Sei sulla pagina 1di 196

NCP Nutrition in Clinical Practice

Volume 33 • Number 6 • December 2018

IMPLICATIONS OF MEDICAL
TECHNOLOGY ON NUTRITION
SUPPORT
Nutrition Support in Adult Patients Receiving
Extracorporeal Membrane Oxygenation

Nutrition Support During Pediatric Extracorporeal


Membrane Oxygenation

Metabolic Support of the Patient on Continuous


Renal Replacement Therapy

Use of Intradialytic Parenteral Nutrition in Patients


Undergoing Hemodialysis

Exploring the Potential Effectiveness of Combining


Optimal Nutrition With Electrical Stimulation to Maintain
Muscle Health in Critical Illness: A Narrative Review

Technology in Parenteral Nutrition Compounding

Standards for Nutrition Support: Adult Hospitalized


Patients

Pediatric Nasogastric Tube Placement


and Verification: Best Practice Recommendations
From the NOVEL Project

wileyonlinelibrary.com/journal/ncp
Peer-reviewed, practical solutions in clinical nutrition ISSN: 0884-5336

NCP_33_6_cover.indd 1 31/10/18 3:45 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 J. Klein, PhD
Interim Chief Executive Officer 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 6 December 2018

Editorial
Editor’s Note 736
Jeanette M. Hasse, PhD, RD, LD, FADA, CNSC

Invited Reviews
Nutrition Support in Adult Patients Receiving Extracorporeal Membrane Oxygenation 738
Danielle E. Bear, RD, MRES; Elizabeth Smith, RD, MSc; and Nicholas A. Barrett, FCICM
Nutrition Support During Pediatric Extracorporeal Membrane Oxygenation 747
Bethany J. Farr, MD; Samuel E. Rice-Townsend, MD; and Nilesh M. Mehta, MD
Metabolic Support of the Patient on Continuous Renal Replacement Therapy 754
Erin M. Nystrom, PharmD, BCNSP and Andrea M. Nei, PharmD, BCPS, BCCCP
Use of Intradialytic Parenteral Nutrition in Patients Undergoing Hemodialysis 767
Menaka Sarav, MD and Allon N. Friedman, MD
Exploring the Potential Effectiveness of Combining Optimal Nutrition With Electrical Stimulation to Maintain 772
Muscle Health in Critical Illness: A Narrative Review
Selina M. Parry, PT, PhD; Lee-anne S. Chapple, PhD; and Marina Mourtzakis, PhD
Methods of Enteral Nutrition Administration in Critically Ill Patients: Continuous, Cyclic, Intermittent, 790
and Bolus Feeding
Satomi Ichimaru, PhD, RD, CNSC
Technology in Parenteral Nutrition Compounding 796
Caitlin Curtis, PharmD, BCNSP

Reviews
Effect of Early vs Late Start of Oral Intake on Anastomotic Leakage Following Elective Lower Intestinal Surgery: 803
A Systematic Review
Boudewijn J. J. Smeets, MD; Emmeline G. Peters, MD; Eelco C. J. Horsten, BSc; Teus J. Weijs, PhD;
Harm J. T. Rutten, PhD; Willem A. Buurman, PhD; Wouter J. de Jonge, PhD; and Misha D. P. Luyer, PhD
The Prognostic Role of Phase Angle in Advanced Cancer Patients: A Systematic Review 813
Mayane Marinho Esteves Pereira, BS; Mariana dos Santos Campello Queiroz, BS;
Nathália Masiero Cavalcanti de Albuquerque, BS; Juliana Rodrigues, PhD;
Emanuelly Varea Maria Wiegert, MD; Larissa Calixto-Lima, MD; and Livia Costa de Oliveira, PhD

Clinical Research
Effect of Vitamin D Level on Clinical Outcomes in Patients Undergoing Left Ventricular Assist Device Implantation 825
Fadi Abou Obeid, MD; Gardner Yost, MS; Geetha Bhat, PhD, MD; Erin Drever, MD; and Antone Tatooles, MD
Diagnostic Accuracy of Bioelectrical Impedance Analysis Parameters for the Evaluation of Malnutrition in Patients 831
Receiving Hemodialysis
Angela Teodósio da Silva, MSc; Daniela Barbieri Hauschild, MSc; Yara Maria Franco Moreno, PhD;
João Luiz Dornelles Bastos, PhD; and Elisabeth Wazlawik, PhD
Comparison of Two Methods for Estimating the Tip Position of a Nasogastric Feeding Tube: A Randomized 843
Controlled Trial
Tim Torsy, RN, MSc; Renée Saman, RN, CNS, MSc; Kurt Boeykens, RN, CNS, MSc; Ivo Duysburgh, MD;
Nele Van Damme, RN, MSc; and Dimitri Beeckman, RN, MSc, PhD
Long-Term Use of Mixed-Oil Lipid Emulsion in Adult Home Parenteral Nutrition Patients: A Case Series 851
Manpreet S. Mundi, MD; Megan T. McMahon, PA-C; Jennifer J. Carnell, PharmD; and Ryan T. Hurt, MD, PhD
Nutrition Status Among HIV-Positive and HIV-Negative Inpatients with Pulmonary Tuberculosis 858
Tássia Kirchmann Lazzari, MSc; Gabriele Carra Forte, PhD; and Denise Rossato Silva, MD, PhD
Associations Between Enteral Nutrition and Acute Respiratory Infection Among Patients in New York Metropolitan 865
Region Pediatric Long-Term Care Facilities
Marissa Burgermaster, PhD; Meghan Murray, MPH, RN; Lisa Saiman, MD, MPH;
David S. Seres, MD, ScM, PNS, FASPEN; and Elaine L. Larson, RN, PhD, CIC, FAAN
Reducing Blood Glucose Testing Is Safe in Patients Receiving Parenteral Nutrition 872
Ashley Ratliff, MS, RD, LD, CNSC; Amy Nishnick, RD, LD, CNSC; Robert DeChicco, MS, RD, LD, CNSC;
and Rocio Lopez, MS, MPH
Risk of Malnutrition Evaluated by Mini Nutritional Assessment and Sarcopenia in Noninstitutionalized Elderly People 879
Ilaria Liguori, MD; Francesco Curcio, MD; Gennaro Russo, MD; Michele Cellurale, MD; Luisa Aran, MD;
Giulia Bulli, MD; David Della-Morte, MD, PhD; Gaetano Gargiulo, MD; Gianluca Testa, MD, PhD;
Francesco Cacciatore, MD, PhD; Domenico Bonaduce, MD; and Pasquale Abete, MD, PhD
Height Prediction From Ulna Length of Critically Ill Patients 887
Micheli S. Tarnowski, RD; Estela I. Rabito, RD, PhD; Daieni Fernandes, RD, MSc; Mariane Rosa, RD;
Manoela L. Oliveira, RD; Vânia N. Hirakata, MSc; and Aline Marcadenti, RD, PhD

Clinical Observations
Venovenous Extracorporeal Membrane Oxygenation in an Adult Patient With Prader-Willi Syndrome: 893
A Nutrition Case Report
Stacy Pelekhaty, RD, LDN, CNSC and Jay Menaker, MD
Stoned—A Syndrome of D-Lactic Acidosis and Urolithiasis 897
Casey M. Berman, MD and Russell J. Merritt, MD, PhD
Avoidance of Overt Precipitation and Patient Harm Following Errant Y-Site Administration of Calcium Chloride 902
and Parenteral Nutrition Compounded With Sodium Glycerophosphate
Collin Anderson, PharmD, PhD, BCPS, BCPPS; Chanelle Stidham, PharmD, BCPS;
Sabrina Boehme, PharmD, BCPS, BCPPS; and Jared Cash, PharmD, BCPS

Standards of Practice
Standards for Nutrition Support: Adult Hospitalized Patients 906
Andrew Ukleja, MD, AGAF; Karen Gilbert, RN, MSN, CNSC, ACNP; Kris M. Mogensen, MS, RD-AP, LDN, CNSC;
Renee Walker, MS, RD, LD, CNSC, FAND; Ceressa T. Ward, PharmD, BCPS, BCNSP, BCCCP;
Joe Ybarra, PharmD, BCNSP; Beverly Holcombe, PharmD, BCNSP, FASHP, FASPEN; and Task Force on Standards for
Nutrition Support: Adult Hospitalized Patients, the American Society for Parenteral and Enteral Nutrition

Special Report
Pediatric Nasogastric Tube Placement and Verification: Best Practice Recommendations From the NOVEL Project 921
Sharon Y. Irving, PhD, CRNP, FCCM, FAAN; Gina Rempel, MD, FRCPC; Beth Lyman, RN, MSN, CNSC, FASPEN;
Wednesday Marie A. Sevilla, MD, MPH, CNSC; LaDonna Northington, DNS, RN, BC; Peggi Guenter, PhD, RN, FAAN,
FASPEN; and The American Society for Parenteral and Enteral Nutrition

Cover Art Note


This issue of Nutrition in Clinical Practice is devoted to medical technology that can be used to advance nutrition support or
other aspects of medical care thereby impacting nutrition therapies.
Cover art credit: everythingpossible 
C 123RF.COM

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 6
Editor’s Note December 2018 736

C 2018 American Society for

Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10220
wileyonlinelibrary.com

Medical technology continues to be introduced into clin- authors outline some of the benefits and risks of this
ical practice at a rapid pace. The use of some medical therapy.
technologies may affect when, what, and how we feed our Parry and colleagues introduce the concept of using
patients. Other technologies may be used to help nutrition electrical muscle stimulation in critically ill patients. The
support practitioners individualize nutrition support for authors describe skeletal muscle atrophy that occurs in
their patients. This issue of Nutrition in Clinical Practice critical and prolonged illness and then review the principles
features articles on advances in medical technology and and potential effects of electrical muscle stimulation in
how they affect nutrient needs and the delivery of nutrition preserving muscle health when combined with adequate
support. nutrition.
Recent advances in the use of extracorporeal membrane Other technological improvements are seen in
oxygenation (ECMO) have expanded its use in intensive parenteral nutrition (PN) compounding. In a review
care units (ICUs) for adults and children with severe of PN compounding, Caitlin Curtis discusses how
respiratory or circulatory failure. A review by Danielle technological improvements such as barcode-assisted
Bear and colleagues focuses on delivery of enteral nu- medication preparation systems and electronic health
trition support and overcoming barriers to feeding adult record (EHR)-to-compounder interfaces provide better
patients undergoing ECMO. In a related article, Farr, safeguards and encourages planning for back-up systems
Rice-Townsend, and Mehta address special considerations in the event of EHR failures or electric outages.
for feeding critically ill infants and children undergoing There are 2 American Society for Parenteral and Enteral
ECMO and how to optimize their weight gain and growth Nutrition (ASPEN) documents in this issue. Important
over time. For additional information on this topic, be consensus recommendations from the ASPEN NOVEL
sure to listen to the podcasts with Danielle Bear and project are published in this issue; these recommendations
Dr. Samuel Rice-Townsend discussing nutrition issues re- describe best practices for placing nasogastric tubes (NGT)
lated to ECMO in adult and pediatric populations. In in pediatric patients and verifying location to avoid pa-
addition, the article by Bear and colleagues is the CE article tient harm. Also in this issue are the updated ASPEN
for this issue. “Standards for Nutrition Support for Adult Hospitalized
Continuous renal replacement therapy (CRRT) is often Patients”. These practice-based standards are intended for
utilized in critically ill patients with renal failure. Infor- use by healthcare professionals charged with the care of
mation on the CRRT prescription and implications for patients in any hospital with or without a formal nutrition
nutrition support was reviewed by Erin Nystrom, who support service or team. The Standards address professional
delves into replacement and dialysate fluids and provides responsibilities as they relate to patient assessment, diag-
a case example and an algorithm for estimating how nosis, education, care plan development, implementation,
these solutions and anticoagulation treatment may impact clinical monitoring, evaluation, and other issues of nutrition
caloric delivery. Sarav and Friedman undertook a review support such as nutrition therapy at end-of-life.
of the benefits and continuing controversies of providing
intravenous infusion of essential nutrients to malnour-
ished patients during hemodialysis treatments, known as
intradialytic parenteral nutrition (IDPN). Although its
long-term nutrition benefits need further study, IDPN is
one option for supplementing nutrition in patients re- Jeanette M. Hasse, PhD, RD, LD, FADA, CNSC
quiring hemodialysis who have poor nutrient intake. The Editor-in-Chief, Nutrition in Clinical Practice
Invited Review

Nutrition in Clinical Practice


Volume 33 Number 6
Nutrition Support in Adult Patients Receiving Extracorporeal December 2018 738–746

C 2018 American Society for

Membrane Oxygenation Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10211
wileyonlinelibrary.com

Danielle E. Bear, RD, MRES1,2,3,4 ; Elizabeth Smith, RD, MSc1,2 ;


2,3,4
and Nicholas A. Barrett, FCICM

Abstract
The use of extracorporeal membrane oxygenation (ECMO) for both severe respiratory and cardiac failure is increasing. Because
these patients are some of the sickest in the intensive care unit, a multidisciplinary approach to their treatment, including appropriate
nutrition therapy, is warranted. Currently, limited data exist on the optimal timing, type, and amount of nutrition to be provided.
This review focuses on describing the current nutrition practices in patients receiving ECMO, details research that is currently being
undertaken, and lists important research questions that require exploration in this field. Observational data suggest that early enteral
nutrition is safe and that although nutrition targets can be met, underfeeding is still common. Until further research is available,
these patients should be fed according to guidelines for the general critically unwell population. (Nutr Clin Pract. 2018;33:738–746)

Keywords
critical illness; enteral nutrition; extracorporeal membrane oxygenation; nutrition assessment; nutrition requirements; nutrition
support

Introduction (CESAR) study.2 ECMO has 2 configurations: venovenous


(VV), which is used for respiratory support, and veno-
Extracorporeal membrane oxygenation (ECMO) is a life- arterial (VA), which is used for cardiac support. To provide
saving supportive therapy used for patients with severe respi- ECMO, blood is drained from the great veins and passed
ratory and/or cardiac failure. ECMO for adult cardiorespi- through a gas exchange membrane that adds oxygen and
ratory failure has seen significant growth in the last decade, removes carbon dioxide; the blood is then returned to the
following the influenza A (H1N1) pandemic of 2009–20111 circulation into either the right atrium (VV ECMO) or aorta
and the conventional ventilatory support vs extracorporeal (VA ECMO).3,4
membrane oxygenation for severe adult respiratory failure

From the 1 Department of Nutrition and Dietetics, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom; 2 Department of
Critical Care, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom; 3 Lane Fox Research Clinical Respiratory Physiology
Research Centre, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom; and 4 Centre for Human and Applied Physiological
Sciences, King’s College London, London, United Kingdom.
Financial disclosure: D.E.B. is funded by a Health Education England/National Institute for Health Research (NIHR) ICA Clinical Doctoral
Fellowship (ICA-CDRF-2015-01-047). The views expressed are those of the authors and are not necessarily those of the National Health Service
(NHS), the NIHR, or the Department of Health.
Conflicts of interest: None declared.
This article originally appeared online on October 15, 2018.

Podcast available
Listen to a discussion of this manuscript with NCP Editor-in-Chief Jeanette M. Hasse, PhD, RD, LD, FADA, CNSC, and author Danielle E.
Bear, RD, MRES. This and other NCP podcasts are available at: https://onlinelibrary.wiley.com/page/journal/19412452/homepage/podcasts

Corresponding Author:
Danielle E. Bear, RD, MRES, HEE/NIHR Clinical Doctoral Fellow and Critical Care Dietitian, St Thomas’ Hospital, Westminster Bridge Road,
London SE1 7EH, United Kingdom.
Email: danielle.bear@gstt.nhs.uk
Bear et al 739

Despite the fact that these patients are some of the sickest support with ECMO, or contributed to by the EN strategy.
and most resource-intensive patients in the intensive care However, a recent retrospective observational study of pa-
unit (ICU), there is a paucity of data regarding the optimum tients receiving VA ECMO for at least 2 days and receiving
timing, type, and amount of nutrition support that should early EN reported that early EN was associated with both
be provided. Indeed, the Extracorporeal Life Support Or- lower in-hospital (hazard ratio [HR] 0.78, 95% CI: 0.62–
ganization, the umbrella organization of ECMO providers, 0.98, P = .032) and 28-day mortality (HR 0.74, 95% CI:
includes only 1 line regarding nutrition support in their 0.56–0.97, P = .031).18 Despite this finding, prospective
Guidelines for Adult Respiratory Failure, which states “full randomized controlled trials are warranted because only
energy and protein support is essential.”5 This lack of 12% of patients received early EN, and reasons for feeding
guidance is reflected in the significant variation in feed- decisions cannot be elucidated. Indeed, in a recent large
ing practices reported both in the United Kingdom and trial of EN vs early parenteral nutrition (PN) in critically ill
internationally.6,7 However, it is acknowledged that investi- patients with shock who were receiving vasopressor support,
gating nutrition support practices to improve outcome and there was no difference in the primary outcome of 28-day
recovery in these patients is warranted.8 all-cause mortality.19 However, those in the EN group had
There is much debate surrounding optimum nutrition higher rates of adverse gastrointestinal events.19 Notably,
support practices for the general critically ill patient; how- more patients in the EN group suffered bowel ischemia
ever, the importance of avoiding overfeeding in the early (19 [2%] vs 5 [<1%], 3.84 [1.43–10.3], P = .007). Although
stages of critical illness is agreed on.9 Given patients re- a secondary outcome, this may be worth considering in
ceiving ECMO are some of the most acutely unwell in the patients receiving VA ECMO who are in severe shock
ICU, this point may be even more relevant, and differences and receiving high-dose vasopressor support whereby it
in responses to feeding strategies between patients receiving may not be unreasonable to delay EN for a short time
VV and VA ECMO may be present. with daily review to assess a more appropriate time to
We aim to provide a comprehensive review of the commence.
evidence surrounding the provision and management of Nasogastric feeding is the most common route of nutri-
nutrition support in adult patients receiving ECMO, and tion support reported in all studies. Use of postpyloric feed-
provide some questions to direct future research in this area. ing appears to be infrequent overall, although rates of up to
33% have been reported.16 Postpyloric feeding tube place-
ment is more difficult and takes a longer time to achieve
Timing and Route of Nutrition Support than gastric placement; however, successful insertion rates
Concerns about the safety of early enteral nutrition (EN) of >90% using a bedside placement technique with an
in critically ill patients exist, particularly in those patients electromagnetic device have been reported.20 The relative
who are in significant states of shock (inadequate organ acceptability and perceived utility of gastric compared with
perfusion) and receiving high-dose vasopressors (commonly postpyloric feeding appears to vary between institutions
adrenergic drugs).9 For these reasons, historical practices and countries, with an international survey of feeding
encouraged withholding, or at least delaying, EN during practices in adult patients receiving ECMO reporting that
ECMO support.10 However, recent European guidelines11 postpyloric feeding is most commonly used in the United
have recommended that early EN is safe and feasible in these States compared with other countries.7 It may also indicate
patients, albeit based on expert opinion. that this method of feeding may be more common in centers
Seven observational studies were found describing nu- who are experienced at bedside placement techniques in
trition practices in patients receiving ECMO (both VV their general ICU cohort.
and VA).10,12-17 In these studies (Table 1), EN was most The reported use of PN, either alone or in combination
frequently commenced within 24 hours of starting ECMO, with EN, ranges from 4% to 30% of patients receiving
with several studies reporting a mean or median time to ECMO support. The use of PN in patients receiving ECMO
feeding of around 13 hours. In the largest study,16 96% remains controversial because of the possibility of lipid
of patients were fed within 24 hours. The reporting of infiltration into the oxygenator causing oxygenator failure.21
adverse events is rare and likely to be impacted on by the However, data supporting this are limited and although it
predominantly retrospective nature of the studies, but in a is clearly a possibility, it is not currently possible to make
prospective multicenter observational study of 107 patients, evidence-based recommendations as to the appropriate use
5 patients (4.5%) were reported to have bowel ischemia.15 of PN during ECMO.22 In addition, newer generation
This was a population of 60% VA ECMO and 40% VV membranes used in the circuit may now negate this risk.8
ECMO patients, but it is not known which type of ECMO Given this, if PN is deemed to be in the best interest of the
those patients who experienced bowel ischemia were receiv- patients on nutrition grounds, it is worth ensuring that close
ing. Thus, it cannot be determined whether this occurrence monitoring of the ECMO circuit is undertaken when pa-
was due solely to the underlying disease, due in part to tients are receiving PN, especially if other lipid-containing
740
Table 1. All Observational Studies Describing Nutrition Practices in Patients Receiving ECMO.

Authors Lu et al MacGowan Ridley et al Ferrie et al Umezawa Lukas et al Scott et al


(2018)17 et al (2018)16 (2015)15 (2013)13 Makikado (2010)12 (2004)10
et al (2013)14
Patient population 102a 203 107 86 7 48 27
Study design Retrospective Retrospective Prospective, Retrospective Prospective Retrospective Retrospective
observational observational multicenter observational observational observational observational
observational
Age, y (median)b 55.4 (mean) 44 42 44.4 (mean) NR 44 NR
APACHE II score Survived: 22 18 20 26 (mean) NR NR NR
(median)b Deceased: 26
(mean)
Type of ECMO VA: 77 VV VA: 61% VA: 31 patients VA VA: 35 patients VV
VV: 25 VV: 39% VV: 55 patients VA VV: 13 patients
Provision of energy Survived: 97% Median energy target Median daily Mean daily nutrition Mean cumulated Mean nutrition NR
during ECMO Deceased: 88% delivered: 89.8% delivery: 20 delivery ࣙ80% in deficit by end of adequacy
Kcal goal achieved kcal/kg 52/86 patients study −3264.94 during
kcal/d ECMO: 55%
(VV: 67%, VA:
50%)
Provision of protein >100% protein Median target Median daily 73% of daily protein NR NR NR
during ECMO requirement delivered: 84.7% delivery: 0.9 g/kg goal tolerated in
the first 2 weeks
Definition and 73% of surviving Prokinetics 66% of patients ࣙ2 aspirates Day 1: GRVs 4-hourly GRVs ×2 consecutive
incidence of GI patients tolerated considered after 2 × received >200 mL and checked every >200 mL GRVs >150 mL
intolerance bolus feeding, GRVs >300 mL prokinetic abdominal 6 hours indicated erythromycin
27% tolerated Prokinetics used in medications distension/ Day 2: every intolerance started
continuous 52.2% of patients discomfort 12 hours GRV >150 mL 95% received
NG feedingc in 33 patients (38%) From day 3 daily prokinetics prokinetics by
No postpyloric started 48 hours
tubes or 71% of patients
prokinetics used received
prokinetics

(continued)
Table 1. (continued)

Time taken to initiate Survived: 1.6 days 13.5 13 13.1 (mean) 2 patients within NR 25 patients within
nutrition support, Deceased: 0.8 days 24 hours 24 hours
hours (median)b (mean) 7 patients within 27 patients within
48 hours 36 hours
Method used to Energy: Energy: 25 kcal/kg/d Energy: Schofield Energy: Schofield Energy: 25 Energy: Schofield Energy: 25 kcal/kg
estimate nutrition Harris-Benedict Protein: 1.2 g/kg/d equation + SF equation + SF kcal/kg/d equation + SF Protein: 1.2–1.5
targets during equation +AF (minimum) (51% of patients) (1.1–1.2) Protein: NR (1.2–1.5) g/kg/d
ECMO (1.2) +SF Weight based Protein: 1.2 g/kg/d Protein:
(1.2–1.5) (33%) (minimum) 1.2–1.5 g/kg/d
Protein: 1.2 g/kg/d Other (16%) 1.5–2 g/kg (RRT)
(minimum) Protein: NR
Route of nutrition NG: 80% of patients Gastric: 60.6% 1602 study Gastric: all patients EN in all patients EN: 69% EN: 18
support Nil other routes Postpyloric: 34.5% days:EN: 1342 started gastric PN: 4% PN: 1
reported PN: 4.9% (84%)d feeds EN and PN: 25% EN and PN: 8
PN: 111 (7%)d Postpyloric: nil 1 patient received no
ON: 155 (10%)d PN: 18 patients nutrition
No nutrition: 83
(5%)d

AF, Activity Factor; APACHE II, Acute Physiology and Chronic Health Evaluation; ECMO, extracorporeal membrane oxygenation; EN, enteral nutrition; GI, gastrointestinal; GRV, gastric
residual volume; NG, nasogastric; NR, not reported; ON, oral nutrition; PN, parenteral nutrition; RRT, renal replacement therapy; SF, stress factor; VA, veno-arterial; VV, venovenous.
a Population divided into survived (n = 41) and deceased (n = 61).
b Median unless otherwise specified.
c Definition of tolerance not provided.
d Reported as number and percent of study days.

741
742 Nutrition in Clinical Practice 33(6)

medications are used concomitantly. Regular monitoring consuming nature of this method, the authors report that
of triglycerides may also help to ascertain risk and the they used an in-house, custom-made adaptor to connect
point at which lipid-free PN may be warranted. In our the metabolic cart to the ECMO circuit, which is not avail-
practice, for all patients, transoxygenator gas exchange with able elsewhere. Wollersheim et al26 presented an alternative
regular serum lipids is monitored once daily if patients are method that still requires measurement of gas exchange
receiving PN. For patients with triglycerides level >3 g/L, using IC at the native lung, but rather than connecting
consideration is given to using lipid-free PN with weekly the IC to the ECMO circuit, blood gas is measured before
lipids and daily fat-soluble vitamin administration. and after the membrane oxygenator, with carbon dioxide
and oxygen content being subsequently calculated using
Dose of Nutrition Support a referenced equation.28 The Weir equation is then used
to determine EE. Both protocols provide an opportunity
The optimal dose of nutrition support in terms of energy
for EE to be measured in patients receiving ECMO, but
and protein in the critically ill population is unknown,9 and
require prospective validation, particularly at different time
this is also true for patients receiving ECMO. The American
points during the ECMO run because EE is not static and
Society for Parenteral and Enteral Nutrition guidelines23
different blood flow rates may influence the measurement.
recommend the use of a nutrition risk scoring tool in all
However, these methods will most certainly allow further
critically ill patients to determine the level of attention paid
investigations into the optimum energy targets for patients
to the dose of both energy and protein. However, given that
receiving both VA and VV ECMO.
severity of illness scoring systems are reliant on physiology
As with general ICU patients, when IC is not available,
and age, it is likely that they underestimate the severity
predictive or weight-based equations are the method of
of illness on ECMO because physiology is manipulated,
choice for determining energy targets in patients receiving
patients are commonly younger than the average ICU
ECMO,10,12-17 although these have not been derived from
patient, patients may have been in the ICU for some days
patients receiving ECMO, and it is not known whether
before commencing ECMO, and patients tend to have a
they are truly appropriate. Studies of clinical practice de-
longer ICU stay. Consequently, it is likely that current
scribe the main equations used to be 25 kcal/kg12,16-18 or
nutrition risk scores may systematically underestimate the
Schofield with added stress factor12,13,15 ranging from 1.1 to
actual risk, and individualized nutrition support regimens
1.5.12,13 When compared with measured energy expenditure
should be provided to these patients.
(MEE), Wollersheim et al26 demonstrated that common
prediction equations significantly underfed or overfed pa-
Energy tients, with 48% overestimated by >500 kcal, when com-
Indirect calorimetry (IC) is considered the gold standard for pared with MEE. Importantly, the weight-based equation,
determining energy expenditure (EE) in critically ill adults 25 kcal/kg,23,24 was shown to significantly underestimate
and is recommended as first-line choice in evidence-based EE when compared with MEE. This finding contradicts de
nutrition guidelines.23,24 IC determines EE by measuring Waele et al,29 who found that MEE in patients receiving
the amount of oxygen inhaled and carbon dioxide exhaled, ECMO was on average 19 kcal/kg, albeit in a group of only
and relies on several assumptions including a stable dis- 6 patients. The differences in these 2 studies may be because
tribution of carbon dioxide and knowledge of nitrogen of different methods used to measure EE or differences in
loss.25 Measuring IC in patients receiving ECMO needs to ECMO; one included only VV ECMO patients,26 whereas
take into account gas exchange via the native lung and via the other included both VV and VA ECMO patients.29 This
the membrane lung, and also the impact of recirculation. underscores the importance of undertaking further research
Recirculation adds an error into the calculation because in this area and using clinical judgment at the bedside when
both the carbon dioxide and oxygen exchange across the estimating energy targets in these patients.
membrane lung can be underestimated. Recirculation also In the absence of nutrition guidelines specifically for
varies based on the state of intravascular blood volume, as patients receiving ECMO, it appears reasonable to follow
well as thoraco-abdominal compliance and cardiac output. currently available guidelines for general critically unwell
Recirculation fraction can be measured using ultrasonic patients that recommend IC where available or weight-based
techniques. equations ranging from 20 to 25 kcal/kg.23,24 European So-
Two studies26,27 have demonstrated it is possible to mea- ciety for Parenteral and Enteral Nutrition guidelines24 go on
sure EE in patients receiving ECMO in 2 ways. De Waele to further recommend 25–30 kcal/kg when the patient has
et al27 described a method whereby EE is first measured in moved into the recovery phase of critical illness. With more
the native lung via the ventilator; then the metabolic cart is and more patients being liberated from the ventilator and
connected to the ECMO circuit to measure at the artificial mobilizing out of bed while still receiving ECMO,30,31 it may
lung. Both numbers are then added together and inserted be suitable to increase energy provision to 30 kcal/kg when
into the Weir equation for calculation. Along with the time- a patient is in this phase of recovery despite still receiving
Bear et al 743

ECMO. However, currently, no studies have measured EE muscle size (total muscle thickness) was associated with
in ambulatory patients receiving ECMO. lower strength and mobility score.36
Improved outcomes have been demonstrated in general Only 1 study aiming to investigate optimum protein in-
ICU patients receiving around 80% of their estimated en- takes in patients receiving ECMO exists. This small study in-
ergy targets32 and 70% of measured energy targets,33 which vestigated the nitrogen balance in obese and nonobese adult
is likely to be comparable in patients receiving ECMO. patients receiving VV ECMO.37 Protein targets were set
Although often the most unwell patients in an ICU, studies at 1.5–2.0 g/kg/d for nonobese patients and 2.0–2.5 g/kg/d
have shown it is possible for patients receiving ECMO for obese patients, with around 85% of this target be-
to achieve and tolerate this target. Ferrie et al13 reported ing delivered. Results were compared between obese and
that 60% of their patients receiving ECMO received >80% nonobese individuals, and it was concluded that obese
of the estimated energy targets, and more recently Mac- compared with nonobese patients had a more negative
Gowan et al16 showed that 89.8% of target energy was nitrogen balance (−1.7 ± 5.7 vs −11.5 ± 9.6). Despite
delivered to patients receiving VV ECMO. However, in the the fact that nitrogen balance has inherent limitations that
latter study, underfeeding was still common with patients make interpretation in the critically ill population difficult,38
receiving <80% of their energy targets on one-quarter of these data provide some initial insights into the nitrogen
feeding days. Interestingly, both studies found a difference balance and potential protein requirements in these patients,
in the energy delivered in the ECMO and post-ECMO and this topic should certainly be explored further.
periods, with those in the post-ECMO period receiving From existing observational data undertaken in patients
more. It is thought that the reason for this relates to receiving ECMO, a minimum 1.2 g/kg/d is targeted for pro-
both gastric/enteral feeding intolerance and to the greater tein in clinical practice. Although there is a discrepancy be-
number of procedures requiring cessation of EN during the tween prescribed and delivered protein intakes, it is possible
ECMO run.16 to meet >80% of the prescribed target,16,17 which has been
When considering the relationship of the dose of energy shown to lead to lower mortality in critically ill, non-ECMO
to clinical outcome in adult patients with ECMO, little data patients.33,39 Whether meeting current recommendations for
exist. However, receiving adequate energy intake (>80%) protein intake in this patient population improves outcomes
was associated with longer length of stay in 1 study.16 It is not related to muscle wasting is unknown, but higher protein
not known whether this is simply due to the patients who intakes were associated with a longer length of stay in
remained in the ICU longer being able to reach nutrition the largest observational study.16 However, as mentioned
targets, or whether there is a true burden associated with previously, these data were not adjusted for length of stay.
nutrition that increases length of stay. In contrast, in a Until further data are available, it would not seem unrea-
retrospective study of Taiwanese patients receiving ECMO, sonable to aim for protein intakes in line with the general
achieving >80% of nutrition targets was associated with critically ill population accounting for clinical condition and
improved mortality.17 It is clear that prospective random- factors such as the use of continuous renal replacement
ized controlled trials are required to further investigate these therapy.
findings.
Barriers to the Delivery of EN
Protein Delayed gastric emptying is frequently cited as a
Recommendations for protein intakes in the critically ill complication associated with enteral feeding in patients
range from 1.2 g/kg/d for the general ICU patient up to receiving ECMO.6,12,15,16 This was confirmed in a United
2.5 g/kg/d for those who are obese or admitted with burns Kingdom–wide survey of nutrition practices in patients
or after trauma.23,24 However, these recommendations are receiving VV ECMO, where 69% of responders reported
based on limited, mainly observational data. The assumed that ࣙ50% of their patients require prokinetics while
benefit of providing adequate protein is a reduction in skele- receiving VV ECMO.6
tal muscle wasting, which may lead to improved physical However, the definition of delayed gastric emptying
function post-ICU.34 Providing adequate protein to patients is based on gastric residual volumes (GRVs), and there
receiving ECMO may be particularly important because are a variety of thresholds ranging from 150 to 300 mL.
of the significant amount of muscle wasting experienced. Furthermore, in the observational studies reported to date,
Recently, the trajectory of muscle wasting, measured using those with a lower acceptable GRV report a higher incidence
muscle ultrasound, in patients receiving both VV and VA of medications used to promote gastric emptying than those
ECMO was reported. In line with critically ill, non-ECMO with a higher GRV threshold. Where a GRV of 150 mL is
patients, rectus femoris cross-sectional area had decreased accepted and postpyloric feeding not used, prokinetic use
by 19% at day 10 and 30% at day 20.35 Other quadriceps is 71%–95%.10,12 Furthermore, there are other approaches,
muscles also decreased in size to a similar degree. Smaller including use of postpyloric feeding, on an ad hoc or
744 Nutrition in Clinical Practice 33(6)

planned basis, which alter the utility of GRV measurements. relationship between total nutrition received and
It is not currently known what an ideal GRV threshold is, or change in muscle quantity and quality.
indeed whether they should be measured at all.40 3. Characterizing changes in muscle quantity and
The largest observational study to date indicated that quality in patients requiring ECMO during criti-
prokinetics/postpyloric feeding was used in >50% of their cal illness (ECMO USS) (NCT02995811) aims to
patients with the result being that >80% of the energy identify changes in different muscle groups (trunk,
and protein targets were delivered.16 This indicates that respiratory, and skeletal muscle) in patients receiv-
with a flexible approach to nutrition support, underfeeding ing ECMO. The association between nutrition and
can be avoided. Although patients in this study were all changes in muscle quality and quantity will also be
receiving VV ECMO, Ferrie et al13 reported no difference addressed.
in the incidence of enteral feeding intolerance between 4. Progressive Rehabilitation Therapy in Patients with
those receiving VA and VV ECMO, so the common be- Advanced Lung Disease (NCT 03562728) aims to
lief that VA ECMO reduces gut perfusion and therefore investigate the impact of a multimodal rehabilitation
reduces motility may not hold true in modern-day clinical program in ameliorating the loss of muscle mass and
practice. strength, and lower extremity balance, strength, and
Delayed gastric emptying is not the only barrier to coordination in patients requiring lung transplant or
delivering EN in these patients. Both MacGowan et al16 ECMO as a bridge to transplant. The multimodal
and Ridley et al15 reported bedside and operating room rehabilitation program includes neuromuscular elec-
procedures to be the most frequent reason for enteral tric stimulation, strength and mobility training, and
feeding interruptions, occurring in >30% of patients. This nutrition supplementation with amino acids 3 times
is also a frequently cited reason for feeding interruptions daily.
in the general critically ill population, and strategies such
as volume-based feeding have proven useful in prevent- Potential Research Questions
ing the associated poor delivery of nutrition.41 However,
No prospective randomized controlled trials exist examin-
volume-based feeding has not been studied specifically in
ing the optimum route, timing, or adequacy of nutrition
patients receiving ECMO, but should not be discounted as
support in patients receiving ECMO, but it is clear that
an option, particularly in the more stable patient. Certainly
these are urgently required. Because VA and VV patients
guidelines directing when EN should be stopped and started
may be different in terms of their metabolic sequelae, we
for procedures have been shown to increase nutrition targets
recommend that VA and VV patients are recruited and
in non-ECMO centers42 and should be considered as stan-
investigated separately, or at least subgroup analysis is
dard practice for all critically ill patients.
considered for each group where they have been recruited
together.
Studies Planned or in Progress The Intensive Care Medicine research agenda for extra-
A search of clinical trials databases was undertaken to deter- corporeal life support acknowledges the limited evidence
mine details of any studies that are in progress investigating surrounding the optimal feeding strategy for these patients.8
aspects of nutrition in patients receiving ECMO. Only 3 Although nutrition did not feature in the top 10 recom-
studies were found. mended research trials for these patients, early rehabili-
tation/mobilization did. We would propose that nutrition
and rehabilitation go hand in hand,34,43 and encourage
investigators to report appropriate nutrition parameters in
1. Measuring Energy Expenditure in ECMO
any of these future studies.
(Extracorporeal Membrane Oxygenation) Patients
We have outlined 5 research questions that we feel are
(MEEP) (NCT01992237) aims to describe a
priorities to answer:
calculation to determine nutrition targets in patients
receiving ECMO. Secondary aims include describing
levels of nutrition needs for different mechanical 1. What is the optimum energy intake in patients receiv-
ventilation states, and to use oxygen and carbon ing VV and VA ECMO?
dioxide elimination to estimate cardiac output. 2. What is the optimum protein intake in patients
2. The use of Computed Tomography (CT) to Measure receiving VV and VA ECMO?
Skeletal Muscle Quantity and Quality in Patients 3. What is the impact of nutrition support on the phys-
Receiving ECMO (NCT03269825) aims to investi- ical and functional recovery of patients receiving VA
gate the clinical predictive value of and change in and VV ECMO?
muscle quantity and quality in patients receiving 4. What are the optimum energy and protein targets
VV ECMO. Secondary aims are to determine any during rehabilitation in patients receiving ECMO?
Bear et al 745

5. Are nutrient losses (micronutrients and macronutri- 7. Nandwani V, Tauber A, McCarthy P, Herr D. 333. Nutrition practice
ents) present across the ECMO membrane, and are patterns in adult ECMO patients: results of an international survey.
Crit Care Med. 2014;42(12 suppl 1):A1440.
these losses clinically meaningful?
8. Combes A, Brodie D, Chen YS, et al. The ICM research agenda on
extracorporeal life support. Intensive Care Med. 2017;43(9):1306-1318.
Conclusion 9. Preiser JC, van Zanten AR, Berger MM, et al. Metabolic and nutri-
tional support of critically ill patients: consensus and controversies. Crit
The use of ECMO as a lifesaving treatment for both Care. 2015;19:35. https://doi.org/10.1186/s13054-015-0737-8.
severe respiratory and cardiac failure is increasing. It is 10. Scott LK, Boudreaux K, Thaljeh F, Grier LR, Conrad SA. Early en-
clear that these patients are some of the sickest in the teral feedings in adults receiving venovenous extracorporeal membrane
ICU and require a multidisciplinary approach to treatment oxygenation. JPEN J Parenter Enteral Nutr. 2004;28(5):295-300.
11. Reintam Blaser A, Starkopf J, Alhazzani W, et al. Early enteral
that includes appropriate nutrition therapy. Currently, there
nutrition in critically ill patients: ESICM clinical practice guidelines.
are limited data to guide nutrition therapy, but it appears Intensive Care Med. 2017;43(3):380-398.
that early EN is safe. Until further data are available, 12. Lukas G, Davies AR, Hilton AK, et al. Nutritional support in adult
nutrition should be provided in line with current guidelines patients receiving extracorporeal membrane oxygenation. Crit Care
for critically ill patients. Prospective randomized controlled Resusc. 2010;12(4):230-234.
13. Ferrie S, Herkes R, Forrest P. Nutrition support during extracorporeal
trials investigating optimal nutrition in these patients are
membrane oxygenation (ECMO) in adults: a retrospective audit of 86
urgently required. patients. Intensive Care Med. 2013;39(11):1989-1994.
14. Umezawa Makikado LD, Flordelis Lasierra JL, Perez-Vela JL, et al.
Statement of Authorship Early enteral nutrition in adults receiving venoarterial extracorporeal
membrane oxygenation: an observational case series. JPEN J Parenter
D. E. Bear, N. A. Barrett, and E. Smith contributed to Enteral Nutr. 2013;37(2):281-284.
conception/design of the manuscript; D. E. Bear, N. A. 15. Ridley EJ, Davies AR, Robins EJ, et al. Nutrition therapy in adult pa-
tients receiving extracorporeal membrane oxygenation: a prospective,
Barrett, and E. Smith contributed to acquisition, analysis,
multicentre, observational study. Crit Care Resusc. 2015;17(3):183-189.
or interpretation of the data; D. E. Bear, N. A. Barrett, 16. MacGowan L, Smith E, Elliott-Hammond C, et al. Adequacy
and E. Smith drafted the manuscript; D. E. Bear, N. A. of nutrition support during extracorporeal membrane oxygenation
Barrett, and E. Smith critically revised the manuscript; and [published online ahead of print January 26, 2018]. Clin Nutr.
D. E. Bear, N. A. Barrett, and E. Smith agree to be fully https://https://doi.org/10.1016/j.clnu.2018.01.012
17. Lu MC, Yang MD, Li PC, et al. Effects of nutritional intervention
accountable for ensuring the integrity and accuracy of the
on the survival of patients with cardiopulmonary failure undergoing
work. All authors read and approved the final manuscript. extracorporeal membrane oxygenation therapy. In Vivo. 2018;32(4):
829-834.
Supplementary Information 18. Ohbe H, Jo T, Yamana H, et al. Early enteral nutrition for cardiogenic
Additional supporting information may be found online in the or obstructive shock requiring venoarterial extracorporeal membrane
oxygenation: a nationwide inpatient database study. Intensive Care
Supporting Information section at the end of the article.
Med. 2018;44(8):1258-1265.
19. Reignier J, Boisrame-Helms J, Brisard L, et al. Enteral versus
parenteral early nutrition in ventilated adults with shock: a ran-
References domised, controlled, multicentre, open-label, parallel-group study
1. Noah MA, Peek GJ, Finney SJ, et al. Referral to an extracorporeal (NUTRIREA-2). Lancet. 2018;391(10116):133-143.
membrane oxygenation center and mortality among patients with 20. Bear DE, Champion A, Lei K, Camporota L, Barrett NA. Electro-
severe 2009 influenza A(H1N1). JAMA. 2011;306(15):1659-1668. magnetically guided bedside placement of post-pyloric feeding tubes
2. Peek GJ, Mugford M, Tiruvoipati R, et al. Efficacy and economic in critical care. Br J Nurs. 2017;26(18):1008-1015.
assessment of conventional ventilatory support versus extracorporeal 21. Buck ML, Ksenich RA, Wooldridge P. Effect of infusing fat emulsion
membrane oxygenation for severe adult respiratory failure (CESAR): a into extracorporeal membrane oxygenation circuits. Pharmacotherapy.
multicentre randomised controlled trial. Lancet. 2009;374(9698):1351- 1997;17(6):1292-1295.
1363. 22. Lee HM, Archer JR, Dargan PI, Wood DM. What are the adverse
3. Brodie D. The evolution of extracorporeal membrane oxygenation for effects associated with the combined use of intravenous lipid emulsion
adult respiratory failure. Ann Am Thorac Soc. 2018;15(suppl 1):S57- and extracorporeal membrane oxygenation in the poisoned patient?
S60. Clin Toxicol (Phila). 2015;53(3):145-150.
4. Sherren PB, Shepherd SJ, Glover GW, et al. Capabilities of a mobile 23. McClave SA, Taylor BE, Martindale RG, et al. Guidelines for the
extracorporeal membrane oxygenation service for severe respiratory provision and assessment of nutrition support therapy in the adult
failure delivered by intensive care specialists. Anaesthesia. 2015;70(6): critically ill patient: Society of Critical Care Medicine (SCCM) and
707-714. American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.).
5. Extracorporeal Life Support Organization (ELSO). ELSO JPEN J Parenter Enteral Nutr. 2016;40(2):159-211.
Guidelines for Adult Respiratory Failure, Version 1.4. August 2017. 24. Kreymann KG, Berger MM, Deutz NE, et al. ESPEN guide-
https://www.elso.org/Portals/0/ELSO%20Guidelines%20For%20Adult lines on enteral nutrition: intensive care. Clin Nutr. 2006;25(2):
%20Respiratory%20Failure%201_4.pdf 210-223.
6. Bear DE, Halslam J, Camporota L, Shankar-Hari M, Barrett NA. An 25. Brandi LS, Bertolini R, Calafa M. Indirect calorimetry in criti-
international survey of nutrition practices in adult patients receiving cally ill patients: clinical applications and practical advice. Nutrition.
veno-venous ECMO. Intensive Care Med Exp. 2015;3(suppl 1):A295. 1997;13(4):349-358.
746 Nutrition in Clinical Practice 33(6)

26. Wollersheim T, Frank S, Muller MC, et al. Measuring energy ex- 36. Puthucheary ZA, Rawal J, McPhail M, et al. Acute skeletal muscle
penditure in extracorporeal lung support Patients (MEEP)—protocol, wasting in critical illness. JAMA. 2013;310(15):1591-1600.
feasibility and pilot trial. Clin Nutr. 2018;37(1):301-307. 37. Pelekhaty S, Galvagno SM Jr, Hochberg E, et al. Nitrogen balance
27. De Waele E, van Zwam K, Mattens S, et al. Measuring resting during venovenous extracorporeal membrane oxygenation support:
energy expenditure during extracorporeal membrane oxygenation: pre- preliminary results of a prospective, observational study [published
liminary clinical experience with a proposed theoretical model. Acta online ahead of print May 25, 2018]. JPEN J Parenter Enteral Nutr.
Anaesthesiol Scand. 2015;59(10):1296-1302. https://https://doi.org/10.1002/jpen.1176
28. Dash RK, Bassingthwaighte JB. Erratum to: Blood HbO2 and HbCO2 38. Kopple JD. Uses and limitations of the balance technique. JPEN J
dissociation curves at varied O2, CO2, pH, 2,3-DPG and temperature Parenter Enteral Nutr. 1987;11(5 suppl):79s-85s.
levels. Ann Biomed Eng. 2010;38(4):1683-1701. 39. Nicolo M, Heyland DK, Chittams J, Sammarco T, Compher C. Clin-
29. De Waele E, Staessens K, Demol J, La Meir M, Sapen HD. SUN-P027: ical outcomes related to protein delivery in a critically ill population: a
the energy expenditure of patients on ECMO is not elevated: beware multicenter, multinational observation study. JPEN J Parenter Enteral
when you feed them! Clin Nutr. 2017;36:S62. Nutr. 2016;40(1):45-51.
30. Fan E, Gattinoni L, Combes A, et al. Venovenous extracorporeal mem- 40. Reignier J, Mercier E, Le Gouge A, et al. Effect of not mon-
brane oxygenation for acute respiratory failure: a clinical review from itoring residual gastric volume on risk of ventilator-associated
an international group of experts. Intensive Care Med. 2016;42(5):712- pneumonia in adults receiving mechanical ventilation and early
724. enteral feeding: a randomized controlled trial. JAMA. 2013;309(3):249-
31. Marhong JD, DeBacker J, Viau-Lapointe J, et al. Sedation and mo- 256.
bilization during venovenous extracorporeal membrane oxygenation 41. Heyland DK, Murch L, Cahill N, et al. Enhanced protein-energy
for acute respiratory failure: an international survey. Crit Care Med. provision via the enteral route feeding protocol in critically ill patients:
2017;45(11):1893-1899. results of a cluster randomized trial. Crit Care Med. 2013;41(12):2743-
32. Heyland DK, Cahill N, Day AG. Optimal amount of calories for 2753.
critically ill patients: depends on how you slice the cake! Crit Care Med. 42. Segaran E, Barker I, Hartle A. Optimising enteral nutrition in crit-
2011;39(12):2619-2626. ically ill patients by reducing fasting times. J Intensive Care Soc.
33. Zusman O, Theilla M, Cohen J, et al. Resting energy expenditure, 2016;17(1):38-43.
calorie and protein consumption in critically ill patients: a retrospective 43. Heyland DK, Stapleton RD, Mourtzakis M, et al. Combining nutrition
cohort study. Crit Care. 2016;20(1):367. and exercise to optimize survival and recovery from critical illness:
34. Bear DE, Wandrag L, Merriweather JL, et al. The role of nutritional conceptual and methodological issues. Clin Nutr. 2016;35(5):1196-
support in the physical and functional recovery of critically ill patients: 1206.
a narrative review. Crit Care. 2017;21(1):226.
35. Hayes K, Holland AE, Pellegrino VA, Mathur S, Hodgson CL. Acute
skeletal muscle wasting and relation to physical function in patients
requiring extracorporeal membrane oxygenation (ECMO). J Crit Care.
2018;48:1-8.
Invited Review

Nutrition in Clinical Practice


Volume 33 Number 6
Nutrition Support During Pediatric Extracorporeal December 2018 747–753

C 2018 American Society for

Membrane Oxygenation Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10212
wileyonlinelibrary.com

Bethany J. Farr, MD1,2 ; Samuel E. Rice-Townsend, MD1,2 ;


and Nilesh M. Mehta, MD2,3,4

Abstract
Providing adequate nutrition to critically ill pediatric patients is essential and positively impacts outcomes. Critically ill infants and
children receiving extracorporeal membrane oxygenation (ECMO) therapy are nutritionally vulnerable, yet there are challenges to
reliable assessment of nutrition requirements and to the delivery of optimal nutrition in this cohort. In this review of the relevant
literature, we present the current evidence and guidelines for the optimal prescription and delivery of nutrition for pediatric patients
receiving ECMO. We also discuss nutrient delivery considerations in ECMO survivors and identify areas where further study is
needed. (Nutr Clin Pract. 2018;33:747–753)

Keywords
critical illness; enteral nutrition; extracorporeal membrane oxygenation; nutrition requirements; nutrition assessment; nutrition
support; parenteral nutrition; pediatrics

Introduction population is heterogenous, encompassing neonates with


congenital heart disease who are receiving veno-arterial
Critically ill patients in the pediatric intensive care unit (VA) ECMO to young adults with cystic fibrosis awaiting
(PICU) require adequate nutrition support to achieve the lung transplant who are receiving venovenous (VV) ECMO,
best outcomes. Patients with respiratory or cardiorespira- with varied needs and considerations. However, as use of
tory failure who require support with extracorporeal mem- ECMO has expanded over the last 2 decades, now with
brane oxygenation (ECMO) are, by definition, a severely ill >15,000 cases annually, our understanding of nutrition
and vulnerable subset of the critically ill population who requirements and optimal nutrient delivery practices for this
would benefit from optimal nutrition. Yet, the assessment patient group needs to be reviewed.1 We aimed to examine
of energy and protein requirements and the delivery of the existing literature on prescription and delivery of nutri-
these nutrients can be a particular challenge because of the tion therapy in these critically ill patients. In addition, we
unclear metabolic demands and potential safety concerns sought to identify gaps in knowledge where future research
of enteral nutrition (EN) in these patients. Moreover, the efforts are needed.

From the 1 Department of Surgery, Boston Children’s Hospital, Boston, Massachusetts, USA; 2 Harvard Medical School, Boston, Massachusetts,
USA; 3 Division of Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston,
Massachusetts, USA; and 4 Center for Nutrition, Boston Children’s Hospital, Boston, Massachusetts, USA.
Financial disclosure: None declared.
Conflicts of interest: None declared.
This article originally appeared online on October 16, 2018.

Podcast available
Listen to a discussion of this manuscript with NCP Editor-in-Chief Jeanette M. Hasse, PhD, RD, LD, FADA, CNSC, and author Samuel E.
Rice-Townsend, MD. This and other NCP podcasts are available at: https://onlinelibrary.wiley.com/page/journal/19412452/homepage/podcasts

Corresponding Author:
Nilesh M. Mehta, MD, Division of Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s
Hospital, 300 Longwood Avenue, Bader 634, Boston, MA 02115, USA.
Email: nilesh.mehta@childrens.harvard.edu
748 Nutrition in Clinical Practice 33(6)

Nutrition Requirements During Pediatric the protein breakdown seen in neonates receiving ECMO;
ECMO however, this practice has not been adopted because further
research is needed.10
Macronutrient Requirements
Our current understanding of and ability to accurately mea- Macronutrient Intake and Outcomes
sure energy needs in patients receiving ECMO is limited. Although data are limited to guide the prescription of
Few studies have explored this question, and results have nutrition therapy, the importance of the nutrition state
been contradictory. In a study where energy expenditure in at the onset of ECMO therapy and of the delivery of
neonates receiving ECMO was measured by stable isotope adequate nutrition during therapy is clear. Anton et al11
tracer techniques, patients were hypermetabolic both during reported in 2016 that being underweight at the time of
ECMO and in the post-ECMO phase.2 Although patients ECMO cannulation was an independent factor for mor-
receiving ECMO are often presumed to be hypermetabolic tality. Of 491 patients, 24% were underweight at the time
because of the inflammatory burden of an extracorporeal of ECMO initiation and had an increased odds ratio of
circuit combined with underlying severe illness, this may not 1.99 for in-hospital mortality as compared with normal-
always be true. Using respiratory mass spectroscopy, energy weight and obese children.11 The importance of adequate
expenditure in infants who underwent Norwood surgery nutrient delivery for these patients may be extrapolated
was reported to decline rapidly within 8 hours after surgery.3 from studies performed on other critically ill patients, the
In infants who underwent Fontan surgery with cardiopul- sickest of whom may ultimately require ECMO. For such
monary bypass, the immediate postoperative metabolic patients, effective delivery of nutrition while in the intensive
measurements revealed hypometabolism.4 In another study care unit (ICU) has been shown to impact outcomes, even
using an intravenous isotope-labeled infusion technique to mortality. For mechanically ventilated patients in 31 PICUs
estimate energy requirements in neonates supported with in 8 countries, receiving less than a third of the prescribed
ECMO, energy needs were not significantly greater than energy during the first 10 days after admission to the PICU
age-matched counterparts.5 was associated with significantly higher 60-day mortality.12
Due to the technical difficulties of traditional methods Another single-center retrospective study of PICU patients
to accurately measure gas exchange in a patient attached with acute respiratory distress syndrome demonstrated that
to an ECMO circuit, data on energy expenditure are scant adequate delivery of energy (>80% predicted REE) and
in children receiving ECMO.6 Without indirect calorimetry, protein (>1.5 g/kg/d) was associated with improved survival
intensivists must rely on standard equations to estimate to ICU discharge.13 In patients with a >4-day PICU stay,
resting energy expenditure (REE). These equations, how- receiving at least 25% of goal calories via the enteral route
ever, were developed using data from healthy children and over the first 48 hours of admission resulted in a lower
are therefore frequently inaccurate in estimating REE in mortality overall compared with those who did not meet
critically ill children.7 Therefore, the reliance on standard this goal.14 The role of nutrient delivery route in patients
equations to determine energy needs may result in un- receiving ECMO is discussed later in this review.
intended underestimation or overestimation of REE and Adequate protein delivery was also associated with more
result in caloric overfeeding or underfeeding. A reliable ventilator-free days and reduced mortality after adjusting
and practical method to measure energy expenditure in for illness severity.13 Similarly, in a multicenter study of
patients receiving ECMO is needed. Until then, existing 1245 mechanically ventilated children, delivery of ࣙ60% of
best practices established in the general critically ill pediatric prescribed protein requirement was associated with lower
and neonatal populations should be used to guide energy 60-day mortality. Mortality for patients with <20% ade-
delivery goals for patients supported with ECMO. quate protein delivery was 9.3%, and decreased to 5.6% in
Neonatal and pediatric patients receiving ECMO those receiving 20%–60% recommended protein, and 3.2%
have increased protein requirements because of profound in those receiving >60% recommended protein, demonstrat-
catabolism. It is essential to offset the protein losses ing a dose-dependent relationship.15 The route of nutrient
from this catabolic state with provision of adequate delivery may be an important consideration, and early
protein.2,8 In neonates, whole-body protein breakdown enteral feeding is generally preferred in critically ill children.
was found to be 100% higher than in age-matched healthy
neonates.2 In neonates treated with ECMO, >1.5 g/kg/d
protein is needed to achieve a positive nitrogen balance.9 Micronutrient Requirements
Nutrition guidelines of neonatal patients receiving ECMO Children who are receiving extended ECMO may expe-
recommend provision of up to 3 g/kg/d protein to offset rience aggravated losses of micronutrients. In an ex vivo
catabolic losses.8 It has been suggested that administration model of ECMO, circuit losses of essential amino acid
of insulin, as an anabolic hormone, may negate some of isoleucine, vitamin A, and vitamin E were demonstrated.16
Farr et al 749

In an animal model of smoke-induced acute lung injury, study was not randomized. The first 9 patients were treated
ECMO application was associated with exaggerated losses with PN only, whereas the next 7 patients received EN,
of selenium.17 The mechanisms of these alterations in initiated between days 3 and 9 of ECMO, introducing a
micronutrients are not clear, but their impact on outcomes potential for bias and confounding.
in pediatric ECMO may be important and require fur- Several retrospective studies have reported that enteral
ther investigation. In patients receiving ECMO who also feeding can be tolerated without excessive risk in patients
require dialysis, micronutrient losses may be further com- receiving VA and VV ECMO. One retrospective chart review
pounded, because continuous renal replacement therapy over a 5-year period reported success in feeding in 67 of 77
has been associated with loss of ionized calcium, inorganic neonatal patients receiving ECMO.24 In patients who were
phosphorus, and selenium, among other trace elements.18,19 fed, there were no reports of bilious emesis, bloody stools, or
These observations may have implications for micronutrient abdominal distention; however, feeds were frequently held
supplementation in the pediatric ECMO population, and at- for a variety of reasons. Notably in this study, 80% of the
tempts to monitor micronutrients should be made. However, infants being enterally fed were receiving inotropic support.
the assessment of the micronutrient status of critically ill Another cohort study of enterally fed pediatric patients
patients is challenging because of the confounding effects of receiving VA and VV ECMO reported no increase in
inflammation on micronutrient levels in plasma. In patients sepsis, abdominal pathology, or respiratory infections, while
receiving ECMO, these challenges are further compounded demonstrating the cost-effectiveness of this route relative to
by the dilutional effects of the extracorporeal circuit blood PN.23 A retrospective single-institution study showed that
volume. Overall, micronutrient distribution, losses, and neonates receiving VA ECMO who were enterally fed did
requirements in pediatric ECMO remain important areas not have any increase in bacteremia with gut pathogens as
for future investigations. compared with the infants receiving PN, concluding that
there was no increase in translocation associated with EN.25
Similarly, in another study including 49 infant and pediatric
Timing and Route of Nutrient Delivery patients receiving ECMO, there was no association between
EN and increased infection or abdominal pathology.26
Timing of Initiation Multiple retrospective studies including adults receiving
Current guidelines for critically ill pediatric patients recom- ECMO have also shown that enteral delivery is safe and well
mend initiation of EN within 24–48 hours of admission tolerated, with no adverse complications.27-29
whenever possible.7 In patients receiving ECMO, there are In light of these reports, a recent survey of 96 in-
multiple potential barriers to EN, and this route is often not stitutions characterizes the current institutional practice
sufficient to meet daily nutrient delivery goals.20 The use of patterns around nutrition in pediatric and neonatal patients
parenteral nutrition (PN) to supplement insufficient EN is receiving ECMO.30 Eighty-four percent reported using EN
therefore prevalent in this group. However, a recent study during ECMO, and 385 of these sites used an algorithm for
has questioned the role of early PN in critically ill children, initiating and advancing EN. The use of EN was reported by
although patients receiving ECMO were not included.21 a higher proportion of respondents during VV (71%) vs VA
Early discussions around the safe and efficient route of (55%). EN delivery was practiced by 23% during escalating
nutrient delivery are crucial in children receiving ECMO, doses of vasopressor and 60% when vasopressors were being
particularly those with poor nutrition status at baseline or weaned. It is evident that retrospective data have demon-
with a protracted course of illness. strated safety with the use of EN, and recommendations
favor its use. Current practice has also followed this trend
in increasing use of EN in patients with ECMO.
Safety of EN
The safety of enteral delivery of nutrition with ECMO has
long been a concern. It was initially argued that enteral
Timing of PN
feeding in the setting of decreased intestinal perfusion Most studies of nutrition in patients receiving ECMO
would increase the risk for gut ischemia, translocation of demonstrate significant use of PN as a supplemental or
bacteria, and sepsis.22,23 Thus, PN has been preferred in primary source of nutrition within the first week. There
many centers as a means to achieve nutrient delivery goals are conflicting data on the appropriate timing of initiation
during the acute phase. Using differential carbohydrate of PN, and none specific to the ECMO population. A
absorption, gut permeability was shown to be increased recent study in critically ill pediatric patients reported harm
in neonatal patients receiving ECMO relative to healthy from early initiation of PN (within 24 hours of admission)
infants. However, providing minimal enteral feeding to compared with a strategy of withholding PN during the
this group did not negatively alter permeability or lead to first week in the ICU.21 A decrease in incidence of new
complications.22 It should be noted that this retrospective infections and decreased length of ICU and hospital stay
750 Nutrition in Clinical Practice 33(6)

were reported in those who received late initiation of PN Still, EN averaged only 10% of energy needs in the first
(after 1 week). Infants and children receiving ECMO were week of ECMO. Younger age, diagnosis of congenital
not included in this study. In addition, there were several diaphragmatic hernia (CDH), and use of VA ECMO, but
aspects of the study design that limit external validity.21,31 not use of vasoactive agents, were factors associated with
In the absence of data, a reasonable approach in infants low EN use.20 In a review of 491 pediatric patients receiving
and children receiving ECMO may be to emphasize early ECMO, PN was initiated in 88% of patients at a median
and cautious advancement of EN and to initiate PN within time of day 1 from cannulation. EN was initiated in 30%
3–5 days in children with malnutrition and within 5–7 days of patients, and 35% of those reached full enteral feeds. Of
in well-nourished children if nutrient delivery goal has not note, 17% had feeds stopped for a concern of pneumatosis
been achieved by EN alone.32 intestinalis.11
In early years of ECMO, lipid emulsion for patients We have described multiple reports of the safety and
receiving PN was delivered either via a separate intravenous tolerance of enteral feeds in patients receiving ECMO
(IV) catheter or directly into the circuit. In an in vitro after initial resuscitation and hemodynamic stability is
study, layering and agglutination of lipid emulsion in the achieved, even while receiving vasoactive infusions.23,24,26
circuit was noted by 30 minutes of the infusion. A survey The benefits of initiating enteral feeds in an infant with
conducted at that time revealed that 73% of ECMO centers ongoing hemodynamic instability need to be balanced
preferentially used a separate IV access for lipid infusions, with the potential for gut ischemia, bacterial translocation,
and 18% used the ECMO circuit for infusion, with all cen- and sepsis secondary to poor perfusion and splanchnic
ters reporting complications such as agglutination, cracking vasoconstriction. The clinical apprehension for gut-related
of stopcocks, membrane malfunction, and clot formation.33 worsening after introduction of enteral feeding in infants
Based on such studies that suggested potential risk to receiving ECMO is prevalent, resulting in a reliance on
the circuit from direct infusion, lipid infusion through PN during the acute phase.30 Critically ill children who
a separate IV catheter was recommended.34 More recent are not receiving ECMO support have tolerated EN while
experience suggests that intralipid infusions are not associ- receiving vasoactive medications with no adverse abdominal
ated with oxygenator failure with newer polymethylpentene complications, while adjusting for illness severity and degree
membranes.35 of vasoactive infusion use.36 These findings further support
that vasoactive use should not necessarily prohibit initiation
Practical Aspects of Nutrient Delivery: of EN in pediatric patients receiving ECMO.
We have included a suggested nutrient delivery algorithm
Suggested Algorithm (Figure 1) for pediatric patients receiving ECMO. Because
In 2010, the American Society for Parenteral and Enteral there is wide variability in patient factors and underlying
Nutrition published guidelines for delivery of nutrition to disease processes, it is meant as a starting place as considera-
neonates receiving ECMO.8 The guidelines recommended tion is given to the individual patient’s needs and condition.
protein requirements of up to 3 g/kg/d and energy require-
ments equivalent to healthy subjects. Early introduction Nutrition-Related Outcomes in Neonatal
of enteral feeding was recommended once the patient had
stabilized. In practice, a majority (84.2%) of providers re-
ECMO Survivors
ported use of EN during the ECMO course, and postpyloric There are limited studies relevant to nutrition in patients
feeding was preferred over gastric. An institutional feeding who survive ECMO, and most of this research, all on
protocol for patients receiving ECMO is used in a minority neonates, has focused on the CDH population. There have
of centers.30 In general, the delivery of nutrients by EN been reports of feeding difficulties in neonates after ECMO.
during the early ECMO course is limited, with a reliance One study found that antroduodenal motility was impaired
on PN.20,26 In a single-center report of 49 neonatal and in 10 neonatal ECMO survivors (excluding congenital heart
pediatric patients receiving ECMO, PN was used in 55% disease and CDH), and this was associated with failure to
of patients, any EN in 45% of patients, and an estimated achieve full feeds, longer hospitalization, and diagnosis of
average of 67% of goal energy needs were supplied by day reflux, and predicted the need for feeding tube placement.
5 of ECMO. For patients receiving some EN, it accounted Abnormalities were noted on head computed tomographic
for 38% of total energy supplied, on average.26 In another scans in the majority of this cohort, which may suggest a
institutional report of 54 neonatal and pediatric noncardiac central mechanism for feeding difficulty in some ECMO
patients receiving ECMO, energy and protein delivery was neonatal survivors.37
adequate by the end of 1 week receiving ECMO. PN was Among newborns who receive ECMO therapy, those
the major source of nutrition (94%); however, 85% of with CDH have longer duration of ECMO and more
patients were receiving a combination of PN and EN at complicated hospital courses with higher rates of bron-
day 7, with 44% receiving greater than trophic EN volumes. chopulmonary dysplasia, gastroesophageal reflux, feeding
Farr et al 751

Detailed nutritional Enteral advancement plan:


• Start with continuous feeds at
assessment within 24h: 10 mL/kg/day and advance
• Calculate estimated energy every 6-12 h
requirement using • Goal delivery of 80%
At 24-48h assess hemodynamic stability: estimated energy requirement
Schofield or WHO
• Stable vasoactive medications by one week
equation.
• Fluid resuscitation complete • Consider PN if unlikely to
• Protein minimum 1.5
• No concern for NEC achieve 50% of EN goal by
g/kg/day day 3-5
• Stable lactate levels

Yes
Signs of EN intolerance:
• Increased girth, distension or
No emesis
• Hold feeds for 2h then
Contraindications for EN: Consider reassess, restart if
• Unrepaired CDH starting PN improvement
• Significant ileus • Consider use of pro-kinetic
• Other prohibitive agents and bowel regimen
• If gastric feeds not tolerated or
abdominal pathology
high risk of aspiration
consider post-pyloric feeds
Consider starting EN:
Yes No • Trophic feeds with slow
advancement
• Monitor for intolerance
Consider PN if unable to
tolerate goal EN:
Consider • By day 3-5 for neonates
starting PN and undernourished
• By day 5-7 for others

Figure 1. Our proposed algorithm: a rational approach to nutrition support during pediatric extracorporeal membrane
oxygenation. CDH, congenital diaphragmatic hernia; EN, enteral nutrition; NEC, necrotizing enterocolitis; PN, parenteral
nutrition; WHO, World Health Organization.

problems, and hypotonia. At 1 year of age, the CDH needs requires further investigation. The optimal timing
population had adequate weight gain but had a feeding of PN use in patients receiving ECMO needs to be
tube requirement in 36%, as opposed to the patients with determined. In a subgroup of patients receiving chronic
meconium aspiration who had all transitioned to full oral ECMO support, such as those receiving VV ECMO as a
feeds.38 Recently, a prospective longitudinal study reported bridge to lung transplant, there is an increasing recognition
growth outcomes up to 12 years in 172 patients with CDH, of the role of physical therapy along with nutrition to
43 of whom required ECMO. Growth was persistently optimize rehabilitation.40,41 Attention to optimal nutrient
lagging in both groups, and the ECMO group had the delivery, preferably via the enteral route, along with active
poorest growth. This trend continued into adolescence, rehabilitation has been safely achieved in these patients.42
demonstrating stunting in both groups that was most severe Many centers have developed strategies to minimize
in the ECMO population. Notably, none of the patients immobilization, provide passive physical therapy at the
were feeding tube dependent by 8 years of age.39 This study bedside, and provide active therapy once feasible in patients
highlights the ongoing nutrition challenges of children with receiving ECMO. The safety and effect of exercise with
CDH and the need for close follow-up and multidisciplinary nutrition on muscle mass and strength in ECMO survivors
interventions. needs to be further studied.

Directions for Future Research Summary


The accurate assessment of energy and protein needs in Optimal nutrition is an important goal for infants and
children receiving ECMO is an important area for future children who are receiving ECMO therapy. Although
research. The impact of the mode of ECMO support specific studies in this vulnerable group of patients are
and underlying diagnoses on REE and macronutrient scant, there have been some important observations made.
752 Nutrition in Clinical Practice 33(6)

There is potential for energy overfeeding from inaccurate 13. Wong JJ-M, Han WM, Sultana R, Loh TF, Lee JH. Nutrition delivery
estimates of energy requirement and inadequate protein affects outcomes in pediatric acute respiratory distress syndrome.
JPEN J Parenter Enteral Nutr. 2016;41(6):1007-1013.
delivery in the setting of higher requirements in this
14. Mikhailov TA, Kuhn EM, Manzi J, et al. Early enteral nutrition
group. The safety of EN has been demonstrated, and an is associated with lower mortality in critically ill children. JPEN
algorithmic approach with low-volume feeds early, followed J Parenter Enteral Nutr. 2014;38(4):459-466.
by cautious advancement as tolerated is prudent. Future 15. Mehta NM, Bechard LJ, Zurakowski D, Duggan CP, Heyland DK.
studies must explore the impact of bundled interventions, Adequate enteral protein intake is inversely associated with 60-d
mortality in critically ill children: a multicenter, prospective, cohort
including optimal nutrition and physical therapy, on clinical
study. Am J Clin Nutr. 2015;102(1):199-206.
and functional outcomes. 16. Estensen K, Shekar K, Robins E, McDonald C, Barnett AG, Fraser
Statement of Authorship JF. Macro- and micronutrient disposition in an ex vivo model of
extracorporeal membrane oxygenation. Theor Chem Acc. 2014;2(1):29.
B. J. Farr, S. E. Rice-Townsend, and N. M. Mehta equally https://icm-experimental.springeropen.com/articles/10.1186/s40635-
contributed to the conception and design of the manuscript; 014-0029-7
B. J. Farr, S. E. Rice-Townsend, and N. M. Mehta contributed 17. McDonald CI, Fung YL, Shekar K, et al. The impact of acute lung
to the acquisition and analysis of the data; B. J. Farr drafted the injury, ECMO and transfusion on oxidative stress and plasma selenium
manuscript. All authors critically revised the manuscript, agree levels in an ovine model. J Trace Elem Med Biol. 2015;30:4-10.
to be fully accountable for ensuring the integrity and accuracy 18. Datzmann T, Träger K, Reinelt H, Von Freyberg P. Elimination rates
of electrolytes, vitamins, and trace elements during continuous renal re-
of the work, and read and approved the final manuscript.
placement therapy with citrate continuous veno-venous hemodialysis:
influence of filter lifetime. Blood Purif. 2017;44:210-216.
References 19. Broman M, Bryland A CO. Trace elements in patients on continuous
1. Butt W, MacLaren G. Extracorporeal membrane oxygenation 2016: an renal replacement therapy. Acta Anaesthesiol Scand. 2017;61(6):650-
update. F1000Research. 2016;5:F1000 Faculty Rev-750. 659.
2. Keshen TH, Miller RG, Jahoor F, Jaksic T. Stable isotopic quantitation 20. Armstrong LB, Ariagno K, Smallwood CD, Hong C, Arbuthnot
of protein metabolism and energy expenditure in neonates on- and M, Mehta NM. Nutrition delivery during pediatric extracorporeal
post-extracorporeal life support. J Pediatr Surg. 1997;32(7):958-963. membrane oxygenation therapy. JPEN J Parenter Enteral Nutr.
3. Li J, Zhang G, Herridge J, et al. Energy expediture and caloric and 2018;42(7):1133-1138.
protein intake in infants following the Norwood procedure. Pediatr Crit 21. Fivez T, Kerklaan D, Mesotten D, et al. Early versus late parenteral
Care Med. 2008;9(1):55-61. nutrition in critically ill children. N Engl J Med. 2016;374(12):1111-
4. Mehta NM, Costello JM, Bechard LJ, et al. Resting energy expenditure 1122.
after fontan surgery in children with single-ventricle heart defects. 22. Piena M, Albers MJIJ, Van Haard PMM, Gischler S, Tibboel D.
JPEN J Parenter Enteral Nutr. 2012;36(6):685-692. Introduction of enteral feeding in neonates on extracorporeal mem-
5. Jaksic T, Shew SB, Keshen TH, Dzakovic A, Jahoor F. Do critically brane oxygenation after evaluation of intestinal permeability changes.
ill surgical neonates have increased energy expenditure? J Pediatr Surg. J Pediatr Surg. 1998;33(1):30-34.
2001;36(1):63-67. 23. Pettignano R, Heard M, Davis R, Labuz M, Hart M. Total enteral nu-
6. Li X, Yu X, Cheypesh A, Li J. Non-invasive measurements of energy trition versus total parenteral nutrition during pediatric extracorporeal
expenditure and respiratory quotient by respiratory mass spectrometry membrane oxygenation. Crit Care Med. 1998;26(2):358-363.
in children on extracorporeal membrane oxygenation—a pilot study. 24. Hanekamp MN, Spoel M, Sharman-Koendjbiharie I, Peters JWB,
Artif Organs. 2015;39(9):815-819. Albers MJIJ, Tibboel D. Routine enteral nutrition in neonates
7. Mehta NM, Skillman HE, Irving SY, et al. Guidelines for the provision on extracorporeal membrane oxygenation. Pediatr Crit Care Med.
and assessment of nutrition support therapy in the pediatric critically 2005;6(3):275-279.
ill patient: Society of Critical Care Medicine and American Society 25. Wertheim HFL, Albers MJIJ, Piena-Spoel M, Tibboel D. The inci-
for Parenteral and Enteral Nutrition. JPEN J Parenter Enteral Nutr. dence of septic complications in newborns on extracorporeal mem-
2017;41(5):706-742. brane oxygenation is not affected by feeding route. J Pediatr Surg.
8. Jaksic T, Hull MA, Modi BP, Ching YA, George D, Compher C. 2001;36(10):1485-1489.
A.S.P.E.N. clinical guidelines: nutrition support of neonates supported 26. Greathouse KC, Sakellaris KT, Tumin D, et al. Impact of early ini-
with extracorporeal membrane oxygenation. JPEN J Parenter Enteral tiation of enteral nutrition on survival during pediatric extracorporeal
Nutr. 2010;34(3):247-253. membrane oxygenation. JPEN J Parenter Enteral Nutr. 2018;42(1):205-
9. Weber TR, Shah M, Stephens C TT. Nitrogen balance in patients 211.
treated with extracorporeal membrane oxygenation. J Pediatr Surg. 27. Scott LK, Boudreaux K, Thaljeh F, Grier LR, Conrad SA. Early en-
1993;28(7):906-908. teral feedings in adults receiving venovenous extracorporeal membrane
10. Agus MSD, Javid PJ, Ryan DP, Jaksic T. Intravenous insulin decreases oxygenation. JPEN J Parenter Enteral Nutr. 2004;28(5):295-300.
protein breakdown in infants on extracorporeal membrane oxygena- 28. Umezawa Makikado LD, Flordelı́s Lasierra JL, Pérez-Vela JL, et al.
tion. J Pediatr Surg. 2004;39(6):839-844. Early enteral nutrition in adults receiving venoarterial extracorporeal
11. Anton M, Papacostas M, Lee E, Nakonezn P, Green M. Underweight membrane oxygenation. JPEN J Parenter Enteral Nutr. 2013;37(2):281-
status is an independent predictor of in-hospital mortality in pediatric 284.
patients on extracorporeal membrane oxygenation. JPEN J Parenter 29. Ferrie S, Herkes R, Forrest P. Nutrition support during extracorporeal
Enteral Nutr. 2016;201(1):1-8. membrane oxygenation (ECMO) in adults: a retrospective audit of
12. Mehta N, Bechard L, Cahill N, Wang M. Nutritional practices and 86 patients. Intensive Care Med. 2013;39(11):1989-1994.
their relationship to clinical outcomes in critically ill children—an in- 30. Desmarais TJ, Yan Y, Keller MS, Vogel AM. Enteral nutrition in
ternational multicenter cohort study. Crit Care Med. 2012;40(7):2204- neonatal and pediatric extracorporeal life support: a survey of current
2211. practice. J Pediatr Surg. 2015;50(1):60-63.
Farr et al 753

31. Mehta NM. Parenteral nutrition in critically ill children. N Engl J Med. 38. Bernbaum J, Schwartz IP, Gerdes M, et al. Survivors of extracorporeal
2016;374(12):1190-1192. membrane oxygenation at 1 year of age: the relationship of primary
32. Jimenez L, Mehta NM, Duggan CP. Timing of the initiation of diagnosis with health and neurodevelopmental sequelae. Pediatrics.
parenteral nutrition in critically ill children. Curr Opin Clin Nutr Metab 1995;96(5 Pt 1):907-913.
Care. 2017;20(3):227-231. 39. Leeuwen L, Mous DS, van Rosmalen J, et al. Congenital diaphragmatic
33. Buck ML, Ksenich RA, Wooldridge P. Effect of Infusing Fat Emulsion hernia and growth to 12 years. Pediatrics. 2017;140(2):e20163659.
into Extracorporeal Membrane Oxygenation Circuits. Pharmacother- https://https://doi.org/10.1542/peds.2016-3659
apy. 1997;17(6):1292-1295. 40. Ulerich L. Nutrition implications and challenges of the transplant
34. Buck ML, Wooldridge P, Ksenich RA. Comparison of methods for patient undergoing extracorporeal membrane oxygenation therapy.
intravenous infusion of fat emulsion during extracorporeal membrane Nutr Clin Pract. 2014;29(2):201-206.
oxygenation. Pharmacotherapy. 2005;25(11):1536-1540. 41. Hayes D, Tobias JD, Galantowicz M, Preston TJ, Tzemos KK, Mc-
35. Brogan TV, Lequier L, Lorusso R, MacLaren G, Peek G, eds. Extra- Connell PI. Video fluoroscopy swallow study and nutritional support
corporeal Life Support: The ELSO Red Book. 5th ed. Ann Arbor, MI: during ambulatory venovenous extracorporeal membrane oxygenation
Extracorporeal Life Support Organization; 2017. as a bridge to lung transplantation. World J Pediatr Congenit Hear Surg.
36. Panchal AK, Manzi J, Connolly S, et al. Safety of enteral feedings 2014;5(1):91-93.
in critically ill children receiving vasoactive agents. JPEN J Parenter 42. Zebuhr C, Sinha A, Skillman H, Buckvold S. Active rehabilitation
Enteral Nutr. 2016;40(2):236-241. in a pediatric extracorporeal membrane oxygenation patient. PM R.
37. Jadcherla SR, Berseth CL. Antroduodenal motility and feeding out- 2014;6(5):456-460.
come among neonatal extracorporeal membrane oxygenation sur-
vivors. J Pediatr Gastroenterol Nutr. 2005;41(3):347-350.
Invited Review

Nutrition in Clinical Practice


Volume 33 Number 6
Metabolic Support of the Patient on Continuous Renal December 2018 754–766

C 2018 American Society for

Replacement Therapy Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10208
wileyonlinelibrary.com

Erin M. Nystrom, PharmD, BCNSP; and Andrea M. Nei, PharmD, BCPS, BCCCP

Abstract
Continuous renal replacement therapy (CRRT) is the modality of choice in critically ill patients with hemodynamic instability
requiring renal replacement therapy. The goal of this review is to discuss an overview of CRRT types, components, and important
considerations for nutrition support provision. Evidence basis for guidelines and our recommendations are reviewed. Nutrition
support–related implications include the possibility of calorie gain with citrate-based anticoagulation, calorie loss with glucose-
free replacement fluids and dialysate, and significant amino acid losses in effluent. We challenge nutrition support clinicians to
develop a keen understanding of the specific CRRT modalities that are employed in their intensive care units and to be able to
determine how the CRRT prescription may impact a patient’s nutrition support prescription. (Nutr Clin Pract. 2018;33:754–766)

Keywords
critical care; dialysis; intensive care unit; nutritional support; renal replacement therapy

Introduction The CRRT circuit lends significant complexity given im-


portant macronutrient, micronutrient, and electrolyte losses
Acute kidney injury (AKI) is a common complication in or gains that must be accounted for when formulating a nu-
critically ill patients, with a reported incidence of >30%, trition support regimen. A general understanding of CRRT
depending on definitions used.1-3 Approximately 4%–6% of types and prescription variables is thus essential for the
patients with AKI in the intensive care unit (ICU) require a clinician responsible for the nutrition prescription. While
form of renal replacement therapy (RRT).1,4-6 Continuous guidelines have attempted to provide a general framework
renal replacement therapy (CRRT) offers multiple theoreti- for practice, it is challenging to extrapolate dated studies of
cal advantages over intermittent hemodialysis (IHD), but is macronutrient disposition in CRRT to contemporary clini-
primarily used in the presence of hemodynamic instability cal practice given advancements in CRRT technology, dos-
and/or need for aggressive fluid removal (ultrafiltration) in ing, and types of fluids used. Whereas electrolytes and fluids
the setting of massive hypervolemia.7 have more specific indicators to guide requirements via lab,
Malnutrition has been estimated to afflict up to 42% of physical exam, or other clinical parameters, macronutrient
critically ill patients with AKI.8 Factors contributing to mal- needs are more difficult to define in the absence of surrogate
nutrition in critically ill individuals on RRT are multifacto- markers that correlate with clinical outcomes.
rial. The acute stress response of critical illness leads to a The goal of this review is to discuss an overview of
release of regulatory endocrine hormones (catecholamines, CRRT types, components of the CRRT prescription,
cortisol, and glucagon) and proinflammatory cytokines that and important considerations related to nutrition support
contribute to net gluconeogenesis and glycogenolysis.9-11
This ultimately results in net skeletal muscle breakdown and
lipolysis to provide necessary substrates for cellular function From the Department of Pharmacy, Mayo Clinic, Rochester,
and contributes to tissue insulin resistance and inhibition Minnesota, USA.
of lipoprotein lipase. So, although substrate plasma levels Financial disclosure: None declared.
are elevated, use by peripheral tissues may be reduced.11,12 Conflicts of interest: None declared.
Additional complicating factors affecting nutrition status in
This article originally appeared online on October 15, 2018.
patients with renal failure requiring RRT include decreased
Corresponding Author:
intake prior to and during admission, concomitant organ
Erin M. Nystrom, PharmD, BCNSP, Mayo Clinic, 200 First Street
failure, and nutrient losses from sources such as RRT, SW, Rochester, MN 55902.
drains, and wounds. Email: nystrom.erin@mayo.edu
Nystrom and Nei 755

Figure 1. Diagram of diffusion and convection removal of solutes in hemofilter. Differences in mechanism of solute removal
comparing diffusion and convection across the hemofilter membrane are represented. The hemofilter has multiple hollow-fiber
tubules running blood through the device to allow maximal contact with hemofilter membrane. In diffusion, dialysate runs
countercurrent to the blood flow using a concentration gradient to allow removal of small solutes. In convection, the
transmembrane pressure, higher in the blood compartment and lower in the effluent/ultrafiltrate compartment, drives fluid across
the membrane, dragging small and medium solutes with it.

provision. We challenge nutrition support clinicians to un- (Table S1). Contemporary practice uses high-flux mem-
derstand the specific CRRT modalities and types of CRRT branes, allowing better clearance of middle molecules. The
anticoagulation, dialysate, and/or replacement fluids em- term sieving coefficient in convection is the ratio of solute
ployed in their ICUs, as well as to determine how a patient’s in effluent to blood, and is the primary determinant of how
CRRT prescription may impact his or her nutrition support readily a solute is cleared. Sieving coefficients range from 0
regimen. to 1, with a value of 1 indicating free passage of a molecule
through the membrane.
Continuous VV hemodialysis (CVVHD) is most sim-
CRRT Overview ilar to IHD in that it uses diffusion to achieve solute
Multiple CRRT modalities are available, each designated removal, but in a continuous manner, with dialysate run
by location of vascular access and mechanism of solute countercurrent to blood flow. Continuous VV hemofiltra-
removal (Table 1). Vascular access is designated as either tion (CVVH) differs from CVVHD in its use of convec-
veno-venous (VV) or arterio-venous (AV), indicating the lo- tion for fluid and solute removal. Due to the need for
cation of circuit entry and exit from the patient’s circulation. a high ultrafiltration rate to achieve removal of solutes,
AV CRRT, which uses a patient’s arterial blood pressure to replacement fluids are required to replenish volume and
drive blood through the filter, has largely been abandoned electrolytes. Continuous VV hemodiafiltration (CVVHDF)
in favor of VV CRRT given the availability of CRRT uses both diffusion and convection for solute clearance and
pumps. thus requires use of both replacement and dialysate fluids.
The 2 mechanisms for solute removal in CRRT are No CRRT modality has been shown to be superior to
diffusion and convection (Figure 1). In diffusion, a dialysate another.
solution runs countercurrent to blood in the dialysis filter, Other modalities of RRT that are administered in a
creating a concentration gradient across the dialysis mem- prolonged (but not continuous) manner in comparison
brane and driving solute removal from the blood. Solute with typical intermittent dialysis sessions include sustained
removal is dependent on molecular weight, with smaller low-efficiency dialysis (SLED) and sustained continuous
molecules more readily removed, eg, creatinine, urea, and ultrafiltration (SCUF). SLED uses diffusion to achieve
electrolytes. Convection, also referred to as ultrafiltration, solute removal, but over a longer interval than IHD. SCUF
uses a high transmembrane pressure gradient in the hemofil- uses convection at a lower ultrafiltration rate than CVVH,
ter to force plasma water across a semipermeable membrane, primarily to achieve volume removal. Selection of modality
dragging solute with it (solvent drag). Convection is effec- is dependent on institution preference, fluid status, and
tive at removing both small-size and medium-size solutes metabolic/electrolyte abnormalities.
756 Nutrition in Clinical Practice 33(6)

Table 1. RRT Modalities.

Type Solute Clearance Use of RF Use of Dialysate Duration Anticoagulation

CVVH Convection Yes No 24 hours Yes


CVVHD Diffusion No Yes 24 hours Yes
CVVHDF Convection and Diffusion Yes Yes 24 hours Yes
SLED Diffusion No Yes 6–12 hours Usually not
SCUF Convection No No 6–12 hours Usually not

CVVH, continuous veno-venous hemofiltration; CVVHD, continuous veno-venous hemodialysis; CVVHDF, continuous veno-venous
hemodiafiltration; RF, replacement fluids; RRT, renal replacement therapy; SCUF, slow continuous ultrafiltration; SLED, sustained
low-efficiency dialysis.

CRRT Prescription and Implications r Anticoagulation: Citrate anticoagulation can be a


for Nutrition Support significant source of carbohydrate calories from cit-
rate and dextrose. Citrate solutions available for
Since the early 1970s when CRRT was first administered, CRRT anticoagulation include acid citrate dextrose,
significant advancements have been made in pump and filter formula-A ([ACD-A], 2.45% dextrose and 2.2% cit-
technology, replacement fluids, and anticoagulation. It is rate) and 4% trisodium citrate (TSC). Delivery of
vital to understand how CRRT has evolved to appropri- citrate and glucose is dependent on the rate of
ately evaluate available CRRT literature related to nutri- infusion.
tion support. Additionally, understanding the components r Replacement or dialysate fluids: The dextrose (glu-
that make up a CRRT prescription and relevant solutes cose) composition of these fluids, ranging from 0 to
removed or gained (glucose [dextrose], citrate, amino acids) 110 mg/dL, impacts overall net delivery of glucose.
is fundamental to applying literature to clinical practice. Fluids without dextrose may contribute to net losses
Recognizing advancements in CRRT technology in the last of glucose in the effluent, while fluids that contain
decade,13 specific variables for consideration include the dextrose will have minimal impact on net glucose gain
following: or loss in the effluent. Also, replacement fluids may
r Filter type: Contemporary high-flux, high-
be administered prefilter, postfilter, or both, which
could impact overall glucose delivery; higher prefilter
permeability filters allow larger molecules to glucose concentrations may lead to greater net loss of
pass through the membrane and also provide more glucose in the CRRT circuit.
efficient solute removal.
r CRRT dose: CRRT dose is defined as the effluent
flow rate. The effluent volume is comprised of the Select variables with recognized impact on nutrient dis-
total ultrafiltration (convection) and dialysate (dif- position will herein be reviewed.
fusion) volume. For example, in CVVH the CRRT
dose is defined only by the ultrafiltration rate, while in
Replacement and Dialysate Fluids
CVVHDF, it is a combination of ultrafiltration and Replacement fluids are used with CVVH and CVVHDF to
dialysate fluid rate. Guidelines generally recommend replenish electrolytes and plasma volume removed during
a delivered rate of 20–25 mL/kg/hr.7 The higher the convection. Replacement fluids are typically infused pre-
CRRT dose, the higher the solute removal. filter, postfilter, or both (Figure 2). Prefilter administration
r Blood flow rate: Typically 100–300 mL/min, change of replacement fluids helps minimize circuit clotting, but
in blood flow has a relatively small impact on solute may decrease efficiency of solute removal from the blood
clearance unless the effluent rate is increased pro- due to prefilter plasma dilution. Postfilter administration
portionately, as hemofiltration solution is otherwise allows direct delivery of desired replacement solute to
saturated. systemic circulation.14
r CRRT mode: Use of diffusion, convection, or both Dialysate solutions are used in CVVHD and CVVHDF
determines the size and quantity of solutes removed to achieve diffusive clearance of solute. Dialysate is gener-
as well as the need for replacement fluids (con- ally infused into the dialysis filter countercurrent to plasma
vection). Convective clearance allows removal of blood flow (Figures 1 and 2). Efficacy of solute removal,
larger solutes compared with diffusive clearance. The in addition to solute characteristics, is dependent on both
glucose composition of replacement fluids required the degree of concentration gradient across the membrane
to replace fluid removal with convection can signifi- and the dialysate rate, with an increased rate associated with
cantly affect energy gain or loss. higher solute clearance.
Nystrom and Nei 757

Figure 2. CRRT modalities. (A) CVVH, (B) CVVHD, and (C) CVVHDF. In all 3 modalities, anticoagulation is infused early in
the venous side of the CRRT circuit. Replacement fluids are administered in CVVH and CVVHDF and may be given prefilter,
postfilter, or both. Total replacement fluid rate is approximate to effluent removal rate (CRRT dose). In CVVHD and CVVHDF,
dialysate fluids enter the hemofilter and run countercurrent to blood flow. In all 3 modalities the effluent bag contains fluid and
solutes removed from the CRRT circuit. Glucose losses in the effluent are variable and depend on glucose content of
replacement/dialysate fluids in addition to components of the CRRT prescription. *There are multiple types of anticoagulation; 1
example is ACD-A, and others include TSC, heparin, or no anticoagulation. Typical ACD-A rate is 100–300 mL/h, but may be
altered depending on institutional-specific protocols and alterations to the CRRT prescription. ACD-A, acid citrate dextrose,
formula-A; CRRT, continuous renal replacement therapy; CVVH, continuous veno-venous hemofiltration; CVVHD, continuous
veno-venous hemodialysis; CVVHDF, continuous veno-venous hemodiafiltration; TSC, trisodium citrate.
758 Nutrition in Clinical Practice 33(6)

Figure 2. Continued.

Table 2. Electrolyte Composition of Dialysate and ids with specific electrolyte content. Bicarbonate-based
Replacement Fluids. replacement fluids and dialysate solutions are now pre-
ferred over lactate-based solutions due to improved correc-
Component Concentration
tion of acidosis, lack of lactate accumulation, and better
Sodium 136–140 mmol/L hemodynamic tolerance.15,16 Lactate is a calorie source,
Potassium 0–4 mmol/L and, as such, studies demonstrating high caloric gain
Calcium 0–3.5 mEq/L with lactate-based replacement fluids must be taken into
Magnesium 0.5–1.5 mEq/L context.
Phosphate 0–1.25 mmol/L Note that the dextrose component of replacement and
Chloride 107.5–120.5 mmol/L
Bicarbonate 25–35 mmol/L
dialysate solutions is variable, ranging from 0 to 110 mg/dL,
Lactate 0–38 mmol/L which may result in net glucose (energy) loss due to unre-
Dextrose 0–110 mg/dL placed effluent losses of glucose if glucose-free fluids are
used.

