Sie sind auf Seite 1von 120

NCP Nutrition in Clinical Practice

Volume 33 • Number 4 • August 2018

MANAGING GI DISORDERS
Bacteria, Bones, and Stones: Managing
Complications of Short Bowel Syndrome

Dietary Fermentable Oligosaccharides, Disaccharides,


Monosaccharides, and Polyols (FODMAPs)
and Gastrointestinal Disease

Nutrition Management of Necrotizing Enterocolitis

Enteral Nutrition in the Management of Crohn’s


Disease: Reviewing Mechanisms of Actions
and Highlighting Potential Venues for Enhancing
the Efficacy

Enteral Nutrition for Pediatric Crohn’s Disease:


An Underutilized Therapy

Case Report of Wernicke’s Encephalopathy After


Sleeve Gastrectomy

Copper Deficiency Myelopathy After Upper


Gastrointestinal Surgery

Peer-reviewed, practical solutions in clinical nutrition wileyonlinelibrary.com/journal/ncp


ISSN: 0884-5336

NCP_33_4_cover000.indd 1 29/06/18 6:59 PM


Nutrition in Clinical Practice
EDITOR-IN-CHIEF
Jeanette M. Hasse, PhD, RD, LD, FADA, CNSC
Dallas, TX
ASSOCIATE EDITORS
Roland N. Dickerson, PharmD, BCNSP Mary S. McCarthy, PhD, RN, CNSC, FAAN
Memphis, TN Tacoma, WA
Mary Marian, DCN, RDN, CSO, FAND Jayshil Patel, MD
Tucson, AZ Milwaukee, WI

CONTRIBUTING EDITOR
Ryan T. Hurt, MD, PhD
Rochester, MN

ASPEN STAFF
Colleen K. Harper, CAE Catherine Wattenberg Catherine J. Klein, PhD
Interim Chief Executive Officer Director of Publications Publications Manager

EDITORIAL BOARD
Teruyoshi Amagai, MD, PhD Ronald L. Koretz, MD Carol J. Rollins, MS, RD, CNSC,
Nishinomiya, Japan Granada Hills, CA PharmD, BCNSP
Tucson, AZ
Deborah A. Andris, MSN, APNP Debra S. Kovacevich, RN, MPH
Milwaukee, WI Ann Arbor, MI Mary K. Russell, MS, RDN, LDN
David A. August, MD, FACS, CNSP Kenneth A. Kudsk, MD Chicago, IL
New Brunswick, NJ Madison, WI
Denise Baird Schwartz, MS, RD,
Albert Barrocas, MD, FACS Laura E. Matarese, PhD, RD, LDN, CNSC, FADA, FAND, FASPEN
East Point, GA FADA, FASPEN Studio City, CA
David L. Burns, MD, FACG Greenville, NC
David S. Seres, MD, ScM, PNS
Burlington, MA Carol McGinnis, DNP, RN, CNS, CNSC New York, NY
Yvon Carpentier, MD Sioux Falls, SD
Brussels, Belgium Pierre Singer, MD
Remy Meier, MD Tel Aviv, Israel
Isabel Correia, MD, PhD Liestal, Switzerland
Minas Gerais, Brazil Annalynn Skipper, PhD, RD, FADA
Sarah J. Miller, PharmD, BCNSP
Chicago, IL
Gail Cresci, PhD, RD, LD, CNSC Missoula, MT
Cleveland, OH Krishnan Sriram, MD, FRCS(C),
Charles M. Mueller, PhD, RD, CNSC, CDN
Robert S. DeChicco, MS, RD, New York, NY FACS, FCCM
LD, CNSC Chicago, IL
Cleveland, OH Gerard E. Mullin, MD, CNSC
Baltimore, MD Dan Waitzberg, MD, PhD, FASPEN
Rose Ann DiMaria-Ghalili, PhD, RN, CNSC São Paulo, Brazil
Philadelphia, PA Lynne M. Murphy, MSN, RN
Washington, DC Jacqueline Wessel, MEd, RDN, CNSC, CSP,
Beverly J. Holcombe, PharmD, BCNSP,
Luis Nin, MD CLE, LD
FASHP
Montevideo, Uruguay Cincinnati, OH
Ocean Isle Beach, NC
Caroline M. Kiss, RD, DCN Kim Robien, PhD, RD, CSO Patricia H. Worthington, RN, MSN, CNSC
Basel, Switzerland Washington, DC Philadelphia, PA
NUTRITION IN CLINICAL PRACTICE (Print ISSN: 0884-5336; Online ISSN: 1941-2452) is published bimonthly in
February, April, June, August, October, and December on behalf of the American Society for Parenteral and Enteral Nutrition
(ASPEN) by Wiley Subscription Services, Inc., a Wiley Company, 111 River Street, Hoboken, NJ 07030-5774 USA. Periodicals
postage paid at Hoboken, NJ and additional mailing offices. Postmaster: Send all address changes to Nutrition in Clinical Practice,
John Wiley & Sons Inc., C/O The Sheridan Press, PO Box 465, Hanover, PA 17331 USA.
COPYRIGHT  C 2018 by ASPEN. All rights reserved. No part of this publication may be reproduced, stored or transmitted

in any form or by any means without the prior permission in writing from the copyright holder. Authorization to copy
items for internal and personal use is granted by the copyright holder for libraries and other users registered with their local
Reproduction Rights Organization (RRO), e.g. Copyright Clearance Center (CCC), 222 Rosewood Drive, Danvers, MA 01923,
USA (www.copyright.com), provided the appropriate fee is paid directly to the RRO. This consent does not extend to other kinds
of copying such as copying for general distribution, for advertising or promotional purposes, for republication, for creating new
collective works or for resale. Permissions for such reuse can be obtained using the RightsLink “Request Permissions” link on
Wiley Online Library. Special requests should be addressed to: permissions@wiley.com
INFORMATION FOR SUBSCRIBERS Nutrition in Clinical Practice is published in 6 issues per year. Institutional subscription
prices for 2018 are:
Print & Online: US$607 (US and Rest of World), €563 (Europe), £470 (UK).
Prices are exclusive of tax. Asia-Pacific GST, Canadian GST/HST and European VAT will be applied at the appropriate rates. For
more information on current tax rates, please go to www.wileyonlinelibrary.com/tax-vat. The price includes online access to the
current and all online backfiles to January 1st 2014, where available. For other pricing options, including access information and
terms and conditions, please visit www.wileyonlinelibrary.com/access.
DELIVERY TERMS AND LEGAL TITLE Where the subscription price includes print issues and delivery is to the recipient’s
address, delivery terms are Delivered at Place (DAP); the recipient is responsible for paying any import duty or taxes. Title to
all issues transfers Free of Board (FOB) our shipping point, freight prepaid. We will endeavour to fulfil claims for missing or
damaged copies within six months of publication, within our reasonable discretion and subject to availability.
BACK ISSUES Single issues from current and recent volumes are available at the current single issue price from cs-
journals@wiley.com.
DISCLAIMER The Publisher, ASPEN and Editors cannot be held responsible for errors or any consequences arising from the
use of information contained in this journal; the views and opinions expressed do not necessarily reflect those of the Publisher,
ASPEN and Editors, neither does the publication of advertisements constitute any endorsement by the Publisher, ASPEN and
Editors of the products advertised.
PUBLISHER Nutrition in Clinical Practice is published by Wiley Periodicals, Inc., 350 Main St., Malden, MA 02148. E-mail:
cs-journals@wiley.com
JOURNAL CUSTOMER SERVICES: For ordering information, claims and any enquiry concerning your journal subscription
please go to www.wileycustomerhelp.com/ask or contact your nearest office.
Americas: Email: cs-journals@wiley.com; Tel: +1 781 388 8598 or +1 800 835 6770 (toll free in the USA & Canada).
Europe, Middle East and Africa: Email: cs-journals@wiley.com; Tel: +44 (0) 1865 778315. Asia Pacific: Email: cs-
journals@wiley.com; Tel: +65 6511 8000.
Japan: For Japanese speaking support, Email: cs-japan@wiley.com.
Visit our Online Customer Help available in 7 languages at www.wileycustomerhelp.com/ask
PRODUCTION EDITOR email: NCPprod@wiley.com.
Wiley’s Corporate Citizenship initiative seeks to address the environmental, social, economic, and ethical challenges faced in our
business and which are important to our diverse stakeholder groups. Since launching the initiative, we have focused on sharing
our content with those in need, enhancing community philanthropy, reducing our carbon impact, creating global guidelines and
best practices for paper use, establishing a vendor code of ethics, and engaging our colleagues and other stakeholders in our
efforts. Follow our progress at www.wiley.com/go/citizenship
Wiley is a founding member of the UN-backed HINARI, AGORA, and OARE initiatives. They are now collectively known as
Research4Life, making online scientific content available free or at nominal cost to researchers in developing countries. Please
visit Wiley’s Content Access – Corporate Citizenship site: http://www.wiley.com/WileyCDA/Section/id-390082.html
American Society for Parenteral and Enteral Nutrition (ASPEN), 8401 Colesville Road, Suite 510, Silver Spring, MD 20910.
Printed in the USA by The Sheridan Group.
Nutrition in Clinical Practice accepts articles for Open Access publication. Please visit http://olabout.wiley.com/WileyCDA/
Section/id-828081.html for further information about OnlineOpen.

This journal is a member of the Committee on Publication Ethics (COPE) (http://publicationethics.org/about)


Nutrition in Clinical Practice
Volume 33 Issue 4 August 2018

Editorial
Editor’s Note 452
Jeanette M. Hasse, PhD, RD, LD, FADA, CNSC

Invited Reviews
Bacteria, Bones, and Stones: Managing Complications of Short Bowel Syndrome 454
Erika Johnson, MS, RD, CSR, LD, CNSC; Long Vu, MD;
and Laura E. Matarese, PhD, RDN, LDN, CNSC, FADA, FASPEN, FAND
Dietary Fermentable Oligosaccharides, Disaccharides, Monosaccharides, and Polyols (FODMAPs) 468
and Gastrointestinal Disease
Nimish Vakil, AGAF, FACG, FASGE
Nutrition Management of Necrotizing Enterocolitis 476
Vikram J. Christian, MBBS; Elizabeth Polzin, MBA, RD, CD, CNSC; and Scott Welak, MD

Reviews
Enteral Nutrition in the Management of Crohn’s Disease: Reviewing Mechanisms of Actions and Highlighting 483
Potential Venues for Enhancing the Efficacy
Moftah H. Alhagamhmad, MBBCH, DCH UK, PhD
Enteral Nutrition for Pediatric Crohn’s Disease: An Underutilized Therapy 493
Ala K. Shaikhkhalil, MD; and Wallace Crandall, MD, MMM

Clinical Observations
Case Report of Wernicke’s Encephalopathy After Sleeve Gastrectomy 510
Leslie A. Hamilton, PharmD, BCPS, BCCCP; Sarah H. Darby, BS; Allan J. Hamilton, MD;
Matthew H. Wilkerson, MD; and Kabel A. Morgan, MD
Copper Deficiency Myelopathy After Upper Gastrointestinal Surgery 515
Dominic King, MBChB; Keith Siau, MBChB; Latha Senthil, MBBS; Katherine F. Kane, MD;
and Sheldon C. Cooper, MD

Clinical Research
Reduction of Parenteral Nutrition and Hydration Support and Safety With Long-Term Teduglutide Treatment in Patients 520
With Short Bowel Syndrome−Associated Intestinal Failure: STEPS-3 Study
Douglas L. Seidner, MD, AGAF, FACG, CNSC; Ken Fujioka, MD; Joseph I. Boullata, PharmD, RPh, BCNSP, FASPEN;
Kishore Iyer, MBBS, FRCS, FACS; Hak-Myung Lee, PhD; and Thomas R. Ziegler, MD
Survey of Nutrition Management Practices in Centers for Pediatric Intestinal Rehabilitation 528
Anita M. Nucci, PhD, RD, LD; Kipp Ellsworth, MS, RD, CSP, CNSC; Austin Michalski, RD, LD, CNSC;
Emily Nagel, MS, RD, CNSC; and Jackie Wessel, MEd, RD, CNSC, CSP, CLE; on behalf of the ASPEN Pediatric
Intestinal Failure Section
Soluble Fiber Use in Pediatric Short Bowel Syndrome: A Survey on Prevailing Practices 539
Meredith Linley Harvie, MD; Margaret Alyssa Tucker Norris, MS, RD, LDN, CLC;
and Wednesday Marie A. Sevilla, MD, MPH, CNSC
Use of a Gastroschisis Feeding Guideline to Improve Standardization of Care and Patient Outcomes at an Urban 545
Children’s Hospital
R. Colby Passaro, MPH; Kate B. Savoie, MD, MS; and Eunice Y. Huang, MD, MS, FACS, FAAP
Impact of Gastrostomy Feeding Tube Placement on the 1-Year Trajectory of Care in Patients After Stroke 553
Janina Wilmskoetter, PhD; Annie N. Simpson, PhD; Sarah L. Logan, PhD; Kit N. Simpson, DrPH;
and Heather S. Bonilha, PhD, CCC-SLP
Strategies for the Enhancement of Nutrition Practice in a New York State Level 1 Trauma Center: A Hospital’s Journey 567
Lisa Musillo, MS, RDN, CNSC; Laryssa Marie Grguric, RDN, CNSC; Edward Coffield, PhD; Frank Aversano, MD;
Jeremy Bosworth, MD; and Richard Batista, MD
Gastrostomy Tube Feeding in Children With Developmental or Acquired Disorders: A Longitudinal Comparison 576
on Healthcare Provision and Eating Outcomes 4 Years After Gastrostomy
Ellen Backman, MSc; Ann-Kristin Karlsson, PhD; and Lotta Sjögreen, PhD

Letter to the Editor


Four-Oil Lipid Emulsion (Smoflipid) as a Tool in Managing Parenteral Nutrition Shortages 584
Lisa Mostafavifar, PharmD, BCPS, BCNSP; and David C. Evans, MD, FACS

Cover Art Note


The cover art depicts the human gastrointestinal tract; this issue of NCP tackles the nutrition management of some difficult
gastrointestinal disorders.
Cover art credit: 
C Getty Images

The ASPEN Rhoads Research Foundation depends on gifts from caring individuals to continue its work. Honor the memory of
a loved one or celebrate a special occasion by giving a gift to the ASPEN Rhoads Research Foundation.

We have now made it easier to support the foundation by providing a secure online donation form on our website:
www.nutritioncare.org. In addition to the online donation form, you can also charge your gift to a Visa, Mastercard, or American
Express by phoning our office, or you can mail a personal check directly to the ASPEN Rhoads Research Foundation.
Nutrition in Clinical Practice
Volume 33 Number 4
Editor’s Note August 2018 452–453

C 2018 American Society for

Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10183
wileyonlinelibrary.com

Serious digestive disorders and other conditions that im- necrotizing enterocolitis (NEC) among premature infants.
pede nutrient absorption often require intervention with This paper summarizes recommendations for PN support
nutrition support to restore and optimize nutrition status. when NEC occurs, recommended EN after uncomplicated
This issue of NCP features articles that tackle the nutri- NEC, and provides considerations for EN after bowel
tion management of some difficult gastrointestinal (GI) resection. The readers are encouraged to listen to the NCP
disorders. podcast featuring a discussion of this paper with 2 of its
Patients who have had extensive bowel resection often authors.
develop short bowel syndrome (SBS), accompanied by Symptoms of abdominal pain, distention, and bloating
disruptions in the production of bile acids and digestive in adults have been treated by modifying the content of
enzymes, and dilation and dysmotility of the small bowel. short-chain carbohydrates in the patient’s diet. Dr. Vakil
In this issue, Johnson, Vu, and Matarese discuss how these reviews the emerging literature on FODMAP, an acronym
conditions increase the risk of small intestinal bacterial for fermentable oligosaccharides, disaccharides, monosac-
overgrowth, further complicating nutrient absorption. The charides and polyols, as a treatment for GI disorders
authors also delve into how metabolic changes that occur in and raises awareness of a lack of information available
SBS can lead to bone demineralization and nephrolithiasis. to identify low-FODMAP-containing US enteral formu-
Two studies in this issue focus on SBS-related intestinal las. Continuing education credits are available for this
failure. Seidner and colleagues undertook the third (STEPS- article.
3) in a series of studies to monitor the safety and efficacy of Crohn’s disease (CD) causes inflammation in various
teduglutide as a therapy for a cohort of parenteral nutrition segments of the digestive tract, which can be painful and ac-
(PN)-dependent patients. They observed that treatment for companied by diarrhea. Over time, these symptoms lead to
up to 42 months was associated with sustained efficacy and poor intake, poor nutrient absorption, weight loss, and mal-
a reduction in the PN support needed to maintain fluid nutrition. Two papers in this issue address EN in the man-
balance or nutrition intake. agement of CD. Alhagamhmad reviewed the pathogenesis
In a Survey of Nutrition Management Practices in Centers of this disease and summarizes studies of the mechanisms
for Pediatric Intestinal Rehabilitation, Nucci et al collected of action for how EN may suppress intestinal inflammation
information to describe nutrition strategies used by special- and enhance mucosal healing, including modulation of
ists in pediatric intestinal rehabilitation at 14 medical centers gut microbiota. Similarly, Drs. Shaikhkhalil and Crandall
to promote gut adaptation and manage complications that reviewed the pediatric literature on the efficacy, advantages,
have not been previously summarized. Although many and methods of administration of EN in the treatment
enteral nutrition (EN) and PN management strategies were of CD. In addition, they discussed barriers to use of EN
relatively consistent, there was variability in laboratory test and suggested methods practitioners could undertake to
selection and monitoring frequency among practitioners. increase usage.
The authors encourage collaboration among centers to Nutrient deficiencies related to GI surgery are explored in
establish biochemical monitoring standards for pediatric 2 articles. Historically, Wernicke’s Encephalopathy (WE), a
patients with intestinal failure. Also surveying pediatric condition of thiamine deficiency, has been most commonly
practitioners, Harvie et al collected information to describe associated with chronic alcoholism. Recently, thiamine de-
the use of fiber to treat chronic high stool output in pediatric ficiency has also emerged as a concern following bariatric
patients with SBS treated in the US and abroad. For prema- surgery. Hamilton and colleagues describe a case of WE that
ture infants, most respondents (53.4%) did not initiate fiber developed in a young woman, 3 months after she underwent
until the patient was >40 weeks corrected age. Supplemental sleeve gastrectomy. Further concerning was the differential
fiber was added in the form of pectin (52.4%) or from pectin- diagnosis for symptoms of acute ischemic stroke, which
rich foods (green beans, sweet potatoes and bananas) and improved with the administration of IV thiamin. The au-
was discontinued if abdominal distention (67%), increased thors suggest there may be a role for prophylactic thiamin
emesis (43%), or bloody stools (38%) were observed or in the bariatric surgery patient population to prevent WE.
if stool output increased (37%) or significantly decreased King et al present a case of copper deficiency myelopa-
(33%). In a review of pediatric literature, Christian, Polzin, thy, which developed in a chronically malnourished, 50-
and Welak discuss how EN factors into recovery from year-old woman who had undergone upper GI surgery.
453

Magnetic resonance image with T2-weighted enhancement These and other nutrition support articles in this issue
revealed a subacute combined degeneration of the spinal summarize current practice and raise awareness of emerging
cord, which is characteristically associated with vitamin B12 research into how you might restore nutrients to overcome
deficiency. The patient was initially treated with copper- the incidence of malnutrition in patients with severe GI
containing PN followed by 3 monthly hydroxocobalamin disorders.
intramuscular injections and enteral supplements in the
form of copper amino acid chelate. To prevent deleterious
neurologic impairment, the authors suggest that follow
up monitoring of blood levels of copper after upper GI
surgery to be a prudent means of recognizing copper Jeanette M. Hasse, PhD, RD, LD, FADA, CNSC
deficiency. Editor-in-Chief, Nutrition in Clinical Practice
Invited Review

Nutrition in Clinical Practice


Volume 33 Number 4
Bacteria, Bones, and Stones: Managing Complications August 2018 454–466

C 2018 American Society for

of Short Bowel Syndrome Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10113
wileyonlinelibrary.com

Erika Johnson, MS, RD, CSR, LD, CNSC1 ; Long Vu, MD1 ;
and Laura E. Matarese, PhD, RDN, LDN, CNSC, FADA, FASPEN, FAND2

Abstract
Short bowel syndrome (SBS) occurs in patients who have had extensive resection. The primary physiologic consequence is
malabsorption, resulting in fluid and electrolyte abnormalities and malnutrition. Nutrient digestion, absorption, and assimilation
may also be diminished by disturbances in the production of bile acids and digestive enzymes. Small bowel dilation, dysmotility,
loss of ileocecal valve, and anatomical changes combined with acid suppression and antimotility drugs increase the risk of small
intestinal bacterial overgrowth, further contributing to malabsorption. Metabolic changes that occur in SBS due to loss of colonic
regulation of gastric and small bowel function can also lead to depletion of calcium, magnesium, and vitamin D, resulting in
demineralization of bone and the eventual development of bone disease. Persistent inflammation, steroid use, parenteral nutrition,
chronic metabolic acidosis, and renal insufficiency may exacerbate the problem and contribute to the development of osteoporosis.
Multiple factors increase the risk of nephrolithiasis in SBS. In the setting of fat malabsorption, increased free fatty acids are available
to bind to calcium, resulting in an increased concentration of unbound oxalate, which is readily absorbed across the colonic mucosa
where it travels to the kidney. In addition, there is an increase in colonic permeability to oxalate stemming from the effects of
unabsorbed bile salts. The risk of nephrolithiasis is compounded by volume depletion, metabolic acidosis, and hypomagnesemia,
resulting in a decrease in renal perfusion, urine output, pH, and citrate excretion. This review examines the causes and treatments
of small intestinal bacterial overgrowth, bone demineralization, and nephrolithiasis in SBS. (Nutr Clin Pract. 2018;33:454–466)

Keywords
short bowel syndrome; small intestinal bacterial overgrowth; metabolic bone disease; nephrolithiasis

Introduction increase and there is a gradual transition from aerobic to


anaerobic organisms that occur in the more distal sections
Short bowel syndrome (SBS) is the most common form of the bowel. Once across to the ileocecal valve, bacte-
of intestinal failure. It is a debilitating, complex disorder rial counts increase from 107 –109 organisms/mL in the
with multiple complications, some of which are potentially terminal ileum to approximately 1010 –1012 organisms/mL
life threatening.1,2 SBS results from the loss of portions of in the colon.5 In the proximal section of the small bowel
the intestines due to surgical resection or loss of intestinal (SB), only a few acid-resistant bacteria, mainly Lactobacilli,
function.2,3 Regardless of the reasons for the resection, the Streptococci, and Escherichia coli exist.6 In the jejunum and
result is an inability to maintain fluid and nutrient balances
through the consumption of a normal diet.2 The result-
ing malabsorption can cause numerous catheter, liver and From the 1 Center for Human Nutrition, Digestive Disease and
Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA; and
biliary, metabolic, renal, bone mineralization, or financial 2 Department of Internal Medicine and Surgery, Brody School of
and quality-of-life complications.1,3,4 This review focuses Medicine, East Carolina University, Greenville, North Carolina,
on bacterial overgrowth including d-lactic acidosis (DLA), USA.
metabolic bone disease (MBD), and nephrolithiasis. Financial disclosure: None declared.
Conflicts of interests: None declared.

Bacteria This article originally appeared online on June 21, 2018.


Corresponding Author:
Microbiota Changes in SBS Laura E. Matarese, PhD, RDN, LDN, CNSC, FADA, FASPEN,
FAND, Department of Internal Medicine and Surgery, Brody School
There are changes to the intestinal microbiotia of pa- of Medicine, East Carolina University, 600 Moye Blvd., Vidant MA
tients with SBS when compared with that of healthy nor- 342, Mail Stop 734, Greenville, NC 27834, USA.
mal adults. In the nonresected intestine, bacterial counts Email: mataresel@ecu.edu
Johnson et al 455

colon, numerous and predominantly anaerobic microbial in production of lactate by the bacteria overwhelm the
organisms reside, such as Bacteroides, Bifidobacteria, and buffering capacity of the exocrine secretions of the pan-
Clostridia, with Lactobacilli making up <1%.7 creas, resulting in a low pH throughout the entire resected
In patients with SBS, the overall amount of bacteria bowel.6
are found to be quantitatively similar, but the difference The loss of the ileocecal valve was once thought to
is noticed in the variety of microbial species. The changes promote SIBO because it acted as a barrier to the reflux of
observed in a resected intestine is that the bacterial diversity material from the colon into the SB. However, it has been
is noticeably reduced, and there is a shift between dominant suggested that the ileocecal valve does not independently
and subdominant organisms. For example, after a small affect the risk of SIBO.9 Instead, the increased risk for
bowel resection, the microbiota of fecal matter and the SIBO is thought to be related to reduced ileal length,
remaining distal intestine becomes depleted in the variety impaired peristalsis, and gastrointestinal acidity.5 The use
of Bacteroides and Clostridium species; Lactobacilli become of antimotility medications may also play a role.10,11
more predominant in the entire remaining intestine.7,8 An-
other observed change is the presence of some strains such Clinical significance. SIBO may contribute to the onset of
as Lactobacillus mucosae, which have never been accounted malnutrition, mucosal injury, and onset of symptoms. One
for in the intestinal microbiota of healthy human adults, of the most significant nutrition implications is the risk
but have been found in SBS patients.8 In the setting of of developing malnutrition associated with malabsorption.
SBS, the microbial contents of the colon and the metabolic This can occur when deconjugation of bile acids in the
capabilities of the microbiota have an influence on a variety proximal SB disrupt fat digestion and lead to the production
of intestinal functions and become an essential component of lithocholic acid.12 Under normal conditions, conjugated
in the course of management. bile acid enter the gastrointestinal tract in the duodenum
and are important in the emulsification, digestion, and ab-
Small Intestinal Bacterial Overgrowth sorption of dietary fat that occur in the SB. Deconjugation
and subsequent dehydroxylation of the bile acids result in
Pathophysiology. In SBS, anatomical changes, fast tran- the formation of the secondary bile acids deoxycholic acid
sit, malabsorption, and hyperacidity occur after massive and lithocholic acid.13 Bacteria such as Lactobacilli are able
resection, and this creates an optimal condition for the to deconjugate bile acids, and as previously stated, there
alteration of bacteria throughout the intestine. Small in- is an increased amount of Lactobacilli in the intestines of
testinal bacterial overgrowth (SIBO) may occur when there patients with SBS. Therefore, conjugated bile acids entering
is a disproportional or excess of a particular species of the SB will be rapidly deconjugated.6 As a result of the
bacteria in the proximal SB, this does not necessarily equate deconjugation of bile acids by Lactobacilli, there is only a
to harmful pathological consequences. In fact, there are small amount of conjugated bile acid available, leading to
some benefits to having microbiota, even in excess. Take, insufficient concentrations for fat absorption.
for instance, the role of bacteria in the energy salvage Another major pathophysiologic consequence of SIBO
of a patient with SBS. In the presence of a colon in relates to the inflammatory epithelial changes that sub-
continuity, undigested carbohydrates pass into the colon sequently occur in the mucosa. Lithocholic acid, which
and are fermented by colonic bacteria into short chain is produced from deconjugation of bile acids, is toxic to
fatty acids and other organic acids. These fatty acids are enterocytes. Mucosal injury may also result from bacterial
absorbed by the colon and provide additional energy as well adherence, production of enterotoxins, and other metabolic
as enhance absorption of sodium and water. In pathologic products produced by bacteria. Enterocyte injury leads to a
cases of SIBO, elevated bacterial counts have been found in loss of activity of brush-border activity, villous atrophy, and
the proximal SB, commonly with bacterial species including altered permeability, the latter predisposing to the devel-
Streptococci, Bacteroides, Escherichia, and Lactobacilli.5 In opment of a protein-losing enteropathy, impaired immune
SBS patients, the dominant bacteria becomes Lactobacilli, defense, and bacterial translocation. Biopsy specimens of
which is rather harmless, but an invasion of the SB with the SB may provide the best evidence of the harmful impact
an excessive or imbalanced population does carry a risk of of the bacteria present. Biopsy provides a visual of the
developing pathological SIBO. This can cause inflammation inflammatory changes, villus blunting, and the presence of
and exacerbate malabsorption. adherents or intracellular bacteria, and it can also support
With the initial aid of gastric acid, Lactobacilli survive the diagnosis of pathologic SIBO.5
past the stomach and are present in the proximal SB. As a The consequences of malnutrition and inflammation
result of the malabsorption of nutrients, carbohydrates are caused by SIBO are largely responsible for the variety
more abundant for Lactobacilli to ferment into lactic acid of symptoms that occur. Common symptoms associated
and further contribute to the acidic environment optimal with SIBO include gas, bloating, and abdominal discomfort
for the sustained survival of Lactobacilli. Even the increase caused by the fermentation of undigested carbohydrates by
456 Nutrition in Clinical Practice 33(4)

Table 1. Symptoms Associated With Small Intestinal An increase in hydrogen in the breath sample after oral
Bacterial Overgrowth. ingestion of the carbohydrate substrate, typically 10–20
r ppm, indicates SIBO.5,12,14 However, the test results will
Increase flatulence
r Bloating
be flawed in the setting of malabsorption because of the
r Abdominal discomfort delivery of undigested substrate to the colon, making it im-
r Foul smelling stools possible to differentiate between SB and colonic production
r Explosive diarrhea of hydrogen. Despite their popular use, hydrogen breath
r Steatorrhea tests are not recommended for use in testing for SIBO in the
r Frothy, foamy stools SBS population because of the number of high false positive
r Decrease in oral intake
r and false negative rates.
Micronutrient deficiencies (vitamins A, B1, B12, D,
and E and iron)
Because the majority of patients with SBS present with
multiple predisposing factors to SIBO, clinicians often forgo
the diagnostic testing and presume SIBO based on the
bacteria (Table 1). Other symptoms that occur as a result of presence and strength of symptoms and will often treat
malabsorption include steatorrhea and fat-soluble vitamin empirically.
deficiencies. Vitamin B12 is another nutrient deficiency that
can occur because of the use of the vitamin by anaerobic DLA
bacteria. This deficiency can result in megaloblastic anemia
or neurological symptoms. Secretory diarrhea may occur Pathophysiology. DLA is a rare syndrome that occurs in
when bile acid absorption is disrupted, which happens when individuals with SBS and an intact colon. Alteration of
lactobacilli deconjugate the bile acid.13 the colonic microbiota plays a major role in the production
of D-lactate. Development of this syndrome occurs when
Diagnosis. SIBO is defined as a bacterial colony count there is an excess production of D-lactate that exceeds the
>100,000 colony-forming units per millimeter (105 clearance of D-lactate and leads to an accumulation of this
CFU/mL) from the direct measurement of jejunal fluid or substance.
the presence of bacteria from colon or oropharynx in the SBS can cause an excess production of D-lactate due
SB.14 Nonspecific symptoms may suggest the presence of to the malabsorption of carbohydrates. This occurs when a
SIBO such as increase in watery or frothy stool output, large quantity of undigested carbohydrates reach the colon,
especially if associated with bloating, flatulence, and the bacteria in the colon ferment the undigested carbohy-
abdominal pain. A physical exam can show abdominal drates to SCFA, lactic acid and other organic acids. Due
distention with an audible succussion splash, which can to the larger than normal amount of carbohydrates that
indicate fluid and gas in the distended bowel loops.5 There reach the colon, there is a greater than normal production
is, as of yet, no widespread agreement as to the optimal of organic acids which lower the luminal pH of the colon.
test for SIBO, especially in the setting of SBS. The gold The low pH alters the colonic microbiota and supports the
standard, although rarely used in practice, for diagnosing survival of acidophilic bacteria that favor production of
SIBO is a quantitative culture of aspirated SB fluid.5 lactic acid. As populations of both D-lactic-acid producing
However, this test is not without significant limitations. and L-lactic-acid producing bacteria increase in the colon,
The test is relatively expensive, as it involves endoscopy or the pH continues to decline, progressively promoting the
fluoroscopy to place the aspiration catheter; there is a risk growth and survival of lactic acid producing bacteria,
for contamination of the specimen during its transit into including bacteria such as Lactabillus fermenti, Lactobillus
the SB; and it requires a detailed bacteriologic analysis, acidophilus, and Streptococcus, all of which produce D-
which is especially difficult with anaerobic organisms. It has lactate. Eventually, the gut flora is predominated by lactic
been reported that only about 40% of the total gut flora acid producing bacteria, and in some cases primarily those
can be identified using conventional culture methods.11 that produce D-lactate. Once the intestinal flora has a
As a result of the invasiveness of the direct method of high concentration of D-lactate producing bacteria, large
measurement, several indirect tests have been developed to amounts of carbohydrate, particularly readily fermentable
test for SIBO. These tests are widely used as a surrogate simple sugars, reaching the colon will result in a high
to direct aspiration. Hydrogen breath tests have become production of D-lactate.15 Carbohydrate malabsorption
the most commonly used because of their low cost and can alter gut microbiota, predisposing patients with SBS
relative simplicity. The hydrogen test uses a carbohydrate to SIBO, and SIBO can cause malabsorption of carbo-
as a substrate, such as glucose, lactulose, or xylose. How hydrates further supporting the development of D-lactate
the test works is that the excessive bacteria in the SB production.
will metabolize the substrate releasing hydrogen that is In addition to excess production of D-lactate, in the
subsequently absorbed and then released in expired air. setting of SBS, there is potential for impaired clearance of
Johnson et al 457

Table 2. Symptoms of D-Lactic Acidosis.13,15 available but assays are sent out since so infrequently used
and it can take weeks for results to be available. It is not
r Encephalopathy
r recommended to wait for the results to confirm elevated D-
Slurred speech
r Gait disturbance
lactate levels before treating. A thorough work up should
r Weakness rule out other indications that might be implicated such as
r Tachypnea diabetic ketoacidosis, hepatic encephalopathy, iron toxicity,
ingestion of toxic substances, or uremic acidosis in advance
chronic kidney disease.
It might be difficult to measure elevated D-lactate levels
it. D-Lactate is metabolized to pyruvate through the enzyme when a patient is having neurological symptoms because the
D-2-hydroxy acid dehydrogenase (D-2-HDH). The highest symptoms are transient and the patient may not be available
concentration of D-2-HDH is found in the kidney and liver. to collect a urine or blood sample. Often times a diagnosis
These 2 organs are often compromised in SBS patients, is suspected during an office visit, after a patient reports the
potentially reducing D-2-HDH concentration and D-lactate symptoms during a discussion with a clinician.
metabolism. Another potential for impaired D-lactate
metabolism, involves oxalate, which is a potent inhibitor of Treatment for SIBO and DLA
D-2-HDH and is often elevated in the blood of patients
with SBS.15-18 The inhibition of D-2-HDH by oxalate Nutrition therapy. The diet for SBS remains an important
and the potential impact of oxalate upon renal function, component for managing SIBO and DLA. The usual dietary
may reduce metabolism and impair urinary excretion of modifications to reduce stool output should be emphasized
D-lactate. with the patient, and maintaining adequate hydration is
important. Patients are still recommended to limit simple
Clinical significance. A key neurological symptom experi- sugars such as cakes, cookies, and candies. Restriction of
enced in patients with DLA is encephalopathy and this may readily fermentable simple carbohydrates should combine
pose a diagnostic challenge for clinicians since the presenta- with a modest amount of complex carbohydrates with 4–6
tion can be mistaken for another neurological condition.18 small frequent meals to avoid exposure of the gut flora to
Other neurological symptoms include altered aggressive large boluses of undigested carbohydrates. Fats should not
behavior, slurred speech, and ataxia, with patients often ap- be restricted, unless they provoke steatorrhea and watery
pearing drunk. The actual etiology of neurologic symptoms diarrhea. In which case, a moderate-fat diet should be
seen in DLA is somewhat unclear. It appears that acidosis implemented and combined with medical therapies. For
and elevation of blood D-lactate levels alone are not capable patients with significant bile acid loss, from a terminal ileum
of producing the observed neurological manifestations. The resection, and a colon in continuity, bile acid resins could ac-
neurological symptoms do not always correlate with the company a moderate-fat diet. Pancreatic enzymes could be
level of D-lactate, and it is unsettled whether it is D-lactate combined with a moderate fat diet in patients with or with-
or a simultaneously produced metabolite that causes the out a colon present. In extreme cases, SIBO may complicate
syndrome of intoxication19 (Table 2). From our clinical adequate oral intake and pathologic bacterial may cause
experience, SIBO and DLA appears to affect their quality mucosal damage, so nutrition support may be required.
of life and ability to wean parenteral nutrition (PN). For patients who experience DLA, the recommendation
to restrict simple sugars is a preventive therapy. During
Diagnosis. A high level of suspicion is key in diagnosing an acute episode of DLA, treatment involves withholding
DLA in patients with SBS, an intact colon in continuity, that enteral sources of carbohydrate and remaining nil per os or
present with classic neurological disturbances. The level of nothing by mouth until severe neurologic symptoms resolve.
suspicion should be raised if the patient recently received To correct acidosis, intravenous bicarbonate and fluid can
a course of antibiotic therapy or started a probiotic, both be administered, but lactated ringer’s solution should be
of which can alter the intestinal flora. A diet history will avoided because they contain both D-lactate and L-lactate.
frequently reveal ingestion of relatively large amounts of If the patient is at risk or has severe malnutrition, with
carbohydrates in the form of simple sugars prior to the onset an anticipated prolonged oral food restriction, then PN
of symptoms. support is warranted; carbohydrate provided parenterally
If a patient presents with the described clinical fea- does not increase D-lactate levels.20,21 Once patients recover
tures and is acidotic, a diagnosis can be confirmed with a from neurologic symptoms, PN should be transitioned to
normal L-lactate level and in increase in urine or serum enteral feedings.
D-lactate level > 3mmol/L.16 Conventional lab assays for Micronutrient deficiency and their associated symptoms
lactate usually only measure L-lactate, which is why the are common in patients with SBS, and supplementation will
test is usually normal in DLA. D-lactate measuring kits are be necessary. For reasons described previously, in the setting
458 Nutrition in Clinical Practice 33(4)

of SIBO, the replacement of fat-soluble vitamins A, D, and of developing DLA. However, these claims are made in the
E are likely as well as vitamin B12 and iron. Because of the absence of quality scientific data.22
possibility that a problem with pyruvate metabolism may One of the goals of treating SIBO is to reduce the risk
be involved in the development of neurologic symptoms in of an overproduction of D-lactic acid, which in turn would
DLA, thiamine status should be assessed and supplemented reduce the risk of developing an episode of DLA. This is
if indicated.16 An oral calcium supplement is recommended done by attempting to reduce the number of pathogenic
for patients with SBS with a colon in continuity, not to bacteria present and to reduce the count of D-lactic acid
correct deficiencies but, rather, as an oxalate binder to producers. Although DLA could occur without being ex-
reduce the risk of increased free oxalate in circulation. acerbated by a course of antibiotics or the initiation of
a probiotic, the use of these therapies has been linked to
DLA episodes. Although it is reasonable to treat SIBO and
Medical therapy. SIBO is implicated in most cases of DLA, DLA with antibiotic and probiotic therapies, the results
therefore the prevention of this overgrowth is important with chronic preventive therapy have not been consistent
in the long-term management and prevention of DLA. and are unpredictable.
Medications associated with or known to inhibit intestinal The following antibiotic therapies are used to treat SIBO
motility or the inhibition of gastric acid secretion should and acute flares of DLA: tremethoprim-sulfamethoxazole,
be eliminated or substituted when treating SIBO. This is neomycin, ciprofloxacin, vancomycin, and metronidazole.
difficult in the SBS population because a majority of the However, each of these antibiotics has also been associated
treatment involves antimotility, antidiarrheal, and antise- with the onset of DLA,15,23 so use with caution is advised
cretory medications to manage malabsorption and diarrhea (Table 3).
and control gastric acid hypersecretion after a SB resection. Oh et al24 reported the beneficial effects with the antibi-
However, after resection, the remaining bowel progresses otic neomycin in treating DLA. In this case, the patient was
through several phases of adaptation, and the remaining able to remain free of recurrent episodes for at least 1 year.
bowel begins to increase absorptive function. During this In a different report, neomycin was not effective in reducing
time, absorption improves and it may be necessary to the clinical symptoms of DLA nor reducing Lactobacilli.6
reassess the need for antimotility and antisecretory medi- Rifaximin is increasingly used in the treatment of SIBO
cations and discontinue use if possible because this could and has been shown in controlled studies to be a good choice
reduce the risk of developing SIBO for some individuals. for management.11 In at least 1 trial, rifaximin showed a
When medication correction is not an option, microbial higher SIBO decontamination rate than metronidazole.25
modification is attempted with antibiotic and probiotic However, even rifaximin appears to come with a degree
therapies. Currently, there are very few well controlled of doubt because it has been discussed that lactic acid
trials for the treatment of SIBO, so recommendations are bacteria can grow at high levels, even in the presence of high
based on clinical experience, and they are purely empirical concentration of intraluminal rifaximin.17
approaches. This is done with caution because antibiotic In 1 of the original cases of DLA reported in a hu-
therapy can increase or decrease D-lactate production. man, a probiotic, specifically Lactobacillus acidophilus, was
A suggestion is to start with a single 7–14 day course of started prior to the onset of the episode.24 Since that first
antibiotic, preferably one that is nonabsorbable and covers report was published, other reports have been published
both aerobic and anaerobic organisms. When patients ex- implicating probiotics in cases of DLA and also being used
perience a reoccurrence of symptoms, a repeat course of successfully in the prevention of recurrent episodes.26,27 In
antibiotics is needed. Rotating among different antibiotic fact, some newer studies are showing promising results of
regimens is recommended to prevent the development of a combination of antibiotics and probiotic therapies.28,29
resistance. There is also a growing number of studies that are pro-
Because there is concern regarding antimicrobial resis- viding preliminary evidence that support probiotics as an
tance, antibiotic allergic reactions, and Clostridium difficile effective prevention and treatment option for SIBO.10,27
diarrhea associated with the prolong use of antibiotics, there With all that potential comes the need for well-designed,
is growing interest in the use of probiotics. Despite current randomized controlled trials testing for strain-specific ef-
popularity and a growing number of experimental studies fects that may support specific claims. In addition to
that have shown potential benefits to probiotics, the role of finding an effective therapeutic agent, it is also important
these agents in the management of SIBO remains unclear. to determine the duration and frequency of treatment
There is considerable discrepancies when it comes to the administration.22
use of probiotics to enhance outcomes of managing SIBO Surgical options. Among patients with SBS, medical and
and DLA. Among clinicians, there is a common practice antibiotic therapies may fail, indicating the need for surgical
to avoid Lactobacillus strains of probiotics in SBS with a strategies. Options include bowel tapering, lengthening, or
colon in continuity because of the belief that there is a risk resection (Table 4).
Johnson et al 459

Table 3. Antibiotic Use With Small Intestinal Bacterial Overgrowth and D-Lactic Acidosis in Patients With Small Bowel
Syndrome.

General Guidelines Antibiotic Adult Dose

r No standard protocol Single-course regimens (7–14 days)


r May increase or decrease d-lactate Amoxicillin-clavulanate 500 mg 3x/day
r Use with caution Ciprofloxacin 500 mg 2x/day
r Alternate between 2 different Rifaximin 550 mg 3x/day
single antibiotic regimens Metronidaxole 500 mg 3x/day
Trimethoprim-sulfamethoxazole 1 double-strength tablet 2x/day

Table 4. Treatment Options for Small Intestinal Bacterial Table 5. Risk Factors for Metabolic Bone Disease.30
Overgrowth and D-Lactic Acidosis in Patients With Short
Bowel Syndrome. Nonmodifiable Modifiable

Diet modification r Female sex r Dietary deficiencies


r 4–6 small meals r Older age in vitamin D,
r Simple sugar restriction (particularly calcium, phosphorus
r Low to moderate fat menopause) r Tobacco use
r Lactose free, as needed r Race (White and r Alcohol use
r Replacement of micronutrient deficiencies Asian) r Elevated body mass
(fat-soluble vitamins, B1, B12, iron) r Family history of index
r Calcium supplement with colon in continuity metabolic bone r Decreased physical
Probiotics and antibiotics disease activity
r Empirical trials
r Role of probiotics remain unproven
r Use cautiously in patients with a history of D-lactic constitute a dynamic organ that is constantly being broken
acidosis down and remade anew. Osteoclasts of the mononuclear
Other medications monocyte–macrophage precursor lineage break down and
r Re-evaluate need for proton pump inhibitor 6
resorb old bone, preventing long-term build-up of stress
months postoperatively small bowel resection
r Re-evaluate need for antidiarrheal, antisecretory, and damage. Meanwhile, osteoprogenitor cells develop the os-
antimotility agents from time to time teoblasts that generate new protein matrices that form a
Surgical framework for bone mineralization. This bone remodeling
r When no medical or dietary modification treatments takes place in both the dense outer cortical bone as well as
are effective the spongy inner trabecular bone. The overall bone mass
r Bowel tapering, lengthening, or colon removal and density are the major determinants of bone strength.32

Epidemiology. Decreased bone strength leads to an in-


creased likelihood of bone fractures, which are a major
Bones cause of morbidity and mortality particularly in the elderly
MBD population. The overall prevalence of osteoporosis in the
U.S. population aged older than 50 years is 10.3% for
A spectrum of disease. MBD affects bone strength through 10.2 million adults, and an additional 43.4 million adults
a spectrum of disease from compromised bone density are thought to have osteopenia.33 These fragile patients
as seen in osteopenia and osteoporosis to defective bone generate >1.5 million fractures annually.34 Patients with
mineralization as seen in osteomalacia. Risk factors for hip fractures in particular have a 3-fold increase in mor-
MBD include both nonmodifiable and modifiable charac- tality risk linked to fracture-related complications such as
teristics (Table 5).30 The poor bone mineralization seen in thromboembolism, infections, and heart failure.35 Those
osteomalacia is usually caused by vitamin D deficiency or patients that survive continue to have increased morbidity
another pathologic process that ultimately results in vitamin with limitations in mobility, basic activities of daily living,
D deficiency such as phosphate wasting disorders, renal self-care, and quality of life as up to one-third require
losses, metabolic acidosis, and malabsorption.31 placement in assisted living or nursing home support.34,36
The treatment of osteoporosis and recovery from fracture
Bone physiology. Rather than forming a completely rigid also comprises a significant financial burden in the U.S.
and unchanging structure, the bones of the skeletal system elderly, estimated at 16 billion dollars in 2002.34
460 Nutrition in Clinical Practice 33(4)

Diagnosis. Although conventional plain-film radiographs often undergo bowel resections with abdominal malignancy,
can help detect complications of low bone mineral density which can impact their ability to intake enough calcium
(BMD) such as fractures, it has poor sensitivity to detecting from anorexia and malnutrition as well as direct mal-
early changes of MBD. The dual-energy X-ray absorpti- absorption. Other malignancies can cause hypogonadism
metry scan has long been considered the gold standard and paraneoplastic syndromes that can directly affect cal-
for diagnosing osteoporosis. It is precise, accurate, has low cium metabolism and worsen MBD.41 For patients with
radiation exposure, and is relatively inexpensive.8 The test inflammatory bowel disease, mucosal inflammation of the
generates 2 measures, a t-score and z-score, for several intestines can lead to malabsorption, and the elevation of
different bone sites. The t-score is the number of standard cytokines is believed to stimulate osteoclast activity, causing
deviations between the patient’s BMD and the BMD of a more bone turnover.42 The avoidance of trigger foods such
healthy 30-year-old. The z-score is a similar but compares as dairy products could lead to vitamin D and calcium
the patient’s BMD to the BMD of people similar in age, deficiency as well. Furthermore, corticosteroid use has been
race, and sex. The t-score is the most relevant number strongly linked to the development of osteoporosis in the
for osteoporosis, which is defined as a t-score ࣘ −2.5. general population and certainly puts patients prescribed
Osteopenia is diagnosed with a t-score between −1.0 and with frequent steroid tapers for inflammatory bowel disease
−2.5. The t-scores >−1 are considered normal BMD.37,38 flares at risk for MBD.41,44

MBD in SBS Deficiencies. SBS can lead to deficiencies in protein, cal-


cium, magnesium, and vitamin D, which are all important
Prevalence. Although there are some congenital causes of for optimal bone health.42 There is a high prevalence of
SBS, the majority of adult cases are caused by extensive vitamin D deficiency as noted by Ellegård et al45 in a
surgical resection for various diseases, including mesen- Swedish cohort of 106 patients with intestinal insufficiency
teric infarction, Crohn’s disease, radiation enteritis, and or intestinal failure in which 67% of the patients were
intestinal volvulus. The decreased anatomical length of deficient (defined as <50 nmol/L) and 88% were found to
bowel creates a reduced intestinal absorptive surface area have low bone density. These results are consistent with a
that can lead to increased losses of fluids and electrolytes, similar Danish study of 167 patients that found vitamin
compensatory restriction of enteral nutrition to reduce D deficiency in approximately 50% of the patients and
losses, increased gastrointestinal tract transit time, and the osteoporosis in 56.9%.40 This relationship of vitamin D
development of SIBO.39 The failure of the intestines to deficiency and low BMD also holds in different populations
absorb an adequate amount of vitamin D and calcium around the world as seen in studies from Canada and
predisposes the SBS patient to osteoporosis, and that risk China, and often the vitamin D levels remain low despite
is higher than for the general population. In a 2017 study, supplementation.46,47
Nygaard et al40 examined a population of 167 patients Deficiencies in other related vitamins and minerals such
with chronic intestinal failure, of whom 80% had SBS. as vitamin K, vitamin C, copper, and fluoride can play a role
The prevalence of osteoporosis in these patients was 56.9% in SBS leading to MBD. Vitamin K is typically synthesized
compared with 24.2% in the control group for an odds ratio by normal gut flora that can be disrupted in SBS. Vitamin K
of 4.2; a further 31.7% of patients had osteopenia.40 Almost is more commonly known for its role in blood coagulation,
90% of these patients with SBS had MBD. but the vitamin K-dependent proteins osteocalcin, matrix
Gla protein, and protein S are all involved in the process of
Risk factors. The vast majority of patients with SBS have bone formation, so a deficiency in vitamin K can also impact
some form of MBD from osteopenia to osteoporosis. Mal- bone metabolism and decrease bone mineralization.42 In a
absorption of vitamin D and calcium certainly is an integral retrospective study of 189 Canadian patients on home PN
component, but other aspects of the disease play crucial (HPN), a trend was noted for higher hip and lumbar spine
roles in the development of MBD. The short bowel length t-scores for patients who had vitamin K supplementation
decreases fluid absorption time, leading to chronic diarrhea when compared with those who had no supplementation.48
and loss of bicarbonate-rich fluid, and can cause a nonanion
gap metabolic acidosis. This situation can be compounded MBD With HPN
further by the development of SIBO causing anion gap
DLA. In addition, renal calcium wasting can further ex- Prevalence. For patients with SBS, malabsorption and di-
acerbate calcium depletion via an extraintestinal route.41,42 arrhea often limit giving nutrition via enteral route. The use
The binding of calcium to fatty acids via saponification and of HPN is common and allows for the provision of calories,
generating Ca-soap can inhibit calcium absorption as well.43 vitamins, and other nutrients via intravenous route as a
Other disease states requiring intestinal surgery that bridge to intestinal surgery or transplant or even continued
leads to SBS can also be risk factors for MBD. Patients indefinitely. A cross-sectional and retrospective multicenter
Johnson et al 461

study of 165 European patients on long-term HPN for a guidelines recommend keeping aluminum to a minimum,
duration >6 months found a high prevalence of reduced ideally <25 mcg/L.51
BMD, with a t-score at any site <−1 in 84% and t-scores The European Society for Clinical Nutrition and
ࣘ−2.5 in 41%.49 MBD is common among patients using Metabolism guidelines also suggest adding vitamin D to
HPN and often may go underdiagnosed without adequate PN for patients with intestinal failure as so many of them
screening in this patient population. have MBD or are at high risk for MBD; the recommended
intravenous dose is 200 international units, which is typically
provided in the form of a multivitamin supplement.52
Risk factors. In addition to the numerous risk factors for
Additional supplementation of vitamin D is indicated in
MBD from SBS, there are risk factors for MBD from PN
patients with low 25-hydroxyvitamin D levels and high
as well that affect calcium excretion. Adequate amounts of
PTH.45 Unfortunately, there are no intravenous formula-
calcium and phosphorus are required, although the levels
tions of vitamin D available in the United States that can
that can be provided in solution are limited by calcium-
be added to PN aside from that found in the multivitamin,
phosphate precipitation.41 The proteins supplied by amino
so additional supplementation is given by enteral route. It
acids constitute a titratable acid, and their low pH will
should be noted that vitamin D is a negative acute phase
also increase urinary calcium excretion. Higher levels of
reactant and care should be taken when checking levels in
sodium will increase the renal glomerular filtration rate and
patients with acute inflammation.53 Interestingly, in patients
thus increase urinary calcium excretion, and higher levels
with low BMD, low PTH, and low 1,25-hydroxyvitamin D
of dextrose will increase serum insulin concentrations and
levels, but a normal 25-hydroxvitamin D level, the vitamin
increase calcium excretion as well.42 In the home setting,
D supplement can be temporarily withdrawn with levels
patients often perform cycling of their HPN infusion,
rechecked at yearly intervals. The removal of vitamin D
reducing duration from 24 hours to >12 hours, which has
from PN did not have adverse effects and is actually shown
been shown to increase urinary calcium excretion. Although
to significantly increase bone mineral content.54
the exact mechanism is unknown, it is likely related to
the increased rate of HPN infusion when the duration is
Long-term HPN. Despite the presence of risk factors for
shortened.41
MBD from PN, many studies have shown that long-term
Other factors in PN that affect BMD include those that
PN does not exacerbate osteoporosis. A Danish study
adversely alter bone metabolism. Magnesium has an effect
followed 75 patients receiving HPN from 1995 to 2003.
on parathyroid hormone (PTH) secretion and renal activity
The mean duration of HPN was 4 years, and patients
that plays an important role in calcium homeostasis.50
received an average of 4.4 dual-energy X-ray absorptimetry
Heparin-associated osteoporosis can also occur, particu-
scans during the study period. There was a moderate but
larly for those who require treatment for deep venous
statistically significant 1% decrease in BMD, but this loss
thrombosis, which is a common complication with central
was not significantly greater than that seen in age-matched
intravenous lines used for PN access.42 Aluminum is a
and sex-matched healthy controls.55 In some studies, there
common element found in many components of PN, and
was actually an increase in BMD, such as the multicenter
its toxicity can impair PTH secretion and decrease levels of
study by Pironi et al,56 which followed 65 patients on HPN
activated vitamin D.41
during an average of 18 months and found a statistically
significant increase in z-score at the lumbar spine. A French
PN preparation guidelines. Given the risk factors for MBD prospective study followed 56 patients with intestinal failure
from PN, it is imperative that PN formula preparations are on HPN for 5 years of whom 67% had osteoporosis at
adjusted to avoid worsening of MBD. Calcium gluconate baseline and found an improvement in trabecular BMD,
should be provided at 15 mEq/day and with phosphate in a particularly in patients whose onset of intestinal failure was
ratio of 1:2, for 15 mEq calcium to 30 mmol phosphorus in adulthood.57
in the PN solution. Magnesium should be dosed around
15–20 mEq/day, depending on the volume of diarrhea.41 Management of MBD
Amino acids should not exceed 1.5 g/kg/day for short
term and upon resolution of critical illness, should be Patient evaluation. Evaluation of MBD should incorporate
reduced to 0.8–1.0 g/kg/day to avoid metabolic acidosis and a thorough history and physical examination as well as
hypercalciuria.42 Acetate levels in the PN solution are kept relevant blood work. Many patients with osteoporosis do
to a goal serum bicarbonate in the mid-range of normal not have symptoms, but a history of bone fractures from
to also avoid acidosis.41 Aluminum toxicity was a larger minor trauma or vertebral compression fractures is suspect.
problem in the 1980s with the use of casein hydrolysate as a Patients can complain of skeletal bone pain or proximal
protein source, although some PN additives still have a small muscle weakness. A family history of osteoporosis may also
amount of aluminum contamination.42 For all patients, the be present. Social history including tobacco and alcohol
462 Nutrition in Clinical Practice 33(4)

use must also be elicited. Secondary causes of osteoporo- forearm, but there was a trend for increasing BMD in
sis include Cushing’s syndrome, hyperparathyroidism, and other bone sites.60 A Canadian case series of 11 patients
hypogonadism in men, so signs of these conditions should also noted an improvement in hip t-score with the use of
be investigated on physical exam. A medication history is intravenous palmidronate.61
essential as corticosteroids, estrogens, and loop diuretics can For patients who are intolerant of bisphosphonates, an
have an impact in MBD.41 alternative therapy is teriparatide, which is recombinant
Laboratory work should consist of studies to support the PTH. This medication is given by subcutaneous route and
history and physical including basic metabolic panel, mag- has a different mechanism of action; rather than inhibit
nesium, and phosphorus levels. PTH, thyroid-stimulating osteoclasts, it stimulates osteoblasts to increase bone
hormone, and 25-hydroxyvitamin D levels can be checked, anabolism. There is a case report of a patient intolerant
particularly if patients have malabsorption. Commonly of oral alendronate on teriparatide for 18 months whose
the recommendation is for a 24-hour urine calcium to be spinal t-score increased from −2.5 to 0.62 The patient’s
monitored every 6–12 months to check for excess urinary BMD normalized from the osteoporotic range.
calcium excretion.41 However, many impracticalities exist Newer pharmacologic therapy includes the RANKL
in the home setting with adequate urine collection and re- inhibitor denosumab, so named because it targets the
moving calcium supplements prior to collection. In addition RANKL, a cytokine that is responsible for osteoclast func-
to biochemistry, European Society for Clinical Nutrition tion and differentiation. By binding RANKL, denosumab
and Metabolism guidelines recommend routine monitoring interferes with its ability to bind the receptor RANK,
of BMD with dual-energy X-ray absorptimetry starting at shutting down osteoclasts and inhibiting bone resorption.63
baseline and then yearly intervals, although this scanning Denosumab has been shown to reduce hip fractures in post-
frequency is not well supported by studies at this time.52 menopausal women and has also been studied in patients on
Patients should ideally be followed longitudinally using the HPN with intestinal failure. A total of 15 patients assessed
same scanner every time to increase precision and minimize on denosumab after 1 year were found to have significant
variation in measurement and interpretation.37 improvement in t-scores and z-scores of the lumbar spine
and femur compared with no difference found for the 17
Nonpharmacologic treatment. It is important for patients patients in the control group.58 The treatment was generally
with MBD or at risk of MBD to continue to treat their well tolerated.
underlying disease, infections, and metabolic acidosis and
reduce chronic inflammation where possible. The treatment Nephrolithiasis (Stones)
of the secondary causes of osteoporosis involves diagnos-
ing and managing hyperthyroidism, hypogonadism, and Individuals with SBS are susceptible to numerous potential
hyperparathyroidism.41 Healthy lifestyle changes should complications. One of these is the development of renal
also be counseled for patient to discontinue tobacco use, stones. This can result in frequent episodes of renal colic,
minimize alcohol intake, and encourage regular physical obstructive uropathy, and urinary tract infections. In some
activity, particularly weight-bearing exercise. Not only do cases, the deposition of stones can lead to the progressive
these measures increase BMD but also decrease fall rates.31 impairment of renal function.

Medications. There are several classes of medications that Prevalence


are approved by the Food and Drug Administration for the The prevalence of symptomatic nephrolithiasis, renal
treatment of osteoporosis. These drugs include conjugated stones, ranges from 42% to 50% in patients with SBS
estrogens, selective estrogen receptor modulators, bispho- compared with 8.4% in the general population.64-66 The inci-
sphonates, calcitonin, and receptor activator of nuclear dence of stones for patients with a jejunostomy or ileostomy
factor-κB ligand (RANKL) inhibitors. The pharmacologic ranges from 5% to 15%.67,68 If the colon is retained such
mechanism of action is to inhibit osteoclasts and re- as those with a jejuno-colic anastomosis, they have a 25%
duce bone resorption.41,58 Bisphosphonates are considered chance of developing symptomatic nephrolithiasis.69
the first-line therapy for osteoporosis in postmenopausal
women based on efficacy, safety, and cost. Unfortunately,
Types, Causes, and Prevention
they are poorly absorbed from the gastrointestinal tract
and commonly cause gastrointestinal intolerance, which The composition of the stones will vary depending on the
makes them difficult to use in patients with SBS.59 However, type of surgical resection and several other predisposing
intravenous preparations exist and an early randomized factors. Both uric acid and calcium oxalate renal stones are
controlled trial of 20 patients taking clodronate found it common in patients with a jejunostomy or ileostomy68,70-73
inhibited bone resorption and increased BMD. However, and are thought to be related to chronic dehydration,
the BMD increase was only statistically significant in the low urine volume, and reduced urine sodium.74-76 This
Johnson et al 463

Table 6. High Oxalate Foods.a,83-88

Food Type Examples

Fruits Blackberries, black raspberries, blueberries, red currants, dewberries, figs, grapes, gooseberries,
kiwi, lemon peel, lime peel, orange peel, red raspberries, rhubarb, strawberries, tangerines,
any juice made from above fruits
Vegetables Beans (green, wax, dried), beets (tops, roots, greens), celery, chives, collards, dandelion,
eggplant, escarole, kale, leeks, mustard greens, okra, parsley, parsnips, peppers (green),
pokeweed, rutabagas, sorrel, spinach, summer squash, sweet potatoes, Swiss chard, tomato
soup, vegetable soup, watercress, yams
Meat substitutes, beans, nuts, Almonds, baked beans canned in tomato sauce, cashews, green beans (waxed and dried),
seeds peanut butter, peanuts, pecans, sesame seeds, sunflower seeds, tofu (soybean curd), walnuts
Beverages Any juice made from high oxalate fruits, draft beer, chocolate (plain), chocolate milk, cocoa,
coffee powder (instant), Ovaltine, tea (brewed)
Starches Fig Newtons, fruit cake, graham crackers, grits, white corn, kamut, marmalade, soybean
crackers, wheat germ
Condiments Ground cinnamon, raw parsley, pepper, >1 tsp/day, ginger, soy sauce
a Partial listing.

Table 7. Methods to Decrease Risk of Stone Formation in Short Bowel Syndrome.

