplantation with ste-
Studies of mTOR inhibitors as monotherapy roids and azathioprine prior to the introduction of
have demonstrated the possibility of an CNI. Polyclonal antibodies are currently used as
increased risk of hepatic artery thrombosis and an induction agent, a steroid-sparing agent or as
poor wound healing. There is also a higher inci- treatment of steroid-resistant rejection. Their
dence of rejection. Current practice is for intro- action is on multiple T-cell antigens, B-cell anti-
duction as combination therapy with tacrolimus gens, HLA class 1 and 2, macrophages and NK
in patients requiring broader immunosuppres- cells causing lymphocyte depletion [49].
sion or as a replacement monotherapy for Adverse effects include fever, hypotension,
patients intolerant of CNIs. In particular, early headache, aseptic meningitis, ARDS, pulmonary
440 E. Gonsalkorala et al.

edema, graft thrombosis. Steroids, antihistamines Induction


and acetaminophen are given as pretreatment to
counteract these adverse effects. High dose intravenous steroids are administered
during transplantation and continued for 2–3 days
until the patient is able to tolerate oral intake.
Monoclonal Antibodies Induction immunosuppression post liver trans-
plantation consists of intravenous methylpred-
Anti IL-2 (CD 25) receptor antibodies such as nisolone at doses of 500–1000 mg followed by a
daclizumab or basiliximab are used as induction taper according to local practice. Once the patient
therapy to prevent rejection, especially in cases is able to tolerate oral intake, methylprednisolone
with renal dysfunction peri-transplantation as is ceased and prednisolone is commenced, usu-
they allow lower or later start of nephrotoxic CNI ally at a dose of approximately 20 mg daily.
[50]. Various protocols are in use. Typically, anti T-cell depleting (anti-thymocyte globulin
IL-2 (CD 25) receptor antibodies are adminis- (ATG), alemtuzumab) and non-depleting (basilix-
tered on the first post-operative day and then imab, daclizumab) antibodies can be used as an
4–7 days post transplant and they remain in circu- induction agent, either in conjunction or as an
lation for several weeks. There are few adverse alternative to steroids. These strategies are not
effects and they are generally very well tolerated. common and are used in select cases under the
OKT3 (muromonab-CD3) binds to the CD3 guidance of an experienced transplant physicians.
receptor on mature T-cells, preventing signal 1
activation and depletion of lymphocytes by T-cell
lysis and cytokine release [51]. Adverse effects Maintenance
are similar to ATG but OKT3 is less well tolerated
with a higher incidence of post-transplant lym- Maintenance immunosuppression with a calcineu-
phoproliferative disease (PTLD). Administration rin inhibitor (CNI) (tacrolimus or cyclosporine) is
is by intravenous infusion and onset of action is commenced within the first 24–48 h post-trans-
within minutes, lasting 1 week. It is commonly plantation. In the absence of acute cellular rejection
used to treat steroid-resistant acute rejection and in the immediate post-transplant period, predniso-
requires premedication antibodies with steroids, lone is weaned and often ceased 3 months post
antihistamines and acetaminophen, similar to transplantation. Patients are then frequently main-
polyclonal antibodies. tained on a CNI as sole immunosuppression agent.
Campath (Alemtuzumab) is a humanized anti Tacrolimus is the preferred CNI over cyclosporine
CD52 monoclonal antibody that causes lympho- due to reduced incidence of graft loss, acute cellu-
cyte depletion from the circulation and peripheral lar rejection and steroid resistant rejection [52]. In
nodes. Its role in immunosuppressive regimens is the event of significant CNI toxicity (Table 32.2), a
not yet identified, but it can be used as induction strategy involving reduction in the dose of CNI and
therapy to facilitate lower doses of CNI and in commencement of an anti-metabolite agent (myco-
conjunction with sirolimus. phenolate or azathioprine) or mTOR inhibitor
(sirolimus or everolimus) is adopted.

 pproach to Immunosuppression
A
Post Liver Transplantation Special Situations

Each liver transplant center will have their own Hepatitis C


established protocols on immunosuppression
regimes. Below is an overview of a broad approach In industrialized countries hepatitis C is now the
according to timing post liver transplantation, spe- single most common reason for liver transplanta-
cial situations and treatment of graft rejection. tion. Re-infection of the graft is almost universal
32 Immunosuppression 441

[53] and occurs in the immediate post-transplant mortality [63]. Longitudinal studies of liver
period. High dose steroid therapy for acute rejec- transplant patients with chronic renal insuffi-
tion causes an increase in viremia and more rapid ciency demonstrate that CNI toxicity is clinically
progression of disease recurrence [54]. Ten to and histologically the most common cause in
thirty percent develop cirrhosis at 5 years post patients who progress to end stage renal disease
transplantation [55]. Strategies used include early [23]. A number of strategies have been employed
steroid withdrawal and the combination of induc- to minimize the dose of CNI in the immediate
tion therapy with IL-2 blockade [56]. Some post transplant period in patients at risk of devel-
­in-­vitro studies suggest that cyclosporine instead oping renal injury, principally those with pre-­
of tacrolimus has an inhibitory effect on replica- existing renal dysfunction. Minimizing early
tion but the concentrations used in these replica- acute CNI-induced renal injury will reduce the
tion studies were greater than 1000 times of incidence of acute and chronic renal disease later
physiological concentration [57]. Furthermore after transplant. Induction immunosuppression
cyclosporine is less diabetogenic than tacrolimus with IL-2 receptor blockers or ATG, and delayed
and diabetes is considered a risk factor for fibro- or reduced dose start of CNI is commonly part of
sis progression post transplant for hepatitis C renal protective protocols. Some centers adopt
[58]. Treatment of hepatitis C either pre or post-­ the approach of converting from CNI to mTOR
transplant setting is now occurring with new oral-­ inhibitors in patient with acute kidney injury.
only direct acting agents (so called DAA agents).
These regimes have the advantage of very high
cure rates with few side effects. Some DAA Hepatocellular Carcinoma
regimes have minimum effect on immunosup-
pression levels and reinfection due to high levels Liver transplantation is a curative management
of immunosuppression may be less concerning if option for patients with hepatocellular carcinoma
hepatitis C is promptly treated. (HCC) and is an indicator for transplantation.
Model for end stage liver disease score (MELD)
of 15 is one of the minimum criteria required for
Autoimmune Hepatitis listing for liver transplantation and exception
points are allocated to those with HCC. Tumor
Incidence of recurrence of autoimmune hepatitis recurrence post liver transplantation is approxi-
in the new graft is approximately 25% [59, 60]. A mately 10–20% and the type of immunosuppres-
maintenance immunosuppression regime that sive agent used is one of many factors which
includes prednisolone may reduce the risk of influences recurrence. High serum levels of CNI,
recurrence [61] and therefore prednisolone is prolonged steroid use, anti-lymphocytic antibody
often continued in addition to CNIs. and ATG has been associated with increased risk
of HCC recurrence. mTOR inhibitors, which
have anti-neoplastic effects, may improve recur-
Renal Dysfunction rence rates when compared with non-mTOR
inhibitor regimes [64–66]. A randomized control
Renal dysfunction and acute kidney injury after study to address this very question is currently
liver transplantation is common and has impor- underway [67].
tant implications for subsequent patient morbid-
ity and survival. Ten to sixty percent of liver
transplant recipients develop postoperative acute Graft Rejection
kidney injury and 10–25% require postoperative
renal replacement therapy [62]. The need for Once acute cellular rejection has been diagnosed
postoperative renal replacement is associated treatment is instituted with high dose intravenous
with a two to sixfold increased risk of 1-year prednisolone (usually methylprednisolone at
442 E. Gonsalkorala et al.

500–1000 mg per day) and continued for 3 days. 3. Crespo G, Marino Z, Navasa M, Forns X. Viral
hepatitis in liver transplantation. Gastroenterology.
Patients are then switched to high oral steroids.
2012;142(6):1373–83.e1.
Approximately 80% of ACR will respond to high 4. Neuberger J. Incidence, timing, and risk factors for
dose prednisolone [68, 69]. The rest may require acute and chronic rejection. Liver Transpl Surg.
alternatives such as ATG or alemtuzumab. 1999;5(4 Suppl 1):S30–6.
5. Beaudreuil S, Samuel D, Rouas-Freiss N, Durrbach
Following resolution of ACR a regime that
A. New aspect of immunosuppressive treatment in
involves adding either an anti-metabolite or liver transplantation. How could you induce toler-
mTOR inhibitor to CNI may be adopted. ance in liver transplantation? Transpl Immunol.
Initial management of AMR is the same as 2007;17(2):98–107.
6. Wiesner RH, Ludwig J, Krom RA, Hay JE, van Hoek
that for ACR. Due to the paucity of evidence,
B. Hepatic allograft rejection: new developments in
presently there are no established protocols spe- terminology, diagnosis, prevention, and treatment.
cifically for treatment of AMR [70]. Instead indi- Mayo Clin Proc. 1993;68(1):69–79.
vidual centers will have their own approach to 7. Demetris AJ, Murase N, Lee RG, Randhawa P, Zeevi
A, Pham S, Duquesnoy R, Fung JJ, Starzl T. Chronic
management of AMR. The concepts underlying
Rejection. A general overview of histopathology
treatment of acute AMR are suppression of T-cell and pathophysiology with emphasis on liver, heart
response, elimination of circulating antibodies, and intestinal allografts. Annals of Transolantation
inhibition of residual antibodies and suppression 1997;2(2):27–44.
8. Johnston JA, Wang LM, Hanson EP, Sun XJ, White
or depletion of B-cells [71]. Limited studies have
MF, Oakes SA, et al. Interleukins 2, 4, 7, and 15
shown varying degrees of success with plasma- stimulate tyrosine phosphorylation of insulin receptor
phoresis, IV immunoglobulins, rituximab and substrates 1 and 2 in T cells. Potential role of JAK
proteasome inhibitors [70]. Due to paucity of kinases. J Biol Chem. 1995;270(48):28527–30.
9. Wells AD. Cell-cycle regulation of T-cell responses—
research there are presently no guidelines for
novel approaches to the control of alloimmunity.
management of chronic AMR. Immunol Rev. 2003;196:25–36.
In an era when organ shortage is amplified by 10. Kim EY, Lee EN, Lee J, Park HJ, Chang CY, Jung
increasing use of grafts for patients with significant DY, et al. Two-signal blockade with anti-CD45RB
and anti-CD154 monoclonal antibodies inhibits
co-morbidities or for retransplantations, marginal
graft rejection via CD4-dependent mechanisms
(so called) extended criteria grafts are frequently in allogeneic skin transplantation. Exp Mol Med.
utilized. This necessitates individualized immuno- 2006;38(3):284–94.
suppression protocols. Whilst the main aim of 11. Del Bello A, Congy-Jolivet N, Danjoux M, Muscari
F, Kamar N. Donor-specific antibodies and liver trans-
immunosuppression is to prevent graft rejection,
plantation. Hum Immunol. 2016;77(11):1063–70.
there is an emphasis now on minimizing immuno- 12. O’Leary JG, Cai J, Freeman R, Banuelos N, Hart
suppression-induced complications. It is now rec- B, Johnson M, et al. Proposed diagnostic criteria
ognized that “less is more” with lower levels of for chronic antibody-mediated rejection in liver
allografts. Am J Transplant. 2016;16(2):603–14.
immunosuppression as the long-term target.
13. Demetris AJ, Bellamy C, Hubscher SG, O’Leary J,
Bearing this in mind, there is increasing interest Randhawa PS, Feng S, et al. 2016 Comprehensive
and research how to achieve complete withdrawal update of the Banff working group on liver allograft
of immunosuppression in patients who have been pathology: introduction of antibody-mediated rejec-
tion. Am J Transplant. 2016;16:2816–35.
stable for years after liver transplantation.
14. Stites E, Le Quintrec M, Thurman JM. The comple-
ment system and antibody-mediated transplant rejec-
tion. J Immunol. 2015;195(12):5525–31.
References 15. Stegall MD, Chedid MF, Cornell LD. The role

of complement in antibody-mediated rejec-
tion in kidney transplantation. Nat Rev Nephrol.
1. Banff schema for grading liver allograft rejection:
2012;8(11):670–8.
an international consensus document. Hepatology.
16.
Borel JF, Feurer C, Gubler HU, Stahelin
1997;25(3):658–63.
H. Biological effects of cyclosporin A: a new
2. Wiesner RH, Demetris AJ, Belle SH, Seaberg EC,
antilymphocytic agent. 1976. Agents Actions.
Lake JR, Zetterman RK, et al. Acute hepatic allograft
1994;43(3–4):179–86.
rejection: incidence, risk factors, and impact on out-
17. Geissler EK, Schlitt HJ. Immunosuppression for liver
come. Hepatology. 1998;28(3):638–45.
transplantation. Gut. 2009;58(3):452–63.
32 Immunosuppression 443

18. Goto T, Kino T, Hatanaka H, Nishiyama M, Okuhara once-­daily tacrolimus administration in liver trans-
M, Kohsaka M, et al. Discovery of FK-506, a novel plant patient. Transplant Proc. 2010;42(4):1322–4.
immunosuppressant isolated from Streptomyces tsu- 34. Greig P, Lilly L, Scudamore C, Erb S, Yoshida E,
kubaensis. Transplant Proc. 1987;19(5 Suppl 6):4–8. Kneteman N, et al. Early steroid withdrawal after
19.
Taylor AL, Watson CJ, Bradley JA. liver transplantation: the Canadian tacrolimus versus
Immunosuppressive agents in solid organ transplanta- microemulsion cyclosporin A trial: 1-year follow-up.
tion: mechanisms of action and therapeutic efficacy. Liver Transpl. 2003;9(6):587–95.
Crit Rev Oncol Hematol. 2005;56(1):23–46. 35. Lennard L, Van Loon JA, Weinshilboum RM.

20. Mukherjee S, Mukherjee U. A comprehensive review Pharmacogenetics of acute azathioprine toxicity:
of immunosuppression used for liver transplantation. relationship to thiopurine methyltransferase genetic
J Transplant. 2009;2009:701464. polymorphism. Clin Pharmacol Ther. 1989;46(2):
21. Undre NA. Pharmacokinetics of tacrolimus-based
149–54.
combination therapies. Nephrol Dial Transplant. 36. Klupp J, Bechstein WO, Platz KP, Keck H, Lemmens
2003;18(Suppl 1):i12–5. HP, Knoop M, et al. Mycophenolate mofetil added to
22.
Shihab FS. Cyclosporine nephropathy: patho- immunosuppression after liver transplantation—first
physiology and clinical impact. Semin Nephrol. results. Transplant Int. 1997;10(3):223–8.
1996;16(6):536–47. 37. Allison AC, Eugui EM. Purine metabolism and

23. Velidedeoglu E, Crawford MD, Desai NM, Campos immunosuppressive effects of mycophenolate mofetil
L, Abt PL, Markmann JW, et al. Predictors of late kid- (MMF). Clin Transpl. 1996;10(1 Pt 2):77–84.
ney dysfunction post-liver transplantation. Transplant 38. Bullingham RE, Nicholls AJ, Kamm BR. Clinical
Proc. 2002;34(8):3315–6. pharmacokinetics of mycophenolate mofetil. Clin
24. Ojo AO, Held PJ, Port FK, Wolfe RA, Leichtman Pharmacokinet. 1998;34(6):429–55.
AB, Young EW, et al. Chronic renal failure after 39. Sollinger HW. Mycophenolates in transplantation.

transplantation of a nonrenal organ. N Engl J Med. Clin Transpl. 2004;18(5):485–92.
2003;349(10):931–40. 40. Pfitzmann R, Klupp J, Langrehr JM, Uhl M, Neuhaus
25. Luke RG. Mechanism of cyclosporine-induced hyper- R, Settmacher U, et al. Mycophenolatemofetil for
tension. Am J Hypertens. 1991;4(5 Pt 1):468–71. immunosuppression after liver transplantation: a
26. Eidelman BH, Abu-Elmagd K, Wilson J, Fung JJ, follow-up study of 191 patients. Transplantation.
Alessiani M, Jain A, et al. Neurologic complications 2003;76(1):130–6.
of FK 506. Transplant Proc. 1991;23(6):3175–8. 41. Wiesner R, Rabkin J, Klintmalm G, McDiarmid S,
27. Reznik VM, Jones KL, Durham BL, Mendoza
Langnas A, Punch J, et al. A randomized double-blind
SA. Changes in facial appearance during cyclosporin comparative study of mycophenolate mofetil and aza-
treatment. Lancet. 1987;1(8547):1405–7. thioprine in combination with cyclosporine and corti-
28. U.S. Multicenter FK506 Liver Study Group. A com- costeroids in primary liver transplant recipients. Liver
parison of tacrolimus (FK 506) and cyclosporine Transpl. 2001;7(5):442–50.
for immunosuppression in liver transplantation. The 42. Stewart SF, Hudson M, Talbot D, Manas D, Day

U.S. Multicenter FK506 Liver Study Group. N Engl J CP. Mycophenolate mofetil monotherapy in liver
Med. 1994;331(17):1110–5. transplantation. Lancet. 2001;357(9256):609–10.
29. O’Grady JG, Burroughs A, Hardy P, Elbourne D, 43. Knight SR, Morris PJ. Does the evidence support
Truesdale A, UK and Republic of Ireland Liver the use of mycophenolate mofetil therapeutic drug
Transplant Study Group. Tacrolimus versus micro- monitoring in clinical practice? A systematic review.
emulsified ciclosporin in liver transplantation: Transplantation. 2008;85(12):1675–85.
the TMC randomised controlled trial. Lancet. 44. Sehgal SN. Sirolimus: its discovery, biological prop-
2002;360(9340):1119–25. erties, and mechanism of action. Transplant Proc.
30. Pichlmayr R, Winkler M, Neuhaus P, McMaster P, 2003;35(3 Suppl):7S–14S.
Calne R, Otto G, et al. Three-year follow-up of the 45. Haydar AA, Denton M, West A, Rees J, Goldsmith
European Multicenter Tacrolimus (FK506) Liver DJ. Sirolimus-induced pneumonitis: three cases
Study. Transplant Proc. 1997;29(5):2499–502. and a review of the literature. Am J Transplant.
31. Bottiger Y, Undre NA, Sawe J, Stevenson PJ, Ericzon 2004;4(1):137–9.
BG. Effect of bile flow on the absorption of tacroli- 46. Fung J, Marcos A. Rapamycin: friend, foe, or misun-
mus in liver allograft transplantation. Transplant Proc. derstood? Liver Transpl. 2003;9(5):469–72.
2002;34(5):1544–5. 47. Zimmerman MA, Trotter JF, Wachs M, Bak T,

32. Rodriguez-Peralvarez M, Germani G, Darius T, Lerut Campsen J, Skibba A, et al. Sirolimus-based
J, Tsochatzis E, Burroughs AK. Tacrolimus trough immunosuppression following liver transplanta-
levels, rejection and renal impairment in liver trans- tion for hepatocellular carcinoma. Liver Transpl.
plantation: a systematic review and meta-analysis. 2008;14(5):633–8.
Am J Transplant Off J Am Soc Transplant Am Soc 48. Neuhaus P, Klupp J, Langrehr JM. mTOR inhibitors:
Transplant Surg. 2012;12(10):2797–814. an overview. Liver Transpl. 2001;7(6):473–84.
33. Merli M, Di Menna S, Giusto M, Giannelli V, Lucidi 49. Michallet MC, Preville X, Flacher M, Fournel S,

C, Loria I, et al. Conversion from twice-daily to Genestier L, Revillard JP. Functional antibodies to
444 E. Gonsalkorala et al.

leukocyte adhesion molecules in antithymocyte glob- autoimmune hepatitis after liver transplantation. Liver
ulins. Transplantation. 2003;75(5):657–62. Transpl. 2009;15(10):1254–61.
50. Yoshida EM, Marotta PJ, Greig PD, Kneteman NM, 60. Gautam M, Cheruvattath R, Balan V. Recurrence

Marleau D, Cantarovich M, et al. Evaluation of renal of autoimmune liver disease after liver trans-
function in liver transplant recipients receiving dacli- plantation: a systematic review. Liver Transpl.
zumab (Zenapax), mycophenolate mofetil, and a 2006;12(12):1813–24.
delayed, low-dose tacrolimus regimen vs. a standard-­ 61. Milkiewicz P, Hubscher SG, Skiba G, Hathaway M,
dose tacrolimus and mycophenolate mofetil regimen: Elias E. Recurrence of autoimmune hepatitis after liver
a multicenter randomized clinical trial. Liver Transpl. transplantation. Transplantation. 1999;68(2):253–6.
2005;11(9):1064–72. 62. McCauley J, Van Thiel DH, Starzl TE, Puschett

51. Wilde MI, Goa KL. Muromonab CD3: a reappraisal JB. Acute and chronic renal failure in liver transplan-
of its pharmacology and use as prophylaxis of solid tation. Nephron. 1990;55(2):121–8.
organ transplant rejection. Drugs. 1996;51(5):865–94. 63. Fan PY. Renal replacement therapy after liver trans-
52. Haddad EM, McAlister VC, Renouf E, Malthaner R, plantation. Transplantation. 2005;80(3):425–6. author
Kjaer MS, Gluud LL. Cyclosporin versus tacrolimus reply
for liver transplanted patients. Cochrane Database 64. Welker MW, Bechstein WO, Zeuzem S, Trojan

Syst Rev. 2006;4:CD005161. J. Recurrent hepatocellular carcinoma after liver
53. Gane EJ, Portmann BC, Naoumov NV, Smith HM, transplantation—an emerging clinical challenge.
Underhill JA, Donaldson PT, et al. Long-term out- Transpl Int. 2013;26(2):109–18.
come of hepatitis C infection after liver transplanta- 65. Cabrera R, Nelson DR. Review article: the manage-
tion. N Engl J Med. 1996;334(13):815–20. ment of hepatocellular carcinoma. Aliment Pharmacol
54. Berenguer M, Prieto M, Cordoba J, Rayon JM,
Ther. 2010;31(4):461–76.
Carrasco D, Olaso V, et al. Early development of 66. Yanik EL, Chinnakotla S, Gustafson SK, Snyder JJ,
chronic active hepatitis in recurrent hepatitis C virus Israni AK, Segev DL, et al. Effects of maintenance
infection after liver transplantation: association with immunosuppression with sirolimus after liver trans-
treatment of rejection. J Hepatol. 1998;28(5):756–63. plant for hepatocellular carcinoma. Liver Transpl.
55. Berenguer M, Palau A, Aguilera V, Rayon JM, Juan 2016;22(5):627–34.
FS, Prieto M. Clinical benefits of antiviral therapy 67. Schnitzbauer AA, Zuelke C, Graeb C, Rochon J,

in patients with recurrent hepatitis C following liver Bilbao I, Burra P, et al. A prospective randomised,
transplantation. Am J Transplant. 2008;8(3):679–87. open-labeled, trial comparing sirolimus-containing
56. Moonka DK, Kim D, Kapke A, Brown KA, Yoshida versus mTOR-inhibitor-free immunosuppression in
A. The influence of induction therapy on graft and patients undergoing liver transplantation for hepato-
patient survival in patients with and without hepa- cellular carcinoma. BMC Cancer. 2010;10:190.
titis C after liver transplantation. Am J Transplant. 68. Adams DH, Neuberger JM. Patterns of graft rejec-
2010;10(3):590–601. tion following liver transplantation. J Hepatol.
57. Watashi K, Hijikata M, Hosaka M, Yamaji M,
1990;10(1):113–9.
Shimotohno K. Cyclosporin A suppresses replication 69. Adams DH, Neuberger JM. Treatment of acute rejec-
of hepatitis C virus genome in cultured hepatocytes. tion. Semin Liver Dis. 1992;12(1):80–8.
Hepatology. 2003;38(5):1282–8. 70. Kim PT, Demetris AJ, O’Leary JG. Prevention and
58. Foxton MR, Quaglia A, Muiesan P, Heneghan MA, treatment of liver allograft antibody-mediated rejec-
Portmann B, Norris S, et al. The impact of dia- tion and the role of the ‘two-hit hypothesis’. Curr
betes mellitus on fibrosis progression in patients Opin Organ Transplant. 2016;21(2):209–18.
transplanted for hepatitis C. Am J Transplant. 71. Singh N, Pirsch J, Samaniego M. Antibody-mediated
2006;6(8):1922–9. rejection: treatment alternatives and outcomes.
59. Montano-Loza AJ, Mason AL, Ma M, Bastiampillai Transplant Rev. 2009;23(1):34–46.
RJ, Bain VG, Tandon P. Risk factors for recurrence of
Acute Kidney Injury After Liver
Transplantation 33
Raymond M. Planinsic, Tetsuro Sakai,
and Ibtesam A. Hilmi

Keywords episodes occur within the first 2 postoperative


Renal injury · Chronic kidney disease · days. Earlier studies found that mortality at
Dialysis · Renal replacement therapy · 30 days was 50% in patients who developed
Abdominal compartment syndrome · AKI and 29% in non-AKI patients [2]. AKI
Hemolytic uremic syndrome necessitating renal replacement therapy has
been associated with mortality rates from 55 to
90% [3]. More recent data confirmed that AKI
Introduction is associated with increased risk of graft failure
(but not mortality) [4]. Risk factors for AKI
Acute kidney injury after liver transplantation is a include preoperative renal dysfunction repre-
frequent complication that is caused by a multi- sented with a higher preoperative serum creati-
tude of perioperative renal insults often in addi- nine (SCrea), greater requirements for
tion to pre-existing renal insufficiency. Its intraoperative blood transfusion, more frequent
definition is problematic mostly because the basis episodes of intraoperative hypotension and
of most definitions, serum creatinine is a poor and other preexisting co-­morbidities [2]. Recently,
insensitive marker of renal injury. This chapter other predisposing factors associated with
will review the epidemiology and common causes development include female sex, weight
of acute kidney injury after liver transplantation. (>100 kg), severity of liver disease, diabetes
mellitus, number of units of FFP transfused and
the diagnosis of NASH [4].
Epidemiology

The incidence of postoperative renal insuffi- Definitions of AKI


ciency and acute kidney injury (AKI) in patients
undergoing liver transplantation ranges from AKI was traditionally defined as an acute reduc-
20% up to 90% [1] and more than 80% of these tion in glomerular filtration rate sufficient to cause
azotemia and multiple at times conflicting defini-
tions existed in the literature that made compari-
R. M. Planinsic, MD (*) • T. Sakai, MD, PhD, sons of studies difficult. In 2004 the s­econd
MHA • I. A. Hilmi, MD, ChB
Department of Anesthesiology, University of
international consensus conference of the Acute
Pittsburgh Medical Center, Pittsburgh, PA, USA Dialysis Quality Initiative (ADQI) Group pro-
e-mail: planinsicrm@anes.upmc.edu posed a new classification scheme for AKI, that

© Springer International Publishing AG, part of Springer Nature 2018 445


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_33
446 R. M. Planinsic et al.

Fig. 33.1 RIFLE GFR Criteria Urine Output Criteria


criteria (risk, injury,
failure, loss of function,
Increased SCreat x1.5 or UO < .5ml/kg/h
endstage kidney
Risk GFR decrease > 25% x 6 hr
disease). With
permission: Bellomo High
et al. Critical Care 2004 Sensitivity
8:R204 Increased SCreat x2 UO < .5ml/kg/h
or GFR decrease > 50% x 12 hr
Injury

Increased SCreat x3 UO < .3ml/kg/h

ria
or GFR decrease 75% x 24 hr or

Oligu
Failure OR SCreat ≥4mg/dl Anuria x 12 hrs
Acute rise ≥0.5mg/dl

High
Specificity
Persistent ARF = complete loss
Loss
of kidney function >4 weeks

End Stage Kidney Disease


ESKD (>3 months)

includes separate criteria for ­SCrea/glomerular et al. divided ATN into ischemic and nephrotoxic
filtration rate (GFR) and urine output [5]. These causes and attributed 52% of ATN to ischemia
RIFLE (Risk of renal dysfunction, Injury to the and 18% of ATN to nephrotoxic causes [7]. Other
kidney, Failure of kidney function, Loss of kidney significant causes of post-operative ATN include
function and End-stage kidney disease) criteria contrast nephropathy, sepsis, and rarely
define AKI either by SCrea/GFR (increase of rhabdomyolysis.
SCrea ≥3 times of the base line or GFR decrease Early diagnosis of AKI is of critical impor-
of 75% of the base line, or SCrea ≥4 mg/dL) or by tance as only early intervention can potentially
urine output (urine output <0.3 mL/h × 24 h or affect outcome. However serum creatinine is a
anuria × 12 h) (Fig. 33.1). A more recent defini- very slow and insensitive marker that reflects
tion by the Acute Kidney Injury Network (AKIN) renal function but not injury. It may take days
proposes “An abrupt (within 48 h) reduction in after a renal injury for serum creatinine to
kidney function currently defined as an absolute increase and any intervention at this time would
increase in serum creatinine of more than or equal be too late. Furthermore, the decision when and
to 0.3 mg/dL (≥26.4  μmol/L), a percentage how to start immunosuppression with nephro-
increase in serum creatinine of more than or equal toxic immunosuppressive often needs to be made
to 50% (1.5-fold from baseline), or a reduction in before an increase of serum creatinine increases
urine output (documented oliguria of less than reveals substantial renal injury.
0.5 mL/kg per hour for more than 6 h)” [6] as a Recently novel biomarkers of renal injury
definition of AKI, reflecting the fact that even have been discovered and tested as predictors of
small changes of serum creatinine affect outcome renal injury after liver transplantation. Of these
for example after cardiac surgery. Neutrophil Gelatinase-associated Lipocalin
(NGAL) is one of the most promising. NGAL is
a 23 kD protein that can be detected in urine and
Causes of AKI blood within hours after renal injury and is a sen-
sitive marker in multiple scenarios of kidney
Post-transplant AKI can be attributed to several injury. After liver transplantation Wagener et al.
causes (Table 33.1). Acute tubular necrosis showed that NGAL increases rapidly in blood
(ATN) appears to be a major cause of AKI. Fraley and urine and can predict acute kidney injury
33  Acute Kidney Injury After Liver Transplantation 447

Table 33.1  Causes of postoperative renal failure in liver transplantation


Acute tubular necrosis Calcineurin inhibitor toxicity Other
Ischemic Tacrolimus Abdominal compartment syndrome
Nephrotoxicity Cyclosporine A Hemolytic uremic syndrome
Nephropathy Thrombotic thrombocytopenic purpura
Sepsis Infection
Rhabdomyolysis

Table 33.2  Agents associated with renal failure


Pre-renal hemodynamic changes Acute tubular necrosis Acute interstitial nephritis
Cyclosporin Aminoglycosides Penicillins
Tacrolimus Amphotericin B Cephalosporins
Radiocontrast agents Cisplatin Sulfonamides
Amphotericin B Cephalosporins Rifampin
ACE inhibitors Radiocontrast agents NSAIDs
ACE receptor blockers COX-2 inhibitors
NSAIDs Interferon
COX-2 inhibitors Interleukin-2
From: Coffman TM. Renal failure caused by therapeutic agents, in Primer on Kidney Diseases. Greenberg A, Editor.
Academic Press: San Diego, 1998, 260–65

with good sensitivity and specificity. Cystatin vasopressor requirements possibly in conjunction
(CyC) along with NGAL and APACHE II scores with caval crossclamp (and renal venous obstruc-
were shown by Portal to predict AKI within the tion) and use of nephrotoxic drugs such as calci-
first 48 h after LT with high sensitivity and speci- neurin inhibitors all contribute to renal injury and
ficity. Additionally serum IL-8 and urine IL-8, may precipitate AKI. Common postoperative
NGAL, IL-6 and IL-8 have been shown to be causes of AKI are explained in more detail below.
elevated in AKI within 24 h after LT. Use of these
biomarkers may allow earlier intervention to pre-
dict and hence intervene to minimize AKI post Calcineurin Inhibitors
LT [8–11]. Further studies are required to confirm
the clinical utility of these and other biomarkers. Both tacrolimus and cyclosporine A contribute to
In patients with ATN, muddy-brown casts are the development of chronic renal failure in the
usually seen in the urinary sediment and an post transplant period in liver transplantation
increased fractional excretion of sodium is evi- patients and the use of both tacrolimus or cyclo-
dent. Treatment of ATN is usually supportive and sporine has been associated with acute increases
there is no intervention that is able to prevent or in creatinine due to changes in renal hemody-
ameliorate AKI [12]. However it is important to namics [13]. Non-progressive and dose depen-
avoid further renal insults by maintaining blood dent renal dysfunction may be observed with
pressure and renal perfusion and minimizing elevations in serum creatinine levels paralleling
nephrotoxic drugs. Pharmacologic agents that the elevations of serum levels of the calcineurin
may cause AKI are listed in Table 33.2. inhibitor. Lowering the dose of calcineurin inhib-
AKI can usually not be attributed to a single itors may ameliorate deteriorating renal function.
cause and multiple renal insults are required to In addition, there have been a number of studies
cause clinically overt AKI. Pre-existing renal suggesting that chronic nephrotoxicity may be
insufficiency and hepato-renal syndrome, intra- alleviated by use of rapamycin as the primary
and post-operative hypotension, hypovolemia and immunosuppressive agent instead of calcineurin
448 R. M. Planinsic et al.

inhibitors [13, 14]. Late changes of calcineurin renal failure (Fig. 33.2). Chronic renal failure
inhibitor use include renal tubular atrophy and after non-renal solid organ transplantation is
renal interstitial fibrosis [14–16] that may lead to associated with a 4.55 times higher risk of death
irreversible renal failure requiring hemodialysis. compared to patients with no chronic renal fail-
Strategies to reduce the dose of calcineurin inhib- ure [19]. Therefore preventing chronic renal fail-
itors by using alternate forms of immunosuppres- ure by reducing calcineurin inhibitors to the
sion have been attempted. Induction of tolerance lowest possible dose and avoiding other injuries
in liver transplantation where calcineurin inhibi- is paramount to ensure long term success of the
tors are slowly weaned to very low doses may transplant.
significantly diminish or eliminate the renal tox-
icity related to these agents while still providing
adequate immunosuppression [17]. Thrombotic Thrombocytopenic
Careful monitoring of calcineurin inhibitor Purpura: Hemolytic Uremic
levels is essential to avoid major toxicity. And Syndrome
decreasing doses when supra-therapeutic levels
are observed may lessen the incidence of chronic Thrombotic thrombocytopenic purpura (TTP)
renal failure. Other supportive treatments include and hemolytic uremic syndrome (HUS) have
strict control of blood pressure, control of hyper- been described in patients after liver transplanta-
lipidemia, and control of post-transplant diabetes tion and are often attributed to immunosuppres-
mellitus [18]. Often, however, renal failure is sive drugs. Both cyclosporine A and tacrolimus
unrelenting and renal replacement therapy is nec- have been associated with TTP-HUS [20–24].
essary. Liver transplantation has the second high- HUS is characterized by fever, microangiopathic
est incidence of renal failure requiring renal hemolytic anemia and thrombocytopenia. In
replacement therapy of solid non-renal trans- TTP, these symptoms are accompanied by neuro-
plants (after intestinal transplants). Twelve, 36 logic changes and acute renal failure. TTP-HUS
and 60 months after liver transplantations 8.0%, may be associated with malignant hypertension
13.9% and 18.1% respectively developed chronic and subsequent arteriolar injury. The diagnosis is

0.35
0.30
of Chronic Renal Failure

Liver
Cumulative Incidence

Intestine
0.25 Lung

0.20
Heart
0.15

0.10
Heart-Lung
0.05
0.00
0 12 24 36 48 60 72 84 96 108 120
Months since Transplantation
No. at Risk
Heart-lung 576 375 295 219 194 156 133 107 72 46 30
Heart 24,024 19,885 17,238 14,687 12,341 10,022 7997 6104 4526 3096 1991
Intestine 228 152 110 84 57 33 23 13 8 5 5
Liver 36,849 28,495 24,041 19,508 15,724 12,564 9844 7345 5292 3614 2261
Lung 7,643 5,633 4,316 3,184 2,327 1,629 1136 745 468 258 133

Fig. 33.2  Cumulative incidence of chronic renal failure and December 31, 2000. (With permission: Ojo AO et al.
among 69,321 persons who received nonrenal organ N Engl J Med 2003;349:931–940)
transplants in the United States between January 1, 1990,
33  Acute Kidney Injury After Liver Transplantation 449

usually made on clinical grounds alone but may transplant lymphoproliferative disorder (PTLD)
be confirmed by renal biopsy. Plasmapheresis has may occur secondary to EBV and has been shown
been successfully used with or without holding in autopsy studies to infiltrate the kidney [31].
the toxic drug however usually changing immu- Although there has not been a clear correlation
nosuppression is required to treat this condition. between renal infiltration in PTLD and renal fail-
ure, this should be considered in the differential
diagnosis of AKI.
Abdominal Compartment Syndrome There have been case reports of John
Cunningham (JC) and BK polyoma viruses caus-
Increased intra-abdominal pressure is a contrib- ing hemorrhagic cystitis and renal failure in bone
uting factor to AKI after liver transplantation. marrow transplant recipients [32] and renal
Progressive and abrupt increases in intra-­ allograft recipients [33–35]. No reports of these
abdominal pressure reduce cardiac output, con- viruses causing renal failure in liver transplant
tribute increased inspiratory pressures when patients have been reported even though the
ventilated and decreases in splanchnic, hepatic, viruses have been found in the urine of liver
and renal perfusion. These changes are collec- transplant patients [36] and should be considered
tively referred to as “abdominal compartment when other causes are not found.
syndrome” [24–27]. Biancofiore et al. [28], using
urinary bladder manometry, has shown that up to
32% of patients undergoing liver transplantation Summary
have intra-abdominal pressures greater than
20–25 mmHg. This elevation in intra-abdominal Renal failure after liver transplantation is a seri-
pressure was associated with renal failure, lower ous and life threatening complication. Early iden-
filtration gradient, and prolonged ventilation in tification of high-risk patients is essential to
the post-transplant period and may exacerbate minimize the development of this problem. Early
renal injury to a degree that renal replacement diagnosis of renal dysfunction and optimal medi-
therapy is necessary. Renal failure ensues not cal management postoperatively in the intensive
only because of decreased renal arterial flow but care unit is required to ameliorate further renal
also because renal venous drainage is affected. injury. If irreversible renal failure develops, renal
Increased intra-abdominal pressure further replacement therapy with hemodialysis may be
impedes blood flow to the liver and graft function required and possible renal transplantation should
[29]. Frequent measurements of intra-abdominal also be considered if ARF is not reversible.
pressure and possibly re-exploration if the intra-­
abdominal pressure is sustained high may help
alleviate this problem. References
1. McCauley J, et al. Acute and chronic renal failure in
liver transplantation. Nephron. 1990;55(2):121–8.
Infectious Complications 2. Pascual E, et al. Incidence and risk factors of early
acute renal failure in liver transplant patients.
Postoperative infections can progress to sepsis Transplant Proc. 1993;25(2):1827.
and septic shock and cause substantial renal 3. Ishitani M, et al. Outcome of patients requiring hemo-
dialysis after liver transplantation. Transplant Proc.
injury that may progress to renal failure requiring 1993;25(2):1762–3.
renal replacement therapy. Specific infection that 4. Hilmi IA, et al. Acute kidney injury following ortho-
cause direct renal injury are often caused by topic liver transplantation: incidence, risk factors, and
viruses. Epstein-Barr virus (EBV) has been effects on patient and graft outcomes. Br J Anaesth.
2015;114(6):919–26. https://doi.org/10.1093/bja/
reported to cause renal failure in patients after aeu556.
liver transplantation [30]. If detected, appropriate 5. Bellomo R, et al. Acute Dialysis Quality Initiative
antiviral therapy should be initiated. Post-­ workgroup. Acute renal failure—definition, out-
450 R. M. Planinsic et al.

come measures, animal models, fluid therapy 21. Evens AM, et al. TTP/HUS occurring in a simultane-
and ­ information technology needs: the Second ous pancreas/kidney transplant recipient after clopi-
International Consensus Conference of the Acute dogrel treatment: evidence of a nonimmunological
Dialysis Quality Initiative (ADQI) Group. Crit Care. etiology. Transplantation. 2002;74(6):885–7.
2004;8:R204–12. 22. Yango A, et al. Successful treatment of tacrolimus-­
6. Mehta RL, et al. Acute Kidney Injury Network: report associated thrombotic microagiopathy with siroli-
of an initiative to improve outcomes in acute kidney mus conversion and plasma exchange. Clin Nephrol.
injury. Crit Care. 2007;11(2):R31. 2002;58(1):77–8.
7. Fraley DS, et al. Impact of acute renal failure on mor- 23. Medina PJ, Sipols JM, George JN. Drug-associated
tality in end-stage liver disease with or without trans- thrombotic thrombocytopenia purpura-hemo-
plant. Kidney Int. 1998;54:518–24. lytic uremic syndrome. Curr Opin Hematol.
8. Wagener G, et al. Urinary neutrophil gelatinase-­ 2001;8(5):286–93.
associated lipocalin as a marker of acute kidney injury 24.
Holman MJ, et al. FK506 associated throm-
after orthotopic liver transplantation. Nephrol Dial botic thrombocytopenic purpura. Transplantation.
Transplant. 2011;26(5):1717–23. 1993;55(1):205–6.
9. Niemann CU, et al. Acute kidney injury dur- 25. Burch J, Moore F, Franciose R. The abdominal

ing liver transplantation as determined by neutro- compartment syndrome. Surg Clin North Am.
phil gelatinase-associated lipocalin. Liver Transpl. 1996;76:833–42.
2009;15(12):1852–60. 26. Malbrain M. Abdominal pressure in the critically ill
10. Portal AJ, et al. Neutrophil gelatinase-associated
patients. Intensive Care Med. 1999;25:1453–8.
lipocalin predicts acute kidney injury in patients 27. Cullen DJ, et al. Cardiovascular, pulmonary and

undergoing liver transplantation. Liver Transpl. renal effects of massively increased intra-abdominal
2010;16(11):1257–66. pressure in critically ill patients. Crit Care Med.
11. Sirota J, et al. Urine IL-18, NGAL, IL-8 and serum 1989;17:118–21.
IL-8 are biomarkers of acute kidney injury follow- 28. Biancofiore G, et al. Intra-abdominal pressure moni-
ing liver transplantation. BMC Nephrol. 2013;14:17. toring in liver transplant recipients: a prospective
https://doi.org/10.1186/1471-2369-14-17. study. Intensive Care Med. 2003;29(1):30–6.
12.
Esson ML, Schrier RW. Diagnosis and treat- 29. Mogilner J, et al. Effect of elevated intra-abdominal
ment of acute tubular necrosis. Ann Intern Med. pressure on portal vein and superior mesenteric artery
2002;137(9):744–52. blood flow in a rat. J Laparoendosc Adv Surg Tech A.
13. Cotterell AH, et al. Calcineurin inhibitor-induced 2009;19(Suppl 1):S59–62.
chronic nephrotoxicity in liver transplant patients is 30. Tsai JD, et al. Epstein-Barr virus-associated acute
reversible using rapamycin as the primary immuno- renal failure: diagnosis, treatment and follow-up.
suppressive agent. Clin Transplant. 2002;16(Suppl Pediatr Nephrol. 2003;18(7):667–74.
7):49–51. 31. Collins MH, et al. Autopsy pathology of pediat-

14. Gonwa TA, et al. Improved renal function in
ric posttransplant lymphoproliferative disorder.
sirolimus-treated renal transplant patients after Pediatrics. 2001;107(6):E89.
early cyclosporine elimination. Transplantation. 32. Iwamoto S, et al. BK virus-associated fatal renal fail-
2002;74(11):1560–7. ure following late-onset hemorrhagic cystitis in an
15. Benigni A, et al. Nature and mediators of renal lesions unrelated bone marrow transplant. Pediatr Hematol
in kidney transplant patients given cyclosporine for Oncol. 2002;19(4):255–61.
more than one year. Kidney Int. 1999;55(2):674–85. 33. Hirsch HH, et al. Prospective study of poly-

16. Jurewicz WA, et al. Tracolimus versus cyclospo-
omavirus type BK replication and nephropa-
rine immunosuppression: long-term outcome in thy in renal-­ transplant recipients. N Engl J Med.
renal transplantation. Nephrol Dial Transplant. 2002;347(7):488–96.
2003;18(Suppl 1):I7–11. 34. Elli A, et al. BK polyomavirus interstitial nephritis in
17. Starzl TE, et al. Tolerogenic immunosuppression
a renal transplant patient with no previous acute rejec-
for organ transplantation. Lancet. 2003;361(9368):​ tion episodes. J Nephrol. 2002;15(3):313–6.
1502–10. 35. Hirsch HH, et al. Polyomavirus BK nephropathy: a
18. Cardarelli F, et al. The problem of late allograft
(re-)emerging complication in renal transplantation.
loss in kidney transplantation. Minerva Urol Nefrol. Am J Transplant. 2002;2(1):25–30.
2003;55(1):1–11. 36. Marshall WF, et al. Survey of urine from trans-

19. Ojo AO, et al. Chronic renal failure after trans-
plant recipients for polyomavirus JC and BK using
plantation of a nonrenal organ. N Engl J Med. the polymerase chain reaction. Mol Cell Probes.
2003;349(10):931–40. 1991;5(2):125–8.
20. Rerolle JP, et al. Tacrolimus-induced hemolytic ure-
mic syndrome and end stage renal failure after liver
transplantation. Clin Transpl. 2000;14(3):262–5.
Early Graft Failure
34
Avery L. Smith, Srinath Chinnakotla,
and James F. Trotter

Keywords the incidence of primary non-function affects 4 to


Liver function · Acidosis · Liver blood flow · 7% of deceased donor liver grafts [1, 2]. With liv-
Small for size syndrome · Ischemia-­ ing donor transplantation, cold and warm isch-
reperfusion injury · Early allograft emia times are minimal and donor quality
dysfunction typically excellent and, therefore, primary non-­
function is less common with a 3% incidence
reported in the A2ALL study [3]. However,
Introduction recipients of living donor liver transplants have a
higher incidence of SFSS as a cause of early graft
The increasing organ donor shortage and widen- failure. Early graft failure in both deceased donor
ing indications for transplants has forced the use and living donor liver transplant recipients has a
of liver allografts with expanded criteria that are major impact on the prognosis and clinical out-
allocated to sicker and more decompensated come after liver transplantation (LT). This chap-
recipients, allowing a greater volume of trans- ter will outline the risk factors for early allograft
plants. Furthermore, advances in surgical tech- dysfunction and provide a guide to early diagno-
nique have allowed the use of partial liver grafts sis and management strategies of graft failure and
both split and living donor in order to increase to SFSS.
the donor pool. Consequently, graft dysfunction,
graft failure, and small for size syndrome (SFSS)
have become more important than ever. Clinically, Assessment of Liver Graft Function
post-liver transplant graft failure is a continuum
ranging from an ambiguous and easily reversible Operating Room
graft dysfunction to a complete absence of func-
tion called primary non-­function. The incidence Signs of graft function are apparent in the operat-
of graft dysfunction varies from 13 to 27% and ing room after reperfusion of the liver allograft.
Very soon after completion of the portal vein and
hepatic artery anastomosis, the liver should pink
A. L. Smith, MD • J. F. Trotter, MD (*) up uniformly, start producing bile, metabolic
Baylor University Medical Center, Dallas, TX, USA acidosis should correct itself, and coagulation
­
e-mail: James.Trotter@baylorhealth.edu abnormalities should improve. Bile production
S. Chinnakotla, MD during the transplant procedure itself is an excel-
Department of Surgery, University of Minnesota, lent prognostic sign and the bile flow rate was one
Minneapolis, MN, USA