Selection of replacement fluids and dialysate solu-


Anticoagulation
tions is based on a patient’s specific metabolic, acid– Due to prolonged contact time between blood and the extra-
base, and electrolyte profile. A variety of commercially corporeal CRRT circuit, anticoagulation is recommended
available replacement fluids and dialysate solutions, with to prevent filter clotting and optimize permeability and dial-
varying electrolyte and buffer components, are available ysis efficiency. The type of anticoagulation used in CRRT
(Tables 2 and S2). Some institutions may compound flu- varies among institutions, with the most common options
Nystrom and Nei 759

being regional citrate anticoagulation (RCA), heparin, or carbon dioxide production, and increased infection risk.21-23
none.17 Current guidelines recommend RCA as first-line Additionally, the energy prescription should be based on
therapy for anticoagulation in the absence of contraindica- a dry weight, which may be challenging to determine
tions to citrate.7 RCA has been shown to prolong filter life, in the hypervolemic patient. Hyperglycemia, high insulin
decrease incidence of spontaneous filter failure, and reduce requirements, and hypertriglyceridemia may be indicators
bleeding compared with systemic heparin.18 Citrate achieves of intolerance to the dextrose and/or lipid emulsion load
local anticoagulation by chelating calcium within the circuit, and warrant reassessment of the nutrition prescription with
thus inactivating an essential component of the clotting an accounting of the possible contributions from CRRT
cascade. A postfilter infusion of calcium into the circuit is fluids.
necessary to regain a normal systemic calcium concentra-
tion. When blood from the CRRT circuit is infused back Carbohydrate Gains and Losses With CRRT
into the systemic circulation, citrate is rapidly metabolized
When determining the amount of calories to provide via
by the liver, muscle, and kidney, producing bicarbonate.
enteral nutrition (EN) or parenteral nutrition (PN), clini-
Citrate is typically infused at a fixed rate with monitoring
cians should routinely assess for dextrose-containing fluids
of systemic total and ionized calcium levels for evidence of
administered with intravenous (IV) medications or as bolus
citrate toxicity, suspected when the total calcium to ionized
or continuous crystalloid infusions as well as the potential
calcium ratio is >2.5,19 or titrated to a low intracircuit
for energy gain via fluids used in the CRRT circuit. Carbo-
ionized calcium goal. Possible side effects of RCA include
hydrate sources in the CRRT circuit include:
metabolic alkalosis and hypocalcemia. Patients with liver
failure or profound shock with decreased muscle perfusion r Citrate (3 kcal/g24,25 ) from RCA of ACD-A (2.2%
are at highest risk of citrate accumulation and subsequent citrate) and TSC (4% trisodium citrate);
side effects.19 r Glucose (3.4 kcal/g) from ACD-A (2.45% dextrose)
Products available for RCA include ACD-A, which is and replacement and dialysate fluids (0–110 mg/dL);
2.2% citrate formulated in a 2.45% dextrose solution, and r Lactate (3.62 kcal/g26 ), of lower importance in con-
4% TSC. Given that citrate and dextrose are sources of temporary practice in replacement and dialysate
carbohydrate, energy contributed with use of RCA can be fluids.
clinically significant.
If citrate is not an option for anticoagulation, heparin Table 3 summarizes studies evaluating delivery of citrate,
anticoagulation is recommended in patients who are not lactate, and glucose associated with CRRT.27-29 Notice that
at high risk for bleeding. In the setting of heparin-induced energy gain can be substantial, depending on the type and
thrombocytopenia, alternative agents such as direct throm- rate of fluids used, with 1 study reporting up to 1300 kcal/d
bin inhibitors or fondaparinux may be considered after with high-lactate replacement fluids and anticoagulation
weighing risks and benefits of therapy.7 If both citrate and with ACD-A.27 Variability in energy gain was noted in the
heparin are contraindicated, no anticoagulation may be 2 studies by Balik et al, depending on the lactate content of
used with close monitoring for circuit and filter thrombosis. replacement fluids and type of anticoagulation.27,28 Note-
worthy is the specific impact of replacement fluids, with
or without glucose, and citrate anticoagulation, on calorie
Energy Requirements gain.
Defining energy requirements in critical illness has been the The most recent study published on this topic confirmed
subject of much debate, lacking the input of high-quality the potential for significant caloric contribution, specifically
study data. Similarly, no randomized controlled studies have with CVVH. New et al measured energy uptake with
defined optimal energy provision in CRRT patients, for use of guideline-recommended citrate anticoagulation as
whom energy requirements have not been distinguished to ACD-A and bicarbonate-based replacement fluids (glucose
be different than patients without AKI. In a secondary 110 mg/dL). Of the 10 patients studied, 8 received ACD-
analysis of 1456 patients with AKI requiring CRRT, neither A at a rate of 300 mL/h (1 patient received ACD-A
higher daily caloric intake of at least 25 kcal/kg/d nor calorie at 200 mL/h and 1 patient received both rates during the
intake itself was associated with differences in RRT-free, study time period), with an average ultrafiltration rate of
ICU-free, ventilation-free, or hospital-free days.20 30 mL/kg/h and blood flow rate of 200 mL/min. The study
Thus, we turn our attention to the avoidance of over reported an average daily delivery of 513 kcal/d (citrate
feeding, as CRRT fluids can be a significant source of 218 kcal/d, glucose 295 kcal/d).30 Investigators were unable
calories. Because metabolic alterations of critical illness and to determine differences in energy uptake comparing differ-
AKI complicate the body’s ability to use non protein sources ent rates of ACD-A due to low numbers in the 200 mL/h
of calories, it is especially important to avoid overfeeding, group. Additionally, the impact of glucose-free replacement
which can result in accumulation of liver fat, increased fluids on energy gain was not assessed. These factors limit
760 Nutrition in Clinical Practice 33(6)

Table 3. Carbohydrate Studies in CRRT.

Study CRRT Type CRRT Fluids Key Study Conclusions

Balik et al, CVVH ACD-A/lactate fluidsa r Average calorie gain 1318 kcal/d.
201227 (n = 18) r Net loss noted with bicarbonate fluidsa +
CVVHDF heparin (control).
(n = 23) r No difference in citrate, lactate, or glucose
delivery between CVVH and CVVHDF.
r Significant caloric gain noted; however, high-
lactate fluids are no longer recommended by
guidelines, and heparin was used for
anticoagulation in the control group, limiting
clinical applicability.
Balik et al, CVVHDF ACD-A/lactate fluidsa (n = 29) r Calorie gain (per day):
201328 (n = 81) vs TSC/bicarbonate fluidsa ◦ ACD-A/lactate fluids 1078 kcal
(n = 34) vs heparin/lactate ◦ TSC/bicarbonate fluids 115 kcal
fluidsa (n = 18) ◦ Heparin/lactate fluids 361 kcal
◦ Calculated ACD-A/bicarbonate fluids
457 kcal/d
r Kcal delivery in heparin group was solely
related to lactate fluids.
r Different blood flows used in each group
affected citrate rate.
r Net caloric gain noted in all groups, but
delivery was impacted by type of
anticoagulation and fluid used; ACD-A and
lactate-based fluids provided highest gain.
r Use of lactate fluids limits clinical application
in current practice.
Stevenson et al, CVVH Bicarbonate dialysate fluids r In vitro study comparing glucose kinetics
201329 CVVHD with and without glucose among CRRT types.
CVVHDF r Calculations in CVVH and CVVHDF
assumed replacement fluids without glucose.
r Net glucose losses noted with all CRRT
modalities when bicarbonate fluids were used
without glucose.
r Glucose loss decreased with inclusion of
glucose 100 mg/dL in dialysate fluids.
r ACD-A with CVVHD + bicarbonate dialysate
without glucose ˗120 to +470 kcal/d.
r Citrate delivery was not assessed (only
glucose).
r Glucose losses noted with non-glucose-
containing dialysate and replacement fluids;
glucose losses decreased with addition of
glucose to dialysate fluids and with citrate
anticoagulation.
New et al, CVVH ACD-A/bicarbonate r Average 513 kcal/d
201730 (n = 10) replacement fluidsa r Contemporary CVVH regimen using
guideline-recommended fluids.
r Notable caloric delivery from ACD-A
anticoagulation and bicarbonate replacement
fluids.a

ACD-A, acid citrate dextrose, formula-A; CRRT, continuous renal replacement therapy; CVVH, continuous veno-venous hemofiltration;
CVVHD, continuous veno-venous hemodialysis; CVVHDF, continuous veno-venous hemodiafiltration; TSC, trisodium citrate.
a Glucose-containing fluids (100–110 mg/dL). The term “fluids” in this table designates either replacement or dialysate fluids or both depending on

the type of CRRT studied.


Nystrom and Nei 761

extrapolation to clinical practice for CVVH when lower the CRRT circuit. Lastly, the method of citrate titration is
ACD-A rates and/or glucose-free replacement fluids are variable among institutions, possibly leading to differences
employed. in citrate rate, which will also impact overall delivery of
It is important to highlight how the glucose content of citrate and glucose.
replacement and dialysate solutions may impact net delivery
of carbohydrates with CRRT. Non-glucose-containing flu- Case Example
ids have the potential to contribute to glucose losses be-
Patient A.B., a 70-year-old female weighing 66 kg, is ad-
cause effluent losses of glucose are left unreplaced with
mitted with septic shock secondary to bowel perforation,
use of glucose-free replacement fluids. Stevenson et al
complicated by AKI requiring CRRT. She is 7 days into
confirmed this with in vitro models of CVVH, CVVHD,
her ICU stay, and due to strict nothing by mouth status,
and CVVHDF.29 The authors suggested an equation based
she is being considered for PN. CVVH has been prescribed
on logistic regression that could be used to calculate glucose
with an ultrafiltration rate of 30 mL/kg/h (2L/h), blood
losses for any method of CRRT with any glucose concentra-
flow rate 200 mL/min, and ACD-A anticoagulation at
tion in dialysate, assuming use of non-dextrose-containing
300 mL/h. Replacement fluids are high-bicarbonate with
replacement fluids in CVVH or CVVHDF:
glucose concentration of 110 mg/dL. Based on recent study
data (New et al), we could expect ࣈ500 carbohydrate kcal/d
y = 0.81 [QD × EC × (GPre − GD ) + QUf × EC × GPre ]
from her current convection-based CVVH regimen.30
+ 4.16 A.B. later improves clinically, and her replacement fluid
is switched to a lower bicarbonate solution that does not
y = total kcal/d removed; QD = dialysis flow rate (L/hr); include glucose. As such, effluent losses of glucoses are no
EC = extraction coefficient; GPre = glucose concentration longer being replaced via glucose-containing replacement
prefilter (mg/dL); GD = glucose concentration in dialysate fluids, so we subtract estimated effluent glucose losses from
(mg/dL); QUf = ultrafiltration rate (L/hr) the 500 kcal estimated above, using the following method.
With an ultrafiltration rate of 2 L/h, we would expect
Several considerations should be made if using this
about 48 L removed over 24 hours. Effluent loss of 48 L with
equation in clinical practice. Noting glucose EC = Ef /Pre
an assumed prefilter glucose concentration of 100 mg/dL or
(Ef = concentration in effluent leaving the dialysis filter,
1g/L totals 48 g of estimated glucose losses or ࣈ160 kcal,
Pre = concentration in the prefilter blood), this value (EC)
rounded to 150 kcal; 500 kcal (as previously demonstrated)
could be considered 1 for practical application given glucose
subtract 150 kcal = a rough estimate of ࣈ350 kcal/d net gain
is freely filtered in both dialysis and ultrafiltration with
with this CVVH program.
contemporary filters. Indeed, the value calculated in the
Alternatively, if A.B. had been receiving CVVHDF with
study was 1.04 (95% confidence interval [CI]: 1.03–1.05).
citrate anticoagulation (ACD-A at 300 mL/h), an ultrafiltra-
Additionally, it is not routine practice to sample immediate
tion rate of 1 L/h, and dialysate rate of 1L/h with replace-
prefilter glucose concentrations, which will likely be altered
ment/dialysate fluids that did not contain dextrose, we could
compared with systemic glucose values if replacement fluids
estimate losses according to the Stevenson equation, where
are administered prefilter and if citrate anticoagulation
EC is estimated to be 1, GPre is 100 mg/dL (highly variable),
is administered. Third, this in vitro study used a fixed
and y is the number of kcal removed per day:
blood flow rate of 200 mL/min. Finally, the equation does
not account for use of replacement fluids (in CVVH or
y = 0.81 [QD ×EC × (GPre − GD ) + QUf × EC × GPre ]
CVVHDF) that contain dextrose. Acknowledging these
limitations and in the absence of more specific data to guide + 4.16
practice, this equation can provide a starting estimate for
bedside clinicians to estimate glucose losses in effluent. 166 kcal/d = 0.81[1 L/h × 1 × (100 mg/dL − 0)
Figure 3 illustrates a simple algorithm for estimating how
anticoagulation and replacement/dialysate solutions may +1 L/h × 1 × 100 mg/dL] + 4.16
impact caloric delivery from carbohydrates in the CRRT
circuit, highlighting the prominent variables of citrate- Since we need to account for citrate anticoagulation as a
based anticoagulation and glucose content of replacement source for calories (not accounted for with the Stevenson
fluids. Also for consideration is the CRRT dose (effluent equation), we could extrapolate data from CVVH (New
flow rate); the higher the dose, the higher the removal of et al) with total initial calories of 500 kcal and subtract
glucose and citrate. Additionally, replacement fluids may 166 kcal (net loss calculated above) to total net gain of
be administered prefilter, postfilter, or both, which could 334 kcal/d. This caloric gain is fairly similar to the estima-
impact overall glucose delivery. Higher prefilter glucose tion above using only ultrafiltration (CVVH), but with a
concentrations may lead to greater net loss of glucose in similar total CRRT dose (ultrafiltration 1 L/h + dialysis flow
762 Nutrition in Clinical Practice 33(6)

Figure 3. Algorithm for energy gain and/or loss assessment. Flow chart depicts CRRT variables that can contribute to energy
gain or loss. *CRRT modalities of CVVHDF and CVVHD use diffusion and, thus, dialysate (not shown). Dialysate that is
dextrose free theoretically could result in calorie loss. ACD-A, acid citrate dextrose, formula-A; CRRT, continuous renal
replacement therapy; CVVH, continuous veno-venous hemofiltration; CVVHD, continuous veno-venous hemodialysis;
CVVHDF, continuous veno-venous hemodiafiltration; TSC, trisodium citrate.

rate 1 L/h = 2 L/h). It is important to note that this is an CRRT circuit and individual prescription, beyond the fact
estimation, given lack of in vivo data specific to CVVHDF that amino acids, with a sieving coefficient of around 1, are
with citrate anticoagulation. readily filtered from the blood into effluent.33,35,40
In conclusion, contemporary CRRT practices are likely CRRT-specific factors that determine amino acid clear-
to contribute a net gain of calories from carbohydrates when ance, like that of other solutes, include use of convec-
anticoagulation of ACD-A is used, noting this contribution tion (CVVH or CVVHDF) and/or diffusion (CVVHD or
may vary depending on the citrate rate. This could also CVVHDF), blood flow rate, dialysis flow rate, effluent rate,
be the case with other citrate-based anticoagulation, ie, and filter membrane properties.38,40 As such, these variables
TSC. This energy gain is buffered with use of glucose- are essential to consider when translating research into
free replacement fluids, as glucose losses in effluent are not recommendations for protein dosing in CRRT patients in
replaced. Notably, if citrate-based anticoagulation is not contemporary clinical practice. While core studies refer-
used, an overall net energy loss may occur if glucose-free enced in guidelines41-43 have been limited by lack of ran-
replacement or dialysate fluids are employed. While we have domization and power and surrogate outcomes assessed (eg,
some literature to help guide estimation of energy gain nitrogen balance), extrapolation is further constrained by
or loss with CRRT, it remains challenging to describe an the evolution of CRRT technology since their publication.
approach to incorporate all CRRT prescriptions given the Notably, newer CRRT technology allows higher blood flow
wide variability in practice. It is therefore critical to consider and effluent removal rates that easily double those studied
individual institutional CRRT practices when determining by Bellomo36,37 and Sheinkestel.38,44 Additionally 3 of these
application of current study findings to clinical practice. 4 studies evaluated CRRT modalities that used diffusion
(continuous arteriovenous hemodiafiltration [CAVHD] or
CVVHD), leaving only 7 adult patients studied on a CRRT
Protein (Amino Acids) modality that employed convective clearance (CVVHDF).37
The catabolism that characterizes critical illness is com- Maxvold and colleagues, in a study of 6 pediatric patients
pounded in AKI by multiple factors, chief among them randomized to either CVVH or CVVHD and crossed over
amino acid losses in ultrafiltrate and/or dialysate with to the alternate modality after 24 hours, demonstrated
CRRT.31-39 Quantifying absolute amino acid loss is com- a 30%–40% higher amino acid clearance with convection
plicated by the multitude of variables that make up the (CVVH) than with diffusion (CVVHD).35
Nystrom and Nei 763

To our knowledge, the most recent study evaluating concentrated, minimum-sodium PN formula to allow fluids
amino acid disposition in CRRT is that by Zappitelli et al, to be managed primarily outside of PN. Significant sources
who studied 15 critically ill children receiving CVVHD of sodium in the ICU include crystalloid administered with
and found a 10% loss of total amino acid intake and, medications and for resuscitation and as needed well beyond
more specifically, a 17%–22% loss when amino acids were the daily required sodium intake, which can exacerbate
provided by PN.39 This suggests the route of nutrition hypervolemic states. Notably, dialysate and replacement
may have implications for amino acid loss in dialysate. A solutions contain sodium of 130–140 mEq/L. Hypona-
relationship between blood levels of amino acids, rather tremia that accompanies AKI is predominantly due to a
than amino acid intake, and effluent losses has been demon- state of hypervolemia rather than a sodium deficit in the
strated by others, suggesting nutrition parameters, such as absence of conditions of sodium loss, as with significant
rate of parenteral amino acid infusion, may be a factor in gastrointestinal output. While significant fistula, ostomy, or
losses with CRRT.33,34,38 The clinical implications of this are wound losses of sodium warrant sodium replacement, this
unclear. is to be considered separately from the hyponatremia due
Research questions on protein provision in critical ill- to hypervolemia that is common in AKI. It is, therefore,
ness along with outcomes for evaluation have recently essential that volume status be assessed carefully whenever
been proposed.45 Additional questions related to protein there is hyponatremia. In hypervolemia, total sodium body-
provision in patients receiving CRRT are in dire need of sodium stores may be increased along with volume (edema),
investigation. Studies measuring amino acid loss in effluent thus requiring restriction of both fluid (assuming the ab-
with contemporary CRRT modalities would be instructive sence of a fluid-responsive shock state) and sodium intake.23
in the absence of clinical outcomes, although studies of
effluent losses should not be considered equal to those that Electrolytes and Minerals
evaluate the effect of protein provision on clinical outcomes
Defining electrolyte needs is comparatively more straight-
in patients receiving CRRT. Specific queries evaluating
forward than determining energy and protein needs. Crit-
variables of convection (CVVH), diffusion (CVVHD), or
ically ill patients on CRRT are closely monitored with
combination convection/diffusion (CVVHDF), amino acid
frequent laboratory tests to guide the CRRT prescrip-
intake, rate of parenteral administration of amino acid, and
tion, including dialysate and replacement-fluid electrolyte
route of amino acid administration are warranted.
composition; protocol-based or provider-specific orders for
In the meantime, the published research and our clinical
potassium, magnesium, and phosphate supplementation;
experience is reassuring in that a high amino acid dose
and separate calcium infusion to replace calcium bound
(ie, 2.0–2.5 g/kg) as recommended41,42 can be adminis-
to and lost with citrate when RCA is employed. Nutrition
tered successfully without increasing blood urea nitrogen
support plays mostly a supporting role with regard to the
levels or generating disproportionate amino acid loss (ie,
management of electrolytes, acknowledging the partnership
waste).33,34,36,37 Given the prevalent use of convection-
with primary ICU and nephrology teams when modifica-
based modalities of CVVH and CVVHDF, prescription
tions to PN are needed.
of higher blood flow and effluent doses, and advances in
hemofilter technology since the publication of the above-
referenced studies, it is reasonable to suspect amino acid
Hypophosphatemia
losses in CRRT are being underestimated. Generous protein Hypophosphatemia is an important complication of CRRT
provision of 2.5 g/kg with PN or via addition of protein and may have a negative impact on patient outcomes.46
modules to high-protein tube feeding is warranted. CRRT continuously clears phosphate from the blood, while
replacement and dialysate fluids have historically excluded
Fat phosphate. Close monitoring of phosphate levels is essen-
tial for the duration of CRRT. A variety of approaches
Alterations to IV lipid emulsion for CRRT patients are not
to prevent and treat hypophosphatemia that occurs with
recommended beyond that which would be in response to
CRRT are employed, including the more recent use of
hypertriglyceridemia, a common complication in the ICU,
commercial phosphate-containing CRRT solutions, Phox-
or for administration of non nutrition lipid emulsion with
illum (Gambro Renal Products, Daytona Beach, FL) and
clevidipine (2 kcal/mL) or propofol (1.1 kcal/mL).
multiPlus (Fresenius Kabi, Bad Homberg, Germany), which
can reduce the need for supplemental phosphate. When
Volume and Sodium Considerations phosphate-free fluids are used, additional supplementation
CRRT is a highly efficient means of fluid removal. Ac- is necessary, administered either by mouth or per tube
cordingly, it is not necessary to provide a concentrated or via the IV route. Sodium phosphate may be infused
tube-feeding formula, which may limit optimal protein intermittently, continuously, or added to the PN. Selection
provision. It is, however, our practice in the ICU to provide a of route of phosphate supplementation should consider the
764 Nutrition in Clinical Practice 33(6)

risk of diarrhea with enteral administration and availability in patients on prolonged courses of CRRT is warranted.
of parenteral products, which have recently been subject The reader is referred to the excellent reviews cited in this
to national shortages. Low-phosphate, renal-formula tube section for further information on vitamin and trace element
feedings are not recommended for patients receiving CRRT. disposition in CRRT.

Calcium Conclusion
RCA works locally within the CRRT circuit to inactivate The patient receiving CRRT presents a unique nutrition
the coagulation cascade and platelets by chelating calcium. challenge for clinicians in the ICU. Current evidence and
Since a portion of the citrate–calcium complex remains after practical experience suggests CRRT can be a significant
extracorporeal elimination, calcium must be infused on the source of calories, in particular with the use of citrate-based
venous (return) side of the circuit prior to reentering the anticoagulation. Clinicians should observe patients for signs
patient’s circulation to achieve a normal systemic calcium of overfeeding, such as hyperglycemia, high insulin re-
concentration.47 The amount of parenteral calcium neces- quirements, and hypertriglyceridemia. Glucose losses from
sary to do this is exponentially higher than standard doses blood into effluent are buffered by the use of dextrose-
that would be provided with PN. As such, a stable and containing replacement fluids (CVVH and CVVHDF) and
unchanged amount of calcium may be provided in the PN, dialysate fluids (CVVHD and CVVHDF), but the clinician
if any. should be aware that these losses can be significant with
the use of newer, dextrose-free dialysate and replacement
Potassium and Magnesium fluids. Amino acid losses are significant with CRRT, and
Potassium and magnesium are provided in replacement and restriction of amino acids should be avoided in an attempt
dialysate fluids. Additional potassium and magnesium may to limit the need for RRT. Special considerations exist for
be prescribed as part of protocol orders or as needed. Rec- volume and electrolytes, the crux of which is managed
ognizing wide variation among institutions, it is our practice within the CRRT circuit and established institution-specific
to zero the electrolytes in PN for patients receiving CRRT protocols. We urge working collaboratively with nephrology
and incrementally add back potassium or magnesium based on fluid and electrolyte provision in the nutrition program,
on recurrent and regular need for potassium or magnesium recognizing the ability to tailor PN more readily than EN.
outside of the RRT circuit, in close communication with Transitions off CRRT should be anticipated with improved
our nephrology colleagues. As with phosphorus, the risk of hemodynamics, as discontinuing CRRT may warrant ad-
diarrhea with enteral administration of potassium should justments to calorie, protein, and electrolyte provision. We
be weighed. Because enteral magnesium is poorly absorbed recommend that nutrition support clinicians practicing in
and is dose-limited by diarrhea, IV replacement of magne- the ICU work to understand the specific CRRT modalities
sium sulfate to treat hypomagnesemia is favored. used in their practice, the specific components of the CRRT
In addition to understanding the CRRT modality used, prescription, and patient-specific variables that may affect
convection-based, diffusion-based, or both, nutrition sup- macronutrient and micronutrient disposition.
port clinicians should be familiar with the electrolyte con-
centrations in replacement and dialysate fluids routinely Statement of Authorship
prescribed at their institutions. See Table 2 for ranges E. M. Nystrom and A. M. Nei equally contributed to the
of electrolyte concentrations used in available products. conception and design of the manuscript; E. M. Nystrom and
The reader is also referred to product-specific labeling for A. M. Nei equally contributed to the acquisition and analysis
electrolyte content of commercial replacement and dialysate of the data; E. M. Nystrom and A. M. Nei equally contributed
to the interpretation of the data; and E. M. Nystrom and A.
fluids.
M. Nei drafted the manuscript. Both authors critically revised
the manuscript, agree to be fully accountable for ensuring the
Vitamins and Trace Elements
integrity and accuracy of the work, and read and approved the
Optimal dosing of vitamins and trace minerals in critical final manuscript.
illness is unknown.48 RRT adds a level of complexity,
potentially increasing the risk for deficiencies via filter Supplementary Information
adsorption or effluent losses, and additional needs beyond Additional supporting information may be found online in the
amounts administered with nutrition programs are gener- Supporting Information section at the end of the article.
ally not well understood.49,50 A review of the literature
suggests losses of selenium with CRRT to a degree that References
may warrant additional selenium beyond that provided 1. Hoste EA, Clermont G, Kersten A, et al. RIFLE criteria for acute
by standard multiple trace element products in PN.39,51-54 kidney injury are associated with hospital mortality in critically ill
Recognizing blood assay limitations, additional monitoring patients: a cohort analysis. Crit Care. 2006;10(3):R73.
Nystrom and Nei 765

2. Herrera-Gutierrez ME, Seller-Perez G, Sanchez-Izquierdo-Riera JA, 21. Wolfe RR. The 2017 Sir David P Cuthbertson lecture. Amino acids and
Maynar-Moliner J. Prevalence of acute kidney injury in intensive muscle protein metabolism in critical care. Clin Nutr. 2018;37(4):1093-
care units: the “COrte de prevalencia de disFuncion RenAl y DEpu- 1100.
racion en criticos” point-prevalence multicenter study. J Crit Care. 22. Butler SO, Btaiche IF, Alaniz C. Relationship between hyperglycemia
2013;28(5):687-694. and infection in critically ill patients. Pharmacotherapy. 2005;25(7):963-
3. Koeze J, Keus F, Dieperink W, van der Horst IC, Zijlstra JG, van 976.
Meurs M. Incidence, timing and outcome of AKI in critically ill 23. Mundi MS, Nystrom EM, Hurley DL, McMahon MM. Management
patients varies with the definition used and the addition of urine output of parenteral nutrition in hospitalized adult patients. JPEN J Parenter
criteria. BMC Nephrol. 2017;18(1):70. Enteral Nutr. 2017;41(4):535-549.
4. Iwagami M, Yasunaga H, Noiri E, et al. Current state of continuous 24. Oudemans-van Straaten HM, Kellum JA, Bellomo R. Clinical review:
renal replacement therapy for acute kidney injury in Japanese intensive anticoagulation for continuous renal replacement therapy—heparin or
care units in 2011: analysis of a national administrative database. citrate? Crit Care. 2011;15(1):202.
Nephrol Dial Transplant. 2015;30(6):988-995. 25. Lanckohr C, Hahnenkamp K, Boschin M. Continuous renal replace-
5. Nash K, Hafeez A, Hou S. Hospital-acquired renal insufficiency. Am ment therapy with regional citrate anticoagulation: do we really know
J Kidney Dis. 2002;39(5):930-936. the details? Curr Opin Anaesthesiol. 2013;26(4):428-437.
6. Park S, Lee S, Jo HA, et al. Epidemiology of continuous renal replace- 26. Marino PL. The ICU Book. Baltimore, MD: Williams & Wilkins; 2014.
ment therapy in Korea: results from the National Health Insurance 27. Balik M, Zakharchenko M, Otahal M, et al. Quantification of sys-
Service claims database from 2005 to 2016. Kidney Res Clin Pract. temic delivery of substrates for intermediate metabolism during citrate
2018;37(2):119-129. anticoagulation of continuous renal replacement therapy. Blood Purif.
7. Kidney Disease: Improving Global Outcomes (KDIGO) Acute Kidney 2012;33(1-3):80-87.
Injury Work Group. KDIGO clinical practice guideline for acute 28. Balik M, Zakharchenko M, Leden P, et al. Bioenergetic gain of
kidney injury. Kidney Int. 2012;2(suppl):1-138. citrate anticoagulated continuous hemodiafiltration—a comparison
8. Fiaccadori E, Lombardi M, Leonardi S, Rotelli CF, Tortorella G, between 2 citrate modalities and unfractionated heparin. J Crit Care.
Borghetti A. Prevalence and clinical outcome associated with preex- 2013;28(1):87-95.
isting malnutrition in acute renal failure: a prospective cohort study. 29. Stevenson JM, Heung M, Vilay AM, Eyler RF, Patel C, Mueller BA.
J Am Soc Nephrol. 1999;10(3):581-593. In vitro glucose kinetics during continuous renal replacement therapy:
9. Weissman C. Nutrition in the intensive care unit. Crit Care. implications for caloric balance in critically ill patients. Int J Artif
1999;3(5):R67-R75. Organs. 2013;36(12):861-868.
10. Ilias I, Vassiliadi DA, Theodorakopoulou M, et al. Adipose tissue 30. New AM, Nystrom EM, Frazee E, Dillon JJ, Kashani KB, Miles JM.
lipolysis and circulating lipids in acute and subacute critical illness: Continuous renal replacement therapy: a potential source of calories in
effects of shock and treatment. J Crit Care. 2014;29(6):1130.e1135- the critically ill. Am J Clin Nutr. 2017;105(6):1559-1563.
e1139. 31. Fiaccadori E, Regolisti G, Maggiore U. Specialized nutritional support
11. Carlson GL. Insulin resistance and glucose-induced thermogenesis in interventions in critically ill patients on renal replacement therapy. Curr
critical illness. Proc Nutr Soc. 2001;60(3):381-388. Opin Clin Nutr Metab Care. 2013;16(2):217-224.
12. Patkova A, Joskova V, Havel E, et al. Energy, protein, carbohydrate, 32. Davenport A, Roberts NB. Amino acid losses during continuous
and lipid intakes and their effects on morbidity and mortality in high-flux hemofiltration in the critically ill patient. Crit Care Med.
critically ill adult patients: a systematic review. Adv Nutr. 2017;8(4):624- 1989;17(10):1010-1014.
634. 33. Davies SP, Reaveley DA, Brown EA, Kox WJ. Amino acid clearances
13. Ronco C. Continuous renal replacement therapy: forty-year anniver- and daily losses in patients with acute renal failure treated by con-
sary. Int J Artif Organs. 2017:40(6):257-264. tinuous arteriovenous hemodialysis. Crit Care Med. 1991;19(12):1510-
14. Murugan R, Hoste E, Mehta RL, et al. Precision fluid management 1515.
in continuous renal replacement therapy. Blood Purif. 2016;42(3):266- 34. Frankenfield DC, Badellino MM, Reynolds HN, Wiles CE 3rd,
278. Siegel JH, Goodarzi S. Amino acid loss and plasma concentration
15. Barenbrock M, Hausberg M, Matzkies F, de la Motte S, Schaefer RM. during continuous hemodiafiltration. JPEN J Parenter Enteral Nutr.
Effects of bicarbonate- and lactate-buffered replacement fluids on car- 1993;17(6):551-561.
diovascular outcome in CVVH patients. Kidney Int. 2000;58(4):1751- 35. Maxvold NJ, Smoyer WE, Custer JR, Bunchman TE. Amino acid
1757. loss and nitrogen balance in critically ill children with acute renal
16. Tian JH, Ma B, Yang K, Liu Y, Tan J, Liu TX. Bicarbonate- versus failure: a prospective comparison between classic hemofiltration and
lactate-buffered solutions for acute continuous haemodiafiltration or hemofiltration with dialysis. Crit Care Med. 2000;28(4):1161-1165.
haemofiltration. Cochrane Database Syst Rev. 2015(3):CD006819. 36. Bellomo R, Seacombe J, Daskalakis M, et al. A prospective com-
17. Uchino S, Bellomo R, Morimatsu H, et al. Continuous renal replace- parative study of moderate versus high protein intake for criti-
ment therapy: a worldwide practice survey. The beginning and ending cally ill patients with acute renal failure. Renal Failure. 1997;19(1):
supportive therapy for the kidney (B.E.S.T. kidney) investigators. 111-120.
Intensive Care Med. 2007;33(9):1563-1570. 37. Bellomo R, Tan HK, Bhonagiri S, et al. High protein intake during
18. Bai M, Zhou M, He L, et al. Citrate versus heparin anticoagulation continuous hemodiafiltration: impact on amino acids and nitrogen
for continuous renal replacement therapy: an updated meta-analysis of balance. Int J Artif Organs. 2002;25(4):261-268.
RCTs. Intensive Care Med. 2015;41(12):2098-2110. 38. Scheinkestel CD, Adams F, Mahony L, et al. Impact of increasing
19. Meier-Kriesche HU, Gitomer J, Finkel K, DuBose T. Increased total to parenteral protein loads on amino acid levels and balance in critically
ionized calcium ratio during continuous venovenous hemodialysis with ill anuric patients on continuous renal replacement therapy. Nutrition.
regional citrate anticoagulation. Crit Care Med. 2001;29(4):748-752. 2003;19(9):733-740.
20. Bellomo R, Cass A, Cole L, et al. Calorie intake and patient outcomes 39. Zappitelli M, Juarez M, Castillo L, Coss-Bu J, Goldstein SL. Con-
in severe acute kidney injury: findings from the Randomized Evaluation tinuous renal replacement therapy amino acid, trace metal and folate
of Normal vs. Augmented Level of Replacement Therapy (RENAL) clearance in critically ill children. Intensive Care Med. 2009;35(4):698-
study trial. Crit Care. 2014;18(2):R45. 706.
766 Nutrition in Clinical Practice 33(6)

40. Btaiche IF, Mohammad RA, Alaniz C, Mueller BA. Amino acid 47. Morabito S, Pistolesi V, Tritapepe L, Fiaccadori E. Regional citrate
requirements in critically ill patients with acute kidney injury anticoagulation for RRTs in critically ill patients with AKI. Clin J Am
treated with continuous renal replacement therapy. Pharmacotherapy. Soc Nephrol. 2014;9(12):2173-2188.
2008;28(5):600-613. 48. Vanek VW, Borum P, Buchman A, et al. A.S.P.E.N. position paper:
41. Brown RO, Compher C. A.S.P.E.N. clinical guidelines: nutrition sup- recommendations for changes in commercially available parenteral
port in adult acute and chronic renal failure. JPEN J Parenter Enteral multivitamin and multi-trace element products. Nutr Clin Pract.
Nutr. 2010;34(4):366-377. 2012;27(4):440-491.
42. McClave SA, Taylor BE, Martindale RG, et al. Guidelines for the 49. Oh WC, Gardner DS, Devonald MA. Micronutrient and amino acid
provision and assessment of nutrition support therapy in the adult losses in acute renal replacement therapy. Curr Opin Clin Nutr Metab
critically ill patient: Society of Critical Care Medicine (SCCM) and Care. 2015;18(6):593-598.
American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.). 50. Tucker BM, Safadi S, Friedman AN. Is routine multivitamin sup-
JPEN J Parenter Enteral Nutr. 2016;40(2):159-211. plementation necessary in US chronic adult hemodialysis patients? A
43. Cano NJ, Aparicio M, Brunori G, et al. ESPEN guidelines on par- systematic review. J Renal Nutr. 2015;25(3):257-264.
enteral nutrition: adult renal failure. Clin Nutr. 2009;28(4):401-414. 51. Berger MM, Shenkin A, Revelly JP, et al. Copper, selenium, zinc, and
44. Scheinkestel CD, Kar L, Marshall K, et al. Prospective randomized thiamine balances during continuous venovenous hemodiafiltration in
trial to assess caloric and protein needs of critically Ill, anuric, critically ill patients. Am J Clin Nutr. 2004;80(2):410-416.
ventilated patients requiring continuous renal replacement therapy. 52. Ben-Hamouda N, Charriere M, Voirol P, Berger MM. Massive copper
Nutrition. 2003;19(11-12):909-916. and selenium losses cause life-threatening deficiencies during prolonged
45. Hurt RT, McClave SA, Martindale RG, et al. Summary points and continuous renal replacement. Nutrition. 2017;34:71-75.
consensus recommendations from the International Protein Summit. 53. Gervasio JM, Garmon WP, Holowatyj M. Nutrition support in acute
Nutr Clin Pract. 2017;32(1 suppl):142S-151S. kidney injury. Nutr Clin Pract. 2011;26(4):374-381.
46. Heung M, Mueller BA. Prevention of hypophosphatemia during 54. Wooley JA, Btaiche IF, Good KL. Metabolic and nutritional aspects
continuous renal replacement therapy—an overlooked problem. Semin of actue renal failure in critically ill patients requiring continuous renal
Dial. 2018;31(3):213-218. replacement therapy. Nutr Clin Pract. 2005;20(2):176-191.
Invited Review

Nutrition in Clinical Practice


Volume 33 Number 6
Use of Intradialytic Parenteral Nutrition in Patients December 2018 767–771

C 2018 American Society for

Undergoing Hemodialysis Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10190
wileyonlinelibrary.com

Menaka Sarav, MD1 ; and Allon N. Friedman, MD2

Abstract
Intradialytic parenteral nutrition (IDPN) is a form of supplemental nutrition used to treat patients with malnutrition who receive
hemodialysis. Once the diagnosis of malnutrition is made in such patients, encouragement of oral intake is the first-line treatment. If
this fails, then enteral or parenteral nutrition may be needed. This review examines the literature on the use of IDPN and summarizes
the current recommendations. There is considerable controversy over indications and benefits of IDPN, and well-controlled, long-
term studies are needed to help tease out these issues. In the interim, clinical judgment should be used when considering IDPN for
individual patients. (Nutr Clin Pract. 2018;33:767–771)

Keywords
chronic kidney failure; hemodialysis; intradialytic parenteral nutrition; malnutrition; parenteral nutrition

Background Technical Aspects


The presence of protein energy malnutrition (PEM) can There are 2 main types of IDPN. In compounded
be quite common1-3 among the hundreds of thousands of admixture-based IDPN, all-in-one IDPN bags are mixed by
patients in the United States who receive hemodialysis, and a pharmacy based on individual patient needs. In commer-
it independently predicts higher hospitalization rates, lower cial admixture-based IDPN, premixed bags are provided for
quality of life, and death.4 Causes of PEM in hemodialysis generic use. Of the 2, commercial premixed bags are far
patients include reduced food intake from uremia, concur- more popular because the process of compounding single
rent illnesses, alterations in the balance between hunger and bags based on specific patient needs is time consuming and
satiety hormones, dietary restrictions, financial limitations, adds cost.5 IDPN is administrated via IV infusion with
depression/anxiety, and nutrient and energy losses that an infusion pump during hemodialysis. Typically the most
occur during the hemodialysis session.3-5 concentrated IDPN formula is used to reduce the risk for
Prevention of PEM requires a multidisciplinary effort volume overload and fit the treatment within the usual time
involving the entire healthcare team. Essential steps to im- constraints of a standard hemodialysis session.
proving nutrition status include liberalizing any dietary re- A typical IDPN infusion provides 800–1200 kcal in the
strictions, encouraging food intake, providing oral nutrition form of glucose, lipids, and amino acids (usually 30–60 g
supplements, and offering nutrition education. However, of the latter).6 However, some amino acids can be lost
these interventions are not always effective in helping the in the dialysate depending on the hemodialysis filter used
patient achieve his or her nutrition goals. In such cases,
intradialytic parenteral nutrition (IDPN), which involves From the 1 Division of Nephrology and Hypertension, Department of
an intravenous (IV) infusion of essential nutrients during Medicine, NorthShore University HealthSystem, Evanston, Illinois,
hemodialysis treatments, may be helpful as a component of USA; and the 2 Department of Medicine, Indiana University School
nutrition support. of Medicine, Indianapolis, Indiana, USA.
This discussion will focus on the use of IDPN for patients Financial disclosure: None declared.
who are receiving in-center maintenance hemodialysis, who Conflicts of interest: None declared.
comprise the great majority of dialysis patients in the This article originally appeared online on September 12, 2018.
United States, and will review technical aspects and admin-
Corresponding Author:
istration, the appropriate selection of patients, risks and Menaka Sarav, MD, Division of Nephrology and Hypertension,
benefits, and evidence supporting its use. The use of IDPN Department of Medicine, 2650 Ridge Avenue, Evanston,
in patients receiving home and peritoneal dialysis will not IL 60201, USA.
be covered in this report. Email: msarav@northshore.org
768 Nutrition in Clinical Practice 33(6)

Table 1. National Kidney Foundation–Recommended Measures for Monitoring Nutrition Status in Patients Receiving
Maintenance Dialysis.

Frequency of
Parameters Measurements Comments Limitations

Predialysis serum albumin level Monthly Target level is normal >4 g/dL Affected by nonnutrition factors
such as inflammation,
dehydration, infection,
acid-base status
Percentage change in Monthly Trends from edema-free body Getting a true dry weight may be
postdialysis body weight weight need to be monitored challenging
Subjective global assessment Every 6 months A simple technique based on Does not include visceral protein
subjective and objective aspects levels in the assessment; it is
of the medical history and subjective and has not been
physical examination studied well in hemodialysis
populations
Dietary interview and/or dairy Every 6 months 3-Day dietary records followed by Time-consuming and limited by
interviews and calculating accuracy of documentation by
nutrient intake by an patient and/or interview and
experienced registered renal calculation by dietitian
dietitian
nPNA Monthly In the clinically stable patient, nPNA is not valid in catabolic or
nPNA can be used to estimate anabolic state; nPNA may
protein intake fluctuate from day to day as a
function of protein intake, and a
single nPNA measurement may
not reflect usual protein intakes

nPNA, normalized protein nitrogen appearance.

and if they have been reused.7 Electrolyte-free admixtures alone.17 In addition, all of the randomized studies have
(ie, without sodium, potassium, and phosphorus) are also been open label in design, which increases the opportunity
available.5 When taking into account amino acids and en- for bias. Moreover, even though most studies used serum
ergy that can be lost with each hemodialysis session (ie, 200– albumin level as an inclusion criterion or surrogate outcome
480 kcal of energy and 10–12 g of amino acids),8 not >3000 representing nutrition improvement, serum albumin level
kcal of energy and 150 g of amino acids will be provided by is primarily an indicator of inflammation or illness, thus
IDPN with thrice weekly dialysis. Thus, in a 70-kg patient, making interpretation of these studies more challenging.18
approximately 6 kcal/kg/d (target being 30–35 kcal/kg/d) Clearly, longer-term prospective clinical trials with appro-
of energy9,10 and 0.30 g/kg/d (target being 1.2 g/kg/d) of priate control groups are needed to identify which type of
amino acids9,10 are provided by IDPN, highlighting the fact patient is most likely to benefit from IDPN.12
that IDPN should be used only as supplemental nutrition in
patients with a spontaneous oral nutrition intake of at least Selection of Patients for IDPN
25 kcal/kg/d of energy and 0.9 g/kg/d of amino acids.5,9,11,12
The process of selecting patients for IDPN first requires
identifying patients who are malnourished. Table 1 includes
Evidence Supporting the Use of IDPN a list of parameters from the National Kidney Foundation
Several retrospective and prospective randomized studies Disease Outcome Quality Initiative (NKF KDOQI) that
have examined the effects of IDPN on clinical end points. can be used to identify malnutrition.11 As Table 1 notes,
Benefits noted included improvements in anthropometric each of the parameters have limitations, sometimes serious
parameters and serum albumin and serum prealbumin albu- ones, when applied to the hemodialysis population. There-
min levels.12-15 One human study that infused stable isotopes fore, identifying PEM in hemodialysis patients requires a
to assess muscle turnover during IDPN identified increases large dose of clinical judgment based on a careful history
in regional and total body protein synthesis.16 However, in and physical examination.
the largest randomized study of IDPN (n = 186), the addi- Once PEM is identified, IDPN should not be the initial
tion of IDPN to oral nutrition supplementation over 1 year treatment choice. Rather, encouraging increased oral nu-
did not improve 2-year mortality rates, hospitalization rates, trition or supplements like protein drinks or bars through
or general well-being as compared with oral supplements dietary counseling and education is an important first step.
Sarav and Friedman 769

Table 2. Society Recommendations on Indications for IDPN.

Kidney Disease Outcomes Quality Initiative, 1. Evidence of protein or energy malnutrition and inadequate dietary
2000 protein and/or energy intake
2. Inability to administer or tolerate adequate oral nutrition, including food
supplements or tube feeding
3. Oral or enteral intake that, when combined with IDPN, meets the
recommended dietary intake
European Society of Parenteral and Enteral 1. IDPN improves nutrition status in undernourished patients who are
Nutrition, 2009 receiving hemodialysis
2. In outpatients, if nutrition counseling and oral nutrition supplements are
unsuccessful, IDPN should be proposed
American Society of Parenteral and Enteral IDPN should not be used as a nutrition supplement in malnourished
Nutrition, 2010 chronic kidney disease stage 5 hemodialysis patients

IDPN, intradialytic parenteral nutrition.

Every effort should also be made to ease dietary restrictions Interestingly, NKF KDOQI recommends IDPN only after
and financial constraints that may work to limit food intake. EN fails, whereas ESPEN recommends IDPN as first-line
Pharmacological agents such as megestrol and ghrelin may therapy when placement of a feeding tube is deemed to be
stimulate appetite, and increase serum albumin level and higher risk than IDPN. In contrast, the American Society
weight; however, these drugs have not been studied sys- of Parenteral and Enteral Nutrition does not recommend
tematically, and large-scale prospective studies are needed IDPN because of lack of adequate supporting data.10 The
in patients who are receiving hemodialysis.19,20 Moreover, differences in recommendations between societies is likely
the physician should also search for identifiable causes for due to a lack of high-grade clinical evidence, thus making
PEM. A complete gastrointestinal workup is needed to any judgment about the benefits and risks primarily opinion
make sure there are no physical or structural impediments based. These conflicting recommendations highlight the
(eg, poor dentation, dysphagia, gastroparesis) to oral intake. need for clinical judgment in determining which patients
Other causes of anorexia such as depression, dementia, or may benefit from IDPN.
inadequate dialysis should also be excluded. When should IDPN be discontinued once initiated?
If despite the efforts described earlier the patient is Stopping is reasonable if there is sustained improvement
unable to meet the nutrition intake goals, then the option in nutrition parameters, if adverse effects of IDPN are
of IDPN should be discussed.9 Notably, in patients who are observed (see the following section), or if there is sponta-
not meeting daily nutrition goals even with oral nutrition neous improvement of energy/protein intake, thus making
supplements and IDPN, placement of a feeding tube for IDPN unnecessary. In contrast, lack of improvement after
enteral nutrition (EN) or daily parenteral nutrition (PN) 3–6 months of IDPN should also lead to discontinuation
may be the appropriate next step. Another subgroup of of IDPN.5 Ultimately the decision to discontinue IDPN,
hemodialysis patients in whom EN or PN may be beneficial like the decision to initiate it, is based primarily on clinical
are critically ill individuals. These persons should be treated judgment.
as intensive care patients.9
Several nephrology and nutrition societies have com-
mented on the management of PEM, including the use
Risks and Advantages of IDPN
of intensive nutrition counseling, dietary oral supplements, There are several benefits and drawbacks of IDPN, all of
and IDPN as shown in Table 2 and Figure 1. NKF which are listed in Table 3. Of particular interest in the
KDOQI guidelines recommend using IDPN in malnour- known drawbacks are associated metabolic and electrolyte
ished patients who are receiving dialysis who are not able derangements. For example, IDPN is contraindicated if
to increase oral ingestion with dietary counseling, do not triglyceride levels are >300 mg/dL because lipids present
tolerate oral supplementation or enteral tube feeding, and in the IDPN could exacerbate hypertriglyceridemia. Hyper-
have a sufficient spontaneous intake that combined with glycemia has also been identified as a possible complication
the nutrients provided by IDPN can reach the nutrition of IDPN. Because of the risk for electrolyte disturbances,
targets.11 European Society of Parenteral and Enteral Nu- stringent monitoring of the electrolytes, especially during
trition (ESPEN) guidelines suggest using IDPN in mal- the first weeks of IDPN support, is highly recommended.9
nourished dialysis patients who had unsuccessful responses Depending on the compounding of the nutrient solution
to nutrition counseling and oral nutrition supplements.9 blood glucose, triglycerides and liver function tests need to
770 Nutrition in Clinical Practice 33(6)

Figure 1. Algorithm for intradialytic parenteral nutrition use in patients who are receiving hemodialysis. ASPEN, American
Society of Parenteral and Enteral Nutrition; ESPEN, European Society of Parenteral and Enteral Nutrition; KDOQI, Kidney
Disease Outcomes Quality Initiative.

Table 3. Advantages and Disadvantages of IDPN. a similar rate to the control group (which was consuming
oral supplementation).17 One potential nonmedical compli-
Advantages 1. No need for a dedicated enteral feeding cation of IDPN involves its financial impact because cost
tube or parenteral nutrition vascular
access
varies based on insurance coverage. Previously, Medicare
2. Ultrafiltration during dialysis will help Part B required proof of gastrointestinal impairment before
minimize the risks for fluid overload coverage. However, because Medicare Part D has covered
3. No demands on patient time or effort IDPN under “drug therapies” since 2006, it is now easier to
Disadvantages
obtain coverage once medical necessity is demonstrated.22
1. Provides insufficient energy and protein
to support long-term daily needs
2. Does not address the problem of
improving patient’s eating behavior Conclusion
3. Side effects such as metabolic and
electrolyte abnormalities Patients requiring hemodialysis are at risk for PEM, which is
4. Medicare reimbursement for IDPN is linked to a greater likelihood of adverse clinical outcomes.4
complex and extremely time-consuming IDPN is a relatively safe and efficacious option for
IDPN, intradialytic parenteral nutrition. delivering nutrients to hemodialysis patients with reduced
spontaneous oral intake. However, it is not designed to be
the only or even the major source of nutrition sustenance. It
be monitored on a routine basis.21 Overall, IDPN has been is also not indicated for severely malnourished or critically
demonstrated in hemodialysis patients to be safe with a low ill patients who are receiving hemodialysis. No studies have
complication rate. In the largest randomized study of IDPN demonstrated that IDPN improves major clinical outcomes,
ever performed, the most common adverse effects identified although few randomized trials in this field exist.23 There-
were digestive symptoms, hypotension, and muscle cramps fore, additional studies are needed to establish the benefits
that occurred during treatment, although they occurred at of IDPN. In the meantime, clinical judgment and acumen
Sarav and Friedman 771

remain key to diagnosing PEM and determining the need for 11. Clinical practice guidelines for nutrition in chronic renal failure.
IDPN. K/DOQI, National Kidney Foundation. American journal of kidney
diseases: the official journal of the National Kidney Foundation 2000;
35(6 suppl 2):S1-S140.
Statement of Authorship 12. Dukkipati R, Kalantar-Zadeh K, Kopple JD. Is there a role for intradi-
M. Sarav and A. Friedman contributed to the conception/ alytic parenteral nutrition? A review of the evidence. Am J Kidney Dis.
2010;55(2):352-364.
design of the manuscript; M. Sarav and A. Friedman
13. Cano N, Labastie-Coeyrehourq J, Lacombe P, et al. Perdialytic
contributed to acquisition, analysis, or interpretation of the parenteral nutrition with lipids and amino acids in malnourished
data; M. Sarav and A. Friedman drafted the manuscript; hemodialysis patients. Am J Clin Nutr. 1990;52(4):726-730.
all authors critically revised the manuscript and agree to 14. Marsen TA, Beer J, Mann H. Intradialytic parenteral nutrition in main-
be fully accountable for ensuring the integrity and accuracy tenance hemodialysis patients suffering from protein-energy wasting.
Results of a multicenter, open, prospective, randomized trial. Clin Nutr.
of the work; and all authors read and approved the final
2017;36(1):107-117.
manuscript. 15. Chertow GM, Ling J, Lew NL, Lazarus JM, Lowrie EG. The
association of intradialytic parenteral nutrition administration with
References survival in hemodialysis patients. Am J Kidney Dis. 1994;24(6):
1. Kopple JD, McCollum Award Lecture, 1996: protein-energy malnutri- 912-920.
tion in maintenance dialysis patients. Am J Clin Nutr. 1997;65(5):1544- 16. Pupim LB, Flakoll PJ, Ikizler TA. Nutritional supplementation acutely
1557. increases albumin fractional synthetic rate in chronic hemodialysis
2. Kalantar-Zadeh K, Block G, McAllister CJ, Humphreys MH, Kopple patients. J Am Soc Nephrol. 2004;15(7):1920-1926.
JD. Appetite and inflammation, nutrition, anemia, and clinical out- 17. Cano NJ, Fouque D, Roth H, et al. Intradialytic parenteral nutrition
come in hemodialysis patients. Am J Clin Nutr. 2004;80(2):299-307. does not improve survival in malnourished hemodialysis patients: a 2-
3. Gracia-Iguacel C, Gonzalez-Parra E, Perez-Gomez MV, et al. Preva- year multicenter, prospective, randomized study. J Am Soc Nephrol.
lence of protein-energy wasting syndrome and its association with 2007;18(9):2583-2591.
mortality in haemodialysis patients in a centre in Spain. Nefrologia. 18. Friedman AN, Fadem SZ. Reassessment of albumin as a nutritional
2013;33(4):495-505. marker in kidney disease. J Am Soc Nephrol. 2010;21(2):223-230.
4. Kalantar-Zadeh K, Rhee C, Sim JJ, Stenvinkel P, Anker SD, Kovesdy 19. Yeh SS, Marandi M, Thode HC Jr, et al. Report of a pilot, double-
CP. Why cachexia kills: examining the causality of poor outcomes in blind, placebo-controlled study of megestrol acetate in elderly dialysis
wasting conditions. J Cachexia Sarcopenia Muscle. 2013;4(2):89-94. patients with cachexia. J Ren Nutr. 2010;20(1):52-62.
5. Sabatino A, Regolisti G, Antonucci E, Cabassi A, Morabito S, Fiac- 20. Wynne K, Giannitsopoulou K, Small CJ, et al. Subcutaneous ghrelin
cadori E. Intradialytic parenteral nutrition in end-stage renal disease: enhances acute food intake in malnourished patients who receive
practical aspects, indications and limits. J Nephrol. 2014;27(4):377-383. maintenance peritoneal dialysis: a randomized, placebo-controlled
6. Cano N. Nutritional supplementation in adult patients on hemodialy- trial. J Am Soc Nephrol. 2005;16(7):2111-2118.
sis. J Ren Nutr. 2007;17(1):103-105. 21. Druml W, Kierdorf HP; Working group for developing the guidelines
7. Ikizler TA, Flakoll PJ, Parker RA, Hakim RM. Amino acid and for parenteral nutrition of The German Association for Nutritional
albumin losses during hemodialysis. Kidney Int. 1994;46(3):830-837. Medicine. Parenteral nutrition in patients with renal failure – Guide-
8. Liu Y, Xiao X, Qin DP, et al. Comparison of intradialytic parenteral lines on Parenteral Nutrition, Chapter 17. Ger Med Sci. 2009;7:
nutrition with glucose or amino acid mixtures in maintenance hemo- Doc11.
dialysis patients. Nutrients. 2016;8(6):e220. 22. Fuhrman MP. Intradialytic parenteral nutrition and intraperitoneal
9. Cano NJ, Aparicio M, Brunori G, et al. ESPEN Guidelines on nutrition. Nutr Clin Pract. 2009;24(4):470-480.
Parenteral Nutrition: adult renal failure. Clin Nutr. 2009;28(4):401-414. 23. Pupim LB, Flakoll PJ, Brouillette JR, Levenhagen DK, Hakim RM,
10. Brown RO, Compher C. A.S.P.E.N. clinical guidelines: nutrition sup- Ikizler TA. Intradialytic parenteral nutrition improves protein and en-
port in adult acute and chronic renal failure. JPEN J Parenter Enteral ergy homeostasis in chronic hemodialysis patients. J Clin Invest. 2002;
Nutr. 2010;34(4):366-377. 110(4):483-492.
Invited Review

Nutrition in Clinical Practice


Volume 33 Number 6
Exploring the Potential Effectiveness of Combining Optimal December 2018 772–789

C 2018 American Society for

Nutrition With Electrical Stimulation to Maintain Muscle Parenteral and Enteral Nutrition
DOI: 10.1002/ncp.10213
Health in Critical Illness: A Narrative Review wileyonlinelibrary.com

Selina M. Parry, PT, PhD1,∗ ; Lee-anne S. Chapple, PhD2,∗ ;


3,∗
and Marina Mourtzakis, PhD

Abstract
Muscle wasting occurs rapidly within days of an admission to the intensive care unit (ICU). Concomitant muscle weakness and
impaired physical functioning can ensue, with lasting effects well after hospital discharge. Early physical rehabilitation is a promising
intervention to minimize muscle weakness and physical dysfunction. However, there is an often a delay in commencing active
functional exercises (such as sitting on the edge of bed, standing and mobilizing) due to sedation, patient alertness, and impaired
ability to cooperate in the initial days of ICU admission. Therefore, there is high interest in being able to intervene early through
nonvolitional exercise strategies such as electrical muscle stimulation (EMS). Muscle health characterized as the composite of
muscle quantity, as well as functional and metabolic integrity, may be potentially maintained when optimal nutrition therapy is
provided in complement with early physical rehabilitation in critically ill patients; however, the type, dosage, and timing of these
interventions are unclear. This article explores the potential role of nutrition and EMS in maintaining muscle health in critical
illness. Within this article, we will evaluate fundamental concepts of muscle wasting and evaluate the effects of EMS, as well as the
effects of nutrition therapy on muscle health and the clinical and functional outcomes in critically ill patients. We will also highlight
current research gaps in order to advance the field forward in this important area. (Nutr Clin Pract. 2018;33:772–789)

Keywords
critical illness; nutrition support; intensive care unit; muscle stimulation; muscle wasting; protein intake

Introduction environments.”8 Traditionally, physical rehabilitation com-


mences once the patient is awake and able to engage with
There is a growing population of survivors of critical illness, therapy. There is promising data to suggest that commenc-
with almost 90% of patients surviving the initial insult of ing within the first few days of an ICU admission may
critical illness.1 However, approximately 50% of survivors2 be critical to minimizing the plethora of impairments that
may develop intensive care unit–acquired weakness (ICU- can ensue.9,10 However, in the early stages of the ICU
AW) and suffer ongoing significant morbidity in terms of admission there are numerous barriers, which can mean
physical, cognitive, and psychological health.2-5 ICU-AW
refers to clinically detectable global muscle weakness that
From the 1 Department of Physiotherapy, The University of
develops as a result of no specific etiology other than being
Melbourne, Victoria, Australia; the 2 Intensive Care Research, Royal
critically unwell.6 Individuals may have evidence of my- Adelaide Hospital, South Australia, Australia; and the 3 Department
opathy, polyneuropathy, and/or significant muscle atrophy. of Kinesiology, University of Waterloo, Ontario, Canada.
ICU-AW contributes to significant impairments in physical Financial Disclosure: Department of Health, Australian Government,
functioning and decreased health-related quality of life National Health and Medical Research Council (1111640).
(HRQoL), which persist years after hospital discharge.2-5 Conflicts of Interest: None declared.
Thus, it is essential to devote attention to the quality of ∗ All joint first author in this manuscript due to equal contribution.
survivorship in terms of both clinical and physical function
This article originally appeared online on October 25, 2018.
outcomes.7
The World Health Organization defines rehabilitation Corresponding Author:
Selina M. Parry, PT, PhD, The University of Melbourne, Department
as “a set of measures that assist individuals who experi-
of Physiotherapy, School of Health Sciences, Level 7, Alan Gilbert
ence or are likely to experience disability, to achieve and Building, 161 Barry St Carlton VIC 3010, Victoria, Australia.
maintain optimum functioning in interaction with their Email: selina.parry@unimelb.edu.au
Parry et al 773

that it is challenging to provide “active” rehabilitation.11,12 Muscle Wasting in Critical Illness


We now look to technology that can be used to enable
patients to exercise without the need for volitional and direct Skeletal muscle is a highly plastic and adaptive tissue, which
patient engagement.13 The evidence is emerging in terms responds to changes in mechanical loading and is 1 of
of the potential utility of assistive technologies such as the largest tissue groups in the human body.21-23 Skeletal
bedside cycle ergometry and electrical muscle stimulation muscle is an integral tissue in predicting clinical, physical,
(EMS).10,14 Muscle stimulation in particular is an attractive and metabolic outcomes.17,24-26 To evaluate muscle health,
intervention because it enables targeted, artificial activation an aggregate of measures are essential, including muscle
and loading of individual muscles, with parameters being mass and its integrity (ie, metabolic and physical features)
adapted to provide strength or endurance load to the (Figure 1).15 Low muscle mass is reported in 20%–70%
muscle. of ICU patients at the time of ICU admission,25,26 and a
Muscle health can be characterized as a composite of further ࣈ30% reduction in quadriceps muscle mass may
muscle quantity (measured as muscle mass, muscle cross- occur within 10 days of an ICU admission.27,28 Remarkably,
sectional area, etc) as well as muscle quality (physical and low muscle mass is also reported up to 6 and 12 months
metabolic function of skeletal muscle).15 In critically ill following ICU discharge.24
patients, muscle health may be supported when optimal Skeletal muscle atrophy is associated with morbidity,
nutrition therapy is provided in addition to early physical re- increased hospital length of stay (LOS), and mortality.
habilitation; however, the type, dosage, and timing of these Lower limb muscle mass is commonly associated with
interventions are unclear.16-18 Nutrition support delivered strength, suggesting that reduction in muscle mass may
to ICU patients forms part of routine care for the majority also contribute to poorer physical performance,28-32 and
of ventilated patients. Marked catabolism occurs during these functional deficits may persist for years following
critical illness, and hence protein in particular may assist in ICU discharge3 (Figure 1). Muscle atrophy has been
reducing at least some of the muscle wasting experienced linked to elevated tumor necrosis factor-α and interleukin-
in the ICU setting. However, the delivery of nutrition does 6 concentrations,33,34 and these have been further associ-
not equate directly to utilization of energy in critical illness. ated with increased risk of infection in older adults.35,36
In other words, although one may be providing nutrition Given that skeletal muscle comprises >75% of glucose
support to ICU patients, patients’ tissues may develop disposal,37,38 it is anticipated that glucose and fat dysregula-
resistance to sufficiently taking up nutrients from enteral or tion may occur. For example, muscle atrophy as a result of
parenteral feeds to meet their energy demands. Critically ill acute immobility has been associated with impaired glucose
patients demonstrate a vast heterogeneity in energy needs tolerance,34 impaired glucose uptake,35 and reduced fat
as well as magnitude in mitochondrial dysfunction and oxidation.34 Although most of the weight lost during critical
dysregulated fat oxidation that unfavorably influence energy illness is typically regained within 1 year following ICU
generation in skeletal muscle.19 As such, we need to better discharge,39 there is evidence of sustained muscle atrophy
understand the metabolic energy demands of exercise in with reduced muscle satellite cell content40 and substantial
critically ill patients to understand its bioenergetic impli- fat gain24 up to 6 months and 1 year, respectively, following
cations and ultimately reduce muscle wasting and improve ICU discharge. Thus, understanding the timeframe and
functional recovery.20 mechanisms of muscle atrophy will lead to developing meth-
Exercise is known to improve blood flow to skeletal ods that aim to preserve muscle mass and its physiological
muscle in noncritically ill populations, which would enhance functions.
nutrient and oxygen uptake by the muscle to maintain its There are multifaceted factors that lead to skeletal muscle
integrity and function. Optimal protein intake and exercise atrophy in critically ill patients, including prolonged bed
could synergistically facilitate muscle protein deposition to rest,21 catabolic signaling (including proinflammation and
maintain muscle health. However, can these same concepts insulin resistance),41 and undernourishment via low caloric
be applied to a critically ill patient? Can muscle health be and protein intakes.42,43 Insufficient caloric intake generally
preserved with EMS in an ICU patient? Can additional compromises protein intake, leading to reduced protein
protein be delivered and used by muscle in a critically synthesis. Amino acids provide the substrate for protein
ill patient in the same manner as a healthy individual? synthesis44 ; thus, reduced untake may lead to reduced
To address these questions, we will evaluate fundamen- protein synthesis.
tal concepts of muscle wasting and evaluate the effects However, in the presence of reduced activity and insulin
of EMS, as well as the effects of nutrition therapy on resistance, it is likely that critically ill patients also exhibit
muscle health and the clinical and functional outcomes anabolic resistance, which is the reduced ability of the mus-
in critically ill patients. We will also highlight current cle to take up and use amino acids for protein synthesis.45
research gaps in order to advance the field in this important To compensate for the body’s high protein needs during
area. critical illness, skeletal muscle degradation is enhanced to
774 Nutrition in Clinical Practice 33(6)

Figure 1. Factors affecting muscle health in pre-ICU, ICU trajectory, and ICU survivorship. ICU, intensive care unit.

provide amino acids for the synthesis of other proteins acute, the consequential muscle atrophy that results may
that may be needed for immune function, for example, be longer term. Targeted rehabilitative approaches that
resulting in muscle atrophy. On the other hand, bed rest, require muscle contraction to preserve muscle mass or re-
independent of illness, can result in muscle loss.21 Reduced covery from muscle atrophy would be essential to regaining
blood flow is related to reduced delivery of amino acids to proper neural processes in rehabilitation of muscle strength.
skeletal muscle.46 Given the extent of bed rest combined However, given the unstable condition of many critically
with accelerated catabolic processes in a critically ill patient, ill patients, we need to consider a continuum of exercise
reduced delivery of amino acids to skeletal muscle would interventions that may begin with cost-effective and feasible
likely contribute extensively to muscle wasting. strategies in which participation is nonvolitional in nature
It is unequivocally important to assess the implications and then progresses to functional activities that require vol-
of morphological and metabolic deficits that evolve dur- untary contraction as patient alertness and ability to engage
ing ICU stay. Loss of muscle quality may be evidenced improve.
with infiltration of fat into skeletal muscle,47 which would
likely impair muscle glucose and fat metabolism. Muscle
characteristics and architecture including pennation angle,
Principles of EMS
echogenicity, and fascicle length are fundamental to force The quadriceps muscle is the largest muscle group in the
production and consequently strength. During critical ill- human body and integral to performance of activities such
ness, these features deteriorate27,28 and result in functional as standing from a chair, stair climbing, and locomotion.
and strength deficits at ICU discharge.28 Skeletal muscle Given its size and importance in weight-bearing activities,
relies on proper regulation of the electrical excitability it is the most widely examined muscle within the muscle
for adequate muscle contraction.48-50 Lower activation of stimulation literature.
voltage-gated Na+ channels can reduce membrane ex- An individual muscle consists of hundreds to thousands
citability, which leads to attenuated muscle contraction of fibers arranged into functional groups called motor units.
and conseuqently muscle weakness.51 This acute neuropa- The motor unit is the final common pathway in motor
thy is reported to be reversible—not degenerative—and processing, which would result in a physical action.52,53
may be related to electrolyte abnormalities during critical Small motor units have small diameter axons and typically
illness.48,49 Although this neuropathic mechanism may be innervate slow twitch (type I) muscle fibers, which are largely
Parry et al 775

Table 1. Parameters of Relevance in Muscle Stimulation.14


Parameter Description

Amplitude Quantity of energy flowing per second measured in


(intensity) milliamplitude (mA)
Pulse width Duration of electrical impulse measured in
(duration) microseconds (usecs)
Frequency Number of electrical impulses per second (Hertz);
affected by twitch summation phenomenon,
commonly frequency is set between 35–50 Hz.
With higher frequencies >100 Hz tetany of the
muscle will occur and there is risk of faster
fatigability.
Ramp-up Current intensity will increase (ramp-up) to a preset
maximum over a defined period of time
Ramp-down Current intensity will decrease (ramp-down) to a
preset minimum over a defined period of time
On:Off Length of time over which each individual electrical
duration impulse is delivered versus no stimulation can be
preset with some muscle stimulators. For example
an On:Off duration of 5:1 means that the electrical
Figure 2. Functional electrical stimulation-cycling machine impulse is delivered for 5 seconds and off for
(RT-300 supine model and SAGE stimulator; Restorative 1 second.
Therapies, Ltd, Baltimore, MD). Reprinted from Parry SM,
Berney S, Warrillow S. et al. Functional electrical stimulation
with cycling in the critically ill: A pilot case–matched control
study. J Crit Care. 2014:29:695.e1-e7, with permission from muscle recruitment potentially may enable longer
Elsevier. time for contraction to be sustained prior to reaching
a point of fatigue.57
fatigue-resistant. Large motor units have large diameter
With both NMES and FES, there are a range of param-
axons and typically innervate fast twitch (type II) muscle
eters that can be adapted to achieve muscle contraction.14
fibers, which are less fatigue-resistant but enable greater
Table 1 provides an overview of these parameters. Sufficient
force production to be generated quickly.52,54 Physiologi-
quadriceps tension may be achieved with frequencies of 5–
cally, during volitional contraction muscle recruitment is
100 Hz and a pulse width of 100–400 usecs.55 With lower
asynchronous and thought to follow the Hennemann’s size
pulse width, there is preferential activation of sensory fibers,
principle with smaller units (ie, slow twitch, fatigue-resistant
which can result in higher levels of sensory discomfort for
fibers) recruited first. Because more force is needed, larger
the patient. There is a direct inverse relationship between
units are recruited.23 The mechanism of recruitment with
pulse width and amplitude. Higher amplitudes are required
EMS remains contentious.
when using a lower pulse width in order to evoke muscle
Artificial muscle activation aims to preserve muscle mass
contraction. With higher tissue impedance, as can occur
and/or recover/improve muscle function.54,55 Skeletal mus-
with presence of edema and adipose tissue, longer pulse
cle can be artificially stimulated to induce a visible and
widths may be required. The size and location of surface
palpable muscle contraction in the absence of volitional
electrodes can influence the efficacy of muscle stimulation.
control using 2 different methodologies:
Ideally, electrodes should be placed close to the motor point
of a muscle, which is the point at which the motor units are
1. Neuromuscular electrical stimulation (NMES),
most superficial to the skin surface.
which often involves stimulation of isolated muscle
groups in a nonfunctional manner (ie, stimulated in
supine)55
EMS in the ICU: What Do We Know?
2. Functional electrical stimulation (FES), which gen- There are a number of systematic and narrative reviews
erally involves stimulation of multiple muscle groups published in the last 10 years on the topic of EMS within
while undertaking a combined functional activity the ICU setting13,14,58,59 in relation to muscle health.
such as bedside cycling55,56 (Figure 2). The muscles During the writing of this article, we conducted a lit-
are artificially stimulated to contract at the point erature search and identified at least 23 distinct studies
in range at which volitional activation would occur published within the ICU literature specifically evaluating
during the functional activity. For example, during the safety, feasibility, and/or efficacy of muscle stimulation
knee extension while cycling the quadriceps muscle (NMES or FES) since 1987 (Table 2). More than half of the
would be stimulated to contract, and on knee flexion literature (52%, n = 12/23) has been published since 2015,
the hamstrings would be activated. This alternating highlighting the rapid and emerging interest in this area.
776
Table 2. Summary of Studies Examining EMS in Intensive Care.
Author Year, Time to First Muscles
Location N Population Session Stimulated EMS Parameters Main Findings

NMES
Bouletreau 1987, 10 Hospitalized for 8 or 12 days Gastroc Patient Position Significant reduction in excretion of
France110 at least 8 days (depending on Quads Not specified 3-MH in NMES
in the ICU allocation) Stimulation Parameters
0–120 V, 1.75 Hz, 3000 usecs
On:Off time: 5:5 sec
Training Parameters
30 min 2 × day for 4 days
Gerovasili 52 ICU patients 2 days Quads Patient Position Preservation of RF and VI muscle
2009,111 Routsi with APACHE peroneus Not specified thickness observed in NMES group
2010,61 II > 13 longus Stimulation Parameters (RF: −8%; VI: −13%) vs control
Karatzanos stratified based Mean mA for quads 38 (10) and 37 (11) mA for (RF: −14%; VI: −22%)111
2012112 Greece on age and peroneus longus No significant absolute or relative
gender 45 Hz, 400 usecs differences in handgrip strength112
On:Off time: 12 sec/6 sec Higher MRC scores and less ICU-AW
Ramp time: 0.8 sec with NMES group61,112
Training Parameters
55 min/day until ICU DC
Meesen 2010, 19 Hospitalised in Unclear Quads Patient Position Reduced thigh circumference loss in
Belgium113 ICU with MV Supine with half-roll under knee (to enable knee NMES vs control limb
> 1 day flexion)
Stimulation Parameters
0–5 min: 35–85 mA, 5 Hz, 250 usecs; Stimulation
intensity (mA) gradually increased in 2–10 mA
steps proportional to stimulation intensity in
warm-up trial
5–11 min: 60 Hz, 330 usecs
11–19 min: 100 Hz, 250 usecs
19–25 min: 80 Hz, 300 usecs
25–30 min: 2 Hz, 250 usecs
On:Off time: 90:30 sec (5 min): 10:20 sec (6 min):
10:20 sec (8 min): 7:14 sec (6 min): 90:30 sec
(5 min)
Training Parameters
Daily 30-min session as long as intubated/sedated
(NMES: right leg; sham: left leg)
Gruther 2010, 16 Short (< 7 days) ST group 3 (2) Quads Patient Position Significant increase in quadriceps
Austria62 and long-term days; Not specified muscle thickness (+4.9%) vs sham
ICU patients LT group 33 (15) Stimulation Parameters (−3.2%) for long-term group.
(> 14 days) days Max tolerated: mA not specified Significant loss of quadriceps muscle
50 Hz, 350 usecs thickness in both the short-term
On:Off time: 8 sec/24 sec NMES and sham groups
Training Parameters (ࣈ37%–39%).
30 min/day week 1
60 min/day week 2-week 4
5 sessions/week for 4 weeks
(continued)
Table 2. (continued)
Author Year, Time to First Muscles
Location N Population Session Stimulated EMS Parameters Main Findings

Rodriguez 2012, 14 ICU patients 2 (1–2) days Biceps Patient Position No change in biceps thickness or
Argentina63 with sepsis Quads Not specified circumference with NMES
Stimulation Parameters Higher MRC scores for biceps and
100 Hz, 300 usecs quadriceps with NMES
On:Off time: 2 sec:4 sec
Training Parameters
30 min 2 × day continued until successful
extubation
Poulsen 2011, 8 ICU patients NR; baseline Quads Patient Position No preservation of quadriceps muscle
Denmark64 with septic measures Not specified volume observed with NMES. Both
shock assessed 26 Stimulation Parameters group had significant reduction
(16–52 hrs) 31 (23–48) mA for VM and 42 (37–54) mA for within 1 week (NMES 20%; control
VL 16%).
35 Hz, 300 usecs
On:Off time: 4 sec:6 sec
Ramp time: 0.5 secs
Training Parameters
60 min daily for 1 week
Angelopoulos 31 Acute group: Unclear Quads Patient Position Both medium and high-frequency
2013, presence of Peroneus Not reported stimulation induced
Greece114 SIRS criteria longus Stimulation Parameters microcirculatory changes to skeletal
or diagnosis of High-Frequency Protocol muscle
sepsis for 3–5 57–87 mA; 75 Hz, 400 usecs
days at time of On:Off time: 5 sec/21 sec
session Ramp time: 1.5 sec up/0.8 down
ICU-AW Group: Medium Frequency Protocol
individuals 45 Hz, 400 usecs
who had On:Off time: 5 sec/12 sec
confirmed Ramp time: 1.5 sec up/0.8 down
ICU-AW using Training Parameters
MRC-SS 30 min (with additional 5 min warm-up and 5
min recovery phase using 10 Hz current of 400
usecs), single session (pre-/postevaluation)
Hirose 2013, 25 Coma within first >7 days after Quads Patient position Preservation of leg muscle mass
Japan 115 24 hr of hospi- admission Tib ant Not specified observed with computed
talization; Hamstrings Stimulation Parameters tomography in EMS group
first-time Gastroc 40 mA, Hz and usecs not specified (compared with control)
stroke or TBI On:Off time 10:10 sec
injury; 16–75 Training Parameters
years of age 30 min daily starting 7 days after admission (3
with paralysis min warm-up; 25 min training; 2 min
of both or 1 cool-down)
limb Ceased after 14 days

(continued)

777
778
Table 2. (continued).

Author Year, Time to First Muscles


Location N Population Session Stimulated EMS Parameters Main Findings

Flavigna 2014, 11 Adult ICU Quads Patient Position Goniometry measurement of


Brazil116 patients MV Tib Anterior Not specified ankle dorsiflexion range was
for up to 48 Stimulation Parameters higher in the EMS limb
hours 18–60 mA, 50 Hz, 400 usecs compared with control
On:Off time: 9:9 sec No significant difference in
On time: 2 sec rise time, 5 sec MRC scores for muscle
contraction, 2 sec fall time strength
Training Parameters No significant difference in
20 min/daily until patient attained circumference measures
grade 4 strength for muscle except at 10-cm distance for
stimulated the thigh; lower scores in
control group
Segers 2014, 50 SICU/MICU Day 3 Quads Patient Position Nonresponders (unable to elicit
Belgium117 patients Supine, head-up 30° muscle contraction) were
enrolled on Legs in neutral: solid knee support more likely to have sepsis,
days 3–5 of roll placed under knees to achieve edema, and receipt of
admission approx. 15° hip flex and 30° knee vasopressors
whom had flex Inverse relationship between
an expected Stimulation Parameters edema quantity and type of
prolonged 0–80 mA, 50 Hz, 300–500 usecs muscle contraction
LOS for at Series time: 8 sec Series pause: 20 sec Could not predict adequate
least 3 more Ramp time: 2 sec contraction using NCS or
days Training Parameters EMG
5 times/week (Mon-Fri) With 50% of sessions erythema
25 min/day until ICU DC (5-min evident under the electrodes
warmup) but disappeared immediately
post sessions. No significant
cardiovascular or respiratory
response changes with
NMES

(continued)
Table 2. (continued).

Author Year, Time to First Muscles


Location N Population Session Stimulated EMS Parameters Main Findings

Dirks 2015, 9 Expected sedation <4.5 days Quads Patient Position NMES leg showed no atrophy in
Netherlands65 time of >3 days Not specified type I or II muscle fibers
and admitted to Stimulation Parameters compared with control leg,
the ICU Intensity set to level at which full contractions were which had a significant decline
visible and palpable, intensity every 3 min of 16(9%) and 24(7%) in type I
Averaged 29.9 mA in first session and progressively and II muscle fiber CSA.
increased to 32.3 mA in final session
Warm-up 5 min (5 Hz, 250 usecs)
Stimulation period (30 min 100 Hz, 400 usecs, 5 sec
on (0.75 sec rise, 3.5 sec contraction, 0.75 sec fall)
and 10 sec off
Cool-down 5 mins (5 Hz, 250 usecs)
Training Parameters
2 sessions per day over period of 3–10 days (NMES
leg and control leg)
Akar 2015, 30 Diagnosis of Unspecified Deltoid Patient Position NMES and NMES + exercise
Turkey118 COPD and quads Not specified groups had significant
respiratory Stimulation Parameters improvements in lower
failure 20 mA-25 mA, 50 Hz, pulse width not reported, 6-sec extremity strength, whereas
treatment with duration, 1.5-sec increase and 0.75-sec decrease upper limb strength increased
IMV without Training Parameters across all groups
comorbidities 5 times/week for a total of 20 sessions duration not No difference for weaning time or
specified functional milestones
Kho 2015, 34 Adult ICU 4.6 (1.8) days Quads Patient Position 63% (n = 146/230) sessions
USA66 patients MV Tib ant Not specified completed
> 24 hrs and Gastroc Stimulation Parameters No significant difference in leg
expected to Ramp up/down: 2 sec: < 1 sec, 50 Hz muscle strength at hospital
require > 2 days Quads: 400 usecs, On:Off time: 5 sec:10 sec discharge (however, studies
more in the ICU Tib ant/gastroc: 250 usecs discontinued before reaching a
On:Off time: 5 sec:5 sec (alternating recruitment of priori sample size)
Tib ant and gastroc) Patients receiving NMES had
Training Parameters larger increase in leg strength
60 mins/day (either single or 2 × 30 mins sessions) between ICU awakening and
9.1 (8.7) days of NMES delivered ICU discharge, and between
ICU awakening and hospital
discharge. NMES group walked
>2 times further at hospital
discharge compared with
control group.

(continued)

779
Table 2. (continued).

780
Author Year, Time to First Muscles
Location N Population Session Stimulated EMS Parameters Main Findings

Fischer 2016, 54 Adult ICU patients Day 1 Quads Patient position 94% sessions delivered
Austria67 undergoing Not specified (n = 136/145)
cardiothoracic Stimulation Parameters NMES no significant effect on
surgery and Median NMES intensities was 40–40.5 mA MLT quadriceps; NMES group
expected to remain range (2–120) mA regained strength 4.5 times
in ICU >48 hrs 0.4 ms, 66 Hz faster than patients in control
On:Off time 3.5 sec: 4.5 sec group
Ramp-up/-down: 0.5 sec
Training Parameters
2 × 30 mins, 7 days/week during entire ICU
stay (but not more than 14 days)
Dall’Acqua 25 Adults hospitalized < 48 hrs of Pec major Patient Position No complications or significant
2017, for no longer than IMV Rectus Supine changes in vital signs
Brazil60 15 days and > 24 abdominas Stimulation Parameters Significant difference with NMES
hrs of invasive MV Pec major: 53 (15) mA in terms of muscle thickness
Rec abdominas: 68 (18) mA and shorter ICU LOS
50 Hz, 300 usecs Preservation of muscle mass in
Ramp-up/-down: 1 sec NMES group (compared with
On:Off time: 3 sec:10 sec control group with significant
Training Parameters decrease in muscle mass across
NMES 30 min daily, ceased on day 7 or time)
when patient extubated or if deceased
(whichever occurred first)
Patsaki 2017, 128 ICU patients MV > ICU discharge Quads Patient Position No difference in MRC scores,
Greece 119 72 hrs and level of Peroneus Not specified handgrip strength, functional
consciousness longus Stimulation Parameters status, and hospital LOS
adequate to 45 Hz, 400 usecs between NMES and control
respond to at least On:Off time: 12 sec:6 sec groups
3 of 5 commands Rise/Fall time: 0.8 sec:0.8 sec In patients with ICU-AW the
stratified based on Training Parameters NMES group had significantly
age and diagnosis 55 mins daily 7 days/wk until hospital higher MRC scores at 2 weeks
of ICU-AW (MRC discharge compared with control
sum score ࣘ 48 or
> 48)
Shen 2017, 25 ICU patients MV > Enrolled on Biceps Patient position 68% of patients unable to
Taiwan 72 hrs ICU Day 3 Quadriceps Not specified complete handgrip strength
Stimulation Parameters No significant difference in
0–75 mA, 15,000 Hz, pulse width NR handgrip strength and MV time
Training Parameters between groups
32 mins daily 5 days/ week

(continued)
Table 2. (continued).