Methods Rationale

Adequate fluid and salt intake In short bowel syndrome, urine volume and urine sodium are reduced due to malabsorption.
Increase fluid and salt intake to increase urine output to a goal of 2 liters per day prevent
stone formation
Low oxalate diet Reduce oxalate intake for those with a colon to reduce oxalate excretion
Low-fat diet Reduce steatorrhea that leads to binding of intraluminal calcium by fatty acids, which leads to
nonavailability of calcium to bind oxalate, which leads to increased absorption of dietary
oxalate
Increase foods with vitamin C Vitamin C converted to citrate which prevents oxalate and calcium crystal formation
or citrate supplements
Calcium supplementation Bind oxalate in the gut
Magnesium supplements Hypomagnesemia leads to reduced citrate excretion and urine pH
Probiotics Oxalate degrading
Cholestyramine Bind oxalate in the colon

highlights the importance of adequate fluid and salt intake stone formation by competing for calcium binding in the
to increase urine output and prevent stone formation. Urine urine.81 Hypomagnesemia leads to reduced citrate excretion
volume should be increased as much as possible so that and urine pH. Thus, it is important to maintain adequate
the urine output exceeds 2 liters per day.68 This can be magnesium levels.
challenging in patients with SBS and may require the use Patients with SBS with the colon or portions of the colon
of supplemental intravenous fluids or even PN. In addition, in circuit are more likely to experience stone formation;
patients with the largest resections or the most extensive these are generally calcium oxalate in nature. Normally,
disease have the greatest degree of hyperoxaluria and are calcium binds to oxalate in the intestine allowing it to
more likely to form renal stones.77,78 Individuals with an be excreted in the stool. In SBS, unabsorbed fat bind to
ileal pouch anal anastomosis have a slightly smaller risk calcium. This leaves oxalate free to be absorbed by the
for renal stones, presumably due to better fluid and salt colon. More oxalate is absorbed, resulting in a higher risk
balance.79 Calcium oxalate stone formation results from of developing kidney stones.82 These may occur as early as
hypocitraturia and hypomagnesuria; uric acid stones are 2 months after surgical resection or as late as 67 months
generally related to low urine volume and low pH.66 SBS following an operation.64 Consumption of an oxalate-free
patients with hyperglycemia due to diabetes or poorly diet will reduce urinary oxalate excretion.77 Oxalate is preva-
managed PN are at risk for developing stones.80 Diets high lent in many foods, making the diet difficult to comply with
in citrate-containing fruits and vegetables such as citrus for some patients (Table 6).83-88 Alternatively, increasing
fruits, tomatoes, and Jerusalem artichokes reduce oxalate dietary calcium intake in ranges of 600–2000 mg/day with
464 Nutrition in Clinical Practice 33(4)

meals has been shown to decrease urinary oxalate excretion manuscript; E. Johnson, L. Vu, and L. E. Matarese critically
by competing with luminal oxalate and limiting intestinal revised the manuscript; and E. Johnson, L. Vu, and L. E.
absorption.89-91 Matarese agree to be fully accountable for ensuring the integrity
Patients with SBS often have excess fecal fat due to and accuracy of the work. All authors read and approved the
steatorrhea. Unabsorbed fatty acids compete with oxalate final manuscript.
for calcium resulting in greater oxalate absorption.77 A
low-fat diet should be used to reduce the fecal output that References
leads to binding of intraluminal calcium, which in turn 1. Hofstetter S, Stern L, Willet J. Key issues in addressing the clinical and
results in nonavailability of calcium to bind oxalate. The humanistic burden of short bowel syndrome in the US. Curr Med Res
situation is often further complicated by the presence of Opin. 2013;29(5):495-504.
2. O’Keefe SJ, Buchman AL, Fishbein TM, Jeejeebhoy KN, Jeppesen PB,
unabsorbed bile salts as well as long-chain fatty acids.74
Shaffer J. Clin Gastroenterol Hepatol. 2006;4(1):6-10.
Cholestyramine has been shown to reduce oxaluria by 3. Nightingale JMD. The short bowel. In: Nightingale JMD, ed. Intestinal
chelating bile salts and preventing stone formation.79 How- Failure. London, England: Greenwich Medical Media; 2001:177-198.
ever, cholestyramine also binds and impedes the absorption 4. Buchman AL, Scolapio J, Fryer J. AGA technical review on short
of other nutrients including fat-soluble vitamins and may bowel syndrome and intestinal transplantation. Gastroenterology.
2003;124(4):1111-1134.
even worsen diarrhea in some patients.
5. Dibaise JK, Young RJ, Vanderhoof JA. Enteric microbial flora,
Certain vitamins have been implicated in the devel- bacterial overgrowth, and short-bowel syndrome. Clin Gastroenterol
opment and treatment of oxalate stones. Pyridoxine and Hepatol. 2006;4(1):11-20.
thiamine deficiency has been associated with enhanced 6. Bongaerts GPA, Tolboom JJ, Naber AHJ, et al. Role of bacteria in the
metabolic generation of oxalate or enhanced ileal absorp- pathogenesis of short bowel syndrome-associated d-lactic academia.
Microb Pathog. 1997;22(5):285-293.
tion. As a result, pyridoxine in doses of 150 mg per day
7. Kaneko T, Bando Y, Kurihara H, Satomi K, Nonoyama K, Matsuura
has been used to reduce endogenous oxalate production.92 N. Fecal microflora in a patient with short-bowel syndrome an identi-
Thiamine deficiency has been shown to cause hyperoxaluria fication of dominant lactobacilli. J Clin Microbiol. 1997;35(12):3181-
in animal models.93 3185.
Oxalate has been shown to be degraded by bacteria 8. Joly F, Mayeur C, Bruneau A, et al. Drastic changes in fecal and
mucosa-associated microbiota in adult patients with short bowel syn-
in the colon.94,95 As a result, there has been interest in
drome. Biochimie. 2010;92(7):753-761.
using probiotics to change the composition of the gut 9. Matarese L, O’Keefe SJ, Kandil H, Bond G, Costa G, Abu-Elmagd K.
microbiota. Certain probiotics have been used to reduce Short bowel syndrome: clinical guidelines for nutrition management.
oxalate absorption.96 The best probiotic and specific dosage Nutr Clin Pract. 2005;20(5):493-5020.
to use still needs to be determined. The oxalate-degrading 10. Cresci G, Bawden E. Gut microbiome: what we do and don’t know.
Nutr Clin Pract. 2015;30(6):734-746.
activity of different probiotics varies, thus more research
11. Quigley E, Abu-Shanab A. Small intestinal bacterial overgrowth. Infect
is indicated to determine the exact strain and dose.97 Pre- Dis Clin N Am. 2010;24(4):943-959.
vention and treatment of both uric acid and oxalate stones 12. Quigley EMM, Quera R. Small intestinal bacterial overgrowth: roles
is based on the gastrointestinal anatomy and may require of antibiotics, prebiotics, and probiotics. Gastroenterology. 2006;130(2
a multipronged approach to include dietary interventions, Suppl 1):S78-S90.
13. Bongaerts GPA, Severijnen R, Tangerman A, Verrips A, Tolboom JJM.
medications, and fluid management (Table 7).
Bile acid deconjugation by lactobacilli and its effects on patients with a
short small bowel. J Gastroenterol. 2000;35(11):801-804.
14. Schiller L. Evaluation of small bowel bacterial overgrowth. Curr
Gastroenterol Rep. 2007;9(5):373-377.
Summary 15. Peterson C. D-lactic acidosis. Nutr Clin Pract. 2005;20(6):634-645.
SBS can result in significant acute and chronic complica- 16. Uribarri J, Oh M, Carroll H. D-lactic acidosis: a review of clinical
presentation, biochemical features, and pathophysiologic mechanisms.
tions resulting from changes in gastrointestinal anatomy
Medicine. 1998;77(2):73-82.
and the results of malabsorption. These complications 17. White L. D-lactic acidosis: more prevalent than we think. Practical
can have a negative impact on the patient’s quality of gastroenterology. https://med.virginia.edu/ginutrition/articles-from-
life. SIBO, bone demineralization, and nephrolithiasis are practical-gastroenterology/. Accessed March 8, 2018.
common and debilitating in these patients. Comprehensive 18. Kowlgi NG, Chhabra L. D-lactic acidosis: an underrecognized
complication of short bowel syndrome. Gastroenterol Res Pract.
monitoring and treatment can minimize these complications
2015;2015:476215.
and the progression of further disease. 19. Goulet O, Joly F. The pathological microbiota: intestinal microbiota in
short bowel syndrome. Gastroenterol Clinical Bio. 2010;34(1):S37-S43.
Statement of Authorship 20. Ramakrishnan T, Stokes P. Beneficial effects of fasting and low
carbohydrate diet in D-lactic acidosis associated with short bowel
E. Johnson, L. Vu, and L. E. Matarese contributed to con- syndrome. JPEN J Parenter Enteral Nutr. 1985;9(3):361-363.
ception/design of the article; E. Johnson, L. E. Vu, and L. E. 21. Bongaerts GPA, Severijnen R. Arguments for a lower carbohydrate–
Matarese contributed to acquisition, analysis, or interpretation higher fat diet in patients with a short small bowel. Med Hypothesis.
of the data; E. Johnson, L. Vu, and L. E. Matarese drafted the 2006;67(2):280-280.
Johnson et al 465

22. Vitetta L, Coulson S, Thomsen M, Nguyen T, Hall S. Probiotics, D- 42. Ferrone M, Geraci M. A review of the relationship between parenteral
lactic acidosis, oxidative stress and strain specificity. Gut Microbes. nutrition and metabolic bone disease. Nutr Clin Pract. 2007;22(3):329-
2017;8(4):311-322. 339. https://doi.org/10.1177/0115426507022003329
23. Attar A, Flourie B, Rambaud JC, et al. Antibiotic efficacy in small 43. Gacs G, Barltrop D. Significance of Ca-soap formation for calcium
intestinal bacterial overgrowth–related chronic diarrhea: a crossover, absorption in the rat. Gut. 1977;18(1):64-68. https://doi.org/10.1136/
randomized trial. Am Gastro Assoc. 1999;117(4):794-797. gut.18.1.64
24. Oh MS, Phelps KR, Traube M, Barbosa-Saldivar J, Boxhill C, Carroll 44. Abitbol V, Roux C, Chaussade S, et al. Metabolic bone assess-
HJ. D-lactic acidosis in a man with the short bowel syndrome. N Engl ment in patients with inflammatory bowel disease. Gastroenterology.
J Med. 1979;301(5):249-52. 1995;108(2):417-422. https://doi.org/10.1016/0016-5085(95)90068-3
25. Lauritano EC, Gabrielli M, Scarpellini E, et al. Antibiotic therapy in 45. Ellegård L, Kurlberg G, Bosaeus I. High prevalence of vitamin D
small intestinal bacterial overgrowth: rifaximin versus metronidazole. deficiency and osteoporosis in out-patients with intestinal failure. Clin
Eur Rev Med Pharmacol. 2009;13(2):111-116. Nutr. 2013;32(6):983-987. https://doi.org/10.1016/j.clnu.2013.02.005
26. Munakata S, Arakawa C, Kohira R, Fujita Y, Fuchigami T, 46. Fan S, Ni X, Wang J, et al. High prevalence of suboptimal vitamin
Mugishima H. A case of D-lactic acid encephalopathy associated with D status and bone loss in adult short bowel syndrome even after
use of probiotics. Brain Develop. 2010;32(8):691-694. weaning off parenteral nutrition. Nutr Clin Pract. 2017;32(2):258-265.
27. Vanderoof Y, Murray K. Treatment strategies for small bowel bac- https://doi.org/10.1177/0884533616665784
terial overgrowth in short bowel syndrome. J Med Gastro Nutrition. 47. Napartivaumnuay N, Gramlich L. The prevalence of vitamin D
1998;27(2):155-160. insufficiency and deficiency and their relationship with bone min-
28. Gigante A, Sardo L, Gasperini ML. D-lactic acidosis 25 years eral density and fracture risk in adults receiving long-term home
after bariatric surgery duet to salmonella enteritidis. Nutrition. parenteral nutrition. Nutrients. 2017;9(5):481. https://doi.org/10.3390/
2012;28(1):108-111. nu9050481
29. Takahashi K, Terashima H, Kohno K, Ohkohchi N. A stand-alone 48. Aljarallah B, Fernandes G, Jeejeebhoy KN, et al. The Canadian
symbiotic treatment for the prevention of D-lactic acidosis in short Home Total Parenteral Nutrition (HTPN) registry: vitamin K
bowel syndrome. Int Surg. 2013;98(2):110-113. supplementation and bone mineral density. JPEN J Parenter
30. NIH Consensus Development Panel on Osteoporosis Prevention, Di- Enteral Nutr. 2012;36(4):415-420. https://doi.org/10.1177/
agnosis, and Therapy. Osteoporosis prevention, diagnosis, and therapy. 0148607111431983
J Am Med Assoc. 2001;285(6):785-795. https://doi.org/10.1001/jama. 49. Pironi L, Morselli Labate AM, et al. Prevalence of bone disease in
285.6.785 patients on home parenteral nutrition. Clin Nutr. 2002;21(4):289-296.
31. Goltzman D. Functions of vitamin D in bone. Histochem Cell Biol. https://doi.org/10.1054/clnu.2002.0548
2018;149(4):305-312. https://doi.org/10.1007/s00418-018-1648-y 50. Rodriguez-Ortiz ME, Canalejo A, Herencia C, et al. Magnesium
32. Clarke B. Normal bone anatomy and physiology. Clin J Am modulates parathyroid hormone secretion and upregulates parathyroid
Soc Nephrol. 2008;3(suppl 3):S131-S139. https://doi.org/10.2215/CJN. receptor expression at moderately low calcium concentration. Nephrol
04151206 Dial Transplant. 2014;29(2):282-289. https://doi.org/10.1093/ndt/gft400
33. Wright NC, Looker AC, Saag KG, et al. The recent prevalence of 51. Klein GL. Aluminum in parenteral solutions revisited—again. Am J
osteoporosis and low bone mass in the United States based on bone Clin Nutr. 1995;61(3):449-456. https://doi.org/10.1093/ajcn/61.3.449
mineral density at the femoral neck or lumbar spine. J Bone Miner Res. 52. Pironi L, Arends J, Bozzetti F, et al. ESPEN guidelines on
2014;29(11):2520-2526. https://doi.org/10.1002/jbmr.2269 chronic intestinal failure in adults. Clin Nutr. 2016;35(2):247-307.
34. Blume SW, Curtis JR. Medical costs of osteoporosis in the el- https://doi.org/10.1016/j.clnu.2016.01.020
derly Medicare population. Osteoporos Int. 2011;22(6):1835-1844. 53. Waldron JL, Ashby HL, Cornes MP, et al. Vitamin D: a
https://doi.org/10.1007/s00198-010-1419-7 negative acute phase reactant. J Clin Pathol. 2013;66(7):620-622.
35. Panula J, Pihlajamäki H, Mattila VM, et al. Mortality and https://doi.org/10.1136/jclinpath-2012-201301
cause of death in hip fracture patients aged 65 or older—a 54. Verhage AH, Cheong WK, Allard JP, Jeejeebhoy KN. Increase
population-based study. BMC Musculoskelet Disord. 2011;12(1):105. in lumbar spine bone mineral content in patients on long-term
https://doi.org/10.1186/1471-2474-12-105 parenteral nutrition without vitamin D supplementation. JPEN
36. Dyer SM, Crotty M, Fairhall N, et al. A critical review of the J Parenter Enteral Nutr. 1995;19(6):431-436. https://doi.org/10.
long-term disability outcomes following hip fracture. BMC Geriatr. 1177/0148607195019006431
2016;16(1):158. https://doi.org/10.1186/s12877-016-0332-0 55. Haderslev K, Tjellesen L, Haderslev P, Staun M. Assessment of the
37. Lewiecki EM, Lane NE. Common mistakes in the clinical use of bone longitudinal changes in bone mineral density in patients receiving home
mineral density testing. Nat Clin Pract Rheumatol. 2008;4(12):667-674. parenteral nutrition. JPEN J Parenter Enteral Nutr. 2004;28(5):289-
https://doi.org/10.1038/ncprheum0928 294. https://doi.org/10.1177/0148607104028005289
38. Nayak S, Edwards DL, Saleh AA, Greenspan SL. Systematic review 56. Pironi L, Tjellesen l, De Francesco A, et al. Bone mineral density in
and meta-analysis of the performance of clinical risk assessment instru- patients on home parenteral nutrition: a follow-up study. Clin Nutr.
ments for screening for osteoporosis or low bone density. Osteoporos 2004;23(6):1288-1302. https://doi.org/10.1016/S0261-5614(04)00061-5
Int. 2015;26(5):1543-1554. https://doi.org/10.1007/s00198-015-3025-1 57. Cohen-Solal M, Baudoin C, Joly F, et al. Osteoporosis in patients
39. Pironi L, Arends J, Baxter J, et al. ESPEN endorsed recommendations. on long-term home parenteral nutrition: a longitudinal study.
Definition and classification of intestinal failure in adults. Clin Nutr. J Bone Miner Res. 2003;18(11):1989-1994. https://doi.org/10.
2015;34(2):171-180. https://doi.org/10.1016/j.clnu.2014.08.017 1359/jbmr.2003.18.11.1989
40. Nygaard L, Skallerup A, Olesen SS, et al. Osteoporosis in patients with 58. Szczepanek K, Pedziwiatr M, Klek S. Denosumab improves bone
intestinal insufficiency and intestinal failure: prevalence and clinical mineral density in patients with intestinal failure receiving home
risk factors [published online ahead of print August 2017]. Clin Nutr. parenteral nutrition: results from a randomized, controlled clinical trial
https://doi.org/10.1016/j.clnu.2017.07.018 [published online ahead of print March 2017]. JPEN J Parenter Enteral
41. Seidner DL. Parenteral nutrition-associated metabolic bone dis- Nutr. https://doi.org/10.1177/0148607117695247
ease. JPEN J Parenter Enteral Nutr. 2002;26(5 suppl):S37-S42. 59. Lin JH. Bisphosphonates: a review of their pharmacokinetic properties.
https://doi.org/10.1177/014860710202600511 Bone. 1996;18(2):75-85. https://doi.org/10.1016/8756-3282(95)00445-9
466 Nutrition in Clinical Practice 33(4)

60. Haderslev K, Tjellesen L, Sorensen H, Staun M. Effect of cyclical 79. Smith LH, Fromm H, Hofmann AF. Acquired hyperoxaluria,
intravenous clodronate therapy on bone mineral density and nephrolithiasis and intestinal disease. Description of a syndrome. N
markers of bone turnover in patients receiving home parenteral Engl J Med. 1972;286(26):1371-1375.
nutrition. Nutr Clin Pract. 2003;18(2):176-176. https://doi.org/ 80. Weinberg AE, Patel CJ, Chertow GM, Leppert JT. Diabetic severity
10.1177/0115426503018002176 and risk of kidney stone disease. Eur Urol. 2014;65(1):242-247. https://
61. Raman M, Aghdassi E, Baun M, et al. Metabolic bone disease doi.org/10.1016/j.eururo.2013.03.026
in patients receiving home parenteral nutrition: a Canadian study 81. Goldfarb DS, Asplin JR. Effect of grapefruit juice on urinary
and review. JPEN J Parenter Enteral Nutr. 2006;30(6):492-496. lithogenicity. J Urol. 2001;166(1):263-267.
https://doi.org/10.1177/0148607106030006492 82. Hylander E, Jarnum S. Jensen HJ, Thale M. Enteric hyperoxaluria:
62. Pazianas M, Compher C, Schiavone-Gatto P, Kinosian BP. In- dependence on small intestinal resection, colectomy, and steator-
testinal failure-associated metabolic bone diseases and response to rhoea in chronic inflammatory bowel disease. Scand J Gastroenterol.
teriparatide. Nutr Clin Pract. 2006;21(6):605-609. https://doi.org/ 1978;13(5):577-588.
10.1177/0115426506021006605 83. Ney DM, Hofmann AF, Fischer C, Stubblefield N. The Low Oxy-
63. Cummings SR, Martin JS, McClung MR, et al. Denosumab for preven- late Diet Book. San Diego, CA: University of California Press;
tion of fractures in postmenopausal women with osteoporosis. N Engl 1981.
J Med. 2009;361(8):756-765. https://doi.org/10.1056/NEJMoa0809493 84. Massey LK, Roman-Smith H, Sutton RA. Effect of dietary oxalate and
64. Nightingale JM, Lennard-Jones JE, Gertner DJ, et al. Colonic preser- calcium on urinary oxalate and risk of formation of calcium oxalate
vation reduces need for parenteral therapy, increases incidence of renal kidney stones. J Am Diet Assoc. 1993;93(8):901-906.
stones, but does not change high prevalence of gall stones in patients 85. Holmes RP, Kennedy M. Estimation of the oxalate content of foods
with a short bowel. Gut. 1992;33:1493-1497. and daily oxalate intake. Kidney Int. 2000;57(2000):1662-1667.
65. Scales CD Jr, Smith AC, Hanley JM, Saigal CS. Prevalence of kidney 86. Zarembski PM, Hodgkinson A. The oxalic acid content of English
stones in the United States. Eur Urol. 2012;62(1):160-165. diets. Br J Nutr. 1962;16:627-634.
66. Teichman JM. Clinical practice. Acute renal colic from ureteral calcu- 87. Kasidas GP, Rose GA. Oxalate contents of some common foods; deter-
lus. N Engl J Med. 2004;350(7):684. mination of by an enzymatic method. J Hum Nutr. 1980;34(4):255-266.
67. Clarke AM, McKenzie RG. Ileostomy and the risk of urinary uric acid 88. Siener R, Honow R, Voss S, Seidler A, Hesse A. Oxalate content of
stones. Lancet. 1969;2(7617):395-397. cereals and cereal products. J Agric Food Chem. 2006;54(8):3008.
68. Tomson CRV. Nephrocalcinosis and nephrolithiasis. In: Nightingale 89. Lemann J, Pleuss JA, Worcester EM, Hornick L, Schrab D, Hoffmann
JMD, ed. Intestinal Failure. London, England: Greenwich Medical RG. Urinary oxalate excretion increases with body size and decreases
Media Limited; 2001:227-242. with increasing dietary calcium intake among healthy adults. Kidney
69. Nightingale JMD. Hepatobiliary, renal and bone complications of Int. 1996;49(1):200-208.
intestinal failure. Best Pract Res Clin Gastroenterol. 2003:17(6):907- 90. Barilla DE, Notz C, Kennedy D, Pak CYC. Renal oxalate excretion
929. following oral oxalate loads in patients with ileal disease and with renal
70. Deren JJ, Porush JG, Levitt MF, Khilnani MT. Nephrolithiasis as a and absorptive hypercalciurias: effect of calcium and magnesium. Am
complication of ulcerative colitis and regional enteritis. Ann Inter Med. J Med. 1978;64(4):579-585.
1962;56(6):843-853. 91. Hylander E, Jarnum S, Nelsen K. Calcium treatment of enteric
71. Maratka Z, Nedbal J. Urolithiasis as a complication of the surgical hyperoxaluria after JIB for morbid obesity. Scand J Gastoenterol.
treatment of ulcerative colitis. Gut. 1964;5:214-217. 1980;15(3):349-352.
72. Bennett RC, Jepson RP. Uric acid stone formation following ileostomy. 92. Lyon ES, Borden TA, Ellis JE, Vermeulen CW. Calcium oxalate
ANZ J Surg. 1966;36(2):153-158. lithiasis produced by pyridoxine deficiency and inhibition by high
73. Kennedy HJ, Lee ECG, Claridge G, Truelove SC. The health of magnesium diets. Invest Urol. 1966;4(2):133-142.
subjects living with a permanent ileostomy. Q J Med. 1982;51(203):341- 93. Sidhu H, Gupta R, Thind SK, Nath R. Oxalate metabolism in
357. thiamine-deficient rats. Ann Nutr Metab. 1987;31(6):354-361.
74. Gallagher ND, Harrison DD, Skyring AP. Fluid and electrolyte distur- 94. Argenzio RA, Liacoz JA, Allison MJ. Role of intestinal oxalate
bances in patients with long established ileostomies. Gut. 1962;3(3):219- degrading bacteria in the pathogenesis of enteric hyperoxaluria. Gas-
223. troenterology. 1985;88:1309.
75. Clarke AM, Chirnside A. Chronic dehydration and sodium depletion 95. Allison MJ, Cook HM, Milne DB, Gallagher S, Clayman RV. Ox-
in patients with established ileostomies. Lancet. 1967;2(7519):740- alate degradation by gastrointestinal bacteria from humans. J Nutr.
743. 1986;116(3):455-460.
76. Kennedy HJ, Al-Dujaili EAS, Edwards CRW, Truelove SC. Water 96. Okombo J, Liebman M. Probiotic-induced reduction of gastrointesti-
and electrolyte balance in subjects with a permanent ileostomy. Gut. nal oxalate absorption in healthy subjects. Urol Res. 2010;38(3):169-
1983;24(8):702-705. 178.
77. Chadwick VS, Modha K, Dowling RH. Mechanism for hyperoxaluria 97. Mogna L, Pane M, Nicola S, Raiteri E. Screening of different pro-
in patients with ileal dysfunction. N Engl J Med. 1973;289(4):172-176. biotic strains for their in vitro ability to metabolize oxalates: any
78. Dobbins JW, Binder HJ. Effect of bile salts and fatty acids on the prospective use in humans? J Clin Gastroenterol. 2014;48(Suppl 1):
colonic absorption of oxalate. Gastroenterology. 1976;70(6):1096-1100. S91-S95.
Invited Review

Nutrition in Clinical Practice


Volume 33 Number 4
Dietary Fermentable Oligosaccharides, Disaccharides, August 2018 468–475

C 2018 American Society for

Monosaccharides, and Polyols (FODMAPs) and Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10108
Gastrointestinal Disease wileyonlinelibrary.com

Nimish Vakil, AGAF, FACG, FASGE1,2

Abstract
FODMAP is an acronym for fermentable oligosaccharides, disaccharides, monosaccharides, and polyols. Dietary modification of
FODMAPs has been shown to have significant effects on the physiology of the gastrointestinal tract and improves
symptoms of abdominal pain, distention, and bloating in patients with irritable bowel syndrome. Structured withdrawal and
reintroduction of FODMAPs supervised by a dietitian is the optimal practice for dietary FODMAP modification in irritable bowel
syndrome. FODMAPs are present in enteral feeding formulas and may have a role in diarrhea and bloating in tube-fed patients.
Emerging areas of research include the effects of dietary modification of FODMAPs on the microbiome, micronutrient absorption,
and caloric intake. FODMAP dietary modification is an emerging area in other gastrointestinal disorders and is of relevance to all
practicing dietitians. (Nutr Clin Pract. 2018;33:468–475)

Keywords
disaccharides; enteral nutrition; FODMAP; irritable bowel syndrome; inflammatory bowel diseases; microbiota; monosacccha-
rides; oligosaccharides; polysaccharides; sugar alcohols

Introduction glucosidases break down the dextrin and maltose further


and with other disaccharidases (lactase and sucrase) break
FODMAP is an acronym for fermentable oligosaccharides, sugars down further into monosaccharide units. Lactose
disaccharides, monosaccharides and polyols. FODMAPs is a disaccharide composed of glucose and galactose and
are short-chain carbohydrates that have the following 3 is broken down into its component monosaccharides by
characteristics: they are poorly absorbed in the small intes- lactase. In the absence of the lactase enzyme, lactose is
tine, they are fermentable, and they are osmotically active. carried to the distal small bowel and colon where it is
The FODMAP content of foods may be found in Table 1. used by bacteria and can cause symptoms of bloating and
diarrhea. Glucose, galactose, and fructose are absorbed
Carbohydrates and Their Absorption through the epithelium and are transported by the portal
vein to the liver. At low concentrations, glucose is absorbed
Carbohydrates consist of sugars, including monosaccha- using a sodium-dependent active transporter. At higher
rides and disaccharides; polyols, which are sugar alco- concentrations, a second facilitated transporter becomes
hols such as sorbitol and xylitol; and oligosaccharides, active. Galactose is absorbed using the same transporters.
which are carbohydrates containing 3–10 monosaccharides Fructose is an important monosaccharide that is present
found in plants such as onions. Polysaccharides are large in a free from in the diet, as a subunit of a disaccharide
chains of monosaccharides. Polysaccharides may be de-
signed for energy storage, such as glycogen or starch or From the 1 University of Wisconsin School of Medicine and Public
serve a structural function such as cellulose or chitins. A Health, Madison, Wisconsin, USA; and 2 Aurora Health Care,
fructan is a polysaccharide made up of fructose. Fructo- Summit, Wisconsin, USA.
oligosaccharides are fructans with a short chain length. A Financial disclosure: None declared.
detailed description of carbohydrates and their absorption Conflicts of interests: None declared.
may be found elsewhere.1 This article originally appeared online on June 5, 2018.
The digestion of carbohydrates begins with the break-
Corresponding Author:
down of complex carbohydrates by salivary and pancreatic Nimish Vakil, MD, Aurora Summit Medical Center, 36500 Aurora
amylase into simpler sugars such as dextrin and maltose. Drive, Summit, WI 53066.
Enzymes in the brush border of the small intestine called Email: nvakil@wisc.edu
Vakil 469

Table 1. High FODMAP Foods by Component.

Fructans and Galacto


Fructose Lactose oligo-saccharides Polyols

Fruits: apples, cherries, figs, Cheese: soft cheeses (ricotta, Legumes: beans (black, fava, Sweetening agents: sorbitol,
mango, pears, watermelon cottage, mozzarella) kidney, navy), soy (beans, mannitol, xylitol, isomal
flour, milk)
Vegetables: artichokes, Milk: sheep, cow, goat and Nuts and grains: wheat, rye, Fruits: nectarines, peaches,
asparagus, dried tomatoes buffalo barley, pistachios and pears, blackberries, cherries,
and sugar snap peas cashews, Almonds (>10) plums, prunes, watermelon,
lychee
Sweeteners: honey, high Milk products: yogurt, Vegetables: garlic, leek, Sweetners: isomalt, sorbitol,
fructose corn syrup, agave ice-cream, custard, scallion, onion or garlic mannitol, xylitol, maltitol,
nectar condensed and evaporated powder lactitol
milk
Alcohol: sherry, port, rum Fruit: grapefruit, watermelon. Vegetables: cauliflower, celery,
sweet wines Plums, prunes, peaches, figs, snow peas, sweet potato
dates, currants and banana
Other: teas (oolong, Other: hard candies, toffee,
chamomile and fennel), jams and preserves, chewing
inulin (also appears as gum, chocolates, protein
chicory root extract), carob powders, baked goods, cough
drops and throat lozenges

FODMAP, fermentable oligosaccharides, disaccharides, monosaccharides, and polyols.

and as a component of the polysaccharide fructan. Fructose content of luminal content.7 Poorly absorbed carbohydrates
consumption has increased in the U.S. diet in recent years. such as the FODMAPs increase intestinal water by their
The prevalence of high-fructose corn syrup used as a osmotic activity.8
sweetener in various foods increased from 16% in 1977 to
42% in 1998 and has remained stable at that rate.2 Fructose Effects on colonic gas. Passage of undigested or unab-
is absorbed by 2 transporters GLUT-2 and GLUT-5, which sorbed short-chain carbohydrates into the colon provides
are located in the brush border epithelium of the small a substrate for colonic bacteria to produce hydrogen and
intestine. Absorption through the GLUT-2 pathway is facil- methane. Studies in healthy individuals and patients with
itated by ingestion of glucose, but the GLUT-5 pathway is irritable bowel syndrome using magnetic resonance imag-
independent of glucose coadministration.3 Approximately ing show a significant increase in colonic gas with high
half the population cannot tolerate 25 grams of fructose.4 FODMAP diets. In patients with irritable bowel syndrome,
With the average consumption of fructose ranging from 11– the increased gas is associated with the development of
54 grams a day, the increasing use of fructose as a sweetener abdominal symptoms.9,10 Studies using magnetic resonance
exposes many patients to high doses of fructose. imaging show that polysaccharides such as inulin commonly
An exception to the absorptive mechanisms described derived from plant products such as chicory root increase
previously is the handling of dietary fiber. Fiber is a complex colonic gas in both healthy individuals and patients with
polysaccharide that cannot be absorbed and passes through irritable bowel syndrome and are associated with symptoms
the gastrointestinal (GI) tract into the stool. Colonic bacte- in patients with irritable bowel syndrome.5 A double-blind,
ria may metabolize some parts of dietary fiber. placebo-controlled trial of the administration of a fructan
(maltodextrin) to children with irritable bowel syndrome
Physiologic Effects of FODMAPs showed a significant worsening of bloating, flatulence, and
abdominal pain symptoms with fructan exposure.11
Effect on intestinal water. The effect of high FODMAP
diets has been studied in humans with an ileostomy by ex- Effects on short-chain fatty acids and visceral hypersensitivity.
amining the fluid effluent from the ileostomy. Other studies In addition to the production of methane and hydrogen,
have measured intestinal water by using magnetic resonance the fermentation of short-chain carbohydrates in the colon
imaging to quantify intestinal water.5,6 High FODMAP results in the production of short-chain fatty acids. An
diets increase intestinal water. This may contribute to pain important feature of functional gastrointestinal disorders
because of intestinal distention in patients with visceral hy- such as irritable bowel syndrome is the heightened sensi-
persensitivity and diarrhea as a result of the increased water tivity of these patients to gut stimulation or distention, a
470 Nutrition in Clinical Practice 33(4)

condition termed visceral hypersensitivity. Short-chain fatty and their constituents. In a randomized, controlled, single-
acids in luminal contents have been shown create visceral blind trial, McIntosh et al19 evaluated the effect of low and
hypersensitivity in animal studies.12,13 Low FODMAP diets high FODMAP diets on the food-related metabolome by
are associated with a decrease in liposaccharides (a complex measuring histamine, p Hydroxybenzoic acid, and azelaic
of fat and polysaccharides) in the luminal content of the acid in the urine of patients with irritable bowel syndrome.
colon.11 Intracolonic administration of fecal supernatant In patients on a high FODMAP diet, urinary levels of
from patients with irritable bowel syndrome induces visceral histamine increased significantly, whereas histamine levels
hypersensitivity in rats.11 Administration of a lipopolysac- dropped in patients randomized to the low FODMAP
charide antagonist blocked the increase and fecal super- diet. Histamine is an endogenous amine that is thought
natant from healthy individuals, and patients with irritable to play a role in irritable bowel syndrome and studies
bowel syndrome on a low FODMAP diet did not have blocking histamine have shown a benefit in patients with
an effect on visceral hypersensitivity.11 Low FODMAP this disorder.20 A number of other agents may be involved
diets may therefore alter visceral sensitivity by changing in the genesis of symptoms in irritable bowel syndrome, but
the composition of luminal contents (decreasing luminal additional research is necessary in this area.
short-chain fatty acids and liposaccharides). Short-chain
fatty acids have a beneficial effect on epithelial function, Disease States
and therefore concerns remain about the prolonged use
of low FODMAP diets on colonic health and the risk of Irritable Bowel Syndrome
colon cancer.8 There are a number of limitations with the
Irritable bowel syndrome is a common and debilitating
studies that have been performed to date, including small
medical condition that is associated with abdominal pain,
sample sizes, conflicting results, and difficulties with the
bloating distention, and changes in bowel habit. No single
measurement of short-chain fatty acids in the ascending
curative treatment exists, and patients often report an asso-
colon.8
ciation of symptoms with certain foods. A detailed review
of the criteria for a diagnosis may be found elsewhere.21
Effect on the microbiome. The gastrointestinal tract harbors
a large number of organisms in an ecosystem that is
unique for the individual and relatively stable over time.14 Short-term studies (4–6 weeks) on FODMAP restriction in
This ecosystem consists of luminal bacteria and bacteria irritable bowel syndrome. Of the patients in various studies,
that are adherent to the mucosal surface. The microbiota 50%–80% report an improvement of their symptoms on
communicates with wall of the organ and can influence a low FODMAP diet. A recent meta-analysis evaluated 6
function. The 2 main families or phyla of organisms are randomized controlled trials and 16 nonrandomized tri-
Firmicutes and Bateroidetes that together make up 90% als and reported substantial improvements in abdominal
of the microbiome.15 Alterations in the microbiome could pain, bloating, and overall symptoms with odds ratios
change the cross-talk between the bacteria and intestine, ranging from 1.75–1.81.22 Dietary intervention with low
altering function and creating symptoms. Diets that differ FODMAP dietary education when compared with sham
in FODMAP content have been shown to have an effect on (placebo) dietary intervention has shown a substantial
the gut microbiome of the host.16 In patients with irritable benefit for dietary advice given by a dietitian when com-
bowel syndrome, symptoms improved on a low FODMAP pared with placebo with regard to irritable bowel syndrome
diet, but concentrations of bifidobacter (a beneficial or- symptoms.15
ganism in the colon) decreased.17,18 A probiotic coadmin-
istered with the low FODMAP diet restored bifidobacter Long-term results of FODMAP restriction on irritable bowel
concentrations.15 A small study showed that a 3-week low syndrome. The evidence for long-term efficacy of a low
FODMAP diet when compared with a high FODMAP diet FODMAP diet in irritable bowel syndrome is limited in
resulted in a higher abundance of Adlercreutzia, a genus quality and in the number of studies available. A dietary
that uses hydrogen.17 Investigation into the effects on the strategy of reducing fructose and fructans was effective in
FODMAP diet on the microbiome are in their infancy. alleviating symptoms in 74% of patients at 14 months.23
The study was small, and recall bias limits the validity of
Effect on the metabolome. The metabolome refers to the the observed benefit. A randomized trial comparing gut-
complete set of small-molecule chemicals found within a directed hypnotherapy with a low FODMAP diet evaluated
biological sample. Within the human body there is an en- patients at 6 months and reported that 82% of patients in the
dogenous metabolome and also a food-related metabolome, diet therapy group had improved symptoms when compared
which is of growing interest. The food metabolome is with their baseline. Hypnotherapy achieved similar results,
defined as the part of the human metabolome directly but the numbers of patients in each group were small (n =
derived from the digestion and biotransformation of foods 25).24 A retrospective study of patients with irritable bowel
Vakil 471

syndrome followed for 16 months on a low FODMAP diet tients with these disorders report aggravation of symptoms
suggested that one third of patients continued to adhere with certain foods. There are limited data on the FODMAP
to the diet, whereas 84% of patients used a modified low diet in inflammatory bowel disease. In a small study of
FODMAP diet. Of the patients, 54% used the diet on and patients with quiescent Crohn’s disease, a low FODMAP
off based on symptom severity; 54% of patients reported diet was associated with changes in the fecal microbiome
a partial improvement in symptoms and 32% reported when compared with a normal Australian diet.30 Symp-
complete resolution.25 The British Dietetic Association rec- toms of irritable bowel syndrome coexist in patients with
ommends the low FODMAP diet as a second-line strategy inflammatory bowel disease. A recent meta-analysis found
with dietary advice to be provided by a dietitian.26 that patients with inflammatory bowel disease who were
still symptomatic despite adequate control of their disease
Nonceliac gluten sensitivity. Nonceliac gluten sensitivity by objective tests (markers of inflammation or endoscopic
is a disorder characterized by abdominal symptoms that evidence of active disease) had significant improvement in
improve after gluten withdrawal in the absence of celiac abdominal symptoms with a low FODMAP diet.30,31
disease. This is one of the fastest growing segments of the
food industry in North America, and the sale of gluten- Predicting a Response to the FODMAP Diet
free foods is expected to reach $7.5 billion in 2020.27 In a
Because of the cost and inconvenience associated with
double-blind crossover trial of patients with irritable bowel
the low FODMAP diet, a number of strategies are being
syndrome and nonceliac gluten sensitivity, Biesiekierski et
evaluated to determine if a response to the diet can be
al28 administered a low FODMAP diet for 2 weeks followed
predicted by diagnostic tests. A recent study suggests that
by high-gluten (16 g gluten/day), low-gluten (2 g gluten/day
fecal bacterial profiles using a commercially available assay
and 14 g whey protein/day), or control (16 g whey pro-
may predict a response to the FODMAP diet in irritable
tein/day) diets for 1 week. In all patients, symptoms im-
bowel syndrome. Bacterial abundance was higher in nonre-
proved significantly on a low FODMAP diet and worsened
sponders to the low FODMAP diet when compared with
equally when gluten or whey protein was added to the
responders.32,33 In a small recent study, the measurement
diet, suggesting the lack of a specific effect for gluten in
of volatile organic compounds in stool using a low-cost
aggravating symptoms. Wheat contains several potential
assay predicted a response to a low FODMAP diet in 100%
symptom inducers including gluten, fructans, and soluble
of patients.34 Fructose and lactose breath testing are not
proteins. Wheat-containing foods are a major source of
predictive of a response to a low FODMAP diet.35
fructans in the U.S. diet. To determine the effects of gluten
compared with fructans, Skodje et al29 studied patients
who did not have celiac disease by diagnostic tests and
Instructing Patients on the FODMAP Diet
who were on a self-imposed, gluten-free diet for relief of The management of a FODMAP diet in clinical practice has
abdominal symptoms.29 Participants were randomized in a been proposed as consisting of the following 4 stages (Figure
blinded manner to diets containing gluten (5.7 g), fructans 1): (a) initial dietitian visit for FODMAP restriction, (b)
(2.1 g), or placebo concealed in muesli bars for 7 days. 4–6 week dietitian visit for FODMAP reintroduction and
After a washout period, patients were re-randomized until (c) elective third visit for FODMAP personalization, and
every patient received each of the dietary interventions. (d) long-term follow-up of irritable bowel syndrome and
Symptoms were recorded using validated questionnaires. nutrition status.36 During the first visit, there is a detailed
Symptoms of bloating and overall symptoms of irritable dietary assessment including an evaluation of dietary pref-
bowel syndrome were substantially worse in patients given erences and foods known to cause symptoms. A nutrition
fructans, but there was no difference in symptoms between assessment is also useful as some patients may lose weight
those given placebo or gluten-enriched bars. These data on the FODMAP diet and the long-term nutrition conse-
suggest that fructans may be the cause of symptoms in many quences await definition. A description of the FODMAP
patients who report gluten sensitivity in the absence of celiac diet, the association of FODMAPs with symptoms, and
disease. From a clinical standpoint, it is reasonable to treat the results of FODMAP withdrawal are provided along
patients who present with gluten sensitivity and who do not with counseling on FODMAP restriction. Table 1 provides
have celiac disease with the structured dietary approach for a list of common foods containing FODMAPs. The initial
the low FODMAP diet that is described later because their phase of management consists of removal of all the po-
symptoms may be related to fructans in wheat and they may tential sources of FODMAPs to reduce their concentration
be sensitive to other FODMAPs as well. in the diet below the threshold of symptom generation.
It is important to emphasize to the patient that the diet
Inflammatory bowel disease. Inflammatory bowel disease is not permanent but compliance with the initial phase
(Crohn’s disease and ulcerative colitis) is known to be is important to determine if the patient is going to be
affected by changes in the fecal microbiome, and some pa- a responder to dietary intervention. The importance of
472 Nutrition in Clinical Practice 33(4)

Figure 1. Timeline for fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAP) intervention.

fructans in wheat (as opposed to gluten sensitivity) should drawal. A 4-week period of complete FODMAP withdrawal
be explained. Some patients need a discussion of other is considered adequate.35 Supplementing with a probiotic
diets that they may have tried with varying benefits. A containing bifidobacter has been shown to be effective in
number of diets are described in books and on the internet, replenishing the microbiome and should be discussed with
including the specific carbohydrate diet, the paleo diet, and the patient.
the Candida diet. Some of these diets restrict FODMAPs
and can result in symptom improvement, but the scientific
basis for these diets is inadequate and there is no structured
Structured Reintroduction
mechanism to reintroduce foods into the diet. Our approach There is substantial evidence from randomized controlled
to the first visit for dietary intervention has evolved from trials for the FODMAP withdrawal phase, but little struc-
giving the patient a handout (which was ineffective) to a tured evidence on the best method to reintroduce foods or
structured hour-long visit when the patient meets with the the order in which they should be reintroduced. Individual
dietitian and the gastroenterologist. The existing therapeu- centers have developed their own strategies for the struc-
tic relationship with the physician is an important part of tured addition of foods back to the diet. Our technique
the management of irritable bowel syndrome and can be is illustrated in Table 2, and the instructions provided
used to reassure the patient that they can follow the diet and to the patient are summarized in Figure 2. We typically
to enhance compliance with diet therapy.37,38 Patients fre- schedule a 30-minute dietitian visit to explain the strategy of
quently ask questions regarding the use of alcohol. Alcohol gradual dose escalation of foods shown in Table 2. A daily
can cause gastrointestinal symptoms by itself. Beer is low symptom chart is provided to the patient, and symptoms
in FODMAPs despite its origins in barley and rye because are recorded every day. For example, with lactose addition,
the fermentation process destroys many fructans. Rum and a half cup is ingested on Monday, 1 cup on Tuesday, and
fortified wines such as sherry and port contain high amounts 1½ cups on Wednesday. If the patient develops symptoms
of fructose and are forbidden in the initial phase. Dry wines on Monday, the experiment is stopped for that week and
(defined as <4 g of sugar per liter) are safe during the lactose is listed as a substance the patient cannot tolerate.
initial phase and include varietals such as sauvignon blanc, If the patient is able to tolerate lactose, the experiment is
albarino, and chardonnay in the recommended dose of 150 ended on Wednesday. From Thursday–Sunday, the patient
mL. Whiskey, vodka, and gin are low FODMAP alcohols returns to the low FODMAP diet that was prescribed for
in a dose of 30 mL per day. The major risk of FODMAP the preceding 4 weeks. On Monday, the next agent on the
exposure comes from the mixers used with these drinks, list is tried using the same protocol. At the end of the
such as tonic water and soda. Exercise enthusiasts need to structured reintroduction, the patient will know the foods
be made aware of chicory root in energy bars, which often that cannot be tolerated even in low doses. Other foods may
cause distress during exercise because of their high fructan be tolerated but in limited amounts, and some foods may
content (Table 1). The duration of the initial restrictive generate no symptoms at all. Based on these responses, a
phase is in a state of evolution because of the effects of diet for long-term use that avoids some foods, limits others,
the low FODMAP diet on the microbiota. Randomized and permits others is crafted for the patient. Symptoms
controlled trials have generally restricted FODMAPs for are recorded every day, and it is immediately apparent to
6 weeks or longer, but recent descriptions of a reduction the patient whether lactose can be tolerated and at what
in bifidobacter in the colon have prompted many groups dose. The strategy is repeated in subsequent weeks until
to reevaluate the duration of complete FODMAP with- complete. A recent study showed that patients who were
Vakil 473

Table 2. Structured FODMAP Reintroduction Plan.

Challenge week What to eat on challenge days

Week 1: lactose ½ cup milk or ¾ cup plain yogurt (without sweeteners or other FODMAPs)
Week 2: fructose ½ mango or 1–2 tablespoons of honey
Week 3: polyols-sorbitol Blackberries 3–10 or ¼ avocado
Week 4: polyols-mannitol 1/3 cup cauliflower or ¾ cup sweet potato
Week 5: fructans-wheat 2 slices of whole wheat bread or 1 cup cooked pasta
Week 6: fructans-onion 1 tablespoon diced onion
Week 7: fructans-garlic 1 clove of garlic
Week 8: galacto oligo-saccharides ½ cup kidney beans, black beans, or thawed peas or 15–25 almonds

FODMAP, fermentable oligosaccharides, disaccharides, monosaccharides, and polyols.

unaffected, but calcium intake decreased and overall caloric


• Test only one FODMAP group at a time
• Continue avoiding other FODMAPs intake declined in some patients.14,40 In our experience, some
• Test with a food that only contains that one FODMAP patients lose a small amount of weight in the initial 4–
• Eat the prescribed amount of the test food (don’t over 6 weeks of dietary FODMAP restriction. Some patients
do it)
• Try to test on 3 days during the week allowing a rest
choose to follow a very restricted diet, and future research
day between challenges will help determine if micronutrient deficiencies occur when
• Use the same test food on each of the 3 days but these diets are administered long term. Monitoring for
increase the dose by 50% (e.g. go from ½ cup to one
cup to 1½ cup) micronutrient deficiencies should be individualized based
• Track symptoms every day (not just the challenge on the degree of dietary restriction.
days)

FODMAPs in Enteral Feeding


Figure 2. Principles of food reintroduction for patients. FODMAPs may be important in enteral feeding. Diarrhea
FODMAP, fermentable oligosaccharides, disaccharides, is a commonly encountered problem with enteral feeding
monosaccharides, and polyols. in clinical practice. The FODMAP content of the enteral
feeding solution may play a role in the genesis of symptoms.
A retrospective study of patients with diarrhea caused by
initially treated with a low FODMAP diet and were then enteral feeding in Australia suggested that a longer hospital
allowed to liberalize their diet using a structured approach stay and a longer duration of enteral feeding were risk
were able to maintain the symptom improvement seen in the factors for diarrhea. Formula FODMAP levels in that
highly restrictive phase and were able to increase the amount study ranged from 10.6–36.5 g/day. A low FODMAP
of fiber in the diet. Interestingly, the microbiome was not enteral formula was associated with a 5-fold reduction in
affected in these patients.39 diarrhea rates.41 In a randomized, controlled trial of a low,
moderate, and high FODMAP enteral feeds in Korea, Yoon
Patient Support et al42 demonstrated a significant reduction in diarrhea
and improvement in nutrition parameters and clinical
Some patients welcome support tools and applications that
outcome in patients randomized to the low FODMAP
help them to choose low FODMAP foods. Table 3 lists re-
enteral feeding formula. Quantifying FODMAPs in enteral
sources that our patients have found useful in helping them
formulas has proven difficult because of the interference
navigate the FODMAP diet. They include an app that can
with in vitro assays of fructans and raffinose caused by
be used on mobile devices to identify the FODMAP content
the maltodextrin content of the formula.43 At the present
of foods, charts that identify low and high FODMAP-
time, there is no comprehensive list of low FODMAP-
containing foods, recipes, and support from others on the
containing enteral formulas in the United States, and the
same journey.
FODMAP content of Australian enteral feeding formulas
described by Halmos et al41 may not apply to North
Long-Term Monitoring of Nutrition Status America. Some enteral feeding formulas contain fructose
The effects the modified personalized FODMAP diet on and others contain inulin, and these are likely to have a
the gut microbiome are being actively studied as is the high FODMAP content. Controversy persists regarding the
benefit of adding probiotics. There is little information from use of fiber supplements in enteral tube feeding, and this
carefully conducted trials regarding the long-term nutrition debate has recently been summarized elsewhere.44 It should
impact of the FODMAP diet. In one study, iron intake was be noted that many enteral feeding formulas supplemented
474 Nutrition in Clinical Practice 33(4)

Table 3. Resources for Patients Being Started on the FODMAP Diet.

Resource Cost Link Comment

Monash University Mobile App $12.99 Apple and android app stores Links to an extensive database
of FODMAP containing
foods
Low FODMAP recipes & support Free https://www.facebook.com/groups/7435 Recipes and support group
group 58315679493/
Low FODMAP foods Variable https://www.fodyfoods.com Sites for low FODMAP foods
http://trueselffoods.com including energy bars
Reintroducing FODMAPs book $17.99 https://reintroducingfodmaps.com/ A book on FODMAP
reintroduction of patients
who need additional
information
A little bit yummy Free https://app.alittlebityummy.com/meal-plan/ Multiple FODMAP resources
FODMAP cookbooks Variable Online book stores FODMAP Cookbooks written
by registered dietitians
Low and high FODMAP checklists Free http://www.katescarlata.com/low Checklist for low and high
fodmapdietchecklists/ FODMAP containing foods

FODMAP, fermentable oligosaccharides, disaccharides, monosaccharides, and polyols.

with fiber have another FODMAP added to the solution References


(fructo-oligosaccharides).43 Further work is urgently 1. Kelm N, Levin R, Havel P. Carbohydrates in Modern Nutrition in
needed in this area. Health and Diseases. 11th ed. Philadelphia: Lippincott Williams &
Wilkins; 2014:36-57.
2. Marriott B, Cole N, Lee N. National estimates of dietary fructose
Future Directions With FODMAP and Dietary intake increase from 1977 to 2004 in the United States. J Nutr.
Therapy 2009;139(6):1228S-1235S.
3. Noelting J, DiBaise J. Mechanisms of fructose absorption. Clin Transl
Future research is directed at test strategies to determine Gastroenterol. 2015;6(11):e120.
who will benefit from the low FODMAP diet. We need more 4. Gibson PR, Newnham E, Barrett JS, Shepherd SJ, Muir JG. Review ar-
ticle: fructose malabsorption and the bigger picture. Aliment Pharmacol
data on the long-term outcome of treatment with the low
Ther. 2007;25(4):349-363.
FODMAP diet and its effects on the microbiome and the 5. Barrett JS, Gearry RB, Muir JG, et al. Dietary poorly absorbed, short-
nutrition status of the individual. An important area of chain carbohydrates increase delivery of water and fermentable sub-
research is in food science. The development of enzymes that strates to the proximal colon. Aliment Pharmacol Ther. 2010;31(8):874-
break down fructans in food could help enormous numbers 882.
6. Marciani L, Cox EF, Hoad CL, et al. Postprandial changes in small
of people who currently purchase gluten-free products at
bowel water content in healthy subjects and patients with irritable bowel
substantial cost. syndrome. Gastroenterology. 2010;138(2):469-477.
7. Major G, Pritchard S, Murray K, et al. Colon hypersensitivity to dis-
Acknowledgments tension, rather than excessive gas production, produces carbohydrate-
related symptoms in individuals with irritable bowel syndrome. Gas-
I am grateful to Amanda Motl, RD, for the care of our troenterology. 2017;152(1):124-133.
patients and for the information on the reintroduction phase 8. Shepherd S, Lomer M, Gibson P. Short-chain carbohydrates and func-
of FODMAP dietary therapy. tional gastrointestinal disorders. Am J Gastroenterol. 2013;108(5):707-
717.
Statement of Authorship 9. Ong DK, Mitchell SB, Barrett JS, et al. Manipulation of dietary short
chain carbohydrates alters the pattern of gas production and genesis
N. Vakil contributed to conception/design of the research; N. of symptoms in irritable bowel syndrome. J Gastroenterol Hepatol.
Vakil contributed to acquisition, analysis, or interpretation 2010;25(8):1366-1373.
of the data; N. Vakil drafted the manuscript; Nimish Vakil 10. Staudacher HM, Whelan K. The low FODMAP diet: recent advances
critically revised the manuscript; and N. Vakil agree to be fully in understanding its mechanisms and efficacy in irritable bowel syn-
accountable for ensuring the integrity and accuracy of the drome. Gut. 2017;66(8):1517-1527.
work. 11. Chumpitazi BP, McMeans AR, Vaughan A, et al. Fructans exacerbate
symptoms in a subset of children with irritable bowel syndrome. Clin
Supplementary Information Gastroenterol Hepatol. 2018;16(2):219-225.
12. Bourdu S, Dapoigny M, Chapuy E, et al. Rectal instillation of butyrate
Additional supporting information may be found online in the provides a novel clinically relevant model of noninflammatory colonic
Supporting Information section at the end of the article. hypersensitivity in rats. Gastroenterology. 2005;128(7):1996-2008.
Vakil 475

13. Zhou SY, Gillilland M 3rd, Wu X, et al. FODMAP diet mod- 30. Halmos EP, Christophersen CT, Bird AR, Shepherd SJ, Muir JG,
ulates visceral nociception by lipopolysaccharide-mediated intestinal Gibson PR. Consistent prebiotic effect on gut microbiota with altered
inflammation and barrier dysfunction. J Clin Invest. 2018;128(1):267- FODMAP intake in patients with Crohn’s disease: a randomised,
280. controlled cross-over trial of well-defined diets. Clin Transl Gastroen-
14. Gut Microbiota for Health. http://www.gutmicrobiotaforhealth.com/ terol. 2016;7:e164. https://doi.org/10.1038/ctg.2016.22
en/about-gut-microbiota-info/. Accessed March 24, 2018. 31. Zhan YL, Zhan YA, Dai SX. Is a low FODMAP diet beneficial
15. Staudacher H, Whelan K. Altered gastrointestinal microbiota in ir- for patients with inflammatory bowel disease? A meta-analysis and
ritable bowel syndrome and itsmodification by diet: probiotics, pre- systematic review [published online ahead of print May 24, 2017]. Clin
biotics and the low FODMAP diet. Proc Nutr Soc 2016;75(3):306- Nutr. https://doi.org/10.1016/j.clnu.2017.05.019
318. 32. Bennet SMP, Böhn L, Störsrud S, et al. Multivariate modelling of fae-
16. Halmos EP, Christophersen CT, Bird AR, Shepherd SJ, Gibson PR, cal bacterial profiles of patients with irritable bowel syndrome predicts
Muir JG. Diets that differ in their FODMAP content alter the colonic responsiveness to a diet low in FODMAPs [published online ahead of
luminal microenvironment. Gut. 2015;64(1):93-100. print April 17, 2017]. Gut. https://doi.org/10.1136/gutjnl-2016-313128
17. Staudacher HM, Lomer MCE, Farquharson FM, et al. A diet 33. Valeur J, Småstuen MC, Knudsen T, Lied GA, Røseth AG. Exploring
low in FODMAPs reduces symptoms in patients with irritable gut microbiota composition as an indicator of clinical response to
bowel syndrome and a probiotic restores bifidobacterium species: dietary FODMAP restriction in patients with irritable bowel syndrome
a randomized controlled trial. Gastroenterology. 2017;153(4):936- [published online ahead of print January 4, 2018]. Dig Dis Sci.
947. https://doi.org/10.1007/s10620-017-4893-3
18. Staudacher HM, Lomer MC, Anderson JL, et al. Fermentable car- 34. Rossi M, Aggio R, Staudacher HM, et al. Volatile organic
bohydrate restriction reduces luminal bifidobacteria and gastrointesti- compounds in feces associate with response to dietary intervention
nal symptoms in patients with irritable bowel syndrome. J Nutr. in patients with irritable bowel syndrome [published online
2012;142(4):1510-1518. ahead of print October 7, 2017]. Clin Gastroenterol Hepatol.
19. McIntosh K, Reed DE, Schneider T, et al. FODMAPs alter https://doi.org/10.1016/j.cgh.2017.09.055
symptoms and the metabolome of patients with irritable bowel 35. Wilder-Smith CH, Olesen SS, Materna A, Drewes AM. Predictors
syndrome: a randomised controlled trial. Gut. 2017;66(7):1241- of response to a low-FODMAP diet in patients with functional
1251. gastrointestinal disorders and lactose or fructose intolerance. Aliment
20. Fabisiak A, Włodarczyk J, Fabisiak N, Storr M, Fichna J. Targeting Pharmacol Ther. 2017;45(8):1094-1106.
histamine receptors in irritable bowel syndrome: a critical appraisal. 36. Whelan K, Martin LD, Staudacher HM, Lomer MCE. The low
J Neurogastroenterol Motil. 2017;23(3):341-348. FODMAP diet in the management of irritable bowel syndrome: an
21. Lacy B, Mearin F, Chang L, et al. Bowel disorders. Gastroenterology. evidence-based review of FODMAP restriction, reintroduction and
2016;150(6):1393-1407. personalisation in clinical practice [published online ahead of print
22. Marsh A, Eslick EM, Eslick GD. Does a diet low in FODMAPs January 15, 2018]. J Hum Nutr Diet. https://doi.org/10.1111/jhn.12530
reduce symptoms associated with functional gastrointestinal disorders? 37. Halpert A, Godena E. Irritable bowel syndrome patients’ perspectives
A comprehensive systematic review and meta-analysis. Eur J Nutr. on their relationships with healthcare providers. Scand J Gastroenterol.
2016;55(3):897-906. 2011;46(7-8):823-830.
23. Shepherd SJ, Gibson PR. Fructose malabsorption and symptoms of 38. Hulme K, Chilcot J, Smith MA. Doctor-patient relationship and
irritable bowel syndrome: guidelines for effective dietary management. quality of life in irritable bowel syndrome: an exploratory study of the
J Am Diet Assoc. 2006;106(10):1631-1639. potential mediating role of illness perceptions and acceptance. Psychol
24. Peters SL, Yao CK, Philpott H, et al. Randomised clinical trial: the Health Med. 2017;1-11. https://doi.org/10.1017/S0033291717003324.
efficacy of gut directed hypnotherapy is similar to that of the low [Epub ahead of print]
FODMAP diet for the treatment of irritable bowel syndrome. Aliment 39. Harvie RM, Chisholm AW, Bisanz JE, et al. Long-term irritable
Pharmacol Ther. 2016;44(5):447-459. bowel syndrome symptom control with reintroduction of selected
25. Maagaard L, Ankersen DV, Végh Z, et al. Follow-up of patients with FODMAPs. World J Gastroenterol. 2017;23(25):4632-4643.
functional bowel symptoms treated with a low FODMAP diet. World 40. Böhn L, Störsrud S, Liljebo T, et al. Diet low in FODMAPs
J Gastroenterol. 2016;22(15):4009-4019. reduces symptoms of irritable bowel syndrome as well as tradi-
26. McKenzie YA, Bowyer RK, Leach H, et al. British Dietetic Association tional dietary advice: a randomized controlled trial. Gastroenterology.
systematic review and evidence-based practice guidelines for the dietary 2015;149(6):1399-1407.e2
management of irritable bowel syndrome in adults (2016 update). 41. Halmos EP, Muir JG, Barrett JS, Deng M, Shepherd SJ, Gibson PR.
J Hum Nutr Diet. 2016;29(5):549-575. Diarrhoea during enteral nutrition is predicted by the poorly absorbed
27. Gluten-Free Products Market Analysis by Product (Bakery, Dairy short-chain carbohydrate (FODMAP) content of the formula. Aliment
Alternatives, Desserts & Ice-Creams, Prepared Foods, Pasta & Rice), Pharmacol Ther. 2010;32(7):925-933.
By Distribution (Grocery Stores, Mass Merchandiser, Club Stores), 42. Yoon SR, Lee JH, Lee JH, et al. Low-FODMAP formula improves
and Segment Forecasts, 2018–2025. https://www.grandviewresearch. diarrhea and nutritional status in hospitalized patients receiving enteral
com/industry-analysis/gluten-free-products-market. Accessed March nutrition: a randomized, multicenter, double-blind clinical trial. Nutr J.
3, 2018. 2015;14:116.
28. Biesiekierski JR, Peters SL, Newnham ED, Rosella O, Muir JG, Gib- 43. Halmos EP, Bogatyrev A, Ly E, Liels KL, Muir JG, Gibson PR.
son PR. No effects of gluten in patients with self-reported non-celiac Challenges of quantifying FODMAPs in enteral nutrition formulas:
gluten sensitivity after dietary reduction of fermentable, poorly ab- evaluation of artifacts and solutions. JPEN J Parenter Enteral Nutr.
sorbed, short-chain carbohydrates. Gastroenterology. 2013;145(2):320- 2017;41(8):1262-1271.
328. 44. Tarleton S, Kraft C, DiBaise J. Fiber-enriched enteral formulae:
29. Skodje GI, Sarna VK, Minelle IH, et al. Fructan, rather than gluten, advantageous or adding fuel to the fire? Pract Gastroenterol. https://
induces symptoms in patients with self-reported non-celiac gluten med.virginia.edu/ginutrition/wp-content/uploads/sites/.../Parrish_Dec
sensitivity [published online ahead of print November 2, 2017]. Gas- _13.pdf accessed 2/4/2018
troenterology. https://doi.org/10.1053/j.gastro.2017.10.040
Invited Review

Nutrition in Clinical Practice


Volume 33 Number 4
Nutrition Management of Necrotizing Enterocolitis August 2018 476–482

C 2018 American Society for

Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10115
wileyonlinelibrary.com

Vikram J. Christian, MBBS1 ; Elizabeth Polzin, MBA, RD, CD, CNSC2 ;


and Scott Welak, MD3

Abstract
Necrotizing enterocolitis (NEC) is one of the most significant causes of morbidity and mortality among premature infants. The
exact cause is considered multifactorial and related to gastrointestinal immaturity, inflammation and enteral feeding. The role of
nutrition is vitally important in NEC. The main modifiable risk factor is the introduction and advancement of enteral feedings.
After an infant has recovered from NEC, enteral feeds should be cautiously resumed to prevent injury from prolonged use of
parenteral nutrition. The logistics of how, when, and what to feed are somewhat unclear and often depend on the severity of the
disease. For patients with an enterostomy, refeeding the distal intestine with the small-intestinal ostomy output may improve bowel
growth and prevent long-term complications. (Nutr Clin Pract. 2018;33:476–482)

Keywords
enteral nutriton; necrotizing enterocolitis; parenteral nutrition; premature infant

Introduction and Background Nutrition Before NEC


Necrotizing enterocolitis (NEC) is one of the most signif- NEC is a disease that is almost exclusively found in prema-
icant causes of morbidity and mortality among extremely ture infants. The gastrointestinal tract develops throughout
premature infants.1 Although the disease may affect infants pregnancy, both in tissue growth and maturity. When an
of any gestational age (0.2/1000 live births), the disease infant is born prematurely, that development is altered. Be-
occurs in 10% of infants born <1.5 kg.1 Mortality occurs cause of the complications with prolonged PN use, enteral
in 3% of all cases of NEC, but increases to 30% among nutrition (EN) is required during the ex utero maturation of
those who require surgical intervention.2 Survivors of NEC the intestine.
are at risk for many long-term morbidities, most notably
gastrointestinal complications such as short gut syndrome
and dependence on parenteral nutrition (PN) for survival.
From the 1 Division of Gastroenterology, Department of Pediatrics,
In addition, these infants face an increased risk for neurode- Medical College of Wisconsin, Milwaukee, Wisconsin, USA;
velopmental impairment and chronic lung disease.2 2 Clinical Nutrition Department, Children’s Hospital of Wisconsin,

Despite decades of basic scientific and clinical research, Milwaukee, Wisconsin, USA; and 3 Division of Neonatology,
the pathophysiology of NEC is still incompletely under- Department of Pediatrics, Medical College of Wisconsin, Milwaukee,
Wisconsin, USA.
stood. The exact cause is considered multifactorial and is
related to gastrointestinal immaturity, inflammation and Financial disclosure: None declared.
enteral feeding. Treatments such as ventilator support, Conflicts of interest: None declared.
maintenance of hemodynamic stability, and intravenous This article originally appeared online on June 25, 2018.
fluid resuscitation are not specific to treatment of NEC, but
are aimed at reducing its effects. Podcast available
The role of nutrition is vitally important in NEC. The Listen to a discussion of this manuscript with NCP Editor-in-Chief
main modifiable risk factor is the introduction and advance- Jeanette M. Hasse, PhD, RD, LD, FADA, CNSC, and authors
Vikram J. Christian, MBBS, and Scott Welak, MD. This and other
ment of enteral feedings. After recovery from NEC, many
NCP podcasts are available at: https://onlinelibrary.wiley.com/
infants have complications related to nutrition, including page/journal/19412452/homepage/podcasts
malabsorption, surgical resection of the small intestine, and
special nutrition needs. This article reviews the relationship
Corresponding Author:
between NEC and nutrition, including the effects of feeding
Scott Welak, MD, Neonatology, Medical College of Wisconsin, 8701
before the disease and changes that occur after infants Watertown Plank Road, Milwaukee, WI 53226, USA.
recover from the disease. Email: swelak@mcw.edu
Christian et al 477