© Springer International Publishing AG, part of Springer Nature 2018 451


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_34
452 A. L. Smith et al.

of the most useful predictors of postoperative sands or levels that are steadily increasing imply
function in many studies. Bile production may severe organ damage and unlikely recovery. In
reflect the recovery of adenosine triphosphate one study, serum AST levels of >5000 U/L
synthesis in the graft. Anecdotally, the color of the resulted in a primary non-function rate of 41% as
bile may be equally important, with golden brown opposed to a rate of 10% in those with peak AST
color considered ideal. Signs and symptoms of levels of 2000–5000 U/L. [5] Elevated prothrom-
graft failure can be recognized very early during bin time and alanine transaminase levels may
surgery: unusual or discolored appearance of the have similar predictive value. Persistent lactic
liver, worsening coagulopathy after reperfusion acidosis, hypoglycemia, hyperkalemia, increas-
of the liver graft, abnormal CO2 production, per- ing hyperbilirubinemia and persistent hypopro-
sistent lactic acidosis, inadequate urine output, thrombinemia are all prominent signs of poor
instability of patients to raise the core body tem- function however rather than the absolute value
perature, hemodynamic instability with persistent of any of these tests the trend is of even greater
or increasing vasopressor requirements, abnor- importance. Multiorgan system failure is the
malities in glucose, and hyperkalemia are all indi- inevitable result in the absence of a functioning
cators that should alert the anesthesiologist to the liver and poor outcome is not necessarily associ-
possibility of early graft failure. ated with any particular individual organ system
but rather the number of organ systems involved.
In some patients graft failure may be more insidi-
Early Postoperative Period ous in presentation especially well-compensated
patients with low preoperative Model for
Postoperatively, the diagnosis of early graft fail- Endstage Liver Disease (MELD) score.
ure and primary non-function can be made on the Therefore, early detection of post-operative graft
basis of clinical findings and laboratory values. dysfunction is essential. Multiple different defini-
Good postoperative mental status and good urine tions of early allograft dysfunction using various
output and renal function are indicative of satis- laboratory cut-off values have been described. In
factory allograft function. Progressively increas- 2010, Olthoff et al. defined early allograft dys-
ing encephalopathy, low urine output, worsening function using the following specific criteria with
metabolic acidosis, and hypotension should alert the following cutoff values: serum bilirubin
to the possibility of graft failure. Liver function >10 mg/dL and an INR > 1.6 on postoperative
tests obtained immediately after surgery are more day 7 or AST levels >2000 U/L within the first
reflective of the blood products and coagulation 7 days and found that patients with early allograft
factors the patient received in the operating room. dysfunction (EAD), using this definition, have a
However, after 12-hours post-transplant, these tenfold higher chance of death within 6 months
may be more indicative of the function of the new [6]. A further evaluation of EAD has reported
liver allograft. Prothrombin time and interna- outcomes on a continuous graded function [7].
tional normalized ratio (INR) are good indicators Aside from its obvious effects on liver function,
of synthetic function of the new liver because the EAD independently increases the risk of acute
biological half-life of factor VII is only 4–6 h kidney injury and endstage renal disease by
and, without adequate liver synthesis, plasma OR = 3.5 and 3.1, respectively, p < 0.05 [8].
concentrations will fall rapidly [4]. The serum
transaminases reflect the degree of preservation
injury and usually peak 24 hours post-transplant Management of Early Graft Failure
and then decrease by approximately 30% every
24 hours post-transplant for the first few postop- It is imperative that vascular (hepatic artery, por-
erative days. Peak serum transaminases less than tal vein, hepatic vein outflow) or other technical
2000 U/L are indicative of minimal preservation complications are expeditiously excluded.
injury and serum transaminases in tens of thou- Vascular patency can be quickly evaluated by a
34  Early Graft Failure 453

doppler ultrasound. Doppler ultrasound often the US), our practice has been to maintain ade-
reveals a low resistive index in the hepatic artery. quate cardiac output and perfusion to the liver
Often, a surgical re-­exploration is the most expe- and initiate continuous venovenous hemodialysis
ditious way to exclude a wide variety of vascular (CVVHD). Primary non-function will almost
and mechanical complications and allow “hands always precipitate acute renal failure and fluid
on” assessment and a safe biopsy of the graft. If overload from large volume blood transfusions to
there is a technical problem, it can be rectified at correct aberrant coagulation that will necessitate
the time of re-exploration. In the case of mechan- CVVHD. These are several additional advan-
ical compression of the liver due to a size mis- tages of initiating CVVHD including lowering
match (large liver in small recipient), the ammonia levels, correcting acid-base and fluid-­
abdominal cavity should be expanded leaving the electrolyte disturbances, lowering CVP and albeit
fascia open or by closing the abdomen with a reducing the congestion of the liver graft, improv-
synthetic mesh. ing pulmonary congestion and clearing lactic aci-
The best treatment of graft failure is avoiding dosis. This will often stabilize the patient until a
the use of grafts that carry a significant risk of new liver graft becomes available. There is some
primary non-function (see Table 34.1), careful evidence that prostaglandin E1 infusion may be
selection of recipients, and a good technical, helpful to increase hepatic blood flow and patient
logistical, and anesthesiologic technique during outcomes [10]. In cases of severe hemodynamic
the liver transplant operation. Once the diagnosis or pulmonary instability due to toxic metabolites
of graft failure is established, the critical question released from the necrotic liver graft, a hepatec-
is, if graft failure is reversible or not. In general if tomy and/or temporary portocaval shunt may be
there is increasing serum transaminases (in the required [11]. This is obviously a drastic step but
thousands), poor synthetic function with elevated can result in temporary stabilization in a patient
prothrombin time and INR despite continuous on the verge of cardiopulmonary collapse. The
administration of fresh frozen plasma, persistent authors’ experience has shown that although
metabolic acidosis, renal failure, and worsening patients with primary non-function are very sick
encephalopathy, it is very unlikely that the graft and in multi-organ failure excellent outcomes
will improve and the only remaining treatment is can be achieved if they are carefully stabilized
urgent re-transplantation [9]. Once the patient is and then re-transplanted in time [9].
listed for urgent re-transplantation (status Ia in

Small for Size Syndrome


Table 34.1  Risk factors for early graft failure
Procurement If a partial liver graft (liver donor or split) is
Donor related factors Recipient unable to meet the functional demands of the
Age > 60 years Non-heart Hemodynamically recipient, liver graft failure may ensue manifest-
beating unstable patient on ing itself as coagulopathy, ascites, prolonged
donors multiple pressors
cholestasis, and encephalopathy, often associated
Steatosis >30% Prolonged
cold ischemia with renal and respiratory failure. These patients
time are further at increased risk for sepsis and gastro-
>16 hours intestinal bleeding. This ill-defined clinical pic-
Hypernatremia ture is considered to be primarily linked to
>165 insufficient graft size and is hence termed “small
Multiple high
for size syndrome.” A liver biopsy performed on
dose pressors of
the donor such grafts often shows cholestasis with bile
Prolonged plugs, and areas of regeneration and ischemia
hospitalization with patchy necrosis [12]. The etiology of SFSS
of the donor is likely to be multifactorial. Donor factors asso-
454 A. L. Smith et al.

ciated with an increased risk for SFSS after liver 3. Olthoff KM, Merion RM, Ghobrial RM, et al.
transplantation included graft to recipient weight Outcomes of 385 adult-to-adult living donor liver
transplant recipients. Ann Surg. 2005;242:314–25.
ratio (GRWR) of <0.8% and preexisting steatosis 4. Roloff JS. Disseminated intravascular coagulation
of the donor graft [13]. The recipient factors and other acquired bleeding disorders. In: Levin DL,
include a decompensated recipient with a high Morris FC, editors. Essentials of pediatric intensive
MELD score and severe portal hypertension. The care. St. Louis: Quality Medical Publishing; 1990.
5. Rosen HR, Martin P, Goss J, et al. Significance of
exact pathogenic mechanisms of SFSS are still early aminotransferase elevation after liver transplan-
unclear; critical graft size and function, graft tation. Transplantation. 1998;65:68–72.
injury, regeneration, and recovery may all con- 6. Olthoff KM, Kulik L, Samstein B, et al. Validation of
tribute to SFSS [14]. Recent clinical studies have a current definition of early allograft dysfunction in
liver transplant recipients and an analysis of risk fac-
focused on the reduction of portal venous inflow tors. Liver Transpl. 2010;16:943–9.
to the small graft to prevent development of 7. Pareja E, Cortes M, Hervás D, et al. A score model for
SFSS, thereby implicating portal hyperperfusion the continuous grading of early allograft dysfunction
as a cause of SFSS [15]. Spontaneous improve- severity. Liver Transpl. 2015;21:38–46.
8. Wadei HM, Lee DD, Croome KP, et al. Early allograft
ment of liver function may occur over time but dysfunction after liver transplantation is associated
approximately 50% of recipients with SFSS will with short-and long-term kidney function impairment.
die of sepsis or other complication within Am J Transplant. 2016;16:850–9.
4–6 weeks after transplantation [12]. Therefore, 9. Uemura T, Randall HB, Sanchez EQ, Ikegami T,
Narasimhan G, McKenna GJ, et al. Liver retrans-
prevention of SFSS after transplantation is key. plantation for primary nonfunction: analysis of a
Once SFSS is established after partial liver graft 20-year single-center experience. Liver Transpl.
transplantation and technical complications such 2007;13:227–33.
as bile leak or vascular thrombosis have been 10. Henley KS, Lucey MR, Normolle DP, et al. A double-­
blind, randomized, placebo-controlled trial of pros-
excluded by imaging studies further treatment is taglandin E1 in liver transplantation. Hepatology.
supportive. If the patient has evidence of a large 1995;21:366–72.
spleen or severe portal hypertension, splenic 11. Oldhafer KJ, Bornscheuer A, Fruhauf NR, et al. Rescue
artery ligation to reduce portal vein blood flow hepatectomy for initial graft non-­function after liver
transplantation. Transplantation. 1999;67:1024–8.
has been reported in one study of seven patients 12.
Demetris AJ, Kelly DM, Eghtesad B, et al.
[15, 16]. If there is no response to intervention, Pathophysiologic observations and histopathologic
the prudent course would be to retransplant the recognition of the portal hyper perfusion or small for
patient before sepsis and multiorgan failure size syndrome. Am J Surg Pathol. 2006;30:986–93.
13. Dahm F, Georgiev P, Clavien P. Small for size syn-
develops [13]. drome after partial liver transplantation: definition,
mechanisms, of disease and clinical implications. Am
J Transplant. 2005;5:2605–10.
References 14. Dutkowski P, Linecker M, DeOliveira ML, et al.

Challenges to liver transplantation and strate-
gies to improve outcomes. Gastroenterology.
1. Maring JK, Klompmaker IJ, Zwaveling JH, 2015;148:307–23.
Kranenburg K, Ten Vergert EM, Slooff MJ. Poor ini- 15. Humar A, Beissel J, Crotteau S, et al. Delayed splenic
tial graft function after orthotopic liver transplanta- artery occlusion for treatment of established small for
tion: can it be predicted and does it affect outcome? size syndrome after partial liver transplantation. Liver
An analysis of 125 adult primary transplantations. Transpl. 2009;15:163–8.
Clin Transpl. 1997;11:373–9. 16. Lee SG. A complete treatment of adult living donor
2. Deschenes M. Early allograft dysfunction: causes, liver transplantation: a review of surgical technique
recognition, and management. Liver Transpl. and current challenges to expand indication of
2013;19S:SS6–8. patients. Am J Transplant. 2015;15:17–38.
Sepsis and Infection
35
Fuat Hakan Saner

Keywords infections occurring within the first 2 months. In


Bacterial infections · Pneumonia · Fungal the last three decades, there was a significant
infection · Selective bowl decontamination · increase of sepsis and septic shock [3] and gram
Septic shock · Antibiotics positive bacteria. Particularly, methicllin-resis-
tant staphylococcus aureus (MRSA) and vanco-
mycin-resistant enterocci (VRE) [4] are now very
Introduction common, due to extended and prolonged use of
third generation cephalosporines and chinolones.
Thomas Starzl reported the results of the first The extensive use of third generation cephaloso-
liver transplant program in 1976 [1]: only 29% of pines and imipenem/cilastation furthermore
the transplanted patients survived 1 year after induces the growth of Acinetobacter, yeast, and
transplantation and the main cause of death, at VRE [5] (Fig. 35.1).
that time, was uncontrolled bleeding due to Pulmonary complications such as pneumonia
severe coagulopathy and acute and chronic rejec- are a major cause of death in liver transplant
tion. Infection was considered less common. patients [6] and the majority of pneumoniae are
Only when cyclosporine was introduced as an caused by bacteria (50–70%) [7–10] especially
immunosuppressant drug to avoid acute and gram-negative rods. Aspergillus species are the
chronic rejection, the reported 1-year survival most common non-­bacterial organisms causing
increased to 80–90%. But with improved sur- pneumonia in the first month after transplanta-
vival, infectious complications after liver trans- tion. In the early post transplant setting the ICU
plantation were more commonly reported. physician is mainly confronted with bacterial and
Infection is now one of the leading causes of fungal infections and this review will therefore
morbidity and mortality in liver transplant focus on these microbes.
patients. More than 50% of liver transplant recip-
ients develop infections during the first year after
transplantation [2], with the majority of bacterial Risk Factors for Bacterial Infections

After liver transplantation, bacteremia has been


F. H. Saner documented in 20–40% of patients [7–10] with a
Department of General-, Visceral- and Transplant reported mortality of 15–36% [7, 9]. Early after
Surgery, Medical Center University Essen, liver transplantation it is difficult to estimate the
Essen, Germany likelihood of infection in a setting of fever and/or
e-mail: fuat.saner@uni-due.de

© Springer International Publishing AG, part of Springer Nature 2018 455


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_35
456 F. H. Saner

Penicillin [1940] Methicillin [1960]

Penicillin-resistant Methicillin-resistant
S. aureus S. aureus (MRSA)
S. aureus
Vancomycin [1980]
3’rd generation
[2002] Cephalosporines
Vancomycin-
resistant Ciprofloxacin [1987]
S. aureus Vancomycin
(Glykopeptide) Vancomycin-resistant
intermediate-resistant Enterococci (VRE)
S. aureus

Fig. 35.1  Development of resistance in gram-positive 1963 Methicillin-resistant Staphylococcus aureus


cocci. Chain et al. published in 1940 about penicillin as a (MRSA) occurred. With the extended use of cephalospo-
chemotherapeutic agent (Lancet ii:226–228). The same rine and chinolones, and Vancomycin, vancomycin resis-
group reported already in the same year an enzyme from tant enterococci and Vancomycin resistant S. aureus
bacteria able to destroy penicillin [11]. Methicillin was occurred
the first penicillinase-resistant antbiotic. However in

altered laboratory markers like C-reactive protein patients with life threatening infections [16]. In a
or procalcitonin. Markers that reliably predict the univariate analysis, peak PCT and peak CRP were
probability of bacteremia in febrile patients significantly higher in patients with infections com-
would allow a judicious and more appropriate pared to patients without infections. A multivariate
use of antibiotic while culture results are analysis involving male sex, BMI < 20 kg/m2
pending. (vs. BMI > 25 kg/m2), with acute liver failure as
Singh et al. [12] evaluated the risk factors for an indication for liver transplantation and pro-
bacterial infections in 59 liver transplant patients longed cold ischemia time (>420 min) were inde-
with clinical signs of infection. When comparing pendent risk factors for infection, while peak PCT
patients who had post transplant bacteremia with was not an independent risk factor. Furthermore,
patients with non-bacteremic systemic inflamma- anti-thymocyte globulin (ATG) can markedly
tion, bacteremic patients were significantly more increase procalcitonin in patients before hemato-
likely to have renal dysfunction, diabetes melli- poetic stem cells [17], When ATG is part of
tus, higher APACHE II scores, lower serum albu- immunosuppressive protocol as treatment for
min levels, and were more likely in the ICU at rejection, symptoms can be very similar to sepsis
time of onset of symptoms. White blood cell (fever, elevated liver enzymes, low dose catechol-
count, bilirubin, or prothrombin time were not amine requirement) and the diagnosis of infection
significantly different, as was temperature that is very difficult.
was previously assumed to be an early and non-
specific marker for infection in surgical patients.
Procalcitonin (PCT) is a prognostic marker for Risk Factors for Fungal Infection
infection in non-transplant patients and it is sug-
gested that guiding the antibiotic treatment to the Invasive fungal infections have been reported in
level of procalcitonin may reduce the antibiotic 5–42% of liver transplant recipients with an asso-
exposure with a comparable cure rate [13–15]. ciated mortality of 25–71% [18–21]. The unique
There is little data supporting the use of procalci- susceptibility of liver transplant patients to inva-
tonin as a marker of infection after liver transplan- sive fungal infection is well recognized; the main
tation. Van den Broek et al. evaluated procalcitonin cause of fungal infections is candidemias [22].
(and C-reactive protein) as a prognostic marker Pre- and intra-operative risk factors for invasive
for infectious complications in liver transplant fungal infections in liver transplant patients
35  Sepsis and Infection 457

include previous liver transplantation (within 30 patients who received placebo (from 60 to 90%)
days), renal failure (when dialysis is required), and the incidence of fungal colonization in
extended use of antibiotic prophylaxis, reopera- patients who received fluconazole (from 70 to
tion due to bleeding or bile leak and treatment of 28%) was decreased. Confirmed fungal infec-
rejection with pulsed dose of steroids [7, 23, 24]. tions occurred in 45 of 104 placebo recipients
In the recent years, more and more non-albicans (43%) but only in 10 of 108 fluconazole recipi-
strains are evident and azole resistant albicans areents (9%) (P < 0.001). Fluconazole prevented
frequently isolated [24]. both superficial infection (29 of 104 patients
receiving placebo recipients [28%] compared to
4 of 108 patients receiving fluconazole [4%];
The Role of Selective Bowl P < 0.001) and invasive infection (24 of 104
Decontamination, Antibiotic patients receiving placebo [23%] compared to 6
Prophylaxis and Antimycotic of 108 patients receiving fluconazole [6%];
Prophylaxis P < 0.001).
N. Singh et al. [32] evaluated the efficacy of
To prevent infections most centers extend antibi- liposomal amphotericin B as prophlaxis for inva-
otic prophylaxis for 2 or 3 days. However, there sive fungal infections in high risk liver transplant
is no evidence to support this approach and anti- patients requiring hemodialysis. They compared
biotic prophylaxis is only able to prevent wound this cohort with a historical control group with-
infections, not pneumonia, bile leak, or abscess. out antifungal prophylaxis. Antifungal prophy-
Extended use of antibiotics will increase multi- laxis with liposomal amphotericin B reduced the
drug-resistant (MDR) bacterials [4] and is a risk incidence of fungal infections from 36% in the
factor for fungal infection [24]. historical control group to 0% in the treatment
In 1983, Stoutenbeek described selective group. Moreover, antifungal prophylaxis with
digestive tract decontamination (SDD) [25] to liposomal amphotericin B protected against fun-
prevent nosocomial infection and since then mul- gal infections independent of covariates.
tiple studies have evaluated the use of SDD after However, both studies failed to demonstrate a
liver transplantation. There were four prospective difference in outcome after liver transplantation
randomized trials with conflicting results [26– with antifungal prophylaxis. These results were
29]. Safdar et al. [30] undertook a systematic confirmed by a meta-analysis [33]: prophylaxis
review and meta-analysis of randomized trials of reduced colonization and confirmed fungal infec-
SDD versus placebo after liver transplantation tions and mortality attributable to fungal infec-
and found SDD to be effective in reducing gram-­ tion but did not affect the overall mortality or
negative infections. However, due to more fre- empiric treatment for suspected fungal infection.
quent gram-positive infections, the effect on the The beneficial effect of antifungal prophylaxis
overall infection rate was limited. The relative was predominantly associated with the reduction
risk of infection with SDD was 0.88 (95% CI, of Candida albicans infection and the mortality
0.7–1.1) indicating no statistically significant attributable to C. albicans. Compared to controls,
reduction in infection with the use of SDD. however, patients receiving antifungal prophy-
There are two cornerstone studies of antimy- laxis experienced a higher proportion of episodes
cotic prophylaxis after liver transplantation: of non-albicans Candida, and in particular of C.
Bussetil et al. [31] evaluated the efficacy and glabrata. No beneficial effect on invasive
safety of prophylactic fluconazole in liver trans- Aspergillus infection was observed.
plant recipients in a randomized, double-blind, In conclusion, although antifungal prophy-
placebo-controlled trial. More than 200 liver laxis has been widely studied and practiced, no
transplant recipients received fluconazole consensus exists on which patients should receive
(400 mg/d) or placebo up to 10 weeks after trans- prophylaxis, with which agent, and for what
plantation. Fungal colonization increased in duration. Depending on the local epidemiology
458 F. H. Saner

and incidence of fungal infection transplant cen- is ≥2  μg/mL [42] are required to prevent the
ters with an incidence <8% do not require pro- development of resistance. In critically ill patients
phylaxis. Concerns about selection of a loading dose of 25–30 mg/kg should be consid-
triazole-resistant Candida strains are realistic and ered [43]. However, there is an incremental risk
a potential disadvantage of prophylaxis. It may of nephrotoxicity from 12 to 42.7% associated
be more reasonable to identify patients who are at with higher vancomycin doses. The risk increases
risk for fungal infections and treat assumed fun- with higher vancomycin trough levels, longer
gal infections pre-emptively instead of using duration of vancomycin use, concomitant use of
general antifungal prophylaxis on all transplant other nephrotoxic agents and in patients who are
recipients. Due to shift towards non-albicans critically ill or with previously compromised
strains, use of echinocandin instead of fluconazol renal function [44–49]. Data on the degree of
[34] may be indicated. renal recovery are scarce and the mechanisms of
vancomycin toxicity have only partially been
evaluated. Animal data suggest that vancomycin
 ultiply Resistant Gram-Positive
M stimulates oxidative phosphorylation in renal
and Gram-Negative Bacteria proximal tubule epithelial cells, thus acting as an
oxidative stressor [49]. Severe vancomycin neph-
Staphylococcus aureus (MRSA) rotoxicity may present histologically as a tubulo-­
interstitial nephritis, sometimes with granulomas
Methicillin-resistant Staphylococcus aureus [50]. Liver transplant patients who are already at
(MRSA) is an important cause of infection in an increased risk for kidney failure, particular
liver transplant patients. The incidence of MRSA due to the use of calcineurin inhibitors and preop-
is continuously increasing and a significant cause erative impaired kidney function should be
of blood stream infections (BSI). In some inten- treated with alternative drugs without nephro-
sive care units in the USA, MRSA has a preva- toxic side effects.
lence of >64% [35]. However, in Europe, even Linezolid is bacteriostatic and approved for
after liver transplantations the MRSA rate for skin- and soft tissue infections (SSTI) and noso-
BSI was only 13% [7]. In the absence of trans- comail pneumonia but not approved for the treat-
plantation, risk factors associated with MRSA ment of MRSA sepsis or endocarditis. With
infection are prolonged illness, co-morbitidies, long-term use (>28 days) thrombocytopenia and/
especially diabetes mellitus and renal failure or peripheral and optic neuropathy may occur.
requiring dialysis, longer ICU and hospital stay There is no interaction with the cytochrome P
and extended exposure to third generation cepha- 450 enzymatic system.
losporins and chinolons [4, 36]. Colonization Daptomycin is bactericidal and used to treat
with MRSA increases the risk of later infection for SSSI, MRSA bacteremia and right- heart
usually by the colonizing strain [37]. Liver trans- endocarditis but it should not be used for MRSA
plant patients who are colonized with MRSA pneumonia, because it is inactivated by lung
have a higher incidence of MRSA infections, ­surfactant. There is also no interaction with the
ranging from 31 to 78% [38, 39]. Carriage of cytochrome P 450 enzymatic system.
MRSA does not increase the mortality rate, how- Tigecycline is bacteriostatic and approved for
ever once MRSA infection is evident the mortal- SSSI and intra-abdominal infections (IAI). There
ity risk is clearly increased [38]. is no data about treating bacteremia or endocardi-
Vancomycin is the drug of choice for infec- tis with tigecycline. Caution should be used in the
tions caused by sensitive MRSA [40] and should presence of confirmed bacteremia because serum
be given to obtain adequate trough levels of at concentrations can rapidly decrease between
least 10 μg/mL. Trough levels above 10 μg/mL doses [51].
when the minimum inhibitory concentration Quinopristin-dalfopristin is bactericidal and
(MIC) is 1 μg/mL [41] and 15–20 μg/mL, if MIC used for the treatment of SSSI. The use is limited
35  Sepsis and Infection 459

due to adverse effects such as athralgia and plant patients are often severe and are associated
myalgia. with prolonged ICU- and hospital stay [61] and
Ceftopirole is a beta-lactam antibiotic active higher risk of death [62].
against MRSA. It is bactericidal, however there
is no data with regard to the treatment of bactere-
mia and endocarditis. Ceftopirole is currently Treatment of  VRE
only approved in 13 European countries and
Canada. Linezolid has become the drug of choice for
many types of VRE infection. Linezolid has been
used in the treatment of serious VRE infections,
 revention and Infection of MRSA
P including VRE bacteremia, VRE endocarditis
in Liver Transplant Patients and skin and soft tissue infections (SSTI) [63–
65]. The most serious adverse effect of linezolid
Several guidelines have been proposed to reduce treatment is bone marrow suppression usually
the rate of MRSA colonization and infections occurring after more than 14 days treatment.
[52, 53]. These guidelines were not evaluated for Daptomycin, a lipopeptide, has in vitro bacte-
liver transplant patients, however due to the ricidal activity against the most relevant Gram-­
absence of data in liver transplant patients, we positive organisms, including VRE. The initially
suggest to use recommendations for the general proposed dose of 4 mg/kg is likely insufficient
ICU population. Liver transplant candidates and daptomycin is approved for 6 mg/kg in
should be identified by routine nasal swab and patients with MRSA bacteremia and right-heart
MRSA in colonized patients should be eradicated endocarditis. Studies evaluating higher doses
using local mupiricine ointment. (8–12 mg/kg) are ongoing. During daptomycin
The use of antibiotics should be judicious and treatment creatininkinase and myoglobin should
appropriate. We recommend to limit the empiri- daily be monitored, because of the risk of dapto-
cal use of antibiotics and avoid long periopera- mycin induced rhabdomyolysis.
tive prophylaxis therapy, adopt the narrowest Tigecycline, a gylcopeptide, is a broad-­
spectrum therapy for documented infections, spectrum antibiotic with high in vitro activity
limit the use of antibiotics to 7 days and avoid against VRE. There is little clinical data about the
treating contaminations [54, 55]. use of tigecycline for VRE. A retrospective study
of ICU patients after major abdominal surgery
(mean APACHE II score of 27) found that 16%
Vancomycin-Resistant of the patients were infected with MRSA and
Enterococcus (VRE) 27% with VRE. Tigecycline was used in combi-
nation in 76% of these cases and alone in 24%.
Vancomycin-resistant enterococci (VRE) have The mortality rate was 30%, significantly lower
emerged as a relevant pathogen in liver transplant than the expected mortality of 55% ­considering
recipients. VRE colonization and infection was the mean APACHE II score of 27 in this group
first described at the Mayo Clinic in 1995 [56]. [66]. Due to rapidly declining serum levels tige-
The rate of colonization is center-dependent and cycline cannot be recommended as the first line
more common in the USA than in Europe. One of treatment for enterococcal sepsis. However, a
the main risk factors for VRE colonization is the case report demonstrated clinical cure in a patient
extended and inappropriate use of Vancomycin with severe enterococcal sepsis treated with tige-
and second or third generation cephalosporin cycline alone [67].
(Fig. 35.2) [57]. Most common infections caused Prevention and Infection control for VRE can
by VRE in liver transplant patients are blood be achieved by using antibiotics especially
stream, intra-abdominal, biliary tract and wound Vancomycin and third generation cephalosporins
infections [58–60]. VRE infections in liver trans- only for a clear indication. If infection is sus-
460 F. H. Saner

Cephalosporine 3’rd generation

Overuse (→ selection)

ESBL Enterococci
Klebsiella spp.
E. coli
Enterobacter cloacae
No coverage

Resistant to Cephalosporine Vancomycin


3’rd generation

Imipenem
Selection

Selection

VRE

Acinetobacter Yeast
spp.

Modified to Bernstein Chest 1999.

Fig. 35.2  Extended use of Cephalosporines causes the enterococci (VRE). On the other hand the overuse of third
selection of resistant bacteriae. The overuse of third genera- generation cephalosporines stimulates the formation of the
tion cephalosporines induces the growth of bacterials that enzyme extended-spectrum-beta-lactamase (ESBL) in bac-
are not sensitive to cephalosporines. The growth of entero- teria that is able to destroy all amino- and ureidopenicil-
cocci is increased that were treated initially with lines. The use of imipenem/cilastatin stimulates the growth
Vancomycin, inducing the growth of Vancomycin resistant of acenitobacter and increases the risk of yeast infections

pected, a thorough search for a focus should be score, extended and prolonged antibiotic
initiated, for example by CT scan of chest and ­exposure are the main risk factors for multidrug
abdomen or transesophageal echocardiography resistant gram-negative bacteria. Prompt recog-
to exclude endocarditis (very rare in liver trans- nition and adequate treatment are critical for a
plant patients). successful outcome.

 ultidrug Resistant (MDR) Gram-­


M
Negative Bacteria in Liver Transplant Extended-Spectrum Beta-Lactamase
Patients (ESBL) Producing Enterobacteriaceae
In the recent two decades the rate of MDR gram-­
negative bacteriae has increased [68]. These are ESBL producing enterobacteriaceaes are not
extended-spectrum beta-lactamase (ESBL) pro- uncommon in liver transplant patients. The
ducing bacteria, carbapenem-resistant Klebsiella reported incidence varies between 5.5 and 7%
and E. coli and MDR Acinetobacter baumanii. [69, 70]. A French group assessed in a retrospec-
The risk factor of infection with these bacteria is tive study three independent risk factors for
very similar to the risk with gram-positive ­postoperative infection [70]. Preoperative fecal
­bacteria. Prolonged ICU stay, higher APACHE II carriage, MELD score > 25 and reoperation after
35  Sepsis and Infection 461

transplantation were associated with increased Carbapenemase-Producing


risk of infection. During the study time the inci- Enterobacteriaceae
dence of infections increased significantly from
1.6% (period: 2001–2003) to 12.8% (2009– Carbapenem-resistent Klebsiella pneumonia
2010). Exclusive mortality data for liver trans- (CRKP) is the key player among this subgroup of
plant patients infected with ESBL are lacking. gram-negative bacteria’s, that shows an increas-
However, in a retrospective study evaluating of ing rate since the new millennium [78]. In one
ESBL bacterial blood stream infection in a mixed study CRKP accounts for 23% of bloodstream
cohort (mainly kidney and liver transplant infections for all liver transplant patients with an
patients) the mortality was 26% within the first associated mortality of 86% compared to 29%
30 days after transplantation [71]. Treatment of for non-CRKP sepsis [79]. General risk factors
choice for ESBL producing Enterobacteriacae for CRKP infections were identified for extensive
are carbepenemes [72] however it should be use of broad-spectrum antibiotics and severity of
guided by antimicrobial susceptibility testing. illness with long-term ICU and invasive proce-
The most used are imipenem/cilastatin and dure requirement (e.g. dialysis, ventilation) [80].
meropenem [57]. Other studies identified additional risk factors
specific for liver transplant recipients. They
found in a large cohort that high MELD score,
MDR Pseudomonas aeroginosa hepatocellular carcinoma, Roux-en-Y anastomo-
sis, post-operative bile leak, dialysis, prolonged
The prevalence of MDR Pseudomonas, particular ventilator support, and recurrence of hepatitis C
in patients, with recurrent hospital stay continues virus were independent risk factors for CRKP
to increase [73]. Pseudomonas account for 6.5% infections [81, 82]. The allocation of livers in
blood stream infection in liver transplant patients UNOS and some Eurotransplant countries is
[74]. However, newer data reports an incidence linked to the MELD score. The higher the MELD
of about 32.3% [75]. In a French study in a mixed score the higher to chance to receive an organ.
transplant population, mainly kidney- and liver However, high MELD patients are prone to
transplant recipients’ 15.4% of bacteremia epi- increased colonization risk with CRKP. An
sodes were caused by pseudomonas strains (49 Italian group evaluated the incidence and impact
out of 318) [75]. Among these pseudomonas of pre-LTx colonization with CRKP on postop-
strains 63% (31/49) was resistant to at least one erative outcome [81]. In a 30 months period all
of “typical” antipseudomonas antibiotics. LTx candidate were monitored up to 180 days
Mortality among patients with Pseudomonas posttransplant. A total of 237 patients were
sepsis was significantly higher than in non-­ recruited and of these 41 patients (17.3%) were
pseudomonas sepsis (38 vs. 16%). colonized with CRKP (11 at time of transplant,
Many experts recommend a combination 30 after transplantation). Infections occurred in
treatment for Pseudomonas infections, although 20 patients (18 bloodstream and 2 pneumonia)
this approach remains controversial [76]. within 6 weeks post transplant. When comparing
Ceftazidime/avibactam and ceftolozane/tazo- non-colonized, colonized at transplant and colo-
bactam were recently approved for complicated nized after transplantation patients, infection rate
intraabdominal infections [77]. In vitro activities was highest in colonized after transplantation
indicates sufficient activity of avibactam against (46.7 vs. 18.2% -colonized at transplant and 2%
extended-spectrum beta-lactamase type TEM-1, for non-colonized patients).
TEM-2, TEM-3 AmpC and OXA-48, however The treatment of this organism remains a
lacking activity against NDM-1, and VIM-1. major challenge. There are new antibiotics in the
Tazobactam covers only TEM-1–TEM-3 [77]. pipeline, however, convincing clinical data, eval-
Although clinical studies about efficacy are uated for these type of bacteria’s are lacking.
­available for both new agents, pathogen-specific, Treatment options include colistin alone [83]
e.g. KPC producing Pseudomonas is lacking. or in combination with tigecycline [84].
462 F. H. Saner

A recent review [85] discussed the current mortality of 90% for ICU patients with CRAB-­
gold standard of pharmacotherapy for infection, infection [94].
including combination treatment and the new The treatment of CRAB remains a major chal-
developed ceftazidime/avibacatm (cefta/Avi) and lenge. The most commonly used drug is colistin
imipenem/relebactame (imi/rele). Ceftazidime/ but the use of colistin is associated with increased
avibacatm and relebactame can be used if carb- colistin resistance. The combination of colistin
anemem resistant is based on KPC or OXA-48. with carbapenem was associated with a better 28
However, when carbanemem resistant is medi- days survival in a small study of 60 patients [93].
ated with MBL (metallo-beta-lactamase) these In another study of non-transplant patients the
agents are ineffective. A combination treatment combination treatment of colistin with rifampicin
could be used, but lacks for robust data. Since we had no impact in survival [95].
have very limited treatment options for CRKP
bacteriae, we should be very cautious with trans-  ungal Infections in Liver Transplant
F
plantation of CRKP colonized patients depend- Patients
ing on how CRKP colonization occurred. A Candida species are the most common cause for
patient whose CRKP colonization occurs only fungal infections in liver transplant patients [7]
after contact with a CRKP carrier may be a fea- and frequently occur within the first 3 months
sible transplant candidate compared to patients following transplantation [96]. In recent years
who contracted CRKP due to recurrent long-term earlier occurrence (within the first 4 weeks) has
antibiotic exposure with recurrent septic epi- been reported. Candida albicans is the dominant
sodes. Colonization that occurs only after contact pathogen that is responsible for about 65% of
with a CRKP carrier may result in successful candida infections, followed by Candida gla-
transplant after eradication using oral gentamicin brata with 21% [24]. Candida krusei which is
and oral colistin [86]. However patients with common after stem cell transplant is far less
CRKP due to recurrent long-term antibiotic common in liver transplant patients [97]. The
exposure should not be transplanted because of risk of candidemia is already discussed in detail
high-risk for postoperative infection and death. above. The diagnosis of invasive candidiasis is
dependent on detection of the organism from a
usually sterile body site such as the bloodstream
Carbapenem-Resistant or intra-­abdominal fluid. Candida detected in the
Actinetobacter Baumanii (CRAB) broncho-­alveolar lavage (BAL) must be consid-
ered a contamination and should not be treated,
Acinetobacter baumanii (AB) are susceptible to even after liver transplantation. Liver transplant
carbapenems (exception ertapenem) [87]. The patients with fever of unknown origin (negative
occurrence of CRAB is most likely linked to CT Scan of chest and abdomen, endocarditis and
extensive use of carbapenems. The incidence of viral infections have been ruled out and treat-
CRAB is significantly increasing and in some ment with broad spectrum antibiotics e.g. imin-
centers CRAB accounts for 95% among AB iso- penem + Linezolid has been unsuccessful)
lates [88, 89]. AB is mainly related to nosocomial should be treated preemptively with antifungal
pneumonia. Among LT-patients with sepsis and/ therapy [98]. Immunosuppressed patients such
or septic shock AB accounts for 24% of all septic as liver transplant recipients can initially receive
patients. The incidence of CRAB among an echinocandin for 14 days (weak evidence
AB-isolation is reported to be at least 50%. level of II C-benefits and risks closely balanced
Main infection sites were the lungs and around and/or uncertain). Fluconzole should only be
24% of the isolates for sepsis were AB, while used for patients, who are not critically ill and
50% was CRAB [90–92]. Preoperative coloniza- only for superficial candidiasis. Treatment of
tion with CRAB seem to be a risk factor for post- invasive candidiasis in liver transplant recipients
operative infection [93]. CRAB infections are should follow the same principles as other
associated with a high mortality with a reported patients and is extensively discussed in the
35  Sepsis and Infection 463

Table 35.1  General susceptibility patterns of Candida species


Species Flucoanzol Itraconazol Voriconazol Posaconazol L-Ampho B Echinocandin
C. albicans S S S S S S
C. tropicalis S S S S S S
C. parapsilosis S S S S S S-R
C. glabrata S-DD to R S-DD to R S-DD to R S S S
C. krusei R R S S S S
C. lusetania S S S S S S
R resistant, S susceptible, S-DD susceptible dose-dependent. C. parapsilosis isolates resistant to echinocandins are
uncommon

“Clinical practice guidelines for the manage- selected cases. Flucoanzole, itraconazole and
ment of candidiasis” [98]. Table 35.1 gives a voriconazole demonstrate significant drug-drug
general overview of susceptibility of Candida interactions and special attention must be given
species. to dose adjustments of co-administered drugs,
especially the calcineurin inhibitors [101].

Amphotericin B
Echinocandin
Amphotericin B is a polyene antifungal drug, It
was originally extracted from Streptomyces nodo- Caspofungin, micafungin and anidulafungin are
sus, a filamentous bacterium, in 1955 at the only available as parenteral preparations. They
Squibb Institute for Medical Research from cul- all have excellent in vitro activity against most
tures from an undescribed streptomycete isolated. Candida species including C. glabrata and C.
Amphotericin B interacts with ergosterol, the krusei [102]. C. parapsilosis and C. guilliermon-
main component of fungal cell membranes, form- dii demonstrate less in vitro susceptibility to the
ing a transmembrane channel that leads to mon- echinocandins [103]. The echinocandins have
ovalent ion (K+, Na+, H+, Cl−) leakage, the primary few adverse effects, do not require dose adjust-
effect leading to fungal cell death. Amphotericin ment for renal insufficiency or dialysis and are
B use has serious adverse effects. Very often seri- rarely associated with drug–drug interactions
ous acute reactions occursafter the 1–3 h after [102]. It is recommended to reduce the dose of
infusion and consist of fever, shaking chills, and caspofungin to 35 mg in patients with liver fail-
headache. Nephrotoxicity is a frequently reported ure. However, in a retrospective study with liver
adverse effect, and can be severe and even irre- transplant patients with poor graft function the
versible. Liposomal amphotericin B exhibits use of caspofungin with 50 mg had no adverse
fewer adverse effects, particular less nephrotoxic- effects [104] and there have been no reports of
ity [99]. The recommended dose is 3 g/kg BW. hepatoxicity.
A black-box warning for micafungin has been
issued in Europe, based upon an increased num-
Triazoles ber of liver tumors observed in rat models. No
such black-box warning has been included in the
Fluconazole, itraconazole, voriconazole and US label or Japan. In a prospective randomized,
posaconazole demonstrate similar activity double blind study, where safety and efficacy of
against most Candida species, although they are micafugin was evaluated against amphotericin B,
less active against Candida glabrata and Candida 202 patients were treated with micafungin. In
krusei [100]. Posaconazole demonstrates excel- none of these patients a liver tumor was diag-
lent in vitro activity against most Candida species nosed. In Japan the drug has been used for nearly
but requires oral administration, that can be 10 years, with no reported increased incidence of
impossible in critically ill patients in some liver tumors.
464 F. H. Saner

Invasive Aspergillosis Caspofungin is currently the only echinocan-


din that is approved by the FDA for treatment of
Invasive aspergillosis occurs in 1–10% in liver aspergillosis. Caspofungin was successful used
transplant patients [7, 105]. Most invasive fungal as first line therapy in heart-lung transplant
infections in these high-risk patients occur within patients [112] and as salvage therapy in invasive
the first month posttransplant; the median time to aspergillosis as single agent [113]. In a non-­
onset of invasive aspergillosis after renal replace- comparative study micafungin could safely used
ment therapy and retransplantation was 13 and 28 as a primary and salvage drug treatment for
days, respectively in one study [106, 107]. aspergillosis [114].
Mortality rate in liver transplant recipients with Data about the use of anidulafungin as treat-
invasive aspergillosis has ranged from 88 to ment of aspergillosis in clinical trials are absent.
100% [7, 108]. Posaconazole a new extended-spectrum triazole
A substantial delay in establishing an early has been successful been used in bone marrow
diagnosis remains a major impediment to the suc- transplant patients with graft-versus-host disease
cessful treatment of invasive aspergillosis. and in neutropenic hemato-oncologic as prophy-
Isolation of aspergillosis from the respiratory tract laxis to avoid invasive aspergillosis [115, 116].
in liver transplant patients is rare (1.5%), however In a prospective open-label study posacon-
with a high predictive value ranging from 41 to azole was successfully used as rescue treatment
72% for developing invasive aspergillosis [109]. for patients who are refractory or intolerant to
The ability of galactomannan to predict inva- conventional therapy [117].
sive aspergillosis was assessed in a prospective There are only a few data about combined
study with 154 liver transplant patients. The doc- aspergillus treatment. The new upcoming IDSA
umented specificity was 98% and false-positive guidelines [118] recommend the combination
galactomannan tests were found in up to 13% as salvage treatment. In a prospective study of
[110]. Liver transplant patients undergoing trans- solid organ transplant recipients that included
plantation for autoimmune liver disease and liver transplant patients, the combination ther-
those requiring dialysis were significantly more apy consisting of caspofungin and voriconazol
likely to have false-positive galactomannan tests was prospectively assessed in patients with
[110]. confirmed invasive aspergillosis and compared
Since 1990 liposomal Amphotericin B to a historical control group treated with liposo-
replaced the classic Amphotericin B as the main- mal amphotericn B [107]. The overall 90-day
stay for treatment of invasive aspergillosis, survival rate was not different in both groups,
because of fewer side effects, particular less however patients infected with Aspergillus
nephrotoxicity. Newer azols and echinocandins fumigates and renal failure showed a significant
with anti-aspergillus activity and a better toler- better 90-day survival with the combination
ance profile expanded the pool of available anti-­ therapy of caspofungin and voriconazol.
aspergillus drugs. Although definitive clinical trials are pending
In a prospective randomized trial the sur- that demonstrates a benefit of combined ther-
vival rate with voriconazol was significantly apy it should be considered as a rescue treat-
higher compared to amphotericin B deoxycho- ment in selected cases.
late. Voriconazol treated patients had fewer
adverese effects (except transient visual distur- Conclusion
bances) [111]. Voriconazole is now regarded as Patients following liver transplantation need
the drug of choice for the primary treatment of meticulous attention for signs of infections and
invasive aspergillosis, a recommendation prompt treatment. The use of antibiotics should
endorsed by the Infectious Diseases Society of be restrictive, an extended and inappropriate
America (IDSA) for the treatment of invasive use confers the risk of multi-drug resistant bac-
aspergillosis [98]. terial and fungal infections. Antimycotic pro-
35  Sepsis and Infection 465

phylaxis is recommended in high-risk patients antibiotic stewardship in infections--hope for hype?