Author Year, Time to First Muscles


Location N Population Session Stimulated EMS Parameters Main Findings

Silva 2017, 11 Adult APACHE II ICU Day 1 Glut max 56 (15) mA for glut max Feasibility and Safety:
Brazil120 score > 13, MV Gastroc 55 (13) mA quads N = 83 (85%) of sessions
24–48 hr with Tib ant 52 (13) mA hamstrings completed
prediction to Hamstrings 57 (13) mA tib ant N = 99 (100%) of sessions a
remain MV for Quads 57 (11) mA gastroc visible contraction present
at least 3 days During the 3 days pulse width No skin burns
by senior ICU 550 (170) usecs glut max Setup time: 107 (24) min
physician on 450 (150) hamstrings Other:
admission 500 (200) usecs quads NS change in CPK levels
550 (150) usecs tib ant
450 (150) usecs gastroc
Daily for 3 days
Once a day for 15 mins (90 contractions)
pulse width equal to chronaxie, 100 Hz
On:Off time: 5 sec:5 sec
No rise time or decay
No warm-up or cool- down period
3 times total of 45 minuntes
Woo 2017, 10 Patients aged 20 Unclear Quads Patient Position No difference in circumference
Korea121 years or older Supine knee extended and quads CSA between legs
admitted to Stimulation Parameters (no additive benefit of NMES
ICU and Lowest intensity with visible muscle with cycling)
required MV for contraction
at least 24 hrs 35 Hz, 250 sec
ON:off time: 10 sec:12 sec
Training Parameters
Cycling provided prior and separately to
isolated muscle stimulation. 10-min rest
prior to NMES, which was applied for 20
min on left thigh. Single session
FES
Parry 2015, 16 Adults admitted Median of Quads Patient Position 73% of sessions delivered. One
Australia56 with a diagnosis 15.3 Hamstrings Supine head-up 30° transient episode of
of sepsis or (12-31.5) hr Gluteals Stimulation Parameters desaturation; nil other safety
severe sepsis between Gastroc 0–140 mA, 300–400 usecs, 30–50 Hz events.
and predicted to recruit to Training Parameters Trend toward earlier and faster
be MV > 48 hrs first 20–60 min/day, 5 × week until ICU recovery of functional
and remain in treatment discharge milestones in intervention group
ICU > 4 days compared with control

(continued)

781
Table 2. (continued).

Author Year, Time to First Muscles

782
Location N Population Session Stimulated EMS Parameters Main Findings

Fewer individuals required inpatient


rehabilitation in the intervention
group (43%) vs control (86%).
Lower frequency of delirium vs
control (25:87%) although not
significant. Duration of
delirium was significantly
shorter in intervention group
(0 [0–3] days) vs control
(6 [3.3–13.3])
Medrinal 19 Patients > 18 Unclear Quadriceps Patient Position Safety:
2018, years MV at During FES cycling, NMES synchronized with Only FES cycling increased CO
France68 least 24 hrs and knee extension, otherwise supine and produced sufficient
ventilated with Stimulation Parameters intensity of muscle work
pressure Stimulation intensity aimed to obtain a palpable Increase in CO by 1 L/min after 9
support contraction min of FES cycling
300 usecs, 35 Hz, No change in CO over time with
no ramp time NMES
Training Parameters Significant increase in HR,
4 consecutive 10-min sessions of bed exercise (10 TAPSE, and MAP during FES
mins PROM, 10 mins NMES, 10 mins cycle cycling. FES cycling increased
ergometry, 10 mins FES cycling), order of CO and physiological
intervention/sec Randomized. 30-min rest cardiorespiratory response and
between each intervention. Single session. reduced muscle HbO2
Fossat 2018, Patients > 18 31 (19-46) hr Quads Patient Position Safety: 1 skin allergy to electrode
France69 years, admitted Supine pads for NMES
to ICU < 72 hrs Stimulation Parameters No difference in combined cycling
prior to Based on Routsi61 + NMES in addition to usual
randomization Training Parameters care in terms of global muscle
and expected to 15 mins cycling + 50 mins NMES 5 days/week strength ICU discharge
need another 48 until ICU discharge No significant difference in
hrs in the ICU, ventilator-free days or
independent self-reported health-related
mobility quality-of-life at 6 months

APACHE II, Acute Physiologic Assessment and Chronic Health Evaluation II; CO, cardiac output; COPD, chronic obstructive pulmonary disease; CPK, creatine phosphokinase test; CSA,
cross sectional area; EMG, electromyography; EMS, electrical muscle stimulation; FES, functional electrical stimulation; Gastroc, Gastrocnemius; Glut max, gluteus maximus; HR, heart rate;
hr, hour(s); Hz, hertz; ICU, intensive care unit; ICU-AW, intensive care unit–acquired weakness; ICU DC, ICU discharge; IMV, invasive mechanical ventilation; LOS, length of stay; LT, long
term; mA, milliamplitude; MAP, mean arterial pressure; min, minute(s); MICU, medical ICU; MLT, muscle length thickness; MRC, Medical Research Council; MRC-SS, Medical Research
Council sum-score; MV, mechanical ventilation; NCS, nerve conduction study; NMES, neuromuscular electrical stimulation; n, number; NR, not reported; NS, not significant; Pec major,
pectoralis major; Rec abdominas, Rectus abdominas; RF, rectus femoris; Rx, treatment; ST, short term; Tib ant, tibialis anterior; TBI, traumatic brain injury; Quads, Quadriceps; sec,
second(s); V, voltage; VI, vastus intermedius; VL, vastus lateralis; VM, vastus medialis; SICU, surgical ICU; SIRS, systemic inflammatory response syndrome; usecs, microseconds; 3-MH,
3-methylhistidine; %, percentage; >, greater than; <, less than.
Parry et al 783

Whereas the majority of the research has been conducted in can be present in ICU patients, will negatively affect the
Europe (61%, n = 13/23), publications from South America ability to deliver the intended stimulation intensity. In these
(n = 3/23), Asia (n = 3/23), North America (n = 1/23), and circumstances, a higher pulse width and/or intensity will be
Australia (n = 1/23) are also evident. required in order to induce a muscle contraction. However,
Although the quadriceps muscle is the most widely this often results in poorer tolerance due to increased
evaluated muscle group, there was 1 study that reported on discomfort with sensory-evoked pain. There is emerging
the preservation of muscle mass for pectoralis major and interest in the role of thermally enhanced muscle stimu-
rectus abdominus muscles after NMES, as well as reduced lation, which may assist with some of the issues around
ICU LOS.60 There is significant heterogeneity within the edema and sensory discomfort while optimizing stimulation
studies in terms of stimulation parameters, intervention parameters.71 Importantly, given that optimal muscle health
duration, and timing of delivery relative to time in the may be achieved with combined muscle contraction and
ICU (Table 2). The evidence is currently inconclusive in enhanced nutrition deliver, there is a need to investigate
terms of providing definitive guidance on the efficacy of the potential synergism between NMES and optimized
muscle stimulation within the ICU setting. In the studies, nutrition intervention in future research.
which reported the time to first intervention session, the Although there are several merits to the use of EMS,
majority commenced within the first 5 days of the admission various issues need to be addressed, which include:
period (65%, n = 15 of 23) (Table 2). There is conflicting
evidence for the preservation of muscle mass using NMES, r Developing optimal stimulation parameters (inten-
particularly in individuals with higher severity of illness.60-64 sity, pulse width, frequency) without compromising
Muscle biopsy data in 1 study demonstrated absence of muscle force production due to fatigability
atrophy of type I and II muscle fibers.65 Data suggesting r Identifying those who respond versus those who do
a potential for accelerated gains in strength and physical not respond to muscle stimulation; does this modality
function have been reported at the time of ICU and hospital work for all patients?
discharge.66,67 However, there is limited evaluation of the r Developing an objective means of quantifying the
potential longer-term benefits of muscle stimulation beyond quality of muscle contraction
hospital discharge, which is an important area for further r Understanding the longer-term impact of muscle
evaluation. stimulation on patient-oriented outcomes
FES literature first emerged in 2015 with a pilot case r Further evaluate the safety and feasibility of muscle
control FES cycling study in the ICU that primarily focused stimulation
on determining the safety and feasibility of early interven-
tion (ie, within a couple of days of ICU admission).56 The Is There a Role for Protein to Improve Muscle
study demonstrated safety and feasibility with 1 transient
adverse event (desaturation) and delivery of three-quarters
Health?
of potential intervention sessions.56 There was a trend Protein delivery is considered vital for muscle maintenance
toward faster recovery of functional milestones, reduced in both health and disease. Because critical care manage-
delirium, and reduced need for rehabilitation posthospital ment shifts to focus on strategies to maintain muscle mass
discharge.56 Medrinal et al68 explored the impact of 4 and function, attention has been placed on the role of
different assistive rehabilitation strategies (passive range of protein in maintaining muscle health in this population.72
motion exercises with therapist, NMES, cycle ergometry, Accordingly, several narrative reviews advocate increased
and FES) on cardiorespiratory response to exercise. The protein delivery to critically ill patients,73-76 most notably
study demonstrated that FES cycling was the main in- by Hoffer and Bistrian, which strongly suggested that
tervention, which increased cardiac output and produced providing 2–2.5 g/kg/d is safe and could be optimal during
sufficient intensity of muscle work; however, a recent study critical illness.77 Delivery of greater protein doses is also
that combined NMES and cycling reported no difference recommended in a number of international guidelines: the
in outcomes in terms of muscle strength and ventilator-free 2016 update of the American Society for Parenteral and En-
days or HRQoL at 6 months.69 teral Nutrition critical care nutrition guidelines recommend
Beyond the heterogeneity in muscle stimulation param- delivery of protein doses of 1.2–2.0 g/kg/day or higher,78
eters, training duration, and timing to intervene, there and the European Society of Parenteral and Enteral Nu-
are also potential confounding factors unique to the trition Parenteral Nutrition Guidelines for Critical Care
ICU population, which may contribute to the conflicting recommend providing 1.3–1.5 g protein/kg/day.79 These
evidence regarding the efficacy of muscle stimulation. recommendations are, however, based on very low quality
Tissue impedance will impact the intensity and delivery of evidence.
of current flow toward the underlying motor units.54,55,70 Early physiological studies show nitrogen balance is
Increased edema and high adipose tissue, 2 factors that negative during states of inflammation.80 More recently,
784 Nutrition in Clinical Practice 33(6)

Figure 3. Potential effects of combined EMS and increased protein intake on muscle health during the ICU trajectory and
survivorship. (Text in light gray font indicates factors that are likely to be unaffected by EMS.) EMS, electrical muscle stimulation;
ICU, intensive care unit.

stable isotope-labeled amino acid tracer techniques have lationship between protein delivery and muscle health.
been used to provide an estimate of protein metabolism Puthucheary et al conducted an observational study in 63
(synthesis, breakdown, and oxidation) at the whole-body critically ill adults and reported an association between
level. Rooyackers et al used this technique to demonstrate greater protein delivery and increased loss in ultrasound-
that critically ill patients with multiorgan failure have an derived quadriceps muscle.27 Two RCTs of differing protein
increase in both protein synthesis and breakdown.81 Further doses have been conducted thus far, which both were
work from Sweden has shown that the addition of par- single-centered and evaluated the effect of protein dose
enteral nutrition (PN) or enteral nutrition (EN) results in an on ultrasound-derived muscle size. The first, by Ferrie
overall small improvement in whole-body protein balance. et al, used PN to deliver augmented protein (1.1 g/kg/day)
Whereas nutrient provision appears to stimulate protein compared with standard care (0.9 g/kg/day) in 119 critically
breakdown, it also stimulates protein synthesis to a greater ill adults. Greater protein was associated with an increased
degree, resulting in an overall positive protein balance.82-84 forearm muscle thickness, with no difference in measures of
However, these studies only measure incorporation of la- strength.89 Another Australian group showed that greater
beled amino acids into the plasma, and hence it is unknown enterally delivered protein to 60 critically ill patients was
whether additional amino acids are delivered, taken up, associated with attenuation of quadricep muscle loss at ICU
and utilized by the muscle during critical illness. An expert discharge.90 It is important to consider that there is much to
review published in 2017 thoroughly discusses the complex- be learned by the distinct study designs used in these studies,
ity of protein kinetics in critical illness, demonstrating that which may have led to discrepant results. Future work
a greater understanding of the physiological response to exploring protein intervention on specific muscle outcomes
protein supplementation during different clinical conditions is necessary to better understand the role of protein delivery
is required.85 during critical illness (Figure 3).
A number of observational studies show an association There is also work being conducted on other nutrition
between greater protein delivery and improved clinical strategies that may influence muscle health during critical
outcomes, including mortality and time to discharge from illness. Bear et al are exploring the role of intermittent versus
ICU alive.86-88 However, few studies have shown a re- continuous EN in critical illness on ultrasound-derived
Parry et al 785

rectus femoris cross-sectional area, for which results are group receiving NMES and nutrition support had a 2-fold
expected this year (ClinicalTrials.gov NCT02358512). reduction in loss of muscle CSA. In addition, there are 2
This same group is exploring the role of β-hydroxy- registered trials currently recruiting that combine exercise
β-methylbutyrate, a derivative of leucine, on the same and nutrition interventions: 1) NEXIS (ClinicalTrials.org
outcome (ClinicalTrials.gov NCT03464708). Although NCT03021902), combining cycle ergometry and amino
physiological studies demonstrate improved protein balance acid supplementation versus standard care; and 2) ExPrEs
with nutrition support during critical illness, and pilot (ClinicalTrials.org NCT02509520), combining NMES and
studies show potential attenuation of muscle wasting with leucine supplementation in elderly ICU patients.
protein delivery, it is unknown whether nutrition therapy In designing studies that provide both exercise and
alone is able to reduce the significant muscle atrophy that nutrition early in critical illness, a number of factors need
occurs during critical illness, particularly early after injury to be carefully considered:
for which protein catabolism is at its greatest.91 It also needs
to be considered whether early physical rehabilitation dur-
ing critical illness places a subsequent demand on increased 1. The route of nutrition support is important. We know
nutrient availability to facilitate muscle synthesis. Therefore, that in critical illness delayed gastric motility100
it is thought that the combination of nutrition support and and impaired nutrient absorption of both fat101
exercise therapy during critical illness may provide the great- and carbohydrate102 affect nutrient uptake from
est benefit on muscle attenuation and functional recovery. enterally delivered nutrition. Whether enterally de-
livered protein can be adequately absorbed and
How Could We Combine Exercise and Protein incorporated into muscle during critical illness is
yet to be determined (ANZCTR Trials Registration;
for Best Patient Outcomes? ACTRN12616001652460). Whereas EN may be the
In health, the combination of exercise and amino acid preferred and commonly used route when compared
delivery has been shown to enhance the capacity of the with PN, it is important to consider how much of
skeletal muscle to promote protein synthesis.92 In non- the nutrition delivered is available for muscle uptake.
ICU studies, combining protein and exercise demonstrate In the elderly, postprandial splanchnic extraction
far greater benefits in terms of muscle mass, strength, and (uptake of amino acids for the splanchnic organs)
physical functioning compared with either therapy on their is increased, reducing the availability of amino acids
own.92,93 Pennings et al showed low-intensity cycling and for muscle synthesis.103,104 Whether an exacerbation
resistance-type tasks prior to amino acid consumption in- of splanchnic sequestration during critical illness
creased muscle protein synthesis rates when compared with occurs requires quantification.
rest.94 Similarly, Biolo et al showed amino acid transport 2. The duration of intervention should be contemplated.
after exercise is 30%–100% greater than following rest,95 and The majority of large randomized controlled trials
this may be related to exercise-induced nutrient-stimulated of nutrition interventions during critical illness are
vasodilation.96 Bed-rest studies demonstrate the benefit of only delivered for a short period of time early during
multimodal interventions. Trappe et al97 conducted a 3-arm the ICU admission. This is despite studies report-
study in 24 healthy women. During 60 days of bed rest, ing reduced nutrition intake in survivors of critical
participants underwent bed rest only, bed rest plus exercise illness,105,106 which is likely to influence functional
regime, or bed rest plus a leucine-enhanced high-protein recovery.
diet to 1.6 g/kg/day. Exercise therapy, which included a 3. Timing of both nutrient and exercise therapy should be
combination of aerobic and resistance exercises, was shown considered. In critical illness, it is common practice
to have a positive impact on muscle attenuation, whereas it to provide nutrition continuously, whereas exercise
was reported that the greatest muscle loss occurred in those interventions are likely to be administered at set
patients who received the high-protein diet.97 timepoints. As discussed by Bear et al in their review
In critical illness, despite the coupling of protein and on the pros and cons of feeding modalities, there
exercise being identified as a key priority area during the may be cause to believe that bolus or intermittent
International Summit of Critical Care experts,98 there are delivery of EN will have a greater influence on
few studies with observational or controlled interventions, muscle protein synthesis.107 Based on this premise,
that report data on both physical function and nutrition the effect of intermittent versus continuous EN on
intake. In a poster abstract, Wappel et al presented a ultrasound-derived muscle wasting is being explored
subanalysis of a pilot study that compared exercise alone (ClinicalTrials.gov NCT02358512). Timing relative
with an exercise-nutrition intervention that also included to the exercise (ie, pre- vs postexercise nutrition)
NMES on the outcome of cross-sectional area of the may also have implications on the extent of protein
quadriceps.99 Although not powered to show an effect, the synthesis and degradation.17,94
786 Nutrition in Clinical Practice 33(6)

4. Bioenergetics of critically ill patients need to be of the data; and S. M. Parry, L-A. S. Chapple, and M. Mourtza-
comprehensively investigated. We need to understand kis drafted the manuscript. All authors critically revised the
the bioenergetics and physiological ability of the manuscript, agree to be fully accountable for ensuring the
muscle to respond to muscle stimulation, particu- integrity and accuracy of the work, and read and approved
larly in high severity of illness in which there may be the final manuscript.
compromised muscle membrane function and mito-
chondrial dysfunction.20 The metabolic demands of References
exercise are poorly understood within the ICU 1. Kaukonen KM, Bailey M, Suzuki S, Pilcher D, Bellomo R. Mortality
setting.20 Pilot data demonstrates that energy re- related to severe sepsis and septic shock among critically ill patients in
Australia and New Zealand, 2000–2012. JAMA. 2014;311(13):1308-
quirements are likely to be higher in individu-
1316.
als with critical illness compared with the healthy 2. Appleton RTD, Kinsella J, Quasim T. The incidence of intensive care
population.108 We postulate that energy demands unit-acquired weakness syndromes: A systematic review. J Intensive
will be lower with muscle stimulation compared with Care Soc. 2015;16(2):126-136.
use of technology such as cycle ergometry or func- 3. Herridge MS, Tansey CM, Matte A, et al. Functional disability
5 years after acute respiratory distress syndrome. N Engl J Med.
tional rehabilitation, but this needs to be explored.
2011;364(14):1293-1304.
We also need to better understand whether amino 4. Hermans G, Van Mechelen H, Clerckx B, et al. Acute outcomes and
acid uptake and protein turnover maintain their 1-year mortality of intensive care unit-acquired weakness. A cohort
capacity during critical illness to preserve muscle study and propensity-matched analysis. Am J Respir Crit Care Med.
integrity. 2014;190(4):410-420.
5. Fan E, Dowdy DW, Colantuoni E, et al. Physical complications in
5. The patient population needs to be clearly defined.
acute lung injury survivors: a two-year longitudinal prospective study.
ICU studies involve a highly heterogenous group Crit Care Med. 2014;42(4):849-859.
of individuals. However it is important to recognize 6. Stevens R, Marshall S, Cornblath D, et al. A framework for diagnos-
that the response to nutrition and exercise interven- ing and classifying intensive care unit-acquired weakness. Crit Care
tions is likely to be influenced by a number of patient Med. 2009;37(10):S299-308.
7. Needham DM, Davidson J, Cohen H, et al. Improving long-
characteristics, including sarcopenia, age, comor-
term outcomes after discharge from intensive care unit: report
bidities, and obesity. Recent work has also proposed from a stakeholders’ conference. Crit Care Med. 2012;40(2):
that genetic predisposition may predict the extent 502-509.
of muscle wasting.109 Therefore, consideration and 8. (WHO), W.H.O. Concept Paper – WHO Guidelines on Health-
categorization of the patient population are vital. Related Rehabilitation (Rehabilitation Guidelines), 2011.
9. Tipping CJ, Harrold M, Holland A, Romero L, Nisbet T, Hodgson
CL. The effects of active mobilisation and rehabilitation in ICU
Conclusions on mortality and function: a systematic review. Intensive Care Med.
2017;43(2):171-183.
Muscle dysfunction is common for survivors of critical 10. Hodgson CL, Tipping CJ. Physiotherapy management of intensive care
illness and remains a significant issue; it may influence unit-acquired weakness. J Physiother. 2017;63(1):4-10.
strength and physical performance. Current research sug- 11. Parry SM, Nydahl P, Needham DM. Implementing early physi-
cal rehabilitation and mobilisation in the ICU: institutional, clin-
gests that early intervention within the first few days may ician, and patient considerations. Intensive Care Med. 2018;44(4):
be the critical window for improvement of strength and 470-473.
functional recovery outcomes. However, our understanding 12. Parry SM, Remedios L, Denehy L, et al. What factors affect imple-
of the physiological changes that occur in muscle during mentation of early rehabilitation into intensive care unit practice? A
critical illness needs to be further explored. This work would qualitative study with clinicians. J Crit Care. 2017;38:137-143.
13. Needham DM, Truong AD, Fan E. Technology to enhance physical
inform the potential effectiveness of combining optimal rehabilitation of critically ill patients. Crit Care Med. 2009;37(10
protein nutrition with electrical stimulation. To preserve Suppl):S436-S441.
muscle health and potentially improve clinical and patient- 14. Parry SM, Berney S, Granger CL, Koopman R, El-Ansary D,
oriented outcomes, there is a continual and significant need Denehy L. Electrical muscle stimulation in the intensive care setting:
to understand the mechanistic effects of combined therapies a systematic review. Crit Care Med. 2013;41(10):2406-2418.
15. Mourtzakis M, Parry S, Connolly B, Puthucheary Z. Skeletal Muscle
in conjunction with consideration of the dosage, timing, and Ultrasound in Critical Care: A Tool in Need of Translation. Ann Am
delivery methods. Thorac Soc. 2017;14(10):1495-1503.
16. Bear DE, Wandrag L, Merriweather JL, Connolly B, Hart N, Grocott
MPW. The role of nutritional support in the physical and func-
Statement of Authorship
tional recovery of critically ill patients: a narrative review. Crit Care.
S. M. Parry, L-A. S. Chapple, and M. Mourtzakis equally 2017;21(1):226.
contributed to the conception and design of the research; 17. Heyland DK, Stapleton RD, Mourtzakis M, et al. Combining nutri-
S. M. Parry, L-A. S. Chapple, and M. Mourtzakis contributed tion and exercise to optimize survival and recovery from critical illness:
to the acquisition and analysis of the data; S. M. Parry, L.-A. S. Conceptual and methodological issues. Clin Nutr. 2016;35(5):1196-
Chapple, and M. Mourtzakis contributed to the interpretation 1206.
Parry et al 787

18. Denehy L, Granger CL, El-Ansary D, Parry SM. Advances in 38. DeFronzo RA, Jacot E, Jequier E, Maeder E, Wahren J, Felber
cardiorespiratory physiotherapy and their clinical impact. Expert Rev JP. The effect of insulin on the disposal of intravenous glucose.
Respir Med. 2018;12(3):203-215. Results from indirect calorimetry and hepatic and femoral venous
19. Puthucheary ZA, Astin R, McPhail MJW, et al. Metabolic phenotype catheterization. Diabetes. 1981;30(12):1000-1007.
of skeletal muscle in early critical illness. Thorax. 2018. 39. Herridge MS, Cheung AM, Tansey CM, et al. One-year outcomes in
20. Bear DE, Parry SM, Puthucheary ZA. Can the critically ill patient survivors of the acute respiratory distress syndrome. N Engl J Med.
generate sufficient energy to facilitate exercise in the ICU? Curr Opin 2003;348(8):683-693.
Clin Nutr Metab Care. 2018;21(2):110-115. 40. Dos Santos C, Hussain SN, Mathur S, et al. Mechanisms of Chronic
21. Parry SM, Puthucheary ZA. The impact of extended bed rest on Muscle Wasting and Dysfunction after an Intensive Care Unit Stay. A
the musculoskeletal system in the critical care environment. Extrem Pilot Study. Am J Respir Crit Care Med. 2016;194(7):821-830.
Physiol Med. 2015;4:16. 41. Glass DJ. Signaling pathways perturbing muscle mass. Curr Opin Clin
22. de Boer MD, Maganaris CN, Seynnes OR, Rennie MJ, Narici MV. Nutr Metab Care. 2010;13(3):225-229.
Time course of muscular, neural and tendinous adaptations to 23 day 42. Rubinson L, Diette GB, Song X, Brower RG, Krishnan JA. Low
unilateral lower-limb suspension in young men. J Physiol. 2007;583 caloric intake is associated with nosocomial bloodstream infections
(Pt 3):1079-1091. in patients in the medical intensive care unit. Crit Care Med.
23. Sherwood L. Human Physiology: From Cells to Systems, ed. CL Inc. 2004;32(2):350-357.
Vol. 9th Ed. 2016. 43. Heyland DK, Schroter-Noppe D, Drover JW, et al. Nutrition
24. Chan KS, Mourtzakis M, Aronson Friedman L, et al. Evaluat- support in the critical care setting: current practice in canadian
ing Muscle Mass in Survivors of Acute Respiratory Distress Syn- ICUs–opportunities for improvement? JPEN J Parenter Enteral Nutr.
drome: A 1-Year Multicenter Longitudinal Study. Crit Care Med. 2003;27(1):74-83.
2018;46(8):1238-1246. 44. Drummond MJ, Bell JA, Fujita S, et al. Amino Acids are Necessary
25. Moisey LL, Mourtzakis M, Cotton BA, et al. Skeletal muscle predicts for the Insulin-Induced Activation of mTOR/S6K1 Signaling and
ventilator-free days, ICU-free days, and mortality in elderly ICU Protein Synthesis in Healthy and Insulin Resistant Human Skeletal
patients. Crit Care. 2013;17(5):R206. Muscle. Clin Nutr. 2008;27(3):447-456.
26. Weijs PJ, Looijaard WG, Dekker IM, et al. Low skeletal muscle area 45. Morton RW, Traylor DA, Weijs PJM, Phillips SM. Defining anabolic
is a risk factor for mortality in mechanically ventilated critically ill resistance: implications for delivery of clinical care nutrition. Curr
patients. Crit Care. 2014;18(2):R12. Opin Crit Care. 2018;24(2):124-130.
27. Puthucheary ZA, Rawal J, McPhail M, et al. Acute skeletal muscle 46. Rasmussen BB, Fujita S, Wolfe RR, et al. Insulin resistance of muscle
wasting in critical illness. JAMA. 2013;310(15):1591-1600. protein metabolism in aging. Faseb j. 2006;20(6):768-769.
28. Parry SM, El-Ansary D, Cartwright MS, et al. Ultrasonography in 47. Looijaard WG, Dekker IM, Stapel SN, et al. Skeletal muscle quality
the intensive care setting can be used to detect changes in the quality as assessed by CT-derived skeletal muscle density is associated with
and quantity of muscle and is related to muscle strength and function. 6-month mortality in mechanically ventilated critically ill patients.
J Crit Care. 2015;30(5):1151.e9-14. Crit Care. 2016;20(1):386.
29. de Bruin PF, Ueki J, Watson A, Pride NB. Size and strength of the 48. Z’Graggen WJ, Lin CS, Howard RS, Beale RJ, Bostock H. Nerve
respiratory and quadriceps muscles in patients with chronic asthma. excitability changes in critical illness polyneuropathy. Brain. 2006;129
Eur Respir J. 1997;10(1):59-64. (Pt 9):2461-2470.
30. Seymour JM, Ward K, Sidhu PS, et al. Ultrasound measurement 49. Teener JW, Rich MM. Dysregulation of sodium channel gating in
of rectus femoris cross-sectional area and the relationship with critical illness myopathy. J Muscle Res Cell Motil. 2006;27(5-7):291-
quadriceps strength in COPD. Thorax. 2009;64(5):418-423. 296.
31. Young A, Stokes M, Crowe M. Size and strength of the quadriceps 50. Novak KR, Nardelli P, Cope TC, et al. Inactivation of sodium
muscles of old and young women. Eur J Clin Invest. 1984;14(4):282- channels underlies reversible neuropathy during critical illness in rats.
287. J Clin Invest. 2009;119(5):1150-1158.
32. Hayashida I, Tanimoto Y, Takahashi Y, Kusabiraki T, Tamaki J. 51. Koch S, Bierbrauer J, Haas K, et al. Critical illness polyneuropathy in
Correlation between muscle strength and muscle mass, and their as- ICU patients is related to reduced motor nerve excitability caused by
sociation with walking speed, in community-dwelling elderly Japanese reduced sodium permeability. Intensive Care Medicine Experimental.
individuals. PLoS One. 2014;9(11):e111810. 2016;4:10.
33. Visser M, Pahor M, Taaffe DR, et al. Relationship of interleukin- 52. Sheffler LR, Chae J. Neuromuscular electrical stimulation in neurore-
6 and tumor necrosis factor-alpha with muscle mass and muscle habilitation. Muscle Nerve. 2007;35(5):562-590.
strength in elderly men and women: the Health ABC Study. J Gerontol 53. Edstrom L, Grimby L. Effect of exercise on the motor unit. Muscle
A Biol Sci Med Sci. 2002;57(5):M326-M332. Nerve. 1986;9(2):104-126.
34. Schaap LA, Pluijm SM, Deeg DJ, et al. Higher inflammatory marker 54. Maffiuletti NA. Physiological and methodological considerations for
levels in older persons: associations with 5-year change in muscle mass the use of neuromuscular electrical stimulation. Eur J Appl Physiol.
and muscle strength. J Gerontol A Biol Sci Med Sci. 2009;64(11):1183- 2010;110(2):223-234.
1189. 55. Maffiuletti NA, Gondin J, Place N, Stevens-Lapsley J, Vivodtzev I,
35. Cosqueric G, Sebag A, Ducolombier C, Thomas C, Piette F, Weill- Minetto MA. Clinical Use of Neuromuscular Electrical Stimulation
Engerer S. Sarcopenia is predictive of nosocomial infection in care of for Neuromuscular Rehabilitation: What Are We Overlooking? Arch
the elderly. Br J Nutr. 2006;96(5):895-901. Phys Med Rehabil. 2018;99(4):806-812.
36. Brandt C, Pedersen BK. The role of exercise-induced myokines in 56. Parry SM, Berney S, Warrillow S, et al. Functional electrical stimula-
muscle homeostasis and the defense against chronic diseases. J Biomed tion with cycling in the critically ill: a pilot case-matched control study.
Biotechnol. 2010;2010:520258. J Crit Care. 2014;29(4):695.e1-7.
37. Shulman GI, Rothman DL, Jue T, Stein P, DeFronzo RA, Shulman 57. Decker MJ, Griffin L, Abraham LD, Brandt L. Alternating stim-
RG. Quantitation of muscle glycogen synthesis in normal subjects and ulation of synergistic muscles during functional electrical stimula-
subjects with non-insulin-dependent diabetes by 13C nuclear magnetic tion cycling improves endurance in persons with spinal cord injury.
resonance spectroscopy. N Engl J Med. 1990;322(4):223-228. J Electromyogr Kinesiol. 2010;20(6):1163-1169.
788 Nutrition in Clinical Practice 33(6)

58. Maffiuletti NA, Roig M, Karatzanos E, Nanas S. Neuromuscular 76. Weijs PJ. Protein delivery in critical illness. Curr Opin Crit Care.
electrical stimulation for preventing skeletal-muscle weakness and 2016;22(4):299-302.
wasting in critically ill patients: a systematic review. BMC Med. 77. Hoffer LJ, Bistrian BR. Appropriate protein provision in criti-
2013;11:137. cal illness: a systematic and narrative review. Am J Clin Nutr.
59. Williams N, Flynn M. A review of the efficacy of neuromuscular 2012;96(3):591-600.
electrical stimulation in critically ill patients. Physiother Theory Pract. 78. McClave SA, Taylor BE, Martindale RG, et al. Guidelines for the
2014;30(1):6-11. Provision and Assessment of Nutrition Support Therapy in the Adult
60. Dall’ Acqua AM, Sachetti A, Santos LJ, et al. Use of neuromuscular Critically Ill Patient: Society of Critical Care Medicine (SCCM) and
electrical stimulation to preserve the thickness of abdominal and chest American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.).
muscles of critically ill patients: A randomized clinical trial. J Rehabil JPEN J Parenter Enteral Nutr. 2016;40(2):159-211.
Med. 2017;49(1):40-48. 79. Singer P, Berger MM, Van den Berghe G, et al. ESPEN Guidelines on
61. Routsi C, Gerovasili V, Vasileiadis I, et al. Electrical muscle stim- Parenteral Nutrition: intensive care. Clin Nutr. 2009;28(4):387-400.
ulation prevents critical illness polyneuromyopathy: a randomized 80. Plank LD, Hill GL. Sequential metabolic changes following induction
parallel intervention trial. Crit Care. 2010;14(2):R74. of systemic inflammatory response in patients with severe sepsis or
62. Gruther W, Kainberger F, Fialka-Moser V, et al. Effects of neu- major blunt trauma. World J Surg. 2000;24(6):630-638.
romuscular electrical stimulation on muscle layer thickness of knee 81. Rooyackers O, Kouchek-Zadeh R, Tjader I, Norberg A, Klaude M,
extensor muscles in intensive care unit patients: a pilot study. J Rehabil Wernerman J. Whole body protein turnover in critically ill patients
Med. 2010;42(6):593-597. with multiple organ failure. Clin Nutr. 2015;34(1):95-100.
63. Rodriguez PO, Setten M, Maskin LP, et al. Muscle weakness in 82. Liebau F, Sundstrom M, van Loon LJ, Wernerman J, Rooyackers O.
septic patients requiring mechanical ventilation: protective effect of Short-term amino acid infusion improves protein balance in critically
transcutaneous neuromuscular electrical stimulation. J Crit Care. ill patients. Crit Care. 2015;19:106.
2012;27(3):319.e1-8. 83. Liebau F, Wernerman J, van Loon LJ, Rooyackers O. Effect of
64. Poulsen JB, Moller K, Jensen CV, Weisdorf S, Kehlet H, Perner initiating enteral protein feeding on whole-body protein turnover in
A. Effect of transcutaneous electrical muscle stimulation on muscle critically ill patients. Am J Clin Nutr. 2015;101(3):549-557.
volume in patients with septic shock. Crit Care Med. 2011;39(3):456- 84. Sundstrom Rehal M, Liebau F, Tjader I, Norberg A, Rooyackers
461. O, Wernerman J. A supplemental intravenous amino acid infusion
65. Dirks ML, Hansen D, Van Assche A, Dendale P, Van Loon LJ. sustains a positive protein balance for 24 hours in critically ill patients.
Neuromuscular electrical stimulation prevents muscle wasting in Crit Care. 2017;21(1):298.
critically ill comatose patients. Clin Sci (Lond). 2015;128(6):357-365. 85. Martindale RG, Heyland DK, Rugeles SJ, et al. Protein Kinetics and
66. Kho ME, Truong AD, Zanni JM, et al. Neuromuscular electrical Metabolic Effects Related to Disease States in the Intensive Care Unit.
stimulation in mechanically ventilated patients: a randomized, sham- Nutr Clin Pract. 2017;32(1 suppl):21s-29s.
controlled pilot trial with blinded outcome assessment. J Crit Care. 86. Nicolo M, Heyland DK, Chittams J, Sammarco T, Compher C.
2015;30(1):32-39. Clinical Outcomes Related to Protein Delivery in a Critically Ill
67. Fischer A, Spiegl M, Altmann K, et al. Muscle mass, strength Population: A Multicenter, Multinational Observation Study. JPEN
and functional outcomes in critically ill patients after cardiothoracic J Parenter Enteral Nutr. 2016;40(1):45-51.
surgery: does neuromuscular electrical stimulation help? The Catastim 87. Allingstrup MJ, Esmailzadeh N, Wilkens Knudsen A, et al. Provision
2 randomized controlled trial. Crit Care. 2016;20:30. of protein and energy in relation to measured requirements in inten-
68. Medrinal C, Combret Y, Prieur G, et al. Comparison of exercise sive care patients. Clin Nutr. 2012;31(4):462-468.
intensity during four early rehabilitation techniques in sedated and 88. Weijs PJ, Looijaard WG, Beishuizen A, Girbes AR, Oudemans-van
ventilated patients in ICU: a randomised cross-over trial. Crit Care. Straaten HM. Early high protein intake is associated with low mortal-
2018;22(1):110. ity and energy overfeeding with high mortality in non-septic mechan-
69. Fossat G, Baudin F, Courtes L, et al. Effect of In-Bed Leg Cycling ically ventilated critically ill patients. Crit Care. 2014;18(6):701.
and Electrical Stimulation of the Quadriceps on Global Muscle 89. Ferrie S, Allman-Farinelli M, Daley M, Smith K. Protein Require-
Strength in Critically Ill Adults: A Randomized Clinical Trial. JAMA. ments in the Critically Ill: A Randomized Controlled Trial Using
2018;320(4):368-378. Parenteral Nutrition. JPEN J Parenter Enteral Nutr. 2016;40(6):795-
70. Quittan M. Aspects of physical medicine and rehabilitation in the 805.
treatment of deconditioned patients in the acute care setting: the role 90. Fetterplace K, Deane AM, Tierney A, et al. Targeted Full Energy
of skeletal muscle. Wien Med Wochenschr. 2016;166(1-2):28-38. and Protein Delivery in Critically Ill Patients: A Pilot Randomized
71. Callaghan M, Nouri B. Performance Of Thermal-Enhanced Electrical Controlled Trial (FEED Trial). JPEN J Parenter Enteral Nutr. 2018.
Muscle Stimulation In Challenging Subjects: A Randomized Controlled 91. Gamrin-Gripenberg L, Sundstrom-Rehal M, Olsson D, Grip J, Wern-
Trial, in C29. MUSCLES, EXERCISE ASSESSMENT, AND RE- erman J, Rooyackers O. An attenuated rate of leg muscle protein
HABILITATION. p. A6664-A6664. depletion and leg free amino acid efflux over time is seen in ICU long-
72. Hurt RT, McClave SA, Martindale RG, et al. Summary Points stayers. Crit Care. 2018;22(1):13.
and Consensus Recommendations From the International Protein 92. Morton RW, Murphy KT, McKellar SR, et al. A systematic review,
Summit. Nutr Clin Pract. 2017;32(1 suppl):142s-151s. meta-analysis and meta-regression of the effect of protein supple-
73. Heyland DK, Weijs PJ, Coss-Bu JA, et al. Protein Delivery in mentation on resistance training-induced gains in muscle mass and
the Intensive Care Unit: Optimal or Suboptimal? Nutr Clin Pract. strength in healthy adults. Br J Sports Med. 2018;52(6):376-384.
2017;31(1):58S-71S. 93. Heyland DK, Stapleton R, Compher C. Should We Prescribe More
74. Hoffer LJ. Protein requirement in critical illness. Appl Physiol Nutr Protein to Critically Ill Patients? Nutrients. 2018;10(4).
Metab. 2016;41(5):573-576. 94. Pennings B, Koopman R, Beelen M, Senden JM, Saris WH, van Loon
75. Gunst J, Vanhorebeek I, Thiessen SE, Van den Berghe G. LJ. Exercising before protein intake allows for greater use of dietary
Amino acid supplements in critically ill patients. 2018;130:127-131. protein-derived amino acids for de novo muscle protein synthesis in
https://www.ncbi.nlm.nih.gov/pubmed/29223645. both young and elderly men. Am J Clin Nutr. 2011;93(2):322-331.
Parry et al 789

95. Biolo G, Tipton KD, Klein S, Wolfe RR. An abundant supply of 110. Bouletreau P, Patricot MC, Saudin F, Guiraud M, Mathian B.
amino acids enhances the metabolic effect of exercise on muscle Effects of intermittent electrical stimulations on muscle catabolism in
protein. Am J Physiol. 1997;273(1 Pt 1):E122-E129. intensive care patients. JPEN J Parenter Enteral Nutr. 1987;11(6):552-
96. Timmerman KL, Dhanani S, Glynn EL, et al. A moderate acute 555.
increase in physical activity enhances nutritive flow and the muscle 111. Gerovasili V, Stefanidis K, Vitzilaios K, et al. Electrical muscle
protein anabolic response to mixed nutrient intake in older adults. Am stimulation preserves the muscle mass of critically ill patients: a
J Clin Nutr. 2012;95(6):1403-1412. randomized study. Crit Care. 2009;13(5):R161.
97. Trappe TA, Burd NA, Louis ES, Lee GA, Trappe SW. Influence of 112. Karatzanos E, Gerovasili V, Zervakis D, et al. Electrical Muscle
concurrent exercise or nutrition countermeasures on thigh and calf Stimulation: An Effective Form of Exercise and Early Mobilization
muscle size and function during 60 days of bed rest in women. Acta to Preserve Muscle Strength in Critically Ill Patients. Critical Care
Physiol (Oxf). 2007;191(2):147-159. Research and Practice. 2012;2012:432752.
98. Arabi YM, Casaer MP, Chapman M, et al. The intensive care 113. Meesen RL, Dendale P, Cuypers K, et al. Neuromuscular electrical
medicine research agenda in nutrition and metabolism. Intensive Care stimulation as a possible means to prevent muscle tissue wasting
Med. 2017;43(9):1239-1256. in artificially ventilated and sedated patients in the intensive care
99. Wappel S, Ali O, Serra M, et al. The effect of an exercise, nutrition unit: A pilot study. Neuromodulation. 2010;13(4):315-320; discussion
and neuromuscular electrical stimulation intervention on acute muscle 321.
wasting in critically ill patients using mechanical ventilation. AJR- 114. Angelopoulos E, Karatzanos E, Dimopoulos S, et al. Acute mi-
CCM. 2017;195(A2747). crocirculatory effects of medium frequency versus high frequency
100. Chapman M, Fraser R, Vozzo R, et al. Antro-pyloro-duodenal motor neuromuscular electrical stimulation in critically ill patients – a pilot
responses to gastric and duodenal nutrient in critically ill patients. Gut. study. Ann Intensive Care. 2013;3(1):39.
2005;54(10):1384-1390. 115. Hirose T, Shiozaki T, Shimizu K, et al. The effect of electrical muscle
101. Ali Abdelhamid Y, Cousins CE, Sim JA, et al. Effect of Critical stimulation on the prevention of disuse muscle atrophy in patients
Illness on Triglyceride Absorption. JPEN J Parenter Enteral Nutr. with consciousness disturbance in the intensive care unit. J Crit Care.
2015;39(8):966-972. 2013;28(4):536.e1-7.
102. Chapman MJ, Fraser RJ, Matthews G, et al. Glucose absorption and 116. Falavigna L, Silva MG, AL F, et al. Effects of electrical muscle
gastric emptying in critical illness. Crit Care. 2009;13(4):R140. stimulation early in the quadriceps and tibialis anterior muscle
103. Boirie Y, Gachon P, Beaufrere B. Splanchnic and whole-body leucine of critically ill patients. Phyiother Theory Pract. 2014;30(4):223-
kinetics in young and elderly men. Am J Clin Nutr. 1997;65(2):489- 228.
495. 117. Segers J, Hermans G, Bruyninckx F, Meyfroidt G, Langer D,
104. Jonker R, Engelen MP, Deutz NE. Role of specific dietary amino acids Gosselink R. Feasibility of neuromuscular electrical stimulation in
in clinical conditions. Br J Nutr. 2012;108 (Suppl 2):S139-S148. critically ill patients. J Crit Care. 2014;29(6):1082-1088.
105. Chapple LS, Deane AM, Heyland DK, et al. Energy and protein 118. Akar O, Gunay E, Sarinc Ulasli S, et al. Efficacy of neuromuscular
deficits throughout hospitalization in patients admitted with a trau- electrical stimulation in patients with COPD followed in intensive care
matic brain injury. Clin Nutr. 2016;35(6):1315-1322. unit. Clin Respir J. 2017;11(6):743-750.
106. Ridley EJ, Parke RL, Davies AR, et al. What Happens to Nutrition 119. Patsaki I, Gerovasili V, Sidiras G, et al. Effect of neuromuscular
Intake in the Post-Intensive Care Unit Hospitalization Period? An stimulation and individualized rehabilitation on muscle strength in
Observational Cohort Study in Critically Ill Adults. JPEN J Parenter Intensive Care Unit survivors: A randomized trial. J Crit Care.
Enteral Nutr. 2018. 2017;40:76-82.
107. Bear DE, Hart N, Puthucheary Z. Continuous or intermittent feeding: 120. Silva PE, Babault N, Mazullo JB, et al. Safety and feasibility of
pros and cons. Curr Opin Crit Care. 2018;24(4):256-261. a neuromuscular electrical stimulation chronaxie-based protocol in
108. Black C, Singer M, Grocott M. The oxygen cost of rehabilitation in critical ill patients: A prospective observational study. J Crit Care.
mechanically ventilated patients. AJRCCM. 2017;195(A2742). 2017;37:141-148.
109. Garros RF, Paul R, Connolly M, et al. MicroRNA-542 Promotes 121. Woo K, Kim J, Kim HB, et al. The Effect of Electrical Muscle
Mitochondrial Dysfunction and SMAD Activity and Is Elevated in Stimulation and In-bed Cycling on Muscle Strength and Mass of
Intensive Care Unit-acquired Weakness. Am J Respir Crit Care Med. Mechanically Ventilated Patients: A Pilot Study. Acute Crit Care.
2017;196(11):1422-1433. 2018;33(1):16-22.
Invited Review

Nutrition in Clinical Practice


Volume 33 Number 6
Methods of Enteral Nutrition Administration in Critically Ill December 2018 790–795

C 2018 American Society for

Patients: Continuous, Cyclic, Intermittent, and Bolus Feeding Parenteral and Enteral Nutrition
DOI: 10.1002/ncp.10105
wileyonlinelibrary.com

Satomi Ichimaru, PhD, RD, CNSC

Abstract
There are several methods of enteral nutrition (EN) administration, including continuous, cyclic, intermittent, and bolus techniques,
which can be used either alone or in combination. Continuous feeding involves hourly administration of EN over 24 hours assisted
by a feeding pump; cyclic feeding involves administration of EN over a time period of <24 hours generally assisted by a feeding
pump; intermittent feeding involves administration of EN over 20–60 minutes every 4–6 hours via pump assist or gravity assist;
and bolus feeding involves administration of EN over a 4- to 10-minute period using a syringe or gravity drip. In practice, pump-
assisted continuous feeding is generally acceptable for critically ill patients to prevent EN-related complications. However, a limited
number of studies have been conducted to support this practice. In addition, regarding muscle protein synthesis and gastrointestinal
hormone secretion, intermittent or bolus feeding may be more beneficial than continuous EN feeding for critically ill patients.
For medically stable patients with feeding tubes terminating in the stomach, bolus feeding is favored with respect to practical
factors, such as cost, convenience, and patient mobility. However, few studies have shown whether intermittent or bolus feeding is
beneficial in a critical care setting at present. Additional randomized controlled studies comparing intermittent with bolus feeding
are required. (Nutr Clin Pract. 2018;33:790–795)

Keywords
cost and cost analysis; critical illness; enteral nutrition; nutrition support; patient safety; protein synthesis; respiratory aspiration;
tube feeding

Introduction Continuous Feeding


Enteral nutrition (EN) is defined as nutrition provided Continuous feeding provides EN by electric enteral feeding
through the gastrointestinal (GI) tract via a tube, catheter, pump over 24 hours, which is generally initiated at a rate of
or stoma that delivers nutrients distal to the oral cavity.1 20–50 mL/h and advanced to goal rate by 10–25 mL/h every
The modes of EN administration include continuous, cyclic, 4–24 hours.4 This method is selected for patients who are
intermittent, and bolus techniques, which can be used either critically ill, who have been intubated for respiratory failure,
alone or in combination. To determine the most appropriate who are fed through a postpyloric tube, or who cannot
method of administration, the clinician should consider tolerate intermittent or bolus feedings.5
numerous factors such as patient’s age, preexisting and Although continuous feeding is preferred by most ICUs,
current medical condition, nutrition status and requirement, it is supported by only a few relatively outdated studies.
enteral route for feeding (gastric vs small bowel), GI tol- A study on 76 adult burn patients reported that patients
erance, formula type used, patient mobility, feeding pump who received continuous feeding had lower stool frequency
availability, and cost. Continuous feeding seems to be the
standard in the intensive care unit (ICU)2 ; however, there From the Department of Nutrition Management, Osaka Saiseikai
is currently insufficient data to choose the best method Nakatsu Hospital, Osaka, Japan.
to improve patient outcomes of critically ill patients. This This article was modified on 2018-06-25 after initial online publication
narrative review summarizes EN administration methods, to correct the article title and to make some other editing changes.
their advantages and disadvantages, and the indications of Financial disclosure: None declared.
intermittent and/or bolus feeding in critically ill patients. Conflicts of interest: None declared.
This article originally appeared online on June 20, 2018.
Methods of EN Administration Corresponding Author:
Satomi Ichimaru, PhD, RD, CNSC, Department of Nutrition
The 4 methods of EN administration include continu- Management, Osaka Saiseikai Nakatsu Hospital, 2-10-39 Shibata
ous, cyclic, intermittent, and bolus feedings as shown in Kitaku, Osaka 530-0012, Japan.
Figure 1.3 Email: satomi.ichimaru@gmail.com
Ichimaru 791

Figure 1. Methods of delivering enteral tube feeding. (Reprinted by permission from Springer Nature: Diet and Nutrition in
Critical Care, Intermittent and Bolus Methods of Feeding in Critical Care, Ichimaru S and Amagai T, Springer Science+Business
Media New York 2015)

and reached their nutrition goals sooner than those who Bonten et al17 and Tamowicz et al18 hypothesized
received bolus feeding.6 In adult patients with neurological that cyclic feeding beneficially affected gastric acidity and
impairment who received continuous feeding, aspiration bacterial colonization of the stomach and respiratory tract,
was observed less frequently (1/17) than in those who thereby helping to prevent ventilator-associated pneumonia
received intermittent feeding (3/17).7 In a study of 18 (VAP). In these studies, a significant decrease in gastric
postoperative patients, continuous feeding groups showed pH was observed when EN was discontinued in the cyclic
improved cumulative nitrogen balance for 5 days compared feeding groups. However, only the study by Tamowicz
with cyclic feeding groups.8 Whether continuous feeding et al18 showed reduced rate of gastric colonization, and the
provides better glycemic control than intermittent/bolus incidence of VAP was relatively similar between the cyclic
feeding remains controversial.9-11 and continuous feeding groups in both studies.
Continuous feeding is frequently interrupted in the With regard to mortality in the ICU, the cyclic and con-
ICU.12 The most common reasons for discontinuation of tinuous feeding groups showed no significant differences.17
feeding include surgery (27%) and diagnostic procedures In 1 study, the length of hospital stay was found to be
(15%), and minor reasons for EN interruptions include significantly shorter in patients receiving cyclic feeding than
mechanical feeding tube problems (8%), pharmacy delivery in those receiving continuous feeding.19
delay (4%), and miscellaneous factors (3%). Because of the
frequency with which enteral feeds are interrupted, resulting Intermittent Feeding
in delivery of only 50%–60% of prescribed EN volume on a
Intermittent EN feeding is usually delivered over 20–60
daily basis,13 some institutions have implemented a volume-
minutes by infusion pump or by the gravity drip method. In
based feeding protocol to ensure that the volume of EN
the gravity drip method, formula flows out from a feeding
prescribed to their patients is actually provided.14-16
bag and into a feeding tube by gravity. The rate of infusion
is regulated by adjusting a roller clamp.5 Usually, the gravity
Cyclic Feeding drip feeding is tolerated when infused into the stomach.1 In
Cyclic feeding involves feeding by electric enteral feeding intermittent feeding, a volume of 240–720 mL of feeding is
pump over a period of <24 hours, in which the goal infusion administered 4–6 times per day depending on the patient’s
rate is determined by dividing the desired formula volume nutrition needs.5 This feeding method is more physiological
by the number of hours of administration. The infusion than continuous/cyclic feeding because it permits greater
time may vary between 24 and 8 h/day depending on the patient mobility between feedings. If tolerated, the volume
patient’s volume tolerance. This method can be used for of each feeding can be increased and the total number of
patients with feeding tubes terminating in the stomach or feeds can be decreased to improve quality of life.5 According
small bowel. During the course of recovery, patients may to old studies, intermittent feeding has been believed to
transition from the continuous feeding to nocturnal cyclic have some disadvantages, such as risk for aspiration7 and
feeding to stimulate patient’s appetite during the day. It also diarrhea20 ; however, in a recent study of ICU patients
increases patients’ mobility by freeing them from a feeding receiving intermittent or continuous feeding, there was no
set or pump.5 difference in outcomes, including the rate of aspiration
792 Nutrition in Clinical Practice 33(6)

and diarrhea.21 In 1 study, intermittent and continuous and an increase in the rate of MPS. MPS has been found
feeding groups of critically ill trauma patients showed no to peak at 90 minutes, decrease thereafter, and return to
significant differences in mortality in the ICU or incidence baseline at 180 minutes.38 In a study using neonatal pigs,
of pneumonia.22 In 2 studies that compared intermittent significant increases in intramuscular AKT and mTOR, as
and 16-hour cyclic feeding in elderly patients, no significant well as increased MPS, were observed with bolus compared
differences were noted between the groups in mortality, with continuous feeding.39
incidence of diarrhea, or pneumonia.23,24 After the intake of a meal, several hormones, such as
glucagon-like peptide-1, gastric inhibitory peptide, chole-
Bolus Feeding cystokinin, ghrelin, and peptide YY, are secreted from the
enteroendocrine cells lining the lumen of the GI tract.
Bolus feeding is administered via syringe or gravity drip These hormones regulate GI motility, gallbladder con-
over a short period, usually 4–10 minutes. Generally, the traction, pancreatic function, and nutrient absorption40 ;
patient is fed a volume of 240 mL of feeding 3–6 times most of these hormones are secreted within minutes of
daily.5 Feeding provided by this rapid infusion method may feeding, rise transiently, and decline to basal levels after
result in diarrhea and/or aspiration.5,6,25 Therefore, bolus feeding. In continuous tube feeding, this enterohormonal
feeding is usually reserved for the medically stable patients response to nutrition is almost completely absent.41-44
with feeding tubes ending in the stomach.1 For patients with Furthermore, bolus feeding may stimulate small-intestinal
a gastrostomy tube, not only commercial liquid formulas growth. In a study using neonatal pigs, greater small-
but also blended food or viscosity-thickened formula can intestinal mucosal weight and ileal protein mass have been
be administered in the bolus feeding.26,27 One advantage observed fed via bolus feeding compared with continuous
of bolus feeding is that medication can be separately ad- feeding.45
ministered from feeding. Furthermore, this method closely Although the studies introduced earlier are conducted
resembles normal eating patterns, increases the time away in non-ICU settings, we cannot deny the possibility that
from feeding, and provides freedom of movement and a those results may be applicable to ICU patients. Thus,
more normal life. regarding MPS and GI hormone secretion, intermittent
To the best of our knowledge, no study has evaluated or bolus feeding may be more beneficial than continuous
the outcomes of mortality or length of hospital stay be- EN feeding for critically ill patients. Further randomized
tween patients receiving bolus and continuous feeding. The controlled studies comparing continuous with intermittent
bolus feeding group showed improved nitrogen balance in and bolus feeding are required to confirm this hypotheses.
1 study,28 whereas the continuous feeding group showed
significantly improved body weight and arm circumference
in another study.29 Respiratory quotient, resting energy Intermittent vs Bolus Feeding
expenditure, and blood sugar were comparable between Several studies have been conducted to compare the out-
cyclic and bolus feedings in head-injured patients with comes of feeding methods in the ICU; however, to our
mechanical ventilation.30 knowledge, no study has compared intermittent with bolus
Table 1 shows the advantages, disadvantages, and indica- feeding. One reason for this could be that pump-assisted
tions for each method.3 continuous feeding is generally considered acceptable for
patients in the ICU to prevent EN intolerance.2,46 Another
Potential Advantages of Intermittent/ reason could be that intermittent and bolus feeding are often
Bolus Feeding: Muscle Protein Synthesis treated as the same feeding method because of the lack of a
clear definition of them.
and GI Hormone Secretion Currently, no clinical guidelines strongly recommend
In ICU patients, muscle breakdown often exceeds muscle a specific method of feeding for either critically ill or
synthesis, and skeletal muscle wasting leads to functional stable patients. According to the Canadian Critical Care
impairment in most survivors of critical illness.31-33 Insulin Nutrition Guidelines in 2015, there are insufficient data to
and leucine enhance muscle protein synthesis (MPS) by make recommendations on whether EN should be given
activating AKT/protein kinase B and the mammalian target continuously or via other methods in critically ill patients.47
of rapamycin (mTOR).34-36 The continuous infusion of The American guidelines of Nutrition Support Therapy
amino acids for 30–60 minutes has been found to lead in the Adult Critically Ill Patient in 2016 suggest that
to stimulated MPS; however, after 120 minutes, MPS was for high-risk patients or those intolerant to bolus gastric
found to have declined to baseline despite continuation EN, the delivery of EN should be switched to continuous
of the amino acid infusion.37 An oral whey protein bolus infusion.2 The National Institute for Health and Care
has been found to cause a pulsatile release of insulin, a Excellence Clinical Guidelines recommend that EN should
pulsatile increase in intramuscular leucine concentration, usually be continuously administered over 16–24 hours daily
Ichimaru 793

Table 1. Advantages, Disadvantages, and Indications of Each Feeding Method.

Feeding Method Advantages Disadvantages Indications

Continuous May improve tolerance Feeding pump required Initiation of feeding in


May reduce risk of aspiration May restrict ambulation critically ill patients
Increased time for nutrient absorption More expensive Promote tolerance
Compromised gastric
function
Feeding into small bowel
Intolerance to other
feeding methods
Cyclic Facilitates transition of support to oral diet Feeding pump required Transitioning from EN to
Allows daytime ambulation May require high oral nutrition (enhance
Encourages patient to eat normal meals and infusion rates appetite during the day)
snacks May promote intolerance Supplement inadequate
oral intake
Free patient from enteral
feedings during the day
Intermittent Feeding pump may not be required Increased risk for Intolerance to bolus
May enhance quality of life aspiration administration
Allows greater mobility between feedings Gastric distention Initiation of EN without
More physiological Delayed gastric emptying feeding pump
May be better tolerated than bolus feeding
Bolus More physiological Increased risk of Recommended for gastric
Feeding pump not required aspiration feeding
Inexpensive and easy administration Hypertonic, high-fat, or Normal gastric function
Limits feeding time high-fiber formulas
Patient is free to move about, participate in may delay gastric
rehabilitation therapies, and live a emptying or result in
relatively normal life osmotic diarrhea
More likely patient will receive all of formula

EN, enteral nutrition.


Reprinted by permission from Springer Nature: Diet and Nutrition in Critical Care, Intermittent and Bolus Methods of Feeding in Critical Care,
Ichimaru S and Amagai T, Springer Science+Business Media New York 2015.

for patients in the ICU, and if insulin administration is Summary


required, continuous feeding over 24 hours is safe and more
practical.48 The major points of this review are summarized as follows:

r There are 4 methods of EN administration: continu-


ous, cyclic, intermittent, and bolus feedings.
Application of Intermittent and/or Bolus r At present, there is no evidence that suggests that any
Feeding to Other Conditions particular feeding method is superior to others.
In the administration of EN at a lower acuity hospital
r In the ICU, pump-assisted continuous feeding is
unit, a long-term care facility, or the home, it is important generally acceptable to prevent EN-related complica-
to address practical factors, such as cost, convenience, tions.
and patient mobility. Compared with other methods, bolus
r Regarding MPS and GI hormone secretion, inter-
feeding is inexpensive because it requires less equipment mittent or bolus, rather than continuous, EN feeding
such as feeding bags, administration sets, and pumps. In may be favorable even for critically ill patients.
addition, bolus feeding requires the least amount of time,
r In a lower acuity hospital unit, a long-term care fa-
and it is easy to perform for caregivers at home. In patients cility, or the home, bolus feeding is preferred because
who frequently tug and dislodge nasogastric tubes, bolus it is inexpensive, easy to perform, requires the least
feeding may be safer than intermittent feeding. Especially amount of time, and mimics normal eating patterns.
for alert and active patients, bolus feeding can allow them to
r Further research is required to determine whether in-
live a relatively normal life, because of shorter feeding time termittent or bolus feeding is beneficial for critically
and freedom from mechanical devices between feedings. ill patients.
794 Nutrition in Clinical Practice 33(6)

References patients treated under conditions of intensive therapy. Adv Clin


Exp. 2007;16:365-373.
1. Bankhead R, Boullata J, Brantley S, et al. Enteral nutrition practice
19. van Berge Henegouwen MI, Akkermans LM, van Gulik TM, et al.
recommendations. JPEN J Parenter Enteral Nutr. 2009;33(2):122-167.
Prospective, randomized trial on the effect of cyclic versus continu-
2. McClave SA, Taylor BE, Martindale RG, et al. Guidelines for the
ous enteral nutrition on postoperative gastric function after pylorus-
provision and assessment of nutrition support therapy in the adult
preserving pancreatoduodenectomy. Ann Surg. 1997;226:677-685.
critically ill patient: Society of Critical Care Medicine (SCCM) and
20. Ciocon JO, Galindo-Ciocon DJ, Tiessen C, Galindo D. Continuous
American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.).
compared with intermittent tube feeding in the elderly. JPEN J Parenter
JPEN J Parenter Enteral Nutr. 2016;40:159-211.
Enteral Nutr. 1992;16:525-528.
3. Ichimaru S, Amagai T. Intermittent and bolus methods of feeding in
21. Serpa LF, Kimura M, Faintuch J, Ceconello I. Effects of continuous
critical care. In: Rajendram R, Preedy VR, Patel VB, eds. Diet and
versus bolus infusion of enteral nutrition in critical patients. Rev Hosp
Nutrition in Critical Care. New York: Springer; 2014:533-548.
Clin Fac Med Sao Paulo. 2003;58:9-14.
4. Parrish CR. Enteral feeding: the art and the science. Nutr Clin Pract.
22. MacLeod JB, Lefton J, Houghton D, et al. Prospective randomized
2003;18:76-85.
control trial of intermittent versus continuous gastric feeds for critically
5. Brantley SL, Mills ME. Overview of enteral nutrition. In: Mueller CM,
ill trauma patients. J Trauma. 2007;63:57-61.
ed. The A.S.P.E.N. Nutrition Support Core Curriculum. 2nd ed. Silver
23. Lee JS, Auyeung TW. A comparison of two feeding methods in
Spring, MD: American Society for Parenteral and Enteral Nutrition;
the alleviation of diarrhoea in older tube-fed patients: a randomised
2012:170-184.
controlled trial. Age Ageing. 2003;32:388-393.
6. Hiebert JM, Brown A, Anderson RG, Halfacre S, Rodeheaver GT,
24. Lee JS, Kwok T, Chui PY, et al. Can continuous pump feeding
Edlich RF. Comparison of continuous vs intermittent tube feedings
reduce the incidence of pneumonia in nasogastric tube-fed patients? A
in adult burn patients. JPEN J Parenter Enteral Nutr. 1981;5:73-75.
randomized controlled trial. Clin Nutr. 2010;29:453-458.
7. Kocan MJ, Hickisch SM. A comparison of continuous and intermit-
25. Steevens EC, Lipscomb AF, Poole GV, Sacks GS. Comparison of con-
tent enteral nutrition in NICU patients. J Neurosci Nurs. 1986;18:333-
tinuous vs intermittent nasogastric enteral feeding in trauma patients:
337.
perceptions and practice. Nutr Clin Pract. 2002;17:118-122.
8. Campbell IT, Morton RP, Macdonald IA, Judd S, Shapiro L, Stell
26. Epp L, Lammert L, Vallumsetla N, Hurt RT, Mundi MS. Use of
PM. Comparison of the metabolic effects of continuous postoperative
blenderized tube feeding in adult and pediatric home enteral nutrition
enteral feeding and feeding at night only. Am J Clin Nutr. 1990;52:1107-
patients. Nutr Clin Pract. 2017;32:201-205.
1112.
27. Ichimaru S, Amagai T, Wakita M, Shiro Y. Which is more effective
9. Sanz Parı́s A, Lázaro J, Guallar A, Gracia P, Caverni A, Albero R.
to prevent enteral nutrition-related complications, high- or medium-
Continuous enteral nutrition versus single bolus: effects on urine C
viscosity thickened enteral formula in patients with percutaneous
peptide and nitrogen balance. Med Clin (Barc). 2005;124:613-615.
endoscopic gastrostomy? A single-center retrospective chart review.
10. Shahriari M, Rezaei E, Bakht LA, Abbasi S. Comparison of the effects
Nutr Clin Pract. 2012;27:545-552.
of enteral feeding through the bolus and continuous methods on blood
28. Campbell IT, Morton RP, Cole JA, Raine CH, Shapiro LM, Stell PM.
sugar and prealbumin levels in ICU inpatients. J Educ Health Promot.
A comparison of the effects of intermittent and continuous nasogastric
2015;4:95.
feeding on the oxygen consumption and nitrogen balance of patients
11. Evans DC, Forbes R, Jones C, et al. Continuous versus bolus tube
after major head and neck surgery. Am J Clin Nutr. 1983;38:870-878.
feeds: does the modality affect glycemic variability, tube feeding
29. Pichard C, Roulet M. Constant rate enteral nutrition in bucco-
volume, caloric intake, or insulin utilization? Int J Crit Illn Inj Sci.
pharyngeal cancer care. A highly efficient nutritional support system.
2016;6:9-15.
Clin Otolaryngol Allied Sci. 1984;9:209-214.
12. Morgan LM, Dickerson RN, Alexander KH, Brown RO, Minard G.
30. Maurya I, Pawar M, Garg R, Kaur M, Sood R. Comparison of
Factors causing interrupted delivery of enteral nutrition in trauma
respiratory quotient and resting energy expenditure in two regimens of
intensive care unit patients. Nutr Clin Pract. 2004;19:511-517.
enteral feeding-continuous vs. intermittent in head-injured critically ill
13. Heyland DK, Dhaliwal R, Wang M, Day AG. The prevalence of ia-
patients. Saudi J Anaesth. 2011;5:195-201.
trogenic underfeeding in the nutritionally “at-risk” critically ill patient:
31. Puthucheary ZA, Rawal J, McPhail M, et al. Acute skeletal muscle
results of an international, multicenter, prospective study. Clin Nutr.
wasting in critical illness. JAMA. 2013;310:1591-1600.
2015;34(4):659-666.
32. Stevens RD, Dowdy DW, Michaels RK, et al. Neuromuscular dysfunc-
14. Heyland DK, Murch L, Cahill N, et al. Enhanced protein-energy
tion acquired in critical illness: a systematic review. Intensive Care Med.
provision via the enteral route feeding protocol in critically ill patients:
2007;33:1876-1891.
results of a cluster randomized trial. Crit Care Med. 2013;41:2743-
33. Herridge MS, Tansey CM, Matté A, et al. Functional disability 5 years
2753.
after acute respiratory distress syndrome. N Engl J Med. 2011;364:1293-
15. Taylor B, Brody R, Denmark R, Southard R, Byham-Gray L. Im-
1304.
proving delivery through the adoption of the “Feed Early Enteral
34. Bolster DR, Jefferson LS, Kimball SR. Regulation of protein synthesis
Diet Adequately for Maximum Effect (FEED ME)” protocol in a
associated with skeletal muscle hypertrophy by insulin-, amino acid-
surgical trauma ICU: a quality improvement review. Nutr Clin Pract.
and exercise-induced signalling. Proc Nutr Soc. 2004;63:351-356.
2014;29:639-648.
35. Katsanos CS, Kobayashi H, Sheffield-Moore M, Aarsland A, Wolfe
16. Declercq B, Deane AM, Wang M, Chapman MJ, Heyland DK.
RR. A high proportion of leucine is required for optimal stimulation
Enhanced Protein-Energy Provision via the Enteral Route Feeding
of the rate of muscle protein synthesis by essential amino acids in the
(PEPuP) protocol in critically ill surgical patients: a multicentre
elderly. Am J Physiol Endocrinol Metab. 2006;291:E381-E387.
prospective evaluation. Anaesth Intensive Care. 2016;44:93-98.
36. Anthony JC, Yoshizawa F, Anthony TG, Vary TC, Jefferson LS,
17. Bonten MJ, Gaillard CA, van der Hulst R, et al. Intermittent enteral
Kimball SR. Leucine stimulates translation initiation in skeletal muscle
feeding: the influence on respiratory and digestive tract colonization in
of postabsorptive rats via a rapamycin-sensitive pathway. J Nutr.
mechanically ventilated intensive-care-unit patients. Am J Respir Crit
2000;130:2413-2419.
Care Med. 1996;154:394-399.
37. Bohé J, Low JF, Wolfe RR, Rennie MJ. Latency and duration of
18. Tamowicz B, Mikstacki A, Grzymislawski M. The influence of
stimulation of human muscle protein synthesis during continuous
the Feeding Therapy Model on pulmonary complications in
infusion of amino acids. J Physiol. 2001;532:575-579.
Ichimaru 795

38. Atherton PJ, Etheridge T, Watt PW, et al. Muscle full effect after oral concentrations in healthy adults: a randomized crossover study. Ann
protein: time-dependent concordance and discordance between human Surg. 2016;263:450-457.
muscle protein synthesis and mTORC1 signaling. Am J Clin Nutr. 44. Jawaheer G, Shaw NJ, Pierro A. Continuous enteral feeding impairs
2010;92:1080-1088. gallbladder emptying in infants. J Pediatr. 2001;138:822-825.
39. Gazzaneo MC, Suryawan A, Orellana RA, et al. Intermittent bo- 45. Shulman RJ, Redel CA, Stathos TH. Bolus versus continuous feedings
lus feeding has a greater stimulatory effect on protein synthesis in stimulate small-intestinal growth and development in the newborn pig.
skeletal muscle than continuous feeding in neonatal pigs. J Nutr. J Pediatr Gastroenterol Nutr. 1994;18:350-354.
2011;141:2152-2158. 46. Mentec H, Dupont H, Bocchetti M, Cani P, Ponche F, Bleichner G.
40. Gutierrez-Aguilar R, Woods SC. Nutrition and L and K- Upper digestive intolerance during enteral nutrition in critically ill
enteroendocrine cells. Curr Opin Endocrinol Diabetes Obes. 2011;18:35- patients: frequency, risk factors, and complications. Crit Care Med.
41. 2001;29:1955-1961.
41. Ledeboer M, Masclee AA, Biemond I, Lamers CB. Gallbladder motil- 47. Heyland DK, Dhaliwal R, Drover JW, et al. Continuous vs.
ity and cholecystokinin secretion during continuous enteral nutrition. other methods of administration. In: Canadian Critical Care Nu-
Am J Gastroenterol. 1997;92:2274-2279. trition Guidelines in 2015. https://www.criticalcarenutrition.com/docs/
42. Stoll B, Puiman PJ, Cui L, et al. Continuous parenteral and enteral CPGs%202015/6.3%202015.pdf. Accessed November 4, 2017.
nutrition induces metabolic dysfunction in neonatal pigs. JPEN J 48. National Collaborating Centre for Acute Care. Enteral tube feeding
Parenter Enteral Nutr. 2012;36:538-550. in hospital and the community. In: Nutrition Support for Adults:
43. Chowdhury AH, Murray K, Hoad CL, et al. Effects of bolus and Oral Nutrition Support, Enteral Tube Feeding and Parenteral Nutri-
continuous nasogastric feeding on gastric emptying, small bowel water tion. London: National Institute for Health and Care Excellence;
content, superior mesenteric artery blood flow, and plasma hormone 2006.
Invited Review

Nutrition in Clinical Practice


Volume 33 Number 6
Technology in Parenteral Nutrition Compounding December 2018 796–802

C 2018 American Society for

Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10086
wileyonlinelibrary.com

Caitlin Curtis, PharmD, BCNSP

Abstract
Technology is constantly being used in novel ways, and its use in the practice of medicine is no exception. Examples of this
include computer physician order entry, barcode-medication scanning, electronic health records (EHRs), and bedside patient charts,
to name a few. Compounding parenteral nutrition has been included in this technological revolution, with improvements such
as barcode-assisted medication preparation systems and EHR-to-compounder interfaces. Along with some of these electronic
advancements come the inevitable improvements and challenges, which are explored in this article. (Nutr Clin Pract. 2018;33:796–
802)

Keywords
parenteral nutrition; parenteral nutrition solutions; patient safety; medication errors; drug compounding

Compounding parenteral nutrition (PN) easily lends itself refer to PN that is compounded with fat emulsion. PN are
to a discussion regarding technology, as PN remains 1 of referred to as “formula” or “formulations” unless otherwise
the most complex medications prescribed, prepared, and specified.
administered in an inpatient or ambulatory setting. As a
result of its complexity, PN appropriately remains listed as Reducing Errors
a high-risk medication by the Institute for Safe Medication
As compounding PN is a complex process with multiple
Practices (ISMP).1 With every medication, there are several
steps leading to a finished product, it is prone to errors.1,2
steps in the medication-use process in which errors can
Unfortunately, these errors can lead to severe consequences
occur. With PN, the risk of error is high because of the
for patients. One event particularly stands out, as it involves
number of components in each formulation.2 Technology
technology yet it can be solved by technology.3 In 2007, a
seeks to decrease the risk of error by accomplishing the
reported error involves a zinc overdose administered via PN.
following: (1) reducing the need for order transcription with
The prescription was for a neonatal PN, with an additive
computer-prescriber order entry (CPOE) and electronic
of zinc correctly written for the dose of 330 mcg per
health record (EHR)-to-compounder interfaces; (2) intro-
100 mL of PN formula. Although the dose was prescribed
ducing a “photo-finish” check for hand-added ingredients
in units of mcg per 100 mL, the automated compounding
with barcode-assisted medication preparation (BCMP) sys-
device (ACD) required zinc to be entered in a mcg/kg dose.
tems; (3) introducing quality-assurance checks with refrac-
The pharmacist made the adjustment; however, when she
tometry and laboratory; and (4) introducing new techniques
entered it into the ACD, she accidentally entered it as a mg
to detect micro-organisms and endotoxin. CPOE also offers
dose, not mcg dose. This resulted in a final concentration
a unique opportunity to alert prescribers to medication
of 330 mg per 100 mL, which is a 1000 times more than
interactions as well as to medication dose adjustments
was prescribed (Figure 1). Although another pharmacist
needed because of changes in weight or renal function.
These clinical decision-making tools can be applied to PN to
alert prescribers to incompatibilities, volume limits, and the Financial disclosure: None declared.
appropriate dosing of ingredients. In summary, technology Conflicts of interests: None declared.
is able to greatly improve the safety of PN prescribing and This article originally appeared online on April 17, 2018.
compounding.
Corresponding Author:
For the use of this article, PN will be differentiated into Caitlin Curtis, PharmD, BCNSP, University of Wisconsin Hospital
on the following 2 groups: 2-in-1 PN and 3-in-1 PN. The and Clinics, Nutrition Support Pharmacist, Department of
terms 2-in-1 PN or 2-in-1s refer to PN that is compounded Pharmacy, 600 Highland Avenue, Madison, WI 53792, USA.
without fat emulsion, and the terms 3-in-1 PN or 3-in-1s Email: ccurtis@uwhealth.org
Curtis 797

including limits for electrolytes such as sodium, calcium,


and phosphate, are programmed into the software for the
ACD. These safeguards are visible to the compounding
pharmacist, but unavailable to the prescriber. It was not
until the advent of EHR-to-compounder interfaces that real
steps for safety could occur. These interfaces allow real-time
safeguards to be built into order entry so that prescribers can
see the limitations of PN mixture compatibility as they are
entering the orders.
A group in Salt Lake City, UT, implemented such an
EHR-to-compounder interface system that set soft-stop
and hard-stop limits as well as education prompts.5 These
limits and education prompts were set for macronutrients,
electrolytes, micronutrients (trace minerals and carnitine).
Figure 1. Intended dose was 330 mcg per 100 mL or 0.33 mg The “soft-limit prompts” allow the prescriber to override
per 100 mL. The actual dose was 330,000 mcg per 100 mL or
them and then allow the verifying pharmacist to review the
330 mg per 100 mL.
override on verification. The verifying pharmacist is then
able to discuss the override with the prescriber and then they
can decide together if the override is reasonable. The “hard-
checked the work, she missed the entry error. The pharmacy
limit prompt” is not able not able to be overridden by the
technician mixing PN with the ACD needed to refill the zinc
prescriber or the pharmacist. For example, in the author’s
syringe on the compounder—for a total of 11 times. A third
institution, the sodium has been preset to a hard limit of
pharmacist checked the PN solution, not knowing about
154 mEq/liter. (This was an institutional decision.)
the 11 syringe refills. The PN infusion was started on the
Recommendations for PN component ordering and la-
neonate. When an oncoming technician arrived to work, the
beling followed American Society for Parenteral and En-
previous technician discussed the odd situation of needing
teral Nutrition guidelines.6-9 As this system was designed
to change the zinc syringe so many times. This technician
for a pediatric hospital, the choices for PNs were rather
quickly realized the error, and alerted the pharmacist, who
elaborate, as the group determined “there were 8 natural
had the PN stopped and took measures to reverse the
breakpoints for weight categories.” This means the provider
zinc overdose with a chelate. However, sadly, the patient
initially chooses the correct PN order based on the weight
perished. The ISMP concluded that this fatal event was due
of the patient and then the PN limits are built into the
to “systems-based causes” that, if corrected, may prevent
order according to each weight category. Each PN weight
another such error from occurring. Could technology have
“category” (weight 0–1.5 kg, 1.6–3 kg, etc.) is different, and
prevented this event? At the end of this section, this event
therefore the soft and hard limits for each weight category
will be revisited to explore which issues could have been
are different as well. After implementation of the system,
prevented by the use of the technologies available today.
the group revealed a rate of medications errors related to
PN of 2.7 per 1000 PN solutions (a 0.27% error rate).
CPOE and EHR-to-Compounder Interfaces The researchers categorized the errors according to the
CPOE and EHR-to-compounder interfaces go hand in following traditional steps of the medication use process:
hand to providing the safest systems for compounding prescribing, transcription, preparation, and administration.
PNs. (Note: During this discussion, it is implied that the Transcription errors were eliminated, and most errors (95%)
electronic medical record contains CPOE.) When EHR is occurred during administration.
implemented, it eliminates many of the errors associated This resulting decrease in errors is impressive. A prospec-
with illegible handwriting and incorrect units (mcg or mg, tive observational study by Sacks et al2 in 2004 (pre-EHR)
etc.). However, many EHRs do not interface with the reported a frequency of 15.6 errors per 1000 PN prescrip-
ACDs used to compound PN. A survey of hospitals in tions or a 1.6% error rate. Most of the errors reported
2014 by Vanek et al4 showed that 28% of hospitals use occurred during the transcription and administration steps
an EHR-to-ACD interface when compounding PN. If the of the medication use process, 39% and 35%, respectively.
EHR does not interface with the ACD, then the EHR The Salt Lake City group eliminated transcription, thus
prints a label with the PN formula in central pharmacy. eliminating a large source of error. This is likely to responsi-
Then, the compounding pharmacist or technician still must ble for the lower error rate when compared with the study
transcribe the PN formula into the ACD software before by Sacks et al.2 The Salt Lake City group also reported
the PN is compounded. This step allows for transcription that 95% of the errors after the interface were related to
errors. Also, many of the safeguards for compounding PN, administration errors. This is similar to the report by Sacks
798 Nutrition in Clinical Practice 33(6)

et al,2 as they reported that transcription and administration the finished sterile product. For instance, if insulin is added
were the steps that were most prone to error. to a PN, then the technician takes the following actions:
Of course, there are some challenges that come with (1) picks the insulin vial; (2) scans the insulin vial; (3) draws
interfaces. For the interface to work properly, there must be up the correct insulin dose; (4) takes a digital photo of
robust technical support in the setting of product shortages the label, the insulin vial, and the syringe drawn up to
and formulary additions. It is also important that the the correct dose; (5) adds the insulin to the PN. Then the
verifying pharmacist role remains intact to review the PN pharmacist can check in the system and per the digital
order for appropriateness regarding age, weight, disease photos that the correct strength of insulin was picked and
state, and other medications (correct dose in PN, inter- the correct dose of insulin was drawn up. With this system,
actions/therapeutic duplication, and Y-site compatibility). there is extra assurance that the correct products are picked
Also, there is concern is regarding downtime and emergency and within expiration because the technician is required to
procedures. What happens when the EHR goes down or if scan the barcodes before sterile product preparation. Also,
there is a power outage and all orders must be converted to there is more assurance that correct doses are added because
paper—the limits are then not available to guide prescribers. the pharmacist can see a photo of the dose before it is added
These are all challenges to consider when implementing an instead of seeing what the technician thought he/she added
EHR-to-ACD interface. after the fact.
Improvement in error rate by implementation of a
BCMP system is well documented. Speth et al10 first studied
BCMP Systems the system in 2009 and showed that after 2 years of
Although ACDs mix 90%–95% of the ingredients contained implementation, their error detection rate was 0.8%. The
in PN, there are some components that are, or should be,3 researchers compared the error rate with the BCMP system
added by hand. These components, such as insulin, thiamin, of 0.8% to a previously reported error rate of a traditional
zinc, selenium, copper, folic acid, pyridoxine, and so on, paper system of 9%11 and concluded that the BCMP
are too small in volume or weight to be measured by the system results in fewer preparation errors. Of the barcode-
ACD and must be drawn up and added manually. Because intercepted errors, 60% were due to the technician scanning
pharmacy technicians usually draw these components up, the wrong drug, 28% due to the wrong fluid or diluent,
a pharmacist must check them. Traditionally, the “pull- and 12% due to the wrong concentration of drug. Of the
down” method is used, whereas the pharmacy technician pharmacist-intercepted errors, 75% were due to an incorrect
adds the medication or component to the PN and then pulls amount of drug or diluent, which were errors that would not
the syringe plunger “down” to the dose that was added. The have been detected in the traditional paper system. The rest
technician then includes this syringe and the medication vial of pharmacist-intercepted errors were attributed to unclear
in with the bin for the pharmacist to check. This method is digital photos or due to an improper sterile technique.
prone to error because the pharmacist is not 100% certain Another advantage that these authors identified was the
that the medication is added in the dose that was “pulled decrease in medication turnaround times. Turnaround times
down” on the syringe.10 decreased for new orders, stat orders, and chemotherapy
The new BCMP system allows for safer compounding. orders. The most dramatic decrease was for chemotherapy
When an intravenous medication is compounded, the phar- orders from 52 minutes prior to BCMP down to 34 minutes
macy technician scans the label into the system. The BCMP after BCMP implementation. The researchers posit this is
system will show, on a computer screen, the intravenous due to the following aspects of the BCMP system: (1) the
fluid, medication, and dose of each (if applicable) needed system notifies the pharmacist when a sterile product is
to compound the finished product. The technician will then finished and ready to be checked, (2) the pharmacist can
pick and scan each component into the system. The BCMP check the sterile product remotely by viewing the digital
system will detect if the technician has chosen the incorrect photos on a personal computer, (3) many errors are detected
medication, diluent, concentration, or expired medication, and intercepted prior to mixing (i.e., when the technician
and will not allow the technician to proceed to the next step picks and scans the incorrect product), and wasted doses are
in the system until the technician scans the correct products. greatly reduced. The researchers reported a waste reduction
Then the technician draws the amount of medication from of approximately $30,000 during the first 12 months of
the vial into a syringe and the system takes a digital photo implementation. They attribute this to avoidance of errors
of all the components, including the label, medication, as well as due to the system’s ability to track doses discarded
intravenous fluid, reconstituted medication (if applicable), after they have past the beyond-use date.
and “drawn up dose.” The technician then compounds the Moniz et al12 implemented a BCMP system in their
sterile product, and the system takes another digital photo hospital pharmacy and collected data regarding the number
of the finished preparation. The pharmacist is then able to of doses prepared in the pharmacy, the number of errors
check each digital photo to observe what is actually added to detected by pharmacists, and the number of reworked
Curtis 799

or rejected doses.12 The time of data collection was 5 only” additions of low-volume ingredients. Although this
months. The errors were reviewed for error detectability is a low-tech fix, it leads to a high-tech fix. If the zinc is
(would the errors have been detected in the traditional added as a manual ingredient, then it can go through the
system?) and were also scored for harm. Using the BCMP BCMP workflow. Even if all of the other systems had failed,
system, 425,683 medication doses were prepared during the the technician would then have scanned the zinc and the 11
13-month study period. The error-detection rate by the syringes and taken digital photos. Hopefully, when seeing
entire system (barcode and pharmacist final checks) was the 11 syringes full of zinc, the pharmacist would have
0.68%, and the number of reworked or rejected doses was known something was wrong at that point! Regardless, this
1223 and 1667, respectively. Of the rejected doses, 375 doses example illustrates how technology could have halted this
(22.4%) involved errors that were deemed “undetectable” error before it reached the patient.
before implementation of the BCMP—that is, the pharma-
cist would not have been able to detect these errors due to the
lack of the digital photos they now had at their disposal with Quality Assurance
BCMP. The other rejected doses involved errors deemed to
be detectable with the traditional system (prior to BCMP
Refractometry
system implementation) or errors introduced by the BCMP Maintaining quality assurance of ACDs is recommended
system. Of the “undetectable” errors, 21 were judged to by United States Pharmacopeia (USP) Chapter <797>
be of a “severe harm” severity. The error rate with the as well as by the American Society for Hospital Phar-
BCMP system of 0.68% was compared with a previously macists and the American Society for Parenteral and
reported error rate of a traditional paper system of 9%.11 Enteral Nutrition.9,14,15 In USP Chapter <797>, quality
The researchers concluded that the BCMP system results in assurance can be accomplished by way of gravimetric or
fewer preparation errors. volumetric tests. The chapter also suggests that “additional
Deng et al13 also implemented a BCMP in their pediatric tests of accuracy may be employed that determine the
hospital pharmacy and had similar results. After a year content of certain ingredients in the final volume of the
of data collection, 421,730 doses were analyzed for errors. PN admixture.”14 Some hospitals are using refractometry
The error detection rate was 0.74%. Of the 3101 errors, the or refractive index (RI) to aid in the quality assurance
barcode scanning technology detected 2241 (72.27%) of of PN formulations. However, refractometry can only be
the errors before the pharmacist inspected the product on used for 2-in-1 PN formulations, that is, non-fat-containing
the digital photos. The pharmacist was needed to detect the formulations. In the following 2 articles, pharmacists or
remainder of the errors (27.73%). nurses use a predictive equation to calculate refractive index
All of these examples of BCMP were employed in and then compare it to the refractive index measured by a
hospital pharmacies that compound sterile products. None small amount of PN formula dropped onto a refractometer
of these articles specifically mention data regarding the use and then compare the 2 values. If the provider finds that the
of BCMP in the compounding of PN. However, BCMP can values do not match (within a standard deviation), then the
be applied to compounding PNs, as it is currently used in provider asks the pharmacy to compound the PN again.
the author’s institution. Refractive index of a substance is “the ratio of the
velocity of light in air to the velocity of light in the sub-
stance.” Refractive index increases with the concentration
Systems-Based Errors? of solutes in solution. Refractive index has been used to
Returning to the fatal error in compounding3 described verify concentrations of enteral nutrition and drugs.16-18
earlier, how would the aforementioned technologies helped Chang and Yeh19 devised an equation to predict refractive
to prevent them? For the zinc-overdose event, the first error index for 2-in-1 PN formulations and validated the equation
was a paper–ACD dose mismatch, as zinc was prescribed with 500 prepared PNs. Chang and Yeh19 tested 4 predictive
as 330 mcg per 100 mL, but the ACD required the dose equations and compared them with the values measured
to be in a mcg/kg dose. This error would have been solved by the refractometer. They used a hand-held refractometer,
by both the CPOE and EHR-to-compounder interfaces, which required 1 or 2 drops of the PN formulation for
as the ACD requires all doses to be entered correctly. measurement. Of the predictive equations, the first 3 were
The next was a pharmacist calculation error, resulting in variations based on concentrations of the PN ingredients
a concentration of 330 mg per 100 mL. If a prescriber (equation 1 = amino acids (g/L) × 8 + glucose g/L × 7 +
entered this in error into a system with limits, the prescriber sodium (mEq/L) × 2 + phosphate (mg/L) × 0.2 − 50), and
would not be able to continue and the PN would not be equation 4 used Brix values (equation 4 = 81.05 × Brix value
mixed. Next, the technician mixing the PN refilled the zinc − 116.33). Brix values are the same as measured refractive
syringe in the compounder a total of 11 times. The safe index. Chang and Yeh19 used Brix values, measured by their
practice recommendation from ISMP is to “allow manual- refractometer, of known mixes of amino acid +dextrose
800 Nutrition in Clinical Practice 33(6)

concentrations (eg, amino acid 4%, dextrose 6%, Brix Sterility Detection
value reported is 11.8 ± 0.1) Of the 500 PNs, the authors
found a very high correlation of equation 4 with measured Keeping PN sterile using aseptic technique is essential
refractometry. Equation 4, which uses a Brix value, was in delivering safe and effective nutrition support.9,15 A
successful in predicting 100% of the refractive index for painful consequence of interrupting sterile procedure is
PNs within a measured osmolality range of 600 to 900 the outbreak of Serratia marcescens infection due to the
mOsm/kg for a CI range of 90%–110%. The authors were contaminated bags of PN in 2011.21 As a result of the out-
confident in using the predictive equation and hand-held break, 9 patients died. Although the pharmacy providing
refractometer as a quality assurance tool to check PNs for PN in this example was checking for contamination and
compounding accuracy. The authors state that limitations endotoxin, the sample size was too small to detect bacterial
of using refractometry as a quality assurance method is the growth and the results were not available until after the PNs
time and labor needed to calculate predictive equations and were administered. New technologies are emerging to aid
use the refractometer on the final PN formula. in detecting bacterial and endotoxin contamination much
Nelson et al20 also derived and validated an equation sooner so that this tragedy is not repeated.
for refractive index. Nelson et al20 used a refractometer Traditional sterility testing for compounded sterile prod-
of a “reported accuracy of 0.00004” in an environment ucts required by USP <797> is performed by taking a sam-
controlled for temperature, as recommended by USP and ple of the compounded sterile product, passing it through
the manufacturer of the refractometer. They first measured at 0.45 μm filter, and dividing the filter into 2 parts.14
a refractive index for every ingredient used in the PN formu- One part of the filter is transferred to tryptic soy broth
lations compounded at their institution. They found that the medium so as to grow aerobic microorganisms, and the
ingredient indices were additive; that is, if 1 mL of sodium other part is transferred to fluid thioglycollate medium so as
chloride 2 mEq/mL had a refractive index of 1, then 2 mL of to grow both aerobic and anaerobic microorganisms. Both
sodium chloride 2 mEq/mL had a refractive index of 2. The media solutions are stored at the appropriate temperatures
equation they tested for the refractive index of PN formula- (defined by USP) and incubated for 14 days. Of course, this
tions, when 1.333 is the RI of sterile water, was the following: type of testing is not feasible for PN for several reasons.
RIfinal = 1.333 + ࢣ(RIconcentrate [volume of concentrate First, PN would need to be administered before the results
(mL)/total volume of PN solution (mL)]). They validated from testing returned, given the beyond-use date for PN
this equation for refractive index on 1057 pediatric PN stored at room temperature (20–25°C) is 24 hours and
solutions. Of the 1057 PNs, 99.8% of the refractometer at refrigerated temperature (2–8°C) is 9 days. (If the PN
measurements fell within the acceptable range (within 2 is stored at refrigerated temperature, the beyond-use date
standard deviations) of the predictive RI. The authors note is 9 days only if a multivitamin has not been added to
that limitations of the refractometer include cost, personnel the PN.). Also, this method would only be feasible for
training, and that the main factors that drive refractive index 2-in-1 PNs, as 3-in-1 PNs cannot be filtered through a
are concentrations of dextrose and amino acids. Electrolyte 0.45 μm filter. A faster detection method is available and
changes are not readily picked up by refractometry. The based on the detection of carbon dioxide production using
authors now use refractometry testing in their pharmacy colorimetric methods.22 This system uses a 10 mL sample,
routinely for the first and last PN bag compounded each incubates the microorganisms, shakes the samples, and takes
day, as well as after each time the dextrose bag is changed on measurements every 10 minutes. This system can detect
the ACD. The authors state this reduced their error rate to growth of certain microorganisms within 24 hours and is
“2 compounding errors in every 10,000” PNs compounded; approved by the U.S. Food and Drug Administration for
however, they did not list the error rate before the refractom- detecting microorganisms in blood and body fluids. The
etry test was initiated. system has limitations, however, as it is not approved by
the USP for verifying sterility in pharmaceuticals. Because
it uses a 10 mL sample that is not filtered, it may be
Laboratory Testing
appropriate for testing both 2-in-1 and 3-in-1 formulations.
PN formulations may be tested by an institution’s laboratory It is used to test blood and body fluids; however, there are
for quality assurance.14 Per USP Chapter <797>, this test no examples in the literature of this system’s use in testing
may only be used for quality assurance if the institution emulsions.
standardizes its methods to USP references and procedures. A brand of this system, the BacT/Alert automated
Laboratories are accustomed and calibrated to test blood system (bioMérieux, Durham, USA) has been used in South
and body fluids. Thus if the laboratory is to test a PN, Africa to detect microorganisms in PN.23 In this instance,
the laboratory may need to validate to the high range of 2-in-1 PNs are commercially mixed, irradiated, and sent to
dextrose that is typically found in a PN, instead of the range the National Health Laboratory Services for testing. The
found in blood. BacT/Alert system was used to monitor the pre-irradiated
Curtis 801

PNs for microorganisms in addition to soy broth. The for a specific target. However, both antibody sensors and
system isolated Candida parapsilosis, which was further cul- aptamers are too costly and labor intensive to use for testing
tured. After further investigation, this organism was isolated PN on a daily basis. These tests are all in their infancy, and
from work stations within the pharmacy. This report shows it is also too early to know if they might be used for 2-in-1,
that the BacT/Alert system is able to detect microorganisms 3-in-1, or both kinds of PNs.
in PN solutions; however, many important barriers prevent
this system from being implemented in daily use. First, PNs Summary
in this situation were being commercially produced, which
In conclusion, technology has solved many issues related to
means that the PNs presumably had a long shelf life. The
compounding PN, including transcription errors and direct-
BacT/Alert system may be useful when the end product
to-prescriber safeguards. However, more challenges remain,
has a shelf life of 6 months or more, but perhaps not
including the need for back-up systems in the event of EHR
useful if the product needs to be used immediately. However,
failures or electric outages. Also, there is much opportunity
the BacT/Alert system might have a future role in home-
for wider installation of EHR-to-compounder systems in
infusion pharmacies. As home-infusion pharmacies batch
the coming years.
PNs then refrigerate them (beyond-use date is 9 days), in
With regard to quality assurance, refractometry has been
the future pharmacists might be able to use results from
used in some hospitals as an additional quality assurance
the BacT/Alert system in time to dispose of a contaminated
check, but the cost of and equipment and training may
batch. There is currently no data available that tests the use
prohibit widespread application of use. Laboratory checks
of BacT/Alert system in home-infusion pharmacies for PN
of PN for quality assurance checks are available in select
sterility, although this is an opportunity for future research.
hospitals, if the laboratory has the equipment and is able
to calibrate per USP Chapter <797> requirements. The
Endotoxin Detection availability and use of this method of as a quality assurance
check would be an opportunity for future research.
Currently, PN is not tested for endotoxin. However, as
Although the faster detection of microorganisms is
many components are added to the PN, it is possible that
available, it is not approved for testing of sterility of
endotoxins are introduced. If a quick test for endotoxin
compounded medications. Endotoxin testing remains an
was available, it would be desirable to use for PN to
expensive and labor-intensive endeavor. There is much
prevent avoidable fevers in the patient. The oldest endotoxin
opportunity for future research in sterility and endotoxin
detection approved by the USP is the rabbit pyrogen test
testing of PN formulations. Technology is advancing expo-
based on the rise in temperature of rabbits after injection
nentially faster, and further research may address all of these
of a solution containing endotoxin.24 This method has
challenges more quickly than expected.
slowly been replaced by a more scientific and feasible test,
the limulus amoebocyte lysate test. This test is based on a
substance derived from the blood of the horseshoe crab. Statement of Authorship
The substance, limuluspolyphemus, reacts in an enzymatic C. Curtis contributed to conception/design of the research;
process with endotoxin and visibly clots when the endotoxin contributed to acquisition, analysis, or interpretation of
reaches a certain concentration. However, due to the enzy- the data; drafted the manuscript; critically revised the
matic nature of the process, some forms of glucose have manuscript; and agrees to be fully accountable for ensuring
activated limulus amoebocyte lysate . Therefore, it may not the integrity and accuracy of the work.
be suitable to test PN for endotoxin.
Newer ways of detection of endotoxin are on the fore-
front of technology. One way is the use of CD14 to bind References
endotoxin. Through the use of recombinant CD14 and 1. Institute for Safe Medication Practices. ISMP’s list of high-alert med-
the development of an electrochemilumninescent assay, it ications. 2017. http://www.ismp.org/Tools/highalertmedications.pdf.
is possible to detect endotoxin. However, many challenges Accessed September 26,2017.
2. Sacks GS, Rough S, Kudsk KA. Frequency and severity of harm of
exist with this detection system, including the tendency of
medication errors related to the parenteral nutrition process in a large
CD14 to bind to a range of molecules other than endotoxin. university teaching hospital. Pharmacotherapy. 2009;29(8):966-974.
Other ways to detect endotoxin can be specific and sensitive, 3. Institute for Safe Medication Practices. Fatal 1,000-fold overdoses
such as the use of antibody sensors, similar to the use can occur, particularly in neonates, by transposing mcg and mg.
of immunoassay. This method has been used to develop https://www.ismp.org/newsletters/acutecare/articles/20070906.asp. Ac-
cessed October 31, 2017.
monoclonal antibodies specific and sensitive to endotoxin.
4. Vanek VW, Ayers P, Charney P, et al. Follow-up survey on function-
Aptamers are also highly specific and sensitive to detecting ality of nutrition documentation and ordering nutrition therapy in
endotoxin, even in miniscule amounts. Aptamers are 3- currently available electronic health record systems. Nutr Clin Pract.
dimensional molecules designed from deoxyribonucleic acid 2016;31(3):401-415.
802 Nutrition in Clinical Practice 33(6)

5. MacKay M, Anderson C, Boehme S, Cash J, Zobell J. Frequency 15. American Society of Health-System Pharmacists. ASHP guidelines
and severity of parenteral nutrition medication errors at a large on the safe use of automated compounding devices for the prepa-
children’s hospital after implementation of electronic ordering and ration of parenteral nutrition admixtures. Am J Health-Syst Pharm.
compounding. Nutr Clin Pract. 2016;31(2):195-206. 2000;57:1343-1348.
6. Boullata JI, Gilbert K, Sacks G, et al. A.S.P.E.N. clinical guidelines: 16. Cheung JF, Chong S, Kitrenos JG. Use of refractometer to detect
parenteral nutrition ordering, order review, compounding, labeling, controlled substance tampering. Am J Hosp Pharm. 1991;48:1488-
and dispensing. JPEN J Parenter Enteral Nutr. 2014;38(3)334-377. 1492.
7. Druyan ME, Compher C, Boullata JI, et al. Clinical guidelines for the 17. Frankenfield DL, Johnson RE. Refractometry of controlled sub-
use of parenteral and enteral nutrition in adult and pediatric patients: stances. Am J Hosp Pharm. 1991;48:2129-2130.
applying the GRADE system to development of A.S.P.E.N. clinical 18. Chang WK, Chen MZ, Chao YC. Use of the refractometer as a tool
guidelines. JPEN J Parenter Enteral Nutr. 2012;36(1)77-80. to monitor dietary formula concentration in gastric juice. Clin Nutr.
8. Ayers P, Adams S, Boullata J, et al. A.S.P.E.N. parenteral nutrition 2002;21:521-525.
safety consensus recommendations. JPEN J Parenter Enteral Nutr. 19. Chang WK, Yeh MK. Prediction of parenteral nutrition osmolarity
2014;38(3):296-333. by digital refractometry. JPEN J Parenter Enteral Nutr. 2011;35:412-
9. Mirtallo J, Canada T, Johnson D, et al. Safe practices for parenteral 418.
nutrition. JPEN J Parenter Enteral Nutr. 2004;28(6)S39-S70. 20. Nelson S, Barrows J, Haftmann R, Helm M, MacKay M. Calculating
10. Speth SL, Fields DB, Schlemmer CB, Harrison C. Optimizing i.v. the refractive index for pediatric parenteral nutrient solutions. Am J
workflow. Am J Health-Syst Pharm. 2013;70:2076-2080. Health-Syt Pharm. 2013;70:350-355.
11. Flynn EA, Pearson RE, Barker K. Observational study of accuracy 21. Gupta N, Hocevar S, Moulton-Meissner H, et al. Outbreak of Serratia
in compounding i.v. admixtures in five hospitals. Am J Health-Syst marcescens bloodstream infections in patients receiving parenteral
Pharm. 1997;54:904-912. nutrition prepared by a compounding pharmacy. Clin Infect Dis.
12. Moniz TT, Chu S, Tom C, et al. Sterile product compounding using an 2014;59:1-8.
i.v. compounding workflow management system at a pediatric hospital. 22. Kaiser SJ, Mutters N, Backhaus J, et al. Sterility testing of in-
Am J Health-Syst Pharm. 2014;71:1311-1317. jectable products: Evaluation of the growth-based BacT/ALERT 3D
13. Deng Y, Lin AC, Hing J, et al. Risk factors for i.v. compounding errors dual T culture system. PDA J Pharm Sci and Tech. 2016;70:568-
when using an automated workflow management system. Am J Health- 576.
Syst Pharm. 2016;73:887-893. 23. Marais E, Stewart R, Dusé AG. Candida parapsilosis detected in TPN
14. Pharmaceutical compounding—sterile preparations (chapter <797>). using the BacT/Alert system and characterized by randomly amplified
In: The United States Pharmacopeia, 38th Rev., and the National polymorphic DNA. J Hosp Infect. 2004;56:291-296.
Formulary, 33nd ed. Rockville, MD: United States Pharmacopeial 24. Su W, Ding X. Methods of endotoxin detection. J Lab Autom.
Convention, 2015:43-87. 2015;20:354-364.
Review

Nutrition in Clinical Practice


Volume 33 Number 6
Effect of Early vs Late Start of Oral Intake on Anastomotic December 2018 803–812

C 2017 American Society for

Leakage Following Elective Lower Intestinal Surgery: Parenteral and Enteral Nutrition
DOI: 10.1177/0884533617711128
A Systematic Review wileyonlinelibrary.com

Boudewijn J. J. Smeets, MD1,2 ; Emmeline G. Peters, MD1,3 ;


Eelco C. J. Horsten, BSc1 ; Teus J. Weijs, PhD1 ; Harm J. T. Rutten, PhD1,2 ;
Willem A. Buurman, PhD4 ; Wouter J. de Jonge, PhD3 ; and Misha D. P. Luyer, PhD1

Abstract
Background: Experimental and clinical studies have demonstrated a beneficial effect of early enteral nutrition (EN) on anastomotic
leakage following colorectal surgery. Early oral intake is a common form of early EN with various clinical benefits, but the effect
on anastomotic leakage is unclear. This systematic review investigates the effect of early vs late start of oral intake on anastomotic
leakage following lower intestinal surgery. Methods: A systematic literature search was performed using the PubMed, Embase,
Medline, and Cochrane databases. Randomized controlled trials were included that compared early (within 24 hours) vs late start
of oral intake following elective surgery of the small bowel, colon, or rectum. Meta-analysis was performed for anastomotic leakage,
overall complications, length of stay, and mortality. Sensitivity analysis was performed in which studies of inferior methodological
quality were excluded. Results: Nine studies including 879 patients met eligibility criteria. Early start of oral intake significantly
reduced overall complications (odds ratio [OR], 0.65; 95% confidence interval [CI], 0.46–0.93; P = .02), length of stay (mean
difference, −0.89; 95% CI, −1.22 to −0.57; P < .001), and anastomotic leakage (OR, 0.40; 95% CI, 0.17–0.95; P = .04) compared
with late start of oral intake. However, in the sensitivity analysis only the overall reduction of length of stay remained significant.
Conclusion: The effect of early oral intake on anastomotic leakage is unclear as existing studies are heterogeneous and at risk of bias.
High-quality studies are needed to study the potential benefit of EN on anastomotic healing. (Nutr Clin Pract. 2018;33:803–812)

Keywords
enteral nutrition; surgery; wound healing; nutritional support; colorectal surgery; anastomotic leak; length of stay; mortality; meta-
analysis

Anastomotic leakage (AL) is a severe complication fol- systematic reviews included various types of GI surgery or
lowing colorectal surgery as it is associated with increased included studies that combined other elements of fast-track
morbidity, mortality, and cancer recurrence rates.1-4 Despite protocols in the intervention group but not in the control
ongoing efforts, strategies that effectively reduce AL are group.15-18 To further investigate the potential beneficial
lacking and the incidence has remained stable over the effects of EN on anastomotic healing, this systematic review
years.5
Several experimental studies have demonstrated that en-
teral nutrition (EN) may improve anastomotic healing.6-12 From the 1 Department of Surgery, Catharina Hospital, Eindhoven,
Moreover, in 2 recent randomized controlled trials, AL the Netherlands; 2 GROW School of Oncology and Developmental
following colorectal surgery was significantly reduced by Biology, Maastricht University, Maastricht, the Netherlands; 3 Tytgat
Institute for Intestinal and Liver Research, Academic Medical Center,
means of early postoperative EN13 and direct perioperative Amsterdam, the Netherlands; and the 4 School for Mental Health and
sham feeding.14 Taken together, these results suggest that Neuroscience, Maastricht University, Maastricht, Limburg, the
EN administered close to surgery may provide new thera- Netherlands.
peutic opportunities to reduce AL. Financial disclosure: None declared.
A common method to provide EN close to surgery Conflicts of interest: None declared.
is the early postoperative start of oral intake. Systematic
This article originally appeared online on June 19, 2017.
reviews on the effects of early start of oral intake following
gastrointestinal (GI) surgery have demonstrated clear ben- Corresponding Author:
Boudewijn J. J. Smeets, MD, Department of Surgery, Catharina
efits, including a reduction in length of stay (LOS), overall
Hospital Eindhoven, Michelangelolaan 2, Eindhoven, Noord-Brabant
complications, and mortality.15-18 However, in these reviews, 5623 EJ, the Netherlands.
early oral intake did not affect AL.15-18 Importantly, these Email: boudewijn.smeets@catharinaziekenhuis.nl
804 Nutrition in Clinical Practice 33(6)

compares the effect of early vs late start of oral intake on AL Data Extraction and Outcomes
following elective lower intestinal surgery.
The relevant published data were collected in pilot-tested
Methods tables. Extracted information from each study included (1)
study information, including name of first author, year of
This systematic review was performed according to publication, number of participants in each group, and
the Cochrane Handbook for Systematic Reviews for reported outcomes; (2) patient information, including type
Interventions19 and the Preferred Reporting Items for of surgery, disease, sex, age, and perioperative protocols
Systematic Reviews and Meta-Analyses (PRISMA) used affecting AL21 ; and (3) postoperative feeding proto-
statement guidelines.20 The entire review process (ie, article cols. Furthermore, we extracted data on the incidence and
search, critical appraisal, data extraction, and analysis) was definition of AL, overall complications, mortality, and LOS.
conducted by 3 independent researchers (B.J.J.S., E.G.P.,
and E.C.J.H.). Any disagreements were resolved through
discussion.
Risk of Bias in Individual Studies
All studies included in the review were investigated for risk
Eligibility Criteria of bias with the Cochrane collaboration’s tool for assessing
risk of bias.19 Risk of bias was assessed on the follow-
We included only randomized controlled clinical trials that
ing items: randomization method, allocation concealment,
reported on the effects of early vs late start of oral intake
blinding of participants and personnel, blinding of outcome
on AL following elective surgery of the small bowel, colon,
assessment, incomplete outcome data, selective reporting,
or rectum in patients aged ࣙ18 years. We defined early start
and any other item in study design.
of oral intake as any caloric intake started within 24 hours
after surgery. Late start of oral intake was defined as nil by
mouth until resolution of postoperative ileus (ie, passage of Data Analysis
flatus or stool without presence of nausea or vomiting). To
Meta-analysis was performed for AL, overall complications,
assess the true effect of early oral intake alone, we excluded
LOS, and mortality. We performed data analysis using
studies that combined early oral intake with other elements
Review Manager Software version 5.3 as recommended
of fast-track protocols in the intervention group but not in
by the Cochrane Handbook for Systematic Reviews for
the control group.
Interventions.19,22 Dichotomous results were analyzed using
the random-effects model in the Mantel-Haenszel method
Information Sources and Search Strategy and are presented as odds ratio (ORs) with corresponding
The PubMed, Medline, Embase, and Cochrane databases 95% confidence intervals (CIs). Continuous results were an-
were systematically searched. The search strategy combined alyzed using the inverse variance method and are presented
all synonyms regarding the intervention “early oral intake” as mean difference (MD) with corresponding 95% CI. A P
and the domain “lower intestinal surgery” with the Boolean value <.05 was considered to be statistically significant. We
operator “AND.” All synonyms were combined with the assessed presence and amount of statistical heterogeneity
Boolean operator “OR.” An example set of search terms using the I2 statistic. Furthermore, a sensitivity analysis
is provided in Supplementary Table S1. We tested the was performed in which studies of inferior methodological
sensitivity of the search strategy by screening all references quality were excluded.
of included articles for relevant publications that were not
retrieved in the initial search. Furthermore, we screened Results
all citing articles and related articles using Web of Science
version 5.15.1. Identification of additional eligible articles Description of Studies
led to evaluation and improvement of the search strategy
Figure 1 presents the search results and study selection
until it retrieved all eligible articles. We contacted authors
process. The search revealed 80 potentially relevant stud-
by email if articles were not available in full text. The search
ies, of which 9 randomized controlled trials fitted in-
was updated until September 28, 2016.
clusion criteria. Study characteristics are shown in Ta-
ble 1. Types of surgical procedures were evenly matched
Study Selection between groups in all studies. Laparoscopic surgery was
Three authors screened all records on title and abstract. performed only in 1 study23 ; in other studies, open surgery
Records were excluded if they clearly did not address the was performed.24-31 When reported, there was substantial
domain and intervention under investigation. The remain- heterogeneity between studies in the use of perioperative
ing articles were screened in full text. Only articles fulfilling protocols, including use of epidural anesthesia, preopera-
all eligibility criteria were included. tive bowel lavage, nonsteroid anti-inflammatory drugs, and
Smeets et al 805

Figure 1. Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) flowchart for search results and study
selection process.

opioids. Postoperative feeding protocols are described in start of oral intake significantly reduced AL compared with
Table 2. late start of oral intake (OR, 0.40; 95% CI, 0.17–0.95; P =
AL was a rare event in most studies and rarely a .04). However, when trials with risk of bias were excluded
primary outcome. Four studies provided definitions for AL in the sensitivity analysis, the overall effect on AL was no
(Table 3).25,26,28,31 longer significant (OR, 0.39; 95% CI, 0.09–1.76; P = .22)
(Figure 2B).
Methodological Quality of Included Studies
Due to the nature of the intervention, blinding could not Overall Complications
be applied in any study. Three studies gave no explicit
All studies provided data on overall complications. As
description of the randomization method, resulting in
shown in Figure 3A, early start of oral intake significantly
an unclear risk of bias.26,27,30 One open-label study used
reduced overall complications compared with late start
blocked randomization with a fixed block size of 6; hence,
of oral intake (OR, 0.65; 95% CI, 0.46–0.93; P = .02).
risk of selection bias was present.29 In the same study, it was
However, when trials with risk of bias were excluded in the
unclear whether the nasogastric tube was postoperatively
sensitivity analysis, the overall effect on overall complica-
removed in both groups at similar time points. In 1 study,28
tions was no longer significant (OR, 0.74; 95% CI, 0.42–
patients allocated to delayed start of oral intake had a
1.31; P = .30) (Figure 3B).
significantly greater amount of intraoperative blood loss
(early oral intake median 300 mL vs delayed oral intake
median 800 mL, P = .002), which is a known risk factor for LOS
AL.21 The summary of risk of bias assessment is shown in
Seven studies provided data on LOS as mean ± standard
Table 4. Overall, 3 studies were identified to have the best
deviation and were entered in the meta-analysis.23-26,28-30 As
available methodological quality and were entered in the
shown in Figure 4A, early start of oral intake significantly
sensitivity analysis.23,24,31
reduced LOS compared with late start of oral intake (MD,
−0.89; 95% CI, −1.22 to −0.57; P < .001). Excluding
AL studies with risk of bias increased the overall effect of
First, all studies were entered in the meta-analysis regardless LOS (MD, −3.47; 95% CI, −4.73 to −2.21; P < .001)
of methodological quality. As shown in Figure 2A, early (Figure 4B).
806
Table 1. Study Characteristics.a

Mean Age, y Sex (Male/Female), No.