It is believed that NEC occurs from a combination of GI cantly different intestinal bacteria composition than term
immaturity, infection, inflammation, and enteral feeding.3 infants. These differences include a reduced diversity and
Of those risk factors, only enteral feeding is modifiable. In- number of bacteria, and higher proportion of pathogenic
deed, 90% of NEC occurs after infants have received enteral bacteria. Both laboratory and clinical studies have shown
feedings. There are methods of enteral feeding that have that the supplementation of probiotics reduces the incidence
been shown to reduce the risk for NEC. One of the most of NEC.14
important strategies is an early introduction to enteral feeds The mechanism of protection for probiotics is still being
following a protocol of steady advancement of feedings. studied, but there is strong evidence that it affects the
Most clinicians will start infants on feeds within the first signaling of Toll-Like Receptor-4 (TLR4).15 TLR4 is found
few days of life and introduce feeds at 10–30 mL/kg/d.4 in many different tissues but is known to be abundant in
Subsequent advancements are in 10–30 mL/kg/d intervals the small-intestinal epithelium during fetal development,
until the infant achieves his or her goal feeding volume. especially during the period premature infants are most at
When an infant demonstrates an intolerance to feeding risk for NEC. Laboratory studies have shown that TLR4
advancements, enteral feedings are often held at the current activation increases intestinal injury and inflammation in
volume or discontinued. animal models of NEC.16 The administration of probiotics
The most proven intervention to reduce the risk for has been shown to reduce TLR4 activation and prevent
NEC is a diet of exclusively human expressed breast subsequent inflammation and tissue injury.17
milk (EBM).5-8 The protective factors of EBM are not Although there is significant evidence showing beneficial
completely understood. EBM has a unique composition effects of probiotics, their use for the prevention of NEC
compared with cow’s milk formula, including a different in neonatal intensive care units (ICUs) is still limited.
casein/whey ratio that allows for easier digestion. EBM Even though probiotics are available over the counter in
also contains many potentially protective immunological the United States as a dietary supplement, using them to
factors that have been thought to protect infants against prevent or treat an illness requires additional regulation.
NEC. These factors include secretory immunoglobulin A, Currently, the U.S. Food and Drug Administration has
lysozyme, and lactoferrin, which participate in host defense; not approved any probiotic for use in premature infants
growth factors such as epidermal growth factors, insulin, in the prevention of NEC. There are obvious concerns
and insulin-like growth factor; anti-inflammatory cytokines that a provider may have regarding the administration of
such as interleukin-10; and several antioxidant enzymes to a bacteria and the subsequent risk of that species causing
prevent free radical damage.9 bacteremia. The preterm neonatal intestine is known to
Although many mothers are able and willing to provide have poor barrier function, and introducing bacteria into
EBM for their infants, some are unable to due to several the intestinal lumen could lead to a bacterial translocation.
factors including medical conditions, maternal medication There have been individual case reports of culture-positive
use, and inadequate free time. One study showed that only bacteremia in patients receiving lactobacillus. However,
30% of mothers were able to provide their own EBM to in the larger meta-analysis, there have been no increases
their premature infant throughout the entire NICU course.7 in culture-proven sepsis.18 Additional studies are needed
Due to the benefits of human milk for the premature to determine the proper method of probiotic adminis-
infants, the use of donor human breast milk (DHBM) tration, including the strain, dose, and timing of the
has increased in recent years.10 Studies have shown that probiotic.
the use of DHBM reduces the rates of NEC compared
with cow’s milk formula.5,11,12 One study demonstrated that
infants who received DHBM as their sole source of EN
PN During NEC
had a 79% reduced risk for NEC, and that the number to When an infant is diagnosed with NEC, all enteral feedings
treat to prevent NEC with DHBM is 18.5.13 In addition are halted, typically for 7–10 days. Although there is little
to preventing NEC, DHBM has been shown to have long- evidence for the length of holding enteral feedings, most
term benefits, such as an improved lipoprotein profile in clinicians use this time frame as a standard of practice.
adolescents. The time until enteral feeds can be resumed depends on
Despite substantial research, few interventions have the severity and extent of the disease. During this time, the
shown promise in reducing the risk for NEC in preterm infant must receive all hydration and nutrition parenterally.
infants. Many studies have shown improvement in the Due to the need for PN as nutrition support for at
prevention of NEC in the laboratory setting, but have not least 7 days, central access is typically necessary. Infants
translated into the clinical setting by significantly reducing who develop extensive disease often have multiorgan com-
the incidence or severity of the disease. The most promising promise and may require mechanical ventilation. Infants
development in the last decade has been use of probiotics. with significant disease often experience cardiovascular
Studies have shown that preterm neonates have a signifi- compromise and hypotension, requiring frequent volume
478 Nutrition in Clinical Practice 33(4)

resuscitation and inotropic support secondary to poor vas- overfeeding, cycling lipid and PN infusions, or use of lipid
cular tone and excessive capillary leak. Edema is very com- emulsion that includes fish oil.
mon and will cause an increase in total fluid requirements.
It is not uncommon to see infants receiving fluids in excess
of 180–200 mL/kg/d with severe NEC.
Because of excessive fluid administration, multiorgan
Management of EN After Uncomplicated NEC
failure, and subsequent renal insufficiency, infants with Once an infant recovers from NEC, reintroduction of EN is
NEC are at risk for electrolyte abnormalities. Excessive considered. The importance of enteral feeding to intestinal
capillary leak leads to edema of the intestine and soft tissues, growth and maturation is well established. Enteral feeding
and hyponatremia can subsequently occur. Although total prevents gut atrophy, enhances mucosal adaptation, and
body sodium may be relatively normal, their intravascular stimulates intestinal motility and growth.20 Mucosal villous
sodium levels are low, and infants with severe NEC often atrophy occurs within days in the absence of EN.4 This fact
require significant sodium administration. Clinicians should highlights the importance of EN and the need to restart
be aware of increasing requirements of sodium during this feeds when clinically appropriate.
time. There is a lack of consensus on when EN can be reintro-
Acidosis can also be a significant sequela of NEC. In duced post-NEC. Frantz21 in 1975 reported recurrence of
severe cases of NEC, intestinal tissue becomes insufficiently NEC in several patients who had enteral feedings reestab-
perfused and may become necrotic. Lactic acidosis can be lished <10 days after NEC. Recurrence of NEC is the
overwhelming, further worsening vascular tone and cardiac primary concern of resuming enteral feedings. Dimmitt22
output. It is common for infants to require acetate in their recommends bowel rest for 7–14 days, depending on the
PN to partially correct acidosis. The amount of acetate severity of the NEC episode. Most clinicians wait at least
delivered depends on the amount of sodium and potassium 1 week before resuming enteral feeds, reaching the goal of
in the PN. However, the main method for improving acidosis complete enteral feeds 7–10 days later.20
is to stop tissue injury and necrosis. Bohnhorst et al23 studied early vs late reintroduction
If the infant has severe intestinal injury, the risk for of enteral feeding after the diagnosis of NEC. Three con-
hyperkalemia increases. In most cases, hyperkalemia is not secutive days of lack of portal venous gas on abdominal
common. However, in patients with rapidly progressing ultrasounds was considered a prerequisite to initiation of
NEC, the risk increases. There is a subset of NEC patients enteral feeds. The median time of initiation of feeds in the
with Thomsen-Freidenrich (T) cryptantigen activation, early introduction group was 4 days after onset of NEC
which causes a rapid hemolysis and progressive intestinal for medically treated NEC and 7 days for surgically treated
injury. These patients have a very quick decline and are more NEC. The median time of initiation of feeds in the late in-
likely to require surgery for their disease.19 The mortality troduction group was 10 days after onset of NEC. Late rein-
risk is also increased. Given that infants with severe NEC troduction was associated with delayed completion of EN.
are often hypotensive and have poor renal perfusion, the Early introduction was associated with decreased duration
excretion of potassium can be compromised. Therefore, of central venous access, less catheter-related septicemia,
providers should be aware of the risk for hyperkalemia and and less time to hospital discharge when compared with
monitor electrolytes and urine output. the late introduction group. The authors concluded that
Patients with NEC are at risk for hypertriglyceridemia. early feeding after NEC is feasible and associated with fewer
Poor hepatic perfusion leading to poor fat metabolism can complications.23 The authors used a method of determining
occur in severe NEC. Administration of lipid in the PN can readiness for feeds (abdominal ultrasound) that is not
cause elevated triglyceride levels. Although some lipids are universally followed. In 1993, Stringer et al24 described
needed to prevent essential fatty acid deficiency, care should 12 neonates with recurrent NEC. The authors found no
be used when determining lipid dosage. consistent association between recurrent NEC and type or
Once patients have adequately recovered from NEC (at timing of enteral feeds.24
least 7 days), clinicians will resume enteral feedings. PN Given the lack of further studies supporting early
is continued until enteral feeds have reached the goal. In reintroduction of feeds, the practice of waiting at least
severe cases of NEC, especially when intestinal resection 7–14 days post-NEC may still be considered standard of
is required, some infants may take weeks to recover and care. The decision to introduce feeds may be based on
may not tolerate full enteral feedings for several months. clinical indicators and abdominal evaluations.20 Nonnu-
Prolonged PN use, defined as PN use for >21 days, may be tritive sucking (pacifier usage), which has been shown
required. Providers should be aware that prolonged PN use to increase mesenteric blood flow25 and trophic feeds
is a significant risk factor for cholestasis and PN-associated (<20 mL/kg/d), may be started initially to prepare the gut
liver disease. Some modifications can be made with long- for advancement of feeds. We recommend waiting at least 7
term PN use to reduce the risk for liver injury, such as not days before initiation of trophic feeds. Further advancement
Christian et al 479

of EN can be done when the following criteria have been sidered to be hypoallergenic and are expected to be well
met: tolerated in an infant with cow’s milk protein allergy. No
literature was found to support usage of elemental for-
• Adequate hemodynamics off vasoactive agents mulas in a post-NEC infant. Moreover, because elemental
• Reassuring abdominal examination formulas are likely more expensive than EHFs, we recom-
• Stable ventilation requirements mend usage of EHF in a post-NEC infant when cow’s
• Minimal to no electrolyte abnormalities milk protein allergy is presumed contributory or causative.
• Discontinuation of empiric antibiotics Despite lack of supporting data, there are specific situations
• Reassuring abdominal radiograph in which elemental formulas may be preferred over EHF.
These situations include the development/persistence of
Breast milk is the best EN for premature infants and symptoms resembling cow’s milk protein allergy and/or
is also the preferred source of EN as infants recover from the development of recurrent NEC in an infant who is
NEC.20,26 Breast milk contains growth hormones that pro- fed EHF.
mote adaptation of the gut and lactase that aids in digestion Reasonable advancement of feeds has been shown to
of lactose.26 However, because of maternal indications or decrease the incidence of NEC.26 While reintroducing EN,
parental preference, a cow’s milk formula may be used. following this practice may prevent recurrence of NEC.
In 2017, Embleton and Zalewski20 systematically reviewed Perks and Abad-Jorge26 recommended the bolus method of
the literature for data on reintroduction of feeds post- feeding, advancing at a rate of 10–35 mL/kg/d in cases of
NEC. No relevant trials focusing on the choice of enteral medical NEC. We recommend starting bolus feeds initially,
feeding were identified at the time. Perks and Abad-Jorge26 to provide 20 mL/kg/d, gradually advancing at the rate
recommended initiation of cow’s milk preterm formulas of 10–20 mL/kg/d, as tolerated. Feeds may be provided
with a higher caloric density because of the decreased by mouth or by nasogastric/gastrostomy tube. If bolus
lactose and increased medium-chain triglycerides (MCT) feeds are not tolerated, feeds may be run continuously
and long-chain triglycerides (LCT) in these products. We via nasogastric/gastrostomy tube. As enteral feeds are
recommend usage of these formulas for preterm infants. advanced, PN should be gradually weaned, until eventually
Standard term formula may be used in term infants after discontinued.
NEC.
Cow’s milk protein allergy has been implicated in the
pathogenesis of NEC. Gordon and colleagues27 in 2013
Management of EN After Bowel Resection
reclassified NEC based on risk factors that predisposed Reintroduction of feeds after surgical NEC is dependent
an infant to development of NEC. In doing so he de- on length of remaining bowel and patient’s clinical status.
fined “cow’s milk associated NEC” as a subset of NEC.27 Indications to start and advance feeds are the same as those
Chuang et al28 studied serum and intestinal markers of of uncomplicated NEC.
allergic-related inflammatory markers in patients with NEC Human breast milk remains the feed of choice in this
and compared them with specimens from infants with- population as well. Lapillonne et al32 conducted a na-
out inflammatory disease. They concluded that infants tionwide cross-sectional questionnaire-based study among
with NEC demonstrated significant cow’s milk protein French neonatal ICUs to describe the frequency and reasons
sensitization, as evidenced by markers in the peripheral for using EHF. The authors found that among 1969 hos-
blood. Minimal mucosal activation was also noted in some pitalized infants, 12% were receiving EHF. Eleven percent
cases with NEC.28 Faber et al29 reported a case of severe of these EHF prescriptions were due to previous NEC.
cow’s milk allergic colitis resembling necrotizing colitis in According to the survey, the main reasons for using an
a premature infant. Similar cases of NEC attributed to EHF when feeding post-NEC neonates were the absence of
cow’s milk allergy have been reported.30,31 These studies human milk (75%) and the need for surgical management of
suggest that cow’s milk allergy should be considered as NEC (17%). Because these formulas do not typically con-
a cause in cases of recurrent NEC or the occurrence of tain lactose and may contain significant amounts of MCTs,
NEC in the absence of typical risk factors. This also has the authors postulate that they may be better absorbed by
implications in selecting the enteral feed post-NEC. We an infant recovering from NEC.32 This advantage afforded
recommend usage of an extensively hydrolyzed formula to infants recovering from NEC may be unnecessary. This
(EHF) in these specific circumstances to prevent recurrence was determined by Schaart et al33 in 2007. In this study, the
of NEC in an infant who is likely allergic to cow’s milk protein content of small-intestinal enterostomy effluent was
protein. quantified in neonates who underwent bowel resection for
Elemental formulas or amino acid–based formulas, in different reasons (NEC, atresia, perforation). The authors
contrast with EHFs, consist of predominantly free amino concluded that the protein absorptive capacity of neonates
acids as the source of nitrogen. They are therefore con- after bowel resection is intact (70%–90% of total enteral
480 Nutrition in Clinical Practice 33(4)

Table 1. Summary of Recommendations.

Nutrition Before NEC

1. For stable ELBW infants, enteral feeding should be started within the first 48 hours of life.
2. Every effort should be made to provide ELBW infants with their own mother’s EBM.
3. If the mother does not provide EBM, donor human breast milk should be used.
4. Although there are promising studies showing a reduced rate of NEC by using probiotics, clinical trials are still
needed to determine which strain, dose, and timing of probiotics is best used for ELBW infants.

PN During NEC
1. Strongly consider central venous access at the diagnosis of NEC.
2. Provide adequate nutrition support through PN.
3. Anticipate infants having an increased need for fluid requirements secondary to capillary leak.
4. Closely monitor serum sodium and appropriately correct hyponatremia.
5. Anticipate metabolic acidosis and give adequate amounts of acetate.

EN After Uncomplicated NEC

1. Trophic feeds (<20 mL/kg/d) may be started via nasogastric/gastrostomy tube after at least 7 days of bowel rest.
2. EN may be advanced based on clinical indicators:
r Adequate hemodynamics off vasoactive agents
r Reassuring abdominal examination
r Stable ventilation requirements
r Minimal to no electrolyte abnormalities
r Discontinuation of empiric antibiotics
r Reassuring abdominal radiograph
3. Human breast milk (mother or donor-sourced) is the preferred feed.
4. If breast milk is unavailable, preterm formula with higher MCT and LCT should be fed to preterm infants.
5. Standard term formula may be fed to term infants.
6. Extensively hydrolyzed formulas may be considered if cow’s milk protein allergy is suspected (in patients with
recurrent NEC or in the absence of typical risk factors).
7. Bolus feeds may be started by mouth or by nasogastric/gastrostomy tube to provide 20 mL/kg/d.
8. Volume of feeds should be advanced gradually at a rate of 10–20 mL/kg/d.

EN After Bowel Resection


1. Indications to start trophic feeds and advance EN are the same as those of uncomplicated NEC.
2. Human breast milk (mother or donor-sourced) is the preferred feed.
3. Semi-elemental or amino acid–based formula may be considered if the patient is intolerant to conventional
preterm/term formulas.
4. Continuous feeds may be started by nasogastric/gastrostomy tube to provide 20 mL/kg/d.
5. Volume of feeds should be advanced gradually at a rate of 10–20 mL/kg/d.

EBM, human expressed breast milk; ELBW, extremely low birth weight; EN, enteral nutrition; LCT, long-chain triglycerides; MCT,
medium-chain triglycerides; NEC, necrotizing enterocolitis; PN, parenteral nutrition.

protein intake was absorbed).33 Perks and Abad-Jorge26 The authors also recommended advancing feeds at a rate
recommended the use of breast milk or semi-elemental or of 10–35 mL/kg/d.26 Continuous feeding is thought to be
amino acid–based formula initially. We recommend human nonphysiological, because it does not allow for the normal
breast milk, when available, or semi-elemental or amino pattern of gastrointestinal hormone release observed with
acid–based formulas should the patient be intolerant to bolus feedings. Despite this, we recommend starting with
conventional preterm/term formulas. continuous feeds via nasogastric/gastrostomy tube to
Reintroduction of feeds should be a gradual process. provide 20 mL/kg/d, gradually advancing at the rate of
Trophic feeds (<20 mL/kg/d) may be started when the 10–20 mL/kg/d. As the infant approaches the goal volume
patient is deemed ready to start feeds. Perks and Abad- of feeds, PN may be gradually weaned and eventually
Jorge26 recommended continuous feeds delivered on a discontinued.
pump over 24 hours, followed by gradually compressing the Accretion of micronutrient stores occurs in the third
continuous feeds as tolerated to a bolus-feeding schedule. trimester.34 Preterm infants are therefore at risk for
Christian et al 481

micronutrient deficiency and require supplementation. needed to identify other risks and benefits of this practice
Preterm infants recovering from NEC are thought to have and before a refeeding protocol can be recommended to best
increased needs for minerals, vitamins, electrolytes, zinc, and care for these patients.
iron.20 There is a higher risk for deficiency in infants who
have had a bowel resection. This deficiency would depend on Conclusion
the length and portion of bowel resected, presence and loca-
tion of ostomy, and ostomy output. Post-NEC infants may NEC is a complex disease with significant mortality and
also require additional supplementation of levo-carnitine if morbidity. Those who survive the disease are still at risk
there is prolonged use of PN and/or significant ostomy/stool for complications. Resuming enteral feeds can be difficult,
losses.26 with many short- and long-term complications. Enteral
feeds should be cautiously resumed once the infant has
recovered, to prevent long-term injury from prolonged PN
Refeeding of Ostomy Output use. The logistics of how and what to feed are somewhat
In the surgical infant with NEC where bowel resection unclear, and often depends on the severity of the disease.
is necessary, an enterostomy and mucous fistula may be For patients with an enterostomy, refeeding the distal
created.35 In an infant with an enterostomy, enteral feeds are intestine may improve bowel growth and prevent long-term
digested by the proximal intestine, and the effluent empties complications. Recommendations have been summarized
into an ostomy bag. This fluid is theoretically similar to in Table 1. Future studies should be aimed at clarifying the
that which would enter the large intestine, where nutrients best modalities in enteral feeding after NEC, and hopefully
and fluid would be absorbed by the colon. Therefore, provide improved methods of caring for infants who survive
patients with an enterostomy are not able to fully benefit this terrible disease.
from enteral feedings. However, some patients may benefit
from refeeding the ostomy effluent. Refeeding refers to Statement of Authorship
the ostomy output being collected and then refed through V. J. Christian, E. Polzin, and S. Welak contributed to the
the distal mucous fistula. This practice has been shown conception and design of the manuscript. V. J. Christian, E.
to decrease fluid loss and to improve electrolyte balance, Polzin, and S. Welak contributed to acquisition, analysis, or
bowel adaptation, and growth. The practice of refeeding interpretation of the data. V. J. Christian, E. Polzin, and S.
of proximal stoma effluent through a distal mucous fistula Welak drafted the manuscript. V. J. Christian, E. Polzin, and
uses the absorptive surface of the distal bowel for nutrient S. Welak critically revised the manuscript. V. J. Christian,
absorption, stimulates mucosal growth and intestinal adap- E. Polzin, and S. Welak agree to be fully accountable for
tation, and prevents atrophy of the distal bowel.36-38 Infants ensuring the integrity and accuracy of the work, and all
who undergo mucous fistula refeeding have a shorter time authors read and approved the final manuscript.
from initiation of enteral feeds to attainment of goal feeds
and PN discontinuation in the interoperative period.39
The practice of refeeding the output to the mucous fistula References
is thought to improve bowel adaptation and decrease time
1. Abdullah F, Zhang Y, Camp M, et al. Necrotizing enterocolitis
to full enteral feeds, but it does come with some risk.40 in 20,822 infants: analysis of medical and surgical treatments. Clin
The safety of refeeding has posed concern for risk for Pediatr. 2010;49(2):166-171.
perforation, GI bleed, sepsis, and mortality.35 Refeeding 2. Schnabl KL, Van Aerde JE, Thomson AB, Clandinin MT. Necrotizing
is thought to be safe and beneficial when administered enterocolitis: a multifactorial disease with no cure. World J Gastroen-
terol. 2008;14(14):2142-2161.
in an ICU using a protocol that specifies the equipment
3. Neu J, Walker WA. Necrotizing enterocolitis. N Engl J Med.
and techniques used for mucous fistula cannulation and 2011;364(3):255-264.
refeeding parameters.39 Gause et al39 described a refeeding 4. Kliegman RM, Walker WA, Yolken RH. Necrotizing enterocolitis:
protocol using a 6.0 Fr Foley catheter placed by a surgeon research agenda for a disease of unknown etiology and pathogenesis.
while monitoring for signs of vascular compromise. The Pediatr Res. 1993;34(6):701-708.
5. Sullivan S, Schanler RJ, Kim JH, et al. An exclusively human milk-
authors’ protocol consisted of refeeding the succus entericus
based diet is associated with a lower rate of necrotizing enterocolitis
collected from the proximal enterostomy and instilling this than a diet of human milk and bovine milk-based products. J Pediatr.
into the mucous fistula via an electronic feeding pump at a 2010;156(4):562-567.e561.
rate of 1:1.39 The authors recommended replacement of the 6. Quigley M, McGuire W. Formula versus donor breast milk for feeding
catheter in case of dislodgement in a patient who had been preterm or low birth weight infants. Cochrane Database Syst Rev.
2014(4):CD002971.
tolerating refeeding. Forming an institution-specific proto-
7. Schanler RJ, Lau C, Hurst NM, Smith EO. Randomized trial of
col for mucous fistula refeeding is a necessary starting point donor human milk versus preterm formula as substitutes for mothers’
before initiation of refeeding in a neonatal unit. There are own milk in the feeding of extremely premature infants. Pediatrics.
no studies comparing refeeding protocols. More research is 2005;116(2):400-406.
482 Nutrition in Clinical Practice 33(4)

8. Lucas A, Cole TJ. Breast milk and neonatal necrotising enterocolitis. 25. Huang CF, Tsai MC, Chu CH, Lee MY, Shih YL, Cheng SN. The
Lancet. 1990;336(8730):1519-1523. influence of pacifier sucking on mesenteric blood flow in infants. Clin
9. Hanson L, Silfverdal SA, Stromback L, et al. The immunological Pediatr. 2003;42(6):543-546.
role of breast feeding. Pediatr Allergy Immunol. 2001;12(suppl 14): 26. Perks P, Abad-Jorge A. Nutritional management of the infant with
15-19. necrotizing enterocolitis. Pract Gastroenterol. 2008;32(2):46-60.
10. McGuire W, Anthony MY. Donor human milk versus formula for pre- 27. Gordon P, Christensen R, Weitkamp JH, Maheshwari A. Mapping the
venting necrotising enterocolitis in preterm infants: systematic review. new world of necrotizing enterocolitis (NEC): review and opinion. EJ
Arch Dis Child Fetal Neonatal Ed. 2003;88(1):F11-F14. Neonatol Res. 2012;2(4):145-172.
11. Cristofalo EA, Schanler RJ, Blanco CL, et al. Randomized 28. Chuang SL, Hayes PJ, Ogundipe E, Haddad M, MacDonald TT, Fell
trial of exclusive human milk versus preterm formula diets in JM. Cow’s milk protein-specific T-helper type I/II cytokine responses
extremely premature infants. J Pediatr. 2013;163(6):1592-1595. in infants with necrotizing enterocolitis. Pediatr Allergy Immunol.
e1591. 2009;20(1):45-52.
12. Kantorowska A, Wei JC, Cohen RS, Lawrence RA, Gould JB, Lee HC. 29. Faber MR, Rieu P, Semmekrot BA, Van Krieken JH, Tolboom JJ,
Impact of donor milk availability on breast milk use and necrotizing Draaisma JM. Allergic colitis presenting within the first hours of
enterocolitis rates. Pediatrics. 2016;137(3):e20153123. premature life. Acta Paediatr. 2005;94(10):1514-1515.
13. Boyd CA, Quigley MA, Brocklehurst P. Donor breast milk versus in- 30. de Peyer E, Walker-Smith J. Cow’s milk intolerance presenting as
fant formula for preterm infants: systematic review and meta-analysis. necrotizing enterocolitis. Helv Paediatr Acta. 1978;32(6):509-515.
Arch Dis Child Fetal Neonatal Ed. 2007;92(3):F169-F175. 31. Walther FJ, Kootstra G. Necrotizing enterocolitis as a result of cow’s
14. AlFaleh K, Anabrees J. Probiotics for prevention of necrotiz- milk allergy? Z Kinderchir. 1983;38(2):110-111.
ing enterocolitis in preterm infants. Cochrane Database Syst Rev. 32. Lapillonne A, Matar M, Adleff A, Chbihi M, Kermorvant-Duchemin
2014(4):CD005496. E, Campeotto F. Use of extensively hydrolysed formula for refeed-
15. Leaphart CL, Cavallo J, Gribar SC, et al. A critical role for TLR4 in ing neonates postnecrotising enterocolitis: a nationwide survey-based,
the pathogenesis of necrotizing enterocolitis by modulating intestinal cross-sectional study. BMJ Open. 2016;6(7):e008613.
injury and repair. J Immunol. 2007;179(7):4808-4820. 33. Schaart MW, de Bruijn AC, Tibboel D, Renes IB, van Goudoever JB.
16. Gribar SC, Anand RJ, Sodhi CP, Hackam DJ. The role of epithelial Dietary protein absorption of the small intestine in human neonates.
Toll-like receptor signaling in the pathogenesis of intestinal inflamma- JPEN J Parenter Enteral Nutr. 2007;31(6):482-486.
tion. J Leukoc Biol. 2008;83(3):493-498. 34. Shaikhkhalil AK, Curtiss J, Puthoff TD, Valentine CJ. Enteral zinc
17. Patel RM, Underwood MA. Probiotics and necrotizing enterocolitis. supplementation and growth in extremely-low-birth-weight infants
Semin Pediatr Surg. 2018;27(1):39-46. with chronic lung disease. J Pediatr Gastroenterol Nutr. 2014;58(2):183-
18. Sawh SC, Deshpande S, Jansen S, Reynaert CJ, Jones PM. Prevention 187.
of necrotizing enterocolitis with probiotics: a systematic review and 35. Haddock CA, Stanger JD, Albersheim SG, Casey LM, Butterworth
meta-analysis. PeerJ. 2016;4:e2429. SA. Mucous fistula refeeding in neonates with enterostomies. J Pediatr
19. Hall N, Ong EG, Ade-Ajayi N, et al. T cryptantigen activation is Surg. 2015;50(5):779-782.
associated with advanced necrotizing enterocolitis. J Pediatr Surg. 36. Koike Y, Uchida K, Nagano Y, et al. Enteral refeeding is useful for
2002;37(5):791-793. promoting growth in neonates with enterostomy before stoma closure.
20. Embleton ND, Zalewski SP. How to feed a baby recovering from J. Pediatr Surg. 2016;51(3):390-394.
necrotising enterocolitis when maternal milk is not available. Arch Dis 37. Richardson L, Banerjee S, Rabe H. What is the evidence on the practice
Child Fetal Neonatal Ed. 2017;102(6):F543-F546. of mucous fistula refeeding in neonates with short bowel syndrome?
21. Frantz ID 3rd, L’Heureux P, Engel RR, Hunt CE. Necrotizing J Pediatr Gastroenterol Nutr. 2006;43(2):267-270.
enterocolitis. J Pediatr. 1975;86(2):259-263. 38. Al-Harbi K, Walton JM, Gardner V, Chessell L, Fitzgerald PG.
22. Dimmitt RA, Moss RL. Clinical management of necrotizing entero- Mucous fistula refeeding in neonates with short bowel syndrome.
colitis. NeoReviews. 2001;2(5):110-117. J Pediatr Surg. 1999;34(7):1100-1103.
23. Bohnhorst B, Muller S, Dordelmann M, Peter CS, Petersen C, Poets 39. Gause CD, Hayashi M, Haney C, et al. Mucous fistula refeeding
CF. Early feeding after necrotizing enterocolitis in preterm infants. decreases parenteral nutrition exposure in postsurgical premature
J Pediatr. 2003;143(4):484-487. neonates. J Pediatr Surg. 2016;51(11):1759-1765.
24. Stringer MD, Brereton RJ, Drake DP, Kiely EM, Capps SN, Spitz 40. Lau EC, Fung AC, Wong KK, Tam PK. Beneficial effects of mucous
L. Recurrent necrotizing enterocolitis. J Pediatr Surg. 1993;28(8): fistula refeeding in necrotizing enterocolitis neonates with enteros-
979-981. tomies. J Pediatr Surg. 2016;51(12):1914-1916.
Review

Nutrition in Clinical Practice


Volume 33 Number 4
Enteral Nutrition in the Management of Crohn’s Disease: August 2018 483–492

C 2018 American Society for

Reviewing Mechanisms of Actions and Highlighting Potential Parenteral and Enteral Nutrition
DOI: 10.1002/ncp.10004
Venues for Enhancing the Efficacy wileyonlinelibrary.com

Moftah H. Alhagamhmad, MBBCH, DCH UK, PhD

Abstract
Crohn’s disease (CD) is a chronic condition that affects the gut and has adverse effects on growth and development. There is a
global increase in the incidence and prevalence rates, and several factors are believed to contribute to this rise, including dietary
habits. In contrast, the use of enteral nutrition (EN) as an exclusive source of nutrition is increasingly becoming the preferred
induction treatment of pediatric CD patients in part to address the nutrition complications. However, EN therapy is considered
less effective in adults with CD. A better understanding of the molecular mechanisms of enteral therapy will help improve the
clinical management of CD. It is increasingly becoming evident that the therapeutic utility of EN is in part due to the reversal of
the microbial changes and the direct immunomodulatory effects. Moreover, there is a potential tendency for enhancing the efficacy
of EN therapy by improving the palatability of the given formulas and, more important, by magnifying the anti-inflammatory
properties. Recent observations have shown that the immunomodulatory effects of EN are mediated at least in part by blocking
nuclear factor-κB. Furthermore, it is likely that several ingredients of EN contribute to this activity, in particular glutamine and
arginine amino acids. In addition, manipulating the composition of EN therapy by altering concentrations of the key ingredients
is found to have the potential for more efficient therapy. In this review, the underlying mechanisms of EN actions will be discussed
further with a focus on the potential methods for enhancing the efficacy. (Nutr Clin Pract. 2018;33:483–492)

Keywords
Crohn’s disease; enteral nutrition; nutrition therapy; inflammation; immunomodulation

Crohn’s disease (CD) is an entity of inflammatory bowel sion, addressing the nutrition complications is a vital goal
diseases (IBD) and can affect any part of the gastrointestinal in CD management.8 It is noteworthy that enteral nutrition
tract.1 It is a chronic, lifelong disorder that can potentially (EN) with a polymeric formula (PF) and/or elemental
compromise the quality of life and is characterized by formula given as a sole nutrition source is increasingly be-
alternating periods of remission and relapse, imposing a coming the first choice as an induction treatment of young
considerable negative impact on all aspects of life.2 In patients with active CD.9 In addition to its advantage in
addition to the chronic and current incurable nature of CD, terms of correcting the nutrition imbalances, enteral diet in
there is an increase in the global incidence and prevalence the form of exclusive enteral nutrition (EEN) is considered
rates.3 Recent reports show that CD is no longer confined, a safe therapy with a therapeutic efficacy incomparable with
as previously known, to the Western countries; rather, it that of corticosteroids in inducing remissions and, more
is increasingly becoming more prevalent in other parts of
the world with a concomitant rise in CD diagnoses among
From the Faculty of Medicine, University of Benghazi (Al-Arab
children.4 Changes in dietary habits along with the other
Medical University), Benghazi, Libya.
environmental triggers are believed to account for the recent
Financial disclosure: None declared.
changes in the patterns of CD epidemiology.4
Further challenges involved in the management of CD Conflict of interest: None declared.
include the accompanying nutrition complications, includ- Received for publication April 21, 2017; accepted for publication
ing impairments in growth and development.5 It has been October 10, 2017.
reported that around 50% of CD patients may be under- This article originally appeared online on January 11, 2018.
weight and stunted at the time of diagnosis.6 Furthermore, Corresponding Author:
the compromised nutrition status in these children may fur- Moftah Alhagamhmad, MBBCH, DCH UK, PhD, Benghazi
ther deteriorate with the use of immunosuppressive drugs.7 University (Al-Arab Medical University), Medical School, P.O. Box
18251, Hawari Road, Benghazi, Libya.
Thus, it appears that in addition to the induction of remis-
Email: sufrani82@yahoo.com
484 Nutrition in Clinical Practice 33(4)

Figure 1. The changes accompanying Crohn’s disease, including gut microbial changes and loss of integrity of intestinal epithelial
barrier with subsequent overactivation of mucosal immune responses.

important, superior efficacy in enhancing mucosal healing.6 a loss of function of the intestinal epithelial barrier al-
This review discusses the underlying mechanisms of action lows trafficking various agents, including luminal bacteria,
of enteral therapy and highlights the potential means for initiating uncontrolled compensatory immune responses
enhancing the efficacy. with concomitant overexpressions and the release of the
immunoregulatory cytokines.19

Pathogenesis of Crohn’s Disease Mechanisms of Action


For a better understanding of the mode of action of EN and Although there is a considerable gap in our understanding
its role in suppressing intestinal inflammation, the basic con- about the exact mechanisms of action of EN, there is
cepts of CD aetiopathogenesis are briefly described in this substantial evidence indicating that EN exerts therapeutic
section (Figure 1). The pathogenesis is considered multifac- benefits by interfering with the integral components of IBD
torial with genetic, microbial, immunologic, environmental, pathology.20 Immunomodulation, profound changes in the
and other adjuvant factors playing a role.10 Altered immune gut microbiota, and enhanced mucosal healing of the leaky
regulation is one of the suggested underlying mechanisms gut appear to be the key factors beyond the attenuation of
of CD, leading to the ongoing inflammatory processes with inflammation and induction of clinical remissions following
subsequent tissue destruction.11 An exaggerated immune treatment with EN21 (Figure 2).
response is characterized by a rise in the proinflammatory
cytokines levels along with a drop in the anti-inflammatory
mediators.12 Gene expression of the proinflammatory me-
Anti-Inflammatory Property
diators is modulated through activation of the complex The initial observation on the anti-inflammatory effects
intracellular pathways, most notably nuclear factor (NF)- of EN was made on a group of CD patients with resis-
κB13,14 and mitogen protein kinase cascades.15 tant disease who had undergone remission while receiv-
Furthermore, it has been established that changes in the ing elemental formula as preoperative nutrition support.22
gut microbiota also play a crucial role in the pathogenesis Thereafter, several trials provided stronger evidence of the
and development of CD.16 Dysbiosis, or bacterial imbal- immunomodulatory effects of EN.23 Certainly, in numerous
ance, is a leading factor that disrupts the host–immune clinical trials involving CD patients, the EEN regimen
system cross-talk with subsequent perturbation of mucosal exerted a remarkable immunosuppressive effect, resulting
homeostasis.17 Moreover, an increase in the intestinal per- in inhibition of cytokines’ production with a concomitant
meability is an additional contributory factor involved in the fall in the mucosal levels.24-26 In a trial involving 21 patients
disruption of the host–microbiome harmony.18 Certainly, with active CD, treatment with 8 weeks of EEN using PF
Alhagamhmad 485

Figure 2. Mode of action of enteral therapy in attenuating intestinal inflammation by reversing the pathological changes
characterizing Crohn’s disease. NF-κB, nuclear factor-kappa B; Iκκ, Inhibitor of kappa-B kinase; IκB, inhibitor of kappa-B; P,
phosphorylation.

resulted in a significant drop in the cytokine expression in of several genes linked to the NF-κB pathway, indicating
the colonic and ileal biopsies.27 Similar observations were that EN could exert its effect by altering the activity of the
made in another trial that used elemental formula as an intracellular signaling pathways.30 However, this study was
induction therapy in 28 patients with active CD.25 The unable to determine the determinants of PF activity and
mucosal concentrations of the key inflammatory mediators the exact nature of the interaction with the NF-κB pathway.
in the collected biopsies of CD patients were not compa- Thereafter, additional in vitro experiments were conducted
rable with those in the biopsies of matched controls.25 It is to further explore the previous observations.31 Glutamine,
notable that in another trial on 12 CD patients undergoing arginine, vitamin D3 , and α linolenic acid, present in PF,
a 6-week EEN protocol, the anti-inflammatory effects of were identified as potential immunoactive nutrients, and
the given EN therapy proceeded its nutrition values.28 The their effects were studied on the stimulated epithelial cells.31
disease activity score and raised inflammatory markers were The authors noted that at concentrations equivalent to those
corrected in the initial days of treatment, whereas changes used in PF, glutamine, arginine, and vitamin D3 (at final con-
in the nutrition parameters, including weight scores and centrations of 12.7 mM, 1.8 mM, and 3.8 nM, respectively,
subcutaneous fat thickness, were only visible by the second in the culture media) can suppress inflammation, indicating
and third weeks of treatment initiation.28 that these components are likely at least in part to contribute
EN can alter the exaggerated immune responses and to the immunomodulatory effects observed following EEN
thereby suppress inflammation. However, the exact mecha- treatment.31 However, the observed effect of PF with its
nisms of its interaction with the mucosal immune system to whole components was superior to that of combined glu-
attenuate intestinal inflammation remain unclear. De Jong tamine, arginine, and vitamin D3 in blocking cytokine pro-
et al29 investigated the effect on 3 different immortalized duction from the stimulated cells.31 This indicates that there
intestinal epithelial cells cultured as an in vitro model are other currently unidentified ingredients of PF that might
of intestinal inflammation and showed that the therapy be contributing to its activity.32 The authors also showed
exerted a direct suppressing effect. The treatment with PF that the glutamine and arginine ingredients of PF can block
blocked tumor necrosis factor (TNF)-α induced interleukin phosphorylation, a prerequisite process for the activation
(IL)-8 responses in the stimulated cells. The block in IL-8 of the key active components of NF-κB and P38 mitogen
production was consistent with a delay in the degradation of protein kinase cascades,31 thereby blocking the major source
the inhibitory κ-B subunit, the major regulatory molecule of of proinflammatory cytokine production. Glutamine and
NF-κB transcription factor. Subsequently, using the same arginine can interfere with phosphorylation directly through
model, Nahidi et al30 further examined the interactions of potential competitive adenosine 5’-triphosphate binding
PF with the NF-κB pathway. PF modulated the expressions in the signaling components and indirectly by enhancing
486 Nutrition in Clinical Practice 33(4)

Table 1. Studies on the Role of EN in Suppressing Inflammation.

Date, first author, reference, journal


name Study description and main findings

1973 Voitk et al22 Observational study: First report on the immunosuppressive effects of EN made on a
Archives of Surgery group of IBD patients waiting for surgery while receiving elemental formula as
preoperative nutrition support.
1994 Beattie et al26 Descriptive study: Effect of PF on 7 children with CD was evaluated. All children
Alimentary Pharmacology & experienced reduction in the disease activity and 6 of the total patients showed a
Therapeutics reduction in mucosal cytokines levels.
2000 Fell et al27 Prospective cohort study: Efficacy of EN in inducing remission was investigated on
Alimentary Pharmacology & 21 pediatric CD patients. Remission rate was 79% following 8-week regimen of PF
Therapeutics together with a significant reduction in mRNA expression of colonic and ileal
proinflammatory cytokines.
2007 de Jong et al29 In vitro model of IBD utilized intestinal epithelial cells cultured in lab: Treatment
Digestive Diseases and Sciences with PF suppressed TNF-α induced IL-8 productions from the stimulated cells.
2015 Nahidi et al30 In vitro model of IBD as above: Altered immune responses following the treatment
Genes & Nutrition with PF were shown to be mediated through modulation of the NF-κB pathway.
2016 Alhagamhmad et al31 In vitro model of IBD as above: Glutamine, arginine, and vitamin D3 part of PF
Journal of Parenteral and Enteral prompted a reduction in IL-8 production from the activated cells (potential key
Nutrition components of PF). Glutamine and arginine exerted their effects through
interfering with phosphorylation of NF-κB and P38 MAPK pathways. Amino
acids enriched PF prompted superior immunomodulating effects over standard PF.

CD, Crohn’s disease; EN, enteral nutrition; IBD, inflammatory bowel disease; IL, interleukin; MAPK, mitogen protein kinase cascades; NF,
nuclear factor; PF, polymeric formula; TNF, tumor necrosis factor.

production of intermediary active metabolites such as nitric also observed that 7 patients of the EEN group achieved
oxide that is involved in nitrosylation, blocking the tran- complete mucosal healing vs none in the corticosteroids
scription molecules.31 Collectively, the therapeutic utility group.38
of EN appears to be mediated at least in part by the However, the exact mechanism by which EN restores
immunomodulatory effects exerted through the deactiva- the epithelial barrier function has been poorly investigated
tion of the major intracellular signaling pathways NF-κB and remains unclear. It is well accepted that alterations
and P38 mitogen protein kinase cascades. Furthermore, it in the intestinal epithelial barrier function are caused by
is likely that several individual components contribute to an overproduction of the proinflammatory mediators such
this property, most notably glutamine and arginine amino as TNF-α.39,40 TNF-α enhances intestinal epithelial per-
acids. Key studies on the role of EN in attenuating immune meability by modulating the protein expression of myosin
responses are summarized in Table 1. II regulatory light-chain kinase (MLCK).41 Nahidi et al42
investigated the effects of PF on intestinal tight junction
barrier function in the presence of TNF-α in an in vitro
Enhancing Mucosal Healing model using Caco-2 epithelial monolayers. Monolayer per-
Disruption of the integrity of the intestinal epithelial bar- meability was studied, and MLCK gene expression was eval-
rier, a prominent feature of CD, accounts for trafficking uated. In these experiments, TNF-α exposure increased the
of the luminal bacteria and other antigenic agents, re- monolayer permeability, upregulated MLCK expression,
sulting in disruption of the gut microbial homoeostasis and reduced tight junction integrity.42 It is interesting to
and in turn stimulating the mucosal immune system.33 note that these changes were reversed completely with the
Thus, promoting mucosal healing has become a key goal use of PF in association with the inhibition of MLCK.42
in IBD management.34 It is noteworthy that numerous These observations were further supported by studies on
clinical studies have shown that enteral diet, administered a mouse model of colitis, characterized by increased gut
as EEN, is capable of enhancing mucosal healing in treated inflammatory markers and reduced gut barrier function.43
CD patients.35,36 Furthermore, EN is superior to corticos- EEN treatment, given as PF, maintained the gut barrier
teroids in terms of induction of higher mucosal remission integrity and function, reversed the inflammatory changes,
rates.37 In a trial involving 47 young CD patients, the and ameliorated colitis. The authors also found that these
investigators noted that around two-thirds of the children changes were correlated with the down-regulation of the
receiving EEN achieved mucosal healing compared with gene expression of proinflammatory cytokines involved in
<50% of those who received corticosteroids.38 The authors the disruption of tight junction proteins.43
Alhagamhmad 487

Table 2. Studies on the Role of EN in Mucosal Healing.

Date, first author, reference, journal


name Study description and main findings

2006 Borrelli et al37 Randomized controlled trial: A group of 37 children with active CD who were
Clinical Gastroenterology and randomized to receive either EN in THE form of PF or corticosteroids and healing
Hepatology of gut mucosa was assessed. PF showed superior efficacy over corticosteroids in
healing of gut inflammatory lesions.
2012 Nahidi et al42 In vitro model using Caco-2 epithelial monolayers: Effect of EN upon TNF-α
Journal of Gastroenterology induced disrupted intestinal tight junction function was assessed. PF enhanced the
integrity and reduced permeability in conjunction with reduced expression of
myosin II regulatory light-chain kinase protein expression.
2013 Nahidi et al43 Mouse model of colitis: Gut barrier function was assessed following treatments with
Biomed Research International EEN and corticosteroids. PF was superior to corticosteroids, given as
hydrocortisone, in maintaining epithelial barrier integrity and reversing
accompanied inflammatory changes.
2014 Grover et al35 Prospective open label study: Efficacy of EN in inducing endoscopic remission was
Journal of Gastroenterology investigated. Treatment with EEN was effective in inducing mucosal and
transmural remissions and reduced endoscopic relapses at 1 year.

CD, Crohn’s disease; EEN, exclusive enteral nutrition; EN, enteral nutrition; PF, polymeric formula; TNF, tumor necrosis factor.

The TNF-α induced increase in the MLCK gene activity were correlated with disease activity during and after EEN
is mediated by activation of NF-κB and vice versa.40,44 therapy.48
Although it was not directly investigated in the previous How precisely enteral therapy modulates changes in the
in vitro study,42 the downregulation of TNF-α-induced gut microbiota, however, remains unknown. It has been
MLCK protein expression by PF treatment is a potential proposed that enteral diets have prebiotic properties that
indicator of the inhibition of NF-κB activity by EEN enable the modification of microbiota.46 One study has
treatment. Overall, it appears that the anti-inflammatory also proposed limiting nutrient access during the EEN
effects and enhanced mucosal healings observed after EN regimen to starve the bacteria residing in the colon.49
treatment are mediated by attenuating production of the According to an interesting finding reported by a recent
proinflammatory mediators via blocking of the major in- observational study, EN can alter the metabolism of colonic
tracellular signaling pathways. Major studies that have gut microbiota.50 Significant reductions in the fecal concen-
explored the role of EN in mucosal healing are summarized trations of microbial metabolites (short-chain fatty acids, 1-
in Table 2. propanol, 1- butanol, and esters of short-chain fatty acids)
were noted following 2 weeks of EN treatment in CD
patients. These changes were consistent with improvements
Gut Microbiota Modification in the patients’ symptoms along with a fall in the levels of
Additional factors that contribute to the therapeutic ben- their measured inflammatory markers.50 Key studies that
efits of EN in CD management are mediated through explored the influence of EN on microbiota and dysbiosis
modifications in the gut microbiota.45 Using the tempera- are summarized in Table 3.
ture gradient gel electrophoresis technique, the microflora An additional mechanism of action of enteral therapy
in the stool samples of CD patients who received an involves resting the bowel51 and excluding offensive dietary
enteral diet for 8 weeks and those of comparatively healthy factors involved in exacerbations of immune responses,
controls were investigated.46 Major modifications in the thereby interrupting the so-called “bacterial penetration
microflora of the collected samples were observed following cycle.”52 Previous findings provided evidence that there are
the EEN treatment.46 Significant alterations were also noted a variety of diets and food preservatives that can compro-
in the fecal bacterial composition following EN treatment mise the integrity of the intestinal epithelial barrier.53,54
in another study that used an alternative technique (de- In the “bacterial penetration cycle” hypothesis, it has been
naturing gradient gel electrophoresis).47 Furthermore, in proposed that the loss of intestinal epithelial barrier func-
a recent study that used whole-genome high-throughput tion, secondary to consumed dietary factors, allows the
sequencing techniques, the microbial diversity in EEN- adherence and trafficking of bacteria and other antigenic
treated CD patients was significantly lower than that in the substances, triggering the mucosal immune system.55 This
matched controls.48 The authors were also able to identify in turn results in exaggerated immune responses and sub-
several bacterial species within the Firimictus family that sequent tissue destruction with further loss in the integrity
488 Nutrition in Clinical Practice 33(4)

Table 3. Studies on the Role of EN in Microbiota Modifications.

Date, first author, reference, journal


name Study description and main findings

2005 Lionetti et al46 Prospective study: Fecal microflora were collected from 9 children and adults with
Journal of Parenteral and Enteral active CD treated with PF and 5 healthy children and assessed with 16S rRNA
Nutrition gene and temperature gradient gel electrophoresis. The microflora were modified in
all treated CD patients.
2008 Leach et al47 Prospective study: Fecal samples of 6 CD patients and matched healthy controls were
Alimentary Pharmacology & collected before, during, and after EN treatment, and the diversity was evaluated
Therapeutics using denaturing gradient gel electrophoresis. Significant changes in the
composition of bacteria was noted with EN treatment and persisted for 4 months.
2015 Kaakoush et al48 Prospective study: Fecal samples collected from 5 CD patients treated with EN and
Clinical and Translational matched healthy controls were assessed using 16S rRNA gene and whole-genome
Gastroenterology high throughput sequencing. Microbial diversity in CD patients was significantly
lower than that in matched controls and the number of operational taxonomic
units, reflecting microbial diversity and decreased substantially upon starting EN
treatment and correlated with a drop in disease activity. A number of bacterial
species within the Firimictus family was found correlating with disease activity.
2016 Walton et al50 Observational study: Fecal samples collected from CD patients treated with EN and
European Journal of Clinical Nutrition healthy controls were studied by gas chromatography and mass spectrometry for
investigating effects of enteral therapy on gut microbiota metabolism. A reduction
in the fecal concentrations of microbial metabolites, including SCAFs, 1-propanol,
1-butanol, and esters of SCAFs, was noted following the treatment with EN.

CD, Crohn’s disease; EN, enteral nutrition; PF, polymeric formula; SCAFs, short-chain fatty acids.

of mucosal barrier, resulting in a continuous cycle of diet- in maintaining surgically and medically induced remissions
induced damage of the mucosal barrier, bacterial penetra- in CD patients.61,62
tion, and triggered mucosal immune responses.52 Therefore, However, EEN is believed to be less effective in adult
the underlying mechanism by which EEN therapy induces CD patients than in children with CD.63,64 This is partially
therapeutic benefits for the patients might at least in part be attributed to poor compliance in adults.65 Thus, current
associated to an interruption of this vicious cycle of diet- clinical practice does not support the use of EN therapy
facilitated bacterial penetration. as an induction treatment for adult CD patients, except
in those willing to undergo this therapy.66 An additional
EEN as a Treatment Option for CD drawback for EN is the long duration of therapy that can
be extended for several weeks as an induction of remission
Induction and Maintenance of Remission and even longer if used as a maintenance therapy.67,68
Several clinical trials involving pediatric CD patients treated
with EEN or steroids have shown that EEN is as efficient Exclusive or Partial Nutrition
as corticosteroids in inducing remission.35,56,57 However, The enteral diet can be administered as EEN or in con-
the observed supported growth and enhanced development junction with a normal diet as partial enteral nutrition
following EN treatment with no reported adverse effects (PEN).69 However, PEN in combination with a free diet
as those with corticosteroid use indicate superiority of appears to be less effective in inducing remissions than as
EEN over conventional medications as a first-line therapy when given exclusively.70 Sigall-Boneh et al71 investigated
for treating children with active CD.58 In a meta-analysis the effect of PEN together with a structured CD exclusion
involving 7 randomized controlled trials of pediatric CD diet and showed favorable outcomes. Nevertheless, PEN has
patients, Dziechciarz et al59 also reported that the efficacy not been established as an induction treatment60 ; however,
of EN was incomparable with that of steroids in inducing it may have a role as a maintenance therapy.72
remissions. Consistently, as per the 2014 consensus guide-
lines of the European Crohn’s and Colitis Organization
and the European Society of Pediatric Gastroenterology,
Disease Location and Treatment Duration
Hepatology and Nutrition for the medical management Disease location is perceived as a potential factor in
of pediatric CD, an enteral diet given as EEN is the determining the efficacy of EEN for inducing remissions in
preferred induction therapy for children with active CD.60 CD patients.73 EEN treatment has been shown to induce
Furthermore, there is an emerging role for enteral therapy remissions at higher rates in patients with small bowel
Alhagamhmad 489

involvement than in those with isolated colonic stoma creation, postoperative complications, and urgent
involvement.74 In contrast, in a different trial, the need of surgical intervention operation were significantly
disease location did not influence efficacy.57 Regarding lower in patients who received the combination treatment
the treatment duration, as an induction treatment, enteral than in those in the other groups. The authors also noted
therapy is usually administered during a period of up to that the combination treatment group exhibited in addition
8 weeks in pediatric patients with active CD.60 However, an extended preoperative drug-free interval.81 Furthermore,
an optimal treatment duration for enteral therapy, if favorable outcomes were also observed in a recent meta-
used as maintenance therapy, remains unknown and is analysis that included 4 studies that investigated the efficacy
influenced by patient compliance.75 EN was administered of infliximab as a monotherapy for CD patients or in
as a nocturnal supplement for 1 year after the induction combination with an enteral diet.82 Concomitant therapy
of remissions using EEN.76 In contrast, the follow-up was more effective for both, inducing and maintaining
duration was longer (mean ࣘ 2.6 years) in patients who had clinical remission than biological therapy alone.82 However,
undergone surgical interventions for inducing remission.77 it should be noted that in these few trials, EEN was not
used as a monotherapy; therefore, whether enteral diet
Discussing Potential Venues for Enhancing works better as part of a concomitant therapy or when
the Efficacy administered solely warrants further investigations.

Improving Compliance
Magnifying Immunomodulatory Effect
Despite the current limitations that discourage wide utiliza-
Enhancing the anti-inflammatory activity of EN by manip-
tion of enteral diet, improving the palatability might be a
ulating the concentrations of its active components is an
realistic option to improve compliance. Guo et al78 investi-
additional, promising option for enhancing the efficacy.83
gated the efficacy of EEN using more palatable formulas
Recent studies that used colonic epithelial cells cultured in
in adult patients with active CD and assessed its effects.
the laboratory as an in vitro model of IBD have shown that
Remission rate was 85% in the treated patients following a
the glutamine and arginine components of a polymeric diet,
4-week PF regimen.78 Furthermore, there was a significant
at concentrations used in the commercial formula (PF), are
improvement in the quality-of-life score, social function,
capable of suppressing the TNF-α-induced IL-8 production
and emotional status after the treatment. In addition,
from the stimulated cultured cells.31,84 Furthermore, these
around 61% of the recruited participants expressed their
constituents exhibited a dose-dependent anti-inflammatory
willingness to undergo treatment again to induce remission
effect mediated through the synergistic interactions against
if their disease relapsed.78 Thus, these initial observations
the I-κB kinase activity of the NF-κB pathway, indi-
indicate that enhancing compliance through the use of
cating the feasibility of using higher concentrations of
more palatable formulas might be a viable option for better
these active nutrients.31,84 Interestingly, manipulating the
utilization of the therapy; however, further clinical studies
glutamine and arginine components of the standard PF
are warranted to validate these preliminary observations.
enhanced the anti-inflammatory properties.31,84 Certainly,
PF enriched with glutamine and arginine amino acids (at
Combined Therapy final concentrations of 50 mM and 20 mM, respectively,
Commonly, the enteral therapy regimen for treating CD in the culture media) was superior to the standard PF in
patients involves EEN6 or, as recently proposed, PEN is terms of amelioration of TNF-α-induced IL-8 response
given with a CD exclusion diet.71 However, there is a trend in the stimulated intestinal epithelial cells.31 Interestingly,
of using enteral diet as a concomitant therapy given with the addition of curcumin, an additional immunoactive
the other immunosuppressive medications used in IBD nutrient, to glutamine-enriched and arginine-enriched PF
management,79 which might provide an additional potential further enhanced the anti-inflammatory activity in these
setting for better utilization of EN therapy in the man- experiments.83 Moreover, these initial observations were
agement of CD.80 Previous research enrolled 498 patients investigated further in an in vivo mouse model of coli-
with CD patients and investigated the effects of combined tis developed by exposing C57BL/6 mice to dextran sul-
EEN with immunosuppressive drugs.81 The recruited pa- fate sodium to induce intestinal inflammation.85 Amino
tients were randomly grouped into 1 of the following 3 acid–enriched PF substantially prevented dextran sulfate
groups: those who were undergoing preoperative immuno- sodium–induced weight loss, reduced myeloperoxidase ac-
suppressive treatments alone, those undergoing preoper- tivity, suppressed the expression of proinflammatory cy-
ative immunosuppressive treatments in combination with tokines, and partially ameliorated local colonic tissue injury
EEN treatment, and those who received none of these with no reported adverse effects. Although the treatment
treatments. This study reported better outcomes in patients with the enriched formula did not show measurable superi-
who underwent the combination treatment. The rates of ority over the treatment with the standard PF, the reversal of
490 Nutrition in Clinical Practice 33(4)

inflammatory changes was similar to that in standard PF but including NF-κB transcription factor. Furthermore, it is
with 15% lower total given volume.85 Thereafter, additional likely that several components of EN, including glutamine
experiments involving colonic biopsies collected from 10 and arginine amino acids account for that activity. Al-
children with active CD and matched controls investigated tering the composition of the enteral diet seems effective
further the efficacy of the enriched formula in attenuating in enhancing the anti-inflammatory property and thereby
intestinal inflammations.85 The new formula enriched with providing the opportunity for better utilization in clinical
amino acids showed superiority over the standard PF in practice.
suppressing inflammation in the cultured biopsies with no
adverse effects on tissue viability.85 These findings suggested
Acknowledgments
that altering the composition of the enteral diet might
provide a more effective and safe therapy with enhanced This review article is part of fulfillments for a clinical master’s
degree in pediatrics.
immune effects.
Collectively, despite the proven efficacy of the currently
Statement of Authorship
used enteral diet for the treatment of CD in clinical
practice, this therapy has limitations in ameliorating the M.H. Alhagamhmad contributed to the conception and de-
sign of the research; M.H. Alhagamhmad contributed to the
exaggerated immune responses and thereby in attenuating
acquisition, interpretation and analysis of the data; M.H.
intestinal inflammation. Nevertheless, the recent preclinical
Alhagamhmad drafted and critically revised the manuscript.
studies have shown that manipulating concentrations of the M.H. Alhagamhmad agrees to be fully accountable for en-
glutamine and arginine components of the polymeric diet suring the integrity and accuracy of the work, and read and
magnifies the immunomodulatory effects, which might have approved the final manuscript.
the potential to enhance the therapeutic utilities of EEN
in treating patients with CD. However, it should be noted References
that the translation into a real therapy in actual clinical 1. Hanauer SB. Inflammatory bowel disease: epidemiology, pathogenesis,
practice remains ambiguous. Indeed, a glutamine-fortified and therapeutic opportunities. Inflamm Bowel Dis. 2006;12:S3-S9.
polymeric diet did not show any measurable advantages over 2. Beasley G. P-203 disease activity, depression, and health related quality
of life in adolescents with inflammatory bowel disease attending a
the standard formula in a trial involving patients with CD.86
support group. Inflamm Bowel Dis. 2016;22:S70.
Furthermore, it has been claimed that enteral glutamine 3. Molodecky NA, Soon S, Rabi DM, et al. Increasing incidence and
and arginine supplementations did not provide significant prevalence of the inflammatory bowel diseases with time, based on
clinical benefits over the standard enteral formulas in tri- systematic review. Gastroenterology. 2012;142:46-54.
als involving intensive care unit and burn patients.87,88 In 4. Alhagamhmad MH, Leach ST, Lemberg DA, et al. Changing patterns
in the epidemiology of Crohn disease. J Gastroenterol Hepatol Res.
contrast, in several other trials, enteral diets enriched with
2015;4:1805-1809.
glutamine and/or arginine showed several positive clinical 5. Limketkai BN, Parian AM, Shah ND, et al. Short bowel syndrome and
outcomes in critically ill and surgical patients (acceler- intestinal failure in Crohn’s disease. Inflamm Bowel Dis. 2016;22:1209-
ated wound healings, reductions in nosocomial infection- 1218.
associated complications, and hospital mortality rate).89-91 6. Alhagamhmad MH, Day AS, Lemberg DA, et al. An update of the
role of nutritional therapy in the management of Crohn’s disease. J
In addition, considering the recent findings of enhanced
Gastroenterol. 2012;47:872-882.
activity of the polymeric diet following optimization of 7. Day A, Whitten K, Sidler M, et al. Systematic review: nutritional
the amino acids component, enriched enteral therapy ap- therapy in paediatric Crohn’s disease. Aliment Pharmacol Therap.
pears to possess superior therapeutic utility in treating 2008;27:293-307.
IBD patients over the standard formulations. This potential 8. Ezri J, Marques-Vidal P, Nydegger A. Impact of disease and treatments
on growth and puberty of pediatric patients with inflammatory bowel
superiority of enriched diets, however, might not be evident
disease. Digestion. 2012;85:308-319.
if the key ingredients are not supplemented in the optimal 9. Day AS, Whitten KE, Lemberg DA, et al. Exclusive enteral feeding
active concentrations. Thus, further studies are required to as primary therapy for Crohn’s disease in Australian children and
validate these preliminary observations. adolescents: a feasible and effective approach. J Gastroenterol Hepatol.
2006;21:1609-1614.
10. Cashman KD, Shanahan F. Is nutrition an aetiological factor for
Summary inflammatory bowel disease. Eur J Gastroenterol Hepatol. 2003;15:607-
613.
Enteral diet therapy plays an important role in the man- 11. Hume G, Radford-Smith GL. The pathogenesis of Crohn s disease in
agement of CD and can offer several therapeutic bene- the 21st century. Pathology. 2002;34:561-567.
fits. Attenuating exaggerated immune responses, enhanc- 12. Rogler G, Andus T. Cytokines in inflammatory bowel disease. World J
Surg. 1998;22:382-389.
ing mucosal healing and counteracting dysbiosis appear
13. Schottelius A, Baldwin A Jr. A role for transcription factor NF-kB in
to be the underlying mechanisms of actions of EN. The intestinal inflammation. Inter J Colorec Dis. 1999;14:18-28.
immunomodulatory effects of EN are attributed at least in 14. Fiocchi C. Inflammatory bowel disease: etiology and pathogenesis.
part to blocking the major intracellular signaling pathways, Gastroenterology. 1998;115:182-205.
Alhagamhmad 491

15. Waetzig GH, Seegert D, Rosenstiel P, et al. p38 mitogen-activated pro- 36. Grover Z, Burgess C, Muir R, et al. Early mucosal healing with
tein kinase is activated and linked to TNF- signaling in inflammatory Exclusive Enteral Nutrition is associated with improved outcomes in
bowel disease. J Immunol. 2002;168:5342-5351. newly diagnosed children with luminal Crohn’s disease. J Crohn’s and
16. Pagliari D, Piccirillo CA, Larbi A, et al. The interactions between Colitis. 2016;10:1159-1164.
innate immunity and microbiota in gastrointestinal diseases. J Immunol 37. Borrelli O, Cordischi L, Cirulli M, et al. Polymeric diet alone versus
Res. 2015;2015:1-3. corticosteroids in the treatment of active pediatric Crohn’s disease:
17. Alhagamhmad MH, Day AS, Lemberg DA, et al. An overview of the a randomized controlled open-label trial. Clin Gastroenterol Hepatol.
bacterial contribution to Crohn disease pathogenesis. J Med Microbiol. 2006;4:744-753.
2016;65:1049-1059. 38. Berni Canani R, Terrin G, Borrelli O, et al. Short-and long-term ther-
18. Goll R, van Beelen Granlund A. Intestinal barrier homeostasis in apeutic efficacy of nutritional therapy and corticosteroids in paediatric
inflammatory bowel disease. Scand J Gastroenterol. 2015;50:3-12. Crohn’s disease. Dig Liver Dis. 2006;38:381-387.
19. Pastorelli L, De Salvo C, Mercado JR, et al. Central role of the gut 39. Wang F, Graham WV, Wang Y, et al. Interferon-γ and tumor necrosis
epithelial barrier in the pathogenesis of chronic intestinal inflamma- factor-α synergize to induce intestinal epithelial barrier dysfunction
tion: lessons learned from animal models and human genetics. Front by up-regulating myosin light chain kinase expression. Am J Pathol.
Immunol. 2013;4:1-12. 2005;166:409-419.
20. Tan X, Mao J, Tang H, et al. Mechanisms underlying clinical efficacy 40. Al-Sadi R, Boivin M, Ma T. Mechanism of cytokine modulation of
of enteral nutrition in inflammatory bowel disease. Intern J Clini Expe epithelial tight junction barrier. Front Biosci. 2009;14:2765-2778.
Med. 2017;10: 2026-2035. 41. Ma TY, Boivin MA, Ye D, et al. Mechanism of TNF-α modulation of
21. Day AS, Lopez RN. Exclusive enteral nutrition in children with Crohn’s Caco-2 intestinal epithelial tight junction barrier: role of myosin light-
disease. World J Gastroenterol.2015;21:6809-6816. chain kinase protein expression. Am J Physiol Gastro Liver Physiol.
22. Voitk AJ, Echave V, Feller JH, et al. Experience with elemental diet in 2005;288:G422-G430.
the treatment of inflammatory bowel disease: is this primary therapy. 42. Nahidi L, Day AS, Lemberg DA, et al. Differential effects of nutritional
Arch Surg. 1973;107:329-333. and non-nutritional therapies on intestinal barrier function in an in
23. Heuschkel R. Enteral nutrition in Crohn disease: more than just vitro model. J Gastroenterol. 2012;47:107-117.
calories. J Paediatr Gastroenterol Nutr 2004; 38: 239-241. 43. Nahidi L, Leach ST, Mitchell HM, et al. Inflammatory bowel disease
24. Yamamoto T, Nakahigashi M, Saniabadi AR, et al. Impacts of long- therapies and gut function in a colitis mouse model. BioMed Res Intern.
term enteral nutrition on clinical and endoscopic disease activities and 2013;2013:1-15.
mucosal cytokines during remission in patients with Crohn’s disease: a 44. Ye D, Ma I, Ma TY. Molecular mechanism of tumor necrosis factor-α
prospective study. Inflamma Bowel Dis. 2007;13:1493-1501. modulation of intestinal epithelial tight junction barrier. Am J Physiol-
25. Yamamoto T, Nakahigashi M, Umegae S, et al. Impact of elemental Gastrointest Liver Physiol. 2006;290:G496-G504.
diet on mucosal inflammation in patients with active Crohn’s disease: 45. Lionetti P, Callegari M, Cavicchi M, et al. P0127 PP Enteral nutrition-
cytokine production and endoscopic and histological findings. Inflamm induced remission is associated with profound modificatioin of in-
Bowel Dis. 2005;11:580-588. testinal microflora in Crohn’s disease. J Pediatr Gastroenterol Nutr.
26. Beattie R, Schiffrin E, Donnet-Hughes A, et al. Polgmeric nutrition 2004;39:S106.
as the primary therapy in children with small bowel Crohn’s disease. 46. Lionetti P, Callegari ML, Ferrari S, et al. Enteral nutrition and
Aliment Pharmacol Therap. 1994;8:609-615. microflora in pediatric Crohn’s disease. JPEN J Parenter Enteral Nutr.
27. Fell J, Paintin M, Arnaud-Battandier F, et al. Mucosal healing and a 2005;29:S173-S178.
fall in mucosal pro-inflammatory cytokine mRNA induced by a specific 47. Leach S, Mitchell H, Eng W, et al. Sustained modulation of intestinal
oral polymeric diet in paediatric Crohn’s disease. Aliment Pharmacol bacteria by exclusive enteral nutrition used to treat children with
Therap. 2000;14:281-290. Crohn’s disease. Aliment Pharmacol Therap. 2008;28:724-733.
28. Bannerjee K, Camacho-Hübner C, Babinska K, et al. Anti- 48. Kaakoush NO, Day AS, Leach ST, et al. Effect of exclusive enteral
inflammatory and growth-stimulating effects precede nutritional nutrition on the microbiota of children with newly diagnosed Crohn’s
restitution during enteral feeding in Crohn disease. J Pediatr disease. Clin Trans Gastroenterol. 2015;6:1-11.
Gastroenterol Nutr. 2004;38:270-275. 49. Nahidi L, Day AS, Lemberg DA, et al. Paediatric inflammatory
29. de Jong NS, Leach ST, Day AS. Polymeric formula has direct anti- bowel disease: a mechanistic approach to investigate exclusive enteral
inflammatory effects on enterocytes in an in vitromodel of intestinal nutrition treatment. Scientifica. 2014;2014:1-9.
inflammation. Dig Dis Sci. 2007;52:2029-2036. 50. Walton C, Montoya MPB, Fowler DP, et al. Enteral feeding reduces
30. Nahidi L, Corley SM, Wilkins MR, et al. The major pathway by which metabolic activity of the intestinal microbiome in Crohn’s disease: an
polymeric formula reduces inflammation in intestinal epithelial cells: a observational study. Eur J Clin Nutr. 2016;70:1052-1056.
microarray-based analysis. Genes Nutr. 2015;10:1-9. 51. Greenberg G, Fleming C, Jeejeebhoy K, et al. Controlled trial of bowel
31. Alhagamhmad MH, Day AS, Lemberg DA, et al. Exploring and rest and nutritional support in the management of Crohn’s disease. Gut.
enhancing the anti-inflammatory properties of polymeric formula. 1988;29:1309-1315.
JPEN J Parenter Enteral Nutr. 2016;41:436-445. 52. Levine A, Wine E. Effects of enteral nutrition on Crohn’s disease:
32. Meister D, Bode J, Shand A, et al. Anti-inflammatory effects of enteral clues to the impact of diet on disease pathogenesis. Inflamm Bowel Dis.
diet components on Crohn’s disease-affected tissues in vitro. Dig Liver 2013;19:1322-1329.
Dis. 2002;34:430-438. 53. Shen W, Gaskins HR, McIntosh MK. Influence of dietary fat on
33. McGuckin MA, Eri R, Simms LA, et al. Intestinal barrier dysfunction intestinal microbes, inflammation, barrier function and metabolic
in inflammatory bowel diseases. Inflamma Bowel Dis. 2009;15:100-113. outcomes. J Nutr Biochem. 2014;25:270-280.
34. Shah S, Colombel JF, Sands B, et al. Systematic review with meta- 54. Lerner A, Matthias T. Changes in intestinal tight junction permeability
analysis: mucosal healing is associated with improved long-term out- associated with industrial food additives explain the rising incidence of
comes in Crohn’s disease. Aliment Pharmacol Therap. 2016;43:317-333. autoimmune disease. Autoimmun Rev. 2015;14:479-489.
35. Grover Z, Muir R, Lewindon P. Exclusive enteral nutrition induces 55. Barnich N, Denizot J, Darfeuille-Michaud A. E. coli-mediated gut in-
early clinical, mucosal and transmural remission in paediatric Crohn’s flammation in genetically predisposed Crohn’s disease patients. Pathol
disease. J Gastroenterol. 2014;49:638-645. Biol. 2013;61:e65-e69.
492 Nutrition in Clinical Practice 33(4)