but does not improve hospital mortality. Swiss Med Wkly. 2009;139(23–24):318–26.
15. Schuetz P, et al. Effect of procalcitonin-based
An early and preemptive antifungal treat- guidelines vs standard guidelines on antibi-
ment with echinocandins in patients suspected otic use in lower respiratory tract infections: the
of candidemia or voriconazole when aspergil- ProHOSP randomized controlled trial. JAMA.
losis is suspected should be favored over using 2009;302(10):1059–66.
16. van den Broek MA, et al. Procalcitonin as a prog-
general antifungal prophylaxis. nostic marker for infectious complications in liver
transplant recipients in an intensive care unit. Liver
Transpl. 2010;16(3):402–10.
17. Brodska H, et al. Marked increase of procalcitonin
References after the administration of anti-thymocyte globulin
in patients before hematopoietic stem cell transplan-
1. Starzl TE, et al. Orthotopic liver transplantation tation does not indicate sepsis: a prospective study.
in ninety-three patients. Surg Gynecol Obstet. Crit Care. 2009;13(2):R37.
1976;142(4):487–505. 18. Kusne S, et al. Infections after liver transplantation.
2. Rubin RH. The direct and indirect effects of infec- An analysis of 101 consecutive cases. Medicine
tion in liver transplantation: pathogenesis, impact, (Baltimore). 1988;67(2):132–43.
and clinical management. Curr Clin Top Infect Dis. 19. Neofytos D, et al. Epidemiology and outcome of
2002;22:125–54. invasive fungal infections in solid organ transplant
3. Martin GS, et al. The epidemiology of sepsis in the recipients. Transpl Infect Dis. 2010;12(3):220–9.
United States from 1979 through 2000. N Engl J 20. Singh N, et al. Invasive fungal infections in liver
Med. 2003;348(16):1546–54. transplant recipients receiving tacrolimus as the
4. Paterson DL. “Collateral damage” from cephalospo- primary immunosuppressive agent. Clin Infect Dis.
rin or quinolone antibiotic therapy. Clin Infect Dis. 1997;24(2):179–84.
2004;38(Suppl 4):S341–5. 21. Tollemar J, et al. The incidence and diagnosis of
5. Bernstein JM. Treatment of community-acquired invasive fungal infections in liver transplant recipi-
pneumonia--IDSA guidelines. Infectious dis- ents. Transplant Proc. 1990;22(1):242–4.
eases society of America. Chest. 1999;115(3 22. Pappas PG, et al. Invasive fungal infections in
Suppl):9S–13S. low-risk liver transplant recipients: a multi-center
6. Jensen WA, et al. Pulmonary complications of prospective observational study. Am J Transplant.
orthotopic liver transplantation. Transplantation. 2006;6(2):386–91.
1986;42(5):484–90. 23. Ergin F, et al. Invasive aspergillosis in solid-organ
7. Saner FH, et al. Pulmonary and blood stream infec- transplantation: report of eight cases and review of
tions in adult living donor and cadaveric liver trans- the literature. Transpl Int. 2003;16(4):280–6.
plant patients. Transplantation. 2008;85(11):1564–8. 24. Husain S, et al. Changes in the spectrum and
8. Singh N, et al. Infectious complications in liver trans- risk factors for invasive candidiasis in liver
plant recipients on tacrolimus. Prospective analysis transplant recipients: prospective, multicenter,
of 88 consecutive liver transplants. Transplantation. case-controlled study. Transplantation. 2003;
1994;58(7):774–8. 75(12):2023–9.
9. Wade JJ, et al. Bacterial and fungal infections after 25. Miranda DR, et al. Environment and costs in surgi-
liver transplantation: an analysis of 284 patients. cal intensive care unit. The implication of selective
Hepatology. 1995;21(5):1328–36. decontamination of the digestive tract (SDD). Acta
10. Wagener MM, Yu VL. Bacteremia in transplant Anaesthesiol Belg. 1983;34(3):223–32.
recipients: a prospective study of demographics, 26. Arnow PM. Prevention of bacterial infection
etiologic agents, risk factors, and outcomes. Am J in the transplant recipient. The role of selective
Infect Control. 1992;20(5):239–47. bowel decontamination. Infect Dis Clin N Am.
11. Abraham EP, Chain E. An enzyme from bacteria 1995;9(4):849–62.
able to destroy penicillin. 1940. Rev Infect Dis. 27. Bion JF, et al. Selective decontamination of the
1988;10(4):677–8. digestive tract reduces gram-negative pulmonary
12. Singh N, et al. Predicting bacteremia and bactere- colonization but not systemic endotoxemia in
mic mortality in liver transplant recipients. Liver patients undergoing elective liver transplantation.
Transpl. 2000;6(1):54–61. Crit Care Med. 1994;22(1):40–9.
13. Nobre V, et al. Use of procalcitonin to shorten 28. Hellinger WC, et al. A randomized, prospective,
antibiotic treatment duration in septic patients: double-blinded evaluation of selective bowel decon-
a randomized trial. Am J Respir Crit Care Med. tamination in liver transplantation. Transplantation.
2008;177(5):498–505. 2002;73(12):1904–9.
14. Schuetz P, Christ-Crain M, Muller B. Procalcitonin 29. Zwaveling JH, et al. Selective decontamination of
and other biomarkers to improve assessment and the digestive tract to prevent postoperative infection:
466 F. H. Saner

a randomized placebo-controlled trial in liver trans- 44. Hidayat LK, et al. High-dose vancomycin therapy
plant patients. Crit Care Med. 2002;30(6):1204–9. for methicillin-resistant Staphylococcus aureus
30. Safdar N, Said A, Lucey MR. The role of selec- infections: efficacy and toxicity. Arch Intern Med.
tive digestive decontamination for reducing infec- 2006;166(19):2138–44.
tion in patients undergoing liver transplantation: a 45. Hutschala D, et al. Influence of vancomycin on
systematic review and meta-analysis. Liver Transpl. renal function in critically ill patients after cardiac
2004;10(7):817–27. surgery: continuous versus intermittent infusion.
31. Winston DJ, Pakrasi A, Busuttil RW. Prophylactic Anesthesiology. 2009;111(2):356–65.
fluconazole in liver transplant recipients. A random- 46. Ingram PR, et al. Risk factors for nephrotoxicity
ized, double-blind, placebo-controlled trial. Ann associated with continuous vancomycin infusion in
Intern Med. 1999;131(10):729–37. outpatient parenteral antibiotic therapy. J Antimicrob
32. Singh N, et al. Preemptive prophylaxis with a lipid Chemother. 2008;62(1):168–71.
preparation of amphotericin B for invasive fun- 47. Jeffres MN, et al. A retrospective analysis of pos-
gal infections in liver transplant recipients requir- sible renal toxicity associated with vancomycin in
ing renal replacement therapy. Transplantation. patients with health care-associated methicillin-­
2001;71(7):910–3. resistant Staphylococcus aureus pneumonia. Clin
33. Cruciani M, et al. Antifungal prophylaxis in liver Ther. 2007;29(6):1107–15.
transplant patients: a systematic review and meta-­ 48. Lodise TP, et al. Larger vancomycin doses (at least
analysis. Liver Transpl. 2006;12(5):850–8. four grams per day) are associated with an increased
34. Pappas PG, Silveira FP. Candida in solid organ trans- incidence of nephrotoxicity. Antimicrob Agents
plant recipients. Am J Transplant. 2009;9(Suppl Chemother. 2008;52(4):1330–6.
4):S173–9. 49. Lodise TP, et al. Relationship between initial van-
35. National Nosocomial Infections Surveillance System. comycin concentration-time profile and nephrotox-
National Nosocomial Infections Surveillance icity among hospitalized patients. Clin Infect Dis.
(NNIS) system report, data summary from January 2009;49(4):507–14.
1992 through June 2004, issued October 2004. Am J 50. Hong S, et al. Vancomycin-induced acute granulo-
Infect Control. 2004;32(8):470–85. matous interstitial nephritis: therapeutic options. Am
36. Asensio A, et al. Colonization and infection with J Med Sci. 2007;334(4):296–300.
methicillin-resistant Staphylococcus aureus: asso- 51. Micek ST. Alternatives to vancomycin for the
ciated factors and eradication. Infect Control Hosp treatment of methicillin-resistant Staphylococcus
Epidemiol. 1996;17(1):20–8. aureus infections. Clin Infect Dis. 2007;45(Suppl
37. Wertheim HF, et al. The role of nasal carriage in 3):S184–90.
Staphylococcus aureus infections. Lancet Infect Dis. 52. Calfee DP, et al. Strategies to prevent transmission
2005;5(12):751–62. of methicillin-resistant Staphylococcus aureus in
38. Russell DL, et al. Outcomes of colonization acute care hospitals. Infect Control Hosp Epidemiol.
with MRSA and VRE among liver transplant 2008;29(Suppl 1):S62–80.
candidates and recipients. Am J Transplant. 53. Coia JE, et al. Guidelines for the control and preven-
2008;8(8):1737–43. tion of meticillin-resistant Staphylococcus aureus
39. Singh N, et al. Methicillin-resistant Staphylococcus (MRSA) in healthcare facilities. J Hosp Infect.
aureus: the other emerging resistant gram-positive 2006;63(Suppl 1):S1–44.
coccus among liver transplant recipients. Clin Infect 54. Siegel JD, et al. Management of multidrug-resistant
Dis. 2000;30(2):322–7. organisms in health care settings, 2006. Am J Infect
40. Chambers HF. Methicillin resistance in staphylococci: Control. 2007;35(10 Suppl 2):S165–93.
molecular and biochemical basis and clinical impli- 55. Singh N, et al. Short-course empiric antibiotic ther-
cations. Clin Microbiol Rev. 1997;10(4):781–91. apy for patients with pulmonary infiltrates in the
41. Rosenthal VD, Maki DG, Graves N. The intensive care unit. A proposed solution for indis-
International Nosocomial Infection Control criminate antibiotic prescription. Am J Respir Crit
Consortium (INICC): goals and objectives, descrip- Care Med. 2000;162(2 Pt 1):505–11.
tion of surveillance methods, and operational 56. Patel R, et al. Natural history of vancomycin-
activities. Am J Infect Control. 2008;36(9):e1–12. resistant enterococcal colonization in liver and
42. Pallin DJ, et al. Increased US emergency department kidney transplant recipients. Liver Transpl. 2001;
visits for skin and soft tissue infections, and changes in 7(1):27–31.
antibiotic choices, during the emergence of commu- 57. Paterson DL, et al. Antibiotic therapy for Klebsiella
nity-associated methicillin-­resistant Staphylococcus pneumoniae bacteremia: implications of production
aureus. Ann Emerg Med. 2008;51(3):291–8. of extended-spectrum beta-lactamases. Clin Infect
43. Rybak M, et al. Therapeutic monitoring of vanco- Dis. 2004;39(1):31–7.
mycin in adult patients: a consensus review of the 58. Asensio A, et al. Effect of antibiotic prophylaxis on
American Society of Health-System Pharmacists, the risk of surgical site infection in orthotopic liver
the Infectious Diseases Society of America, and the transplant. Liver Transpl. 2008;14(6):799–805.
Society of Infectious Diseases Pharmacists. Am J 59. Newell KA, et al. Incidence and outcome of infec-
Health Syst Pharm. 2009;66(1):82–98. tion by vancomycin-resistant enterococcus follow-
35  Sepsis and Infection 467

ing orthotopic liver transplantation. Transplantation. 75. Bodro M, et al. Extensively drug-resistant Pseudomonas
1998;65(3):439–42. aeruginosa bacteremia in solid organ transplant recipi-
60. Nusair A, et al. Infection control experience in a ents. Transplantation. 2015;99(3):616–22.
cooperative care center for transplant patients. Infect 76. Tamma PD, Cosgrove SE, Maragakis
Control Hosp Epidemiol. 2008;29(5):424–9. LL. Combination therapy for treatment of infections
61. McNeil SA, et al. Vancomycin-resistant enterococ- with gram-negative bacteria. Clin Microbiol Rev.
cal colonization and infection in liver transplant 2012;25(3):450–70.
candidates and recipients: a prospective surveillance 77. van Duin D, Bonomo RA. Ceftazidime/Avibactam
study. Clin Infect Dis. 2006;42(2):195–203. and Ceftolozane/Tazobactam: second-generation
62. Ghanem G, et al. Outcomes for and risk factors beta-lactam/beta-lactamase inhibitor combinations.
associated with vancomycin-resistant enterococcus Clin Infect Dis. 2016;63(2):234–41.
faecalis and vancomycin-resistant enterococcus fae- 78. van Duin D, van Delden C, A.S.T.I.D.C.o. Practice.
cium bacteremia in cancer patients. Infect Control Multidrug-resistant gram-negative bacteria infec-
Hosp Epidemiol. 2007;28(9):1054–9. tions in solid organ transplantation. Am J Transplant.
63. Babcock HM, et al. Successful treatment of 2013;13(Suppl 4):31–41.
vancomycin-­resistant enterococcus endocarditis with 79. Kalpoe JS, et al. Mortality associated with
oral linezolid. Clin Infect Dis. 2001;32(9):1373–5. carbapenem-­resistant Klebsiella pneumoniae infec-
64. McNeil SA, et al. Successful treatment of tions in liver transplant recipients. Liver Transpl.
vancomycin-­resistant enterococcus faecium bacte- 2012;18(4):468–74.
remia with linezolid after failure of treatment with 80. Swaminathan M, et al. Prevalence and risk fac-
synercid (quinupristin/dalfopristin). Clin Infect Dis. tors for acquisition of carbapenem-resistant
2000;30(2):403–4. Enterobacteriaceae in the setting of endemicity.
65. Munoz P, et al. Linezolid therapy for infective endo- Infect Control Hosp Epidemiol. 2013;34(8):809–17.
carditis. Clin Microbiol Infect. 2007;13(2):211–5. 81. Giannella M, et al. Risk factors for infection with
66. Swoboda S, et al. Tigecycline for the treatment of carbapenem-resistant Klebsiella pneumoniae after
patients with severe sepsis or septic shock: a drug liver transplantation: the importance of pre- and
use evaluation in a surgical intensive care unit. J posttransplant colonization. Am J Transplant.
Antimicrob Chemother. 2008;61(3):729–33. 2015;15(6):1708–15.
67. Swoboda S, et al. Septic shock due to vancomycin-­ 82. Pereira MR, et al. Risk factors and outcomes of
resistant enterococci infection. Tigecycline mono- carbapenem-resistant Klebsiella pneumoniae infec-
therapy. Anaesthesist. 2007;56(2):169–74. tions in liver transplant recipients. Liver Transpl.
68. Dowzicky MJ, Park CH. Update on antimicrobial sus- 2015;21(12):1511–9.
ceptibility rates among gram-negative and gram-pos- 83. Souli M, et al. An outbreak of infection due to beta-­
itive organisms in the United States: results from the lactamase Klebsiella pneumoniae Carbapenemase
Tigecycline Evaluation and Surveillance Trial (TEST) 2-producing K. pneumoniae in a Greek University
2005 to 2007. Clin Ther. 2008;30(11):2040–50. Hospital: molecular characterization, epidemiology,
69. Linares L, et al. Klebsiella pneumoniae infection and outcomes. Clin Infect Dis. 2010;50(3):364–73.
in solid organ transplant recipients: epidemiol- 84. Maltezou HC, et al. Outbreak of infections due to
ogy and antibiotic resistance. Transplant Proc. KPC-2-producing Klebsiella pneumoniae in a hos-
2010;42(8):2941–3. pital in Crete (Greece). J Infect. 2009;58(3):213–9.
70. Bert F, et al. Pretransplant fecal carriage of 85. Perez F, et al. Treatment options for infections caused
extended-spectrum beta-lactamase-producing by carbapenem-resistant Enterobacteriaceae: can we
Enterobacteriaceae and infection after liver trans- apply “precision medicine” to antimicrobial chemother-
plant, France. Emerg Infect Dis. 2012;18(6):908–16. apy? Expert Opin Pharmacother. 2016;17(6):761–81.
71. Aguiar EB, et al. Outcome of bacteremia caused 86. Saidel-Odes L, et al. A randomized, double-blind,
by extended-spectrum beta-lactamase-producing placebo-controlled trial of selective digestive decon-
Enterobacteriaceae after solid organ transplantation. tamination using oral gentamicin and oral poly-
Transplant Proc. 2014;46(6):1753–6. myxin E for eradication of carbapenem-resistant
72. Jones RN, Biedenbach DJ, Gales AC. Sustained Klebsiella pneumoniae carriage. Infect Control
activity and spectrum of selected extended-spectrum Hosp Epidemiol. 2012;33(1):14–9.
beta-lactams (carbapenems and cefepime) against 87. Munoz-Price LS, Weinstein RA. Acinetobacter
Enterobacter spp. and ESBL-producing Klebsiella infection. N Engl J Med. 2008;358(12):1271–81.
spp.: report from the SENTRY antimicrobial surveil- 88. Freire MP, et al. Polymyxin use as a risk factor for
lance program (USA, 1997–2000). Int J Antimicrob colonization or infection with polymyxin-resistant
Agents. 2003;21(1):1–7. Acinetobacter baumannii after liver transplantation.
73. van Duijn PJ, Dautzenberg MJ, Oostdijk EA. Recent Transpl Infect Dis. 2014;16(3):369–78.
trends in antibiotic resistance in European ICUs. 89. Zhong L, et al. Multidrug-resistant gram-negative
Curr Opin Crit Care. 2011;17(6):658–65. bacterial infections after liver transplantation – spec-
74. Moreno A, et al. Bloodstream infections among trum and risk factors. J Infect. 2012;64(3):299–310.
transplant recipients: results of a nationwide surveil- 90. Ye QF, et al. Frequency and clinical outcomes of
lance in Spain. Am J Transplant. 2007;7(11):2579–86. ESKAPE bacteremia in solid organ transplantation
468 F. H. Saner

and the risk factors for mortality. Transpl Infect Dis. 104. Saner F, et al. Safety profile of concomitant use of
2014;16(5):767–74. caspofungin and cyclosporine or tacrolimus in liver
91. Liu H, et al. Predictors of mortality in solid-organ transplant patients. Infection. 2006;34(6):328–32.
transplant recipients with infections caused by 105. Brown RS Jr, et al. Incidence and significance of
Acinetobacter baumannii. Ther Clin Risk Manag. Aspergillus cultures following liver and kidney
2015;11:1251–7. transplantation. Transplantation. 1996;61(4):666–9.
92. Shi SH, et al. Multidrug resistant gram-negative 106. Kusne S, et al. Factors associated with invasive
bacilli as predominant bacteremic pathogens in lung aspergillosis and the significance of positive
liver transplant recipients. Transpl Infect Dis. Aspergillus culture after liver transplantation. J
2009;11(5):405–12. Infect Dis. 1992;166(6):1379–83.
93. Shields RK, et al. Epidemiology, clinical char- 107. Singh N, et al. Combination of voriconazole
acteristics and outcomes of extensively drug-­ and caspofungin as primary therapy for invasive
resistant Acinetobacter baumannii infections aspergillosis in solid organ transplant recipients:
among solid organ transplant recipients. PLoS One. a prospective, multicenter, observational study.
2012;7(12):e52349. Transplantation. 2006;81(3):320–6.
94. Otan E, et al. Acinetobacter infection in a liver 108. Denning DW. Therapeutic outcome in invasive
transplantation intensive care unit. Transplant Proc. aspergillosis. Clin Infect Dis. 1996;23(3):608–15.
2013;45(3):998–1000. 109. Paterson DL, Singh N. Invasive aspergillosis
95. Aydemir H, et al. Colistin vs. the combination in transplant recipients. Medicine (Baltimore).
of colistin and rifampicin for the treatment of 1999;78(2):123–38.
carbapenem-­ resistant Acinetobacter baumannii 110. Kwak EJ, et al. Efficacy of galactomannan antigen
ventilator-associated pneumonia. Epidemiol Infect. in the Platelia Aspergillus enzyme immunoassay for
2013;141(6):1214–22. diagnosis of invasive aspergillosis in liver transplant
96. Patterson JE. Epidemiology of fungal infections in recipients. J Clin Microbiol. 2004;42(1):435–8.
solid organ transplant patients. Transpl Infect Dis. 111. Herbrecht R, et al. Voriconazole versus amphoteri-
1999;1(4):229–36. cin B for primary therapy of invasive aspergillosis.
97. Marr KA, et al. Candidemia in allogeneic blood and N Engl J Med. 2002;347(6):408–15.
marrow transplant recipients: evolution of risk fac- 112. Groetzner J, et al. Caspofungin as first-line therapy
tors after the adoption of prophylactic fluconazole. J for the treatment of invasive aspergillosis after tho-
Infect Dis. 2000;181(1):309–16. racic organ transplantation. J Heart Lung Transplant.
98. Pappas PG, et al. Clinical practice guidelines for 2008;27(1):1–6.
the management of candidiasis: 2009 update by the 113. Carby MR, Hodson ME, Banner NR. Refractory
Infectious Diseases Society of America. Clin Infect pulmonary aspergillosis treated with caspofun-
Dis. 2009;48(5):503–35. gin after heart-lung transplantation. Transpl Int.
99. Groll AH, et al. Lipid formulations of amphotericin 2004;17(9):545–8.
B: clinical perspectives for the management of inva- 114. Denning DW, et al. Micafungin (FK463), alone or in
sive fungal infections in children with cancer. Klin combination with other systemic antifungal agents,
Padiatr. 1998;210(4):264–73. for the treatment of acute invasive aspergillosis. J
100. Espinel-Ingroff A, et al. International and multi- Infect. 2006;53(5):337–49.
center comparison of EUCAST and CLSI M27-A2 115. Cornely OA, et al. Posaconazole vs. fluconazole or
broth microdilution methods for testing suscepti- itraconazole prophylaxis in patients with neutrope-
bilities of Candida spp. to fluconazole, itraconazole, nia. N Engl J Med. 2007;356(4):348–59.
posaconazole, and voriconazole. J Clin Microbiol. 116. Ullmann AJ, et al. Posaconazole or fluconazole for
2005;43(8):3884–9. prophylaxis in severe graft-versus-host disease. N
101. Lipp HP. Antifungal agents--clinical pharmacokinet- Engl J Med. 2007;356(4):335–47.
ics and drug interactions. Mycoses. 2008;51(Suppl 117. Walsh TJ, et al. Treatment of invasive aspergillosis
1):7–18. with posaconazole in patients who are refractory to
102. Kauffman CA, Carver PL. Update on echino- or intolerant of conventional therapy: an externally
candin antifungals. Semin Respir Crit Care Med. controlled trial. Clin Infect Dis. 2007;44(1):2–12.
2008;29(2):211–9. 118. Walsh TJ, et al. Treatment of aspergillosis: clinical
103. Bennett JE. Echinocandins for candidemia practice guidelines of the Infectious Diseases Society
in adults without neutropenia. N Engl J Med. of America. Clin Infect Dis. 2008;46(3):327–60.
2006;355(11):1154–9.
Respiratory Failure and ARDS
36
James Y. Findlay and Mark T. Keegan

Keywords tent or late pulmonary edema, and acute


Restrictive lung disease · Hydrothorax respiratory distress syndrome (ARDS) develop.
Ventilation · Non-invasive ventilation These may require prolonged intensive care unit
Ventilator associated pneumonia · Pulmonary (ICU) management and mechanical ventilation
edema with consequent increases in patient hospital
stay, cost, and mortality [4].
In this chapter, we shall address the etiolo-
Introduction gies of perioperative respiratory failure, man-
agement strategies for short-term and prolonged
Pulmonary complications are common in the mechanical ventilatory support, approaches to
period immediately after liver transplantation ventilator weaning, and care of the patient with
(LTx) to the point that they are almost an expected specific causes of respiratory failure including
consequence of the procedure. Approximately ARDS.
one-fifth of patients with end-­stage liver disease
are hypoxemic pretransplant and hypoxemia may
worsen in the post-operative period because of
alterations in the mechanics of respiration. The  epatic Hydrothorax and Alteration
H
published prevalence of pleural effusion, atelec- in Respiratory Mechanics
tasis and interstitial pulmonary edema is up to
87% [1–3]. Fortunately, the majority of these Hepatic hydrothorax occurs in approximately 5%
cases are of little clinical consequence with reso- of patients with severe liver disease. It is present
lution occurring rapidly without the need for on the right side more often than the left. Pleural
complex interventions and without adversely effusions may impair a patient’s ability to wean
affecting outcome [1]. from mechanical ventilation and may need post-­
In some cases, however, more serious pulmo- operative drainage [5]. Although ascitic fluid in
nary complications such as pneumonia, persis- the abdomen is usually drained during the surgi-
cal procedure, residual or persistent ascitic fluid
can reduce functional residual capacity and vital
J. Y. Findlay, MB, ChB, FRCA (*) capacity. Implantation of a new liver in the upper
M. T. Keegan, MB, MRCPI, MSc, D ABA part of the right abdomen to take the place of a
Department of Anesthesiology and Critical Care
Medicine, Mayo Clinic, Rochester, MN, USA shrunken cirrhotic liver may also lead to a reduc-
e-mail: findlay.james@mayo.edu tion in vital capacity by pushing up the right

© Springer International Publishing AG, part of Springer Nature 2018 469


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_36
470 J. Y. Findlay and M. T. Keegan

d­ iaphragm. Furthermore, LTx leads to disruption Immediate Post-operative


of diaphragmatic function and, as in any major Extubation?
upper abdominal procedure, post-operative atel-
ectasis can develop. Immediate post-operative extubation (IPE) after
LTx in the majority of cases is advocated by some
clinicians. In addition to reducing the risk of ven-
Weaning from Mechanical tilator associated pneumonia (VAP), early extu-
Ventilatory Support After “Routine” bation may have beneficial effects on splanchnic
Liver Transplantation and liver blood flow. IPE is often performed with
a view to avoiding ICU admission and subse-
There has been considerable discussion and con- quently eliminating ICU-associated costs and
troversy regarding the time of extubation after reducing hospital length of stay [8, 14].
LTx [6, 7]. Progressive improvement in surgical Arguments against IPE include the contention
and anesthetic techniques, coupled with increased that a period of post-operative ventilation allows
experience mean that prolonged mechanical ven- graft function to be consolidated with less sym-
tilation following LTx is no longer necessary in pathetic activation and protects the recipient from
the majority of patients [8]. Multiple surgical the risks of atelectasis, aspiration or reintubation
practices (e.g. cardiac, thoracic) have embraced a for surgical re-exploration if required [7, 8, 15].
“fast-track” concept that targets early extubation Once hemodynamic stability, hemostasis and the
with subsequent reduction in costs but without presence of a functioning graft have been ascer-
compromising safety [9, 10]. This paradigm has tained, extubation can proceed. A short delay in
been extended to liver transplantation [11]. In the extubation may also allow for better treatment of
majority of centers, efforts are made to achieve early post-operative pain that may otherwise be
extubation of most LTx recipients soon after the compromised for fear of hypoventilation of air-
surgical procedure. The intraoperative use of way compromise. In a multi-center US and
short acting medications and lower dosing of opi- European study, 391 patients were extubated
ates has facilitated rapid post-operative ventilator within 1 h of completion of surgery [16]. Adverse
weaning [12, 13]. events occurred in 7.7% of them within 72 h of
Numerous studies have demonstrated the ben- surgery, although most of these adverse events
efit of the involvement of nurses and respiratory were relatively minor. There was considerable
therapists in implementation of ventilator wean- inter-center variability. In some centers, early
ing protocols. These protocols have allowed extubation is performed in 60–70% of LTx cases
assessment of weaning readiness and progression with avoidance of ICU admission in many of
along a ventilator weaning pathway independent those cases resulting in a reduction in costs [13,
of physician involvement, and have been shown 14, 17, 18]. When selecting IPE appropriate
to decrease the duration of mechanical ventila- patient selection then available resources for sub-
tion. Ventilatory support is incrementally reduced sequent management must be taken into account.
to the point of readiness for extubation and only To date, there has not been a randomized trial of
the final decision to extubate needs to be made by immediate versus early versus delayed extuba-
a physician. Figure 36.1 illustrates the post-­ tion after LTx.
operative weaning protocol at our institution.
Early liberation from mechanical ventilation in
the ICU may or may not decrease ICU length of Patients Who Require Prolonged
stay, depending on ICU workflow and established Ventilatory Support
protocols [12]. Involvement of bedside paramed-
ical staff and use of protocols are also useful in A significant number of patients will not be
the management of longer-term ventilation (see suitable candidates for early extubation after
below). LTx. The introduction of the Model for End
36  Respiratory Failure and ARDS 471

Fig. 36.1  Post liver Initial ventilator settings


transplantation ventilator • Synchronized Intermittent Mandatory Ventilation (SIMV)
8-10 breaths per minute (bpm)
weaning protocol used • Fraction of Inspired Oxygen (FIO2) to keep oxygen
saturations (SpO2) >92% as needed
at the authors’ institution • Tidal Volume (VT) at 8-10 mL/kg Ideal Body Weight (IBW)
• Positive End Expiratory Pressure (PEEp) at 5 cm H 2O
• Inspiratory to Expiratory ratio (I:E) at 1:2-1:3
• Obtain Arterial Blood Gas (ABG) in 30 minutes

Adjust ventilation to maintain Adjustment ranges


pH: 7.35-7.45 FiO2: 0.4-1.0
PaCO 2: 35-50 mm Hg SIMV: 8-15 bpm
PaCO 2: 90-120 mm Hg PEEP: 5-10 cm H 2O
SpO2 ≥92% Note: notify primary service if unable
to achieve above parameters

Assess weaning criteria

Patient weaning criteria section


Hemodynamic ststus
• Systolic blood pressure >80 mm Hg
• Heart rate is between 50-110 bpm
• Cardiac rhythm unchanged from pre-operative ECG recording
• Temperature is between 36 and 38ºc
Recovery from surgery
• Documented recovery from neuromuscular blockade
(4 twitches on train of 4 monitoring)
• Active bleeding is < 100 mL/hr
• Hemoglobin is >8.0 g/dL
Chest x-ray (if no x-ray within the last 24 hr, notify the
primary service)
• Enodotracheal tube above carina
• Lung expanded (some atelectasis may be preasent).
pneumothorax not present
If all criteria met, processed to Weaning track
If any criteria not met, reevaluate on an hourly basis or PRN
until patient meets criteria

Weaning track
• Initiate 30-min trial with patient
on PSV 5/5 cm H2O

Weaning criteria
• Patient is awake
• Patient follows commands
• FiO2 <0.5
• Spontaneous respiratory rate
between 6-20 bpm
• Minute ventilation (VE) >50 mL/kg/min
and <150 mL/kg/min
• Can achieve voluntary breath of
≥12mL/kg

Does patient No Is respiratory No Return to previous


meet above rate between SIMV settings
criteria? 6-20 bpm?

Yes Yes

• Notify physician PSV 5/5 cm H2O Re-evaluate for


• Proceed with ectubation another trial in 1 hour
• Oxygen by continuous facemask to
maintain SpO2 >92%
• Increase PS by 5 cm
H2O until VE is
Is VE No >50 mL/kg/min
>50 mL/kg/min? Note: maximum PS is
15 cm H2O
• If the VE goal of
50 mL/kg/min is not
Yes achieved at PS 15 cm
H2O then the patient is
returned o SIMV
Re-evaluate for another settings as prior to
trial in 1 hour weaning attempt than
reassess in 1 hour
472 J. Y. Findlay and M. T. Keegan

Stage Liver Disease (MELD) system for organ  pproach to the Difficult-to-Wean


A
allocation was designed to prioritize patients of Patient
higher illness severity or need for transplant
when considering allocation of donor organs. Weaning from ventilatory support may take
As a result, the acuity of illness of recipients days or even weeks. In general, ICUs weaning
increased substantially in the last decade. Such from the ventilator may account for more than
individuals may be poorer candidates for imme- half of total ventilator time. Based on a large
diate extubation and even their eligibility for body of literature concerning the methodology
“fast-track” protocols may be questionable. and best practices for ventilator weaning, the
Furthermore, in cases of intraoperative difficul- critical care community has refined the approach
ties with large transfusion requirements, early to liberating patients from the ventilator.
extubation may be unwise because of ongoing Guidelines for ventilator weaning have been
bleeding, significant acidosis, volume overload, published by a North American collaborative
and airway and pulmonary edema. Once hemo- group facilitated by the American College of
dynamic stability has been achieved, such Chest Physicians, the American Association for
patients may require diuresis before liberation Respiratory Care and the American College of
from mechanical ventilatory support can be Critical Care Medicine [24]. An International
considered. Citrate used as a preservative in Consensus Conference convened in 2005 and
packed red blood cells is metabolized to bicar- subsequently a number of recommendations for
bonate by the newly-functioning liver and the ventilator weaning were published [25].
resulting metabolic alkalosis may further Weaning should be considered as part of daily
impair respiratory drive. Acetazolamide causes ventilator management with early consideration
diuresis and bicarbonate loss and may amelio- given to a weaning plan in combination with
rate the situation [19, 20]. interruption of sedation [26]. Spontaneous
When pre-transplant encephalopathy exists, breathing trials (SBTs) have been demonstrated
patients often take longer to awaken, leading to to be useful to assess for weaning readiness
a delay in the timing of extubation. Underlying [27]. A 30-min T-piece or low pressure support
lung disease (e.g. alpha-1-antitrypsin defi- trial has been advocated. Patients may be cate-
ciency), pre-operative sepsis, malnourishment gorized into three groups based on the difficulty
and debilitation may require pre-operative ven- and duration of the weaning process: patients
tilation, potentially prolonging the postopera- who pass the initial SBT and are extubated at
tive duration of ventilatory support. Faenza the first attempt, those who require up to three
and colleagues have identified the presence of SBTs or up to 7 days of weaning after the first
early postoperative impairment of PaO2/FiO2 attempt, and patients who require longer than 7
as a predictor of prolonged postoperative ven- days of weaning. Suitable modes of ventilation
tilation [21]. In a study of 10,517 LTx recipi- for weaning remain somewhat controversial, but
ents transplanted between 2002 and 2008, pressure support or assist control modes are
Yuan et al. reported an increased likelihood of considered superior to synchronized intermit-
prolonged mechanical ventilation was associ- tent mandatory ventilation [24, 28–33].
ated with recipients who were older (age > 50 Furthermore, non-invasive ventilatory support
years), female, required pretransplant dialysis, may be very useful in selected patients [34]. In
or had ascites [22]. In those transplanted for addition to the use of protocols for extubation of
acute liver failure, advanced pre-­ operative the “routine” LTx recipient, many ICUs
encephalopathy, severe intraoperative hemody- empower respiratory therapists or nurses to ini-
namic derangements, and renal dysfunction are tiate a daily T-piece trial to assess suitability for
associated with prolonged postoperative venti- weaning. Such an approach decreases the dura-
lation [23]. tion of mechanical ventilation [35].
36  Respiratory Failure and ARDS 473

Some patients will require tracheostomy. The  edation During Mechanical


S
timing of tracheostomy has been a subject of Ventilation
investigation, and in modern critical care prac-
tice, early tracheostomy is advocated when it The appropriate management of sedation during
appears that a ventilated patient will be difficult mechanical ventilation is controversial. In gen-
to wean, although there is still some controversy eral the use of pharmacologic sedation should not
regarding the timing [36–40]. The vast majority be a default in all ventilated patients. Sedation
of LTx recipients do not require tracheostomy should be employed if required to allow appro-
and there is a certain reluctance to introduce priate ventilation and the in the minimum dose
another potential source of infection in an immu- necessary. The Society of Critical Care Medicine
nosuppressed patient. If required, tracheostomy has published guidelines for sedation in the ICU
in this patient population is usually delayed until [48]. These recommend the use of non-­
2 or 3 weeks of mechanical ventilation have been benzodiazepine agents (propofol or dexmedeto-
required. Percutaneous tracheostomy has gained midine) over benzodiazepines for sedation and
acceptance among the ICU community, and there the use of opiates as the first line choice for anal-
is increasing experience in liver transplant recipi- gesia. Periodic—at least daily—interruptions of
ents [41–43]. In patients with a tracheostomy, sedation allow for the evaluation of the continu-
daily unassisted spontaneous breathing trials ing need for sedation and also evaluation of the
have documented beneficial effects on the wean- patient’s neurological status. These “sedation
ing process [44]. holidays” decrease both ventilator time and ICU
stay [49] but do not lead to a clinically important
incidence of post-traumatic stress disorder or
Protocols myocardial ischemia [50, 51]. A combination of
daily spontaneous awakening trials and daily
In addition to the involvement of paramedical SBTs was shown to be superior than daily SBTs
staff in ventilator weaning, other aspects of alone [26].
ventilatory management may be protocolized.
The use of “ventilator bundles” decreases com-
plications—including VAP—and improves out- Non-invasive Ventilation
come. The most widely studied (and probably
implemented) ventilator bundle consists of four The development of ventilators and masks
items: peptic ulcer prophylaxis, deep venous capable of providing non-invasive mechanical
thrombosis prophylaxis, elevation of the head ventilation has considerably added to the arma-
of the bed to at least 30° and the use of daily mentarium of the modern intensivist. The use
sedation ‘holidays’ [45, 46]. The individual of non-invasive ventilatory support (NIV) in
components of the bundle have been criticized the ICU has considerably increased over the
due to the lack of strong evidence and a more past two decades [52]. Continuous positive air-
“evidence supported” bundle was suggested (no way pressure (CPAP) improves oxygenation
unnecessary ventilator tubing changes, alcohol and decreases work of breathing, especially in
based hand hygiene, staff training, sedation and patients with pulmonary edema and/or left
weaning protocols and oral care) [47]. ventricular dysfunction. Biphasic positive air-
Nevertheless, it is most important to have a pro- way pressure (BiPAP) is a non-invasive tech-
tocol that addresses VAP prophylaxis (see nique of ventilatory support in which a flow
below) and allows for the early identification of and pressure generator applies both an expira-
the patient who can be weaned. Such a protocol tory positive airway pressure (EPAP) and an
should be used in every ventilated patient and inspiratory positive airway pressure (IPAP).
assessed every day. BiPAP can improve both oxygenation and ven-
474 J. Y. Findlay and M. T. Keegan

tilation. It is unusual to i­nitiate NIV in LTx Pulmonary Edema


recipients early in the ­post-­operative period.
However, difficult-to-wean patients not yet Pulmonary edema is common in the early post-­
ready for complete withdrawal of ventilatory transplant period. In one series of 300 patients,
support, may be extubated to BiPAP to limit X-ray findings consistent with pulmonary edema
the duration of invasive mechanical ventilation were seen in 45% of patients. This pulmonary
[53]. Furthermore, NIV may be useful in edema was mostly interstitial and associated
patients in whom there is a desire to avoid intu- with other signs of fluid overload. Resolution
bation or re-intubation while a reversible prob- occurred within 3–4 days with fluid restriction
lem (e.g. pulmonary edema due to volume and diuretic use; the outcome was not adversely
overload, or opiate-induced hypoventilation) is affected [1]. Aduen and colleagues reported the
treated [54]. incidence and outcomes of pulmonary edema
resulting in a PaO2/FiO2 ratio of less than 300 in
a series of 100 consecutive liver transplants [60].
The overall prevalence of pulmonary edema was
 entilator Associated Pneumonia
V 52%. Further analysis of the patients with pul-
(VAP) monary edema revealed that those with immedi-
ate pulmonary edema (present immediately
VAP is a common nosocomial problem and a post-­operatively and resolving within 24 h) had
major cause of ICU morbidity [55, 56]. It occurs outcomes that were no different from those with-
in 8–28% of patients receiving invasive mechani- out pulmonary edema. While patients with per-
cal ventilation and is related to the duration of sistent pulmonary edema (18%) or who
ventilation. Associated mortality is high. The developed pulmonary edema in the post-opera-
predominant organisms responsible for infection tive period (9%), had prolonged duration of
are Staphylococcus aureus, Pseudomonas aeru- mechanical ventilation and lengths of ICU stay.
ginosa, and Enterobacteriaceae, but etiologic A higher pre-­operative MELD score was associ-
agents can vary widely depending on the popula- ated with persistent or late pulmonary edema.
tion. VAP is unlikely to develop in the immediate Half of the persistent group and most of the late
postoperative period in patients who are rapidly group had a pulmonary capillary wedge pressure
weaned from the ventilator but the risk is much (PCWP) < 18 mmHg, implying altered pulmo-
higher in patients who are ventilated for longer nary capillary permeability rather than a hydro-
periods of time, especially in the setting of static mechanism as a cause for pulmonary
administration of immunosuppressants. edema. This suggestion is supported by findings
Guidelines aimed at reducing the incidence of in a smaller series where late onset pulmonary
VAP have been published [57]. In addition to edema was not related to fluid volume adminis-
limiting the duration of mechanical ventilation, tered, whereas early onset pulmonary edema was
prevention of VAP involves reducing coloniza- [61]. When pulmonary edema fluid from liver
tion of the aerodigestive tract with pathogenic transplant patients was analyzed, it was found to
bacteria, and preventing aspiration. Investigation be consistent with permeability edema [62].
and treatment of suspected VAP should proceed Suggestions for the precipitant for the capillary
along established guidelines [55, 58]. injury include cytokine release associated with
Bronchoscopy and bronchoalveolar lavage reperfusion of the liver graft and transfusion
(BAL) may be required to guide antimicrobial related lung injury (TRALI) [60, 62, 63]. It may
therapy [59]. Consultation with a transplant well be that different mechanisms are responsi-
infectious disease specialist should be consid- ble for pulmonary edema in different patients.
ered. The diagnosis and treatment of postopera- From a clinical standpoint, once pulmonary
tive pneumonia are discussed in more detail edema is found to be non-hydrostatic, there is no
elsewhere in this book. specific therapy and the usual supportive
36  Respiratory Failure and ARDS 475

­ anagement should be pursued. If the TRALI


m (ECMO). An algorithm for the management of
component is significant, then current practice severe post-transplant hypoxemia in HPS patients
moves towards limiting the use of blood and has recently been proposed [69]. Prolonged post-
blood products during liver transplantation. transplant oxygen therapy (up to almost 2 years)
Likewise, measures to reduce the extent of reper- may be required in these patients and a higher
fusion injury would be expected to reduce the degree of shunting assessed by pre-transplant
occurrence of pulmonary edema. albumin macro-aggregate scanning was predic-
Cardiogenic pulmonary edema, secondary to tive of a slower rate of post-transplant improve-
post-transplant dilated cardiomyopathy, is seen ment [66].
in a small number of patients [64]. It usually
presents a few days after transplantation, often
when the patient has already left the ICU. A Portopulmonary Hypertension
decrease in left ventricular ejection fraction leads
to hydrostatic pulmonary edema and respiratory As discussed elsewhere in this book, the manage-
failure requiring readmission to the ICU with the ment and candidacy of patients with porto-­
need for non-invasive or invasive mechanical pulmonary hypertension for liver transplantation
ventilation. This entity is usually reversible and is controversial. However, the majority of current
supportive treatment includes inotropes, pressors opinion is that these patients are suitable trans-
and diuretics. plant candidates if acceptable hemodynamic
parameters are present [70]. In the immediate
post-transplantation period, these patients may
Hepatopulmonary Syndrome be at a higher risk of pulmonary complications as
well as right ventricular failure and have longer
Liver transplantation is the only successful treat- ICU stays [71]. Initial management should
ment for patients with hepatopulmonary syn- include the continuation of pre-transplant therapy
drome (HPS). These patients may require for pulmonary hypertension and aggressive man-
preoperative oxygen therapy to maintain satisfac- agement of conditions that could worsen pulmo-
tory oxygenation and are at risk of deterioration nary vasoconstriction. A pulmonary artery
in the peri-transplant period. In a review of older catheter is required to adequately manage these
case series, Krowka and colleagues identified a patients. Additionally, pulmonary hemodynam-
pre-transplant resting PaO2 of less than 50 mmHg ics and right ventricular function should be
as a risk factor for poor post-transplant outcome closely monitored. If pulmonary artery pressures
[65]. More recent series report improved survival rise or right heart failure occurs, treatment should
for this group, with outcomes similar to those be advanced to provide pulmonary vasodilators
with less severe HPS as well as liver transplant and right ventricular support. Inhaled nitric oxide
recipients without HPS [66, 67]. The median may be useful during the acute phase [72].
time for post-transplant mechanical ventilation
was less than 1 day, however, a proportion of
patients developed hypoxemic respiratory failure  cute Respiratory Distress
A
requiring ventilatory support of up to 60 days. Syndrome
The development of severe post-transplant
hypoxemia was associated with a 45% mortality ARDS represents an inflammatory response of
[68]. Techniques used to achieve satisfactory the lungs resulting from either a primary lung
oxygenation have included Trendelenburg posi- insult or a systemic insult [73, 74]. Both mecha-
tioning (to counteract orthodeoxia), the use of nisms may occur in patients with liver disease.
inhaled pulmonary vasodilators, methylene blue The Berlin Definition of ARDS, published in
infusion, high frequency oscillatory ventilation 2012, has replaced the 1994 American-European
and extracorporeal membrane oxygenation consensus definition [75, 76]. A diagnosis of
476 J. Y. Findlay and M. T. Keegan

ARDS by the Berlin Definition requires that each tion of anesthesia, giving rise to ARDS [62].
of the following be present: respiratory symp- Months or years after transplantation, the immu-
toms within 1 week of a known clinical insult; nocompromised recipient may present with
bilateral pulmonary opacities on chest radiograph pneumonia or septic shock and ARDS may
or CT scan not fully explained by effusions, lobar develop. Graft failure, whether caused by pri-
or lung collapse or nodules; respiratory failure mary non-function, acute rejection or vascular
not fully explained by cardiac failure or fluid occlusion, may also lead to the development of
overload. An objective assessment (e.g. echocar- ARDS. Furthermore, treatment of rejection using
diography) must be performed to exclude hydro- monoclonal antibody therapy (e.g. basiliximab)
static edema, if there is no risk factor for may cause an inflammatory lung insult leading to
ARDS. The PaO2/FiO2 (P/F) ratio is then used to diffuse alveolar edema or hemorrhage and/or
differentiate between mild, moderate and severe ARDS [83].
impairment of oxygenation as follows: mild—PF Zhao and colleagues evaluated the incidence
ratio > 200 but ≤300, on ventilator settings that of ARDS (defined by the Berlin criteria) with the
include positive end expiratory pressure (PEEP) associated risk factors and implications in adult
or continuous positive airway pressure (CPAP) of patients who underwent LTx at a single center
5 cm H2O or more; moderate—PF ratio > 100 but between 2004 and 2013 [84]. Of 1726 patients,
≤200 with PEEP ≥5 cm H2O; severe—PF ARDS occurred in 4.1% and was associated with
ratio ≤ 100, with PEEP ≥5 cm H2O. (The term preoperative encephalopathy, the requirement for
“acute lung injury”, used in the 1994 definition, pre-transplant intubation, an increased bilirubin
has been dropped.) concentration, and high intraoperative pressor
The frequency with which ARDS is seen in requirements. The development of ARDS was
the ICU has prompted much investigation into associated with increased mortality, duration of
the causes, prevention and treatment of this life-­ mechanical ventilation, and length of hospital
threatening syndrome. The National Institutes of stay. In addition to the use of low tidal volume
Health in the United States has funded the multi-­ ventilation as a management strategy for patients
center ARDSNet group through the National with ARDS, PEEP is routinely used to prevent
Heart Lung and Blood Institute. This group per- de-recruitment and atelectasis and to improve
formed a landmark clinical trial (ARMA) that oxygenation. By keeping the lungs “open”, PEEP
demonstrated that ventilation with low tidal vol- decreases shear stresses that would worsen lung
umes (6 mL/kg ideal body weight) was associ- injury. Despite a number of well-designed stud-
ated with decreased morbidity and mortality ies, the optimum level of PEEP remains to be elu-
compared with ventilation using tidal volumes of cidated, perhaps because of the heterogeneity of
12 mL/kg ideal body weight [33, 77]. Patients patients in the clinical trials [85–89].
with severe liver disease were excluded from this There are theoretical concerns regarding the
study. Although the use of low tidal volume ven- use of PEEP—especially at high levels—in
tilation and other advances in ICU care have led patients with liver disease. Increased intratho-
to an improvement in hospital survival over time, racic pressure that occurs as a result of PEEP
ARDS-related mortality remains high, and survi- application causes a decrease in venous return
vors can develop long-lasting cognitive, psycho- and can lead to hepatic venous engorgement.
logical and physical impairments [78–80]. This may cause ischemic liver damage, to which
ARDS may develop prior to LTx in the setting the newly engrafted liver is especially vulnerable.
of sepsis or acute liver failure. Peri-operatively, However, Saner and colleagues have demon-
ARDS may develop due to the inflammatory strated that although application of PEEP to
stimulus of the surgical insult or allograft reper- mechanically ventilated LTx recipients increased
fusion. In addition, TRALI that is a form of central venous and pulmonary capillary wedge
ARDS—may develop [81, 82]. Aspiration pneu- pressure, hepatic inflow and outflow of the
monitis may develop pre-operatively or at induc- transplanted livers (both cadaveric and living
­
36  Respiratory Failure and ARDS 477

donor) were not affected by PEEP levels up to with critical hypoxemia after LTx in whom use of
about 15 cm H2O [90, 91]. It is unknown whether the prone position and application of 15 cm H2O
higher levels of applied PEEP cause ischemic of PEEP were lifesaving [103].
hepatic damage, however, these are rarely neces- “Rescue” therapies for critical hypoxemia in
sary. When PEEP is required to achieve accept- ARDS include the use of inhaled nitric oxide and
able oxygenation in patients with ARDS, it inhaled prostaglandins. These therapies have not
should be applied, recognizing that adequate sys- been proven in randomized clinical trials to
temic oxygenation is essential for optimum improve outcome, though they may be life-­saving
hepatic function. In addition, lungs that have in selected cases [104–107]. Both agents selec-
been injured demonstrate a reduction in compli- tively vasodilate pulmonary arterioles in aerated
ance that will offset transmission of pressure to lung units, thus improving ventilation: perfusion
the liver. matching and subsequently improving
The use of recruitment maneuvers in ARDS is oxygenation.
controversial [92]. Systematic reviews of the Developments in the technology of extracor-
available data have been inconclusive [93, 94]. poreal membrane oxygenation (ECMO) over the
Although unable to definitively advise for or past two decades have made ECMO more practi-
against recruitment maneuvers, the authors of cal and accessible. It is increasingly used as sal-
these reviews suggested consideration of recruit- vage therapy for patients with a variety of cardiac
ment maneuvers for life-threatening hypoxemia and respiratory conditions, including ARDS
in ARDS. They did not specifically address the [108]. Park et al. described the use of veno-­
impact of recruitment maneuvers on hepatic venous ECMO in 18 adult patients who devel-
perfusion. oped respiratory failure (12 with pneumonia, 6
When tidal volumes are limited as a lung pro- with ARDS) after LTx that was refractory to
tective strategy, the respiratory rate may be mechanical ventilation and concurrent inhaled
increased to compensate and maintain adequate nitric oxide. Eight of the patients survived [109].
minute ventilation. When respiratory rates in Other reports describing the use of ECMO in
the high 20s and 30s are required, dynamic patients with respiratory failure after LTx are
hyperinflation (“auto-PEEP”) can develop. Such anticipated.
high respiratory rates also increase the shear
stresses on the lungs because of the high fre- Conclusion
quency of opening and closing of lung units. Although pulmonary complications are com-
Therefore, the technique of permissive hyper- mon in the immediate post-liver transplant
capnia may be used [95]. The elevation of car- period, the majority of these do not lead to
bon dioxide levels may also have implications significant morbidity. However, there are
for the liver graft, although there is minimal patients who will develop post-transplant
data in this regard. respiratory failure secondary to either pre-
A variety of other techniques have been used existing conditions or as a consequence of the
in the management of patients with ARDS, espe- procedure. Whilst there is little literature spe-
cially when hypoxemia is severe and refractory cific to the management of the liver transplant
[96]. They have been met with varying levels of patient with respiratory failure, there is a con-
success and none have demonstrated as dramatic siderable body of evidence relating to the
an impact as the low tidal volume strategy. These ventilatory and ICU management of respira-
techniques include the prolonged use of non-­ tory failure. Clinicians caring for these
depolarizing muscle relaxants, high frequency patients should follow current best practices
oscillation, airway pressure release ventilation, for the ICU management of respiratory fail-
and prone positioning [97–102]. Experience with ure, integrating approaches specific to liver
most techniques in the LTx recipient is minimal, transplant patients as and when sufficient evi-
although Sykes and colleagues describe a patient dence is available.
478 J. Y. Findlay and M. T. Keegan

References c­riteria in a liver transplant recipient population.