Author and Publication Early Oral Late Oral Early Oral Late Oral
Year Site of Surgery Disease Intake Intake Intake Intake Reported Postoperative Outcomes

da Fonseca et al,23 2011 Colon NR 57 52 8/16 10/16 LOS, recovery of GI function, overall
morbidity, anastomotic leakage,
readmission, surgical
reintervention after discharge,
mortality
Dag et al,24 2011 Colon, rectum Neoplasia 62 61 52/47 61/39 LOS, recovery of GI function,
complications
El Nakeeb et al,25 2009 Colon, rectum Neoplasia 52 56 39/21 42/18 LOS, recovery of GI function,
complications, readmission,
mortality
Hartsell et al,26 1997 Colon, rectum Neoplasia, diverticular 66 68 NR NR LOS, recovery of GI function,
disease, inflammatory complications
bowel disease
Lucha et al,27 2005 Colon, rectum NR 51 51 17/9 16/9 LOS, recovery of GI function,
aspiration pneumonia,
anastomotic leakage, antiemetic
medication, admission costs
Minig et al,28 2009 Small bowel, Gynecological neoplasia 54 58 0/18 0/22 LOS, recovery of GI function,
colon, rectum complications, intensity of
abdominal pain, patient
satisfaction level, quality of life,
antiemetic and analgesic
medication
Pragatheeswarane et al,29 Small bowel, Neoplasia, familial 47 47 33/27 32/28 LOS, recovery of GI function,
2014 colon, rectum adenomatous complications
polyposis, stricture
Reissman et al,30 1995 Small bowel, NR 51 56 34/46 43/38 LOS, recovery of GI function,
colon, rectum complications
Stewart et al,31 1998 Colon, rectum NR 58 59 19/21 18/22 LOS, recovery of GI function,
complications, antiemetic and
analgesic medication, mobilization

GI, gastrointestinal; LOS, length of stay; NR, not reported.


a All values are presented as patient numbers unless indicated otherwise.
Smeets et al 807

Table 2. Postoperative Feeding Protocols. Table 3. Definitions for Anastomotic Leakage.

Author and Late Oral Author and Publication


Publication Year Early Oral Intake Intake Year Definition

da Fonseca et al,23 POD1 oral liquid diet, SC El Nakeeb et al,25 2009 Symptoms such as fever and
2011 advance to regular diet leakage of intestinal contents
within 24 hours as Hartsell et al,26 1997 Resulting in sepsis and eventual
tolerated death
Dag et al,24 2011 12 hours postoperatively, SC Minig et al,28 2009 Requiring surgical reexploration
start fluids, advance to Stewart et al,31 1998 Fecal discharge from drain tube,
solid diet as tolerated which settled without
El Nakeeb et al,25 POD1 fluids, advance to SC intervention
2009 regular diet within
24–48 hours as
tolerated
Hartsell et al,26 1997 POD1 full liquid diet, SC Experimental studies have suggested that EN can im-
advance to regular diet
prove anastomotic healing via several mechanisms.6-12
if >1000 mL was
consumed within 24 These experimental findings may be corroborated by 2
hours randomized clinical trials of good methodological qual-
Lucha et al,27 2005 8 hours after surgery, SC ity that demonstrated a reduction of AL by means of
start regular diet perioperative sham feeding14 (ie, gum-chewing) and early
Minig et al,28 2009 POD0 CL, advance to SC postoperative enteral tube feeding.13 Early start of oral
regular diet on POD1 intake is a more common form of early EN and has
as tolerated
been extensively described as part of fast-track protocols
Pragatheeswarane et POD1 CL, advance to SC
al,29 2014 full fluid diet within 48 in colorectal surgery. Individual randomized trials have
hours, start solid diet described no effect of early oral intake on AL, but most
over next 24 hours studies had a relatively small sample size and were there-
Reissman et al,30 1995 POD1 CL, advance to SC fore inadequately powered to detect a potential effect on
regular diet as AL.23-31 Previous systematic reviews on early EN also did
tolerated not support an effect on AL.15-18 However, these reviews
Stewart et al,31 1998 4 hours postoperatively, SC
may have been inadequate to assess the true effect of EN,
start free fluids,
advance to solid diet since they included studies with patients undergoing upper
as tolerated on POD1 GI surgery,32 studies that applied immunonutrition,33 or
studies that applied other aspects of fast-track protocols
CL, clear liquids; POD, postoperative day; SC, standard care (nil by only in the treatment group but not in the control group (eg,
mouth until resolution of ileus).
early nasogastric tube removal).34 Furthermore, no review
performed a sensitivity analysis to minimize the risk of
bias.15-18 This study therefore aimed to provide an update
of the available literature and to perform a rigorous critical
Mortality appraisal and sensitivity analysis to examine the true effect
Seven studies provided data on mortality and were entered of EN on anastomotic healing in a clinical setting. In the
in the meta-analysis.23,25,26,28-31 As shown in Figure 5A, early current meta-analysis, the pooling of all 9 randomized trials
start of oral intake did not affect mortality compared with regardless of methodological quality resulted in several
late start of oral intake (OR, 0.61; 95% CI, 0.17–2.22; P = beneficial effects in favor of early oral intake. However,
.45). Excluding studies with risk of bias did not alter the the strict exclusion of 6 studies with a modest to high risk
overall effect of early start of oral intake on mortality (OR, of bias25-30 in the sensitivity analysis significantly reduced
1.04; 95% CI, 0.10–10.35; P = .97) (Figure 5B). overall sample size and made the effect on AL and overall
complications no longer significant. As such, the results
from our study suggest that early start of oral intake is only
Discussion associated with a reduction of length of stay (LOS).
The current review demonstrates that the effect of early The overall reduction of LOS following early oral intake
oral intake on AL is unclear in a clinical setting. This is ranges from almost 1 day in the general meta-analysis to
mainly due to a lack of high-quality evidence, since existing approximately 3 days in the sensitivity analysis. This may
randomized trials are clinically heterogeneous and at risk of be explained by a faster return of bowel function in the
bias. early feeding group, as demonstrated by various indicators
808
Table 4. Critical Appraisal of Included Studies.

da Fonseca Dag et al,24 El Nakeeb Hartsell Lucha Minig Pragatheeswarane Reissman Stewart
Item et al,23 2011 2011 et al,25 2009 et al,26 1997 et al,27 2005 et al,28 2009 et al,29 2014 et al,30 1995 et al,31 1998

Randomization? + + + ? ? + + ? +
Allocation concealment? + + ? ? ? + − ? +
Blinding of participants and personnel? − − − − − − − − −
Blinding of outcome assessment? − − − − − − − − −
Complete outcome data? + + + + + + + + +
No selective reporting? + + + + + + + + +
No other bias? + + + + + − ? + +

+, yes; –, no; ?, unclear.


Smeets et al 809

Figure 2. (A) Forest plot for studies that examined the effect of early vs late start of oral intake for anastomotic leakage. (B)
Sensitivity analysis for studies that examined the effect of early vs late start of oral intake for anastomotic leakage. M-H,
Mantel-Haenszel.

Figure 3. (A) Forest plot for studies that examined the effect of early vs late start of oral intake for overall complications. (B)
Sensitivity analysis for studies that examined the effect of early vs late start of oral intake for overall complications. M-H,
Mantel-Haenszel.
810 Nutrition in Clinical Practice 33(6)

Figure 4. (A) Forest plot for studies that examined the effect of early vs late start of oral intake for length of stay. (B) Sensitivity
analysis for studies that examined the effect of early vs late start of oral intake for length of stay. IV, inverse variance; M-H,
Mantel-Haenszel.

Figure 5. (A) Forest plot for studies that examined the effect of early vs late start of oral intake for mortality. (B) Sensitivity
analysis for studies that examined the effect of early vs late start of oral intake for mortality. M-H, Mantel-Haenszel.
Smeets et al 811

of GI motility (eg, time to first flatus or defecation) in 3. Krarup PM, Nordholm-Carstensen A, Jorgensen LN, Harling H.
multiple studies.23-25,29 However, differences in discharge Anastomotic leak increases distant recurrence and long-term mortality
after curative resection for colonic cancer: a nationwide cohort study.
criteria between the included studies may have confounded
Ann Surg. 2014;259(5):930-938.
the effect of early oral intake on LOS, as suggested by the 4. Mirnezami A, Mirnezami R, Chandrakumaran K, Sasapu K, Sagar
high statistical heterogeneity. P, Finan P. Increased local recurrence and reduced survival from
Despite the attempt to minimize risk of bias by means colorectal cancer following anastomotic leak: systematic review and
of a sensitivity analysis, several other limitations remain meta-analysis. Ann Surg. 2011;253(5):890-899.
5. Daams F, Luyer M, Lange JF. Colorectal anastomotic leakage: as-
present in the current systematic review. First, perioperative
pects of prevention, detection and treatment. World J Gastroenterol.
protocols varied greatly between studies, including the use 2013;19(15):2293-2297.
of nonsteroid anti-inflammatory drugs and preoperative 6. Demetriades H, Botsios D, Kazantzidou D, et al. Effect of early
bowel lavage. However, while differences in these protocols postoperative enteral feeding on the healing of colonic anastomoses
can affect various clinical outcomes, the specific effect on in rats: comparison of three different enteral diets. Eur Surg Res.
1999;31(1):57-63.
AL may be limited except for nonsteroid anti-inflammatory
7. Guven A, Pehlivan M, Gokpinar I, Gurleyik E, Cam M. Early
drugs.21 Second, no study was blinded; however blinding glutamine-enriched enteral feeding facilitates colonic anastomosis heal-
is difficult to apply due to the nature of the intervention. ing: light microscopic and immunohistochemical evaluation. Acta
Third, the included studies involved various sites of lower Histochem. 2007;109(2):122-129.
intestinal surgery; it is well known that the a priori risks of 8. Khalili TM, Navarro RA, Middleton Y, Margulies DR. Early postop-
erative enteral feeding increases anastomotic strength in a peritonitis
AL vary in the small bowel, colon, and rectum. However,
model. Am J Surg. 2001;182(6):621-624.
in the sensitivity analysis, only studies including colorectal 9. Fukuzawa J, Terashima H, Ohkohchi N. Early postoperative oral
surgery were included. Last, a clear definition for AL feeding accelerates upper gastrointestinal anastomotic healing in the
lacked in most studies and varied between studies when rat model. World J Surg. 2007;31(6):1234-1239.
provided. While this review attempts to summarize best 10. Tadano S, Terashima H, Fukuzawa J, Matsuo R, Ikeda O, Ohkohchi
N. Early postoperative oral intake accelerates upper gastrointestinal
available evidence, the generalizability remains limited by
anastomotic healing in the rat model. J Surg Res. 2011;169(2):202-208.
the heterogeneity of the included studies. 11. Kiyama T, Efron DT, Tantry U, Barbul A. Effect of nutritional
In conclusion, the results of this systematic review route on colonic anastomotic healing in the rat. J Gastrointest Surg.
suggest that early oral intake is associated with a reduction 1999;3(4):441-446.
in LOS, while the effect on anastomotic healing remains 12. Kiyama T, Onda M, Tokunaga A, Yoshiyuki T, Barbul A. Effect
of early postoperative feeding on the healing of colonic anastomoses
unclear as existing literature is clinically heterogeneous
in the presence of intra-abdominal sepsis in rats. Dis Colon Rectum.
and at risk of bias. More well-designed, high-quality 2000;43(10)(suppl):S54-S58.
randomized studies are needed to further study the potential 13. Boelens PG, Heesakkers FF, Luyer MD, et al. Reduction of postop-
benefit of EN, since alternative strategies that reduce AL are erative ileus by early enteral nutrition in patients undergoing major
lacking. rectal surgery: prospective, randomized, controlled trial. Ann Surg.
2014;259(4):649-655.
14. van den Heijkant TC, Costes LM, van der Lee DG, et al. Randomized
Statement of Authorship
clinical trial of the effect of gum chewing on postoperative ileus and
B. J. J. Smeets and M. D. P. Luyer equally contributed to the inflammation in colorectal surgery. Br J Surg. 2015;102(3):202-211.
conception and design of the research; T. J. Weijs contributed 15. Andersen HK, Lewis SJ, Thomas S. Early enteral nutrition within
to the design of the research; and B. J. J. Smeets, E. G. 24h of colorectal surgery versus later commencement of feed-
Peters, and E. C. J. Horsten contributed to the acquisition and ing for postoperative complications. Cochrane Database Syst Rev.
analysis of the data and drafted the manuscript. All authors 2006;(4):CD004080.
16. Lewis SJ, Andersen HK, Thomas S. Early enteral nutrition within 24 h
contributed to the interpretation of the data, critically revised
of intestinal surgery versus later commencement of feeding: a system-
the manuscript, agree to be fully accountable for ensuring the
atic review and meta-analysis. J Gastrointest Surg. 2009;13(3):569-575.
integrity and accuracy of the work, and read and approved the 17. Osland E, Yunus R, Khan S, Memon MA. Early enteral nutrition
final manuscript. within 24 h of intestinal surgery versus later commencement of
feeding: a systematic review and meta-analysis. J Gastrointest Surg.
Supplementary Information 2009;13(6):1163-1167.
18. Zhuang CL, Ye XZ, Zhang CJ, Dong QT, Chen BC, Yu Z. Early versus
Additional supporting information may be found online in the
traditional postoperative oral feeding in patients undergoing elective
Supporting Information section at the end of the article.
colorectal surgery: a meta-analysis of randomized clinical trials. Digest
Surg. 2013;30(3):225-232.
References 19. Higgins JPT, Green S. Cochrane Handbook for Systematic Reviews
1. Jannasch O, Klinge T, Otto R, et al. Risk factors, short and long of Interventions. Version 5.1.0 [updated March 2011]. Chichester,
term outcome of anastomotic leaks in rectal cancer. Oncotarget. United Kingdom: The Cochrane Collaboration; 2011. http://www.
2015;6(34):36884-36893. cochrane-handbook.org. Accessed February 13, 2017.
2. Kang CY, Halabi WJ, Chaudhry OO, et al. Risk factors for anasto- 20. Moher D, Liberati A, Tetzlaff J, Altman DG ; PRISMA Group.
motic leakage after anterior resection for rectal cancer. JAMA Surg. Preferred reporting items for systematic reviews and meta-analyses: the
2013;148(1):65-71. PRISMA statement. J Clin Epidemiol. 2009;62(10):1006-1012.
812 Nutrition in Clinical Practice 33(6)

21. McDermott FD, Heeney A, Kelly ME, Steele RJ, Carlson GL, 28. Minig L, Biffi R, Zanagnolo V, et al. Early oral versus “traditional”
Winter DC. Systematic review of preoperative, intraoperative and postoperative feeding in gynecologic oncology patients undergoing
postoperative risk factors for colorectal anastomotic leaks. Br J Surg. intestinal resection: a randomized controlled trial. Ann Surg Oncol.
2015;102(5):462-479. 2009;16(6):1660-1668.
22. Review Manager (RevMan) [Computer program]. Version 5.3. Copen- 29. Pragatheeswarane M, Muthukumarassamy R, Kadambari D, Kate V.
hagen, Denmark: The Nordic Cochrane Centre, The Cochrane Collab- Early oral feeding vs. traditional feeding in patients undergoing elective
oration; 2014. open bowel surgery—a randomized controlled trial. J Gastrointest
23. da Fonseca LM, Profeta da, Luz MM, Lacerda-Filho A, Correia MI, Surg. 2014;18(5):1017-1023.
Gomes da, Silva R. A simplified rehabilitation program for patients 30. Reissman P, Teoh TA, Cohen SM, Weiss EG, Nogueras JJ, Wexner SD.
undergoing elective colonic surgery—randomized controlled clinical Is early oral feeding safe after elective colorectal surgery? A prospective
trial. Int J Colorectal Dis. 2011;26(5):609-616. randomized trial. Ann Surg. 1995;222(1):73-77.
24. Dag A, Colak T, Turkmenoglu O, Gundogdu R, Aydin S. A random- 31. Stewart BT, Woods RJ, Collopy BT, Fink RJ, Mackay JR,
ized controlled trial evaluating early versus traditional oral feeding after Keck JO. Early feeding after elective open colorectal resections:
colorectal surgery. Clinics. 2011;66(12):2001-2005. a prospective randomized trial. Aust N Z J Surg. 1998;68(2):125-
25. El Nakeeb A, Fikry A, El Metwally T, et al. Early oral feed- 128.
ing in patients undergoing elective colonic anastomosis. Int J Surg. 32. Beier-Holgersen R, Boesby S. Influence of postoperative enteral nutri-
2009;7(3):206-209. tion on postsurgical infections. Gut. 1996;39(6):833-835.
26. Hartsell PA, Frazee RC, Harrison JB, Smith RW. Early postoperative 33. Heslin MJ, Latkany L, Leung D, et al. A prospective, randomized
feeding after elective colorectal surgery. Arch Surg. 1997;132(5):518- trial of early enteral feeding after resection of upper gastrointestinal
521. malignancy. Ann Surg. 1997;226(4):567-580.
27. Lucha PA Jr, Butler R, Plichta J, Francis M. The economic impact of 34. Ortiz H, Armendariz P, Yarnoz C. Is early postoperative feeding
early enteral feeding in gastrointestinal surgery: a prospective survey of feasible in elective colon and rectal surgery? Int J Colorectal Dis.
51 consecutive patients. Am Surg. 2005;71(3):187-190. 1996;11(3):119-121.
Review

Nutrition in Clinical Practice


Volume 33 Number 6
The Prognostic Role of Phase Angle in Advanced Cancer December 2018 813–824

C 2018 American Society for

Patients: A Systematic Review Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10100
wileyonlinelibrary.com

Mayane Marinho Esteves Pereira, BS; Mariana dos Santos Campello Queiroz, BS;
Nathália Masiero Cavalcanti de Albuquerque, BS; Juliana Rodrigues, PhD;
Emanuelly Varea Maria Wiegert, MD; Larissa Calixto-Lima, MD; and Livia Costa de
Oliveira, PhD

Abstract
Phase angle (PA) is a ratio between the reactance and resistance obtained by bioelectric impedance analysis and has been interpreted
as a cell membrane integrity indicator and a predictor of total body cell mass. A low PA may suggest deterioration of the cell
membrane, which in advanced cancer patients may result in a reduced overall survival (OS). This systematic review sought to
investigate the current evidence regarding whether there is an association between PA and OS in patients with advanced cancer (ie,
metastatic disease). The search was conducted on electronic databases in August 2017. A total of 34 articles were identified in the
initial literature search. Nine studies reporting on 1496 patients were deemed eligible according to our inclusion criteria. PA data
were analyzed as continuous variables or according to different cutoffs, under a frequency of 50 Khz. Low PA was associated with
worse nutrition status evaluated by body mass index, serum albumin level, transferrin, and fat-free mass. The median OS of the
included papers varied from 25.5–330 days, and all studies analyzed showed a significant association between PA and OS, in that
patients with low PA had worse OS. Future studies are necessary to justify the use of PA in therapeutic decisions for this population
and to evaluate whether nutrition status can influence the association between PA and survival. (Nutr Clin Pract. 2018;33:813–824)

Keywords
bioelectric impedance; cancer; neoplasms; nutrition assessment; phase angle; survival

Introduction negatively associated with resistance.11 Lower PA suggests


cell death or decreased cell integrity, while higher PA
Cancer is recognized as a global public health problem. In suggests large quantities of intact cell membranes. Current
developing countries, at the time of diagnosis, the majority evidence presents PA as an indicator of nutrition status,
of patients with cancer have late-stage disease.1 Malnutri- a predictive factor for risk of complications and death
tion is a frequent manifestation in patients with advanced in patients suffering from different clinical conditions8
cancer and is correlated with poor prognosis and high and an independent prognostic factor in advanced cancer
mortality.2-5 patients.7,12-14 In the present study, we conducted a system-
Bioelectrical impedance analysis (BIA) has been increas- atic review of the literature (SRL) to investigate the evidence
ingly used to assess nutrition status.6,7 BIA consists of a regarding whether there is an association between PA and
rapid, relatively inexpensive, non-invasive, and reproducible overall survival (OS) in patients with advanced cancer (ie,
method for providing indirect estimates of the body’s metastatic disease).
compositional compartments, as well as the distribution
of fluids in the intracellular and extracellular spaces.6,8
In this context, BIA can be useful in clinical practice to
assess changes in body composition.9 BIA measures the From the Palliative Care Unit, National Cancer Institute José Alencar
parameters of body resistance (opposition offered by the Gomes da Silva (INCA), Rio de Janeiro, RJ, Brazil.
body to the flow of an alternate electrical current) and Financial disclosure: None declared.
reactance (resistive effect produced by the tissue interfaces Conflicts of interest: None declared.
and cell membranes).10 This article originally appeared online on May 22, 2018.
The phase angle (PA) is a ratio between reactance and
Corresponding Author:
resistance, and it has been suggested as a marker of cel- Larissa Calixto-Lima, MD, 274 Visconde de Santa Isabel Street, Vila
lular function and, consequently, of nutrition status.11 By Isabel, 20560-120, Rio de Janeiro, RJ, Brazil; +55-21-991729948.
definition, PA is positively associated with capacitance and Email: larissa_calixto@hotmail.com
814 Nutrition in Clinical Practice 33(6)

Table 1. Keywords Used in Search Strategy in Electronic Data Extraction


Databases.
Data extraction tables were specifically developed for this
Electronic Databases Keywords SRL, and the following information was selected indepen-
dently by 2 reviewers: the general characteristics of the stud-
MEDLINE/PubMed ("advanced cancer") AND
ies (first author, year of publication, study design, sample
(https://www.ncbi.nlm. ("electric impedance" OR
nih.gov/pubmed) "phase angle") AND size, country, study aims, and statistical test), participant
("survival" OR characteristics (age and cancer population), information
"outcomes" OR about the BIA measurements (model, current frequency,
"mortality" OR and PA thresholds) and main results about associations
"prognosis") between PA and nutrition status and OS rates.
Scopus (advanced cancer) AND
(https://www.scopus.com) (electric impedance)
AND (survival OR Quality Assessment
outcomes OR mortality
OR prognosis) The quality assessment was performed by 2 independent
LILACS (advanced cancer) AND reviewers using the Newcastle-Ottawa Scale (NOS)16 (Ap-
(http://lilacs.bvsalud.org/) (electric impedance OR pendix 1). The scale consists of 3 quality criteria: selection,
phase angle) AND comparability, and outcome. The maximum score is 9 points
(survival OR outcomes (4 for selection, 2 for comparability, and 3 for outcome).
OR mortality OR Study quality was defined as poor when the score was 1–3,
prognosis)
fair when the score was 4–6, and good when the score was
Cochrane Library (advanced cancer) AND
(http://onlinelibrary. (electric impedance OR 7–9 points.
wiley.com/cochranelibrary/ phase angle) AND Regarding the comparability domain, in addition to age
search) (survival OR outcomes and sex, other confounding factors considered for the sta-
OR mortality OR tistical analysis controls were prognostic evaluation through
prognosis) the Palliative Prognostic Index (PPI) or Palliative Prognostic
(PAP) Score, Karnofsky Performance Score (KPS), and the
Eastern Cooperative Oncology Group (ECOG) Scale of
Performance Status. The score assigned for each paper is
described in Figure 1.
Inter-reviewer reliability was determined using Cohen’s
κ statistics. The interpretation of the coefficient was based
Methods
on the proposal of Shrout.17 The inter-reviewer reliability
Literature Search and Study Selection presented an optimal accuracy (κ = 0.89), representing an
agreement of 92.5%.
A comprehensive search of the literature was conducted ac-
cording to the Preferred Reporting Items for Systematic Re-
views and Meta-Analyses (PRISMA) criteria15 using well- Results
known indexed databases, including MEDLINE/PubMed, Literature Search and Characteristics of the
Scopus, LILACS, and the Cochrane Library in August
2017; a combination of search terms is described in
Included Studies
Table 1. No restrictions were made regarding language or The search resulted in a total of 34 papers. After the
publication date. We selected the studies by the following exclusion for study design, 27 papers were selected for title
inclusion criteria: 1) abstract available on-line, 2) original and abstract review. Subsequently, 18 papers were selected
articles, 3) cohort or case-control design, 4) performed in for full-text review (Figure 2). Papers were excluded by
humans, 5) participants aged ࣙ18 years, and 6) presented the careful reading of titles and abstracts, with reasons for
the relationship between PA and OS. The reference lists of exclusion listed in Table S1. In addition, the examination of
related and included papers were also screened to search for the reference lists of the papers did not recover any further
additional potential studies. studies. Finally, 9 studies reporting on 1496 patients were
Two authors independently reviewed search results. Re- deemed eligible for this SRL.
viewers assessed each title and abstract and considered each The selected papers were all published within the
study for a full-text review. Any disagreements in either last 14 years,7,12,14,18-23 and most were performed
title/abstract or in the full-text paper review phases were on outpatients14,19,21-23 and conducted in the United
resolved by consensus. The opinion of a third reviewer was States,12,14,20-23 Mexico,18,19 and South Korea.7 Five
sought when necessary. studies were prospective,7,12,18-20 and the others were
Pereira et al 815

Figure 1. Evaluation of the methodological quality of selected papers.

Figure 2. Flow chart of studies selection.


816
Table 2. Description of Studies Regarding Authors, Year of Publication, Origin, Age of Participants, Study Design, Sample Size, Cancer Type, Objectives, and Type of
Bioelectrical Impedance Analysis.

Sample
Author/Year Origin Study Design Size Cancer Population Age Objectives BIA

Gupta et al., 200422 USA Retrospective 52 Outpatients, stage IV colorectal 55.8 ± 10.8 Investigate prognostic role BIA-101Q analyzer (RJL
cohort cancer yearsa of PA in advanced Systems, Clinton
colorectal cancer. Township, MI, USA),
single frequency (50
kHz)
Gupta et al., 200423 USA Retrospective 42 Outpatients, stage IV 56.2 ± 10.7 Investigate prognostic role BIA-101Q analyzer (RJL
cohort pancreatic cancer yearsa of PA in advanced Systems, Clinton
pancreatic cancer. Township, MI, USA),
single frequency (50
kHz)
Davis et al., 200920 USA Prospective 50 Inpatients, without defining 63.0 ± 12.0 Determine if BIA correlates (RJL Systems, Clinton
cohort advanced cancer. Cancer yearsa before hydration or Township, MI, USA),
type: pancreatic (12.0%), changes during hydration single frequency (50 kHz)
lung (12.0%), breast (12.0%), and determine if these
myeloma (6.0%), renal changes were
(6.0%), colon (6.0%), gastric prognostically important.
(6.0%).
Gupta et al., 200921 USA Retrospective 165 Outpatients, stages IIIB and IV 56.0 ± 9.1 Investigate prognostic role SFB7 BioImp v1.55
cohort with non-small-cell lung yearsa of BIA-derived PA in analyzer
cancer, treated at Cancer patients with advanced (ImpediMed,
Treatment Centers of non-small-cell lung Brisbane, Australia),
America. cancer. single frequency (50
kHz)
Sánchez-Lara et al., Prospective 119 Outpatients, diagnosis of 60.5 ± 12.5 Evaluate association of Bodystat Quadscan
201219 Mexico cohort histopathologic-confirmed yearsa nutrition parameters in 4000,
or cytologic-confirmed stage health-related quality of multi-frequency
III or IV non-small-cell lung life and OS in patients
cancer. with advanced
non-small-cell lung
cancer.
Lee et al., 20147 South Prospective 28 Inpatients, without defining >70 years Investigate BIA-derived PA Biodynamics model 450
Korea cohort advanced cancer. Cancer = 53.6% as prognostic indicator (Biodynamics Co.,
type: digestive tract (39.2%), 50–70 years for survival in advanced Seattle, WA, USA),
lung (10.7%), hematology = 35.7% cancer patients. single frequency (50
(10.7%), bladder/renal 30–50 years kHz)
(10.7%), and other (28.5%). = 10.7%

(continued)
Table 2. (continued)

Sample
Author/Year Origin Study Design Size Cancer Population Age Objectives BIA

Hui et al., 201412 USA Prospective 222 Inpatients, without defining 55.0 Determine association of Quantum IV (RJL
cohort advanced cancer. Cancer (22.0–79.0) PA, hand grip strength, Systems, Clinton
type: digestive tract most yearsb and maximal inspiratory Township, MI, USA),
common (33.0%), pressure with OS in single frequency (50
respiratory (16.0%), breast patients with advanced kHz)
(13.0%), gynecologic cancer.
(11.0%), genitourinary
(9.0%), head and neck
(5.0%), hematologic (5.0%),
and other (9.0%).
Hui et al., 201714 USA Retrospective 366 Outpatients with advanced 58.0 Determine association of Inbody 720
cohort cancer defined as locally (21.0–90.0) PA obtained from
advanced, recurrent or yearsb multi-frequency BIA with
metastatic disease for solid OS in patients with
tumors, or progres- advanced cancer.
sive/refractory/incurable
disease for hematologic
tumors. Cancer type:
gastrointestinal (30.0%),
breast (14.0%), head and
neck (13.0%), respiratory
(11.0%), gynecologic (9.0%),
and other (15.0%).
(Inbody, Cerritos,
CA, USA),
multi-frequency
(5, 50, and 250
kHz)
Camargo et al., Prospective 452 Inpatients, without defining 57.6 ± 14.6 Associate PA and OS in Inbody 720
201718 Mexico cohort advanced cancer. Cancer yearsa palliative patients at
type: gastric (39.2%), lung National Cancer Institute
(18.8%), gynecologic of Mexico.
(16.6%), and other (25.4%).
(Inbody, Cerritos,
CA, USA),
single frequency
(50 kHz)

BIA, bioelectrical impedance analysis; OS, overall survival; PA, phase angle.
a Mean (± standard deviation).
b Median (interquartile range).

817
818 Nutrition in Clinical Practice 33(6)

retrospective.11,18-20 The age of the population included in general, patients with a PA ࣘ5.6° presented a significantly
the studies varied between the fifth and sixth decade of life. shorter survival time than those with a PA >5.6°.
Four studies included a population with specific types of The median OS of the included papers varied from 25.5–
cancer. One study was exclusively with colorectal cancer 330 days, and the papers that compared the median OS
patients,22 1 with pancreatic cancer patients,23 and 2 studies among those with low and high PA showed decreased values
with lung cancer patients.19,21 For studies that included in OS for those with low PA (Table 3).12,18-23
different cancer types,7,12,14,18,20 digestive tract cancer was It was observed that patients with colorectal cancer
the most common type (Table 2). presented a median OS of 8.6 months when the PA was
ࣘ5.6°,22 lung cancer patients presented a median OS of
Type of BIA Used to Assess PA 7.6 months when PA was ࣘ5.4°,16 and in pancreatic cancer
patients, the median OS was 6.3 months among those with
Regarding the BIA equipment, different models (Biody- PA ࣘ5.0°.20 For studies that included a population with
namics 450, Inbody 720, Bodystat quadscan 400, SFB7 different types of cancer, median OS varied from 35–162
Bioimp v1.55 analyzer, and RJL systems) were used to days with the previously mentioned cutoffs (Table 3).7,12,14,18
assess PA; most were manufactured in the U.S. Only 2 In the Cox analyses, the hazard ratio was approximately
studies used a multi-frequency BIA,14,20 but only 1 study 20% higher for each unit increase of PA.7,12,14,20-23 When
presented PA data using these different frequencies (5, 50, different cutoffs were tested, the hazard ratio for mortality
and 250 kHz).14 Further studies were conducted at a single varied between 1.8-fold–10.7-fold risk for death.18,19,22 In
frequency of 50 kHz (Table 2).7,12,18-23 addition, it was observed that all the studies included in this
SRL showed a significant association between PA and OS
PA Association With Nutrition Status (Table 3).
The association between PA and nutrition status or body
composition was included only in 3 of the selected papers Discussion
and evaluated by pre–serum albumin level23 : 1 for body
mass index (BMI),18 1 for subjective global assessment This is the first SRL that evaluated the association be-
(SGA),23 and 1 for fat-free mass.12 With the exception tween PA and OS in advanced cancer patients. Our results
of SGA, these nutrition markers showed a negative and confirmed that a lower PA value was associated with OS
significant association with PA, indicating that subjects with in advanced cancer patients. These findings suggest PA
worse nutrition status had lower values of PA (Table 3). as a relevant indicator for unfavorable outcomes, with a
decrement in the survival rate. To determine reliable and
useful prognostic factors that can be used in clinical practice,
PA Association With OS the improvement of therapeutic approaches for this type
Survival analyses were performed using Cox proportional of population is necessary.24,25 In this context, PA is an
hazards models with bivariate and multivariate logistic objective and non-invasive method that can be used for
regression in all the studies. Kaplan-Meier curves were prognosis.
constructed to analyze the probability of OS according to Patients with advanced cancer may present a PA lower
PA,12,14,18-23 and the log-rank test was used7,12,14,18,20-23 to than those observed in a healthy population.26 This reduced
verify survival differences between the groups. PA may be related to the metabolic disarrangement that
Regarding the confounder variables used for controls this population experiences, such as malnutrition and cancer
in Cox regression analyses, a high variability among the cachexia.27 In fact, a poor nutrition status can result in
studies was observed: 6 studies controlled for age,7,14,18-21 3 a body fluid imbalance and cell membrane changes.12,27,28
controlled for gender,14,18,20 and only 1 controlled for race.17 Thus, the relationship of PA with body composition and
None of the studies controlled for physical activity. The nutrition status makes it an indicator for the predictive risk
nutrition status of patients was not considered in 3 of the 9 of morbidity and mortality.12
papers.11,20,21 Some studies controlled for different markers, In this context, a significant association was found be-
such as BMI,7,15,19 SGA,19,22 pre–serum albumin level,23 tween PA and the nutrition markers such as pre–serum albu-
and transferrin,23 and only 2 of these studies adjusted for min level, BMI, and fat-free mass. Pre–serum albumin level
body composition, such as fat-free mass.12,22 Four studies is a reliable indicator of acute changes in nutrition status,
included information about useful tools for predicting sur- and it is unaffected by hydration status. On the other hand,
vival; 3 publications11,19,20 included the ECOG scale, and 17 the use of BMI has been vastly discussed in the literature
included the KPS and PPI (Table 3). as a nutrition marker that does not take into account the
Different PA cutoffs were used varying from 4.0°→6.0°, evaluation of fat-free mass and body fat. Hui et al. tested
and in 3 of the studies, no cutoffs were used for the Cox the association between PA and fat-free mass, and found
analysis; the PA was used as a continuous variable.7,12,20 In a positive and significant association, emphasizing the idea
Table 3. Description of Statistical Tests, Cutoffs for Phase Angle, Survival, Potential Confounding, and Main Findings Applied in Studies.

Association
Cutoff for PA (50 Potential Between PA and Association Between PA and
Author/Year Statistical Test kHz) Confounding Nutrition Status OS Main Findings

Gupta et al., Cox regression; 5.57° Diagnostic time; – Patients with PA ࣘ5.6° had PA and age at diagnosis presented
200422 Kaplan-Meier; body weight; median survival of 8.6 positive correlation with OS.
log-rank test. lean mass; months, while others Furthermore, SGA was not
serum albumin presented median OS of 40.4 associated with OS. PA was
level; pre–serum months. In multivariate prognostic indicator in patients
albumin level; analysis, PA (ࣘ5.6°) was a with advanced colorectal cancer.
transferrin; prognostic indicator in The risk was approximately 11-fold
SGA; stage of patients with advanced higher for those with PA ࣘ5.6°.
tumor; prior colorectal cancer (HR =
treatment. 10.7; 95% CI: 1.9–60.2;
P-value = 0.007).
Gupta et al., Spearman 5.0° Pre–serum PA was associated Patients with PA ࣘ5.0° had PA associated positively with OS.
200423 correlation; albumin level; with pre–serum median OS of 6.3 months, Pre–serum albumin level
Pearson serum albumin albumin level (r while others presented a (<150mg/L) was also associated
correlation; Cox level; prior = 0.32; P = median OS of 10.2 months with OS. PA was strong prognostic
regression; treatment. 0.04), (P-value = 0.02). indicator in advanced pancreatic
Kaplan-Meier; transferrin (r = Multivariate analysis after cancer patients. Therefore, each
log-rank test. 0.19; P = 0.25) adjusting for pre–serum unit increase on PA showed
and SGA (r = albumin level demonstrated reduction of mortality of 31%.
-0.26; P = 0.10). relative risk reduction (=
0.69; 95% CI: 0.49; 0·97) that
was associated with each unit
increase in PA. PA continued
to be statistically significant
(P-value = 0.02) after jointly
controlling for serum
albumin level, pre–serum
albumin level, and previous
treatment history.

(continued)

819
820
Table 3. (continued)

Association
Cutoff for PA (50 Potential Between PA and Association Between PA and
Author/Year Statistical Test kHz) Confounding Nutrition Status OS Main Findings

Davis et al., Spearman Survival Resistance; total – Higher PA before hydration Weight loss was associated with
200920 correlation; curves: body fluid; predicted a borderline and shorter OS. PA positively predicted
Pearson Decreased or sodium; gender; significantly lower OS (HR borderline and significantly lower
correlation; Cox not altered age; skin color; = 0.8; 95% CI: 0.6–1.0; OS in advanced cancer patients on
regression; vs increased ECOG. P-value = 0.057); first day of their hospital stay,
Kaplan-Meier; PA paradoxically, an increase in indicating that each unit increase
log-rank test. Regression: PA during hydration on PA presented reduction of 20%
Continuous predicted lower OS (HR = in mortality.
variable 1.2; 95% CI: 1.1–1.4; P-value
= 0.007).
Gupta et al., Cox regression; 5.3° Age; stage of – Patients with PA ࣘ5.3 had Stage at diagnosis and treatment
200921 Kaplan-Meier; tumor; prior median OS of 7.6 months, history was associated with OS.
log-rank test. treatment. while those >5.3 had 12.4 BIA-derived PA was positive and
months (P = 0.02). In independently prognostic indicator
multivariate analysis, each of OS in patients with stage IIIB
degree of increase in PA, and IV non-small-cell lung cancer.
adjusted for age, stage at Nutrition interventions targeted at
diagnosis, and prior improving PA could potentially
treatment, history was lead to improved OS in patients
associated with OS (HR = with advanced non-small-cell lung
0.8; 95% CI: 0.6–1.0; P-value cancer. Therefore, each unit
= 0.020). increase on PA presented reduction
of 20% in mortality.
Sánchez-Lara Mann-Whitney U 5.8° Gender; age; – Median follow-up was 6 (± 5) Malnutrition measured by SGA,
et al., 201219 test; Cox ECOG; stage of months. In multivariate weight loss >10%, and BMI >20
regression; tumor; weight analysis, patients with was associated with lower related
Kaplan-Meier. loss; BMI; ECOG = 2 (HR = 2.7; 95% quality of life. Patients with ECOG
SGA; serum CI: 1.5–4.7; P-value = = 2, high content serum IL-6, and
albumin level; 0.001), PA ࣘ5.8° (HR = 3.0; lower PA showed lower OS. PA was
PLR; NLR; 95% CI: 1.2–7.1; P-value = presented to be an independent
CRP; IL-6; 0.011) and malnutrition (HR prognostic factor in advanced
TNF-α. = 2.7; 95% CI: 1.3–5.5; non-small-cell lung cancer, where
P-value = 0.005) had worse those with PA ࣘ5.8° presented risk
OS. 3-fold higher than those with
higher values.

(continued)
Table 3. (continued)

Association
Cutoff for PA (50 Potential Between PA and Association Between PA and
Author/Year Statistical Test kHz) Confounding Nutrition Status OS Main Findings

Lee et al., Spearman Survival Age; BMI; PPI; – Median OS time was 25.5 days. PA showed positive and significant
20147 correlation; Cox curves: KPS. PA ࣙ4.4° showed a longer association with OS, where each
regression; 4.4° survival time compared with unit increase on PA presented a
log-rank test. Regression: PA <4.4° (100 vs 125 days, reduction of 36% in mortality.
Continuous respectively; P-value = 0.01). However, age, PPS, and BMI did
variable In multivariate analysis, each not show significant relationship
degree of increase in PA, with OS. BIA-derived PA may
adjusted for age, PPI, BMI, serve as independent prognostic
and PA was associated with indicator in advanced cancer
OS (HR = 0.64; 95% CI: patients.
0.42–0.95; P-value = 0.028).
Hui et al., Spearman Survival PAP Score; serum PA was associated Median OS was 106 days (95% PA was positively associated with OS,
201412 correlation; Cox curves: albumin level; with fat free CI: 71–128 days). Patients where each unit increase on PA
regression; 4.4° vs >4.4° fat-free mass. mass (r = 0.29; with lower PA (<3°) had presented a reduction of 14% in
Kaplan-Meier; 2.0–2.9° vs P <0.001) and lower survival (35 vs 220 mortality. Furthermore, patients
log-rank test. 3.0–3.9° vs the fat free mass days) when compared with with lower KPS, PPS,
4.0–4.9° vs index (r = 0.33; those with higher values (PA hypoalbuminemia (<3.0 g/dL), and
5.0–5.9° vs P <0.001). ࣙ6.0°). In multivariate lower fat-free mass (ࣘ51.6 kg) had
ࣙ6.0° analysis, each degree of poor survival. PA was shown to be
<P5 vs >P95 increase in PA was an independently established
Regression: associated with OS (HR = prognostic factor in advanced
Continuous 0.86; 95% CI: 0.74–0.99; cancer setting during hospital stay.
variable P-value = 0.04).
Hui et al., Spearman Survival Gender; age; race; – Median OS was 250 days. PA was positively associated with OS,
201711 correlation; curves: tumor type; Patients with PA ࣘ4.5° where each unit increase on PA
Contal & Right side of ECOG; weight showed a significant lower presented reduction of 15% in
O’Quigley; Cox body: 4.5° loss in 6 median of OS (162 days) mortality. In addition,
regression; Left side of months; serum when compared with those hypoalbuminemia (<4.0g/dL)
Kaplan-Meier; body: 4.3° albumin level; with higher (>4.5°) values elevated lactate dehydrogenase
log-rank test. Regression: calcium; (403 days). In multivariate (>618 unit), serum albumin levels,
Right side of hemoglobin; analysis, each degree increase and neutrophil-lymphocyte ratio
body: 4.4° leukocytes; in PA was associated with (>3) were correlated with poor
Left side of lactate; OS (HR = 0.85; 95% CI: survival. PA represented novel
body: 4.2° lymphocytes; 0.72–0.99; P-value = 0.048). objective prognostic factor in an
neutrophils. outpatient palliative cancer-care
setting, regardless of frequency and
body sides.

(continued)

821
822
Table 3. (continued)

Association
Cutoff for PA (50 Potential Between PA and Association Between PA and
Author/Year Statistical Test kHz) Confounding Nutrition Status OS Main Findings

Camargo et al., Spearman 4.0° Gender; age; Patients with BMI Average survival was 86 vs 163 PA and BMI were positively
201715 correlation; Cox diagnose; BMI. <24.9kg/m² had days for patients with PA correlated with OS. Therefore, risk
regression; significantly ࣘ4.0° and >4.0°, for mortality was approximately
Kaplan-Meier; lower mean PA respectively (P >0.0001). In 2-fold higher for those with PA
log-rank test. (3.73° ±0.92) the multivariate analysis, ࣘ4.0°.
than those with patients with PA ࣘ4.0° had
BMI ࣙ 25 kg/m² HR = 1.8; 95% CI: 1.4–2.3;
(4.37° ±0.98; P P-value >0.0001.
= 0.011).

BIA, bioelectrical impedance analysis; BMI, body mass index; CI, confidence interval; CRP, C-reactive protein; ECOG, Eastern Cooperative Oncology Group; HR, hazard ratio; IL-6,
interleukin 6; KPS, Karnofsky Performance Status; NLR, neutrophils/lymphocytes ratio; OS, overall survival; PA, phase angle; PAP Score, Palliative Prognostic Score; PLR,
platelets/lymphocytes ratio; PPI, Palliative Prognostic Index; PPS, Palliative Performance Scale; SGA, subjective global assessment; TNF-α, tumor necrosis factor-α; vs, versus.
Pereira et al 823

that body composition has an important influence on PA (<50 kHz) are associated with a flow through the extra-
values.12 cellular compartment, whereas higher frequency currents
It is worth noting that despite all the papers included in (>200 kHz) penetrate the cell membranes and pass through
this SRL that assessed PA by using the BIA, most studies thin tissues.33 This differential tissue penetration at a higher
did not evaluate body composition in their study popu- frequency current allows for better accuracy in assessing
lation. Based on the assumption that body composition the body’s composition in healthy individuals.34 Still, more
may influence OS, it is reasonable to say that controlling studies are needed to evaluate the usefulness of different
for this potential confounder in these analyses is necessary. frequencies in the evaluation of PA in advanced cancer
However, most of the studies controlled for at least 1 marker patients.
of nutrition status in the Cox regression. With regard to strengths, the SRL methodology was
Other potential confounders used in the Cox analyses in consistent with the PRISMA statement.15 The search was
these studies should be discussed. The first is performance conducted in different databases and in the reference lists of
status (KPS) or prognostics scales (PPI and PAP score), the included papers; there were also no restrictions made for
which are useful tools for predicting survival of palliative language, the year of publication, or the place of execution.
subjects.13 Only 4 studies, however, included this informa-
tion. Nevertheless, these confounders were considered in Conclusions
the evaluation of quality assessment in this SRL. Other
Considering that survival prediction remains a challenge in
confounders that should be included in analyses that are
individuals with advanced cancer, the results of this SRL
influences on PA values are ethnicity/race, gender, age,
show that PA, derived from BIA, is an important objective
weight and height.12,29,30 Therefore, these aspects should
predictive factor of OS for this population. Nonetheless,
be considered in the study design when involving mea-
there is still a lack of information regarding the use of
surements of PA because these confounders may lead to a
thresholds in this population. Future studies are necessary
prejudice in the associations between PA and OS.
to identify cutoffs of PA to guide therapeutic decisions
Another factor that may also play a role in these sta-
and to evaluate whether nutrition status can influence the
tistical analyses could be the population included in the
association between PA and survival. While no specific
studies; some studies included subjects with different cancer
threshold can be used as a prognostic factor in this popu-
types, and no statistic control was observed for cancer
lation, the reduction in values of PA, evaluated routinely,
type. The lack of control for this variable might influence
could indicate a worse OS.
the PA analysis and its association with OS. Thus, further
investigation regarding this issue is necessary.
Another important issue is related to the different PA Statement of Authorship
cutoffs in the analyses of the studies. Currently, there is L. Calixto-Lima and L.C. Oliveira equally contributed to
no known precise PA value capable to identify the length the conception and design of the research; L.C. Oliveira,
of survival in individuals with advanced cancer. In general, M.M.E. Pereira, M.S.C. Queiroz, E.M. Wiegert, and J.
the mean, median, or lower PA quartiles found in the Rodrigues contributed to the acquisition, analysis, and in-
groups studied were used as a parameter. However, it can terpretation of the data; L.C. Oliveira and L. Calixto-Lima
be affirmed that sick individuals present lower values of PA, drafted the manuscript; M.M.E. Pereira, M.S.C. Queiroz,
which correlates with the severity of the disease.7,12,14,18-23 N.M.C. de Albuquerque, J. Rodrigues, E.M. Wiegert,
In all the studies, the data were collected in a sin- L. Calixto-Lima, and L.C. Oliveira critically revised the
gle care center and were related to inpatient and/or out- manuscript; M.M.E. Pereira, M.S.C. Queiroz, N.M.C. de
patient follow-ups.7,12,14,18-23 It is worth mentioning that Albuquerque, J. Rodrigues, E.M. Wiegert, L. Calixto-Lima,
survival may differ substantially between inpatients and and L.C Oliveira agree to be fully accountable for ensuring
outpatients.14 Inpatients present specific features that de- the integrity and accuracy of the work. All authors read and
mand specialized treatment, including better symptom con- approved the final manuscript.
trol, which could limit the generalization of these findings.
The use of different methods to evaluate PA was influ- Supplementary Information
enced by the BIA frequency that was used to evaluate it, Additional supporting information may be found online in the
i.e., single-frequency or multi-frequency.31,32 In this SRL, Supporting Information section at the end of the article.
different methods of BIA were used in the included studies,
while only 2 studies used a multi-frequency BIA.14,19 How-
ever, only 1 of these studies showed results that were related References
to different frequencies in patients with advanced cancer 1. WHO. World Health Organization. National Cancer Control Programs:
in palliative care; this study was the first with differential Policies and Managerial Guidelines. 2nd ed. Geneva: World Health
analyses relating PA and OS.14 Lower frequencies of PA Organization; 2002.
824 Nutrition in Clinical Practice 33(6)

2. Gangadharam A, Choi SE, Hassan A, et al. Protein calorie malnutri- 19. Sánchez-Lara K, Turcott JG, Juárez E, et al. Association of nutrition
tion, nutritional intervention and personalized cancer care. Oncotarget. parameters including bioelectrical impedance and systemic inflam-
2017;8(14):24009-24030. matory response with quality of life and prognosis in patients with
3. Aktas A, Walsh D, Galang M, et al. Under-recognition of malnutrition advanced non-small-cell lung cancer: a prospective study. Nutr Cancer.
in advanced cancer: the role of the dietitian and clinical practice 2012;64(4);526-534.
variations. Am J Hosp Palliat Care. 2017;34(6):547-555. 20. Davis MP, Yavuzsen T, Khoshknabi D, et al. Bioelectrical impedance
4. Arends J, Bachmann P, Baracos V, et al. ESPEN guidelines on phase angle changes during hydration and prognosis in advanced
nutrition in cancer patients. Clin Nutr. 2017;36(1):11-14. cancer. Am J Hosp Palliat Care. 2009;26(3):180-187.
5. Santarpia L, Marra M, Montagnese C, Alfonsi L, Pasanisi F, Contaldo 21. Gupta D, Lammersfeld CA, Vashi PG, et al. Bioelectrical impedance
F. Prognostic significance of bioelectrical impedance phase angle in phase angle in clinical practice: implications for prognosis in stage IIIB
advanced cancer: Preliminary observations. Nutrition. 2009;25(9):930- and IV non-small cell lung cancer. BMC Cancer 2009;9(37):1-6.
931. 22. Gupta D, Lammersfeld CA, Burrows JL, et al. Bioelectrical impedance
6. Gupta D, Lis CC, Dahlk SL, et al. The relationship between bio- phase angle in clinical practice: implications for prognosis in advanced
electrical impedance phase angle and subjective global assessment in colorectal cancer. Am J Clin Nutr. 2004;80(6):1634-1638.
advanced colorectal cancer. Nutr J. 2008;7(19):1-6. 23. Gupta D, Lis CG, Dahlk SL, Vashi PG, Grutsch JF, Lammersfeld
7. Lee SY, Lee YJ, Yang J, Kim C, Choi W. The association between CA. Bioelectrical impedance phase angle as a prognostic indicator in
phase angle of bioelectrical impedance analysis and survival time advanced pancreatic cancer. Br J Nutr. 2004;92(6):957-962.
in advanced cancer patients: preliminary study. Korean J Fam Med. 24. Krishnan M, Temel JS, Wright AA, Bernacki R, Selvaggi K, Balboni T.
2014;35(5):251-256. Predicting life expectancy in patients with advanced incurable cancer:
8. Llames L, Baldomero V, Iglesias ML, Rodota LP. Values of the phase a review. J Support Oncol. 2013;11(2):68-74.
angle by bioelectrical impedance; nutritional status and prognostic 25. Partridge M, Fallon M, Bray C, McMillan D, Brown D, Laird B. Prog-
value. Nutr Hosp. 2013;28(2): 286–295 nostication in advanced cancer: a study examining an inflammation-
9. Sarhill N, Mahmoud FA, Christie R, Tahir A. Assessment of nutri- based score. J Pain Symptom Manage. 2012;44(2):161-167.
tional status and fluid deficits in advanced cancer. Am J Hosp Palliat 26. Mulasi U, Kuchnia AJ, Cole AJ, Earthman CP. Bioimpedance at the
Care. 2003,20:465-473. bedside: current applications, limitations, and opportunities. Nutr Clin
10. Zarowitz BJ, Pilla AM. Bioelectrical impedance in clinical practice. Pract. 2015;30:180-193.
DICP. 1989; 23:548-555. 27. Grundmann O, Yoon SL, Williams JJ. The value of bioelectrical
11. Baumgartner RN, Chumlea WC, Roche AF. Bioelectric impedance impedance analysis and phase angle in the evaluation of malnutrition
phase angle and body composition. Am J Clin Nutr. 1988,48: and quality of life in cancer patients – a comprehensive review. Eur
16-23 J Clin Nutr. 2015;69(12):1290-1297.
12. Hui D, Bansal S, Morgado M, Dev R, Chisholm G, Bruera E. Phase 28. Norman K, Stobaus N, Zocher D, et al. Cutoff percentiles of bioelec-
angle for prognostication of survival in patients with advanced cancer: trical phase angle predict functionality, quality of life, and mortality in
preliminary findings. Cancer. 2014;120(14):2207-2214. patients with cancer. Am J Clin Nutr. 2010:92(3):612-619.
13. Hui D. Prognostication of survival in patients with advanced 29. Barbosa-Silva MCG, Barros AJD, Wang J, Heymsfield SB, Pierson
cancer: predicting the unpredictable? Cancer Control. 2015;22(4): RN Jr. Bioelectrical impedance analysis: population reference values
489-497. for phase angle by age and sex. Am J Clin Nutr. 2005;82(1):49-52.
14. Hui D, Dev R, Pimentel L, et al. Association between multi-frequency 30. Gonzalez MC, Heymsfield SB. Bioelectrical impedance analysis for
phase angle and survival in patients with advanced cancer. J Pain diagnosing sarcopenia and cachexia: what are we really estimating?
Symptom Manage. 2017;53(3):571-577. J Cachexia Sarcopenia Muscle. 2017;8(2):187-189.
15. Moher D, Liberati A, Tetzlaff J, Altman DG; PRISMA Group. 31. Kyle UG, Bosaeus I, De Lorenzo AD, et al. Bioelectrical impedance
Preferred reporting items for systematic reviews and meta- analysis-part I: review of principles and methods. Clin Nutr.
analyses: The PRISMA statement. J Clin Epidemiol. 2009;62(10): 2004;23(5):1226-1243.
1006-1012. 32. Kyle UG, Bosaeus I, De Lorenzo AD, et al. Bioelectrical impedance
16. Wells GA, Shea B, O´Connell D, et al. The Newcastle-Ottawa Scale analysis-part II: utilization in clinical practice. Clin Nutr. 2004;
(NOS) for assessing the quality of non-randomized studies in meta- 23(6):1430-1453.
analyses. http://www.ohri.ca/programs/clinical_epidemiology/oxford. 33. Utter AC, Nieman DC, Mulford GJ, et al. Evaluation of leg-to-leg BIA
asp. Accessed March 15, 2017. in assessing body composition of high-school wrestlers. Med Sci Sports
17. Shrout PE. Measurement reliability and agreement in psychiatry. Stat Exerc. 2005;37(8):1395-1400.
Methods Med Res. 1998;7:301-317. 34. Gaba A, Kapus O, Cuberek R, Botek M. Comparison of multi-
18. Camargo DAP, Pérez SRA, Franco MMR, Licona NEA, Ceniceros and single-frequency bioelectrical impedance analysis with dual-energy
VIU, Baldenegro LEF. Phase angle of bioelectrical impedance analysis X-ray absorptiometry for assessment of body composition in post-
as prognostic factor in palliative care patients at the national cancer menopausal women: effects of body mass index and accelerometer-
institute in Mexico. Nutr Cancer. 2017;69(4):601-606. determined physical activity. J Hum Nutr Diet. 2015;28(4):390-400.
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 6
Effect of Vitamin D Level on Clinical Outcomes in Patients December 2018 825–830

C 2018 American Society for

Undergoing Left Ventricular Assist Device Implantation Parenteral and Enteral Nutrition
DOI: 10.1002/ncp.10078
wileyonlinelibrary.com

Fadi Abou Obeid, MD1 ; Gardner Yost, MS2 ; Geetha Bhat, PhD, MD2 ;
3 4
Erin Drever, MD ; and Antone Tatooles, MD

Abstract
Background: Vitamin D is an important hormone that regulates cardiac myocyte function. Low levels contribute to the
development of cardiovascular disease and have been implicated in immune function and the inflammatory cascade. Patients who
undergo left ventricular assist device (LVAD) implantation are at risk for driveline infection, stroke, and gastrointestinal (GI)
bleeding. We investigated whether serum 25-hydroxy (25-OH) vitamin D levels affect clinical outcomes after LVAD. Methods:
212 patients who underwent LVAD implantation between 2010 and 2015 were included. We measured preoperative 25-OH
vitamin D level and postoperative adverse events during the first year. Vitamin D level was classified into 3 categories: normal
(>30 ng/mL), insufficient (20–30 ng/mL), and deficient (<20 ng/mL). Clinical outcomes in both insufficient and deficient categories
were compared with the normal category. Results: The odds ratio (OR) of being admitted ࣙ2 times was 2.46 (95% confidence
interval [CI]: 1.067–5.769) for deficiency and 2.5 (95% CI: 0.970–6.443) for insufficiency. The OR of driveline infection was 6.185
(95% CI: 0.80–49.2; P = .07) for insufficiency and 11.467 (95% CI: 1.204–109.26; P = .03) for deficiency. Vitamin D levels were
not associated with GI bleeding, length of stay, or stroke. Conclusions: In patients with LVAD, both deficiency and insufficiency
of 25-OH vitamin D levels are independently associated with increased postoperative driveline infection risk and higher rate of
readmission. Further trials are needed to confirm whether a repletion regimen could be a promising means of decreasing the risk
for these postoperative adverse events. (Nutr Clin Pract. 2018;33:825–830)

Keywords
heart assist devices; heart failure; vitamin D; vitamin D deficiency

Introduction is associated with a doubled risk for stroke compared with


those with normal levels.6 This increase in risk may be
Vitamin D is a steroidal hormone that functions to maintain related to the role of vitamin D in regulation of neu-
bone health and calcium homeostasis. Recently, vitamin D ronal death, as well as its function as an antithrombotic
has been found to optimize the function of multiple organs, agent.
including the heart.1 Vitamin D has important roles in regulation of the
Vitamin D deficiency is very prevalent in patients with immune system, influencing proliferation and differentia-
congestive heart failure, and may contribute to the devel- tion of B and T cells and immunoglobulin production.7
opment of heart failure and other cardiovascular diseases
by activating the renin-angiotensin-aldosterone system, in-
ducing chronic inflammation, and causing endothelial dam- From the 1 Department of Cardiology, Advocate Christ Medical
age that can lead to hypertension, obesity, and diabetes Center, Oak Lawn, IL, USA; 2 Center for Heart Transplant and Assist
mellitus.2,3 At present, some data suggest that vitamin D Devices, Heart Institute, Advocate Christ Medical Center, Oak Lawn,
IL, USA; 3 Department of Endocrinology, Advocate Christ Medical
deficiency is associated with a poor prognosis and low Center, Oak Lawn, IL, USA; and 4 Department of Cardiothoracic
survival rate among patients with chronic heart failure Surgery, Advocate Christ Medical Center, Oak Lawn, IL, USA.
(CHF).4 Financial disclosure: None declared.
Liu et al5 reported that, among 548 patients, lower
Conflicts of interest: None declared.
vitamin D levels were associated with higher levels of brain
natriuretic peptide (BNP) and higher plasma renin activity, This article originally appeared online on March 30, 2018.
as well as increased CHF hospitalization and overall greater Corresponding Author:
incidence of all-cause mortality. Vitamin D deficiency is Geetha Bhat, PhD, MD, Medical Director, Center for Heart
Transplant and Assist Devices, Heart Institute, Advocate Christ
also associated with a greater stroke incidence. A recent Medical Center, 4440 W 95th Street, OPP 6409P, Oak Lawn, IL 60453.
study of healthy adults indicated that vitamin D deficiency Email: geetha.bhat@advocatehealth.com
826 Nutrition in Clinical Practice 33(6)

In addition, vitamin D may have an important role in were collected before LVAD implantation. Vitamin D levels
moderating the severity of bloodstream infections and were not adjusted for shifts in fluid status or administration
sepsis by binding to and inhibiting bacterial lipopolysac- of intravenous (IV) fluids. Clinical outcomes of interest
charides, and by reducing the sepsis cascade in fungal were collected for a 1-year period after LVAD implantation.
infection by controlling the production of proinflammatory The outcome measures included length of stay after LVAD,
cytokines.8,9 readmission rate, driveline infection, stroke, and GI bleeding
These important functions of vitamin D are particularly events.
germane to patients with advanced heart failure who un-
dergo left ventricular assist device (LVAD) implantation.
Candidates for LVAD implantation are patients with end- Outcome Measure Stratification and Statistical
stage heart failure who are at high risk for vitamin D deple- Analysis
tion and concomitant neurohormonal dysregulation.2-5,10
Postoperative length of stay was categorized as <3 weeks
Further, after implantation of an LVAD, patients are at
of postoperative hospital stay or >3 weeks based on mean
increased risk for infection of the surgical device pocket,
length of stay for LVAD implantation, as established by
driveline exit site, and bloodstream. Recognizing the role
Hasin et al.11
of vitamin D in regulation of neurohormonal activation
The number of total readmissions in the first year after
and immune-related processes, we sought to evaluate the
LVAD implantation was recorded. A recent article that
relationship of pre-LVAD vitamin D levels with postop-
analyzed readmissions after LVAD implantation found that
erative clinical outcomes including length of stay after
the overall readmission rate was 1.64 ± 1.97 during a 1.4 ±
LVAD, readmission rate, driveline infection, stroke, and
0.9-year follow-up period.12 In this study, we thus divided
gastrointestinal (GI) bleeding.
the number of readmissions within a year after LVAD into
2 categories: 0–1 admission and ࣙ2 admissions.
Methods Driveline infection was defined as any event identified as
exit site driveline infection in postoperative year 1. Stroke
Study Design events included both ischemic and hemorrhagic stroke, evi-
We conducted a retrospective chart review of 212 consecu- dent clinically and radiologically, observed in the first year
tive patients who underwent implantation of a continuous after LVAD implantation. GI bleeding events were defined
flow LVAD between January 1, 2010, and January 1, 2015. as any unexpected decline in hemoglobin level with evidence
This study was approved by a local ethics committee and of melena, hematemesis, or hematochezia, regardless of the
conforms with the principles outlined in the Declaration of source.
Helsinki. We conducted all analyses using bivariate logistic regres-
sion and calculated odds ratio (OR) with 95% confidence
interval (CI). We made no imputation for missing values.
Participants The main independent variable, serum 25-OH vitamin D,
Patients ࣙ18 years of age who underwent LVAD was categorized, as defined by the Endocrine Society, into
implantation between January 1, 2010, and January 1, 3 categories: normal 25-OH vitamin D with a level >30
2015, were included. Patients who required extracorporeal ng/mL, insufficient 25-OH vitamin D with a level ranging
membrane oxygenation or emergent biventricular support between 20 and 30 ng/mL, and deficient 25-OH vitamin D
were excluded from this study. with a level <20 ng/mL.13 Dependent variables and outcome
measures were compared across the groups with insufficient
and deficient vitamin D levels. The model was adjusted for
Data Collection baseline characteristics that differed between the deficient,
insufficient, and normal vitamin D level groups with P < .05
The study included a total of 212 patients who were eligible
by univariate analysis. Statistical operations were performed
by inclusion and exclusion criteria. Data from eligible pa-
using SAS 9.4 (SAS, Cary, NC, USA).
tients’ charts were collected from electronic medical records
beginning from the time of each patient’s consent for LVAD
implantation until 1 year after LVAD implantation.
Demographic features including age, sex, race, and type
Results
of device were collected. 25-Hydroxy-vitamin D (25-OH From January 1, 2010, through January 1, 2015, 212 pa-
vitamin D) level was collected within a week of implantation tients with LVAD were eligible for chart review. Of these
or within the last hospitalization for pre-LVAD workup. patients, 29 were excluded from analysis because of missing
Other laboratory tests and biomarkers such as creatinine, preoperative 25-OH vitamin D levels. Data on the outcomes
blood urea nitrogen, calcium, serum albumin, and BNP measures were available for the entire population.
Obeid et al 827

Table 1. Demographics for the Study Population (N = 183). insufficient in 51 (27.87%), and deficient in 100 (54.64%).
Although patients with vitamin D deficiency were more
Demographics n %
likely to be African American than white, there were no
Race other significant differences in preoperative demographics
White 95 51.91 or laboratory values between the 3 groups (Table 3).
Black 75 40.98 A total of 102 (55.7%) patients remained in the hos-
Hispanic 7 3.82 pital >3 weeks after LVAD. The frequency of having ࣙ2
Asian 1 0.54 readmissions within the first year was higher in those pa-
Other 5 2.73 tients with vitamin D insufficiency or deficiency. In patients
Sex
Female 46 24.14
with normal vitamin D levels preoperatively, 50% had
Male 137 75.86 ࣙ2 admissions in the first year after LVAD implantation,
Age (Mean ± SD) 58.84 ± 13.43 compared with 70.6% in the insufficient and 71.0% in
Type of device the deficient groups, respectively (P = .0737) (Table 4). A
HeartMate II 146 79.79 logistic regression model determined that the risk for >2
HeartWare HVAD 37 20.21 readmissions in the first postoperative year was 2.46 times
higher for the vitamin D–deficient group and 2.5 times
higher for the vitamin D–insufficient group compared with
Table 2. Preoperative Laboratory Values for the Study the normal vitamin D group (adjusted OR: 2.46, 95% CI:
Population (N = 183). 1.067–5.769, and adjusted OR: 2.5, 95% CI: 0.97–6.433,
Laboratory Tests n Mean SD respectively).
Of the 183 patients in the study, 31 (16.9%) patients had
Vitamin D (ng/mL) 183 20.33 11.70 at least 1 episode of driveline infection in the first year
Parathyroid hormone (IU) 83 18.81 8.25 after LVAD. Of these 31 patients, 17 of 100 (17.2%) were
Calcium (mEq/L) 183 8.99 4.05 in the vitamin D–deficient category, 13 of 51 (26%) in the
Brain natriuretic peptide (pg/mL) 183 771.01 816.65
insufficient category, and only 1 of 32 (3.1%) in the normal
Serum albumin (g/dL) 183 2.89 0.52
Thyroid-stimulating hormone (mIU/mL) 183 3.88 8.77 category. A significantly larger percentage of patients who
Total bilirubin (mg/dL) 183 1.73 6.41 were vitamin D deficient had driveline infections compared
with those who had a normal vitamin D level (17.2% vs
3.1%, P = .027, respectively; Table 4). A logistic regression
model indicated that patients with vitamin D deficiency
Baseline characteristics for the study population are were at an 11.47 times greater risk for development of
summarized in Tables 1 and 2. The population was divided driveline infection compared with patients with normal
into 3 groups based on their 25-OH vitamin D level. 25- vitamin D level (95% CI: 1.204–109.26, P = .03), and that
OH vitamin D level was normal in 32 patients (17.49%), patients with vitamin D insufficiency were at 6.185 times

Table 3. Comparison of Demographic and Laboratory Data for Patients with Normal, Insufficient, and Deficient Preoperative
Vitamin D.

Preoperative Vitamin D

Demographics Normal (n = 32) Insufficient (n = 51) Deficient (n = 100) P Value

Race, n (%) .036


White 22 (22.68) 33 (34.00) 42 (43.30)
Black 10 (13.16) 16 (21.05) 50 (65.79)
Male sex, n 24 36 76 .768
Age, y 64.62 ± 10.35 59.22 ± 12.18 58.4 ± 13.53 .0532
White cell count, 1000/μL 7.441 ± 2.92 8.09 ± 3.00 8.50 ± 3.98 .297
Neutrophils, 1000/μL 5.17 ± 2.13 5.77 ± 2.55 5.89 ± 3.59 .504
Lymphocytes, 1000/μL 1.41 ± 1.49 1.52 ± 0.68 1.27 ± 0.67 .207
Sodium, mg/dL 135.13 ± 3.83 135.59 ± 3.42 134.77 ± 4.59 .240
Creatinine, mg/dL 1.74 ± 1.58 1.44 ± 0.42 1.64 ± 2.51 .551
Serum calcium, mEq/L 8.76 ± 0.40 8.73 ± 0.40 8.76 ± 68 .964
Serum albumin, g/dL 2.91 ± 0.51 2.98 ± 0.51 2.85 ± 0.50 .354
Total bilirubin, mg/dL 1.30 ± 1.17 1.12 ± 1.00 1.24 ± 1.38 .805
828 Nutrition in Clinical Practice 33(6)

Table 4. Comparison of Outcome Measures for Patients With Normal Preoperative Vitamin D, Insufficient Preoperative Vitamin
D, and Deficient Preoperative Vitamin D.

Preoperative Vitamin D

Outcome Measures Normal (n = 32) Insufficient (n = 51) Deficient (n = 100) P Value

1-Year mortality, n (%) 6 (18.75) 1 (1.96) 10 (10.00) .0349


Driveline infection, n (%) 1 (3.1) 13 (26.0) 17 (17.2) .027
Gastrointestinal bleed, n (%) 11 (34.38) 11 (21.57) 30 (30.0) .3950
Length of stay, n (%) 10 (31.25) 14 (27.45) 39 (39.0) .3381
Readmission,a n (%) 16 (50) 36 (70.59) 71 (71.0) .0737
Cerebrovascular accident, n (%) 3 (9.4) 2 (4.0) 10 (10.0) .430

All outcome measures indicate first year of postoperative left ventricular assist device support.
a ࣙ2 readmissions.

greater risk for having driveline infection when compared patients, it was found that 51 of 183 patients (27.88%)
with patients with a normal vitamin D level (95% CI: 0.80– were vitamin D insufficient and that 100 (54.64%) were
49.2, P = .07). deficient. Recognizing the wide range of pleiotropic effects
In their first year of LVAD support, 52 patients (28.4%) associated with vitamin D, we sought to investigate the role
had at least 1 episode of GI bleeding. Among those of hypovitaminosis D with several of the most common
with normal vitamin D levels, 11 (34.38%) experienced GI adverse events in patients with LVAD, including infection,
bleeding, whereas 51 (21.57%) and 30 (30%) of those with GI bleeding, and stroke.
insufficient and deficient levels experienced GI bleeding,
respectively. The rates of GI bleed were not significantly
different between these groups (Table 4). Driveline Infection
Finally, 15 patients (8.19%) experienced a cerebrovascu- Device-associated infection remains 1 of the most common
lar accident (CVA) within the first year. Among those with complications in long-term LVAD support with a preva-
normal vitamin D levels, 3 patients (9.38%) had a stroke, lence rate ranging up to 50% in the first 3 years after
whereas 2 (3.92%) and 10 (10%) of those in the insuffi- implantation.17 Infection is associated with higher mortality
cient and deficient groups, respectively, had a stroke. The and morbidity. Recent studies have elucidated the role of
rates of CVA were not significantly different between these vitamin D receptors in activating nearly all types of immune
groups. cells, including CD4+ and CD8+ T cells, B cells, neutrophils,
macrophage, and dendritic cells.18 In addition, as part of
adaptive immunity, vitamin D plays a major role in the pro-
Discussion liferation and differentiation of B and T cells, and modulates
Vitamin D deficiency is highly prevalent among patients immunoglobulin production.7 Further studies have revealed
with heart failure. The 3-year National Health and the importance of vitamin D in the endothelial response
Nutrition Examination Survey completed in 2004 reported to lipopolysaccharide (LPS). Specifically, endothelial cells
an 89% prevalence rate of hypovitaminosis D (defined as treated with 1,25(OH) vitamin D generated a significantly
vitamin D level <30 ng/mL), after controlling for age, race, lower response when stimulated with LPS compared with
and sex.13,14 A study of 154 patients with LVAD reported untreated cells.8 These findings, which indicate the impor-
34.4% had vitamin D levels <10 ng/mL, relatively lower tant role of vitamin D in immune system function, suggest
than the percentage of patients in our study with levels vitamin D depletion may be associated with greater risk
<10 ng/mL, which was 21.6%.15 for infections. Indeed, the recently published CALCITOP
Despite its prevalence in the heart failure population, few study, an analysis of 3340 cardiac surgery patients, identified
studies are analyzing vitamin D status and its relationship 2.57 times greater risk for postoperative infection in patients
to important clinical outcomes in patients with LVAD. A with vitamin D deficiency compared with those with higher
recent study analyzing patients undergoing elective cardiac levels.19
surgery for coronary artery bypass and valve replacement This trend was clearly seen in our LVAD population in
did not find a relationship between preoperative vitamin D those patients with deficient or insufficient vitamin D levels.
levels and several postoperative outcomes including length The risk for LVAD driveline infections, when compared
of stay.16 with patients with normal vitamin D level, was 6.185
Our study is the largest to evaluate the clinical impact times greater with vitamin D insufficiency and 11.467 times
of vitamin D levels in patients with LVAD. Among all greater with vitamin D deficiency.
Obeid et al 829

Readmission Rate with significantly higher risk for driveline infection and
readmission within 1 year. These effects may be a result of
Strategies to reduce hospital readmission rates are essential the ubiquitous presence of vitamin D receptors on immune
to improve quality of life for patients with LVAD. Multiple cells and the role these receptors play in leukocyte differ-
studies have identified several causes leading to recurrent entiation and proliferation. Consequently, vitamin D levels
readmissions. One large study showed that recurrent heart may alter the immune response to infectious organisms and
failure (33.3%) and GI bleeding (22.2%) were the most contribute to the risk for driveline infection.
common causes of LVAD readmissions.20 A separate study This study identified preoperative vitamin D depletion
of 126 patients indicated that GI bleeding and LVAD- as an important risk factor in those patients undergoing
related infections were the leading causes of readmissions.21 LVAD implantation. Further study is required to determine
Our analysis identified vitamin D level as an independent whether a preoperative repletion regimen will result in
predictor for increased postoperative readmission rate in protective effects or risk reduction. Limitations to this study
patients with LVAD. Patients with insufficient or deficient include that it was a single-center, retrospective study and
vitamin D levels had 2.5 times increased risk for being that longitudinal vitamin D levels were not available. In
readmitted in the year after LVAD implantation. A sig- addition, recent work has shown that vitamin D levels may
nificant percentage of hospital readmissions occur because be diluted in patients who receive bolus IV fluids, an effect
of driveline infections. The increased rate of infection in that may be significant in patients with advanced heart
patients with low vitamin D level may, in part, drive the failure. Future investigation should control for this potential
increased readmission rate in that group. confounding variable.28

Stroke Statement of Authorship


The presence of 25-OH vitamin D receptors within central F. A. Obeid, G. Bhat, E. Drever, and A. Tatooles contributed
nervous system tissues indicates the capability of vitamin to conception/design of the research; F. A. Obeid, G.
D to cross the blood–brain barrier and to interact with Yost, and G. Bhat contributed to acquisition, analysis, or
neural tissue. Several studies have suggested that vitamin interpretation of the data; F. A. Obeid and G. Yost drafted
D may have a neuroprotective effect, reducing the rate of the manuscript; F. A. Obeid, G. Yost, G. Bhat, E. Drever,
stroke and cognitive decline.22,23 In addition, data from and A. Tatooles critically revised the manuscript; and F.
prospective observational studies indicate that vitamin D A. Obeid, G. Yost, G. Bhat, E. Drever, and A. Tatooles
deficiency is an independent risk factor for stroke. In 1 meta- agree to be fully accountable for ensuring the integrity and
analysis, after adjusting for cardiovascular risk factors, the accuracy of the work. All authors read and approved the
risk for cerebrovascular disease was significantly reduced in final manuscript.
individuals with higher 25-OH vitamin D levels compared
with those who were deficient.22,23 Vitamin D levels have
also been shown to be associated with stroke severity and References
worsened early functional capacity after stroke.24-27 1. Shor R, Tirosh A, Shemesh L, et al. 25 Hydroxyvitamin D levels in
Known predictors of stroke in patients with LVAD patients undergoing coronary artery catheterization. Euro J Int Med.
include diabetes, aortic cross-clamping with cardioplegic 2012;23(5):470-473.
arrest, duration of LVAD support, and international nor- 2. Wang TJ, Pencina MJ, Booth SL, et al. Vitamin D deficiency and risk
of cardiovascular disease. Circulation. 2008;117(4):503-511.
malized ratio.27 Recently, a study by Zittermann et al,15
3. Kunadian V, Ford GA, Bawamia B, Qiu W, Manson JE. Vitamin D
analyzing 154 patients with LVAD, indicated that those with deficiency and coronary artery disease: a review of the evidence. Am
25-OH vitamin D level <10 ng/mL had an increased risk Heart J. 2014;167(3):283-291.
for stroke and mortality within 1 year of implantation. In 4. Liu L, Chen M, Hankins SR, et al; Drexel Cardiovascular Health
our study, despite the increased risk for readmission and Collaborative Education, Research and Evaluation Group. Serum 25-
hydroxyvitamin D concentration and mortality from heart failure
driveline study, there was no significant association between
and cardiovascular disease and premature mortality from all-cause in
vitamin D levels and risk for stroke. This discrepancy in United States adults. Am J Cardiol. 2012;110(6):834-839.
relationship of hypovitaminosis D to risk for postoperative 5. Liu LC, Voors AA, van Veldhuisen DJ, et al. Vitamin D status and
stroke may be because of the relatively larger number of outcomes in heart failure patients. Eur J Heart Fail. 2011;13:619-625.
patients with very low preoperative vitamin D levels in 6. Makariou SE, Michel P, Tzoufi MS, Challa A, Milionis HJ. Vitamin
D and stroke: promise for prevention and better outcome. Curr Vasc
Zittermann et al’s15 study compared with ours.
Pharmacol. 2014;12(1):117-124.
7. Bartley J. Vitamin D: emerging roles in infection and immunity. Expert
Conclusions Rev Anti Infect Ther. 2010;8:1359-1369.
8. Equils O, Naiki Y, Shapiro AM, et al. 1,25-Dihydroxyvitamin D
Our study suggests that deficient and insufficient levels of inhibits lipopolysaccharide-induced immune activation in human en-
25-OH vitamin D before LVAD implantation are associated dothelial cells. Clin Exp Immunol. 2006;143:58-64.
830 Nutrition in Clinical Practice 33(6)

9. Khoo AL, Chai LY, Koenen HJ, et al. 1,25-dihydroxyvitamin D3 19. Zittermann A, Kuhn J, Ernst JB, et al. Circulating 25-hydroxyvitamin
modulates cytokine production induced by Candida albicans: impact D and 1,25-dihydroxyvitamin D concentrations and postoperative
of seasonal variation of immune responses. J Infect Dis. 2011;203:122- infections in cardiac surgical patients: the CALCITOP-study. PLoS
130. ONE. 2016;11(6):e0158532.
10. Sayer G, Bhat G. The renin-angiotensin-aldosterone system and heart 20. Tsiouris A, Paone G, Nemeh HW, Brewer RJ, Morgan JA. Fac-
failure. Cardiol Clin. 2014;32(1):21-32. tors determining post-operative readmissions after left ventricular
11. Hasin T, Marmor Y, Kremers W, et al. Readmissions after implan- assist device implantation. J Heart Lung Transplant. 2014;33(10):1041-
tation of axial flow left ventricular assist device. J Am Coll Cardiol. 1047.
2013;61(2):153-163. 21. Akhter SA, Badami A, Murray M, et al. Hospital readmissions
12. Cotts WG, McGee EC Jr, Myers SL, et al. Predictors of hospital after continuous-flow left ventricular assist device implantation: inci-
length of stay after implantation of a left ventricular assist device: dence, causes, and cost analysis. Ann Thorac Surg. 2015;100(3):884-
an analysis of the INTERMACS registry. J. Heart Lung Transplant. 889.
2014;33(7):682-688. 22. Pilz S, Tomaschitz A, Drechsler C, Zittermann A, Dekker JM, März
13. Holick MF, Binkley NC, Bischoff-Ferrari HA, et al; Endocrine Society. W. Vitamin D supplementation: a promising approach for the pre-
Evaluation, treatment, and prevention of vitamin D deficiency: an vention and treatment of strokes. Curr Drug Targets. 2011;12:88-
Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 96.
2011;96(7):1911-1930. 23. Buell JS, Dawson-Hughes B. Vitamin D and neurocognitive dys-
14. Kim DH, Sabour S, Sagar UN, Adams S, Whellan DJ. Prevalence function: preventing “D” ecline? Mol Aspects Med. 2008;29:415-
of hypovitaminosis D in cardiovascular diseases (from the National 422.
Health and Nutrition Examination Survey 2001 to 2004). Am J Cardiol. 24. Sun Q, Pan A, Hu FB, Manson JE, Rexrode KM. 25-hydroxyvitamin
2008;102(11):1540-1544. D levels and the risk of stroke: a prospective study and meta-analysis.
15. Zittermann A, Morshuis M, Kuhn J, et al. Vitamin D metabolites Stroke. 2012;43:1470-1477.
and fibroblast growth factor-23 in patients with left ventricular assist 25. Chowdhury R, Stevens S, Ward H, Chowdhury S, Sajjad A, Franco
device implants: association with stroke and mortality risk. Eur J Nutr. OH. Circulating vitamin D, calcium and risk of cerebrovascular
2016;55(1):305-313. disease: a systematic review and meta-analysis. Eur J Epidemiol.
16. Sriram K, Perumal K, Alemzadeh, et al. The relationship between im- 2012;27:581-591.
mediate preoperative serum 25-hydroxy-vitamin D3 levels and cardiac 26. Daubail B, Jacquin A, Guilland JC, et al. Serum 25-hydroxyvitamin
function, dysglycemia, length of stay, and 30-d readmission in cardiac D predicts severity and prognosis in stroke patients. Eur J Neurol.
surgery patients. Nutrition. 2015;31:820-826. 2013;20:57-61.
17. Leuck AM. Left ventricular assist device driveline infections: recent 27. Morgan JA, Brewer RJ, Nemeh HW, et al. Stroke while on long-
advances and future goals. J Thorac Dis. 2015;7(12):2151-2157. term left ventricular assist device support: incidence, outcome, and
18. Baeke F, Takiishi T, Korf H, Gysemans C, Mathieu C. Vitamin D: predictors. ASAIO J. 2014;60(3):284-289.
modulator of the immune system. Curr Opin Pharmacol. 2010;10:482- 28. Sriram K, Meguid M. The ongoing story of vitamin D. Nutrition.
496. 2015;31:1293-1294.
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 6
Diagnostic Accuracy of Bioelectrical Impedance Analysis December 2018 831–842

C 2018 American Society for

Parameters for the Evaluation of Malnutrition in Patients Parenteral and Enteral Nutrition
DOI: 10.1002/ncp.10098
Receiving Hemodialysis wileyonlinelibrary.com

Angela Teodósio da Silva, MSc1 ; Daniela Barbieri Hauschild, MSc1 ;


1
Yara Maria Franco Moreno, PhD ; João Luiz Dornelles Bastos, PhD2 ;
and Elisabeth Wazlawik, PhD1

Abstract
Background: In the absence of a gold standard technique for assessing nutrition status in patients receiving hemodialysis
(HD), we aimed to determine the diagnostic accuracy of single-frequency (50 kHz) bioelectrical impedance analysis parameters,
resistance/height (R/H), reactance/height (Xc/H), and impedance/height (Z/H), and their cutoff points for malnutrition. Methods:
The reference standards, Subjective Global Assessment (SGA), Malnutrition Inflammation Score, and Nutritional Risk Screening
2002, were performed at baseline and then once a year for 2 years. At least 2 assessments for each reference standard were performed
during the monitoring period, and those patients who were assessed as malnourished on at least 2 consecutive occasions were
classified as malnourished. Results: A total 101 patients receiving HD were evaluated. R/H and Z/H demonstrated low to moderate
accuracy to diagnose malnutrition in men and low accuracy in women, whereas the accuracy of Xc/H was uncertain. The cutoff
points of bioelectrical impedance vector analysis (BIVA) parameters, determined based on the SGA to maximize sensitivity and
specificity simultaneously, were: R/H ࣙ330.05 and ࣙ420.92 ohms/m for men and women, respectively; Z/H ࣙ332.71 and ࣙ423.19
ohms/m for men and women, respectively. In men, sensitivity based on the cutoff points of R/H and Z/H together ranged from
73% to 89% and specificity ranged from 49% to 50%. In women, sensitivity ranged from 58% to 80% and specificity from 48% to
55%. Conclusion: BIVA parameters demonstrated low to moderate accuracy in men and low accuracy in women for the diagnosis
of malnutrition. (Nutr Clin Pract. 2018;33:831–842)

Keywords
bioelectrical impedance; chronic renal insufficiency; nutritional assessment; renal dialysis

Background Bioelectrical impedance analysis (BIA) is based on the


principle that body tissues offer different opposition to the
Protein-energy wasting is characterized as the depletion of passage of electrical current. Impedance (Z) refers to this
body protein and fat and is 1 of the most prevalent nutrition
disorders in patients with chronic kidney disease receiving
hemodialysis (HD).1 Moreover, this condition is associated From the 1 Federal University of Santa Catarina, Health Sciences
with an increase in morbidity and mortality rates as well as Center, Post-Graduate Program in Nutrition, Trindade Florianópolis,
Santa Catarina, Brazil; 2 Federal University of Santa Catarina, Health
reduction in physical function and quality of life.2,3
Sciences Center, Public Health Department, Trindade Florianópolis,
The nutrition status assessment and monitoring of pa- Santa Catarina, Brazil.
tients receiving HD are fundamental to prevention, diag-
Financial disclosures: This study was carried out with financial
nosis, and treatment of malnutrition.1 However, a precise support from the Brazilian governmental agency CAPES
assessment is complex due to frequent changes in the (Coordination for the Improvement of Higher Education Personnel)
body hydration, occurrence of comorbidities, and chronic by way of scholarships awarded to the students involved in the project.
inflammation.4,5 In the absence of a gold standard for Conflicts of interest: None declared.
the assessment of nutrition status, efforts have been made This article originally appeared online on May 22, 2018.
to identify indicators capable of an accurate diagnosis of
Corresponding Author:
malnutrition, such as handgrip strength (HGS), 50 kHz Elisabeth Wazlawik, PhD, Federal University of Santa Catarina,
phase angle, and 50 kHz bioelectrical impedance vector Health Sciences Center, Post-Graduate Program in Nutrition,
analysis (BIVA).5-9 However, few studies have evaluated the Campus Universitário, Trindade Florianópolis, Santa Catarina,
validity of each indicator and this remains unclear.10 Brazil 88040–970; 55-48-3721-5138.
Email: e.wazlawik@ufsc.br
832 Nutrition in Clinical Practice 33(6)

opposition and is composed of 2 vectors: resistance (R) and serum albumin level), and determine the prevalence of
and reactance (Xc).11 The R component of BIA represents malnutrition diagnosed using BIVA.
the opposition to flow of an alternating current through
ionic solutions and is inversely related to intracellular Materials and Methods
and extracellular water volume. Muscle tissue is highly
conductive of electrical current due to a large amount Study Design and Subjects
of water and electrolytes, and presents low resistance to A prospective, longitudinal, diagnostic validation study was
the passage of electric current.12 In situations of illness, a conducted with a convenience sample recruited from 2 HD
higher R value may be related to malnutrition,13 and so clinics. The sample consisted of 101 patients aged ࣙ19
a higher R value may be related to the lower amount of years, undergoing HD 3 times a week, and who agreed to
muscle mass and, consequently, of water and electrolytes. participate from April to September 2011, June to Septem-
Higher Xc values suggest a greater quantity of body cell ber 2012, and July to October 2013. Exclusion criteria
mass and lean body mass.6,11 The parameters R, Xc, and were patients receiving HD for <3 months; those with
Z were standardized by the height (H) in meters of each a BMI >34 kg/m²; patients with amputated or atrophied
patient (resistance/height [R/H], reactance/height [Xc/H], limbs; those wearing a pacemaker; patients with a current
and impedance/height [Z/H]) to account for the effect of diagnosis of cancer, stroke sequelae, or unable to respond;
the length of the conductor because the impedance of a and those hospitalized for any reason and not undergoing at
conductor is related to its length, cross-sectional area, and least 2 evaluations with the reference standards during the
frequency of the electrical current applied.14 The phase evaluation period.
angle is calculated based on R and Xc and is considered an Demographic and clinical data were obtained from the
indicator of both nutrition status and functional state; thus, medical record. In the beginning of the study, a nutrition
the phase angle determined at 50 kHz has been explored status assessment was performed after a session of HD
as a prognostic indicator in patients receiving HD.15,16 using different indicators: BIVA parameters (R/H, Xc/H,
The BIVA is based on the evaluation of the values of and Z/H), BMI, %FM, MUAC, MUAMC, and HGS. The
normalized R and Xc for H. After normalization, the BIVA reference standard indicators (SGA, MIS, and NRS 2002)
parameters (R/H, Xc/H, Z/H) are plotted as non-graph were determined at the beginning of the study as well as
resistance-reactance (RXc) vectors. The position is thought once a year throughout the follow-up period. The minimum
to provide information about hydration status, cell mass, follow-up period was 10 months (1 patient) and the mean
and cell membrane integrity.17 Some limitations of the use duration was 19 months (Figure 1). The clinical informa-
of BIA to evaluate nutrition status in clinical populations tion, the BIVA parameters, and the reference standards were
have been reported and have led researchers to explore the available to different evaluators, and the data were analyzed
use of raw impedance data in nutrition evaluation.18-20 by another appraiser without influence on data collection.
None of the nutrition indicators currently used are This study received approval by the local Research Ethics
considered complete for patients receiving HD.21 Besides Committee under process number No. 1821 and CAAE
BIA, the International Society of Renal Nutrition and 14375113.8.0000.0121; all participants signed a statement
Metabolism recommends the use of anthropometric in- of informed consent.
dicators, such as body mass index (BMI), percentage of
fat mass (%FM), mid-upper arm circumference (MUAC)
BIA
and mid-upper arm muscle circumference (MUAMC), and
biochemical indicators, such as serum albumin level.1 The SF-BIA was performed for the determination of R and Xc
single-frequency (SF) BIA approach is more affordable using a portable tetrapolar device (Biodynamics R
model
and available than bioimpedance spectroscopy. Considering 310e, Biodynamics Corporation, Seattle, WA) with a current
the scarcity of studies and the necessity to explore the intensity of 800 μA and at a single frequency of 50 kHz. The
application of BIVA parameters (R/H, Xc/H, Z/H) for electrodes were placed on the midline between the promi-
the evaluation of malnutrition in patients receiving HD, nent ends of the radius and ulna of the wrist and midline
this study aimed to determine the diagnostic accuracy of between the medial and lateral malleoli of the ankle. The
SF-BIA parameters in the evaluation of malnutrition in patients were evaluated approximately 20 minutes following
patients receiving HD using 3 reference standards: Subjec- HD on the side of the body without vascular access,22 as
tive Global Assessment (SGA), Malnutrition Inflammation recommended by the U.S. National Institutes of Health.23
Score (MIS), and Nutritional Risk Screening (NRS) 2002. The SF-BIA was chosen because it presents a single result
We also aimed to establish cutoff points for BIVA parame- for the entire body of R (ohm), Z (ohm), and Xc (ohm).
ters, investigate correlations with other clinically important
nutrition indicators (BMI, %FM, MUAC, MUAMC, HGS, BIA parameters. R (ohm) and Xc (ohm) to obtain the
Z (ohm)24 :
da Silva et al 833

Figure 1. Flowchart of sample selection process.