56. Critch J, Day AS, Otley A, et al. Use of enteral nutrition for the 75. Yamamoto T, Nakahigashi M, Umegae S, et al. Enteral nutrition for
control of intestinal inflammation in pediatric Crohn disease. J Pediatr the maintenance of remission in Crohn’s disease: a systematic review.
Gastroenterolo Nutr. 2012;54:298-305. Eur J Gastroenterol Hepatol. 2010;22:1-8.
57. Buchanan E, Gaunt W, Cardigan T, et al. The use of exclusive 76. Duncan H, Buchanan E, Cardigan T, et al. A retrospective study
enteral nutrition for induction of remission in children with Crohn’s showing maintenance treatment options for paediatric CD in the first
disease demonstrates that disease phenotype does not influence clinical year following diagnosis after induction of remission with EEN: sup-
remission. Aliment Pharmacol Therap. 2009;30:501-507. plemental enteral nutrition is better than nothing. BMC Gastroenterol.
58. Heuschkel RB, Menache CC, Megerian JT, et al. Enteral nutrition and 2014;14:1-7.
corticosteroids in the treatment of acute Crohn’s disease in children. J 77. Esaki M, Matsumoto T, Hizawa K, et al. Preventive effect of nutri-
Pediatr Gastroenterol Nutr. 2000;31:8-15. tional therapy against postoperative recurrence of Crohn disease, with
59. Dziechciarz P, Horvath A, Shamir R, et al. Meta-analysis: enteral reference to findings determined by intra-operative enteroscopy. Scan J
nutrition in active Crohn’s disease in children. Aliment Pharmacol Gastroenterol. 2005;40:1431-1437.
Therap. 2007;26:795-806. 78. Guo Z, Wu R, Zhu W, et al. Effect of exclusive enteral nutrition on
60. Ruemmele F, Veres G, Kolho K-L, et al. Consensus guidelines of health-related quality of life for adults with active Crohn’s disease. Nutr
ECCO/ESPGHAN on the medical management of pediatric Crohn’s Clin Pract. 2013;28:499-505.
disease. J Crohn’s Colitis. 2014;8:1179-1207. 79. Wu X-L, Chen R-P, Tao L-P, et al. Infliximab combined with enteral
61. El-Matary W, Otley A, Critch J, et al. Enteral feeding therapy for nutrition for managing Crohn’s disease complicated with intestinal
maintaining remission in Crohn’s disease: a systematic review. JPEN fistulas. Gastroenterol Res Pract. 2016;2016:1-6.
J Parenter Enteral Nutr. 2015;41: 550-561. 80. Triantafillidis JK, Vagianos C, Papalois AE. The role of enteral
62. Yamamoto T, Nakahigashi M, Umegae S, et al. Impact of long-term nutrition in patients with inflammatory bowel disease: current aspects.
enteral nutrition on clinical and endoscopic recurrence after resection BioMed Res Intern. 2015;2015:1-12.
for Crohn’s disease: a prospective, non-randomized, parallel, controlled 81. Li Y, Zuo L, Zhu W, et al. Role of exclusive enteral nutrition in the
study. Aliment Pharmacol Therap. 2007;25:67-72. preoperative optimization of patients with Crohn’s disease following
63. Hartman C, Eliakim R, Shamir R. Nutritional status and nutri- immunosuppressive therapy. Medicine. 2015;94:1-9.
tional therapy in inflammatory bowel diseases. World J Gastroenterol. 82. Nguyen DL, Palmer LB, Nguyen ET, et al. Specialized enteral nutrition
2009;15:2570-2578. therapy in Crohn’s disease patients on maintenance infliximab therapy:
64. Zachos M, Tondeur M, Griffiths A. Enteral nutritional therapy for a meta-analysis. Therap Advan Gastroenterol. 2015;8:168-175.
induction of remission in Crohn’s disease. Cochrane Database Syst Rev. 83. Alhagamhmad M, Leach S, Day AS, et al. Mo1232 development of
2007;1:1-50. an enhanced nutritional therapy for the treatment of Crohn’s disease.
65. Kansal S, Wagner J, Kirkwood C, et al. Enteral nutrition in Crohn’s Gastroenterol. 2015;148:S-644.
disease: an underused therapy. Gastroenterol Res Pract. 2013;2013:1- 84. Alhagamhmad M, Leach S, Day AS, et al. Mo1231 attenuation
11. of intestinal inflammation by co-supplementation of glutamine and
66. Wall CL, Day AS, Gearry RB. Use of exclusive enteral nutrition arginine. Gastroenterol. 2015;148:S-644.
in adults with Crohn’s disease: a review. World J Gastroenterol. 85. Alhagamhmad MH, Lemberg DA, Day AS, et al. Advancing nutri-
2013;19:7652-7660. tional therapy: a novel polymeric formulation attenuates intestinal in-
67. Whitten KE, Rogers P, OoI CY, Day AS. International survey of flammation in a murine colitis model and suppresses pro-inflammatory
enteral nutrition protocols used in children with Crohn’s disease. J Dig cytokine production in ex-vivo cultured inflamed colonic biopsies. Clin
Dis. 2012;13:107-112. Nutr. 2017;36:497-505.
68. Gråfors JM, Casswall TH. Exclusive enteral nutrition in the treatment 86. Akobeng AK, Miller V, Stanton J, et al. Double-blind randomized
of children with Crohn’s disease in Sweden: a questionnaire survey. controlled trial of glutamine-enriched polymeric diet in the treatment
Acta Paediatr. 2011;100:1018-1022. of active Crohn’s disease. J Pediatr Gastroenterol Nutr. 2000;30:78-84.
69. Kang Y, Kim S, Kim SY, et al. Effect of short-term partial enteral 87. Peterik A, Milbrandt EB, Darby JM. Immunonutrition in critical
nutrition on the treatment of younger patients with severe Crohn’s illness: still fishing for the truth. Crit Care. 2009;13:1-3.
disease. Gut Liver. 2015;9:87-93. 88. Van Zanten AR, Dhaliwal R, Garrel D, et al. Enteral glutamine
70. Johnson T, Macdonald S, Hill SM, et al. Treatment of active Crohn’s supplementation in critically ill patients: a systematic review and meta-
disease in children using partial enteral nutrition with liquid formula: a analysis. Crit Care. 2015;19:1-16.
randomised controlled trial. Gut. 2006;55:356-361. 89. Garrel D, Patenaude J, Nedelec B, et al. Decreased mortality and
71. Sigall-Boneh R, Pfeffer-Gik T, Segal I, et al. Partial enteral nutrition infectious morbidity in adult burn patients given enteral glutamine
with a Crohn’s disease exclusion diet is effective for induction of supplements: a prospective, controlled, randomized clinical trial. Crit
remission in children and young adults with Crohn’s disease. Inflamm Care Med. 2003;31:2444-2449.
Bowel Dis. 2014;20:1353-1360. 90. Lorenz KJ, Schallert R, Daniel V. Immunonutrition–the influence of
72. Schulman JM, Pritzker L, Shaoul R. Maintenance of remission with early postoperative glutamine supplementation in enteral/parenteral
partial enteral nutrition therapy in pediatric Crohn’s disease: a retro- nutrition on immune response, wound healing and length of hospital
spective study. Can J Gastroenterol Hepatol. 2017;2017:1-7. stay in multiple trauma patients and patients after extensive surgery.
73. de Bie C, Kindermann A, Escher J. Use of exclusive enteral nutrition GMS Interdiscip Plast Reconstr Surg DGPW. 2015;4:1-10.
in paediatric Crohn’s disease in The Netherlands. J Crohn’s Colitis. 91. Caparrós T, Lopez J, Grau T. Early enteral nutrition in critically ill
2013;7:263-270. patients with a high-protein diet enriched with arginine, fiber, and
74. Afzal NA, Davies S, Paintin M, et al. Colonic Crohn’s disease in antioxidants compared with a standard high-protein diet. The effect
children does not respond well to treatment with enteral nutrition if on nosocomial infections and outcome. JPEN J Parenter Enteral Nutr.
the ileum is not involved. Dig Dis Sci. 2005;50:1471-1475. 2001;25:299-309.
Clinical Observations

Nutrition in Clinical Practice


Volume 33 Number 4
Case Report of Wernicke’s Encephalopathy After August 2018 510–514

C 2017 American Society for

Sleeve Gastrectomy Parenteral and Enteral Nutrition


DOI: 10.1177/0884533617722758
wileyonlinelibrary.com

Leslie A. Hamilton, PharmD, BCPS, BCCCP1 ; Sarah H. Darby, BS1 ;


Allan J. Hamilton, MD2 ; Matthew H. Wilkerson, MD3 ; and Kabel A. Morgan, MD3

Abstract
Background: We report a case of a patient who was 3 months post–sleeve gastrectomy and presented with acute stroke symptoms
ultimately due to Wernicke’s encephalopathy (WE) after bariatric surgery. A 20-year-old white female presented to an outside
hospital 3 months after sleeve gastrectomy complaining of nausea and vomiting. She initially underwent a cholecystectomy and later
became less responsive and required intubation. Magnetic resonance imaging changes, presumed to be an acute stroke, prompted
her transfer to our facility. Intravenous (IV) thiamin was administered, and the patient’s symptoms improved over the course of her
hospital stay. Results: Thiamin levels were markedly low, and the patient rapidly improved with the administration of IV thiamin.
The patient was discharged to inpatient rehabilitation. Conclusion: Bariatric surgery is a less common cause of WE but can lead
to acute WE due to malabsorption of thiamin. In patients undergoing bariatric surgery, clinicians should be vigilant about the
potential for WE to occur. In addition, based on history, WE should be considered in the differential diagnosis for symptoms of
acute ischemic stroke. (Nutr Clin Pract. 2018;33:510–514)

Keywords
bariatric surgery; sleeve gastrectomy; thiamin; thiamin deficiency; Wernicke encephalopathy

Historically associated with alcoholism, Wernicke’s en- for complications related to malabsorption postsurgery, be-
cephalopathy (WE) is an acute neurologic development due cause inadequate absorption of necessary vitamins and min-
to thiamin deficiency, with a classic triad of symptoms erals can lead to preventable and dangerous deficiencies.7
consisting of ophthalmologic dysfunction and nystagmus, Here we describe a case of a young woman presenting
ataxia, and mental status changes.1 The human body does with abnormal neurologic findings 3 months after sleeve
not produce any or store large amounts of thiamin, thus gastrectomy and later diagnosed with WE. A literature
requiring its consumption through the diet.2 Thiamin serves review of PubMed with the search terms “Wernicke’s en-
as a cofactor for numerous pathways of carbohydrate cephalopathy,” “gastric sleeve,” and “bariatric surgery” and
metabolism, which when altered leads to mitochondrial of Medline with the terms “bariatric,” “bariatric surgery,”
damage and potential necrosis, apoptosis, and oxidative “encephalopathy,” “surgery,” “Wernicke,” and “Wernicke’s
stress.3 encephalopathy” returned a substantial list of articles in
Because thiamin is obtained through the diet, deficiency
due to malnutrition is more likely in developing nations, From the 1 Department of Clinical Pharmacy, College of Pharmacy,
where proper nutrition may be more difficult to obtain. University of Tennessee Health Science Center, Knoxville, Tennessee,
In the Western world, where access to food may be less USA; 2 Department of Anesthesiology and Pain Management, UT
difficult to obtain, WE is typically connected to alcoholism. Southwestern, Dallas, Texas, USA; and 3 Department of
Anesthesiology, Graduate School of Medicine, University of
However, the number of nonalcoholic WE cases is likely to Tennessee Medical Center, Knoxville, Tennessee, USA.
increase due to malabsorption as an adverse result of the
Financial disclosure: None declared.
growing number of bariatric operations being performed.
With more than one-third of the U.S. adult population Conflicts of interest: None declared.
currently categorized as obese, it is not surprising to see This article originally appeared online on September 27, 2017.
that the number of bariatric operations has increased from Corresponding Author:
approximately 16,000 per year in the early 1990s to almost Leslie A. Hamilton, PharmD, BCPS, BCCCP, Department of Clinical
180,000 in 2012.4-6 Considering that these numbers repre- Pharmacy, College of Pharmacy, University of Tennessee Health
Science Center, 1924 Alcoa Highway, Box 117, Knoxville, TN 37909,
sent only 1% of surgery-eligible patients, they are sure to
USA.
continue to grow and require clinicians to be on the lookout Email: lhamilt4@uthsc.edu
Hamilton et al 511

which a relative few were case reports of WE occurring after


bariatric surgery. The unifying link among the case reports is
that no patient example presented in quite the same manner.

Case Report
The patient is a 20-year-old white woman who was approx-
imately 3 months post–sleeve gastrectomy and presented to
an outside hospital in late August 2015 with 6 weeks of nau-
sea and vomiting due to gallstone pancreatitis. During those
6 weeks, she presented to an outside hospital 3 times and
reported a subjective history of subsisting on rehydration
solution secondary to this nausea and vomiting, with an 83-
lb weight loss in the 3 months since the procedure. During
her last presentation, she presented with hypotension to
the outside hospital’s emergency department, and elevated
lipase and amylase were found on laboratory tests. She was
admitted to its intensive care unit for volume resuscita-
tion, which was completed with lactated Ringer solution.
Her pancreatic enzymes were elevated due to suspected
gallstones, and she had a laparoscopic cholecystectomy
performed on outside hospital day 8 (HD 8). She was noted Figure 1. Magnetic resonance imaging: T1 weighted
to be neurologically intact after the procedure. Social history periaqueductal.
was negative for alcohol and tobacco use.
On the morning of outside HD 9, staff found that she
was markedly less responsive, although she was able to move
all extremities to command. This neurologic change was
evaluated with magnetic resonance imaging (MRI), which
showed bilateral diffusion restriction within the posterior
thalamus (1.5 × 0.5 cm) that extended into the posterior
midbrain adjacent to the aqueduct of Sylvius. This was
accompanied by increased signal on T2 weighting within
these areas of diffusion restriction (Figures 1–4). These
findings were consistent with an acute infarct, with top-of-
the-basilar syndrome. She was transferred to our facility
for evaluation and treatment of a suspected acute stroke.
The patient was intubated, sedated, and paralyzed prior to
arrival at our facility, with outside records reporting that she
was moving all extremities prior to intubation. No mention
was made about ophthalmoplegia or nystagmus in the
records from the outside hospital. Computer tomography
angiography of the head at our facility showed patency
of the basilar artery, bilateral posterior communicating
arteries, and bilateral posterior cerebral arteries, without ev-
idence of basilar occlusion or clot at the artery of Percheron.
The patient’s body mass index was 48 kg/m2 on ad-
Figure 2. Magnetic resonance imaging: T1 weighted.
mission (height, 1.68 m; weight, 135 kg). Pertinent abnor-
mal laboratory results on admission include the following:
sodium, 129 mEq/dL (reference range, 136–147); chloride, international normalized ratio, 1.38 (0.8–1.2); thiamin,
93 mEq/dL (98–109); glucose, 137 mg/dL (70–99); phospho- 29 nMol/L (66.5–200); thyroid-stimulating hormone, 0.441
rus, 2.1 mg/dL (2.7–4.5); magnesium, 1.5 mg/dL (1.6–2.6); IU/mL (0.45–4.5); serum albumin level, 3 g/dL (3.5–5.2);
amylase, 269 U/L (5–125); lipase, 229 U/L (8–78); aspartate prealbumin, 13 mg/dL (16–38); total cholesterol, 79 mg/dL
aminotransferase, 87 U/L (5–34); alanine aminotransferase, (100–199); HDL, 11 mg/dL (>40); and LDL, 29 mg/dL
63 U/L (0–55); prothrombin time, 14.8 seconds (9.1–12); (<100). Of note, although her thiamin level was drawn on
512 Nutrition in Clinical Practice 33(4)

transthoracic echocardiogram was also performed on HD


1 and showed a normal left ventricular ejection fraction,
normal left atrial size, and no evidence of aortic valve
septal defects. Bilateral upper and lower extremity venous
duplex scans were negative for deep venous thrombosis.
Family members at bedside on HD 2 reported seizure-like
activity, at which time she was loaded with fosphenytoin
and levetiracetam, with levetiracetam continued for main-
tenance therapy. The electroencephalogram appreciated no
epileptiform abnormalities.
Inpatient medications included aspirin (81 mg per tube
daily), ceftriaxone (1 g, intravenous [IV], daily for HDs
8–15), enoxaparin (40 mg, subcutaneous, twice daily), es-
omeprazole (40 mg per tube daily), folic acid (1 mg per
tube daily), insulin (sliding scale, subcutaneous, daily),
levetiracetam (1000 mg, IV, twice daily), polyethylene glycol
3350 (17 g per tube daily), and thiamin (500 mg, intravenous,
× 1 dose on HD 3, followed by 100 mg, IV, daily). On HD
4, cholecalciferol (800 IU per tube daily) and multivitamin
(5 mL per tube daily) were initiated, with cyanocobalamin
(1000 mcg per tube daily) and pyridoxine (50 mg per tube
daily) started on HD 6. Modafinil (200 mg by mouth daily)
Figure 3. Magnetic resonance imaging: T2 fluid-attenuated was added on HD 10.
inversion recovery, periaqueductal.
Electrolyte deficiencies were aggressively replaced, and
enteral nutrition (EN) was started slowly with consideration
of the risks of the possibility of refeeding syndrome. Her ini-
tial EN goals were 1700 kcal and 121 g of protein (1.8 g/kg
based on ideal body weight). EN was a polymeric 1 kcal/mL
of fiber-containing formula titrated to goal rate over 3 days.
Her initial sodium of 129 mEq/L was corrected to 136 mE/L
over 3 days. After extubation on HD 5, a mechanical soft
diet was started on HD 7 with oral nutrition supplements.
Follow-up MRI done on HD 7 showed fluid-attenuated
inversion recovery (FLAIR) hyperintensities in bilateral
medial thalami on postcontrast enhancement consistent
with WE. Physical examination prior to extubation showed
a disconjugate gaze. Physical examination after extubation
showed limited horizontal eye movements with nystagmus,
dysarthric speech without aphasia, bilateral dysmetria on
finger-nose-finger, and bilateral upper extremity ataxia. The
patient did complain of vision and hearing difficulty, but
she reported that this was present prior to admission. She
did have mild improvement with nystagmus and dysarthria
throughout the rest of her hospital course. Levetiracetam
was stopped on HD 7, as it was thought that there was no
Figure 4. Magnetic resonance imaging: T2 fluid-attenuated
inversion recovery. evidence that she had experienced a seizure. She remained
in the hospital for 8 more days for rehabilitation with
the day of admission to our institution, it took 3 days for physical and occupational therapy and awaited inpatient
the results to return from an outside laboratory. A urine rehabilitation placement. She was discharged on HD 15.
toxicology screen was performed at the outside hospital
and was positive for acetaminophen, benzodiazepines, and
cannabinoids.
Discussion
A magnetic resonance venogram was performed on HD Bariatric surgery includes Roux-en-Y gastric bypass, sleeve
1 and did not show evidence of a dural venous thrombus. A gastrectomy, gastric band placement, and biliopancreatic
Hamilton et al 513

diversion, and each of these procedures carries with it with nystagmus, ataxia, confusion, and ophthalmoplegia 2
varying degrees of risk of later malabsorption.8 Roux-en- months after his sleeve gastrectomy. His MRI also showed
Y gastric bypass is considered a malabsorptive interven- hyperintense signals on T2 and FLAIR image in thalamus,
tion because it decreases contact time between food and periaqueductal area, and mammillary bodies.16
biliary and pancreatic enzymes, thus limiting digestion and The documented case reports discussed here vary greatly
absorption.9 Other procedures, such as the sleeve gastrec- in terms of how and when patients present after bariatric
tomy, are considered restrictive and may lead clinicians to surgery. Postoperative complications have been shown to
inappropriately consider vitamin deficiencies as less of a have an early or late onset and vary among symptoms such
threat. Without proper intake of thiamin, patients are at as vomiting, ataxia, eye movement disorders, dysarthria,
risk for developing WE, with reports showing that up to confusion, and even urinary incontinence. There is debate
30% of patients develop thiamin deficiency postsurgery.9 over the number of patients who actually present with the
Our patient initially presented to an outside hospital that classic triad of WE symptoms, with numbers ranging from
was unfamiliar with bariatric surgery, which led to a delay 16% to 40%, meaning that the trio of symptoms cannot
in treatment and subsequent worsening of symptoms over be relied on for diagnosis.1,11,17 A systematic review con-
the course of her stay. Another case that faced delays ducted by Kröll and colleagues found 13 well-documented
due to misdiagnosis occurred in a patient after Roux- cases of WE after sleeve gastrectomy. Of these cases, 8
en-Y bypass surgery10 ; however, our patient underwent patients reported experiencing vomiting postsurgery, and
sleeve gastrectomy. This illustrates that practitioners cannot the length of time until WE after surgery varied from 2
assume that any 1 type of bariatric surgery is safe from weeks to 7 months. Other symptoms experienced by patients
complications. Surgery not only induces structural changes included peripheral polyneuropathy, muscle weakness, and
to the gastrointestinal tract, which decreases absorption dysarthria, but these symptoms were not consistent among
capabilities, but also leaves patients with a reduced appetite, patients.11
further complicating the risk of vitamin deficiencies.9 Our Computed tomography scan and MRI findings varied
patient reported nausea and vomiting, which is not unex- as well, while some patients did not undergo scans at all.
pected given that vomiting is considered to be an important With a variety of vague symptoms and an incomplete
predisposing risk factor for postbariatric WE with poor patient history, the diagnosis of WE may be overlooked
thiamin intake.11 Our patient’s combination of vomiting until MRI is performed. Expected MRI findings for WE
and relying on a rehydration solution for nutrition made her include hyperintense signals on T2-weighted and FLAIR
extremely susceptible to WE-type symptoms. images in the periaqueductal regions and bilateral medial
While vomiting and poor oral intake are contributing thalami.18 Mammillary body and cerebellar vermis atrophy
factors to the development of WE, patients present with may also be seen but is unlikely in malnutrition patients
a variety of signs and symptoms once they report to a without alcoholism, because this is often the first thiamin-
facility for medical attention. One case reports a 24-year-old related deficiency causing neurologic dysfunction.17 Our
man presenting 7 months postsurgery with complaints of patient’s positive MRI findings, initially misinterpreted as
difficulty walking, diplopia, and dysarthria. The patient had an acute stroke, were what prompted her transfer from an
normal MRI results.12 Another case describes a 30-year-old outside hospital to our facility. MRI should be considered
woman whose nonadherence with vitamin supplementation for patients who report WE-like symptoms after bariatric
led to the development of diplopia and oscillopsia 2 months surgery; imaging is considered confirmatory due to 93%
after sleeve gastrectomy.13 Others report that a 38-year-old specificity.15 However, image findings as described here may
woman complained of weakness, nausea, and vomiting 1 appear in only 58% of patients and result in misleading MRI
week postsurgery. The following week she presented again results.15 In 1 literature review, MRI findings consistent with
with mental status changes, hypokinesis, diplopia, and an WE were found in just 6 of the 10 patients tested.11 A similar
inability to speak logically, though her MRI results were review states that 7 of 10 patients who underwent MRI
normal.14 In another report, a 27-year-old woman was showed positive findings congruent with WE.19 Thiamin
diagnosed with WE 3 weeks after her surgery with mild levels should be evaluated to solidify diagnosis, and a posi-
mental status changes. She denied vomiting, and computed tive patient response to thiamin therapy will also support a
tomography scan and MRI results were both normal. Her diagnosis. Dosing of thiamin for WE is variable, and while
symptoms resolved approximately 4 hours after admis- the recommendation is to acutely administer IV thiamin
sion and administration of thiamin.15 Two months after (given erratic absorption in malnourished patients), there is
gastric banding, a 40-year-old woman experienced vom- no standard dosing recommendation.20
iting, strabismus, urinary incontinence, and apathy. MRI Of note, our patient was only 20 years of age, which is
showed high-intensity lesions in the dorsomedial thalami, the youngest reported development of WE after bariatric
hypothalami, mammillary bodies, mesencephalic tectum, surgery. At this age, brain maturation may be incomplete,
and periaqueductal region.1 A 31-year-old man presented and further research is necessary to determine if she may
514 Nutrition in Clinical Practice 33(4)

be at increased risk of long-term complications. Although 5. National Institute of Diabetes and Digestive and Kidney Diseases.
the number of case reports in the literature is growing, it Longitudinal assessment of bariatric surgery. http://www.niddk.nih.
gov/health-information/health-topics/weight-control/Bariatric-
is unlikely that many practitioners have witnessed this type
Surgery. Published January 2010. Accessed December 17, 2015.
of case firsthand; thus, other practitioners play a crucial 6. American Society for Metabolic and Bariatric Surgery. New procedure
role in suggesting differential diagnoses as well. Prophylactic estimates for bariatric surgery: what the numbers reveal. http://connect.
thiamin administration may also be suggested to avoid de- asmbs.org/may-2014-bariatric-surgery-growth.html. Published May
ficiency complications if possible. Risk of thiamin toxicity 2014. Accessed December 17, 2015.
7. Nguyen NT, Vu S, Kim E, et al. Trends in utilization of bariatric
is minimal due to rapid renal elimination. Pharmacists can
surgery, 2009-2012. Surg Endosc. 2016;30:2723-2727.
also provide support with preoperative and postoperative 8. Athanasiou A, Angelou A, Diamantis T. Wernicke’s encephalopathy
medication regimens. after sleeve gastrectomy: where do we stand today? A reappraisal. Surg
Obes Relat Dis. 2014;10:563.
Conclusions 9. Tack J, Deloose E. Complications of bariatric surgery: dumping
syndrome, reflux, and vitamin deficiencies. Best Pract Res Clin Gas-
This report describes a case of WE in a young patient with a troenterol. 2014;4:741-749.
sleeve gastrectomy mimicking an acute stroke. Practitioners 10. Iannelli A, Addeo P, Novellas S, Gugenheim J. Wernicke’s en-
cephalopathy after laproscopic Roux-en-Y gastric bypass: a misdiag-
should be vigilant in these patients for signs of thiamin and
nosed complication. Obes Surg. 2010;20:1594-1596.
other nutrient malabsorption and potential WE. There may 11. Kröll D, Laimer M, Borbély YM, Laederach K, Candinas K, Nett
be a role of prophylactic thiamin in the prevention of WE PC. Wernicke encephalopathy: a future problem even after sleeve
among bariatric surgery patients. gastrectomy? A systematic literature review [published online October
17, 2015]. Obes Surg.
Statement of Authorship 12. Jeong HJ, Park JW, Kim YJ, et al. Wernicke’s encephalopathy after
sleeve gastrectomy for morbid obesity. Ann Rehabil Med. 2011;35:583-
L. A. Hamilton, M. H. Wilkerson, and K. A. Morgan equally 586.
contributed to the conception and design of the manuscript; 13. Jenkins PF. Wernicke encephalopathy. Am Orthopt J. 2015;65:104-108.
A. J. Hamilton and S. H. Darby contributed to the acquisition 14. Makarewicz W, Kaska L, Kobiela J, et al. Wernicke’s syndrome after
of the data. All contributed to the analysis and interpretation sleeve gastrectomy. Obes Surg. 2007;17:704-706.
of the data, drafted the manuscript, critically revised the 15. Saab R, El Khoury M, Farhat S. Wernicke’s encephalopathy three
manuscript, agree to be fully accountable for ensuring the weeks after sleeve gastrectomy. Surg Obes Relat Dis. 2014;10:992-994.
integrity and accuracy of the work, and read and approved the 16. Landais A, Saint-Georges G. Wernicke’s encephalopathy following
sleeve gastrectomy for morbid obesity. Rev Méd Interne. 2014;35:760-
final manuscript.
763.
17. Zuccoli G, Pipitone N. Neuroimaging findings in acute Wernicke’s en-
References cephalopathy: review of the literature. Am J Roentgenol. 2009;192:501-
1. Kühn AL, Hertel F, Boulanger T, Diederich NJ. Vitamin B1 in 508.
the treatment of Wernicke’s encephalopathy due to hyperemesis after 18. Rathakrishnan R, Smith EW. MRI in non-alcoholic Wernicke’s en-
gastroplasty. J Clin Neurosci. 2012;19:1303-1305. cephalopathy. J Clin Neurosci. 2008;15:478-480.
2. Voet D, Voet JG, Pratt CW. Fundamentals of Biochemistry: Life at the 19. Milone M, Di Minno MND, Lupoli R, et al. Wernicke encephalopathy
Molecular Level. 4th ed. Hoboken, NJ: John Wiley & Sons; 2012:495. in subjects undergoing restrictive weight loss surgery: a systematic
3. Martin PR, Singleton CK, Hiller-Sturmhöfel S. The role of thiamine review of literature data. Eur Eat Disord Rev. 2014;22:223-229.
deficiency in alcoholic brain disease. Alcohol Res Health. 2003;27:134. 20. Day E, Bentham PW, Callaghan R, Kuruvilla T, George S. Thi-
4. Ogden CL, Carroll MD, Kit BK, Flegal KM. Prevalence of child- amine for prevention and treatment of Wernicke-Korsakoff syn-
hood and adult obesity in the United States, 2011-2012. JAMA. drome in people who abuse alcohol. Cochrane Database Syst Rev.
2014;311:806-814. 2013;(7):CD004033.
Clinical Observations

Nutrition in Clinical Practice


Volume 33 Number 4
Copper Deficiency Myelopathy After Upper August 2018 515–519

C 2017 American Society for

Gastrointestinal Surgery Parenteral and Enteral Nutrition


DOI: 10.1177/0884533617713955
wileyonlinelibrary.com

Dominic King, MBChB; Keith Siau, MBChB; Latha Senthil, MBBS;


Katherine F. Kane, MD; and Sheldon C. Cooper, MD

Abstract
A well-functioning alimentary canal is required for adequate nutrient absorption. Disruption to the upper gastrointestinal tract
through surgery can lead to micronutrient malnourishment. Copper deficiency has been noted in up to 10% of those undergoing
Roux-en-Y gastric bypass surgery, but sequalae are not frequently reported. The resultant deficiency states can have profound and
long-term consequences if not realized early and managed appropriately. Here we present a case of copper deficiency myelopathy,
a condition indistinguishable from subacute combined degeneration of the spinal cord, following upper gastrointestinal bypass
surgery for gastric ulceration, further complicated by inadequate nutrition. (Nutr Clin Pract. 2018;33:515–519)

Keywords
bariatric surgery; copper deficiency; gastric bypass; micronutrients; myelopathy; spinal cord diseases; vitamins

A variety of micronutrient deficiencies are well known to gastrojejunostomy in August 2010. The surgery had been
cause disorders of the nervous system.1 The importance of complicated by recurrent ulceration at the gastrojejunal
a functioning digestive tract is therefore cruicial. Disruption anastamosis and gastric outlet obstruction, requiring
to the gut, whether through surgical approach or disease further surgical intervention. She underwent subtotal
state, inevitably interferes with micronutrient absorption gastrectomy with Roux-en-Y gastric bypass in May 2012
and can have devastating sequalae. The prevalence of cop- with subsequent jejunostomy feeding. This was further
per deficiency following Roux-en-Y gastric bypass surgery complicated by poor remnant emptying, and in June 2013, a
in 1 cross-sectional study was found to be 9.6%,2 and further laparotomy was performed where 55 cm of jejunum
in a series of obese women undergoing bariatric surgery was resected and a surgical jejunostomy was placed for
with Roux-en-Y gastric bypass, 15.4% were found to be which the patient became dependent for feeding. Until refer-
copper deficient.3 Asymptomatic copper deficiency is com- ral to our hospital and the current admission, there had been
mon in a disrupted upper gastrointestinal tract; though no PN. There were recurrent complications with enteral
rare, symptoms can also be seen.3,4 We present a case of feeding tubes (eg, blocked tube and feed leakage), leading to
symptomatic copper deficiency in a patient following upper malnutrition.
gastrointestinal surgery. This produced a myelopathy and Ulcer biopsies ruled out malignancy, and investiga-
clinical mimic of the subacute combined degeneration of tion for gastrin-producing tumors and inflammatory bowel
the spinal cord (SACDSC), characteristically associated disease was unremarkable. Investigations for rarer causes
with vitamin B12 deficiency.
From University Hospitals Birmingham, Birmingham, United
Case Kingdom.
Financial disclosure: None declared.
History of Surgical Events and Nutrition State
Conflicts of interest: Dr Sheldon Cooper has the following conflicts of
Leading Up to Admission interest to declare: Covidien, Advisory Board and educational
sponsorship for an international meeting; Eli Lilly, Advisory Board;
A 50-year-old woman was referred to our tertiary center for Baxter/Willow, speakers’ fee.
completion gastrectomy in January 2015, requiring preoper-
This article originally appeared online on June 28, 2017.
ative parenteral nutrition (PN) containing all essential trace
Corresponding Author:
elements and vitamins (provided in Additrace, Solivito, and
Sheldon C. Cooper, MD, Department of Gastroenterology, QEHB,
Vitlipid) due to severe malnutrition. UHB NHS Foundation Trust, Edgbaston, Birmingham, West
The patient had nonhealing gastric ulcerations that Midlands B15 2GW, UK.
had been treated with laprascopic distal gastrectomy and Email: sheldon.cooper@nhs.net
516 Nutrition in Clinical Practice 33(4)

Figure 1. Magnetic resonance image with T2-weighted enhancement. Subacute combined degeneration of the spinal cord is seen,
with high signal changes in the dorsal columns, represented by the arrows.

of gastric ulcerations were also negative, and serum and In the upper limbs, power was Medical Research Council
urinary nonsteroidal anti-inflammatory drugs were absent. (MRC) grade 4 of 5 (reduced power against resistance) in
High-grade gastric outlet obstruction continued to be finger and wrist extension and flexion and normal more
an issue as the patient continued to eat and drink, and proximally. In the lower limbs, power was MRC grade 4
repeat gastroscopy confirmed further ulceration at the anas- in extension and flexion of hip and knee joints bilaterally.
tomosis. The patient suffered from complex chronic pain More distally, power was reduced further with MRC grade
and found relief in venting her jejunostomy. She developed 3 (active movement against gravity, without resistance) in
hyponatremia, hypoalbuminemia, and ongoing weight loss dorsiflexion and plantarflexion. By February 2015, power
with malnutrition, leading to the admission in 2015 to our in the lower limbs had progressed to MRC grade 2 (active
tertiary center. movement but only if gravity eliminated) in hip and knee
flexion and extension and remained 3 to 4 at the ankle.
Plantars were up going, and reflexes were bilaterally brisk,
Assessment, Examination, and Imaging although ankle jerks were absent. Sensation was reduced to
On initial assessment, the patient was found to be cachectic, pinpoint and light touch in a stocking-and-glove distribu-
with her lowest weight being 38 kg with a body mass tion to the midthighs and forearms, respectively. The first
index (BMI) of 13.15. She was wheelchair bound, later symptoms of distal numbness had been reported in 2013,
becoming bed bound and dependent for activities of daily but progression occurred during the 2015 admission to hos-
living. A year history of progressive finger and toe paraes- pital. Vibration sense and proprioception were reduced in
thesia had been noted and investigated at the original the lower limbs, and continence was maintained. Neurology
referring hospital with nutritient testing of vitamin B12 advice suggested a diagnosis of SACDSC with a peripheral
and folate and by noncontrasted magnetic resonance image neuropathy, secondary to nutrition deficiency.
(MRI) of the brain and spine but with no pathology An MRI scan of the spine with T2 weighting found
found. characteristic findings of SACDSC with high signal changes
Examination revealed normal tone with bilateral upper in the dorsal columns, represented by a charcteristic “inverse
limb and lower limb weakness that was more severe distally. V sign” (Figure 1).
King et al 517

Table 1. Relevant Blood Laboratory Investigations. withdrawn after 1 year from commencement. Nutrition
status has since been maintained, both serologically (in-
Laboratory Investigation Values (Reference Range)
cluding serum copper levels comfortably within the normal
Hemoglobin, g/L 113 (115–165) range) and in terms of a stable BMI. Neurologic assessment
White cell count, ×109 /L 8.7 (4–11) found little in the way of neurophysiologic or functional
Platelets, ×109 /L 517 (150–450) improvement; the patient is currently chair bound, having
Mean corpuscular volume, fL 89.6 (80–99) previously been bed bound. Spinal MRI following admis-
Mean corpuscular hemoglobin, pg 30 (27–33) sion, at 1 month and at 1 year, showed no significant changes
Mean corpuscular hemoglobin 335 (315–365) to the high signal abnormalities seen in Figure 1. Her most
concentration, g/L
Ferritin, μg/L 42 (10–320)
recent copper levels were normal at 16.2 μmol/L, and she
Vitamin B12, ng/L 180 (>200) continued to receive enteral supplements in the form of
Homocysteine, μmol/L 9.2 (6.7–15.2) copper amino acid chelate, 1 tablet per day (the compound is
Copper, μmol/L 5.9 (11–25) 20 mg, which is equivalent of 2 mg of elemental copper). In
Selenium, μmol/L 0.46 (0.9–1.7) the longer term, Forceval has been sufficient to maintain her
Zinc, μmol/L 8.8 (11–24) copper levels with cessation of specific copper supplements,
but she remains under close nutrition supervision.

Discussion and Literature Review


Results of Laboratory Investigations Copper is absorbed from the duodenum and the stomach via
The patient was noted to be mildly cytopenic (Table 1). the Ctr1 apical membrane transporter. Following transport
Vitamin B12 levels at this time were found to be mildly to the liver hepatocytes, either it is utilized, or it binds
low at 180 ng/L (reference, >200 ng/L) but had been within to caeruloplasmin (not measured in this case as usually
the normal range and were thought to be insufficiently low measured in diagnostic workup for copper overload) for
enough to cause SACDSC, following review by our hema- systemic circulation to other tissues. Copper is a vital
tologists; furthermore, homocysteine levels were normal at micronutrient required for essential enzymatic processes in
9.2 μmol/L (6.7–15.2 μmol/L). the human body, including neurotransmitter metabolism,
Vitamins A, E, and B6 levels were normal; antitissue iron transport, oxidative phosphorylation, purine synthesis,
transglutaminase antibodies and vasculitic screen yielded and connective tissue synthesis.5,6 Deficiency of ATPase
negative results; and cerebrospinal fluid investigation re- transporters from the gastrointestinal cells to the portal
vealed normal values (white cell count, 1/cmm; glucose, circulation or the hepatocytes to bile, leads to the copper
3.2 mmol/L; protein, 0.14 g/L; polymerase chain reaction, deficiency and excess states of Menke’s disease (kinky hair
negative). Nerve conduction studies demonstrated sen- disease) and Wilson’s disease, respectively.7-9 Excess copper
sory/motor axonal neuropathy. Copper levels were sent and will bind to metallothioneins to prevent toxic levels and
revealed a low level, 5.9 μmol/L (reference, 11–25 μmol/L). has a higher affinity for binding than does zinc. Zinc is
Borderline-low zinc levels (8.8 μmol/L) and selenium de- known to upregulate these proteins in the enteric mucosa
ficiency (0.46, μmol/L; Table 1) were corrected with PN. and is the mechanism by which copper is lost, leading to
The PN copper component consisted of copper chloride, the deficiency states seen in zinc excess.10,11 Bile secretion
initially at 40 μmol per PN bag (24 hours), in the form achieves homeostasis, with much reabsorbed, but main
of 20 mL of Additrace, double the standard amounts in losses occur via feces and small amounts in the urine.6
24 hours of PN (10 mL or 1 vial), which was reduced to Copper deficiency as a cause of cytopenia has long been
maintenance of 1 vial (10 mL) of Additrace (in addition accepted,6,10 but it is only more recently that a myelo-
to Solivito and Vitlipid), providing 20 μmol in all PN pathic process has been described.12 Copper is normally
thereafter. Inpatient PN was converted to home PN 5 nights seen in high levels within the central nervous system. It
of the week some 4.5 months after commencement. is thought that the copper-deficient myelopathy, analogous
The predominant copper deficiency produced this in- to the SACDSC seen in vitamin B12 deficiency, is related
distinguishable syndrome of SACDSC. Copper-containing to copper’s role in the methylation cycle.13 Methionine
PN and restoration of general nutrition status with a BMI synthase (a part of the cycle) may be the common disrupted
of 21.94 have led to some resolution of symptoms. Three pathway of nerve protein sythesis in copper and vitamin
monthly hydroxocobalamin intramuscular injections (1 mg; B12 deficiency. SACDSC is an insidious progressive combi-
following loading doses of 1 mg on alternate days for 2 nation of dorsal column signs manifesting in ataxia, with
weeks) were also provided per British National Formulary combinations of upper and lower motor signs, classically
guidance. The patient is now soley fed via jejunostomy. PN absent ankle jerks, and up-going plantar response,14 as seen
was weaned down to just 2 nights per week, then finally in this case. Vitamin B12 and copper deficiency may also
518 Nutrition in Clinical Practice 33(4)

cause peripheral neuropathy, confirmed in this case, which 47% of these cases had had upper gastrointestinal surgery.6
can further complicate diagnosis. Kumar and colleagues’ review of Roux-en-Y gastric bypass
Although an underlying cause of copper deficiency surgery as treatement for obesity examined symptomatic
may be found—commonly, upper gastrointestinal surgery and asymptomatic cases and described up to 10% of pa-
(as contributed in this case), malabsorption, zinc excess tients as having copper deficiency postprocedure; however,
(sometimes as a result of denture fixative), copper chelation having symptoms was rare, with only 34 cases after bariatric
through penacillamine—sometimes no cause can be found. surgery found in the literature.4
Plantone et al described 2 cases of women with gait ataxia It is pertinent to mention that the neurologic conse-
(1 mild and 1 moderate) aged 50 and 53 years with MRI quences of copper deficiency can occur many years after
dorsal column findings similar to those seen in our case, surgery. Indeed, in 1 review, the time for neuorolgic onset
with low copper levels but with no cause for the copper following gastric surgery was 5–26 years and 10–46 years
deficiency found. Interestingly, oral suplementation at 8 for bariatric and nonbariatric patients, respectively.6 In 2
mg/d of copper led to significant clinical and radiologic case reports, men in their 70s with neurologic symptoms
improvement.15 Kumar et al described 3 cases of copper secondary to copper deficiency, akin to the patient in this
deficiency myelopathy; for the 1 man (72 years old, who case, had had gastric bypass surgery in their 20s and
had undergone partial gastrectomy at age 26), MRI findings 30s.11,16 Copper deficiency leading to neurologic sequaelae is
showed hyperintense signal change on T2 weighting in being recognized more and more in the literature. Although
the dorsal columns. He had a vitamin B12 deficiency, but surgery for gastric ulceration has become less and less
symptoms of ascending paraesthesia, increased limb weak- common, there is increased bariatric surgery, which can
nes, and ataxia progressed, despite replacement. Copper equally lead to malabsorption, and indeed many obese
deficiency was eventually discovered and treated, though patients may be copper deplete prior to surgery.17,18
clinical improvement was not commmented on. The other The UK recommended dietry intake of copper is 1.2
2 cases were of women in their late 40s where the cause mg/d, which should be easily achieved given the ubiquitous
of the copper deficiency was not clear. One patient had nature of copper in many foodstuffs.19 The literature has
MRI changes, the other did not, and both had ataxic gaits very different treatement suggestions, with Kumar and
and reduced vibration and proprioception sense. All 3 cases colleagues’ review of bariatric surgery finding suggested
had evidence of sensory/motor axonal neuropathy, not seen ranges after Roux-en-Y gastric bypass surgery from 0.45
in the Plantone cases but seen in our case. One woman to 3.0 mg/d (which includes values lower than the daily
received parentral and then oral copper supplementation recommendation).4 The suggestion by Jaiser and Winston is
and achieved slight clinical improvement; the other woman that 8 mg/d orally is suitable to try to prevent neurologic re-
had only oral supplementation (at 2 mg/d), did not achieve laspe/progression following deficiency-induced myelopathy;
normal copper levels, and her syptoms deteriorated.11 this case series found treatment ranges from 2 mg escalating
A review of 16 cases of Scottish patients with severe to 4–6 mg and 8 mg, and in some cases, 9 mg/d were used.
copper deficiency by Gabreyes et al showed that replace- The tolerable upper intake level is thought to be 10 mg/d.6
ment of copper and removal of exccess zinc improved Optimal duration, dose, and form of copper replacement
blood cytopaenias, though neurologic improvement was are not clear from the literature; in this case, long-term
seen much less commonly. Causes of copper deficiency in supplementation is likely to be required as the underlying
this retrospective study included zinc excess supplementa- cause remains. The British Obesity and Metabolic Surgery
tion in the form of dental fixatives, for proximal bowel Society advises that copper be measured annually following
resection, for Wilson’s disease, and from topical zinc use gastric bypass surgery as a standard, with more common
in patients with burns. Two patients, whose cases were not measurements if poor wound healing is seen. More common
related to excess zinc, had celiac disease or penacillamine as measurements were felt unecessary, as routine supplemen-
treatment for systemic sclerosis. Of the 16, 12 had neurologic tation at a minimum of 2 mg/d should be commenced
symptoms with gait disturbance, and 10 had paraesthesias. as recommended by American and British guidelines.4,20
Eight patients had spinal imaging, of which 6 had dorsal In severe deficiency, parentrally administered copper at
column signs, and 2 of the 7 patients who had nerve 2–4 mg/d for 6 days was advised.21
conduction studies had sensory/motor neuropathy. Only 3
of 12 patients had any improvement, and 4 progressed to
be wheelchair bound. All patients received 5 mg/d of oral
Conclusions
copper sulphate.10 This case demonstrates the need for clinicians to be aware of
Copper deficiency myelopathy has a female preponder- nutrition deficiencies and the importance of copper malab-
ance, as demonstrated in Jaiser and Winston’s review of sorption as a cause of neurologic impairment. The literature
55 case reports. The authors demonstrated an age range suggests that the neurologic deterioration secondary to cop-
peaking in the fifth and sixth decades, as seen in this case; per deficiency can only be stabilized or partially improved by
King et al 519

supplementaion; therefore, swift recognition and treatment 7. Gollan JL, Deller DJ. Studies on the nature and excretion of biliary
are paramount.6 This point is further emphasized as the copper in man. Clin Sci. 1973;44:9-15.
8. Gandhi R, Kakkar R, Rajan S, Bhangale R, Desai S. Menkes kinky
consequences of copper deficiency may not be seen until
hair syndrome: a rare neurodegenerative disease. Case Rep Radiol.
many years after the causative insult (eg, gastric bypass 2012;2012:684309.
surgery).10,22 9. Menkes JH, Alter M, Steigleder GK, Weakley DR, Sung JH. A sex-
We suggest that in the context of a malabsorbative state, linked recessive disorder with retardation of growth, peculiar hair, and
in particular through upper gastrointestinal surgery com- focal cerebral and cerebellar degeneration. Pediatrics. 1962;29:764-779.
10. Gabreyes AA, Abbasi HN, Forbes KP, McQuaker G, Duncan A,
pounded by poor nutrition intake, vigilance for a clinical
Morrison I. Hypocupremia associated cytopenia and myelopathy: a
picture of SACDSC is vital. If it is suspected, T2-weighted national retrospective review. Eur J Haematol. 2013;90:1-9.
MRI of the whole spine is the investigation of choice. 11. Kumar N, Crum B, Petersen RC, Vernino SA, Ahlskog JE. Copper
We also suggest that follow-up blood tests that include deficiency myelopathy. Arch Neurol. 2004;61:762-766.
copper levels are a prudent low-cost means of recognizing 12. Schleper B, Stuerenburg HJ. Copper deficiency-associated myelopathy
in a 46-year-old woman. J Neurol. 2001;248:705-706.
copper deficiency before it has any deleterious effect, and
13. Winston GP, Jaiser SR. Copper deficiency myelopathy and subacute
oral supplementation in concordance with recommended combined degeneration of the cord—why is the phenotype so similar?
guidelines can prevent deficiencies before they occur. Med Hypotheses. 2008;71:229-236.
14. Kumar P, Clark M. Kumar and Clark’s Clinical Medicine. New York,
Statement of Authorship NY: Elsevier; 2016.
15. Plantone D, Primiano G, Renna R, et al. Copper deficiency myelopa-
D. King, K. Siau, and S. C. Cooper equally contributed to
thy: a report of two cases. J Spinal Cord Med. 2015;38:559-562.
the conception and design of the case report; all authors 16. Chhetri SK, Mills RJ, Shaunak S, Emsley HC. Copper deficiency. BMJ.
contributed to the acquisition and analysis of the data; all 2014;348:g3691.
authors contributed to the interpretation of the data; and 17. de Luis DA, Pacheco D, Izaola O, Terroba MC, Cuellar L, Cabezas G.
D. King drafted the manuscript. All authors critically revised Micronutrient status in morbidly obese women before bariatric surgery.
the manuscript, read and approved the final manuscript, and Surg Obes Relat Dis. 2013;9:323-327.
agree to be accountable for ensuring integrity and accuracy of 18. Papamargaritis D, Aasheim ET, Sampson B, le Roux CW. Copper,
the work. selenium and zinc levels after bariatric surgery in patients recommended
to take multivitamin-mineral supplementation. J Trace Elem Med Biol.
2015;31:167-172.
References 19. Panel on Dietary Reference Values of the Committee on Medical
1. Lanska DJ. Chapter 30: historical aspects of the major neurological Aspects of Food Policy. Dietary reference values for food energy and
vitamin deficiency disorders: the water-soluble B vitamins. Handb Clin nutrients for the United Kingdom. Rep Health Soc Subj (Lond).
Neurol. 2010;95:445-476. 1991;41:1-210.
2. Gletsu-Miller N, Broderius M, Frediani JK, et al. Incidence and 20. O’Kane M, Pinkney J, Aasheim ET, Barth JH, Batterham RL,
prevalence of copper deficiency following Roux-en-Y gastric bypass Welbourn R. BOMSS Guidelines on Perioperative and Postoperative
surgery. Int J Obes (Lond). 2012;36:328-335. Biochemical Monitoring and Micronutrient Replacement for Patients
3. Ernst B, Thurnheer M, Schultes B. Copper deficiency after gastric Undergoing Bariatric Surgery. London, UK: British Obesity and
bypass surgery. Obesity (Silver Spring). 2009;17:1980-1981. Metabolic Surgery Society; 2014.
4. Kumar P, Hamza N, Madhok B, et al. Copper deficiency after 21. Mechanick JI, Youdim A, Jones DB, et al. Clinical practice guidelines
gastric bypass for morbid obesity: a systematic review. Obes Surg. for the perioperative nutritional, metabolic, and nonsurgical support of
2016;26:1335-1342. the bariatric surgery patient—2013 update: cosponsored by American
5. Rech M, To L, Tovbin A, Smoot T, Mlynarek M. Heavy metal in Association of Clinical Endocrinologists, The Obesity Society, and
the intensive care unit: a review of current literature on trace element American Society for Metabolic and Bariatric Surgery. Obesity (Silver
supplementation in critically ill patients. Nutr Clin Pract. 2014;29:78- Spring). 2013;21:S1-S27.
89. 22. Duddy J. Copper deficiency masquerading as subacute combined
6. Jaiser SR, Winston GP. Copper deficiency myelopathy. J Neurol. degeneration of the cord and myelodysplastic syndrome. Adv Clin
2010;257:869-881. Neurosci Rehabil. 2007;7:20-12.
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 4
Reduction of Parenteral Nutrition and Hydration Support and August 2018 520–527

C 2018 The Authors. Nutrition in

Safety With Long-Term Teduglutide Treatment in Patients Clinical Practice published by


Wiley Periodicals, Inc. on behalf
With Short Bowel Syndrome−Associated Intestinal Failure: of American Society for
Parenteral and Enteral Nutrition
STEPS-3 Study DOI: 10.1002/ncp.10092
wileyonlinelibrary.com

Douglas L. Seidner, MD, AGAF, FACG, CNSC1 ; Ken Fujioka, MD2 ;


Joseph I. Boullata, PharmD, RPh, BCNSP, FASPEN3 ; Kishore Iyer, MBBS, FRCS,
FACS4 ; Hak-Myung Lee, PhD5 ; and Thomas R. Ziegler, MD6

Abstract
Background: Patients with intestinal failure associated with short bowel syndrome (SBS–IF) require parenteral support (PS)
to maintain fluid balance or nutrition. Teduglutide (TED) reduced PS requirements in patients with SBS–IF in the random-
ized, placebo (PBO)-controlled STEPS study (NCT00798967) and its 2-year, open-label extension, STEPS-2 (NCT00930644).
Methods: STEPS-3 (NCT01560403), a 1-year, open-label extension study in patients with SBS–IF who completed STEPS-2, further
monitored the safety and efficacy of TED (0.05 mg/kg/day). Baseline was the start of TED treatment, in either STEPS or STEPS-2.
At the end of STEPS-3, patients treated with TED in both STEPS and STEPS-2 (TED–TED) received TED for ࣘ42 months, and
patients treated with TED only in STEPS-2 (no TED treatment [NT]/PBO–TED) received TED for ࣘ36 months. Results: Fourteen
patients enrolled (TED–TED, n = 5; NT/PBO–TED, n = 9) and 13 completed STEPS-3. At the last dosing visit, mean (SD) PS was
reduced from baseline by 9.8 (14.4 [50%]) and 3.9 (2.8 [48%]) L/week in TED–TED and NT/PBO–TED, respectively. Mean (SD)
PS infusions decreased by 3.0 (4.6) and 2.1 (2.2) days per week from baseline in TED–TED and NT/PBO–TED, respectively. Two
patients achieved PS independence; 2 additional patients who achieved independence in STEPS-2 maintained enteral autonomy
throughout STEPS-3. All patients reported ࣙ1 treatment-emergent adverse event (TEAE); 3 patients had TEAEs that were reported
as treatment related. No patient had a treatment-related treatment-emergent serious AE. Conclusions: Long-term TED treatment
yielded a safety profile consistent with previous studies, sustained efficacy, and a further decline in PS requirements. (Nutr Clin
Pract. 2018;33:520–527)

Keywords
glucagon-like peptide-2; intestinal failure; parenteral nutrition; short bowel syndrome; teduglutide

In adults, intestinal failure associated with short bowel Although PS provides vital fluid and nutrient support
syndrome (SBS−IF) is an uncommon but life-altering con- for patients with SBS−IF, this therapeutic approach is
dition that occurs as a consequence of surgical resection associated with potentially serious or life-threatening com-
or disease with or without partial or complete colonic plications (eg, risk for catheter-related infection and liver
resection, typically in patients with inflammatory bowel failure).3 Patients with SBS−IF also have impaired health-
disease, intestinal obstruction, ischemic events, fistula resec- related quality of life characterized by feelings of social
tion, or malignancy.1 As a result of reduction in residual isolation, a disruption of activities of daily living, and
small-bowel length and absorptive function, patients with gastrointestinal (GI) symptoms (eg, diarrhea, abdominal
SBS−IF experience generalized protein-energy malnutri- pain).4-6
tion, dehydration, and micronutrient depletion due to a loss Teduglutide (TED) is a synthetic analog of endoge-
in intestinal absorptive capacity and often require long-term nous glucagon-like peptide-2, which is synthesized in the
parenteral support (PS) in the form of parenteral nutrition ileum and colon of healthy individuals.7 Subcutaneous
and/or intravenous hydration, depending on the extent of TED administration promotes intestinal adaptation, en-
bowel resection and the presence of colon-in-continuity.2 hances intestinal absorptive capacity, and enables reduc-
Patients with SBS−IF require PS to maintain hydration and tion of PS in patients with SBS−IF.8 TED is approved
nutrition status, including energy, protein sources (amino in the United States (adults) and Europe (adults and
acids), vitamins, trace elements, and electrolytes.1 children aged ࣙ1 year) for the treatment of patients
Seidner et al 521

who are PS dependent.9,10 In the pivotal, phase III, Conference on Harmonisation and Good Clinical Practice,
24 week, multinational, placebo (PBO)-controlled STEPS and the study protocol was approved by institutional review
study (NCT00798967; EudraCT: 2008-006193-15), the pri- boards.
mary study endpoint (ࣙ20% reduction in PS volume at Eligible adult participants in STEPS-3 had completed
Week 20 that was maintained at Week 24) was met in 24 months of TED treatment in STEPS-2, regardless of
63% of adult patients treated with subcutaneous TED whether they had been weaned from PS, and they had
0.05 mg/kg/day compared with 30% of patients treated provided written informed consent before the initiation
with PBO (P < 0.01).8 In the open-label, 2-year, multi- of study-related procedures. All patients received TED
national extension study (STEPS-2; NCT00930644; Eu- 0.05 mg/kg/day by subcutaneous injection.
draCT: 2009-011679-65), treatment with subcutaneous The STEPS-3 study population was stratified into
TED 0.05 mg/kg/day for ࣘ24 months was well tolerated and 2 subgroups for the purpose of data analysis, depending
resulted in additional reductions in weekly PS volume and on length of TED treatment (Figure 1). The TED–TED
number of days of PS infusions per week while maintain- subgroup was composed of patients who received TED
ing patients’ nutrition status.11 The objective of STEPS-3 in the initial PBO-controlled trial (STEPS) and in the
(NCT01560403) was to further assess the long-term safety open-label STEPS-2 study. The no teduglutide treatment
and efficacy of TED in patients with SBS−IF who had (NT)/PBO–TED subgroup included patients who received
completed the STEPS-2 study. NT (entered STEPS-2 directly) or PBO in the initial PBO-
controlled trial (STEPS) and TED in STEPS-2. Compliance
Methods with the dosing regimen, a prespecified endpoint, was
monitored by counting used and unused vials of study drug
Study Design and Population on return patient visits and was considered to have been
STEPS-3 was an additional 1-year, open-label extension achieved if ࣙ80% of planned doses were administered (ie,
of the STEPS-2 open-label TED extension study, designed [vials dispensed–vials returned intact]/days on treatment).
primarily to assess the long-term safety of TED in patients Baseline demographics, patient characteristics, and efficacy
with SBS−IF. In addition, efficacy outcome data using parameters were based on the intent-to-treat (ITT) popula-
measures from the original STEPS study were collected. tion. Baseline for the parameters assessed in STEPS-3 was
STEPS-3 was conducted in the United States, and 5 of the defined as the start of TED treatment in the STEPS study
7 STEPS-2 study sites participated (ie, of the 88 patients for patients in TED–TED or the start of TED treatment
who participated in STEPS-2, 24 patients came from the in STEPS-2 for patients in the NT/PBO–TED subgroup
United States [7 sites; 18 patients] and Canada [3 sites; (Figure 1). As such, observations collected for the patients
6 patients]); the majority were from Europe (7 countries; who received PBO in the initial STEPS trial were not
64 patients). The study was conducted in accordance included in the analysis of the NT/PBO–TED subgroup.
with the Declaration of Helsinki and the International The safety analysis population included all patients who

From the 1 Vanderbilt University Medical Center, Nashville, TN, USA; 2 Scripps Clinic, La Jolla, CA, USA; 3 Hospital of the University of
Pennsylvania and Drexel University, Philadelphia, PA, USA; 4 Mount Sinai Medical Center, New York, NY, USA; 5 Shire Human Genetic
Therapies, Inc., Lexington, MA, USA; and 6 Emory University School of Medicine, Atlanta, GA, USA.
Financial disclosure: The clinical trial was funded by NPS Pharmaceuticals, Inc., Lexington, MA, USA. NPS Pharmaceuticals, Inc., is a wholly
owned indirect subsidiary of Shire. Editorial support, funded by Shire, was provided by Complete Healthcare Communications, LLC (West
Chester, PA, USA). The sponsor of the study participated in study design, data collection, data analysis, data interpretation, review, and approval
of the final report, and provided the study drug.
Conflicts of interest: D.L. Seidner received research support from and served as a study investigator, consultant, and advisory board member for
NPS Pharmaceuticals, Inc., and as a consultant for Shire; K. Fujioka served as a consultant and study investigator for NPS Pharmaceuticals, Inc.,
and as a speaker for Shire; J.I. Boullata served as a study investigator and advisory board member for NPS Pharmaceuticals, Inc.; K. Iyer served
as a consultant, study investigator, and advisory board member for and received research support from NPS Pharmaceuticals, Inc., and has served
as a consultant for Shire; H.-M. Lee is an employee of Shire; and T.R. Ziegler served as a study investigator for NPS Pharmaceuticals, Inc.
Received for publication August 18, 2017; accepted for publication March 5, 2018.
This article originally appeared online on May 15, 2018.
Corresponding Author:
Douglas L. Seidner, MD, AGAF, FACG, CNSC, Vanderbilt University Medical Center, 1211 21st Avenue South,
Ste 514 MAB, Nashville, TN 37232-2713, USA.
Email: douglas.seidner@vanderbilt.edu
This is an open access article under the terms of the Creative Commons Attribution-NonCommercial License, which permits use, distribution and
reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.
522 Nutrition in Clinical Practice 33(4)

Data Analysis
Completed Completed
STEPS STEPS-2 STEPS-3 Descriptive statistics were used to summarize the baseline
27 global sites 25 global sites 5 U.S. sites
randomized, open-label open-label and demographic characteristics, efficacy endpoints, and
double-blind extension extension TEAEs; the study was not designed or sufficiently powered
TED 0.05 mg/kg/day TED 0.05 mg/kg/day to determine the statistical significance of safety or efficacy
24 weeks ≤24 months ≤12 months
endpoints.
TED
0.05 mg/kg/day TED–TED TED–TED Results
(n=43) (n=37) (n=5)
Of the 14 patients who completed treatment with TED
Baseline in STEPS-2 and enrolled in STEPS-3 (ITT population),
13 patients completed the study (ie, were receiving TED
PBO PBO–TED at the time the study concluded). One patient was lost
(n=43) (n=39)
to follow-up after being treated with TED for 8 months;
Baseline
NT/PBO–TED however, available data from this patient were included in
(n=9)
Direct the analysis. The confluence of the rolling start dates and
enrollment the study end date meant that all patients did not receive
NT–TED
into STEPS-2
Not treated
(n=12) 12 months of TED treatment. The mean (SD) duration
in STEPS* of exposure to TED during STEPS-3 was 38.9 (9.8) weeks
Baseline
for the overall population, 41.5 (8.4) weeks for NT/PBO–
TED, and 34.3 (11.3) weeks for TED–TED. Combined
with the TED treatment in the STEPS-2 study, the total
Figure 1. Flow diagram of patients across the STEPS studies.
NT/PBO–TED received NT or PBO in initial PBO-controlled TED exposure time was ࣘ36 months for NT/PBO–TED
trial (STEPS) and TED in STEPS-2. TED–TED received and ࣘ42 months for TED–TED. Of the 14 patients enrolled
TED in initial PBO-controlled trial (STEPS) and in STEPS-2. in STEPS-3, 8 (57%) received ࣙ80% of the planned dose:
*Patients who completed fluid optimization and stabilization 67% (n = 6/9) in the NT/PBO–TED and 40% (n = 2/5) in the
but were not randomized in STEPS because of full study TED–TED subgroups. Patient demographics and baseline
enrollment were eligible for direct enrollment into STEPS-2. characteristics are summarized in Table 1.
NT, no teduglutide treatment; PBO, placebo; TED,
teduglutide.
Efficacy
At baseline, the mean (SD) PS volume requirements were
received ࣙ1 dose of study drug in this extension study and
13.4 (11.6) and 10.5 (7.5) L/week for patients in the
was identical to the ITT population.
TED−TED and NT/PBO−TED subgroups, respectively.
From study baseline to the final dosing visit, mean (SD)
Efficacy and Safety Measures total PS volume was reduced during the STEPS-3 study
Study visits were conducted every 3 months. Prespecified period by 9.8 (14.4) and 3.9 (2.8) L/week for patients in
efficacy parameters included absolute and relative change the TED–TED and NT/PBO–TED subgroups, respectively.
from baseline in actual PS volume received, reduction in The mean (SD) percentage reduction from baseline in PS
days of PS infusions per week, and number of patients who volume was 49.7% (72.4%) for the TED−TED subgroup
achieved independence from PS in the STEPS-3 study. Pre- and 47.8% (42.9%) for the NT/PBO−TED subgroup. In ad-
specified safety parameters included duration of exposure dition, patients in the TED−TED subgroup compared with
to TED; incidence of treatment-emergent adverse events the NT/PBO−TED subgroup exhibited greater reductions
(TEAEs) and treatment-emergent serious adverse events in actual mean PS volume at all visits (Figure 2). In addition
(TESAEs); physical examinations; vital signs; electrocardio- to the mean volume reduction with time during STEPS-3
gram results; colonoscopy evaluations; clinical laboratory compared with baseline, patients reduced the frequency
testing (including serum chemistries for liver and kidney of required PS infusions. For patients in the TED–TED
biochemical values, pancreatic enzymes, and electrolytes); subgroup, the reduction from baseline in mean (SD) days
and assessment of TED-specific antibody formation. Body per week receiving PS at the last dosing study visit in
weight and body mass index (BMI) were determined as this extension study was 3.0 (4.6) days. Patients in the
post hoc analyses because of a data correction to 1 pa- NT/PBO–TED subgroup had a reduction of 2.1 (2.2) days
tient’s weight. Electrocardiograms and colonoscopies were per week receiving PS. Eight of 14 patients had a ࣙ1-day
performed at the first and final study visits; all other safety reduction in PS, and 6 of 14 patients had a ࣙ3-day
parameters were assessed at every study visit. reduction. At the completion of the STEP-3 study,
Seidner et al 523

Table 1. Demographics and Baseline Characteristics.