Liver Transpl. 2001;7:777–82.
18. Glanemann M, Hoffmeister R, Neumann U, et al.
1. Golfieri R, Giampalma E, Morselli Labate AM, Fast tracking in liver transplantation: which patient
et al. Pulmonary complications of liver trans- benefits from this approach? Transplant Proc.
plantation: radiological appearance and statistical 2007;39:535–6.
evaluation of risk factors in 300 cases. Eur Radiol. 19. Carton EG, Plevak DJ, Kranner PW, Rettke SR,
2000;10:1169–83. Geiger HJ, Coursin DB. Perioperative care of
2. Hong SK, Hwang S, Lee SG, et al. Pulmonary the liver transplant patient: Part 2. Anesth Analg.
complications following adult liver transplantation. 1994;78:382–99.
Transplant Proc. 2006;38:2979–81. 20. Carton EG, Rettke SR, Plevak DJ, Geiger HJ,
3. Panfili E, Nicolini D, Polverini V, Agostini A, Kranner PW, Coursin DB. Perioperative care of
Vivarelli M, Giovagnoni A. Importance of radiologi- the liver transplant patient: Part 1. Anesth Analg.
cal detection of early pulmonary acute complica- 1994;78:120–33.
tions of liver transplantation: analysis of 259 cases. 21. Faenza S, Ravaglia MS, Cimatti M, Dante A, Spedicato
Radiol Med. 2015;120:413–20. S, Labate AM. Analysis of the causal factors of pro-
4. Ammori JB, Pelletier SJ, Lynch R, et al. Incremental longed mechanical ventilation after orthotopic liver
costs of post-liver transplantation complications. J transplant. Transplant Proc. 2006;38(4):1131.
Am Coll Surg. 2008;206:89–95. 22. Yuan H, Tuttle-Newhall JE, Chawa V, et al.
5. Garcia N Jr, Mihas AA. Hepatic hydrothorax: patho- Prognostic impact of mechanical ventilation after
physiology, diagnosis, and management. J Clin liver transplantation: a national database study. Am J
Gastroenterol. 2004;38:52–8. Surg. 2014;208:582–90.
6. Mandell MS, Campsen J, Zimmerman M, Biancofiore 23. Lee S, Jung HS, Choi JH, et al. Perioperative risk
G, Tsou MY. The clinical value of early extubation. factors for prolonged mechanical ventilation after
Curr Opin Organ Transplant. 2009;14:297–302. liver transplantation due to acute liver failure.
7. Steadman RH. Con: immediate extubation for Korean J Anesthesiol. 2013;65:228–36.
liver transplantation. J Cardiothorac Vasc Anesth. 24. MacIntyre NR, Cook DJ, Ely EW Jr, et al. Evidence-­
2007;21:756–7. based guidelines for weaning and discontinuing
8. Ozier Y, Klinck JR. Anesthetic management of ventilatory support: a collective task force facilitated
hepatic transplantation. Curr Opin Anaesthesiol. by the American College of Chest Physicians; the
2008;21:391–400. American Association for Respiratory Care; and the
9. Myles PS, Daly DJ, Djaiani G, Lee A, Cheng DC. A American College of Critical Care Medicine. Chest.
systematic review of the safety and effectiveness 2001;120:375S–95S.
of fast-track cardiac anesthesia. Anesthesiology. 25. Boles JM, Bion J, Connors A, et al. Weaning
2003;99:982–7. from mechanical ventilation. Eur Respir J.
10. Campos JH. Fast track in thoracic anesthesia and 2007;29:1033–56.
surgery. Curr Opin Anaesthesiol. 2009;22:1–3. 26. Girard TD, Kress JP, Fuchs BD, et al. Efficacy and
11. Wu J, Rastogi V, Zheng SS. Clinical practice of early safety of a paired sedation and ventilator wean-
extubation after liver transplantation. Hepatobiliary ing protocol for mechanically ventilated patients
Pancreat Dis Int. 2012;11:577–85. in intensive care (awakening and breathing con-
12. Findlay JY, Jankowski CJ, Vasdev GM, et al. Fast trolled trial): a randomised controlled trial. Lancet.
track anesthesia for liver transplantation reduces 2008;371:126–34.
postoperative ventilation time but not intensive care 27. Esteban A, Alia I, Tobin MJ, et al. Effect of spontane-
unit stay. Liver Transpl. 2002;8:670–5. ous breathing trial duration on outcome of attempts
13. Aniskevich S, Pai SL. Fast track anesthesia for liver to discontinue mechanical ventilation. Spanish Lung
transplantation: review of the current practice. World Failure Collaborative Group. Am J Respir Crit Care
J Hepatol. 2015;7:2303–8. Med. 1999;159:512–8.
14. Mandell MS, Lezotte D, Kam I, Zamudio S. Reduced 28. Brochard L, Thille AW. What is the proper approach
use of intensive care after liver transplantation: to liberating the weak from mechanical ventilation?
patient attributes that determine early transfer to sur- Crit Care Med. 2009;37:S410–5.
gical wards. Liver Transpl. 2002;8:682–7. 29. Epstein SK. Weaning from ventilatory support. Curr
15. Xia VW, Taniguchi M, Steadman RH. The changing Opin Crit Care. 2009;15:36–43.
face of patients presenting for liver transplantation. 30. Eskandar N, Apostolakos MJ. Weaning from
Curr Opin Organ Transplant. 2008;13:280–4. mechanical ventilation. Crit Care Clin. 2007;23:263–
16. Mandell MS, Stoner TJ, Barnett R, et al. A mul- 74. x
ticenter evaluation of safety of early extuba- 31. MacIntyre N. Discontinuing mechanical ventilatory
tion in liver transplant recipients. Liver Transpl. support. Chest. 2007;132:1049–56.
2007;13:1557–63. 32. Mauri T, Pivi S, Bigatello LM. Prolonged mechanical
17. Biancofiore G, Romanelli AM, Bindi ML, et al. Very ventilation after critical illness. Minerva Anestesiol.
early tracheal extubation without predetermined 2008;74:297–301.
36  Respiratory Failure and ARDS 479

33. The Acute Respiratory Distress Syndrome Network. 47. Rello J, Lode H, Cornaglia G, Masterton R. A
Ventilation with lower tidal volumes as compared European care bundle for prevention of ventilator-­
with traditional tidal volumes for acute lung injury associated pneumonia. Intensive Care Med.
and the acute respiratory distress syndrome. N Engl 2010;36:773–80.
J Med. 2000;342:1301–8. 48. Barr J, Fraser GL, Puntillo K, et al. Clinical practice
34. Burns KE, Meade MO, Premji A, Adhikari guidelines for the management of pain, agitation,
NK. Noninvasive positive-pressure ventilation and delirium in adult patients in the intensive care
as a weaning strategy for intubated adults with unit. Crit Care Med. 2013;41:263–306.
respiratory failure. Cochrane Database Syst Rev. 49. Kress JP, Pohlman AS, O’Connor MF, Hall JB. Daily
2013;12:CD004127. interruption of sedative infusions in critically ill
35. Ely EW, Meade MO, Haponik EF, et al. Mechanical patients undergoing mechanical ventilation. N Engl
ventilator weaning protocols driven by nonphysician J Med. 2000;342:1471–7.
health-care professionals: evidence-based clinical 50. Kress JP, Gehlbach B, Lacy M, Pliskin N,
practice guidelines. Chest. 2001;120:454S–63S. Pohlman AS, Hall JB. The long-term psycho-
36. Frustos-Vivar F, Esteban A, Paezteguia C, Anzueto A, logical effects of daily sedative interruption on
et al. Outcome of mechanically ventilated patients who critically ill patients. Am J Respir Crit Care Med.
require a tracheostomy. Crit Care Med. 2005;33:290–8. 2003;168:1457–61.
37. King C, Moores LK. Controversies in mechanical 51. Kress JP, Vinayak AG, Levitt J, et al. Daily seda-
ventilation: when should a tracheotomy be placed? tive interruption in mechanically ventilated patients
Clin Chest Med. 2008;29:253–63. vi at risk for coronary artery disease. Crit Care Med.
38. Andriolo BN, Andriolo RB, Saconato H, Atallah 2007;35:365–71.
AN, Valente O. Early versus late tracheostomy for 52. Nava S, Hill N. Non-invasive ventilation in acute
critically ill patients. Cochrane Database Syst Rev. respiratory failure. Lancet. 2009;374:250–9.
2015;1:CD007271. 53. Ferrer M. Non-invasive ventilation as a weaning
39. Szakmany T, Russell P, Wilkes AR, Hall JE. Effect of tool. Minerva Anestesiol. 2005;71:243–7.
early tracheostomy on resource utilization and clini- 54. Antonelli M, Conti G, Bufi M, et al. Noninvasive
cal outcomes in critically ill patients: meta-­analysis ventilation for treatment of acute respiratory failure
of randomized controlled trials. Br J Anaesth. in patients undergoing solid organ transplantation: a
2015;114:396–405. randomized trial. JAMA. 2000;283:235–41.
40. Siempos II, Ntaidou TK, Filippidis FT, Choi 55. Chastre J, Fagon JY. Ventilator-associated pneumo-
AM. Effect of early versus late or no tracheos- nia. Am J Respir Crit Care Med. 2002;165:867–903.
tomy on mortality and pneumonia of critically ill 56. Hunter JD. Ventilator associated pneumonia. BMJ.
patients receiving mechanical ventilation: a system- 2012;344:e3325.
atic review and meta-analysis. Lancet Respir Med. 57. Coffin SE, Klompas M, Classen D, et al. Strategies
2015;3:150–8. to prevent ventilator-associated pneumonia in acute
41. Pirat A, Zeyneloglu P, Candan S, et al. Percutaneous care hospitals. Infect Control Hosp Epidemiol.
dilational tracheotomy in solid-organ transplant 2008;29(Suppl 1):S31–40.
recipients. Transplant Proc. 2004;36:221–3. 58. America ATSIDSo. Guidelines for the manage-
42. Waller EA, Aduen JF, Kramer DJ, et al. Safety of ment of adults with hospital-acquired, ventilator-­
percutaneous dilatational tracheostomy with direct associated, and healthcare-associated pneumonia.
bronchoscopic guidance for solid organ allograft Am J Respir Crit Care Med. 2005;171:388–416.
recipients. Mayo Clin Proc. 2007;82:1502–8. 59. Canadian Critical Care trials Group. A randomized
43. Royo-Villanova Reparaz M, Andreu Soler E, Sánchez trial of diagnostic techniques for ventilator-­associated
Cámara S, et al. Utility of percutaneous dilatational pneumonia. N Engl J Med. 2006;355:2619–30.
tracheostomy in the immediate postoperative period 60. Aduen JF, Stapelfeldt WH, Johnson MM, et al.
of liver transplant. Cir Esp. 2015;93:91–6. Clinical relevance of time of onset, duration, and
44. Jubran A, Grant BJ, Duffner LA, et al. Effect of type of pulmonary edema after liver transplantation.
pressure support vs unassisted breathing through a Liver Transpl. 2003;9:764–71.
tracheostomy collar on weaning duration in patients 61. Snowden CP, Hughes T, Rose J, Roberts
requiring prolonged mechanical ventilation: a ran- DR. Pulmonary edema in patients after liver trans-
domized trial. JAMA. 2013;309:671–7. plantation. Liver Transpl. 2000;6:466–70.
45. Resar R, Pronovost P, Haraden C, Simmonds 62. Yost CS, Matthay MA, Gropper MA. Etiology of
T, Rainey T, Nolan T. Using a bundle approach acute pulmonary edema during liver transplantation:
to improve ventilator care processes and reduce a series of cases with analysis of the edema fluid.
ventilator-­associated pneumonia. Jt Comm J Qual Chest. 2001;119:219–23.
Patient Saf. 2005;31:243–8. 63. Mueller AR, Platz KP, Haak M, et al. The release
46. Dodek P, Keenan S, Cook D, et al. Evidence-based of cytokines, adhesion molecules, and extracellu-
clinical practice guideline for the prevention of lar matrix parameters during and after reperfusion
ventilator-associated pneumonia. Ann Intern Med. in human liver transplantation. Transplantation.
2004;141:305–13. 1996;62:1118–26.
480 J. Y. Findlay and M. T. Keegan

64. Sampathkumar P, Lerman A, Kim BY, et al. Post-­ 80. Herridge MS, Tansey CM, Matte A, et al. Functional
liver transplantation myocardial dysfunction. Liver disability 5 years after acute respiratory distress syn-
Transpl Surg. 1998;4:399–403. drome. N Engl J Med. 2011;364:1293–304.
65. Krowka MJ, Porayko MK, Plevak DJ, et al. 81. Toy P, Popovsky MA, Abraham E, et al. Transfusion-­
Hepatopulmonary syndrome with progressive related acute lung injury: definition and review. Crit
hypoxemia as an indication for liver transplantation: Care Med. 2005;33:721–6.
case reports and literature review. Mayo Clin Proc. 82. Triulzi DJ. Transfusion-related acute lung injury:
1997;72:44–53. current concepts for the clinician. Anesth Analg.
66. Gupta S, Castel H, Rao RV, et al. Improved sur- 2009;108:770–6.
vival after liver transplantation in patients with 83. Massart A, Heenen S, Bejjani G, et al. Does basil-
hepatopulmonary syndrome. Am J Transplant. iximab induction trigger lifethreatening ARDS and
2010;10(2):354–63. shock in young patients after kidney transplantation?
67. Iyer VN, Swanson KL, Cartin-Ceba R, et al. Clin Nephrol. 2015;83:61–70.
Hepatopulmonary syndrome: favorable out- 84. Zhao W, Ge X, Sun K, et al. Acute respiratory dis-
comes in the MELD exception era. Hepatology. tress syndrome after orthotopic liver transplantation.
2013;57:2427–35. J Crit Care. 2016;31:163–7.
68. Nayyar D, Man HS, Granton J, Gupta S. Defining 85. Brower RG, Lanken PN, MacIntyre N, et al. Higher
and characterizing severe hypoxemia after liver versus lower positive end-expiratory pressures in
transplantation in hepatopulmonary syndrome. Liver patients with the acute respiratory distress syn-
Transpl. 2014;20:182–90. drome. N Engl J Med. 2004;351:327–36.
69. Nayyar D, Man HS, Granton J, Lilly LB, Gupta 86. Villar J, Kacmarek R, Perez-Mendez L, Aguirre-­
S. Proposed management algorithm for severe hypox- Jaime A. A high positive-end expiratory pressure,
emia after liver transplantation in the hepatopulmo- low tidal volume ventilatory strategy improves out-
nary syndrome. Am J Transplant. 2015;15:903–13. come in persistent acute respiratory distress syn-
70. Swanson KL, Wiesner RH, Nyberg SL, Rosen CB, drome: a randomized control trial. Crit Care Med.
Krowka MJ. Survival in portopulmonary hyperten- 2006;34:1311–8.
sion: Mayo Clinic experience categorized by treat- 87. Mercat A, Richard JC, Vielle B, et al. Positive end-­
ment subgroups. Am J Transplant. 2008;8:2445–53. expiratory pressure setting in adults with acute lung
71. Bozbas SS, Eyuboglu FO, Arslan NG, Ergur FO, injury and acute respiratory distress syndrome: a ran-
Karakayali H, Haberal M. The prevalence and the domized controlled trial. JAMA. 2008;299:646–55.
impact of portopulmonary hypertension on postop- 88. Meade MO, Cook DJ, Guyatt GH, et al. Ventilation
erative course in patients undergoing liver transplan- strategy using low tidal volumes, recruitment
tation. Transplant Proc. 2009;41:2860–3. maneuvers, and high positive end-expiratory pres-
72. Ramsay M. Portopulmonary hypertension and right sure for acute lung injury and acute respiratory
heart failure in patients with cirrhosis. Curr Opin distress syndrome: a randomized controlled trial.
Anaesthesiol. 2010;23:145–50. JAMA. 2008;299:637–45.
73. Wheeler AP, Bernard GR. Acute lung injury and 89. Briel M, Meade M, Mercat A, et al. Higher vs lower
the acute respiratory distress syndrome: a clinical positive end-expiratory pressure in patients with
review. Lancet. 2007;369:1553–64. acute lung injury and acute respiratory distress syn-
74. Ware LB, Matthay MA. The acute respiratory dis- drome: systematic review and meta-analysis. JAMA.
tress syndrome. N Engl J Med. 2000;342:1334–49. 2010;303:865–73.
75. Ranieri VM, Rubenfeld GD, Thompson BT, et al. 90. Saner FH, Olde Damink SW, Pavlakovic G, et al.
Acute respiratory distress syndrome: the Berlin How far can we go with positive end-expiratory
Definition. JAMA. 2012;307:2526–33. pressure (PEEP) in liver transplant patients? J Clin
76. Bernard GR, Artigas A, Brigham KL, et al. Report Anesth. 2010;22:104–9.
of the American-European Consensus Conference 91. Saner FH, Olde Damink SW, Pavlakovic G, et al.
on acute respiratory distress syndrome: definitions, Positive end-expiratory pressure induces liver con-
mechanisms, relevant outcomes, and clinical trial gestion in living donor liver transplant patients: myth
coordination. Consensus Committee. J Crit Care. or fact. Transplantation. 2008;85:1863–6.
1994;9:72–81. 92. Brower RG, Morris A, MacIntyre N, et al. Effects
77. Malhotra A. Low-tidal-volume ventilation in the of recruitment maneuvers in patients with acute lung
acute respiratory distress syndrome. N Engl J Med. injury and acute respiratory distress syndrome venti-
2007;357:1113–20. lated with high positive end-expiratory pressure. Crit
78. Erickson SE, Martin GS, Davis JL, Matthay MA, Care Med. 2003;31:2592–7.
Eisner MD. Recent trends in acute lung injury mor- 93. Fan E, Wilcox ME, Brower RG, et al. Recruitment
tality: 1996–2005. Crit Care Med. 2009;37:1574–9. maneuvers for acute lung injury: a systematic review.
79. Villar J, Blanco J, Anon JM, et al. The ALIEN study: Am J Respir Crit Care Med. 2008;178:1156–63.
incidence and outcome of acute respiratory distress 94. Suzumura EA, Figueiro M, Normilio-Silva K, et al.
syndrome in the era of lung protective ventilation. Effects of alveolar recruitment maneuvers on clinical
Intensive Care Med. 2011;37:1932–41. outcomes in patients with acute respiratory distress
36  Respiratory Failure and ARDS 481

syndrome: a systematic review and meta-analysis. 103. Sykes E, Cosgrove JF, Nesbitt ID, O’Suilleabhain
Intensive Care Med. 2014;40:1227–40. CB. Early noncardiogenic pulmonary edema
95. Kavanagh BP, Laffey JG. Hypercapnia: per- and the use of PEEP and prone ventilation after
missive and therapeutic. Minerva Anestesiol. emergency liver transplantation. Liver Transpl.
2006;72:567–76. 2007;13:459–62.
96. Pipeling MR, Fan E. Therapies for refractory hypox- 104. Afshari A, Brok J, Moller AM, Wetterslev

emia in acute respiratory distress syndrome. JAMA. J. Inhaled nitric oxide for acute respiratory dis-
2010;304:2521–7. tress syndrome and acute lung injury in adults
97. Papazian L, Forel JM, Gacouin A, et al. and children: a systematic review with meta-anal-
Neuromuscular blockers in early acute respiratory ysis and trial sequential analysis. Anesth Analg.
distress syndrome. N Engl J Med. 2010;363:1107–16. 2011;112:1411–21.
98. Ferguson ND, Cook DJ, Guyatt GH, et al. High-­ 105. Griffiths MJ, Evans TW. Inhaled nitric oxide therapy
frequency oscillation in early acute respiratory dis- in adults. N Engl J Med. 2005;353:2683–95.
tress syndrome. N Engl J Med. 2013;368:795–805. 106. Spieth PM, Zhang H. Pharmacological therapies for
99. Young D, Lamb SE, Shah S, et al. High-frequency acute respiratory distress syndrome. Curr Opin Crit
oscillation for acute respiratory distress syndrome. Care. 2014;20:113–21.
N Engl J Med. 2013;368:806–13. 107. Jain R, DalNogare A. Pharmacological therapy for
100. Maung AA, Kaplan LJ. Airway pressure release ven- acute respiratory distress syndrome. Mayo Clin
tilation in acute respiratory distress syndrome. Crit Proc. 2006;81:205–12.
Care Clin. 2011;27:501–9. 108. Brodie D, Bacchetta M. Extracorporeal membrane
101. Gattinoni L, Taccone P, Carlesso E, Marini JJ. Prone oxygenation for ARDS in adults. N Engl J Med.
position in acute respiratory distress syndrome. 2011;365:1905–14.
Rationale, indications, and limits. Am J Respir Crit 109. Park YH, Hwang S, Park HW, et al. Effect of pul-
Care Med. 2013;188:1286–93. monary support using extracorporeal membrane
102. Guerin C, Reignier J, Richard JC, et al. Prone posi- oxygenation for adult liver transplant recipi-
tioning in severe acute respiratory distress syn- ents with respiratory failure. Transplant Proc.
drome. N Engl J Med. 2013;368:2159–68. 2012;44:757–61.
Part V
Critical Care Medicine for Liver Surgery
Postoperative Care of Living
Donor for Liver Transplant 37
Sean Ewing, Tadahiro Uemura,
and Sathish Kumar

Keywords and in 2014, 1,821 liver patients died while wait-


Donor risk · Left-lobe graft · Liver transplan- ing for a transplant [1]. In 2017, 8,082 liver trans-
tation · Living donor · Mortality · Perioperative plants were performed in the US, of which 367
care · Epidemiology · Ethics · Bile leak were living donor liver transplants. To overcome
Pulmonary embolism · Liver failure the lack of deceased donors, living donor liver
transplantation (LDLT) is becoming increasingly
common since it was introduced into clinical
practice in 1989.
Introduction This chapter reviews the perioperative care of
living donor liver transplantation including the
Liver transplantation is a standard treatment epidemiology of living donation and its variabil-
modality for patients with end-stage liver disease ity internationally. International guidelines and
but the scarcity of cadaveric donors has led to US guidelines regarding preoperative assessment
long waiting times for transplant procedures in are included to stress the importance of patient
this severely ill group of patients and many, selection with living donation. Trends in surgical
unfortunately, die before they ever receive a approach including laparoscopic liver harvesting
transplant. Current UNOS (United Network for and their implication for donor morbidity and
Organ Sharing) data highlights this problem. postoperative care are addressed. Common com-
According to UNOS data, 15,000 patients were plications and an overview of routine postopera-
waiting for a liver transplant in the United States tive care are reviewed and novel strategies for
postoperative pain management and monitoring
of coagulopathy have been highlighted.
S. Ewing, MD
Department of Anesthesiology, University of
Michigan Health System, Ann Arbor, MI, USA
T. Uemura, MD, PhD Epidemiology
Abdominal Transplantation and Hepatobiliary
Surgery, Allegheny General Hospital, Seven thousand one hundred and twenty seven
Pittsburgh, PA, USA
liver transplants were performed in the USA in
S. Kumar, MBBS (*) 2015. Only 359 were LDLT, an 22% (n = 79)
Department of Anesthesiology, University of
Michigan, Ann Arbor, MI, USA increase from the previous year. The number of
e-mail: ssathish@med.umich.edu LDLT performed in the USA reached a peak in

© Springer International Publishing AG, part of Springer Nature 2018 485


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_37
486 S. Ewing et al.

Fig. 37.1 Liver 8000


transplantation from
1990 to 2015 in the US 7000
comparing decease and
living donor liver 6000
transplant (UNOS data)
5000

4000 “Deceased Donor”

3000 “Living Donor”

2000

1000

0
1990
1992
1994
1996
1998
2000
2002
2004
2006
2008
2010
2012
2014
2001 and then decreased dramatically after case donor remnant liver volume. Based on graft vol-
reports of donor deaths and only in 2015 started ume and remnant liver volume, right or left lobe
to increase again (Fig. 37.1) [1–3]. In contrast, graft donation is determined. The remnant liver
the Toronto Live Donor Liver Transplant Program volume, generally, should be more than 35% of
and certain Asian programs have continued to the whole volume [9]. Right or extended right
expand and even doubled LDLT to address a lack lobe donation is associated with more frequent
of deceased donation over the same timeframe, and severe complications than non-right lobe
with no deaths reported in certain high-volume resections [3]. Though attempts have been made
centers [4, 5]. to improve patient screening and surgical tech-
nique, there has been limited reduction in mor-
bidity associated with right lobe donation to
Current Surgical Technique donors and increasingly left lobe grafts are used
to shift risk away from the donor, if a suitable
An adult-to-adult living donor liver transplanta- donor is available [10–12]. Though technically
tion (A2ALL) recipient requires about 30–60% demanding, right posterior sector grafts may be
of the donor total liver mass, necessitating an performed under certain conditions: satisfactory
entire right or left donor lobe resection [6]. To minimum volume requirement for the recipient
provide sufficient liver mass to the recipient, (40% standard liver volume); graft size greater
resection of the left lateral lobe, entire right or than the left lobe plus caudate; extrahepatically
left lobe of the liver is required. Early during the branching portal vein, hepatic artery; and the
development of A2ALL, it became evident that procurement of RPS graft accompanies mini-
the left lateral lobe was insufficient to provide mum morbidity [8].
adequate liver mass and hence an entire right or Multiple centers now report donor hepatec-
left lobe resection was required for A2ALL [7]. tomy performed using complete laparoscopic
Of note, left lateral segment remains an option (graft removed through small incision such as a
for pediatric recipients due to excellent graft Pfannenstiel), hand-assisted (through mid or
quality in living donation, ease with laparo- low transverse incisions), or hybrid (through
scopic approach and smaller required graft size, mini laparotomy) approaches for either right or
and lower risk to donor [8]. The decision to use left hepatectomies [13]. Laparoscopic donor
a right or left lobe is often based on Computed surgery requires a high degree of expertise and
Tomography (CT) volumetry (Fig. 37.2) and the skill and therefore the Second International
37  Postoperative Care of Living Donor for Liver Transplant 487

a b

c d

Fig. 37.2  3D-CT image of a liver. Using software, the 3-dimensional image shows the drainage area of middle
volume of each vessel branch can be automatically calcu- hepatic vein (MHV) tributaries (V5, V8; indicated by
lated before an operation. (a, b) Construction of a red). (Adapted from Yonemura et al. Liver Transplantation
3-dimensional image shows the perfusion area (orange 2005; 11:1556–62), page 1558, this figure)
color) of the right portal vein. (c, d) The construction of a

Consensus Conference on Laparoscopic Liver are discussed in detail in the later part of the
Resections in 2015 stated: “On the basis of chapter.
potential and unknown risk to donor and level
of surgical skills required, this procedure can-
not be recommended for wide introduction at Ethical Issues
this time [14].” In centers experienced with
laparoscopic hepatobiliary surgery, the laparo- One of the most important tenets of medicine is
scopic approach produces equivalent results for non-maleficence or “First, do no harm.” In order to
the recipient and may improve donor postoper- properly weigh the ethical issues, a precise under-
ative pain, hospital length of stay and is cos- standing of risks and benefits to the donor and
metically more acceptable [8]. Postoperative recipient is needed. The ethical challenge in LDLT
complications and pain management in donors is that a healthy individual undergoes a lengthy
488 S. Ewing et al.

major operation with no personal health benefit but infections in donors who received blood
significant surgical, medical psychosocial and transfusion [6]. However, a causative rela-
financial risk with the possibility of a poor outcome tionship is difficult to prove as this could
for the recipient [15, 16]. Evaluation should include possibly be related to complex surgical dis-
a multidisciplinary approach involving psychologi- section, leading to increased blood loss
cal evaluation of both recipient and donor. Detailed necessitating blood transfusion.
guidelines are available through the United (d) History of smoking: There has been an asso-
Network of Organ Sharing regarding donor psy- ciation bile leak with a history of smoking in
chological assessment, provision of an independent a recent study [21]. This association may be
living donor advocate (IDLA), donor reimburse- related to impaired healing related to
ment and informed consent [17, 18]. Preserving the impaired anastomotic blood flow, but the
health of the donor and excluding a donor if they exact cause is also unclear as smoking affects
are not an optimal candidate are crucial and should multiple organ systems.
supersede any other concerns of the transplant
team [16]. Recipients of LDLT should meet the
same listing criteria required for deceased donor Postoperative Care
liver transplantation (DDLT). MELD scores
(Model for End-stage Liver Disease) are used In an effort to minimize postoperative complica-
along with approval of the multidisciplinary trans- tions, close surveillance of the donor is essential.
plant team. Recently published international guide- Optimal management will ensure early ambula-
lines have highlighted major considerations related tion and discharge. Even though these patients are
to donor and recipient safety, though national and healthy when they enter the operating room, they
institutional policies may vary [8, 19]. need careful and intensive monitoring for the first
24 h postoperatively. Experience with this specific
patient population is necessary to allow rapid diag-
Risk Factors for Postoperative nosis and treatment of any postoperative problems
Complication in Living Donors to prevent morbidity. To ensure close surveillance,
most centers admit donors to an intensive care unit
Several independent risk factors have been asso- (ICU) for over-night monitoring [7, 11, 22] but a
ciated with an increased incidence of post- opera- step down unit, intermediary care unit or postanes-
tive complications such as biliary leaks or thesia care unit (PACU) are acceptable alterna-
strictures, bacterial infections, incisional hernias, tives. For example, in New York State the
pleural effusions, wound infections, and intra-­ Department of Health requires that the “live adult
abdominal abscesses [6]. In particular, the fol- liver donors should receive intensive care”.
lowing risk factors had a significant association
with biliary complications such as [3],
Routine Postoperative
(a) Donor age: Older donors are more likely to Management
suffer from complications such as delayed
liver regeneration and poor long-term sur-  urveillance for Deep Vein
S
vival of the graft. Delayed regenerative Thrombosis
capacity can predispose to risk of liver fail-
ure [20]. The risk of thromboembolic complications is
(b) Surgical technique: Right and extended right increased after major surgery and prophylaxis is
lobe resection has been associated with more indicated [23]. Early mobilization is the key to
frequent complications [3]. prevent these complications. Loss of significant

(c) Intraoperative blood transfusion: There is liver volume in the donor can lead to decreased
higher incidence of biliary complications and synthetic capacity and may cause a subclinical or
37  Postoperative Care of Living Donor for Liver Transplant 489

insidious coagulopathy but also hypercoagulabil- and call for larger studies to determine which test
ity due to decreased production of anticoagulant is best suited for directing management of
factors (protein C, protein S, or antithrombin). prophylaxis.
Importantly, conventional coagulation assays Prior to starting anticoagulation, the potential
measure procoagulants only [24] and may differ for coagulopathy should be taken into consider-
from a viscoelastic test of coagulation such as ation. Deep vein thrombosis (DVT) prophylaxis
thromboelastography or rotational thromboelas- is usually accomplished pharmacologically with
tometry. These measure clot formation resulting either subcutaneous low molecular weight
from all factors including natural anticoagulant (LMWH) [8, 22, 28] or unfractionated heparin.
factors, platelets and fibrinogen. In particular, Unfractionated heparin allows the greatest flexi-
apparently hypocoagulable patients with elevated bility with regards to the removal of epidural
INR/aPTT following liver donation or others catheters, if these are used for postoperative pain
with known chronic liver failure will have normal management [29]. LMWH offers the ease of
or even hypercoagulable results on ROTEM/TEG once or twice daily administration. Non-­
and other assays of thrombin generation pharmacological measures include sequential
(Fig. 37.3) [24–27]. Rotational thromboelastom- compression device (SCD) or thromboembolic
etry (ROTEM) testing of fibrinogen function stockings.
(FIBTEM) have detected elevated maximum clot
firmness (MCF) that correlates with fibrinogen
activity, a known acute phase reactant and poten- Antibiotic Prophylaxis
tial contributor to hypercoagulability [27]. Most
studies of ROTEM/TEG have been small, caution Single dose broad-spectrum antibiotic prophy-
against use of a single assay for decision-making laxis is given prior to surgical skin incision.

1 FIBTEM 2 extem
mm ST: 10:34:51 mm ST: 09:59:20
RT: 00:34:44 RT: 01:10:14
CT: 53 s CT: 68 s
60 60
[0043 --0082]
40 CFT: 27 s 40 CFT: 56 s
[0048 --0127]
20 20
α: 84° α: 79°
[0065 --0080]
A10: 58 mm A10: 69 mm
20 20

40 A20: 61 mm 40 A20: 73 mm
[0007--0024] [0050--0070]
60 MCF: 61 mm 60 MCF: 73 mm
[0007--0024] [0052--0070]
ML: * 2% ML: * 11 %
10 20 30 40 50 min 10 20 30 40 50 min
3 intem 4 FIBTEM
mm ST: 10:00:17 mm ST: 10:01:20
RT: 01:09:18 RT: 01:08:16
CT: 143 s CT: 52 s
60 60
[0122 -- 0208]
40 CFT: 27 s 40 CFT: 29 s
[0045 -- 0110]
20 20
α: 85° α: 84°
[0070 -- 0081]
A10: 81mm A10: 61 mm
20 20

40 A20: 82 mm 40 A20: 63 mm
[0051--0072] [0007--0024]
60
60 MCF: 82 mm MCF: 64 mm
[0051--0072] [0007--0024]
ML: * 9% ML: * 12 %

Fig. 37.3  Preoperative baseline rotational thromboelastometry of a cirrhotic patient with synthetic liver dysfunction.
Elevated A20 and maximum clot firmness (MCF) values suggest hypercoaguability
490 S. Ewing et al.

Routine antibiotic prophylaxis is not generally blood count (CBC), electrolyte and metabolic
required postoperatively unless specifically indi- panel, coagulation profile, lactate levels and liver
cated. Although infrequent, donors may develop function tests (LFTs). These laboratory studies
wound infections, intra-abdominal abscess and are recommended every day for first three post-
pneumonia. Prevention of these complications is operative days (PODs), followed by testing on
multimodal including strict barrier precautions, alternate days until discharge [29]. If discharge
avoidance of perioperative hypothermia; pro- occurs within 7 days of surgery, these tests can be
longed, prophylactic antibiotics will not prevent performed on an outpatient basis if necessary.
these complications.

Radiological Evaluation
Intravenous Fluids and Nutrition
A postoperative Chest X-ray is usually obtained
Intravenous (IV) fluid is titrated to urine output to check the position of the central venous cath-
and balanced against oral intake. The goal is to eter (CVC) and to rule out CVC-related compli-
maintain adequate tissue perfusion and oxygen- cations such as misplacement or pneumothorax,
ation. To avoid remnant liver congestion, IV flu- if a CVC was placed intraoperatively. Ultrasound
ids are judiciously used to avoid high central or duplex imaging of the remnant liver is not rou-
venous pressures. Generally crystalloids are tinely performed [3] unless abnormal LFTs or the
used. If there is evidence of hypovolemia or clinical course raise concerns for vascular com-
blood loss not necessitating blood transfusion, plications, such as portal vein thrombosis (PVT)
colloids may be used but there is no evidence of and hepatic artery thrombosis (HAT).
superiority of colloids in this situation. Blood or In the normal course of events, the arterial
blood products are not generally required unless line, CVC and nasogastric tube (NG) tube (if
there is hemodynamic instability due to bleeding. placed) can be removed on the first POD. If an
Most of the donors are young and all of them are epidural catheter is used for postoperative pain
healthy; they should therefore tolerate some management, the urinary catheter is removed
degree of anemia (Hb >7–7.5 g/dL) well as long coincident with discontinuation of the epidural
as they are not hypovolemic. IV fluids are contin- analgesia. If oxygenation is adequate on trials of
ued until full enteral feeding is commenced. Fan room air, oxygen supplementation can also be
et al. reported the use of parenteral nutritional discontinued. The patient is expected to be out of
support in the immediate postoperative period to bed on POD 1. On POD 2, clear liquids are often
enhance liver remnant regeneration, although this started. If tolerated and bowel sounds are normal,
is not a common practice in most centers [30]. a soft diet can be then introduced on POD 3.

Laboratory Testing Postoperative Analgesia

Major hepatic resection causes a measurable Adequate analgesia along with early mobiliza-
decrease in coagulation factors due to transient tion, chest physiotherapy and incentive spirome-
synthetic insufficiency [31]. It is not uncommon try is vital to prevent respiratory complication in
to see evidence of hepatocyte injury (increased the postoperative period. The analgesic plan
transaminases) and a decrement in liver synthetic should be tailored to the surgical approach (open,
functions (increased bilirubin and an abnormal ­laparoscopic, etc). Epidural analgesia, abdominal
coagulation profile) that begins to improve as wall blocks or wound catheter placement are safe
liver regeneration occurs. Complete liver regen- and effective options that have been evaluated as
eration usually occurs between 1 week and 2 part of open hepatectomy enhanced recovery pro-
months after resection [32]. Postoperative labora- tocols [33–36]. In a nationwide analysis of over
tory surveillance usually includes a complete 50,000 patients receiving hepatopancreatic proce-
37  Postoperative Care of Living Donor for Liver Transplant 491

dures, the odds of sepsis, respiratory failure, post- underreported due to lack of a global database or
operative pneumonia, and in-hospital mortality registry and possibly a reluctance to report com-
were lower with epidural use [37]. However, after plications. Based on the current literature, the
major hepatic resections, the potential for a post- complication rate of donors from single center
operative coagulopathy exists and should warrant analysis varies widely from as low as 9% to as
frequent neurological exams in patients with epi- high as 67% [43–52]. The European Liver
dural catheters until the coagulation profile (pro- Transplant Registry has reported a 0.5% mortal-
thrombin time (PT), partial thromboplastin time ity and 21% postoperative morbidity [53]. The
(PTT), international normalized ratio (INR), and Japanese Liver Transplant Society reported no
platelet count) is normal. The coagulation profile mortality and 12% postoperative morbidity [54].
should be normal before the removal of epidural The variability in morbidity and mortality is
catheter and this may delay mobilization and/or likely due to the lack of a standardized system for
discharge. Therefore, many clinicians will preop- classifying complications. In an effort to over-
eratively place a single dose of intrathecal mor- come this classification shortfall, Clavien’s clas-
phine that will provide pain relief for 18–24 h sification of complications of surgery, which has
after surgery instead of epidural analgesia [38, been used for general surgery, can also been
39]. If an epidural is used, analgesia can be transi- applied to transplantation (Table 37.1) [55, 56].
tioned to parenteral and oral analgesics before Using this classification, the NIH funded A2ALL
removal on POD 3 or 4. Intravenous patient-con-
trolled analgesia (IVPCA) is also an acceptable
Table 37.1  Modified Clavien system for classification of
alternative. Tranversus abdominis plane blocks negative outcomes in general surgery and solid organ
have been evaluated as part of multimodal analge- transplantation
sia plans following laparoscopic liver resection
Grade Any alteration from the ideal postoperative
and other upper abdominal procedures [40–42]. 1 course, with complete recovery or
These adjunctive techniques may be useful to complications which can be easily controlled
reduce IVPCA, if neuraxial techniques are contra- and which fulfills the following general
characteristics:
indicated or for patients undergoing laparoscopic
Not life threatening:
donation. More details about post- operative pain not requiring use of drugs other than
management can be found elsewhere in this book. immunosuppressant; analgesics; antipyretics;
antiinflammatory agents; antiemetics; drugs
required for urinary retention or lower urinary
tract infection, arterial hypertension,
Long-Term Follow Up hyperlipidemia, or transient hyperglycemia;
requiring only therapeutic procedures that can
Based on retrospective studies, most centers fol- be performed at the bedside;
Postoperative bleeding requiring ≤3 units of
low patients up at 1, 4, and 12 months after dona-
blood transfusion; and
tion [22]. International guidelines recommend Never associated with a prolongation of
regular clinical monitoring and follow-up for 2 intensive care unit stay or total hospital stay
years [8]. This includes clinical evaluation, liver to more than twice the median stay for the
procedure in the population of the study.
functions and platelet count testing for 1 year,
Grade Any complication that is potentially life
and radiological evaluation if needed. All donors 2 threatening or results in intensive care unit
should have lifetime annual primary care exami- stay >5 days, hospital stay >4 weeks for the
nations for health maintenance. recipient, but which does not result in
residual disability or persistent disease
Grade Any complication with residual or lasting
3 functional disability or development of
Postoperative Complications malignant disease
Grade Complications that lead to retransplantation
Postoperative complications can be broadly clas- 4 (grade 4a) or death (grade 4b)
sified as surgical and medical. Unfortunately, Adapted from Ghobrial et al. with permission,
complications in living donors are probably Gastroenentorology 2008; 135:468–76
492 S. Ewing et al.

Table 37.2  Donor mortality rates [3, 6, 39] Surgical Technique and Bile Leakage
European liver transplant registry 0.5%
Taku Lida et al. (Kyoto group) 0.08% Because most bile leaks occurs from bile duct
Survey of liver transplantation in living 0.2% stumps, secondary to ischemia, it has been recom-
donors in the United States [41] mended to minimize the use of electrocautery
around the bile ducts during liver resection. Some
authors suggest that a biliary decompression tube
Cohort Study reported an overall complication is effective in reducing bile leakage [3] but this is
rate of 38% in nine transplant centers [6]. Using not routinely placed in many centers. Bile leak
the same system, Yi et al. reported a complication from the caudate lobe is problematic and can be
rate as high as 78.3% whereas Patel et al. and refractory to conservative treatment. Careful atten-
more recently Candido et al. reported a lower tion should be paid to the bile ducts in these lobes
overall complication rate of 29.1% and 28%, and may require continuous suture or ligation [57].
respectively [11, 21, 57]. The reason for these
differences, despite of the use of the Clavien sys-
tem, could be due to transplant center surgical  igns, Symptoms, and Management
S
experience as well as the use of retrospective data of Bile Leakage
[6]. The A2ALL study concluded that although
most living donors for liver transplants had, in When bile leakage occurs, patients can present
general, low-grade severity complications, a sub- with fever, abdominal or shoulder pain, and bil-
stantial number had severe or life-threatening ious drainage from drains and the incision. Bile
complications. Quantification of complications leakage can be diagnosed by physical exam,
would improve postoperative care of these ultrasound, CT scan, diagnostic paracentesis, or
patients and improve the informed consent pro- endoscopic retrograde cholangiopancreatogra-
cess [6] and may lead to improved care and long-­ phy (ERCP). Bile leakage may resolve after con-
term outcome of LDLT (Table 37.2). servative treatment or may require interventional
therapy such as continuous drainage, endoscopic
retrograde bile drainage, percutaneous drainage,
Bile Leakage or surgical repair [3].