*Body mass index >34 kg/m2 , limbs amputated or atrophied, pacemaker, cancer, heart attack, inability to respond, hospitalized
or positive for acquired immunodeficiency syndrome.
**Death, underwent kidney transplant, change in treatment to peritoneal dialysis, transference from dialysis center.


Z= (R2 + Xc2 ). measurement parameter in the evaluation of the nutrition
status. They have the advantage of being easily applicable
in clinical practice and were used as a reference standard in
R, Xc, and Z were standardized by H in meters: R/H, Xc/H, another of our research studies.8 Patients were classified as
Z/H (ohm/m). malnourished when diagnosed with malnutrition at least 2
consecutive times by the same indicator during the follow-
Bioelectrical impedance vector analysis (BIVA 2002). The up period (2011–2013). The SGA was based on clinical
BIVA 2002 software developed by Piccoli and Pastori.11 history and clinical examination and classifies the patients
was used for the determination of BIVA, with the creation as: A) well nourished, B) moderately malnourished or sus-
of 50%, 75%, and 95% tolerance ellipses as well as confi- pected of being malnourished, or C) severely malnourished.
dence ellipses. The normalization of R and Xc by H was Categories B and C were grouped for the analysis.27 The
expressed as z-scores (z-R/H and z-Xc/H) with the reference MIS considers the SGA components as well as the duration
population described by Piccoli et al.25 Patients with vectors of HD, comorbidities, BMI, serum albumin level, and total
outside the 75% ellipsis in the upper quadrant (underweight) iron binding capacity.28 It was performed according to
and lower quadrant (cachexia) on the right side of the graph Kalantar-Zadeh et al, and the patients were classified as well
were classified as malnourished (Figure 2A). nourished (<6) and malnourished (ࣙ6).29 NRS 2002 was
Phase angle. R and Xc for the calculation of: evaluated according to Kondrup et al and the patients were
phase angle (◦ ) = arctangent [(Xc()/R()) × (180/π)]26 classified as without (<3) or at nutrition risk (ࣙ3).30

Nutrition Screening
We used 3 reference standards; 2 tools are used to do a
Anthropometric Variables
complete nutrition assessment, SGA and MIS, and 1 tool
is meant to identify nutrition risk, NRS 2002. The reference Weight (kg), H (m), MUAC (cm), and skin-fold thickness
standards are composite indicators, which include objective (mm) were determined after the session of HD. MUAC
and subjective criteria in the evaluation, and thus use >1 and skin-fold thickness were measured on the arm without
834 Nutrition in Clinical Practice 33(6)

Figure 2. (A) Graph with regions of elliptic probability on RXc plane normalized by height (R/H and Xc/H, in /m) 11 ;
(B) Position of vector mean for men (•) and women () after transformations of impedance measures into z-scores; (C)
Distribution of men on resistance-reactance graph; (D) Distribution of women on resistance-reactance graph; (E) Confidence
ellipses for men (black line) and women (dashed line).

vascular access. All data were collected by trained nutrition- suprailiac skin-fold measurements were performed follow-
ists following reference procedures.31 ing the recommendations proposed by Lohman et al.31
Weight (kg) was determined using an electronic scale %FM was determined using the formula described by Siri.33
(Marte R
, Marte Balanças e Aparelhos de Precisão Ltda, The equation proposed by Durnin and Womersley was used
Santa Rita do Sapucaı́, MG, Brazil) and height (cm) was de- to calculate body density.34
termined using a portable stadiometer (Sanny R
, American MUAC (cm) was measured using a flat steel measuring
Medical do Brasil, São Bernardo do Campo, SP, Brazil).32 tape (Cescorf 
R
, Cescorf Equipamentos para Esporte Ltda.,
Skin-fold thicknesses (mm) were determined using Porto Alegre, RS, Brazil) following the method described
calipers (Lange R
adipometer, Beta Technology Incorpo- by Frisancho.35 MUAMC was determined by the following
rated, Cambridge, MD). Biceps, triceps, subscapular, and equation:
da Silva et al 835

[MUAMC (cm) = MUAC (cm) − π × [triceps skin accuracy.42 Using the SGA as the reference standard, the
best cutoff points for R/H and Z/H were obtained for gender
fold (mm)/10]35 because it maximizes sensitivity and specificity simultane-
ously when compared with NRS 2002 and MIS.
HGS Using the cutoff of R/H and Z/H together, sensitivity,
specificity, positive predictive value (PPV) and negative
HGS (kg) was measured using a hydraulic dynamometer predictive value (NPV) were determined for the usefulness
(Saehan R
model SH 5001, Saehan Corporation, Yangdeok- of the BIVA parameters. Individuals with R/H and Z/H
Dong, Masan, Korea). The patients were instructed about below the cutoff points were considered nourished and
the use of the dynamometer; explanation and demonstra- those with R/H and Z/H above the cutoff points were con-
tion were given on the need to tighten the handle to their sidered malnourished. Statistical analyses were performed
full strength. At the moment of assessment, each participant using software (Stata version 11, Stata Corporation, College
was seated with hips and knees at 90° flexion, adducted Station, TX). The vector means of the resistance-reactance
shoulder close to the trunk, flexed elbow at 90°, with graph were analyzed using Hotelling’s T2 test and univariate
the forearm in neutral position (between supination and analysis (F test). P < 0.05 was considered statistically
pronation), and wrist between 0° and 30° of extension and significant.
0° and 15° of ulnar deviation.36
HGS was evaluated on the arm side without vascular
access. Three measurements were performed, with the pe- Results
riod of maximum continuous contraction of 3 seconds, A total of 101 individuals (60 men) participated in the
with a 30-second interval between trials.37 For the analysis, present study. Mean age was 53.5 ± 15.7 years (range,
the highest value of the 3 measurements was taken into 22–81 years) and median duration receiving HD was 40
consideration.38,39 months (range, 3 months–40 years). Almost one-third of
the patients (n = 28, 28%) were aged ࣙ65 years. Figure 1
Biochemical Variable displays the flow chart of the sample selection process.
The causes of chronic kidney disease were systemic arte-
Serum albumin level (g/dL) was determined using the col-
rial hypertension (42%), diabetes mellitus (24%), glomeru-
orimetric method with bromocresol green.40
lonephritis (13%), polycystic kidney disease (5%), and other
or undetermined causes (17%). Table 1 shows the main
Statistical Analysis clinical characteristics of the patients stratified by gender.
Description of the sample was performed by absolute The mean HGS, weight, height, and MUAMC were higher
and relative frequencies, mean and standard deviation for in men, while women showed higher %FM, R/H, Xc/H, and
parametric variables, or median and interquartile range for Z/H (P < 0.001).
non-parametric variables. To check the difference between The results of the BIVA are displayed in Figure 2. The
these variables according to gender, the χ 2 test was used vector mean for the women was near the 75% ellipsis of
for categoric variables and the Student’s t-test or Mann- tolerance in the lower right quadrant (malnourished), and
Whitney test was used for numeric variables. the vector mean for the men was near the 95% ellipsis of
Pearson (parametric variables) or Spearman (non- the lower right quadrant (malnourished; Figure 2B). Based
parametric variables) correlation was performed to assess on the resistance-reactance graph, the malnutrition rate was
the relationship between the BIVA parameters and other 71.6% (n = 43) among the men (Figure 2C) and 51.2% (n =
nutrition status indicators (SGA, MIS, NRS 2002, phase 21) among the women (Figure 2D). Figure 2E displays the
angle, BMI, %FM, MUAC, MUAMC, HGS, and serum confidence intervals of the R/H and Xc/H for the women
albumin level). For dichotomous variables (SGA, MIS, (ellipsis with dashed line) and men (ellipsis with black
NRS 2002): 0, malnourished; or 1, not malnourished. The line). The Hotelling test (T² = 54.7) and F test (F = 27.1)
coefficients were interpreted as follows: 0–0.29 = weak revealed significant differences between sexes (P < 0.001),
correlation; 0.30–0.69 = moderate correlation; and 0.70–1.0 demonstrating the importance of establishing specific cutoff
= strong correlation.41 points for the BIA variables (R/H, Xc/H, and Z/H) for each
To verify the diagnostic accuracy of BIVA parameters in sex. The prevalence of malnutrition (patients classified as
the identification of patients with malnutrition according to malnourished on at least 2 consecutive evaluations) based
SGA, MIS, and NRS 2002, receiver operating characteristic on SGA was 23.8% (n = 24) among the total sample, 23.3%
(ROC) curves were constructed and areas under the curve (n = 14) among the men, and 24.4% (n = 10) among the
(AUC) were obtained. When AUC was >0.90, accuracy was women. Based on the MIS, malnutrition was 26.7% (n = 27)
considered high, while 0.70–0.90 was considered moderate; among the total sample, 25.0% (n = 15) among the men,
0.50–0.69 was considered low and <0.50 meant uncertain and 29.2% among the women (n = 12), and based on the
836 Nutrition in Clinical Practice 33(6)

Table 1. Clinical Characteristics of Patients Receiving Dialysis Stratified by Gender: Florianópolis, Brazil, 2011–2012.

Characteristics Total (n = 101) Men (n = 60) Women (n = 41) P-Value

Age (years) 53.5 ± 15.7 53.2 ± 16.1 53.7 ± 15.5 0.884b


Nutrition variables
HGS (kg) 26.5 ± 11.3 31.0 ± 11.4 19.9 ± 7.20 <0.001c
Weight (kg) 65.4 ± 12.9 69.0 ± 11.5 60.2 ± 13.3 <0.001b
Height (m) 1.63 ± 0.10 1.68 ± 0.09 1.56 ± 0.09 <0.001b
BMI (kg/m²) 24.5 ± 3.62 24.4 ± 3.41 24.6 ± 3.95 0.715b
FM (%) 32.6 ± 6.54 30.7 ± 5.81 35.4 ± 6.63 <0.001b
MUAC (cm) 27.8 ± 4.04 27.5 ± 3.25 28.2 ± 5.01 0.756c
MUAMC (cm) 23.3 ± 3.00 23.9 ± 2.63 22.5 ± 3.32 0.012b
Phase angle (°) 6.35 ± 1.36 6.43 ± 1.26 6.24 ± 1.50 0.501b
Serum albumin level 4.00 ± 0.28 4.01 ± 0.34 3.99 ± 0.18 0.507c
BIA parameters
R/H (ohm/m) 372.4 ± 73.0 336.6 ± 53.0 424.8 ± 66.6 <0.001b
Xc/H (ohm/m) 41.3 ± 11.6 37.8 ± 8.73 46.6 ± 13.2 <0.001c
Z/H (ohm/m) 374.8 ± 73.4 338.8 ± 53.2 427.5 ± 67.0 <0.001b
Comorbidities (n, [%])*
Diabetes mellitus 30 (29.7) 15 (25.0) 15 (36.6) 0.211d
Systemic arterial hypertension 84 (83.2) 47 (78.3) 37 (90.2) 0.116d
Heart disease 27 (26.7) 15 (25.0) 12 (29.3) 0.634d
Kidney function
Time on dialysis (months)a 40.4 (16;84) 28.4 (12;76) 50.6 (29;97) 0.027c

BIA, bioelectrical impedance analysis; BMI, body mass index; FM, fat mass; HGS, handgrip strength; MUAC, mid-upper arm circumference;
MUAMC, mid-upper arm muscle circumference; R/H, resistance for height; Xc/H, reactance for height; Z/H, impedance for height.
*Absolute and relative frequency for categoric variables.
a Median and interquartile range.
b Student’s t-test.
c Mann-Whitney test.
d χ 2 test.

NRS 2002, it was 16.8% (n = 17) among the total sample, The AUCs for Z/H demonstrated low accuracy for the
15.0% (n = 9) among the men, and 19.5% (n = 8) among total sample, and the highest was that of the NRS 2002
the women. (0.68). For men, Z/H demonstrated moderate accuracy with
Positive correlations were found between R/H and Z/H the NRS 2002 (0.75) and low accuracy with the SGA and
with SGA (r = 0.20, P ࣘ 0.05) and NRS 2002 (dichotomous MIS. For women, the accuracy of Z/H was low for all
variables: 0, malnourished; or 1, not malnourished): (r = reference standards, with the highest AUC found for the
0.24, P <0.05). R/H and Z/H were negatively correlated with SGA (0.66; Table 3).
MUAC (r = −0.27, P <0.05), BMI (r = −0.31, P <0.05), Table 3 displays the cutoff points of R/H and Z/H. The
MUAMC (r = −0.48, P <0.05), and HGS (r = −0.61, P SGA was the reference standard for the classification of
<0.05). Xc/H exhibited a positive correlation with % FM malnutrition because it maximizes sensitivity and specificity
(r = 0.27, P <0.05) and phase angle (r = 0.72, P <0.05; simultaneously. The cutoff points established using SGA
Table 2). were as follows: R/H ࣙ 330.05 ohms/m for the total sample
The AUC for R/H demonstrated low accuracy for the and for men, and ࣙ 420.92 ohms/m for women; Z/H
total sample, and the highest was the AUC of the NRS 2002 ࣙ340.47 ohms/m for the total sample, ࣙ 332.71 ohms/m for
(0.68). For men, R/H demonstrated moderate accuracy with men, and ࣙ423.19 ohms/m for women.
the NRS 2002 (0.75) and low accuracy with the SGA and Figure 3 shows the ROC curves with the values of
MIS. For women, the accuracy of R/H was low using all diagnostic accuracy of R/H and Z/H in the identification
3 reference standards, with the highest AUC found for the of men and women with malnutrition according to SGA,
SGA (0.66; Table 3). where R/H and Z/H showed low accuracy among women
The AUCs for Xc/H demonstrated accuracy <50%, and men.
ranging from 42% to 46% for the total sample, 43% to The estimated prevalence of malnutrition based on the
49% for men, and 33% to 44% for women, demonstrating cutoff points of R/H and Z/H together, established using the
uncertain accuracy (data not shown). SGA, was 65.4% (n = 66) among the total sample, 56.7%
da Silva et al 837

Table 2. Correlation Between Bioelectrical Impedance Analysis Parameters and Nutrition Indicators Among Patients Receiving
Hemodialysis: Florianópolis, Brazil, 2011–2013.

Parameter R/H P Xc/H P-Value Z/H P-Value

SGA 0.1975 0.048a –0.085 0.398a 0.1955 0.050a


MIS 0.1661 0.097a –0.068 0.500a 0.1657 0.098a
NRS 2002 0.2364 0.017a –0.1048 0.297a 0.2351 0.0180a
Phase angle (°) –0.0701 0.486b 0.7161 <0.001b –0.0560 0.578b
BMI (kg/m²) –0.3156 0.001b –0.0798 0.428b –0.3135 0.001b
FM (%) 0.0990 0.325b 0.2664 0.007b 0.1024 0.308b
MUAC (cm) –0.2746 0.006b 0.1092 0.277b –0.2698 0.006b
MUAMC (cm) –0.4886 <0.001b –0.0402 0.690b –0.4841 <0.001b
HGS (kg) –0.6169 <0.001b –0.1492 0.137b –0.6129 <0.001b
Serum albumin level (g/dL) 0.0011 0.991b 0.0738 0.466b 0.0026 0.979b

BMI, body mass index; FM, fat mass; H, height; HGS, handgrip strength; MIS, Malnutrition Inflammation Score; MUAC, mid-upper arm
circumference; MUAMC, mid-upper arm muscle circumference; NRS 2002, Nutritional Risk Screening 2002; R, resistance; SGA, Subjective
Global Assessment; Xc, reactance; Z, impedance.
a Spearman correlation; significant values in bold (P ࣘ 0.05).
b Pearson correlation; significant values in bold (P ࣘ 0.05).

Table 3. Resistance/Height (R/H) and Impedance/Height (Z/H) Cutoff Points for Identification of Nourished and Malnourished
Patients Based on SGA, MIS, and NRS 2002, Stratified by Gender.

Reference Standard Sensitivitya Specificitya AUCa Cutoff Point (ohm/m)

Total sample (n = 101) R/H


SGA 87.50 32.37 63.39 ࣙ330.05
MIS 88.89 31.08 60.84 ࣙ326.18
NRS 2002 94.12 32.14 68.24 ࣙ330.05
Men (n = 60) R/H
SGA 78.57 50.00 66.23 ࣙ330.05
MIS 80.00 46.67 64.37 ࣙ326.18
NRS 2002 100.0 33.33 75.49 ࣙ317.65
Women (n = 41) R/H
SGA 80.00 54.84 66.13 ࣙ420.92
MIS 58.33 72.41 56.90 ࣙ437.18
NRS 2002 62.50 72.73 64.39 ࣙ443.40
Total sample (n = 101) Z/H
SGA 83.33 40.26 63.26 ࣙ340.47
MIS 88.89 31.08 60.81 ࣙ329.24
NRS 2002 94.12 32.14 68.14 ࣙ332.71
Men (n = 60) Z/H
SGA 78.57 50.00 65.84 ࣙ332.71
MIS 80.00 46.67 64.00 ࣙ329.24
NRS 2002 100.0 33.33 74.95 ࣙ319.29
Women (n = 41) Z/H
SGA 80.00 54.84 66.13 ࣙ423.19
MIS 58.33 72.41 56.90 ࣙ438.92
NRS 2002 62.50 72.73 64.39 ࣙ445.00

AUC, area under curve; MIS, Malnutrition Inflammation Score; NRS 2002, Nutritional Risk Screening 2002; SGA, Subjective Global
Assessment.
a Data expressed as percentage.

(n = 34) among the men, and 53.7% (n = 22) among the gether established for the identification of malnutrition. In
women. the total sample, the sensitivity ranged from 78% to 88%,
Table 4 shows the diagnostic accuracy of the BIVA specificity 39% to 40%, PPV 23% to 32%, and NPV 38%
parameters based on the R/H and Z/H cutoff points to- to 94%. In men, sensitivity ranged from 73% to 89% and
838 Nutrition in Clinical Practice 33(6)

Figure 3. Receiver operating characteristic (ROC) curves of R/H with ASG in men (A) and women (B), and of Z/H in men (C)
and women (D).

specificity from 49% to 50%. In women sensitivity ranged membranes, and organelles, and it is thought by some to be
between 58% and 80%, and specificity between 48% and related to the amount of soft tissue structures.11
55%. Based on the difference in the mean SF-BIVA parameters
(R/H, Xc/H, and Z/H) between men and women, and the
expression of sexual dimorphism in body composition, the
Discussion highest values of R/H, Xc/H, and Z/H in the women in our
In this study we compared BIA with a risk tool and assess- sample were indicative of a higher amount of %FM than
ment tools. Screening tools, such as NRS 2002, indicated in men (P < 0.001). Highest values of the BIVA parameters
risk factors for a deprived nutrition condition. Also, these were similar to those of the healthy American population,7
tools are the first step in determining nutrition problems,43,44 Italian elderly individuals,45 Indians with chronic kidney
and assessment tools (SGA and MIS) allow the clinician disease,46 and elderly patients hospitalized in the United
to formulate a nutrition diagnosis.44 Our results show that Kingdom.13
BIVA parameters RH and Z/H offer low to moderate Phase angle appeared to be higher in males than in
accuracy in men and low accuracy in women for the diag- females, although this did not reach statistical significance.
nosis of malnutrition using the risk and assessment tools The higher phase angle in men has been reported in the
as reference standards. BIVA parameters were correlated literature,7,13,45-47 suggesting a higher amount of body cell
with SGA, NRS 2002, anthropometric variables, HGS, and mass,17 and particularly greater skeletal muscle mass.7 In
phase angle. R/H and Z/H together carry high sensitivity our sample, males had higher mean values of MAUMC (P
but low specificity compared with the reference standards. = 0.021) and HGS (P <0.001) than women, which may be
Z is a combination of R and Xc. R is the opposition related to the greater amount of muscle mass, resulting in
to flow of an alternating current through intracellular and greater muscle strength.8
extracellular ionic solutions. Xc has been suggested to A weak positive correlation was found between the
represent the capacitive component of tissue interfaces, cell BIVA parameters R/H and Z/H and the reference standards
da Silva et al 839

Table 4. Diagnostic Accuracy of Bioelectrical Impedance Analysis Parameters (R/H and Z/H) for Identification of Nourished
and Malnourished Patients Based on SGA, MIS, and NRS 2002, Stratified by Gender.

Reference Standard Sensitivitya Specificitya PPVa NPVa

Total sample (n = 101)


(R/H ࣙ330.05; Z/H ࣙ 340.47)
SGA
Value 83.33 40.26 30.30 88.57
95% CI 62.62–95.26 29.23–52.06 19.59–42.85 73.26–96.80
MIS
Value 77.78 39.19 31.82 82.86
95% CI 57.74–91.38 28.04–51.23 20.89–44.44 66.35–93.44
NRS 2002
Value 88.24 39.29 22.73 94.29
95% CI 63.56–98.54 28.80–50.55 13.31– 34.70 80.84–99.30
Men (n = 60)
(R/H ࣙ330.05; Z/H ࣙ332.71)
SGA
Value 78.57 50.00 32.35 88.46
95% CI 49.20–95.34 34.90–65.10 17.39–50.53 69.85–97.55
MIS
Value 73.33 48.89 32.35 84.62
95% CI 44.90–92.21 33.70–64.23 17.39–50.53 65.13–95.64
NRS 2002
Value 88.89 49.02 23.53 96.15
95% CI 51.75–99.72 34.75–63.40 10.75– 41.17 80.36–99.90
Women (n = 41)
(R/H ࣙ420.92; Z/H ࣙ423.19)
SGA
Value 80.00 54.84 36.36 89.47
95% CI 44.39–97.48 36.03–72.68 17.20–59.34 66.86–98.70
MIS
Value 58.33 48.28 31.82 73.68
95% CI 27.67–84.83 29.45–67.47 13.86–54.87 48.80–90.85
NRS 2002
Value 62.50 48.48 22.73 84.21
95% CI 24.49–91.48 30.80–66.46 7.82–45.37 60.42–96.62

CI, confidence interval; MIS, Malnutrition Inflammation Score; NPV, negative predictive value; NRS, Nutritional Risk
Screening; PPV, positive predictive value; R/H, resistance/height; SGA, Subjective Global Assessment; Z/H, impedance/height.
a Data expressed as percentage.

SGA and NRS 2002. Moreover, a weak/moderate negative and the phase angle is calculated based on the Xc/R
correlation was found between anthropometric variables ratio.6
and HGS. In an elderly Spanish population with dementia, The AUC of the cutoff points of R/H, Xc/H, and Z/H for
R/H and Z/H were found to have a moderate negative the evaluation of malnutrition ranged from 0.63 to 0.68 (low
correlation with BMI only in women.48 Regarding the accuracy), 0.42 to 0.46 (uncertain accuracy), and 0.6 to 0.68
correlations between BMI and both R/H and Z/H, the (low accuracy), respectively, in the total sample. A similar
data observed in this study are in agreement with data finding was reported in a study involving 1590 American
reported for the American population.7 In the present adults in which the accuracy of the BIVA for the evaluation
study, Xc/H demonstrated a weak positive correlation with of %FM (AUC: 0.49–0.61) was tested using dual-energy X-
%FM. In the American population, a weak and moderate ray absorptiometry.7 In the present study, the accuracy of
negative correlation was found between Xc/H and %FM Xc/H for the identification of malnutrition was <50% in
in men and women, respectively, and the same result was the total sample as well as for men and women separately.
found regarding correlations between %FM and both R/H In a recent study, age was the most important determinant
and Z/H.7 The strong positive correlation between Xc/H of phase angle variation, and a greater age was associated
and the phase angle may be explained by the fact that with a lower phase angle in 1442 healthy subjects.49 Thus,
both parameters reflect the integrity of cell membranes, the reduction in Xc seems to be physiologic, accompanying
840 Nutrition in Clinical Practice 33(6)

the aging process. Because approximately one-third (n = excluded for not undergoing at least 2 evaluations with
28) of the patients in the present investigation were aged the reference standards may have diminished the statistical
ࣙ65 years, it is likely that this factor exerted an influence power of some results. However, the age and gender of
on the results.13 It is possible that the vector length by the individuals who remained in the study were similar to
BIVA allows a better comprehension of body composition those who were excluded. Nonetheless, comparisons of the
than does the consideration of only PA independent of data should be made with caution, considering adult and
vector length, or the resistance component independent of elderly patients undergoing HD 3 times a week. Moreover,
reactance.50 Moreover, it may be suggested that the occur- the sociodemographic characteristics and health conditions
rence of comorbidities, such as diabetes and high blood of the patients evaluated should be taken into account.
pressure, also contributes to the reduction in Xc/H. Previous The validity of the present findings is strengthened by the
studies with specific BIVA,48 classic BIVA,51 and both52 fact that all researchers were trained for the establishment
with elderly individuals report a significant difference in R of intraexaminer and interexaminer agreement and the ad-
values between well-nourished and malnourished patients, equate standardization of the anthropometric measures and
with the malnourished demonstrating higher values of R, administration of the reference standards for the screening
whereas no difference was found in Xc.48,51,52 of malnutrition.
This is the first study to define cutoff points of the To the best of our knowledge, there are no previous
R/H and Z/H parameters of BIA for the identification of prospective longitudinal studies on the diagnostic accuracy
malnutrition in patients undergoing HD. The SGA was of BIVA parameters (R/H, Xc/H). Thus, the present in-
the reference standard based on the highest sensitivity and vestigation is important because the patients classified as
specificity values. The U.S. National Kidney Foundation, malnourished based on the SGA, MIS, and NRS 2002 had
Kidney Disease/Dialysis Outcomes and Quality Initiative at least 2 consecutive evaluations in which the classification
has recommended the use of the SGA for the evaluation of of malnutrition was given by the same indicator during
nutrition status in patients receiving HD.53 The SGA was the study follow-up, which is strong evidence that these
used as the reference standard in 1 other study that aimed individuals truly were and remained malnourished.
to evaluate the diagnostic accuracy of the BIVA for the
evaluation of malnutrition in this group of patients.5 Conclusions
In the present study, the BIVA parameters R/H and
This study showed BIVA parameters R/H and Z/H offer
Z/H demonstrated greater sensitivity than specificity in the
low to moderate accuracy in men and low accuracy in
identification of malnutrition. Sensitivity ranged from 78%
women for the diagnosis of malnutrition using SGA as a
to 88% in the total sample, which is similar to that described
reference standard. However, BIVA parameters were cor-
in a study conducted in Italy with patients receiving HD.5
related with SGA, NRS 2002, anthropometric variables,
In contrast, specificity ranged from 39% to 40%, which
HGS, and phase angle. R/H and Z/H carry high sensitivity
differs significantly from the figure reported in the Italian
but low specificity compared with the reference standards.
study on the use of BIVA for the diagnosis of malnutri-
Further studies using the specific BIVA approach can be
tion in patients undergoing HD (79%).5 A sensitive test
performed. The validation with physiologically relevant
needs to be chosen when the consequences of overlooking
reference standards (eg, dual-energy X-ray absorptiometry,
a diagnosis are considerable.54 Malnutrition is common
lean tissue measures) is necessary to determine if the BIVA
in patients with chronic kidney disease, and it has been
parameters R/H, Xc/H, and Z/H are appropriate bedside
identified as an important risk factor for complications and
tools for the assessment of nutrition status receiving HD
mortality.53 Thus, it is important to identify patients who
patients.
are truly malnourished. Accurate diagnosis can lead to a
prompt nutrition intervention to minimize the occurrence Acknowledgments
of clinical complications and lower the risk of death. BIA
We are grateful to the patients and staff of Apar Vida and
parameters are potential prognostic indicators, as changes
Clinirim clinics in the region of Florianópolis, Southern Brazil,
on the cellular level may occur prior to anthropometric and
where data collection was performed, and the Coordination for
biochemical changes,55 and they could be an important tool the Improvement of Higher Education Personnel (CAPES) for
for evaluating clinical outcome or for monitoring disease the scholarship to the researcher.
progression.47
The SGA is not a specific indicator for patients on HD, Statement of Authorship
but it has been used in different studies involving HD A. T. da Silva, D. B. Hauschild, E. Wazlawik, and Y. M. Franco
and is recommended for this population.53 SGA assesses Moreno contributed to conception/design of the research; A.
subjective aspects of nutrition status that are different T. da Silva, D. B. Hauschild, E. Wazlawik, J. L. Dornelles
from the objective BIVA parameters, and that can be a Bastos, and Y. M. Franco Moreno contributed to acquisition,
limitation. The small sample size and the number of patients analysis, or interpretation of the data; A. T. da Silva and
da Silva et al 841

E. Wazlawik drafted the manuscript; A. T. da Silva, D. B. 17. Piccoli A, Piazza P, Noventa D, Pillon L, Zaccaria M. A new method
Hauschild, Y. M. Franco Moreno, J. L. Dornelles Bastos, and for monitoring body fluid variation by bioimpedance analysis: the RXC
E. Wazlawik critically revised the manuscript; A. T. da Silva, graph. Kidney Int. 1994;46(2):534-539.
D. B. Hauschild, Y. M. Franco Moreno, J. L. Dornelles Bastos, 18. Kuchnia AJ, Teigen LM, Cole AJ, et al. Phase angle and impedance
ratio: reference cut-points from the United States National Health
and E. Wazlawik agree to be fully accountable for ensuring
and Nutrition Examination Survey 1999–2004 from bioimpedance
the integrity and accuracy of the work. All authors read and
spectroscopy data (published online ahead of print September
approved the final manuscript. 26, 2016). JPEN J Parenter Enteral Nutr. 2016. https://www.
ncbi.nlm.nih.gov/pubmed/27670250. Accessed April 17, 2017.
References 19. Bosy-Westphal A, Danielzik S, Dorhofer R-P, Later W, Wiese S,
1. Fouque D, Kalantar-Zadeh K, Kopple J, et al. A proposed nomen- Muller M. Phase angle from bioelectrical impedance analysis: popula-
clature and diagnostic criteria for protein–energy wasting in acute and tion reference values by age, sex, and body mass index. JPEN J Parenter
chronic kidney disease. Kidney Int. 2008;73(4):391-398. Enteral Nutr. 2006;30(4):309-316.
2. Beberashvili I, Sinuani I, Azar A, et al. Serum uric acid as a clinically 20. Mulasi U, Kuchnia AJ, Cole AJ, Earthman CP. Bioimpedance at the
useful nutritional marker and predictor of outcome in maintenance bedside: current applications, limitations, and opportunities. Nutr Clin
hemodialysis patients. Nutrition. 2015;31(1):138-147. Pract. 2015;30(2):180-193.
3. Komatsu M, Okazaki M, Tsuchiya K, Kawaguchi H, Nitta K. Geriatric 21. Fürstenberg A, Davenport A. Comparison of multifrequency bio-
Nutritional Risk Index is a simple predictor of mortality in chronic electrical impedance analysis and dual-energy X-ray absorptiometry
hemodialysis patients. Blood Purif. 2015;39(4):281-287. assessments in outpatient hemodialysis patients. Am J Kidney Dis.
4. Mancini A, Grandaliano G, Magarelli P, Allegretti A. Nutritional 2011;57(1):123.
status in hemodialysis patients and bioimpedance vector analysis. J Ren 22. Di Iorio BR, Scalfi L, Terracciano V, Bellizzi V. A systematic eval-
Nutr. 2003;13(3):199-204. uation of bioelectrical impedance measurement after hemodialysis
5. Piccoli A, Codognotto M, Piasentin P, Naso A. Combined evalua- session. Kidney Int. 2004;65(6):2435-2440.
tion of nutrition and hydration in dialysis patients with bioelectrical 23. National Institute of Health (NIH). Bioelectrical impedance analysis
impedance vector analysis (BIVA). Clin Nutr. 2014;33(4):673-677. in body composition measurement: National Institutes of Health tech-
6. Norman K, Stobäus N, Pirlich M, Bosy-Westphal A. Bioelectrical nology assessment conference statement. Am J Clin Nutr. 1996;64(3
phase angle and impedance vector analysis—clinical relevance and Suppl.):524S-532S.
applicability of impedance parameters. Clin Nutr. 2012;31(6):854-861. 24. Kushner RF. Bioelectrical impedance analysis: a review of principles
7. Buffa R, Saragat B, Cabras S, Rinaldi AC, Marini E. Accuracy of and applications. J Am Coll Nutr. 1992;11(2):199-209.
specific BIVA for the assessment of body composition in the United 25. Piccoli A, Pillon L, Dumler F. Impedance vector distribution by sex,
States population. PLoS One. 2013;8(3):e58533. race, body mass index, and age in the United States: standard reference
8. Garcia MF, Wazlawik E, Moreno YMF, Führ LM, González-Chica intervals as bivariate Z scores. Nutrition. 2002;18(2):153-167.
DA. Diagnostic accuracy of handgrip strength in the assessment of 26. Baumgartner RN, Chumlea WC, Roche AF. Bioelectric impedance
malnutrition in hemodialyzed patients. e-SPEN Journal. 2013;8:e181- phase angle and body composition. Am J Clin Nutr. 1988;48:
e186. 6-43.
9. Buffa R, Mereu E, Comandini O, Ibanez ME, Marini E. Bioelec- 27. Detsky AS, McLaughlin JR, Baker JP, et al. What is subjective
trical impedance vector analysis (BIVA) for the assessment of two- global assessment of nutritional status? JPEN J Parenter Enter Nutr.
compartment body composition. Eur J Clin Nutr. 2014;68(11):1234- 1987;11:8-13.
1240. 28. Kalantar-Zadeh K, Kopple JD, Block G, Humphreys MH. A
10. Locatelli F, Fouque D, Heimbürguer O, et al. Nutritional status malnutrition-inflammation score is correlated with morbidity and
in dialysis patients: a European consensus. Nephrol Dial Transplant. mortality in maintenance hemodialysis patients. Am J Kidney Dis.
2002;17(4):563-572. 2001;38(6):1251-1263.
11. Piccoli A, Pastori G. BIVA software. In: Sciences DoMaS. Padova, 29. Yamada K, Furuya R, Takita T, et al. Simplified nutritional screening
Italy: University of Padova; 2002. tools for patients on maintenance hemodialysis. Am J Clin Nutr.
12. Eickemberg M, Oliveira CC, Roriz AKC, Sampaio LR. Bioelec- 2008;87:106-113.
tric impedance analysis and its use for nutritional assessments 30. Kondrup J, Rasmussen HH, Hamberg O, Stanga Z; Ad Hoc
(Bioimpedância elétrica e sua aplicação em avaliação nutricional). Rev ESPEN Working Group. Nutritional risk screening (NRS 2002): a new
Nutr. 2011;24(6):873-882. method based on an analysis of controlled clinical trials. Clin Nutr.
13. Slee A, Birc D, Stokoe D. Bioelectrical impedance vector analysis, 2003;22(3):321-336.
phase-angle assessment and relationship with malnutrition risk in 31. Lohman, TG. Advances in body composition assessment. IL: Human
a cohort of frail older hospital patients in the United Kingdom. Kinetics Publishers; 1992.
Nutrition. 2015;31(1):132-137. 32. World Health Organization. Global database on Body Mass Index
14. Pupim LCB, Ribeiro CB, Kent P, et al. Update on dialysis: Use of http://apps.who.int/bmi/index.jsp?introPage=intro_3.html, 2008.
bioelectrical impedance analysis in dialysis patients (Atualização em 33. Siri WE. Body composition from fluid spaces and density: analysis
diálise: Uso da Impedãncia Bioelétrica em pacientes em diálise). J Bras of methods. In: Brozek J, Henschel A. Techniques for Measuring
Nefrol. 2000;22(4):249-256. Body Composition. Washington DC: National Academy of Sciences;
15. Maggiore Q, Nigrelli S, Ciccarelli C, Grimaldi C, Rossi GA, Michelassi 1961:223-224.
C. Nutritional and prognostic correlates of bioimpedance indexes in 34. Durnin JV, Womersley J. Body fat assessed from total body density
hemodialysis patients. Kidney Int. 1996;50(6):2103-2108. and its estimation from skinfold thickness: measurements on 481
16. Segall L, Moscalu M, Hogaş S, et al. Protein-energy wasting, as men and women aged from 16 to 72 years. Br J Nutr. 1974;32:
well as overweight and obesity, is a long-term risk factor for mor- 77-97.
tality in chronic hemodialysis patients. Int Urol Nephrol. 2014;46(3): 35. Frisancho AR. Triceps skin-fold and upper arm muscle size norms for
615-621. assessment of nutritional status. Am J Clin Nutr. 1974;27:1052-1058.
842 Nutrition in Clinical Practice 33(6)

36. Leal VO, Mafra D, Fouque D, Anjos LA. Use of handgrip strength 46. Jha V, Jairam A, Sharma MC, et al. Body composition analysis with
in the assessment of the muscle function of chronic kidney disease bioelectric impedance in adult Indians with ESRD: comparison with
patients on dialysis: a systematic review. Nephrol Dial Transplant. healthy population. Kidney Int. 2006;69(9):1649-1653.
2010;26(4):1354-1360. 47. Barbosa-Silva MC, Barros AJ, Wang J, Heymsfield SB, Pierson RN Jr.
37. Headley S, Germain M, Mailloux P, et al. Resistance training improves Bioelectrical impedance analysis: population reference values for phase
strength and functional measures in patients with end-stage renal angle by age and sex. Am J Clin Nutr. 2005;82(1):49-52.
disease. Am J Kidney Dis. 2002;40(2):355-364. 48. Camina Martı́n MA, de Mateo Silleras B, Barrera Ortega S,
38. Leal VO, Stockler-Pinto MB, Farage NE, et al. Handgrip strength Domı́nguez Rodrı́guez L, Redondo del Rı́o MP. Specific bioelectrical
and its dialysis determinants in hemodialysis patients. Nutrition. impedance vector analysis (BIVA) is more accurate than classic BIVA
2011;27(11-12):1125-1129. to detect changes in body composition and in nutritional status in
39. Schlüssel MM, Anjos LA, Vasconcellos MT, Kac G. Reference values institutionalised elderly with dementia. Exp Gerontol. 2014;57:264-271.
of handgrip dynamometry of healthy adults: a population-based study. 49. Gonzalez MC, Barbosa-Silva TG, Bielemann RM, Gallagher D,
Clin Nutr. 2008;27(4):601-607. Heymsfield SB. Phase angle and its determinants in healthy subjects:
40. Doumas BT, Watson WA, Biggs HG. Albumin standards and the influence of body composition. Am J Clin Nutr. 2016;103:712-716.
measurement of serum albumin with bromcresol green. Clin Chim 50. Mereu E, Buffa R, Lussu P, Marini E. Phase angle, vector
Acta. 1971;31(1):87-96. length, and body composition. Am J Clin Nutr. 2016;104(3):845-847.
41. Arango HG. Bioestatı́stica: Teórica e Computacional. 2nd ed. Rio de https://doi.org/10.3945/ajcn.116.137513.
Janeiro: Guanabara Koogan; 2005. 51. Norman K, Smoliner C, Valentini L, Lochs H, Pirlich M. Is bioelectri-
42. Swets JA. Measuring the accuracy of diagnostic systems. Science. cal impedance vector analysis of value in the elderly with malnutrition
1988;240(4857):1285-1293. and impaired functionality? Nutrition. 2007;23(7-8):564-569.
43. Charney P. Nutrition screening vs nutrition assessment: how do 52. Marini E, Buffa R, Saragat B, et al. The potential of classic and
they differ? Nutr Clin Pract. 2008;23(4):366-372. https://doi.org/ specific bioelectrical impedance vector analysis for the assessment of
10.1177/0884533608321131. sarcopenia and sarcopenic obesity. Clin Interv Aging. 2012;7:585-591.
44. Correia MITD. Nutrition screening vs nutrition assessment: what’s 53. National Kidney Foundation. K/DOQI clinical practice guidelines for
the difference? Nutr Clin Pract. 2017;(1):https://doi.org/10.1177/ nutrition in chronic renal failure. Am J Kidney Dis. 2000;35(2):S1-S140.
0884533617719669. 54. Fletcher RH, Fletcher SW, Fletcher GS. Diagnóstico. In: Epidemiolo-
45. Saragat B, Buffa R, Mereu E, et al. Specific bioelectrical impedance gia Clı́nica: Elementos Essenciais. Porto Alegre: Artmed; 2014:116-139.
vector reference values for assessing body composition in the Italian 55. Barbosa-Silva MC. Subjective and objective nutritional assessment
elderly. Exp Gerontol. 2014;50:52-56. methods. Curr Opin Clin Nutr Metab Care; 2008;11(3):248-254.
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 6
Comparison of Two Methods for Estimating the Tip Position December 2018 843–850

C 2018 American Society for

of a Nasogastric Feeding Tube: A Randomized Controlled Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10112
Trial wileyonlinelibrary.com

Tim Torsy, RN, MSc1 ; Renée Saman, RN, CNS, MSc1,2 ; Kurt Boeykens, RN, CNS,
MSc2 ; Ivo Duysburgh, MD2 ; Nele Van Damme, RN, MSc3 ;
and Dimitri Beeckman, RN, MSc, PhD3

Abstract
Background: Several studies have shown that the nose-earlobe-xiphoid distance (NEX) is inadequate to estimate the insertion length
of nasogastric tubes. An alternative approach tested in these studies, which leads to > 90% well-placed nasogastric tubes, used a
corrected calculation of the NEX: (NEX × 0.38696) + 30.37. The primary aim of this study was to determine whether using the
corrected NEX was more successful than the NEX in determining the insertion length. The secondary aim was to investigate the
likelihood to successfully obtain gastric aspirate. Methods: Adult patients in a general hospital (N = 215) needing a nasogastric tube
were randomized to the control (NEX) or intervention group (corrected NEX). Tip positioning was verified using X-ray. Correct
tip positioning was defined as between 3 and 10 cm under the lower esophageal sphincter (LES). Results: In >20% of all patients,
both methods underestimated the required tube length for correct positioning. The NEX showed an overestimation (17.2%) of the
insertion length (>10 cm under the LES) compared with the corrected NEX (4.8%). There was no difference (P = 0.938) between the
2 groups in obtaining gastric aspirate (55.6% vs 56%). However, correct tip positioning resulted in a fourfold increase of obtaining
gastric aspirate. Conclusions: Both methods resulted in a high risk of incorrectly placed tubes due to malposition of the tip near the
LES or distal esophagus. This may increase the risk of reflux and pulmonary aspiration. Based on these results, the development
of more reliable methods requires consideration. (Nutr Clin Pract. 2018;33:843–850)

Keywords
enteral nutrition; gastrointestinal intubation; nasogastric tube; nutritional support; patient safety; X-rays

Introduction data about aspiration are difficult to determine due to vague


definitions, poor assessment methods, and varying levels of
Enteral feeding via nasogastric tube is a frequently used clinical recognition. Pulmonary aspiration of feeding can
short-term method for patients who cannot eat or whose be caused by either pulmonary misplacement of the tube
oral nutrition intake is insufficient. In adults, the stomach or incorrect positioning of the tube in the gastrointesti-
is a J-shaped organ with a greater curvature of 34 cm, a nal tract. In 2005, incorrectly placed nasogastric tubes in
maximum transverse diameter of 10 cm, a pyloric sphincter general were first recognized in England by the National
diameter of 1.2 cm, and a capacity of 0.9 litres.1 A correct Patient Safety Agency (NPSA) as a patient safety issue.
position of the tip of the nasogastric tube inside the stomach Between September 2005 and March 2010, there were 21
is required to ensure the safe administration of nutrients.2,3 deaths and 79 reported cases in England of harm related
Correct positioning will also prevent reflux and aspiration
of nutrients into the lungs. An incorrect gastric tip position
due to underestimation of the tube may lead to feeding From the 1 Department of Nursing, Odisee University College,
Brussels, Belgium; 2 AZ Nikolaas General Hospital, Sint-Niklaas,
remaining in the esophagus because most tubes have several Belgium; and 3 University Centre for Nursing and Midwifery, Ghent
lateral openings proximal of the distal tip opening. Con- University, Ghent, Belgium.
versely, overestimation (leading to a tube segment located Financial disclosure: None declared.
too deep into the stomach) can cause curling of the tube
Conflicts of interest: None declared.
inside the stomach or even migrating the tube back into the
esophagus or beyond the pylorus into the duodenum. Incor- This article originally appeared online on June 30, 2018.
rect positioning may also prevent obtaining an aspirate for Corresponding Author:
pH measurement or gastric residual volume.4,5 Torsy Tim, RN, MSc, Odisee University College, Department of
Health (Nursing and Midwifery), Hospitaalstraat 23, 9100
Although pulmonary aspiration is 1 of the most common Sint-Niklaas, Belgium.
complications in enterally fed patients, reliable prevalence Email: tim.torsy@odisee.be
844 Nutrition in Clinical Practice 33(6)

to feeding through misplaced nasogastric tubes. In 2009, inside the stomach, the presence of the lateral openings
inadequate feeding related to a misplaced nasogastric tube was taken into account (as described in the interventions
became a never event, which is defined as a serious, largely and outcomes section of this article). The target popu-
preventable patient safety incident that should not occur if lation of this study were adults because a method for
the available preventative measures have been implemented newborns and children (using the distance between the tip
by healthcare providers. Despite all guidance, during 2009– of the nose to the earlobe to the mid-umbilicus) already
2010 there were 41 never events reported to the NPSA in exists.9,10
which a misplaced nasogastric tube was not detected prior The primary aim of this first prospective randomized
to use. Even at this time, misplaced nasogastric tubes are trial was to determine whether the use of the Hanson
never events: Between September 2011 and March 2016, a formula (NEX × 0.38696) + 30.37 was more successful
total of 95 incidents were reported. These incidents show than using the NEX method. The secondary aim was to
that, despite the sensitization of all stakeholders, risks to investigate the likelihood to obtain a gastric aspirate to
patient safety persist.6 perform pH measurement.
Most often, the nose–earlobe–xiphoid (NEX) method is
used to determine the insertion length of a nasogastric tube. Methods
This measures the length from the tip of the nose to the
earlobe to the xiphoid.7,8 This simple and popular method Study Design
was introduced by Royce, Tepper, Watson, and Day in 1951. This study was designed as a blinded prospective random-
They described a 6-month experience of estimating the ized trial. The data were collected between December 2015
appropriate tube length using external anatomical reference and September 2016 in a Belgian general hospital on both
points in neonates. Subsequently, the NEX method was intensive care units (ICU) and non ICUs.
recommended as a gold standard for neonates, children,
and adults in nursing textbooks without further research Participants
or reflection. The reliability of the NEX method in both
adults and pediatric populations, however, has been ques- Patients were eligible to participate if they met the following
tioned several times.8-10 Using the NEX, Ziemer and Carroll criteria: adult (aged ࣙ 18 years) and a medical indication
noticed that a deeper insertion was often needed to obtain for nasogastric tube feeding. Patients were excluded when
a gastric aspirate.7 the xiphoid was not palpable or if they had documented
After a study on healthy volunteers and a postmortem surgical or anatomical abnormalities of the esophagus or
study including 99 cases, Hanson described alternative stomach (eg, hiatus hernia). Written informed consent was
formulas to calculate the nasogastric tube length in a more obtained from all patients or their legal representatives in
reliable way.2 Two formulas were proposed as a correction case of diminished awareness or coma.
of the NEX (method 1 and 2), and 1 formula was based on Participants were randomized to either the control or the
body length (method 3): intervention group using block randomization.11 Random-
ization was performed by an investigator with no clinical
Method 1: ([NEX – 50 cm]/2) + 50 cm involvement in the trial. A randomization list with 2 groups
Method 2: (NEX × 0.38696) + 30.37 (control and intervention group), block sizes of 6 patients,
Method 3: (body length × 0.20239) + 17.07 and an actual list length of 240 participants (control group
Method 2 resulted in more nasogastric tubes being and intervention group) was created.
placed correctly inside the stomach (92.3% vs 91.35%), with After assignment to 1 of the study groups, blinding was
a tip position ranging from 1 to 10 cm under the lower performed at the following levels:
esophageal sphincter (LES) being optimal.2 For that reason,
method 2 was selected as the experimental intervention
Included patients were kept blinded to their allocation.
in this current randomized controlled trial. Ellett et al
Data collectors (clinical nutrition nurse specialists) could
already had reported that the minimum insertion length of
not be blinded. They were aware of the allocated arm
the nasogastric tube corresponds to the distance from the
to perform the appropriate intervention.
tip of the nose to the gastroesophageal junction, thereby
Outcome adjudicators (radiologists) were also kept
adding the distance from the tip of the tube to the most
blinded to minimize bias and maximize the validity
proximal lateral opening on the tube to ensure that each
of the results.
lateral opening was located inside the stomach.3 The ac-
tual required length of the nasogastric tube according to
either control or intervention group was determined just
Interventions and Outcomes
before the placement procedure starting from the distal tip Two experienced clinical nutrition nurse specialists indepen-
opening of the tube. To consider a tube as correctly placed dently measured the NEX distance (Figure 1) in all patients.
Torsy et al 845

Figure 1. NEX distance.


NEX, nose-earlobe-xiphoid distance.

The 2 measurements were taken within a short space of


time according to the following procedure: the patient was Figure 2. Flowchart for evaluating the X-ray images.
in supine position with the chest at an angle of 30° and Cm, centimeter; VOD, vault of the diaphragm.
facing forward. The chest was uncovered, and the xiphoid
was marked. A measuring tape with an accuracy up to half
a centimeter was directed in a straight line from the tip of removal of the guidewire. Gastric aspirate was tested for pH
the nose to the earlobe and then to the marked xiphoid using pH color-indicator strips (pH 2.0–9.0) with a 0.5 pH
(Figure 1). A nasogastric tube was subsequently inserted. interval scale (Merck KGaA, Darmstadt, Germany). If an
The nasogastric tube used in the study was a aspirate could be obtained, a pH value ࣘ 5.5 indicated that
polyurethane radiopaque feeding tube (Nutricia Flocare it was indeed gastric aspirate.12
ENFit; CH10 or CH14, total length 110 cm), manufactured A chest X-ray of the patient, positioned in supine posi-
by Danone Trading B.V., Schiphol, The Netherlands and tion as described above, was taken in situ to determine the
consisting following characterisitcs: An internal guidewire, tip position of the tube. Preferably, the guidewire was left
a distal opening in the tip, and lateral openings up to 2 cm inside or put back inside the tube to increase the visibility
from the distal tip. The guidewire ended 5 cm before the of the nasogastric tube on X-ray. The chest X-ray images
distal tip opening. Newly placed tubes were inserted by covered the area from the apex to the base of the lungs,
the 2 clinical nutrition nurse specialists according to the including the diaphragm and the stomach below. All of the
randomization criteria. If the tubes were already in situ but images were retrieved from the hospital picture archiving
not yet used for tube feeding, they were only repositioned and communication system, processed and anonymized
but not replaced, again according to the randomization using DicomCleaner (PixelMed Publishing, Bangor, PA),
criteria. A safety margin of 1 cm was added so a correct and then evaluated on medical imaging monitors.
tip position of the nasogastric tube inside the stomach was
defined as somewhere between 3 and 10 cm under the LES.
After insertion or repositioning, the tube was fixed to the
Instruments
nose with a fixation tape for nasal tubes manufactured by The chest X-ray images were independently evaluated
ConvaTec, Deeside, UK (ConvaTec Naso-Fix). by 3 different radiologists using a predefined evaluation
A gastric aspirate was obtained immediately after inser- flowchart (Figure 2). The authors and the 3 participating
tion or repositioning of the nasogastric tube because pH radiologists used the vault of the diaphragm as the reference
measurement is considered an alternative bedside method point for the location of the LES in the assessment of
to X-ray to check the position of a nasogastric tube.11 the images because the LES itself is not very clear on
A large syringe (60 mL) with an ENFit (Danone Trading an X-ray.
B.V., Schiphol, The Netherlands) connection was used for “Obtainment of gastric aspirate” was considered as neg-
aspiration. An initial attempt of obtaining gastric aspirate ative after 2 attempts (as described above) without retrieving
occurred before the removal of the guidewire. If no aspirate gastric content within 1 hour after insertion. A few drops of
could be obtained, an extra attempt was made 1 hour after the aspirate were placed on a pH indicator strip.
846 Nutrition in Clinical Practice 33(6)

Statistical Methods Characteristics of the control and intervention group were


comparable (P > 0.05), as shown in Table 1.
Statistical analyses were conducted using SPSS 24.0 (IBM
Corporation, Armonk, NY). Categorical variables were Tip Position and Gastric Aspirate
presented as frequencies (percentages); normally distributed
continuous variables were described using means and stan- An overall significant difference (P = 0.032) was demon-
dard deviations (SD); and between-group comparisons were strated between the 2 groups regarding the positioning of
performed using independent samples t test. Comparison the tip. The tip of the nasogastric tube was situated <3 cm
of categorical data between groups was performed using under the LES in 20.2% of the tubes in the control group
χ 2 test. and in 22.6% in the intervention group (P = 0.691). A
The interobserver agreement between the 3 independent significant difference (P = 0.009) concerning overestimation
radiologists was assessed using κ statistics. Observer agree- of the tip position was found between the control group
ment was defined as 2 or more concurring interpretations (17.2%) and the intervention group (4.8%). There was
of the X-ray being tip <3 cm under the LES, between 3 no statistically significant difference (P > 0.05) between
and 10 cm under the LES, > 10 cm under the LES, or both methods for external length determination and the
tip not visible on X-ray. To obtain an overall κ value of chance of successfully obtaining gastric aspirate through
the assessments, Fleiss κ was calculated. The classification the nasogastric tube. Absolute values, percentages, and P
system used by Coblentz et al was used to define the strength values are shown in Table 2. The interobserver reliability of
of agreement for the κ coefficient.13 the X-ray evaluation between the 3 radiologists indicated
The association between the study group and the out- acceptable agreement (κ = 0.44, 95% confidence interval
come was controlled for age, gender, body length, and cogni- [CI], 0.39–0.49).
tive awareness using a multivariate logistic binary regression Figure 3 shows the distribution of the nasogastric tube
analysis. The variable “gastric aspirate obtained” was added insertion lengths using either the NEX method or Hanson’s
to the multivariate model to analyze how this variable was formula-corrected NEX distance. Using the NEX method,
related to the correct positioning of the nasogastric tube. the mean insertion length was 51.62 cm (SD 3.79) and the
Hosmer-Lemeshow statistic was calculated as a measure of interquartile range (IQR) was 4 with a minimum insertion
model fit. A significance level of P < 0.05 was applied. length of 40.5 cm and a maximum insertion length of
59.5 cm. Using (NEX × 0.38696) + 30.37, the mean
insertion length was 50.50 cm (SD 1.62) and an IQR of 1.95
Ethical Statement and Trial Registering
with a minimum insertion length of 43.5 cm and a maximum
All patients (or their legal representative in case of dimin- insertion length of 53.0 cm. The distribution of the insertion
ished awareness or coma) gave informed consent. Collecting lengths was significantly lower (P = 0.009) using Hanson’s
the informed consents was done by the clinical nutrition formula-corrected NEX distance.
nurse specialists. Patients or their representatives who were
unable or unwilling to sign the written informed consent Influencing Factors
were excluded from the study.
Five potential influencing factors in aiming for correct gas-
The study protocol was approved by the Review Com-
tric tip positioning were analyzed in a multivariate binary
mittee for Medical Ethics from AZ Nikolaas (East Flan-
logistic regression analysis: age, gender (male/female), body
ders, Belgium) (EC15053). The study was registered at
length, method of length determination (“NEX”/“(NEX
ISRCTN.com (ISRCTN18322407).
× 0.38696) + 30.37”) and cognitive awareness level
(aware/diminished awareness/comatose) (Table 3). The
Results binary outcome variable was “tip position of the nasogastric
tube inside the stomach” (<3 cm under LES = incorrect
Patient Characteristics position; ࣙ 3 cm under LES = correct position). The
A total of 215 patients participated in the study. The quality variables “weight” and “body mass index (BMI)” were not
of the X-ray images of 32 patients was insufficient to included in the model due to the large number of missing
allow conclusive determination of the position of the tip of values (81 of 183, 44.3%) and their mutual multicollinearity.
the nasogastric tube. Although the nasogastric tubes were The variable “gastric aspirate after placement (yes/no)”
visible on all X-ray images, the images had poor margin was also included in the model to explore the differences
settings and thus the tip was not visible on the image between the 2 groups concerning this variable. The model
and could not be evaluated. Therefore, after the procedure resulted in a significant association (P = 0.001) between
of taking X-ray images, data of only 183 persons were the outcome variable “tip position” and the obtainment
used for further analysis: 99 patients were included in the of gastric aspirate (odds ratio 4.109, 95% CI 1.785–9.458).
control group and 84 patients in the intervention group. There is no discrepancy between the model predicted and
Torsy et al 847

Table 1. Characteristics of Participants (N = 183).a

Control Group Intervention Group No Data Available


Characteristics (n = 99) (n = 84) P Value no. (%)

Demographic variables
Age, mean ± SD, years 72.39 ± 12.33 70,62 ± 12.65 0.339
Gender
Male, no. (%)a 52 (52.5) 48 (57.1) 0.532
Female, no. (%)a 47 (47.5) 36 (42.9) 0.532
Clinical variables
Body length, mean ± SD, cm (no.) 165.37 ± 9.09 (97) 164.83 ± 8.22 (84) 0.675 2 (1.1)
Weight, mean ± SD, kg (no.) 70.59 ± 17.99 (61) 66.36 ± 14.52 (41) 0.193 81 (44.2)
BMI, mean ± SD, kg/m² (no.) 25.63 ± 5.74 (61) 24.35 ± 5.01 (41) 0.249 81 (44.2)
Use of antacids, no. (%)b 78 (78.8) 61 (72.6) 0.407 3 (1.6)
Unit
ICU, no. (%)b 50 (50.5) 44 (52.4) 0.800
Non-ICU, no. (%)b 49 (49.5) 40 (47.6) 0.800
Cognitive awareness level
Aware, no. (%)b 43 (43.4) 29 (34.5) 0.219
Diminished awareness, no. (%)b 8 (8.1) 14 (16.7) 0.075
Comatose, no. (%)b 48 (48.5) 41 (48.8) 0.965

BMI, body mass index; ICU, intensive care unit; no., number; SD, standard deviation.
a Data collected by clinical nutrition nurse specialists
b Percentages within group

Table 2. Localization of Nasogastric Tubes in Both Groups and Obtainment of Gastric Aspirate (N = 183).

Control Group (n = 99) Intervention Group (n = 84) P Value

Tip positioning
< 3 cm under LES, no. (%) 20 (20.2) 19 (22.6) 0.691
3–10 cm under LES, no. (%) 62 (62.6) 61 (72.6) 0.151
> 10 cm under LES, no. (%) 17 (17.2) 4 (4.8) 0.009
Obtaining gastric aspirate,a no. (%) 55 (55.6) 47 (56.0) 0.938

LES, lower esophageal sphincter; no., number.


a After placement of the tube.

the effectively observed probabilities (Hosmer-Lemeshow


2.228, degrees of freedom 8, P = 0.973).

Discussion
This study is the first prospective randomized trial in which
the reliability of the NEX method and Hanson’s formula
has been tested. Both methods are inaccurate when aiming
for a correct tip position of 3 or more cm under the LES.
When using the NEX, the range of tip positions is larger
compared with Hanson’s formula (Figure 3). The chance of
obtaining an aspirate was low (around 56%) in both groups
(Table 2).
One of the key findings from this study was that in both
trial groups in approximately 1 of 5 patients (20.2% in the
control and 22.6% in the intervention group), the tip of the
tube was located <3 cm under the LES. This means that at Figure 3. Boxplot describing the distribution of insertion
least 1 lateral opening of the nasogastric tube was located lengths (in centimeters).
in the esophagus or at the same height as the LES. This can NEX, nose-earlobe-xiphoid distance.
848 Nutrition in Clinical Practice 33(6)

Table 3. Multivariate Binary Logistic Regression Between Possible Influencing Factors in Aiming for a Correct Tip Position.

β Coefficient Standard Error Wald Statistic df OR (95% CI) P Value

Age −0.007 0.020 0.135 1 0.993 (0.956 – 1.032) 0.714


Gender 0.614 0.555 1.222 1 1.847 (0.622 – 5.481) 0.269
Malea
Female
Body length 0.031 0.035 0.779 1 1.031 (0.963 – 1.103) 0.377
Gastric aspirate 1.413 0.425 11.037 1 4.109 (1.785 – 9.458) 0.001
Yes
Noa
Method of length determination 0.332 0.402 0.684 1 1.394 (0.634 – 3.063) 0.408
NEX
(NEX × 0.38696) + 30.37a
Awareness levela 0.634 2 0.728
Awareness level(1) 0.191 0.442 0.187 1 1.211 (0.509 – 2.883) 0.665
Awareness level(2) 0.542 0.695 0.609 1 1.720 (0.441 – 6.711) 0.435
Constant −4.400 6.438 0.467 1 0.012 0.494

CI, confidence interval; df, degrees of freedom; NEX, nose-earlobe-xiphoid distance; OR, odds ratio.
a Reference category.

lead to regurgitation and an increased risk of pulmonary 24 potentially predicting variables were included in the
aspiration.14 data collection and analysis in order to develop a new
The NEX method is the most commonly used technique method for external length determination. This resulted in a
for measuring the nasogastric tube length but has been 3-variable model of calculation based on gender, weight,
questioned in the literature.8-10 After a postmortem study and the distance between nose and umbilicus in supine
including 99 cases, Hanson concluded that the NEX method position with the head flat (GWNUF), in which gender was
resulted in 72% well-placed nasogastric tubes, with the ideal “1” if the participant was male and “0” if the participant
tip position between 1 and 10 cm under the LES.2 Using was female: 29.38 + (4.53 × gender) + (0.34 × NUF) –
the 2 Hanson formulas adjusting the NEX, the number of (0.06 × weight). The GWNUF resulted in 85.3% of correct
properly placed tubes increased to around 92%; however, positions (vs 72% for the NEX and 84.2% for Hanson)
approximately 3% still were located in the distal esophagus, but here also 5 of 11 malpositioned tubes were in the
which was about the same for the NEX (2%). Note that esophageal danger zone. In addition to increasing the risk
Hanson’s study did not mention any lateral openings in the of feeding in the esophagus due to a malpositioned tip
nasogastric tubes, now common in most tubes. Therefore, of the nasogastric tube, the feasibility of the GWNUF in
the tube must be inserted sufficiently deep into the stomach clinical practice can also be questioned for other reasons: the
to ensure that not only the tip but also all lateral openings difficulty of obtaining the correct weight of bedridden pa-
are located inside the stomach to prevent tube feeding tients; discomfort for the patients to maintain the required
entering the esophagus. In our study, the presence of lateral position; and higher likelihood to make clinical errors when
openings may explain why the percentage of insufficiently using different parameters to estimate the insertion length
deep placed nasogastric tubes exceeded Hanson’s. Hanson in clinical practice. The combination of these elements,
also stated that the NEX might lead to an overestimation together with the large number of included ICU patients
of the required length, resulting in a placement >10 cm in the study, ensured that this method was not selected as
under the LES in 25% of cases compared with <6% using the preferred intervention in the study. It may not possible
Hanson’s formulas. Our study reached the same conclusion, to obtain an accurate weight in critically ill patients who
with 17.2% of the tubes located >10 cm under the LES in usually are also ventilated. In addition, the positioning of
the control group and 4.8% in the intervention group. ICU patients as described above is not always possible due
In a study with a cross-sectional design, Ellett et al to their physical condition.
tested the reliability of 1 of the formulas proposed by The study of Boeykens et al. (2014) mainly fo-
Hanson (([NEX – 50 cm]/2) + 50 cm) compared with cused on pH measurement as an alternative method for
the NEX.3 A correct tip position was defined as between X-ray to confirm the correct position of the nasogastric
3 and 10 cm under the LES. In the NEX group, 9 out tube.12 In Boeykens’ study, the probability of a correctly
of 21 malpositioned tubes were placed insufficiently deep placed nasogastric tube was 98.9% when taking pH mea-
vs 11 of 19 tubes in the Hanson group. Additionally, surement as criterion. Metheny et al stated that obtaining
Torsy et al 849

gastric aspirate and pH measurement is not useful to detect this study again emphasizes the importance of strict and
placement of the nasogastric tube in the esophagus because clear guidance on preventing potential incidents that may
of possible gastric reflux.15,16 Therefore, caution should lead to harm or death. Therefore, further research should
be exercised in interpreting a pH measurement on gastric focus on determining other possible influencing factors
aspirate. Our study showed that a correctly positioned tip (e.g.k anatomical characteristics) for a more correct gastric
inside the stomach increased the possibility to obtain gastric tip positioning in order to develop an easy, safe, and
aspirate by almost 4 times. In our study, >20% of all clinically feasible method for estimating the insertion length
nasogastric tubes were placed too shallow whether the NEX of nasogastric tubes.
or Hanson’s formula was used to determine the insertion
length. Therefore, it is recommended to combine either
method for external length determination with X-ray to Conclusions
verify correct placement of the tip.
Both the NEX-method and Hanson’s formula are not reli-
Taylor et al suggested the use of NEX + 10 cm as
able to determine the appropriate length of the nasogastric
an alternative to the NEX method, which would lead to
tube in order to obtain a correct tip position between 3
a reduction from 16% to 7% of misplaced nasogastric
and 10 cm under the LES. At the same time, differences
tubes, with the tip located into the esophagus. This small
in the likelihood of obtaining gastric aspirate could not
study (N = 36) using electromagnetic guidance and not
be demonstrated between the 2 methods. In >20% of
X-ray was solely conducted in an intensive care setting in
patients, the tip of the nasogastric tube was located in
a population of mainly neurosurgery and trauma patients
the esophageal danger zone. To reduce the risk of mis-
with gastric emptying problems whose stomachs possibly
placement, and consequently also reduce the inconvenience
dilate more than in other patients.5 This method made
caused to patients due to repositioning of the tube, a more
it possible to guide the tip of the tube in the stomach
adequate method of determining the appropriate length of
immediately. In earlier studies2,3 as well as in this current
the nasogastric tube should be developed. Meanwhile, it is
study, an overestimation of the NEX was observed; thus,
strongly recommended to confirm placement of the tube tip
the NEX + 10 cm potentially could result in tubes migrating
through X-ray and reposition the tube when necessary. This
postpyloric, curling up/kinking inside the stomach—or even
will minimize the risk of feeding remaining in the esophagus
worse, migrating up and re-entering the esophageal danger
and of possible reflux.
zone.
There are some limitations in this prospective random-
ized trial. Firstly, there was no registration of the number of
Statement of Authorship
excluded patients with previously documented hiatus hernia
during the period of data collection. After including all T. Torsy, R. Saman, K. Boeykens and I. Duysburgh equally
patients in the study, there was also a 14.9% patient dropout contributed to the conception and design of the research; D.
Beeckman and N. Van Damme contributed to the design of the
due to the poor quality of the chest X-ray images (tube or
research; T. Torsy, R. Saman, K. Boeykens and N. Van Damme
tip not visible). Secondly, there was no separate registration
contributed to the acquisition and analysis of the data; T. Torsy,
of data obtained by the 2 clinical nutrition nurse specialists D. Beeckman and N. Van Damme contributed to the inter-
measuring the NEX distances, and thus no conclusions pretation of the data; and T. Torsy and R. Saman drafted the
can be drawn about interrater variability. Finally, the κ manuscript. All authors critically revised the manuscript, agree
value (κ = 0.44) of the interobserver agreement between the to be fully accountable for ensuring the integrity and accuracy
radiologists can also be questioned. The clinical significance of the work, and read and approved the final manuscript.
of this value depends on its context. In a review17 of medical
imaging literature regarding imaging for expressing observer Aknowledgements
agreement with regard to categorical data, the conclusion The authors would like to thank Leo Verguts, Karline
was made that a κ value ࣙ 0.4 could be considered an Schutyser, Dirk De Backer, Kurt Van Belle and Patrick Palmans
acceptable level of observer variability. For this reason, for their valuable contribution to this study and Leen Trommel-
the classification system used by Coblentz at al., in which mans for assisting with the final draft. They also would like to
agreement is quantified as poor, fair, moderate, good, or thank Odisee University College and the general hospital AZ
excellent, was followed.13 Nikolaas for facilitating this study.
Despite the fact that Hanson’s formula in our study led
References
to 72.6% correctly placed nasogastric tubes, with the tip
1. Ferrua MJ, Singh RP. Modeling the fluid dynamics in a human
located somewhere between 3 and 10 cm under the LES,
stomach to gain insight of food digestion. J Food Sci 2010;75:
there were still outliers (both over- and underestimation 151–162.
of the insertion length) that could not be associated with 2. Hanson RL. Predictive criteria for length of nasogastric tube insertion
any of the variables of the data collection. Despite all, for tube feeding. JPEN J Parenter Enteral Nutr 1979;3:160–163.
850 Nutrition in Clinical Practice 33(6)

3. Ellett MLC, Beckstrand J, Flueckiger J, Perkins SM, Johnson CS. 10. Ellett MLC, Cohen MD, Perkins SM, Smith CE, Lane KA, Austin JK.
Predicting the insertion distance for placing gastric tubes. Clin Nurs Predicting the insertion length for gastric tube placement in neonates.
Res 2005;14:11–27. J Obstet Gynecol Neonatal Nurs 2011;40:412–421.
4. Chen Y, Wang L, Chang Y et al. Potential risk of malposition 11. Sealed Envelope Ltd. Create a blocked randomi-
of nasogastric tube using nose-ear-xiphoid measurement. PLoS One sation list. Updated January 1, 2015. Available at:
2014;9:e88046. https://www.sealedenvelope.com/simple-randomiser/v1/lists. Accessed
5. Taylor SJ, Allan K, McWilliam H, Toher D. The ‘NEX’ guideline is October 30, 2015.
incorrect. Br J Nurs 2014;23:641–644. 12. Boeykens K, Steeman E, Duysburgh I. Reliability of pH measurement
6. National Patient Safety Agency (NPSA). Patient Safety Alert: and the auscultatory method to confirm the position of a nasogastric
Nasogastric tube misplacement: continuing risk of death and severe tube. Int J Nurs Stud 2014;51:1427–1433.
harm. Updated July 22, 2016. Available at: https://improvement.nhs.uk/ 13. Coblentz CL, Babcook CJ, Alton D, Riley BJ, Norman G. Observer
news-alerts/nasogastric-tube-misplacement-continuing-risk-of-death-severe-harm.
variation in detecting the radiologic features associated with bronchi-
Accessed March 2, 2018. olitis. Invest Radiol 1991;26:115–118.
7. Fitzpatrick JJ, Wallace M. Encyclopaedia of Nursing Research 3rd ed. 14. Metheny NA, Titler MG. Assessing placement of feeding tubes. Am
New York, NY: Springer Publishing; 2011. J Nurs 2001;101:36–45.
8. Santos SC, Woith W, Freitas MI, Zeferino EB. Methods to determine 15. Metheny NA, Reed L, Wiersema L, McSeeney M, Wehrle MA,
the internal length of nasogastric feeding tubes: an integrative review. Clark J. Effectiveness of pH measurements in predicting feeding tube
Int J Nurs Stud 2016;61:95–103. placement: an update. Nurs Res 1993;42:324–331.
9. Beckstrand J, Ellett MLC, McDaniel A. Predicting the internal dis- 16. Metheny NA, Clouse RA, Clarck JM, Reed L, Wehrle MA, Wiersema
tance to the stomach for positioning nasogastric and orogastric feeding L. pH testing of feeding-tube aspirates to determine placement. Nutr
tubes in children. J Adv Nurs 2007;59:274–289. Clin Pract 1994;9:185–190.
17. Kundel HL, Polansky M. Measurement of observer agreement. Radi-
ology 2003;228:303–308.
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 6
Long-Term Use of Mixed-Oil Lipid Emulsion in Adult Home December 2018 851–857

C 2018 American Society for

Parenteral Nutrition Patients: A Case Series Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10014
wileyonlinelibrary.com

Manpreet S. Mundi, MD1 ; Megan T. McMahon, PA-C2 ;


Jennifer J. Carnell, PharmD2 ; and Ryan T. Hurt, MD, PhD1,2,3,4

Abstract
Background: Despite providing significant benefits, home parenteral nutrition (HPN) can be associated with complications such
as infections, intestinal failure–associated liver disease, and metabolic abnormalities. Soybean oil (SO)–based intravenous lipid
emulsion (ILE) has been noted to contribute to some of these complications, leading to the development of alternative sources
of ILE. Mixed-oil (MO) ILE has recently been approved for use in adults with short-term studies revealing a benefit over SO
ILE. Currently there is a paucity of data regarding long-term use in the HPN population. Methods: The current study reports our
experience with MO ILE in 9 HPN patients. Results: A total of 9 patients (3 women and 6 men) with average age of 48.1 ± 15.1 years
and a median HPN use of 9.9 years (9.0 months–30.7 years) were transitioned from SO ILE to MO ILE as a result of intolerance.
The 9 patients tolerated MO ILE well for 140.7 ± 29.7 days. The percentage of calories provided through ILE increased from 7.6 ±
6.5% to 18.4 ± 8.2% (P = .003), whereas the dextrose decreased from 66.9 ± 8.4% to 56.9 ± 5.5% (P = .0007). Although statistical
significance was not reached, there was a trend toward improvement in alkaline phosphatase from 138.0 (52–884) to 106 (47–512;
P = .09). Conclusion: MO ILE was well tolerated in this small cohort and led to improvement in the macronutrient composition
of HPN while providing a trend toward improvement in liver studies. These results are promising; however, additional randomized
control trials are needed to delineate the true benefit. (Nutr Clin Pract. 2018;33:851–857)

Keywords
fatty acids; home parenteral nutrition; intravenous fat emulsions; intravenous lipid emulsions; parenteral nutrition; parenteral
nutrition solutions

Introduction liest signs of IFALD can be lab abnormalities, including


elevations in bilirubin and alkaline phosphatase, followed by
Studies have estimated between 25,000–35,000 patients use elevations in aspartate aminotransferase (AST) and alanine
parenteral nutrition (PN) at home as a result of intestinal aminotransferase (ALT).9,12 Although the exact etiology
failure.1,2 With appropriate training, home PN (HPN) can is not known, factors such as infections, inflammation,
be provided safely by patients or family members, allowing
them to receive adequate nutrition and electrolytes outside
From the 1 Division of Endocrinology, Diabetes, Metabolism and
the hospital setting. However, with long-term use, compli-
Nutrition, Mayo Clinic, Rochester, Minnesota, USA; 2 Division of
cations such as infection, lipid abnormalities, and intesti- General Internal Medicine, Mayo Clinic, Rochester, Minnesota,
nal failure–associated liver disease (IFALD) can arise.3,4 USA; 3 Division of Gastroenterology and Hepatology, Mayo Clinic,
Lipid profile changes such as a decrease in high-density Rochester, Minnesota, USA; and the 4 Division of Gastroenterology,
lipoprotein and an increase in low-density lipoprotein and Hepatology and Nutrition, University of Louisville, Louisville,
Kentucky, USA.
triglyceride levels can also be seen after the initiation of
HPN.5 Liposomes, which are created from excess phospho- Financial disclosure: None declared.
lipid emulsifier, as well as triglyceride content may play a Conflicts of interest: Ryan T. Hurt is a consultant for Nestlé.
significant role given that 10% intravenous lipid emulsions Manpreet S. Mundi, Megan T. McMahon, and Jennifer Carnell have
no financial disclosure or conflict of interest to report.
(ILEs) tend to have greater lipid profile changes when
compared with 20% ILE.3,6 This article originally appeared online on February 13, 2018.
IFALD may present as hepatic steatosis, cholestasis, Corresponding Author:
cholelithiasis, and hepatic fibrosis and can be seen in as high Manpreet S. Mundi, MD, Division of Endocrinology, Diabetes,
Metabolism, and Nutrition, Mayo Clinic, 200 First St. SW, Rochester,
as 30%–60% of children and 15%–40% of adults requiring
MN 55905.
long-term PN.7-11 Especially in children, some of the ear- Email: mundi.manpreet@mayo.edu
852 Nutrition in Clinical Practice 33(6)

decreased bile flow, and excess calories especially from high tests are obtained on a routine basis (weekly initially, fol-
dextrose and lipids have been felt to be contributory.5,9,13 lowed by biweekly, then monthly, and depending on stabil-
In addition to excess calories, the high n-6 fatty acid and ity, potentially quarterly). If indications of ILE intolerance
phytosterol content of soybean oil (SO)–based ILE are such as an elevation of liver enzymes or bilirubin level
also thought be associated with IFLAD.3 Based on this, 1.5 times baseline is noted, the frequency of ILE infusion
strategies for treatment have included use of ursodiol, is reduced. Typically, reduction proceeds from 3 times per
cycling of HPN, and a reduction or discontinuation of SO week ILE to once per week with the reassessment of labs.
ILE. In fact, many programs including ours provide SO ILE If there is no improvement or stabilization in liver enzymes
3 times per week as a preventive measure.3,13 or bilirubin levels, the ILE is reduced further to once every
In addition, attempts have been made with subsequent other week, and then once monthly. The reduction in ILE
generations of ILE to reduce the SO lipid content. Novel calories is compensated for with an increase in calories from
ILEs have also focused on increasing the n-3 to n-6 fatty dextrose or potentially amino acids if not optimized.
acid ratio through the use of alternative fatty acid sources With FDA approval and commercial availability of MO
such as medium-chain triglycerides, olive oil, and most ILE in the United States, our program reviewed our adult
recently fish oil (FO). In terms of FO containing ILE, a HPN cohort to identify patients who were intolerant to
formulation composed entirely of FO (Omegaven, Frese- SO ILE and require a reduction in the frequency of ILE
nius Kabi, Bad Homburg, Germany) is available in Europe administration. These patients were contacted and placed
but is not approved for use or commercially available in the on MO ILE if they approved and had no contraindications
United States. This ILE is available under a compassionate for use such as hypersensitivity to egg, soybean proteins,
use allowance through the Food and Drug Administration fish, or peanut proteins. Patients were started on the same
(FDA) for the treatment of IFLAD. The FDA has recently frequency and grams of MO ILE as their last SO ILE dose.
approved a mixed-oil (MO) ILE for adults (Smoflipid, As an example, if patient was receiving 50 g of SO ILE once
Fresenius Kabi, Bad Homburg, Germany), which is com- a week, they would be started on 50 g once a week of MO
posed of a combination of fatty acid sources (30% SO, ILE. Labs including liver enzymes and bilirubin levels were
30% medium-chain triglycerides, 25% olive oil, and 15% assessed frequently (every 2–4 weeks) to ensure tolerance. If
FO). Short-term trials (14–28 days) in adults and pediatric patients were tolerating MO ILE indicated by stability or
patients from various cohorts have shown benefit over SO improvement in labs as well as a lack of adverse reactions,
ILE in terms of inflammatory markers, fatty acid profile, then the dose was increased with a corresponding reduction
and liver function studies as well as bilirubin levels.14-18 in dextrose dose every 2 weeks. Dose escalation depended
Despite these promising results, the impact of FO ILE on specific clinical scenario, however, typically if a patient
beyond 28 days is not fully known. To assess tolerability was on ILE once every other week, ILE frequency would be
and impact on liver function studies and total bilirubin, we increased to every week while the dose in grams was kept the
prospectively followed adult patients placed on MO ILE same. If they tolerated once a week, they would be increased
as a result of an intolerance of SO ILE. In addition, we to 2–3 times per week followed by further increase to 4–7
assessed the impact of transitioning to MO ILE on the times per week.
macronutrient composition of PN, with the hypothesis that The results from this case series are presented below.
transitioning to MO ILE containing PN would be better All data are presented as mean ± standard deviation for
tolerated based on previous studies and would allow for normally distributed data and median (range) for nonpara-
improved macronutrient composition. metric data. The statistical analysis was performed using
JMP, Version 10 (SAS Institute Inc., Cary, NC).
Methods
Our program initiates HPN in 90–100 adult patients annu-
Results
ally and closely follows 110–140 patients on HPN per year. A total of 9 adult patients (3 women and 6 men) with an
Patients are usually inpatients at the time of training and average age of 48.1 ± 15.1 years were transitioned from
initiation of HPN. On discharge, patients typically receive SO ILE to MO ILE as a result of intolerance (Table 1).
PN with total daily calories between 20–35 kcal/kg depend- They had been on HPN for a median of 9.9 years with
ing on current body mass index based on major guidelines.19 a range of 9.0 months to 30.7 years. The most common
Amino acids range between 0.8–1.0 g/kg for unstressed indications for HPN were short bowel syndrome (44%) and
patients to as high as 1.5–2.0 g/kg for patients with signifi- bowel obstruction or dysmotilty (33%) as well as radiation
cant fistulae, wounds, or renal dialysis dependence. Overall enteritis (11%) and enterocutaneous fistula (11%). A major-
composition of macronutrients typically leads to 50%–60% ity of the patients were transitioned to MO ILE because
of calories from dextrose, 15%–20% from amino acids, and of an intolerance of SO ILE that presented in the form of
20%–25% from ILE. Per our current protocol, laboratory elevated liver studies. One patient subsequently developed
Mundi et al. 853

Table 1. Baseline Demographics. a catheter-related blood stream infection 30 days prior to


and during MO ILE use. Although some patients such as
Variable Results, n = 9
patient 1 (Table 3) did have a significant decline in alkaline
Age, y 48.1 ± 15.1 phosphatase, the overall group revealed only a trend toward
Gender, women/men 3/6 improvement with median value changing from 138.0 (52–
Duration of HPN, y 9.9 (0.9–30.7) 884) to 106 (47–512; P = .09). Similarly, AST, ALT, and total
Duration of MO ILE at analysis, d 140.7 ± 29.7 (range of bilirubin values also showed trends toward improvement,
75–180 days) again with some individuals showing clinically significant
Indication for HPN, n (%) improvements (Table 3). As expected, vitamin E levels im-
- Short bowel syndrome 4 (44)
proved significantly with MO ILE administration increasing
- Bowel obstruction/motility 3 (33)
disorder 1 (11) from 6.6 ± 1.9 to 10.3 ± 4.3 (P = .02). In addition, there was
- Radiation enteritis 1 (11) no significant change in triglyceride (117.9 ± 45.3 to 107.3
- Entrocutaneous fistula ± 46.5, P = .8) or glucose levels (93.0 ± 14.6 to 98.6 ± 18.9,
P = .37).

Data are presented as mean ± standard deviation if normal


distribution or as median (range) for nonparametric data. HPN, home Discussion
parenteral nutrition; MO ILE, mixed-oil intravenous lipid emulsion.
The present case series outlines our HPN program’s experi-
ence with our first 10 patients transitioned from 100% SO
intolerance of the high dextrose formula with hyperglycemia ILE to MO ILE. These patients were transitioned to MO
requiring insulin in HPN. ILE because of an intolerance of SO ILE that presented
At the time of analysis, the 9 patients were on MO ILE predominantly as liver function test abnormalities. One
for an average of 140.7 ± 29.7 days. Their initial body mass patient with neuroendocrine tumor in the abdomen com-
index was 23.6 ± 4.6 kg/m2 , and they were receiving an plained of pain with SO ILE and thus was transitioned to
average 25.7 ± 8.0 kcal per kg of body weight per day in MO ILE to assess if there would be an improvement in
their HPN formula (Table 2). All 9 patients tolerated MO symptoms given the lack of alternative options for nutrition.
ILE well, and we were able to increase the percentage of Unfortunately, despite 2 weeks of MO ILE, she did not
calories provided through ILE from 7.6 ± 6.5% to 18.4 experience an improvement and thus was removed from
± 8.2% (P = .003) while decreasing the dextrose from further analysis. The remaining patients tolerated MO ILE
66.9 ± 8.4% to 56.9 ± 5.5% (P = .0007; Figure 1). The well without adverse reactions and with trends toward
increase in lipids corresponded to an increase from 14.7 improvement in liver studies. In addition, the percent of
± 13.5 g/day (0.2 ± 0.2 g/kg/day) to 34.0 ± 21.3 g/day calories delivered as fats were significantly increased and
(0.6 ± 0.4 g/kg/day; P = .01). Only 1 patient developed dextrose decreased when compared with baseline.

Table 2. Pre–MO ILE and Post–MO ILE Values From 9 Patients.

Variable Pre–MO ILE Latest Value on MO ILE P Value

BMI 23.6 ± 4.6 22.9 ± 4.1 .39


Total calories per day, kcal 1751.2 ± 491.4 1762.9 ± 444.2 .92
Total calories per kg per day, kcal 25.7 ± 8.0 27.0 ± 11.1 .43
% Calories from amino acid, % 25.5 ± 5.5 24.8 ± 7.6 .63
% Calories from dextrose, % 66.9 ± 8.4 56.9 ± 5.5 .0007
% Calories from fats, % 7.6 ± 6.5 18.4 ± 8.2 .003
ILE g/day, g/kg/day 14.7 ± 13.5 (0.2 ± 0.2) 34.0 ± 21.3 (0.6 ± 0.4) .01
Frequency of ILE, days per week 1.4 ± 1.2 3.4 ± 1.8 .01
Alkaline phosphatase, U/L 138.0 (52–884) 106 (47–512) .09
AST, U/L 64.3 ± 33.9 46.2 ± 19.9 .18
ALT, U/L 108.9 ± 105.0 63.8 ± 35.9 .31
Total bilirubin, mg/dL 1.4 ± 0.7 1.2 ± 0.9 .07
Vitamin E, mg/L, n = 8 6.6 ± 1.9 10.3 ± 4.3 .02
Triglyceride, mg/dL, n = 8 117.9 ± 45.3 107.3 ± 46.5 .8
Glucose, mg/dL 93.0 ± 14.6 98.6 ± 18.9 .37

Data are presented as mean ± standard deviation if normal distribution or as median (range) for nonparametric data. ALT, alanine
aminotransferase; AST, aspartate aminotransferase; BMI, body mass index; ILE intravenous lipid emulsion; MO ILE, mixed-oil intravenous lipid
emulsion. p-value that reached significance indicated by bolding.
854 Nutrition in Clinical Practice 33(6)

Figure 1. Change in macronutrient composition after initiation of mixed-oil intravenous lipid emulsion (MO ILE).

Table 3. Liver Studies for Each Patient Before and After MO ILE.

Alkaline Phosphatase, U/L AST, U/L ALT, U/L Total Bilirubin, mg/dL

Pre– Post– Pre– Post– Pre– Post– Pre– Post–


Patient MO ILE MO ILE MO ILE MO ILE MO ILE MO ILE MO ILE MO ILE

1 884 512 116 62 91 47 1.1 0.7


2 125 103 41 45 61 66 1.6 1.2
3 125 127 27 29 52 63 0.9 1.0
4 139 131 22 25 46 42 2.3 2.3
5 138 63 59 27 66 29 0.7 0.4
6 52 47 79 46 82 59 1.7 1.9
7 200 195 55 92 70 155 1.1 0.3
8 104 79 56 53 111 80 2.8 2.8
9 384 131 124 44 401 55 0.8 0.4

ALT, alanine aminotransferase; AST, aspartate aminotransferase; MO ILE, mixed-oil intravenous lipid emulsion.