NT/PBO–TEDa TED–TEDb All Patients


Characteristic (n = 9) (n = 5) (n = 14)

Mean (SD) age,c years 55.9 (12.2) 55.8 (10.7) 55.9 (11.3)
Age range, n (%)
<45 years 2 (22) 1 (20) 3 (21)
45–<65 years 6 (67) 3 (60) 9 (64)
ࣙ65 years 1 (11) 1 (20) 2 (14)
Women, n (%) 6 (67) 4 (80) 10 (71)
Race, n (%)
White 7 (78) 5 (100) 12 (86)
Black 2 (22) 0 2 (14)
Ethnicity, n (%)
Not Hispanic or Latino 9 (100) 5 (100) 14 (100)
Mean (SD) body weight, kg 68.5 (14.2) 58.4 (14.2) 64.9 (14.5)
Mean (SD) BMI, kg/m2 24.4 (4.2) 21.8 (3.2) 23.5 (3.9)
Reason for resection, n (%)
Vascular disease 1 (11) 3 (60) 4 (29)
Crohn’s disease 2 (22) 0 2 (14)
Injury 1 (11) 1 (20) 2 (14)
Volvulus 1 (11) 0 1 (7)
Cancer 0 0 0
Other 4 (44) 1 (20) 5 (36)
Colon remaining, n (%) 7 (78) 3 (60) 10 (71)
Colon-in-continuity, n (%) 5 (56) 3 (60) 8 (57)
Mean (SD) percentage of colon 52.6 (39.9) 50.0 (0.0) 51.8 (32.6)
remainingd
Median (range) estimated remaining 55 (17–100) 76 (30–190) 66 (17–190)
small intestine,e cm
Stoma, n (%) 2 (22) 3 (60) 5 (36)
Mean (SD) time since start of PS 6.5 (9.1) 5.0 (3.5) 6.0 (7.4)
dependence, years

BMI, body mass index; NT, no teduglutide treatment; PBO, placebo; PS, parenteral support (parenteral nutrition and/or intravenous hydration);
TED, teduglutide.
a NT/PBO–TED received NT or PBO in initial PBO-controlled trial (STEPS) and TED in STEPS-2.
b TED–TED received TED in initial PBO-controlled trial (STEPS) and in STEPS-2.
c Age at informed consent in initial PBO-controlled trial (STEPS).
d Includes only patients with a colon (NT/PBO−TED, n = 7; TED−TED, n = 3; all patients, n = 10).
e Includes only patients with known residual small intestine length (NT/PBO−TED, n = 7; TED−TED, n = 5; all patients, n = 12).

4 patients were independent from PS. Two female patients for such antibodies. Subsequent sample results collected
(aged 43 and 41 years) with no stoma, colon-in-continuity, at Months 6 and 9 did not detect neutralizing TED
and baseline PS volumes of 4.5 and 4.7 L/week achieved antibodies.
enteral autonomy after 126 and 130 weeks, respectively, TEAEs were reported by 100% of patients; most were
with TED treatment. The other 2 patients had achieved mild to moderate in severity and none led to study discon-
independence from PS during the STEPS-2 study and tinuation. There were no deaths during the study. There
maintained long-term enteral autonomy in STEPS-3. These were no reports of pancreatic-related events or GI steno-
2 patients were women (aged 61 and 60 years) with no stoma sis/obstruction in this extension study. Among the overall
and baseline PS volumes of 4.1 and 6.3 L/week, respectively. study population, the most common TEAEs were asthenic
conditions (an adverse event [AE] grouping consisting of
medically similar terms including asthenia, decreased ac-
Safety tivity, fatigue, and lethargy) and diarrhea (Table 2). Five
TED-specific antibody-positive samples were reported in patients (36%) reported ࣙ1 TESAE during the study.
3 of 14 patients (21%), all of whom were in the NT/PBO– None of the 14 TESAEs reported were considered by the
TED subgroup. In 1 of these patients, neutralizing anti- study investigators to be related to treatment with TED
bodies specific for TED were detected at the first visit in (Table S1). There were no electrocardiogram abnormalities
STEPS-3, but the Month 3 visit sample was negative reported as a TEAE or TESAE. There was 1 reported
524 Nutrition in Clinical Practice 33(4)

serious cardiac AE in a patient with bilateral pneumo-


18
Mean (SE) PS Volume, L/week

NT/PBO–TED nia and acute congestive heart failure; the events were
16 TED–TED
13.4 deemed by the investigator to be unrelated to treatment
14
with TED.
12
Colonoscopy procedures were scheduled at the first
10
10.5 7.9
STEPS-3 study baseline visit (ie, the end of STEPS-2)
8
6.7 6.2 and at the end of STEPS-3 study visit (ie, Month 12 or
6 6.3 6.0
6.0
early termination). Per the study protocol, polyps were
4
removed at the baseline study visit before the patient could
2
continue in the STEPS-3 extension study. Four of the
0
0. .0 10 patients with remnant colon had colonoscopies at both
–2
BL 3 9 2 the first visit and the end-of-study visit. Of these 4 pa-
Months tients, 1 patient had hyperplastic colon polyps at the first
study visit and colon polyps at the final study visit (no
NT/PBO–TED 9 9 8 7 4
TED–TED 5 5 5 1 1
additional histology reported), and 1 patient had a tubular
adenoma at the final study visit only (normal baseline
colonoscopy). Three of the 10 patients with remnant colon
Figure 2. Mean (SE) reduction in parenteral support.
had only the first visit colonoscopy; 1 patient had cecal
NT/PBO–TED received NT or PBO in initial PBO-controlled
trial (STEPS) and TED in STEPS-2. TED–TED received polyps with histology of tubular adenoma. The remaining
TED in initial PBO-controlled trial (STEPS) and in STEPS-2. 3 of 10 patients with remnant colon had only a single,
The table below the graph has the number of patients for each unscheduled visit colonoscopy; 1 of these patients was
study visit that corresponds to each time point. BL, baseline; initially evaluated for possible polyps, but follow-up his-
NT, no teduglutide treatment; PBO, placebo; PS, parenteral tology showed that no polyp was present. Collectively, of
support (parenteral nutrition and/or intravenous hydration); the 10 patients with remnant colon, 3 patients had ade-
TED, teduglutide.
noma or polyps based on the colonoscopy and/or histology
evaluations.
Patients maintained their nutrition status, as
demonstrated by the lack of substantial changes in
Table 2. Treatment-Emergent Adverse Events Reported in ࣙ2
Patients Sorted by Incidence in Overall Population. mean (SD) body weight (TED−TED, −3.0 [6.7] kg;
NT/PBO−TED, −0.9 [4.6] kg) or BMI (TED−TED,
Adverse Event NT/PBO–TEDa TED–TEDb −1.0 [2.4] kg/m2 ; NT/PBO−TED, −0.3 [1.6] kg/m2 ) at
Group or (n = 9) (n = 5) the last dosing visit compared with baseline (Table S2).
Preferred Term n (%), Events n (%), Events In addition, patients exhibited stable electrolyte levels
(ie, calcium, magnesium, and phosphate), as reflected in
Asthenic 2 (22), 2 1 (20), 1
conditions the absence of meaningful changes from baseline at the
Diarrhea 1 (11), 1 2 (40), 3 end of study visit. There were no clinically meaningful
Abdominal pain 2 (22), 2 0 changes in mean (SD) concentrations for blood urea
Benign neoplasms 2 (22), 2 0 nitrogen (TED−TED, 3.4 [15.7] mmol/L; NT/PBO−TED,
gastrointestinal 0.7 [2.3] mmol/L), serum creatinine (TED−TED, 23.0
Catheter sepsis 0 2 (40), 2 [61.5] μmol/L; NT/PBO−TED, −3.4 [12.7] μmol/L), serum
Cognition and 2 (22), 3 0
sodium (TED−TED, −1.6 [7.3] mmol/L; NT/PBO−TED,
attention
disorders and 0.6 [3.0] mmol/L), or urine sodium (TED−TED, 56.2
disturbances [43.2] mmol/L; NT/PBO−TED, −22.3 [64.8] mmol/L) in
Dyspnea 0 2 (40), 2 patients at the end of study visit compared with baseline.
Hypersensitivity 2 (22), 2 0 One patient in the NT/PBO–TED subgroup reported an
Viral infection 2 (22), 2 0 elevation in serum creatinine levels (126 μmol/L; upper
Weight decreased 0 2 (40), 3 limit of normal, 115 μmol/L) that returned to 116 μmol/L
NT, no teduglutide treatment; PBO, placebo; TED, teduglutide.
by the 6 month visit and remained within normal limits
a NT/PBO–TED received NT or PBO in initial PBO-controlled trial through the end of study visit; this event was considered
(STEPS) and TED in STEPS-2. to be unrelated to treatment by the site investigator. Stable
b TED–TED received TED in initial PBO-controlled trial (STEPS)
serum albumin levels were maintained in all patients,
and in STEPS-2.
and mean decreases in serum concentrations of alkaline
phosphatase (ALP), alanine aminotransferase (ALT), and
aspartate aminotransferase (AST) were reported in all
Seidner et al 525

Table 3. Change in Serum Albumin Level and Liver Enzymes From Baseline to Last Dosing Study Visit.

Liver Function Tests/Liver Enzymes NT/PBO–TEDa (n = 9) TED–TEDb (n = 5)

Serum albumin level (normal range, 32–50 g/L)


Baseline, g/L, mean (SD) 39.1 (3.2) 41.4 (4.3)
Change from baseline, g/L, mean (SD) 0.7 (3.2) −2.8 (3.6)
>10% increase from baseline at EOT, n (%) 1 (11) 0
>10% decrease from baseline at EOT, n (%) 1 (11) 1 (20)
Alkaline phosphatase (normal range, 37–147 U/L)
Baseline, U/L, mean (SD) 124.8 (50.1) 130.4 (50.8)
Change from baseline, U/L, mean (SD) −18.7 (25.7) −17.0 (39.9)
>10% increase from baseline at EOT, n (%) 0 1 (20)
>10% decrease from baseline at EOT, n (%) 4 (44) 2 (40)
ALT (normal range, 0–55 U/L)
Baseline, U/L, mean (SD) 34.7 (22.6) 24.8 (4.9)
Change from baseline, U/L, mean (SD) −4.0 (14.3) −2.6 (10.7)
>10% increase from baseline at EOT, n (%) 3 (33) 2 (40)
>10% decrease from baseline at EOT, n (%) 6 (67) 2 (40)
AST (normal range, 0–45 U/L)
Baseline, U/L, mean (SD) 31.0 (10.8) 25.4 (5.6)
Change from baseline, U/L, mean (SD) −2.2 (6.6) −1.4 (8.1)
>10% increase from baseline at EOT, n (%) 2 (22) 1 (20)
>10% decrease from baseline at EOT, n (%) 5 (56) 2 (40)
Bilirubin (normal range, 5.1–25.7 μmol/L)
Baseline, μmol/L, mean (SD) 8.7 (7.5) 8.5 (4.0)
Change from baseline, μmol/L, mean (SD) −2.5 (6.2) −3.4 (4.4)
>10% increase from baseline at EOT, n (%) 1 (11) 0
>10% decrease from baseline at EOT, n (%) 4 (44) 3 (60)
GGT (normal range, 0–30 U/L)
Baseline, U/L, mean (SD) 79.0 (56.1) 57.8 (82.4)
Change from baseline, U/L, mean (SD) −9.9 (26.7) −4.4 (11.7)
>10% increase from baseline at EOT, n (%) 3 (33) 1 (20)
>10% decrease from baseline at EOT, n (%) 5 (56) 3 (60)

ALT, alanine aminotransferase; AST, aspartate aminotransferase; EOT, end of treatment; GGT, γ -glutamyl transferase; NT, no teduglutide
treatment; PBO, placebo; TED, teduglutide.
a NT/PBO–TED received NT or PBO in initial PBO-controlled trial (STEPS) and TED in STEPS-2.
b TED–TED received TED in initial PBO-controlled trial (STEPS) and in STEPS-2.

but 2 patients at the end of study visit compared with Discussion


baseline (Table 3). Furthermore, for each liver enzyme
measured, more patients experienced a >10% decrease The findings from the open-label STEPS-3 extension study
between baseline and end of treatment than experienced a provide general support for the safety, tolerability, and
>10% increase in these measures (Table 3). clinical utility of TED in the treatment of adults with
Two patients had mild hepatobiliary-related laboratory SBS−IF for ࣙ36 months.
findings reported at baseline that continued throughout the The safety data in patients with extended exposure to
study. The increases in ALT and γ -glutamyl transferase TED indicated that the drug was well tolerated, and data
(GGT) reported in these 2 patients were not deemed to be were in line with the expectations based on previous studies.
clinically significant and were not considered by the site No indication of new adverse safety signals emerged in the
investigator to be related to treatment with TED. During STEPS-3 long-term extension study. The development of
the study, increased serum lipase concentrations (>3 times anti-TED-specific antibodies was reported in some patients,
upper limit of normal) were reported in 3 patients, and but this finding had no apparent effect on the efficacy
increased serum amylase concentrations (ࣙ350 U/L) were of TED, as determined by PS adjustments during the
reported in 1 patient, without reported clinical evidence of course of this study among these individuals. Although
pancreatitis. An additional patient had decreased amylase the intestinotrophic properties of TED may account for
levels (ࣘ15 U/L) postbaseline. the favorable effects of intestinal adaptation observed in
526 Nutrition in Clinical Practice 33(4)

patients with SBS,12 the development of GI polyps is a whole. In the subgroup of patients who experienced ࣙ50%
previously identified safety concern.8,11 In this small open- reductions in PS volume, liver enzymes decreased by a mean
label, 1-year STEPS-3 study, 10 patients had remnant colon of 6%–44% from baseline values.11 The improvement in
and underwent colonoscopy procedures. In total, 3 patients liver biochemical values seen in STEPS-2 continued in this
were reported to have polyps at either the first study visit study, with mean decreases from baseline in ALP, ALT, AST,
and/or the final study visit, 2 of whom had histologically GGT, and bilirubin levels observed at the end of treatment.
confirmed adenoma. In the preceding open-label, 2-year In addition, for each liver enzyme measured, more patients
STEPS-2 study, 9 patients had reported polyps with no experienced >10% decreases from baseline than experi-
case of overt malignancy related to polyps reported, 5 of enced >10% increases from baseline at end of treatment.
whom had histologically confirmed adenoma.11,13 Collec- Together, the results of this study support previous findings
tively, when the overlap in the STEPS-2 final visit and indicating that in patients receiving TED, chronic PS can
the STEPS-3 baseline visit is considered, 10 patients were be reduced with corresponding improvements in liver
reported to have polyps throughout the STEPS study series. function.
Together, these colonoscopy results support the safety- TED has been shown to enhance intestinal absorption
related recommendation in the U.S. and European pre- in patients with SBS−IF.18 Therefore, it is important that
scribing information stating that patients receiving TED patients who achieve improvements in intestinal fluid ab-
have regular colonoscopy for signs of neoplastic growth.9,10 sorption during treatment with TED also achieve balanced
The ongoing global registry study for patients with SBS reductions in the need for PS to minimize the risk for
(ClinicalTrials.gov identifier: NCT01990040) will provide fluid overload. In this study, long-term treatment with TED
needed data regarding the incidence of colorectal polyps for ࣘ42 months was associated with sustained efficacy and a
in the SBS population overall, as well as among patients further decline in PS requirements in patients with SBS−IF.
treated with TED, during a follow-up period of at least It is also noteworthy that some patients who achieved
10 years. Any available results from routine colonoscopies PS autonomy in STEPS-2 maintained such independence
will be captured as part of the clinical outcomes data with continuous long-term TED in STEPS-3. However, the
collection registry, and may permit a more comprehensive finding that other patients reached enteral autonomy for the
analysis of the potential risk of polyp formation associated first time after approximately 2 years of treatment with TED
specifically with TED therapy. in STEPS-3 suggests that some patients may need longer
Patients maintained and improved nutrition status in exposure to TED to achieve PS independence.
the presence of PS volume reductions, as evidenced by the Limitations of the study include the open-label design
reported vital signs and chemistry laboratory tests during and lack of a control arm; additionally, our study popu-
the course of the study. Although 2 patients experienced lation was a small selected cohort that met inclusion re-
episodes of decreased body weight that were reported as quirements for longer-term follow-up. This extension study,
TEAEs, these episodes appeared to be transient; mean body undertaken at the end of the clinical development program
weight and BMI remained relatively stable during the course for drug approval, had a study design and a data analysis
of the study. Furthermore, electrolyte levels remained steady plan that was descriptive and was not intended to be suf-
or improved during the course of the study, and mean ficiently powered for statistical significance analysis. Even
serum kidney biochemical values generally remained stable with this limitation, this type of study design can provide
between baseline and the end of the study. In addition, mean important information on outcomes, and as reported here
serum albumin levels were relatively unchanged at the end for the STEPS-3 data, observed results that were in line with
of the study compared with baseline, indicating that stable those of previous studies.8,11 These factors may limit the
nutrition status was maintained during the course of the applicability of the current findings to patients in the real-
study.14,15 It is notable that stable or improved nutrition world clinical setting with a broader range of disease and
status found in this 1-year extension study is similar to that clinical characteristics and require further study. Although
reported in the previous 2-year STEPS-2 study,11 further the ongoing global registry study will add valuable real-
supporting the conclusion that extended exposure to TED world experience in broader populations of patients with
is well tolerated. SBS, this small extension study provides additional new
Long-term PS is associated with increased risk of liver data that long-term treatment with TED has a safety profile
damage, and advanced liver disease has been reported in similar to that reported in previous studies and supports
0%–50% of patients receiving chronic PS; the greatest risk the overall conclusion that TED is well tolerated. However,
is in patients with ultrashort bowel (<50 cm).16,17 During the ideal time for response was not determined and may,
the preceding STEPS-2 study, serum concentrations of in fact, be undeterminable because, as this study shows,
liver enzymes either declined or remained stable during PS reductions can be further enhanced with continuous
2.0–2.5 years of TED treatment in the study group as a treatment.
Seidner et al 527

Acknowledgments 4. Carlsson E, Bosaeus I, Nordgren S. Quality of life and concerns in


patients with short bowel syndrome. Clin Nutr. 2003;22:445-452.
The authors wish to thank all participating patients and their
5. Kelly DG, Tappenden KA, Winkler MF. Short bowel syndrome:
families and the network of clinical investigators, study coor- highlights of patient management, quality of life, and survival. JPEN
dinators, and operations staff at all participating centers for J Parenter Enteral Nutr. 2014;38:427-437.
their contributions to the entire STEPS clinical trial program. 6. Kalaitzakis E, Carlsson E, Josefsson A, Bosaeus I. Quality of life in
The authors also thank Peter Nagy, MD, and Clément Olivier, short-bowel syndrome: impact of fatigue and gastrointestinal symp-
MD, of Shire International GmbH (Zug, Switzerland), for their toms. Scand J Gastroenterol. 2008;43:1057-1065.
insights during the finalization of the manuscript. 7. Janssen P, Rotondo A, Mule F, Tack J. Review article: a comparison
of glucagon-like peptides 1 and 2. Aliment Pharmacol Ther. 2013;37:18-
Statement of Authorship 36.
8. Jeppesen PB, Pertkiewicz M, Messing B, et al. Teduglutide reduces need
D. L. Seidner, K. Fujioka, J. I. Boullata, K. Iyer, and T. R. for parenteral support among patients with short bowel syndrome with
Ziegler contributed to the conception and design of the re- intestinal failure. Gastroenterology. 2012;143:1473-1481.
 R
search; D. L. Seidner, K. Fujioka, J. I. Boullata, K. Iyer, 9. GATTEX (teduglutide [rDNA origin]). Full prescribing information.
and T. R. Ziegler contributed to the acquisition, analysis, Shire-NPS Pharmaceuticals, Inc., Lexington, MA; 2016.

R

and interpretation of the data; H.-M. Lee contributed to the 10. Revestive (teduglutide). EMA summary of product characteristics.
analysis and interpretation of data; D. L. Seidner, K. Fujioka, Shire Pharmaceuticals Ireland Limited, Dublin, Ireland; 2016.
11. Schwartz LK, O’Keefe SJ, Fujioka K, et al. Long-term teduglutide for
J. I. Boullata, K. Iyer, H.-M. Lee, and T. R. Ziegler drafted the
the treatment of patients with intestinal failure associated with short
manuscript; D. L. Seidner, K. Fujioka, J. I. Boullata, K. Iyer,
bowel syndrome. Clin Transl Gastroenterol. 2016;7:e142.
H.-M. Lee, and T. R. Ziegler critically revised the manuscript; 12. Jeppesen PB, Sanguinetti EL, Buchman A, et al. Teduglutide (ALX-
D. L. Seidner, K. Fujioka, J. I. Boullata, K. Iyer, H.-M. Lee, 0600), a dipeptidyl peptidase IV resistant glucagon-like peptide 2 ana-
and T. R. Ziegler agree to be fully accountable for ensuring the logue, improves intestinal function in short bowel syndrome patients.
integrity and accuracy of the work; and all authors read and Gut. 2005;54:1224-1231.
approved the final manuscript. 13. Rudzki S, Armstrong D, Jeppesen PB, Forbes A, Lee H-M. Post hoc
analysis of polyps in 9 short bowel syndrome patients treated with
Supplementary Information teduglutide. Clin Nutr. 2016;35(Suppl. 1):S183-S184.
14. Don BR, Kaysen G. Serum albumin: relationship to inflammation and
Additional supporting information may be found online in the nutrition. Semin Dial. 2004;17:432-437.
supporting information tab for this article. 15. Hankins J. The role of albumin in fluid and electrolyte balance. J Infus
Nurs. 2006;29:260-265.
References 16. Wiles A, Woodward JM. Recent advances in the management of
intestinal failure-associated liver disease. Curr Opin Clin Nutr Metab
1. Jeppesen PB. Spectrum of short bowel syndrome in adults: intestinal
Care. 2009;12:265-272.
insufficiency to intestinal failure. JPEN J Parenter Enteral Nutr.
17. Dibb M, Teubner A, Theis V, Shaffer J, Lal S. Review article: the
2014;38:8S-13S.
management of long-term parenteral nutrition. Aliment Pharmacol
2. Matarese LE. Nutrition and fluid optimization for patients with short
Ther. 2013;37:587-603.
bowel syndrome. JPEN J Parenter Enteral Nutr. 2013;37:161-170.
18. Jeppesen PB, Hartmann B, Thulesen J, et al. Glucagon-like peptide
3. Winkler MF, Smith CE. Clinical, social, and economic impacts of
2 improves nutrient absorption and nutritional status in short-bowel
home parenteral nutrition dependence in short bowel syndrome. JPEN
patients with no colon. Gastroenterology. 2001;120:806-815.
J Parenter Enteral Nutr. 2014;38:32S-37S.
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 4
Survey of Nutrition Management Practices in Centers August 2018 528–538

C 2017 American Society for

for Pediatric Intestinal Rehabilitation Parenteral and Enteral Nutrition


DOI: 10.1177/0884533617719670
wileyonlinelibrary.com

Anita M. Nucci, PhD, RD, LD1 ; Kipp Ellsworth, MS, RD, CSP, CNSC2 ;
Austin Michalski, RD, LD, CNSC3 ; Emily Nagel, MS, RD, CNSC4 ;
and Jackie Wessel, MEd, RD, CNSC, CSP, CLE5 ;
on behalf of the ASPEN Pediatric Intestinal Failure Section

Abstract
Background: Nutrition management of pediatric intestinal failure (IF) requires interdisciplinary coordination of parenteral
nutrition (PN) and enteral nutrition (EN) support. Nutrition strategies used by specialists in pediatric intestinal rehabilitation
to promote gut adaptation and manage complications have not been previously summarized. Methods: A practice survey was
distributed to members of the dietitian subgroup of the American Society for Parenteral and Enteral Nutrition Pediatric Intestinal
Failure Section. The survey included 24 open-ended questions related to PN and enteral feeding strategies, nutrition management
of PN-associated liver disease, and laboratory monitoring. Results: Dietitians from 14 centers completed the survey. Management
components for patients at risk for cholestasis were consistent and included fat minimization, trace element modification, avoiding
PN overfeeding, and providing EN. Parenteral amino acid solutions designed for infants/young children are used in patients <1 or
2 years of age. Trace minerals are dosed individually in 10 of 14 centers. Eleven centers prescribe a continuous infusion of breast
milk or elemental formula 1–2 weeks after resection while 3 centers determine the formula type by the extent of resection. Most
(86%) centers do not have a protocol for initiating oral/motor therapy. Laboratory panel composition varied widely by center. The
selection and frequency of use depended on clinical variables, including cholestatic status, exclusive vs partial PN dependence,
postrepletion verification vs routine monitoring, intestinal anatomy, and acuity of care. Conclusion: EN and PN management
strategies are relatively consistent among U.S. centers. Collaborative initiatives are necessary to define better practices and establish
laboratory monitoring guidelines. (Nutr Clin Pract. 2018;33:528–538)

Keywords
enteral nutrition; liver failure; parenteral nutrition; pediatrics; short bowel syndrome; surveys and questionnaires

Short bowel syndrome (SBS) may occur after massive fluid and electrolyte balance as well as enteral nutrition
resection of the small bowel due to necrotizing enterocolitis (EN) and PN support.7 Pediatric nutrition specialists rely on
or conditions such as small bowel atresia, malrotation established nutrition guidelines and interventions intended
with midgut volvulus, or gastroschisis and is the most to promote adaptation of the remnant small bowel and facil-
common cause of intestinal failure (IF) in children.1 IF itate weaning of PN and achievement of enteral autonomy
can also occur due to impaired intestinal absorption or before complications occur.8
altered motility in children with microvillus atrophy or
intestinal pseudo-obstruction. Pediatric IF is a clinical
condition characterized by malabsorption, malnutrition, From 1 Georgia State University, Atlanta, Georgia, USA; 2 Children’s
Healthcare of Atlanta, Atlanta, Georgia, USA; 3 University of
and growth retardation.2-4 Management of children with
Michigan, C.S. Mott Children’s Hospital, Grand Rapids, Michigan,
IF is complex but can be optimized by interdisciplinary USA; 4 Helen DeVos Children’s Hospital, Grand Rapids, Michigan,
coordination of nutrition support, medical therapies, and USA; and 5 Cincinnati Children’s Hospital Medical Center,
surgical interventions.5 Many children with IF experience Cincinnati, Ohio, USA.
progressive adaptation of their remaining small bowel over Financial disclosure: None declared.
a few months to years with eventual independence from Conflicts of interest: None declared.
parenteral nutrition (PN). However, hepatobiliary disease
This article originally appeared online on July 21, 2017.
is a chronic complication of long-term use of PN and is
Corresponding Author:
a major contributor to the high morbidity and mortality
Anita M. Nucci, PhD, RD, LD, Department of Nutrition, Georgia
of children with IF.5,6 Nutrition management during the State University, PO Box 3005, Atlanta, GA 30302-3995, USA.
period of intestinal adaptation includes maintenance of Email: anucci@gsu.edu
Nucci et al 529

Nutrition practice guidelines for the general manage- subgroup members between January and September 2015.
ment of children receiving PN have existed for several The survey included 23 primarily open-ended questions
decades.9-11 In 2004, a 2-part review of intestinal reha- related to IR center composition, PN and enteral feeding
bilitation (IR) and SBS management by DiBaise et al1 strategies, nutrition management of IFALD, and laboratory
was published. Part I included an overview of the patho- monitoring (Table 1). The study was determined to be
physiology of SBS and a discussion of luminal nutrients, exempt from federal regulations by university institutional
hormones, growth factors, and other substances that may review boards.
enhance adaptation. Part II provided nutrition management
guidelines for SBS with dietary guidelines by the presence or Results
absence of a colon, suggestions for micronutrient supple-
mentation, and a discussion of the clinical benefits of man- IR Center Structure
agement by an interdisciplinary IR team.5 More recently, Dietitians from 14 centers completed the survey. The cen-
detailed macronutrient and micronutrient recommenda- ters were located in Atlanta, Georgia; Aurora, Colorado;
tions for the management of children with IF have been Baltimore, Maryland; Boston, Massachusetts; Cleveland,
published. These reports include initial postoperative as well Ohio; Cincinnati, Ohio; Columbus, Ohio; Grand Rapids,
as short-term and long-term PN and EN recommendations Michigan; Indianapolis, Indiana; Little Rock, Arkansas;
and laboratory monitoring guidelines12,13 with an emphasis New Haven, Connecticut; St Louis, Missouri; and Seattle,
on the prevention of nutrient deficiencies and complica- Washington. Seven of the 14 centers (50%) have an or-
tions, such as IF-associated liver disease (IFALD).14 In ganized interdisciplinary IR service that manages children
addition, multiple cohort studies have reported benefits of with IF upon discharge from the neonatal intensive care
restricting intravenous (IV) soy-based fats as a means of pre- unit (NICU). While in the NICU, children with IF are
venting IFALD,15 as a treatment for infants diagnosed with managed by the NICU service with consultation from the
IFALD,16 and after reversal of cholestasis using fish oil– IR team. Centers without an organized IR team use both
based IV emulsion.17 Although clinical nutrition guidelines the gastroenterology and pediatric surgery services for care
for the management of children with IF are available, most management.
research that has been conducted in this population has been
descriptive or semi-experimental in design. Clinical trials are
costly and often require multicenter collaboration to obtain
PN Management
an adequate sample size. However, experimental studies Parenteral amino acid solutions designed for infants and
to establish evidence-based criteria for care are needed.12 young children are used in patients <1 year of age in all
No published review of current nutrition practice for this but 1 center, which uses the product up to 2 years of age. A
population has been reported. The purpose of this study is standard parenteral amino acid solution is used thereafter.
to (1) summarize the nutrition strategies used by physicians Twelve of 14 centers (86%) include protein calories when
and dietitians in pediatric IR to promote gut adaptation and calculating total caloric intake. While excluding protein
manage complications in children with IF and (2) compare calories when calculating total caloric intake has not
current nutrition practice with existing recommendations. been a standard of care for many years, we assessed
whether dietitians working in IR centers still follow this
practice. The general practice of dosing trace elements and
Methods cycling/tapering of PN solutions is shown in Table 2. Eleven
The Pediatric Intestinal Failure Section of the American of the 14 centers (79%) reported monitoring serum levels
Society for Parenteral and Enteral Nutrition (ASPEN) was of trace elements as a result of reduced intake, specifically
initiated in 2014. The section includes physician, dietitian, copper, selenium, and zinc. Dietitians from 10 centers
and pharmacy subgroups. The dietitian subgroup includes reported that the decision to initiate iron supplementation
members from 27 pediatric hospitals within the United is dependent upon laboratory determination of deficiency.
States and 4 international facilities. The short-term goals of The general consensus was that enteral iron should be
the dietitian subgroup are (1) to prepare nutrition education provided when there is a sufficient length of proximal small
materials for use by healthcare professionals and care- bowel and tolerance to enteral feedings is established. IV
giver(s) of children with IF and (2) to create a network of iron is selected in the case of an extremely short bowel
practitioners with a specialty in pediatric IF for the purpose and feeding intolerance where PN is the primary source of
of sharing resources and discussing nutrition practice issues. nutrition. PN support standards of care did not significantly
A long-term goal of the subgroup is to develop nutrition differ for children who receive long-term PN (>1 month)
research questions and form multicenter research collabo- at the centers surveyed. However, 4 center dietitians (29%)
rations. A qualitative nutrition practice survey developed by noted that laboratory monitoring differed by the length
the dietitian subgroup was electronically distributed to all of time on PN with the inclusion of parameters for
530 Nutrition in Clinical Practice 33(4)

Table 1. Pediatric Intestinal Failure Nutrition Practice Survey Questions.

Number Question

1 What medical services/teams manage your patients with IF?


2 What is your standard protein source for PN?
3 Do you use the same protein source for all patients (yes/no)?
4 If you use the same protein source for all patients, under what circumstances would you use a specialized
parenteral amino acid solution?
5 Do you use a trace mineral package or dose minerals individually for patients on PN?
6 For patients with IF on PN, when would you provide supplemental iron?
7 If supplemental iron is provided, how is it given and under what circumstances would each method be chosen?
8 Do you have any protocols for patients receiving long-term (>1 month) PN without EN intake?
9 Do you include protein calories when calculating the caloric intake of patients on PN?
10 What is your practice regarding cycling PN?
11 What is your practice regarding tapering of PN?
12 If you are caring for a patient with IF who is at risk for cholestasis, what is your general philosophy for
nutrition management?
13 If you are caring for a patient with IF who has cholestasis, what is your standard practice for nutrition
management?
14 If you are caring for a patient with cholestasis, what is your institution policy with regard to the provision of
parenteral trace minerals?
15 How many days are your patients with IF typically ordered “nothing by mouth” status post resection?
16 What is your formula of choice for children with IF status postresection?
17 What is your typical mode of feeding after resection?
18 What oral supplements do you typically recommend for patients with IF?
19 Do you have a protocol for starting oral/motor therapy for oral feedings after resection?
20 How soon are oral feedings initiated in children with IF at your center?
21 Do you use urine sodium to determine whether a patient with IF requires sodium supplementation?
22 What laboratory tests do you routinely follow in patients with IF on PN? Include macronutrients and
micronutrients and interval for follow-up (eg, daily, weekly, monthly).
23 Which of the following parameters do you follow in patients receiving PN?
a. Carbohydrate infusion rate (select: mg/kg/min or mg/kg/h)
b. Serum triglyceride
c. Serum cholesterol
d. Triene/tetraene ratio
e. Nonprotein nitrogen/calorie ratio
f. Other:

EN, enteral nutrition; IF, intestinal failure; PN, parenteral nutrition.

trace elements and essential fatty acid deficiency to the Most centers (n = 12) do not have a formal protocol
monitoring schedule for those on long-term PN. for the initiation of oral feedings after small bowel resec-
tion. Nine of these centers do consult with occupational
or speech therapy for the purpose of encouraging age-
Enteral/Oral Management appropriate complementary food intake when the child is
Eight of the center dietitians (57%) reported a length of developmentally ready. Two centers reported that they use a
time that children with IF are typically unable to receive protocol for starting oral intake. Of these, 1 center begins
enteral/oral feeding after initial surgery (range, 5 days to 2 with nonnutritive oral feedings of human milk or infant
weeks). The type of EN and oral supplementation provided formula and advances to small-volume oral boluses when
to children with SBS after resection varied between centers trophic feedings are tolerated. This center also initiates solid
(Table 3). Most centers (n = 13) prefer the administration of foods at 6 months of age beginning with vegetables followed
continuous feeds via nasogastric tube (NG) or gastrostomy by bananas, grains, meats, and fruit. The second center
tube (GT) postresection, with 2 also recommending small begins with 2 oral feedings per day under the supervision of
oral feedings as tolerated. One center prefers to provide an occupational or speech therapist when enteral feedings
oral feeding in the absence of feeding intolerance and if have reached 3 mL/h. The timing of initiation of oral feed-
the child is developmentally ready for oral intake. Vitamin, ings varies by center. Most centers did not report an exact
trace element, and fiber supplements are the products most time point for the initiation of feedings but all provided
commonly prescribed. qualifications for initiation of oral feedings (Table 3).
Nucci et al 531

Table 2. Practice Responses to Questions Related to Parenteral Nutrition Management.

Number of Centers
Survey Question Response Rate Practice Response per Response

Do you use a trace element 14 of 14 r Trace element package only 0


package or dose minerals r Individual dosing 9
individually for patients on r Both trace element package and individual 5
PN? dosing are used

What is your practice regarding 11 of 14 Cycling criteriaa


cycling of PN? r Medical stability 6
r Minimum 3 months corrected age 3
r Weight >5 kg 1
r Intake of >50 kcal/kg/d from EN 1
r Tolerance of continuous enteral feeding 1
14 of 14 Cycling strategy
r 1- to 2-hour increments 3
r 2- to 4-hour increments 5
r No more than 4 hours off PN (infants) 1
r No specific strategy reported 5

What is your practice regarding 14 of 14 Tapering strategy


tapering of PN? r minutes up and down
1
r 30 minutes up and 1 hour down
1
r 1 hour up and down
8
r 2 hours down only
1
r hours up and down
1
r No specific strategy reported
2

EN, enteral nutrition; PN, parenteral nutrition.


a Centers may have reported >1 response.

Prevention and Management of IFALD Two centers provide a reduced amount of copper and 3
centers remove manganese entirely from the PN solution.
The center dietitians were queried regarding general
philosophies toward the nutrition management of patients
at risk for IFALD. Limiting IV fat was a major component Laboratory/Biochemical Monitoring
of the nutrition management of children with IF at risk An overview of the IF center dietitian practice survey
for developing cholestasis at all centers (Table 4). Among responses to questions related to laboratory/biochemical
the centers employing some form of fat restriction, typical monitoring in children with IF is shown in Table 5. Most
dosing was limited to 1–2 g/kg/d. The second major theme centers (93%) use urine sodium to determine whether a
among the centers participating in the survey was providing patient requires sodium supplementation. Dietitians from
EN in the form of trophic feedings or aggressive advance- all centers listed a variety of panels, vitamins, trace ele-
ment of feedings. Monitoring parameters included glucose ments, and miscellaneous laboratories whose selection and
infusion rate (GIR), triene/tetraene ratio, trace elements, frequency of use appeared to depend largely on a patient’s
and weight gain and linear growth. The center dietitians clinical variables such as noncholestatic vs cholestatic status,
were also asked to describe their standard practice for the exclusive vs partial PN dependence, postrepletion verifica-
nutrition care of a child with IF as well as IFALD. Fat tion vs routine monitoring, intestinal anatomy, and acuity
minimization or restriction is the most common element of of care (ie, NICU vs general floor vs outpatient). Dietitians
nutrition support practice among all centers included in the from all centers use the GIR (mg/kg/min) as a clinical
survey (Table 4). Each center typically limits IV fat to 1 parameter for patients receiving PN.
g/kg/d in patients with cholestasis. However, some centers
(n = 3) reduce fat dose to <1 g/kg/d either by providing
a restricted daily dose or providing 1 g/kg 2 or 3 days
Discussion
per week. Four dietitians reported that trace elements were While the provision of PN has resulted in a reduced
dosed individually at their centers for children with IFALD. mortality rate for infants and children with IF, serious and
532 Nutrition in Clinical Practice 33(4)

Table 3. Practice Responses to Questions Related to Enteral/Oral Nutrition Management.

Response Number of Centers


Survey Question Rate Practice Response per Response

What is your formula of choice 14 of 14 r Human milk or elemental infant formula 8


for children with intestinal r Amino acid–based infant formula 3
failure status postresection? r Selection based on the extent of small bowel resectiona 3

What oral supplements do you 14 of 14 r Vitamin and trace element supplements 13


typically recommend for r Carbohydrate/fat or fat modular 4
patients with intestinal r Fiber supplements
failure?b ◦ Green beans 2
◦ Pectin 3
◦ Other fiber supplements 3

How soon are oral feedings 14 of 14 r As soon as possible 4


initiated in children with r Depends on stool/ostomy output 2
intestinal failure at your r Varies individually 2
center? r Within 7–10 days 1
r Medically stable for 2 weeks on EN 1
r Age appropriate (>37 weeks) 1
r Developmentally appropriate; 2–3 days after EN initiated 1
r After EN is initiated 1
r After EN tolerated at 5–10 mL/h 1

EN, enteral nutrition.


a Human milk or an elemental infant formula with significant resection; polymeric infant formula with minimal resection.
b Centers may have reported >1 response.

potentially life-threatening complications may still occur as dietitians surveyed reported that cycling of the PN solution
a result of long-term use of PN. Growth failure continues is a treatment goal in their practices and that advancement
to be observed in a high percentage of children with IF,18 (reduction in PN infusion hours) is determined by blood
the risk of nutrient deficiencies has been shown to increase glucose tolerance. Providing a dextrose solution over fewer
during the transition from PN to EN,19-21 and multiple hours may result in a GIR greater than what is currently
years of PN therapy can result in the development of recommended for children without cholestasis (<12–14
IFALD.14 The nutrition care of children with IF should mg/kg/min).23 Therefore, monitoring for hyperglycemia or
be individualized based upon the intestinal anatomy and hypoglycemia is essential during the cycled infusion and
medical condition of each patient. The EN and PN man- while the infusion is off. Abrupt cessation of PN has been
agement strategies reported by the pediatric IF practitioners reported to be well tolerated in children >2 years of age.24
who participated in this survey are relatively consistent. However, many children with IF are <1 year of age at the
Only half of the dietitians surveyed function as part of an time of diagnosis.25 Tapering of the dextrose solution over
organized IR program. The strategies currently being used 30–60 minutes before discontinuation may prevent rebound
are designed to support intestinal adaptation and reverse hypoglycemia and is a common practice reported by the
or slow the progression of complications. However, clinical dietitians who completed our survey.
and laboratory monitoring practices vary considerably be- Most centers provide human milk or amino acid–based
tween centers. Collaborative initiatives such as the Pediatric infant formula as the primary source of nutrition for chil-
Intestinal Failure Section of ASPEN are ideally suited to dren with SBS. This practice aligns with recommendations
define better practices and establish monitoring guidelines from the current literature. In a retrospective medical record
through multicenter research efforts. review of 30 neonates with SBS, Andorsky et al26 reported
PN is an established treatment for children with IF that the use of either human milk or an amino acid–based
that must be carefully managed to prevent or delay the formula was associated with a shorter duration of PN.
development of PN-associated complications.22 Methods Human milk contains long-chain fatty acids, free amino
currently recommended for the prevention of IFALD in- acids such as glutamine, and growth factors that paired
clude avoiding overfeeding, providing a balanced distri- with immune-enhancing properties may assist in intestinal
bution of macronutrients in the PN solution, cycling the adaptation. An amino acid–based formula may decrease
PN infusion, and providing EN as tolerated.23 All of the the duration of PN since it contains a high percentage of
Nucci et al 533

Table 4. Practice Responses to Questions Related to Prevention and Management of Intestinal Failure–Associated Liver Disease.

Number of Centers
Survey Question Response Rate Practice Response per Response

If you are caring for a patient 14 of 14 r IV fat restriction 12


with short bowel syndrome r Initiate/advance EN 9
who is at risk for cholestasis, r Cycle PN 4
what is your general r Avoid overfeeding 4
philosophy for nutrition r Dose trace elements individually 2
management?a r Wean PN 2
r Monitor serum levels of trace elements 2
r Provide parenteral protein at age-appropriate 1
level

If you are caring for a patient 14 of 14 r IV fat restriction 12


with short bowel syndrome r Initiate/advance enteral nutrition 9
who has cholestasis, what is r Cycle PN 6
your standard practice for r Dose trace elements individually 4
nutrition management? r Provide fish oil–based IV fat emulsion 4
r Monitor serum levels of trace elements 4
r Avoid overfeeding 2
r Monitor for essential fatty acid deficiency 2
(triene/tetraene ratio)
r Limit parenteral protein dose 2
r Do not exceed maximum recommended glu- 1
cose infusion rate
r Provide enteral formula with higher MCT oil 1
content

EN, enteral nutrition; IV, intravenous; MCT, medium-chain triglyceride; PN, parenteral nutrition.
a Centers may have reported >1 response.

long-chain fatty acids, which may promote mucosal et al29 recommended the use of human milk or standard
adaptation.27 Although long-chain fatty acids promote polymeric formula depending on the age of the child.
adaptation and provide a source of essential fatty acids, Most centers initiate continuous feedings postresection
medium-chain triglycerides are advantageous because they since they are believed to be better tolerated than bolus
can be directly absorbed by enterocytes. Most amino feeds and promote adaptation via continual saturation of
acid–based formulas contain a mixture of long-chain and the intestinal lumen. However, limited research evidence
medium-chain triglycerides, which may explain why most exists on the topic. A frequently cited study to support the
centers prefer these formulas if human milk is not available. use of continuous feeds was conducted by Parker et al30
Some researchers argue that there is insufficient evidence to over 30 years ago. The researchers reported that continuous
support 1 type of formula over another for patients with feedings yielded a significant increase in body weight and
SBS. Ksiazyk et al28 conducted a prospective, randomized, enteral balance (enteral retention of formula and nutrients)
crossover, double-blind study of 10 children receiving infant compared with oral bolus feeds. Unfortunately, the sample
formula containing either hydrolyzed or intact protein. size was very small (n = 9), and only 2 of the infants had
Children were trialed on each formula for 30 days (crossover SBS. A more recent randomized crossover study of nutrient
on day 31), and nitrogen balance and lactulose/mannitol absorption by mode of nutrition therapy (tube feeding
excretion ratio (to measure intestinal permeability) were alone, oral feeding alone, combination of tube and oral
evaluated on days 1, 31, and 61 of the study. The researchers feeding) was conducted in adults with SBS (n = 15). Results
reported that the 2 types of formula did not yield a sig- revealed significantly greater absorption of protein, fats, and
nificant difference in intestinal permeability, caloric/energy energy when a tube feeding was used alone or in conjunction
intake, weight gain, or nitrogen balance. However, feeding with oral feeding compared with when oral feeding was used
tolerance was not assessed, and all infants were receiving alone.31 Although continuous feedings may be better tol-
30% of calorie intake from hydrolyzed enteral feeds before erated and improve nutrient absorption, bolus feedings are
beginning the study protocol. In a review of published data desirable as they mimic the typical infant eating pattern and
relevant to feeding strategies in children with SBS, Olieman are therefore more physiological. Bolus feedings allow for
534 Nutrition in Clinical Practice 33(4)

Table 5. Practice Responses to Questions Related to Laboratory/Biochemical Monitoring.

Number of Centers
Survey Question Response Rate Practice Response per Response

Do you use urine sodium to determine 14 of 14 r Yes 10


whether a patient with short bowel r Yes, occasionally 2
syndrome requires sodium r Yes, as well as serum osmolarity 1
supplementation? r No 1

What laboratory tests do you routinely 13 of 14b Panels (daily to monthly): CBC, CMP, Varying responses
follow in patients on PN with short BMP by 13 centers
bowel syndrome or intestinal failure? Vitamins (every 3–12 months): serum
Include macronutrients and retinol, retinol binding protein, RBC
micronutrients and interval for folate, vitamin B-12, methylmalonic
follow-up (eg, daily, weekly, acid, vitamin D, vitamin E
monthly).a Trace elements (every 3–12 months):
serum copper, serum zinc, selenium
(RBC and serum), serum iron, total
iron binding capacity, ferritin
Other (every 3–12 months): magnesium,
phosphorous, serum triglycerides, C-
reactive protein, prothrombin time, in-
ternational normalized ratio, thyroid-
stimulating hormone, serum citrulline,
triene/tetraene ratio

Which of the following parameters do 13 of 14b r Glucose infusion rate 14


you monitor in patients receiving r Serum triglyceride 13
PN?a r Serum cholesterol 4
r Triene/tetraene ratio 12
A. Glucose infusion rate r 5
Nonprotein calorie/nitrogen ratio
B. Serum triglyceride
C. Serum cholesterol
D. Triene/tetraene ratio
E. Nonprotein calorie/nitrogen ratio

BMP, basic metabolic panel; CBC, complete blood count; CMP, comprehensive metabolic panel; PN, parenteral nutrition; RBC, red blood cell.
a Centers may have reported >1 response.
b Question was not answered by 1 center.

periods of fasting, which may prevent hyperinsulinemia32 ence of a colon.29 Pectin, a water soluble dietary fiber,
and promote appropriate bacterial clearance.33 may slow the motility of gastric contents and increase the
Vitamins and trace elements were the supplements most contact of nutrients with the intestinal lumen. However,
often reported to be prescribed. Several dietitians also the dose used (1%–3% solution) is based on a single case
reported use of a carbohydrate and fat modular product, series of 2 formerly premature infants who had small bowel
purees, and pectin/fiber. Vegetable oils such as canola, resections.36 Drenckpohl et al37 documented the benefits of
safflower, or flaxseed can be added to tube feedings to using stage 2 baby food green beans to infant feedings in a
increase caloric intake or supplement essential fatty acids case series. After the addition of green beans to formula (4-
without adding significant volume. Fiber is often added to oz jar of green beans per 8-oz formula), stool consistency
feedings to slow bowel transit time and reduce stool output, improved in all of the infants. The researchers concluded
which assists with nutrient absorption and subsequently that the mixture of 32% soluble and 68% insoluble dietary
growth.34 Soluble fiber is beneficial because it is fermented fiber present in the green beans (2–3 g fiber per 4-oz jar)
into short-chain fatty acids, which can be metabolized by assisted in improving stool consistency.
colonocytes for energy.35 In a 2010 practice paper published While most dietitians did not report using a protocol for
in the Journal of the Academy of Nutrition and Dietetics, initiating oral/motor therapy, most centers do recommend
the authors noted that dietary fiber should be used when therapy when appropriate. Oral aversion is common in
developmentally appropriate (age 4–6 months) in the pres- patients who receive nutrition support for a prolonged
Nucci et al 535

period of time. Consequently, oral motor therapy and early mean of 50 days (range, 11–712 days).16 Deficiencies were
oral feedings are important to the rehabilitation process. mild and resolved within 1–2 months after the number of
These therapies may also provide beneficial support for days of fat infusion were increased. The authors noted
families who become frustrated with lack of progress with the importance of monitoring markers of essential fatty
oral feeds.38 Although most centers did not report an exact acid deficiency during fat minimization and suggested that
time for initiation of complementary oral feedings, some absolute values of linoleic and α-linolenic acid be examined
use criteria to assess readiness or generally stated that they vs the triene/tetraene ratio, which is frequently used.16 Given
would begin oral feeds as soon as possible. The initiation the importance of dietary fat on infant brain development,
of oral feeds as soon as developmentally and medically there is concern about the effect of fat minimization on
appropriate is important to prevent oral aversion.38 The the long-term neurodevelopment of infants with IFALD
first year of life is the most critical period for the de- treated with low-dose IV fat. Few studies have assessed this
velopment of oral feeding skills.33 Oral feeding promotes risk. However, preliminary findings at 1 institution showed
epidermal growth factor release from salivary glands and that infants treated with IV fat emulsion reduction had
increases gastrointestinal secretion of trophic factors.33 mostly normal neurodevelopmental outcomes at ages rang-
Small bolus feedings by mouth (equal to or less than the ing from 2–5 years.40 Another institution recently published
volume tolerated continuously per hour) should be trialed data showing no adverse effect of low-dose IV soy-based
to encourage development of swallowing. Solid food should fat emulsion on neurodevelopment or growth at 2 years of
be introduced at a developmentally appropriate age under age in a cohort of 15 premature infants compared with 15
the guidance of a feeding therapist.12 Unfortunately, no premature infants who received standard dosing.41
clinical studies have been conducted to identify the preferred The use of ω-3 or fish oil–based fat emulsions is associ-
time of initiation of complementary solid foods or the ated with resolution of biochemical markers of IFALD42-44
optimal oral diet for children with SBS.29 Current nutrition but lacks rigorous clinical studies in the pediatric IF pop-
recommendations are subdivided by colon status. A high- ulation. Puder et al45 conducted a trial of fish oil–based
carbohydrate (50%–60% of energy), lower fat (20%–30% fat emulsion in 42 infants with SBS and cholestasis. Of
of energy) diet may be indicated for patients with a colon the 38 patients who survived or were not transplanted, 19
while a higher fat (30%–40% of energy), lower carbohydrate (50%) experienced reversal of cholestasis (direct bilirubin
(40%–50% of energy) diet may be preferred when the colon ࣘ2 mg/dL). The fish oil–based fat emulsions are thought
is absent.5 Complex carbohydrates may be better tolerated to improve liver function due to the anti-inflammatory
than simple sugars, and patients with a colon may need to properties of the ω-3 fatty acids and improved triglyceride
avoid or restrict oxalate intake to decrease the risk of oxalate clearance.42 In addition, they lack the phytosterols of soy-
renal stones.5 based fats, which are thought to be relatively hepatotoxic.42
Several components of nutrition management for chil- However, there are reports of continued liver disease de-
dren at risk for IFALD were consistent among all centers spite resolution of cholestasis,46-48 and similar reductions
and were similar if not the same as those principles used in cholestasis are achieved by restricting traditional soy-
for patients at risk for developing cholestasis. The literature based IV fats. Fish oil–based fat emulsions are not cur-
related to minimization of soy-based fat emulsion (ࣘ1 rently approved by the Food and Drug Administration
g/kg/d) is primarily observational. Although restriction of (FDA) for use in the United States but remain available
IV fat emulsion has been shown to reduce total bilirubin for use under research or compassionate use protocols.
levels in children with IFALD,16 the effect of the accom- Alternatively, Smoflipid (Fresenius Kabi, Bad Homburg,
panying caloric reduction on growth in the IF population Germany), an IV fat emulsion that contains soybean oil,
is unknown.14 Levit et al39 conducted a clinical trial that medium-chain triglycerides, olive oil, and fish oil, is FDA
examined the safety and efficacy in preventing IFALD using approved for use in adults. Although the safety and efficacy
a low-dose soy-based fat emulsion (1 g/kg/d) vs a control of using Smoflipid in pediatric patients have not yet been
dose (ࣈ3 g/kg/d) in preterm infants (n = 136) after 14 established,49 randomized controlled trials have examined
days of PN. The authors reported that after 28 days of Smoflipid vs soy-based fat emulsion with encouraging re-
life, no difference in growth or reduction of cholestasis sults, including decreased serum γ -glutamyl transferase50
was observed between the randomized groups. However, and total bilirubin51 in the Smoflipid groups. In a small
this was not a surgical population requiring prolonged PN retrospective review, conjugated bilirubin decreased within
support, and the determination of cholestasis (ࣙ15% of the 2 weeks after a change to Smoflipid from a soy-based fat
total bilirubin at 28 days of life) differed from the definition emulsion in children with IFALD.52
used in other fat minimization studies (conjugated bilirubin Reducing or removing trace elements from PN solutions
>2 mg/dL).15,16 Essential fatty acid deficiency was observed is a proposed strategy to decrease the risk of IFALD.13 Since
in infants with IFALD who were placed on a soy-based fat copper is eliminated via biliary excretion, parenteral intake
emulsion reduction protocol (1 g/kg/d twice weekly) for a of copper may need to be reduced when IFALD is present,
536 Nutrition in Clinical Practice 33(4)

while increased provision of copper may be necessary in Two case studies and 1 retrospective cohort analysis re-
patients with a jejunostomy or other biliary output.10,53 Of vealed varied use of iodine biomarkers (thyroid-stimulating
note, copper deficiency has been reported with copper-free hormone, T4, serum vs urine iodine) and varied frequency
PN.53 Recent studies examining the practice of reducing of monitoring59-61 in investigating iodine deficiency in their
or removing copper from PN solutions of cholestatic pa- chronic PN pediatric patients. The high variability in labora-
tients have shown that a standard intake of 20 mcg/kg/d tory test selection and monitoring frequency may be reflec-
for infants may be safe, and it is suggested that copper tive of a lack of national guidelines. Our survey has revealed
levels be assessed prior to reducing or removing copper the importance of continual collaboration among centers to
from PN solutions.54-56 Manganese supplementation may establish biochemical monitoring standards, particularly in
not be necessary for children with IFALD as manganese the absence of the large numbers of subjects needed for well-
is a contaminant in PN solutions.38 Therefore, use of executed clinical trials in this subspecialty area of practice.
individual trace element preparations vs a packaged trace Areas in which collaboration may be beneficial include the
element preparation may be the best practice in patients following:
with IFALD.10,38 Careful monitoring of trace element status
when micronutrients are removed from PN is warranted.
r Designating the relevant micronutrients for monitor-
Provision of EN in some form continues to be a sig- ing and the most reliable indicators (eg, erythrocyte
nificant component of the nutrition support practice for vs plasma, serum vitamin B-12 or serum methyl-
patients with IFALD. Expert opinion based on experience malonic acid or both)
and available research seems to focus on advancing enteral
r Establishing the monitoring frequency and indi-
feedings to promote intestinal adaptation and weaning PN cations for any protocol variance (eg, intestinal
support while maintaining proportional growth. EN is be- anatomy, liver function, PN dependent vs nondepen-
lieved to promote intestinal adaptation by stimulating mu- dent)
cosal hyperplasia, gastrointestinal hormone secretion, and
r Facilitating monitoring across the inpatient/home
pancreaticobiliary secretions.1 These mechanisms may also health spectrum of care, particularly in regard to
protect against the development of IFALD by improving the management of parenteral trace element product
biliary flow and decreasing the risk of bacterial overgrowth, shortages62
which may further inhibit bile secretions.23 Most experts
The design of the practice survey has the advantage of
agree on starting EN as early as medically feasible and
permitting collection of data that might not be captured in
advancing slowly while taking advantage of available strate-
a closed-ended questionnaire. However, this type of study
gies as previously discussed, including continuous feeds,
survey also has several limitations. Questions are subject
refeeding of ostomy output into distal mucous fistula, and
to individual interpretation (eg, what supplements do you
fiber supplementation.13,22,38
typically recommend for patients with IF?), and specific
Our survey revealed considerable variability in labo-
components may not have been reported due to the open-
ratory test selection and monitoring frequency among
ended format (eg, what laboratory tests do you routinely
pediatric IF practitioners. Most dietitians responding to
follow in patients with IF on PN?).
the survey indicated the use of specific laboratory panels
at defined intervals to address the reported high preva-
lence of micronutrient deficiencies in patients with pedi-
Conclusion
atric IF.4,19,21,57 The laboratory monitoring practices of EN and PN management strategies in the presence and
most centers are generally reflective of the evidence-based absence of cholestasis are relatively consistent among U.S.
literature. Most centers routinely monitor urine sodium practitioners caring for children with IF. However, labora-
due to the known association of growth failure with a tory monitoring practices vary widely among centers. Al-
low urine sodium level, particularly in those patients with though only half of the dietitians surveyed function as part
jejunostomies, ileostomies, and colostomies.58 Many centers of an organized IR program, interdisciplinary management
have laboratory monitoring protocols, similar to published and early referral of children dependent on PN to an IR pro-
recommendations from children’s hospitals with established gram can result in positive outcomes, including cessation of
IR centers, to address the high risk of micronutrient de- PN support, accelerated growth, and improved survival.10,14
ficiency in their patient populations.12,57 Although infre- Multicenter clinical trials to identify best nutrition practices
quently mentioned by survey respondents, the monitoring and to establish laboratory monitoring guidelines across
of iodine status in pediatric IF patients has garnered much the spectrum of care for children with IF would be opti-
attention in the literature since the submission of survey mal. Collaboration among centers should be encouraged
results, particularly in light of numerous studies demon- considering the difficulty of conducting large-scale clinical
strating the potential increased risk of iodine deficiency in trials in this population. Collaborative initiatives such as the
pediatric patients requiring long-term PN supplementation. Pediatric Intestinal Failure Section of ASPEN appear well
Nucci et al 537

positioned to pragmatically address the myriad variables 14. Wales PW, Allen N, Worthington P, George D, Compher C, the
affecting micronutrient status in pediatric IF patients and American Society for Parenteral and Enteral Nutrition, Teitelbaum
D. A.S.P.E.N. clinical guidelines: support of pediatric patients with
to issue evidence-based laboratory monitoring guidelines.
intestinal failure at risk of parenteral nutrition–associated liver disease.
Moreover, the section can foster collaboration and take an JPEN J Parenter Enteral Nutr. 2014;38:538-557.
active role in conducting clinical research to determine best 15. Sanchez SE, Braun LP, Mercer LD, Sherrill M, Stevens J, Javid PJ.
practices for enhancing intestinal adaptation and preventing The effect of lipid restriction on the prevention of parenteral nutrition–
or delaying nutrition support-related complications. associated cholestasis in surgical infants. J Pediatr Surg. 2013;48:573-
578.
16. Cober MP, Killu G, Brattain A, Welch KB, Kunisaki SM, Teit-
Statement of Authorship elbaum DH. Intravenous fat emulsions reduction for patients with
A. M. Nucci contributed to the conception and design of the parenteral nutrition–associated liver disease. J Pediatr. 2012;160:
research, as well as the acquisition, analysis, and interpretation 421-427.
17. Khan FA, Fisher JG, Sparks EA, et al. Preservation of biochem-
of the data. K. Ellsworth, A. Michalski, E. Nagel, and J.
ical liver function with low-dose soy-based lipids in children with
Wessel equally contributed to the design of the research. K.
intestinal failure–associated liver disease. J Pediatr Gastroenterol Nutr.
Ellsworth, A. Michalski, and E. Nagel contributed to the 2015;60:375-377.
analysis and interpretation of the data. J. Wessel contributed to 18. Nucci A, Burns RC, Armah T, et al. Interdisciplinary management
the interpretation of the data. A. M. Nucci, K. Ellsworth, A. of pediatric intestinal failure: a 10-year review of rehabilitation and
Michalski, and E. Nagel drafted the manuscript. All authors transplantation. J Gastrointest Surg. 2008;12:429-436.
critically revised the manuscript, gave final approval of the 19. Ubesie AC, Kocoshis SA, Mezoff AG, Henderson CJ, Helmrath MA,
manuscript, and agree to be fully accountable for ensuring the Cole CR. Multiple micronutrient deficiencies among patients with
integrity and accuracy of the work. intestinal failure during and after transition to enteral nutrition. J
Pediatr. 2013;163:1692-1696.
20. Yang CJ, Duro D, Zurakowski D, Lee M, Jaksic T, Duggan C. High
References prevalence of multiple deficiencies in children with intestinal failure: a
1. DiBaise JK, Young RJ, Vanderhoof JA. Intestinal rehabilitation and longitudinal study. J Pediatr. 2011;159:39-44.
the short bowel syndrome: part 1. Am J Gastroenterol. 2004;99:1386- 21. Gonzalez HF, Perez NB, Malpeli A, Martinez MI, Buono BD, Viteri
1395. FE. Nutrition and immunological status in long-term follow up of
2. Warner BW, Ziegler MM. Management of the short bowel syndrome children with short bowel syndrome. JPEN J Parenter Enteral Nutr.
in the pediatric population. Pediatr Clin North Am. 1993;40:1335-1350. 2005;29:186-191.
3. Barksdale EM, Stanford A. The surgical management of short bowel 22. Squires RH, Iyer KR. Intestinal failure. In: Corkins MR, ed. The
syndrome. Curr Gastroenterol Rep. 2002;4:229-237. ASPEN Pediatric Nutrition Support Core Curriculum. 2nd ed. Silver
4. Wu J, Tang Q, Feng Y, et al. Nutrition assessment in children with short Spring, MD: American Society for Parenteral and Enteral Nutrition;
bowel syndrome weaned off parenteral nutrition: a long-term follow-up 2015:427-440.
study. J Pediatr Surg. 2007;42:1372-1376. 23. Slicker J, Vermilyea S. Pediatric parenteral nutrition: putting the
5. DiBaise JK, Young RJ, Vanderhoof JA. Intestinal rehabilitation and microscope on macronutrients and micronutrients. Nutr Clin Pract.
the short bowel syndrome: Part 2. Am J Gastroenterol. 2004;99:1823- 2009;24:481-486.
1832. 24. Werlin SL, Wyatt D, Camitta B. Effect of abrupt discontinuation
6. Duro D, Kamin D, Duggan C. Overview of pediatric short bowel of high glucose infusion rates during parenteral nutrition. J Pediatr.
syndrome. J Pediatr Gastroenterol Nutr. 2008;47:S33-S36. 1994;124:441-444.
7. Vanderhoof JA, Young RJ, Thompson JS. New and emerging thera- 25. Squires RH, Duggan C, Teitelbaum DH, et al. Natural history of
pies for short bowel syndrome in children. Pediatr Drugs. 2003;5(8): pediatric intestinal failure: initial report from the pediatric intestinal
525-531. failure consortium. J Pediatr. 2012;161:723-728.
8. Goulet O, Sauvat F. Short bowel syndrome and intestinal transplanta- 26. Andorsky D, Lund D, Lillehei C, et al. Nutritional and other postop-
tion in children. Curr Opin Clin Nutr Metab Care. 2006;9:304-313. erative management of neonates with short bowel syndrome correlates
9. Kerner JA, ed. Manual of Pediatric Parenteral Nutrition. New York, with clinical outcomes. J Pediatr. 2001;139:27-33.
NY: John Wiley; 1983. 27. Vanderhoof J, Langnas A. Short-bowel syndrome in children and
10. Greene HL, Hambidge KM, Schanler R, Tsang RC. Guidelines for the adults. Gastroenterology. 1997;113:1767-1778.
use of vitamins, trace elements, calcium, magnesium, and phosphorus 28. Ksiazyk J, Piena M, Keirkus J, Lyszkowska M. Hydrolyzed versus
in infants and children receiving total parenteral nutrition: report of nonhydrolyzed protein diet in short bowel syndrome in children. J
the Subcommittee on Pediatric Parenteral Nutrient Requirements from Pediatr Gastroenterol Nutr. 2002;35:615-618.
the Committee on Clinical Practice Issues of the American Society for 29. Olieman J, Penning C, Ijsselstijn H. Enteral nutrition in children with
Clinical Nutrition. Am J Clin Nutr. 1988;48:1324-1342. short-bowel syndrome: current evidence and recommendations for the
11. Corkins MR, ed. The ASPEN Pediatric Nutrition Support Core Cur- clinician. J Am Diet Assoc. 2010;110(3):420-426.
riculum. 2nd ed. Silver Spring, MD: American Society for Parenteral 30. Parker P, Stroop S, Greene H. A controlled comparison of continuous
and Enteral Nutrition; 2015. intermittent feeding in the treatment of infants with intestinal disease.
12. Cole R, Kocoshis SA. Nutrition management of infants with sur- J Pediatr. 1981;99:360-364.
gical short bowel syndrome and intestinal failure. Nutr Clin Pract. 31. Joly F, Dray X, Corcos O, Barbot L, Kapel N, Messing B. Tube feed-
2013;28:421-428. ing improves intestinal absorption in short bowel syndrome patients.
13. Mayer O, Kerner JA. Management of short bowel syndrome in Gastroenterology. 2009;136:824-831.
postoperative very low birth weight infants. Semin Fetal Neonatal Med. 32. Batra A, Beattie RM. Management of short bowel syndrome in infancy.
2017;22:49-56. Early Hum Dev. 2013;89:899-904.
538 Nutrition in Clinical Practice 33(4)