Bile leak is one of the most common complica-


tions in living donors and has been called the Infection
“achilles heel” of LDLT [15]. Its incidence ranges
from 4.7 to 10.6% [3, 6]. The right donor hepa- Despite careful surveillance and meticulous oper-
tectomy tends to have higher incidence of bile ative techniques the incidence of postoperative
leakage because the biliary system in the right infection rate ranges from 5.0 to 12.5% [3, 6] .
lobe has more anatomical variation than the left These include pneumonia, urinary tract infection,
lobe. Biliary complications are due to bile leak cellulitis, sepsis, Clostridium difficile enterocoli-
from stumps and due to biliary ischemia. tis, wound infection, and ­intra-­abdominal abscess
Unrecognized anatomical variations can predis- [3, 57]. Infectious complications are often sec-
pose to these complications. Preoperative CT ondary to other complication such as respiratory
cholangiography or intraoperative cholangio- failure, delayed graft function, or bile leak.
grams are helpful in assessing the anatomy of
bile ducts. After reporting a decreased incidence
of biliary complication with left lobe grafts, Hemorrhage
Taketomi et al. recommended that the left lobe
should be considered first in choosing segments Yi et al. reported 3.8% of donors experienced tran-
for A2ALL [58]. sient bleeding from surgical drains that improved
37  Postoperative Care of Living Donor for Liver Transplant 493

without blood transfusion [11]. Patel et al. reported complications of the donors. Although the over-
one case out of 433 (0.23%) that returned to the all risk of PVT is low, it can be life threatening
operating room for hemorrhage [57]. The A2ALL for a healthy donor who may then end up needing
study did not report any bleeding complications. a liver transplant. There have been two case
The overall risks of bleeding complications reports of PVT and inferior vena cava thrombosis
reported in these studies were very low. reported by the A2ALL study [3, 6].

Pulmonary Embolism Liver Failure Post-Donation

The risk of thromboembolic complications fol- Postoperative liver failure in the donor is often
lowing LDLT has been highlighted in many due to inadequate remnant liver volume. To avoid
papers [59, 60]. The incidence of pulmonary this catastrophic complication, preoperative eval-
embolism has been reported to be between 0.2 uation is critical. Three-dimensional computed
and 0.8% [3, 6, 54]. Hypercoagulability in living tomography (3D–CT) or magnetic resonance
donors is likely the result of an increase in throm- imaging (MRI) volumetry is used preoperatively
bin–antithrombin complexes and P selectin fol- to estimate liver graft volume as well as the donor
lowing surgery. This has also been observed in remnant liver volume (Fig. 37.2). Donor remnant
patients undergoing hepatic resection for benign liver volume needs to be at least 30–35% of total
tumors [61]. As mentioned previously, hyperco- preoperative liver volumes. Two unusual cases of
agulability may be missed by convention coagu- donor hepatic failure have been attributed to non-
lation assays (INR/aPTT) as they only detect the alcoholic steatohepatitis in one and to
activity of procoagulants in patients with liver Berandinell-Seip syndrome (lipodystrophy syn-
dysfunction [24]. Viscoelastic assays such as drome) in the other [53, 63, 64]. These complica-
ROTEM and TEG have gained increasing sup- tions were due to unrecognized pathology in the
port for global assessment of coagulation follow- donor liver. To avoid these problems, some cen-
ing liver donation [25–27]. Preoperatively, ters routinely perform a liver biopsy as a part of
Bustelos et al. screened 188 potential donors for routine preoperative work-up for donors, particu-
bleeding or procoagulant and found that about larly in the presence of biochemical, serological,
20% of them have at least one abnormality. The or imaging evidence of liver disease [19]. Risk
donors in this study were screened for factor V factors for posthepatectomy liver failure include
Leiden, prothrombin mutation, deficiencies of advanced donor age, diabetes and postoperative
protein C, S, and anti- thrombin [62]. They rec- thrombocytopenia [65–67]. Supportive care is
ommend preoperative screening of all potential the mainstay treatment and may generally follow
donors for bleeding and hypercoagulable states. recommendations of AASLD for acute liver fail-
Taking into account the potential risk to donor ure including monitoring for changes in coagula-
and recipient, screening should be considered tion, bilirubin, encephalopathy, infection,
even though the cost-effectiveness of performing hemodynamic failure, renal failure and metabolic
these tests is yet to be determined. DVT prophy- disorders with treatment as early as possible [68].
laxis and early ambulation are the cornerstones in Hepatotoxic and nephrotoxic medications should
preventing rare but life-threatening thromboem- be avoided. Failure of supportive care may ulti-
bolic complications. mately require listing for liver transplant.

Vascular Complications Donor Mortality

The incidences of PVT, HAT, and reversal of por- Living-related liver donation can be performed
tal venous flow are rare but possibly catastrophic with relatively low risk of significant p­ erioperative
494 S. Ewing et al.

morbidity and mortality [57]. The overall mortal- References


ity based on the current literature is between 0.08
and 0.5% (Table 37.2) [8]. Trotter et al., in a com- 1. http://optn.transplant.hrsa.gov/.
2. Trotter JF, Adam R, Lo CM, Kenison
prehensive review of the medical and lay litera- J. Documented deaths of hepatic lobe donors for
ture, reported 19 deaths of live liver donors of living donor liver transplantation. Liver Transpl.
which 13 were related to surgery [2]. The causes 2006;12(10):1485–8.
of death were sepsis (5 cases), liver failure (2 3. Iida T, Ogura Y, Oike F, Hatano E, Kaido T,
Egawa H, et al. Surgery-related morbidity in liv-
cases), myocardial infarction, cerebral hemor- ing donors for liver transplantation. Transplantation.
rhage, pulmonary embolism, peptic ulcer dis- 2010;89(10):1276–82.
ease, and unknown cause (2 cases). As experience 4. Hwang S, Lee SG, Lee YJ, Sung KB, Park KM,
in performing these complex procedures Kim KH, et al. Lessons learned from 1,000 liv-
ing donor liver transplantations in a single center:
increases, improved outcomes and decreased how to make living donations safe. Liver Transpl.
mortality and morbidity should be seen. In the 2006;12(6):920–7.
event of donor death, hospitals and teams 5. Levy GA, Selzner N, Grant DR. Fostering liver liv-
ing donor liver transplantation. Curr Opin Organ
involved in LDLT should have basic plan for cri-
Transplant. 2016;21(2):224–30.
sis management for appropriate public response 6. Ghobrial RM, Freise CE, Trotter JF, Tong L, Ojo
as no institution can eliminate risk of donor mor- AO, Fair JH, et al. Donor morbidity after living
tality [8]. Any donor death or major morbidity is donation for liver transplantation. Gastroenterology.
2008;135(2):468–76.
a catastrophe and should prompt a thorough
7. Broering DC, Wilms C, Bok P, Fischer L, Mueller L,
investigation of the cause and potential ways to Hillert C, et al. Evolution of donor morbidity in living
avoid this in the future. related liver transplantation: a single-center analysis
of 165 cases. Ann Surg. 2004;240(6):1013–24. dis-
cussions 24–6
Conclusion 8. Miller CM, Durand F, Heimbach JK, Kim-Schluger
LDLT decreases wait lists for transplants, L, Lee SG, Lerut J, et al. The International Liver
Transplant Society Guideline on living liver donation.
and is a feasible and potentially lifesaving
Transplantation. 2016;100(6):1238–43.
alternative for select patients. LDLT has 9. Yonemura Y, Taketomi A, Soejima Y, Yoshizumi
become the most effective alternative to T, Uchiyama H, Gion T, et al. Validity of preop-
DDLT. As this technique continues to evolve, erative volumetric analysis of congestion volume
in living donor liver transplantation using three-­
steps to improve perioperative outcomes are
dimensional computed tomography. Liver Transpl.
required including strategies to categorize 2005;11(12):1556–62.
and standardize the definitions of surgical 10. Marsh JW, Gray E, Ness R, Starzl TE. Complications
complications and report them in centralized of right lobe living donor liver transplantation. J
Hepatol. 2009;51(4):715–24.
registries. The development of a valid and
11. Yi NJ, Suh KS, Cho JY, Lee HW, Cho EH, Yang
useful prognostic scoring system to improve SH, et al. Three-quarters of right liver donors expe-
donor safety will undoubtedly improve peri- rienced postoperative complications. Liver Transpl.
operative outcomes and facilitate the selec- 2007;13(6):797–806.
12. Braun HJ, Dodge JL, Roll GR, Freise CE, Ascher NL,
tion of suitable donors. Minimizing the risk
Roberts JP. Impact of graft selection on donor and
for the donor will continue to be of para- recipient outcomes after living donor liver transplan-
mount importance as the use of LDLT tation. Transplantation. 2016;100(6):1244–50.
expands. Because donor undergoes major 13. Park JI, Kim KH, Lee SG. Laparoscopic living

donor hepatectomy: a review of current status. J
surgery with no physiologic indication, usu-
Hepatobiliary Pancreat Sci. 2015;22(11):779–88.
ally out of an altruistic aim to help others, 14. Wakabayashi G, Cherqui D, Geller DA, Buell JF,
they should be treated with the best possible Kaneko H, Han HS, et al. Recommendations for
care and utmost respect. laparoscopic liver resection: a report from the second
international consensus conference held in Morioka.
Ann Surg. 2015;261(4):619–29.
Acknowledgments  We would like to thank Dr. Patrick 15. Brown RS Jr. Live donors in liver transplantation.
M. Mcquillan M.D. for helping review the Gastroenterology. 2008;134(6):1802–13.
manuscript.
37  Postoperative Care of Living Donor for Liver Transplant 495

16. Shiffman ML, Brown RS Jr, Olthoff KM, Everson G, 31. Borromeo CJ, Stix MS, Lally A, Pomfret EA. Epidural
Miller C, Siegler M, et al. Living donor liver trans- catheter and increased prothrombin time after right
plantation: summary of a conference at the National lobe hepatectomy for living donor transplantation.
Institutes of Health. Liver Transpl. 2002;8(2):174–88. Anesth Analg. 2000;91(5):1139–41.
17. https://optn.transplant.hrsa.gov/media/1200/optn_ 32. Marcos A, Fisher RA, Ham JM, Shiffman ML, Sanyal
policies.pdf AJ, Luketic VA, et al. Liver regeneration and function
18. Brown RS Jr, Higgins R, Pruett TL. The evolution in donor and recipient after right lobe adult to adult
and direction of OPTN oversight of live organ dona- living donor liver transplantation. Transplantation.
tion and transplantation in the United States. Am J 2000;69(7):1375–9.
Transplant. 2009;9(1):31–4. 33. Khan J, Katz J, Montbriand J, Ladak S, McCluskey
19. Manas D, Burnapp L, Andrews PA. Summary of the S, Srinivas C, et al. Surgically placed abdominal
British Transplantation Society UK Guidelines for wall catheters on postoperative analgesia and out-
living donor liver transplantation. Transplantation. comes after living liver donation. Liver Transpl.
2016;100(6):1184–90. 2015;21(4):478–86.
20. Olthoff KM. Hepatic regeneration in living donor
34. Ntinas A, Kardassis D, Konstantinopoulos I, Kottos
liver transplantation. Liver Transpl. 2003;9(10 Suppl P, Manias A, Kyritsi M, et al. Duration of the tho-
2):S35–41. racic epidural catheter in a fast-track recovery pro-
21. Candido HL, da Fonseca EA, Feier FH, Pugliese R, tocol may decrease the length of stay after a major
Benavides MA, Silva ED, et al. Risk factors associ- hepatectomy: a case control study. Int J Surg.
ated with increased morbidity in living liver donation. 2013;11(9):882–5.
J Transp Secur. 2015;2015:949674. 35. Revie EJ, McKeown DW, Wilson JA, Garden OJ,
22. Gruttadauria S, Marsh JW, Vizzini GB, di Francesco Wigmore SJ. Randomized clinical trial of local infil-
F, Luca A, Volpes R, et al. Analysis of surgical and tration plus patient-controlled opiate analgesia vs.
perioperative complications in seventy-five right epidural analgesia following liver resection surgery.
hepatectomies for living donor liver transplantation. HPB (Oxford). 2012;14(9):611–8.
World J Gastroenterol. 2008;14(20):3159–64. 36. Savikko J, Ilmakunnas M, Makisalo H, Nordin A,
23. Risk of and prophylaxis for venous thrombo-
Isoniemi H. Enhanced recovery protocol after liver
embolism in hospital patients. Thromboembolic resection. Br J Surg. 2015;102(12):1526–32.
Risk Factors (THRIFT) Consensus Group. BMJ. 37. Amini N, Kim Y, Hyder O, Spolverato G, Wu CL,
1992;305(6853):567–74. Page AJ, et al. A nationwide analysis of the use
24. Tripodi A, Salerno F, Chantarangkul V, Clerici M, and outcomes of perioperative epidural analgesia in
Cazzaniga M, Primignani M, et al. Evidence of nor- patients undergoing hepatic and pancreatic surgery.
mal thrombin generation in cirrhosis despite abnor- Am J Surg. 2015;210(3):483–91.
mal conventional coagulation tests. Hepatology. 38. Aydogan MS, Bicakcioglu M, Sayan H, Durmus M,
2005;41(3):553–8. Yilmaz S. Effects of two different techniques of post-
25. Mohammed M, Fayed N, Hassanen A, Ahmed F,
operative analgesia management in liver transplant
Mourad W, El Sheikh M, et al. Rotational thrombo- donors: a prospective, randomized, double-blind
elastometry and standard coagulation tests for live study. Transplant Proc. 2015;47(4):1204–6.
liver donors. Clin Transpl. 2013;27(2):E101–8. 39. De Pietri L, Siniscalchi A, Reggiani A, Masetti M,
26. Cerutti E, Stratta C, Romagnoli R, Schellino MM, Begliomini B, Gazzi M, et al. The use of intrathe-
Skurzak S, Rizzetto M, et al. Thromboelastogram cal morphine for postoperative pain relief after liver
monitoring in the perioperative period of hepatec- resection: a comparison with epidural analgesia.
tomy for adult living liver donation. Liver Transpl. Anesth Analg. 2006;102(4):1157–63.
2004;10(2):289–94. 40. Niraj G, Kelkar A, Jeyapalan I, Graff-Baker P,

27. Gouvea G, Diaz R, Auler L, Toledo R, Soluri A, Williams O, Darbar A, et al. Comparison of anal-
Haack AG, et al. Perioperative coagulation profile in gesic efficacy of subcostal transversus abdominis
living liver donors as assessed by rotational thrombo- plane blocks with epidural analgesia following upper
elastometry. Liver Transpl. 2010;16(3):387–92. abdominal surgery. Anaesthesia. 2011;66(6):465–71.
28. Mismetti P, Laporte S, Darmon JY, Buchmuller A, 41. Serag Eldin M, Mahmoud F, El Hassan R, Abdel
Decousus H. Meta-analysis of low molecular weight Raouf M, Afifi MH, Yassen K, et al. Intravenous
heparin in the prevention of venous thromboembolism patient-controlled fentanyl with and without transver-
in general surgery. Br J Surg. 2001;88(7):913–30. sus abdominis plane block in cirrhotic patients post
29. Itamoto T, Emoto K, Mitsuta H, Fukuda S, Ohdan H, liver resection. Local Reg Anesth. 2014;7:27–37.
Tashiro H, et al. Safety of donor right hepatectomy 42. Abdelsalam K, Mohamdin OW. Ultrasound-guided
for adult-to-adult living donor liver transplantation. rectus sheath and transversus abdominis plane blocks
Transpl Int. 2006;19(3):177–83. for perioperative analgesia in upper abdominal sur-
30. Fan ST, Lo CM, Liu CL, Yong BH, Chan JK,
gery: a randomized controlled study. Saudi J Anaesth.
Ng IO. Safety of donors in live donor liver trans- 2016;10(1):25–8.
plantation using right lobe grafts. Arch Surg. 43. Trotter JF, Talamantes M, McClure M, Wachs M, Bak
2000;135(3):336–40. T, Trouillot T, et al. Right hepatic lobe donation for
496 S. Ewing et al.

living donor liver transplantation: impact on donor 57. Patel S, Orloff M, Tsoulfas G, Kashyap R, Jain A,
quality of life. Liver Transpl. 2001;7(6):485–93. Bozorgzadeh A, et al. Living-donor liver transplanta-
44. Renz JF, Busuttil RW. Adult-to-adult living-donor
tion in the United States: identifying donors at risk
liver transplantation: a critical analysis. Semin Liver for perioperative complications. Am J Transplant.
Dis. 2000;20(4):411–24. 2007;7(10):2344–9.
45. Pomfret EA, Pomposelli JJ, Lewis WD, Gordon FD, 58. Taketomi A, Kayashima H, Soejima Y, Yoshizumi
Burns DL, Lally A, et al. Live donor adult liver trans- T, Uchiyama H, Ikegami T, et al. Donor risk in
plantation using right lobe grafts: donor evaluation and adult-to-­adult living donor liver transplanta-
surgical outcome. Arch Surg. 2001;136(4):425–33. tion: impact of left lobe graft. Transplantation.
46. Miller CM, Gondolesi GE, Florman S, Matsumoto C, 2009;87(3):445–50.
Munoz L, Yoshizumi T, et al. One hundred nine living 59. Durand F, Ettorre GM, Douard R, Denninger

donor liver transplants in adults and children: a single-­ MH, Kianmanesh A, Sommacale D, et al. Donor
center experience. Ann Surg. 2001;234(3):301–11. safety in living related liver transplantation:
discussion 11–2 underestimation of the risks for deep vein throm-
47. Ibrahim S, Chen CL, Lin CC, Yang CH, Wang CC, bosis and pulmonary embolism. Liver Transpl.
Wang SH, et al. Intraoperative blood loss is a risk 2002;8(2):118–20.
factor for complications in donors after living donor 60. Wakeno-Takahashi M, Nakao S, Ikeda S, Kaibori

hepatectomy. Liver Transpl. 2006;12(6):950–7. M, Yamamoto H, Yanagida H, et al. A case of sus-
48. Grewal HP, Shokouh-Amiri MH, Vera S, Stratta R, pected severe pulmonary embolism in a living-­
Bagous W, Gaber AO. Surgical technique for right related liver transplantation donor. J Clin Anesth.
lobe adult living donor liver transplantation with- 2005;17(3):218–20.
out venovenous bypass or portocaval shunting and 61. Bezeaud A, Denninger MH, Dondero F, Saada V,
with duct-to-duct biliary reconstruction. Ann Surg. Venisse L, Huisse MG, et al. Hypercoagulability
2001;233(4):502–8. after partial liver resection. Thromb Haemost.
49. Ghobrial RM, Saab S, Lassman C, Lu DS, Raman 2007;98(6):1252–6.
S, Limanond P, et al. Donor and recipient outcomes 62. Bustelos R, Ayala R, Martinez J, Martin MA, Toledo
in right lobe adult living donor liver transplantation. T, Grande S, et al. Living donor liver transplanta-
Liver Transpl. 2002;8(10):901–9. tion: usefulness of hemostatic and prothrombotic
50. Marcos A, Fisher RA, Ham JM, Shiffman ML,
screening in potential donors. Transplant Proc.
Sanyal AJ, Luketic VA, et al. Right lobe liv- 2009;41(9):3791–5.
ing donor liver transplantation. Transplantation. 63. Akabayashi A, Slingsby BT, Fujita M. The first donor
1999;68(6):798–803. death after living-related liver transplantation in
51. Beavers KL, Sandler RS, Fair JH, Johnson MW,
Japan. Transplantation. 2004;77(4):634.
Shrestha R. The living donor experience: donor health 64. Malago M, Testa G, Frilling A, Nadalin S, Valentin-­
assessment and outcomes after living donor liver Gamazo C, Paul A, et al. Right living donor liver
transplantation. Liver Transpl. 2001;7(11):943–7. transplantation: an option for adult patients: single
52. Beavers KL, Sandler RS, Shrestha R. Donor morbid- institution experience with 74 patients. Ann Surg.
ity associated with right lobectomy for living donor 2003;238(6):853–62. discussion 62–3
liver transplantation to adult recipients: a systematic 65. Margonis GA, Amini N, Buettner S, Besharati S,

review. Liver Transpl. 2002;8(2):110–7. Kim Y, Sobhani F, et al. Impact of early postoperative
53. Adam R, McMaster P, O’Grady JG, Castaing
platelet count on volumetric liver gain and periopera-
D, Klempnauer JL, Jamieson N, et al. Evolution tive outcomes after major liver resection. Br J Surg.
of liver transplantation in Europe: report of the 2016;103(7):899–907.
European liver transplant registry. Liver Transpl. 66. Han S, Park HW, Song JH, Gwak MS, Lee WJ,

2003;9(12):1231–43. Kim G, et al. Association between intraoperative
54. Umeshita K, Fujiwara K, Kiyosawa K, Makuuchi
platelet transfusion and early graft r­egeneration
M, Satomi S, Sugimachi K, et al. Operative mor- in living donor liver transplantation. Ann Surg.
bidity of living liver donors in Japan. Lancet. 2016;264(6):1065–72.
2003;362(9385):687–90. 67. Tanemura A, Mizuno S, Wada H, Yamada T, Nobori
55. Clavien PA, Camargo CA Jr, Croxford R, Langer T, Isaji S. Donor age affects liver regeneration during
B, Levy GA, Greig PD. Definition and classifica- early period in the graft liver and late period in the
tion of negative outcomes in solid organ transplanta- remnant liver after living donor liver transplantation.
tion. Application in liver transplantation. Ann Surg. World J Surg. 2012;36(5):1102–11.
1994;220(2):109–20. 68. Polson J, Lee WM, American Association for the
56. Clavien PA, Sanabria JR, Strasberg SM. Proposed Study of Liver Disease. AASLD position paper:
classification of complications of surgery with the management of acute liver failure. Hepatology.
examples of utility in cholecystectomy. Surgery. 2005;41(5):1179–97.
1992;111(5):518–26.
Liver Surgery: Early
Complications—Liver Failure, Bile 38
Leak and Sepsis

Albert C. Y. Chan and Sheung Tat Fan

Keywords is in the range of 20–30%. Liver failure, bile leak


Indocyanine green (ICG) clearance · Liver func- and sepsis are serious complications that could
tion reserve · Hepatectomy · Portal vein emboli- lead to a fatal outcome. In this chapter, we will
zation · Liver failure · Biliary complications present our approach for prevention, diagnosis
and management of these complications.
With adequate remnant liver volume and
Introduction absence of co-morbid illness, liver failure is the
most common cause of mortality after major hep-
Liver resection remains to be the curative treat- atectomy. Early signs of liver failure include
ment of choice for both malignant and benign hypotension, lactate acidosis, respiratory depres-
liver tumors. With advances in hepatic surgery sion, oliguria, jaundice, hepatic encephalopathy
and operative technique, liver resection has and coagulopathy. Patients may improve in the
evolved from a rough and hasty procedure to a initial postoperative period but deteriorate after-
fine and delicate operation. Such surgical wards with an onset of drowsiness, jaundice,
advances have resulted in a dramatic reduction of flapping tremor, ascites, pleural effusion, and oli-
operative mortality, from over 50% in early series, guria. Early recognition of these symptoms
to less than 10% in recent decades. Targeting a allows treatment to be promptly implemented
‘zero’ mortality has even become a realistic goal before the patient becomes unsalvageable.
to achieve. However, the postoperative complica- Extensive resection in patients with pre-existing
tions rate remains largely unchanged over the liver dysfunction is often the cause but many
years despite reduction in operative mortality and patients with borderline liver function can still
undergo major hepatectomy and make a full
recovery in an experienced center. A comprehen-
sive preoperative assessment and optimal periop-
erative care are critical to select the appropriate
A. C. Y. Chan, MBBS, FRCS patients for partial hepatectomy.
Department of Surgery, The University of Hong
Kong, Queen Mary Hospital,
Hong Kong, People’s Republic of China
e-mail: acchan@hku.hk Patient Selection
S. T. Fan, MD, PhD, DSc (*)
Liver Surgery Centre, Hong Kong Sanatorium and The presence of co-morbid illness such as car-
Hospital, Hong Kong, People’s Republic of China diovascular disease [1] and renal impairment [2]
e-mail: stfan@hksh.com

© Springer International Publishing AG, part of Springer Nature 2018 497


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_38
498 A. C. Y. Chan and S. T. Fan

increases the risk of hepatectomy. Biological this problem, the aspartate aminotransferase/
age is not a contraindication for hepatectomy platelet count ratio index, which is a biochemi-
but surgery in elderly patients can be challeng- cal surrogate for histological fibrogenesis in cir-
ing because exposure of the liver is limited by rhosis, may be alternatively used [4]. Another
rigidity of the rib cage and the possible diffi- approach that has been recently adopted in some
culty in lowering the central venous pressure. centers is the use of the model for end-stage
Severe co-­ morbid illness such as congestive liver disease (MELD) score, that includes the
heart failure and chronic renal failure, however, international normalized ratio, serum creatinine
should be considered as contraindication for and serum bilirubin. However, given that most
major hepatectomy. Patients with chronic renal of the patients selected for major hepatectomy
failure may be suitable for hepatectomy pro- have normal renal function and international
vided that perioperative hemodialysis or con- normalized ratio, the resultant MELD score can
tinuous renal replacement therapy is available. be expected to be low and the range of MELD
The American Society of Anesthesiologist score among most of the patients would be lim-
(ASA) score is frequently used for postopera- ited, rendering it impractical for clinical
tive risk assessment and is reliable in predicting application.
morbidity after hepatectomy [3]. With refined Indocyanine green (ICG) clearance test is a
liver function assessment in recent years, pres- sophisticated quantitative test for functional
ence of co-morbid illness has become a more assessment of the hepatocytes. ICG is an innocu-
important factor in predicting postoperative out- ous dye with rare allergic reactions in some
come of hepatectomy. patients as the only adverse effect. After intrave-
nous administration, it binds to albumin and
β-lipoprotein and is exclusively metabolized by
 reoperative Assessment of Liver
P the liver and excreted unchanged in bile without
Function Reserve any entero-hepatic circulation. Its elimination is
affected by liver blood flow and reflects intrahe-
Liver function could be assessed readily by lab- patic portovenous shunting and sinusoidal capil-
oratory blood tests. Serum bilirubin and albu- larization. ICG levels can be measured either by
min are reflections of excretory and synthetic blood draw or non-invasively using a probe, simi-
functions. White cell counts and platelets counts lar to a pulse oximeter that measures the concen-
are surrogate markers for portal hypertension. tration of ICG in arterial blood using colorimetry.
Serum alanine aminotransferase and aspartate In a healthy subject, the ICG retention value at
aminotransferase are elevated when hepatocytes 15 min (ICGR-15) after intravenous administra-
undergo apoptosis. Raised serum ammonia level tion of the dye is about 10% and plasma disap-
is observed when hepatic encephalopathy sets in pearance rate (PDR) is >18%. The ICG clearance
and hypoglycemia is an indication of fulminant test can be used alone or in combination with
hepatic failure. The Child-Pugh classification other clinical parameters for preoperative assess-
(A, B, C) is commonly used to categorize liver ment of liver function. The higher the ICGR-15
function reserve. Patients with Child-Pugh class value, the higher the hospital mortality rate is
A liver function are considered suitable for observed. Our experience indicated that a cutoff
major hepatectomy, whereas those with Child- value of ICGR-15 for a safe major hepatectomy
Pugh class B liver function are only eligible for is 14% and for minor hepatectomy is 22% [5].
minor hepatic resection in selected cases. With experience, the limit could be extended to
Nonetheless, the accuracy of this scoring sys- 17% for major hepatectomy. More importantly,
tem is undermined by the fact that subjective the ICG clearance test is a more sensitive test
clinical parameters, i.e. ascites and encephalop- than the Child-Pugh score in preoperative
athy, are included in the assessment, rendering it ­assessment of liver function. A wide range of
susceptible to inter-­observer variation. To solve ICGR-15 values exist among patients with Child
38  Liver Surgery: Early Complications—Liver Failure, Bile Leak and Sepsis 499

A and B cirrhosis. Considering ICGR-15 is a tive complications and mortality. In view of this,
reflection of liver function, liver function and evaluation of the size of future liver remnant by
reserve could be quite heterogeneous even among computed tomography (CT) volumetry forms an
patients with Child A or B cirrhosis. Nonetheless, essential part of the preoperative assessment of
ICGR-15 value should be interpreted with cau- liver function. In patients with normal liver, a
tion, as falsely high values could be observed in minimum of 20–25% of standard liver volume is
portal vein obstruction, bile duct obstruction, sig- essential to ensure survival [8]. In cirrhotic livers,
nificant intrahepatic arteriovenous shunting or a future liver remnant ≥40% is required to ensure
Gilbert syndrome. a safe major hepatectomy. When the future liver
remnant <40%, preoperative portal vein emboli-
zation (PVE) can be used to induce hypertrophy
Liver Scintigraphy of the liver remnant in 3–6 weeks’ time. PVE
blocks blood flow to the liver ipsilateral to the
Another quantitative liver function test that is of tumor so portal inflow is diverted to the contralat-
clinical interest is the dynamic 99mTc-mebrofenin eral lobe and induces clonal expansion and
scintigraphy coupled with SPECT imaging. hypertrophy of the hepatocytes [9]. PVE has
99
Tc-mebrofenin is taken up but not metabolized been shown to reduce the incidence of liver fail-
by hepatocytes, and is completely excreted into ure after hepatectomy [10]. However, there
bile by adenosine-triphosphate driven export awaits randomized control trials to show the ben-
pumps, which makes it an ideal agent to assess the efit of PVE in terms of reduction in hospital mor-
function of hepatocytes. Moreover, it has the abil- tality and prolongation of the overall survival of
ity to measure segmental liver function as well as cancer patients.
total liver function, which is different from ICG
that measures total liver function/flow only.
Because of this unique property, there has been an  iver Protective Strategy During
L
enormous interest in Western centers to evaluate Hepatectomy
the role of 99mTc-mebrofenin scintigraphy in pre-
operative evaluation of future remnant liver func- Meticulous surgical technique is an important
tion prior to major hepatectomy. One major factor to prevent liver injury due to bleeding and
advantage of this imaging technique is the ability ischemia. Excessive bleeding induces organ isch-
to provide a single cut-off value for the prediction emia that in turn predisposes to sepsis and sys-
of liver failure in both diseased and normal liver temic inflammatory reaction, resulting in
parenchyma. Recent evidence suggested that the multi-organ failure and potentially death.
uptake cutoff value for liver failure and liver fail- Hyperdynamic injury to small remnant liver and
ure mortality in non-cirrhotic livers was 2.5– liver congestion as a result of inadvertent damage
2.69%/min/body surface, and 2.2%/min/body to major hepatic veins are other factors that could
surface respectively [6, 7] Data on cirrhotic livers, lead to liver failure (Fig. 38.1). As a result, sur-
however, is still lacking. Due to the financial cost geons play an important role in liver protection
and logistical reason to arrange the scan, it during major hepatectomy. From a technical
remains to be seen if this imaging technique will point of view, the operation should begin with a
become part of the routine assessment of preop- generous skin incision, i.e. bilateral subcostal
erative liver function in the future. incision with midline extension. Adequate expo-
sure is the key element to facilitate mobilization
of the right liver and dissection along the plane
CT Volumetry between the anterior surface of inferior vena cava
(IVC) and the posterior surface of the caudate
The volume of liver remnant after hepatectomy is lobe. It also provides more space to allow rota-
an important element in determining postopera- tion of the liver to the left side and reduces the
500 A. C. Y. Chan and S. T. Fan

Fig. 38.1 Operative
causes of liver failure Massive hemorrhage

Hyperdynamic injury to small Liver ischemia Sepsis


liver remnant

Systemic inflammatory
Liver failure response

Liver congestion

Mortality

chance of avulsion of the left hepatic vein when a adopted right at the beginning of parenchymal
right hepatectomy is performed. Thoracotomy transection. Therefore, we recommend that the
should be advocated to give better exposure when hanging maneuver should be applied to provide
a large tumor is located in segment 7 or 8 of the direction for transection only when the middle
liver. It is important to remove all packs and hepatic vein is exposed and passed by.
gauzes in the liver hilum before excessive rota- Maintaining a low central venous pressure (CVP)
tion of the right liver as this could potentially helps to minimize blood loss during hepatic
cause extrinsic compression on the inflow ves- resection, [12] as it facilitates venous drainage
sels. Excessive rotation of the right liver would from the hepatic sinusoids and thus reducing
also cause compression of the left lateral segment venous backflow and hepatic congestion. A CVP
against the left subcostal wound, leading to pres- in the range of 3–5 cmH2O is preferable though
sure ischemia. For large tumors, using the ante- the risk of air embolism becomes a concern if
rior approach would avoid excessive rotation of CVP drops below this range. Simple physical
the liver as the hepatic transection commences measures include stopping intravenous fluid and
down towards the anterior surface of the IVC tilting the operative table in a reverse
once the inflow vessels are divided and ligated. Trendelenburg position to help bring down the
Bleeding volume, blood transfusion requirement CVP. If these measures fail, a bolus of low dose
and oncologic outcomes were all shown to diuretic can be administered. A recent report sug-
improve after adoption of the anterior approach gested that the use of Milrinone, a phosphodies-
[11]. However, control of hemorrhage deep in the terase three inhibitor, is effective in reducing the
parenchyma can sometimes be difficult with this CVP during donor hepatectomy by causing dia-
approach. To address this problem, the use of a stolic relaxation of the heart, which in turn
hanging maneuver may allow exposure of the improves venous return from the IVC and reduces
deep parenchyma better and improve surgical venous backflow into the liver [13]. Apart from
hemostasis. The hanging maneuver entails blind keeping a low CVP, control of inflow vessels, i.e.
passage of a long instrument with a tape into the Pringle maneuver, is another effective way to
anterior surface of the IVC and the posterior sur- minimize blood loss during hepatectomy, as
face of the caudate lobe. Inadvertent injury of the blood supply from the hepatic arterial and portal
IVC upon blind passage of the instrument can circulation would become temporarily occluded
happen and will lead to profuse hemorrhage. by clamping. Intermittent Pringle maneuver was
Another pitfall of this maneuver is that the plane shown to be effective in reducing blood loss from
of transection can be deviated from the original the transection surface in a randomized control
plane, guided by the middle hepatic vein if this is trial [14]. It has also been shown in various clini-
38  Liver Surgery: Early Complications—Liver Failure, Bile Leak and Sepsis 501

cal studies to be protective against ischemic acids as it is anti-catabolic and promotes protein
injury to the liver. However, it does not guarantee synthesis in cirrhotic patients. Medium-chain tri-
a bloodless operative field as bleeding from glycerides form part of the PN regimen as it
hepatic veins can still occur. In this situation, depends less than long-chain triglycerides on
total vascular occlusion, i.e. clamping of both binding to serum albumin which is advantageous
inflow and outflow vessels, could be employed for cirrhotic patients. No other intravenous fluid,
but hemodynamic disturbance is expected. The other than PN, are given in order to avoid fluid
ultrasonic dissector is our preferred device for overload and liver congestion. We were able to
parenchymal transection. It is effective in reduc- demonstrate that the use of PN in the form of
ing perioperative blood loss and facilitates expo- branched-chain amino acids reduces postopera-
sure of the major hepatic vein so as to guide the tive septic complications, ascites formation and,
direction of transection. With judicious use of the more importantly, leads to quicker recovery of
ultrasonic dissector, perioperative blood loss has liver function [16]. This, however, is not rou-
been reduced year by year in our center [15]. tinely done in many centers worldwide. Early
oral intake is encouraged as soon as bowel sounds
resume to maintain intestinal integrity, avoid bac-
 ostoperative Management After
P terial translocation, stimulate production of hepa-
Hepatectomy tocyte growth factor and enhance portal blood
flow, all of which are important elements for liver
All patients at risk of liver failure should be regeneration.
admitted to the intensive care unit after hepatec-
tomy with continuous monitoring of hemody-
namics, body temperature and urine output.  anagement of Liver Failure After
M
Monitoring for emergence of liver failure is the Hepatectomy
most important aspect of postoperative manage-
ment. Early clinical signs of liver failure, i.e. Early referral to a tertiary center with experience,
depressed sensorium, hypotension, respiratory possible availability of liver support devices and
depression and oliguria, and laboratory signs liver transplant service is crucial to improve the
such as prolonged prothrombin time, hypoglyce- chance of survival in these patients. The aim of a
mia, metabolic acidosis should be carefully mon- liver support device is to remove accumulated
itored. Oliguria is one of the earliest sign of liver toxic substance that cannot be metabolized by the
insufficiency and excessive inflammatory failing liver using an extracorporeal circulation
response. When intravenous fluids fail to increase system. There are two main types of dialysis-
urine output, liver failure needs to be ruled out. based liver support systems: bioartificial and arti-
Excessive fluid overloading is potentially hazard- ficial. While the development of bioartificial
ous, as it induces liver congestion and further livers is still in its infancy, various forms of artifi-
deterioration of liver function. Early hemody- cial livers, such as the molecular adsorbent recir-
namic support and possibly hemodialysis should culating system (MARS), liver dialysis unit and
be implemented early. the Prometheus device, have been approved for
Routine parenteral nutrition (PN) has been clinical use in liver failure. In fact, MARS treat-
advocated by our center as an important aspect of ment has been shown to be effective in reducing
postoperative management, particularly for cir- serum bilirubin and ammonia levels in our cohort
rhotic patients undergoing major hepatectomy, as of 74 patients with liver failure. Though the
it reduces the body net catabolic response to sur- 30-day mortality rate remains over 70%, about a
gery and enhances protein synthesis that is essen- fifth of the patients were able to receive bridging
tial to maintain immunological and metabolic transplantation [17]. However, a large random-
functions. The choice of PN in our center is a ized study of MARS treatment for acute-chronic
solution enriched with branched-chain amino liver failure, failed to demonstrate any survival
502 A. C. Y. Chan and S. T. Fan

benefit compared to standard medical therapy and hence predispose to bile leak. Alternatively,
[18]. High volume plasma exchange has been collection of bile in the dead-space forms a favor-
promising for the treatment of acute liver failure; able environment for micro-organisms to grow.
a recent randomized trial demonstrated a survival The clinical presentation of bile leak includes an
of 58.7% in patients receiving high volume onset of fever on postoperative day 4–7. For
plasma exchange compared to 47.8% with stan- immuno-compromised patients, persistent tachy-
dard medical therapy alone [19]. cardia after hepatectomy could be the tell-tale
sign of occult bile leak. Other clinical symptoms
include chills and rigors, abdominal distension,
Bile Leak malaise, nausea, and vomiting. A bilo-cutaneous
fistula is manifested as bile discharges from the
Despite a reduction in operative mortality, the main wound. If left untreated, a continuous bile
incidence of biliary complications has not leak will lead to bacterial peritonitis, and even
changed over the years with an incidence ranging reactionary hemoperitoneum secondary to intra-­
from 4.0 to 8.1% in various large series [20–22]. abdominal sepsis.
Therefore, it is of paramount importance to Laboratory blood tests show leucocytosis and
implement measures that allow early detection of an abnormal liver enzymes profile with predomi-
bile leak and thereby reduce the adverse effects nant elevation of serum bilirubin and alkaline
of biliary complications on postoperative mor- phosphatase. Serum gamma-glytamyl transpepti-
bidity and mortality. dase can be high but this is non-specific, as it
could also signal a hepatocyte damage on the
liver transection surface. A high-resolution CT
 athophysiology of Bile Leak, Sepsis
P scan of the abdomen can detect fluid accumula-
and Liver Failure tion in the dead space or adjacent to the raw sur-
face of the liver. The source of bile leak is
The presence of bile and blood clots in the dead confirmed by a cholangiogram obtained either by
space after hepatectomy provides a good medium endoscopic retrograde cholangiopancreatogra-
to harbor bacterial growth. Infection provokes a phy (ERCP) or percutaneous transhepatic chol-
systemic inflammatory response that is character- angiography (PTC). Any active contrast
ized by the release of various cytokines including extravasation confirms the location of bile leak.
tumor necrosis factor-alpha, interleukin-1 and However, it is noteworthy to highlight that the
interleukin-6 [23–26]. These cytokines in turn cholangiogram may not be able to detect leaks
cause dysfunction of the host defense immune arising from damage to a segregated bile duct
system and subsequently multi-organ failure. that is not communicating with the main biliary
Patients with loss of liver mass after hepatectomy system. It is important to look for any fluid accu-
are more susceptible to the development of liver mulation adjacent to the caudate lobe, as this is a
failure once this cascade of inflammatory reac- common site for leak from transected bile ducts
tions is triggered by biliary complications. in the caudate lobe segregated from the main bili-
ary system. In this situation, a cholangiogram via
 linical Presentation of Bile Leak
C ERCP or percutaneous transhepatic biliary drain-
Advancing age is a risk factor for postoperative age cannot detect the site of leak and its diagnosis
biliary complications [20]. Although the caus- and treatment depend on image-guided percuta-
ative association between age and bile leak is less neous drainage. The aspirated bile should be sent
clear, it has been shown that intra-abdominal sep- for bacteriological culture. The common caus-
sis is more common in elderly patients after hep- ative microorganisms are Staphylococcus aureus,
atectomy [26]. It was thought that infection and Escherichia coli and Candida species. Other
bile leak correlate with each other. Infection may microorganisms involved are Streptococcus,
induce tissue devitalization around the bile duct Pseudomonas, Morganella, and bacillus. When
38  Liver Surgery: Early Complications—Liver Failure, Bile Leak and Sepsis 503

patients present with generalized peritonitis or Tissue Aspirator (CUSA; Valley Lab, Boulder,
when non-operative treatments fail, a re-­operation CO) for parenchymal transection. The advantage
is indicated. of CUSA is to allow good exposure of blood ves-
Definition of post-hepatectomy bile leak is sels and bile duct inside the liver parenchyma and
defined as follows: reduce devitalization of liver tissues. When the
dissection is performed near the hilar plate, the
1. Presence of bile from output in the abdominal power of CUSA is reduced to avoid denudation
drain of the hepatic ducts. After completion of liver
2. Intra-abdominal collection of bile confirmed transection, hemostasis is ensured and a bile leak
on re-operation or from percutaneous test is performed. Ten milliliters of diluted meth-
drainage ylene blue solution is injected slowly via the cys-
(a) Radiological evidence of bile leak on tic duct into the common bile duct. If there is leak

cholangiogram obtained either by ERCP in the raw surface of the liver, it will be shown by
or PTC extravasation of the methylene blue solution that
is then plicated by fine absorbable sutures.
Bile Leak Common sites of bile leak include the bile
In contrast to blood, bile does clot readily and can duct stump, minor ducts over the transection sur-
leak through tiny defects in a divided bile duct or face and the caudate lobe. Compression on the
from suture lines in the bile duct stump. Therefore, supraduodenal portion of the common bile duct
meticulous surgical technique is the key to pre- may facilitate detection of any bile leak, but such
vent bile leak. In order to appreciate how bile leak maneuver would undoubtedly increase the intra-
is detected and to be prevented, one has to gain ductal pressure, predisposing to bile seepage
some insight into the operative technique of hepa- through the needle holes and resulting in a ‘false
tectomy. The following paragraph describes the positive’ result. As the biliary tract is a low pres-
technique of hepatectomy adopted by our center. sure system, it is insensible to create a high-­
pressure condition artificially and such maneuver
is therefore not recommended. For those patients
Surgical Techniques of Hepatectomy with a prior history of cholecystectomy, when
for Liver Tumors the cystic duct stump is no longer identifiable it
may not be feasible to perform the methylene
The operation begins with a bilateral subcostal blue test via the cystic duct approach. In this
incision with midline extension for optimal expo- situation, bile leak can be tested by gently press-
sure and access to the liver. An intraoperative ing the raw surface of the liver with a piece of
liver ultrasonography is then performed to iden- gauze and inspecting for any traces of yellow
tify the location of the tumor and its anatomical bile stain afterwards. When an external stent is
relationship with the middle or right hepatic vein, placed across the anastomosis in a hepaticojeju-
so as to define the parenchymal transection line. nostomy, an intraoperative bile leak test can be
The gallbladder is then removed and the cystic performed by injection of methylene blue solu-
duct is cannulated by a Fr 3.5 Argyle infant feed- tion into the external stent and the surgeon can
ing tube. An intraoperative cholangiogram is then check for any leak from the anastomotic line. A
obtained to detect anatomical variation of the cholangiogram is then performed at around 7–10
biliary system. Hilar dissection is performed to days after the operation to detect for any occult
isolate the ipsilateral hepatic artery and portal leak from this bilio-enteric anastomosis and the
vein, and is subsequently divided and suture-­ stent can only be removed at around 4–6 weeks
ligated. By this time, the liver parenchyma that is after the operation to allow formation of a mature
supplied by the ipsilateral inflow vessels would fistula tract. Other operative measures that could
become discolored and the transection line will prevent bile leak include the application of a
be demarcated. We use the Cavitron Ultrasonic hemostatic agent such as fibrin glue to form a
504 A. C. Y. Chan and S. T. Fan

plug within the leak site. An alternative measure required to repair the site of bile leak.
is to deploy a piece of greater omentum to cover Avoidance of excessive dissection at the hilar
the raw surface of the liver. plate and denudation of the hepatic duct are
key factors to prevent postoperative bile leak.
 ethylene Blue Test and Postoperative
M
Bile Leak
The methylene blue test is a sensitive method to References
rule out bile leak. In our previous retrospective
analysis of 304 patients who had a methylene blue 1. Miyagawa S, Makuuchi M, Kawasaki S, Kakadu
test, 60 patients had a positive test, a 3.6% bile T. Criteria for safe hepatic resection. Am J Surg.
leak rate [27]. The leak site was sutured intraop- 1995;169(6):589–94.
2. Wei AC, Poon RT, Fan ST, Wong J. Risk factors for
eratively but 10% of these patients still developed perioperative morbidity and mortality after extended
postoperative bile leak. One possibility was that hepatectomy for hepatocellular carcinoma. Br J Surg.
the defect was not sutured adequately. Another 2003;90(1):33–41.
possibility was that the bile duct might have been 3. Schroeder RA, Marroquin CE, Bute BP, Khuri S,
Henderson WG, Kuo PC. Predictive indices of mor-
denuded by excessive dissection at the hilar region bidity and mortality after liver resection. Ann Surg.
and the friable tissues around the bile duct did not 2006;243(3):373–9.
hold sutures well. Furthermore, the persistent bile 4. Ichikawa T, Uenishi T, Takemura S, et al. A sim-
leak could be a manifestation of major duct injury ple, noninvasively determined index predicting
hepatic failure following liver resection for hepato-
and the ligatures at the bile duct stump were dis- cellular carcinoma. J Hepatobiliary Pancrat Surg.
lodged by the increased ductal pressure as it built 2009;16(1):42–8.
up in the postoperative period. On the other hand, 5. Fan ST. Liver functional reserve estimation: state
among those who had a negative bile leak test, of the art and relevance for local treatments: the
Eastern perspective. J Hepatobiliary Pancreat Sci.
only 2% developed this complication [27]. In 2010;17(4):380–4.
other words, a negative methylene blue test is reli- 6. Dinant S, de Graaf W, Verwer BJ, et al. Risk assess-
able to rule out bile leak. ment of posthepatectdomy liver failure using hepato-
biliary scintigraphy and CT volumetry. J Nucl Med.
2007;48(5):685–92.
 anagement of Bile Leak
M 7. de Graaf W, van Lienden KP, Dinant S, et al.
Adequate fluid resuscitation and the use of broad-­ Assessment of future remnant liver function using
spectrum antibiotics follow the basic principle hepatobiliary scintigraphy in patients undergo-
for treatment of intra-abdominal sepsis. A patient ing major liver resection. J Gastrointest Surg.
2010;14(2):369–78.
should be fasted for a sufficient period of time 8. Chan SC, Liu CL, Lo CM, et al. Estimating liver
before further intervention is implemented. Any weight of adults by body weight and gender. World J
sizeable intra-abdominal bile collection should Gastroenterol. 2006;12(14):2217–22.
be drained either percutaneously or by laparot- 9. Kubota K, Makuuchi M, Kusaka K, et al. Measurement
of liver volume and hepatic functional reserve as a
omy. If there is persistent drainage of bile, endo- guide to decision-making in resectional surgery for
scopic papillotomy and stenting are necessitated. hepatic tumors. Hepatology. 1997;26(5):1176–81.
Re-operation is indicated when there are signs of 10.
Hemming AW, Reed AI, Howard RJ, et al.
generalized peritonitis or hemoperitoneum or the Preoperative portal vein embolization for extended
hepatectomy. Ann Surg. 2003;237(5):681–91; dis-
leak is from a segregated hepatic duct. The mor- cussion 691–683
tality rate due to biliary complications is high and 11. Liu CL, Fan ST, Cheung ST, et al. Anterior approach
ranges from 20 to 30% [20]. versus conventional approach right hepatic resec-
tion for large hepatocellular carcinoma: a pro-
spective randomised controlled study. Ann Surg.
Conclusion 2006;244(2):194–203.
With the use of methylene blue test after paren- 12. Jones RM, Moulton CE, Hardy KJ. Central venous
chymal transection, the incidence of biliary pressure and its effect on blood loss during liver resec-
complications has declined from 9.8 to 3.5% in tion. Br J Surg. 1998;85(8):1058–60.
recent years. Meticulous surgical technique is
38  Liver Surgery: Early Complications—Liver Failure, Bile Leak and Sepsis 505

13. Ryu HG, Nahm FS, Sohn HM, et al. Low central 21. Yamashita Y-i, Hamatsu T, Rikimaru T, Tanaka S,
venous pressure with milrinone during living donor Shirabe K, Shimada M, Sugimachi K. Bile leakage
hepatectomy. Am J Transplant. 2010;10(4):877–82. after hepatic resection. Ann Surg. 2001;233(1):45–50.
14. Man K, Fan ST, Ng IO, et al. Prospective evaluation 22. Nagano Y, Togo S, Tanaka K, et al. Risk factors and
of Pringle manoeuvre in hepatectomy for liver tumors management of bile leakage after hepatic resection.
by a randomised study. Ann Surg. 1997;226(6):704– World J Surg. 2003;27(6):695–8.
11; discussion 711–703 23. Calandra T, Baumgartner JD, Grau GE, et al.