IFALD, which can present as hepatic steatosis, cholesta- nutrient excess and toxicity along with infection are felt to
sis, cholelithiasis, and potentially hepatic fibrosis, is a be risk factors.3,9,20 Treatment options are limited, with case
common problem affecting patients on HPN.7-11 In fact, series and small trials showing some benefit with use of
Cavicchi et al10 followed 90 patients supported with HPN antibiotics and ursodeoxycholic acid.21-24 In addition, cyclic
in the setting of intestinal failure and noted that 65% HPN (12-hour infusion) along with decreased frequency of
had developed chronic cholestasis after a median of 6 ILE (3 days per week) have also shown some promise to help
months. A total of 41.5% developed some form of IFALD improve IFALD.25,26
after median of 17 months, and 18.9% developed fibrosis Unfortunately, despite these treatments and preventive
with 5.6% progressing to cirrhosis with long-term HPN options, patients can still progress to IFALD requiring
use. Although the precise etiology is not fully understood, further reduction in ILE frequency (<3 days per week). This
Mundi et al. 855

often leads to a significant mismatch in composition of liver studies. In some cases, this increase would work for a
HPN macronutrients, with the majority of calories being few months, but given sufficient time liver studies became
provided as dextrose to compensate for the reduction in abnormal once again, leading to a reduction in SO ILE use.
ILE. This was evident in our case series as >65% of calories With the transition to MO ILE, the calories from ILE were
on average were being provided from dextrose with only increased from 7.6% to 18.4%, with the frequency of ILE
7.6% being provided from ILE prior to initiating MO ILE. increasing to 3.4 days per week from the initial 1.4 days per
This strategy may not be sufficient in many cases as the week. This allowed the reduction in dextrose calories from
high dextrose formula can lead to complications of its own, 66.9% to 56.9%.
including hyperglycemia requiring insulin to be added to Our cohort also experienced an increase in vitamin E
PN, as was seen in 1 of our patients. In addition to hyper- levels from 6.6 to 10.4, similar to other trials.16-18 This is
glycemia, high-dextrose PN formulas that were commonly expected given that most FO ILE contain approximately
used prior to SO ILE being available in the United States 200 mg/L of α-tocopherol.3 α-tocopherol is an antioxidant
were also associated with hepatic enzyme elevations, steato- from the vitamin E family that can scavenge free radicals
sis, and essential fatty acid deficiency (EFAD).3 In addition, that bind to deoxyribonucleic acid and proteins resulting in
our group has also noted that these metabolic complications cell damage and death. In addition to raising α-tocopherol
from high-dextrose formulas limit the number of overall levels, FO ILE also has higher ratio of n-3 poly-unsaturated
calories that can be provided, resulting in the underfeeding fatty acids (PUFAs) when compared with n-6 PUFAs. The
of often malnourished patients. n-6 PUFAs can be further metabolized to give rise to
These issues have led to the exploration of the use of proinflammatory eicosanoids, with ILEs that have a high
alternative ILE with small case series or short-term (5– ratio of n-6 to n-3 PUFA being felt to be proinflammatory
29 days) trials showing promising results.3 Piper et al15 and thus contributing to some of the long-term complica-
randomized 44 postoperative patients who required PN to tions associated with HPN.27-30 Although this was beyond
either MO ILE (n = 22) or olive oil/SO containing ILE the capability of this current study, other randomized trials
(20% ClinOleic) for 5 days. They noted that at days 2 and 5, have shown that FO ILE use can lead to a more beneficial
the MO ILE group had lower AST and ALT levels despite n-3 to n-6 PUFA ratio as well as decreased inflammatory
there being no difference at day 0. Goulet et al17 randomized markers such as interleukin-6 and tumor necrosis factor-
28 children on HPN to either MO ILE or SO ILE (20% α.14,17,18
intralipid) and followed them for 29 days. Although they The current study did have a number of limitations,
found no difference in liver enzymes between the groups, starting with the fact that it was a prospective case series
they did note that the MO ILE group had a reduction in and not a randomized control trial. As a result, all patients
total bilirubin values, whereas the SO ILE group had an were not blinded to the treatment, which could have resulted
increase. Similarly, Klek et al18 randomized 73 adult patients in some bias occurring in terms of tolerance. In fact, some
on HPN as a result of intestinal failure to either MO ILE patients self-requested initiation of MO ILE as soon as it
or SO ILE (20% intralipid) and followed them for 4 weeks. was available in the United States. These patients may have
They noted that mean concentrations of ALT, AST, and been less likely to report any adverse tolerance issues. The
total bilirubin were significantly lower in the MO ILE group study was also a case series and thus patients served as their
when compared with the control group. own control with comparisons being performed between
Our case series also found similar results with all liver values gathered prior to initiation of MO ILE and at the
studies (alkaline phosphatase, AST, ALT, and total biliru- time of analysis. Certainly other clinical factors such as
bin) showing overall improvement although statistical sig- infection or improvement in clinical status could contribute
nificance was not reached because of the small number of to the findings. In addition, most of the patients in the case
patients. However, as opposed to the previous trials, which series had suffered from elevations in liver studies for many
tended to enroll patients who were being initiated on HPN years with recurrent reduction in SO ILE. As a result, labs
and excluding those with liver disease, our cohort consisted that led to the initial reduction in SO ILE were not attainable
of patients who had previously been on HPN for a median and perhaps could have revealed a significant improvement
duration of 9.9 years and had developed an intolerance with transition to MO ILE.
of SO ILE that presented as abnormal liver studies in the Another limitation pertains to the development of
majority of cases. Based on our protocol, ILE frequency EFAD. Because SO has the highest concentration of es-
was reduced to improve or stabilize liver studies, resulting in sential fatty acids (linoleic acid and α-linolenic acid), a
a significant increase in calories from dextrose. At the time reduction in SO content in MO ILE when compared with
that MO ILE was approved for use in adults, 5 patients had SO ILE does raise increased concern for the develop-
been on HPN for close to 10 years or greater and during ment of EFAD.3 In our present case series, no patients
this time multiple attempts had been made to increase SO reported symptoms consistent with EFAD and thus fatty
ILE after the initial reduction led to an improvement in acid profile was not obtained. Literature has estimated that
856 Nutrition in Clinical Practice 33(6)

0.05–0.14 g/kg/day of SO ILE should be sufficient to prevent 4. Varayil JE, Whitaker JA, Okano A, et al. Catheter salvage af-
EFAD.31 In our case series, on average, patients received ter catheter-related bloodstream infection during home parenteral
nutrition. J Parenter Enter Nutr. May 2015;41(3):481-488. https://
0.6 ± 0.4 g/kg/day of MO ILE. As a result, we felt less
doi.org/10.1177/0148607115587018
concern regarding the development of EFAD. In addition, 5. Meguid MM, Kurzer M, Hayashi RJ, Akahoshi MP. Short-term effects
most of the patients in the case series were consuming of fat emulsion on serum lipids in postoperative patients. JPEN J
and possibly absorbing some nutrition from an oral intake Parenter Enteral Nutr. 1989;13(1):77-80.
of variable amounts. Although it was very difficult for us 6. Tashiro T, Mashima Y, Yamamori H, Sanada M, Nishizawa M, Okui
K. Intravenous intralipid 10% vs. 20%, hyperlipidemia, and increase in
to obtain and estimate how much nutrition was absorbed
lipoprotein X in humans. Nutrition. 1992;8(3):155-160.
through oral intake given the anatomic variability such as 7. Xu Z-W, Li Y-S. Pathogenesis and treatment of parenteral nutrition-
short bowel syndrome, some essential fatty acids could have associated liver disease. Hepatobiliary Pancreat Dis Int. 2012;11(6):586-
been obtained through oral intake. As a result, a long-term 593.
randomized trial with the assessment of fatty acid profile 8. Burns DL, Gill BM. Reversal of parenteral nutrition–associated liver
disease with a fish oil–based lipid emulsion (omegaven) in an adult
will be needed to assess whether MO ILE use is associated
dependent on home parenteral nutrition. J Parenter Enter Nutr.
with a higher risk of EFAD when compared with SO ILE. 2013;37(2):274-280. https://doi.org/10.1177/0148607112450301
Until these results are available, caution should be taken 9. Kelly DA. Intestinal failure–associated liver disease: what do
in those who are at high risk for EFAD such as severely we know today? Gastroenterology. 2006;130(2, suppl):S70-S77.
malnourished adults or children. In fact, MO ILE does https://doi.org/10.1053/j.gastro.2005.10.066
10. Cavicchi M, Beau P, Crenn P, Degott C, Messing B. Prevalence
not currently have FDA indication for use in the pediatric
of Liver Disease and Contributing Factors in Patients Receiving
population. Home Parenteral Nutrition for Permanent Intestinal Failure. Ann
Intern Med. 2000;132(7):525-532. https://doi.org/10.7326/0003-4819-
132-7-200004040-00003.
Conclusions 11. Chan S, McCowen KC, Bistrian BR, et al. Incidence, prognosis,
Overall, our case series did reveal that initiation of MO and etiology of end-stage liver disease in patients receiving home
total parenteral nutrition. Surgery. 1999;126(1):28-34. https://doi.org/
ILE in patients with intolerance to SO ILE predominantly
10.1067/msy.1999.98925
because of elevated liver studies allowed for the increase 12. Bishay M, Pichler J, Horn V, et al. Intestinal failure-associated
in percentage of calories obtained from ILE along with liver disease in surgical infants requiring long-term parenteral
a significant reduction in the percentage of calories from nutrition. J Pediatr Surg. 2012;47(2):359-362. https://doi.org/
dextrose, bringing the overall macronutrient composition 10.1016/j.jpedsurg.2011.11.032
13. Tillman EM. Review and clinical update on parenteral nutrition–
closer to the desired amounts. This change was associated
associated liver disease. Nutr Clin Pract. 2013;28(1):30-39.
with no worsening of liver enzyme and function tests, which https://doi.org/10.1177/0884533612462900
instead showed a slight improvement. Long-term prospec- 14. Metry AA, Abdelaal W, Ragaei M, Refaat M, Nakhla G. SMOFlipid
tive randomized control trials are necessary to elucidate versus intralipid in postoperative ICU patients. Enliven J Anesth Crit
the full impact of MO ILE in HPN patients in terms of Care Med. 2014;1(6):15.
15. Piper SN, Schade I, Beschmann RB, Maleck WH, Boldt J, Röhm KD.
inflammation, fat metabolism, and liver function.
Hepatocellular integrity after parenteral nutrition: comparison of a
fish-oil-containing lipid emulsion with an olive-soybean oil-based lipid
Statement of Authorship emulsion. Eur J Anaesthesiol. 2009;26(12):1076-1082.
16. Tomsits E, Pataki M, Tölgyesi A, Fekete G, Rischak K, Szollár
Manpreet S. Mundi and Ryan T. Hurt contributed to the L. Safety and efficacy of a lipid emulsion containing a mixture of
research; Megan T. McMahon, Jennifer Carnell, and Manpreet soybean oil, medium-chain triglycerides, olive oil, and fish oil: a
S. Mundi contributed to acquisition, analysis, or interpretation randomised, double-blind clinical trial in premature infants requiring
of data. Manpreet S. Mundi drafted the manuscript; Megan T. parenteral nutrition. J Pediatr Gastroenterol Nutr. 2010;51(4):514-521.
McMahon, Jennifer Carnell, and Ryan T. Hurt critically revised https://doi.org/10.1097/MPG.0b013e3181de210c
the manuscript; and all authors agree to be fully accountable for 17. Goulet O, Antébi H, Wolf C, et al. A new intravenous fat emulsion con-
ensuring the integrity and accuracy of the work. All authors taining soybean oil, medium-chain triglycerides, olive oil, and fish oil a
read and approved the final manuscript. single-center, double-blind randomized study on efficacy and safety in
pediatric patients receiving home parenteral nutrition. J Parenter Enter
Nutr. 2010;34(5):485-495. https://doi.org/10.1177/0148607110363614
References 18. Klek S, Chambrier C, Singer P, et al. Four-week parenteral nutrition
1. Howard L, Ament M, Fleming CR, Shike M, Steiger E. Current use and using a third generation lipid emulsion (SMOFlipid)—a double-blind,
clinical outcome of home parenteral and enteral nutrition therapies in randomised, multicentre study in adults. Clin Nutr. 2013;32(2):224-231.
the United States. Gastroenterology. 1995;109(2):355-365. https://doi.org/10.1016/j.clnu.2012.06.011
2. Mundi MS, Pattinson A, McMahon MT, Davidson J, Hurt RT. 19. Staun M, Pironi L, Bozzetti F, et al. ESPEN Guidelines on parenteral
Prevalence of home parenteral and enteral nutrition in the United nutrition: home parenteral nutrition (HPN) in adult patients. Clin Nutr.
States. Nutr Clin Pract. 2017;32(6):799-805. https://doi.org/10.1177/ 2009;28(4):467-479. https://doi.org/10.1016/j.clnu.2009.04.001
0884533617718472 20. Carter BA, Shulman RJ. Mechanisms of disease: update on the
3. Mundi MS, Salonen BR, Bonnes S. Home parenteral nutrition: molecular etiology and fundamentals of parenteral nutrition associated
Fat emulsions and potential complications. Nutr Clin Pract. cholestasis. Nat Clin Pract Gastroenterol Hepatol. 2007;4(5):277-287.
2016;31(5):629-641. https://doi.org/10.1177/0884533616663635 https://doi.org/10.1038/ncpgasthep0796
Mundi et al. 857

21. Luis VAS, Btaiche IF. Ursodiol in patients with parenteral nutrition– ther deterioration of liver functions for the TPN patients with impaired
associated cholestasis. Ann Pharmacother. 2007;41(11):1867-1872. liver function. Hepatogastroenterology. 2000;47(35):1347-1350.
https://doi.org/10.1345/aph.1K229 27. Wanten GJ, Calder PC. Immune modulation by parenteral lipid
22. Lindor KD, Burnes J. Ursodeoxycholic acid for the treatment of home emulsions. Am J Clin Nutr. 2007;85(5):1171-1184.
parenteral nutrition-associated cholestasis. A case report. Gastroen- 28. Tilley SL, Coffman TM, Koller BH. Mixed messages: modulation of
terology. 1991;101(1):250-253. inflammation and immune responses by prostaglandins and thrombox-
23. Beau P, Labat-Labourdette J, Ingrand P, Beauchant M. Is ursodeoxy- anes. J Clin Invest. 2001;108(1):15-23.
cholic acid an effective therapy for total parenteral nutrition-related 29. Lewis RA. Interactions of eicosanoids and cytokines in immune
liver disease? J Hepatol. 1994;20(2):240-244. regulation. Adv Prostaglandin Thromboxane Leukot Res. 1990;20:170-
24. Günşar C, Vatansever S, Var A, et al. Antibiotic treatment is 178.
superior to ursodeoxycholic acid on total parenteral nutrition as- 30. Vanek VW, Seidner DL, Allen P, et al. A.S.P.E.N. position paper
sociated hepatic dysfunction. Pediatr Surg Int. 2010;26(5):479-486. clinical role for alternative intravenous fat emulsions. Nutr Clin Pract.
https://doi.org/10.1007/s00383-010-2578-5 2012;27(2):150-192. https://doi.org/10.1177/0884533612439896
25. Maini B, Blackburn GL, Bistrian BR, et al. Cyclic hyperalimentation: 31. Gramlich L, Meddings L, Alberda C, et al. Essential fatty acid
An optimal technique for preservation of visceral protein. J Surg Res. deficiency in 2015: the impact of novel intravenous lipid emul-
1976;20(6):515-525. https://doi.org/10.1016/0022-4804(76)90085-8 sions. J Parenter Enter Nutr. 2015;39(1 suppl):61S-66S. https://doi.
26. Hwang TL, Lue MC, Chen LL. Early use of cyclic TPN prevents fur- org/10.1177/0148607115595977
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 6
Nutrition Status Among HIV-Positive and HIV-Negative December 2018 858–864

C 2018 American Society for

Inpatients with Pulmonary Tuberculosis Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10006
wileyonlinelibrary.com

Tássia Kirchmann Lazzari, MSc1 ; Gabriele Carra Forte, PhD2 ;


and Denise Rossato Silva, MD, PhD3

Abstract
Background: The association between tuberculosis (TB) and malnutrition is well recognized. Considering the risk of mortality
due to malnutrition in patients with TB, it is necessary to conduct a thorough nutrition assessment to identify individuals at
nutrition risk. The study objective was to assess the nutrition status of hospitalized patients with TB, co-infected or not by human
immunodeficiency virus (HIV). Methods: Patients with confirmed diagnosis of TB were included using a cross-sectional design.
Nutrition assessment parameters included: body mass index (BMI), triceps skin-fold thickness (TSF), bioelectrical impedance
analysis (BIA), mid-upper-arm circumference (MUAC), mid-arm muscle circumference (MAMC), food frequency questionnaire,
Malnutrition Screening Tool (MST), Subjective Global Assessment (SGA), and serum levels of hemoglobin. Results: A total 108
patients completed the study. Forty-four patients (40.7%) were HIV positive. Considering the BMI, 36.1% of the patients met the
criteria for nutrition deficiency. Body fat percentage was low in 27.8% of patients. In addition, more than half of the participants met
criteria for malnutrition according to MUAC, MAMC, TSF, SGA, or MST. Malnutrition measured by MAMC was more frequent
in HIV-positive patients (n=33, 75.0%) than in HIV-negative patients (n=31, 48.4%) (P = 0.010). Regarding the components of diet,
selenium and vitamin C intake among HIV-positive patients was significantly lower than in HIV-negative patients. Conclusions: We
identified a high prevalence of malnutrition in hospitalized patients with pulmonary TB, regardless of the method used to assess
nutrition status. In HIV-positive patients, malnutrition measured by MAMC was more frequent than in HIV-negative patients.
(Nutr Clin Pract. 2018;33:858–864)

Keywords
ascorbic acid; body mass index; electrical impedance; nutritional status; selenium; skin fold thickness; tuberculosis

Tuberculosis (TB) is a major public health problem world- as interleukin-6 (IL-6) and tumor necrosis factor (TNF)-
wide, particularly in low and middle income populations. α, that are anorectic and also responsible for metabolic
It is estimated that one-third of the world population is changes which in most cases result in increased mortality
infected with Mycobacterium tuberculosis. Brazil is in 16th
place among the 22 countries that collectively account for
From the 1 Programa de Pós-Graduação em Ciências Pneumológicas
80% of TB cases globally, with a reported incidence of 30.9
da Universidade Federal do Rio Grande do Sul, Rio Grande do Sul,
cases/100,000 inhabitants/year in 2015.1,2 Porto Alegre is Brazil; 2 Programa de Pós-Graduação em Ciências Pneumológicas da
the second Brazilian capital with the highest number of TB Universidade Federal do Rio Grande do Sul, Rio Grande do Sul,
cases, with an incidence of 88.8 cases/100,000 inhabitants Brazil; 3 Faculdade de Medicina, Universidade Federal do Rio Grande
and a high percentage of TB-human immunodeficiency do Sul, Serviço de Pneumologia, Hospital de Clı́nicas de Porto Alegre,
Programa de Pós-Graduação em Ciências Pneumológicas da
virus (HIV) (25.2%).1 Universidade Federal do Rio Grande do Sul, Rio Grande do Sul,
The association between TB and malnutrition is well Brazil.
recognized; TB can lead to malnutrition and malnutrition Financial disclosure: FIPE-HCPA (Fundo de Incentivo à Pesquisa –
may predispose to TB. The protein-energy malnutrition Hospital de Clı́nicas de Porto Alegre).
observed in patients with TB with or without infection
Conflicts of interest: None declared.
by HIV is characterized by reduction of visceral protein,
Received for publication June 22, 2017; accepted for publication
lean mass and fat, and a significant decrease of some September 9, 2017.
anthropometric and biochemical changes (Hb).3 Moreover,
This article originally appeared online on February 3, 2018.
malnutrition is associated with fatigue of the respiratory
muscles, leading to worsening disease prognosis.4,5 Corresponding author:
Denise Rossato Silva, MD, PhD, Serviço de Pneumologia, Hospital
The systemic response to infection, comprising neuro- de Clı́nicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 2° andar,
humoral and immunologic changes, is characterized by Porto Alegre, RS, CEP 90.035-003, Brazil.
increased production of proinflammatory cytokines, such Email: denise.rossato@terra.com.br
Kirchmann Lazzari et al 859

in TB patients.6-9 Moreover, in TB there is reduction of smoker was defined as smoking at least 100 cigarettes in
liver production of albumin, apolipoprotein, transferrin, their lifetime and at the time of the survey, smoking at least
and thus, Hb. Simultaneously, the synthesis of acute-phase 1 day a week. A former smoker was defined as smoking
proteins increases, including C-reactive protein (CRP).6 at least 100 cigarettes in their lifetime but at the time of
The lower the lymphocyte count, the more susceptible to the survey, did not smoke at all. Never smoked reported
opportunistic infections the patient becomes and greater having smoked <100 cigarettes in their lifetime. Alcohol
will be the energy expenditure, progressing to severe, and abuse was defined as daily consumption of at least 30
in some cases intractable, weight loss.10,11 Another study grams (equivalent to 24 ounces of 4% beer) for men and 24
showed that low Hb levels are related to malnutrition and grams (equivalent to a 175-mL glass of wine) for women.
poor prognosis.12 An independent physician analyzed the chest X-rays and
The effects of malnutrition on hospitalized patients with classified them as typical or compatible with active TB
TB are not well known. Many studies are retrospective and according to previously described guidelines.21
the results are inconsistent.13-17 There are several limitations
in many studies regarding the body composition of patients Nutrition Assessment
with TB, with or without HIV. There is lack of information
Nutrition assessment was performed by body mass index
on food intake. Another limitation is that the lymphocyte
(BMI), triceps skin-fold thickness (TSF), mid-arm circum-
cell count is not carried out to evaluate the effect of HIV
ference, mid-arm muscle circumference (MAMC), bioelec-
on disease severity.18,19 Therefore, the aim of this study is to
tric impedance analysis (BIA), food frequency question-
assess the nutrition status of hospitalized patients with TB
naire (FFQ), Malnutrition Screening Tool (MST), Subjec-
co-infected or not by HIV.
tive Global Assessment (SGA), and laboratory tests. The
same researcher made all of the measurements.
Methods
Study Design and Location BMI. BMI (kg/m2 ) was calculated and the patients were
classified into categories based on the BMI cutoffs for
We conducted a cross-sectional study in a general, tertiary weight categories as recommended by the WHO.22 Weight
care, university-affiliated hospital with 750 beds. The study was measured using a standing scale. No patient had fluid
was approved by the Ethics Committee at the hospital on overload that would affect BMI. Malnutrition was defined
December 19, 2012 (number 13-0002). as a BMI <18.5 kg/m2 .22

Patients TSF. TSF thickness was measured with a skin caliper on


the posterior upper arm midway between the acromion and
Adult patients (age ࣙ18 years) with pulmonary TB who
olecranon processes.23 The values obtained were compared
were hospitalized during the study period were identified
with reference values.24
and invited to participate. Patients with extrapulmonary TB,
those who were receiving treatment for >1 week, those who
were unable to comply with study procedures, and those Mid-upper-arm circumference (MUAC). MUAC was mea-
who refused to sign the consent form were excluded from sured using a non-stretch plastic tape midway between the
this study. We also excluded patients with renal or hepatic acromion and olecranon of the non-dominant arm.25 The
insufficiency and those with peripheral edema. Pulmonary values obtained were compared with reference values.26
TB was diagnosed according to the Brazilian Guidelines for
Tuberculosis.20 MAMC. MAMC was calculated by using MUAC and TSF
measurements (MUAC [cm] – 3.142 x TSF [cm]). The values
obtained were compared with reference values.27
Data Collection
The following data were collected from patient records using BIA. BIA is a simple, non-invasive technique that has been
a standardized data extraction tool: demographic data (sex, recommended for clinical investigation of body composi-
age, race, years of schooling), behavioral data (smoking tion analysis. A single-frequency BIA was performed at 50
status, alcohol abuse, injection drug use), and medical kHz and 800 mA with standard tetrapolar lead placement
history (clinical form of TB, symptoms at admission, to measure fat and lean tissue mass. All BIA measurements
methods of diagnosis, presence of comorbidities, prior TB were performed by 1 trained observer using the same
treatment, drug regimen, interval from hospital admission equipment and recommended standard conditions.28 Fat
until diagnosis, length of hospital stay, intensive care unit mass and fat-free mass (FFM) were estimated from the
[ICU] admission, length of mechanical ventilation, and values of resistance and reactance obtained through BIA,
hospitalization outcome [death or discharge]). A current using sex-specific equations validated by Kotler et al29 in a
860 Nutrition in Clinical Practice 33(6)

sample of white, black, and Hispanic patients that included Table 1. Characteristics of Hospitalized Patients With
HIV-infected individuals. Tuberculosis.

Characteristics n=108
FFQ. To assess daily food intake we used a FFQ that
is reproducible and validated.30 The study’s nutritionists Male sex, n (%) 77 (71.3)
reviewed the diet records and resolved any questions with Age (years), mean ± SD 44.9 ± 15.5
the participant. We calculated the mean daily intake of White race, n (%) 60 (55.6)
macronutrients and micronutrients using NutWin - Nu- Current smoking, n (%) 45 (41.7)
Alcoholism, n (%) 42 (38.9)
trition Support Program software, version 1.5 (Health’s
Drug use, n (%) 37 (34.3)
Informatics Department, Escola Paulista de Medicina, Uni- HIV, n (%) 44 (40.7)
versidade Federal de São Paulo, São Paulo, Brazil). The Smear positive, n (%) 74 (68.5)
FFQ was used to assess usual intake before hospitalization Chest X-ray typicala of TB, n (%) 67 (62.0)
(food intake in the past month) because our idea was to Chest X-ray compatibleb with TB, n (%) 41 (38.0)
see the effects of the disease in nutrition status and not the In-hospital mortality, n (%) 9 (8.3)
effects of hospitalization in nutrition status. Hb (g/dL), mean ± SD 10.4 ± 2.1
BMI (kg/m2 ), mean ± SD 20.7 ± 4.5
Body fat (%, BIA), mean ± SD 24.1 ± 10.6
SGA. SGA is a valid and reliable tool which assesses Body fat (%,TSF), mean ± SD 22.8 ± 9.2
nutrition status based on the features of a medical history SGA A, n (%) 5 (4.6)
and physical examination. Subjects were rated as being well SGA B, n (%) 63 (58.3)
nourished (SGA A); moderately, or suspected of being, SGA C, n (%) 40 (37.0)
malnourished (SGA B); or severely malnourished (SGA C). MST 1, n (%) 7 (6.5)
MST 2, n (%) 101 (93.5)
It is widely used both in hospital care and in studies to assess
Malnutrition by MUAC, n (%) 69 (63.9)
nutrition status.31 Malnutrition by MAMC, n (%) 64 (59.3)
Malnutrition by TSF, n (%) 64 (59.3)
MST. The nutrition risk assessment was carried out using
a Nodular, alveolar, or interstitial infiltrates predominantly affecting
the MST. It incorporates 3 components: the presence of
the zones above the clavicles or upper zones; cavitation affecting the
weight loss (score 0 or 2), the amount of weight lost (score upper zones or the apical segment of the lower lobe. b Enlarged hilar
1–4), and poor food intake or poor appetite (score 0 or 1). nodes, pneumonic lesion, atelectasis, mass lesion, miliary. BMI, body
The total score was calculated for each patient. A score ࣘ2 mass index. BIA, bioelectrical impedance analysis. Hb, hemoglobina.
means that the patient is at risk for malnutrition. It has been HIV, human immunodeficiency virus. MAMC, mid-arm muscle
circumference. MST, malnutrition screeening tool (1: not at risk of
used in patients with TB as a tool capable of predicting malnutrition; 2: at risk of malnutrition). MUAC, mid upperarm
prognosis due to the nutrition status of these patients. It has circumference. SD, standard deviation. SGA, subjective global
the advantage of being easy, fast, and simple to use and does assessment (A: well nourished; B: moderately, or suspected of being,
not require calculations. Its sensitivity and specificity is 93% malnourished; C: severely malnourished). TB, Tuberculosis. TSF,
triceps skin-fold thickness.
compared with the SGA, but both can and should be used
together for optimal nutrition assessment.32-34
confidence level, we estimated a sample size of at least 21
Laboratory testing. Blood samples were collected for Hb patients per group.
dosage. Anemia was determined when the Hb levels were
<13 g/dL (men) and <12 g/dL (women).35 Results
During the study period, 108 patients met the inclusion
Statistical Analysis criteria and were included in the analysis. The characteristics
Data analysis was performed using SPSS 18.0 (Statistical of the study population are shown in Table 1. The mean
Package for the Social Sciences, Chicago, Illinois). Data age of all patients was 44.9 ± 15.5 years, 71.3% were males,
were presented as number of cases, mean ± standard devi- and 55.6% were white. Forty-four patients (40.7%) were HIV
ation (SD), or median with interquartile range (IQR). Cat- positive. BMI was ࣘ18.5 kg/m2 in 39 (36.1%) patients. The
egoric comparisons were performed by χ 2 test using Yates’ majority of patients were considered malnourished or at
correction if indicated or by Fisher’s exact test. Continuous risk of malnutrition by SGA, MST, MUAC, MAMC, and
variables were compared using the t-test or Wilcoxon test. TSF evaluations.
A 2-sided P value <0.05 was considered significant for Table 2 shows the comparisons between HIV-positive
all analyses. Sample size calculation for the comparison and HIV-negative patients. Malnutrition measured by
between HIV-positive and HIV-negative groups was based MAMC was more frequent in HIV-positive patients (n =
on a previous study.36 Considering 90% power and a 95% 33, 75.0%) than in HIV-negative patients (n = 31, 48.4%)
Kirchmann Lazzari et al 861

Table 2. Comparison Between HIV Positive and HIV Negative Patients With Tuberculosis.

Characteristics HIV positive n=44 HIV negative n=64 p value

Male sex, n (%) 27 (61.4) 50 (78.1) 0.094


Age (years), mean ± SD 39.2 ± 10.3 48.9 ± 17.2 <0.0001
White race, n (%) 15 (34.1) 45 (70.3) <0.0001
Current smoking, n (%) 22 (50.0) 23 (35.9) 0.208
Alcoholism, n (%) 19 (43.2) 23 (35.9) 0.577
Drug use, n (%) 23 (52.3) 14 (21.9) 0.002
Smear positive, n (%) 29 (65.9) 45 (70.3) 0.785
Chest X-ray typicala of TB, n (%) 24 (54.5) 43 (67.2) 0.259
Chest X-ray compatibleb with TB, n (%) 21 (47.7) 20 (31.3) 0.126
In-hospital mortality, n (%) 3 (6.8) 6 (9.4) 0.906
Hb (g/dL), mean ± SD 9.6 ± 1.7 10.9 ± 2.2 0.001
Selenium (μg), mean ± SD 0.011± 0.053 0.027 ± 0.069 0.02
Vitamin C (mg), median (P25-P75) 17.9 (6.3-59.0) 38.5 (24.4-107.7) 0.005
BMI (kg/m2 ), mean ± SD 19.87 ± 3.8 21.3 ± 4.9 0.079
Body fat (%, BIA), mean ± SD 24.2 ± 11.4 24.1 ± 10.1 0.951
Body fat (%,TSF), mean ± SD 21.2 ± 8.2 23.8 ± 9.7 0.143
SGA A, n (%) 2 (4.5) 3 (4.7) 0.999
SGA B, n (%) 23 (52.3) 40 (62.5) 0.389
SGA C, n (%) 19 (43.2) 21 (32.8) 0.371
MST 1, n (%) 4 (9.1) 3 (4.7) 0.440
MST 2, n (%) 40 (90.9) 61 (95.3) 0.440
Malnutrition by MUAC, n (%) 32 (72.7) 37 (57.8) 0.167
Malnutrition by MAMC, n (%) 33 (75.0) 31 (48.4) 0.010
Malnutrition by TSF, n (%) 27 (61.4) 37 (57.8) 0.865
a Nodular, alveolar, or interstitial infiltrates predominantly affecting the zones above the clavicles or upper zones; cavitation affecting the upper

zones or the apical segment of the lower lobe. b Enlarged hilar nodes, pneumonic lesion, atelectasis, mass lesion, miliary. BIA, bioelectrical
impedance analysis. BMI, body mass index. CRP, C-reactive protein. Hb, hemoglobina. HIV, human immunodeficiency virus. MAMC, mid-arm
muscle circumference. MST, malnutrition screeening tool (1: not at risk of malnutrition; 2: at risk of malnutrition). MUAC, mid upper arm
circumference. P25-P75: percentile 25-percentile 75. SD, standard deviation. SGA, subjective global assessment (A: well nourished; B: moderately,
or suspected of being, malnourished; C: severely malnourished). TB, Tuberculosis. TSF, triceps skin-fold thickness.

(P = 0.010). Regarding diet components, only selenium India, the prevalence of malnutrition by BMI was ˃85%.38
and vitamin C intake were lower in HIV-positive patients There is much discussion about the use of BMI as the only
compared with HIV-negative patients (P = 0.02 and P = method for nutritionl assessment due to its limitations.39.40
0.005, respectively). The other components of the diet were Thus, a variety of objective and subjective measures have
not statistically different between groups (data not shown). been used to diagnose malnutrition or identify patients at
risk of malnutrition. Using some of these measures, such
as BIA, TSF, MUAC, and MAMC, we found a higher
Discussion prevalence of malnutrition than by using only BMI. Still,
In this study, we found a high prevalence of malnutrition in using the SGA, previously evaluated in patients with TB,41
hospitalized patients with newly diagnosed pulmonary TB, 95.7% of patients in our sample had moderate or severe
regardless of the parameter used to assess nutrition status. malnutrition. We also used the MST to assess nutrition
Considering the BMI, more often used in studies, 36.1% of status, a tool that already was shown to be useful in the
the patients met the criteria for malnutrition. In addition, evaluation of TB patients,33 and it showed that 93.5%
27.8% of patients had a low body fat percentage. More than of patients in the present study were malnourished or at
half of the individuals studied had criteria for malnutrition risk of malnutrition. Therefore, many patients considered
according to MUAC, MAMC, TSF, SGA, or MST. having normal weight, overweight, or obesity by BMI were
The prevalence of malnutrition varies across studies. Two classified as malnourished or at risk of malnutrition by
other studies also conducted in Brazil, 1 in São Paulo37 other nutrition assessment methods. We then consider that
and another in Rio de Janeiro,8 showed results similar to BMI has limitations for nutrition assessment also in patients
those found in the present study, with a prevalence of 34.9% with TB and that other measures should be used together.
and 32.0%, respectively, for malnutrition according to BMI. Among the other measures used in our study, it is difficult
However, in a study conducted in a rural population of to say which ones are the best or if all of them are needed in
862 Nutrition in Clinical Practice 33(6)

future studies. We don’t think it will be possible to establish between diet and TB, nutrients examined vary, but include
a gold standard for the evaluation of malnutrition in TB proteins; vitamins A, C, D, and E; zinc; selenium; iron;
patients. Unfortunately, the present study was not designed copper; cholesterol; and polyunsaturated fatty acids.43,52
to calculate sensitivity and specificity of each tool. In spite Micronutrient deficiency is considered the most frequent
of that, a single parameter probably will not be able to reflect cause of morbidity and secondary immunodeficiency asso-
nutrition deficits among inpatients with TB. ciated with infection, including TB.43 In our study, HIV-
Malnutrition and TB are both problems of developing positive patients had a lower intake of selenium and vita-
countries. TB can lead to malnutrition and malnutrition min C than HIV-negative patients, and both were below
may predispose to TB. Poor nutrition leads to protein- the RDA. The essential trace element selenium plays an
energy malnutrition and micronutrient deficiencies that lead important role in maintaining the immune process and
to immunodeficiency. This secondary immunodeficiency thus may play a critical role in the removal of mycobacte-
increases the susceptibility of the host to infections and ria. Deficiencies in micronutrients, including selenium, are
thus increases the risk of developing TB. TB alone leads widespread in HIV-infected individuals, most notably under
to reduced appetite, malabsorption of nutrients and mi- resource-limited conditions.53 Selenium has been described
cronutrients, and altered metabolism causing poor nutrition as a significant risk factor in the development of disease
status.42 by mycobacteria in HIV-positive patients.54 Vitamin C also
Nutrition status appears to be an important determinant has a key function in pulmonary antioxidant defense,55 and
of clinical outcomes during TB treatment. Malnutrition studies have reported low concentrations of this vitamin in
can adversely affect treatment outcomes, being linked to TB patients.55,56 Orally administered vitamin C has been de-
excess deaths43-45 and increased risk of relapse.44,46 Severe scribed to have beneficial effects in TB,57,58 even increasing
malnutrition at diagnosis was associated with a 2-fold higher the antibacterial activity of rifampin and isoniazid against
risk of death.38 In a study in Malawi, a high degree of M. tuberculosis.59
malnutrition was a risk factor for early mortality among This study has some methodologic limitations. First, this
TB patients.45 One study suggests that malnutrition inter- was a single-center study and included only hospitalized
vention could have a substantial impact on TB mortality patients, probably the most severe cases of TB. Second, the
in areas with high prevalence of malnutrition, even if only reduced number of deaths does not allow the evaluation
small gains in malnutrition can be achieved.47 In addition, of nutrition parameters as predictors of mortality. Also,
restoration of nutrition could also lead to immunologic single-frequency BIA is less accurate than multifrequency
changes that could enhance the clearance of mycobacteria BIA; however, it was used and also validated previously in
and reduce infectiousness of patients.43 One study showed hospitalized patients with other diseases.60,61 In addition,
that nutrition support can improve both sputum smears or we used FFQ to determine micronutrients; however, FFQ is
culture-negative conversion rate and BMI, shortening the a reasonable instrument for assessing the intake of most mi-
time of sputum conversion.48 In another study, patients re- cronutrients when compared with nutrition biomarkers. The
ceiving better nutrition tended to show more rapid clearance advantages of this method are that the burden on examiners
of bacteria and radiographic changes in addition to greater and examinees is small, the cost is low, and self-recording
weight gain.49 We collected data on mortality in our study; is feasible. On the other hand, it is limited because of the
however, we had only 9 deaths and could not evaluate risk limited food categories and intake amount per meal; thus,
factors for mortality, including malnutrition. the measurement of the intake amount is less accurate than
High prevalence of HIV infection in underdeveloped with the dietary record method or the recall method.62-64
countries further aggravates the problems of malnutrition Therefore, considering the advantages and shortcomings of
and TB. In a study that compared nutrition status of these methods, we used the FFQ method in the initial period
individuals with HIV alone, TB-HIV, and HIV-negative, to assess the routine general intake pattern over a long
the authors found that malnutrition was most pronounced period of time because it is considered to be a useful method
among patients with TB-HIV coinfection.50 Net protein for examining the relationship between diet and diseases.
anabolism is impaired in patients in HIV-positive adults Nevertheless, despite these limitations, our study reaches its
with TB, and this impairment is significantly greater than goal to show nutrition deficiencies in patients hospitalized
that seen in patients with HIV or TB alone.43 We found with pulmonary TB. These results are applicable to other
differences in malnutrition between HIV-positive and HIV- regions with similar TB and HIV incidences. Considering
negative patients only by MAMC. A study conducted in that the complications of malnutrition increase the length
Guinea-Bissau also showed a low MAMC in HIV-infected of hospital stay and costs,65 it is important to conduct
TB patients, and this variable was associated with greater an adequate nutrition assessment of patients at hospital
mortality.51 admission. Nutrition supplementation may be an approach
The interaction between nutrition deficiencies and infec- to rapid recovery and improve outcomes in patients with
tion is well known. In studies evaluating the relationship TB.66,67
Kirchmann Lazzari et al 863

Conclusion 11. Ramos LM, Sulmonett N, Ferreira CS et al. Functional profile of


patients with tuberculosis sequelae in a university hospital. J Bras
This study identified a high prevalence of malnutrition in Pneumol. 2006;32(1):43-47.
patients with pulmonary TB admitted in a tertiary hospital, 12. Van LM, Kumwenda JJ, Harries AD, et al. Malnutrition and the
regardless of the method used to assess nutrition status. In severity of lung disease in adults with pulmonary tuberculosis in
Malawi. Int J Tuberc Lung Dis. 2004;8(2):211-217.
HIV-positive patients, malnutrition measured by MAMC 13. Cakir B, Yonem A, Guler S, et al. Relation of leptin and tumor necrosis
was more frequent than in HIV-negative patients. Further- factor alpha to body weight changes in patients with pulmonary
more, we demonstrated that selenium and vitamin C intake tuberculosis. Horm Res. 1999;52(6):279-283.
were significantly lower in HIV-positive patients than in 14. Kim JH, Lee CT, Yoon HI, et al. Relation of ghrelin, leptin and inflam-
HIV-negative patients. Further studies with a larger sample matory markers to nutritional status in active pulmonary tuberculosis.
Clin Nutr. 2010; 29(4):512-518.
size are needed to clarify these findings. 15. Langhans W. Anorexia of infection: current prospects. Nutrition. 2000;
16(10):996-1005.
Acknowledgments 16. Schwenk A, Hodgson L, Rayner CF et al. Leptin and energy
metabolism in pulmonary tuberculosis. Am J Clin Nutr. 2003;77(2):392-
We would like to acknowledge the support from the Interna- 398.
tional Clinical Operational Health Services Research Training 17. van Crevel R, Karyadi E, Netea MG, et al. Decreased plasma
Award (ICOHRTA/Fogarty International Center/National In- leptin concentrations in tuberculosis patients are associated with
stitutes for Health-NIH) and Johns Hopkins University (Johns wasting and inflammation. J Clin Endocrinol Metab. 2002;87(2):758-
Hopkins Bloomberg School of Public Health). 763.
18. Ando M, Mori A, Esaki H, et al. The effect of pulmonary re-
habilitation in patients with post-tuberculosis lung disorder. Chest.
Statement of Authorship 2003;123(6):1988-1995.
T. Kirchmann Lazzari is responsible for literature search, study 19. Maguire GP, Anstey NM, Ardian M, et al. Pulmonary tuberculosis,
impaired lung function, disability and quality of life in a high-burden
design, data collection, and review of the manuscript. G. Carra
setting. Int J Tuberc Lung Dis. 2009;13(12):1500-1506.
Forte did literature search, study design, analysis of data, and 20. Conde MB, Melo FA, Marques AM, et al. III Brazilian Thoracic Asso-
review of the manuscript. D. Rossato Silva did literature search, ciation Guidelines on tuberculosis. J Bras Pneumol. 2009;35(10):1018-
study design, analysis of data, manuscript preparation, and 1048.
review of the manuscript. 21. Diagnostic Standards and Classification of Tuberculosis in Adults and
Children. This official statement of the American Thoracic Society
and the Centers for Disease Control and Prevention was adopted
References by the ATS Board of Directors, July 1999. This statement was en-
1. Secretariat of Health Surveillance – Brazilian Ministry of Health. Epi- dorsed by the Council of the Infectious Disease Society of America,
demiological Bulletin 2016. Available at: www.saude.gov.br. Accessed: September 1999. Am J Respir Crit Care Med. 2000;161(4 Pt 1):1376-
June 2017. 1395.
2. World Health Organization. Global tuberculosis control: WHO report 22. World Health Organization. BMI classification – Global Database on
2015. Available at: www.who.int. Accessed: June 2017. Body Mass Index. 2015. Available at: www.who.int. Accessed: June
3. Brazilian Ministry of Health. National Tuberculosis Control Program 2017.
2011. Available at: www.saude.gov.br. Accessed: June 2017. 23. Heymsfield SB, Baugmgartier RN, Sheau-Fang R. Avaliação nutri-
4. Range N, Andersen AB, Magnussen P, et al. The effect of micronutrient cional da desnutrição por métodos antropométricos. In: Shils ME,
supplementation on treatment outcome in patients with pulmonary OJSMRAC, ed. Tratado de nutrição moderna na saúde e na doença. São
tuberculosis: a randomized controlled trial in Mwanza, Tanzania. Trop Paulo: Manoel; 2003;1168-1190.
Med Int Health. 2005; 10(9):826-832. 24. de Onis M, Habicht JP. Anthropometric reference data for interna-
5. Visser ME, Grewal HM, Swart EC, et al. The effect of vitamin A and tional use: recommendations from a World Health Organization Expert
zinc supplementation on treatment outcomes in pulmonary tuberculo- Committee. Am J Clin Nutr. 1996;64(4):650-658.
sis: a randomized controlled trial. Am J Clin Nutr. 2011;93(1):93-100. 25. National Health and Nutrition Examination Survey (NHANES).
6. Costa AE. Resposta imune humoral ao antı́geno registros do my- Anthropometric procedures manual. 2007. Available at: http://
cobacterium tuberculosis em pacientes com tuberculose e seus contatos www.cdc.gov/nchs/data/nhanes/nhanes_07_08/manual_an.pdf.
domiciliares. Rev Patol Trop. 2011;40(1):23-34. Accessed: June 2017.
7. Cruz RC, De Albuquerque MF, Campelo AR, et al. Pulmonary 26. Tang AM, Dong K, Deitcher M, Chung M, Maalouf-Manasseh Z,
tuberculosis: association between extent of the residual pulmonary Tumilowicz A, et al. Food and Nutrition Technical Assistance III Project
lesion and alteration in the lung function [in Portuguese]. Rev Assoc (FANTA) Washington, DC: United States Agency for International
Med Bras. 2008;54(5):406-410. Development (USAID); 2013. Use of cutoffs for Mid-Upper Arm Cir-
8. Ramalho RA. Avaliação Nutricional de pacientes com tuberculose cumference (MUAC) as an Indicator or Predictor of Nutritional and
pulmonar antendidos na UISHL. Bol Pneumol Sanitaria. 2000;8(2):13- Health-Related Outcomes in Adolescents and Adults: A Systematic
19. Review. 2016:1-39.
9. Teixeira HC, Abramo C, Munk ME. Immunological diagnosis of 27. Weber JKJ. Assessing nutrition. In: Nieginski E, ed. Health As-
tuberculosis: problems and strategies for success. J Bras Pneumol. sessment in Nursing. Philadelphia: Lippincott Williams & Wilkins;
2007;33(3):323-334. 2003.
10. Nogueira CR. Aspectos antropométricos, bioquı́micos e sintoma- 28. Kyle UG, Genton L, Karsegard L, et al. Single prediction equation for
tológicos em mulheres com tuberculose pulmonar. Rev Ciênc Med. bioelectrical impedance analysis in adults aged 20–94 years. Nutrition.
2006;15(4):281-288. 2001; 17(3):248-253.
864 Nutrition in Clinical Practice 33(6)

29. Kotler DP, Burastero S, Wang J, et al. Prediction of body cell mass, fat- 50. Swaminathan S, Padmapriyadarsini C, Sukumar B, et al. Nutritional
free mass, and total body water with bioelectrical impedance analysis: status of persons with HIV infection, persons with HIV infection and
effects of race, sex, and disease. Am J Clin Nutr. 1996;64(3 Suppl):489S- tuberculosis, and HIV-negative individuals from southern India. Clin
497S. Infect Dis 2008;46(6):946-949.
30. Molina MC, Bensenor IM, Cardoso LO, et al. Reproducibility and 51. Gustafson P, Gomes VF, Vieira CS, et al. Clinical predictors for death
relative validity of the Food Frequency Questionnaire used in the in HIV-positive and HIV-negative tuberculosis patients in Guinea-
ELSA-Brasil [in Portuguese]. Cad Saude Publica. 2013;29(2):379-389. Bissau. Infection. 2007;35(2):69-80.
31. Detsky AS, McLaughlin JR, Baker JP, et al. What is subjective 52. Cegielski JP, McMurray DN. The relationship between
global assessment of nutritional status? JPEN J Parenter Enteral Nutr. malnutrition and tuberculosis: evidence from studies in humans
1987;11(1):8-13. and experimental animals. Int J Tuberc Lung Dis. 2004;8(3):286-
32. Ferguson M, Capra S, Bauer J, et al. Development of a valid and 298.
reliable malnutrition screening tool for adult acute hospital patients. 53. Irlam JH, Siegfried N, Visser ME, et al. Micronutrient supplemen-
Nutrition. 1999; 15(6):458-464. tation for children with HIV infection. Cochrane Database Syst Rev.
33. Miyata S, Tanaka M, Ihaku D. Usefulness of the Malnutrition 2013;(10):CD010666.
Screening Tool in patients with pulmonary tuberculosis. Nutrition. 54. Shor-Posner G, Miguez MJ, Pineda LM, et al. Impact of selenium
2012;28(3):271-274. status on the pathogenesis of mycobacterial disease in HIV-1-infected
34. Reilly HM, Martineau JK, Moran A, et al. Nutritional screening– drug users during the era of highly active antiretroviral therapy.
evaluation and implementation of a simple Nutrition Risk Score. Clin J Acquir Immune Defic Syndr. 2002; 29(2):169-173.
Nutr. 1995;14(5):269-273. 55. Bakaev VV, Duntau AP. Ascorbic acid in blood serum of patients
35. Bottoni A, Oliveira GPC, Ferrini MT, Waitzberg DL, Avaliação with pulmonary tuberculosis and pneumonia. Int J Tuberc Lung Dis.
nutricional: exames laboratoriais . In: Waitzberg DL, ed. Nutrição oral, 2004;8(2):263-266.
enteral e parenteral na prática clı́nica. 3rd. Edition. São Paulo: Atheneu, 56. Andosca JB, Foley JA. Calcium ribonate and vitamin C (Nu 240-10) in
2000:279-294. the treatment of tuberculosis. Dis Chest. 1948;14(1):107-114.
36. Ramakrishnan K, Shenbagarathai R, Kavitha K, et al. Serum zinc and 57. Hemila H, Kaprio J, Pietinen P, et al. Vitamin C and other compounds
albumin levels in pulmonary tuberculosis patients with and without in vitamin C rich food in relation to risk of tuberculosis in male
HIV. Jpn J Infect Dis. 2008;61(3):202-204. smokers. Am J Epidemiol.1999;150(6):632-641.
37. Souza ARA, Tudisco ES. Avaliação nutricional de pacientes tubercu- 58. McConkey M, Smith DT. The Relation Of Vitamin C Deficiency To
losos em tratamento ambulatorial. J Pneumol. 1992;18(4):167-170. Intestinal Tuberculosis In The Guinea Pig. J Exp Med. 1933;58(4):503-
38. Bhargava A, Chatterjee M, Jain Y, et al. Nutritional status of adult 512.
patients with pulmonary tuberculosis in rural central India and its 59. Khameneh B, Fazly Bazzaz BS, Amani A, et al. Combination of
association with mortality. PLoS One. 2013;8(10):e77979. anti-tuberculosis drugs with vitamin C or NAC against different
39. Bhurosy T JR. Pitfalls Of Using Body Mass Index (BMI) In Assess- Staphylococcus aureus and Mycobacterium tuberculosis strains. Microb
ment Of Obesity Risk. Curr Res Nutr Food Sci. 2013;1(1):71-76. Pathog. 2016;93:83-87.
40. Garn SM, Leonard WR, Hawthorne VM. Three limitations of the body 60. Selberg O, Selberg D. Norms and correlates of bioimpedance
mass index. Am J Clin Nutr. 1986;44(6):996-997. phase angle in healthy human subjects, hospitalized patients, and
41. Miyata S, Tanaka M, Ihaku D. Subjective global assessment in patients patients with liver cirrhosis. Eur J Appl Physiol. 2002;86(6):509-
with pulmonary tuberculosis. Nutr Clin Pract. 2011;26(1):55-60. 516.
42. Kant S, Gupta H, Ahluwalia S. Significance of nutrition in pulmonary 61. Yalin SF, Gulcicek S, Avci S, et al. Single-frequency and multi-
tuberculosis. Crit Rev Food Sci Nutr. 2015;55(7):955-963. frequency bioimpedance analysis: What is the difference? Nephrol-
43. Gupta KB, Gupta R, Atreja A, et al. Tuberculosis and nutrition. Lung ogy. (Carlton) 2017. htts://doi.org/10.1111/nep.13042. [Epub ahead of
India. 2009; 26(1):9-16. print].
44. Koethe JR, von Reyn CF. Protein-calorie malnutrition, macronutrient 62. Hong S, Choi Y, Lee HJ, et al. Development and validation of
supplements, and tuberculosis. Int J Tuberc Lung Dis. 2016;20(7):857- a semi-quantitative food frequency questionnaire to assess diets of
863. Korean type 2 diabetic patients. Korean Diabetes J. 2010;34(1):
45. Zachariah R, Spielmann MP, Harries AD, et al. Moderate to severe 32-39.
malnutrition in patients with tuberculosis is a risk factor associated with 63. Roman-Vinas B, Ortiz-Andrellucchi A, Mendez M, et al. Is the
early death. Trans R Soc Trop Med Hyg. 2002; 96(3):291-294. food frequency questionnaire suitable to assess micronutrient intake
46. Khan A, Sterling TR, Reves R, et al. Lack of weight gain and relapse adequacy for infants, children and adolescents? Matern Child Nutr.
risk in a large tuberculosis treatment trial. Am J Respir Crit Care Med. 2010;6 Suppl 2:112-121.
2006;174(3):344-348. 64. Senekal M, Steyn NP, Nel J. A questionnaire for screening the
47. Oxlade O, Huang CC, Murray M. Estimating the Impact of Re- micronutrient intake of economically active South African adults.
ducing Under-Nutrition on the Tuberculosis Epidemic in the Central Public Health Nutr. 2009; 12(11):2159-2167.
Eastern States of India: A Dynamic Modeling Study. PLoS One. 65. Linn BS. Outcomes of older and younger malnourished and well-
2015;10(6):e0128187. nourished patients one year after hospitalization. Am J Clin Nutr.
48. Si ZL, Kang LL, Shen XB et al. Adjuvant Efficacy of Nutrition 1984;39(1):66-73.
Support During Pulmonary Tuberculosis Treating Course: Systematic 66. Mehta JB, Fields CL, Byrd RP Jr, et al. Nutritional status and
Review and Meta-analysis. Chin Med J (Engl). 2015;128(23):3219- mortality in respiratory failure caused by tuberculosis. Tenn Med.
3230. 1996;89(10):369-371.
49. Ramakrishnan CV, Rajendran K, Jacob PG, et al. The role of diet in 67. Volosevich GV. Blood T-lymphocyte functional activity in pulmonary
the treatment of pulmonary tuberculosis. An evaluation in a controlled tuberculosis patients undergoing chemotherapeutic agent and vitamin
chemotherapy study in home and sanatorium patients in South India. treatment [in Russian]. Probl Tuberk. 1982;(3):47-50.
Bull World Health Organ. 1961;25:339-359.
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 6
Associations Between Enteral Nutrition and Acute December 2018 865–871

C 2018 American Society for

Respiratory Infection Among Patients in New York Parenteral and Enteral Nutrition
DOI: 10.1002/ncp.10017
Metropolitan Region Pediatric Long-Term Care Facilities wileyonlinelibrary.com

Marissa Burgermaster, PhD1 ; Meghan Murray, MPH, RN2 ;


Lisa Saiman, MD, MPH ; David S. Seres, MD, ScM, PNS, FASPEN5
3,4
;
and Elaine L. Larson, RN, PhD, CIC, FAAN6,7

Abstract
Background: Pediatric long-term care facilities (pLTCF) serve a complicated and resource-intensive patient population with high
usage of nutrition support. However, the relationship between nutrition support and outcomes among pLTCF residents is not well
understood. We described this relationship in three metropolitan New York pLTCF and a subsample of infants from one of these
facilities with a feeding disorders unit. Methods: In this prospective cohort study, we used logistic regression to assess relationships
between enteral nutrition (EN), and acute respiratory infections (ARI) among residents (n = 720, 50% male, mean age = 5.5 years,
mean number comorbidities = 2.1) and infant subsample (<1 year, n = 208, 50% male, mean number comorbidities = 2.0). We
tested these associations in multivariable models controlling for numbers of comorbidities and infections. Results: Many residents
received nutrition via percutaneous (59%) or nasogastric (15%) feeding tubes. In univariate analyses, residents receiving EN had
more comorbidities. In multivariable analyses, EN was associated with ARI (incidence rate ratio = 1.65, p < .001). Among infants
in the specialized unit, greater risk of ARI was associated only with percutaneous (incidence rate ratio = 1.94, p < .01) feeding. EN
was associated with lower odds of being discharged home (OR = 0.45, p < .01). Conclusion: The prevalence of EN, complexity of
cases, and necessity of long-term EN make nutrition support important in pLTCFs. Differences in EN types and adverse outcomes
in the infant subsample suggest different care is necessary for this subpopulation. Results provide context for improving quality of
care and clinician/caregiver education for this population. (Nutr Clin Pract. 2018;33:865–871)

Keywords
enteral nutrition; long-term care; pediatrics; respiratory tract infections; skilled nursing facilities

Pediatric long-term care facilities (pLTCFs) primarily serve tions; and require frequent, ongoing healthcare use.2 They
children who need extensive assistance with activities of often have respiratory, nutrition, and/or neuromuscular
daily living.1 Residents of pLTCFs have substantial, spe- disorders.1 With ongoing advances in critical care for in-
cialized healthcare needs; chronic comorbid conditions fants and children, survival rates of premature infants and
associated with medical fragility; severe functional limita- children with birth defects and genetic disorders continue to

From the 1 Division of Preventive Medicine and Nutrition, Department of Medicine, Columbia University Medical Center, New York, New
York, USA; 2 School of Nursing, Columbia University Medical Center, New York, New York, USA; 3 Department of Pediatrics, Columbia
University Medical Center, New York, New York, USA; 4 Department of Infection Prevention & Control, NewYork-Presbyterian Hospital, New
York, New York, USA; 5 Division of Preventive Medicine and Nutrition, Department of Medicine and Institute of Human Nutrition, Columbia
University Medical Center, New York, New York, USA; 6 Associate Dean for Research, Anna Maxwell Professor of Nursing Research, School of
Nursing, Columbia University, New York, New York, USA; and 7 Department of Epidemiology, Mailman School of Public Health, Columbia
University, New York, New York, USA.
Financial disclosure: This work was supported by the Agency for Healthcare Research and Quality, U.S. Department of Health and Human
Services (Keep It Clean for Kids: The KICK Project, R01HS021470). M.B.’s involvement in the preparation of this manuscript was supported by
National Institutes of Health training grants T32HL007343 and T15LM007079.
Conflicts of interest: None.
This article originally appeared online on February 15, 2018.
Corresponding Author:
David S. Seres, MD, ScM, PNS, FASPEN, Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, P&S 9-501,
New York, NY 10032, USA.
Email: dseres@columbia.edu
866 Nutrition in Clinical Practice 33(6)

improve.3 Likewise, improved treatment of acute illnesses characteristics and outcomes for residents receiving EN,
has increased survival, as well as disability.4 Although we also separately examined a subsample of infants, many
estimates suggest that children with chronic comorbid con- of whom received nutrition support for different reasons
ditions represent <1% of the population and those who than the full sample. We aimed to: (1) describe the use of
reside in pLTCF make up a smaller fraction, this growing EN and clinical characteristics of children at pLTCFs who
and resource-intense healthcare sector has been found to are receiving EN; (2) describe the use of EN and clinical
account for disproportionate healthcare costs.2,5-8 characteristics among residents younger than 1 year in 1 of
Many children with chronic comorbid conditions re- the facilities with a specialized unit for infants with feeding
quire the support of medical technology to maximize their difficulties; and (3) identify relationships between feeding
functioning.2 A 2011 retrospective analysis of data from 4 type and outcomes including infections, hospitalization,
U.S. children’s hospitals indicated that 56% of children who and discharge home in both the whole sample and the infant
received complex care coordination services had gastros- subsample.
tomy tubes.9 Children with chronic comorbid conditions
who require technology assistance, including feeding tubes, Methods
incur 3.5 times the costs of other children with chronic
We conducted a prospective cohort study of residents at 3
comorbid conditions.5 Enteral nutrition (EN; ie, nasal or
pLTCFs from September 2012 to December 2015 as part
percutaneous tube feeding) is, therefore, an important com-
of an intervention study to reduce healthcare-associated
ponent of pLTCF care; however, the prevalence of EN and
infections, the Keep It Clean for Kids (KICK) Project
potential adverse impacts of EN in this unique population
(R01HS021470).15,17 The 3 pLTCFs were independent fa-
has not yet been well described in the literature.
cilities located in the New York City metropolitan area,
Some pLTCFs also admit children who need close ob-
ranging in size from 54 to 137 beds, with therapeutic, medi-
servation, medication administration, rehabilitation, and/or
cal, and school services on-site. This study was approved by
family education for shorter-term stays; many residents are
the BLINDED Institutional Review Board and the ethics
premature infants with acute nutrition needs.1 Some of
boards of the pLTCFs with approval for waiver of written
these infants cannot feed safely or efficiently because of
documentation of consent.
inability to suck, swallow, or coordinate breathing with
swallowing.10 Clinical indications for EN in both very
premature as well as medically complex infants11 and re-
Sample
ported relationships between extrauterine growth restriction Analyses included all residents during 2012–2015 except for
and neurodevelopmental impairment have driven recent those admitted for respite care only. Subanalyses comparing
interest in optimizing EN in this population,12 but there PFT, NGFT, and no EN in infants included residents
is wide variation and lack of consensus for nourishment younger than 1 year at the time of enrollment from a single
strategies.13 facility.
A recent retrospective cohort study found that of 15,642 Residents were enrolled from 1 of 3 pLTCFs in the
children having a gastrostomy tube placed, 8.6% had an New York metropolitan region, including 54-bed, 97-bed,
emergency department visit and 3.9% were readmitted and 137-bed facilities. Each of these pLTCFs is a stand-
within 30 days of discharge; infection, mechanical com- alone facility and provides care for only pediatric residents.
plication, and replacement were the primary gastrostomy- On average, 30% (range 22%–44%) of residents had tra-
related reasons for these visits.14 Children in pLTCFs cheostomies and 7% (range 5%–10%) were mechanically
are at particularly high risk for healthcare-associated in- ventilated. About 25% of residents are hospitalized at
fections due to their underlying conditions; mechanical least once each year. Overall, 70% (range 58%–86%) of
supports; and multiple contacts with medical providers, residents in these facilities have a neurological disorder and
family members, and other children with chronic comorbid 50% (range 36%–65%) have a respiratory disorder. The 3
conditions.15 We performed an epidemiological study of pLTCFs provide similar care, each with in-house physicians,
infections in residents in 3 pLTCFs in the New York City nursing staff, dietitians, and therapists, including physical,
metropolitan area and found use of EN ranged from 77% occupational, music, and art therapies.
to 85% of residents.16 Percutaneous feeding tubes (PFTs),
but not nasogastric feeding tubes (NGFTs), were an inde- Variables of Interest
pendent predictor of acute respiratory infection (ARI).16
In this exploratory study, we present a more detailed Dependent variables. We measured 3 dependent variables:
analysis focused on nutrition support to provide a bet- ARI, infection-related hospitalization, and discharge home.
ter understanding of the relationship between nutrition
support and adverse outcomes among residents in pLTCFs. Acute respiratory infection. Infections were defined based
In addition to a more detailed description of the clinical on clinical diagnosis by a physician or nurse practitioner.
Burgermaster et al 867

Study staff conducted ongoing medical record reviews to each patient was diagnosed with, including infections of the
collect data related to clinician diagnosis, signs and symp- respiratory tract, conjunctivitis/otitis, skin and soft tissue,
toms, and diagnostic testing related to infections. ARIs urinary tract, gastrointestinal, and bloodstream.
included both viral and nonviral acute upper respiratory
infections, as well as pneumonia, pharyngitis, and sinusitis. Analyses
Descriptive statistics were used to characterize the study
Infection-related hospitalization. Hospital admissions re-
sample of all residents and subsample of infants. We
lated to infection were defined as transfers to acute care for
assessed relationships between EN, clinical characteristics,
>24 hours with the indication of an infection diagnosed at
and infections in the entire study sample using binomial
the pLTCF. This information was collected from medical
logistic regression. We assessed relationships among NGFT,
record reviews conducted by study staff.
PFT, clinical characteristics, and infections among the
subsample of infants using multinomial logistic regression.
Discharge home. Discharge disposition was obtained from
For all analyses involving rates, Poisson regression was
the medical records by the study staff. Home was defined as
used. To better understand the independent influence of
the legal guardian’s residence. Discharge home was based
EN among residents and types of EN among infants, we
on a resident’s last discharge and his or her status at the
also tested these associations while controlling for clinically
study’s conclusion; that is, if a resident was discharged home
relevant covariates, including number of comorbidities and
and then readmitted, the resident was not considered to be
number of infections, in multivariable models. Statistical
discharged home.
significance was set a priori at .05. Analyses were conducted
with SAS version 9.3 (SAS Institute, Cary, NC).
Independent variable. In these analyses, nutrition support
was the independent variable. We defined EN to include any
tube feeding (ie, nasal or percutaneous tube feeding). Few
Results
residents enrolled during the study period had parenteral Of 720 pLTCF residents enrolled in the study, 59% had a
nutrition (n = 2); these residents were excluded from analy- PFT and 15% had a NGFT. Among residents older than
ses. Residents rarely received oral feeding in addition to EN; 1 year, only 25 (5%) received feeding via NGFT. Residents
therefore, it was not considered in analyses. For subanalyses, receiving any EN were older and had resided in the facilities
we compared EN via NGFT (including nasogastric and for longer; they also had more comorbidity types, more
nasojejunal feeding tubes) and PFT (including all types of gastrointestinal disorders, and, specifically, more feeding
gastrostomy, jejunostomy, or other feeding enterostomy). disorders. Residents receiving EN had more infections,
Eight residents were documented as having both PFT and and fewer residents receiving EN were discharged home.
NGFT at admission. For this study, they were included with Nutrition, comorbidity types, discharge disposition, and
residents having PFT. infections in residents who did and did not receive EN are
described in Table 1.
Covariates. We included patient characteristics and comor- Among the 208 infants in the subsample, frequency
bidities as covariates in our analyses. Patient demographics of feeding type varied for different disorders. Specifically,
and comorbidities were collected from medical records at infants with feeding disorders had higher odds of being fed
enrollment, including information about medical devices. via NGFT. Infants fed via PFT had more infections, and
Comorbidities were based on the admitting diagnoses for fewer infants with PFT were discharged home. Nutrition,
each resident and included the presence of ࣙ1 neurological comorbidity types, discharge disposition, and infections in
(eg, cerebral palsy, epilepsy), respiratory (eg, bronchopul- infants with NGFT, PFT, or no feeding tube are described
monary dysplasia, respiratory failure), cardiovascular (eg, in Table 2.
congenital heart disease, hypertension), gastrointestinal (eg, Associations between feeding type and outcomes of
short bowel disease, feeding disorder), metabolic (eg, mi- interest were measured in all residents and the subsample
tochondrial disease), or immune disorder (eg, HIV/AIDS). of infants, controlling for number of comorbidity types.
In previous research, our team found that the number of As gender and age were not significant in the bivariable
chronic comorbid conditions rather than the type of condi- models, they were not included in the multivariable models.
tion was predictive of ARI.16 Comorbidities were summed These results are described in Table 3. Residents with
by type for each resident (ie, a resident with a neurological EN had a higher rate of ARI compared with residents
disorder and a respiratory disorder has 2 comorbidities without EN when controlling for number of comorbidity
independent of how many individual neurological and types. Residents with EN were significantly less likely to
respiratory disorders the resident may have); this variable be discharged home as compared with residents without
was called number of comorbidity types throughout the EN when controlling for number of comorbidity types.
manuscript. Number of infections was a count of infections However, residents with EN were not significantly more
868 Nutrition in Clinical Practice 33(6)

Table 1. Characteristics of Residents With and Without Enteral Feeding Tubes at 3 Pediatric Long-Term Care Facilities,
2012–2015, and Unadjusted Comparisons.

Residents Residents Not


Receiving ENa Receiving EN
(n = 526, 73%) (n = 194, 27%) OR/IRRb P Value

Mean age at enrollment, y (range) 5.2 (0.02–22.3) 5.8 (0.02–26.2) 0.98 .23

Male sex, n (%) 254 (48) 107 (55) 0.76 .10

Mean no. of comorbidities (range) 2.3 (0–5) 1.6 (0–4) 2.01 <.001
Respiratory disorder 291 (55) 70 (36) 2.19 <.001
Neurological disorder 398 (76) 107 (55) 2.53 <.001
Gastrointestinal disorder 350 (67) 76 (39) 3.09 <.001
Feeding disorder 139 (26) 28 (14) 2.13 <.001
Dysfunctional suck/swallow 12 (2) 0 (0) — —
Short bowel disease 17 (3) 4 (2) 1.57 .42
Failure to thrive 57 (11) 8 (4) 2.80 .008

Mean length of enrollment, d (range) 606.2 (1–1217) 398.1 (4–1217) 1.001 <.001

Rate of infection-related hospital admissions 1.01 0.67 1.50c .007


per 1000 resident-days

Discharged home,d n (%) 154 (29) 99 (51) 0.40 <.001

Mean no. of infections (range) 3.42 (0–32) 1.33 (0–17) 1.21 <.001
ARI 298 (57) 55 (28) 3.30 <.001
Rate of ARI per 1000 resident-days 3.57 2.02 1.77c <.001
Pneumonia 109 (21) 13 (7) 3.64 <.001
Mean no. of pneumonia infections (range) 0.33 (0–8) 0.11 (0–4) 2.01 <.001

ARI, acute respiratory infection; EN, enteral nutrition; IRR, incidence rate ratio; OR, odds ratio.
a EN includes both nasal (including nasogastric and nasojejunal feeding tubes) and percutaneous feeding tubes (including all types of

gastrostomy, jejunostomy, or other feeding enterostomy).


b OR was determined by logistic regression. IRR was determined by Poisson regression.
c IRR reported for Poisson regression with continuous outcome.
d Based on last discharge only, does not include residents who were discharged home and subsequently returned to the same facility.

likely to be hospitalized for an infection, controlling for of comorbidities suggests that these residents were more
number of comorbidity types. complicated, which is consistent with their lower odds
Among the infants, only those with a PFT had an of being discharged home when controlling for number
increased rate of acute respiratory tract infections compared of comorbidity types. The prevalence of EN, complexity
with those without EN when adjusting for number of co- of cases, and necessity of long-term EN underscore the
morbidity types. Neither infants with an NGFT nor infants importance of nutrition support and clinical experts to
with a PFT had a statistically significantly increased rate of manage it properly for this population.
hospitalizations for infection when compared with infants Our finding that the pLTCF residents in this study who
without EN, nor were they significantly less likely to be dis- received EN had more ARIs and pneumonia is consistent
charged home, controlling for number of comorbidity types. with a study of respiratory infection among a sample of
children with profound intellectual and multiple disability
in day care centers, 42% with gastrostomy, which found that
Discussion feeding difficulty and gastrostomy both increased risk for
Seventy percent of pLTCF residents in our sample received ARI.18 The greater number of mean number of infections
EN, and the frequency of EN via PFT (59%) among our per patient among the patients receiving EN may be par-
sample is consistent with the frequency of gastrostomy tially explained by other types of infections, including skin
(56%) reported in a nationwide study of health resource infections at the insertion site. Superficial skin infections
use among children with chronic comorbid conditions.9 have been related to gastrostomy in pediatric acute care
That residents receiving EN had, on average, more types settings, with rates ranging from 7% to 25%.14,19,20 Future
Burgermaster et al 869

Table 2. Characteristics of Infants Younger Than 1 Year With Nasogastric, Percutaneous, and No Feeding Tubes at a Single
Pediatric Long-Term Care Facility With Specialized Unit for Infants, 2012–2015, and Unadjusted Comparisons.

Infants With Infants With Infants Without a


NGFTsa PFTsb Feeding Tube
(n = 65, 31%) (n = 76, 37%) (n = 67, 32%) P Valuec

Mean age at enrollment, y (range) 0.32 (0.05–0.90) 0.38 (0.02–0.99) 0.32 (0.02–0.92) .17

Male sex, n (%) 27 (42) 38 (50) 38 (56) .22

Mean no. of comorbidities (range) 1.95 (0–4) 2.14 (1–4) 1.82 (0–4) .12
Respiratory disorder 28 (43) 45 (59) 36 (54) .16
Neurological disorder 22 (34) 35 (46) 24 (36) .27
Gastrointestinal disorder 50 (77) 52 (68) 36 (54) .02
Feeding disorder 42 (65) 31 (41) 21 (31) <.001
Dysfunctional suck/swallow 2 (3) 1 (1) 0 (0) —
Short bowel disease 2 (3) 6 (8) 3 (4) .44
Failure to thrive 8 (12) 8 (11) 2 (3) .17

Mean length of enrollment, d (range) 78 (4–1190) 240 (1–1217) 168 (4–1217) .009

Rate of infection-related hospital admissions per 1.18 1.15 0.89d .77


1000 resident-days

Discharged homee 44 (68) 36 (47) 44 (66) .02

Mean no. of infections (range) 0.46 (0–4) 1.58 (0–9) 0.67 (0–6) .002
ARI 11 (17) 24 (32) 14 (21) .11
Rate of ARI per 1000 resident-days 2.56 4.17 2.05d .004
Pneumonia 2 (3) 7 (9) 2 (3) .19
Mean no. of pneumonia infections (range) 0.03 (0–1) 0.11 (0–2) 0.04 (0–2) .28

ARI, acute respiratory infection; NGFT, nasogastric feeding tube; PFT, percutaneous feeding tube.
a NGFTs (including NGFT and nasojejunal feeding tubes) and PFTs.
b PFTs (including all types of gastrostomy, jejunostomy, or other feeding enterostomy).
c Based on logistic regression for all variables except rate of infection-related hospital admissions and rate of ARI, which were based on Poisson

regression.
d Incidence rate ratio reported for Poisson regression with continuous outcome.
e Based on last discharge only, does not include residents who were discharged home and subsequently returned to the same facility.

research should examine the prevalence of EN-related skin experiencing pneumonia was very similar (2 cases in each
infections in pLTCF. condition) compared with 7 cases in infants with PFT, sug-
Although 68% of the infants in our subsample received gesting that NGFTs may not be a risk factor for pneumonia
any type of EN, PFT was less frequent than in our overall compared with PFT in this population. There is limited
sample (37% vs 59%). This may be because some infants are research comparing ARI among patients with NGFT and
admitted to a pLTCF for short-term intensive feeding. Our PFT in pediatric population. A retrospective cohort study
findings that many more infants with NGFT had feeding of children 0–14 years old at a single children’s hospital
disorder–related gastrointestinal comorbidities is consistent revealed similar rates of pneumonia among patients with
with our observation that infants with different feeding tube long-term (>3 months) use of PFT or NGFT, but that
types are admitted to pLTCFs for different reasons and, complications (excluding pneumonia and food refusal) were
therefore, need different care. almost twice as common among patients with PFT (80%)
This is the first study, to our knowledge, to report than NGFT (46%).21
respiratory infection rates in relation to EN among in- Limitations of this study include its observational na-
fants in a pLTCF. The finding that even when controlling ture: causality cannot be inferred from these analyses. Fur-
for number of comorbidities, the incidence rate of ARI, ther, the results may not be generalizable to pLTCF in other
including pneumonia, was significantly higher for infants regions. We only collected information about NGFT or PFT
with PFT contradicts common wisdom associating NGFT at enrollment into the study. Therefore, it is possible that
with an increased incidence of pneumonia relative to PFTs. some residents who received EN at enrollment had tubes
The number of infants with NGFT and no EN who were removed during the study period, and others who initially
870 Nutrition in Clinical Practice 33(6)

Table 3. Comparison of Acute Respiratory Infection, and feeding tube problems have been cited as reasons
Hospitalization, and Home Discharge Outcomes by Type of for cessation or holding of tube feeds.24-26 Due to the
Feeding in All Residents and Infants at a Single Pediatric complexity of nutrition support required by this population,
Long-Term Care Facility With Specialized Unit.
future research should also describe the role of registered
IRRa /ORb P dietitians and other nutrition experts in pLTCFs.
(95% CI) Value Our findings suggest that care providers in pLTCFs
should monitor residents who are receiving EN for respira-
All patients, comparison between ENc and no EN tory infections and take appropriate precaution to prevent
Rate of ARId 1.65a,e (1.39, 1.97) <.001e them. Additional testing is necessary to identify pathophys-
Discharged home 0.61b,e (0.42, 0.89) <.01e
iological pathways that explain the link between EN and
Rate of hospitalizationf 1.17a (0.68, 1.27) .65
Infants, comparison among NGFT, PFT,h and no EN
g respiratory infection, as well as specific risk factors and
Rate of ARI intervention targets; however, because EN and PFT are
NGFT vs no EN 1.19a (0.60, 2.36) .621 typically nonmodifiable risk factors, prevention is crucial.
PFT vs no EN 1.94a,e (1.21, 3.11) .006e Nutrition support care providers can play an important
Discharged home role in reducing infection by engaging in evidence-based
NGFT vs no EN 1.04b (0.50, 2.18) .90 practices and by educating key stakeholders. Residents
PFT vs no EN 0.57b (0.28, 1.14) .11 fed via EN, as well as staff and visitors, should receive
Rate of hospitalization
vaccinations for respiratory viruses. Additional preventive
NGFT vs no EN 1.47a (0.53, 4.09) .46
PFT vs no EN 0.75a (0.32, 1.77) .52 clinical practices (eg, attention to secretions, engagement
in movement and activity) and policies (eg, prohibiting ill
ARI, acute respiratory infection; EN, enteral nutrition; IRR, staff or visitors, enacting transmission reduction protocols)
incidence rate ratio; NGFT, nasogastric feeding tube; OR, odds ratio; should be followed when caring for residents with EN.
PFT, percutaneous feeding tube.
a IRR reported for Poisson regression with continuous outcome.
b OR reported for binomial logistic regression.
c EN includes both nasal (including NGFTs and nasojejunal feeding
Conclusions
tubes) and PFTs (including all types of gastrostomy, jejunostomy, or Our description of nutrition support in pLTCFs indicates
other feeding enterostomy). that most residents, including infants, receive EN, and that
d ARI, including pneumonia.
e Statistically significant result. Multivariable regression was EN is associated with ARI. Among infants, PFTs were
conducted while controlling for number of comorbidity types. For associated with greater risk for ARI. Furthermore, EN was
discharged home and rate of hospitalization outcomes, we also associated with lower odds of being discharged home in the
controlled for number of infections. whole sample. The results presented in this article provide
f Based on last discharge only, does not include residents who were

discharged home and subsequently returned to the same facility. a context for improving quality of care, as well as clinician
g NGFT (including NGFTs and nasojejunal feeding tubes). and caregiver education, for this challenging and growing
h PFT (including all types of gastrostomy, jejunostomy, or other
patient population.
feeding enterostomy).

Acknowledgments
did not receive EN had feeding tubes inserted during the The authors would like to acknowledge the administration and
study period. Further, this precluded any assessment of staff at the 3 contributing facilities, Rebecca Rudel for her
nutrition adequacy. We did not have information about contribution to the literature review, and Bevin Cohen for her
indications for EN or, more specifically, for the choice of consultation on statistical analyses.
PFT or NGFT for individual residents. Finally, because of
the small number of residents older than 1 year who were fed Statement of Authorship
via NGFT, we were not able to compare adverse outcomes M. Burgermaster, L. Saiman, D.S. Seres, and E.L. Larson
between NGFT and PFT in the full sample. contributed to the conception or design; M. Burgermaster, M.
Future research should examine nutrition adequacy and Murray, D.S. Seres, and E.L. Larson contributed to the ac-
mechanical EN complications in pLTCFs. Nutrition sup- quisition, analysis, or interpretation; M. Burgermaster and M.
Murray drafted the manuscript; M. Burgermaster, M. Murray,
port care is especially important for children with chronic
L. Saiman, D.S. Seres, and E.L. Larson critically revised the
comorbid conditions because frequent reassessment of
manuscript, gave final approval, and agree to be accountable
estimated energy requirement is necessary due to growth for all aspects of work ensuring integrity and accuracy.
and disease progression,22 and because many experience
feeding difficulties, malnutrition, and inadequate micronu- References
trient intake.23 In pediatric intensive care units, perceived 1. O’Malley M, Hutcheon RG. Genetic disorders and congenital mal-
intolerance, high gastric residual volumes, nausea and vom- formations in pediatric long-term care. J Am Med Dir Assoc.
iting, fasting around procedures, hemodynamic instability, 2007;8(5):332-334.
Burgermaster et al 871

2. Cohen E, Kuo DZ, Agrawal R, et al. Children with medical complexity: 15. Murray MT, Pavia M, Jackson O, et al. Health care–associated
an emerging population for clinical and research initiatives. Pediatrics. infection outbreaks in pediatric long-term care facilities. Am J Infect
2011;127(3):529-538. Control. 2015;43(7):756-758.
3. Stoll BJ, Hansen NI, Bell EF, et al. Trends in care practices, morbidity, 16. Saiman L, Maykowski P, Murray M, et al. Epidemiology of infections
and mortality of extremely preterm neonates, 1993–2012. JAMA. in pediatric long-term care facilities. JAMA Pediatr. 2017;171(9):872-
2015;314(10):1039-1051. 878. https://doi.org/10.1001/jamapediatrics.2017.1482
4. Hjern A, Lindblad F, Boman KK. Disability in adult survivors of 17. Larson EL, Cohen B, Murray M, Saiman L. Challenges in conducting
childhood cancer: a Swedish national cohort study. J Clin Oncol. research in pediatric long-term care facilities. Clin Pediatr. 53(11):1041-
2007;25(33):5262-5266. 1046.
5. Cohen E, Berry JG, Camacho X, Anderson G, Wodchis W, Guttmann 18. Proesmans M, Vreys M, Huenaerts E, et al. Respiratory morbidity
A. Patterns and costs of health care use of children with medical in children with profound intellectual and multiple disability. Pediatr
complexity. Pediatrics. 2012;130(6):e1463-e1470. Pulmonol. 2015;50(10):1033-1038.
6. Glendinning C, Kirk S, Guiffrida A, Lawton D. Technology-dependent 19. Jacob A, Delesalle D, Coopman S, et al. Safety of the one-step
children in the community: definitions, numbers and costs. Child Care percutaneous endoscopic gastrostomy button in children. J Pediatr.
Health Dev. 2001;27(4):321-334. 2015;166(6):1526-1528.
7. Simon TD, Berry J, Feudtner C, et al. Children with complex chronic 20. Viktorsdóttir MB, Óskarsson K, Gunnarsdóttir A, Sigurdsson L.
conditions in inpatient hospital settings in the United States. Pediatrics. Percutaneous endoscopic gastrostomy in children: a population-based
2010;126(4):647-655. study from iceland, 1999–2010. J Laparoendosc Adv Surg Tech.
8. Berry JG, Hall DE, Kuo DZ, et al. Hospital utilization and character- 2015;25(3):248-251.
istics of patients experiencing recurrent readmissions within children’s 21. Ricciuto A, Baird R, Sant’Anna A. A retrospective review of enteral
hospitals. JAMA. 2011;305(7):682-690. nutrition support practices at a tertiary pediatric hospital: a compari-
9. Berry JG, Agrawal R, Kuo DZ, et al. Characteristics of hospitalizations son of prolonged nasogastric and gastrostomy tube feeding. Clin Nutr.
for patients who use a structured clinical care program for children with 2015;34(4):652-658.
medical complexity. J Pediatr. 2011;159(2):284-290. 22. Mahan L, Escott-Stump S, Raymond J, Krause M. Krause’s Food &
10. Lau C. Development of infant oral feeding skills: what do we know? the Nutrition Care Process, 13th ed. St. Louis, MO: Elsevier/Saunders;
Am J Clin Nutr. 2016;103(2):616S-621S. 2012.
11. Khan Z, Marinschek S, Pahsini K, et al. Nutritional/growth status in 23. Wolff J, Wiltzer Karim J, Ronis S, Grossberg R. Effects of nutritional
a large cohort of medically fragile children receiving long-term enteral care coordination on children with medical complexity. Dev Med Child
nutrition support. J Pediatr Gastroenterol Nutr. 2016;62(1):157-160. Neurol. 2015;57:107.
12. Stevens TP, Shields E, Campbell D, et al. Variation in enteral feeding 24. Skillman HE, Mehta NM. Nutrition therapy in the critically ill child.
practices and growth outcomes among very premature infants: a Curr Opin Crit Care. 2012;18(2):192-198.
report from the New York State Perinatal Quality Collaborative. Am 25. Leong AY, Cartwright KR, Guerra GG, Joffe AR, Mazurak VC,
J Perinatol. 2016;33(1):9-19. Larsen BM. A Canadian survey of perceived barriers to initiation
13. Mills L, Modi N. Clinician enteral feeding preferences for very preterm and continuation of enteral feeding in PICUs. Pediatr Crit Care Med.
babies in the UK. Arch Dis Child Fetal Neonatal Ed. 2015;100(4):F372- 2014;15(2):e49-e55.
F373. 26. Dokken M, Rustøen T, Stubhaug A. Indirect calorimetry reveals
14. Goldin AB, Heiss KF, Hall M, et al. Emergency department visits that better monitoring of nutrition therapy in pediatric intensive
and readmissions among children after gastrostomy tube placement. care is needed. JPEN J Parenter Enteral Nutr. 2015;39(3):344-
J Pediatr. 2016;174:139-145.e132. 352.
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 6
Reducing Blood Glucose Testing Is Safe in Patients Receiving December 2018 872–878

C 2018 American Society for

Parenteral Nutrition Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10020
wileyonlinelibrary.com

Ashley Ratliff, MS, RD, LD, CNSC1 ; Amy Nishnick, RD, LD, CNSC1 ;
Robert DeChicco, MS, RD, LD, CNSC1 ; and Rocio Lopez, MS, MPH2

Abstract
Background: Hyperglycemia is a common adverse event associated with parenteral nutrition (PN); however, there is no consensus
on optimal glucose monitoring. Unnecessary point-of-care (POC) blood glucose (BG) testing can result in additional healthcare
cost and discomfort to the patient. This study’s aim was to determine whether decreasing the frequency of POC glucose testing in
patients receiving PN can reduce costs without increasing the frequency of glycemic events. Methods: This study examined adult,
noncritically ill patients who require PN and are managed by a nutrition support team. Guidelines were developed for the initiation
and discontinuation of POC BG monitoring. Pre-intervention and post-intervention data were collected for each patient, including
daily POC glucose and glucose from the basic metabolic panel (BMP). Equivalency testing was completed for each group. Results:
Data were collected on 171 subjects (86 preguideline, 85 postguideline). The total number of POC tests per patient was significantly
less in the postintervention period (median 23.0 vs 25.5; P = .011), as well as the average number of POC tests per patient per
day (2.9 ± 1.4 vs 3.6 ± 1.4; P < .001). The average BMP glucose (126 mg/dL preintervention vs 129 mg/dL postintervention; P
= .005) and the number of hypoglycemic and hyperglycemic events per 100 tests (both P < .001) were equivalent. Conclusions:
Implementation of new guidelines resulted in decreased frequency of POC BG tests by 20%, which may impact cost savings and
patient comfort in hospitalized patients receiving PN without increasing average BG level or glycemic events. (Nutr Clin Pract.
2018;33:872–878)

Keywords
blood glucose; cost savings; glucose monitoring; hyperglycemia; nutritional support; parenteral nutrition; quality improvement

Hyperglycemia is a common adverse event associated with into quartiles based on their BG levels. Lin et al9 found
parenteral nutrition (PN), occurring in up to 50% of hos- an increased risk for complications (eg, death, infection,
pitalized patients.1 Hyperglycemia-induced complications cardiac complications, or acute renal failure) between the
in patients who are receiving PN increase morbidity and lowest BG levels (<114 mg/dL) and the highest BG levels
mortality.2-5 The optimal blood glucose (BG) level in hos- (>180 mg/dL). All patients who were receiving PN had
pitalized patients who are receiving PN is not clear. Tight BG measurements twice weekly via serum glucose, whereas
BG control (ie, <110 mg/dL) in critically ill patients in a patients with hyperglycemia had BG monitoring every
surgical intensive care unit (ICU) demonstrated improved 6 hours via capillary finger sticks. Cheung et al10 reported
outcomes,6 but these results were not duplicated in patients hyperglycemia increased the risk for complications in pa-
in a medical ICU.7 One potential factor for the lack of tients who were receiving PN. Monitoring was described
improved outcomes with tight BG control in medical ICU
patients was increased episodes of hypoglycemia. For this
reason, the American Society for Parenteral and Enteral From the 1 Nutrition Support Team, Center for Human Nutrition
Nutrition (ASPEN) recommends a target BG range of 140– Cleveland Clinic, Cleveland, Ohio, USA; and 2 Quantitative Health
180 mg/dL in hospitalized patients who are receiving PN Sciences JJN3-01, Cleveland Clinic, Cleveland, Ohio, USA.
nutrition support.8 Financial disclosure: None.
Point-of-care (POC) capillary testing is the standard for Conflicts of interest: None.
monitoring BG because it is fast, easy, and relatively precise.
This article originally appeared online on March 12, 2018.
However, there is no consensus on the optimal frequency,
timing, and duration of POC testing, or when it is appro- Corresponding Author:
Ashley Ratliff, MS, RD, LD, CNSC, Center for Human Nutrition
priate to decrease or discontinue monitoring when it is no
TT-22, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195,
longer necessary. Lin et al9 described monitoring BG based USA.
on the presence of hyperglycemia. Patients were divided Email: ratlifa@ccf.org
Ratliff et al 873

as once-daily BG monitoring, with increased frequency to for <5 days. Eligible patients were identified from daily work
every 4 hours if the patient was hyperglycemic. Neither lists.
study commented on the effect of the frequency of BG Two study groups were defined: preintervention and
monitoring on outcomes. Sarkisian et al2 evaluated BG postintervention or preguideline and postguideline
using glucometer readings and central blood draws as part implementation. Data for each patient were collected
of standard PN laboratory results. Frequency of glucometer retrospectively from November 2014–May 2015 during the
readings ranged from 2 to 7 times in the first 48 hours and preintervention period and August–October 2015 during
3 to 18 times in the following 3–9 days on PN. There was the postintervention period. Data were collected from
no association with the frequency of BG monitoring and initiation of PN for a minimum of 5 days to a maximum
mortality. Pleva et al1 reported hyperglycemia in noncrit- of 10 days or until PN was discontinued for any reason (eg,
ically ill patients using daily laboratory tests for the first transitioned to oral or enteral nutrition, discharged, patient
3 days, which transitioned to every Monday, Wednesday, refused, withdrawal of care), PN was held for >24 hours
and Friday thereafter. POC glucose testing was done every for any reason (eg, IV access problems, surgery, orders not
6 hours. Once the BG was well controlled and the goal written), patient was transferred to an ICU, or the patient
rate of PN was reached, the POC testing was decreased or was discharged on home PN.
discontinued. ASPEN recommends POC BG monitoring The intervention consisted of guidelines for the initi-
every 6 hours, or more frequently based on the degree of ation and discontinuation of POC BG monitoring (Ta-
hyperglycemia, for patients starting on PN, then 3 times ble 1). POC BGs were obtained using glucometers that
daily until BG is controlled.11 During cyclic infusion of were calibrated monthly to ensure accuracy. A BG <70
PN, POC BG testing should be done 30 minutes to 1 hour
postinfusion to assess for rebound hypoglycemia.11
Table 1. Guidelines for Point-of-Care Blood Glucose
The current practice for monitoring patients initiated Monitoring.
on continuous PN at a large medical center followed by
a nutrition support team (NST) includes a daily basic 1. Starting POC testing
metabolic panel (BMP) and POC BG tests every 6 hours a. New central PN
at 6:00 AM, 12:00 PM, 6:00 PM, and 12:00 AM. Patients who i. Continuous infusion: order POC glucose
are receiving cyclic PN receive POC BG tests 3 times daily: 2 monitoring every 6 hours at 6:00 AM,
hours after starting the cycle, halfway through the cycle, and 12:00 PM, 6:00 PM, and 12:00 AM
1 hour postinfusion. POC BG monitoring can be decreased ii. Cyclic infusion: order POC glucose
monitoring 3 times daily 2 hours after
or discontinued at the discretion of the clinician once a beginning cycle, midcycle, and 1 hour after
patient is deemed stable at full calorie PN, with or without completing cycle
cycling, but this is not consistently practiced. As such, there b. New peripheral PN: POC blood glucose
may be opportunities to reduce BG testing in stable patients monitoring not required unless patient has
without affecting patient safety or quality of care. insulin-dependent diabetes and/or evidence of
The purpose of this study was to determine the effects of recent hyperglycemia (ie, blood glucose
implementing guidelines aimed at decreasing the frequency >200 mg/dL)
c. Existing home PN: POC blood glucose
of POC glucose testing in stable patients who are receiving monitoring is not required if patient’s home PN
PN on frequency of adverse glycemic events and healthcare formula does not contain insulin and there is no
costs. evidence of recent hyperglycemia (ie, blood
glucose >200 mg/dL)
2. Discontinuing POC testing
a. Discontinue POC blood glucose monitoring
Methods when blood glucose levels <200 mg/dL for >24
hours and PN at goal calories (continuous
Study Design infusion) or goal calories and cycle hours (cyclic
infusion)
This quality improvement project examined hospitalized b. Discontinue POC blood glucose test if ordered
adult, noncritically ill patients admitted to a quaternary care by another service if patient does not meet
1200-bed urban hospital, who received PN, defined as an criteria for POC blood glucose monitoring
intravenous (IV) formula containing protein and dextrose, 3. Restarting POC testing
initiated and managed by the NST inpatient service. The a. Start/restart POC blood glucose monitoring
every 6 hours for any patient with BMP blood
institutional review board determined the study protocol glucose <70 or >200 mg/dL
to be exempt. Patients were excluded from the study if
they were followed by the endocrinology service for BG BMP, basic metabolic panel; PN, parenteral nutrition; POC, point of
management, had PN initiated in an ICU, or received PN care.
874 Nutrition in Clinical Practice 33(6)

mg/dL was defined as hypoglycemia and >200 mg/dL as calories were generally 25–35 kcal/kg/d but were adjusted
hyperglycemia. The guidelines were based on a review for underweight or obese patients.
of published standards along with expert opinion. The
guidelines were implemented after the preintervention data Statistical Analysis
were collected, and a training program targeting NST
Pilot data were collected to determine sample size. A total
registered dietitians, staff physicians, and nurse practition-
of 86 subjects per period was needed to provide 95% power.
ers/physician assistants was completed. Postintervention
Univariate analysis was performed to compare patient char-
data were not collected until 2 weeks after the new guidelines
acteristics between the 2 time periods. Analysis of variance
were implemented to allow the staff to become accustomed
or the nonparametric Kruskal-Wallis test was used for
to the new practice. Nurse managers were informed about
continuous or ordinal variables. The Pearson’s χ 2 tests or
the new guidelines and educated their staff 1 time at their
Fisher’s exact tests were used for categorical factors.
weekly meeting. Continuing education was provided by
In addition, an equivalence test was used to assess
NST clinicians to bedside nursing when requested. Per
whether the average BMP glucose and the observed
guidelines, NST clinicians had the discretion to discontinue,
proportion of subjects with adverse glycemic events during
reduce, start, restart, or otherwise alter BG monitoring
preintervention and postintervention were equivalent.
based on clinical judgment. The NST clinicians received
Equivalency testing was used to determine whether the
weekly continued education of the new guidelines for
preintervention and postintervention groups were similar.
1 month.
Equivalency test uses a null hypothesis that assumes the
differences between the 2 groups are greater than the
Data Collection noted amount, also known as the “interval of tolerable
differences.” This threshold represents a difference that
Data were collected retrospectively from the electronic med-
has clinically insignificant outcomes. For this study, this
ical record. Daily BMPs and POC BG tests were obtained
difference was noted as postintervention was equivalent
during the specified time range. Preintervention and postin-
to preintervention if there was a difference no >15 mg/dL
tervention data were collected and analyzed separately.
for average glucose level, as determined by the average
Morning BMP glucose levels were used as standard for
variability in BG samples, 5% for low glucose (<70) or 10%
benchmarking BG levels for each patient. Morning BMP
for high glucose (>200). In equivalency testing, P < .05
glucose levels were used to monitor glucose levels, given
means the 2 groups are equivalent, not different.
each patient who was receiving PN had these drawn daily
Also, the adverse glycemic event rate per 100 BMP tests
even when POC glucose tests were discontinued. Other
for each time period was calculated by dividing the total of
data collected included PN indication (eg, ileus/obstruction,
observed events by the total number of tests performed; 2
leak/high-output enterocutaneous fistula, malabsorption,
1-sided tests were used to assess equivalence between the
other), PN route (ie, central, peripheral) at end of data
groups.
collection for each patient, PN infusion schedule (ie, con-
All analyses were performed using SAS version 9.4
tinuous, cyclic) at end of study period, demographics (eg,
(SAS Institute, Cary, NC), and P < .05 was considered
age, gender), diet order (eg, NPO, clear liquid/full liquid,
statistically significant. Data are presented as median or
other) at end of study period, enteral tube feeding (eg, yes,
mean ± standard deviation.
no) at end of study period, diseases/conditions affecting BG
control such as history of type 1 or 2 diabetes mellitus re-
quiring medication before admission, actively being treated
Results
for infection (ie, current medications including at least 1 an- Data were collected on 171 subjects (86 preintervention,
tibiotic), medications potentially contributing to BG abnor- 85 postintervention). Average age was 56 ± 17 years, and
malities (ie, steroids, thiazide diuretics, fluoroquinolones, 53% were male. Table 2 presents a summary of patient
β-blockers, hormones, anticonvulsants, antipsychotics, and demographics and clinical characteristics for each time
miscellaneous medications such as octreotide, isoniazid, and period. There were no significant differences in PN route,
clonidine), and nutrition status (eg, no malnutrition, mild PN infusion schedule, history of diabetes, antibiotic ad-
malnutrition, moderate malnutrition, severe protein-calorie ministration, or administration of medications that could
malnutrition). affect BG between the preintervention and postintervention
PN was individualized for each patient based on clinician groups.
judgments using ASPEN guidelines. Patients were typically Table 3 demonstrates the total number of POC tests
started on 150–200 g dextrose on day 1 of PN adminis- per patient was significantly fewer in the postintervention
tration and advanced to goal over the next 3 days. Protein group (median 23.0 vs 25.5; P = .011). The average number
was generally dosed at 1.5–2.0 g/kg/d and fats at 250 mL of POC tests per patient per day was significantly fewer
of 20% lipid injectable emulsion 3 times per week. Goal in the postintervention than the preintervention group
Ratliff et al 875

Table 2. Demographic and Clinical Characteristics of Patients Who Are Receiving Parenteral Nutrition.

Preintervention (n = 86) Postintervention (n = 85)

Factors n (%) n(%) P Value

Age, y 54.8 ± 16.3 56.5 ± 16.9 .52a


Gender, n (%) .49b
Male 43 (50.0) 47 (55.3)
Female 43 (50.0) 38 (44.7)
Nutrition status, n (%) .77b
None 23 (26.7) 20 (23.5)
Mild 10 (11.6) 13 (15.3)
Moderate 12 (14.0) 15 (17.6)
Severe 41 (47.7) 37 (43.5)
PN indication, n (%) .026b,c
Ileus/obstruction 35 (40.7) 41 (48.2)
Leak/ECF 13 (15.1) 23 (27.1)
Malabsorption 16 (18.6) 12 (14.1)
Other 22 (25.6) 9 (10.6)
PN route, n (%) .95b
Central 81 (94.2) 78 (94.0)
Peripheral 5 (5.8) 5 (6.0)
PN infusion schedule, n (%) .79b
Continuous 34 (39.5) 32 (41.6)
Cyclic 52 (60.5) 45 (58.4)
Diet, n (%) .24b
NPO/Clear liquid 38 (44.2) 26 (35.1)
Full liquid/other 48 (55.8) 48 (64.9)
History of diabetes mellitus, n (%) 16 (18.6) 17 (20.0) .82b
Antibiotics when started, n (%) 38 (44.2) 31 (36.5) .30b
Glucose-increasing medications, n (%) 25 (29.1) 26 (30.6) .83b
Median LOS (Q1, Q3), d 8.0 (5.0, 9.0) 9.0 (6.0, 9.0) .80d

ECF, enterocutaneous fistula; LOS, length of stay; PN, parenteral nutrition; Q1, 25th percentile; Q3,75th percentile.
a Analysis of variance.
b Pearson’s χ 2 test.
c Represents those P values <.05 and signifies the 2 groups are significantly different.
d Kruskal-Wallis test.