33. Goulet O, Olieman J, Ksiazyk J. Neonatal short bowel syndrome as 49. Food and Drug Administration. SMOFLIPID. https://www.acce
a model of intestinal failure: physiological background for enteral ssdata.fda.gov/drugsatfda_docs/label/2016/207648lbl.pdf. Published
feeding. Clin Nutr. 2013;32:162-171. May, 2016. Accessed January 27, 2017.
34. Batra A, Beattie RM. Management of short bowel syndrome in infancy. 50. Tomsits E, Pataki M, Tölgyesi A, Fekete G, Rischak K, Szollár
Early Hum Dev. 2013;89:899-904. L. Safety and efficacy of a lipid emulsion containing a mixture of
35. Nordgaard I, Hasen B, Mortensen P. Importance of colonic support soybean oil, medium-chain triglycerides, olive oil, and fish oil: a
for energy absorption as small-bowel failure proceeds. Am J Clin Nutr. randomised, double-blind clinical trial in premature infants requir-
1996;65:222-231. ing parenteral nutrition. J Pediatr Gastroenterol Nutr. 2010;51:514-
36. Hawkins R, Henry B, Gottschalk ME. Pectin supplemented enteral 521.
feedings in the treatment of short bowel syndrome in two infants. J Am 51. Goulet O, Antébi H, Wolf C, et al. A new intravenous fat emulsion
Diet Assoc. 1995;95:A-28. containing soybean oil, medium-chain triglycerides, olive oil, and fish
37. Drenckpohl D, Hocker J, Shareef M, Vegunta R, Colgan C. Adding oil: a single-center, double-blind randomized study on efficacy and
dietary green beans resolves the diarrhea associated with bowel surgery safety in pediatric patients receiving home parenteral nutrition. JPEN
in neonates: a case study. Nutr Clin Pract. 2005;20:674-677. J Parenter Enteral Nutr. 2010;34:485-495.
38. Wessel J, Kocoshis S. Nutritional management of infants with short 52. Attard MI, Patel N, Simpson J. Change from intralipid to SMOF lipid
bowel syndrome. Semin Perinatol. 2007;31:104-111. is associated with improved liver function in infants with PN associated
39. Levit OL, Calkins KL, Gibson C, et al. Low-dose intravenous soybean liver disease. Arch Dis Child. 2012;97:A54-A55.
oil emulsion for prevention of cholestasis in preterm neonates. JPEN J 53. Hurwitz M, Garcia MG, Poole RL, Kerner JA. Copper deficiency
Parenter Enteral Nutr. 2016;40:374-382. during parenteral nutrition: a report of four pediatric cases. Nutr Clin
40. Blackmer AB, Warschausky S, Siddiqui S, et al. Preliminary find- Pract. 2004;19:305-308.
ings of long-term neurodevelopment outcomes of infants treated 54. Frem J, Sarson Y, Sternberg T, Cole CR. Copper supplementation
with intravenous fat emulsion reduction for the management of par- of cholestatic infants. J Pediatr Gastroenterol Nutr. 2010;50:650-
enteral nutrition–associated cholestasis. JPEN J Parenter Enteral Nutr. 654.
2015;39:34-46. 55. Corkins MR, Martin VA, Szeszycki EE. Copper levels in cholestatic
41. Ong ML, Purdy IB, Levit OL, et al. Two-year neurodevelopment infants on parenteral nutrition. JPEN J Parenter Enteral Nutr.
and growth outcomes for preterm neonates who received low-dose 2013;37:92-96.
intravenous soybean oil [published online October 21, 2016]. JPEN J 56. Blackmer AB, Bailey E. Management of copper deficiency in
Parenter Enteral Nutr. cholestatic infants: review of the literature and a case series. Nutr Clin
42. Gura K, Duggan C, Puder M, et al. Reversal of parenteral nutrition– Pract. 2013;28:75-86.
associated liver disease in two infants with short bowel syndrome using 57. Chi-fu JY, Duro D, Zurakowski D, Lee M, Jaksic T, Duggan C.
parenteral fish oil: implications for future management. Pediatrics. High prevalence of multiple micronutrient deficiencies in children
2006;118:e197-e201. with intestinal failure: a longitudinal study. J Pediatr. 2011;159:
43. Gura K, Lee S, Valim C, et al. Safety and efficacy of a fish-oil-based 39-44.
fat emulsion in the treatment of parenteral nutrition–associated liver 58. Mansour F, Petersen D, De Coppi P, Eaton S. Effect of sodium
disease. Pediatrics. 2008;121:e678-e686. deficiency on growth of surgical infants: a retrospective observational
44. Diamond IR, Sterescu A, Pencharz PB, et al. Changing the paradigm: study. Pediatr Surg Int. 2014;30:1279-1284.
omegaven for the treatment of liver failure in pediatric short bowel 59. Mortensen M, Williamson N, David C, Hsu EK, Javid PJ, Horslen
syndrome. J Pediatr Gastroenterol Nutr. 2009;48:209-215. S. Iodine deficiency in a parenteral nutrition–dependent adolescent
45. Puder M, Valim C, Meisel JA, et al. Parenteral fish oil improves with intestinal pseudo-obstruction. JPEN J Parenter Enteral Nutr.
outcomes in patients with parenteral nutrition associated liver injury. 2016;40:730-733.
Ann Surg. 2009;250:395-402. 60. Golekoh MC, Cole CR, Jones NHY. Severe hypothyroidism from
46. Soden JS, Lovell MA, Brown K, Partrick DA, Sokol RJ. Failure of iodine deficiency associated with parenteral nutrition. JPEN J Parenter
resolution of portal fibrosis during omega-3 fatty acid lipid emulsion Enteral Nutr. 2016;40:1191-1193.
therapy in two patients with irreversible intestinal failure. J Pediatr. 61. Santoro JD, Nespor C, Poole RL, Kerner JA. Iodine supplementation
2010;156:327-331. for pediatric patients receiving long-term parenteral nutrition. Nutr
47. Mercer DF, Hobson BD, Fischer RT, et al. Hepatic fibrosis persists and Clin Pract. 2016;31:245-249.
progresses despite biochemical improvement in children treated with in- 62. A.S.P.E.N. Clinical Practice Committee Shortage Subcommittee. Par-
travenous fish oil emulsion. J Pediatr Gastroenterol Nutr. 2013;56:364- enteral nutrition trace elements product shortage considerations. http://
369. www.nutritioncare.org/News/General_News/Parenteral_Nutrition_
48. Nandivada P, Chang MI, Potemkin AK, et al. The natural history Trace_Element_Product_Shortage_Considerations/. Updated July 26,
of cirrhosis from parenteral nutrition–associated liver disease after 2016. Accessed January 13, 2017.
resolution of cholestasis with parenteral fish oil therapy. Ann Surg.
2015;261:172-179.
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 4
Soluble Fiber Use in Pediatric Short Bowel Syndrome: August 2018 539–544

C 2018 American Society for

A Survey on Prevailing Practices Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10089
wileyonlinelibrary.com

Meredith Linley Harvie, MD1 ; Margaret Alyssa Tucker Norris, MS, RD, LDN,
CLC2 ; and Wednesday Marie A. Sevilla, MD, MPH, CNSC3

Abstract
Background: In pediatric short bowel syndrome (SBS), adding fiber to enteral feedings is 1 treatment method to manage increased
stool output. However, there are no standardized recommendations on the use of fiber in this setting, including type, dosage,
titration strategies, etc. Objective: The aim of this study is to determine current prevailing practices on the use of fiber in
the treatment of chronic high stool output in the pediatric SBS population. Methods: An anonymous electronic survey with 13
questions was sent through health professional electronic mailing lists. The survey was completed by healthcare professionals
including physicians (primary care, subspecialists, and surgeons), nurse practitioners, and registered dietitians. Results: A total of
94 responses were received. The most common supplemental fiber used was pectin (62.8%). The 2 major factors considered when
initiating fiber therapy were consistency of stool (74.5%) and volume of stool output (85.1%). The major factor that determined
discontinuation of fiber was abdominal distention (67%). A majority of providers waited 2 weeks or less to see improvement
following fiber initiation before discontinuing it. Conclusions: The goal of the survey was to gather more information with regard
to fiber use in the management of SBS patients. The data collected can be used to provide future direction on determining best
practices for fiber use in SBS patients. (Nutr Clin Pract. 2018;33:539–544)

Keywords
child; dietary fiber; enteral nutrition; intestinal rehabilitation; short bowel syndrome

Introduction time. On the contrary, viscous-forming or gel-forming


fiber dissolves in water, forms a viscous gel, and is therefore
The primary goal of management in pediatric short bowel able to slow the digestion of complex nutrients by increasing
syndrome (SBS) is to reach enteral autonomy through intestinal transit time.3 Readily fermented fiber may provide
intestinal rehabilitation. Enteral autonomy is clinically de- a “prebiotic” effect, leading to the formation of short-chain
fined as the patient’s ability to achieve growth and main- fatty acids (SCFAs) and reabsorption of sodium and
tain hydration with caloric intake solely administered to water. Non-fermented fiber creates a “stool normalizing”
the gastrointestinal tract without the aid of parenteral effect by staying gelled throughout large bowel transit, caus-
nutrition. One of the barriers to enteral autonomy is the ing bulking of loose stools.3 However, it does not provide
quick transit time of stools, which can lead to poor
nutrient absorption, electrolyte abnormalities, and poor
growth.1,2 One accepted treatment in the management of From the 1 University of Tennessee Health Sciences Center,
loose stools in pediatric SBS is addition of fiber into enteral Department of Pediatrics, Memphis, Tennessee, USA; 2 University of
or oral formula because the standard formula contains little Tennessee Le Bonheur Children’s Hospital, Clinical Nutrition
Department, Memphis, Tennessee, USA; and the 3 Children’s
to none. Hospital of Pittsburgh, Division of Pediatric Gastroenterology,
There are 2 main types of fiber: insoluble and soluble. Hepatology and Nutrition, Pittsburgh, Pennsylvania, USA.
Insoluble fiber does not dissolve in water and can have a Financial disclosure: None declared.
laxative effect, making stooling worse. Soluble fiber can
Conflicts of interest: None declared.
improve stool consistency and is therefore the most readily
used. Soluble fiber can be subdivided based on whether This article originally appeared online on May 16, 2018.
it is viscous-forming or gel-forming, readily fermented, Corresponding Author:
or a combination of both (Table 1).3 Each type has Wednesday Marie A. Sevilla, MD, MPH, CNSC, Children’s Hospital
of Pittsburgh, Division of Pediatric Gastroenterology, Hepatology
its own advantages and disadvantages. A non-viscous and Nutrition, 4401 Penn Avenue, 6th Floor Faculty Pavilion,
fiber dissolves in water but does not increase viscosity Pittsburgh, PA 15224.
of chyme, having little to no effect on luminal transit Email: wednesday.sevilla2@chp.edu
540 Nutrition in Clinical Practice 33(4)

Table 1. Classification of Soluble Fibers Commonly Used. Table 2. Regions Represented by Respondents.

Non-Viscous, Viscous, Viscous, Readily Number of


Readily Fermented Non-Fermented Fermented Region States Respondents (n = 94)

Wheat dextrin Methylcellulose Pectin West WA, OR, CA, NV, UT, n = 14
Inulin Psyllium Guar gum AZ, NM, CO, WY
Oligosaccharides β-glucan Midwest ND, SD, NE, KS, MN, n = 18
IA, MO, IL, WI, IN,
OH, MI
South TX, OK, AR, LA, MS, n = 39
the potential positive benefits achieved in the fermentation AL, GA, FL, SC, NC,
process. TN, KY, WV, VA, MD
While different studies have chosen viscous-forming or Northeast ME, NH, VT, NY, MA, n = 14
gel-forming soluble fiber, there are currently no standard- RI, CT, NJ, PA
Outside Australia, Canada, Oman, n=9
ized recommendations for the use of fiber in pediatric SBS.
the U.S. Mexico
There are no studies that examine and compare the effects
of different types of fiber, initiation and maintenance doses,
titration strategies, or the accuracy of monitoring for clini-
cal response. The aim of this study is to determine current
prevailing practices on the use of fiber in the treatment of
chronic high stool output in the pediatric SBS population.

Methods
Pediatric health professionals, in multiple disciplines man-
aging pediatric patients with SBS, were surveyed. This
population was chosen to gain knowledge on details of the
use of fiber, the goal being to provide future directions for
determining best practices and future research strategies. An
anonymous electronic survey with 13 questions was sent
through health professional electronic mailing lists (see
Appendix). The survey was completed by healthcare pro-
fessionals including physicians (primary care, subspecialists,
and surgeons), nurse practitioners, and registered dieti-
tians. The electronic survey was chosen to reach a wide
variety of healthcare professionals as well as multiple re-
gions across the United States and the globe. Survey results
Figure 1. Percentage of healthcare professionals who
were then summarized. Survey instrument development and responded to survey.
data collection were completed with a statistical program
(Qualtrics, Provo, UT). The data were analyzed through
analytic software (SPSS, IBM, Armonk, NY). The survey outside the United States (Table 2). The majority of respon-
study was approved by the University of Tennessee Health dents were dietitians. Other medical professionals included
Science Center Institutional Review Board. physicians, nurse practitioners/physician assistants, and sur-
geons (Figure 1). A large percentage, 69% of respondents,
Results practiced as part of an intestinal rehabilitation team or
program, most of whom (40.9%) reported an average annual
Respondents SBS patient census of 10–50 patients.
A total of 94 responses were received. There was a good
Type of Fiber
representation of health professionals from various regions
of the United States, with 9 respondents from other coun- Based on the survey, the most common supplemental fiber
tries including Australia, Canada, Mexico, and Oman. The used in the treatment of loose stools in pediatric short bowel
regions included South, Midwest, Northeast, and West. Of patients was pectin (62.8%). Pectin was the most commonly
the 94 responses received, 40% percent of the participants used form of fiber in the Southern and Midwestern states, as
were from the South, 20% from the Midwest, 15% from well as outside the United States. In the survey, an op-
the Northeast, 15% from the West, and 10% were from tion for additional comments was provided. Some responses
Harvie et al 541

failure occurs due to the limited surface area and altered


gut motility. Intestinal adaptation is key to achieving enteral
autonomy. For intestinal adaptation to occur, the gut must
be stimulated by enteral nutrition to enhance digestive
and absorptive capacity. By slowing down intestinal transit
time, this will presumably allow more time for absorption.
Therefore, medical management of increased stool out-
put will occasionally include the use of antimotility agents
such as diphenoxylate-atropine and/or loperamide. These
medications inhibit intestinal peristalsis by slowing tran-
Figure 2. Most common practices on the use of fiber in
pediatric SBS. SBS, short bowel syndrome. sit time,4 but they are not without side effects. Fiber, while its
effects may not be as immediate, is used as an alternative or
adjunct to these medications. Some side effects of fiber in-
included supplemental fiber that was administered in the clude abdominal distention, bloating, and pain. Depending
form of green beans, which contain pectin, prepared as baby on the type of fiber, these side effects can range in intensity.
food. Other pectin-rich foods, such as sweet potatoes and Inulin, a low-digestible carbohydrate, when consumed in
bananas, were also mentioned. A small proportion admin- large quantities is associated with increased gastrointesti-
istered wheat dextrin (35.1%) and guar gum (14.9%). Wheat nal intolerance. On the contrary, wheat dextrin is much
dextrin was the most commonly used form of fiber in the more tolerable in high quantities (30–45 g/day).5 These side
Western and Northeastern states. effects can sometimes be minimized by starting with a small
amount and increasing slowly. There are only few subsets of
Amount of Fiber people who cannot tolerate fiber at all.
The addition of dietary fiber to pediatric formula for
Respondents who used pectin (52.4%) reported the max- non-SBS has shown some potential benefits including de-
imum percentage content of added pectin to feeds was creasing stool output. Khoshoo et al6 performed a prospec-
3%. About 6% of the respondents used a maximum of tive, double-blind, randomized trial comparing tolerance of
4% pectin added to feeds. Regarding guar gum or wheat a peptide-based formula to a peptide-based formula with
dextrin, titration of supplementation was administered a insoluble and soluble fiber in children. The trial showed
teaspoon (ࣈ2g; 20.2%); a tablespoon (ࣈ6g; 8.3%); and improvement of stool consistency in children receiving the
a gram (9.5%) at a time. It is uncertain how the healthcare formula with fiber. There are case studies that report the
provider determined their titrations. use of soluble fiber (pectin and guar gum) to minimize
chronic diarrhea in the pediatric patient.1,7-10 Alam et al8
Initiating and Discontinuing Fiber performed a double-blind, randomized, controlled trial that
showed a statistically significant reduction in duration of
In determining when to initiate fiber therapy, the 2 major
diarrhea in children who received supplementation with sol-
factors were consistency of stool (74.5%) and volume of
uble fiber in the acute setting.8 The aforementioned studies
stool output (85.1%). For premature infants, majority of the
were not conducted in children with SBS. It remains to
respondents (53.4%) did not initiate fiber until the patient
be examined through randomized controlled trials whether
was >40 weeks corrected age. There was not a majority
fiber should be added to the diets of SBS patients. While
consensus on length of time the healthcare provider waited
there are no clinical studies to show the safety and efficacy
to see if there was an improvement with fiber therapy, but
of fiber use in children with SBS, many health professionals
most did not continue >2 weeks if there was no improve-
will use fiber based on experience rather than clinical
ment. The major factor that determined discontinuation of
evidence.11 Current recommendations from the American
fiber was abdominal distention (67%), in addition to in-
Dietetic Association, according to Olieman et al, are to
creased emesis (43%) and bloody stools (38%). Others had
introduce soluble fiber around 4 to 6 months of age in
to discontinue due to increased stool production (37%) or
the SBS patient with an intact colon. The paper mentions
decreased stool production (33%).
pectin, green beans, and guar gum as sources of soluble
A summary of the most common practices on the use of
fiber used, but it did not give details to why those fibers
fiber in pediatric SBS is listed in Figure 2.
were chosen. It also mentions the limited evidence on the
effect of soluble fibers on enteral feeding tolerance, as
Discussion previously mentioned.12 The aim of our survey was to
A major complication of SBS is intestinal failure, which gather information on the current practice of soluble fiber
is the inability to maintain fluid, electrolyte, and nutrient use in treatment of increased stool output in the setting of
balance despite adequate intake. For SBS patients, intestinal SBS. A majority of respondents (62.77%) selected pectin as
542 Nutrition in Clinical Practice 33(4)

their source of soluble fiber. Added to this is the use of relatively healthy individuals (without intestinal pathology)
pectin-rich foods such as green beans, sweet potatoes and may see benefits of soluble fiber at a faster rate, given the
bananas. There is evidence of improved adaptation of the complexity of anatomy and microbiome of the SBS patient,
small intestine and colon in animal models of SBS with the it is unknown how quickly they should respond to fiber
addition of pectin to an elemental diet.11 initiation.
SBS patients are known to have gut microbiota dys- As mentioned previously, there is no standardization of
biosis secondary to small bowel resection and interrup- how much fiber supplementation to use in SBS patients. As
tion of normal developing gut microflora. A majority we learn more about the microbiome of SBS patients, the
of patients are diagnosed clinically with small bacterial dosage may become more individualized. Although it is
overgrowth as an effect of dysbiosis. Treatment often re- known that fiber and prebiotics can affect the intestinal
quires multiple courses of antibiotics, further disrupting microflora, the individual responses can vary. There are
and altering their gut microbiome.13 While soluble fiber some who respond well to certain fibers and some who do
has been postulated to increase intestinal transit time, more not respond at all. This could be secondary to individual
recent studies have noted certain fibers to act as a prebi- genetics, unique microflora, inadequate dosages, and/or lack
otic with the capacity to alter the gut microbiota. A pre- of bacteria to ferment the specific fiber chosen.14 Two stud-
biotic is fermented by intestinal microflora stimulating the ies showed that at lower doses of fructooligosaccharides and
growth of more favorable intestinal bacteria. This increase galactooligosaccarides (2.5 g/day), there was no significant
in bacterial mass leads to increase in fecal mass (stool increase in Bifidobacterium spp (produces SCFAs as end
bulking). The soluble fibers that have prebiotic proper- products from fermentation); however, when doses were
ties include wheat dextrin, pectin, gums, and inulin.5 The increased (10g/day) there was significant increase in Bifi-
fermentation of these fibers leads to production of SCFAs, dobacterium spp.18,19 Based on the data by region, there is
which are thought to be essential in the metabolic activity of consistency among regions in terms of type of fiber used,
colonocytes.14 Piper et al13 performed a study which con- but no consensus across the United States. It is possible that
cluded that the microbiome of healthy children contained people in certain regions have more similar microbiomes
an abundance of anti-inflammatory commensal bacteria, and, therefore, tolerate and/or benefit from certain types of
which produce SCFAs, compared with SBS patients. SCFAs soluble fiber differently.
help the intestinal tract thrive and are considered to play
a key role in immune function. By lowering luminal and Limitations of Current Study
fecal pH, SCFAs impede the growth of potentially harmful
The goal of the survey was to gather more information
bacteria, such as certain Clostridia species.5 Animal studies
regarding fiber use in SBS patients. We did not explore the
show a positive correlation between increased SCFAs and
rationale of choosing 1 type of fiber vs another. The survey
an increased immune response. Rats provided a diet with
was mostly focused on when and why providers initiated
fructooligosaccarides (soluble fiber) continuously for more
fiber therapy and which type was used. It would be helpful to
than a week had increased salivary IgA and SCFAs.15 Other
know why certain fiber was chosen vs another, for example,
studies mirrored these findings as well.16 These results em-
availability, cost, or training experience. The survey did not
phasize the potential benefits of providing fiber in pediatric
provide clarity on whether fiber was used in combination
patients with intestinal failure.
with other therapies to help with significant stool output
From our results, it appears the majority of health
or overall growth. The survey did not indicate whether
professionals surveyed (65%) will wait 2 weeks or less to
fiber therapy improved or resolved the patients’ loose stools.
see improvement with fiber supplementation. According
While a variety of healthcare professionals responded to
to Limketkai et al, intestinal adaptation can occur during 1
the survey, the majority of respondents were dietitians and
to 2 years following bowel resection;4 therefore, the trial of
included very few surgeons. Ideally, in future studies, there
fiber may need to be longer before pursuing other interven-
should be more of a balance in healthcare professionals to
tions. Nakao et al17 completed a study that showed adults
help formulate the best practice.
with chronic watery stools can achieve normal stooling pat-
terns after soluble fiber was given for ࣙ4 weeks. The study
also mentioned that after the administration of soluble
Future Research
fiber, there was a slight increase in iron, copper, and zinc More studies are needed to demonstrate the clinical effec-
levels compared with pretreatment values. It is possible that tiveness of fiber administration in pediatric SBS. Studies
the 2-week period of observation after commencement of exploring the type, dose, and response to fiber therapy
treatment is largely driven by the adverse effects of supple- should be completed. The preferred study would be a
menting with fiber. These adverse effects include symptoms double-blind, randomized, controlled trial. Animal models
of intolerance such as increased stool output, leading to can also be used in regard to effectiveness of fiber in
weight loss and the perception of treatment failure. While SBS subjects. The introduction of soluble fiber has shown
Harvie et al 543

to increase absorption of key elements such as calcium  Do not use guar gum/wheat dextrin (5)
and iron. Carvalho et al20 performed a controlled study  other (6)
in rats that showed improvement of iron deficiency on a
diet containing soluble fiber. Prebiotic fibers, as previously Q3 If you use pectin, what is the maximum percentage you
mentioned, have been shown to improve calcium absorption use?
in both humans and rats,21 including a study showing
increased calcium absorption in pubertal adolescents fol- ◦ 1% (1)
lowing soluble fiber diet.22 While these studies were per- ◦ 2% (2)
formed in subjects without intestinal pathology, it opens ◦ 3% (3)
doors for further research involving children with intestinal ◦ 4% (4)
failure. ◦ Do not use pectin (5)
Research involving the microbiome has become a recent ◦ other (6)
area of interest in gastroenterology. Collecting more data on
the microbiomes of SBS patients would help develop more Q4 For which patients do you provide soluble fiber? Please
targeted therapies for these patients. Most studies that were select all that apply.
reviewed did not specifically mention whether the subjects
had an intact colon. Correlations with remnant anatomy (ie,  Those who have a colon (1)
length of remaining small bowel and large bowel, absence  Those without a colon (2)
of ileocecal valve), type of enteral feed, and frequency are  Both A and B (3)
other complications of SBS that should be studied.  Do not use pectin or guar gum/wheat dextrin (4)

Statement of Authorship Q5 How do you decide when to start supplemental fiber?


Please select all that apply.
W. Sevilla contributed to the conception and design of the
research; A. Norris contributed to the design of the research;
 Volume of stool output (1)
W. Sevilla and A. Norris contributed to the acquisition and
analysis of the data; W. Sevilla and M. L. Harvie equally  Age (2)
contributed to the interpretation of the data; M. L. Harvie  Weight (3)
drafted and critically revised the manuscript; A. Norris drafted  Volume of feeds (4)
the manuscript; and W. Sevilla critically revised the manuscript.  Consistency of stool (5)
All authors agree to be accountable for all aspects of work  Do not use soluble fiber (6)
ensuring integrity and accuracy, and they approved the final  Other (7)
manuscript.
Q6 How early do you begin soluble fiber?

Appendix ◦ <34 weeks gestational age (1)


◦ Between 34 and 40 weeks gestational age (2)
Survey Questions and Answer Choices
◦ >40 weeks gestational age (3)
◦ Other (4)
Fiber Use in Pediatric Short Bowel Patients Questionnaire
◦ Do not use soluble fiber (5)
Q1 What do you use for supplemental fiber in the treatment
Q7 How do you administer fiber? Please select all that
of loose stools (ie, slowing transit time)? Please select all that
apply.
apply.

 Pectin (1)  Mixed with feeds (1)


 Guar gum (ie, Nutrisource) (2)  Seperate from feeds (2)
 Wheat dextrin (ie, Benefiber) (3)  Other (3)
 Other (4)
Q8 Why do you stop supplemental fiber?
Q2 If you use guar gum or wheat dextrin, how do you titrate
up/down? Please select all that apply.  Increased stool output (1)
 Decreased stool output (2)
 By 1 tablespoon (1)  Increased emesis (3)
 By 1 gram (2)  Abdominal distention (4)
 By 1 teaspoon (3)  Bloody stools (5)
 Do not titrate (4)  Other (6)
544 Nutrition in Clinical Practice 33(4)

Q9 How long do you wait to see improvements once 3. McRorie J. Evidence-based approach to fiber supplements and clini-
initiating soluble fiber? cally meaningful health benefits, part 1. Nutr Today 2015; 50(2):82-89.
4. Limketkai B, Parian A, Shah N, Colombel J. Short bowel syn-
◦ 2–5 days (1) drome and intestinal failure in Crohnʼs disease. Inflamm Bowel Dis
2016;22(5):1209-1218.
◦ 1–2 weeks (2)
5. Slavin J. Fiber and prebiotics: mechanisms and health benefits. Nutri-
◦ 2–4 weeks (3) ents 2013;5(4):1417-1435.
◦ >1 month (4) 6. Khoshoo V, Sun S, Storm H. Tolerance of an enteral formula with
◦ Other (5) insoluble and prebiotic fiber in children with compromised gastroin-
testinal function. J Am Diet Assoc 2010;110(11):1728-1733.
Q10 What region is your facility located in? 7. Rabbani G, Teka T, Saha S, et al. Green banana and pectin Improve
small intestinal permeability and reduce fluid loss in Bangladeshi
◦ West (WA, OR, CA, NV, UT, AZ, NM, CO, WY, MT, children with persistent diarrhea. Dig Dis Sci 2004;49(3):475-484.
8. Alam N, Meier R, Schneider H, et al. Partially hydrolyzed guar gum–
ID) (1)
supplemented oral rehydration solution in the treatment of acute
◦ Mid-West (ND, SD, NE, KS, MN, IA, MO, IL WI, diarrhea in children. J Pediatr Gastroenterol Nutr 2000;31(5):503-507.
IN, OH, MI) (2) 9. Becker B, Kuhn U, Hardewig-Budny B. Double-blind, randomized
◦ South (TX, OK, AR, LA, MS, AL, GA, FL, SC, NC, evaluation of clinical efficacy and tolerability of an apple pectin-
TN, KY, WV, VA, MD) (3) chamomile extract in children with unspecific diarrhea. Arzneimit-
telforschung 2006;56(06):387-393.
◦ North East (ME, NH, VT, NY, MA, RI, CT, NJ, PA)
10. Drenckpohl D, Hocker J, Shareef M, Vegunta R, Colgan, C. Adding
(4) dietary green beans resolves the diarrhea associated with bowel surgery
◦ Outside the U.S.; specify (5) in neonates: a case study. Nutr Clin Pract 2005;20(6):674-677.
11. Goulet O, Olieman J, Ksiazyk J, et al. Neonatal short bowel syndrome
Q11 What type of medical professional are you? as a model of intestinal failure: physiological background for enteral
feeding. Clin Nutr 2013;32(2):162-171.
◦ Physician (1) 12. Olieman J, Penning C, Ijsselstijn H, et al. Enteral nutrition in children
◦ Dietitian (2) with short-bowel syndrome: current evidence and recommendations for
the clinician. J Am Diet Assoc 2010;110:420-426.
◦ Surgeon (3)
13. Piper H, Fan D, Coughlin L, et al. Severe gut microbiota dysbiosis is
◦ Nurse practitioner/physician assistant (4) associated with poor growth in patients with short bowel syndrome.
◦ Other (5) JPEN J Parenter Enteral Nutr 2017;41(7):1202-1212.
14. Holscher H. Dietary fiber and prebiotics and the gastrointestinal
Q12 What is your average annual short bowel patient microbiota. Gut Microbes 2017;8(2):172-184.
census? 15. Yamamoto Y, Takahahi T, To M, et al. The salivary IgA flow rate Is
increased by high concentrations of short-chain fatty acids in the cecum
◦ <10 (1) of rats ingesting fructooligosaccharides. Nutrients 2016;8(8):500.
16. Kim M, Qie Y, Park J, Kim C. Gut microbial metabolites fuel host
◦ 10–50 (2)
antibody responses. Cell Host Microbe 2016;20(2):202-214.
◦ 50–100 (3) 17. Nakao M, Ogura Y, Satake S, et al. Usefulness of soluble dietary fiber
◦ >100 (4) for the treatment of diarrhea during enteral nutrition in elderly patients.
◦ Other (5) Nutrition 2002;18(1):35-39.
18. Bouhnik Y, Vahedi K, Achour L, et al. Short-chain fructo-
Q13 Does your facility have a gut/intestinal rehabilitation oligosaccharide administration dose-dependently increases fecal bifi-
dobacteria in healthy humans. J Nutr 1999;129(12):113–116.
team and/or program?
19. Davis L, Martı́nez I, Walter J, Goin C, Hutkins R. Barcoded py-
rosequencing reveals that consumption of galactooligosaccharides
◦ Yes (1)
results in a highly specific bifidogenic response in humans. PLoS One
◦ No (2) 2011;6(9):e25200.
20. Carvalho L, Brait D, Vaz M, et al. Partially hydrolyzed guar gum
increases ferroportin expression in the colon of anemic growing rats.
References Nutrients 2017;9(3):228.
1. Rabbani G, Teka T, Zaman B, Majid N, Khatun M, Fuchs, G. Clinical 21. Wallace T, Marzorati M, Spence L, Weaver C, Williamson P. New fron-
studies in persistent diarrhea: dietary management with green banana tiers in fibers: innovative and emerging research on the gut microbiome
or pectin in Bangladeshi children. Gastroenterology 2001;121(3):554- and bone health. J Am Coll Nutr 2017;36(3):218-222.
560. 22. Abrams S, Griffin I, Hawthorne K, et al. A combination of prebiotic
2. Squires R, Duggan C, Teitelbaum D, et al. Natural history of pediatric short- and long-chain inulin-type fructans enhances calcium absorp-
intestinal failure: initial report from the pediatric intestinal failure tion and bone mineralization in young adolescents. Am J Clin Nutr
consortium. J Pediatr 2012;161(4):723-728.e2. 2005;82(2): 471-476.
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 4
Use of a Gastroschisis Feeding Guideline to Improve August 2018 545–552

C 2018 American Society for

Standardization of Care and Patient Outcomes Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10083
at an Urban Children’s Hospital wileyonlinelibrary.com

R. Colby Passaro, MPH1 ; Kate B. Savoie, MD, MS1,2,3 ; and


Eunice Y. Huang, MD, MS, FACS, FAAP1,2,3

Abstract
Background: This study examined clinical outcomes associated with the use of a gastroschisis-specific (GS) feeding advancement
guideline. Methods: We performed a retrospective study of all simple gastroschisis babies (N = 65) treated between June 2009June
2015. We compared patients treated on a postintestinal surgery guideline using either a 1-day (1D) or 3-day (3D) feeding
advancement from August 2009–August 2013 with infants treated on a GS guideline from September 2013–June 2015. Results:
Patients in the 2 groups were similar in sex, race, gestational age, weight, and comorbidities. Median time to full enteral nutrition
(EN) was 11 days for the 1D group, 22 days for the 3D group, and 18 days for the GS group (P < .01). However, lengths of stay
and estimated weight gain per day were similar among the groups. A total of 3 infants (10%) in the 1D group developed necrotizing
enterocolitis compared with none in the 3D or GS groups. Control chart analysis showed reduced variation in median time to full
EN in the GS group when compared with the 1D and 3D groups. Guideline adherence was significantly better with the GS guideline
when compared with the 1D or 3D guidelines (94% vs 72% vs 90%; P < .01). Conclusion: A GS protocol yielded reduced variation
in median time to full EN, significant improvement in percent adherence to the guideline, and zero cases of necrotizing enterocolitis.
Weight gain and lengths of stay were not adversely affected by slower feeds. (Nutr Clin Pract. 2018;33:545–552)

Keywords
gastroschisis; infant; necrotizing enterocolitis; digestive system; surgical procedures; enteral nutrition; tube feeding; neonate; clinical
protocols

Gastroschisis is a congenital abdominal wall defect requir- trition management.11 For example, a recent study showed
ing emergent surgical intervention and is on the rise in the that time from gastroschisis closure to first feed predicts
United States. According to the Centers for Disease Control outcomes in these infants.12 Despite that, there are few
and Prevention, the prevalence of gastroschisis increased published guidelines prescribing exactly how and when
from 2.3 per 10,000 live births to 4.4 per 10,000 live births enteral feeding should be administered, and how it should
between 1995 and 2005.1 Almost 50% of babies born with be advanced.13,14 Cholestatic jaundice and infection can
gastroschisis are born prematurely, which is a risk factor also be a problem with long-term use of PN; therefore, the
for the development of necrotizing enterocolitis (NEC) and prompt advancement of enteral nutrition (EN) is preferred
longer lengths of hospital stay. Some of these children when safe.15-19
require prolonged parenteral nutrition (PN), which can lead Prior to 2009, there was no standard feeding advance-
to severe cholestasis.2,3 Although gastroschisis is typically ment regimen in our institution; the advancement for
diagnosed via antenatal ultrasound, it is difficult to reliably postintestinal surgery (PIS) infants in the neonatal intensive
predict postnatal morbidity.4
The use of protocols to standardize care has been
shown to be beneficial for critically ill patients, con- From the 1 College of Medicine, University of Tennessee Health
Science Center, Memphis, Tennessee, USA; 2 Division of Pediatric
tributing to improved patient outcomes and provider sat-
Surgery, Le Bonheur Children’s Hospital, Memphis, Tennessee, USA;
isfaction in retrospective studies.5,6 Protocolization also and 3 Department of General Surgery, University of Tennessee Health
helps to identify provider noncompliance to best practices Science Center, Memphis, Tennessee, USA.
and may help identify associated factors that contribute Conflicts of interests: None declared.
to poor outcomes.7 Moreover, the implementation of
This article originally appeared online on April 4, 2018.
evidence-based nutrition management protocols has proven
Corresponding Author:
to improve patient outcomes in critically ill patients.8-10
Eunice Y. Huang, MD, MS, FACS, FAAP, 51 N. Dunlap St., Second
One key element associated with morbidity, mortality, Floor, Memphis, TN 38105.
and health outcomes of gastroschisis patients is their nu- Email: ehuang@uthsc.edu
546 Nutrition in Clinical Practice 33(4)

care unit (ICU) was determined by physician preferences June 2015 as categorized by the International Classification
and individual infant needs. In an effort to standardize of Disease ninth edition diagnosis code 756.73. Inclusion
and improve the quality of care we deliver to our PIS criteria were any infants with a diagnosis of simple gas-
patients, we instituted a feeding advancement guideline for troschisis (N = 65), excluding infants with associated com-
postsurgical patients as a part of a quality improvement plex intestinal problems or intestinal resections (n = 14).
project in August 2009. The guidelines were the product Infants were also excluded if they did not reach goal feeds
of collaboration between pediatric surgeons, neonatologists, or if they died prior to discharge from the hospital (n = 5).
pediatric gastroenterologists, neonatal dietitians, clinical Patients surgically treated at our neonatal ICU unit
pharmacists, and speech therapists. Feeding advancement were sampled in the following 2 cohorts: 1 treated between
was based on residual small-bowel length after intestinal August 1, 2009 and August 31, 2013, on a PIS feeding
surgery and weight at initiation of feeds. We assessed the guideline (n = 43) and another treated between September 1,
safety and efficacy of this guideline in a previous study, 2013 and June 30, 2015, on a GS feeding guideline
finding less severe cholestasis and increased breast milk use (n = 17). Babies treated before September 1, 2013, had
in the postimplementation group, and 67% adherence to enteral intake increases every 1 day (n = 26) or 3 days
the guideline.20 However, during this evaluation, we also (n = 4), or a combination of these (n = 13), depending
noted that our simple gastroschisis patients were being on bowel length and continuity (Table 1). Those infants
categorized into >1 feeding advancement category (either managed on a combination were assigned to a group based
1-day advancement or 3-day advancement) by the clinicians on their starting guideline, yielding only a 1-day group
using this protocol despite the fact that these patients did (n = 29) and a 3-day group (n = 14). Of note, although
not have any bowel resected. In addition, we observed a few no patients included in this study had any bowel resection,
episodes of NEC in our 1-day advancement cohort. 14 infants were fed according to the 3-day PIS guideline
As a result, we developed a gastroschisis-specific (GS) based on the clinical assessment of the patient care team,
feeding guideline. The purpose of this guideline, imple- including the surgeon and neonatologist. All patients
mented in September 2013, was to more accurately meet were assessed every 12 hours, or as clinically appropriate,
the needs of the simple gastroschisis patient population and for signs of intolerance. The babies treated on or after
provide quality care by standardizing management. These September 1, 2013, received enteral intake increases every
infants had the full length of their small intestine, did not other day until 50% of target and then were advanced every
have associated complex intestinal problems and, therefore, day until target (Table 2).
underwent standard abdominal wall closure with or without
initial silo placement. Under this guideline, all postoperative Feeding Advancement Strategy
infants with simple gastroschisis and no contraindications
PN was initiated immediately for postoperative infants.
to enteral feeding are advanced every 48 hours until 50% of
Enteral feeding was initiated, orally or by nasogastric tube
goal feeds, then every 24 hours until goal feeds. Advance-
(based on the infant’s neurodevelopmental and cardiopul-
ment was designed to be every other day at the start of
monary status), when the infant had return of bowel
enteral feeding because of possible early dysmotility from
function (ie, stool or flatus). When feeding was tolerated,
inflammation of the previously exposed intestines.
the overall caloric intake from feeding was calculated, and
The aim of this study was to retrospectively compare
the difference between total caloric needs and those pro-
gastroschisis infants cared for after implementation of the
vided by the feeding was supplemented with PN. Clinicians
PIS guideline to those cared for after implementation of the
were encouraged to use expressed breast milk as the first
GS guideline. Our primary aim was to determine whether
choice for feeding, followed by donor breast milk, then
implementation of the newer guideline resulted in a change
age-appropriate formula. Fortification of breast milk was
in the length of time needed to achieve full enteral nutrition
initiated only after the infant was tolerating >50% of his
(FEN) after surgical closure. Our secondary goals were to
or her target feeding.
determine if the change in guideline resulted in decreased
The GS guideline was developed in September 2013. In
in-hospital complication rates, decreased variation in time
this guideline, all postoperative infants with simple gas-
to full feeds, and/or improved guideline compliance.
troschisis in continuity with the colon and no contraindica-
tions to enteral feeding were advanced every 48 hours until
Methods 50% of goal, then every 24 hours until goal feeds (Table 2).

Study Design Outcome Measures


After receiving approval from the university institutional The primary outcome for our study was time to FEN.
review board, we identified all neonates who underwent Secondary outcomes included time to initiation of feeds
abdominal surgery for gastroschisis between June 2009 and after abdominal wall closure, length of hospital stay from
Passaro et al 547

Table 1. Postintestinal Surgery Enteral Feeding Guidelines.

Patient Type and 1 Day 3 Day


Feeding

Residual small >40% residual small bowel in continuity with 20%–40% residual small bowel in continuity
bowela colon with colon
Patient weight <1500 g ࣙ1500 g <1500 g ࣙ1500 g
Initial enteral 12 mL/kg/d 12–24 mL/kg/d 1 mL every 3 12 mL/kg/d
feedsb continuous continuous OR hours for 3 days continuous
feeds bolus feeds then 12 mL/kg/d OR
continuous intermittent
feeds continuous
feeds
Feeding increases Increase by 12 Increase by 12 Increase by 12 mL/kg/hr every 3 days
mL/kg/d every mL/kg/d every
48 hours 24 hours
Feeding Evaluate every 3 hours. If intolerance, hold Evaluate every 3 hours. If intolerance, hold
intolerancec feeds for 12 hours and reassess feeds for 12 hours and reassess

No infants included in this study were started on the 7-day guideline because they were not eligible for it based on the aforementioned criteria.
a Normal intestinal length: 140 cm for gestational age of 19–27 weeks, 220 cm for gestational age of 27–35 weeks, and 300 cm for gestational age

>35 weeks.22
b Contraindications to enteral feeding include treatment with indomethacin within 48 hours, cardiac conditions that compromise gastrointestinal

perfusion, polycythemia with a hematocrit >65 mg/dL, significant metabolic acidosis, decompensating respiratory status, hemodynamic
instability, exchange transfusion within 48 hours, abdominal signs and symptoms, severe asphyxia within 72 hours, significant apnea, cyanosis or
bradycardia associated with gavage feedings, diagnosis of stage I necrotizing enterocolitis within 10 days or stage II necrotizing enterocolitis
within 14 days, and prior return of bowel function postoperatively.
c Feeding intolerance was defined as a change in the character of gastric residuals, emesis, or ostomy output; hemorrhagic stools; or abdominal

distention with redness.

Table 2. Gastroschisis-Specific Enteral Feeding Guidelines. admission to discharge, number of patients with at least 1
episode of feeding intolerance, number of patients with at
Patient Type and Simple Gastroschisis
least 1 in-hospital complication, frequency of in-hospital
Feeding
complications, including infection, transfusion, additional
Eligible candidates Postoperative infants with simple surgical intervention, or NEC, highest total bilirubin, to-
gastroschisis in continuity with tal bilirubin at discharge, daily weight gain (grams), and
colon and no contraindications to percent adherence to guideline.
enteral feedinga
Initial enteral feeds 12 mL/kg/d continuous or bolus feeds
Feeding increases Increase by 12 mL/kg/d every
48 hours until 50% of goal enteral
volume; then increase in
Definitions
increments of 12 mL/kg/d every FEN was defined as the infant reaching goal kilocalories per
24 hours until goal kilogram per day (kcal/kg/day) feeding, as determined by
b
Feeding intolerance Evaluate every 3 hours. If intolerance,
the neonatologist and dietitian taking care of patients in the
hold feeds for 12 hours and reassess
neonatal ICU. Time to FEN was calculated from time of
a Contraindications to enteral feeding include treatment with start of enteral feeding after abdominal closure to time of
indomethacin within 48 hours, cardiac conditions that compromise reaching FEN, for example, when both target volume and
gastrointestinal perfusion, polycythemia with a hematocrit >65
mg/dL, significant metabolic acidosis, decompensating respiratory
target calories had been reached.
status, hemodynamic instability, exchange transfusion within 48 The percent of remaining small bowel for infants treated
hours, abdominal signs and symptoms, severe asphyxia within 72 on the PIS protocol was determined by dividing the remain-
hours, significant apnea, cyanosis, or bradycardia associated with ing small-bowel length as measured by the operative surgeon
gavage feedings, diagnosis of stage I necrotizing enterocolitis within
10 days or stage II necrotizing enterocolitis within 14 days, and prior
by the expected average neonatal small-bowel length based
return of bowel function postoperatively. on gestational age as determined by a previous publication
b Feeding intolerance was defined as a change in the character of
(19–27 weeks gestational age, 140 cm; 27–35 weeks, 220 cm;
gastric residuals, emesis, or ostomy output; hemorrhagic stools; or >35 weeks, 300 cm), and multiplying that factor by 100.20-22
abdominal distention with redness.
Length of stay was calculated as the number of days
the infant spent in the hospital from their arrival date to
548 Nutrition in Clinical Practice 33(4)

discharge after gastroschisis repair. No infants included in Table 3. Baseline Demographics: Median (Quartile 1,
the study were discharged before reaching full enteral feeds. Quartile 3) or Frequency (%).
NEC was defined as a diagnosis made by the neona-
Baseline 1-Day GL 3-Day GL GS GL
tologist or surgeon treating the infants if they had signs Demographics (n = 29) (n = 14) (n = 17) P Value
consistent with Bell’s stage II or higher based on overall
clinical picture, imaging, or operative findings. Male 14 (48) 10 (71) 9 (53) .35
Adherence to feeding guidelines was calculated by as- Race
sessing the percentage of days that the clinician used the Black or 8 (28) 5 (36) 2 (12) .62
recommended advancement or nonadvancement of feeding African
American
based on the infant’s daily clinical status. This information White 15 (52) 7 (50) 11 (65)
was reviewed in the daily progress notes in each infant’s Other, multi, 6 (20) 2 (14) 4 (24)
medical record, where the EN volume was recorded; if the or
volume delivered was in accordance with the documented unspecified
guideline, that day was considered adherent. If there were Gestational age 36 37.4 36.5 .05
clinical reasons that resulted in delayed advancement, this (34.6, 37.1) (36.6, 37.6) (35.2, 37.4)
was considered “adherence to the guideline” because both Birthweight, g 2310 (1919, 2556 (2200, 2650 (2400, .14
2789) 2900) 2990)
guidelines allowed for change based on clinical status of the Non-GI- 19 (66) 5 (36) 10 (59) .18
infants. For those infants fed with the PIS guideline, adher- related
ence was assessed according to the documented designation, comorbidities
regardless of whether the protocol selected was “correct”
based on the criteria for 1-day or 3-day advancement. GL, guideline; GS, gastroschisis-specific.

Results
Statistical Analysis
Data were summarized using proportions for categorical Cohort Demographics
variables and median (interquartile ranges) for nonpara- There were 60 simple gastroschisis infants included in the
metric continuous variables. To test for differences in the analysis: 43 infants were treated according to the PIS
1-day, 3-day, and GS guidelines for categorical variables, guideline (29 infants on 1-day advancement; 14 on 3-day
the χ 2 test or Fisher’s exact test were used, as appropriate. advancement), and 17 were treated on the GS guideline
The Kruskall-Wallis test was used to test for differences in (Table 3). The median gestational age was 36.5 weeks
continuous variables between the 3 groups. The Wilcoxon (quartile 1, 35.2; quartile 3, 37.45); 1 (1.7%) infant was
Mann–Whitney test was used to test for differences in ࣘ28 weeks gestation, 2 (3.3%) were 29–32 weeks gestation,
continuous variables between the PIS and GS groups. All 29 (48.3%) were 33–36 weeks gestation, and 28 (46.7%)
statistical analyses were performed using Stata SE 12.0 were ࣙ37 weeks gestation. The median birth weight was
(StataCorp, College Station, TX). 2472.5 grams (quartile 1, 2061.5; quartile 3, 2855). The most
To better visualize the results of our process change and common comorbidities were patent foramen ovale, patent
assess the common vs special cause variations, Shewhart ductus arteriosus, intraventricular hemorrhage, laryngoma-
control charts were developed in concert with the hospital lacia, meconium aspiration syndrome, respiratory distress
Department of Quality Improvement and Risk to identify syndrome, hydronephrosis, bladder exstrophy, and anemia
common and special cause variation in time-to-goal enteral of prematurity.
feeds and percent adherence to the guidelines. Control
charts are a standard assessment for quality improvements Analysis of Outcomes, Adverse Events,
that allow simple detection of events that are indicative of
actual process change. Common cause variations are caused
and Guideline Compliance
by unknown factors resulting in a random distribution of There was a significant difference in the median time to
outputs around the mean. They are always present in the FEN between the 3 groups: 11 days for the 1-day guideline,
process. This variation falls between the upper and lower 22 days for the 3-day guideline, and 18 days for the GS
control limits on the chart. Special cause variations are guideline (P < .01). This difference in median time to FEN
caused by factors that result in a nonrandom distribution was not significant when comparing the PIS protocol babies
of output and can be potentially removed by improving the (combined 1-day and 3-day guidelines) to the GS protocol
process.23 All quality improvement analyses were performed babies (14 vs 18; P = .38). Length of stay was also similar
using Minitab 18 (Minitab Inc., State College, PA). between the 3 groups.
Passaro et al 549

Table 4. Outcomes: Median (Quartile 1, Quartile 3) or Frequency (%).

Outcomes 1-Day GL (n = 29) 3-Day GL (n = 14) GS GL (n = 17) P Value

Time to initiation of feeds after 13 (10, 18) 13 (9, 17) 10 (7, 14) .14
reanastomosis
Time to full feeds 11 (8, 19) 22 (14, 32) 18 (15, 23) <.01
Length of stay 37 (25, 52) 51 (36, 59) 40 (29, 56) .15
Patients with at least 1 episode 13 (45) 6 (43) 6 (35) .81
of feeding intolerancea
Number of patients with at 7 (24) 3 (21) 3 (17) .77
least 1 in-hospital
complication
Number of complications, total 9 (–) 3 (–) 3 (–)
Infection 4 (14) 2 (14) 3 (17) 1.00
Transfusion 2 (7) 0 (0) 0 (0) .72
Additional surgical 0 (0) 1 (7) 0 (0) .23
intervention
Necrotizing enterocolitis 3 (10) 0 (0) 0 (0) .33
Highest total bilirubin 2.7 (0.8, 3.9) 2.3 (1.3, 4.8) 3.2 (1.2, 4.9) .63
Total bilirubin at discharge 0.5 (0.4, 1.3) 0.75 (0.4, 2.2) 0.8 (0.4, 3.7) .63
Daily weight gain, g 17.4 (12.5, 21.4) 19.2 (15.3, 26.1) 19.7 (17.7, 22.9) .25
Percent adherence to guideline 72 (45, 89) 90 (79, 100) 94 (87, 100) <.01

Bold text = P value <0.05. GL, guideline; GS, gastroschisis-specific.


a Feeding intolerance = cessation of feeds for at least 24 hours secondary to aspiration, emesis, oral aversion, surgical interventions related to

feeding/gastrointestinal disturbance, diarrhea, abdominal distention, blood in the stool, ileus, necrotizing enterocolitis, infection, or increased
ostomy output.

The number of infants with feeding intolerance was not the upper control limit for time-to-goal enteral feeds and
significantly different among the 3 groups. The number of points below the lower control limit for percent adherence
infants with at least 1 in-hospital complication in the 1-day to the guidelines.24 There was no special cause variation in
group (7; 24%) was higher than that in the 3-day group (2; the GS guideline group.
21%) and GS group (3; 18%); yet this difference was not
statistically significant (P = .77). There were 3 cases of NEC Discussion
(10%) in the group advanced using the 1-day PIS guideline
Although the survival rate for gastroschisis is >90%,
(Table 4).
survivors are frequently affected by varying degrees of
There was no significant difference in the level of highest
gastrointestinal dysfunction, which can lead to significant
total bilirubin while in the hospital or total bilirubin at
morbidity and prolonged need for PN.25 Appropriate nu-
discharge between the 3 groups. Speed of advancement of
trition management is thus a key component of quality
feeds did not affect daily weight gain: 17.4 grams/day for the
hospital care and good health outcomes for these post-
1-day group, 19.2 grams/day for the 3-day group, and 19.7
surgical infants.26 However, enteral feeding practices for
grams/day for the GS group (P = .25).
infants with gastroschisis are highly variable, and there are
Median percent adherence to the guideline was much
few published guidelines addressing how to advance feeds
higher in the GS (94%) and 3-day advancement (90%)
following abdominal wall closure.12 Our results show that
groups than in the 1-day advancement group (72%; P < .01;
implementing a simple GS feeding guideline can improve
Table 4).
the standardization of care by reducing variation in mean
time to FEN and increasing provider adherence to feeding
Quality Improvement Analysis advancement protocol. Moreover, this guideline is safe and
The analysis of time-to-goal feeds using Shewhart control does not significantly change mean time-to-goal enteral
charts showed both decreased common and special cause feeds, length of stay, or daily weight gain.
variation during the GS guideline time period as compared Full enteral feeding at a median of 18 days on the
with the PIS period, both in the 1-day and 3-day groups GS guideline is consistent with the current literature for
(Figure 1). Similar findings were noted when percent feeding advancement in infants with simple gastroschisis.27
adherence was assessed (Figure 2). There were instances of Although the infants managed on the 1-day feeding
special cause variation (according to the Westgard rules) in guideline reached FEN in a shorter period of time (11
both the 1-day and 3-day groups in the form of points above days), we contend that this difference is minimally clinically
550 Nutrition in Clinical Practice 33(4)

Figure 1. Three-group time to enteral feeds.