15. Fan ST, Lo CM, Poon RT, et al. Continuous improve- Prognostic values of tumor necrosis factor/cachectin,
ment of survival outcomes of resection of hepatocel- interleukin-1, interferon-alpha, and interferongamma
lular carcinoma: a 20-year experience. Ann Surg. in the serum of patients with septic shock. Swiss-
2011;253(4):745–58. Dutch J5 Immunoglobulin Study Group. J Infect Dis.
16. Fan ST, Lo CM, Lai EC, et al. Perioperative nutri- 1990;161(5):982–7.
tional support in patients undergoing hepatec- 24. Calandra T, Gerain J, Heumann D, Baumgartner JD,
tomy for hepatocellular carcinoma. N Engl J Med. Glauser MP. High circulating levels of interleukin-6 in
1994;331(23):1547–52. patients with septic shock: evolution during sepsis,
17. Chiu A, Chan LM, Fan ST. Molecular adsorbent
prognostic value, and interplay with other cytokines.
recirculating system treatment for patients with The Swiss-Dutch J5 Immunoglobulin Study Group.
liver failure: the Hong Kong experience. Liver Int. Am J Med. 1991;91(1):23–9.
2006;26(6):695–702. 25. Martin C, Boisson C, Haccoun M, et al. Patterns

18. Banners R, Nevens F, Larsen FS, et al. Extracorporeal of cytokine evolution (tumor necrosis factor-alpha
albumin dialysis with the molecular adsorbent recir- and interleukin-6) after septic shock, hemor-
culating system in acute-on-chronic liver failure: the rhagic shock, and severe trauma. Crit Care Med.
RELIEF trial. Herpetology. 2013;57(3):1153–62. 1997;25(11):1813–9.
19. Larsen FS, Schimidt LE, Bernsmeier C, et al. High 26. Yanaga K, Kanematsu T, Takenaka K, et al.

volume plasma exchange in patients with acute Intraperitoneal septic complications after hepatec-
liver failure: an open randomised controlled trial. J tomy. Ann Surg. 1986;203(2):148–52.
Hepatol. 2016;64(1):69–78. 27. Lam CM, Lo CM, Liu CL, et al. Biliary com-

20. Lo CM, Fan ST, Liu CL, et al. Biliary complications plications during liver resection. World J Surg.
after hepatic resection: risk factors, management, and 2001;25(10):1273–6.
outcome. Arch Surg. 1998;133(2):156–61.
Pain Management in Liver
Transplantation 39
Paul Weyker, Christopher Webb,
and Leena Mathew

Keywords address and discuss in detail the analgesic issues


Methadone · Opioids · Epidural analgesia in liver transplantation and liver resection.
Transverse abdominis plane · Duramorph The goals of analgesia during liver transplan-
Transcutaneous electrical nerve stimulation tation are similar to other types of surgery, but
unique considerations found in patients with end-
stage liver disease impact the overall approach to
Introduction pain management. Altered physiologic parame-
ters including the pharmacokinetics and pharma-
Liver transplantation is a complex surgical proce- codynamics of commonly used analgesics,
dure requiring comprehensive and intensive mul- decreased coagulation factors, abnormal platelet
tidisciplinary involvement in the perioperative function, and altered mental status are some of
period. Over the years there has been substantial these important considerations. Many patients
evolution of the surgical technique and the periop- with end-stage liver disease also have a history of
erative management that resulted in improved alcohol or drug abuse, as 10–12% of patients
outcomes. The anesthesiologist and intensivist undergoing liver transplantation have alcoholic
play a crucial role throughout the perioperative liver disease [1]. These patients often require
period and adequate analgesic delivery is of multiple hospitalizations during which they
utmost importance during this period. Providing receive opioids and develop increased opioid
adequate pain control may prove to be challeng- requirements compared to healthy patients under-
ing and there are unique considerations in patients going similarly extensive operations. It is no sur-
undergoing liver transplantations or resections. In prise that perioperative pain management in
addition to relieving mental suffering associated patients undergoing liver transplantation involves
with pain, appropriate pain control is essential to a multifaceted approach that includes medical
prevent the profound physiologic consequences optimization of the patient followed by the devel-
of inadequate analgesia. This chapter aims to opment of an analgesic plan that extends from the
preoperative setting and continues into the
extended postoperative period.
P. Weyker, MD • C. Webb, MD While this chapter will focus on pain manage-
L. Mathew, MD (*) ment after liver transplantation and living liver
Department of Anesthesiology,
Columbia University Medical Center,
donation, many of its conclusions can be applied
New York, NY, USA for the treatment and prevention of pain after
e-mail: lm370@cumc.columbia.edu hepatic resection as well.

© Springer International Publishing AG, part of Springer Nature 2018 507


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7_39
508 P. Weyker et al.

Metabolism and Clearance phase I metabolism are significantly prolonged


when compared to drugs metabolized through
Biotransformation is the process by which drugs phase II reactions in patients with cirrhosis [4, 7]
are broken down into metabolites that more eas- confirming the commonly accepted belief that
ily eliminated by the body [2, 3]. The liver plays phase I reactions are greatly impaired in patients
a key role in this intricate process such that small with chronic liver disease while phase II are
changes in liver function or liver blood flow can essentially preserved [8].
dramatically change the concentrations of drugs
and their metabolites. Cirrhotic end-stage liver
disease is characterized by the histological pres- Pharmacokinetics in Liver Disease
ence of hepatocellular fibrosis. These histologi-
cal changes, clinically termed cirrhosis, result in Drug metabolism in liver failure is discussed
decreased hepatic blood flow, porto-systemic extensively elsewhere in this book. We will there-
shunting, sinusoidal capillarization, and an over- fore focus the discussion here on the pharmaco-
all reduction in the activity and quantity of hepa- kinetics of commonly used analgesics. Opioids
tocytes. Consequently, these physiologic have long been the foundation of pain manage-
aberrations alter drug absorption, distribution, ment in anesthesiology. The WHO analgesic lad-
and elimination [4]. These changes manifest as der recommends the use of opioid analgesics in
increased oral bioavailability, decreased protein the treatment of moderate to severe pain. A mul-
binding, prolonged duration of action, and an timodal approach is essential in tailoring an anal-
overall reduction in drug metabolism [3, 4]. gesic regimen that is specific to the individual
Drugs used for both the acute and chronic pain patient with distinct comorbidities. To avoid
management are primarily lipid soluble and must under or over treatment, it is imperative to under-
undergo enzymatic metabolism into more soluble stand the medication classes at our disposal.
forms before being excreted by the kidneys [2, Opioids remain the gold standard for the treat-
4–6]. These enzymatic reactions are categorized ment of moderate to severe pain in the acute set-
as phase I and phase II reactions depending on ting, but the use of opioids in the long-term
how the liver alters them. In the case of phase I setting of nonmalignant pain continues to be
reactions, the drugs undergo chemical modifica- controversial.
tions including hydrolysis, oxidation, dealkyl-
ation, and reduction. In the case of phase II
reactions, the drugs undergo conjugation, that Morphine
renders them watersoluble [2, 4–6]. Phase I reac-
tions involve the cytochrome P-450 family of Morphine is the prototypic phenanthrene alka-
enzymes and occur in the smooth endoplasmic loid derived from opium. It undergoes signifi-
reticulum of hepatocytes [5]. More specifically, cant first pass metabolism in the liver, resulting
the isoenzymes primarily involved in phase I in an oral bioavailability of 30–40% [6, 8]. While
metabolism of most drugs, including the opioids, the liver accounts for 60–70% of its metabolism,
involve the CYP3A4 and CYP2D6 subgroups significant extrahepatic metabolism through the
[2, 6]. Phase II reactions involve conjugation of kidneys has also been described [9, 10]. In the
the parent drug by transferases, such as uridine liver, morphine is metabolized through
diphosphate-glucuronosyltransferases (UGTs) glucuronidation forming morphine-3-glucuro-
­
whereby methylation, acetylation, and sulfation nide (M3G), morphine-6-glucuronide (M6G)
renders the drugs more water soluble and thus and to a lesser extent demethylation to normor-
easily excreted [2, 6, 7]. Phase I reactions are phine [6, 8]. M3G is the main metabolite of mor-
more impaired in patients with cirrhosis as com- phine and while it is generally thought to be an
pared to phase II reactions. Sellers et al. have inactive metabolite, some studies suggested that
shown that the half lives of drugs undergoing M3G may act as an anti analgesic [6, 9]. M6G
39  Pain Management in Liver Transplantation 509

remains p­ harmacologically active and while it is hepatic metabolism is excreted unchanged by


produced at much smaller amounts, it can accu- both the kidneys and through the biliary system.
mulate in patients with renal dysfunction [6, 8]. In the context of liver disease, one can infer that
The metabolism of morphine varies depending methadone metabolism is slowed due to the
on the degree of cirrhosis; patients with severe impairment of phase I reactions in patients with
cirrhosis (Child-Pugh class C) have an increased cirrhosis. Interestingly, studies utilizing mass
oral bioavailability due to decreased first pass spectrometry illustrated that the total 24 h urinary
metabolism, lower plasma clearance, and pro- excretion of methadone and its inactive metabo-
longed elimination half-lives due to a decrease in lites was drastically reduced in patients with liver
total body clearance [2, 6, 8, 11–13]. Using disease (32.6%) compared to matched healthy
hepatic vein catheterization to directly measure controls (48.3%) [17]. Additionally, Novick et al.
morphine hepatic extraction in cirrhotic patients, found similar findings in alcoholic patients with
Crotty et al. showed that hepatic extraction ratios cirrhosis with lower peak plasma levels in the cir-
were decreased by 25% in cirrhotic patients rhotic group compared to alcoholic patients with-
compared to healthy controls [12]. Given the out cirrhosis [16]. It seems counterintuitive that
higher free plasma concentrations of morphine peak plasma levels are lower in cirrhotic patients
combined with the reduced metabolism, many when many studies have demonstrated a pro-
physicians not only decrease the total dose of longed elimination half-life in cirrhotic patients
morphine given but also increase the time inter- compared to controls. This may be due to a com-
val between doses [6]. bination of both an increase in the volume of dis-
tribution of methadone and intra- and extrahepatic
storage of methadone that is independent of
Methadone reduced enzymatic capacity [6, 16]. Interestingly,
cirrhotic patients in these studies did not exhibit
Methadone is a synthetic opioid agonist used any signs or symptoms of methadone overdose
most commonly as a treatment for chronic pain due to increased biliary excretion of methadone
as well as in the detoxification treatment from and its metabolites into the gastrointestinal tract
heroin. While methadone maintenance therapy [16]. Nonetheless, many anesthesiologists rec-
(MMT) is a controversial topic in the setting of ommend that methadone is used cautiously in
liver transplantation, it is nonetheless imperative patients with liver dysfunction [15].
that the anesthesiologist understands its role in
the context of end-stage liver disease. Unlike
morphine and many of the other opioids, metha- Hydromorphone
done exhibits low hepatic extraction, resulting in
a high oral bioavailability [14]. Methadone is Hydromorphone, a semi-synthetic opioid, is a
highly bound to plasma proteins, with some stud- phenanthrene opioid similar to morphine [18].
ies suggesting that 90% of the plasma concentra- Acting primarily at the m-opioid receptor, hydro-
tion of methadone is protein bound [6]. This morphone has a half-life of 1–3 h and is 7–10
translates to a prolonged elimination half-life of times more potent at these receptors than mor-
about 30 h (reports ranged from 8.5 to 58 h) [6]. phine [18, 19]. Hydromorphone also undergoes
Interestingly the analgesic half-life of methadone first pass metabolism in the liver through
may be quite short (4–6 h). Therefore, metha- glucuronidation to form hydromorphone-3-­
­
done must be used very judiciously in the treat- glucuronide (H3G), a neuroexcitatory metabolite
ment of acute pain. Methadone undergoes that lacks analgesic effects [2, 19, 20]. Studies
extensive hepatic metabolism through phase I, or involving rats, whose lateral ventricles were
oxidative reactions via demethylation to inactive directly injected with synthetic H3G, have illus-
metabolites that are excreted in the urine and bile trated that the metabolite induces myoclonic
[6, 15, 16]. The remainder of the drug that escapes jerks, allodynia, and seizures in a dose-dependent
510 P. Weyker et al.

manner similar to the neuroexcitatory effects and desproprionfentanyl as metabolites [25]. An


seen with M3G [19]. While both metabolites are initial study of fentanyl in eight patients with
too polar to cross the blood–brain barrier (BBB) mild-to-moderate hepatic insufficiency failed to
in large quantities, a clinically significant portion demonstrate a significant prolongation of half-­
can cross the BBB to elicit the aforementioned life. Haberer et al. showed that the half-life of
effects in patients with impaired elimination [20]. fentanyl in cirrhotic patients was minimally pro-
The neuroexcitatory effects of the hydromor- longed to 304 min compared to 263 min in
phone metabolite are seen clinically in patients healthy controls [6, 18, 26]. Interestingly, the
with renal failure but less so than with morphine elimination half time of fentanyl in patients
metabolites [15]. Retrospective studies of pallia- undergoing abdominal aortic surgery with aortic
tive care patients with renal dysfunction who cross clamping was significantly prolonged
were switched from morphine to hydromorphone (8.7 h [27]) possibly due to decreased hepatic
demonstrated an 80% decrease in cognitive blood flow. Unfortunately, there are very few and
impairment, drowsiness, and nausea [21]. While limited studies analyzing the context-sensitive
there are limited studies of hydromorphone in half time of fentanyl in cirrhotic patients. Many
either liver transplant recipients or in patients clinicians will administer fentanyl to cirrhotic
with cirrhosis, data from models using morphine patients without any dosing reductions, given its
are commonly extrapolated to hydromorphone as lack of active or toxic metabolites.
they share a common metabolic pathway. As with
morphine, many pain physicians recommend
using a decreased dose in patients with hepatic Buprenorphine
impairment [13].
Buprenorphine, another member of the phenan-
threne opioid family, is a highly lipophilic opioid
Fentanyl with combined agonist–antagonist effect. While
buprenorphine is predominately excreted
The phenylpiperidine class of opioids includes unchanged in the bile, one-third undergoes
fentanyl, alfentanil, sufentanil, remifentanil, and hepatic metabolism via both phase I and phase II
meperidine. Fentanyl, the most commonly used reactions to form buprenorphine-3-glucuronide
of these, is a highly potent lipid soluble synthetic and nor-buprenorphine which are inactive and
opioid agonist [6, 13, 18]. Fentanyl is highly active metabolites, respectively [6, 28, 29]. There
selective for the m-opioid receptor, 80–100 times is also evidence for entero-hepatic circulation
more potent than morphine [6, 18, 22]. with a small percentage of both buprenorphine
Intravenous fentanyl has a distribution half-life and nor-buprenorphine being excreted in the
of 1–3 h compared to the transdermal form with feces [28, 29]. In comparing oral to sublingual
a 17-h half-life [13, 23]. The majority of fentanyl and parenteral routes, oral buprenorphine has
(85%) exists in the plasma as the protein bound poor bioavailability due to extensive first pass
form with 60% being bound to albumin and the kinetics while sublingual regimens maintain a
remainder is bound to alpha-1 acid glycoprotein bioavailability of 60–70% of the parenteral dose
[24]. As it is highly lipid soluble, fentanyl must due to its high lipid solubility [30]. Buprenorphine
first undergo reuptake from its lipid storage sites is 30–40 times more potent than morphine. Its
before undergoing phase I (CYP3A4) hepatic metabolite nor-buprenorphine has analgesic
biotransformation via de-alkylation and hydrox- effects that are 15–40 times less than buprenor-
ylation to inactive metabolites with less than 10% phine [6]. While the drug maintains a high affin-
being excreted in the kidneys unchanged [6, 18, ity for the m-opioid receptor, buprenorphine
25]. The major inactive metabolite produced by produces only partial agonistic effects (at doses
human hepatic enzymes is norfentanyl while ani- <0.5 mg/kg), namely supraspinal analgesia,
mal enzyme studies have found both norfentanyl respiratory depression, and meiosis [30, 31].
39  Pain Management in Liver Transplantation 511

Interestingly, studies involving nociceptive stim- Table 39.1  Opioids and equipotent doses
uli in mice showed that a 5–10% receptor occu- Equipotent dose Duration of
pancy produced effective analgesia [32]. Generic name parenteral (mg) action (h)
However, unlike the aforementioned morphine Morphine 10 4–5
derivatives, buprenorphine has the capacity to Hydromorphone 1.5 4–5
antagonize the k- and d-opioid receptors (at doses Oxymorphone 1.0–1.5 4–5
>0.5 mg/kg) resulting in limited spinal analgesia, Codeinea 120 (10–30) (4–6)
dysphoria, hallucinations, and delusions [30, 31]. Hydrocodone (5–10) (4–8)
Another important aspect of buprenorphine is the Oxycodone 10–15 4–5
ceiling effect for both analgesic and respiratory Methadone 7.5–10 3–5
depression [18, 30, 33]. Increasing the dose of Meperidine 80–100 2–4
Fentanyl 100 μg 0.5
buprenorphine beyond the analgesic level will
Sufentanil 15 μg 0.5
produce more dysphoria and other unwanted side
Alfentanil 750 μg 0.25
effects. Buprenorphine exhibits very slow recep-
Buprenorphine 0.4 4–6
tor dissociation from both the m and k receptors
Butorphanol 2–3 4–5
with a half-life of 2–5 h [30, 33, 34]. Clinically,
Nalbuphine 10 4–5
slow receptor dissociation produces fewer with-
Adapted from Wood and Wood. Drugs and anesthesia:
drawal signs and symptoms of withdrawal upon pharmacology for anesthesiologists. 2nd edn.; 1982
completion of buprenorphine treatment. This a
Numbers in parentheses, doses and duration of action for
slow dissociation combined with a high receptor oral doses
affinity also produces a competitive displacement
effect when buprenorphine is combined with
other opioids [18, 30]. Studies comparing recep- 34]. As with all opioid regimens in the transplant
tor assays of fentanyl and buprenorphine showed patient, patients must be closely monitored as
that buprenorphine is only displaced from the changes in hepatic function and perfusion during
opioid receptors once very high plasma concen- the perioperative period will affect opioid dosing.
trations of the other opioids are achieved [34]. Table 39.1 lists equipotent doses and duration of
Additionally, this opioid blocking effect has been action of commonly used opioids.
observed to last as long as 24 h [30]. These find-
ings led many clinicians to use buprenorphine in
addiction medicine where once daily dosing Dexmedetomidine
could be used for the treatment of opioid
withdrawal. Dexmedetomidine, an enantiomer of medetomi-
With the growing clinical use of buprenor- dine, is a highly selective alpha-2 agonist that is
phine and other partial agonists, it is important 1600 times more selective for the alpha-2 recep-
that the anesthesiologist understands how this tor than the alpha-1 receptor [35]. Compared to
drug pertains to the liver transplant patient. While clonidine, dexmedetomidine is 7–8 times more
there are limited studies available, based on potent at the alpha-2 receptor [35–37]. As an
known decreases in phase I metabolism in the alpha-2 agonist, dexmedetomidine binds to both
cirrhotic patient, many experts recommend to central and peripheral alpha-2 receptors.
lower the initial doses of buprenorphine with Postsynaptic alpha-2 receptors are located within
slow and monitored titration [6, 29]. In the peri- the central nervous system with the highest con-
operative setting, patients on stable sublingual centration of receptors found in the locus coeru-
doses of buprenorphine can be managed with a leus [38]. Presynaptic alpha-2 receptors are
divided dose of their maintenance regimen. located within the peripheral nervous system and
Breakthrough pain is best treated with highly various organ tissues [35, 37, 38]. Upon activa-
potent opioids, such as fentanyl due to the opioid tion, presynaptic alpha-2 receptors inhibit the
blocking effects of buprenorphine [29, 30, 33, release of norepinephrine from the nerve endings
512 P. Weyker et al.

[38, 39]. While dexmedetomidine does have a release [49]. Gabapentin is only available as an
supraspinal mechanism for analgesia, the pri- oral preparation and its bioavailability is inversely
mary analgesic response occurs at the level of the proportional to the dose given [50]. There is no
spinal cord by inhibition of nociceptive pathways hepatic metabolism and gabapentin is eliminated
in the dorsal horn [40–43]. Dexmedetomidine unchanged in the urine. Its elimination half-life is
undergoes extensive hepatic biotransformation 5–8 h and as such is often dosed in three times
with 95% of the parent drug being metabolized daily regimen [49]. The most common adverse
by both phase I and phase II reactions [44] to effects of the drug are somnolence and dizziness.
form inactive and nontoxic metabolites that are Multiple studies have found an opioid sparing
excreted in the urine and feces [44]. In healthy effect of preemptive analgesia with preoperative
patients, dexmedetomidine has an elimination oral gabapentin. Doses ranged anywhere from
half-life of 2–2.5 h [44]. In patients with severe 300 to 1200 mg in these studies. A total of seven
hepatic impairment, there is a decrease in plasma meta-analyses have concluded that gabapentin is
protein binding and clearance values while the effective in reducing postoperative pain and has
elimination half-life was increased to 3.9–7.4 h an opioid sparing effect [49]. There are no studies
[44]. While there are limited studies of dexme- to date looking at the efficacy and safety of gaba-
detomidine in liver transplant recipients, one case pentin dosing in end-stage liver disease patients
report described the successful use of a dexme- undergoing liver transplantation. However, given
detomidine infusion for 5 weeks postoperatively the lack of hepatic metabolism and opioid spar-
without any adverse side effects or signs/symp- ing effect, preoperative administration of gaba-
toms of withdrawal upon termination of the infu- pentin in carefully selected patients with
sion [45]. Similarly, studies evaluating the relatively normal renal function can be
prolonged use of dexmedetomidine in adult ICU considered.
patients suggests that adverse events, such as bra-
dycardia, occur only during drug loading while
withdrawal effects such as rebound tachycardia Perioperative Pain Management
and hypertension were absent [46].
Perioperatively, some anesthesiologists take The recognition that patients with end-stage
advantage of the (weak) analgesic effects of dex- liver disease suffer from a multitude of symp-
medetomidine to decrease the intra-operative toms, including nausea, dyspnea, and severe
MAC of anesthetic agents and the post-operative pain, is important but often underappreciated
opioid requirements [38, 47, 48]. [51]. According to a prospective cohort study by
Roth et al., approximately one-third of patients
with end-stage liver disease have at least moder-
Gabapentin ate pain, with pain scores very similar to patients
suffering from end-stage colon or lung cancer.
Originally developed as an antiepileptic drug, The study further found that two-thirds of these
gabapentin has become a widely used drug in the patients are low-income men with multiple
treatment of pain syndromes, including post-­ comorbidities including alcoholism and drug
therapeutic neuralgia, neuropathic pain, diabetic abuse. These patients often rate their quality of
peripheral neuropathy and to treat acute pain. life as poor to fair [51]. Often patients with a
The FDA originally approved its use in 1994 as ­ history of substance abuse have undertreated
an adjuvant for seizure prophylaxis [49]. pain due to the notion that these are unreliable
Chemically similar to the neurotransmitter patients. These physician biases in combination
GABA [50], multiple studies attempted to eluci- with concern of altered hepatic synthetic func-
date gabapentin’s mechanism of action. It likely tions make it understandable why pain in patients
inhibits specific high voltage activated calcium with end-­ stage liver disease is often
channels therefore reducing neurotransmitter undertreated.
39  Pain Management in Liver Transplantation 513

Ideally analgesic regimens should be contin- length of hospital stay. Although controversies
ued with modifications as needed through the exist regarding the utility of preemptive analge-
course of disease progression and the periopera- sia, more recent systematic reviews suggest that
tive period. The anesthesiologist’s role in analge- there may be some benefit from preemptive anal-
sic management starts with a thorough history gesia as long as appropriate attention is paid to
that includes the patient’s baseline analgesic regi- intraoperative and postoperative analgesia as
men. Knowing the patient’s preoperative analge- well [57, 58].
sic requirement and response to current therapy The use of opioids including fentanyl, hydro-
will allow the anesthesiologist to better predict morphone, methadone and remifentanil is appro-
the analgesic needs throughout the operative and priate during liver transplantation and the
postoperative period. intraoperative analgesic management is similar
to other major abdominal surgeries. However,
there are considerations unique to liver transplan-
Methadone Maintenance Therapy tation patients. Multiple studies have shown that
liver transplantation patients require less mor-
It is not unusual to encounter patients with end-­ phine than other major abdominal surgeries [59–
stage liver disease on methadone maintenance 62]. Eisenach et al. who first demonstrated this
therapy (MMT) for opioid abuse considering that difference in 1989, proposed that the decreased
over 80% of these patients are infected with hep- morphine requirement was due to elevated
atitis C [52]. To date there have been at least four endogenous opioids and not due to altered metab-
retrospective studies with a total 52 patients on olism [61]. Donovan et al. found higher levels of
MMT who have received liver transplants [52– metenkephalin in patients with end-stage liver
55]. Weinrieb et al. found that patients on MMT disease both before surgery and on postoperative
had much higher opioid requirements both intra- days 1–3; further evidence that decreased mor-
operatively and postoperatively compared to a phine requirements were likely due to increased
matched group of patients undergoing liver trans- levels of endogenous opioids [60]. Moretti et al.
plantation not on MMT [52]. Methadone therapy postulated that the decreased opioid requirements
has become a controversial topic in the field of may in part be due to the denervation of the trans-
liver transplantation. Some transplant centers planted organ [62]. Regardless of the exact mech-
require cessation of methadone before a patient is anism, the anesthesiologist should be aware of
allowed on the waiting list [52]. However, the this difference in opioid requirements when pre-
aforementioned studies have demonstrated that scribing, for example, patient-controlled analge-
in patients on MMT who receive liver transplants sia for liver transplant patients. To date there have
substance relapse is rare and survival is similar to been no studies evaluating patient controlled
patients not receiving MMT [53, 56]. We there- analgesia dosing regimens in liver transplant
fore think that patients receiving MMT should patients.
not be excluded from transplant eligibility solely
based on the fact they are on MMT.
Epidural Analgesia

Intraoperative Analgesia An area of substantial controversy in liver trans-


plant anesthesia is the perioperative placement
Perioperative analgesia is important to alleviate and use of thoracic epidural catheters. Decreased
suffering as well as decrease potentially harmful synthetic function in the liver leads to decreased
physiologic consequences. Numerous studies coagulation factors and coexisting renal disease
have assessed the physiologic benefits of analge- may cause platelet dysfunction both affecting
sia including decreased risk of DVT, decreased hemostasis and bleeding. Several studies have
risk of developing chronic pain, and decreased been published that argue for and against
514 P. Weyker et al.

p­ lacement of epidural catheters for liver trans- using a long acting local anesthetic such as bupi-
plant recipients. Trzebicki et al. recounted the use vacaine 0.25–0.5%.
of thoracic epidural catheters in liver transplant
recipients over the course of 10 years. Only
patients with INR < 1.5, aPTT <45, and platelets Postoperative Analgesia
>70 were included. During the 10-year time
period 24% of 67 patients undergoing liver trans- One of the key goals of providing adequate post-­
plantation received a thoracic epidural. The operative analgesia is to minimize physiologic
authors concluded that placement of preoperative complications while maximizing patient comfort.
thoracic epidural allowed early extubation: 84% Multimodal approaches to pain management are
of patients who had an epidural were extubated in an attractive option to reduce the side effects
the operating room. The epidurals were removed accompanied by pharmacologic interventions but
on postoperative day 5 in most patients after nor- also to decrease the metabolic demand on the
malization of coagulation studies and platelet newly transplanted liver. Thus, even though par-
levels [63]. However, Fazakas et al. suggest that enteral opioids continue to be the mainstay of
although complications are rare, when they do acute postoperative pain management, ideally
occur they exceed the benefits provided by anal- analgesia should include a three-pronged
gesia [64]. The most challenging piece to the approach:
puzzle is the unpredictability of normalization of
the coagulation factors and platelets in patients • Pharmacotherapeutic: Opioids and non-opioid
following transplantation. At this time there is adjuvants
not enough evidence to recommend the routine • Non-pharmacotherapeutic: Behavioral approaches
use of thoracic epidurals for perioperative liver and physical modalities
transplantation analgesia; however, it is certainly • Invasive interventions: Peripheral nerve
reasonable to consider a thoracic epidural for blocks, trigger point injections, acupuncture
intraoperative and postoperative analgesia in
carefully selected patients.
Pharmacotherapeutic

Local Infiltration To date there are no adequate postoperative anal-


gesia studies in liver transplantation to elucidate
Infiltration of the wound with subcutaneous local the most effective analgesic modality. As men-
anesthetic has been used for a long time to pro- tioned earlier, this population has decreased opi-
vide postoperative analgesia in large abdominal oid requirements. Though there is no consensus
incisions. This is potentially beneficial in liver statement, the most common practice is to admin-
transplant recipients who have large and poten- ister opioids via patient-controlled analgesia.
tially very painful incisions including subcostal NSAIDS are generally avoided in this population
incisions. Various combinations of injection of due to platelet dysfunction and defunct renal
surgical incisions with local anesthetics have function in patients with end-stage liver disease.
been studied [65–71] and the data has been con- Anticonvulsants like gabapentin and pregablin
flicting regarding the efficacy of this interven- can be used as adjuvants and even though these
tion. Some studies demonstrated a clear benefit agents have not been studied effectively in this
while others showed no effect [70, 71]. Local population, their use can be considered on an
infiltration has not yet been studied in liver trans- individual basis. Acetaminophen is generally
plant patients, however, given the minimal cost, avoided in the immediate post-transplant period,
lack of significant adverse effects [72], and the although once graft function is established and
possibility of opioid sparing and for preemptive hepatic metabolism normalizes acetaminophen is
analgesia, it is reasonable to infiltrate the wound probably safe and should be considered [73].
39  Pain Management in Liver Transplantation 515

Behavioral Approaches pathways is the gate control theory and the


release of endogenous opioids [77]. Though an
Studies have shown that high preoperative anxi- intricate description of this process is beyond the
ety scores correlate with postoperative dissatis- scope of this chapter, it is important to note that
faction with patient-controlled analgesia [74]. primary sensory afferents synapse in the dorsal
Keeping this in mind, psychosocial counseling root ganglion at each spinal level and send pro-
including teaching the patient coping skills and jections to the substantia gelatinosa of the dorsal
anxiety management through distraction, bio-­ horn. The substantia gelatinosa acts as the gate-
feedback, mindfulness therapy, and deep breath- keeper between the periphery and the higher
ing techniques are ideally started in the processing pathways [79, 80]. The electrical
preoperative setting. According to UNOS bylaws stimulation of large diameter afferent fibers
all patients undergoing liver transplantation must competes with smaller diameter afferent pain
have a psychosocial evaluation before transplan- fibers and inhibits transmission of noxious stim-
tation and this evaluation can be an opportunity uli from the first order to second order neurons
to include coping mechanisms to provide insight [79–81]. Activation of large afferent fibers also
into perioperative analgesic needs. leads to the release of GABA and glycine that
bind to receptor sites that inhibit second order
neurons [81–83]. TENS analgesia is also partly
Physical Modalities due to the release of endogenous opioids in
response to electrical stimulation [76]. Cerebral
Rehabilitative techniques focus on early func- spinal fluid concentrations of beta-endorphins,
tional restoration and generally improve the methionine, enkephalin, and dynorphin A are
patient’s global sense of well-being. Among oth- elevated in healthy patients after both high and
ers, one of the easily used modalities includes low frequency electrical stimulation [77, 84–86].
simple neuromodulation techniques such as Activation of opioid receptors in the dorsal root
transcutaneous electrical nerve stimulation ganglion inhibits voltage gated calcium channels
(TENS). The opioid sparing effect of TENS and opens potassium channels, decreasing neu-
therapy is of particular importance to patients ronal excitability [81]. Clinical studies have
with opioid intolerance or in patient populations shown a reduction in postoperative opioid
in whom opioid therapy is complicated due to requirement in patients receiving TENS therapy
impaired metabolism. There are no controlled compared to placebo TENS [87]. Studies look-
studies evaluating the efficacy of TENS therapy ing at postoperative TENS with PCA compared
in liver transplant patients in the perioperative to PCA alone in patients undergoing lower
phase. TENS therapy is a noninvasive, non-­ abdominal surgery found a 53% reduction of
pharmacological tool used in the management of morphine requirements in patients receiving
acute and chronic pain [75, 76]. It utilizes elec- mixed frequency TENS at 2 and 100 Hz com-
trical currents through surface electrodes to pared to PCA only group [76]. These findings
modulate central and peripheral pain pathways confirmed the opioid sparing effect of TENS
that ultimately result in decreased pain percep- therapy and showed a superior effect of mixed
tion [75, 76]. It was introduced to the medical frequency TENS (2 and 100 Hz) over low (2 Hz)
community in the late 1960s by Wall and Sweet or high (100 Hz) frequency TENS in reducing
in their sentinel article on TENS for pain man- postoperative opioid requirements [76, 77]. Not
agement [77, 78]. An interesting finding in this surprisingly, patients who received the TENS
paper which has since been replicated in clinical therapy also exhibited less nausea, dizziness,
trials is that TENS analgesia ranges minutes to and pruritis as compared to the PCA group [76].
an hour after termination of electrical stimula- None of the patients in the studies mentioned
tion in patients with pain [78]. The main mecha- above experienced any adverse effects from the
nisms involved in TENS modulation of the pain TENS treatments [76, 87].
516 P. Weyker et al.

Invasive Pain Interventions the utility of TAP blocks with a continuous cath-
eter in two civilian trauma patients, describing
The use of invasive pain interventions in the post-­ multiple perioperative benefits of the TAP block
operative phase should start prior to surgery by including excellent analgesia, rapid extubation,
considering preoperative epidural placement or early hospital discharge, as well as an alternative
spinal duramorph administration in carefully technique to central neuraxial anesthesia when
selected patients. Since there is no consensus in coagulopathy is present [91]. Though it is
this regard, institutional practices and individual- increasingly used in patients undergoing renal
ization of therapy is key. As mentioned earlier, transplantation, the TAP block has yet to gain
injecting the incision site with a long acting anes- popularity in the post-liver transplantation
thetic may have a role in preemptive analgesia patient and must be used with caution. Risks
with minimal adverse effects. associated with the TAP nerve block technique
Postoperative, if the pain is in a specific dis- should be considered prior to performing the
tribution, perineural injection of local anesthetic procedure including block failure, infection,
can often provide relief. Blocks to consider inadvertent intravascular local anesthetic injec-
include intercostal blocks or transverse abdomi- tion, and bowel perforation. Farooq and Carey
nis plane (TAP) injections or catheters using describe a case report of liver trauma with a
ultrasound guidance for correct visualization. blunt needle while performing the TAP block
TAP block provides analgesia for the skin, sub- [92]. In an attempt to minimize these risks,
cutaneous tissue and peritoneum, while addi- ultrasound-­guided placement by a skilled opera-
tional analgesia is required for visceral pain. tor is essential. The number of studies and
TAP block should therefore be used as a compo- included patients is still insufficient to reliably
nent of multimodal pain treatment. To minimize guide clinical practice. However, TAP blocks
complications, TAP blocks should only be per- may become a valuable tool to optimize analge-
formed by experienced practitioners using ultra- sia if used judiciously in select patients. Trigger
sonography. The aim of a TAP block is to deposit point injections and acupuncture may also be
local anesthetic in the plane between the internal considered in amenable patients with corrected
oblique and transverse abdominis muscles tar- coagulation states.
geting the spinal nerves in this plane. Sensory
innervation to the abdominal wall skin and mus-
cles up to the parietal peritoneum will be inter- Living Donor Hepatectomy
rupted. This plane contains the thoracolumbar
nerves originating from T6 to L1 spinal roots Pain control for living donor hepatectomy is
that supply sensation to the anterolateral abdom- more complex than analgesia for the recipient.
inal wall. These multiple mixed segmental Living liver donors are healthy and have only
nerves branch and communicate as they run very few comorbidities. Since the first living
through the lateral abdominal wall between donor hepatectomy was performed in the United
internal oblique and transverse abdominal (TA) States in 1989, the numbers gradually increased
muscles, within the TA fascial plane. The anal- during the 1990s with just over 500 being done
gesic efficacy of the TAP block compared to pla- in 2001. However, since 2001, that number has
cebo has been demonstrated in prospective decreased by half likely due to several donor
randomized trials of different surgical proce- deaths [93]. Given the elective nature of the pro-
dures such as abdominal surgery [88], hysterec- cedure, every precaution must be taken to mini-
tomy [89], or retropubic prostatectomy [90]. All mize morbidity, mortality and also pain and
these studies have found a superiority of the TAP suffering. A small retrospective study found that
block and a reduction of visual analogue scale patients who underwent a right lobe donor hepa-
scores and morphine consumption. More tectomy had significantly higher pain scores
recently a case report was published describing than patients who underwent major hepatic
39  Pain Management in Liver Transplantation 517

tumor resection [94]. It was postulated that pain more pruritis found in the intrathecal morphine
scores were higher due to length of procedure as group [99].
well as the fact that these patients had no pain Another alternative to neuraxial anesthesia
before surgery. Preoperatively, living donors that is increasingly utilized for patients undergo-
have normal coagulation factors and platelet ing major abdominal surgery and hepatectomy
function. Therefore, placement of thoracic epi- surgery is the placement of peripheral nerve cath-
durals for postoperative pain control is practiced eters to target the thoracolumbar nerves. Two
in many centers. A study by Siniscalchi et al. approaches are described and vary depending on
described a series of 30 donors who received surgical incision and include either subcostal
thoracic epidural catheters. The coagulation sta- TAP catheters (Fig. 39.1) or rectus sheath cathe-
tus and platelet counts of all patients in this ters (Fig. 39.2). Niraj et al. described the use of
study returned to acceptable levels by postoper- subcostal TAP catheters for patients undergoing
ative day number 4, allowing the removal of epi- partial hepatectomy and other major abdominal
dural catheters without any complications [95]. surgeries [100]. In this particular study, patients
Choi et al. describe a similar experience with with either a right subcostal incision (partial hep-
epidural placement in living donors. Of 360 liv- atectomy), bilateral subcostal incisions, or trans-
ing donors, 242 received epidural catheters pre- verse incisions (above the umbilicus) were
operatively. Catheters were removed in 177 of included. The authors compared subcostal TAP
these patients by postoperative day 3–4. None catheters to T7–T9 thoracic epidural catheters. In
of the patients in this series experienced epi- this particular study, there was no difference in
dural hematomas [96]. The most common pain scores with coughing (primary outcome).
adverse effects in all these studies were pruritis However, patients who received subcostal TAP
and nausea associated with epidural opioids. catheters used significantly more tramadol
Ozkardesler et al., reported a series of 100 liv- (400 mg vs. 200 mg, p = 0.002) [100]. It should
ing donor patients receiving thoracic epidurals. be noted that patients treated with epidural anal-
One of their patients suffered from a postdural gesia also received fentanyl 2 mcg/mL as part of
puncture headache [97]. Based on these studies, the epidural solution [100]. As initially described
it can be concluded that as long as coagulation by Hebbard et al., the subcostal oblique approach
status is followed closely postoperatively, place- to the TAP plane deposits local anesthetic in a
ment of thoracic epidurals for living donor hep- medial to lateral fashion and more reliably blocks
atectomy appears to be a safe and effective the higher abdominal (T8–T10) thoracolumbar
method of perioperative analgesia. nerves [101, 102] as compared to the traditional
Another commonly used mode of analgesia posterior approach to the TAP plane. Thus, the
for this patient population is the use intrathecal subcostal approach is more suitable for upper
morphine. A prospective double-blinded study abdominal surgery as compared to the posterior
of 40 donor hepatectomy patients compared TAP approach [103]. Rectus sheath blocks and/or
postoperative morphine consumption in patients catheters are an additional regional anesthetic
who received intrathecal morphine and fentanyl technique that can be employed to improve post-­
to patients receiving a placebo injection. operative pain control. At our institution, it is our
Patients who received intrathecal opioids used practice to place bilateral rectus sheath catheters
significantly less morphine postoperatively for our donor hepatectomy surgeries. In general,
[98]. Another randomized prospective study of the catheters are placed at the T8–T9 dermatomal
donor hepatectomy patients found similar level or at the mid-point of the midline incision.
results: patients who received preoperative It should be noted that variability in the course of
intrathecal morphine used significantly less these nerves (T6–T8) exist, such that the nerves
postoperative fentanyl via a patient-controlled may pierce through the rectus abdominis (RA) at
analgesia delivery system. Both groups have the costal margin without ever traveling between
similar adverse effect profiles with slightly the RA and the posterior rectus sheath. Similarly,
518 P. Weyker et al.

Superficial

EO

RA
IO

Lateral
Medial

TA

Peritoneum

Deep

Fig. 39.1 Using a linear transducer (12-3MHz, RA muscle. The dashed arrow demonstrates the needle
Amsterdam, Netherlands), the probe is placed along the adjustment to enter the TA plane. The needle is then
costal margin so that it sits parallel to the subcostal line. driven medial to lateral (black arrow) as local anesthetic is
The probe is initially placed close to the xiphoid process deposited in the TA plane. EO external oblique, IO inter-
to identify the RA muscle. Once the RA muscle is identi- nal oblique, TA transversus abdominus, RA rectus abdo-
fied, the probe is then moved laterally until the EO, IO, minus. Solid white arrow shows needle location for
and TA come into view. The solid white arrow demarcates depositing local between the RA and TA; white dashed
the initial point of injection to target the thoracolumbar arrow demonstrates needle adjustment to enter the TA
nerves as they enter the lateral border of the RA to travel plane
in the rectus sheath before piercing through to enter the

Superficial

RA
Lateral
Medial

TA
Peritoneum

Deep

Fig. 39.2 Using a linear transducer (12-3MHz, sheath. The lateral border of the RA is targeted not only
Amsterdam, Netherlands), the probe is placed in a trans- because the nerves enter the sheath laterally but also
verse view in the area overlying the abdominal to be anes- because the TA lies underneath the RA at this level,
thetized. The rectus abdominus muscle is identified. In a decreasing the risk of inadvertent peritoneal puncture. RA
medial to lateral direction, the needle is advanced until the rectus abdominus, TA transversus abdominus, solid white
tip is located between the posterior border of the rectus arrow demarcates needle trajectory
abdominus muscle and the posterior border of the rectus
39  Pain Management in Liver Transplantation 519

the T9–T12 nerves also pass through the lateral atric transplant patients not only blunts the neu-
portion of the RA and in certain situations, may roendocrine response to pain that decreases the
even pierce through the RA from this lateral edge risk of vascular thrombosis but also minimizes
[102]. These anatomic variants may account for the amount of intraoperative and postoperative
block or catheter failure when local anesthetic is opioids and other analgesics presented to the
deposited at the medial border of the RA newly transplanted liver [104].
muscle.