(2.9 ± 1.4 vs 3.6 ± 1.4; P < .001). The number of POC tests or forgo testing entirely in patients who are at low risk for
per patient per day was similar between the 2 periods until development of hyperglycemia.
day 3. Afterward, the number of POC tests was lower in The patients were similar in both the preintervention
the postintervention period, and this difference continued and postintervention groups in regard to all demographics
until the end of the study. There was no evidence of a except classification of PN indication, which was signifi-
significant difference in average POC glucose levels between cantly different (P = .026). This could have been caused by
the 2 periods (135 ± 35.2 vs 135 ± 30.9; P = .94) (Table 3). differences in clinical judgment due to separate investigators
Table 4 presents the results of the equivalence tests. collecting data for preintervention and postintervention
The average BMP glucose was equivalent between the 2 periods.
time periods (ie, 126 mg/dL preintervention vs 129 mg/dL POC BG monitoring is the cornerstone of glycemic
postintervention; P = .005). Similarly, the number of hy- management in hospitalized patients. This is especially true
poglycemic and hyperglycemic events per 100 tests was for patients who are receiving PN because hyperglycemia is
equivalent (P < .001). a common metabolic complication. There are guidelines for
the initiation of POC BG monitoring in patients who are
receiving PN,11 but discontinuing testing is not addressed.
Discussion Reducing the frequency of diagnostic tests may improve
This project provides evidence that at a large medical center, the value of healthcare providing the quality of patient
with a dedicated NST managing PN daily, it is safe to care can be maintained. In particular, laboratory tests
discontinue POC BG monitoring once patients are stable, are scrutinized because they are routinely performed in
876 Nutrition in Clinical Practice 33(6)

Table 3. Frequency and Average Blood Glucose Testing.

Preintervention Postintervention
Factors (n = 86) (n = 85) P Value

Total no. of BMP tests, median (min, max) 86 9 (3, 12) 85 9 (5, 11) .84a
Total no. of POC tests, median (min, max) 86 25 (0, 59) 85 23 (0, 47) .011a,b
Average no. of POC tests/day, mean ± SD 86 3.6 ± 1.4 85 2.9 ± 1.4 <.001b,c
Overall average POC, mean ± SD 83 135 ± 35 81 136 ± 31 .94c
Overall minimum POC, mean ± SD 83 84 ± 18 81 95 ± 23 <.001b,c
Overall maximum POC, mean ± SD 83 203 ± 69 81 194 ± 67 .38c

BMP, basic metabolic panel; max, maximum; min, minimum; POC, point of care.
a Kruskal-Wallis test.
b Represents those P values <.05 and signifies the 2 groups are significantly different.
c Analysis of variance.

Table 4. Effect of Point-of-Care Glucose Monitoring on Glycemic Events: Equivalence Testing.

Preintervention Postintervention Equivalence


Factors (n = 86) (n = 85) Test P Value

Average BMP glucose (mg/dL), mean ± SD 86 126.4 ± 30.9 85 128.8 ± 32.6 .005
Hypoglycemia on BMP
Total no. of events, median (min, max) 86 0 (0, 1) 85 0 (0, 3) —
Any event, n (%) 86 3 (3.5) 85 4 (4.7) .1
No. of events/100 tests, rate (95% CI) 86 0.43 (0.14, 1.3) 85 0.99 (0.47, 2.1) <.001a
Hyperglycemia on BMP
Total no. of events, median (min, max) 86 0 (0, 6) 85 0 (0, 6) —
Any event, n (%) 86 18 (20.9) 85 21 (24.7) .16
No. of events/100 tests, rate (95% CI) 86 7.0 (5.3, 9.2) 85 7.5 (5.8, 9.9) <.001a

Two 1-sided tests were used to assess equivalence. Margin of equivalence was defined as 5% for hypoglycemia and 10% for hyperglycemia.
BMP, basic metabolic panel.
a Represents those P values <.05 and signifies the 2 groups are equivalent.

hospitalized patients, many times without a clear indication. cause readings at those levels usually require intervention,
Efforts to reduce the number of unnecessary laboratory tests typically oral or IV carbohydrate for hypoglycemia, and
have involved a variety of approaches with varying degrees subcutaneous sliding scale insulin for hyperglycemia, even in
of success ranging from a 98% decrease to a 28% increase in the absence of symptoms. Although serum BG levels are not
testing.12 a clinical outcome, they were used as a surrogate measure
Reducing laboratory tests is meaningful only if clinical of blood sugar management because they are considered
outcomes are maintained or improved. If there is an increase more precise than capillary BG levels and were available
in adverse events attributable to a reduction in testing, daily throughout the study period.
any potential cost savings may be negated. Unfortunately, The average number of POC BG tests was reduced ap-
the effect of reducing the frequency of laboratory test- proximately 20% in the current project. Potential benefits of
ing on clinical outcomes, such as rate of complications reducing the frequency of POC BG tests include decreases
or hospital length of stay, is difficult to determine due in costs and patient discomfort. This can benefit the patient,
to the number of confounding factors. Most studies in the healthcare team, and the institution. Reducing POC BG
this area assumed clinical equivalence rather than tracking tests can decrease disruption of sleep cycles and patient
outcomes.13,14 discomfort from the finger sticks. With the time savings re-
Results of this quality improvement project demonstrate alized from performing fewer POC tests, healthcare workers
the frequency of POC BG monitoring can be reduced can engage in other activities to benefit patients, and the cost
without affecting the average BG level or the number of savings from decreased testing can be passed along to the
episodes of hypoglycemia or hyperglycemia in hospitalized patient and institution.
patients who are receiving PN. Adverse glycemic events Although a single POC test is relatively inexpensive, it
were defined as blood sugars <70 or >200 mg/dL be- is typically ordered every 6 hours for patients who are
Ratliff et al 877

receiving PN compared with once daily for most laboratory This simplified training of the clinicians increased the
tests. Hospital charges are proprietary, making it difficult to likelihood that guidelines were followed.
calculate actual savings due to decreased POC testing. For Another strength of this project was the availability of
this project, we estimated the cost of a single POC BG test daily serum BG measurements. Serum BG was considered
at $6.00. Based on the patient volume of the NST inpatient the gold standard to determine overall blood sugar man-
service at our institution, a 20% reduction in POC BG tests agement for this project compared with POC BG tests.
would equal 7700 fewer tests per year and an annual savings Although daily serum BG measurements were probably un-
of $46,000. necessary, at least in a portion of the patients, it gave us the
The savings in labor due to nursing assistants performing opportunity to serially monitor blood sugar management in
fewer tests is less clear. In theory, if nursing assistants all patients without gaps in the data.
perform fewer unnecessary POC tests, they would have more
time to perform other, more vital patient care activities. This Conclusions
would likely result in improved patient care, but overall labor
This project demonstrated that the implementation of new
costs would be unchanged.
guidelines reduced the frequency of POC BG tests by 20%
A limitation of the project was the variation among clin-
in hospitalized patients who were receiving PN without
icians in application of the new POC BG testing guidelines.
increasing average BG level or adverse glycemic events. This
Because guidelines are simply suggestions for practice rather
was achieved by discontinuing monitoring in patients once
than policies, they allowed clinicians to vary treatment
they were stable at goal calories and infusion schedule,
based on their clinical judgment, and although the NST is
and forgoing testing in patients who were at low risk for
very experienced overall, this was a new practice for most
development of hyperglycemia. Although these interven-
of the clinicians, so not everyone had the same level of
tions were successful on a small scale with a limited group
comfort reducing the number of POC tests. It is expected,
of clinicians for a limited time in patients receiving PN,
with time, that clinicians will become more accustomed
it is not clear whether they would be as successful on a
to the new guidelines and follow them more uniformly.
larger scale over a larger period with patients who are not
This lack of comfort with reducing POC tests extended to
receiving PN. In addition, the subjects were on regular
the patients’ primary service. Because the primary service
nursing floors, so it is not clear whether the same guidelines
is primarily responsible for submitting orders for their
can be used in ICUs. Regardless, reducing unnecessary
patients, they reordered POC BG monitoring multiple times
POC BG tests should be a goal for all clinicians who
in study patients in good faith, thinking it was inadvertently
are managing patients receiving PN and warrants fur-
discontinued.
ther investigation because it can potentially reduce health-
Another limitation of the project was the lack of a
care costs and patient discomfort, and improve healthcare
measure of patient satisfaction specifically related to the re-
value.
duction in POC BG tests. We assumed reducing POC testing
would improve satisfaction because of fewer interruptions
Statement of Authorship
in the sleep cycle, particularly at midnight and 6:00 AM,
when testing is typically done, and less discomfort because A. Ratliff, B. DeChicco, A. Nishnick contributed to concep-
of the fewer finger sticks. Unfortunately, POC testing is tion/design of the research, acquisition, R. Lopez contributed
to the analysis or the data; All authors contributed to the
only one of many factors that comprise patient satisfaction
interpretation of the data, drafted the manuscript, A. Ratliff,
scores, and it was beyond the scope of this project to tease
B. DeChicco, A. Nishnick critically revised the manuscript;
out these data. and All authors agree to be fully accountable for ensuring
Lastly, a potential weakness of this project is the number the integrity and accuracy of the work. All authors read and
of patients. The goal was to enroll 86 patients in each group approved the final manuscript.
based on the expectation there would be a 25% decrease
in the frequency of POC testing. The actual decrease
References
was closer to 20%, and 1 patient in the postintervention
1. Pleva M, Mirtallo JM, Steinberg SM. Hyperglycemic events in non-
group was an outlier because she had repeated episodes of
intensive care unit patients receiving parenteral nutrition. Nutr Clin
hypoglycemia and hyperglycemia. In spite of this, the data Pract. 2009;24:626-634.
demonstrated equivalence between the groups in terms of 2. Sarkisian S, Fenton TR, Shaheen AA, Raman M. Parenteral nutrition-
the number of adverse events. associated hyperglycemia in noncritically ill inpatients is associated
A strength of this project is that it was conducted with higher mortality. Can J Gastroenterol. 2010;24:453-457.
3. Kumar PR, Crotty P, Raman M. Hyperglycemia in hospitalized
at a single institution by members of a single team of
patients receiving parenteral nutrition is associated with
experienced clinicians who are primarily responsible for increased morbidity and mortality: a review. Gastroenterol Res
managing blood sugars in patients who are receiving PN. Pract. 2011;2011:760720.
878 Nutrition in Clinical Practice 33(6)

4. Pasquel FJ, Spiegelman R, McCauley M, et al. Hyperglycemia during 10. Cheung NW, Zaccaria C, Napier B, Fletcher JP. Hyperglycemia is
total parenteral nutrition. Diabetes Care. 2010;33:739-741. associated with adverse outcomes in patients receiving total parenteral
5. Oliveira G, Tapia MJ, Ocon J, et al. Parenteral nutrition-associated nutrition. Diabetes Care. 2005;28:2367-2371.
hyperglycemia in non-critically ill patients increases the risk of in- 11. Sacks GS, Mayhew S, Peterson C. Parenteral nutrition implementation
hospital mortality (multicenter study). Diabetes Care. 2013;36:1061- and management. In: Merrit R, ed. ASPEN Nutrition Support Practice
1066. Manual. 2nd ed. Silver Spring, MD: American Society for Parenteral
6. Van den Berghe G, Wouters P, Weekers F, et al. Intensive insulin and Enteral Nutrition; 2009:114-115.
therapy in critically ill patients. N Engl J Med. 2001;345:1359-1367. 12. Kobewka D, Ronksley P, McKay J, Forster A, van Walraven
7. Van den Berghe G, Wilmer A, Hermans G, et al. Intensive insulin C. Influence of educational, audit and feedback, system based,
therapy in the medical ICU. N Engl J Med. 2006;354:449-461. and incentive and penalty interventions to reduce laboratory test
8. McMahon MM, Nystrom R, Braunschweig C, Miles J, Compher utilization: a systematic review. Clin Chem Lab Med. 2015;53:
C; American Society for Parenteral and Enteral Nutrition Board of 157-183.
Directors. A.S.P.E.N. clinical guidelines: nutrition support of adult 13. Feldman LS, Shihab HM, Thiemann D, et al. Impact of providing
patients with hyperglycemia. JPEN J Parenter Enteral Nutr. 2013;37: fee data on laboratory test ordering. JAMA Intern Med. 2013;173:
23-36. 903-908.
9. Lin LY, Lin HC, Lee PC, Ma WY, Lin HD. Hyperglycemia correlates 14. Thakkar R, Kim D, Knight AM, et al. Impact of an educational
with outcomes in patients receiving total parenteral nutrition. Am J intervention on the frequency of daily blood test orders for hospitalized
Med Sci. 2007;333:261-265. patients. Am J Clin Pathol. 2015;143:393-397.
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 6
Risk of Malnutrition Evaluated by Mini Nutritional December 2018 879–886

C 2018 American Society for

Assessment and Sarcopenia in Noninstitutionalized Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10022
Elderly People wileyonlinelibrary.com

Ilaria Liguori, MD1 ; Francesco Curcio, MD1 ; Gennaro Russo, MD1 ;


Michele Cellurale, MD1 ; Luisa Aran, MD1 ; Giulia Bulli, MD1 ;
David Della-Morte, MD, PhD2,3 ; Gaetano Gargiulo, MD4 ; Gianluca Testa, MD,
PhD1,5 ; Francesco Cacciatore, MD, PhD1,6 ; Domenico Bonaduce, MD1 ;
and Pasquale Abete, MD, PhD1

Abstract
Background: Malnutrition indices and muscle mass and strength in the elderly are poorly investigated. Moreover, malnutrition seems
to be 1 of the more important factors in the cause of sarcopenia. The presence of sarcopenia and its relationship with malnutrition
indices were studied in noninstitutionalized elderly people who underwent Comprehensive Geriatric Assessment (CGA). Methods:
A total of 473 elderly subjects (mean age, 80.9 ± 6.6 years) admitted to CGA were studied. Malnutrition risk was evaluated with
Mini Nutritional Assessment (MNA) score, whereas muscle mass and muscle strength were evaluated by bioimpedentiometry and
hand grip, respectively. Sarcopenia was assessed as indicated in the European Working Group on Sarcopenia in Older People
(EWGSOP) consensus. Results: Overall prevalence of sarcopenia was 13.1%, and it increased from 6.1% to 31.4% as MNA
decreased (P < .001). MNA score was lower in elderly subjects with sarcopenia (15.4 ± 4.2) than without sarcopenia (22.0 ±
4.0) (P = .024). Linear regression analysis showed that MNA score is linearly related both with muscle mass (r = 0.72; P < .001)
and strength (r = 0.42; P < .001). Multivariate analysis, adjusted for several confounding variables including comorbidity and
disability, confirmed these results. Conclusions: MNA score is low in noninstitutionalized elderly subjects with sarcopenia, and it
is linearly related to muscle mass and muscle strength. These data indicate that MNA score, when evaluated with muscle mass and
strength, may recognize elderly subjects with sarcopenia. (Nutr Clin Pract. 2018;33:879–886)

Keywords
aged; elderly; malnutrition; muscle strength; nutrition assessment; nutritional status; sarcopenia

Sarcopenia has been defined as a phenomenon character- elderly patients, and 22%–28% in this setting are at nutrition
ized by an age-related loss of muscle mass and strength risk.12 However, data on the prevalence of malnutrition in
particularly evident in older people aged >75 years.1-3
Sarcopenia is related to an increased risk for morbidity
and mortality in elderly people,4-6 especially in institution- From the 1 Department of Translational Medical Sciences, University
alized elderly nursing home residents.7 This pathological of Naples “Federico II”, Naples, Italy; 2 Department of Systems
condition is defined as a geriatric syndrome characterized Medicine, University of Rome Tor Vergata, Rome, Italy; 3 IRCCS San
by a loss of independence, a higher risk for falls, a reduc- Raffaele Pisana, Rome, Italy; 4 Division of Internal Medicine, AOU
San Giovanni di Dio e Ruggi di Aragona, Salerno, Italy; 5 Department
tion of the quality of life, and an increase in healthcare of Medicine and Health Sciences, University of Molise, Campobasso,
cost.1-3 From the pathophysiological point of view, several Italy; and 6 Azienda Ospedaliera dei Colli, Monaldi Hospital, Heart
factors may be involved in the pathogenesis of sarcope- Transplantation Unit, Naples, Italy.
nia including hormonal, metabolic, inflammatory state, Financial disclosure: None declared.
physical inactivity, and more importantly, malnutrition
Conflicts of interest: None declared.
state.8
[This article was modified on 2018-06-05 after initial online
Muscle protein loss at advancing age depends on a publication to correct reference 17.]
discrepancy between muscle protein synthesis and muscle
This article originally appeared online on February 13, 2018.
protein breakdown; in fact, muscle protein synthesis rates
are reduced in the older population with age-related decline Corresponding Author:
Pasquale Abete, MD, PhD, Associate Professor of Geriatrics,
at a rate of 3.5% per decade from age 20–90 years.9-11 It
Dipartimento di Scienze Mediche Traslazionali, Università di Napoli
is well known that protein-energy-malnutrition prevalence Federico II, Via S. Pansini, 80131 Naples, Italy.
in the acute care setting ranges between 23% and 60% of Email: p.abete@unina.it
880 Nutrition in Clinical Practice 33(6)

community-dwelling older adults are poor, although some Frailty status was assessed by the Italian Frailty Index
reports indicate a range of 5%–30%.12 derived by Rockwood’s frailty methods, recently validated
Moreover, malnutrition and sarcopenia often coexist in in an Italian cohort of elderly subjects.24 Fried’s frailty
a clinical syndrome characterized by a combination of methods were also assessed according to the Cardiovascular
decreased nutrient intake and decreased body weight, along Health Study Collaborative Research Group.25
with a decrease in muscle mass, strength, and/or physical
function (ie, malnutrition-sarcopenia syndrome [MSS]).13
Malnutrition and sarcopenia, taken together, are associated
Assessment of Nutrition Status
with negative health outcomes. Thus, it is extremely impor- Nutrition status was assessed by MNA. The MNA test
tant to assess malnutrition status, and more importantly, is composed of simple measurements and brief questions
nutrition assessment should be integrated with sarcopenia that can be completed in <10 minutes: (1) anthropometric
screening.13 measurements (weight, height, and weight loss); (2) global
Several malnutrition tools have been used with different assessment (6 questions related to lifestyle, medication, and
results.14 Mini Nutritional Assessment (MNA) represents mobility); (3) dietary questionnaire (8 questions, related to
1 of the more specific tool to assess malnutrition state in number of meals, food and fluid intake, and autonomy of
geriatric settings.15 However, this approach, together with feeding); and (4) subjective assessment (self-perception of
sarcopenia screening, is not well investigated in outpatient health and nutrition). The sum of the MNA score distin-
older adults. guishes between elderly patients: (1) with adequate nutrition
Thus, in this study, we aimed to investigate the rela- status (MNA ࣙ 24), (2) with protein-calorie malnutrition
tionship between muscle mass and strength and malnutri- (MNA >17), and (3) at risk for malnutrition (MNA between
tion evaluated by MNA in outpatients older adults who 17 and 23.5). With this scoring, sensitivity was found to be
underwent Comprehensive Geriatric Assessment (CGA), 96%, specificity 98%, and predictive value 97%.26
including several clinical and biochemical parameters.
Assessment of Muscle Mass and Strength
Methods
Muscle mass was measured by bioelectrical impedance
Study Population analysis (BIA) using a Quantum/S Bioelectrical Body Com-
The study enrolled 473 consecutive elderly outpatients (ࣙ65 position Analyzer (Akern Srl, Florence, Italy). Whole-body
years) admitted to a CGA center. The study received BIA measurements were taken between the right wrist and
full ethical approval from research ethics committees in ankle with the subject in a supine position. Muscle mass
accordance with the ethical standards laid down in the was calculated using the following BIA equation of Janssen
1964 Declaration of Helsinki and its later amendments. All and colleagues.27 Skeletal muscle mass (kg) = ([height2 /BIA
participants signed an informed consent form, and the insti- resistance × 0.401] + [gender × 3.825] + [age × −0.71])
tutional review boards approved the study. Anthropometric + 5.102, where height is measured in centimeters; BIA
measurements including age, sex, body mass index (BMI), resistance is measured in ohms; for gender, men = 1 and
and waist circumference (WC) were performed.16,17 women = 0; and age is measured in years. Absolute skeletal
muscle mass (kg) was converted to skeletal muscle index
standardizing by meters squared (kg/m2 ). Muscle strength
Comprehensive Geriatric Assessment was assessed by grip strength, measured using a hand-grip
Elderly participants underwent a comprehensive geri- dynamometer (Mecmesin Advanced Force Gauge 500N;
atric multidimensional evaluation that included cognitive GDM, Corsico (MI), Italy). The participant squeezed the
function evaluation with Mini-Mental State Examination dynamometer with maximum effort for at least 5 seconds.
(MMSE)18 ; depressive symptoms with Geriatric Depression Hand-grip strength was tested 3 times with a minimum of
Scale (GDS)19 ; comorbidity and comorbidity severity with 60 seconds rest provided for recovery between each attempt.
a Cumulative Illness Rating Scale (CIRS-Comorbidity and Maximum grip strength was recorded as the highest value of
CIRS-Severity)20 and drug number; disability with basic 3 trials. The maximum value was used rather than the mean
and instrumental activities of daily living (BADLs and of 3 trials allowing for a measure that was independent
IADLs, respectively); physical performance with 4-m gait of fatigue, which may have been possible in some of the
speed (cutoff < 0.8 m/s)21 ; physical activity with Physical participants. BMI-adjusted values were used as a cutoff
Activity Scale for the Elderly (PASE)22 ; social support eval- point to classify low muscle strength (BMI ࣘ24, 24.1–28,
uation with Social Support Assessment (SSA) scored from <28 was 29, ࣘ30, and ࣘ32 kg for men; BMI ࣘ23, 23.1–
17 (participants with the lowest support) to 0 (participants 26, 26.1–29, and <29 was 17, ࣘ17.3, ࣘ18, and ࣘ21 kg for
with the highest support)23 and Tinetti Mobility Test or women, respectively).25 Using the cutoff points indicated
Performance-Oriented Mobility Assessment (TMT). in the European Working Group on Sarcopenia in Older
Liguori et al 881

People (EWGSOP) consensus, subjects with a low grip 4-m gait speed decreased as MNA score decreased. GDS
strength presenting low muscle mass (skeletal muscle index score, BADLs and IADLs lost, and low social support
<8.87 and 6.42 kg/m2 in men and women, respectively) were increase as MNA score decreased. Similarly, both frailty
classified as “sarcopenia.”28 by Fried’s score and Rockwood’s score increased as MNA
score decreased. Among biomarkers, lymphocytes and b-
Biochemical Parameters CHE decreased, whereas CRP increased as MNA score
decreased.
Hemoglobin (normal values: 12.0–17.5 g/dL in men and Figure 1 shows the relationship among skeletal muscle
12.0–16.0 g/dL in women), lymphocytes (normal values: mass and strength stratified by MNA score in nonin-
1.0–4.8 × 103 ), serum albumin level (normal values: 3.6– stitutionalized elderly people. Muscle mass progressively
5.2 g/dL), total iron binding capacity (normal values: 218– decreased (from 17.6 ± 3.0 to 10.0 ± 2.9 kg/m2 ), as well as
411 μg/dL), C-reactive protein (CRP; normal values: <5 muscle strength (from 41.2 ± 10.2 to 27.7 ± 8.3 kg), as MNA
mg/dL), and butyryl-cholinesterase (b-CHE; normal values: score decreased.
5.400–13.200 U/L) were routinely obtained. At univariate analysis, age, CIRS-severity score, number
of drugs, BADLs lost, and CRP were negatively correlated
Statistical Analysis with skeletal muscle mass, whereas Tinetti score, 4-m gait
Baseline characteristics of the sample are expressed as speed, PASE score, serum albumin level, hemoglobin, b-
mean ± standard deviation. Participants were stratified CHE, and, more importantly, MNA score were positively
by the presence or absence of sarcopenia and stratified correlated with skeletal muscle mass. Multivariate analysis
by categories of MNA (<17, 17–23.5, ࣙ24). Categorical confirms this correlation except for serum albumin level and
variables were analyzed using χ 2 testing, continuous vari- CRP (Table 3). The positive linear relation between skeletal
ables using a 1-way analysis of variance, and nonparametric muscle mass and MNA score is shown in Figure 2A (y =
variables (ie, PASE score and lymphocyte number) using 0.65x + 2.2; r = 0.72; P < .001).
Wilcoxon-Mann-Whitney. Univariate regression analysis Univariate and multivariate linear regression analysis on
was used to test a correlation among muscle mass and muscle strength is shown in Table 3. Age, CIRS comorbidity
strength, and MNA score with other variables such as and severity, number of drugs, BADLs lost, and CRP were
age, BMI, WC, MMSE, GDS, CIRS-Comorbidity, CIRS- negatively correlated with muscle strength, whereas Tinetti
Severity, drug number, BADLs, IADLs, PASE, 4-m walking score, 4-m gait speed, PASE score, hemoglobin, b-CHE, and
speed, TMT, and social support score. Statistically signifi- MNA score were positively correlated with muscle strength.
cant variables were included into a multivariate regression Except for CRP, all linear relationships were confirmed at
model as potential confounders. All statistical analyses multivariate analysis. The positive linear relation between
were performed with SPSS software (version 15.0; SPSS, muscle strength and MNA score is shown in Figure 2B
Chicago, IL). A P value <.05 was considered statistically (y = 1.1x + 14.2; r = 0.46; P < .001).
significant.
Discussion
Results Our data show that MNA score, 1 of the most specific
tools for the assessment of malnutrition, is lower in non-
A total of 473 elderly subjects with a mean age of 80.9
institutionalized older adults with sarcopenia than in those
± 6.6 years was studied. Table 1 lists the anthropometric
without sarcopenia. Interestingly, MNA score is strongly
measurements, CGA, and biochemical assessment stratified
related to muscle mass and strength both at univariate and
in sarcopenia and nonsarcopenia according to EWGSOP
multivariate analyses. This evidence suggests that malnutri-
definition and diagnostic algorithm. The prevalence rate
tion state is frequently associated with a reduction of muscle
of sarcopenia was 13.1%. CIRS comorbidity and severity
mass and strength; therefore, it should be investigated,
score, drug number, and CRP were higher, whereas 4-minute
especially in the presence of sarcopenia in noninstitution-
gait speed, Tinetti score, and b-CHE were lower in elderly
alized older adults.
subjects with sarcopenia. In addition, MNA score together
with muscle mass and grip strength were lower in elderly
Sarcopenia
subjects with sarcopenia with respect to elderly subjects
without sarcopenia (Table 1). Age-related muscle mass decline is defined “sarcopenia,”
Table 2 lists the same parameters stratified for different and it is primarily due to the progressive atrophy and
levels of MNA score (ࣙ24, 17–23.5, and <17). Prevalence loss of type II muscle fibers and motor neurons.29 This
of sarcopenia progressively increased as MNA decreased phenomenon tends to be reduced at a rate of 1%–2% per
(from 6.1% to 31.4%; P < .001). Elderly subjects were year after the age of 50 years.1-3 Sarcopenia is characterized
progressively older, and MMSE and Tinetti score and by fibrosis and infiltration of adipose tissue determining a
882 Nutrition in Clinical Practice 33(6)

Table 1. Baseline Characteristics of the 473 Elderly Patients Enrolled in the Study Stratified for the Presence and Absence of
Sarcopenia.

Sarcopeniaa

All No (86.9%) Yes (13.1%)


Characteristics (N = 473) (n = 411) (n = 62) Pb

Anthropometric data
Age ± SD, y 80.9 ± 6.6 74.3 ± 8.1 80.5 ± 7.6 .145
Female sex, % 60.9 64.5 37.1 .057
BMI ± SD, kg/m2 27.4 ± 5.6 27.2 ± 3.1 24.2 ± 4.0 .064
Waist circumference ± SD, cm 99.7 ± 13.5 99.0 ± 10.8 96.4 ± 11.4 .007
Geriatric evaluation ± SD
Mini-Mental State Examination score 21.5 ± 6.3 23.2 ± 6.4 21.6 ± 4.4 .059
Geriatric Depression Scale score 7.6 ± 4.4 6.7 ± 4.6 8.6 ± 5.4 .187
CIRS-Comorbidity score 3.9 ± 2.2 3.40 ± 1.79 5.12 ± 2.05 .024
CIRS-Severity score 1.9 ± 0.5 1.60 ± 0.22 2.34 ± 0.55 .030
No. of drugs 6.1 ± 3.2 2.0 ± 1.0 4.0 ± 1.0 .020
BADLs lost, n 2.0 ± 1.8 1.4 ± 1.6 2.6 ± 1.8 .212
IADLs lost, n 4.2 ± 2.8 2.8 ± 1.4 4.0 ± 3.2 .321
Tinetti score 17.3 ± 7.8 22.4 ± 4.0 18.2 ± 2.2 .020
MNA score 20.9 ± 4.3 22.0 ± 4.0 15.4 ± 4.2 .024
4-m gait speed, m/s 0.46 ± 0.4 0.91 ± 0.20 0.60 ± 0.30 <.05
PASE score 37.6 ± 51.1 102.0 ± 40.8 38.4 ± 80.2 <.001
Social support score 8.3 ± 4.9 7.0 ± 2.0 9.5 ± 2.0 <.05
Fried’s frailty score 3.5 ± 1.5 2.2 ± 1.2 3.6 ± 1.2 <.05
Rockwood’s frailty score 21.2 ± 8.7 14.6 ± 9.4 28.2 ± 6.2 <.05
Muscle measurements
Grip strength, kg 26.2 ± 7.6 30.4 ± 8.6 21.8 ± 8.4 <.05
Skeletal muscle mass, kg/m2 8.0 ± 1.6 9.0 ± 1.6 6.6 ± 1.4 <.05
Biochemical measurements ± SD
Serum albumin level, g/dL 4.1 ± 0.7 4.0 ± 0.4 3.4 ± 0.8 .560
Total iron binding capacity, μg/dL 325.6 ± 54.6 296.4 ± 68.2 222.8 ± 74.2 .266
Hemoglobin, g/dL 12.9 ± 1.8 13.2 ± 1.4 12.4 ± 2.0 .059
Lymphocytes, n × 103 4.1 ± 8.2 2.2 ± 1.0 1.7 ± 1.7 .112
C-reactive protein, mg/dL 0.8 ± 0.4 0.40 ± 0.28 0.90 ± 0.44 <.05
Butyryl-cholinesterase, U/L 7480 ± 2320 8600 ± 1450 3400 ± 2850 <.05

BADLs, basic activities of daily living; BMI, body mass index; CIRS, Cumulative Index Rating Scale; IADLs, instrumental activities of daily
living; MNA, Mini Nutritional Assessment; PASE, Physical Activity Scale for the Elderly.
a Sarcopenia was defined in subjects with a low muscle strength presenting low muscle mass (skeletal muscle index <8.87 and <6.42 kg/m2 in men

and women, respectively). BMI-adjusted values were used as a cutoff point to classify low muscle strength (BMI ࣘ24, 24.1–28, and <28 was 29,
ࣘ30, and ࣘ32 kg for men; BMI ࣘ23, 23.1–26, 26.1–29, and <29 was 17, ࣘ17.3, ࣘ18, and ࣘ21 kg for women, respectively).
b P = statistical difference between the presence and the absence of sarcopenia.

reduction of functional capacity, an increase of disability Therefore, elderly adults may lose muscle mass and
and mortality, and therefore an increase of healthcare strength.33 Accordingly, older adults usually eat less protein
costs.3-5,30 Many mechanisms have been hypothesized to than younger adults.31 In fact, community-dwelling older
explain the origin of age-related decline in muscle strength adults (ࣈ10%) and those living in care homes (ࣈ30%) do not
and mass, including inflammation, physical inactivity, and consume the estimated average requirement for daily protein
malnutrition.31,32 intake (0.7 g/kg body weight/day),34,35 which represents the
minimum intake level to maintain muscle integrity for older
Malnutrition and Sarcopenia adults.36,37

The relationship between malnutrition and sarcopenia is Mini Nutritional Assessment as Tool
not well established, especially in elderly outpatients.29 A
disproportion between protein intake and protein needs may
for Malnutrition Detection
determine a loss of skeletal muscle mass because of im- Because malnutrition in the elderly is multifactorial, several
balance between muscle protein synthesis and degradation. measures of malnutrition in geriatric people have been
Liguori et al 883

Table 2. Baseline Characteristics of the 473 Older Adult Participants Enrolled in the Study stratified for Mini Nutritional
Assessment.

Mini Nutritional Assessment score

ࣙ24 (51.0%) 17-23.5 (34.2%) <17 (14.8%) P


Characteristics (n = 241) (n = 162) (n = 70) for Trend

Anthropometric data
Age ± SD, y 79.7 ± 6.5a 81.3 ± 6.7 81.8 ± 6.3 .001
Female sex, % 27.4 46.3 26.3 .432
Body mass index, kg/m2 28.1 ± 5.0a 27.6 ± 6.2 26.1 ± 5.0 <.05
Waist circumference, cm 100.3 ± 9.4a 98.5 ± 11.2 98.2 ± 10.1 <.05
Geriatric evaluation ± SD
Mini-Mental State Examination score 24.3 ± 4.9a 21.0 ± 6.3 18.5 ± 6.8 <.001
Geriatric Depression Scale score 4.8 ± 3.6a 8.3 ± 4.0 10.1 ± 4.1 <.001
CIRS-Comorbidity score 3.1 ± 2.1 4.1 ± 2.2 4.4 ± 2.0 .456
CIRS-Severity score 1.7 ± 0.4 1.9 ± 0.5 2.0 ± 0.4 .455
No. of drugs 5.2 ± 3.3 6.5 ± 3.0 6.6 ± 3.2 .184
BADLs lost, n 1.0 ± 1.5 2.1 ± 1.8 3.2 ± 1.8 <.05
IADLs lost, n 2.5 ± 2.6 4.5 ± 2.6 5.9 ± 2.2 <.05
Tinetti score 21.9 ± 6.0 16.2 ± 7.5 12.8 ± 7.1 <.05
MNA score 25.8 ± 1.5a 20.6 ± 1.8 14.5 ± 2.2 <.001
4-m gait speed, m/s 0.66 ± 0.47 0.46 ± 0.5 0.33 ± 0.5 <.05
PASE score 65.4 ± 60.8a 28.9 ± 43.8 16.1 ± 28.7 <.05
Social support score 5.4 ± 3.9 8.9 ± 4.6 11.4 ± 4.2 <.05
Fried’s frailty score 2.6 ± 1.5 3.6 ± 1.4 4.5 ± 1.3 <.001
Rockwood’s frailty score 15.7 ± 8.0 22.2 ± 7.4 27.2 ± 7.1 <.001
Sarcopenia, n (%) 15 (6.2) 25 (15.4) 22 (31.4) <.001
Muscle measurements ± SD
Grip strength, kg 41.2 ± 10.2 38.1 ± 9.3 27.7 ± 10.3 <.001
Skeletal muscle mass, kg/m2 17.6 ± 1.5 16.8 ± 2.1 10.0 ± 2.9 <.001
Biochemical measurements ± SD
Serum albumin level, g/dL 4.2 ± 0.5 4.2 ± 0.9 3.9 ± 0.4 .052
Total iron binding capacity, μg/dL 350.4 ± 50.4 341.0 ± 60.2 265.0 ± 80.2 .163
Hemoglobin, g/dL 13.2 ± 1.8 13.0 ± 1.7 12.0 ± 1.9 .420
Lymphocytes, n × 103 5.0 ± 9.8 4.9 ± 9.3 1.2 ± 0.6 <.05
C-reactive protein, mg/dL 0.50 ± 0.42a 0.64 ± 0.50 0.95 ± 0.30 .048
Butyryl-cholinesterase, U/L 7950 ± 2150 7650 ± 2300 7000 ± 2100 .042

BADLs, basic activities of daily living; CIRS, Cumulative Index Rating Scale; IADLs, instrumental activities of daily living; MNA, Mini
Nutritional Assessment; PASE, Physical Activity Scale for the Elderly.
a P < .01 vs MNA score <17.

developed. Biochemical and clinical indices are investi- Sarcopenia and Mini Nutritional Assessment
gated by Nutritional Risk Index38 and Geriatric Nutri- Score
tional Risk Index39 ; anthropometry, mobility, cognitive
state, self-perceived health, and nutrition by Short Form In our noninstitutionalized older adults, MNA score pro-
MNA,40 by Malnutrition Universal Screening Tool,41 and gressively decreases as muscle mass and strength are
by unintentional weight loss and poor intake (MST)42 ; and reducing in both univariate (r = 0.72 and r = 0.46, respec-
medical history and clinical and subjective evaluation by tively) and multivariate (r = 0.62 and r = 0.40, respectively)
Nutrition Risk Screening 2002 (NRS-2002).43 In contrast analyses. Interestingly, MNA score is significantly lower in
with nutrition screening tools, nutrition assessment methods subjects with sarcopenia vs without sarcopenia (15.4 ± 4.2
are more comprehensive and are frequently used by geria- vs 22.0 ± 4.0; P < .024). Some reports have investigated the
tricians. In particular, the Subjective Global Assessment44 relationship between MNA and sarcopenia. However, this
and MNA45 are largely used to perform a nutrition di- relationship in noninstitutionalized older adults is poorly
agnosis and begin nutrition management in older adult investigated. In contrast, among acute geriatric patients,
patients.46 malnutrition evaluated with MNA was associated to 30%
884 Nutrition in Clinical Practice 33(6)

Figure 1. Muscle mass and strength stratified by Mini


Nutritional Assessment score (ࣙ24, 17–23.5, <17) in
noninstitutionalized elderly people; P for trend = .001 for
both muscle mass (*P < .05 for <17 vs 17–23.5 and ࣙ24) and
strength (*P < .05 for <17 vs 17–23.5 and ࣙ24).

of the patients with sarcopenia.47 In addition, among male


residents in a nursing home, MNA scores were significantly
lower in residents having low compared with having normal
skeletal muscle mass (17.1 ± 3.4 vs 19.6 ± 2.5; P = .005).48
More importantly, in a prospective study, subjects with
malnutrition risk and sarcopenia have a risk for mortality 4
times higher than subjects without sarcopenia with normal
nutrition.49 Therefore, Vandewoude et al13 recently indi-
cated the MSS to depict the clinical scenario characterized
by both malnutrition and sarcopenia. Finally, it should
be emphasized that malnutrition plays a key role in the
pathogenesis of both frailty and sarcopenia.8 Malnutrition Figure 2. Regression linear relationship among muscle mass
and sarcopenia are frequently present in older adults with (A) and muscle strength (B) and Mini Nutritional Assessment
diabetes and chronic obstructive pulmonary disease that are score for noninstitutionalized elderly people.
characterized by a high degree of frailty.50,51 Interestingly,
the “frailty score” identified by Fried’s and Rockwood’s
methods was 2.6 ± 1.5 and 15.7 ± 8.0 at the highest status should be always evaluated to address therapeutical
MNA and 4.5 ± 1.3 and 27.2 ± 7.1 at the lowest MNA, interventions and lifestyle modifications (ie, increase of
respectively. protein intake and physical activity).3 Thus, it is mandatory
for clinician researchers to develop a reliable and valid
Evaluation of Nutrition Status in Patients With diagnostic algorithm to identify elderly subjects with both
Sarcopenia: Clinical Implications malnutrition and sarcopenia.

As discussed earlier, malnutrition and sarcopenia are con-


Conclusions
ditions frequently found in older adults that lead to nega-
tive outcomes including increased morbidity and mortality MNA score, 1 of the most accepted instruments for the
and increased healthcare costs and rehospitalizations.8,13,49 assessment of malnutrition, is lower in noninstitutionalized
Usually, these conditions have been separately screened, older adults with sarcopenia. Thus, the presence of mal-
but rarely are both conditions simultaneously assessed.8,13 nutrition is strongly related to muscle mass and strength.
Moreover, many patients concurrently show malnutrition These findings support that low MNA score is frequently
and sarcopenia; therefore, patient’s nutrition and functional associated to elderly outpatients with sarcopenia.
Liguori et al 885

Table 3. Univariate and Multivariate Linear Regression Analyses on Skeletal Muscle Mass and Muscle Strength.

Muscle Mass Muscle Strength

Univariate Multivariate Univariate Multivariate

Variables r P r P r P r P

Age −0.14 <.05 −0.15 <.05 −0.18 <.05 −0.16 <.05


Body mass index 0.06 .204 0.04 .220
Waist circumference 0.12 .340 0.06 .320
Mini-Mental State Examination 0.08 .120 0.08 .102
Geriatric Depression Scale −0.14 .347 −0.10 .300
CIRS-Comorbidity −0.10 .340 −0.10 <.05 −0.08 <.05
CIRS-Severity −0.07 .032 −0.06 <.05 −0.10 <.01 −0.09 <.01
No. of drugs −0.08 .030 −0.07 <.01 −0.04 .042 −0.04 <.05
BADLs −0.14 <.05 −0.12 <.05 −0.14 <.05 −0.12 <.05
IADLs −0.08 .342 −0.04 .442
Mini Nutritional Assessment 0.72 <.01 0.62 <.05 0.46 <.01 0.40 <.05
Tinetti score 0.65 <.01 0.051 .03 0.44 <.01 0.39 <.05
4-m gait speed 0.30 <.05 0.29 <.05 0.32 .05 0.28 <.05
PASE 0.57 <.05 0.30 .04 0.41 .001 0.29 <.05
Social support −0.04 .282 −0.07 .541
Serum albumin level 0.18 .022 0.14 .084 0.12 .064
Total iron binding capacity 0.14 .112 0.14 .122
Hemoglobin 0.24 <.04 0.20 <.05 0.30 <.05 0.26 .020
Lymphocytes 0.14 .114 0.15 .75
C-reactive protein −0.24 <.05 −0.08 .421 −0.20 <.05 −0.18 .325
Butyryl-cholinesterase 0.35 <.01 0.30 <.01 0.44 <.01 0.40 .036

BADLs, basic activities of daily living; CIRS, Cumulative Index Rating Scale; IADLs, instrumental activities of daily living; PASE, Physical
Activity Scale for the Elderly.

Acknowledgments 5. Iannuzzi-Sucich M, Prestwood KM, Kenny AM. Prevalence of sar-


copenia and predictors of skeletal muscle mass in healthy, older men
Funding source: Fondazione Roma NCDS-2013-00000331 –
and women. J Gerontol A Biol Sci Med Sci. 2002;57:M772-M777.
Sarcopenia and Insulin Resistance in the Elderly; Age- 6. Metter EJ, Talbot LA, Schrager M, Conwit R. Skeletal muscle strength
Associated Inflammation as a Shared Pathogenic Mechanism as a predictor of all-cause mortality in healthy men. J Gerontol A Biol
and Potential Therapeutic Target (DDM), Rome, ITALY. Sci Med Sci. 2002;57:B359-B365.
7. Landi F, Liperoti R, Fusco D, Mastropaolo S, Quattrociocchi D, Proia
Statement of Authorship A. Sarcopenia and mortality among older nursing home residents. J Am
Med Dir Assoc. 2012;13:121-126.
I. Liguori, F. Cacciatore, D. Bonaduce and P. Abete contributed 8. Cruz-Jentoft AJ, Kiesswetter E, Drey M, Sieber CC. Nutrition, frailty,
to conception and design; D. Della-Morte, G. Gargiulo and and sarcopenia. Aging Clin Exp Res. 2017;29:43-48.
G. Testa contributed to analysis and interpretation of data; 9. Walrand S, Guillet C, Salles J, Cano N, Boirie Y. Physiopathological
F. Curcio, G. Russo, M. Cellurale L. Aran and G. Bulli mechanism of sarcopenia. Clin Geriatr Med. 2011;27:365-385.
contributed to acquisition of data. 10. Cooper C, Fielding R, Visser M, et al. Tools in the assessment of
sarcopenia. Calcif Tissue Int. 2013;93:201-210.
11. Mithal A, Bonjour JP, Boonen S, et al; IOF CSA Nutrition Working
References Group. Impact of nutrition on muscle mass, strength, and performance
1. Baumgartner RN, Koehler KM, Gallagher D, et al. Epidemiology in older adults. Osteoporos Int. 2013;24:1555-1566.
of sarcopenia among the elderly in New Mexico. Am J Epidemiol. 12. Agarwal E, Miller M, Yaxley A, Isenring E. Malnutrition in the elderly:
1998;147:755-763. a narrative review. Maturitas. 2013;76:296-302.
2. Marcell TJ. Sarcopenia: causes, consequences, and preventions. J 13. Vandewoude MF, Alish CJ, Sauer AC, Hegazi RA. Malnutrition-
Gerontol A Biol Sci Med Sci. 2003;58:M911-M916. sarcopenia syndrome: is this the future of nutrition screening and
3. Rolland Y, Czerwinski S, Abellan Van Kan G, et al. Sarcopenia: its as- assessment for older adults? J Aging Res. 2012;2012:651570.
sessment, etiology, pathogenesis, consequences and future perspectives. 14. van Bokhorst-de van der Schueren MA, Guaitoli PR, Jansma EP, et al.
J Nutr Health Aging. 2008;12:433-450. Nutrition screening tools: does one size fit all? A systematic review of
4. Rantanen T, Guralnik JM, Sakari-Rantala R, et al. Disability, physical screening tools for the hospital setting. Clin Nutr. 2014;33:39-58.
activity, and muscle strength in older women: the Women’s Health and 15. Guigoz Y. The Mini Nutritional Assessment (MNA) review of the
Aging Study. Arch Phys Med Rehabil. 1999;80:130-135. literature: what does it tell us? J Nutr Health Aging. 2006;10:466-485.
886 Nutrition in Clinical Practice 33(6)

16. Testa G, Cacciatore F, Galizia G, et al. Waist circumference but not 35. Tieland M, Borgonjen-Van Den Berg K, van Loon LJ, de Groot LC.
body mass index predicts long-term mortality in elderly subjects with Dietary protein intake in community-dwelling, frail, and institutional-
chronic heart failure. J Am Geriatr Soc. 2010;58:1433-1440. ized elderly people: scope for improvement. Eur J Nutr. 2012;51:173-
17. Cacciatore F, Testa G, Langellotto A, et al. Role of ventricular rate 179.
response on dementia in cognitively impaired elderly subjects with atrial 36. Bauer J, Biolo G, Cederholm T, et al. Evidence-based recommenda-
fibrillation: a 10-year study. Dement Geriatr Cogn Disord. 2012;34:143- tions for optimal dietary protein intake in older people: a position paper
148. from the PROT-AGE study group. J Am Med Dir Assoc. 2013;14:542-
18. Testa G, Cacciatore F, Galizia G, et al. Depressive symptoms predict 559.
mortality in elderly subjects with chronic heart failure. Eur J Clin Invest. 37. Deutz NE, Bauer JM, Barazzoni R, et al. Protein intake and exercise
2011;41:1310-1317. for optimal muscle function with aging: recommendations from the
19. Testa G, Cacciatore F, Galizia G, et al. Charlson Comorbidity Index ESPEN Expert Group. Clin Nutr 2014;33:929-936.
does not predict long-term mortality in elderly subjects with chronic 38. Wolinsky F, Coe RM, McIntosh WA, et al. Progress in the development
heart failure. Age Ageing. 2009;38:734-740. of a nutritional risk index. J Nutr. 1990;120:1549-1553.
20. Washburn RA, Smith KW, Jette AM, Janney CA. The Physical Activity 39. Cereda E, Pedrolli C. The geriatric nutritional risk index. Curr Opin
Scale for the Elderly (PASE): development and evaluation. J Clin Clin Nutr Metab Care. 2009;12:1-7.
Epidemiol. 1993;46:153e62. 40. Rubenstein L, Harker J, Salva A, Guigoz Y, Vellas B. Screening
21. Peel NM, Kuys SS, Klein K. Gait speed as a measure in geriatric for undernutritionin geriatric practice: developing the short-form
assessment in clinical settings: a systematic review. J Gerontol A Biol mini nutritional assessment (MNA-SF). J Gerontol. 2001;56A:M366-
Sci Med Sci. 2013;68:39-46. M372.
22. Abete P, Cacciatore F, Ferrara N, et al. Body mass index and preinfarc- 41. Malnutrition Advisory Group (MAG). The ‘MUST’ Explanatory
tion angina in elderly patients with acute myocardial infarction. Am J Booklet. A Guide to the ‘Malnutrition Universal Screening Tool’
Clin Nutr. 2003;78:796-801. (‘MUST’) for Adults: A Standing Committee of the British Association
23. Mazzella F, Cacciatore F, Galizia G, et al. Social support and long- for Parenteral and Enteral Nutrition (BAPEN). Worcestershire (UK):
term mortality in the elderly: role of comorbidity. Arch Gerontol Redditch; 2003.
Geriatr. 2010;51:323-328. 42. Ferguson M, Capra S, Bauer J, Banks M. Development of a valid and
24. Abete P, Basile C, Bulli G, et al. The Italian version of the “frailty reliable malnutrition screening tool for adult acute hospital patients.
index” based on deficits in health: a validation study. Aging Clin Exp Nutrition. 1999;15:458-464.
Res. 2017;29(5):913-926. 43. Kondrup J, Rasmussen H, Hamberg O, et al. An ad hoc ESPEN Work-
25. Fried LP, Tangen CM, Walston J, et al; Cardiovascular Health Study ing Group. Nutritional Risk Screening (NRS 2002): a new method
Collaborative Research Group. Frailty in older adults: evidence for a based on analysis of controlled clinical trials. Clin. Nutr. 2003;22:321-
phenotype. J Gerontol A Biol Sci Med Sci. 2001;56:M146-M156. 336.
26. Vellas B, Guigoz Y, Garry PJ, et al. The Mini Nutritional Assessment 44. Detsky A, McLaughlin J, Baker J. What is subjective global assess-
(MNA) and its use in grading the nutritional state of elderly patients. ment of nutritional status. JPEN J Parenter Enteral Nutr. 1987;11:
Nutrition. 1999;15:116-122. 8-13.
27. Janssen I, Heymsfield SB, Baumgartner RN, Ross R. Estimation 45. Guigoz Y, Vellas B, Garry P. Assessing the nutritional status of
of skeletal muscle mass by bioelectrical impedance analysis. J Appl the elderly: the mini nutritional assessment as part of the geriatric
Physiol. 2000;89:465-471. evaluation. Nutr Rev. 1996;54:S59-S65.
28. Volpato S, Bianchi L, Cherubini A, et al. Prevalence and clinical cor- 46. Young AM, Kidston S, Banks MD, et al. Malnutrition screening tools:
relates of sarcopenia in community-dwelling older people: application comparison against two validated nutrition assessment methods in
of the EWGSOP definition and diagnostic algorithm. J Gerontol A Biol older medical inpatients. Nutrition. 2013;29:101-106.
Sci Med Sci. 2014;69:438-446. 47. Jacobsen EL, Brovold T, Bergland A, et al. Prevalence of factors
29. Cruz-Jentoft AJ, Landi F, Topinkova E, et al. Understanding sarcope- associated with malnutrition among acute geriatric patients in Norway:
nia as a geriatric syndrome. Curr Opin Clin Nutr Metab Care. 2010;13: a cross-sectional study. BMJ Open. 2016;6:e011512.
1-7. 48. Tufan A, Bahat G, Ozkaya H, et al. Low skeletal muscle mass index is
30. Goodpaster BH, Chomentowski P, Ward BK, et al. Effects of physical associated with function and nutritional status in residents in a Turkish
activity on strength and skeletal muscle fat infiltration in older adults: nursing home. Aging Male. 2016;19:182-186.
a randomized controlled trial. J Appl Physiol. 2008;105:1498-1503. 49. Hu X, Zhang L, Wang H, et al. Malnutrition-sarcopenia syndrome
31. Morley JE, Baumgartner RN. Cytokine-related aging process. J Geron- predicts mortality in hospitalized older patients. Sci Rep. 2017;7:
tol A Biol Sci Med Sci. 2004;59:924-929. 3171.
32. Soeters PB, Schols AM. Advances in understanding and assessing 50. Galizia G, Cacciatore F, Testa G, et al. Role of clinical frailty on
malnutrition. Curr Opin Clin Nutr Metab Care. 2009;12:487-494. long-term mortality of elderly subjects with and without chronic
33. Meng X, Zhu K, Devine A, Kerr DA, Binns CW, Prince RL. A 5-year obstructive pulmonary disease. Aging Clin Exp Res. 2011;23:
cohort study of the effects of high protein intake on lean mass and 118-125.
BMC in elderly postmenopausal women. J Bone Miner Res. 2009;24: 51. Abete P, Della-Morte D, Gargiulo G, Basile C, Langellotto A, Galizia
1827-1834. G, Testa G, Canonico V, Bonaduce D, Cacciatore F. Cognitive
34. Volpi E, Campbell WW, Dwyer JT, et al. Is the optimal level of impairment and cardiovascular diseases in the elderly. A heart-brain
protein intake for older adults greater than the recommended dietary continuum hypothesis. Ageing Res Rev. 2014;18:41-52.
allowance. J Gerontol A Biol Sci Med Sci. 2013;68:677-681.
Clinical Research

Nutrition in Clinical Practice


Volume 33 Number 6
Height Prediction From Ulna Length of Critically Ill Patients December 2018 887–892

C 2017 American Society for

Parenteral and Enteral Nutrition


DOI: 10.1177/0884533617716432
wileyonlinelibrary.com
Micheli S. Tarnowski, RD1 ; Estela I. Rabito, RD, PhD2 ; Daieni Fernandes, RD, MSc3 ;
Mariane Rosa, RD4 ; Manoela L. Oliveira, RD5 ; Vânia N. Hirakata, MSc6 ;
and Aline Marcadenti, RD, PhD7

Abstract
Background: Ulna length (UL) has been used in mathematical formulas to predict the body height of healthy and sick individuals.
However, the evaluation of its use with patients admitted to intensive care units (ICU) is scarce. The objective of this study was
to develop a mathematical equation to estimate critically ill patients’ height using the UL measure and to evaluate its agreement
with measured standing height. Methods: This cross-sectional study was performed at the ICU of a tertiary hospital in Brazil. A
total of 100 patients aged ࣙ18 years who had their body height measured before ICU admission were enrolled. The equation was
developed through multiple linear regression, and its agreement was assessed through paired Student’s t test and Bland-Altman
plot. Results: The following formula was obtained: height in cm = 153.492 – (7.97 × sex [sex: male = 1, female = 2]) + (0.974 ×
UL [in cm]). The difference between means of measured height (MH) and height estimated from UL was not significant (166.26
± 8.75 cm and 166.30 ± 5.29 cm, respectively, P = .96), and a significant correlation (r = 0.624, P < .001) was detected. In the
Bland-Altman analysis, UL was in agreement with MH; however, there was a significant bias (P < .001) suggesting that it may be
disproportional and dependent on the average’s height value. Conclusion: The mathematical equation for height estimation using
UL developed in this study matched the MH of critically ill patients. However, we suggest more studies for its validation. (Nutr
Clin Pract. 2018;33:887–892)

Keywords
nutritional assessment; ulna; body height; intensive care units; critical illness

Nutrition assessment is crucial to develop therapeutic plans predictor, easily obtained in bedridden patients, and less
and evaluate their effectiveness, particularly for critically ill affected by the aging process when the standing body height
patients.1 Anthropometric evaluation is generally impaired measure is difficult to obtain.7
for critical patients because of the high complexity of their Studies suggest the use of mathematical equations for de-
clinical state, with limitations and difficulties related to im- termining body height through UL.6,8-14 The Malnutrition
mobilization, the equipments required, and the significant
edema often present in this population.2 From the 1 Health Multidisciplinary Residence Program in Intensive
The Sociedad Española de Nutrición Parenteral y En- Therapy, Federal University of Health Sciences of Porto Alegre,
teral (Spanish Society for Parenteral and Enteral Nutrition) Porto Alegre, Brazil; 2 Department of Nutrition and Postgraduate
Program in Food and Nutrition, Federal University of Parana,
recommends that body mass and height be assessed when Curitiba, Brazil; 3 Division of Nutrition, Brotherhood of the Santa
patients are admitted to intensive care units (ICUs).3 Such Casa of Porto Alegre, Porto Alegre, Brazil; 4 Unimed Grande
assessment can be used for nutrition and medical manage- Florianopolis Hospital, São José, Brazil; 5 Department of Nutrition,
ment procedures, such as drug dose prescription, parame- Federal University of Health Sciences of Porto Alegre, Porto Alegre,
ters for mechanical ventilation, energy needs estimate, and Brazil; 6 Clinics Hospital of Porto Alegre, Porto Alegre, Brazil; and
7 Department of Nutrition, Federal University of Health Sciences of
ideal body weight calculation.3-5 Porto Alegre, and Postgraduate Program in Health Sciences:
The absence of exact anthropometric information is a Cardiology, Institute of Cardiology of the Rio Grande do Sul, Porto
reality in ICUs, where there is no gold standard for nutrition Alegre, Brazil.
assessment in critically ill patients. In this context, the Financial disclosure: None declared.
definition of energy and protein prescription goals must Conflicts of interest: None declared.
be based on plausible estimates of body mass and height,
This article originally appeared online on July 20, 2017.
to avoid malnutrition, overfeeding, and incorrect nutrition
diagnosis.4 Therefore, measurements of different body com- Corresponding Author:
Aline Marcadenti, RD, PhD, Department of Nutrition, Federal
partments, such as the length of long bones (considered
University of Health Sciences of Porto Alegre, Rua Sarmento Leite,
good predictors of body height), are feasible alternatives.6 245 Porto Alegre, 90050-170, Rio Grande do Sul, Brazil.
Ulna length (UL) has been used because it is a reliable height Email: alinemo@ufcspa.edu.br
888 Nutrition in Clinical Practice 33(6)

Universal Screening Tool (MUST) also suggests the use of Height was measured by the hospital nursing and nutri-
UL to estimate standing height for body mass index (BMI) tion staff, with stadiometers assembled with weighing scales
calculation to determine the nutrition risk classification.15 available in inpatient hospital units. The professionals were
However, such methods and equations were developed and trained to follow the same protocol: place the patient in a
validated with distinct population groups, based on sex, standing position, barefooted, wearing the least amount of
age, and race, but not critically ill patients. The use of clothing possible, with arms positioned alongside the body
UL to predict the height of critically ill patients would be and hands turned toward the thighs. MH was recorded in
useful, being a quick and easily applicable method. Besides, centimeters.
alternative measurements to estimate body height, such as The other anthropometric indexes were measured in the
knee height (KH), may have some limitations in the ICU, first 48 hours after ICU admission by 2 trained nutritionists:
particularly with patients who are overweight, have bilateral
amputations, or have femoral venous access.6
UL (cm): Measured with a bone anthropometer caliper
The use of UL to predict standing height was shown in
(WCS: Cardiomed, Curitiba, Brazil; range up to 280
a Brazilian population admitted to an emergency room.16
cm; 1-mm resolution) positioned between the elbow’s
However, in this study, the method suggested by MUST was
tip (olecranon process) and the midpoint of the wrist
used to identify the values of estimated height according
prominent bone (styloid process), according to MUST
to UL. Therefore, the main objective of our study was to
protocol.15 UL was measured preferentially in the left
develop a mathematical formula to estimate the standing
arm of the patient, folded in front of the patient’s chest,
body height of critically ill patients with their UL and to
with fingers pointing to the opposite shoulder.
evaluate its agreement with the measured height (MH). As
KH (cm): Measured with a bone anthropometer caliper
a secondary objective, we evaluated the agreement between
(WCS) with individuals in the supine position and the
the estimated height according to a mathematical formula
right leg forming a 90° angle with knee and ankle.
that uses the KH measurement and the MH of the subjects.
Estimated body height according to KH (KHH) was
calculated according to mathematical formulas proposed
Methods by Chumlea et al20 : for men, 64.19 – (0.04 × age in years)
+ (2.02 × KH in cm); for women, 84.88 – (0.24 × age in
This cross-sectional study was carried out between Septem-
years) + (1.83 × KH in cm).
ber 2015 and August 2016. Patients admitted to a general
ICU were enrolled (Santa Clara Hospital, Brotherhood
of the Santa Casa of Porto Alegre, Porto Alegre, Brazil). Statistical Analysis
The study included individuals of both sexes, aged ࣙ18
The analyses were performed with SPSS 22.0 (IBM,
years, whose standing body height was previously measured
Chicago, IL). Continuous variables were described with
at inpatient hospital units and recorded in their medical
mean and standard deviation (variables with normal distri-
records before being admitted to the ICU. They agreed to be
bution) or median and interquintile interval (nonparametric
part of the research and signed the informed consent form
variables); absolute numbers and frequencies were used to
(or their legal representatives). Individuals with incomplete
describe qualitative variables. Multiple linear regression,
medical records, those without MH previously registered,
in stepwise method, was used to create the body height–
and those whose KH could not be obtained at the ICU were
predictive mathematical formula based on UL. Pearson’s
excluded.
correlation test was used for correlations of the different
The data were recorded in a standardized questionnaire.
heights (MH, UL height [ULH], and KHH). Agreements
Age (years), sex, ICU length of stay (days), and reason
among the heights were evaluated through Student’s t test
for admission in the ICU (cardiovascular, gastrointestinal,
for paired samples, multiple linear regression for assessing
respiratory, postsurgery, infection/sepsis, shock, and other)
the bias significance, and Bland-Altman plot. P < .05 values
were the information collected from the electronic medical
were considered significant.
records to characterize the sample.
The study was approved by the Research Ethics Com-
The nutrition risk and/or nutrition status classification
mittee of the Brotherhood of the Santa Casa of Porto Ale-
was obtained through the Nutrition Risk in the Critically
gre (protocol 40073414.9.0000.5335) and by the Research
Ill (NUTRIC) score,17 Subjective Global Assessment,18 and
Ethics Committee of the Federal University of Health
BMI (kg/m2 ), calculated with the patient’s body mass and
Sciences of Porto Alegre.
MH. The patient’s body mass (kg) was obtained directly
through the electronic medical record (if the patient was
weighed up to 48 hours before ICU admission) or estimated
Results
through a mathematical formula19 (in the first 48 hours after In total, 100 patients who stayed a median of 8 days
of ICU admission). (range 3–15) in the ICU were evaluated. Regarding the
Tarnowski et al 889

Table 1. Characteristics of the Sample.a

Variables Total Sample (N = 100)

Sex
Male 57 (57)
Female 43 (43)
Age, y 62.47 ± 16.60
Body mass index, kg/m2 24.49 ± 4.93
NUTRIC score
High nutrition risk 22 (22)
Low nutrition risk 88 (88)

NUTRIC, Nutrition Risk in the Critically Ill.


a Values are presented as n (%) or mean ± SD.

Table 2. Linear Regression Model Used to Identify the


Predictive Equation for Height.a

Nonstandardized Figure 1. Scatter plot showing the correlation between


Coefficient measured height (MH) and height estimated from ulna length
(ULH; equation proposed by authors). r = 0.624 (P < .001).
Factors B SE t P Value

Constant 153.492 9.467 16.214 <.001


Sex −7.970 1.632 −4.884 <.001
Ulna length, cm 0.974 0.322 3.030 .003

a Dependent variable: standing measured height (cm). R2 = 0.364.

leading causes to ICU admission, 35% of the patients had


postsurgery complications; 21% were in shock; and 12%
had gastrointestinal or respiratory diseases. According to
the Subjective Global Assessment, 28% of the patients
were moderately undernourished, and 20% were severely
undernourished; 7% had a BMI <18.5 kg/m2 . Demographic
and anthropometric data related to the sample are described
in Table 1.
Two models were used for the construction of the predic-
tive mathematical formula: in the first, the linear regression
model included age, sex, and UL; in the second, sex and UL
only. The analysis showed that among all variables used, sex Figure 2. Scatter plot showing the correlation between
measured height (MH) and height estimated from knee height
and UL were the best predictors of MH, the residues of
(KHH; equations proposed by Chumlea et al20 ). r = 0.592
which showed normal distribution and were independent of (P < .001).
one another (Durbin-Watson test = 1.742). Therefore, the
following mathematical formula for height prediction was
obtained: height in cm = 153.492 − (7.97 × sex [sex: male between the averages of MH and ULH (difference = −0.04
= 1, female = 2]) + (0.974 × UL [in cm]). Statistical data cm, P = .96), suggesting an agreement between the methods.
related to the equation of ULH are presented in Table 2. The Bland-Altman plot (Figure 3) shows the limits of
Figures 1 and 2 show scatter plots and the correlations agreement (mean differences ± 2 standard deviations) as
between MH and ULH (Figure 1) and between MH and well as the bias. The result of this was significant in the linear
KHH (Figure 2). Both were statistically significant (P < regression analysis (P < .001), suggesting that the limits of
.001); however, MH and ULH presented a superior corre- agreement depend on the average height values and that
lation (r = 0.624) versus MH and KHH (r = 0.592). the bias may be disproportional: when mean height values
Regarding means of different measures of height, MH are small, the difference between methods is small; when
was 166.26 ± 8.75 cm; ULH, 166.30 ± 5.29 cm; and mean height values increase, so does the difference between
KHH, 157.56 ± 8.73 cm. There was no significant difference methods.
890 Nutrition in Clinical Practice 33(6)

Figure 5. Bland-Altman plot showing the agreement between


Figure 3. Bland-Altman plot showing the agreement between measured height and height estimated from knee height.
measured height and height estimated from ulna length. The Dotted line indicates bias, and continuous lines indicate limits
dotted line indicates bias, and the continuous lines indicate the of agreement. IL, inferior limit; SL, superior limit.
limits of agreement. IL, inferior limit; SL, superior limit.
regression equation, y = −99.6 + 0.6 × x, suggests that the
difference between MH and ULH is dependent on MH. For
example, the difference between the proposed mathematical
formula (ULH) and the standing real height (MH) of an
individual at 155 cm would be −6.6 cm; for an individual
with 167 cm of body height, it would be 0.6 cm; and for an
adult measuring 189 cm, the difference would be 13.8 cm.
A significant difference between MH and KHH averages
was observed (difference: 8.69 cm, P < .001). However, this
bias was not significant in the linear regression analysis (P
= .98), suggesting that it is proportional, as observed in
Figure 5. The limits-of-agreement interval on the Bland-
Altman plot was higher when compared with ULH.

Discussion
This study evaluated the use of UL as a method to estimate
the height of critically ill patients. Because performing the
height measurement through the gold standard (upright
Figure 4. Concordance and confidence intervals for the
posture) is often very difficult in this setting, our main
differences (y) and the means (x) between measured height
and height estimated from ulna length. Linear regression goal was to develop a mathematical formula for height
equation: y = −99.6 + 0.6 × x; R2 = 0.312. estimation with UL.
We decided to exclude both the recumbent height
measurement—because it is unfeasible for many critically
Because there was a significant association between ill patients—and the patient’s reported body height, due to
the differences and the averages (bias: −0.04 cm, P < the trend for height reduction related to the aging process
.001), we decided that the relationship between averages and the fact that many individuals ignore their actual height
and differences (estimates of maximum differences) should (ie, many elderly remember only their height measured at
be expressed according to a line of linear regression. youth).21
Figure 4 shows the concordance and confidence intervals Due to alternative methods for estimating height, some
for the difference (cm) between MH and ULH (y) and equations based on UL have been explored among dif-
the average (cm) between MH and ULH (x). The linear ferent populations, such as the British and Portuguese,14
Tarnowski et al 891

Turks,13 Sudanese,22 Indians and Iraqis,12 and Sri Lankan,11 both. The best regression model was the one based on
as well as with American9,23 and Australian5 pediatric both variables (fibula length + UL), showing a strong and
populations. However, these mathematical formulas were significant correlation (r = 0.87, P < .001). The predictive
created for healthy populations and were based on different formula based on only UL showed moderate correlations
ethnicities.10 Regression analysis is considered a reliable tool for Indians and Iraqis (r = 0.62 and r = 0.59, respectively),
for height estimations, and one of the advantages of this supporting our results.
method is to create an equation with a single body measure. Our study had some limitations. Among them, we men-
However, due to its less accurate predictive capability in the tion the sample size, because the enrollment of a larger
face of great variability among the body proportions of dif- number of individuals could imply a bias decrease. How-
ferent populations, it is suggested that each predictive equa- ever, our results are consistent with other studies assess-
tion be specifically created for the population in question.22 ing healthy populations. In our study, we evaluated only
Using UL, we identified an agreement between the height patients whose MH was listed on the medical record, and
estimated by our mathematical formula and the patient’s such procedures are not always performed in hospitals—
MH, despite some reservations. We observed that the esti- underreporting of patients’ weight and height data is not un-
mation resulting from the mathematical formula tends to be common. Although professionals were previously trained,
influenced by extremes; that is, for tall individuals, it tends we cannot warrant that the protocol for height measure was
to overestimate stature, while for those of very low stature, it homogenized among all who were responsible for measuring
tends to underestimate it. In sum, its utility is limited at the patients’ height in the hospital admission units.
extremes of height. The equation based on KH for height
estimation presented a systematic bias; that is, KHH was
not influenced by extremes, and the bias is uniform among
Conclusion
patients, being less influenced by very tall or very short The mathematical equation based on UL to predict body
individuals. Despite the large limits-of-agreement interval height that was developed in this study seems to provide
observed in our study regarding ULH (13.34 to −13.46 good standing height estimation for critically ill patients.
cm), it is lower than that observed in a study conducted in We suggest, however, that our results be replicated in other
a Brazilian emergency room (14.39 to −13.69 cm), which populations of hospitalized individuals, to reinforce our
used the MUST protocol to evaluate the values of estimated findings. Regarding next steps of our research, we intend to
height with UL.16 validate our equation in a larger population of critically ill
In a study performed in the ICU of a European univer- patients.
sity hospital,6 researchers assessed whether different math-
ematical formulas of height prediction validated among
Acknowledgments
healthy populations would be accurate for critically ill
patients, and their correlation between the height estimated We thank the Brotherhood of the Santa Casa de Porto Alegre,
its nursing and central ICU staff, and the Nutrition and
through UL and MH was lower than the correlation ob-
Dietetics Service, as well as the Federal University of Health
served in our study (r = 0.51). It reinforces the impor-
Sciences of Porto Alegre. We also thank Marcelo Oliveira da
tance of elaboration and validation of specific predictive Silva (Lecttura Traduções) for English language support.
equations for each population. Ahmed22 assessed the height
prediction of a Sudanese adult population through the long Statement of Authorship
bones of upper limbs; with an equation based on UL, a
A. Marcadenti, D. Fernandes, and E. I. Rabito equally con-
positive correlation was observed in relation to MH (r = tributed to the conception and design of the research; M. Rosa
0.73 for men and r = 0.72 for women), showing that the use contributed to the design of the research; M. Rosa, M. L.
of this body compartment may be a feasible alternative for Oliveira, and M. S. Tarnowski contributed to the acquisition
estimating height, for both men and women. of the data; A. Marcadenti, E. I. Rabito, and V. N. Hirakata
In a study with critically ill patients performed in a contributed to the analysis and interpretation of the data; and
Swiss university hospital,4 MH and the Chumlea method M. S. Tarnowski and A. Marcadenti drafted the manuscript.
for estimating height were compared. Similar to our re- All authors critically revised the manuscript, agree to be fully
sults, a satisfactory correlation was observed between body accountable for ensuring the integrity and accuracy of the
heights estimated by KH and MH; however, it presented an work, and read and approved the final manuscript.
overestimation of height values, with a varied and randomly
References
distributed dispersion.
1. Hejazi N, Mazloom Z, Zand F, Rezaianzadeh A, Amini A. Nutritional
Al-Wasfi and Puranik12 assessed the height prediction
assessment in critically ill patients. Iran J Med Sci. 2016;41(3):171-179.
of 196 healthy Indians and Iraqis, using measurements of 2. Ghorabi S, Ardehali H, Amiri Z, Shariatpanahi VZ. Association of the
2 body parts: fibula and ulna. Based on that, 3 equations adductor pollicis muscle thickness with clinical outcomes in intensive
were created: first, using fibula length; second, UL; third, care unit patients. Nutr Clin Pract. 2016;31(4):523-526.
892 Nutrition in Clinical Practice 33(6)

3. Ruiz-Santana S, Sánchez AJA, Abilés J. Guidelines for specialized 14. Barbosa VM, Stratton RJ, Lafuente E, Elia M. Ulna length to predict
nutritional and metabolic support in the critically-ill patient: update. height in English and Portuguese patient populations. Eur J Clin Nutr.
Consensus SEMICYUC-SENPE: nutritional assessment. Nutr Hosp. 2012;66(2):209-215.
2011;26(2):12-50. 15. Elia M. The MUST Report. Nutritional Screening of Adults: A Multi-
4. Berger MM, Cayeux M, Schaller M, Piazza G, Chiolero RL. Stature disciplinary Responsibility. Development and Use of the Malnutrition
estimation using the knee height determination in critically ill patients. Universal Screening Tool (MUST) for Adults. Redditch, England:
e-SPEN. 2008;3:e84-88. BAPEN; 2003.
5. Gauld LM, Kappers JBN, Carlin JB, Robertson. Height prediction 16. Silva FM, Figueira L. Estimated height from knee height or ulna length
from ulna length. Dev Med Child Neurol. 2004;46:475-480. and self-reported height are no substitute for actual height in inpatients.
6. L’her E, Martin Babau J, Lellouch F. Accuracy of height estimation Nutrition. 2017;33:52-56.
and tidal volume setting using anthropometric formulas in an ICU 17. Rosa M, Heyland DK, Fernandes D, Rabito EI, Oliveira ML, Marca-
Caucasian population. Ann Intensive Care. 2016;6(1):55. denti A. Translation and adaptation of the NUTRIC score to identify
7. Dennis DM, Hunt EE, Budgeon CA. Measuring height in recumbent critically ill patients who benefit the most from nutrition therapy. Clin
critical care patients. Am J Crit Care. 2015;24(1):41-47. Nutr ESPEN. 2016;14:31-36.
8. Lorini C, Collini F, Castagnoli M, et al. Using alternative or direct 18. Detsky AS, McLaughlin JR, Baker JP, et al. What is subjective
anthropometric measurements to assess risk for malnutrition in nursing global assessment of nutritional status? JPEN J Parenter Enteral Nutr.
homes. Nutrition. 2014;30(10):1171-1176. 1987;1:8-13.
9. Forman MR, Zhu Y, Hernandez LM, et al. Arm span and ulnar length 19. Chumlea WC, Guo S, Roche AF, Steinbaugh ML. Prediction of body
are reliable and accurate estimates of recumbent length and height in a weight for the nonambulatory elderly from anthropometry. J Am Diet
multiethnic population of infants and children under 6 years of age. J Assoc. 1988;88(5):564-568.
Nutr. 2014;144(9):1480-1487. 20. Chumlea WC, Roche AF, Steinbaugh ML. Estimating stature from
10. Madden AM, Tsikoura T, Stott DJ. The estimation of body height from knee height for persons 60 to 90 years of age. J Am Geriatr Soc.
ulna length in healthy adults from different ethnic groups. J Hum Nutr 1985;33(2):116-120.
Diet. 2012;25(2):121-128. 21. Sorkin JD, Muller DC, Andres R. Longitudinal change in height of
11. Ilayperuma I, Nananyakkara G, Palahepitiya N. A model for the men and women: implications for interpretation of the body mass
estimation of personal stature from the length of forearm. Int J index: the Baltimore longitudinal study of aging. Am J Epidemiol.
Morphol. 2010;28(4):1081-1086. 1999;150(9):969-977.
12. Al-Wasfi AAH, Puranik MG. Estimation of height from length of ulna 22. Ahmed AA. Estimation of stature from the upper limb measurements
and length of fibula in Indian and Iraqi population. Int J Inf Res Rev. of Sudanese adults. Forensic Sci Int. 2013;228(1-3):178.e1-e7.
2015;2:904-908. 23. Haapala H, Peterson MD, Daunter A, Hurvitz EA. Agreement be-
13. Duyar I, Peli C. Estimating body height from ulna length: need of tween actual height and estimated height using segmental limb lengths
a population-specific formula. Eurasian J Anthropol. 2010;1(1):11- for individuals with cerebral palsy. Am J Phys Med Rehabil. 2015;
17. 94(7):539-546.
Clinical Observations

Nutrition in Clinical Practice


Volume 33 Number 6
Venovenous Extracorporeal Membrane Oxygenation December 2018 893–896

C 2018 American Society for

in an Adult Patient With Prader-Willi Syndrome: A Nutrition Parenteral and Enteral Nutrition
DOI: 10.1002/ncp.10069
Case Report wileyonlinelibrary.com

Stacy Pelekhaty, RD, LDN, CNSC1 ; and Jay Menaker, MD2

Abstract
Prader-Willi Syndrome (PWS) is a genetic condition that results in a constellation of symptoms and typically results in hyperphagia
and obesity in adulthood. Critically ill adults with PWS present a unique challenge to the nutrition professional, particularly when
they require support modalities such as extracorporeal membrane oxygenation (ECMO). The purpose of this case study is to review
the nutrition care of a critically ill adult patient with PWS who required venovenous ECMO. The patient was successfully managed
with a hypocaloric, high-protein approach, which did not result in the diagnosis of malnutrition during his hospitalization. The
patient was ultimately transitioned off extracorporeal life support and discharged to a rehabilitation facility. (Nutr Clin Pract.
2018;33:893–896)

Keywords
critical illness; enteral nutrition; extracorporeal membrane oxygenation; nutrition assessment; nutritional support; Prader-Willi
Syndrome

Prader-Willi syndrome (PWS) is a genetic condition that Case Study


results from the lack of expression of paternal genes from
chromosome 15q11.2-q13.1 Characteristic features of PWS A 24-year-old male with known PWS presented to an
include hyperphagia, absent gag reflex, high pain tolerance, outside hospital with several days of upper respiratory
developmental delays, and obesity in adults.1 However, symptoms. The patient was hypoxic and initially tried on
adult patients with PWS may not experience the same noninvasive ventilation; however, his condition continued
metabolic alterations as age-, sex-, and weight-matched con- to deteriorate and required endotracheal intubation. The
trol subjects; therefore, generalized guidelines for obesity patient’s clinical course continued to worsen, and he ex-
should be applied with caution.2-7 Optimizing the nutrition perienced hypotension that required vasoactive support.
care of the critically ill adult patient with PWS therefore Due to this shock state and overwhelming critical illness,
presents a unique challenge. the patient experienced development of acute anuric renal
Venovenous extracorporeal membrane oxygenation (VV failure that required renal replacement therapy (RRT).
ECMO) is a life support method that provides oxygena- Despite maximal therapy provided by the outside hospital,
tion and ventilation to critically ill patients in profound the patient’s respiratory status continued to decline and
respiratory failure.8 The use of ECMO to support adult
From the 1 Department of Clinical Nutrition, University of Maryland
patients has increased significantly in the past 10 years,
Medical Center, Baltimore, Maryland, USA; and 2 Department of
with 2807 cases in 251 centers in the United States reported Surgery, University of Maryland School of Medicine, R Adams
in 2017 according to the Extracorporeal Life Support Cowley Shock Trauma Center, Baltimore, Maryland, USA.
Organization.9 A 2015 study by Kon and colleagues10 Financial disclosure: None.
demonstrated that obese patients who require VV ECMO
Conflicts of interest: None.
had similar outcomes as nonobese patients. In addition, the
This article originally appeared online on March 12, 2018.
PRESERVE score has increased body mass index (BMI)
as protective against mortality in patients who require VV Corresponding Author:
Stacy Pelekhaty, RD, LDN, CNSC, Department of Clinical
ECMO.11 The purpose of this case report is to review the
Nutrition, University of Maryland Medical Center, 22 S. Greene
nutrition management of an adult male with PWS who Street, Baltimore, MD 21409, USA.
requires VV ECMO. Email: spelekhaty@umm.edu
894 Nutrition in Clinical Practice 33(6)

Table 1. Estimated Energy and Protein Requirements.

Nutrient Permissive Underfeeding5 Institutional Practice Reference

Energy (kcal) 22–25 kcal/kg IBW Penn State Equation (Tmax 36.7, Mve 0) 20–30 kcal/kg Mifflin St Jeor equation
1408–1600 kcal/day 2637 × 1.1–1.2 = 2901–3164 kcal/day 3800–5700 kcal/day 2833 kcal/day
Protein ࣙ2.5 g/kg IBW 3–4 g protein/kg IBW 0.8 g/kg actual weight
ࣙ160 g/day 192–256 g/day 152 g/day

IBW, ideal body weight; Mve, minute ventilation (Liters per minute); Tmax, 24-hour maximum temperature in degrees Celsius.

on hospital day 3 he was transferred to our institution gasses or attaching a calorimeter to the oxygenator have
for VV ECMO evaluation. On arrival to our facility, the been published but lack wide validation.15-17 In addition,
patient had a peak airway pressure on the ventilator of these methods require equipment not currently available at
44 cm H2 O with marginal oxygenation. The patient was this institution and/or blood draws that are not part of
cannulated for VV ECMO based on his borderline ratio routine patient care. Protein needs were estimated in the very
of partial pressure of arterial oxygen (PaO2 ) and fraction high range at 3–4 g of protein/kg IBW related to elevated
of inspired oxygen delivered by the ventilator (FIO2 ) and demand during CRRT and VV ECMO support.
high ventilatory needs. A 29 French drainage cannula was Continuous enteral nutrition (EN) was provided with
placed in the right common femoral vein and 23 French a 1.2 kcal/mL formula containing fish oil and 75 g/L of
arterial cannula was placed in the right internal jugular vein protein via a nasojejunal small-bore feeding tube for a goal
for return. After cannulation, the patient was transferred of 1440 mL daily using a rate-based feeding approach.
to our dedicated multidisciplinary intensive care unit for This formula was selected primarily for its high-protein
patients who require VV ECMO. Continues renal replace- and low-fiber content given the patient’s high protein de-
ment therapy (CRRT) was continued through the ECMO mands and critical illness.12 A protein modular providing 15
circuit. Baseline anthropometrics according to the parents g/packet was provided as a bolus of 2 packets 4 times daily.
were weight of 190 kg (420 lb) and height of 66” (167.6 The combined nutrition regimen provided 1728 kcal/day
cm) with a calculated ideal body weight (IBW) of 64 kg (27 kcal/kg IBW) and 228 g of protein/day (3.6 g of
(141 lb) according to the Hamwi equation. On arrival, protein/kg IBW). During the early stage of his admission,
the patient met criteria for class III obesity based on a the patient required sedation with propofol, which uses a
BMI of 68 kg/m2 . Nutrition-focused physical examination 10% soybean–oil based fat emulsion as the carrier fluid and
revealed diffuse fluid accumulation and significant obesity provides 1.1 kcal/mL. The patient’s sedation requirements
limiting the assessment of underlying lean body mass, and provided an additional 600–900 kcal/day. To avoid over-
an abdomen/pelvis computed tomography (CT) scan was feeding during this time, the EN regimen was adjusted to
not available for review. 840 mL/day with 8 packets of protein modular (2 packets
Energy and protein requirements are summarized in administered 4 times daily) to provide 1008 kcal (16 Kcal/kg
Table 1. Permissive underfeeding needs were calculated IBW) and 183 g of protein (2.9 g of protein/kg IBW).
according to the 2016 American Society for Parenteral and On hospital day 21 the patient’s clinical status had
Enteral Nutrition (ASPEN) and Society of Critical Care improved to the point of no longer requiring VV ECMO
Medicine (SCCM) guidelines.12 Energy demand during VV support. After the liberation of ECMO support, the patient
ECMO has yet to be established, therefore institutional required mechanical ventilation for an additional 23 days.
practice is to add an additional 10%–20% to the Penn State Indirect calorimetry was completed on hospital day 27
equation with minute ventilation of 0 L/min. International using the Viasys Vmax ENCORE system. MEE was 2870
practice surveys have established 20–30 kcal/kg as the most kcal/day and respiratory quotient was 0.74. This was 100%
commonly used method of estimating energy needs on VV of estimated basal energy expenditure using Mifflin St Jeor.
ECMO13,14 ; therefore, this is calculated as a cross-reference. EN was adjusted to 1560 mL/day with 2 packets of protein
Due to the novel nature of this patient, basal metabolic rate modular administered 4 times daily, increasing the nutrition
was also calculated. The Mifflin St Jeor equation was used provision to 1872 kcal (29 kcal/kg IBW) and 237 g protein
because this was found to correlate acceptably with mea- (3.7 g protein/kg IBW), providing 65% of MEE according
sured energy expenditure (MEE) in community-dwelling to permissive underfeeding guidelines.12 Nitrogen balance
patients with PWS.2 Indirect calorimetry is not possible was not available due to the patient’s continued renal failure.
during ECMO because gas exchange occurs via the ECMO The amount of goal nutrition support received during the
circuitry and is minimal in the native lung. Methods to patient’s hospitalization is summarized in Table 2.
address this change in the site of ventilation during ECMO During the remainder of the patient’s 53-day hospital
using values from preoxygenator and postoxygenator blood stay, his renal function recovered and no dialysis was
Pelekhaty and Menaker 895

Table 2. Nutrition Support Goals and Delivery During Hospitalization.