Figure 2. Three-group percent adherence to guideline.

significant, especially as it did not impact daily weight gain process. This strategy limits the stress put on the postsurgi-
or length of stay, which are critical indicators of neonatal cal inflamed intestines; gastroschisis infants are at a higher
nutrition status and cost of care, respectively. risk of feeding intolerance shortly after surgery and may
Consistent with previous studies, we found that although not tolerate advancement every day. Our results suggest
increasing the rate of feeding advancement can result in this strategy may be effective as we noted a downward
a shorter time to FEN in infants with gastroschisis, this trend in incidence of feeding intolerance from the PIS
speed brings with it a risk of NEC.28 We designed our infants to the GS infants, and we did not observe any
GS guideline to limit this risk by advancing feeding every cases of NEC in infants managed according to the GS
other day for the first half of the feeding advancement guideline.
Passaro et al 551

Our results contribute to the small but growing body of Acknowledgments


literature showing that standardization of feeding guide- We would like to thank Donna Vickery, administrative director
lines on EN administration improves patient outcomes.26 in the Department for Quality Improvement and Risk, for her
Our GS guideline successfully reduced variation in practice, help with control chart development. We would also like to
yielding increased adherence to the feeding advancement thank Sandra Grimes for her help as our research coordinator.
protocol by providers. This standardization of care resulted
in less variation in time to FEN for the infants managed on Statement of Authorship
this protocol, and therefore fewer isolated cases of infants
R. C. Passaro, K. Savoie, and E. Huang equally contributed
with very long hospital stays. In other words, although the
to the conception and design of the research; R. C. Passaro
1-day guideline may have been very successful for some
contributed to the acquisition and analysis of the data; R.
infants, it was very unsuccessful for others, in contrast to the C. Passaro, K. Savoie, and E. Huang contributed to the
GS guideline, which resulted in a slight increase in days to interpretation of the data; and R. C. Passaro, K. Savoie, and E.
FEN for the group as a whole, but fewer adverse outcomes Huang drafted the manuscript. R. C. Passaro, K. Savoie, and
such as NEC. Our results thus show that it is possible to E. Huang critically revised the manuscript; R. C. Passaro, K.
provide more efficient care without compromising patient Savoie, and E. Huang agree to be fully accountable for ensuring
outcomes with a GS guideline. the integrity and accuracy of the work; and all authors read and
There are several limitations to this study. First, it is a approved the final manuscript.
retrospective study subject to errors caused by inconsisten-
cies or omissions in medical record data. We addressed this References
limitation by strictly defining the outcomes of interest at 1. Kirby RS, Marshall J, Tanner JP, et al. Prevalence and correlates of
the outset of the study and having 1 researcher perform gastroschisis in 15 states, 1995 to 2005. Obstet Gynecol. 2013;122(2 pt
data extraction to maintain consistency of interpretation of 1):275-281.
data. We also performed multiple internal audits of the data 2. Gupta V, Trivedi A, Walker K, Holland JA. Neurodevelopmental
outcome of infants with gastroschisis at one-year follow-up. J Neonatal
following the first extraction. Second, this was a small study,
Surg. 2015;4(2):12.
increasing the risk for type II errors. There may have been 3. Mills JA, Lin Y, Macnab YC, Skarsgard ED. Perinatal predictors of
differences in outcomes between the 2 populations that were outcome in gastroschisis. J Perinatol. 2010;30(12):809-813.
not identified as a result of our small sample size. This is a 4. D’Antonio F, Virgone C, Rizzo G, et al. Prenatal risk factors and out-
common issue in research projects evaluating rare diseases; comes in gastroschisis: a meta-analysis. Pediatrics. 2015;136(1):e159-
e169.
however, we plan to continue monitoring these outcomes in
5. Chang SY, Sevransky J, Martin GS. Protocols in the management of
our expanding population in the coming years. Because the critical illness. Crit Care. 2012;16(2):306.
safety of our patients is of the utmost importance, we chose 6. Wellington A, Perlman JM. Infant-driven feeding in premature infants:
to evaluate the safety and efficacy of this guideline now a quality improvement project. Arch Dis Child Fetal Neonatal Ed.
rather than wait several more years to ensure statistically 2015;100(6):F495-F500.
7. Cotton BA, Dossett LA, Au BK, Nunez TC, Robertson AM, Young
significant results. The use of the Shewhart control charts
PP. Room for (performance) improvement: provider-related factors
also helped us note changes in variation over time in these associated with poor outcomes in massive transfusion. J Trauma.
small populations. Finally, the neonates in the PIS protocol 2009;67(5):1004-1012.
were treated during a different time period than those on the 8. Barr J, Hecht M, Flavin KE, Khorana A, Gould MK. Outcomes in crit-
GS protocol, and healthcare is a rapidly advancing field, so ically ill patients before and after the implementation of an evidence-
based nutritional management protocol. Chest. 2004;125(4):1446-1457.
there may have been additional differences in management
9. Dervan N, Dowsett J, Gleeson E, Carr S, Corish C. Evaluation of over-
that were not accounted for by our analysis, confounding and underfeeding following the introduction of a protocol for weaning
our results. from parenteral to enteral nutrition in the intensive care unit. Nutr Clin
Our results show that standardization of care with a Pract. 2012;27(6):781-787.
simple GS feeding guideline can yield fewer adverse health 10. McCallie KR, Lee HC, Mayer O, Cohen RS, Hintz SR, Rhine WD.
Improved outcomes with a standardized feeding protocol for very low
outcomes without significantly impacting time-to-goal en-
birth weight infants. J Perinatol. 2011;31(suppl 1):S61-S67.
teral feeds, length of stay, or weight gain. Future quality 11. Walter-Nicolet E, Rousseau V, Kieffer F, et al. Neonatal outcome of
improvement projects with a larger sample size are needed to gastroschisis is mainly influenced by nutritional management. J Pediatr
accurately assess the statistical significance of the reduction Gastroenterol Nutr. 2009;48(5):612-617.
in undesired outcomes such as feeding intolerance, in- 12. Aljahdali A, Mohajerani N, Skarsgard ED. Effect of timing of enteral
feeding on outcome in gastroschisis. J Pediatr Surg. 2013;48(5):971-976.
hospital infections, and postsurgical enterocolitis. The next
13. Olieman JF, Penning C, Ijsselstijn H, et al. Enteral nutrition in children
step for our quality improvement project is an increase in the with short-bowel syndrome: current evidence and recommendations for
rate of feeding advancement for bigger babies with simple the clinician. J Am Diet Assoc. 2010;110(3):420-426.
gastroschisis: 24 kcal/kg per advancement for infants >1500 14. Kohler JA Sr, Perkins AM, Bass WT. Human milk versus formula after
grams. The safety and efficacy of this change in our feeding gastroschisis repair: effects on time to full feeds and time to discharge.
J Perinatol. 2013;33(8):627-630.
guideline will be evaluated in future studies.
552 Nutrition in Clinical Practice 33(4)

15. Sondheimer JM, Asturias E, Cadnapaphornchai M. Infection and 22. Touloukian RJ, Smith GJ. Normal intestinal length in preterm infants.
cholestasis in neonates with intestinal resection and long-term par- J Pediatr Surg. 1983;18(6):720-723.
enteral nutrition. J Pediatr Gastroenterol Nutr. 1998;27(2):131-137. 23. Koutras MV, Bersimis S, Maravelakis PE. Statistical process control
16. Spencer AU, Yu S, Tracy TF, et al. Parenteral nutrition-associated using shewhart control charts with supplementary runs rules. Methodol
cholestasis in neonates: multivariate analysis of the potential protective Comput Appl. 2007;9(2):207-224.
effect of taurine. JPEN J Parenter Enteral Nutr. 2005;29(5):337-343; 24. Westgard JO, Barry PL. Cost-Effective Quality Control: Managing the
discussion 43–44. Quality and Productivity of Analytical Processes. Washington, DC:
17. Mungnirandr A, Khorana J, Ruangtrakool R. Correlation between American Association for Clinical Chemistry Press; 1986.
duration of postoperative parenteral nutrition and incidence of post- 25. Overcash RT, DeUgarte DA, Stephenson ML, et al. Factors associated
operative complication in gastroschisis patients. J Med Assoc Thai. with gastroschisis outcomes. Obstet Gynecol. 2014;124(3):551-557.
2010;93(4):443-448. 26. Loomis T, Byham-Gray L, Ziegler J, Parrott JS. Impact of standard-
18. Wessel JJ, Kocoshis SA. Nutritional management of infants with short ized feeding guidelines on enteral nutrition administration, growth
bowel syndrome. Semin Perinatol. 2007;31(2):104-111. outcomes, metabolic bone disease, and cholestasis in the NICU.
19. Fallon EM, Mitchell PD, Potemkin AK, et al. Cholestasis and growth J Pediatr Gastroenterol Nutr. 2014;59(1):93-98.
in neonates with gastroschisis. J Pediatr Surg. 2012;47(8):1529-1536. 27. Miranda da Silva Alves F, Miranda ME, de Aguiar MJ, Bouzada
20. Savoie KB, Bachier-Rodriguez M, Jones TL, et al. Standardization of Viana MC. Nutritional management and postoperative prognosis
feeding advancement after neonatal gastrointestinal surgery: does it of newborns submitted to primary surgical repair of gastroschisis.
improve outcomes? Nutr Clin Pract. 2016;31(6):810-818. J Pediatr (Rio J). 2016;92(3):268-275.
21. Holland AJ, Walker K, Badawi N. Gastroschisis: an update. Pediatr 28. Hay WW Jr. Strategies for feeding the preterm infant. Neonatology.
Surg Int. 2010;26(9):871-878. 2008;94(4):245-254.
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 4
Impact of Gastrostomy Feeding Tube Placement August 2018 553–566

C 2018 American Society for

on the 1-Year Trajectory of Care in Patients After Stroke Parenteral and Enteral Nutrition
DOI: 10.1002/ncp.10015
wileyonlinelibrary.com

Janina Wilmskoetter, PhD1 ; Annie N. Simpson, PhD2,3 ; Sarah L. Logan, PhD4 ;


Kit N. Simpson, DrPH2 ; and Heather S. Bonilha, PhD, CCC-SLP1,3

Abstract
Background: Percutaneous endoscopic gastrostomy (PEG) feeding tubes are commonly placed in acute stroke patients with a
need for enteral nutrition. However, PEG tubes are associated with medical complications and a decrease in quality of life. We
compared the 1-year care trajectory of stroke patients with and without PEG tube placement to enhance knowledge about the
long-term impact of PEG tube placement. Methods: We conducted a retrospective analysis of commercially insured stroke patients
included in the Truven Health MarketScan Research Databases of 2011. We analyzed their index hospital stay and conducted 1-
month, 3-months, 6-months, and 1-year follow-ups. We compared admissions to inpatient rehabilitation facilities, acute hospitals,
skilled nursing facilities, outpatient hospital visits, and home visits for stroke patients with and without PEG tube placement using
unadjusted and adjusted modelling. Results: Of the 8911 included stroke patients, 148 patients (1.7%) had a PEG tube placed during
their index hospital stay. After controlling for age, gender, stroke severity, comorbidities, and stroke type, PEG tube placement was
an independent predictor for admissions to inpatient rehabilitation facilities and skilled nursing facilities. Furthermore, PEG tube
placement was an independent predictor for all-cause, unplanned hospital readmissions in a multivariable logistic model (area under
the receiver operating characteristic curve was .84). Conclusion: Stroke patients who receive a PEG tube can expect a significantly
different care trajectory after being discharged from the acute hospital. Our findings can aide in predicting recovery and planning
resources and identifying gaps and points for improvement in stroke care for patients with PEG tube placement. (Nutr Clin Pract.
2018;33:553–566)

Keywords
stroke; deglutition; deglutition disorders; enteral nutrition; hospitalization; patient readmission

Background decision to place a PEG tube need to carefully weigh the


potential risks and benefits. One of the biggest factors for
Stroke is a leading cause of death, severe long-term dis- consideration is the prognosis of the individual patient.
ability, and healthcare expenditures in the United States,1,2 For this purpose, understanding the trajectory of care for
with 795,000 people annually experiencing a new or recur-
rent stroke.1 Patients commonly depend on enteral and/or
From the 1 Department of Health Sciences and Research, College of
parenteral nutrition because complications from the stroke Health Professions, Medical University of South Carolina,
impact their ability to meet nutrition and hydration needs, Charleston, South Carolina, USA; 2 Department of Healthcare
for example, as a consequence of dysphagia (swallowing Leadership and Management, College of Health Professions, Medical
disorders). When there is an anticipated prolonged need University of South Carolina, Charleston, South Carolina, USA;
3 Department of Otolaryngology–Head and Neck Surgery, Medical
for enteral nutrition, percutaneous endoscopic gastrostomy
University of South Carolina, Charleston, South Carolina, USA; and
(PEG) tubes are placed in stroke patients,3 with about 6% of 4 Department of Neurosciences, College of Medicine, Medical
all stroke patients receiving a PEG tube during their acute University of South Carolina, Charleston, South Carolina, USA.
inpatient hospital stay.4 Financial disclosure: None declared.
Despite nutrition and hydration benefits, PEG tubes are
Conflicts of interest: None declared.
associated with short- and long-term complications, such as
skin/wound or abdominal wall infections, tube obstruction, This article originally appeared online on February 3, 2018.
and gastric hemorrhage.5-7 Also, PEG tubes are commonly Corresponding Author:
associated with a decrease in quality of life because patients Heather Shaw Bonilha, PhD, CCC-SLP, Department of Health
Sciences & Research, College of Health Professions, Medical
and their families experience the limitations in the patients’ University of South Carolina, 77 President St. MSC 700, Charleston,
ability to eat as an emotional and social burden.3 Thus, SC 29425, USA.
patients, families, and clinicians who are involved in the Email: bonilhah@musc.edu
554 Nutrition in Clinical Practice 33(4)

stroke patients with and without a PEG tube is crucial for prevent future readmissions.14 Our findings emphasize that
clinicians to adequately counsel patients and their families stroke patients with a PEG tube are at risk for hospital
about what they can expect if or when a PEG tube is placed, readmissions because of clinically significant postacute care
to allocate resources, and plan follow-up visits, including complications.
reevaluations. Based on the currently available evidence, much remains
Once a PEG tube is placed, stroke patients can ex- unknown about the healthcare trajectory of stroke patients
perience a variety of different healthcare trajectories and with a PEG tube after being discharged from the acute care
health outcomes. Patients may show complete recovery of hospital. For instance, we lack an understanding of the
their swallowing abilities with a successful removal of their impact of PEG tube placement on discharge destinations,
feeding tube, or patients may show little or no improvement access to rehabilitation, and hospital readmissions that oc-
of their swallowing abilities with a sustained need of their cur longer than 2 months after the acute stroke. Specifically,
feeding tube, or patients may die as a consequence of feeding access to inpatient rehabilitation facilities (IRFs) seems to
tube/dysphagia-related or unrelated complications.8-10 The be a crucial catalyst for favorable poststroke outcomes.
proportion of patients with poststroke dysphagia who have The American Heart Association/American Stroke Asso-
their PEG tube removed varies broadly between 16.3% and ciation recommend in their recently published guidelines
75% and likely depends on a multitude of patient factors that—if possible—stroke patients should receive treatment
and practice patterns.11 Despite PEG tube removal being in an IRF rather than an SNF because of the better
a primary goal of stroke rehabilitation,12,13 some stroke overall outcomes and higher rate of returning to live in
patients with a PEG tube will never recover their swallowing communities.17
abilities and will be in the need of ongoing healthcare Understanding the trajectory of the care of stroke
support. patients with PEG tubes is crucial for various reasons.
Our previous research has shown that stroke patients During the inpatient hospital stay, clinicians need to take
who receive a PEG tube during their acute hospital stay are the prognosis of an individual patient into account to
twice as likely to be readmitted within 30–60 days to an acute make informed decisions that can have an impact on the
care hospital when compared with stroke patients who did patient’s postacute trajectory (such as placement of a PEG
not receive a PEG tube.14 For some patients, the primary tube) and to provide recommendations for their postacute
medical reason of their hospital readmission was directly care (such as discharge destinations, treatment planning,
linked to their PEG tube. Reducing hospital readmissions resource allocation). In addition, clinicians need to inform
is not only a main incentive of acute care hospitals who patients and their families about possible postacute care
are penalized for 30-day readmissions through the Medicare trajectories to allow them to form realistic expectations and
Hospital Readmission Reduction Program15 but also in the enable informed decision making. Moreover, information
interest of patients because readmissions are a surrogate for about the continuum of care, such as the occurrence of
quality of care. Although several factors other than PEG complications and nonfavorable outcomes (eg, rehospital-
tube placement may contribute to hospital readmissions in izations following preventable complications), may allow
stroke patients,16 we found in a previous study that PEG caregivers and stakeholders to identify key aspects in the
tube placement was an independent risk factor for hospital care of these patients that warrant improvement.
readmissions in stroke patients.14 Thus, the purpose of our study was to provide informa-
When we selectively investigated stroke patients with tion about the continuum of care of stroke patients with
PEG tube placement regarding their risk factors for read- PEG tube placement when compared with stroke patients
missions, we found that discharge locations play a role without PEG tube placement by evaluating their index
in the likelihood that stroke patients with a PEG tube hospital stay and their 1-year care trajectory. We aimed to
will be readmitted. More than half (53.80%) of all stroke build on our previous research on practice patterns and
patients with a PEG tube were discharged from the acute hospital readmissions by expanding the follow-up time to
hospital stay to a skilled nursing facility (SNF), and the vast 1 year, expanding the patient population, and testing our
majority (71.2%) of all 30-day readmitted stroke patients 30-day readmission model in a new patient population. We
with a PEG tube were originally discharged to a SNF. had 3 main objectives in our study. First, we sought to
We confirmed discharge location as one of several risk determine the demographic characteristics, medical charac-
factors in a prediction model for 30-day readmissions for teristics, and rate of PEG tube placements during the index
stroke patients with PEG tube placement. Besides discharge (acute stroke) hospital stay of commercially insured stroke
location, number of comorbidities, hospital length of stay survivors with no history of prestroke dysphagia. Second,
(LOS), and stroke type were predictive for 30-day readmis- we sought to conduct a 1-year follow-up of these patients to
sions. To the best of our knowledge, this is the only study determine their care trajectory by investigating admissions
that assessed hospital readmissions of stroke patients with to IRFs, hospital readmissions, and admission to other
PEG tube placement and that identified opportunities to facility encounters (SNF, outpatient hospital, and home
Wilmskoetter et al 555

visits). Third, we sought to test our previously developed patient’s condition at discharge from the acute hospital stay.
prediction model for 30-day readmissions for stroke patients SASI includes 7 binary indicator variables (aphasia, coma,
with PEG tube placement.14 For all our aims, we sought dysphagia and/or dysarthria, hemiplegia or monoplegia,
to compare stroke survivors with PEG tube placement to neglect, nutritional infusion, tracheostomy, and/or ventila-
patients without PEG tube placement during their index tion) that can be used separately or as a total weighted
hospital stay to understand the specific characteristics of score to control for stroke severity in administrative
patients with poststroke PEG tube placement. Importantly, databases.
as a major improvement of our previous research studies The characteristics of stroke patients with and without
and most stroke outcome studies that are based on ad- PEG tube placement were compared using χ 2 tests and
ministrative databases, we were able to control for stroke Wilcoxon-rank sum tests. Nonparametric tests were ap-
severity by using a novel, validated, stroke severity index plied because all variables were not normally distributed
recently developed by our group specifically for application when tested with the Shapiro-Wilk test and by visual
in administrative databases.18 examination of distribution. To adjust for confounding
variables, multivariable logistic regression modeling was
performed to predict binary outcome variables, and gen-
Methods eralized linear modeling with negative binominal distribu-
We conducted a retrospective analysis of commercially tions and a log link function was performed to predict
insured stroke patients included in the Truven Health hospital LOS.
MarketScan Research Databases. These databases capture Time points of analysis were the index hospital stay, 1
person-specific clinical utilization, expenditures, and enroll- month (30 days), 3 (90 days), 6 (180 days), and 12 months
ment across inpatient, outpatient, prescription drug, and (360 days) postdischarge. Only patients who were enrolled in
carveout services. We analyzed 3 years of the MarketScan the commercial insurance at the time point of analysis were
databases: 2010 was used as the baseline period, 2011 included in the specific analysis. Because patients drop out
was used to identify the index stroke patient cohort, and of the insurance, we used changing cohorts across all time
2012 was used as the follow-up period. We only included points.
patients with a primary diagnosis of stroke in 2011 that For all 4 follow-up time points, we assessed the trajectory
was either represented by an International Classification of care by analyzing inpatient and facility records regarding
of Diseases, Ninth Revision, Clinical Modification (ICD-9- admission rates and diagnoses, time between facility stays,
CM) code of 434.xx for ischemic or 431.xx for hemorrhagic LOS, and discharge destinations. Specifically, we investi-
stroke. PEG tube placement was identified through the gated (1) admissions to IRFs, (2) hospital readmissions, and
ICD-9-CM procedure code 43.11. The 1-year baseline (year (3) SNF, outpatient hospital, and home visit encounters.
2010) before the index hospital stay was used to exclude For hospital readmissions, we separately investigated 30-
patients with a previous stroke, dysphagia diagnosis, feeding day all-cause, unplanned readmissions as defined by the
tube placement, or speech and language pathology (SLP) Center for Medicare and Medicaid Services (CMS), trans-
utilization. To ensure a 1-year baseline and a minimum ferred admissions, and planned readmissions. CMS defines
follow-up of 1 month for all patients, we excluded all stroke “readmission” as an unplanned admission to an hospital
patients who were not enrolled in their insurance at least 1 within 30 days after discharge from the same or another
year prior and 1 month after their index hospitalization in hospital.15 Hospital admissions that represent planned (in-
2011. cluding in-patient rehabilitation) or transferred admissions
Inpatient records for the index hospital stay in 2011 of are excluded from 30-day all-cause readmissions. To identify
stroke patients with and without PEG tube placement were planned readmissions, we used the CMS planned readmis-
analyzed with univariate analyses for demographic (age, sion algorithm (version 2.1, March 2013). This algorithm
gender) and clinical information (ischemic or hemorrhagic defines whether readmissions are planned admissions, that
stroke, comorbidities measured by the Charlson Comorbid- is, if the primary readmission diagnosis is not acute or
ity Index [CCI],19,20 died during hospital stay, LOS, presence not a complication of care. The algorithm lists diagnoses
of dysphagia, discharge destination). We also employed a and procedures using the Clinical Classification Software.
recently developed validated stroke severity measure for use We used the Clinical Classification Software provided by
in archival data, the Stroke Administrative Severity Index the Agency for Healthcare Research and Quality for back
(SASI).18 SASI was developed using the National Institute translation into ICD-9-CM diagnosis and procedure codes
of Health Stroke Scale as a theoretical framework—a (http://www.hcup-us.ahrq.gov/toolssoftware/ccs/ccs.jsp).
commonly clinically used stroke severity scale—and uses To test our previously published prediction model, we
diagnoses and procedure codes at discharge from the acute used multivariable logistic regression analysis to, first, de-
hospital stay. SASI predicts 30-day postdischarge mortality termine whether PEG tube placement is an independent
or discharge to hospice and thus models severity of the predictor for 30-day unplanned readmissions and, second,
556 Nutrition in Clinical Practice 33(4)

16,461 stroke patients in 2011


(Dx 1 = 431.xx or 434.xx)

134 excluded
- patients <18 years

7375 excluded based on baseline 1 year prior index


hospital stay
- 3 with missing data
- 3842 not enrolled for ≥ 1 year prior the index hospital stay
- 3527 not meeting eligibility criteria (3304 diagnosed with prior
stroke, 379 diagnosed with prior dysphagia, 120 had prior
feeding tube placement, 100 had prior SLP utilization)

8955 index stroke patients

44 excluded
- patients with 2 hospital
admissions at the index stroke date

8911 index stroke patients


- 148 patients with PEG tube placement
- 8763 patients without PEG tube placement

Figure 1. Flowchart for patient enrollment for the index patient cohort. Dx 1, primary admission diagnosis; PEG, percutaneous
endoscopic gastrostomy; SLP, speech and language pathologist.

to predict the outcome of 30-day unplanned readmissions study was reviewed by our institutional review board who
in patients with a stroke who had a PEG tube placement determined the study was not human subject research.
during their index hospital stay. We adopted our previously
published model that included the following 4 predictors:
(1) LOS (expressed as a binary variable with a cutoff
point of, median, 11 days), (2) stroke type (hemorrhagic vs Results
ischemic stroke as the admission diagnosis), (3) comorbidi-
ties (measured by the CCI), and (4) discharge destination
Patients
from the index hospital stay. For the predictor “discharge In the MarketScan databases from 2011, 16,461 patients had
destination” we tested discharges to any destination (eg, at least 1 inpatient stay with a primary diagnosis for ischemic
home, IRF, hospice) against discharges to nursing facilities or hemorrhagic stroke (Figure 1). After excluding patients
(eg, SNF), because nursing facilities were the discharge younger than 18 years, patients not fulfilling our 1-year
destinations for the majority of the patients. In addition, baseline criteria and patients with 2 hospital admissions at
to the previously identified significant predictors, we tested the same index stroke date, our final sample included 8911
whether stroke severity (measured by the SASI) is a signifi- stroke patients. Of this sample, 148 patients (1.66%) had a
cant predictor for hospital readmissions and whether adding PEG tube placement during their index hospital stay.
this predictor would improve our model. We assessed model For the follow-up period, the number of enrolled stroke
fit with the Hosmer and Lemeshow test and the model’s patients decreased from 8911 patients at the time of the
discriminatory ability to differentiate between patients who index hospital stay to 5791 (64.99%) patients at the time of
will and will not experience a 30-day hospital readmission the last follow-up at 12 months postdischarge. Table 1 shows
by the area under the receiver operating characteristic curve how many patients were included in each group at each
(AUROC). follow-up time point. At 12 months postdischarge, 39.19%
All analyses were conducted by using SAS statistical of all patients with a PEG tube placement and 65.42% of all
software (version 9.3; SAS Institute, Inc., Cary, NC). The patients without a PEG tube placement were still enrolled
Wilmskoetter et al 557

Table 1. Enrolled and Eligible Patients.

Stroke Patients With Stroke Patients Without


PEG Tube Placement PEG Tube Placement
All Stroke During Index Hospital During Index Hospital
Time point Patients, n (%) Stay, n (%) Stay, n (%)

Index hospital stay 8911 (100) 148 (100) 8763 (100)


1 month postdischarge 8330 (93.48) 133 (89.86) 8197 (93.54)
3 months postdischarge 7773 (87.23) 112 (75.68) 7661 (87.42)
6 months postdischarge 7025 (78.48) 86 (58.11) 6939 (79.19)
12 months postdischarge 5791 (64.99) 58 (39.19) 5733 (65.42)

PEG, percutaneous endoscopic gastrostomy.

Table 2. Patient Demographics and Characteristics.

Group 1: Stroke Group 2: Stroke Absolute Statistical


Patients With PEG Patients Without Difference Difference
Tube Placement, PEG Tube Between the Between the
Variable n = 148 Placement, n = 8763 Groups Groups, P Value

Age (years) Mean (SD) 55.35 (7.2) 54.36 (8.39) 0.99 .2985b
Range 29–64 18–64 NA
Female gender, (vs male) n (%) 58 (39.19) 3,632 (41.45) 3,574 (2.26) .5803a
Ischemic stroke (vs n (%) 94 (63.51) 7,611 (86.85) 7,517 (23.34) <.001a
hemorrhagic)
Length of stay (days) Mean (SD) 17.08 (10.93) 4.35 (5.61) 12.73 <.001b
Median (Range) 14 (1-60) 3 (1-214) 11
Died during hospital stay n (%) 7 (4.73) 328 (3.75) 321 (1.02) .5368a
Charlson Comorbidity Index Mean (SD) 1.55 (1.56) 0.99 (1.49) 0.56 <.001b
(min 0, max 24) Median (Range) 2 (0–10) 0 (1–12) 2
Coded for dysphagia n (%) 69 (46.62) 437 (4.99%) 368 (41.63) <.001a
Stroke administrative severity Mean (SD) 5.22 (5.63) 2.17 (3.54) 3.05 <.001b
index (min 0, max 56) Median (Range) 5 (0–28) 0 (0–35) 5

max, maximum; min, minimum; NA, not applicable; PEG, percutaneous endoscopic gastrostomy.
a χ 2 test.
b Wilcoxon-rank-sum test.

in insurance and, thus, available for the 12-month follow-up Discharge destinations. Figure 2 shows the proportions
analysis. of patients with and without PEG tube placement being
discharged to specific destinations. The largest proportion,
one third (36.5%), of stroke patients with a PEG tube
Index Hospital Stay placement were directly discharged from the index hospital
stay to IRFs. About a quarter (24.3%) of stroke patients
Patient demographics and characteristics. Table 2 shows the with PEG tube placement were directly discharged to SNFs,
demographic and medical characteristics of patients with and about a sixth (15.5%) were discharged to home or home
and without PEG tube placement during their index hospi- healthcare. The vast majority, two thirds (68.3%), of stroke
tal stay. The average age and gender distribution matched patients without a PEG tube placement were discharged to
between the 2 groups and was not statistically different. home or home healthcare. Furthermore, 14% went to IRFs
About the same proportion of patients in both groups died and 2.9% of all discharged patients without a PEG were
during the index hospital stay. Stroke patients with PEG discharged to SNFs.
tube placement were more likely to have had a hemorrhagic Using multivariable logistic regression, PEG tube place-
stroke, longer hospital stays, more comorbidities, higher ment during the index hospital stay was an independent
stroke severity, and were more often coded for dysphagia positive predictor for discharge to IRFs, SNFs, and long-
when compared with stroke patients without PEG tube term care hospitals, but a negative predictor for discharge
placement. to home after controlling for age, gender, stroke severity
558 Nutrition in Clinical Practice 33(4)

15.5
Home
68.3

36.5
Inpatient rehabilitation facility
14

24.3
Skilled nursing facility
2.9

12.2 Stroke patients with PEG


Other destination
10.7 tube placement (N=148)

6.8 Stroke patients without PEG


Long term care hospital
0.4 tube placement (N=8,763)

4.7
Inhospital death
3.7

0 10 20 30 40 50 60 70 80
Percentages

Figure 2. Discharge destinations from the index hospital stay


for stroke patients with and without percutaneous endoscopic
gastrostomy (PEG) tube placement.

(SASI score), comorbidities (CCI score), and stroke type. Figure 3. Unadjusted and adjusted odds ratios for discharge
destinations from the index hospital stay for patients with
Patients with PEG tube placement had higher odds for
percutaneous endoscopic gastrostomy (PEG) tube placement
being discharged to IRFs, SNFs, or long-term care hos- when compared with patients without PEG tube placement.
pitals, but lower odds for being discharged home when Unadjusted: simple logistic regression with PEG tube
compared with stroke patients without PEG tube placement placement as primary predictor; adjusted: multivariable
(Figure 3). logistic regression with PEG tube placement as primary
Stroke severity varied for patients discharged to different predictor and age, gender, stroke severity (Stroke
destinations (Table 3). Overall patients who were discharged Administrative Severity Index score), comorbidities (Charlson
Comorbidity Index score), and stroke type as control variables
home had the lowest severity and patients discharged to
(length of hospital stay and dysphagia were not included
long-term care hospitals the highest stroke severity. Patients because of moderate correlations with the main predictor
with PEG tube placement who were discharged home had PEG tube placement [correlation coefficient r > 0.2; P <
significantly lower stroke severity than patients discharged .0001]).
to IRFs. Patients without PEG tube placement who were
discharged home had significantly lower stroke severity than
patients discharged to other destinations, such as SNFs,
IRFs, or long-term care hospitals. did not significantly contribute to the model for LOS in
IRFs and, thus, were dropped from the final parsimonious
The 1-Year Care Trajectory model.

IRFs. Within 1 month after discharge from the acute care Hospital readmissions. In total, 35% of all stroke patients
hospital, 45.11% (60 of 133) of all stroke patients who with a PEG tube placement and 13% without a PEG tube
had a PEG tube placement during their index hospital placement were admitted to an inpatient facility—excluding
stay had been to an IRF. This was a significantly larger IRFs—during the first month after discharge (Figure 5).
proportion than patients without a PEG tube placement This included transfer admission, 30-day hospital readmis-
of whom 17.32% (1420 of 8197) had been to an IRF sion as defined by CMS, and planned hospital readmissions.
(Table 4). PEG tube placement increased the odds 2-fold The 30-day all-cause, unplanned readmission rate (exclud-
for patients going to IRFs while controlling for confounders ing rehabilitation, transfer, and planned admissions) was
(Figure 4). The cumulative LOS in IRFs was significantly 18% for patients with PEG tube placement and 9% for
longer for stroke patients with PEG tube placement than for patients without PEG tube placement.
patients without PEG tube placement. PEG tube placement After controlling for age, gender, stroke severity (SASI
was an independent predictor of longer LOS in IRFs, score), comorbidities (CCI score), and stroke type, stroke
even after controlling for stroke severity and stroke type patients with PEG tube placement had 1.8 times higher odds
(adjusted mean rehabilitation LOS estimate for patients for a 30-day all-cause, unplanned hospital readmission and
with PEG tube placement was 3.21 days and was 2.77 days; 3.8 higher odds for being transferred to another hospital
P < .0001 without PEG). Age, gender, and comorbidities when compared with stroke patients without PEG tube
Table 3. Average Stroke Severity (Measured by the Stroke

Statistical Difference Test Between Stroke

months

<.001b
<.001a
Patients With and Without PEG Tube
Administrative Severity Index) of Patients Discharged From

12
the Index Hospital Stay to Different Destinations (Ordered by
Increasing Severity of Group 1).

Placement, P Value

months

<.001a

.001b
Group 2: Stroke

6
Discharge Group 1: Stroke Patients Without
Destination From Patients With PEG PEG Tube
Index Hospital Tube Placement, n = Placement, n =

months

<.001b
<.001a
Stay 148; Mean (SD) 8763; Mean (SD)

3
Home 3.91 (5.92) 1.50 (2.68)
Others 4.34 (6.81) 2.41 (3.73)b

<.001b
<.001a
month
Skilled nursing 4.89 (5.24) 3.46 (4.40)b

1
facility
Inpatient 6.00 (4.73)a 4.25 (3.81)b
rehabilitation

(n = 8197) (n = 7661) (n = 6939) (n = 5733)


Stroke Patients With PEG Tube Placement Stroke Patients Without PEG Tube Placement

months

(17.63)

(13.69)
facility

1,011

18.05

1–96
12
Long-term care 7.90 (9.10) 4.06 (4.41)b
hospital

PEG, percutaneous endoscopic gastrostomy.

months

(17.91)

(13.41)
1–136
1,243

17.86
a Significant difference (P ࣘ .05) in stroke severity of patients

6
discharged to this location when compared with patients of the same
group discharged to home.
b Highly significant difference (P < .001) in stroke severity of patients

discharged to this location when compared with patients of the same

months
Table 4. Stays in Inpatient Rehabilitation Facilities After Discharge From the Index Hospital Stay.

(17.88)

(12.81)

IRF, inpatient rehabilitation facility; LOS, hospital length of stay; PEG, percutaneous endoscopic gastrostomy.
1–136
1,370

17.51
group discharged to home.

3
placement. The likelihood for planned readmissions did not

(17.32)

(11.50)
month

1,420

16.93

1–88
differ between the 2 groups (Figure 6).
The most frequent primary readmission diagnosis for 30- 1
day all-cause, unplanned hospital readmissions for stroke (n = 58)
patients with and without PEG tube placement (n = 779)
months

(23.64)
8–111
(48.2)
32.5
12

was cerebral artery occlusion (27.21%), followed by in- 28


tracerebral hemorrhage (4.88%), transient cerebral ischemia
(3.34%), and cerebral embolism with cerebral infarction
(n = 133) (n = 112) (n = 86)
months months

(47.67)

(21.46)
8–111
33.54
(3.21%). The most frequent primary readmission diagnoses
41
6

were the same for patients with and without PEG tube
placement.
(52.68)

(18.12)

Within 1 month after being discharged from the index


30.24

8–82
59
3

hospital stay, patients with a PEG tube placement were,


on average, readmitted to an acute care hospital after 8.04
days (SD 9.04, minimum 0, maximum 30) when com-
(45.11)

(17.16)
month

29.12

6–82

pared with patients without a PEG tube placement who


60
1

were readmitted after 9.14 days (SD 8.73, minimum 0,


maximum 30). The difference between patients with and
Cumulative LOS Mean (SD)

without PEG tube placement was not statistically significant


Range
n (%)

(P = .5028).
b Wilcoxon-Rank-Sum test.

Once readmitted, stroke patients with PEG tube place-


ment had a significantly longer LOS for their first un-
least 1 IRF stay

planned all-cause hospital readmission within 1 month


Patients with at

after discharge when compared with stroke patients without


IRF stays

PEG tube placement (average 12.17 days and 7.68 days,


respectively; P = .0304). However, after controlling for
for all
Variable

a χ 2 test.

stroke severity and stroke type, PEG tube placement did


not have a statistically significant effect the LOS of the first

559
560 Nutrition in Clinical Practice 33(4)

Figure 4. Unadjusted and adjusted odds ratios for admissions to inpatient rehabilitation facilities within 30 days (1 month) after
discharge from the acute hospital for patients with percutaneous endoscopic gastrostomy (PEG) tube placement when compared
with patients without PEG tube placement. Unadjusted: simple logistic regression with PEG tube placement as primary
predictor; adjusted: multivariable logistic regression with PEG tube placement as primary predictor and age, gender, stroke
severity (Stroke Administrative Severity Index score), comorbidities (Charlson Comorbidity Index score), and stroke type as
control variables (length of hospital stay and dysphagia were not included because of moderate correlations with the main
predictor PEG tube placement [correlation coefficient r > 0.2; P < .0001]).

unplanned all-cause hospital readmission as exemplified


by the data from the first month after discharge (P =
.1897; adjusted means estimate for patients with PEG tube
placement was 2.27 days, for patients without PEG tube
placement 1.98 days).

SNF encounter, outpatient hospital visits, and home visits.


Table 5 shows the number and proportion of stroke patients
with and without a PEG tube placement who were admitted
to a SNF and/or had an outpatient hospital visit and/or
home visit. At all follow-up time points, significantly more
stroke patients with a PEG tube were admitted to a SNF
when compared with stroke patients without a PEG tube

25% Figure 6. Unadjusted and adjusted odds ratios for all-cause,


unplanned readmissions as defined by Center for Medicare
% of all index hospital paents with

and Medicaid Services, transfer admissions and planned


one or more inpaent admission

20%

hospital readmissions within 30 days (1 month) after


15% discharge from the acute hospital for patients with
Transfer admissions percutaneous endoscopic gastrostomy (PEG) tube placement
30-day hospital readmission as when compared with patients without PEG tube placement.
10% defined by CMS
Unadjusted: simple logistic regression with PEG tube
Planned hospital readmissions (other
than inpaent rehabiliaon) placement as primary predictor; adjusted: multivariable
5%
logistic regression with PEG tube placement as primary
predictor and age, gender, stroke severity (Stroke
0% Administrative Severity Index score), comorbidities (Charlson
All stroke paents with PEG All stroke paents without
tube placement PEG tube placement Comorbidity Index score), and stroke type as control variables
Type of hospitalizaon (length of hospital stay and dysphagia were not included
because of moderate correlations with the main predictor
Figure 5. Hospitalizations within 1 month after discharge PEG tube placement [correlation coefficient r > 0.2; P <
from the index hospital stay for all patients who were enrolled .0001]).
at 1 month postdischarge. CMS, Center for Medicare and
Medicaid Services; PEG, percutaneous endoscopic
gastronomy.
Wilmskoetter et al 561

Table 5. Number and Proportion of Patients With and Without PEG Tube Placement Who Had a Skilled Nursing Facility Stay,
Outpatient Visit or Home Visit at 1-Month, 3-Month, 6-Month, or 12-Month Follow-Ups.

Visit Patient Group 1 month, n (%) 3 months, n (%) 6 months, n (%) 12 months, n (%)

Skilled nursing facility stay Stroke patients with 30 (23) 40 (36) 32 (37) 23 (40)
PEG tube placement
Stroke patients without 258 (3) 322 (4) 313 (5) 273 (5)
PEG tube placement
Outpatient hospital visit Stroke patients with 40 (30) 78 (70) 67 (78) 52 (90)
PEG tube placement
Stroke patients without 3607 (44) 4887 (64) 5041 (73) 4,581 (80)
PEG tube placement
Home visit Stroke patients with 15 (11) 40 (36) 38 (44) 24 (41)
PEG tube placement
Stroke patients without 753 (9) 916 (12) 904 (13) 790 (14)
PEG tube placement

PEG, percutaneous endoscopic gastrostomy.

Table 6. Multivariable Logistic Regression Model for the Event of an All-Cause, Unplanned 30-Day Readmission for Stroke
Patients With a PEG Tube Placement During Their Index Hospital Stay.

Variable β SE β P Value OR (95% CI)

Intercept −1.55 0.61 .0110 NA


Hemorrhagic stroke 1.25 0.57 .0271 3.50 (1.15–10.61)
Charlson Comorbidity Index 0.22 0.15 .1544 1.25 (0.92–1.68)
Length of hospital stay (ࣙ11 d) −0.95 0.57 .0960 0.39 (0.13–1.18)
Discharge to home 1.32 0.65 .0412 3.73 (1.05–13.21)
Discharge to rehab facility −0.89 0.61 .1468 0.41 (0.12–1.37)
Discharge to other destinations −1.75 1.13 .1197 0.17 (0.02–1.58)

The variable “discharge to” was not included in the model because only 2 patients were coded for this event. β, regression coefficient; NA, not
applicable; PEG, percutaneous endoscopic gastrostomy.

placement (χ 2 test, P < .0001). After controlling for age, gender, stroke severity, stroke type, and comorbidities, PEG
gender, stroke severity, stroke type, and comorbidities, the tube patients had significantly higher odds for home visits at
odds for being admitted to a SNF were 6–8 times higher the last 3 follow-up time points (3 months: OR = 2.50, 95%
for patients with a PEG tube than for patients without a CI,1.66–3.76; 6 months: OR = 3.37, 95% CI, 2.15–5.30; 12
PEG tube (1 month: OR = 5.84, 95% CI, 3.65–9.34; 3 months: OR = 2.69, 95% CI, 1.55–4.68).
months: OR = 7.23, 95% CI, 4.71–11.11; 6 months: OR =
7.48, 95% CI, 4.64–12.07; 12 months: OR = 7.79, 95% CI, Prediction Model for All-Cause, Unplanned
4.39–13.82). 30-Day Readmissions
Patients with PEG tube placement had significantly more
outpatient hospital visits in the first month (P = .0014), but 30-day readmission prediction model for stroke patients with
did not differ at the 3-month, 6-month, and 12-month follow PEG tube placement. We tested our previously developed
ups. After controlling for age, gender, stroke severity, stroke prediction model for 30-day readmissions of stroke patients
type, and comorbidities, PEG tube patients had significantly with PEG tube placement in the stroke patient cohort of
lower odds for outpatient hospital visits at 1 month (OR = the study presented here. The model showed a good model
0.33, 95% CI, 0.21–0.49) and 3 months follow-up (OR = fit with a Hosmer-Lemeshow test P value of .95 (P > .05).
0.573, 95% CI, 0.369–0.890). Significant predictors in the model were hemorrhagic stroke
The number of home visits did not differ between the and discharge destination (Table 6). The AUROC was 0.75
2 groups for the 1-month follow up (P > .05), but did for (Figure 7).
the 3-month, 6-month, and 12-month follow-up time points, Adding the SASI to the prediction model to control
when significantly more patients with PEG tube placement for stroke severity—and deleting the predictor discharge to
had home visits (P < .001; Table 5). After controlling for age, hospice because SASI itself predicts 1-year mortality or
562 Nutrition in Clinical Practice 33(4)

discharge to hospice—improved the discriminative ability


of the model. Adding all SASI indicators separately re-
vealed the best discriminative ability with an AUROC of
0.85 (Hosmer-Lemeshow test: P = 0.69 [P > .05]; Figure 8)
when compared with adding the total SASI score (AUROC
= 0.77; Hosmer-Lemeshow test: P = .83 [P > .05]).
Significant predictors for 30-day readmission for stroke
patients with a PEG tube placement were comorbidities,
hemorrhagic stroke, and presence of hemiplegia and/or
monoplegia (Table 7).

30-day readmission prediction model for stroke patients


without PEG tube placement. We tested the same model
for all-cause, unplanned 30-day readmissions in patients
without PEG tube placement. The Hosmer-Lemeshow test
showed a good model fit with a P value of .48 (P > .05). The
predictors stroke type, CCI, LOS, and discharge destination
were significant (P < .05; Table 8). The AUROC was 0.61
(Figure 9).
Adding the SASI to the prediction model to control for
stroke severity did not improve the discriminative ability of
the model (adding the total SASI score, Hosmer-Lemeshow
Figure 7. Area under the receiver operating curve (ROC) for
test: P = .81 [P > .05]; AUROC = 0.61; adding all SASI
the model to predict all-cause, unplanned 30-day readmissions
indicators separately, Hosmer-Lemeshow test: P = .42 [P > in stroke patients with a percutaneous endoscopic
.05]; AUROC = 0.61). gastrostomy tube placement during their index hospital stay.

Discussion
The purpose of our study was to better understand the
prognosis and trajectory of care of stroke patients who have
a PEG tube placement during their acute hospital stay. Here,
we sought to improve clinicians’ ability to counsel these
vulnerable and high-risk patients and their families when
deciding whether to place a PEG tube, and to identify needs,
plan resources, and prevent potential complications after
placement.
We found that in total only 1.7% of all stroke patients in
our study received a PEG tube during their index hospital
stay, compared with 5.22% of stroke patients admitted to
community hospitals in Florida during 2012 in a previous
study.4 A higher proportion of hemorrhagic than ischemic
stroke patients received a PEG tube (4.5% and 1.2%,
respectively). This is consistent with our previous find-
ings. However, the proportion of ischemic stroke patients
receiving a PEG tube during their index hospital stay
has been reported higher in other studies, with approx-
imately 8% of all ischemic stroke patients.21 The rather
small proportion in this study may be a result of several
factors. Through the availability and linkage of 3 years of Figure 8. Area under the receiver operating curve (ROC) for
MarketScan data, we were able to apply rigorous inclusion the model including the individual Stroke Administrative
and exclusion criteria (eg, exclusion of patients with a prior Severity Index indicators to predict all-cause, unplanned
stroke or prior diagnosis for dysphagia) that we had not 30-day readmissions in stroke patients with a percutaneous
endoscopic gastrostomy tube placement during their index
been able to apply in our previous study. Furthermore, the
hospital stay.
MarketScan databases only include commercially insured
Wilmskoetter et al 563

Table 7. Multivariable Logistic Regression Model Including the Individual SASI Indicators for the Event of an All-Cause
Unplanned 30-Day Readmission for Stroke Patients With a PEG Tube Placement During Their Index Hospital Stay.

Variable β SE β P Value OR (95% CI)

Intercept −1.66 0.84 .0475 NA


Hemorrhagic stroke 1.44 0.64 .0233 4.22 (1.22–14.65)
Charlson Comorbidity Index 0.48 0.18 .0067 1.62 (1.14–2.28)
Length of hospital stay (ࣙ11 d) −1.12 0. 64 .0817 0.33 (0.09–1.15)
Discharge to home 1.02 0.69 .1377 2.77 (0.72–10.63)
Discharge to rehab facility −1.21 0.72 .0917 0.30 (0.07–1.22)
Discharge to other destinations −1.72 1.18 .1444 0.18 (0.02–1.80)
SASI aphasia −0.75 0.83 .3704 0.47 (0.09–2.43)
SASI coma −0.17 1.27 .8916 0.84 (0.07–10.19)
SASI dysphagia and/or dysarthria 1.13 0.63 .0724 3.08 (0.90–10.52)
SASI hemi- and/or monoplegia −2.29 0.78 .0034 0.10 (0.02–0.47)

The 3 SASI indicators tracheostomy and/or ventilation, nutritional infusion, and neglect were not included in the model because ࣘ2 patients were
coded for these indicators. β, regression coefficient; NA, not applicable; PEG, percutaneous endoscopic gastrostomy; SASI, Stroke Administrative
Severity Index.

Table 8. Multivariable Logistic Regression Model for the Event of an All-Cause Unplanned 30-Day Readmission for Stroke
Patients Without a PEG Tube Placement During Their Index Hospital Stay.

Variable β SE β P Value OR (95% CI)

Intercept −2.27 0.13 <.0001 NA


Hemorrhagic stroke 0.32 0.11 .0028 1.38 (1.12–1.71)
Charlson Comorbidity Index 0.11 0.02 <.0001 1.11 (1.06–1.17)
Length of hospital stay (ࣙ11 d) 0.77 0.13 <.0001 2.16 (1.67–2.79)
Discharge to home −0.30 0.13 .0234 0.74 (0.58–0.96)
Discharge to rehab facility −0.50 0.16 .0018 0.61 (0.44–0.83)
Discharge to hospice −0.76 0.76 .3204 0.47 (0.11–2.09)
Discharge to other destinations 0.41 0.17 .0149 1.50 (1.08–2.08)

β, regression coefficient; NA, not applicable; PEG, percutaneous endoscopic gastrostomy.

patients and do not include patients from other providers, follow-up care, thus, supporting our focus on this group of
such as Medicare or Medicaid. Consequently, the patients patients.
in the cohort of our present study were younger overall For our follow-up analyses, we observed a high dropout
(54.37 years compared with 71.36 years), had fewer co- of patients. At 12 months postdischarge, 35% of all patients
morbidities (CCI was 1.00 compared with 3.04), shorter without and 61% of all patients with PEG tube placement
LOS (mean 4.56 days compared with 5.62 days), and a were no longer enrolled in the private insurance and conse-
smaller proportion died during the index hospital stay quently lost for follow-up. The remarkably higher dropout
(3.77% compared with 5.77%). These differences suggest in the group of PEG tube patients likely reflects the higher
that the MarketScan patient cohort was generally healthier, severity of this group. A dropout of the insurance can, for
which likely contributed to the lower rate of PEG tube example, be caused through job loss, causing the patient to
placements.22,23 switch from the job-provided private insurance to another
In our study, patients with and without PEG tube place- insurance, such as Medicare.
ment during their acute hospital stay differed with respect Our main objective was to investigate the trajectory of
to several medical characteristics. A higher proportion of care for stroke patients with PEG tube placement. The
patients with PEG tube placement had hemorrhagic strokes, trajectory of care after the acute care hospital stay starts
more severe strokes (SASI), more comorbidities, and stayed with the discharge destination. Discharge destinations are
longer in the hospital than patients without PEG tube crucial for the outcome of stroke patients because they can
placement. Patients who received a PEG tube were overall significantly impact patients’ recovery as a result of varia-
in a more severe acute medical condition and, as a result of tions in treatment access. Of the patients with PEG tube
this, may demand higher levels of specialized and intense placement, 60%—compared with only 16.9% of patients
564 Nutrition in Clinical Practice 33(4)

without PEG tube placement—were directly discharged


from the acute care hospital to higher intense care/treatment
facilities, with 36.5% to IRFs and 24.3% being discharged
to SNFs. These discharge patterns differ from our previous
research of stroke patients discharged from community
hospitals in Florida in 2012. In our previous study, only
15.4% of stroke patients with a PEG tube placement were
discharged to IRFs and 54.3% to SNFs.14 Again, we specu-
late that this difference originates from differences in stroke,
age, and overall medical conditions. Whether a patient is
discharged to IRFs or SNFs is influenced by their recovery
potential. Patients with a better prognosis and who can
participate in a daily program of intense therapy (usually at
least 3 hours of rehabilitation training a day) will be more
likely to be discharged to IRFs. Thus, we speculate that
patients from the MarketScan cohort presented here had
a better recovery potential and prognosis based on factors
such as younger age and fewer comorbidities and, thus,
more patients were eligible for IRFs. Future research should
address factors that contribute to the decision making on
discharge destinations for stroke patients with PEG tubes.
When combining patients who were directly discharged
to IRFs and patients who were admitted later, we found that Figure 9. Area under the receiver operating curve (ROC) for
half of all stroke patients with a PEG tube placement had the model to predict all-cause, unplanned 30-day readmissions
a least 1 stay in an IRF within 1 year after their discharge in stroke patients without a percutaneous endoscopic
gastrostomy tube placement during their index hospital stay.
from the acute care hospital. Stroke patients with PEG
tube placement were also twice as likely to have at least
1 stay in an IRF and that stay was significantly longer example, Medicare reimburses SLP treatment for stroke
when compared with stroke patients without PEG tube patients in SNF, rehabilitation facilities, and for home
placement, after controlling for stroke severity, comorbidi- healthcare up to 100 days poststroke (www.medicare.gov).
ties, and other confounders. Thus, our findings suggest that In contrast, patients who are discharged to other destina-
patients with PEG tube placement are preferably discharged tions, had their stroke >100 days ago, or have private insur-
(or later admitted) to IRFs when compared with their ance do not necessarily receive coverage for SLP services.
counterparts without a PEG tube placement. This finding Unfortunately, the MarketScan database does not specify
warrants verification in other studies, but suggests that swallowing treatment—or SLP treatment in general—and,
healthcare providers identify the high need of rehabilitation thus we were not able to assess if and how often patients
in patients with PEG tube placement. However, still 50% received swallowing treatment.
of stroke patients with PEG tube placement did not receive Our findings suggest opportunities for those patients
rehabilitation treatment at an IRF. Further clarification is who may not receive rehabilitation through inpatient or
needed on whether these patients had received rehabilitation outpatient services. Our data showed that 35% of PEG
through another facility (eg, subacute rehabilitation in a tube patients were admitted to some kind of inpatient
SNF) or through outpatient rehabilitation. Because of the hospital stay within 1 month, meaning that 1 of 3 patients
associated risks, complications, and decrease in quality life, with an initial PEG tube placement will have access to
PEG tube removal in conjunction with swallow recovery is a inpatient healthcare professionals within a few weeks after
main goal of poststroke rehabilitation. Swallowing therapy discharge from their index hospital stay. Some of these
is an agreed-on facilitator of feeding tube removal, even in inpatient admissions were a result of complications and
chronic stroke patients.12,13,24-27 Therefore, any patient with worsening of the medical status with a recurrent stroke
a PEG tube who does not receive swallowing therapy is less being the most frequent readmission reason. Therefore,
likely to be weaned of their PEG tube and consequently is patients may not present their full recovery potential at that
at higher risks for PEG tube–associated complications than time. Nevertheless, we believe that the high frequency of
patients who receive swallowing therapy. inpatient encounters of PEG tube stroke patients could
Indeed, not all stroke patients receive swallowing treat- serve as an ideal opportunity to provide reevaluations of
ment because access to SLP treatment depends on multiple their swallowing ability and need of PEG tube. Some
factors, such as insurance and discharge destination. For patients may not receive reevaluations otherwise (as this
Wilmskoetter et al 565

service is often limited outside of hospitals) and, as a con- In a previous study, we were able to develop a prediction
sequence, will not be identified as candidates for swallowing model for 30-day all-cause, unplanned readmissions of
therapy or PEG tube removal. stroke patients who had PEG tube placement during their
Besides transfers to other hospitals, the largest propor- index hospital stay. The model showed a good discriminative
tion of hospital admissions within 1 month after discharge capability as tested in stroke patients admitted to commu-
from the index hospital stay were unplanned all-cause nity hospitals in Florida with an AUROC of 0.81.14 In the
readmissions. Twice as many patients with PEG tube place- study presented here, we were able to test our model in
ments (18%) had an all-cause, unplanned readmission when a patient cohort of commercially insured younger stroke
compared with patients without PEG tube placement (9%). patients with PEG tube placement. The model showed
These readmissions excluded rehabilitation, transfer, and a comparable discriminative capability for 30-day read-
planned admissions. These findings replicate our previous missions with an AUROC of 0.76. Thus, we successfully
study of hospital readmissions of stroke patients who were validated our prediction model in an independent dataset
discharged from community hospitals in Florida in 2012, with a different stroke patient population. Adding SASI to
where we also found that twice as many patients with PEG our prediction model improved its discriminative ability for
tube placement (21.06%) were readmitted within 30 days 30-day all cause unplanned readmissions for stroke patients
when compared with patients without PEG tube placement with PEG tube placement. However, the same model did
(10.84%).14 Building on our previous analysis, we were able not show a satisfying discriminative ability for 30-day all
to show that PEG tube placement is an independent predic- cause unplanned readmissions for stroke patients without
tor increasing the odds 2-fold for 30-day readmissions even PEG tube placement. This was not surprising as the model
after controlling for stroke severity and several other patient was originally developed for stroke patients with PEG
and medical factors. Previous studies had to consider PEG tube placement. Thus, our findings suggest that develop-
tube placement as a surrogate for stroke severity instead of ing prediction models for hospital readmissions should be
an independent predictor because of the lack of a proper population specific to reach the best possible discriminative
stroke severity measure. To our knowledge, our study is ability.
the first study that can verify the independent contribution
of PEG tube placement to 30-day readmissions. The most
common causes for 30-day unplanned readmissions were Limitations
recurrent strokes (cerebral artery occlusions, hemorrhages, Our study has several limitations that are a characteristic
transient ischemic attacks) in both groups of patients to administrative databases. There is a potential for as-
with and without PEG tube placement. In our previous certainment bias with possible mismatches between what
study, cerebral artery occlusion and septicemia followed is coded in the database and true clinical events. As an
by transient ischemic attacks were the leading causes of example, in our study, 5.7% of 8911 stroke patients were
hospital readmission for stroke patients without PEG tube coded for dysphagia at discharge from their index hospital
placement, and septicemia and food/vomit pneumonitis stay. Strikingly, this is in contrast to various clinical studies
followed by cerebral artery occlusion for stroke patients with that have shown that 50%–78% of all acute stroke pa-
PEG tube placement.14 The slight differences in the most tients present dysphagia.28,29 Undercoding of dysphagia in
common readmission diagnoses between our 2 studies indi- archival data is a well-known problem30 that has prevented
cate limitations for generalizations across different patient us from including this variable in any of our outcome
cohorts and datasets. analyses. Another example is the removal of PEG tubes.
The 1-year care trajectory revealed that stroke patients Few patients were coded for PEG tube removal in either
with PEG tube placement were 8 times more likely to have inpatient, outpatient, or facility billing files. We suspect that
a SNF stay after controlling for age, gender, comorbidities, 1 reason for this may be that PEG tube removal might not be
stroke severity, and stroke type. The reasons for this striking coded during an office visit or can happen accidentally apart
difference between the 2 groups remain speculative. It is from a medical encounter. The absence of reliable coding
to be expected that factors such as family support, level for PEG tube removal in administrative data excludes that
of dependency, and recovery prognosis play a role in the variable from analyses.
decision to admit patients to skilled nursing facilities. The Furthermore, we had to use changing cohorts in the 1-
same factors might also affect the initial decision to place month, 3-month, 6-month, and 12-month follow-up analy-
a PEG in the acute care hospital stay. Unfortunately, we ses as a result of patients dropping out of the commercial
were not able to assess these factors within the MarketScan insurance and, thus, being lost to longer term follow-up in
data. Future studies are warranted to investigate medical our analysis. As a consequence, a direct comparison of the
and nonmedical factors contributing to poststroke facility inpatient care that stroke patients received at the different
stays. follow-up time points is not possible in these data.
566 Nutrition in Clinical Practice 33(4)

Conclusions 11. Wilmskoetter J, Herbert TL, Bonilha HS. Factors associated with
gastrostomy tube removal in patients with dysphagia after stroke. Nutr
To our knowledge this is the first study investigating the Clin Pract. 2017;32(2):166-174.
care trajectory for stroke patients with and without PEG 12. Buchholz AC. Weaning patients with dysphagia from tube feeding
to oral nutrition: a proposed algorithm. Can J Diet Pract Res.
tube placement using administrative databases. Our study
1998;59(4):208-214.
provides unique insights into differences between these 2 13. Crary MA, Groher ME. Reinstituting oral feeding in tube-fed adult
patient groups. The study results improve our understand- patients with dysphagia. Nutr Clin Pract. 2006;21(6):576-586.
ing of what clinicians and—more important—patients can 14. Wilmskoetter J, Simpson KN, Bonilha HS. Hospital readmissions
expect within 1 year following a stroke and can aide in of stroke patients with percutaneous endoscopic gastrostomy feeding
tubes. J Stroke Cerebrovasc Dis. 2016;25(10):2535-2542.
predicting recovery, planning resources, and identifying
15. Centers for Medicare & Medicaid Services (CMS). Readmissions
gaps, risks, and points for improvement in stroke care Reduction Program (HRRP) 2016. https://www.cms.gov/medicare/
itself. medicare-fee-for-service-payment/acuteinpatientpps/readmissions-
reduction-program.html. Accessed March 7, 2016.
16. Lichtman JH, Leifheit-Limson EC, Jones SB, et al. Predictors
Statement of Authorship
of hospital readmission after stroke: a systematic review. Stroke.
Janina Wilmskoetter, Annie Simpson and Heather Bonilha 2010;41(11):2525-2533.
contributed to the conception and design of the research; Kit 17. Winstein CJ, Stein J, Arena R, et al. Guidelines for adult stroke
Simpson contributed to the design of the research; Janina rehabilitation and recovery: a guideline for healthcare professionals
Wilmskoetter, Annie Simpson, Sarah Logan and Kit Simp- from the American Heart Association/American Stroke Association.
Stroke. 2016;47(6):e98-e169.
son contributed to the acquisition and analysis of the data;
18. Simpson AN, Wilmskoetter J, Hong I, et al. Stroke administrative
Janina Wilmskoetter, Annie Simposn and Heather Bonilha
severity index: Using administrative data for 30-day poststroke out-
contributed to the interpretation of the data. Janina Wilm- comes prediction. J Comp Eff Res. 2017. https://doi.org/10.2217/cer-
skoetter drafted the manuscript; all authors critically revised 2017-0058 [Epub ahead of print].
the manuscript, agree to be fully accountable for ensuring the 19. Quan H, Li B, Couris CM, et al. Updating and validating the Charlson
integrity and accuracy of the work, and read and approved the Comorbidity Index and score for risk adjustment in hospital discharge
final manuscript. abstracts using data from 6 countries. Am J Epidemiol. 2011;173(6):676-
682.
20. Quan H, Sundararajan V, Halfon P, et al. Coding algorithms for
References defining comorbidities in ICD-9-CM and ICD-10 administrative data.
1. Mozaffarian D, Benjamin EJ, Go AS, et al. Heart disease and stroke Med Care. 2005;43(11):1130-1139.
statistics—2015 update: a report from the American Heart Association. 21. George BP, Kelly AG, Schneider EB, Holloway RG. Current practices
Circulation. 2015;131(4):e29-e322. in feeding tube placement for US acute ischemic stroke inpatients.
2. Centers for Disease Control and Prevention (CDC). Prevalence and Neurology. 2014;83(10):874-882.
most common causes of disability among adults—United States, 2005. 22. Dubin PH, Boehme AK, Siegler JE, et al. New model for predicting
MMWR Morb Mortal Wkly Rep. 2009;58(16):421-426. surgical feeding tube placement in patients with an acute stroke event.
3. Dennis M, Lewis S, Cranswick G, Forbes J. FOOD: a multicentre Stroke. 2013;44(11):3232-3234.
randomised trial evaluating feeding policies in patients admitted to 23. Li J, Zhang J, Li S, Guo H, Qin W, Hu WL. Predictors of percutaneous
hospital with a recent stroke. Health Technol Assess. 2006;10(2):iii-iv, endoscopic gastrostomy tube placement after stroke. Can J Neurol Sci.
ix-x, 1–120. 2014;41(1):24-28.
4. Wilmskoetter J, Simpson AN, Simpson KN, Bonilha HS. Practice 24. Carnaby G, Hankey GJ, Pizzi J. Behavioural intervention for dys-
patterns of percutaneous endoscopic gastrostomy tube placement in phagia in acute stroke: a randomised controlled trial. Lancet Neurol.
acute stroke: are the guidelines achievable. J Stroke Cerebrovasc Dis. 2006;5(1):31-37.
2016;25(11):2694-2700. 25. Huckabee ML, Cannito MP. Outcomes of swallowing rehabilitation in
5. Janes SEJ, Price CSG, Khan S. Percutaneous endoscopic gastros- chronic brainstem dysphagia: A retrospective evaluation. Dysphagia.
tomy: 30-day mortality trends and risk factors. J Postgrad Med. 1999;14(2):93-109.
2005;51(1):23-29. 26. Shaker R, Easterling C, Kern M, et al. Rehabilitation of swallowing by
6. Allison MC, Morris AJ, Park RH, Mills PR. Percutaneous endoscopic exercise in tube-fed patients with pharyngeal dysphagia secondary to
gastrostomy tube feeding may improve outcome of late rehabilitation abnormal UES opening. Gastroenterology. 2002;122(5):1314-1321.
following stroke. J R Soc Med. 1992;85(3):147-149. 27. Elmstahl S, Bulow M, Ekberg O, Petersson M, Tegner H. Treatment of
7. Powell-Tuck J, van Someren N. Enterostomy feeding for patients with dysphagia improves nutritional conditions in stroke patients. Dyspha-
stroke and bulbar palsy. J R Soc Med. 1992;85(12):717-719. gia. 1999;14(2):61-66.
8. Scolapio JS, Romano M, Meschia JF, Tarrosa V, Chukwudelunzu FE. 28. Martino R, Foley N, Bhogal S, Diamant N, Speechley M, Teasell R.
PEG feeding tube placement following a stroke: when to place, when to Dysphagia after stroke: incidence, diagnosis, and pulmonary compli-
wait. Nutr Clin Pract. 2000;15(1):36-39. cations. Stroke. 2005;36(12):2756-2763.
9. Ickenstein GW, Kelly PJ, Furie KL, et al. Predictors of feeding 29. Daniels SK, Foundas AL. Lesion localization in acute stroke patients
gastrostomy tube removal in stroke patients with dysphagia. J Stroke with risk of aspiration. J Neuroimaging. 1999;9(2):91-98.
Cerebrovasc Dis. 2003;12(4):169-174. 30. Gonzalez-Fernandez M, Gardyn M, Wyckoff S, Ky PK, Palmer JB.
10. Yi Y, Yang EJ, Kim J, Kim WJ, Min Y, Paik NJ. Predictive factors for Validation of ICD-9 code 787.2 for identification of individuals with
removal of percutaneous endoscopic gastrostomy tube in post-stroke dysphagia from administrative databases. Dysphagia. 2009;24(4):398-
dysphagia. J Rehabil Med. 2012;44(11):922-925. 402.
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 4
Strategies for the Enhancement of Nutrition Practice August 2018 567–575

C 2017 American Society for

in a New York State Level 1 Trauma Center: A Hospital’s Parenteral and Enteral Nutrition
DOI: 10.1177/0884533617724144
Journey wileyonlinelibrary.com

Lisa Musillo, MS, RDN, CNSC1 ; Laryssa Marie Grguric, RDN, CNSC1 ;
Edward Coffield, PhD2 ; Frank Aversano, MD3 ; Jeremy Bosworth, MD3 ;
and Richard Batista, MD3

Abstract
Background: Provision of enteral nutrition (EN) support is historically inadequate in the critically ill population. An interdisci-
plinary approach utilizing various strategies has been shown to improve initiation of timely EN support. The purpose of this
study was to examine whether the implementation of a series of interventions led by an interdisciplinary team was associated
with changes in the initiation of nutrition support in a level 1 trauma center. Methods: Patients admitted between 2009 and 2013
with isolated closed head trauma injuries were identified through the hospital’s trauma center database. The initial population
consisted of 159 patients; after exclusion criteria, 141 patients were included in the statistical analyses. Two statistical analyses
were conducted. The first calculated the average days to the initiation of nutrition start by admission year. The second estimated
the association between admission year and time to nutrition initiation with a generalized linear model. Results: Time to initiate
nutrition therapy was estimated to decrease by 1.46 days (47.31%) from 2009 to 2013. The time to initiate nutrition in 2013 was
1.63 days. A significant association was found between the time to initiate nutrition and the 2012 and 2013 binary variables while
controlling for confounding variables. The time frame was estimated to be 1.09 (P = .008) and 1.75 (P = .000) days shorter in 2012
and 2013 relative to 2009. Conclusions: An interdisciplinary effort utilizing multiple strategies identified and addressed barriers,
resulting in a reduction of variability and a proactive approach to early EN. (Nutr Clin Pract. 2018;33:567–575)

Keywords
clinical protocols; closed head injuries; critical illness; enteral nutrition; nutritional support

Nutrition during critical illness is 1 of many therapies ternational survey. This facility participated in the 2007
that influence patient outcomes. As described in the 2016 study “Improving the Practice of Nutrition Therapy in the
guidelines for the provision and assessment of nutrition Critically Ill: An International Quality Improvement Project
support therapy, “delivery of early nutrition support ther- (IQIP).”5 Participation in this project allowed an assessment
apy, primarily by the enteral route, is seen as a proac- of our processes for initiating and providing adequate
tive therapeutic strategy that may reduce disease severity, nutrition therapy to our patients, relative to other sites. Our
diminish complications, decreased length of stay in the performance in this survey was used as a springboard for
intensive care unit (ICU) and favorably impact patient change within our facility.
outcomes.”1 The initiation and provision of nutrition during
clinical treatment fall well below established benchmarks.1,2
From the 1 Department of Food and Nutrition, Nassau University
Addressing the deficiencies in critical care nutrition support
Medical Center, East Meadow, New York, USA; 2 Department of
requires universal and local strategies to minimize and Health Professions, Hofstra University, Hempstead, New York, USA;
possibly prevent complications.3,4 and 3 Department of Surgery, Nassau University Medical Center,
From a local or single hospital perspective, the strategies East Meadow, New York, USA.
within our ICU to address nutrition insufficiency during Financial disclosure: None declared.
critical care were developed by an interdisciplinary team Conflicts of interest: None declared.
and implemented and refined over a multiyear period.
This article originally appeared online on September 27, 2017.
After the critical care dietitian identified deficiencies in
Corresponding Author:
nutrition provision, it was apparent that study participation
Lisa Musillo, MS, RDN, CNSC, Department of Food and Nutrition,
was necessary to compare the facility against international Nassau University Medical Center, 2201 Hempstead Tpk, East
nutrition practice. The dietitian then became instrumental Meadow, NY 11554, USA.
in arrangement of survey participation in a multisite in- Email: lmusillo@numc.edu
568 Nutrition in Clinical Practice 33(4)

Table 1. Challenges in Providing Adequate Nutrition Therapy in a Level 1 Trauma Center.

Challenge Description

Knowledge deficit r Inadequate knowledge of nutrition therapy policies and practices across all disciplines
r Unnecessary delays and/or withholding of tube feeding based on gastric residual volume
r Inadequate advancement of tube feeding per provider order of “as tolerated” or “ramp up”
r Infrequent use of promotility agents to enhance tube feeding tolerance
r Insufficient nursing communication between shifts regarding tube feeding tolerance and advancement
r Infrequent usage of rectal tube system, prompting withholding of feeding for diarrhea
r Lack of interest or awareness of the importance of timely nutrition therapy
Time lost to r Lost hours/days to anticipated extubation, lengthy tests and procedures/operations
clinical care r Preoperative/procedure fasting
r Canceled cases resulting in unnecessary fasting
r Tube feeding practices during hemodialysis and sustained low-efficiency dialysis
Equipment issues r Tube feeding pump availability and usage issues
r Lack of availability of jejunally placed feeding tubes

This retrospective study details the processes, challenges, disciplines, a lack of awareness regarding nutrition initiation
and barriers that we encountered while identifying barriers and/or adequacy, access to feeding devices and tools, and the
to adequate nutrition therapy (Table 1) and developing delay/interruption of nutrition provision due to common
and implementing interventions intended to address these ICU events. Examples include patients having undergone
barriers (Table 2) through an interdisciplinary team ap- major surgery or trauma, immediate preextubation and pos-
proach. The association between the interventions and time textubation, patients receiving dialysis, patients with severe
to nutrition initiation was examined with a sample of 141 sepsis requiring vasopressor support, perceived high gastric
admitted patients with isolated closed head trauma injuries residual volumes (GRVs), infrequent use of promotility
from 2009 to 2013. The results from this study illustrate agents, lack of diet advancement, inefficient staff commu-
how an internal focus on quality improvement, led by nication, no postpyloric feeding tube access, and lack of
an interdisciplinary team, has changed our practices to enteral nutrition (EN) pump availability.4 The interventions
improve nutrition support and patient care. intended to address these barriers are listed in Table 2. For
the IQIP study, each participating center identified its goal
provision/adequacy based on its facility’s standards.
Background and Changes One of the first interventions, introduced in 2008, ad-
Nutrition provision challenges within our facility were first dressed the education component. Hospital staff were pro-
identified with IQIP. The IQIP study results were released vided with nutrition lectures conducted by the critical care
to our hospital administration in 2007, which prompted dietitian. These lectures stressed the importance of prompt
the formation of an interdisciplinary team consisting of initiation and advancement of nutrition therapy. In 2008,
members from the nutrition, surgery, medicine, and nursing the hospital also introduced “tube feeding pump centers.”
departments. The critical care dietitian was the lead team The purpose of these centers was to increase access to and
member. This team was tasked with identifying barriers availability of EN pumps and tube feeding products and
to nutrition therapy and then formulating a quality im- accessories. It was hypothesized that facilitated access via
provement plan. After identification of barriers to nu- these pump centers would minimize the amount of time that
trition implementation in 2008, intervention mechanisms staff allocated to search for the required items to initiate and
were applied to improve patient care. The team evaluated maintain nutrition support.
progress as time passed, and it adjusted the improvement The third intervention introduced in 2008 was the pur-
interventions as required. Hospital leadership and team chase, by the Department of Surgery, of an electromag-
members supported this evaluation-intervention-evaluation netic feeding tube placement device to facilitate postpyloric
approach, although it was challenging from time and re- feeding tube placements.6 The purchase of this device was
source perspectives. intended to increase the ease and efficiency of nasojeju-
The team identified a number of barriers to nutrition nal/orojejunal tube placements. The expectation was that
therapy through IQIP (eg, time to initiation of nutrition the device would facilitate accessibility and efficacy and
therapy and adequacy of provision; Table 1). The main thereby increase the quantity of feeding tubes placed at
barriers were inadequate nutrition education across all an earlier time. Placements were done by surgical residents
Musillo et al 569

Table 2. Nutrition Therapy Quality Improvement Tasks and Interventions by Admission Year.