Chronic Pain
Pediatric Liver Transplantation
Studies evaluating the quality of life after liver
Pediatric pain management is particularly chal- transplantation show improvement of functional
lenging, as pediatric patients are often unable to status. Belle et al. showed that patient’s quality of
verbalize their pain or discomfort. While very life improved dramatically after transplantation
few analgesic studies have been conducted in [107]. Management of chronic pain in patients
this population, many authors believe that in who have undergone liver transplantation is simi-
pediatric transplant, similar to adult patients, a lar to management of other patients with chronic
combination of elevated endogenous meten- pain. Pain management physicians use a multi-
kephalins and other neuropeptides combined modal approach taking into account the patient’s
with attenuated sensory input from the dener- liver function, coexisting medical and psycho-
vated liver leads to a decreased need for opioids logical comorbidities.
postoperatively [104]. Not surprising, despite An area of specific interest for the chronic
this decreased need for postoperative narcotics, pain specialist is neuropathic pain. Management
cohort studies have shown that physicians are of these chronic pain patients is particularly
often undertreating postoperative pain in pediat- difficult during the perioperative transplant
ric patients [105]. Communication barriers, fears period, as they are often on stable regimens of
of oversedation, and prolonged intubations and opioids, antidepressants, and anticonvulsants
pediatric ICU stays are believed to be the main to control their pain. Both the tricyclic antide-
reasons causing ineffective pain control for the pressants (TCAs) and serotonin norepineph-
pediatric patient [106]. A cohort study by Sharek rine reuptake inhibitors (SNRIs) undergo
et al. illustrated that a multidisciplinary and mul- extensive hepatic biotransformation via phase I
tifaceted approach combining preoperative edu- reactions into metabolites excreted in the urine
cation of child and parents, immediate [108, 109]. While specific randomized control
postoperative consultations with pain medicine, trials of liver transplant recipients are lacking,
certified child life specialists, and child psychia- studies in patients with varying degree of
try services combined with intravenous mor- hepatic impairment have suggested that the
phine significantly decreases pain scores in dose of TCAs and venlafaxine are to be
pediatric liver transplant patients [106]. While decreased by up to 50% [109]. Duloxetine
randomized control studies are lacking, case should be avoided altogether given numerous
reports have described the use of caudal mor- case reports associated with hepatotoxicity and
phine for acute intraoperative and postoperative fulminant hepatic failure. As ­previously stated,
pain control in pediatric liver transplant patients. gabapentin appears to be safe without any need
As with all types of neuraxial anesthesia, coagu- for dose reductions if the patients has normal
lopathy is a major concern. If clotting factors, renal function. Nonetheless, despite normal
platelet levels and coagulation studies are nor- postoperative graft function, many pain physi-
mal, caudal morphine can be an excellent option cians will still avoid the use of antidepressants
for perioperative pain control in these patients as a treatment for neuropathic pain in liver
[104]. As in adults, neuraxial anesthesia in pedi- transplant recipients.
520 P. Weyker et al.

Conclusions 12. Crotty B, Watson KJ, Desmond PV, Mashford ML,


Wood LJ, Colman J, et al. Hepatic extraction of mor-
Management of pain in patients with liver dis- phine is impaired in cirrhosis. Eur J Clin Pharmacol.
ease raises special concerns. The choice of 1989;36(5):501–6.
appropriate analgesic agents requires a thor- 13. Chandok N, Watt KD. Pain management in the cir-
ough understanding of their pharmacokinetic rhotic patient: the clinical challenge. Mayo Clin
Proc. 2010;85(5):451–8.
and side effect profiles. Hepatic metabolism 14. Nilsson MI, Meresaar U, Anggard E. Clinical phar-
complicates the use of intravenous and oral macokinetics of methadone. Acta Anaesthesiol
analgesics, while the coagulopathy seen with Scand Suppl. 1982;74:66–9.
liver disease limits the use of regional tech- 15. Murphy EJ. Acute pain management pharmacology
for the patient with concurrent renal or hepatic dis-
niques in many patients. However, a multi- ease. Anaesth Intensive Care. 2005;33(3):311–22.
modal pain therapy should aim to minimize 16. Novick DM, Kreek MJ, Arns PA, Lau LL, Yancovitz
the pain associated with liver transplantation SR, Gelb AM. Effect of severe alcoholic liver dis-
or resection and ideally renders the patient ease on the disposition of methadone in maintenance
patients. Alcohol Clin Exp Res. 1985;9(4):349–54.
pain free. 17. Kreek MJ, Bencsath FA, Field FH. Effects of liver
disease on urinary excretion of methadone and
metabolites in maintenance patients: quantitation by
direct probe chemical ionization mass spectrometry.
References Biomed Mass Spectrom. 1980;7(9):385–95.
18. Trescot AM, Datta S, Lee M, Hansen H. Opioid
1. O’Leary JG, Lepe R, Davis GL. Indications pharmacology. Pain Physician. 2008;11(2
for liver transplantation. Gastroenterology. Suppl):S133–53.
2008;134(6):1764–76. 19. Wright AW, Mather LE, Smith MT.
2. Smith HS. Opioid metabolism. Mayo Clin Proc. Hydromorphone-3-­
­ glucuronide: a more potent
2009;84(7):613–24. neuro-excitant than its structural analogue, mor-
3. McLean AJ, Morgan DJ. Clinical pharmacokinetics phine-3-glucuronide. Life Sci. 2001;69(4):409–20.
in patients with liver disease. Clin Pharmacokinet. 20. Smith MT. Neuroexcitatory effects of morphine
1991;21(1):42–69. and hydromorphone: evidence implicating the
4. Verbeeck RK, Horsmans Y. Effect of hepatic insuffi- 3-­glucuronide metabolites. Clin Exp Pharmacol
ciency on pharmacokinetics and drug dosing. Pharm Physiol. 2000;27(7):524–8.
World Sci. 1998;20(5):183–92. 21. Lee MA, Leng ME, Tiernan EJ. Retrospective study
5. Howden CW, Birnie GG, Brodie MJ. Drug of the use of hydromorphone in palliative care
metabolism in liver disease. Pharmacol Ther. patients with normal and abnormal urea and creati-
1989;40(3):439–74. nine. Palliat Med. 2001;15(1):26–34.
6. Tegeder I, Lotsch J, Geisslinger G. Pharmacokinetics 22. Paix A, Coleman A, Lees J, Grigson J, Brooksbank
of opioids in liver disease. Clin Pharmacokinet. M, Thorne D, et al. Subcutaneous fentanyl and
1999;37(1):17–40. sufentanil infusion substitution for morphine
7. Hoyumpa AM, Schenker S. Is glucuronidation truly intolerance in cancer pain management. Pain.
preserved in patients with liver disease? Hepatology. 1995;63(2):263–9.
1991;13(4):786–95. 23. Murphy MR, Hug CC Jr, McClain DA. Dose-­
8. Wood M, Wood AJJ. Drugs and anesthesia: phar- independent pharmacokinetics of fentanyl.
macology for anesthesiologists. 2nd ed. Baltimore: Anesthesiology. 1983;59(6):537–40.
Williams & Wilkins; 1990. 24. Wiesner G, Taeger K, Peter K. Serum protein binding
9. Andersen G, Christrup LL, Sjogren P, Hansen SH, of fentanyl. The effect of postoperative acute phase
Jensen NH. Changing M3G/M6G ratios and phar- reaction with elevated alpha 1-acid glycoprotein and
macodynamics in a cancer patient during long-­ methodologic problems in determination by equilib-
term morphine treatment. J Pain Symptom Manag. rium dialysis. Anaesthesist. 1996;45(4):323–9.
2002;23(2):161–4. 25. Feierman DE, Lasker JM. Metabolism of fentanyl,
10. Mazoit JX, Sandouk P, Zetlaoui P, Scherrmann a syntheticopioidanalgesic, byhumanlivermicro-
JM. Pharmacokinetics of unchanged morphine somes. Role of CYP3A4. Drug Metab Dispos.
in normal and cirrhotic subjects. Anesth Analg. 1996;24(9):932–9.
1987;66(4):293–8. 26. Haberer JP, Schoeffler P, Couderc E, Duvaldestin
11. Hasselstrom J, Eriksson S, Persson A, Rane P. Fentanyl pharmacokinetics in anaesthe-
A, Svensson JO, Sawe J. The metabolism and tized patients with cirrhosis. Br J Anaesth.
bioavailability of morphine in patients with 1982;54(12):1267–70.
severe liver cirrhosis. Br J Clin Pharmacol. 27. Hudson RJ, Thomson IR, Cannon JE, Friesen
1990;29(3):289–97. RM, Meatherall RC. Pharmacokinetics of fentanyl
39  Pain Management in Liver Transplantation 521

in patients undergoing abdominal aortic surgery. 45. Enomoto Y, Kudo T, Saito T, Hori T, Kaneko M,
Anesthesiology. 1986;64(3):334–8. Matsui A, et al. Prolonged use of dexmedetomidine
28. Cone EJ, Gorodetzky CW, Yousefnejad D, Buchwald in an infant with respiratory failure following liv-
WF, Johnson RE. The metabolism and excretion ing donor liver transplantation. Paediatr Anaesth.
of buprenorphine in humans. Drug Metab Dispos. 2006;16(12):1285–8.
1984;12(5):577–81. 46. Guinter JR, Kristeller JL. Prolonged infusions of
29. Kress HG. Clinical update on the pharmacology, dexmedetomidine in critically ill patients. Am J
efficacy and safety of transdermal buprenorphine. Health Syst Pharm. 2010;67(15):1246–53.
Eur J Pain. 2009;13(3):219–30. 47. Lin TF, Yeh YC, Lin FS, Wang YP, Lin CJ, Sun WZ,
30. Heit HA, Gourlay DL. Buprenorphine: new tricks et al. Effect of combining dexmedetomidine and
with an old molecule for pain management. Clin J morphine for intravenous patient-controlled analge-
Pain. 2008;24(2):93–7. sia. Br J Anaesth. 2009;102(1):117–22.
31. Sadee W, Rosenbaum JS, Herz A. Buprenorphine: 48. Aho MS, Erkola OA, Scheinin H, Lehtinen AM,
differential interaction with opiate receptor subtypes Korttila KT. Effect of intravenously administered
in vivo. J Pharmacol Exp Ther. 1982;223(1):157–62. dexmedetomidine on pain after laparoscopic tubal
32. Tyers MB. A classification of opiate receptors that ligation. Anesth Analg. 1991;73(2):112–8.
mediate antinociception in animals. Br J Pharmacol. 49. Dauri M, Faria S, Gatti A, Celidonio L, Carpenedo R,
1980;69(3):503–12. Sabato AF. Gabapentin and pregabalin for the acute
33. Virk MS, Arttamangkul S, Birdsong WT, Williams post-operative pain management. A systematic-­
JT. Buprenorphine is a weak partial agonist that narrative review of the recent clinical evidences.
inhibits opioid receptor desensitization. J Neurosci. Curr Drug Targets. 2009;10(8):716–33.
2009;29(22):7341–8. 50. Goa KL, Sorkin EM. Gabapentin. A review of its
34. Boas RA, Villiger JW. Clinical actions of fentanyl pharmacological properties and clinical potential in
and buprenorphine. The significance of receptor epilepsy. Drugs. 1993;46(3):409–27.
binding. Br J Anaesth. 1985;57(2):192–6. 51. Roth K, Lynn J, Zhong Z, Borum M, Dawson
35. Coursin DB, Maccioli GA. Dexmedetomidine. Curr NV. Dying with end stage liver disease with cir-
Opin Crit Care. 2001;7(4):221–6. rhosis: insights from SUPPORT. Study to under-
36. Scheinin H, Virtanen R, MacDonald E, Lammintausta stand prognoses and preferences for outcomes and
R, Scheinin M. Medetomidine—a novel alpha risks of treat-ment. J Am Geriatr Soc. 2000;48(5
2-adrenoceptor agonist: a review of its pharmaco- Suppl):S122–30.
dynamic effects. Prog Neuro-Psychopharmacol Biol 52. Weinrieb RM, Barnett R, Lynch KG, DePiano M,
Psychiatry. 1989;13(5):635–51. Atanda A, Olthoff KM. A matched comparison study
37. Mantz J, Josserand J, Hamada S. Dexmedetomidine: of medical and psychiatric complications and anes-
new insights. Eur J Anaesthesiol. 2011;28(1):3–6. thesia and analgesia requirements in methadone-­
38. Khan ZP, Ferguson CN, Jones RM. Alpha-2 and imid- maintained liver transplant recipients. Liver Transpl.
azoline receptor agonists. Their pharmacology and 2004;10(1):97–106.
therapeutic role. Anaesthesia. 1999;54(2):146–65. 53. Kanchana TP, Kaul V, Manzarbeitia C, Reich DJ,
39. Langer SZ, Arbilla S. Presynaptic receptors and Hails KC, Munoz SJ, et al. Liver transplantation for
modulation of the release of noradrenaline, dopa- patients on methadone maintenance. Liver Transpl.
mine and GABA. Postgrad Med J. 1981;57(Suppl 2002;8(9):778–82.
1):18–29. 54. Liu LU, Schiano TD, Lau N, O’Rourke M, Min
40. Hayashi Y, Maze M. Alpha 2 adrenoceptor agonists AD, Sigal SH, et al. Survival and risk of recidi-
and anaesthesia. Br J Anaesth. 1993;71(1):108–18. vism in methadone-dependent patients under-
41. Hayashi Y, Rabin BC, Guo TZ, Maze M. Role of going liver transplantation. Am J Transplant.
pertussis toxin-sensitive G-proteins in the analgesic 2003;3(10):1273–7.
and anesthetic actions of alpha 2-adrenergic agonists 55. Hancock MM, Prosser CC, Ransibrahmanakul K,
in the rat. Anesthesiology. 1995;83(4):816–22. Lester L, Craemer E, Bourgeois JA, et al. Liver
42. Guo TZ, Jiang JY, Buttermann AE, Maze transplant and hepatitis C in methadone m­ aintenance
M. Dexmedetomidine injection into the locus ceru- therapy: a case report. Subst Abuse Treat Prev
leus produces antinociception. Anesthesiology. Policy. 2007;2:5.
1996;84(4):873–81. 56. Jiao M, Greanya ED, Haque M, Yoshida EM, Soos
43. Pertovaara A, Kauppila T, Jyvasjarvi E, Kalso JG. Methadone maintenance therapy in liver trans-
E. Involvement of supraspinal and spinal seg- plantation. Prog Transplant. 2010;20(3):209–14;
mental alpha-2-adrenergic mechanisms in quiz 15.
the medetomidine-­ induced antinociception. 57. Campiglia L, Consales G, De Gaudio AR. Pre-­
Neuroscience. 1991;44(3):705–14. emptive analgesia for postoperative pain control: a
44. Karol MD. Pharmacokinetics and interaction phar- review. Clin Drug Investig. 2010;30(Suppl 2):15–26.
macodynamics of dexmedetomidine in humans. 58. Katz J, McCartney CJ. Current status of pre-
Bailleres Best Pract Res Clin Anesthesiol. emptive analgesia. Curr Opin Anaesthesiol.
2000;14(2):261–9. 2002;15(4):435–41.
522 P. Weyker et al.

59. Chen JP, Jawan B, Chen CL, Wang CH, Cheng KW, 72. Brower MC, Johnson ME. Adverse effects of local
Wang CC, et al. Comparison of postoperative mor- anesthetic infiltration on wound healing. Reg Anesth
phine requirements in healthy living liver donors, Pain Med. 2003;28(3):233–40.
patients with hepatocellular carcinoma undergoing 73. Venkataramanan R, Habucky K, Burckart GJ,
partial hepatectomy, and liver transplant recipients. Ptachcinski RJ. Clinical pharmacokinetics in
Transplant Proc. 2010;42(3):701–2. organ transplant patients. Clin Pharmacokinet.
60. Donovan KL, Janicki PK, Striepe VI, Stoica C, 1989;16(3):134–61.
Franks WT, Pinson CW. Decreased patient anal- 74. Gil KM, Ginsberg B, Muir M, Sykes D, Williams
gesic requirements after liver transplantation and DA. Patient-controlled analgesia in postoperative
associated neuropeptide levels. Transplantation. pain: the relation of psychological factors to pain
1997;63(10):1423–9. and analgesic use. Clin J Pain. 1990;6(2):137–42.
61. Eisenach JC, Plevak DJ, Van Dyke RA, Southorn 75. DeSantana JM, Walsh DM, Vance C, Rakel BA,
PA, Danielson DR, Krom RA, et al. Comparison Sluka KA. Effectiveness of transcutaneous electrical
of analgesic requirements after liver transplan- nerve stimulation for treatment of hyperalgesia and
tation and cholecystectomy. Mayo Clin Proc. pain. Curr Rheumatol Rep. 2008;10(6):492–9.
1989;64(3):356–9. 76. Hamza MA, White PF, Ahmed HE, Ghoname
62. Moretti EW, Robertson KM, Tuttle-Newhall JE, EA. Effect of the frequency of transcutaneous
Clavien PA, Gan TJ. Orthotopic liver transplant electrical nerve stimulation on the postoperative
patients require less postoperative morphine than do opioid analgesic requirement and recovery profile.
patients undergoing hepatic resection. J Clin Anesth. Anesthesiology. 1999;91(5):1232–8.
2002;14(6):416–20. 77. Sluka KA, Walsh D. Transcutaneous electrical nerve
63. Trzebicki J, Nicinska B, Blaszczyk B, Jureczko L, stimulation: basic science mechanisms and clinical
Kolacz M, Pacholczyk M, et al. Thoracic epidural effectiveness. J Pain. 2003;4(3):109–21.
analgesia in anaesthesia for liver transplantation: 78. Wall PD, Sweet WH. Temporary abolition of pain in
the 10-year experience of a single centre. Ann man. Science. 1967;155(758):108–9.
Transplant. 2010;15(2):35–9. 79. Melzack R, Wall PD. Pain mechanisms: a new the-
64. Fazakas J, Toth S, Fule B, Smudla A, Mandli T, ory. Science. 1965;150(699):971–9.
Radnai M, et al. Epidural anesthesia? No of course. 80. Wright A, Sluka KA. Nonpharmacological treat-
Transplant Proc. 2008;40(4):1216–7. ments for musculoskeletal pain. Clin J Pain.
65. Ejlersen E, Andersen HB, Eliasen K, Mogensen 2001;17(1):33–46.
T. A comparison between preincisional and postin- 81. DeLeo JA. Basic science of pain. J Bone Joint Surg
cisional lidocaine infiltration and postoperative pain. Am. 2006;88(Suppl 2):58–62.
Anesth Analg. 1992;74(4):495–8. 82. Maeda Y, Lisi TL, Vance CG, Sluka KA. Release
66. Hashemi K, Middleton MD. Subcutaneous bupiva- of GABA and activation of GABA(A) in the spinal
caine for postoperative analgesia after herniorrha- cord mediates the effects of TENS in rats. Brain Res.
phy. Ann R Coll Surg Engl. 1983;65(1):38–9. 2007;1136(1):43–50.
67. Randall JK, Goede A, Morgan-Warren P, Middleton 83. Kalra A, Urban MO, Sluka KA. Blockade of opioid
SB. Randomized clinical trial of the influence of receptors in rostral ventral medulla prevents anti-
local subcutaneous infiltration vs subcutaneous and hyperalgesia produced by transcutaneous electrical
deep infiltration of local anaesthetic on pain after nerve stimulation (TENS). J Pharmacol Exp Ther.
appendicectomy. Color Dis. 2010;12(5):477–9. 2001;298(1):257–63.
68. Ausems ME, Hulsewe KW, Hooymans PM, 84. Han JS, Chen XH, Sun SL, Xu XJ, Yuan Y, Yan
Hoofwijk AG. Postoperative analgesia requirements SC, et al. Effect of low- and high-frequency
at home after inguinal hernia repair: effects of wound TENS on Metenkephalin-Arg-Phe and dynorphin
infiltration on postoperative pain. Anaesthesia. a immunoreactivity in human lumbar CSF. Pain.
2007;62(4):325–31. 1991;47(3):295–8.
69. Lohsiriwat V, Lert-akyamanee N, Rushatamukayanunt 85. Hughes GS Jr, Lichstein PR, Whitlock D, Harker
W. Efficacy of pre-incisional bupivacaine infiltration C. Response of plasma beta-endorphins to transcu-
on postoperative pain relief after appendectomy: pro- taneous electrical nerve stimulation in healthy sub-
spective double-blind randomized trial. World J Surg. jects. Phys Ther. 1984;64(7):1062–6.
2004;28(10):947–50. 86. Salar G, Job I, Mingrino S, Bosio A, Trabucchi
70. Kuan YM, Smith S, Miles C, Grigg M. Effectiveness M. Effect of transcutaneous electrotherapy on CSF
of intra-operative wound infiltration with beta-endorphin content in patients without pain
long-acting local anaesthetic. ANZ J Surg. problems. Pain. 1981;10(2):169–72.
2002;72(1):18–20. 87. Erdogan M, Erdogan A, Erbil N, Karakaya HK,
71. Victory RA, Gajraj NM, Van Elstraete A, Pace NA, Demircan A. Prospective, randomized, placebo-­
Johnson ER, White PF. Effect of preincision versus controlled study of the effect of TENS on postthora-
postincision infiltration with bupivacaine on postop- cotomy pain and pulmonary function. World J Surg.
erative pain. J Clin Anesth. 1995;7(3):192–6. 2005;29(12):1563–70.
39  Pain Management in Liver Transplantation 523

88. McDonnell JG, O’Donnell B, Curley G, Heffernan 99. Ko JS, Choi SJ, Gwak MS, Kim GS, Ahn HJ, Kim
A, Power C, Laffey JG. The analgesic efficacy of JA, et al. Intrathecal morphine combined with intra-
transversus abdominis plane block after abdominal venous patient-controlled analgesia is an effec-
surgery: a prospective randomized controlled trial. tive and safe method for immediate postoperative
Anesth Analg. 2007;104(1):193–7. pain control in live liver donors. Liver Transpl.
89. Carney J, McDonnell JG, Ochana A, Bhinder R, 2009;15(4):381–9.
Laffey JG. The transversus abdominis plane block 100. Niraj G, Kelkar A, Jeyapalan I, et al. Comparison
provides effective postoperative analgesia in patients of analgesic efficacy of subcostal transversus
undergoing total abdominal hysterectomy. Anesth abdominis plane blocks with epidural analgesia
Analg. 2008;107(6):2056–60. following upper abdominal surgery. Anaesthesia.
90. O’Donnell BD, McDonnell JG, McShane AJ. The 2011;66(6):465–71.
transversus abdominis plane (TAP) block in open 101. Hebbard P. Subcostal transversus abdominis plane
retropubic prostatectomy. Reg Anesth Pain Med. block under ultrasound guidance. Anesth Analg.
2006;31(1):91. 2008;106(2):674–5; author reply 675.
91. Allcock E, Spencer E, Frazer R, Applegate G, 102. Hebbard PD, Barrington MJ, Vasey C. Ultrasound-­
Buckenmaier C III. Continuous transversus abdomi- guided continuous oblique subcostal transversus
nis plane (TAP) block catheters in a combat surgical abdominis plane blockade: description of anatomy
environment. Pain Med. 2010;11(9):1426–9. and clinical technique. Reg Anesth Pain Med.
92. Farooq M, Carey M. A case of liver trauma with a 2010;35(5):436–41.
blunt regional anesthesia needle while performing 103. Lee TH, Barrington MJ, Tran TM, Wong D, Hebbard
transversus abdominis plane block. Reg Anesth Pain PD. Comparison of extent of sensory block follow-
Med. 2008;33(3):274–5. ing posterior and subcostal approaches to ultrasound-­
93. Merion RM. Current status and future of liver trans- guided transversus abdominis plane block. Anaesth
plantation. Semin Liver Dis. 2010;30(4):411–21. Intensive Care. 2010;38(3):452–60.
94. Cywinski JB, Parker BM, Xu M, Irefin SA. A com- 104. Kim TW, Harbott M. The use of caudal morphine
parison of postoperative pain control in patients after for pediatric liver transplantation. Anesth Analg.
right lobe donor hepatectomy and major hepatic 2004;99(2):373–4; table of contents.
resection for tumor. Anesth Analg. 2004;99(6):1747– 105. Howard RF. Current status of pain management in
52; table of contents. children. JAMA. 2003;290(18):2464–9.
95. Siniscalchi A, Begliomini B, De Pietri L, Braglia V, 106. Sharek PJ, Wayman K, Lin E, Strichartz D,
Gazzi M, Masetti M, et al. Increased prothrombin Sentivany-Collins S, Good J, et al. Improved pain
time and platelet counts in living donor right hepa- management in pediatric postoperative liver trans-
tectomy: implications for epidural anesthesia. Liver plant patients using parental education and non-­
Transpl. 2004;10(9):1144–9. pharmacologic interventions. Pediatr Transplant.
96. Choi SJ, Gwak MS, Ko JS, Kim GS, Ahn HJ, Yang 2006;10(2):172–7.
M, et al. The changes in coagulation profile and 107. Belle SH, Porayko MK, Hoofnagle JH, Lake JR,
epidural catheter safety for living liver donors: a Zetterman RK. Changes in quality of life after liver
report on 6 years of our experience. Liver Transpl. transplantation among adults. National Institute
2007;13(1):62–70. of Diabetes and Digestive and Kidney Diseases
97. Ozkardesler S, Ozzeybek D, Alaygut E, Unek T, (NIDDK) Liver Transplantation Database (LTD).
Akan M, Astarcioglu H, et al. Anesthesia-related Liver Transpl Surg. 1997;3(2):93–104.
complications in living liver donors: the experience 108. Haanpaa ML, Gourlay GK, Kent JL, Miaskowski C,
from one center and the reporting of one death. Am Raja SN, Schmader KE, et al. Treatment consider-
J Transplant. 2008;8(10):2106–10. ations for patients with neuropathic pain and other
98. Roy JD, Massicotte L, Sassine MP, Seal RF, Roy medical comorbidities. Mayo Clin Proc. 2010;85(3
A. A comparison of intrathecal morphine/fentanyl Suppl):S15–25.
and patient-controlled analgesia with patient-­ 109. Jensen TS, Madsen CS, Finnerup NB. Pharmacology
controlled analgesia alone for analgesia after liver and treatment of neuropathic pains. Curr Opin
resection. Anesth Analg. 2006;103(4):990–4. Neurol. 2009;22(5):467–74.
Index

A circulatory symptoms of, 48–49


Abdominal compartment syndrome, 449 classifications for, 42, 43
Abdominal incision and exposure, 124 clinical features and general management, 47–48
Abdominal surgery, liver disease, 395 coagulation, 56
ABO incompatibility, marginal donors and, 264 complex nature, 48
Acetaminophen, 514 diagnosis and multimodality monitoring, 311
Acetazolamide, 472 etiologies, 43
Achilles heel, 492 gastroenterology, 51
Acidemia, 248, 276 ICP monitoring, 311–312
Acquired disease, 298–299 idiosyncratic drug reaction, 45
Acquired hepatocerebral degeneration (AHD), 325 immunity and bacteremia, 52–53
Actinetobacter baumanii (AB), carbapenem-resistant, inflammation within brain, 309
462 intracranial hypertension
Activated partial thromboplastin time (APTT), 181, 195, pathophysiology, 309–311
398, 399 ischemic hepatitis, 46
Acute cellular rejection (ACR), 431, 433, 434 jugular bulb oximetry, 312
Acute Dialysis Quality Initiative (ADQI) Group, 235, liver transplantation, 55, 57, 58
270, 445 malignancy, 46
Acute interstitial nephritis (AIN), 447 metabolic disorder, 47
Acute kidney injury (AKI), 53–56, 234, 270, 351 metabolisms, 51–52
abdominal compartment syndrome, 449 negative paracetamol level, 47
agents associated with, 447 neurological monitoring, 138
calcineurin inhibitors, 447 neurological problems, 50–51
causes of, 446–449 non-invasive monitoring of ICP, 313
in cirrhosis, 270–275 nutrition, 51
definitions, 445–446 paracetamol overdose, 43
diagnosis, 446 prognosis of, 56–57
epidemiology, 445 respiratory complications, 49–50
hemolytic uremic syndrome, 448 seronegative, 46
infectious complications, 449 specialist center, 51
after liver transplantation, 418 transcranial Doppler, 312–313
preoperative preparation, 402 transplantation in
RIFLE criteria, 446 patient population, 257–258
thrombotic thrombocytopenic purpura, 448 pre-emptive total hepatectomy, 259
Acute Kidney Injury Network (AKIN), 235, 270, 418, preoperative considerations, 258–259
446 recovery, 265
Acute liver failure (ALF), 257–259, 265, 394 vascular insults, 46
AASLD recommendations for, 493 viral hepatitis, 44
acute kidney injury, 53 Acute pain, 515
adrenal insufficiency, 49 management, 508
autoimmune hepatitis, 47 morphine, 519
cerebral blood flow and systemic inflammation, 309 postoperative management, 514
cerebral edema in, 307 treatment, 509, 512
cerebral perfusion pressure, 311 Acute rejection, features, 433

© Springer International Publishing AG, part of Springer Nature 2018 525


G. Wagener (ed.), Liver Anesthesiology and Critical Care Medicine,
https://doi.org/10.1007/978-3-319-64298-7
526 Index

Acute respiratory distress syndrome (ARDS), 273, 314, Anesthetic agents, 351
418, 475–477 Anesthetic induction, 407
ventilator management, 421–422 Anesthetic management
Acute tubular necrosis (ATN), 446, 447 infants and toddlers, 227
Adaptive immune system, 11, 12 liver disease, 405–406
Adenomas, 341 pre-teenager, 229
Adrenal insufficiency, 273 teenager, 229
Adult-to-adult living donor liver transplantation Anesthetic volatile agents, 353
(A2ALL), 212, 486, 491–493 Angiotensinogen, 16
AHD, see Acquired hepatocerebral degeneration (AHD) Anidulafungin, 464
AKI, see Acute kidney injury (AKI) Antibiotic prophylaxis
Alanine aminotransferase (ALT), 79, 351 LDLT, 489–490
Alcohol, 12, 13 role of, 457
Alcohol dehydrogenase (ADH), 12 Antibody mediated rejection (AMR), 434
Alcohol septal ablation, 289 Antibody-based therapies, 439
Alcoholic liver disease, 23 monoclonal antibodies, 440
Alcohol-induced cirrhosis, 403 polyclonal antibodies, 439–440
ALF, see Acute liver failure (ALF) Antidiuretic hormone (ADH), 275
Allograft rejection mechanisms, 433, 434 Anti-epileptic drugs (AEDs), 14
Allograft tolerance, 12 Antifibrinolytics, 262, 380
Alpha-1 antitrypsin deficiency, 297 Antifibrotic agents, targets for, 32–34
Altered mental status, 424 Antifibrotic therapies, 32, 33
Alvimopan, 77 Antigen presenting cells (APCs), 11, 433
Amebic abscess, 340 Anti-lymphocyte globulin, 439
Antimetabolites, 437
adverse effect, 438
American Association for the Study of Liver Diseases clinical use, 438
(AASLD), 493 method of action, 438
American Society of Anesthesiologist (ASA) score, 498 pharmacokinetics and metabolism, 438
American Society of Transplant Surgeons (ASTS), 105 therapeutic drug monitoring, 438
Amino acids, 401 Antimicrobial therapy, 474
Amiodarone, 417 Antimycotic prophylaxis, 457
Amlodipine, 417 Antithrombin (AT), 176
Ammonia, 14, 50, 136, 308 Anti-thymocyte globulin (ATG), 439, 456
Ammonia-reducing strategy, 317 Aortic valve replacement (AVR), 397
Amphotericin B, 463 Aprotinin, 96, 199
Analgesia, 513–516 Aprotinine, 166
epidural (see Epidural analgesia) ARDS, see Acute respiratory distress syndrome (ARDS)
intraoperative, 513 Argyle infant feeding tube, 503
post-operative, 490, 514 Arrhythmia, 289
behavioral approaches, 515 Arterial pressure, invasive, 354
invasive pain interventions, 516 Arterialization of the portal vein, 131
pharmacotherapeutics, 514 Artificial hepatic support, 227
physical modalities, 515 Ascites, 27, 397, 418
sedation and, 422, 425 Aspartate aminotransferase (AST), 79
Analgesia first (A-1) approach, 422 Aspergillosis, 464
Anasarca ascites, 294 Aspergillus fumigates, 464
Anemia, 398 Aspiration pneumonitis, 476
Anesthesia, 516 Associating Liver Partition and Portal Vein Ligation for
advances in, 92 Staged Hepatectomy (ALPPS), 343, 383
neuraxial (see Neuraxial anesthesia) Asterixis, 28
and perioperative care, 95 ATN, see Acute tubular necrosis (ATN)
Anesthesia-induced hepatitis (AIH), 405 ATP binding cassette carriers (ABC), 13
Anesthesiology Atracurium, 405
bleeding prevention, 200 Atrial fibrillation, 289, 417
early extubation, 151–152 Autoimmune disease, 297–298
intraoperative renal replacement therapy, 151 Autoimmune hepatitis, 47, 404, 441
pulmonary artery catheters, 149–150 Autonomic dysfunction, 295
transesophageal echocardiography, 150 Azathioprine, 437
viscoelastic test, 151 adverse effect, 438
Index 527

clinical use, 438 surgical and anesthesiological techniques, 200–201


method of action, 438 Bleeding time, 180
pharmacokinetics and metabolism, 438 Blood flow assessment, 141
therapeutic drug monitoring, 438 Blood products, 198
Blood stream infections (BSI), 458
Blood urea nitrogen (BUN), 401–402
B Blood volume distribution, 66
Bacteremia, 52 Blood–brain barrier (BBB), 308, 318, 510
Bacterial infections, risk factor, 455–456 Brain
Balloon valvuloplasty, 288 inflammation within, 309
Barbiturate coma, 320 death, 92–93
Barcelona Liver Clinic Liver Cancer Group (BCLC), 343 Brain edema, 322
Bariatric surgery, liver disease, 396 Bronchoalveolar lavage (BAL), 462, 474
Baso-lateral membrane, 7 Bronchoscopy, 474
β-Blockers, 285 Budd-Chiari syndrome (BCS), 6, 46, 301
Benign solid liver lesions, 341–342 Buprenorphine, 510–511
Benzodiazepine, 406, 422, 424
Berlin Definition, ARDS, 476
Beta-adrenergic signaling, 164 C
Beta-blocker, 350 CABG, see Coronary artery bypass grafting (CABG)
Bile duct anastomosis, 130 CAC, see Coronary artery calcification (CAC)
Bile leak, 428, 502 CAD, see Coronary artery disease (CAD)
biliary complications, 492 Cadaveric liver grafts, 104
clinical presentation, 502 Calcineurin inhibitors (CNIs), 418, 435
LDLT complication, 492 acute kidney injury, 447
management, 492, 504 adverse effects, 436
methylene blue test, 504 clinical use, 436
pathophysiology, 502, 503 method of action, 435
post-hepatectomy, 503 pharmacokinetics and metabolism, 435
signs and symptoms, 492 therapeutic drug monitoring, 436
surgical technique, 492 Calcium channel blockers, 417
Bile, production, 451, 452 Calcium influx, 212
Biliary atresia, 224 Campath (alemtuzumab), 440
Biliary cirrhosis, 297 Candida species, 462, 463
Biliary complications, 502, 504 C. albicans, 457, 462
adverse effects, 502 C. glabrata, 457, 462
assessment, 428 C. krusei, 462
incidence, 502 Carbapenem-resistant Actinetobacter baumanii (CRAB), 462
pediatric liver transplantation, 230 Carbapenem-resistent Klebsiella pneumonia (CRKP),
risk factor, 502 461–462
Biliary leak, 373 Carbon monoxide, 164
Biliary obstruction, 79 Cardiac dysfunction, 282
Bilirubin, 81 Cardiac mesoderm, 4
Bioartificial livers, 501 Cardiac output, 139
Bio-availability, 12 Cardiac transplantation, in presence of liver failure, 239
Biopsy-proven steatosis, 104 Cardiomyopathy
Biotransformation, 508 cirrhotic, 282
Biphasic positive airway pressure (BiPAP), 473, 474 hypertrophic obstructive, 289
Bispectral (BIS) index, 138 Cardiopulmonary bypass (CPB), 396
Black-box warning, micafungin, 463 Cardiothoracic surgery, liver disease, 396
Bleeding during liver transplantation, 377, 420, 427 Cardiovacular risk assessment, 350–351
adverse effects, 201–202 Cardiovascular changes
blood products, 198–199 cardiac output, 165
causes of, 197–198 in end-stage liver disease, 281–283
fluid restriction, 200 extrinsic factors, 164
laboratory monitoring, 201 hemodynamic changes, 164
pharmacological agents, 199–200 inotropes and vasopressors, 165
prevention/treatment, 202 intrinsic factors, 163
prophylactic strategies, 198 pathogenic mechanisms, 164
rebalanced hemostasis, 196 physiological considerations, 163
528 Index

Cardiovascular system Chronic kidney disease (CKD), 270, 498


caval cross-clamping, 155 development of, 447, 448
response to liver transplantation, 415–417 incidence of, 448
Caspofungin, 464 Chronic liver disease, 394
Catabolism, 315 alcoholic liver disease, 23
Caudate lobe, 127 ascites, 27
Caval anastomosis, 128 cirrhotic cardiomyopathy, 29
end-to-side, 127 clinical presentation and complications, 26
hyperkalemic patient, 128 CTP scoring system, 31
lower, 128 epidemiology, 22
side to side, 122 esophageal varices, 28
Caval clamp trial, 156 etiologies, 22–23
Caval cross-clamping, 155 HBV, 24
cardiovascular system, 155–156 HCC, 30
gastrointestinal system, 157 HCV, 24
neurologic system, 157 Hepatic hydrothorax, 27
physiological effects of, 156 HPS, 28
pulmonary system, 156 HRS, 29
renal system, 156–157 MELD score, 31
Caval preservation technique, 94, 122 NAFLD, 23
Cavitron Ultrasonic Dissector (CUSA), 336 pathophysiology, 24–26
Cavitron Ultrasonic Tissue Aspirator (CUSA), 503 POPH, 29
Cavoportal hemitransposition (CPHT), 131 portal hypertension, 27
CCA, see Cholangiocarcinoma (CCA) portosystemic encephalopathy, 28
Ceftazidime/avibacatm, 462 prognosis, 31
Ceftopirole, MRSA, 459 PVT, 30
Centers for Disease Control and Prevention (CDC), 24 SBP, 27
Central neural activation, 164 synthetic dysfunction, 26
Central pontine myelinolysis (CPM), 137, 324, 419 treatment, 31
Central venous catheter (CVC), 490 Chronic pain, 509, 515, 519
Central venous pressure (CVP), 140, 150, 198, 248, 371, Chronic renal failure, chronic kidney disease (CKD), see
379, 500 Cirrhosis, 389, 394, 508
Central-line associated blood stream infections acute kidney injury in, 270, 272
(CLABSIs), 315 alcoholic liver disease, 23
Cephalosporines, 460 ascites, 27
Cerebral astrocytes, 14 biliary, 297
Cerebral blood flow (CBF), 260, 309 cirrhotic cardiomyopathy, 29–30
Cerebral cytotoxic edema, etiology, 307–309 clinical presentation and complications, 26
Cerebral edema, 308 CTP scoring system, 31
acute liver failure in, 307 epidemiology, 22
types of, 15 esophageal varices, 28
Cerebral ischemia, 313 etiologies, 22
Cerebral manifestation, hepatic disease with, 325 HBV, 24
Cerebral metabolic rate, 316 HCC, 30
Cerebral perfusion pressure (CPP), 160, 259, 311, 319 HCV, 24
Cerebrovascular stability, 259 hemodynamic changes in, 283
Chest X-ray, 490 hepatic hydrothorax, 27
Child-Pugh classification, 498 HPS, 28
Child-Turcotte-Pugh (CTP) score, 31, 74, 75, 111, 112, HRS, 29
392 MELD score, 31
CHLT, see Combined heart and liver transplantation mortality, 390
(CHLT) NAFLD, 23
Cholangiocarcinoma (CCA), 343 pathophysiology, 24
Cholangiocyte, 9 POPH, 29
Cholecystectomy, 128 portal hypertension, 27
laparoscopic vs. open, 396 portosystemic encephalopathy, 28
liver disease, 395 postoperative complications, 390
Cholestatic liver disease, 223 prevalence, 22
Chronic antiplatelet therapy, 351 procedure types, 390
Chronic cellular rejection, 431 prognosis, 31
Index 529

pulmonary function in patients with, 294 Coronary artery bypass surgery, 284
PVT, 30 Coronary artery calcification (CAC), 286
retrospective review, 390 Coronary artery disease (CAD), 283–284
SBP, 27 consequence of, 284–285
synthetic dysfunction, 26 coronary stent placement, 287–288
treatment, 31–34 dobutamine stress echocardiography, 285
Cirrhotic cardiomyopathy, 29, 282 invasive, diagnostic evaluation of, 286–287
Cisatracurium, 352, 405 management, 287
Citrate, 472 myocardial perfusion scan, 286
Citrate chelation, 419 preoperative evaluation, 285
CKD, see Chronic kidney disease (CKD) Coronary stent placement, 287
Clavien’s classification system, LDLT, 491, 492 Corticosteroids, 437
Clichy criteria, 57 adverse effects, 437
CLLT, see Combined lung–liver transplantation (CLLT) clinical use, 437
CNIs, see Calcineurin inhibitors (CNIs) method of action, 437
CNS dysfunction, 323 Couinaud segmental anatomy, 371
Coagulation Cox regression studies, 117
cascade and liver disease, 178 CPP, see Cerebral perfusion pressure (CPP)
inhibition and fibrinolysis, 176 CRAB, see Carbapenem-resistant Actinetobacter
liver transplantation, 420 baumanii (CRAB)
management, viscoelastic testing for, 150 Cranial computed tomography, 138
pathophysiology of, 197 Creatinine, 112
Coagulopathy, 137, 261 Critical illness related corticosteroid insufficiency
liver disease, 398, 399 (CIRCI), 48
liver transplantation, 427 CRRT, see continuous renal replacement therapy (CRRT)
perioperative correction, 399 Cryoglobulinemia, 299
Cold ischemia, 212 Cryoprecipitate (CP), 399
Cold ischemic time (CIT), 124, 208, 209 CTP score, see Child-Turcotte-Pugh (CTP) score
Combined heart and liver transplantation (CHLT), The Culture of Organs, 90
237–238 CVP, see Central venous pressure (CVP)
indication for, 238 Cyclosporine, 435
intra-operative management, 238–239 adverse effects, 436
postoperative course, 240 clinical use, 436
pre-operative evaluation, 238 method of action, 435
Combined liver kidney transplant (CLKT), 275 pharmacokinetics and metabolism, 435–436
Combined lung–liver transplantation (CLLT), 240–241 therapeutic drug monitoring, 436–437
indication for, 241 Cyclosporine A, 221
intra-operative management, 242 Cyst, hydatid, 340
post-operative management, 242–243 Cystic fibrosis, 295–297
pre-operative evaluation, 241 Cytoablative technique, 338
Compartment syndrome, 397 Cytochrome P450, 352
Computed tomography (CT) volumetry 3A4 (CYP3A4), 435
hepatectomy, 499 2E1 (CYP2E1), 13
LDLT, 486 substrates, inducers, inhibitors of, 13–14
Confusion Assessment Method for the ICU (CAM-ICU), Cytokine, 309
422, 423 Cytotoxic edema, 15, 307
Congenital heart disease, 227
Continuous positive airway pressure (CPAP), 473
Continuous renal replacement therapy (CRRT), 52, 151, D
235, 274, 277, 317 DAMPs, see Damage-associated-molecular patterns
Continuous venovenous hemodialysis (CVVHD), 249, (DAMPs)
274, 277, 453 Daptomycin
Continuous veno-venous hemofiltration (CVVH), 259, MRSA, 458
263 VRE, 459
Contrast-enhanced transthoracic echocardiograph, 301 DCD, see Donation after cardiac death (DCD)
Conventional laboratory tests, 135 1-Deamino-8-D-arginine vasopressin (DDAVP), 200
Conventional liver function test, 79–80, 141 Deceased donor liver transplantation (DDLT), 488
Conventional technique, of orthotopic liver Deep vein thrombosis (DVT), 373, 399
transplantation, 157 surveillance for, 488–489
Coronary artery bypass grafting (CABG), 288, 396 Dendritic cells, 9, 10
530 Index