Year Task/Intervention

2007 Participation in “Improving the Practice of Nutrition Therapy in the Critically Ill: An International Quality
Improvement Project” (IPNT)
2008 IPNT results released and disseminated to administration
From IPNT results, identified deficiencies in nutrition therapy practice relative to other sites and critical care guidelines
Multidisciplinary team formed
Identification of barriers to adequate nutrition therapy
Development and implementation of improvement plan
Tube feeding pump centers created
Purchase and introduction of electromagnetic feeding tube placement device to assist in placement of jejunally placed
feeding tubes at bedside
Updated preoperative fasting policy
Nutrition therapy educational lectures for all clinical staff
Preoperative fasting guideline liberalized to 2 h with jejunally placed feeding tubes (Oct 2009)
2009 New physician hired as director of Department of Medicine and Critical Care Committee
New director of Department of Medicine advocated for change in nutrition practices (Nov 2009)
2010 Critical care nutrition snapshot (eg, review of practices) completedResults presented at Critical Care Committee
meetings
Repeat data collection presented at each meeting moving forward (Feb 2010–Mar 2010)
“Pending verification” order status added, allowing registered dietitian nutritionists to place nutrition-specific orders
into the electronic medical record for physician approval (Mar 2010)
2011 Enrollment in the “Enhanced Protein-Energy Provision via the Enteral Route in Critically Ill Patients (PEP uP
Protocol)” trial (Jul 2011)
2012 Official “rollout” of modified feeding protocol to burn center, coronary care unit, medical intensive care unit, surgical
intensive care unit (fall 2011–winter 2012)
Preoperative fasting guideline further liberalized to no tube feeding withholding when provided by jejunally placed
feeding tubes (Oct 2012)
2013 Registered dietitian nutritionists granted privilege to place jejunally placed feeding tubes using electromagnetic feeding
tube placement device (fall 2013)

(in conjunction with a surgical technician) or surgical data collection to compare with preintervention outcomes.
physician’s assistants under the supervision of a surgical Improvement was evident; however, the team identified fur-
attending. ther improvements that were needed to achieve acceptable
The intervention mechanisms implemented in 2008 were benchmarks. One of the first identified steps needed to fur-
maintained once implemented and included continued nu- ther improve nutrition initiation was a more constant data
trition therapy education lectures. The continuation of these surveillance procedure. Moving forward, data collections
education sessions provided new staff with nutrition therapy were completed every 2 months by the Nutrition Service.
education and updated existing staff on any procedure or The key metrics collected were the time to initiate nutrition
policy change. The educational sessions were seen as a key and provision with an EN pump. Nutrition provision data
component to ensure that all staff members worked together were collected with EN pumps with long-term memory
to improve the timeliness and adequacy of nutrition therapy. features, which could be accessed to collect the actual
This culture was further supported by the addition of a volume of EN delivered. The use of the EN pumps for this
new Department of Medicine chair in 2009, who strongly purpose also initiated the need to ensure the accuracy of the
advocated for improvements in nutrition therapy for the data collected. There are discrepancies between documented
critically ill based on our hospital’s performance in the IQIP EN delivery and the actual amount provided to patients.10
study. The final intervention implemented in 2009 was an The bimonthly time to initiation of nutrition therapy data
update of the anesthesia preoperative fasting policy. This was started in 2010 and is ongoing and regularly presented
updated policy shortened the time frame of EN withholding at committee meetings.
to 2 hours prior to any surgical or invasive procedure for In an attempt to further improve outcome metrics, an
patients with jejunally placed feeding tubes.7-9 Prior to this additional change in 2010 was a “pending verification”
update, the withholding time was a minimum of 8 hours. order status in the electronic medical record (EMR). The
In 2010, the team conducted a systematic review of team worked with the information technology department
the nutrition therapy program. This included a repeat to develop an order entry field that allowed registered
570 Nutrition in Clinical Practice 33(4)

dietitian-nutritionists (RDN) to input nutrition-related research.7-9 Coming into 2013, the dietitians were granted
orders (eg diets, tube feedings, and supplements). This privileges to place postpyloric feeding tubes with the elec-
process was expected to improve the timing of diet order tromagnetic feeding tube device. This was implemented to
changes and the implementation of nutrition support and increase the number of feeding tubes placed in a timely
decrease the amount of nutrition-related order errors. manner.
The review of outcomes and implementation of inter- The changes implemented throughout the quality im-
ventions or adjustments to existing practices continued in provement initiative were intended to improve the hospital’s
2011. In this year, permission was granted by the hospital’s nutrition practice. This study examined the timing of nutri-
administration for participation in a multicenter cluster- tion support in an isolated closed head injury population to
randomized trial entitled “Enhanced Protein-Energy Pro- examine if a relationship existed between quality improve-
vision via the Enteral Route in Critically Ill Patients: A ment measures implemented and current nutrition practice.
Single Center Feasibility Trial of the PEP uP Protocol.”11 Specifically, the study examined whether time to initiate
This feeding strategy was different from the method of nutrition was associated with binary year variables, which
EN administration being used at the time in the ICU. presumably measured differences in nutrition practice over
The standard method was a continuous static rate with no time. The interventions were viewed as a collective whole
compensation for lost volume due to EN withholding. The and were examined as such, although days to nutrition
new protocol implemented a volume-based feeding strategy. initiation were estimated annually across the examined
The purpose of the PEP uP protocol was to expedite period.
the initiation of nutrition support and adequate calorie and
protein provision for critically ill patients in an attempt to
improve patient outcomes. The protocol emphasizes early
Methods
enteral access, initiation and formula choice, dosing by This was an expedited study approved by the Nassau
volume with patient weight, trophic EN during inotropic University Medical Center Institutional Review Board. The
therapy, built-in method of infusion rate changes based on sample population was drawn from a 500-bed regional
the actual volume delivered, proactive use of prokinetic level 1 trauma center in Nassau County, New York. For
agents, and the use of GRVs and diarrhea algorithms. The inclusion in this retrospective study, patients had to be
PEP uP protocol also empowered nursing to achieve the admitted between 2009 and 2013 with isolated closed head
desired daily EN goal. Institutional Review Board approval trauma injuries. Patients meeting these criteria were iden-
was obtained, and the study commenced in summer of tified through the hospital’s trauma center database. Once
2011. The medical ICU was the pilot location. The team identified, the medical records of each patient were reviewed
received positive feedback on the PEP uP protocol during its to confirm that each patient’s diet information was complete
testing. The results from the formal PEP uP protocol study and accurate. Required information included type of diet
illustrated improvement in adequacy and total nutrition (ie, oral, nonoral [EN vs parenteral nutrition]) and time to
across all study sites.11 initiation of nutrition therapy.
After completion of the trial, the team decided to use Time to initiation of nutrition was recorded in number
a modified version of the PEP uP protocol with different of days and calculated from the time that patients were
enteral formulas and weight-based volumes.11 The modified admitted to the ICU to the time that nutrition therapy was
enteral feeding protocol (EFP) was implemented and ex- initiated. The time-based nature of this variable entailed
panded to all critical care areas (surgery, coronary care, and that it was in noninteger form (eg, 1.4 days). Clinical and
burn center) after a successful rollout in the medical ICU by demographic variables used in the analyses were extracted
late fall/early winter of 2012. As part of the implementation, from the patients’ medical records. Per admission dates, each
a new EMR order panel was developed to assist doctors in patient was assigned 1 of 5 binary variables indicating the
correctly ordering the EFP. A “nurse champion” is impera- patient’s admission year (2009–2013). Each of the previ-
tive to the success of the protocol. A nurse champion was ously discussed quality improvement measures aligned with
designated to assist with continuous protocol coordination 1 of the admission year periods (Table 2). Accordingly, the
and to help educate staff on appropriate protocol use. A admission year binary variables allowed an examination of
breach in EFP would trigger a reeducation to the staff whether the quality improvement measures were associated
involved to reinforce concepts of the protocol provided with changes in the time to initiate nutrition. Specifically, did
by the nurse champion. As this is largely a nurse-driven days to the initiation of nutrition differ by admission year?
protocol, a strong leader in nursing is imperative to success. Two statistical analyses were conducted. The first anal-
Further changes were made to the anesthesia preopera- ysis calculated the average days to start of nutrition by
tive fasting policy in 2012. The policy was liberalized to state admission year. This analysis allowed a preliminary inves-
that EN withholding is unnecessary with jejunally placed tigation into whether the initiation of nutrition changed
feeding tubes. This change was based on evidence-based annually from 2009 to 2013. The second analysis estimated
Musillo et al 571

the association between the binary admission year variables Table 3. Descriptive Statistics for 141 Patients With Isolated
and time (measured in days) to nutrition initiation through Closed Head Trauma Injuries Who Received Nutrition
a generalized linear model (GLM) with a γ distribution and Therapy.a
identity link. A GLM with a γ distribution and identity Variable Percentageb SD Min Max
link was used in the second analysis; using an ordinary
least squares for this analysis may result in faulty estimates Age, y, mean 50.80 21.14 19 92
due to the strictly positive nature of the response variable <65 73.76 44.15
(days to nutrition initiation). The GLM-based analysis ࣙ65 26.24 44.15
allowed an investigation into whether the annual quality Sex
Female 21.28 41.07
improvement measures (binary admission year variables)
Male 78.72 41.07
were associated with changes in days to nutrition initiation, Race
while controlling for confounding factors.12 African American 17.02 37.72
The primary independent variables in the GLM were the Otherc 3.55 18.56
binary admission years. The base admission year was 2009. White 79.43 40.56
Accordingly, if the annual quality improvement measures Ethnicity
were associated with decreases in time to nutrition therapy Non-Hispanic 82.98 37.72
Hispanic 17.02 37.72
initiation, the coefficient on the other binary admission year
Admission year, mean 2011 1.38 2009 2013
variables should be negative. Covariates included in the 2009 20.57 40.56
GLM were age (ࣙ65 years), race (African American, other, 2010 21.28 41.07
white), Hispanic ethnicity, inpatient mortality, number of 2011 21.28 41.07
comorbidities and complications, diet type (oral, nonoral), 2012 20.57 40.56
Glasgow Coma Score, injury severity score, admission unit 2013 16.31 37.08
(surgical, nonsurgical), and cause of injury (fall, motor Clinical outcome
Alive 73.76 44.15
vehicle accident, other, unknown). The covariates were
Deceased 26.24 44.15
included to control for other factors that may influence time Diet type
to nutrition therapy initiation. All statistical analysis were Oral 18.44 38.92
conducted in STATA 14.12 Nonorald 81.56 38.92
Clinical variables, mean
Days to nutrition 2.67 1.57 0.39 8.65
Results initiatione
Total comorbidities 4.74 1.88 0.00 6.00
The initial population consisted of 159 patients. However,
Total complications 1.47 1.58 0.00 6.00
18 of these patients were removed due to incomplete diet Glasgow Coma Scale 9.71 4.62 3.00 15.00
documentation (n = 10), incorrect classification of admis- Injury Severity Score 26.23 11.79 4.00 75.00
sion to the adult ICU (n = 4), and incomplete covariate Cause of injury
information (n = 4). Statistical differences were found be- Otherf 31.21 46.50
tween the omitted patients and sample patients on a number Fall 36.17 48.22
of factors. As illustrated in Table 3, the majority of the Motor vehicle accidentg 24.82 43.35
Unknown 7.80 26.92
analytic sample was male (73.76%) with an average age of
50.80 years. Approximately 20% of the patients identified as a Statisticswere calculated on a sample with full covariate information
nonwhite and 17.02% as being of Hispanic ethnicity. From (N = 141).
b Values are presented as percentages unless noted otherwise.
a clinical perspective, the average number of comorbidities c Individuals who identified as American Indian/Eskimo/Aleut, Asian
among the sample was 4.74, and 26.24% of the sample died
or Pacific Islander, or other.
while in the facility. d Individuals who received enteral or parenteral feeding.

The majority of patients (81.56%) received EN or par- e The difference in days between when patients were admitted and

enteral nutrition, and the average time to initiation of when they began receiving nutrition.
f Individual’s injury caused by assault, bicyclist/pedestrian struck, boat
nutrition therapy was 2.67 days overall. As illustrated in accident, cutting instrument, fight/brawl, found down, hanging,
Table 4, the average days to nutrition initiation steadily railway accident, sports, penetrating trauma (knife, gun, impalement).
g Individual’s injury caused by motor vehicle collision.
decreased with each succeeding year from 2009 to 2013.
The average days to nutrition initiation was 3.095 in 2009,
while it was 1.631 days in 2013. This 47.31% reduction in
days to the initiation of nutrition therapy could be due to control for many of these factors and isolate the association
a number of factors (eg, differences in patients’ injury types between the changes in nutrition therapy practice (Table 2),
or comorbidities between the admission years) beyond the measured through the binary admission year variables, and
quality improvement measures. The GLM was intended to days to nutrition initiation.
572 Nutrition in Clinical Practice 33(4)

Table 4. Mean Days to Nutrition Initiation for 141 Patients Table 5. Association Between the Number of Days to
With Isolated Closed Head Trauma Injuries Stratified by Nutrition Initiation and the Admission Year and Other
Admission Year.a Characteristics.a

Yearb No. Meanc SD Median Min Max Variable (Referent) Coefficient P Value

2009 29 3.095 1.43 2.91 0.98 7.18 Admission yearb (2009)


2010 30 3.288 1.64 2.74 1.12 6.94 2010 −0.47 .220
2011 30 2.985 1.78 2.70 0.74 8.65 2011 −0.72 .089
2012 29 2.094 1.29 1.78 0.53 5.82 2012 −1.09 .008
2013 23 1.631 0.95 1.54 0.39 4.44 2013 −1.75 .000
Age group (<65 y): ࣙ65 y 0.20 .564
a Nutrition initiation: the difference in days between when patients Sex (male): female −0.21 .465
were admitted and when they began receiving nutrition therapy. Race (white)
Statistics were calculated on a sample with full covariate information African American 0.18 .568
(N = 141). Otherc 0.97 .285
b Admission year or year of treatment is intended to indicate the
Ethnicity (non-Hispanic): Hispanic −0.33 .289
quality improvement interventions that were implemented in the year
that a patient was admitted or treated. Clinical outcome (alive): deceased −0.32 .252
c Means are rounded, which adjusts the percentage change statistic Diet type (nonorald ): oral 1.15 .002
cited in the article between 2009 and 2013. Based on these rounded Clinical variables
figures, the percentage change between these 2 years is 47.30%; Total comorbidities −0.11 .119
unrounded, the percentage change is 47.31%. Total complication 0.06 .535
Glasgow Coma Scale 0.04 .070
Injury Severity Score 0.01 .387
The GLM results are illustrated in Table 5. The re- Cause of injury (fall)
sults are reported in days. For instance, holding all else Othere −0.20 .504
constant, it was estimated that the time to the initiation Motor vehicle accidentf 0.20 .638
of nutrition therapy was 1.09 days (P = .008) shorter in Unknown −0.91 .037
2012 relative to 2009. Likewise, it was estimated that the Constant 3.15 .000
time to the initiation of nutrition therapy was 1.75 days a Coefficients (days) estimated from a generalized linear model with a
(P = .000) shorter in 2013 relative to 2009. These 2 results γ distribution and identity link; the sample included 141 patients with
support the hypothesis that an association exists between isolated closed head trauma injuries who received nutrition therapy.
the implemented quality improvement interventions and Nutrition initiation: the time difference (in days) between when
patients were admitted and when they began receiving nutrition
days to initiation of nutrition. Although the coefficients for
therapy.
the other 2 binary year variables (2010, 2011) were in the b Admission year or year of treatment is intended to indicate the
expected direction, a significant association was not found quality improvement interventions that were implemented in the year
between days to nutrition initiation and these 2 variables. that a patient was admitted or treated.
c Individuals who identified as American Indian/Eskimo/Aleut, Asian
The results also illustrate that diet type was associated with
or Pacific Islander, or other.
days to nutrition initiation. Patients who received their first d Individuals who received enteral or parenteral feeding.

feeding orally were estimated to have a time to nutrition e Individual’s injury caused by assault, bicyclist/pedestrian struck, boat

initiation that was 1.15 days (P = .002) longer than patients accident, cutting instrument, fight/brawl, found down, hanging,
railway accident, sports, penetrating trauma (knife, gun, impalement).
whose first feeding was administered nonorally. f Individual’s injury caused by motor vehicle collision.
The results from the GLM support the trends illustrated
in Table 4; the difference between these 2 analyses (eg, no
statistical differences in days to nutrition therapy initiation
closed head trauma injuries. There was a 47.31% reduction
between 2009 and 2010 or 2009 and 2011 in the GLM mod-
(or 1.46 days) in the time to initiate nutrition therapy from
els) is likely due to the inclusion of confounding factors in
2009 to 2013. The time to initiate in 2013 was 1.631 days, an
the GLM. Results from an ordinary least squares regression
outcome consistent with recommendations for EN to begin
model (Appendix Table A1) were similar to those estimated
within 2 days.2 These changes are also in line with the new
in the GLM. However, the significance of the coefficient on
2016 nutrition critical care guidelines,1 which recommends
the 2012 binary year was sensitive to the patients included
a volume-based feeding or top-down multistrategy protocol
in the ordinary least squares regression.
be used. Protocols such as these empower nursing to alter
the run rates to achieve the goal volume by the end of the day
Discussion
instead of traditional static run rates, which were previously
Our multi-intervention strategy has demonstrated signifi- used in our facility.13,14
cant improvement, starting in 2012, in the timeliness of Over the course of 5 years, different strategies were
initiation of nutrition among a population with isolated implemented and maintained (Table 2), and their effects
Musillo et al 573

here are demonstrated in a cumulative fashion. Although environment. Strengths of this quality improvement process
significant associations were found between the days to include that it was an interdisciplinary effort with numer-
nutrition initiation and the 2012 and 2013 binary admission ous stakeholders assisting to elicit ongoing improvements.
year variables, these associations are likely due to all inter- Implemented changes were supported by the group and
ventions and not just the interventions that occurred in these maintained from inception. In addition, despite the results
2 years. In other words, significant changes that were found of this study and the possible improvement in our hospital’s
are likely due to a comprehensive effect—one that builds nutrition policies and practices, the continuous improve-
on the interventions implemented in previous years, instead ment nature of our system entails that the monitoring, ed-
of just the interventions introduced in 2012 and 2013. ucation, and search for other ways to improve our nutrition
Heyland et al articulated a strategy to begin to motivate policies and practices continues. For instance, deficiencies
surgeons and other providers to overcome the prevailing were identified in the protocol’s order panel in the EMR,
persistence to change traditional or historic practice and and changes were made to optimize protocol compliance
adopt evidence-based perioperative nutrition therapy, thus with the order panel. Working as an interdisciplinary team,
narrowing the knowledge practice gap. The 8 phases of hospital staff continue to identify deficiencies and provide
the action cycle discussed various activities needed to move corrections to illustrate that this is an ongoing and fluid
knowledge into practice. Our interventions were not based process.
on this model, but many interventions and processes mirror While this was a retrospective study to examine the
the cycle discussed here. This would be a good resource in improvement measures performed at this facility, the results
addition to this article to bring change to other facilities’ presented in this analysis are subject to a number of limi-
historic practice.15 Although not examined with these data, tations. Importantly, the results are based on associations,
the largest barrier to overcome when implementing the new not causation. While attempts were made to isolate the
protocol was historic practice by nursing in regard to EN relationship between days to nutrition initiation and the
“ramp up” protocols and use of static run rates. It was interventions, we were unable to directly connect changes in
also challenging to overcome fears regarding perceived high days to initiation to the discussed intervention. In addition,
GRVs and “high” run rates. After a number of education binary admission year variables may serve as indicators
sessions, staff were seemingly less reluctant to implement for factors beyond the interventions, such as unmeasured
change, due to gained experience with the new protocols. changes within the hospital during the examined year.
Stooling has historically been a barrier to EN provision. However, in the absence of a control group or hospital, this
However, with the use of improved fecal management rather noisy method was used. The single-center nature of
systems and diarrhea-specific protocols, there has been a this study is thus an additional limitation.
concomitant decrease in nurses’ hesitation to provide EN. Many of the strategies used here were demonstrated
It is imperative to have the support of nursing and hos- to be effective in other facilities.3,6,8,11 We “fine-tuned”
pital administration to effect change and maintain success the interventions to best fit the needs of our hospital.
while implementing similar protocols. Nurse champions Individual centers should examine their present cultures
who understand and appreciate the importance of timely and own needs prior to implementing new interventions.
and adequate nutrition are essential in the change of culture Overall, it is important to note that the results of this study
within the critical care arena. may be due to this facility and its own evolving nutrition
Another barrier to initiation of EN in the ICU was the culture.
use of antiquated preoperative fasting policies. Patients were
maintained nil per os with a minimum of 8 hours prior
to any interventional procedure. At times, the procedures
Conclusion
were delayed or postponed if nutrition was not withheld Our participation in board-approved research projects has
within a designated 8-hour time frame, thereby increasing shaped our evidence-based best practice, and significant
fasting time. With placement of a postpyloric feeding tube, change has occurred as a result. Barriers to adequate nutri-
invasive procedures were not delayed, due to preoperative tion provision were identified and addressed, resulting in a
fasting protocols by anesthesia. The purchase and use of reduction of variability and a proactive approach to early
a bedside electromagnetic feeding tube device allowed for nutrition initiation. This approach was novel in the fact that
expedited postpyloric feeding tube placement. This in turn the process began with just the identification that an issue
allowed for EN to be provided with minimal need for x- existed in the timing of initiation of nutrition. A cascade
ray confirmation, with no required preoperative fasting, of interventions ensued based on identified needs of the
and with less EN withholding for “high” GRVs or other facility. In retrospect, it may appear the interventions were
intolerance issues. not novel; however, they were in 2007 in this facility. Many
From 2013 to present, the strategies implemented over hospitals continue to struggle with the barriers that we
the examined years culminated into a proactive nutrition have identified, and they are still searching for their specific
574 Nutrition in Clinical Practice 33(4)

solutions. This topic needs to continue to be discussed in 4. The interdisciplinary team approach was important.
the literature to help bring awareness and foster change It is recommended that other systems follow a team-
to abandon historic practices. Optimization of nutrition based approach that develops solutions based on
therapy requires the coordination of multiple disciplines. their needs. The interventions introduced here were
The interdisciplinary effort applied in our facility was a specific to the needs and identified barriers of this
key component of our efforts to improve nutrition therapy examined facility.
practices. Increased communication among team members
regarding nutrition goals and actual support provided Appendix
has increased. A culture change has occurred over time. Table A1. Association Between the Number of Days to
This nurse-directed protocol, with associated changes, has Nutrition Initiation and the Admission Year and Other
become second nature to the critical care team. Elevation of Characteristics.a
the RDN within the institution is imperative for successfully
improving the quality of nutrition care. The RDN’s scope Variable (Referent) Coefficient P Value
of practice needs to be expanded to include contribution to Admission yearb (2009)
evidence-based nutrition practice, feeding tube placements, 2010 −0.31 .496
physical assessment, and nutrition-specific order entry in 2011 −0.46 .314
the EMR for best outcomes. Physician knowledge regarding 2012 −1.08 .042
nutrition has been enhanced throughout this process with 2013 −1.53 .000
resulting improvement in nutrition therapies. The RDN has Age group (<65 y): ࣙ65 y 0.51 .137
Sex (male): female 0.06 .860
also been recognized by other healthcare team members as
Race (white)
an integral part of total patient care. African American 0.18 .654
Nutrition therapy is now viewed in a proactive manner, Otherc 1.75 .044
striving for enhancement in the time frame from the time Ethnicity (non-Hispanic): Hispanic 0.10 .763
of admission to the start of EN. EN provision has been Clinical outcome (alive): deceased −0.47 .222
significantly improved despite the multiple common barriers Diet type (nonorald ): oral 0.97 .016
that historically plague this aspect of care. Simplicity of the Clinical variables
Total comorbidities −0.07 .352
application and tools that were developed can be modified
Total complication 0.07 .442
to meet the requirements of other areas/facilities. Glasgow Coma Scale 0.06 .068
The results presented here were from our institution’s Injury Severity Score 0.01 .665
directive to improve nutrition therapy. This is an example Cause of injury (fall)
of what an institution can do when it looks into its own Othere 0.03 .935
nutrition policies, relative to standards and through knowl- Motor vehicle accidentf 0.44 .234
edge gained through participation in research studies, and Unknown −0.73 .171
Constant 2.46 .003
then implements strategies to address challenges through an
interdisciplinary team-based approach with the support of a Coefficients (days) estimated from an ordinary least squares
hospital administration. regression model; the sample included 141 patients with isolated
closed head trauma injuries who received nutrition therapy. Nutrition
initiation: the time difference (in days) between when patients were
Key Points admitted and when they began receiving nutrition therapy.
b Admission year or year of treatment is intended to indicate the

1. Not just 1 intervention but all of them together likely quality improvement interventions that were implemented in the year
explain our results. that a patient was admitted or treated.
c Individuals who identified as American Indian/Eskimo/Aleut, Asian
2. The results are likely partially explained by buy-
or Pacific Islander, or other.
in from across the hospital and a continuation d Individuals who received enteral or parenteral feeding.
e Individual’s injury caused by accident, accident at home, assault,
of support for adequate and timely nutrition. The
interdisciplinary team approach allowed us to not bicyclist, boat accident, cutting instrument, fight/brawl, found down,
gunshot wound, gunshot, hanging, hit by blunt instrument, impaled
only account for the medical knowledge needed by object, other, pedestrian struck, poison, railway accident, sports,
but also understand the barriers from multiple stabbing, or stab wound.
f Individual’s injury caused by either a motor vehicle collision or a
perspectives.
3. The overall understanding of barriers, as well as pos- motorcycle collision.
sible challenges with implementing the interventions,
allowed the development of solutions to challenges Acknowledgments
that arose from a complete perspective instead of We thank Kathy Hill, MA, RDN, for her ongoing support and
1 solution from only 1 disciplinary or departmental assistance with implementing the nutrition-related strategies
perspective. presented here.
Musillo et al 575

Statement of Authorship 6. Rivera R, Campana J, Hamilton C, Lopez R, Seidner D. Small


bowel feeding tube placement using an electromagnetic tube placement
L. Musillo contributed to the conception/design of the re-
device: accuracy of tip location. JPEN J Parenter Enteral Nutr.
search; all authors contributed to the acquisition, analysis, or 2011;35(5):636-642.
interpretation of the data; L. Musillo, L. M. Grguric, and R. 7. Moncure M, Samaha E, Moncure K et al. Jejunostomy tube feedings
Batista drafted the manuscript; L. Musillo, L. M. Grguric, E. should not be stopped in perioperative patient. JPEN J Parenter Enteral
Coffield, and R. Batista critically revised the manuscript; and Nutr. 1999;23(6):356-359.
L. Musillo and L. M. Grguric agree to be fully accountable for 8. Pousman RM, Pepper C, Panharipande P et al. Feasibility of imple-
ensuring the integrity and accuracy of the work. All authors menting a reduced fasting protocol for critically ill trauma patients
read and approved the final manuscript. undergoing operative and nonoperative procedures. JPEN J Parenter
Enteral Nutr. 2009;33(2):176-180.
9. Parent BA, Mandell SP, Maier RV et al. Safety of minimizing preoper-
References
ative starvation in critically ill and intubated trauma patients. J Trauma
1. McClave SA, Taylor BE, Martindale RG et al. Guidelines for the Acute Care Surg. 2016;80(6):157-163.
provision and assessment of nutrition support therapy in the adult 10. Musillo L, Grguric-Smith LM, Coffield E, Totino K, DiGiacomo JC. Is
critically ill patient: Society of Critical Care Medicine (SCCM) and there a discrepancy? Comparing enteral nutrition documentation with
American Society for Parenteral and Enteral Nutrition (ASPEN). enteral pump volumes [published online September 1, 2016]. Nutr Clin
JPEN J Parenter Enteral Nutr. 2016;40(2):159-211. Pract.
2. Lee ZY, Barakatun-Nisak MY, Airini IN, Heyland DK. Enhanced 11. Heyland DK, Cahill NE, Dhaliwal R et al. Enhanced protein-energy
Protein-Energy Provision via the Enteral Route in Critically Ill Patients provision via the enteral route in critically ill patients: a single center
(PEP uP protocol): a review of evidence. Nutr Clin Pract. 2016;31(1):68- feasibility trial of the PEP uP protocol. Crit Care Med. 2010;14(2):R78.
79. 12. StataCorp. Stata Statistical Software: Release 14. College Station, TX:
3. Wilson S, Madisi NY, Bassily-Marcus A, Manasia A, Oropello J, StataCorp LP; 2015.
Kohli-Seth R. Enteral nutrition administration in a surgical intensive 13. Marshall AP, Cahill NE, Gramlich L, MacDonald G, Alberda C,
care unit: achieving goals with better strategies. World J Crit Care Med. Heyland DK. Optimizing nutrition in intensive care units: empowering
2016;4(3):108-186. critical care nurses to be effective agents of change. Am J Crit Care.
4. Peev MP, Yeh DD, Quraishi SA et al. Causes and consequences of 2012;21(3):186-194.
interrupted enteral nutrition: a prospective observational study in criti- 14. McClave SA, Mohamed SA, Esterle M et al. Volume-based feeding in
cally ill surgical patients. JPEN J Parenter Enteral Nutr. 2015;39(1):21- the critically ill patient. JPEN J Parenter Enteral Nutr. 2015;39(6):707-
27. 712.
5. Cahill NE, Dhaliwal R, Day AG et al. Nutrition therapy in the 15. Heyland DK, Dhaliwal R, Cahill NE et al. Driving perioperative
critical care setting: what is “best achievable” practice? An international nutrition quality improvement processes forward! JPEN J Parenter
multicenter observational study. Crit Care Med. 2010;38(2):395-401.6. Enteral Nutr. 2013;37(1):83S-98S.
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 4
Gastrostomy Tube Feeding in Children With Developmental August 2018 576–583

C 2018 American Society for

or Acquired Disorders: A Longitudinal Comparison Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10056
on Healthcare Provision and Eating Outcomes 4 Years wileyonlinelibrary.com
After Gastrostomy

Ellen Backman, MSc1,2 ; Ann-Kristin Karlsson, PhD3 ; and Lotta Sjögreen, PhD4

Abstract
Background: Studies on long-term feeding and eating outcomes in children requiring gastrostomy tube feeding (GT) are scarce.
The aim of this study was to describe children with developmental or acquired disorders receiving GT and to compare longitudinal
eating and feeding outcomes. A secondary aim was to explore healthcare provision related to eating and feeding. Methods: This
retrospective cohort study reviewed medical records of children in 1 administrative region of Sweden with GT placement between
2005 and 2012. Patient demographics, primary diagnoses, age at GT placement, and professional healthcare contacts prior to
and after GT placement were recorded and compared. Feeding and eating outcomes were assessed 4 years after GT placement.
Results: The medical records of 51 children, 28 boys and 23 girls, were analyzed and grouped according to “acquired” (n = 13) or
“developmental” (n = 38) primary diagnoses. At 4 years after GT placement, 67% were still using GT. Only 6 of 37 (16%) children
with developmental disorders transferred to eating all orally, as opposed to 10 of 11 (91%) children with acquired disorders. Children
with developmental disorders were younger at the time of GT placement and displayed a longer duration of GT activity when
compared with children with acquired disorders. Conclusions: This study demonstrates a clear difference between children with
developmental or acquired disorders in duration of GT activity and age at GT placement. The study further shows that healthcare
provided to children with GT is in some cases multidisciplinary, but primarily focuses on feeding rather than eating. (Nutr Clin
Pract. 2018;33:576–583)

Keywords
enteral nutrition; feeding and eating disorders of childhood; gastrostomy; interdisciplinary; nutritional support; patient care teams;
pediatrics; tube feeding

Introduction “To eat” is defined according to the International


Classification of Functioning, Disability and Health as a
Gastrostomy tube feeding (GT) is necessary when children multifaceted activity including actions of eating food that
with severe feeding or eating disorders require prolonged has been served, bringing it to the mouth, and consuming it
nonoral enteral nutrition (EN) due to acquired or develop- in culturally acceptable ways.10 Eating difficulties can cause
mental disorders.1,2 Indications for GT include digestive dis- an imbalance in the caregiver–child interaction and may
eases inducing a decrease in nutrient absorption, disorders lead to significant distress and frustration in the child and
affecting the ability to eat, or inadequate caloric intake.3
Depending on the underlying disorder, GT can be short
term or long term. A clear difference in the length of GT From the 1 School of Health and Welfare, Halmstad University,
programmes when comparing children in need of GT due to Sweden; 2 Regional Habilitation Center, Region Halland,
Kungsbacka, Sweden; 3 Department of Research and Development,
neurological impairments or to transient organ failure has Region Halland, Halmstad, Sweden; and 4 Mun-H-Center Orofacial
been noted.4,5 However, this difference has only been briefly Resource Center for Rare Diseases, Public Dental Service,
explored in relation to demographic characteristics and in Gothenburg, Sweden.
established healthcare guidelines.6 Financial disclosure: This work was supported by the Department of
Feeding and eating are complex, interrelated activities Research and Development, Region Halland, Sweden.
and for caregivers of children with GT, both of these Conflicts of interests: None declared.
concepts have been shown to be of importance.7 “To feed” This article originally appeared online on March 30, 2018.
can be defined as “to give food to” and relates to nutrition
Corresponding Author:
to sustain vital bodily functions. Undernutrition may result Ellen Backman, MSc, School for Health and Welfare, Halmstad
in poor cognitive development, impaired wound healing, University, Box 823 SE-301 18 Halmstad, Sweden.
reduced immuno-competence, and/or growth inhibition.8,9 Email: ellen.backman@hh.se
Backman et al 577

the caregiver.11 Traditional biomedical approaches focusing This represents a slightly different classification than has
on physical health outcomes is therefore insufficient for been used earlier but was chosen as ICD-10 is the interna-
interventions such as GT with broad and long-term tional standard for reporting diseases and health conditions
consequences.12 A multidisciplinary team approach has for all clinical and research purposes.20 The ICD-10 has
been advised to improve nutrition management in children been used in Sweden since 1997 with a Swedish update in
with GT, including the areas of psychology, nutrition, 2011.21 The primary diagnoses were classified as “develop-
speech-language pathology, and medicine.13-15 mental” (DD), relating to, for example, neurodevelopmental
The use of nonoral EN has increased in the pediatric disabilities or congenital malformations, or “acquired”
population in western Europe during the past 20 years,4,16,17 (AD), relating to conditions that were contracted after
yet the healthcare related to feeding and eating practices birth.2
and follow-up provided are still nonstandardized and highly Data were collected retrospectively using the before-
variable.14,18,19 More important, as little is known about mentioned instrument beginning at 12 months prior to GT
the long-term feeding and eating outcomes correlated with placement and continuing until year 4 following GT place-
underlying disorders, this limits our capacity to provide ment. The medical records of all children were accessed
professional healthcare adapted to whether feeding and through the region’s electronic medical records system.
eating functions have been lost or not yet developed. The descriptions of healthcare actions of each healthcare
The aim of this study was therefore to describe children professional was divided into care provided prior to and
with developmental or acquired disorders receiving GT and after GT placement and was compiled at group level until
to explore healthcare provision related to eating and feed- saturation was reached.
ing. The aim was to also compare longitudinal eating and The head of the clinics responsible for the children’s
feeding outcomes in children with developmental disorders healthcare were informed about the study, and they ap-
with those in children with acquired disorders. proved a review of the medical records. All families eligible
for the study received written information and were given
the possibility to withdraw from the study at any time. The
Methods study adhered to the ethical guidelines of the Declaration
This descriptive and longitudinal study retrospectively re- of Helsinki22 and was ethically approved by the Regional
viewed medical records belonging to children receiving GT. Ethics Committee in Lund (dnr 2016/93).
Data were reviewed up to 4 years after GT placement.
Medical records belonging to 51 children in 1 adminis- Data Analysis
trative region in the western part of Sweden were retrieved
from specialized pediatric healthcare providers in December Data were mainly presented using descriptive statistics and
2016. Approximately 3% of Sweden’s 10 million inhabitants expressed as medians, minimum–maximum, and percent-
live in the region. All children using GT were eligible for ages. Nonparametric statistical tests were used to analyze
taking part in the study, and inclusion criteria were (a) age differences as the data were nonnormally distributed. Com-
at start of GT <19 years, (b) GT duration >1 month, and parison of categorical data was performed using Fisher’s
(c) GT placement between January 1, 2005, to December exact test. Group differences in continuous variables were
31, 2012. The complete search strategy is available online analyzed using the Mann–Whitney U test for 2 indepen-
(Supporting Information Figure S1). dent samples. A P value <.05 was considered statisti-
A protocol was used for data collection from the medical cally significant. The analyses have been performed us-
records prior to and after GT placement (Supporting Infor- ing SPSS software, version 24.0 (IBM Corp., Armonk,
mation Table S1). A pilot protocol was initially tested on the NY). Because of the retrospective nature of the study, the
records of 15 children to evaluate content and its use and data for all variables for every child were not available.
reduce the amount of missing data. Pilot testing proved the The total amount of children for each analysis is clearly
protocol to include ambiguous definitions of variables re- reported.
lated to feeding and healthcare as well as numerous free-text
options. Following pilot testing, many of the free-text op- Results
tions were replaced with tick boxes as saturation of possible
Characteristics of Study Cohort
medications, diagnoses, and number and category of health-
care professionals in contact with the family was established. The medical records of 51 children, 28 boys and 23 girls,
The child’s primary diagnosis (the diagnostic codes from were analyzed in the study. The characteristics of the
the International Statistical Classification of Diseases and children are given in Table 1. The median age at the time
Related Health Problems–10th revision; ICD-1020 ) stated in of GT placement was 35 months (minimum–maximum:
the medical record by the pediatrician in charge of referring 4–196), and 51% of the children (n = 26) were aged 3
to GT surgery was used to classify the underlying condition. or younger. A total of 3 children died during the study
578 Nutrition in Clinical Practice 33(4)

Table 1. Characteristics of Study Cohort.

Developmental Acquired
Category Disorders Disordersa P Value

Number of patients, n (%)b 38 (75) 13 (25)


Males, n (%) 18 (47) 10 (77) .11
Median age at gastrostomy placement in months, n 24 (4–180) 109 (4–196) .041
(minimum–maximum)
Number of children with nasogastric tube prior to 22 (58) 8 (62) >.99
gastrostomy placement, n (%)
Medication, n (%)
Intestinal regulators 19 (50) 9 (69) .34
Antibiotics 11 (29) 11 (85) .001
Anticonvulsants 14 (37) 2 .19
Respiratory medication 11 (29) 3 >.99
Gastroesophageal reflux disease medication 10 (26) 3 >.99
Chemotherapy – 10 (77) <.001
No medication 4 – .56
Other 10 (26) 11 (85) <.001
a Neoplasms and intracranial injuries were classified as acquired disorders.
bN = 51.

Table 2. Primary Diagnoses Comprising the Study Group A total of 17 different indications for commencing GT
According to the Chapters of the International Statistical was stated in the medical records when referring to gas-
Classification of Diseases and Related Health Problems–10th trostomy surgery, with “swallowing disorders” from ICD-
Revision and Allocation to Subgroups, N = 51.
10 chapter R being the most frequent indication for the
Number of study cohort (n = 18, 35%). Examples of other indications
Category Children, n (%) were from chapters C, neoplasm; E, malnutrition; F, other
feeding disorders of infancy and childhood; K, intesti-
Developmental disorders, n = 38 nal malabsorption; and P, feeding problems of newborn.
Chapter E, metabolic disorders (E71) 1 (2) Among the children with DD, “swallowing disorders” from
Chapter F, mental, behavioural, and 3 (5)
neurodevelopmental disorders (F79, F90,
ICD-10 chapter R was the most frequent indication, corre-
F98) sponding to 37% of the indications for commencing GT (n
Chapter G, diseases of the nervous system 12 (24) = 14). Among the children with AD, malignant neoplasm
(G40, G71, G80) from ICD-10 chapter C was the most frequent indication
Chapter P, certain conditions originating in 4 (8) (n = 5, 38%).
the perinatal period (P07)
Chapter Q, congenital malformations, 18 (35)
deformations and chromosomal
abnormalities (Q03, Q04, Q21, Q37, Q75,
Professional Healthcare
Q79, Q85, Q87, Q90, Q92, Q93, Q99) Healthcare professionals represented in the children’s
Acquired disorders, n = 13 medical records in regard to feeding, and/or eating in-
Chapter C, malignant neoplasms (C41, C71, 10 (20)
cluded pediatrician, registered nurse, registered dietitian,
C71, C91, C92)
Chapter D, diseases of the blood and 1 (2) speech-language pathologist, pediatric dentist, occupational
blood-forming organs and certain disorders therapist, physiotherapist, social worker in medical and
involving the immune mechanism (D71) healthcare, psychologist, and special needs educator. The
Chapter S, intracranial injury (S 06) 2 (4) types of healthcare actions are further described in the
supplementary material available online (Supporting Infor-
mation Table S2).
Of the 51 children, 26 children representing with both
period, all attributed to causes other than GT placement. DD and AD also had specialized care outside the admin-
Postoperative details for these children were not included. istrative region, which was sometimes only briefly noted in
A total of 35 primary medical diagnoses in 8 different the provided medical records. This care took place in clinics
chapters of the ICD-10 were noted among the children specialized in cancer treatment or centers for rare diseases,
(Table 2). for example.
Backman et al 579

Table 3. Number of Children in Contact With Each Healthcare Professional.

Prior to Gastrostomy Placement After Gastrostomy Placement

Developmental, Acquired, n = 13; Developmental, Acquired, n = 11;


Healthcare Contacta n = 38; n (%) n (%) n = 37; n (%) n (%)b

Pediatrician 35 (92) 12 (92) 35 (95) 11 (100)


Registered nurse 26 (68) 11 (85) 34 (92) 11 (100)
Registered dietitian 27 (71) 7 (54) 36 (97) 11 (100)
Speech-language pathologist 26 (68) 2 30 (81) 1
Paediatric dentist 14 (38) 5 (38) 24 (65) 9 (82)
Social worker in medical and 13 (34) – 14 (38) –
healthcare and/or psychologistc
Occupational therapist 11 (29) – 14 (38) 1
Physiotherapist 8 (21) – 10 (27) –
Special needs educators 3 – 3 –
Specialized multidisciplinary feeding 1 – 4 1
team
a Healthcare contacts related to feeding and/or eating within the administrative region 12 months prior to gastrostomy placement and for 4 years
after.
b A total of 3 children died during follow-up; data for these children are not included after gastrostomy placement.
c Healthcare provided by social workers or psychologists was grouped as 1 category as the counseling was often given by the 2 professionals

together.

The analysis of the professional healthcare contacts 2–43) after GT placement and was more often seen in
involved in eating and feeding demonstrated a higher de- children with AD (P < .001). The use of nasogastric tube
gree of multidisciplinary care, defined as including areas feeding in the year prior to GT placement was not related
of psychology, nutrition, speech-language pathology and to a specific eating outcome.
medicine in the group of children with DD both prior to Commercially manufactured enteral products were the
and after GT placement when compared with the children source of nutrition for the majority of the children. Home-
with AD as seen in Table 3. made, blenderized food was given to 11 children (23%)
A total of 5 specialized multidisciplinary feeding teams during the study period, all of whom had DD. Homemade
were mentioned in the medical records: 1 in this region, 3 in food was most frequently used in combination with manu-
other administrative regions in Sweden, and 1 abroad. The factured products. GT was permanently removed for 1 child
feeding teams were comprised differently, and the therapy receiving homemade food due to eating sufficiently orally,
was provided in some cases as intensive in-patient care, and and 2 children increased their oral intake.
sometimes as outpatient follow-up. Feeding habits were documented after GT placement
No formal assessments of the children’s eating abilities based on the amount of food eaten orally or in tube and are
were found in the medical records prior to GT place- depicted in Figure 1. The degree of oral intake improved
ment, but 27 children (53%) had undergone an informal for 19 children (40%), remained the same for 18 children
assessment, and 6 children (12%) were assessed using a (38%), and worsened for 6 children (13%). For 3 children,
Videofluoroscopic Swallowing Study; all had DD. Prior to the degree of oral intake varied during the study period.
GT placement, 24 children (47%, 11 DD/13 AD) were not Data on feeding habits were missing for 1 child (1 DD) the
assessed for eating ability. first and fourth year, for 1 child (1 DD) the first year, and 1
After GT placement, 1 child with DD was formally child (1 AD) received all nutrition orally, using GT only for
assessed using a Swedish scale measuring activities of daily additional fluid and/or medication.
living, with eating ability as 1 part of the scale. A total of 26 Further analyses of the changes in degree of oral intake
children (54%, 24 DD/2 AD) were informally assessed. A revealed the following:
total of 21 children (44%, 13 DD/8 AD) were not assessed
for eating ability after GT placement.
r None of the 15 children (14 DD/1 AD) receiving “all
in tube” during the first year after GT placement
transferred to managing “all orally.” Of these chil-
Feeding and Eating Outcomes
dren, 6 increased their oral intake to either “most in
GT activity 4 years after GT placement is depicted in Table tube” (4 DD/1 AD) or “mostly orally”(1 DD).
4. Discontinuation of GT for eating sufficiently orally oc- r A total of 16 children (8 DD/8 AD) received “most
curred after a median of 16 months (minimum–maximum: in tube” in the first year after GT placement, and 9
580 Nutrition in Clinical Practice 33(4)

Table 4. Follow-Up of Gastrostomy Tube Feeding Activity 4 Years After Gastrostomy Placement.

Ongoing Gastrostomy Eating Sufficiently P


Category Tube Feeding Orally Value

Number of children, N = 48, n (%) 32 (67) 16 (33)


Male, n (%) 16 (50) 11 (73) .136
Acquired disorders, n (%)a 1 (3) 10 (63)
Developmental disorders, n (%) 31 (97) 6 (37)
Median age at gastrostomy placement in 24 (4–152) 113 (4–196) .03
months, n (minimum–maximum)
a Neoplasms and traumatic brain injuries were classified as acquired disorders.

100%

90%

80%

70%

60%
All orally
50% Mostly orally
Most in tube
40%
All in tube
30%

20%

10%

0%
Developmental Developmental Acquired disorders, Acquired disorders,
disorders, year 1, n= 36 disorders, year 4, n=37 year 1, n=11 year 4, n=11

Figure 1. Feeding habits first and fourth year after gastrostomy placement distributed by acquired (“neoplasms” or “intracranial
injuries”) or developmental disorders: most in tube, >50% of total intake in the tube; mostly orally, >50% of oral intake from
liquids and solid foods orally. Data are missing for 2 children the first year and for 1 child the fourth year as a result of care
outside of the administrative region.

of these (1 DD/8 AD) transferred to managing “all GT. One main finding is the apparent difference in eating
orally” during the study period. A total of 2 children outcomes for children requiring GT for DD or AD. Only
decreased their oral intake (2 DD), and 1 child (1 6 of 37 (16%) children with DD transferred to eating all
DD) increased the oral intake to “mostly orally.” orally, as opposed to 10 of 11 (91%) children with AD.
r A total of 14 children (13 DD/1 AD) received Diagnoses relating to malignancies dominated in the group
“mostly orally” in the first year after GT placement. with AD (11 of 13 children). The need for supplemental nu-
Of these children, 4 (3 DD/1 AD) transferred to man- trition in this group stemmed from intensive chemotherapy
aging “all orally” during the study period, whereas leading to, for example, loss of appetite, food aversion, and
5 children had decreased oral intake at the end of vomiting, and not from an unsafe swallow or poor eating
follow-up to “most in tube” (3 DD) or “all in tube” skills. The use of GT in this group is consistent with prior
(2 DD). studies showing the use of GT as an important part of the
care to improve somatic health issues and to reduce family
“Oral stimulation using different tastes” was given to frustration during the intensive phase of treatment.23,24 A
nearly all children with “all in tube” (83%–100%) during the return to oral feeding after completion of treatment would
4 years following GT placement. therefore be expected.
Another important difference was that children with
Discussion DD were younger at the time of GT placement. These
This longitudinal follow-up of children receiving GT shows findings relating to age and GT activity are not in line with
that at the end of the study period, 4 years after GT earlier studies reporting that children who were weaned
placement, two thirds of the study population still required from GT were younger at the time of GT placement than
Backman et al 581

children with GT still in place.25 However, the study did contact with different healthcare professionals indicated a
not explore differences related to primary diagnoses,25 a preponderance of actions related to aspects of “feeding”:
factor found to be of great importance in the present medical status, nutrition, and growth. These results corre-
study. The current findings are further strengthened by spond to those of previous studies that documented parent
previous studies indicating that differences in GT activity perceptions of healthcare provided to children with GT as
were related to underlying disease,4,5,18 with neurological primarily focusing on medical, not social, needs.27 Despite
disorders associating with a prolonged need for GT. the indications when referring to GT surgery, another
In the present findings, feeding habits were associated finding of the healthcare reported in the present study,
with long-term eating outcomes. Children receiving “all in was the lack of formal tools used to assess eating abilities.
tube” the first year after GT placement were more likely This corresponds to earlier stated inconsistencies in the use
to continue to require nutrition support by GT after 4 of clinical measures for eating, drinking, and feeding.28
years. Children with GT as a complementary source of Moreover, the children’s participation in mealtimes, whether
nutrition during the first year, that is, receiving “most in eating orally or not, was seldom a documented area of
tube” or “mostly orally,” were more likely to increase their concern.
oral intake. This was particularly true for children with The present study is the first to compare the healthcare
AD, most probably related to the secondary nature of their provided to children with GT as a factor of the type of
feeding difficulties. These findings can also be interpreted primary diagnosis. The results reveal a discrepancy between
as the development of sensory functions and oral skills was the care provided to children with DD and to those with
facilitated for children continuing receiving food orally dur- AD. A multidisciplinary team approach with care focused
ing GT.26 The long-term need for GT in children with DD on the broader concept of eating was more often used in
further highlights the necessity of this nutrition support for the former group. Despite this, the latter group showed a
children who otherwise could be at risk of forced feeding, greater increase in oral intake and in reestablishing adequate
severe malnutrition, or growth impairment, an assumption eating. Increase in oral intake was expected because of the
corresponding to earlier findings on positive outcomes of transient nature of the feeding difficulties for the majority
GT.3,5,19 of children with AD. Nevertheless, healthcare professionals
With regard to the primary medical diagnosis of children need to be aware of the multidimensional aspects of food
with GT, the most frequent category was from ICD-10 also for this patient group as food has been shown to carry
chapter Q, congenital malformations, deformations, and cultural representations of the life outside the hospital aside
chromosomal abnormalities. This was somewhat surprising from mere nutrition aspects.29 This difference also stresses
as earlier studies have described neurological disease as the importance of the primary diagnosis when anticipating
being the most common indication for GT.4,5,17,25 One ex- and communicating expected eating outcomes after GT
planation could be the use of diverse classification systems placement. The results highlight a question of whether
for primary diagnoses previously. The present study is the it is fruitful to strive for the same healthcare guidelines,
first to use the international ICD-10 system for classifying regardless of the child’s primary diagnosis, or if separate
the primary medical diagnoses of children with GT. This guidelines relating to classification of the disorder would be
approach would simplify comparisons between studies in more useful in the clinical context. Lalanne et al5 stated that
the future. as neurological impairment represents the main indication
Complications related to GT including gastric perfora- for prolonged gastrostomy feeding, caregivers should be
tions, tissue granulation, or site infections have been well provided with information regarding not only short term
described in previous studies3,5,20 and have therefore not practical and medical aspects of GT but also the potential
been included in the present study. duration on GT related to the child’s underlying disorder.
A multidisciplinary approach in the care of children This study was performed in 1 administrative region,
with GT has been advised.14,15 Sharp and colleagues15 reflecting that particular demographic, geographic, and
recommend that all care offered to children with severe healthcare setting. This affects the generalizability of the
feeding difficulties should include psychology, nutrition, results, and conclusions must therefore be interpreted with
speech-language pathology, and medicine. This would offer caution. However, restricting the data collection to 1 admin-
the necessary oversight and guidance needed to address the istrative region had the advantage of identifying children
feeding difficulties. The findings from the present study do who were not formally registered in the medical records’
indeed show a wide range of healthcare professionals in- system, by using personal contacts, thus giving a richer data
volved in the care of the children, both prior to and after GT material. Another limitation was the retrospective design
placement. The type of care actions addressed basic abilities that bound the data collection to what was noted in the
such as swallowing, chewing, growth, and positioning, as medical records. This likely affected the variable of “feeding
well as the following aspects of well-being: pain, nausea, habits,” for example, that often had to be estimated based
and feelings related to eating. However, distribution of the on free text because no standardized classification had
582 Nutrition in Clinical Practice 33(4)

been used. Despite the retrospective nature of the data References


collection, the findings are important as they are based on 1. Gauderer MW. Percutaneous endoscopic gastrostomy—20 years later:
legislated, professional documentation, and thereby should a historical perspective. J Pediatr Surg. 2001;36:217-219.
reflect the healthcare provided and communicated across 2. Adams RC, Elias E, Council on Children with Disabilities. Non-
oral feeding for children and youth with developmental or acquired
different healthcare providers.
disabilities. Pediatrics. 2014;134:1745-1762.
The present study found examples of children with AD, 3. Fröhlich T, Richter M, Carbon R, Barth B, Köhler H. Review article:
as well as DD, increasing their oral intake. Future research percutaneous endoscopic gastrostomy in infants and children. Aliment
challenges lie in gaining a deeper understanding this in- Pharmacol Ther. 2009;31:788-801.
crease related to the natural history of different disorders, 4. Diamanti A, Di Ciommo VM, Tentolini A, et al. Home enteral
nutrition in children: a 14-year multicenter survey. Eur J Clin Nutr.
identifying possible predictors of feeding outcomes, and in
2013;67:53.
exploring healthcare that stimulates eating development to 5. Lalanne A, Gottrand F, Salleron J, et al. Long-term outcome of
prevent tube dependency. children receiving percutaneous endoscopic gastrostomy feeding. J
In summary, the findings of the present study show a Pediatr Gastroenterol Nutr. 2014;59:172-176.
clear difference in age at GT placement and eating outcomes 6. Heuschkel RB, Gottrand F, Devarajan K, et al. ESPGHAN position
paper on management of percutaneous endoscopic gastrostomy in
for children with either DD or AD. Children receiving
children and adolescents. J Pediatr Gastroenterol Nutr. 2015;60:131-
GT as a result of AD will most likely begin to eat orally 141.
when the intensive phase of treatment or acute phase of 7. Petersen MC, Kedia S, Davis P, Newman L, Temple C. Eating and
illness is over, whereas children with DD receiving GT at feeding are not the same: caregivers´ perceptions of gastrostomy
early age require healthcare that addresses feeding-related feeding for children with cerebral palsy. Dev Med Child Neurol.
2006;48:5.
and eating-related issues for an extensive period of time. A
8. Engle P, Huffman SL. Growing children’s bodies and minds: max-
more systematic and coherent use of available classification imizing child nutrition and development. Food Nutr Bull. 2010;31(2
systems and multidimensional assessment tools may help suppl):S186.
clinicians and researchers to meet the needs of the indi- 9. Holmes S. Enteral nutrition: an overview. Nurs Stand. 2012;26:41.
vidual child when developing treatment programs, selecting 10. World Health Organization. International Classification of Function-
ing, Disability and Health (ICF). http://apps.who.int/classifications/
functional outcome measures, and communicating across
icfbrowser/. Published 2011. Accessed September 28, 2016.
disciplines. Furthermore, the social aspects of mealtimes, 11. Davies WH, Satter E, Berlin KS, et al. Reconceptualizing feeding and
whether eating orally or not, must not be forgotten given the feeding disorders in interpersonal context: the case for a relational
positive implications this everyday interaction has on child disorder. J Fam Psychol. 2006;20:409-417.
development.30,31 12. Nelson KE, Lacombe-Duncan A, Cohen E, et al. Family experiences
with feeding tubes in neurologic impairment: a systematic review.
Acknowledgments Pediatrics. 2015;136:E140-E151.
13. Agostoni FC, Axelsson WLI, Colomb WLV, et al. The need for
The authors wish to thank Anders Holmén, Ola Andersson,
nutrition support teams in pediatric units: a commentary by the
and Amir Baigi at the Department of Research and Develop-
ESPGHAN Committee on Nutrition. J Pediatr Gastroenterol Nutr.
ment, Region Halland, for input in the design of the study as 2005;41:8-11.
well as for statistical advice. Thank you also to Ebba Sundin, 14. Edwards S, Davis AM, Bruce A, et al. Caring for tube-fed children: a
Halmstad University, and Mats Granlund, Jönköping Univer- review of management, tube weaning, and emotional considerations.
sity, for wise comments greatly improving the manuscript. JPEN J Parenter Enteral Nutr. 2016;40:616-622.
15. Sharp WG, Volkert, VM, Scahill L, McCracken CE, McElhanon B. A
Statement of Authorship systematic review and meta-analysis of intensive multidisciplinary in-
tervention for pediatric feeding disorders: how standard is the standard
E. Backman conceptualized the study, collected data, col-
of care? J Pediatr. 2017;118:116-124.e4
laborated on the analysis and interpretation of data, and
16. Daveluy W, Guimber D, Uhlen S, et al. Dramatic changes in home-
drafted the initial manuscript. A.-K. Karlsson participated in based enteral nutrition practices in children during an 11-year period.
the conception and design of the study, collaborated on the J Pediatr Gastroenterol Nutr. 2006;43:240-244.
analysis of data, and reviewed and revised the manuscript. 17. Pedron-Giner C, Navas-Lopez VM, Martinez-Zazo AB, et al. Analysis
L. Sjögreen participated in the conception and design of the of the Spanish National Registry for Pediatric Home Enteral Nutrition
study, supervised data collection, collaborated on the analysis (NEPAD): implementation rates and observed trends during the past 8
and interpretation of data, and reviewed and revised the years. Eur J Clin Nutr. 2013;67:318-323.
manuscript. All authors read and approved the final manuscript 18. Ricciuto A, Baird R, Sant’Anna A. A retrospective review of enteral
as submitted, and agree to be accountable for all aspects of the nutrition support practices at a tertiary pediatric hospital: a compari-
son of prolonged nasogastric and gastrostomy tube feeding. Clin Nutr.
work.
2015;34:652-658.
19. Dahlseng MO, Andersen GL, Da Graca Andrada M, et al. Gas-
Supplementary Information
trostomy tube feeding of children with cerebral palsy: variation
Additional supporting information may be found online in the across six European countries. Dev Med Child Neurol. 2012;54:938-
supporting information tab for this article. 944.
Backman et al 583

20. World Health Organization. International Statistical Classification of 26. van den Hoek L, Harding C, van Gerven M, Cockerill H. Pediatric
Diseases and Related Health Problems, Tenth Revision (ICD-10). World feeding and swallowing rehabilitation: an overview. J Pediatr Rehabil
Health Organization: Malta; 2011. Med. 2017;10:95-105.
21. The National Board of Health and Welfare. ICD-10-SE, Internationell 27. Brotherton AM, Abbott J, Aggett PJ. The impact of percutaneous
statistisk klassifikation av sjukdomar och relaterade hälsoproblem– endoscopic gastrostomy feeding in children; the parental perspective.
systematisk f örteckning [International statistical classification of Child Care Health Dev. 2007;33:539-546.
diseases and related health problems–a systematic list]. http://www. 28. Sellers D, Pennington L, Mandy A, Morris C. A systematic review
socialstyrelsen.se/klassificeringochkoder/diagnoskodericd-10. of ordinal scales used to classify the eating and drinking abilities of
Published 2011. Accessed June 10, 2016. individuals with cerebral palsy. Dev Med Child Neurol. 2014;56(4):313-
22. World Medical Association. Declaration of Helsinki—ethical princi- 322.
ples for medical research involving human subjects 2013. http://www. 29. Skolin I, Wahlin Y, Broman DA, et al. Altered food intake and
wma.net/en/30publications/10policies/b3/. Published 2013. Accessed taste perception in children with cancer after start of chemotherapy:
May 13, 2016. perspectives of children, parents and nurses. Support Care Cancer.
23. Skolin I, Hernell O, Larsson MV, Wahlgren C, Wahlin YB. Percuta- 2006;14:369-378.
neous endoscopic gastrostomy in children with malignant disease. J 30. Fiese BH, Foley KP, Spagnola M. Routine and ritual elements in family
Pediatr Oncol Nurs. 2002;19:154-163. mealtimes: contexts for child well-being and family identity. New Dir
24. Bauer J, Jürgens H, Frühwald M. Important aspects of nutrition in Child Adolesc Dev. 2006;Spring:67-89.
children with cancer. Adv Nutr. 2011;2:67-77. 31. Harrison ME, Norris ML, Obeid N, Fu M, Weinstangel H, Sampson
25. Åvitsland TL, Kristensen C, Emblem R, Veenstra M, Mala T, M. Systematic review of the effects of family meal frequency on
Bjørnland K. Percutaneous endoscopic gastrostomy in children: a safe psychosocial outcomes in youth. Can Fam Physician. 2015;61:96-
technique with major symptom relief and high parental satisfaction. J 106.
Pediatr Gastroenterol Nutr. 2006;43:624-628.
Letter to the Editor

Nutrition in Clinical Practice


Volume 33 Number 4
Four-Oil Lipid Emulsion (Smoflipid) as a Tool in Managing August 2018 584

C 2018 American Society for

Parenteral Nutrition Shortages Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10106
wileyonlinelibrary.com

Drug shortages call for institutions to be creative to provide tory to insulin therapy. Given this product is new to the U.S.
optimal care to patients. We read with interest the article market, there have been questions regarding its safety and
by Holcombe et al1 reviewing the impact of shortages efficacy. Our institution has been using this product since
and strategies to address them. Our inpatient pharmacy April 2017 and recently completed a retrospective review in-
is using similar approaches to manage the critical amino cluding 24 patients over a 4-month time period. Our results
acid shortage with the addition of incorporating a four-oil were similar to previously published studies that showed a
lipid injectable emulsion (ILE; Smoflipid) into some of our trend toward decreased aspartate aminotransferase (AST;
inpatient care plans. Our institution has found adding four- 165 vs 56 units/L; P = .46), alanine transaminase (ALT;
oil ILE to formulary particularly helpful during times of 96 vs 43 units/L; P = .32), and C-reactive protein (CRP;
parenteral nutrition (PN) shortages. 104 vs 61 mg/L; P = .65). TGs were unchanged before and
All patient protein orders were decreased as a con- after (174 vs 192 mg/dL; P = .65). The duration of four-oil
servation measure institution-wide. In patients who could ILE varied, with 46% of patients receiving 1–7 days, 25%
not tolerate additional dextrose, the dose of four-oil ILE receiving 8–14 days, 17% receiving 15–21 days, and 12%
was increased to >1 g/kg/d and in some cases up to receiving ࣙ21 days. Given the majority of patients received
2 g/kg/d not exceeding 30% total kilocalories. Recom- a short duration of four-oil ILE and still experienced a
mended dosing practices allow higher doses of four-oil ILE decrease in AST/ALT and CRP is promising.
compared with soybean–oil derived lipids. Our institution In summary, incorporating a four-oil ILE into patient
has worked closely with pharmacy information technology care plans to deliver more calories from fat has allowed us
building order panels for PN linked to four-oil ILE and PN to maintain caloric intake despite necessary reductions in
linked to conventional soybean–oil ILE to avoid dispensing protein because of the amino acid shortage. This has been
the wrong ILE product. We also created a report for our one creative approach our institution is taking in response
intravenous (IV) room pharmacist to double-check that to critical PN shortages in addition to those described by
patients are not receiving >1 ILE because having 2 ILE the authors.
products on formulary that look similar could introduce
error. Since these processes have been in place, our institu- Lisa Mostafavifar, PharmD, BCPS, BCNSP
tion has reported zero medication safety events related to Department of Pharmacy, The Ohio State University
dispensing errors. Wexner Medical Center, Columbus, Ohio, USA
There have been no documented adverse effects from
using this higher dose of ILE. At our institution, the four- David C. Evans, MD, FACS
oil ILE is implemented in patients receiving standardized Department of Surgery, The Ohio State University,
commercially available 2-in-1 PN products for initiation or Columbus, Ohio, USA
de-escalation of PN, chronic PN or those deemed chronic
during admission, patients with elevated liver function tests Reference
3 times normal limit, triglycerides (TGs) between 300 and 1. Holcombe B, Mattox TW, Plogsted S. Drug shortages: effect on par-
500 mg/dL, and patients experiencing hyperglycemia refrac- enteral nutrition therapy. Nutr Clin Pract. 2018;33(1):53-61.

Das könnte Ihnen auch gefallen