Desmopressin, 185 Dysfibrinogenemia, 398


Detoxification of ammonia to glutamine, 308 Dysglycaemia, 315
Dexmedetomidine, 75, 406, 424, 511–512
Diabetic keto-acidosis (DKA), 15
Diazepam, 75 E
Difficult-to-wean patient, 472–474 Early allograft dysfunction (EAD), 209, 452
Diffusing capacity of the lung for carbon monoxide Early graft failure
(DLCO) test, 295 early postoperative period, 452
Diffusion-perfusion defect, 295 management, 452
Dilated cardiomyopathy, 417 operating room, 451
Dilutional coagulopathy, 427 risk factors, 453
Direct acting agent (DAA), 441 small for size syndrome, 453–454
Direct-acting antiviral drugs (DAAD), 117 ECD, see Extended criteria donor (ECD)
Disseminated intravascular coagulation (DIC), 179, 398 Echinocandin, 463
Dobutamine, 286 Echinococcal cyst, 340
Dobutamine stress echocardiography (DSE), 285–286 Echinococcosis, 340
Domino liver transplantation, 131–132 Echocardiography, 356, 417
Donation after cardiac death (DCD), 105, 117, 252 Electrocardiogram, 420
protocols, 97 Electrocautery, 492
procurements, 117 Electrolyte management, liver transplantation, 419, 426
utilization of, 210 Emergency surgery, liver disease, 395
Donation Service Area (DSA), 253 Empirical antibiotic therapy, 315
Donor age, 207 Encephalopathy, 314, 403
Donor demographics etiology and pathophysiology of, 307
donation after cardiac death, 210 See also specific encephalopathies
donor age, 207–209 End stage liver disease (ESLD), 396–398, 400, 401
donor characteristics, 208 Endocrine function, 16
donor risk index, 210–211 Endogenous cannabinoids, 164
malignancy, 211 Endothelial cells, 9
number of transplants, 208 Endothelial damage, 175
prolonged cold ischemic time, 209 Endothelial dysfunction, 180
seropositive donors, 209–210 Endothelial nitric oxide synthase (eNOS), 9
steatosis, 209 Endothelial protein C receptor (EPCR), 177
Donor gamma-glutamyl transpeptidase, 103 Endothelin (ET-1), 213
Donor graft quality, 103 End-stage liver disease (ESLD), 31, 94, 508, 509
Donor hepatectomy, 500 cardiovascular changes in, 281
Donor liver MMT, 513
implantation, 127–131 symptoms, 512
distribution, resulting in regional disparities in liver Enflurane, 96
allocation, 113–115 Enhanced recovery after surgery (ERAS) programm, 360
Donor management, prior to procurement, 118 Enteral nutrition, 428
Donor management goals (DMGs), 118 Enterobacteriaceae
Donor Organ Sharing Scheme, 102–103 carbapenemase-producing, 461
Donor pool, 115–118 ESBL producing, 460
Donor population, proportion by age, 111 Epidural analgesia, 97, 490, 491, 513
Donor risk index (DRI), 117, 210, 211, 252 Epidural devices, 138
Donor selection, 103 Epinephrine, 14
Donor Service Area (DSA), 113–115 Epsilon-aminocaproic acid (EACA), 186
Donor’s bile duct, 130 Epstein-Barr virus (EBV), 449
Donors and ABO incompatibility, 264 ESLD, see End-stage liver disease (ESLD)
Donor-specific antibodies (DSA), 434 Esophageal Doppler, 355
Doppler ultrasound, early graft failure, 453 Esophageal varices, 28
DRI, see Donor risk index (DRI) Euro-Collins, 214
Drug-induced liver injury (DILI), 13, 45 European Liver Transplant Registry, 491
Drug-related eosinophilic syndrome (DRESS), 45 Eurotransplant, 103
DSE, see Dobutamine stress echocardiography (DSE) Eurotransplant liver waiting list, 102
D-sorbitol elimination, 84 Everolimus, 438
Duloxetine, 519 adverse effect, 439
Duramorph, 516 clinical use, 439
Dynamic liver function test, 79, 80, 141 method of action, 439
Index 531

pharmacokinetics and metabolism, 439 Glucose metabolism, 136


therapeutic drug monitoring, 439 Glutamine, 50
Excessive blood loss, 377 Glutamine synthetase (GS), 308
Expanded criteria organs, 116 Glycemic control, 315
Expiratory positive airway pressure (EPAP), 473 liver transplantation, 419, 427
Extended criteria donor (ECD), 102–105 Glycogen storage diseases (GSDs), 14, 226, 341
Extended-spectrum beta-lactamase (ESBL) producing Glycoprotein VI, 174
enterobacteriaceae, 460–461 Graft dysfunction, 427, 434
Extracorporeal circuit, 127 Graft failure, 476
Extracorporeal liver assist devices, 317 diagnosis, 453
Extracorporeal membrane oxygenation (ECMO), 210, signs and symptoms, 452
477 treatment of, 453
Extracorporeal perfusion, 105 Graft function
Extrahepatic biliary tract, 4 evaluation, 427
Extrahepatic injury, 225 signs of, 427
Extubation, guidelines, 422 Graft rejection, 441–442
Graft weight to recipient body weight ratio (GWBWR),
370, 371
F Graft-monitoring tools, 136
Falciform ligament, 125 Growth hormone, 16
Familial amyloidotic polyneuropathy (FAP), 131
Fentanyl, 510
FHF, see Fulminant hepatic failure (FHF) H
Fibrillation, atrial, 289 Halothane, 96, 352, 405, 406
Fibrin sealant, 380 Halothane hepatitis, 405
Fibrinogen, 261 Hanging maneuver, 500
Fibrinolysis, 177, 182, 398 Hanging technique, 336
Fibrogenic microbiome, 26 HAT, see Hepatic artery thrombosis (HAT)
Fibrosing alveolitis, 299 HBV, see Hepatitis B virus (HBV)
Fibrosis, 24–26 HCC, see Hepatocellular carcinoma (HCC)
Flapping tremor, 28 HCV, see Hepatitis C virus (HCV)
Fluconazole, 457, 462 Heart failure, 283, 289
Fluid management, 263, 318 Heart-beating donation, 92
Fluid restriction, 200 Hemangiomas, 341
Fluid therapy, 357 Hemodynamic changes
Focal nodular hyperplasia (FNH), 341, 342 anhepatic phase, 167
Forward flow theory, 9, 65 in cirrhosis, 283
Fresh frozen plasma (FFP), 183, 379, 399 dissection phase, 167
Fulminant hepatic failure (FHF), 123, 226, 259, 308 graft reperfusion, 168
Functional residual capacity (FRC), 397 neo-hepatic phase, 169
Fungal infections, 315 pathogenic mechanisms, 164
in liver transplantion, 462–463 Hemodynamic consequences, of vascular occlusions,
risk factor, 456–457 352–353
Future liver remnant (FLR), 82 Hemodynamic disturbance, and management, 166
Hemodynamic management, 357–358
Hemodynamic monitoring, 138
G liver transplantation, 420
Gabapentin, 512, 514 tools, 354
Galactose elimination, 84 Hemodynamics, 352
Gamma-aminobutyric acid (GABA) receptors, 74 Hemolytic uremic syndrome (HUS), 448
Gastro-duodenal artery (GDA), 124 Hemorrhage, 342, 377, 378, 380, 383, 492–493
Gastrointestinal (GI) tract, 5 Hemostasis
Gastrointestinal stress ulcers, 428 activated partial thromboplastin time, 181
Gastrointestinal system, caval cross-clamping, 157 alterations, 195, 196
Genetic polymorphisms, 45 amplification phase, 175
Gestational alloimmune liver disease (GALD), 226 bleeding time, 180
GFR, see Glomerular filtration rate (GFR) coagulation cascade and liver disease, 178
Global enddiastolic (GEDV), 140 desmopressin, 185
Glomerular filtration rate (GFR), 270, 271, 402 endothelial dysfunction, 180
Glucose homeostasis, 14 fibrinolysis monitoring, 182
532 Index

Hemostasis (cont.) Hepatocellular dysfunction, 26


fresh frozen plasma, 183–184 Hepatocytes, 7, 9
hyperfibrinolysis, 179–180 Hepatology, 335
inhibition and fibrinolysis, 176–178 Hepatomegaly, 340
in liver disease, 178 Hepatoprotective method, 353
lysine analogues, 186 Hepatopulmonary axis, 295
platelet function, 179 Hepatopulmonary syndrome (HPS), 28–29, 300–302,
platelet function analyzer-100, 180 400, 475
platelet transfusion, 184–185 Hepatorenal syndrome (HRS), 29, 235, 271, 282,
primary hemostasis, 174 400–401, 418
procoagulant drugs, 185 grades of, 401
propagation phase, 175, 176 mechanisms, 401
prothrombin time, 180–181 stages of, 402
recombinant factor VIIa, 186 Hepatorenal syndrome-acute kidney injury (HRS-AKI)
secondary hemostasis, 174–176 treatment algorithm, 274
thrombin generation test, 181 Hereditary hemochromatosis, 404
thromboelastography, 181 Hereditary hemorrhagic telangiectasia, 298
topical agents in, 187 Heterotaxy syndrome, 227
Hepatectomies, 82 High volume in intensive care (IVOIRE) study, 56
Hepatectomy, 125, 321–322, 370 High volume plasma exchange (HVPE), 318
CT volumetry, 499 Hilar dissection, 123, 126
liver protective strategy during, 499 Histidine-tryptophan-glutarate (HTK) solution, 92
living donor (see Living donor hepatectomy) HOCM, see Hypertrophic obstructive cardiomyopathy
management, 501 (HOCM)
patient selection, 497 HPS, see Hepatopulmonary syndrome (HPS)
postoperative assessment, 501–502 HRS, see Hepatorenal syndrome (HRS)
pre-emptive total, 259 HUS, see Hemolytic uremic syndrome (HUS)
preoperative assessment, 497–501 Hydatid cystic disease, 339, 340
right, 334 Hydatid sand, 341
surgical techniques, 503, 504 Hydromorphone, 77, 509–510
Hepatic arterial buffer response (HABR), 383 Hydromorphone-3-glucuronide (H3G), 509
Hepatic artery buffer, 63 Hydroxyethyl starch (HES), 358
Hepatic artery thrombosis (HAT), 83, 262, 428, 493 Hyperacute rejection, 230
Hepatic blood flow (HBF), 163, 372 Hypercalcemia, 137
Hepatic disease, with cerebral manifestation, 325 Hypercholesterolemia, 283
Hepatic drug metabolism, first pass metabolism, 12 Hypercoagulability, 398, 493
Hepatic encephalopathy, 14, 52, 227, 403, 407, 418, 426 Hyperfibrinogenemia, 398
Hepatic endothelial cells, 8 Hyperfibrinolysis, 16, 179, 197, 261, 262
Hepatic failure, 76 Hyperglycemia, 315, 419
Hepatic hydrothorax, 27, 298, 400, 418, 469–470 Hyperkalemia, 137, 248
Hepatic inflow occlusion, 352 Hyperlactatemia, in LTx patients, 136
Hepatic parenchymal transection, 336 Hypertension, 64
Hepatic resection, 335, 490, 491 CNIs, 436
Hepatic reticulo-endothelial dysfunction, 400 intracranial, 307
Hepatic stellate cell (HSC), pathways of, 25 portal, 294
Hepatic surgery, indications for, 339–340 porto-pulmonary, 475
Hepatic vein catheterization, 509 Hypertrophic obstructive cardiomyopathy (HOCM),
Hepatic vein wedge pressure (HVWP), 64, 345 289–292
Hepatic veins join, 6 Hyperventilation, 314
Hepatic venous pressure gradient (HVPG), 27, 64, 65 Hypervolemic hyponatremia, 324, 419, 426
Hepatitis B virus (HBV), 24, 209 Hypnotics, 352
Hepatitis C virus (HCV), 24, 209, 440–441 Hypocalcemia, 168, 419, 427
Hepatobiliary disease, 296 Hypofibrinogenemia, 199
Hepatobiliary surgery Hypoglycemia, 51, 419
alternatives to standard resections, 338 Hypomagnesemia, 419
hepatic resection technique, 335–338 Hyponatremia, 137, 323–325, 397, 398, 419
liver anatomy, 333–335 Hypoperfusion, 65
minimally invasive hepatic surgery, 338–339 Hypotension, 237
Hepatocaval ligament, 127 differential diagnosis, 416
Hepatocellular carcinoma (HCC), 12, 30–31, 106, 113, vasocontricting agents, 417
343, 345, 441 Hypothermia, 51, 137
Index 533

artificial cellular ambience of, 212 fluid management and


preservation injury, 213 osmotherapy, 318–319
therapeutic, 320 glycemic control, 315
Hypothermic machine perfusion (HMP), 214 ICP-targeted therapy, 313
advantages, 214 infection prophylaxis, 315–316
schematic diagram of, 215 nutrition, 315
Hypovolemia, 273, 275, 289, 290 plasma exchange, 318
Hypovolemic hyponatremia, 419, 426 positioning and environment, 314
Hypoxemia, 294, 296, 300 sedation and neuromuscular blockade, 316
seizure prophylaxis, 316
temperature, 314–315
I therapies targeting CPP, 319–320
ICP, see Intracranial pressure (ICP) ventilation, 314
ICU, see Intensive care unit (ICU) Intracranial pressure (ICP), 138, 259, 260, 307, 313
ICU survival, predicting, 81–82 etiology, 310
Idiosyncratic drug reaction, 45 intra-operative considerations, 322
iHD, see Intermittent hemodialysis (iHD) monitoring, 311, 313
Immediate post-operative extubation (IPE), 470 strategies for treating refractory increases in,
Immunological rejection, 431–434 320–322
Immunosuppression, 240 Intrahepatic biliary tract, 4
liver transplantation, 424–426 Intrahepatic glucose levels, 136
post liver transplantation, 440 Intraoperative monitoring
Immunosuppressive agent, 434–435 adequate oxygen delivery, 140
antimetabolites, 437, 438 blood flow assessment, 141
calcineurin inhibitors, 435, 436 cardiac output, 139
corticosteroids, 437 central venous pressure, 140
mTOR inhibitors, 438, 439 central venous saturation, 140
rejection and discovery, 90 conventional laboratory tests, 135–136
side effects, 435 conventional liver function test, 141
Indocyanine green (ICG) clearance test, 80–82, 498 dynamic liver function test, 141
Indomethacin, 320 electrolyte imbalances, 137
Induction therapy, 437, 440 graft function, 140–141
Infants, anesthetic management, 227 hemodynamic monitoring, 138–139
Infectious Diseases Society of America (IDSA), 464 hemostasis and coagulation, 137–138
Inferior vena cava (IVC), 124, 155, 227 metabolism, 136–137
Infiltration, local, 514 neurological monitoring, 138
Inflammation, within brain, 309 positive pressure ventilation, 140
Inflammatory cytokines, activation of, 136 standard hemodynamic monitoring, 139
Inhaled nitric oxide, 475, 477 transesophageal echocardiography, 140
Inherited disease, 295–297 Intrapulmonary shunting, 295
Initiation, 25 Intrathecal morphine (ITM), 372
Innate immune system, 11, 52 Intrathoracic blood volume (ITBV), 140
Inosine monophosphate dehydrogenase Intravenous (IV) fluid, LDLT, 490
(IMDPH), 438 Intravenous patient-controlled analgesia
Inotropes, 165, 166 (IVPCA), 491
Inspiratory positive airway pressure (IPAP), 473 Invasive arterial pressure, 354
Intensive care unit (ICU) Invasive pain interventions, 516
complication prevention, 428 Ischemia, preservation injury, 213
massive transfusion, 420 Ischemia/reperfusion injury (IRI), 141, 380–381
Intermittent hemodialysis (iHD), 274 management, 353–354
International Club of Ascites (ICA), 271, 274 mechanism of, 212–213
International Consensus Conference, 472 organ preservation and modalities, 213–215
International normalized ratio Ischemic cholecystitis, 128
(INR), 181, 234, 398, 452, 453 Isoflurane, 96, 352
Intra-abdominal pressure (IAP), 49 Ito cells, 8, 9
Intracranial hypertension (ICH), 49, 50, 258
in acute liver failure, 307
pathophysiology, 309 J
preoperative management, 313 Janus Kinase 2 (JAK-2) mutation, 46
ammonia-reducing strategy, 317 Japanese Liver Transplant Society, 491
extracorporeal liver assist devices, 317 Jugular bulb oximetry, 312
534 Index

K oral and allograft tolerance, 12


Kasai portoenterostomy, 224 oxygen tension, 11
Kayser-Fleischer rings, 47 periportal zone, 11
Kidney transplantation, breakthroughs in, 93 portal hypertension, 6
King’s College Criteria (KCC), 48, 56–57 protein metabolism and hepatic encephalopathy,
Kupffer cell, 9, 10, 12 14–15
Kupffer cell dysfunction, 400, 401 Liver abscess
amebic abscess, 340
hydatid cyst, 340–341
L pyrogenic abscess, 340
Lactate, 48, 136 Liver Advisory Group, 103
Laparoscopic cholecystectomy, 396 Liver allocation, in US, 110–113
Laparoscopic donor surgery, LDLT, 486 Liver biopsy, 453
Laparoscopic hepatobiliary surgery, 487 Liver cirrhosis, effect of, 71
Laparoscopic liver resection (LLR), 339, 360 Liver disease
Laparoscopic right hepatectomy, 371 abdominal surgery, 395
Laudanosine, 405 acute, 394
LD, see Living donation (LD) ascites, 397–398
LDLT, see Living donor liver transplantation (LDLT) bariatric surgery, 396
Left hepatectomy (LH), 373 cardiothoracic surgery, 396
Left lateral hepatectomy (LL), 373 cholecystectomy, 395
Left lobe grafts, 486, 492 chronic, 394–395
Left ventricular outflow tract (LVOT) obstruction, 289 coagulopathy, 398, 399
Lesions CTP Classification, 391, 392
benign solid liver, 341 elective surgery, 394
focal nodular hyperplasias, 341 emergency surgery, 395
malignant liver, 342 hemostatic alterations in, 195–197
Lidocaine, 83, 316 hepatorenal syndrome, 400
Ligamentum venosum, 4 hereditary hemochromatosis, 404
LigaSure® device, 336 immediate preoperative preparation, 406–407
Linear transducer, 518 anesthetic induction, 407
Linezolid, MRSA, 458 emergence and postoperative care, 407
Lipid metabolism, 15 intraoperative monitoring/management, 407
Lipopolysaccharide (LPS), 12 impact on perioperative outcome, 389–391
Liposomal amphotericin B, 457, 463 MELD score, 392
Lithium dilution cardiac output (LiDCO™) systems, 165 neurologic dysfunction, 403–404
Liver, 12, 13 OPCAB, 396
age-adjusted rates of, 30 perioperative complication, 391
anatomic lobules and metabolic zones, 10–11 pharmacokinetics, 508
arterial and venous circulation, 5 pharmacologic consideration, 404–405
cellular classification, 6–10 benzodiazepines, 406
cellular microanatomy, 9 dexmedetomidine, 406
coagulation and fibrinolysis, 15–16 neuromuscular blocking agents, 405
embryology, 4 opioids, 406
external anatomy, 6, 7 propofol, 406
functions, 4 volatile anesthetic agents, 405
glucose homeostasis, 14 physiology, 63–64
hepatic drug metabolism portal pressure and hypertension, 64–65
first pass metabolism, 12–13 preoperative consideration, 404
P450 system, 13 preoperative preparation, 402
phase II and III metabolism, 13 prevalence, 391
hepatic endocrine function, 16 pulmonary function, 399, 400
hepatic microanatomy, 8 renal function evaluation, 401
hexagonal hepatic lobule, 11 scoring system, 391, 392, 394
innate and adaptive immunity, 11–12 severity and nature, 394
internal anatomy, 6, 7 splanchnic, portal and hepatic venous circulation, 64
left lobe segments, 6 surgical procedure, 395–397
lipid metabolism and nonalcoholic fatty liver disease, TAVR, 397
15 thoracic surgery, 397
macroscopic anatomy, 4–6 vasopressin in, 65–67
Index 535

Liver failure, 380, 383, 394 AKI after (see Acute kidney injury (AKI))
operative causes, 500 anesthesia and perioperative care, 95–96
pathophysiology, 502 anesthetic considerations, 259
post hepatectomy, 344 anhepatic phase, 129
post-donation, 493 arterial reconstruction, 125
prediction, 383 back table preparation, 124
signs, 497 between 1991 and 2009, 102
Liver function biliary complications, 428
assessment, 74–75 cadaveric liver grafts, 104
early graft failure cardiovascular changes
assessment, 451–453 cardiac output, 165
early postoperative period, 452 extrinsic factors, 164
management, 452–453 hemodynamic changes, 164
operating room, 451–452 inotropes and vasopressors, 165–166
small for size syndrome, 453 intrinsic factors, 163–164
laboratory measures, 394 pathogenic mechanisms, 164–165
scintigraphy, 499 phases, 167
Liver function reserve, patient selection, 498 physiological considerations, 163
Liver function test, 79, 452 cardiovascular response, 415
Liver graft exchange, 103 caval anastomosis, 128
Liver injury, 24 caval replacement vs. piggyback, 94–95
Liver preservation, technical advances, 216 cerebrovascular stability, 259–261
Liver remnant volume (LRV), 382 coagulation response, 420
Liver resection, 82 coagulopathy, 261–262
cardiovacular risk assessment, 350 consequence of CAD, 284
indications, 339 contraindications to, 57
intraoperative management early modern era, 89
anesthetic agents, 351–352 early postoperative care, 96
esophageal Doppler, 355 end to side, 129
fluids type, 358 fast-tracking, 96–97
hemodynamic consequences of vascular glycemic control, 419, 427
occlusions, 352 graft function evaluation, 427–428
hemodynamic management, 357 hemodynamic changes
hemodynamic monitoring tools, 354 during anhepatic phase, 167–168
hemodynamics, 352 during dissection phase, 167
hypnotics, 352 during graft reperfusion, 168
invasive arterial pressure, 354–355 during the neo-hepatic phase, 169
ischemia-reperfusion injury management, 353 hemodynamic disturbance, 166–167
mechanical ventilation, 359–360 hemodynamic monitoring, 420–421
pulmonary artery catheter, 355 hemostatic alterations in, 195
transfusion, 358–359 hepatic artery anastomosis, 125
transoesophageal echocardiography, 356 hepatic encephalopathy, 426
liver function evaluation, 350 history of, 121–123
morbidity, 344 hypotension, 416
nutritional assessment, 351 ICU complication prevention, 428
postoperative analgesia and rehabilitation, 360 immunosuppression, 424
pulmonary function assessment, 351 immunosuppressive agents, 90–91
renal function assessment, 351 indications for, 32, 111
Liver surgery complications interventions
bleeding and excessive blood loss, 377–378 anhepatic phase, 168
fluid management and transfusion, 379 dissection phase, 167
pharmacologic agents, 380 graft reperfusion, 168–169
small-for-size syndrome, 382 neo-hepatic phase, 169–170
surgical technique, 378–379 intraoperative management of renal function,
minimal invasive resection, 384 275–276
perioperative hepatic insufficiency, 380 living donor (see Living donor liver transplantation
ischemia reperfusion injury, 380 (LDLT))
treatment, 381–382 management of coagulopathy and bleeding, 427
Liver transplantation, 82–83, 93–94, 448, 456, 485, 486 mechanical ventilation, 421, 470
abdominal incision and exposure, 124–126 metabolic abnormalities during, 248
536 Index

Liver transplantation (cont.) infection, 492


native liver hepatectomy, 125–127 IV fluids and nutrition, 490
neurological outcomes after, 325 laboratory testing, 490
neurological response, 418 liver failure post-donation, 493
organ preservation techniques and solutions, 91–92 long-term follow up, 491
organ response, 415–420 pulmonary embolism, 493
pediatric, 519 PVT, 493
perioperative fluid balance, 262–264 radiological evaluation, 490
portal vein thrombus, 126 risk factors, 488
postoperative care, 416 surgical technique, 486–488
pre-procurement biopsy, 118 Local infiltration, 514
in presence of newly transplanted heart, 239–240 Lorazepam, 424
in presence of renal failure, 236 L-Ornithine-L-aspartate (LOLA), 317
procedure, 123 Low molecular weight (LMWH), 489
pulmonary response, 417 Low serum sodium, 137
renal replacement therapy, 276 Lung Allocation Score (LAS), 240
renal response, 418 Lymphocyte, 9
rejection, 432
sedation and analgesia, 422, 425
sodium and electrolyte management, 419, 426 M
super-urgent listing for, 55 Macrosteatosis, 117
surgical considerations, 259 Maddrey discriminant function (MDF), 23
survival of patients, 58 Magnetic resonance imaging (MRI), 138, 493
technical and conceptual foundations, 89–90 Major histocompatibility complex (MHC), 433
technical innovations, 97 Major histocompatibility complex I (MHC I) markers, 11
temporary portocaval, 123 Malignant liver lesions, 342–343
vascular complications, 428 Malnutrition, 351
worldwide growth, regulation and academic Mammalian target of rapamycin (mTOR) inhibitors, 438
organizations, 98 adverse effect, 439
Liver tumor, 226, 338 clinical use, 439
Liver volumes, 57 method of action, 439
Liver–kidney transplantation, 249 pharmacokinetics and metabolism, 439
Living donation (LD), 105–106 therapeutic drug monitoring, 439
Living donor hepatectomy, 367–370, 516–519 Mannitol, 166, 318
altruistic nature of, 373 Marginal donors and ABO incompatibility, 264–265
anesthetic management, 371–372 Mass spectrometry, 509
clinical and biological outcome of, 373 Matrix metalloproteinase 9 (MMP-9), 308
complications, 373–374 Mayo Clinic, 391
evaluation criteria, 369 MDR Pseudomonas, 461
post-operative management, 372 Mean pulmonary artery pressure (MPAP), 400
preoperative evaluation, 367 Mechanical ventilation
contraindications to donations, 370 liver transplantation, 421–422
ethical considerations, 369–370 non-invasive, 472, 473
first evaluation phase, 367–368 prolonged, 472
second evaluation phase, 368 sedation during, 473
third evaluation phase, 368–369 weaning protocol, 470, 473
surgical technique, 370–371 Mechanical ventilators, 92
Living donor liver transplantation (LDLT), 97, 105, 118, Metabolic acidosis, 426
252 Metabolic alkalemia, 427
epidemiology, 485–486 Metabolic alkalosis, 403
ethical issues, 487 Metabolic syndrome, 15
mortality rates, 492–494 Metabolism, 508
postoperative care, 488 buprenorphine, 510
postoperative complications, 491–492 dexmedetomidine, 511
analgesia, 490–491 fentanyl, 510
antibiotic prophylaxis, 489 gabapentin, 512
bile leak, 492 hydromorphone, 509
Clavien’s classification system, 491, 492 methadone, 509
DVT, 488 morphine, 508
hemorrhage, 492 Methadone, 509
Index 537

Methadone maintenance therapy (MMT), 509, 513 VRE, 459, 460


Methicillin-resistant Staphylococcus aureus (MRSA), Muscle relaxants, 92
456, 458–459 Mycophenolate mofetil (MMF), 265, 437
Methylene blue test, bile leak, 504 adverse effect, 438
Micafungin, 463, 464 clinical use, 438
Microbubble, 301 method of action, 438
Microdialysis, 136, 137 pharmacokinetics and metabolism, 438
Microwave ablation (MWA), 113 therapeutic drug monitoring, 438
Midazolam, 75 Mycophenolic acid glucuronide (MPAG), 438
Middle hepatic vein (MHV), 333 Myectomy, 289
Minimal invasive resection, 384 Myeloproliferative disorders, 46
Minimally invasive hepatic surgery, 338 Myocardial perfusion scan, 286
MMF, see Mycophenolate mofetil (MMF) Myofibroblasts, 26
Mobile army surgical hospital (MASH) system, 93
Model for end stage liver disease (MELD) score, 57, 82,
97, 101, 294, 308, 380, 392, 393, 441, 452, N
470–472, 498 N-acetylcysteine (NAC), 13, 44, 166
advantages of, 102 N-acetyl-p-benzoquinone-imine (NAPQI), 13
coagulopathy and transfusion, 250–251 NAFLD, see Nonalcoholic fatty
disadvantage, 102 liver disease (NAFLD)
disease severity, 112 National Institute on Alcohol Abuse
formulas for, 112 and Alcoholism, 403
futility, 254 National Institutes of Health, 476
implementation, 247–248 National Institutes of Health (NIH) Consensus
organ allocation, 252–253 Conference on Liver Transplantation, 94
on post-transplant survival, 102 Native liver hepatectomy, 125
predictive accuracy, 137 Natural killer (NK) cells, 10, 11, 358
renal insufficiency, 248–250 Natural killer T (NKT) cells, 11
severity of disease, 251–252 Near infrared spectrophotometry (NIRS), 313
Share 35, 253–254 Near-infrared spectroscopy (NIRS), 138
short term prognosis of patients, 112 Neoangiogenesis, 9
temperature, 137 Neonatal hypoxia, 226
at transplantation, 113 Neonatal liver failures, 226
Modified Ramsay Sedation Scale, 423 Nephropathy, 418
MOF, see Multiorgan failure (MOF) Nephrotoxicity, 458, 463
Molecular adsorbent recirculating system (MARS), 275, Neuraxial anesthesia, 516, 517, 519
317, 501 Neurologic dysfunction, liver disease, 403
Monoclonal antibodies, 440 Neurologic system, caval cross-clamping, 157
Monoclonal antibody therapy, 476 Neurological monitoring, 138
Monoethylglycinexylidide (MEGX) test, 80, 83–84 Neuromuscular blockade, 316
Morphine, 77, 508–509 Neuromuscular blocking agents, 76, 405
Morphine-3-glucuronide (M3G), 508 Neurotoxic ammonia, 136
Morphine-6-glucuronide (M6G), 508 Neurotoxicity, 436
MPAG, see Mycophenolic acid glucuronide (MPAG) Neutrophil function, 53
MRSA, see Methicillin-resistant Staphylococcus aureus Neutrophil gelatinase-associated lipocalin (NGAL), 273,
(MRSA) 446
mTOR inhibitors, see Mammalian target of rapamycin NHS Blood and Transplant, 102
(mTOR) inhibitors Nitric oxide (NO), 164
Multiorgan failure (MOF), 257 Non-absorbable disaccharides, 426
Multiorgan system failure, 452 Non-alcoholic fatty liver disease (NAFLD), 15, 22, 23,
Multiply resistant gram-positive/negative bacteria 396
amphotericin B, 463 Non-alcoholic steatohepatitis (NASH), 23, 284, 396
CRAB, 462 Non-anatomic resections, 334
CRKP, 461 Noncardiogenic pulmonary edema, 418
echinocandin, 463 Non-cirrhotic liver disease, 394
ESBL producing enterobacteriaceaes, 460 Non-dihydropyridine calcium channel blockers, 417
invasive aspergillosis, 464 Non-invasive ventilation (NIV), 472–474
MDR Pseudomonas, 461 Nonparenchymal cells, 7
MRSA, 458, 459 Non-selective cyclo-oxygenase inhibitor, 320
triazoles, 463 Norepinephrine, 29, 276
538 Index

Normoglycemia in Intensive Care Evaluation and multimodal approaches to, 514


Survival Using Glucose Algorithm Regulation pediatric, 519
(NICE-SUGAR), 427 perioperative, 512–513
Normothermia, 314 post-operative analgesia (see Post-operative
Normothermic MP (NMP), 215 analgesia)
Nosocomail pneumonia, 458 Pancuronium, 76
Nutrition, LDLT, 490 Paracentesis, 397
Paracetamol overdose (POD), 43–44
Paracetamol-induced liver injury, 257
O Parenchymal disease, 299
Off-pump coronary artery bypass (OPCAB), 284, Parenteral nutrition (PN), 501
288–289, 396 Partial thromboplastin time (PTT), 15
OKT3 (muromonab-CD3), 440 PD, see Pharmacodynamics (PD)
Oliguria, 501 Pediatric end-stage liver disease model (PELD), 112,
OPCAB, see Off-pump coronary artery bypass (OPCAB) 221, 222
Open cholecystectomy, 396 Pediatric liver transplantation, 227–229, 519
Open laparotomy, liver disease, 395 age distribution, 222–223
Operating room, patients, 258 anesthetic management
Opioids, 77, 424, 508 infants and toddlers, 227–229
equipotent doses, 511 pre-teenager, 229
liver disease, 406 teenager, 229
use of, 513 biliary atresia, 224–225
Oral tolerance, 12 biliary complications, 230
Organ allocation, 97 cholestatic liver disease, 223–224
MELD score, 252 congenital heart disease, 227
pediatric liver transplantation, 221 fulminant hepatic failure, 226
Organ distribution, 102–103 hepatic encephalopathy, 227
Organ donation, 94, 97–98 history of, 221
Organ preservation, 91, 124, 212 indication for, 223
Organ Procurement and Transplant Network (OPTN) infectious complications, 231
data, 234, 253 liver tumors, 226
Organ procurement organizations (OPOs), 102, 113, 114 metabolic disease, 222, 225–226
Organ Procurement Transplantation Network (OPTN), neonatal liver failures, 226
113, 114, 221 organ allocation, 221–222
Organomegaly, 295 outcome, 231
Orthodeoxia, 300, 301 post operative care, 229–230
Orthotopic liver transplantation, 121 primary nonfunction, 231
Osler–Weber–Rendu disease, 298 rejection, 230
Osmolar damage, 104 SPLIT database, 231
Osmotherapy, 318 TPN-induced liver disease, 227
Osmotic demyelinisation, 137 vascular anomalies, 227
Osmotic myelinolysis, 318 vascular complications, 230
Oximetry, jugular bulb, 312 Pentoxifylline, 382
Percutaneous transluminal coronary angioplasty (PCTA),
284
P Perioperative complication, liver disease, 391
Packed red blood cells (PRBC), 379 Peritoneal dialysis, 274
Pain management, 514 Perpetuation, 26
acute, 508, 515 Pharmacodynamics (PD), 71–72
buprenorphine, 510 absorption, 71
chronic, 515, 519 changes in patients, 74
dexmedetomidine, 511 elimination, 72
fentanyl, 510 hepatic dysfunction, 70
gabapentin, 512 metabolism, 72
hydromorphone, 509 neuromuscular blocking agents, 76
intraoperative analgesia, 513–514 opioids, 77
living donor hepatectomy, 516 protein binding and distribution, 72
local infiltration, 514 sedatives, 75
methadone, 509 Pharmacokinetics (PK), 69–71
morphine, 508 absorption, 71
Index 539

classifications, 70 cirrhosis, 390


of drugs, 352 LDLT (see Living donor liver transplantation
elimination, 72–74 (LDLT))
hepatic dysfunction, 70 liver disease, 407
metabolism, 72 Postoperative liver dysfunction (POLD), 405
neuromuscular blocking agents, 76 Post-renal kidney injury, 271
opioids, 77 Postreperfusion syndrome, 417
protein binding and distribution, 72 Post-transplant lymphoproliferative disorder (PTLD),
sedatives, 75–76 440, 449
Pharmacological agents, 199, 380 PPH, see Portopulmonary hypertension (PPH)
Pharmacovigilance Risk Assessment Committee PPV, see Pulse pressure variation (PPV)
(PRAC), 358 Pregablin, 514
Phase I oxidative pathways, 79 Pre-renal kidney injury, 271
Piggyback technique, 94, 122, 123, 128, 259 Pre-teenager, anesthetic management, 229
advantages of, 158 Primary biliary cirrhosis, 297
of orthotopic liver transplantation, 157–159 Primary nonfunction (PNF), 209
placement, 158 Primary sclerosing cholangitis (PSC), 130, 343
Piperidine opioids, 77 Pringle’s maneuver, 337, 352, 353, 500
PK, see Pharmacokinetics (PK) Procalcitonin (PCT), 456
Plasma exchange, 318 Procoagulant drugs, 185
Plasmapheresis, 449 Prolonged cold ischemic time, 209
Plasminogen activator inhibitor-1 (PAI-1), 179 Prolonged hypothermia, 321
Platelet function, in liver disease, 179 Prolonged mechanical ventilation, 470–472
Platelet function analyzer-100 (PFA-100), 180 Prophylactic anticoagulation, 399
Platelet transfusion, 184 Prophylactic hyperventilation, 314
Platelets, 379 Prophylaxis
Platypnea, 400 deep vein thrombosis, 488
Pleural space disease, 298 disadvantage, 458
Pneumonitis, aspiration, 476 role of, 457–458 (see also specific prophylaxis)
POD, see Paracetamol overdose (POD) Propofol, 75, 316, 406, 424
Polyclonal antibodies, 439 Propofol infusion syndrome (PRIS), 424
POPH, see Portopulmonary hypertension (POPH) Protein binding
Portal hypertension, 26, 27, 294, 390, 392, 395 capacity, 73
Portal pressure, 64 and distribution, 72
Portal vein anastomosis, 128 Protein C, 177
Portal vein embolization (PVE), 345, 383, 499 Protein metabolism, 14
Portal vein flow (PVF), 383 Proteins induced by Vitamin K absence (PIVKA), 398
Portal vein thrombosis (PVT), 30, 131, 428, 493 Prothrombin complex concentrates (PCCs), 200, 261,
Portal venous blood flow (PBF), 163 399
Portocaval shunt (PCS), 384 Prothrombin time (PT), 15, 56, 180, 195, 398, 399, 452,
Portopulmonary hypertension (PPH), 29, 150, 239, 298, 453
400, 475 PSC, see Primary sclerosing cholangitis (PSC)
Portosystemic encephalopathy (PSE), 28 PSE, see Portosystemic encephalopathy (PSE)
Posaconazole, 463, 464 Pulmonary artery catheter (PAC), 139, 149, 165, 236,
Positive end expiratory pressure (PEEP), 50, 314, 421, 355–356, 421, 475
476, 477 Pulmonary artery occlusion pressure (PAOP), 150
Post hepatectomy liver failure, 344–347 Pulmonary artery pressures, 150, 475
Post live transplantation, immunosuppression approach, Pulmonary disease, 399
440 hepatic hydrothorax, 400
Post operative care, pediatric liver transplantation, 229 HPS, 400
Posthepatectomy liver failure, risk factors, 493 portopulmonary hypertension, 400
Post-operative analgesia, 514 Pulmonary edema, 474
behavioral approaches, 515 Pulmonary embolism, LDLT, 493
invasive pain interventions, 516 Pulmonary function
pharmacotherapeutics, 514 assessment, 351
physical modalities, 515 in patients with cirrhosis, 294–295
Postoperative care Pulmonary function test, 294, 301
LDLT, 488 Pulmonary hypertension (POPH), 299–300
liver transplantation, 416 Pulmonary response, liver transplantation, 417
Postoperative complications, 488 Pulmonary system, caval cross-clamping, 156
540 Index

Pulmonary vascular resistance (PVR), 400 Reno-portal anastomosis (RPA), 131


Pulmonary vasodilator therapy, 400 Reperfusion, 128
Pulse contour cardiac output (PiCCO™), 165 Reperfusion injury, 213
Pulse oximetry, 139 Respiratory failure, 296
Pulse pressure variation (PPV), 140, 354, 355 Restrictive lung disease, 294
PVE, see Portal vein embolization (PVE) rFVIIa, Recombinant Factor VIIa (rFVIIa)
PVT, see Portal vein thrombosis (PVT) Richmond Agitation-Sedation Scale (RASS), 422, 423
Pyrogenic abscess, 340 Rifaximin, 28
Right heart failure, 6
Right ventricular systolic pressure (RSVP), 299
Q Risk, injury, failure, loss, end-stage (RIFLE) criteria,
Quantitative liver function test, 79 235, 270, 272, 446
Quinopristin-dalfopristin, MRSA, 458–459 Rocuronium, 76
Rotational thromboelastometry (ROTEM®), 251, 261,
489
R Roux-en-Y hepatico-jejuonostomy, 131, 344
RAAS, see Renin angiotensin aldosterone system Roux-en-Y jejunal limb, 131
(RAAS) RRT, see Renal replacement therapy (RRT)
Radio frequency ablation (RFA), 113
Radiology, LDLT, 490
Raft function monitoring, 140 S
Randomized clinical trials (RCT), 200 Salvage therapy, 464
Randomized Evaluation of Normal versus Augmented Sarcopenia, 351
Level (RENAL) study, 56 SBP, see Spontaneous bacterial peritonitis (SBP)
Rapamycin, 447 Scandiatransplant, 102
RASS, see Richmond Agitation-Sedation Scale (RASS) Scientific Registry of Transplant Recipients (SRTR)
Reactive oxygen species (ROS), 213 database, 98, 210, 237, 253
Rebalanced hemostasis, 398 Scintigraphy, liver, 499
Recipient prioritizing, 101–102 Second International Consensus Conference on
Recombinant Factor VIIa (rFVIIa), 186–187, 262, 380 Laparoscopic Liver Resections, 487
Recovery Sedation
from hyperfibrinolysis, 262 and analgesia, 422–425
liver transplantation, 265 during mechanical ventilation, 473
Rectus abdominis (RA), 517–519 scales, 423
Red cell transfusion, 358 Sedative agent, 316
Refractory vasoplegia, 273 Segmental biliary dilation, 340
Regional anesthesia, 404 Selective bowl decontamination, 457
Rejection Selective digestive tract decontamination (SDD), 457
allograft mechanisms, 433, 434 Sepsis
features, 433 pathophysiology, 502
immunological, 431 pseudomonas, 461
liver transplantation, 432 Seronegative ALF, 46
treatment of, 476 Seropositive donors, 209
Remifentanil, 77, 352, 406 Serum albumin, 112
Renal allograft dysfunction, 237 Serum creatinine (SCr), 112, 401
Renal dysfunction, 441 Serum cystatin C, 273
Renal failure, 270 Serum sodium, 112
Renal function Serum-ascites albumin gradient (SAAG), 27
assessment, 351 SFSS, see Small-for-size syndrome (SFSS)
evaluation, 401 Share-35 rule, 114, 115, 253
intraoperative management of, 275 Sickest first model, 97
Renal injury, see Acute kidney injury (AKI) Simultaneous liver–kidney transplantation (SLKT),
Renal insufficiency, 248, 270 233–235
Renal replacement therapy (RRT), 50, 156, 248, 274, eligibility criteria for, 235
276–278, 445, 448, 449 intra-operative management, 236
Renal system, caval cross-clamping, 156 post-operative management, 237
Renal transplantation, in presence of newly transplanted pre-operative evaluation, 235–236
liver, 237 Single pass albumin dialysis (SPAD), 275
Renin angiotensin aldosterone system (RAAS), 282, 283 Single photon emission computed tomography (SPECT),
Renin angiotensin system (RAS), 53 286
Index 541

Sinusoidal constriction, 64 method of action, 435


Sinusoidal endothelial cells (SECs), 212 pharmacokinetics and metabolism, 435
Sirolimus, 438 therapeutic drug monitoring, 436
adverse effect, 439 Target-controlled propofol infusion, 352
clinical use, 439 T-cell activation, 433–434
method of action, 439 99m
Tc-mebrofenin scintigraphy, 499
pharmacokinetics and metabolism, 439 TEE, see Transoesophageal echocardiography (TEE)
therapeutic drug monitoring, 436, 439 Teenager, anesthetic management, 229
Skin- and soft tissue infections (SSTI), 458 Temporary portocaval shunt (TPCS), 122, 123, 127
SLKT, see Simultaneous liver–kidney transplantation Terlipressin, 29, 165
(SLKT) Thalium-persantine angioscintigraphy, 350
Small-for-size syndrome (SFSS), 382–384, 453 Therapeutic hypothermia (TH), 320–321
Smoking, 488 Thiopurine methyltransferase (TPMT), 437
Society of Critical Care Medicine, 473 Thoracentesis, preoperative/postoperative, 400
Sodium bicarbonate, 276 Thoracic surgery, liver disease, 397
Sodium management, liver transplantation, 419, 426 Three-dimensional computed tomography (3D–CT),
Sodium-MELD score, 112 liver, 487, 493
Solid organ transplantation, 491 Thrombectomy, blood loss during, 127
Solute carrier (SLC) transporters, 13 Thrombin activatable fibrinolysis inhibitor (TAFI), 16,
Spanish Model of Organ Donation, 103 178, 179
Spanish transplant system, 103 Thrombin generation test, 181
Spironolactone therapy, 397 Thrombocytopenia, 398, 427
Splanchnic vasodilation, 26 Thromboelastography (TEG), 181, 182, 199, 251, 261,
Splanchnic venous pressure, 198 263, 489
Splenectomy, 383 Thromboembolic prophylaxis, 428
Splenic artery ligation (SAL), 383 Thrombopoietin, 16
Split liver transplant (SLT), 211–212 Thrombotic thrombocytopenic purpura (TTP), 448–449
Spontaneous bacterial peritonitis (SBP), 27 Tigecycline
Spontaneous breathing trials (SBTs), 472, 473 MRSA, 458–459
Squibb Institute for Medical Research, 463 VRE, 459
Standard cold storage (SCS), 214 Tissue factor (TF), 175
Standard criteria donor (SCD), 211 Tissue factor pathway inhibitor (TFPI), 176
Standard exception (SE), 101 Tissue Factor VIIa, 380
Standard hemodynamic monitoring, 139 Tissue plasminogen activator (tPA), 16, 177, 179, 197,
Static liver function test, 80 399
Statins, 350 T lymphocytes, 10
Steatosis, 117, 209 Toddlers, anesthetic management, 227
Stellate cells, 8, 9 Toll-like receptor 4 (TLR 4), 12
Stroke volume index, 150 Topical hemostatic agent, 380
Stroke volume variation, 140 Toronto Live Donor Liver Transplant Program and
Studies of Pediatric Liver Transplantation (SPLIT), 222 certain Asian programs, 486
Subdural devices, 138 Total intravenous anesthesia (TIVA), 353
Succinylcholine, effects, 405 Total parenteral nutrition (TPN), 52, 227
Summagadex, 76 Total vascular isolation (TVI), 336, 341
Supportive care, 493 Toxic liver syndrome, 259
Sustained low efficiency dialysis (SLED), 276 Tracheal extubation, 421
SVR, see Systemic vascular resistance (SVR) Tracheostomy, 473
Sympathetic nervous system (SNS), 282, 283 TRALI, see Transfusion related
Sympatho-adrenal system, 275 lung injury (TRALI)
Synthetic dysfunction, 26 Tranexamic acid (TxA), 186
Systemic inflammation, cerebral blood flow and, 309 Tranexanic acid, 359
Systemic inflammatory process, 50 Trans arterial chemoembolization (TACE), 113
Systemic vascular resistance (SVR), 155, 281–283 Transcapillary filtration pressure, 276
Systolic anterior motion (SAM), 289 Transcatheter aortic valve implantation (TAVI), 288
Transcatheter aortic valve replacement (TAVR), liver
disease, 397
T Transcranial Doppler sonography, 138
Tacrolimus, 435 Transcranial Doppler ultrasound (TCD), 312
adverse effects, 436 Transcutaneous electrical nerve stimulation (TENS)
clinical use, 436 therapy, 515
542 Index

Transesophageal echocardiography (TEE), 140, 141, Vaptans, 426


150, 165, 356, 407, 421 Vascular anomalies, 227
Transfusion, 358 Vascular complications
Transfusion associated circulatory overload (TACO), assessment, 428
183, 202, 399 LDLT, 493
Transfusion related acute lung injury (TRALI), 183, 202, pediatric liver transplantation, 230
427, 474, 475 Vascular occlusion
Transjugular intrahepatic portosystemic shunt (TIPS), hemodynamic consequences of, 352
98, 112, 275, 282, 345, 392, 393, 395, 397, types, 337
400 Vasoconstrictor, 274
Transmitral flow, 283 Vasodilatation, 271
Transoesophageal echocardiography (TEE), 356–357 Vasodilation, 417
Transplantable organs, amount of, 115 Vasodilator, 286
Transpulmonary thermodilution cardiac output monitors, Vasodilatory effect, 282
50 Vasogenic edema, 15
Transthoracic echocardiography, 407 Vaso-mediated pulmonary hypertension, 239
Transverse abdominis plane (TAP), 372, 516 Vasopressin, 65, 165
Triazoles, 463 Vasopressors, 165, 166, 252, 276
Tricyclic antidepressants (TCAs), 519 Vasosol, 214
Trisegmentectomy, 334 Vecuronium, 76
Tris-hydroxymethyl aminomethane (THAM), 276 Vena Cava management, 122
Trojan horse hypothesis, 14 Vena porta, 127
TTP, see Thrombotic thrombocytopenic purpura (TTP) Veno-occlusive disorders, 46
T-tube drain, 130 Venous thromboembolism, 428
Tumor-associated antigens (TAA), 12 Veno-venous bypass (VVB), 122, 159, 160, 259, 322
Tympanic tonometry, 313 Ventilator associated pneumonia (VAP), 470, 474–475
Ventilator bundles, 473
Ventral endoderm, 4
U Viral hepatitis, 44–45
U.S. Nationwide Inpatient Sample (NIS), 397 Visceral circulation, 4
UDP-glucuronyl transferase 1, 79 Viscoelastic assays, 493
Ulinastatin, 382 Visco-elastic coagulation testing, 261
Ultrasonography (US), 339, 341 Viscoelastic testing, 150–151, 201
Unfractionated heparin, 489 Vitamin K-dependent coagulation factors, 398
United Network for Organ Sharing (UNOS), 113 Volatile anesthesics, 352, 382, 405
Uridine diphosphate-glucuronosyltransferases (UGTs), Von Willebrand factor (VWF), 15, 174
508 Voriconazol, aspergillosis, 464
US Preventive Services Task Force (USPS), 24 VRE, see Vancomycin-resistant enterococci (VRE)

V W
Valvular disease, 288 Water-soluble drugs, 72
Vancomycin, 458 Weaning, from mechanical ventilation, 470
Vancomycin-resistant enterococci (VRE), 459, 460 Wernicke’s encephalopathy, 404
VAP, see Ventilator associated pneumonia (VAP) Wilson’s disease (WD), 47, 225, 325–326, 404

Das könnte Ihnen auch gefallen