EN Daily Protein
Hospital Infusion Goal Modular % Goal
Days (mL) Goal Goal Energy Provision Goal Protein Provision Received

2–11 840 8 packets 1008 kcal (16 kcal/kg 183 g of protein (2.9 g of protein/kg 86
IBW) IBW)
12–26 1440 8 packets 1728 kcal (27 kcal/kg 228 g of protein (3.6 g of protein/kg 86
IBW) IBW)
27–34 1560 8 packets 1872 kcal (29 kcal/kg 237 g of protein (3.7 g of protein/kg 88.5
IBW) IBW)

EN, enteral nutrition; IBW, ideal body weight.

required upon discharge. In addition, the patient success- ing of critically ill adult patients with obesity (BMI >
fully weaned off mechanical ventilation on hospital day 44, 30 mg/kg2 ).12 Permissive underfeeding refers to providing
and he passed a speech and language pathology swallow less energy than required to meet total energy expenditure,
evaluation for a pureed diet with nectar thick liquids. At while providing sufficient protein to meet requirements.
the time of discharge to a subacute facility, the patient This feeding strategy may reduce the risk for metabolic
was consuming adequate oral nutrition to meet his estimate complications from overfeeding, which include hypercarbia
energy and protein needs, and no longer required EN and hyperglycemia.12 Despite improved insulin sensitivity
support. The patient’s weight on discharge from the hospital and reduced incidence of diabetes mellitus compared with
was recorded as 185 kg (407 lb), which is 97% of baseline matched controls in patients with PWS, efforts should be
weight. made to minimize hyperglycemia related to critical illness.
In addition, according to a case series of critical illness and
sudden death in PWS, PWS results in abnormal response
Discussion to hypoxia18 ; therefore, reducing respiratory stress from
PWS is a genetic syndrome with a worldwide prevalence overfeeding is an important nutrition management consid-
estimated to be 1 in 10,000–30,000.1 Providers working in eration. Overfeeding can complicate ventilatory manage-
intensive care settings may not be familiar with the features ment by causing CO2 retention12 ; it follows that overfeeding
and unique challenges presented by this patient population. should be avoided while patients require VV ECMO support
PWS presents as poor feeding and hypotonia in the infant, for profound respiratory failure. The 2016 ASPEN and
which transitions to hyperphagia around 8 years of age and SCCM guidelines do not provide recommendations on the
progresses to obesity by adulthood.1 Additional manifes- nutrition care of patients receiving VV ECMO. Based on in-
tations of the syndrome include developmental delays, be- ternational surveys, current practice among ECMO centers
havioral disturbances, dysmorphic features, hypogonadism, is to provide 20–30 kcal/kg and 1.2–2 g protein/kg.13,14 The
and short stature.1 practice at this institution incorporates cross-referencing
Patients with PWS may not have the same risk for this kilocalorie per kilogram (kcal/kg) range with the Penn
comorbidities as patients with obesity who do not have State equation with modifications because of the alterations
PWS. Current research suggests that patients with PWS in minute ventilation on VV-ECMO to reduce the risk for
have improved insulin sensitivity and reduced frequency of overfeeding.
nonalcoholic fatty liver disease when compared with age-, In the absence of disease-specific nutrition guidelines, the
sex-, and weight-matched control subjects.2-4 Treatment of practice at this institution is to rely on general guidelines
hypogonadism with human growth hormone in childhood for the nutrition management of critically ill adult pa-
appears to further improve the metabolic profile and body tients with additional monitoring, such as nitrogen balance
composition in adults with PWS.5 Energy expenditure in and indirect calorimetry. This enhanced monitoring aug-
healthy adults with PWS is less7 than that of matched ments the standard practice of routinely monitoring weight,
controls, and disease-specific predictive equations are not nutrition-focused physical examination, and the amount of
widely validated.2 goal nutrition support received. Routine nutrition-focused
A thorough literature review did not yield guidelines for physical examination conducted during the patient’s stay
the management of critically ill patients with obesity related did not suggest the development of fat or muscle wasting
to PWS, nor do specific guidelines exist for the nutrition with permissive underfeeding; however, exams were limited
care of patients who require ECMO. The 2016 ASPEN by both nonpitting edema and obesity. Imaging tools such
and SCCM guidelines recommend permissive underfeed- as CT scans and bedside ultrasound (US) may provide
896 Nutrition in Clinical Practice 33(6)

additional methods to assess the nutrition status of critically 6. Coupaye M, Lorenzini F, Lloret-Linares C, et al. Growth hormone
ill patients.19 CT or US in this patient may have revealed therapy for children and adolescents with Prader-Willi syndrome is
associated with improved body composition and metabolic status in
fat or muscle loss that could not be identified through
adulthood. J Clin Endocrinol Metab. 2013;98(2):E328-E335. https://
physical examination because of class II obesity and the doi.org/10.1210/jc.2012-2881.
presence of nonpitting edema on arrival. However, this 7. Butler MG, Theodoro MF, Bittel DC, Donnelly JE. Energy ex-
patient did not require an abdomen/pelvis CT on admission, penditure and physical activity in Prader-Willi syndrome: compari-
and assessing the L3 slice for body composition may not be son with obese subjects. Am J Med Genet A. 2007;143A(5):449-459.
https://doi.org/10.1002/ajmg.a.31507.
practical for routine use.19 Bedside US offers a convenient
8. Makdisi G, Wang I. Extra corporeal membrane oxygenation (ECMO)
and noninvasive method of incorporating imaging into review of a lifesaving technology. J Thorac Dis. 2015;7(7):E166-E176.
nutrition assessment; however, there is not currently con- https://doi.org/10.3978/j.issn.2072-1439.2015.07.17.
sensus on the proper technique, especially in obese and 9. Extracorporeal Life Support Organization. ECLS Registry Report.
edematous patients,19 and it cannot be used to diagnose January 2018. https://www.elso.org/Registry/Statistics.aspx. Accessed
September 25, 2017.
nutrition status at this time.
10. Kon ZN, Dahi S, Evans CF, et al. Class III obesity is not a contraindi-
cation to venovenous extracorporeal membrane oxygenation support.
Conclusion Ann Thorac Surg. 2015;100(5):1855-1860.
11. Schmidt M, Zogheib E, Rozé H, et al. The PRESERVE mortality
Hypocaloric, high-protein EN support was adequate to sup- risk score and analysis of long-term outcomes after extracorporeal
port this critically ill adult male with PWS while he required membrane oxygenation for severe acute respiratory distress syndrome.
ECMO and CRRT. Employing this feeding strategy for Intensive Care Med. 2013;39(10):1704-1713.
the nutrition management of this patient did not result in 12. McClave SA, Taylor BE, Martindale RG, et al. Guidelines for
the provision and assessment of nutrition support therapy in
the development of malnutrition during his hospital stay
the adult critically ill patient: Society of Critical Care Medicine
based on serial monitoring of weight and nutrition-focused (SCCM) and American Society of Parenteral and Enteral Nutri-
physical examination. More studies are needed to investi- tion (A.S.P.E.N). JPEN J Parenter Enteral Nutr. 2016;40(2):159-211.
gate the optimal nutrition management and monitoring of https://doi.org/10.1177/0148607115621863.
critically ill patients with PWS, especially those who require 13. Bear DE, Haslam J, Camporota L, Shankar-Hari M, Barrett NA. An
international survey of nutrition practices in adult patients receiving
life support technologies such as CRRT and ECMO.
veno-venous ECMO. Intensive Care Med. Exp. 2015;3(suppl 1):A295.
https://doi.org/10.1186/2197-425X-3-S1-A295.
Statement of Authorship 14. Nandwani V, Tauber A, McCarthy P, Herr D. 333. Nutrition practice
S Pelekhaty and J Menaker equally contributed to the con- patterns in adult ECMO patients: results of an international survey.
ception and design of the research; S Pelekhaty drafted the Crit Care Med. 2014;42(12 suppl 1):A1440. https://doi.org/10.1097/
manuscript. All authors critically revised the manuscript, agree 01.ccm.0000457830.75362.ed.
15. Wollersheim T, Frank S, Müller MC, et al. Measuring energy ex-
to be fully accountable for ensuring the integrity and accuracy
penditure in extracorporeal lung support patients (MEEP)—protocol,
of the work, and read and approved the final manuscript.
feasibility and pilot trial. Clin Nutr. 2018;37(1):301-307. https://doi.org/
16. De Waele A, van Zwam K, Mattens S, et al. Measuring resting
References energy expenditure during extracorporeal membrane oxygenation:
1. Cassidy SB, Schwartz S, Miller JL, Driscoll DJ. Prader-Willi syndrome. preliminary clinical experience with a proposed theoretical model.
Genet Med. 2012;14(1):10-26. https://doi.org/10.1038/gim.0b013e Acta Anaesthesiol Scand. 2015;59(10):1296-1302. https://doi.org/
31822bead0. 10.1111/aas.12564.
2. Lazzer S, Grugni S, Tringali G, Srtoria A. Prediction of basal 17. Li X, Yu X, Cheypesh A, Li J. Non-invasive measurements of en-
metabolic rate in patients with Prader-Willi syndrome. Eur J Clin Nutr. ergy expenditure and respiratory quotient by respiratory mass spec-
2016;70:494-498. trometry in children on extracorporeal membrane oxygenation—a
3. Lacroix D, Moutel S, Coupaye M, et al. Metabolic and adipose pilot study. Artif. Organs. 2015;39:815-819. https://doi.org/10.1111/
tissue signatures in adults with Prader Willi syndrome: a model aor.12465
of extreme adiposity. J Clin Endocrinol Metab. 2015;100(3):850-859. 18. Stevenson DA, Anaya TM, Clayton-Smith J, et al. Unexpected death
https://doi.org/10.1210/jc.2014-3127. and critical illness in Prader-Willi syndrome: report of ten indi-
4. Bedogni G, Grugni G, Nobili V, Agosti F, Saezza A, Sartorio viduals. Am J Med Genet A. 2004;124A(2):158-164. https://doi.org/
A. Is non-alcoholic fatty liver disease less frequent among women 10.1002/ajmg.a.20370.
with Prader-Willi syndrome? Obes Facts. 2014;7:71-76. https://doi.org/ 19. Earthman CP. Body composition tool for assessment of adult mal-
10.1159/000358570. nutrition at the bedside: a tutorial on research considerations and
5. Fintini D, Inzaghi E, Colajacomo M, et al. Non-alcoholic fatty clinical application. JPEN J Parenter Enteral Nutr. 2015;39(7):787-822.
liver disease (NAFLD) in children and adolescents with Prader-Willi https://doi.org/10.1177/0148607115595227.
syndrome (PWS). Pediatr Obes. 2016;11(3):235-238. https://doi.org/
10.1111/ijpo.12052.
Clinical Observations

Nutrition in Clinical Practice


Volume 33 Number 6
Stoned—A Syndrome of D-Lactic Acidosis and Urolithiasis December 2018 897–901

C 2018 American Society for

Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10063
wileyonlinelibrary.com
Casey M. Berman, MD1 ; and Russell J. Merritt, MD, PhD2

Abstract
Short bowel syndrome (SBS) can lead to many complications related to the condition and its therapy. We describe 2 children with
SBS who we believe are the second and third patients documented to have experienced both D-lactic acidosis and urolithiasis. We
review aspects of these SBS complications and recent findings on the microbiome of patients with SBS that may predispose to these
complications. (Nutr Clin Pract. 2018;33:897–901)

Keywords
D-lactate; D-lactic acidosis; dysbiosis; hypocitraturia; hyperoxaluria; lactobacillus; pediatrics; probiotics; short bowel syndrome;
small bowel bacterial overgrowth; urolithiasis

Introduction admission. He was not taking a proton-pump inhibitor or


other medications aside from vitamins and iron.
Short bowel syndrome (SBS) usually results from surgical He presented to the hospital with worsening diarrhea,
resection of a large segment of the small intestine as a low-grade fever, and altered mental status. One day prior
result of congenital or acquired diseases.1 Common causes to admission he became very unbalanced, falling on his side
of SBS in children include necrotizing enterocolitis, midgut while trying to crawl. He was also sleeping more than usual
volvulus, intestinal atresias, and abdominal wall defects.2 and disinterested in playing. He was febrile to 100.8°F at
The incidence of SBS is estimated to be 24.5 per 100,000 home prior to admission. On admission, he was afebrile
live births.2 SBS may lead to serious common seque- and all vital signs were within normal limits. On exam,
lae, including dehydration, electrolyte imbalance, malnutri- he was sleeping but arousable. No focal neurologic deficits
tion, chronic diarrhea, small intestinal bacterial overgrowth were found. His laboratory assessment was remarkable
(SIBO), central line associated bloodstream infections, in- for metabolic acidosis with a total carbon dioxide (CO2)
testinal failure–associated liver disease, and gallstones.1,3 of 8 mMol/L (reference range 22–30 mMol/L) and an
Although these conditions are well described, other serious, anion gap of 22 (reference range 5–15) without elevation of
but less common, complications may also arise from SBS. In blood urea nitrogen (BUN) or creatinine. C-reactive protein
this report, we describe 2 SBS patients who developed both was elevated at 2.6 mg/d (reference range 0.0–0.9 mg/dL).
D-lactic acidosis and urolithiasis. Consent was obtained for Lactate was within normal limits at 15.0 mg/dL (reference
publication of these case reports per local investigational range 6–16 mg/dL). D-lactate was sent due to concern
review board guidelines. for D-lactic acidosis. He was started on broad-spectrum
intravenous (IV) antibiotics for the history of fever with
Case 1 a central venous catheter. The D-lactate sample drawn on
The first patient is a boy who first presented with D-lactic
acidosis at the age of 16 months with a weight of 9.8 kg. He From the 1 New York Presbyterian–Columbia University, New York,
was born prematurely at 33 weeks’ gestation and had SBS New York, USA; and 2 Children’s Hospital Los Angeles, Keck School
as a result of in-utero midgut volvulus with bowel necrosis. of Medicine, University of Southern California, Los Angeles,
Following surgical intervention, he had 5 cm of duodenum California, USA.
remaining with a duodeno-colonic anastomosis. Prior to Financial disclosure: None declared.
presentation with D-lactic acidosis, he was maintaining Conflicts of interests: None declared.
an oral intake of 950 kcal of 26 kcal/oz elemental infant This article originally appeared online on March 23, 2018.
formula/d in addition to 300 ml of parenteral nutrition (PN)
Corresponding Author:
composed of 20% dextrose and 4% amino acids. The high Russell J. Merritt, MD, PhD, Children’s Hospital Los Angeles, 4650
total intake reflected his very limited enteral absorption, but Sunset Boulevard, MS #78, Los Angeles, CA 90027, USA.
good appetite. He had missed 2 nights of PN just prior to Email: rmerritt@chla.usc.edu
898 Nutrition in Clinical Practice 33(6)

admission resulted several days later and was elevated at prim/sulfamethoxazole, alternating at 2-week intervals, for
10.43 mMol/L (reference range 0–0.25 mMol/L), consistent SIBO prophylaxis.
with a diagnosis of D-lactic acidosis. Blood cultures grew She presented with acute-onset of slurred speech, bal-
Staphylococcus epidermidis, and he continued a 14-day ance disturbance, and vertigo. On arrival, she was afebrile
course of IV vancomycin. and her vital signs were within normal limits. Physical exam
Following admission, he was started on IV fluids con- was significant for left facial paralysis, dysarthria, and right-
taining acetate, and his feeds were held to decrease substrate sided dysmetria. She was acidotic with a total CO2 of
for intestinal D-lactate production. He was given neomycin 14 mMol/L and anion gap of 18. A serum specimen for
for presumed SIBO. A contrast upper gastrointestinal series D-lactate level was sent. The neurology service was con-
with follow through did not show bowel dilation or obstruc- sulted in the emergency room for evaluation for a possible
tion. By day 2 of hospitalization, his mental status returned cerebral vascular event. Computerized tomographic scan
to baseline. He restarted oral feeds on day 4 of hospitaliza- of the head showed no abnormalities. Magnetic resonance
tion and was discharged home on day 5 to complete a 2- imaging showed subtle areas of T2 hyperintensity and
week course of neomycin, on his prior home feeding, and restricted diffusion involving the right cerebellar hemisphere
PN. Metronidazole and trimethoprim/sulfamethoxazole, and the right hippocampus. Given her presentation and
alternating at 2-week intervals, were restarted following the suspicion of D-lactic acidosis, she was admitted to
discontinuation of neomycin. the hospital, and IV bicarbonate, oral neomycin, and
At 4 years of age, he presented to clinic with new onset a carbohydrate-free clear liquid diet were started. The
dysuria, lethargy, and loose stools and was admitted to the d-lactate value returned as 1.58 mMol/L. The patient was
hospital. He was afebrile and vital signs were within normal discharged 4 days after admission to complete a 2-week
limits. He was alert and oriented on physical exam, with course of neomycin and was advised to stay on a low
no significant findings. Urinalysis showed oxalate crystals simple-sugar diet. After finishing the course of neomycin,
and hematuria without signs of infection. Chemistry was she began a prophylactic regimen of alternating neomycin
notable for a total CO2 of 9 mMol/L, with an anion gap and metronidazole.
of 19. The D-lactate drawn on admission was later reported One year later, at age 16, she presented with sudden-
as 2.96 mMol/L. Renal ultrasound showed a 1.5-cm cal- onset right flank pain and on computerized tomographic
culus in the bladder with mild right-sided hydronephrosis. scan was found to have a 5 mm calculus at the right
A vitamin D concentration obtained 1 month later was ureterovesicular junction with mild hydronephrosis as well
32 ng/mL (reference range 20–50 ng/mL). He was started on as multiple 1–3 mm nonobstructive renal calculi bilaterally.
oral neomycin. On hospital day 8 lithotripsy was performed. A temporary stent was placed to relieve symptoms. Several
Stone analysis was not completed. He was discharged home weeks later, she underwent ureteroscopy with ureteral stent
and a repeat renal ultrasound 2 months later showed no removal and right retrograde pyelogram, and no persistent
stones. He has not developed gallstones. stones were identified. Calcifications from the stent yielded
At the age of 5 years, while on his prophylactic antibiotic 80% calcium oxalate monohydrate and 20% calcium oxalate
regimen, he experienced another episode of apparent D- dihydrate. Five months later she had a similar episode
lactic acidosis, but the diagnosis was not documented with of a left-obstructing urinary stone, which passed without
laboratory testing. Upon discharge, his cyclic prophylactic intervention. A 24-hour urine stone profile was remarkable
antibiotic regimen was changed to include neomycin, and for elevated oxalate excretion of 47 mg/d (normal 20–
he has not experienced a recurrence. 40 mg/d), supersaturation of calcium oxalate, and remark-
ably low urinary excretion of citrate of 55 mg/d (normal
>550 mg/d)—all predisposing factors to stone formation.
The urine pH was 5.6. A vitamin D concentration ob-
Case 2 tained about 3 months later was 45 ng/mL. She was in-
The second patient is an adolescent girl admitted to the structed to drink at least 2.5 liters of fluids/d and started
hospital at 15 years of age with a history of SBS sec- on 1200 mg/d of calcium carbonate and on potassium-
ondary to major bowel resection at age 13. She experienced citrate supplementation. In the weeks after starting this
bowel necrosis related to a congenital mesenteric defect regimen, she reported passing several small stones, but
and a complex internal hernia. She was left with 56 cm of since then has not had recurrent stones. On repeat 24-
small bowel and a jejuno-cecal anastomosis. She underwent hour urine stone analysis 4 months after initiation of
elective cholecystectomy for symptomatic cholelithiasis 7 potassium citrate and calcium carbonate supplementation,
months later. She was partially dependent on PN for 1 year urinary citrate increased to 468 mg/d, but oxalate excretion
following bowel resection. At the time of D-lactic acidosis remained elevated at 117 mg/d. She has subsequently done
presentation, she was on a reduced oxalate oral diet, a well on a low oxalate diet with calcium and potassium
proton pump inhibitor, and metronidazole and trimetho- citrate supplementation and a low simple-sugar diet with
Berman and Merritt 899

prophylactic alternating neomycin and metronidazole at 1- Table 1. Clinical Features Seen With D-Lactic Acidosis.
week intervals.
Clinical settings
Short bowel syndrome with colon in continuity
Discussion Malabsorption syndromes
Bariatric intestinal bypass surgery
To our knowledge, only 1 prior patient with SBS has been Neurologic manifestations
reported with both D-lactic acidosis and urolithiasis.4 We Altered behavior
believe these findings may be interrelated because of the Somnolence
increased intestinal bacteria lactate production and acetate Drunk appearing
Aggressive
catabolism in SBS dysbiosis and the effect of acidosis
Confused/disoriented/delirious
on renal citrate excretion as described in the following Neurologic examination findings
paragraphs. Gait disturbance/ataxia/clumsiness
Plasma D-lactate of >3 mMol/L is often used to make Weakness
the diagnosis of D-lactic acidosis.5 However, there is not Dysarthria
a good correlation between D-lactate concentration and Ptosis
symptom severity.6,7 SBS puts patients at increased risk for Nystagmus
Blurred vision
harboring large numbers of D-lactate, producing intestinal
Supportive laboratory findings
bacteria related to their reduced carbohydrate absorption, Metabolic acidosis
absence of an ileocecal valve, and SIBO with increased Elevated anion gap
fermentation of lumenal carbohydrates.5,8,9 Organic acid Negative urine ketones
production may exceed the buffering of pancreatic bicar- Normal L-lactate
bonate secretion.8,10 Lactate-producing bacteria, including
Lactobacillus fermenti, Lactobacillus acidophilus, and Lac-
tobacillus mucosae, thrive in a more acidic environment and Table 2. Management of D-Lactic Acidosis in Short Bowel
quickly become the predominant bacteria of the intestinal Syndrome.
flora of patients with SBS.5,8-12 The bacteria present in SBS
can produce large amounts of D-lactate.9 This dysbiosis Acute therapy
is also characterized by deficiencies of Bacteroides and the Intravenous base (acetate or bicarbonate)
Fasting or enteral carbohydrate restriction
Firmicutes, including Clostridia.11,13 Enteral antibiotics (neomycin may be a preferred drug)
The presentation of D-lactic acidosis resembles alcohol Chronic prevention of recurrences
intoxication (see Table 1 for list of common symptoms). Simple carbohydrate restriction as feasible
Encephalopathy appears to be universal on presentation.14 Cyclic antibiotic therapy; neomycin may be especially
Slurred speech is the most common symptom (52% of pa- helpful
tients) observed followed by ataxia (32%), gait disturbance Consider probiotics (not lactobacillus or D-lactate
(29%), weakness (16%), aggressive behavior (6%), feeling producing organisms)
Recalcitrant cases
drunk (3%), and blurry vision (3%).14 The etiology of the Consider:
central nervous system symptoms appears to be multifac- Surgical therapy of underlying anatomy facilitating small
torial. D-lactate is neurotoxic,15 and brain tissue lacks D- intestinal bacterial overgrowth
2-hydroxyacid dehydrogenase.15 Acidosis inhibits pyruvate Fecal transplantation
decarboxylation, which in turn reduces acetyl Coenzyme
A and adenosine triphosphate production and decreases
neurotransmitter production.15 The cerebellum, with less
pyruvate dehydrogenase, is more vulnerable to this effect.15 Although dietary modification alone may be effective,
Other potentially neurotoxic substances are also produced antibiotic treatment is commonly implemented. Neomycin
by enteric bacteria.16 Comorbid nutrition deficiencies in was used successfully in the first documented case of
SBS such as thiamin deficiency may also contribute to the D-lactic acidosis.21 Another case report of a 12-year-old
symptoms.14,17,18 boy with SBS and D-lactic acidosis supports the utility of
Treatment of D-lactic acidosis is described in Table 2 a combination of neomycin with transient withholding of
and begins with IV correction of academia.5,8 Withholding feedings15 as was done in our patients.
of enteral carbohydrates is helpful in acute management Probiotic administration for the treatment of D-lactic
to reduce D-lactate production.8,19 A 9-year-old girl was acidosis is controversial, as these patients already have
managed with dietary polymeric carbohydrate and an exclu- presumed SIBO.22 Some probiotics can colonize the in-
sion of simple sugars with resolution of recurrent D-lactic testine with flora incapable of D-lactate production. A 3-
acidosis.20 year-old boy with SBS and chronic symptoms of D-lactic
900 Nutrition in Clinical Practice 33(6)

acidosis responded to probiotics that changed his fecal Table 3. Management of Short Bowel Syndrome–Associated
flora from predominantly gram-positive to predominantly Oxalate Urolithiasis.
gram-negative with a nearly complete elimination of uri-
Interventions that may be helpful after acute urologic
nary D-lactate excretion.23 Fecal transplantation has also management of stone(s):
been described as successful therapy for recurrent D-lactic Analysis of stone composition
acidosis.24 Quantification of urinary calcium, magnesium, oxalate, and
Long-term therapy to prevent recurrences includes sim- citrate
ple carbohydrate restriction, which may prove challeng- Assure good hydration
ing in a young child. Prophylactic antibiotics to pre- Diet rich in fruits and vegetables, as feasible
Low oxalate diet
vent bacterial overgrowth appear to be helpful. Of inter-
Oral calcium supplementation
est, both of our patients presented with D-lactic acido- Oral potassium citrate supplementation to normalize
sis while they were taking alternating metronidazole and urinary citrate
trimethoprim/sulfamethoxazole prophylaxis. Neither had a Oral magnesium supplementation, as tolerated
recurrence after neomycin was included in their rotating
antibiotic regimens. Incompletely evaluated intervention
Both of our patients with D-lactic acidosis experienced Probiotic therapy with Oxalobacter formigenes or other
oxalate degrading probiotic organism(s)
urolithiasis. Normally, oxalate is bound by calcium in the
intestine, leading to the fecal excretion of calcium oxalate.4
Fat malabsorption in SBS patients leads to excess lumenal
vegetables provide dietary potassium, and low fruit intake
fatty acids and formation of calcium-fatty acid soaps.25
was found in the hypocitraturic group. Addition of fruits
Unbound oxalate is absorbed, and intestinal oxalate perme-
and vegetables to adult nephrolithiasis patients with hypoci-
ability appears to be increased in SBS due to toxic mucosal
traturia significantly increased urinary citrate.33 In addition,
effects of malabsorbed free fatty acids and deconjugated bile
potassium chloride supplementation led to significant in-
salts.4,25 The resultant hyperoxaluria can lead to urinary cal-
creases in citrate excretion,34 and magnesium salts increase
cium oxalate stones. In a general U.S. pediatric population,
urinary pH, which enhances the excretion of citrate.
renal stones were present in about 14.8 per 100,000 children
Intestinal dysbiosis found in SBS may contribute to
in 2003–2008.26 Although data specific to pediatric SBS pa-
hypocitraturia. Some bacteria metabolize citrate.35 A study
tients do not appear to exist, an adult cohort study reported
comparing the microbiome of SBS patients vs healthy con-
that at a mean of 5 years of follow-up, nephrolithiasis was
trols found that the citrate cycle was amplified in the bacteria
present in 24% of SBS patients with a colon.27
of the SBS patients.13 Some lactic acid bacteria metabolize
In children with calcium oxalate stones, hyperoxaluria,
citrate.36-40 It is likely that upregulation of citrate catabolism
hypercalciuria, and hypocitraturia are common4 and were
depletes intestinal citrate and may lead to reduced urinary
documented in our female patient. A low oxalate diet, potas-
citrate.
sium citrate supplementation, and calcium supplementation
Hypocitraturia has also been attributed to acidosis in re-
are beneficial. Calcium therapy for hyperoxaluria deceases
nal tubular acidosis. Intracellular acidosis leads to decreased
oxalate absorption linearly with increasing dietary calcium
urinary citrate excretion.32 This may occur with D-lactate
dose of up to 1200 mg/d.28
acidosis and offers a possible mechanism by which D-lactic
Hyperoxaluria in SBS may be related to intestinal dys-
aciduria may predispose to urolithiasis. Interestingly, when
biosis. Intestinal oxalate is degraded by bacterial enzymes,
our first patient presented with urolithiasis, he had D-lactic
not host enzymes.29 Oxalobacter formigenes, with enzymes
acidosis. Management of calcium oxalate stones in patients
for oxalate degradation, is absent in patients with cal-
with SBS is described in Table 3.
cium oxalate nephrolithiasis.29 O. formigenes is sensitive
to various antibiotics and may be reduced by persistent
antibiotic use, as given to many SBS patients. Urinary
Conclusions
citrate prevents the formation of calcium oxalate crystals We identified 2 pediatric SBS patients who experienced both
and stones. Hypocitraturia occurs in 19%–68% of patients symptomatic D-lactic acidosis and urolithiasis. D-lactic
with urolithiasis.30,31 acidosis should be considered in SBS patients who present
Diet is a factor in hypocitraturia, including low consump- with central nervous system symptoms and hematuria
tion of magnesium and potassium.32 In 63 children with and flank pain suggest possible urolithiasis. Based on the
urolithiasis, daily intake of magnesium and potassium and literature related to both of these SBS complications, we
urinary concentrations of magnesium and potassium were speculate that the dysbiosis in a subset of SBS patients, likely
lower in patients with hypocitraturia when compared with characterized in part by increased lactobacilli, may put
those who had normal urinary citrate.32 Most individuals them at risk for both of these complications via increased
with hypomagnesuria exhibit hypocitraturia.30,32 Fruits and D-lactate production and increased citrate catabolism.
Berman and Merritt 901

Statement of Authorship 20. Mayne AJ, Handy DJ, Preece MA, George RH, Booth IW. Dietary
management of D-lactic acidosis in short bowel syndrome. Arch Dis
RJM contributed to the conception and design of the research.
Child. 1990;65(2):229-231.
CMB and RJM contributed to the acquisition and analysis of 21. Oh MS, Phelps KR, Traube M, Barbosa-Saldivar JL, Boxhill C, Carroll
the data. RJM and CMB contributed to the interpretation of HJ. D-Lactic acidosis in a man with the short-bowel syndrome. N Engl
the data and CMB drafted of the manuscript. Both authors J Med. 1979;301(5):249-252.
critically revised the manuscript, agree to be fully accountable 22. Munakata S, Arakawa C, Kohira R, Fujita Y, Fuchigami T,
for ensuring the integrity and accuracy of the wok and read and Mugishima H. A case of D-lactic acid encephalopathy associated with
approved the final manuscript. use of probiotics. Brain Dev. 2010;32(8):691-694.
23. Schoorel EP, Giesberts MA, Blom W, van Gelderen HH. D-lactic
References acidosis in a boy with short bowel syndrome. Arch Dis Child.
1980;55(10):810-812.
1. Diamond IR, de Silva N, Pencharz PB, Kim JH, Wales PW. Neonatal 24. Davidovics ZH, Vance K, Etienne N, Hyams JS. Fecal transplantation
short bowel syndrome outcomes after the establishment of the first successfully treats recurrent d-lactic acidosis in a child with short bowel
Canadian multidisciplinary intestinal rehabilitation program: prelim- syndrome. JPEN J Parenter Enteral Nutr. 2017;41(5):896-897.
inary experience. J Pediatr Surg. 2007;42(5):806-811. 25. Bhasin B, Urekli HM, Atta MG. Primary and secondary hyperox-
2. Wales PW, de Silva N, Kim J, Lecce L, To T, Moore A. Neonatal aluria: understanding the enigma. World J Nephrol. 2015;4(2):235-244.
short bowel syndrome: population-based estimates of incidence and 26. Dwyer ME, Krambeck AE, Bergstralh EJ, Milliner DS, Lieske JC, Rule
mortality rates. J Pediatr Surg. 2004;39(5):690-695. AD. Temporal trends in incidence of kidney stones among children: a
3. Vanderhoof JA, Langnas AN. Short-bowel syndrome in children and 25-year population based study. J Urology. 2012;188:247-252.
adults. Gastroenterology. 1997;113(5):1767-1778. 27. Nightingale MD, Lennard-Jones JE, Gertner DJ, Wood SR, Bartram
4. Rahman N, Hitchcock R. Case report of paediatric oxalate urolithiasis CI. Colonic preservation reduces need for parenteral therapy, increases
and a review of enteric hyperoxaluria. J Pediatr Urol. 2010;6(2):112- incidence of renal stones, but does not change high prevalence of gall
116. stones in patients with a short bowel. Gut. 1992;33:1493-1497.
5. Fabian E, Kramer L, Siebert F, et al. D-lactic acidosis—case report and 28. Holmes RP, Knight J, Assimos DG. Lowering urinary oxalate
review of the literature. Z Gastroenterol. 2017;55(1):75-82. excretion to decrease calcium oxalate stone disease. Urolithiasis.
6. De Vrese M, Koppenhoefer B, Barth CA. D-lactic acid metabolism 2016;44(1):27-32.
after an oral load of DL-lactate. Clin Nutr. 1990;9:23-28. 29. Vaidyanathan S, von Unruh GE, Watson ID, Laube N, Willens S,
7. Thurn JR, Pierpont GL, Ludvigsen CW, Eckfeldt JH. D-lactate Soni BL. Hyperoxaluria, hypocitraturia, hypomagnesiuria, and lack
encephalopathy. Am J Med. 1985;79(6):717-721. of intestinal colonization by Oxalobacter formigenes in a cervical
8. Bongaerts G, Bakkeren J, Severijnen R, et al. Lactobacilli and acidosis spinal cord injury patient with suprapubic cystostomy, short bowel, and
in children with short small bowel. J Pediatr Gastroenterol Nutr. nephrolithiasis. Sci World J. 2006;6:2403-2410.
2000;30(3):288-293. 30. Kato Y, Yamaguchi S, Yachiku S, et al. Changes in urinary parameters
9. Petersen C. D-lactic acidosis. Nutr Clin Pract. 2005;20(6):634-645. after oral administration of potassium-sodium citrate and magnesium
10. Bongaerts GP, Tolboom JJ, Naber AH, et al. Role of bacteria in the oxide to prevent urolithiasis. Urology. 2004;63(1):7-11.
pathogenesis of short bowel syndrome-associated d-lactic acidemia. 31. Bevill M, Kattula A, Cooper CS, Storm DW. The modern metabolic
Microb Pathog. 1997;22(5):285-293. stone evaluation in children. Urology. 2017;101:15-20.
11. Mayer C, Gratadoux JJ, Bridonneau C, et al. Faecal D/L lactate ratio 32. Kovacevic L, Wolfe-Christensen C, Edwards L, Sadaps M, Laksh-
is a metabolic signature of microbiota imbalance in patients with short manan Y. From hypercalciuria to hypocitraturia—a shifting trend in
bowel syndrome. PLoS One. 2013;8(1):e54335. pediatric urolithiasis? J Urol. 2012;188(4 suppl):1623-1627.
12. Davidovics ZH, Carter BA, Luna RA, Hollister EB, Shulman RJ, 33. Meschi T, Maggiore U, Fiaccadori E, et al. The effect of fruits and
Versalovic J. The fecal microbiome in pediatric patients with short vegetables on urinary stone risk factors. Kidney Int. 2004;66(6):2402-
bowel syndrome. JPEN J Parenter Enteral Nutr. 2016;40(8):1106- 2410.
1113. 34. Domrongkitchaiporn S1, Stitchantrakul W, Kochakarn W. Causes of
13. Piper HG, Fan D, Coughlin LA, et al. Severe gut microbiota dysbiosis hypocitraturia in recurrent calcium stone formers: focusing on urinary
is associated with poor growth in patients with short bowel syndrome. potassium excretion. Am J Kidney Dis. 2006;48(4):546-554.
JPEN J Parenter Enteral Nutr. 2017;41:1202-1212. 35. Edin-Liljegren A, Hedelin HH, Grenabo L, Pettersson S. Impact of
14. Kowlgi NG, Chhabra L. D-lactic acidosis: an underrecognized com- Escherichia coli on urine citrate and urease-induced crystallization.
plication of short bowel syndrome. Gastroenterol Res Pract. 2015: Scanning Microsc. 1995;9(3):901-905.
476215. 36. Ostlie HM, Helland M, Narvhus JA. Growth and metabolism of
15. Zhang DL, Jiang ZW, Jiang J, Cao B, Li JS. D-lactic acidosis sec- selected strains of probiotic bacteria in milk. Int J Food Microbiol.
ondary to short bowel syndrome. Postgrad Med Jl. 2003;79(928):110- 2003;87(1-2):17-27.
112. 37. Palles T, Beresford T, Condon S, Cogan TM. Citrate metabolism in
16. Halperin ML, Kamel KS. D-lactic acidosis: turning sugar into acids in Lactobacillus casei and Lactobacillus plantarum. J Appl Microbiol.
the gastrointestinal tract. Kidney Intl. 1996;49(1):1-8. 1998;85(1):147-154.
17. Hudson M, Pocknee R, Mowat NAG. D-Lactic acidosis in short 38. Hugenholtz J. Citrate metabolism in lactic acid bacteria. FEMS
bowel syndrome—an examination of possible mechanisms. Q J Med. Microbiol Rev.1993;12(1-3):165-178.
1990;74(274):157-163. 39. Diaz-Muniz I, Banavara DS, Budinich MF, Rankin SA, Dudley EG,
18. Puwanant M, Mo-Suwan L, Patrapinyokul S. Recurrent d-lactic aci- Steele JL. Lactobacillus casei metabolic potential to utilize citrate as
dosis in a child with short bowel syndrome. Asia Pac J Clin Nutr. an energy source in ripening cheese: a bioinformatics approach. J Appl
2015;14(2):195-198. Microbiol. 2006;101(4):872-882.
19. Perlmutter DH, Boyle JT, Campos JM, Egler JM, Watkins JB. D- 40. Mortera, P, Pudlik A, Magni C, Alarcon S, Lolkema JS. Ca2+-citrate
Lactic acidosis in children: an unusual metabolic complication of small uptake and metabolism in lactobacillus casei ATCC 334. Appl Enviorn
bowel resection. J Pediatr. 1983;102(2):234-238. Microbiol. 2013;79(15):4603-4612.
Clinical Observations

Nutrition in Clinical Practice


Volume 33 Number 6
Avoidance of Overt Precipitation and Patient Harm December 2018 902–905

C 2017 American Society for

Following Errant Y-Site Administration of Calcium Chloride Parenteral and Enteral Nutrition
DOI: 10.1177/0884533617723865
and Parenteral Nutrition Compounded With wileyonlinelibrary.com
Sodium Glycerophosphate

Collin Anderson, PharmD, PhD, BCPS, BCPPS; Chanelle Stidham, PharmD, BCPS;
Sabrina Boehme, PharmD, BCPS, BCPPS; and Jared Cash, PharmD, BCPS

Abstract
Calcium phosphate precipitates present 1 of many challenges associated with parenteral nutrition (PN) compounding. Extensive
research has led to the establishment of solubility curves to guide practitioners in the prescription and preparation of stable PN.
Concurrent dosing of intravenous products via y-site administration with PN can alter the chemical balance of the solution
and modify solubility. Medications containing calcium or phosphate should not be administered in the same line as PN, due
to the high potential for precipitation. Herein a case is reported from a pediatric cardiac intensive care unit where a physician
ordered the administration of calcium chloride. The bedside nurse added the calcium chloride intermittent infusion as a y-site
administration with the patient’s PN. The patient’s PN had been compounded with sodium glycerophosphate, temporarily available
in the United States during a sodium phosphate shortage. The patient did not experience any observable adverse effects from the
y-site administration with PN. Following this event, the scenario was replicated to investigate any precipitation risk associated with
the y-site administration. Additionally, a separate PN solution containing sodium phosphate rather than glycerophosphate was
compounded and used in a laboratory setting to demonstrate the potential for harm had the patient’s PN been compounded with an
inorganic phosphate source. This replication of the error demonstrates the additional safety gained in relation to precipitation risk
when PN solutions are compounded with sodium glycerophosphate in place of sodium phosphate. (Nutr Clin Pract. 2018;33:902–
905)

Keywords
drug compounding; food-drug interactions; nutritional support; parenteral nutrition

Calcium phosphate precipitation in compounded parenteral are calcium and phosphate concentrations, form of calcium
nutrition (PN) products has been well documented and utilized, amino acid product and concentration, pH, and
researched. Deaths attributed to calcium phosphate pre- temperature.3 Other precipitates, such as zinc phosphate,
cipitates administered within the context of PN led to a can occur in PN. In neonatal and pediatric patient pop-
U.S. Food and Drug Administration safety alert and are ulations, practitioners can be limited in their ability to
referenced in early recommendations by a national advi- provide adequate amounts of calcium and phosphorus due
sory group regarding safe practices for PN formulations.1,2 to volume constraints in the setting of increased mineral
Extensive research established solubility curves for varying needs. Generally, significant time and resources are devoted
PN compositions. Due to their everyday utility, these graphs to ensuring that PN preparations are safe and do not pose a
were compiled in tertiary references and handbooks for easy
access to practitioners involved in prescribing, preparing, From Intermountain Healthcare Primary Children’s Hospital, Salt
and administering PN. Pharmacists initially referred to Lake City, Utah, USA.
printed graphs for each PN prescription to determine if Financial disclosure: None declared.
the formulation would likely precipitate. More recently, Conflicts of interest: None declared.
compounding software has incorporated those graphs into
This article originally appeared online on September 27, 2017.
platforms that automatically suggest when a proposed PN
product may lead to precipitation. Immediate warnings Corresponding Author:
Collin Anderson, PharmD, PhD, BCPS, BCPPS, Pharmacy,
are provided to the compounding personnel, allowing for
Intermountain Healthcare Primary Children’s Hospital, 100 N Mario
adjustments in the formulation prior to production. Many Capecchi Dr, Salt Lake City, UT 84113, USA.
factors contribute to PN solubility. Primary among these Email: collin.anderson@imail.org
Anderson et al 903

for 90 mg of IV calcium chloride, which was obtained


from an automated dispensing cabinet and administered
via y-site administration with PN downstream from the
filter. After administration, during nurse hand-off at shift
change, the oncoming nurse immediately recognized the
error that occurred in administering calcium with PN and
contacted the unit clinical pharmacist. Bedside practitioners
did not observe any adverse effects surrounding the y-site
Figure 1. Sodium glycerophosphate. administration of the calcium chloride and PN. It was noted
that the patient’s PN contained sodium glycerophosphate as
its sole phosphate source.
significant risk for forming calcium phosphate precipitates.
Y-site administration of any other solutions containing
calcium or phosphate with PN should be strictly avoided,
Method
due to the extreme concentrations encountered during such The situation was re-created to study the difference in y-
administration. It is important to remember that in addition site administration of calcium chloride with the patient’s PN
to standard mineral intravenous (IV) replacement products, formulation vs a similar PN containing inorganic phosphate
other medications may contain phosphate in their formula- in place of sodium glycerophosphate. The original PN con-
tion (eg, dexamethasone sodium phosphate). tained 49.8 mEq/L of calcium gluconate and 21.5 mmol/L
Sodium glycerophosphate (Figure 1) is an organically of sodium glycerophosphate, which is within published
bound phosphate that has a significantly improved calcium solubility limits for these agents. This PN was compounded
phosphate solubility profile in comparison with inorganic again to re-create the y-site administration that occurred
phosphate preparations. Phosphate is bound to glycerol in the intensive care unit. An identical PN containing
within the organic phosphate salt. Upon IV infusion, the inorganic phosphate in place of glycerophosphate could
phosphate is readily cleaved from the glycerol component not be compounded at the same concentrations, because it
through an enzymatic process. The pharmacokinetic pro- would exceed the solubility limit. Therefore, a similar in-
files of sodium phosphate and sodium glycerophosphate are organic phosphate-containing PN was compounded within
nearly identical, and the products are both valid options as the solubility curve limit by reducing calcium gluconate
a phosphate source.4 In terms of solubility in the presence to 39.6 mEq/L and sodium phosphate to 17.1 mmol/L
of calcium, sodium glycerophosphate has been shown to (Table 1). With an Alaris infusion pump, each PN was run
be superior. PN solutions containing glycerophosphate are at the ordered rate of 4.8 mL/hr through a CareFusion (San
less likely to precipitate and allow for greater flexibility Diego, CA) 0.2-μm filter infusion set. Downstream from the
in calcium and phosphate dosing. European approval of filter, a bifuse administration set was used to run 90 mg of
the drug first occurred nearly 2 decades ago, and the calcium chloride, 10%, over 30 minutes via a separate pump.
product is readily available in those markets. Due to pre- The contents of the catheter output for both PN solutions
vious inorganic phosphate shortages within the U.S. drug were collected over the time of administration and then im-
distribution system, the Food and Drug Administration al- mediately passed through a 0.8-μm nitrocellulose filter disk.
lowed for temporary importation and utilization of sodium The filter from the y-site infusion of the sodium phosphate–
glycerophosphate. Studies of sodium glycerophosphate and containing PN showed particulate matter presumed to be
calcium were performed following methodology originally calcium phosphate, which was visible to the naked eye.
used to determine PN calcium phosphate solubility curves. The filter from the y-site infusion of the glycerophosphate-
Research has demonstrated a remarkable solubility profile containing PN contained 6 crystals, which were seen upon
for sodium glycerophosphate in solutions with calcium- microscopic evaluation (Figure 2).
containing salts, essentially eliminating concern for precipi-
tation in the range of clinically relevant PN preparations.5,6 Discussion
A sentinel event was averted due to the favorable compatibil-
Situation ity profile of sodium glycerophosphate. Calcium phosphate
A 3.1-kg male was admitted to the cardiac intensive care pulmonary emboli were implicated in the deaths of 2 indi-
unit of our free-standing pediatric institution following viduals in the 1990s. The same publication that reported the
open heart surgery. The patient received PN according to deaths replicated the implicated PN formulation. Infusion
an institutional cardiac nutrition protocol. Blood calcium of the PN in healthy pigs caused death within 4 hours.7
levels were persistently low (ionized calcium <1.2 mmol/L), Simulation of the scenario that occurred at our hospital
requiring correction with IV calcium chloride. Approxi- likewise demonstrates the grave danger associated with
mately 12 hours after PN was started, an order was placed exceeding calcium phosphate solubility limits. Precipitation
904 Nutrition in Clinical Practice 33(6)

Table 1. Original PN Composition With Glycerophosphate Versus Inorganic Phosphate–Containing PN.

Original PN-Containing Glycerophosphate Experimental PN-Containing Sodium Phosphate

Component Weight-Based Dose Concentration Weight-Based Dose Concentration

Dextrose 7.4 g/kg 20% 9.3 g/kg 20%


Trophamine 1.8 g/kg 4.9% 1.8 g/kg 3.9%
Calcium gluconate 1.85 mEq/kg 50 mEq/L 1.85 mEq/kg 40 mEq/L
Sodium glycerophosphate 0.8 mmol/kg 22 mmol/L NA NA
Sodium phosphate NA NA 0.8 mmol/kg 17 mmol/L
Magnesium sulfate 0.4 mEq/kg 10.8 mEq/L 0.4 mEq/kg 8.6 mEq/L
Sodium acetate 0.65 mEq/kg 17 mEq/L 0.85 mEq/kg 18 mEq/L
Sodium chloride 0.65 mEq/kg 17 mEq/L 0.85 mEq/kg 18 mEq/L
Carnitine 10 mg/kg 0.27 g/L 10 mg/kg 0.21 g/L
Total volume 37 mL/kg 115 mL/d 47 mL/kg 145 mL/d

NA, not applicable; PN, parenteral nutrition.

Figure 2. A area of the filter (3 × 3 mm2 ) pictured under 30× magnification. The simulated y-site administration of the sodium
glycerophosphate–containing parenteral nutrition with calcium chloride (left) and the analogous inorganic phosphate–containing
parenteral nutrition with calcium chloride (right).

of the y-site admixture was clearly observable without and calcium chloride in compounded PN have not been
magnification. performed at the replicated y-site concentration reported
Research has demonstrated that within PN formulations, herein, it is not advisable to exceed previously published
sodium glycerophosphate and calcium chloride are solu- limits.
ble at concentrations up to 50 mmol/L and 50 mEq/L, Like other institutions, our hospital has had long-
respectively.5,6 Of note, the concentration of calcium and standing policies, procedures, and education regarding
phosphate in the replicated y-site admixture was calculated the incompatibility surrounding y-site administration of
to be 418 mEq/L of calcium and 15.7 mmol/L of phosphate, calcium and/or phosphate solutions with PN, as recom-
far exceeding concentrations in published solubility studies. mended by the American Society for Parenteral and Enteral
The filtered output from the sodium glycerophosphate– Nutrition.8,9 Electronic and print resources are available to
containing PN and y-site calcium chloride did result in nursing personnel, and pharmacists are regularly consulted
6 crystals that were discovered through microscopic ex- regarding compatibility. In the case reported, human err
amination of the filtered solution. As controlled physical bypassed those safeguards, resulting in potential danger.
compatibility studies involving sodium glycerophosphate Fortunately, the patient’s PN was compounded with sodium
Anderson et al 905

glycerophosphate, and there was no apparent harm to the integrity and accuracy of the work, and read and approved the
patient. The favorable solubility profile of sodium glyc- final manuscript.
erophosphate vs inorganic phosphate leads to a better safety
profile in regard to the formation of harmful precipitates References
in PN. While PN remains a dangerous medication even 1. McKinnon BT. FDA safety alert: hazards of precipitation associated
when compounded with sodium glycerophosphate, the risk with parenteral nutrition. Nutr Clin Pract. 1996;11:59-65.
of calcium phosphate precipitation is dramatically lower 2. Mirtallo J, Driscoll D, Helms R, Kumpf V, McKinnon B. Safe practices
than in standard PN solutions compounded with inorganic for parenteral nutrition formulations. JPEN J Parenter Enteral Nutr.
1998;22:49-66.
phosphate.
3. Newton D, Driscoll D. Calcium and phosphate compatibility: revisited
again. Am J Health Syst Pharm. 2008;65:73-80.
Conclusion 4. Topp H, Hochfeld O, Bark S, et al. Glycerophosphate is interchangeable
with inorganic phosphate in terms of safety and serum pharmacokinet-
When compared with sodium phosphate, sodium glyc- ics. Pharmacology. 2011;88:193-200.
erophosphate minimizes risk of calcium phosphate pre- 5. MacKay M, Anderson C. Physical compatibility of sodium glycerophos-
cipitation within PN. Making sodium glycerophosphate phate and calcium gluconate in pediatric parenteral nutrition solutions.
available within the United States would decrease the pre- JPEN J Parenter Enteral Nutr. 2015;39:725-728.
6. Anderson C, MacKay M. Physical compatibility of calcium chloride
cipitation risks associated with PN. It is recommended
and sodium glycerophosphate in pediatric parenteral nutrition solutions.
that sodium glycerophosphate be used in place of sodium JPEN J Parenter Enteral Nutr. 2016;40:1166-1169.
phosphate in PN. 7. Hill SE, Heldman LS, Goo EDH, Whippo PE, Perkinson JC. Fatal
microvascular pulmonary emboli from precipitation of a total nutrient
Statement of Authorship admixture solution. JPEN J Parenter Enteral Nutr. 1996;20:81-87.
8. Ayers P, Adams S, Boullata J, et al. ASPEN parenteral nutrition
All authors contributed to the conception and design of the
safety consensus recommendations. JPEN J Parenter Enteral Nutr.
research; C. Anderson and C. Stidham contributed to the 2014;38:296-233.
acquisition, analysis, and interpretation of the data; and C. 9. Boullata JI, Gilbert K, Sacks G, et al. ASPEN clinical guidelines:
Anderson drafted the manuscript. All authors critically revised parenteral nutrition ordering, order review, compounding, labeling, and
the manuscript, agree to be fully accountable for ensuring the dispensing. JPEN J Parenter Enteral Nutr. 2014;38:334-377.
Standards of Practice

Nutrition in Clinical Practice


Volume 33 Number 6
Standards for Nutrition Support: Adult Hospitalized Patients December 2018 906–920

C 2018 American Society for

Parenteral and Enteral Nutrition


DOI: 10.1002/ncp.10204
wileyonlinelibrary.com
Andrew Ukleja, MD, AGAF1 ; Karen Gilbert, RN, MSN, CNSC, ACNP2 ;
Kris M. Mogensen, MS, RD-AP, LDN, CNSC3 ; Renee Walker, MS, RD, LD, CNSC,
FAND4 ; Ceressa T. Ward, PharmD, BCPS, BCNSP, BCCCP5 ; Joe Ybarra, PharmD,
BCNSP6 ; Beverly Holcombe, PharmD, BCNSP, FASHP, FASPEN7 ; Task Force
on Standards for Nutrition Support: Adult Hospitalized Patients, the American Society
for Parenteral and Enteral Nutrition

Abstract
The American Society for Parenteral and Enteral Nutrition defines standards as benchmarks representing a range of performance
of competent care that should be provided to assure safe and efficacious nutrition care in most circumstances. Standards
are documents that define the structure needed to provide competent care. These Standards for Nutrition Support for Adult
Hospitalized Patients are an update of the 2010 Standards. These practice-based standards are intended for use by healthcare
professionals charged with the care of adult hospitalized patients receiving nutrition support therapy in any hospital with or
without a formal nutrition support service or team. These Standards address professional responsibilities as they relate to patient
assessment, diagnosis, education, care plan development, implementation, clinical monitoring, evaluation, and professional issues
around nutrition support. (Nutr Clin Pract. 2018;33:906–920)

Keywords
enteral nutrition; hospitalization; nutrition assessment; nutrition support; parenteral nutrition; standard of care

Introduction usually address professional responsibilities as they relate to


patient assessment, diagnosis, education, care plan devel-
The American Society for Parenteral and Enteral Nu- opment, implementation, clinical monitoring, evaluation,
trition (ASPEN) is dedicated to improving patient care and professional issues. ASPEN publishes discipline-based
by advancing the science and practice of clinical nutri- (eg, dietitian, nurse, pharmacist, or physician) and practice-
tion and metabolism. Founded in 1976, ASPEN is an based (eg, adult hospitalized patients, pediatric hospitalized
interdisciplinary organization whose members are involved patients, home and alternate site care) standards. Standards
in the provision of clinical nutrition therapies, including are presented in the most generic terms possible. The details
parenteral and enteral nutrition. With more than 6,500 of specific tests, therapies, and protocols are left to the
members from around the world, ASPEN is a community of
dietitians, nurses, pharmacists, physicians, scientists, stu-
dents, and other health professionals from every facet of From the 1 Beth Israel Deaconess Medical Center, Division of
nutrition support clinical practice, research, and educa- Gastroenterology, Boston, Massachusetts, USA; 2 Thomas Jefferson
tion. ASPEN envisions an environment in which every University Hospital, Philadelphia, Pennsylvania, USA; 3 Department
patient receives safe, efficacious, and high-quality nutrition of Nutrition, Brigham and Women’s Hospital, Boston,
Massachusetts, USA; 4 Michael E. DeBakey Veteran Affairs Medical
care. ASPEN’s mission is to improve patient care by Center, Houston, Texas, USA; 5 Emory Healthcare, Atlanta, Georgia,
advancing the science and practice of clinical nutrition USA; 6 Medical City McKinney, McKinney, Texas, USA; and
and metabolism. These Standards for Nutrition Support 7 American Society for Parenteral and Enteral Nutrition, Silver

for Adult Hospitalized Patients are an update of the 2010 Spring, Maryland, USA.
standards.1 They are intended for use by any hospital with Financial disclosure: None declared.
or without a formal nutrition support service (or team). Conflicts of interest: None declared.
ASPEN defines standards as benchmarks representing This article originally appeared online on October 15, 2018.
a range of performance of competent care that should be
Corresponding Author:
provided to assure safe and efficacious nutrition care in Beverly Holcombe, PharmD, BCNSP, FASHP, FASPEN, ASPEN,
most circumstances.2 Standards are documents that define 8401 Colesville Rd, Suite 510, Silver Spring, MD 20910.
the structure needed to provide competent care. Standards Email: beverlyh@nutritioncare.org
Ukleja et al 907

discretion of individual healthcare facilities. Each health- Chapter I: Organization


care facility shall strive to provide the best nutrition support
care that is possible given the resources of the organization. Standard 1. Nutrition Support Service (or
The standards aim to ensure sound and efficient nutrition Team)
care for those in need of nutrition support therapy.
A nutrition support service (or team) should assess and
in collaboration with patients’ primary teams, manage the
Important Note nutrition support therapy of patients who require or may
These standards do not constitute medical or other pro- require nutrition support therapy. These patients are of-
fessional advice and should not be taken as such. To the ten, but not always, determined to be nutritionally-at-risk
extent that the information published herein may be used at admission or upon subsequent evaluation.3 Organized
to assist in the care of patients, this is the result of the sole nutrition support services (or teams) are associated with
professional judgment of the attending healthcare profes- improved patient outcomes, decreased length of hospital-
sional whose judgment is the primary component of quality ization, and improved cost effectiveness.4-19 If a hospital
medical care. The information presented in these standards does not have a designated nutrition support service (or
is not a substitute for the exercise of such judgment by the team), the care used to provide nutrition support therapy
healthcare professional. Circumstances in clinical settings should be interprofessional. The scope and design of the
and patient indications may require actions different from nutrition support service (or team) and their respective
those recommended in this document and in those cases, the activities vary according to the unique attributes of each
judgment of the treating professional should prevail. hospital. Among various organizations, management of
nutrition support may comprise a spectrum of activities
Audience for Standards including no formal structure, an administrative nutrition
committee only, a consultative nutrition support service (or
These practice-based standards are intended for use by team), or a nutrition support service (or team) that assumes
healthcare professionals charged with the care of adult responsibility for the nutrition care of patients who receive
hospitalized patients receiving nutrition support therapy. nutrition support therapy.

Level of Care 1.1 When an organized nutrition support service (or


team) exists, it shall be directed by a clinician who
As limited by the Important Note above, these Standards
has appropriate education, specialized training, pa-
of Practice present a range of performance of competent
tient care experience, or experience in managing
care that should be provided by healthcare professionals
nutrition support services (teams).
caring for adult hospitalized patients receiving nutrition
1.2 An organized nutrition support service (or team)
support therapy. Terminologies included in each standard
should include a physician, nurse, dietitian, and
are specified as:
pharmacist, each following the standards of prac-
tice for their discipline, as available.20-23
(a) “Shall”: Indicates standards to be followed strictly. 1.3 If a nutrition support service (or team) is not
(b) “Should”: Indicates that among several possibilities established, nutrition support therapy should be
one is particularly suitable, without mentioning or managed with an interprofessional approach that
excluding others, or that a certain course of action includes the patient’s physician, nurse, dietitian,
is preferred but not necessarily required. and pharmacist.
(c) “May”: Indicates a course of action that is permis-
sible within the limits of recommended practice. Standard 2. Policies and Procedures
Written policies and procedures for providing nutrition
These standards have been developed by the ASPEN support therapy shall be current.
Task Force on Standards for Nutrition Support: Adult
Hospitalized Patients, reviewed by the ASPEN Clinical 2.1 The policies and procedures shall be developed with
Practice Committee, and approved by the ASPEN Board the input of and review by all members of the
of Directors on July 25, 2018. These Standards of Prac- nutrition support service (or team) and/or nutrition
tice should be used in conjunction with the previously support committee.
published ASPEN Clinical Guidelines, Standards, Position 2.2 The policies and procedures shall be reviewed
Papers, and other Board Approved documents, which can periodically and revised as appropriate to define
be accessed at the ASPEN Documents Library, http://www. optimal patient care and therapeutic outcomes. (See
nutritioncare.org/Clinical_Practice_Library/. 3.2.)
908 Nutrition in Clinical Practice 33(6)

Standard 3. Performance Improvement baseline nutrition parameters, identify nutrition risk


factors and specific nutrition deficits, determine individual
The nutrition support service (or team) and/or nutrition nutrition needs, and identify medical, psychosocial, and
support committee shall regularly review and report on ser- socioeconomic factors that may influence the prescription
vice performance, quality indicators, patient outcome data, and administration of nutrition support therapy.34,35
and adverse events related to nutrition support therapies.24
These reports shall be shared with all internal stakeholders 5.1 The nutrition assessment shall be performed within
and reported to external agencies as required. the time frame specified by the hospital and by a
dietitian or a clinician with documented specialized
3.1 The nutrition support service (or team) and/or nu- expertise in nutrition.
trition support committee shall recommend policy, 5.2 The nutrition assessment shall include evaluation
procedure, or protocol changes that improve and/or of the patient’s current nutrition status and nutri-
enhance the safety and efficacy of nutrition support tion requirements.
therapy. 5.2.1 A malnutrition diagnosis, if present, and
3.2 The review of service performance should assess degree of malnutrition shall be clearly doc-
the appropriateness and effectiveness of nutrition umented to facilitate appropriate diagnosis
support therapy. coding.
5.2.2 Degree of obesity (ie, class I, class II, or class
Chapter II: Nutrition Care III), if applicable, shall also be documented.
Nutrition care and the administration of nutrition support 5.3 The patient’s nutrition requirements shall be sum-
therapy shall proceed according to a series of steps with marized based on the findings of the nutrition
feedback loops. These steps include nutrition screening, assessment and should include energy, macronutri-
formal nutrition assessment, creation of a nutrition care ent (protein, and as appropriate, carbohydrate and
plan, implementation of the plan, patient monitoring, eval- fat), as well as fluid, electrolyte, and micronutrient
uation of the plan, evaluation of the care setting, and requirements, as appropriate.
reformulation of the plan or termination of therapy. (See 5.4 Nutrition assessment shall include a review and
Figure 1: ASPEN Adult Nutrition Care Pathway.) documentation of factors relevant to delivery of
nutrition support therapy. Relevant factors may
Standard 4. Nutrition Screening include, but are not limited to, the following: ability
to eat safely and adequately, patient’s goals, assess-
Nutrition screening is defined as “a process to identify ment of aspiration risk, functional status of the
an individual who is malnourished or who is at risk for gastrointestinal tract, cognitive function/abilities,
malnutrition to determine if a detailed nutrition assessment enteral and vascular access, and results of tests and
is indicated.”1 Patients who are nutritionally-at-risk shall be invasive procedures.
identified by a validated screening process and by periodic
rescreening per institutional policy or standard.3,25-33 This Chapter III: The Nutrition Care Plan
process should be created, approved, and regularly reviewed
by a group with organizational authority, preferably a Standard 6. Goals
designated nutrition committee.
The process of nutrition care is multifactorial and shall
include multiple levels of intervention including screening
4.1 Results of the nutrition screening shall be docu-
for nutrition risk factors. The nutrition care plan shall
mented and communicated and appropriate inter-
be created from a comprehensive review and analysis of
vention shall be initiated within the time frame
information gathered from many aspects of the patient’s
specified by the hospital or as clinically indicated.
care. The nutrition care plan should include “statements
4.2 A procedure for rescreening of patients not imme-
of nutrition goals and monitoring/evaluation parameters,
diately identified as nutritionally-at-risk should be
the most appropriate route of administration of nutrition
implemented and regularly reviewed.
therapy, method of nutrition access, anticipated duration of
therapy, and training and counseling goals and methods.”2
Standard 5. Nutrition Assessment A formal nutrition assessment provides the basis for
All patients identified as nutritionally-at-risk based the nutrition care plan. The nutrition care plan guides
on the nutrition screening shall undergo a nutrition comprehensive nutrition therapy by defining its ratio-
assessment.3,27-33 This nutrition assessment shall be nale, describing appropriate intervention and monitoring,
documented and made available to all patient care providers. and delineating recommended reassessment and reevalu-
The intent of the nutrition assessment is to document ation parameters. This process facilitates changes in care
Ukleja et al 909

Figure 1. The American Society for Parenteral and Enteral Nutrition adult nutrition care pathway.

appropriate to the clinical setting while considering the Standard 7. Interprofessional Approach
continuum of care. Revision of the nutrition care plan based
on changes in clinical status and achievement of goals of The nutrition care plan should be developed using an inter-
therapy should occur before discontinuation of nutrition professional team approach involving the patient, caregiver
support therapy. (if applicable), the nutrition support service (or team), the
910 Nutrition in Clinical Practice 33(6)

Figure 1. Continued.
Ukleja et al 911

patient’s physician(s), dietitian(s), nurse(s), pharmacist(s), 11.1.2 As delineated by clinical privileges and
and other appropriate healthcare professionals. applicable professional licensure laws, a
nutrition support clinician may enter/write
Standard 8. Patient and Caregiver orders for feeding formulations, laboratory
Communication tests, and adjunctive therapy (eg, intra-
venous [IV] fluids, insulin, IV/oral elec-
The nutrition care plan should include patient and/or care- trolytes) and adjust regimens based on
giver(s) education about nutrition support therapy, goals, response to therapy, changing clinical con-
and expectations and should incorporate the wishes of the dition(s), altered laboratory values, and
patients and/or caregiver(s). Appropriate routes of admin- nutrition assessment parameters.
istration shall be defined, identification of intake goals shall 11.1.3 Hospital policy should include a com-
be included, and estimated duration of therapy as well as petency for nutrition support therapy
criteria for discontinuation of therapy should be addressed. prescribing.39
11.2 Orders for nutrition support therapy shall be
Standard 9. Selection of Route documented in the patient’s medical record before
The route selected to provide nutrition support therapy shall administration.
be appropriate to the patient’s clinical status or condition 11.2.1 A standardized order format and review
and shall periodically be assessed for continued appropri- process for nutrition support therapies
ateness as well as for its adequacy in meeting goals of the orders shall be used to minimize the
nutrition care plan.36 (See Figure 2: Route of administration risk of adverse events and error. This
algorithm.) process shall include standardized elec-
tronic orders (eg, computerized provider
Standard 10. Selection of Formulation order entry [CPOE] system) for prescrib-
ing PN and EN. Handwritten order to
The enteral nutrition (EN) or parenteral nutrition (PN) prescribe PN and EN should be avoided
formulation shall be appropriate for the patient’s disease due to potential for error. Verbal and
process and compatible with the route of access.37,38 telephone orders and text messaging of
orders should be avoided.40,41 See ASPEN
10.1 The EN or PN formulation shall be adjusted as ap- Parenteral Nutrition Safety Consensus
propriate based on the patient’s clinical response. Recommendations39 and ASPEN Safe
10.2 The EN or PN formulation shall be adjusted Practices for Enteral Nutrition Therapy40
accordingly when significant amounts of nutrients for details of the prescribing process for
are provided (eg, parenteral infusions, medica- PN and EN.
tions) through means other than the EN for- 11.3 Nutrition care plans shall be implemented to
mula or PN admixture or lost/eliminated through promote safe, accurate, and effective nutrition sup-
mechanical procedures or anatomical defects port therapy based on the patient’s needs and clin-
(eg, renal replacement therapy, enterocutaneous ical condition and will provide resource-efficient
fistula) . and fiscally responsible care.

Chapter IV: Implementation


Standard 11. Ordering Process Standard 12. Nutrition Support Access
Access for nutrition support therapy shall be achieved and
Implementation of the nutrition care plan shall follow
maintained in a manner that minimizes risk to the patient
nutrition assessment and development of a formal nutrition
and optimizes therapeutic outcome(s).36,40-43
care plan.

11.1 Authority to prescribe nutrition support therapy 12.1 Standard techniques and policies should be estab-
shall be determined by hospital policy and appli- lished and followed for access device insertion and
cable professional licensure laws. routine care. (See section 16.5.)
11.1.1 Hospital policy should clearly articu- 12.1.1 The selection of a venous access site (cen-
late the appropriate credentials, training, tral vs peripheral vein) should depend on
and/or certifications and competencies re- expected duration of therapy, nutrition
quired for clinicians who prescribe nutri- requirements, and patient’s vascular con-
tion support therapy. dition and preferences.40,43,44 When PN is
912 Nutrition in Clinical Practice 33(6)

Figure 2. Route of administration algorithm. GI, gastrointestinal; PN, parenteral nutrition.

administered via a central access device, Table 1. Selection of Enteral Access Based on Gastric
the femoral vein site should be avoided, es- Tolerance and Anticipated Feeding Duration41 .
pecially in the obese, to minimize infection
Duration
risks associated with nontunneled central
venous catheters (CVCs).43,45-48 Periph- Normal Long Term
erally inserted central catheters (PICC) Gastric Short Term (Longer Than
should not be used as a strategy to re- Motility (4–6 Weeks) 6 Weeks)
duce central line-associated bloodstream
Yes Nasogastric Gastrostomy
infection (CLABSI).46 A CVC with the
No Nasoduodenal Jejunostomy
fewest number of lumens or ports required Nasojejunal
for that patient should be used for PN
administration.46
12.1.2 The selection of an enteral access device
(nasoenteric vs enterostomy [ie, gastros- place the specific access device. Guid-
tomy, jejunostomy]) should depend on the ance technology for placement of cen-
patient’s disease state, needs and goals, tral venous access and enteral access de-
ethical situation, gastrointestinal anatomy vices should be used only by clinicians
and function, expected duration of EN who have completed prerequisite training
therapy, and the ability to safely access and credentialing required by the respec-
the gastrointestinal tract via radiologic, tive institution.50,52,53 Professionals with
surgical, endoscopic techniques, or other knowledge in preventing, recognizing, and
guided technology.41,42,49-51 (See Table 1: managing complications associated with
Selection of Enteral Access Based on Gas- the placement and maintenance of the
tric Tolerance and Anticipated Feeding access devices should monitor the use of
Duration.) the access devices.41,43,44,46-48
12.1.3 Appropriate access devices shall be placed 12.1.4 Proper placement of central venous ac-
by a physician, nurse, or trained health- cess devices shall be confirmed using ap-
care professional who is competent to propriate technology and documented in
Ukleja et al 913

the medical record before initial use.43,44 compare it with previous orders when
For enteral access devices, the auscaltatory applicable.40,56
method shall not be relied upon to dif- 13.2 In hospitals that use automated compounding
ferentiate between pulmonary, gastric, and devices (ACDs) for preparation of PN formu-
small bowel placement of a nasoenteric lations, policies and procedures shall be devel-
tube.50 When using enteral access system oped to address responsibilities for operation and
or guidance technology to place enteral maintenance, staff training, and monitoring ACD
access devices, if any difficulty occurs dur- performance (ie, quality assurance).40,57
ing insertion, confirmation of the final 13.2.1 Adequate training of personnel shall in-
tube position should be done per insti- clude use of computer software to as-
tution protocol.53 Radiographic confirma- sist in daily use and trouble shooting of
tion is the gold standard for determining ACDs.40
the exact tube position after insertion and 13.2.2 PN substrate dosing limit alerts shall be ac-
should be used.41,49-52 tivated in the computer software and used
12.1.5 Central venous access should be used for in the assessment of the PN formulation
the delivery of PN admixtures with an prior to compounding.40
osmolarity greater than 900 mOsm/L.54 13.2.3 Documents generated by the ACD or
The catheter tip should be positioned in other electronic devices shall be compared
the lower segment of the superior vena with the ordered PN formulation.40
cava adjacent to the cavo-atrial junction.43 13.2.4 The pharmacist and/or pharmacy techni-
Peripheral PN may be administered, if in- cian shall monitor the equipment during
dicated, through a peripheral access device the preparation process to assure proper
provided the osmolarity of the admixture operation.40
is less than or equal to 900 mOsm/L. Lipid 13.2.5 End-product and validation testing of PN
injectable emulsion (ILE) may be concur- admixtures should be completed.
rently infused.43,44,54 13.3 In hospitals that outsource preparation of PN
12.1.6 Monitoring procedures for nutrition sup- admixtures, policies and procedures shall be de-
port therapy administration shall include veloped for appropriate ordering, storage, prepa-
visual inspection of the patient’s enteral ration, labeling, and dispensing of PN admixtures.
or parenteral access devices and insertion Hospitals should ensure that the outsource agency
site. prepares PN formulations in accordance with
12.2 Complications related to an access device and USP General Chapter <797>: Pharmaceutical
outcome(s) of the interventions to manage the Compounding-Sterile Products.55
complication(s) shall be clearly documented in the 13.4 In hospitals that use standardized, commercially
medical record. available PN products, policies and procedures
shall be developed for appropriate ordering, stor-
Standard 13. PN Admixture Preparation age, preparation, labeling, and dispensing of PN
admixtures.40,56
PN shall be prepared accurately as prescribed and stored
13.5 PN admixtures shall be sterile and free from phys-
safely according to United States Pharmacopeia (USP)
ical contaminants (foreign materials and phys-
General Chapter<797>: Pharmaceutical Compounding-
ical matter) and minimize patient exposure to
Sterile Products.55
aluminum.40,59
13.6 A pharmacist should refer to ASPEN, American
13.1 PN formulations shall be prepared using current Society of Health-system Pharmacists (ASHP),
policies and procedures regarding manufacturing, FDA Drug Shortages, or other appropriate re-
compatibility, and stability. These procedures shall source(s) on managing shortages and outages
be supervised by a licensed pharmacist with ap- of PN components and develop hospital-specific
propriate credentials and experience.40,44 strategies to provide optimal PN therapy during
13.1.1 A hospital-specific standardized process shortages.
for PN preparation shall be used. This 13.7 Nonnutrient medication (eg, insulin) should be
may include the use of standardized PN added to PN only when supported by physiochem-
formulations when appropriate.40,56 ical compatibility and stability data.54
13.1.2 A pharmacist shall review the contents 13.8 A pharmacist shall conduct a visual inspection of
of a PN order for appropriateness and the final PN admixture prior to dispensing.39
914 Nutrition in Clinical Practice 33(6)

13.10 Additions to PN admixture shall not be made r All PN ingredients shall be ordered
outside of the pharmacy sterile compounding as amount per day (ie, grams per day,
environment. mEq per day)
r Name of compounding institution or
Standard 14. EN Formula Preparation pharmacy
15.1.2 Auxiliary labels should be affixed to PN
EN formulas and products shall be prepared accurately
admixture packaging to reduce risk of
and safely as prescribed and stored according to the
error (eg, for central line only).
manufacturers’ directions and published safety consensus
15.1.3 The PN admixture shall be stored in a
recommendations.41
refrigerator (per established guidelines),
unless the admixture will be administered
14.1 EN formulas shall be prepared by trained per-
immediately to the patient.40,45,55
sonnel under professional supervision in a clean
15.2 EN formulas shall be packaged in adminis-
environment. Aseptic technique shall be used in
tration containers, which assure accuracy of
the preparation of EN formulas.41
volume, cleanliness, and minimize the risk for
14.1.1 Preparation equipment shall be sanitized
contamination.41
regularly.
15.2.1 Open-system administration containers
14.1.2 Open-system containers shall be filled with
should be used if the EN formula will be
EN formula using aseptic technique.
modified with modular products. However,
14.2 Any addition of modular products or water to
the addition of modular products to an
the formula shall be ordered by the prescribing
open-system container may result in an
clinician or designee.
unacceptable risk of contamination in
14.2.1 Additions to EN formulas shall not be
hyperthermal environments.41
done at the bedside.
15.2.2 Hospital-prepared EN formulas shall be
14.2.2 Additions to closed-system EN containers
stored in a refrigerator (per established
shall not be made.
guidelines), unless the formula will be ad-
ministered immediately to the patient.
Standard 15. Packaging and Labeling 15.3 EN labels shall be standardized.
PN admixtures and EN formula containers shall be ap- 15.3.1 EN formula containers shall be labeled
propriately packaged and labeled in a standardized fashion accurately with the contents and 2 pa-
according to hospital policy and procedure. tient identifiers (eg, name, medical record
number, date of birth), product name and
15.1 PN admixtures shall be packaged in adminis- strength, additives, volume, and appropri-
tration containers that can assure maintenance ate hang time.41
of sterility and allow visual inspection during 15.3.2 EN formula container labels shall also
preparation, storage, and infusion. contain delivery site/access, route (enteral),
15.1.1 The PN admixtures and ILE administered and method of administration (eg, contin-
as a separate infusion shall be labeled uous, cyclic, bolus).41
with the following as described in the AS- 15.3.3 EN formula container labels shall contain
PEN Parenteral Nutrition Safety Consen- a statement indicating that the product is
sus Recommendations:40 for enteral administration only.41
r Two patient identifiers (eg, name, med- 15.3.4 EN formula container labels shall contain
ical record number, date of birth) a statement indicating that the product is
r Patient location or address not for IV administration.41
r Administration date and time
r Beyond-use date and time Standard 16. Administration of Nutrition
r Route of administration (central vein Support Therapy
vs peripheral vein)
r Prescribed volume EN formulas and PN admixtures shall be administered
r Method of administration (continuous safely and accurately in accordance with the prescribed
vs cyclic) order and consistent with the patient’s tolerance.40,41
r Complete name of all ingredients ex-
pressed in the same units of measure 16.1 Nutrition support therapy shall be administered
as the PN order by or under the supervision of trained personnel.
Ukleja et al 915

16.2 Hospital-specific procedures shall exist regarding be minimized; vascular access devices
techniques used to administer nutrition support used for PN should not be used for blood
therapy. Organizations should use infusion pumps sampling.46,47,64 Coinfusion of fluids or
with the ability to reduce errors.40 medications into the PN system should
16.3 Acute care facilities should establish a policy that be avoided, if possible. If no alternatives
prohibits the use of a PN admixture prepared are available, a pharmacist shall review
for administration at home or in subacute or the compatibility and stability data for
long-term care facilities. PN should be discontin- coinfusion prior to administration.40,44
ued prior to discharge or transport to another 16.9.3 Unit-specific data regarding CLABSI
facility.40 shall be shared with all internal stake-
16.4 Each PN admixture should be inspected prior to holders and reported to external agencies
and during administration. If visual changes are as required.47
present, the admixture shall not be administered, 16.9.4 PN admixtures shall be labeled with the
and the pharmacy shall be notified.40,60 beyond-use date and time and discarded
16.5 Before nutrition support therapy is administered as indicated. Once the delivery system
to the patient, the label on the container shall is accessed, the administration of a PN
be checked against the order and the patient’s admixture shall be completed within 24
identity shall be verified per hospital policy hours.40,44-46,55
to assure the prescribed formulation is deliv- 16.9.5 Administration sets for PN shall be
ered to the appropriate patient and administered changed every 24 hours or with each new
by the correct route at the designated/intended PN container. A 1.2-micron filter shall be
time.40,41,61,62 Administration tubing should be used for all total nutrient admixtures and
attached to PN containers immediately prior a 0.22-micron filter for dextrose/amino
to use.40 acids (2-in-1) admixtures.40,44,65
16.6 The administration rate of the prescribed nutri- 16.9.6 ILE administered separately from PN ad-
tion support therapy shall be checked each time mixtures (dextrose/amino acids) shall be
a new volume is ordered or initiated and peri- infused through a 1.2-micron filter and be
odically during its administration.40,41 Use of an completed within 12 hours of initiating
independent double-check verification should be the infusion.40,44,65
performed by a second clinician prior to beginning 16.10 A policy shall exist regarding the maximal rate
a PN infusion.40,41,62 of administration for ILE. Manufacturers’ recom-
16.7 Procedures shall be written to prevent and manage mendations should be considered in formulating
vascular or enteral access device occlusion and IV this statement.
extravasation.41,42,44,63 16.11 Cycling of PN admixtures should be considered
16.8 EN and PN processes shall be documented in for patients with or at risk for liver dysfunction,
the patient’s medical record including tolerance, on long-term PN, or those who are stable, active,
administration volumes, and hourly rates. The and may benefit from infusion-free time.66,67
amount of nutrition therapy ordered vs amount 16.12 Prevention strategies shall be used to minimize
administered should be noted and reasons for the risk of microbial contamination of EN for-
discrepancies evaluated.41 mulations. (Refer to the Enteral Nutrition Prac-
16.9 Policies and procedures shall exist to prevent, tice Recommendations41 and Guidelines for the
diagnose, manage, and monitor patient infections Provision and Assessment of Nutrition Support
caused by contamination of the PN admixture Therapy in the Adult Critically Ill Patient31 for
or the equipment/devices used in its administra- details.)
tion, as PN is an independent risk factor for 16.13 Procedures and protocols to minimize the risk of
CLABSI.46-48,64 regurgitation and aspiration of EN formulations
16.9.1 Infection prevention strategies shall be should be implemented.41
used to minimize CLABSI including 16.13.1 All patients receiving EN shall be as-
a bundle of targeted, evidence-based sessed for risk of aspiration and steps
catheter insertion and maintenance employed to reduce aspiration risk and
practices.46-48 pneumonia.31,41,68-70
16.9.2 Access ports shall be disinfected with an 16.13.2 The head of the bed should be elevated
appropriate antiseptic prior to catheter 30–45° during EN administration unless
manipulation and manipulation should contraindicated.31,41,69-71
916 Nutrition in Clinical Practice 33(6)

16.14 An enteral feeding protocol should be designed Standard 17. Adverse Events Management
to assure that optimal nutrients are delivered
and unnecessary interruption of feeding mini- An adverse event, including sentinel events related to the
mized. Gastric residual volumes may be used to administration of nutrition support therapy and the equip-
assess EN tolerance as part of a multifaceted ment/access devices, shall be documented and reported
approach.31,41,68 according to hospital protocol to promote a culture of
16.15 Policies and procedures shall exist to minimize the patient safety. Protocols should be developed and followed
risk of enteral misconnections.41,72-74 to decrease the risk of adverse events.
16.15.1 Hospitals should conform to Interna-
tional Organization of Standardization Chapter V: Monitoring and Reevaluating
(ISO) standard 80369-3 that is driving the Nutrition Care Plan
the production of products with incom-
patible connectors by designing features
Standard 18. Parameters and Frequency
that make incorrect connections impos- A plan for monitoring the effect of nutrition support
sible (eg, ENFit).41,74-77 Enteral deliv- therapy interventions should be stated in the nutrition care
ery devices (administration sets, feeding plan.36,40,41
tubes, and enteral syringes) with con- Monitoring parameters are chosen relative to the therapy
nectors that can physically connect with goals of the nutrition care plan. The nutrition care plan shall
other nonenteral connectors shall not be be revised to optimize nutrition support therapy and achieve
purchased. predetermined goals, as indicated.
16.15.2 Standard Luer syringes shall not be used
to administer oral or enteral medications 18.1 The frequency of monitoring should depend on
or EN formula.41,74 severity of illness, level of metabolic stress, nu-
16.15.3 Tubes or catheters shall be traced trition status, as well as the patient’s clinical
from the patient to the point of origin condition.31,36,40,41
before connecting any new device or 18.1.1 Daily or more frequent monitoring should
infusion.41,74,76 be required in patients who are critically ill,
16.15.4 Tubes and catheters having different pur- have debilitating diseases (eg, diabetes mel-
poses should be routed in different, stan- litus) or infection, are at risk for refeeding
dardized directions (eg, IV lines routed syndrome complications, are transitioning
toward the head; enteric lines toward the between PN or EN and oral diet, or have
feet) and labeled at proximal and distal experienced complications associated with
tubing ends.73 nutrition support therapy.
16.16 Protocols shall be established for administering 18.1.2 Weekly or as clinically indicated moni-
medications and modular products through an toring may be needed in patients who
enteral access device.41 are clinically and metabolically stable with
16.16.1 Medications should not be mixed documented stable laboratory parameters.
directly with EN formulas due to 18.2 Monitoring parameters should include the follow-
potential drug-drug and drug-nutrient ing:
interactions.41,78-81 r Physical assessment, including clinical signs of
16.16.2 Medication orders should specify the fluid and nutrient excess or deficiency
route of delivery (eg, PO, NG tube, G r Functional status
tube, J tube) and be administered accord- r Vital signs
ing to current guidelines. Special con- r Actual nutrient intake (oral, enteral, and
siderations include use of proper dosage parenteral)
form, administration of the drug separate r Weight
from the EN formula and other drugs, r Laboratory data
and the location of drug delivery in the r Diagnostic tests
gastrointestinal tract.41,78-83 r Review of all medications
16.16.3 Enteral access device(s) should be r Changes in gastrointestinal function
flushed appropriately before and after r Input and output/fluid balance
each medication administration and 18.3 Appropriate changes in nutrition support therapy
restarting EN administration to help shall be made based on results of monitored
prevent occlusion.41,80,82 parameters. Recommended changes in nutrition
Ukleja et al 917

support therapy including EN formula/PN admix- 21.3 Appropriate education should be provided to pa-
ture or administration route and resulting out- tient and/or caregiver(s) and documented before
comes shall be documented in the nutrition care discharge. Communication with home infusion
plan.84 and healthcare agencies and with the patient’s
18.4 Protocols should be established to maintain blood home nutrition support management team should
glucose control in patients receiving EN or be established prior to hospital discharge.
PN.85-87 21.4 The nutrition support therapy prescription and
administration schedule should be documented
Standard 19. Reevaluation of Nutrition Care and communicated with home infusion and home
Plan health agencies before discharge.36,40,41,45,88 Specif-
ically, with PN, there should be pharmacist-
The patient shall be monitored for progress toward short- to-pharmacist communication to the alternate
and long-term goals as defined in the nutrition care healthcare facilities or home agencies.
plan.36,41 21.5 Periodic monitoring should be recommended de-
pending on patient’s condition.35,39,40,86
19.1 Appropriate parameters should be measured
serially during nutrition support therapy and
documented.36,40,41 Parameters may include
Standard 22. Nutrition Therapy at End-of-Life
weight change, changes in laboratory data, Care
adequacy of intake, ability to transition to oral The decision on nutrition support therapy in an end-of-
diet, functional status performance, and quality life setting should be determined by patient autonomy
of life. and the patient’s family member(s) or surrogate decision
19.2 The monitoring parameters should be compared maker. The patient or the patient’s family member(s) or
with the goals of the nutrition care plan. If goals surrogate decision maker shall decide on acceptance or
are not being met, a new clinical issue or complica- refusal of medical therapy.36,41,86,88-90
tion develops, and/or an adverse event occurs, the
nutrition care plan should be modified.
22.1 The clinician has no obligation to provide nutri-
tion support therapy and hydration to a patient in
Chapter VI: Transition of Therapy the end-of life situation.88-90
Standard 20. Adequacy of Intake 22.2 Decisions at end-of-life are often made based on
healthcare and spiritual literacy of the patient
The transition of nutrition therapies shall be monitored. and his/her family; they shall be involved in the
Recommendations for improving oral and EN intake shall healthcare process of end-of-life.
be documented. Adequacy of energy and nutrient intake
is based on clinical judgment and shall be assessed and
documented before discontinuation of nutrition support References
therapy.31,35,40 (See Figure 2: Route of administration algo- 1. Ulkeja A, Freeman KL, Gilbert K, et al. Standards for nutrition
support: adult hospitalized patients. Nutr Clin Pract. 2010;25(4):403-
rithm.)
414.
2. Robinson D, Walker R, Adams S, et al. American Society for Parenteral
Standard 21. Continuity of Care and Nutrition (A.S.P.E.N.) Definition of Terms, Enteral Style, and
Conventions Used in A.S.P.E.N. Board of Directors–Approved
Continuity of the nutrition support therapy shall oc- Documents. https://www.nutritioncare.org/uploadedFiles/Documents/
cur through active communication with all members of Guidelines_and_Clinical_Resources/ASPEN%20Definition%
the patient care team, the patient, and caregiver(s). (See 20of%20Terms,%20Style,%20and%20Conventions%20Used%20in%
Figure 1: Adult nutrition care pathway.) 20ASPEN%20Board%20of%20Directors-Approved%
20Documents.pdf. Accessed July 1, 2018.
3. The Joint Commission PC.01.02.01, PC 01.02.03. Comprehensive Ac-
21.1 A plan shall be developed for transition of nu-
creditation Manual for Hospitals: The Official Handbook. Oak Brook,
trition support therapy to an alternate healthcare IL: Joint Commission Resources, 2018.
facility or to home care and should include 4. Schneider PJ. Nutrition support teams: an evidence-based practice.
identification of the primary clinician responsi- Nutr Clin Pract. 2006;21(1):62-67.
ble for coordinating, monitoring, providing ed- 5. Winkler MF. Improving safety and reducing harm associated with
specialized nutrition support. Nutr Clin Pract. 2005;20(6):595-596.
ucation, and ordering home nutrition support
6. Gurgueira GL, Leite HP, Taddei JA, de Carvalho WB. Outcomes in a
therapy.31,36,40,41,87,88 pediatric intensive care unit before and after the implementation of a
21.2 Indications for home nutrition support therapy nutrition support team. JPEN J Parenter Enteral Nutr. 2005;29(3):176-
shall be documented. 185.
918 Nutrition in Clinical Practice 33(6)

7. O’Brien DD, Hodges RE, Day AT, Waxman KS, Rebello T. Rec- 26. Kondrup J, Rasmussen H, Hamberg O, et al. Nutritional risk screening
ommendations of nutrition support team promote cost containment. (NRS 2002): a new method based on an analysis of controlled clinical
JPEN J Parenter Enteral Nutr. 1986;10(3):300-302. trials. Clin Nutr. 2003;22(3):321-336.
8. Traeger SM, Williams GB, Milliren G, Young DS, Fisher M, Haug 27. Mueller C, Compher C, Druyan ME, A.S.P.E.N. Board of Directors.
MT. Total parenteral nutrition by a nutrition support team: improved A.S.P.E.N. Clinical Guidelines: nutrition screening, assessment, and
quality of care. JPEN J Parenter Enteral Nutr.1986;10(8):408-412. intervention in adults. JPEN J Parenter Enteral Nutr. 2011;35(1):16-
9. Goldstein M, Braitman LE, Levine GM. The medical and financial 24.
costs associated with termination of a nutrition support nurse. JPEN 28. Lacey K, Pritchett E. Nutrition care process and model: ADA adopts
J Parenter Enteral Nutr. 2000;24(6):323-327. road map to quality care and outcomes management. J Am Diet Assoc.
10. Naylor CJ, Griffiths RD, Fernandez RS. Does a multidisciplinary 2003;103(8):1061-1072.
total parenteral nutrition team improve patient outcomes? A systematic 29. American Dietetic Association. Identifying patients at risk: ADA’s
review. JPEN J Parenter Enteral Nutr. 2004;28(4):251-258. definitions for nutrition screening and nutrition assessment. Council
11. Gales BJ, Riley DG. Improved total parenteral nutrition therapy man- on Practice (COP) Quality Management Committee. J Am Diet Assoc.
agement by a nutritional support team. Hosp Pharm.1994;29(5):469- 1994;94(8):838-839.
470, 473-475. 30. JeVenn AK, Galang M, Hipskind P, Bury C. Malnutrition screening
12. Dalton MJ, Schepers G, Gee JP, Alberts CC, Eckhauser FE, Kirking and assessment. In: Mueller CM, ed. The ASPEN Adult Nutrition
DM. Consultative total parenteral nutrition teams: effect on the Support Core Curriculum (3rd ed.). Silver Spring, MD: American
incidence of total parenteral nutrition-related complications. JPEN J Society for Parenteral and Enteral Nutrition; 2017:185-212.
Parenter Enteral Nutr. 1984;8(2):146-152. 31. McClave SA, Taylor BE, Martindale RG, et al. Guidelines for the
13. Maurer J, Weinbaum F, Turner J, et al. Reducing the inappropriate provision and assessment of nutrition support therapy in the adult
use of parenteral nutrition in an acute care teaching hospital. JPEN J critically ill patient. JPEN J Parenter Enteral Nutr. 2016;40(2):159-211.
Parenter Enteral Nutr. 1996;20(4):272-274. 32. Lim SlL, Ang E, Foo YL, et al. Validity and reliability of nutrition
14. Chris Anderson D, Heimburger DC, Morgan SL, et al. Metabolic screening administered by nurses. Nutr Clin Pract. 2013;28(6):730-736.
complications of total parenteral nutrition: effects of a nutri- 33. Patel V, Roman M, Corkins MR. Nutrition screening and assessment in
tion support service. JPEN J Parenter Enteral Nutr. 1996;20(3): hospitalized patients: a survey of current practice in the United States.
206-210. Nutr Clin Pract. 2014;29(4):483-490.
15. A.S.P.E.N. Practice Management Task Force: DeLegge M, Wooley JA, 34. White JV, Guenter P, Jensen G, et al. Consensus Statement: Academy
Guenter P, et al. The state of nutrition support teams and update on of Nutrition and Dietetics and American Society for Parenteral and
current models for providing nutrition support therapy to patients. Nutr Enteral Nutrition: Characteristics recommended for the identification
Clin Pract 2010;25(1):76-84. and documentation of adult malnutrition (under-nutrition). JPEN J
16. Braun K, Utech A, Velez ME, Walker R. Parenteral nutrition elec- Parent Enteral Nutr. 2012;36(3):275-283.
trolyte abnormalities and associated factors before and after nutrition 35. Guenter P, Jensen G, Patel V, et al. Addressing disease-related malnu-
support team initiation. JPEN J Parenter Enteral Nutr. 2018;42(2):387- trition in hospitalized patients: A call for a national goal. Jt Comm J
392. Qual Patient Saf. 2015;41(10):469-473.
17. Hassell JT, Games AD, Shaffer B, Harkins LE. Nutrition support team 36. Worthington P, Balint J, Bechtold M, et al. When is PN appropriate?
management of enterally fed patients in a community hospital is cost- JPEN J Parenter Enteral Nutr. 2017;41(3):324-377.
beneficial. J Am Diet Assoc. 1994;94(9):993-998. 37. Doley J, Phillips W. Overview of enteral nutrition. In: Mueller CM, ed.
18. Kennedy JF, Nightingale JM. Cost savings of an adult hospital The ASPEN Adult Nutrition Support Core Curriculum (3rd ed.). Silver
nutrition support team. Nutrition. 2005;21(2):1127-1133. Spring, MD: American Society for Parenteral and Enteral Nutrition;
19. DeLegge MH, Kelly AT. The state of nutrition support teams. Nutr 2017:213-226.
Clin Pract. 2013;28(6):691-697. 38. Mirtallo J. Overview of parenteral nutrition. In: Mueller CM, ed.
20. Mascarenhas MR, August DA, DeLegge MH, et al. Standards of prac- The ASPEN Adult Nutrition Support Core Curriculum (3rd ed.). Silver
tice for nutrition support physicians. Nutr Clin Pract. 2012;27(2):295- Spring, MD: American Society for Parenteral and Enteral Nutrition;
299. 2017:285-296.
21. DiMariua-Ghalili RA, Gilbert K, Lord L, et al. Standards of nutrition 39. Guenter P, Boullata JI, Ayers P, et al. Standardized competencies
care practice and professional performance for nutrition support and for PN prescribing: the American Society of Parental and Enteral
generalist nurses. Nutr Clin Pract. 2016;31(4):527-547. Nutrition model. Nutr Clin Pract. 2015;30(4):570-576.
22. Tucker A, Ybarra J, Bingham A, et al. American Society for Parenteral 40. Ayers P, Adams SC, Boullata J, et al. A.S.P.E.N. Parenteral nutrition
and Enteral Nutrition (A.S.P.E.N.) Standards of practice for nutrition safety consensus recommendations. JPEN J Parenter Enteral Nutr.
support pharmacists. Nutr Clin Pract. 2015;30(1):139-146. 2014;38(2):296-333.
23. Brantley SL, Russell MK, Mogensen KM, et al. American Society 41. Boullata JI, Carrera AL, Harvey L, et al. ASPEN safe practices for
for Parenteral and Enteral Nutrition and Academy of Nutrition enteral nutrition therapy. JPEN J Parenter Enteral Nutr. 2017;41(1):15-
and Dietetics: revised 2014 standards of practice and standards of 103.
professional performance for registered dietitian nutritionists (com- 42. Fang JC, Kinikini M. Enteral access devices. In: Mueller CM, ed.
petent, proficient, and expert) in nutrition support. Nutr Clin Pract. The ASPEN Adult Nutrition Support Core Curriculum, (3rd ed.) Silver
2014;29(6):792-828. Spring, MD: American Society for Parenteral and Enteral Nutrition.
24. Mathieson K, Vogt EAM. Quality improvement in clinical practice. In: 2017;251-264.
Mueller CM, ed. The ASPEN Adult Nutrition Support Core Curriculum 43. Neal AM, Drogan K. Parenteral access devices. In: Mueller CM, ed.
(3rd ed.). Silver Spring, MD: American Society for Parenteral and The ASPEN Adult Nutrition Support Core Curriculum (3rd ed.). Silver
Enteral Nutrition; 2017:805-818. Spring, MD: American Society for Parenteral and Enteral Nutrition;
25. Weekes CE, Elia M, Emery PW. The development, validation and reli- 2017:321-344.
ability of a nutrition screening tool based on the recommendations of 44. Gorski LA, Hadaway L, Hagle M, McGoldrick M, Orr M, Doellman
the British Association for Parenteral and Enteral Nutrition (BAPEN). D. Infusion therapy standards of practice. J Infus Nurs. 2016;39(1
Clin Nutr. 2004;23(5):1104-1112. Suppl):S1-S159.
Ukleja et al 919

45. Mirtallo J, Canada T, Johnson D, et al. Task Force for the Revision of 60. Rollins CJ. Total nutrient admixtures: stability issues and their impact
Safe Practices for Parenteral Nutrition. Safe practices for parenteral on nursing practice. J Intraven Nurs. 1997;20(6):299-304.
nutrition [erratum in JPEN J Parenter Enteral Nutr. 2006;30:177]. 61. Pennsylvania Patient Safety Authority. The five rights: not the gold
JPEN J Parenter Enteral Nutr. 2004;28(6):S39-S70. standard for safe medication practices. PA-PSRS Patient Safety
46. O’Grady NP, Alexander M, Burns LA, et al. Guidelines for the preven- Advisory. http://patientsafety.pa.gov/ADVISORIES/Pages/200506_09
tion of intravascular catheter-related infections. https://www.cdc.gov/ .aspx. Accessed March 8, 2018.
infectioncontrol/guidelines/pdf/bsi/bsi-guidelines-H.pdf. Accessed 62. Institute for Safe Medication Practices. Independent double checks:
March 5, 2018. undervalued and misused: selective use of this strategy can play an
47. Marschall J, Mermel L, Fakih M, et al. Strategies to prevent central important role in medication safety. ISMP Medication Safety Alert!
line-associated bloodstream infections in acute care hospitals: 2014 2013;18(12):1-4. https://www.ismp.org/newsletters/acutecare/show
update. Infect Control Hosp Epidemiol. 2014;35(7):753-771. article.aspx?id=51. Accessed March 8, 2018.
48. Chopra V, Krein S, Olmsted R, Safdar N, Saint S. Prevention of central 63. Benedetta B, Andres C. Images in clinical medicine: Extravasation
line-associated bloodstream infections: brief update review. In: Shekelle of peripherally administered parenteral nutrition. N Engl J Med.
PG, Wachter RM, Pronovost PJ, eds. AHRQ. Making Healthcare Safer 2011;364(10):e20.
II: An Updated Critical Analysis of the Evidence for Patient Safety 64. The Joint Commission. Preventing central line–associated bloodstream
Practices. AHRQ Publication No. 13-E001-EF. Rockville, MD: Agency infections: a global challenge, a global perspective. http://www.jointcom
for Healthcare Research and Quality. 2013:88-109. https://www. mission.org/assets/1/18/clabsi_monograph.pdf. Published May 2012.
ahrq.gov/sites/default/files/wysiwyg/research/findings/evidence-based- Accessed March 7, 2018.
reports/services/quality/patientsftyupdate/ptsafetyII-full.pdf. 65. Institute for Safe Medication Practices. IV fat emulsion needs a filter.
Accessed March 7, 2018. ISMP Medication Safety Alert! 2016;21(1):2-3. http://www.ismp.org/
49. Koopman MC, Kudsk KA, Szotkowski MJ, et al. A team-based pro- Newsletters/acutecare/issue.aspx?id=1113. Accessed March 8, 2018.
tocol and electromagnetic technology eliminate feeding tube placement 66. Stout SM, Cober MP. Metabolic effects of cyclic parenteral nutrition
complications. Ann Surg. 2011;253(2):297-302. infusion in adults and children. Nutr Clin Pract. 2010;25(3):277-
50. American Association of Critical Care Nurses. (2016). AACN prac- 281.
tice alert: initial and on-going verification of feeding tube place- 67. Suryadevara S, Celestin J, DeChicco R, et al. Type and prevalence
ment in adults. (applies to blind insertions and placements with of adverse events during the parenteral nutrition cycling process in
an electromagnetic device). http://www.aacn.org/wd/practice/content/ patients being prepared for discharge. Nutr Clin Pract. 2012;27(2):268-
feeding-tube-practice-alert.pcms?menu=practice. Accessed March 7, 273.
2018. 68. Malone A, Seres D, Lord L. Complications of enteral nutrition. In:
51. Powers J, Lubbehusen M, Spitzer T, et al. Verification of an electro- Mueller CM, ed. The ASPEN Adult Nutrition Support Core Curriculum
magnetic placement device compared with abdominal radiograph to (3rd ed.). Silver Spring, MD: American Society for Parenteral and
predict accuracy of feeding tube placement. JPEN J Parenter Enteral Enteral Nutrition; 2017:265-284.
Nutr. 2011:35(4):535-539. 69. American Association of Critical Care Nurses. AACN practice alert:
52. Metheny NA, Meert KL. Effectiveness of an electromagnetic feeding prevention of aspiration in adults. Crit Care Nurse. 2016;36(1):e20-e24.
tube placement device in detecting inadvertent respiratory placement. https://www.aacn.org/clinical-resources/practice-alerts/prevention-of-
Am J Crit Care. 2014;23(3):240-248. aspiration. Accessed March 7, 2018.
53. U.S. Food and Drug Administration. Feeding tube placement systems- 70. DiBardino D, Wunderink R. Aspiration pneumonia: a review of
letter to health care providers. http://www.fda.gov/MedicalDevices/ modern trends. J Crit Care. 2015;30(1):40-48.
Safety/LetterstoHealthCareProviders/ucm591838.htm. Accessed June 71. Tablan OC, Anderson LJ, Besser R, et al. Guidelines for preventing
20, 2018. health-care-associated pneumonia, 2003: recommendations of CDC
54. Boullata JI, Gilbert K, Sacks G, et al. A.S.P.E.N. Clinical guidelines: and the Healthcare Infection Control Practices Advisory Committee.
parenteral nutrition ordering order review, compounding, labeling, MMWR Recomm Rep. 2004;53(RR-3):1-36.
and dispensing. JPEN J Parenter Enteral Nutr. 2014;38(3):334- 72. Guenter P, Hicks RW, Simmons D, et al. Enteral feeding misconnec-
377. tions: a consortium position statement. Jt Comm J Qual Patient Saf.
55. Pharmaceutical compounding—sterile preparations (general informa- 2008;34(5):285-292.
tio chapter 797). In: The United States Pharmacopeia, 31st rev., and 73. Guenter P. New enteral connectors: raising awareness. Nutr Clin Pract.
the National Formulary 26th ed. Rockville, MD: The United States 2014; 29(5):612-614.
Pharmacopeial Convention; 2008:334-351. 74. The Joint Commission. Managing risk during transition to new ISO tub-
56. A.S.P.E.N. Board of Directors and Task Force on Parenteral Nutrition ing connector standards. Sentinel Event Alert 53. http://www.jointcom
Standardization: Kochevar M, Guenter P, Holcombe B, Malone A, mission.org/sea_issue_53/. Published August 20, 2014. Accessed March
Mirtallo J. A.S.P.E.N. statement on parenteral nutrition standardiza- 8, 2018.
tion. JPEN J Parenter Enteral Nutr. 2007;31(5):441-448. 75. Premier Safety Institute. Tubing misconnections. http://www.premier
57. American Society of Health-System Pharmacists. ASHP guidelines safetyinstitute.org/safety-topics-az/tubing-misconnections/tubing-
on the safe use of automated compounding devices for the prepa- misconnections/. Accessed March 8, 2018.
ration of parenteral nutrition admixtures. Am J Health-Syst Pharm. 76. Simmons D, Symes L, Graves K. Tubing misconnections: normaliza-
2000;57(14):1343-1348. tion of deviance. Nutr Clin Pract. 2011;26(3):286-293.
58. American Society of Health-System Pharmacists. ASHP guidelines on 77. Institute for Safe Medication Practices. EnFit enteral devices are
outsourcing sterile compounding services. Am J Health-Syst Pharm. on their way . . . important safety considerations for hospitals. ISMP
2015;72(19):1664-1675. Medication Safety Alert! 2015;20(7):1-7.
59. The American Society for Parenteral and Enteral Nutrition 78. Verdell A, Rollins CJ. Drug-nutrient interactions. In: Mueller CM, ed.
(A.S.P.E.N.) Aluminum Task Force; Charney PJ. A.S.P.E.N. statement The ASPEN Adult Nutrition Support Core Curriculum (3rd ed.). Silver
on aluminum in parenteral nutrition solutions. Nutr Clin Pract. Spring, MD: American Society for Parenteral and Enteral Nutrition;
2004;19(4):416-417. 2017:361-378.
920 Nutrition in Clinical Practice 33(6)

79. Boullata JI. Drug administration through an enteral feeding tube. Am 85. McMahon MM, Nystrom E, Braunschweig C, Miles J, Compher C,
J Nurs. 2009;109(10):34-42. ASPEN Board of Directors. A.S.P.E.N. Clinical Guidelines: nutrition
80. Williams NT. Medication administration through enteral feeding support of adult patients with hyperglycemia. JPEN J Parenter Enteral
tubes. Am J Health-System Pharm. 2008;65(24):2347-2357. Nutr. 2013;38(1):23-36.
81. Institute for Safe Medication Practices. Preventing errors when admin- 86. Mundi MS, Nystrom E, Hurley DL, McMahon MM. Management of
istering drugs via an enteral feeding tube. ISMP Medication Safety parenteral nutrition in hospitalized patients. JPEN J Parenter Enteral
Alert! 2010;15(9)1-4. https://www.ismp.org/newsletters/acutecare/ Nutr. 2017;41(4):535-549.
articles/20100506.asp. Accessed March 8, 2018. 87. Durfee SM, Adams SC, Arthur E, et al. A.S.P.E.N. Standards for
82. McIntyre CM, Monk H. Medication absorption considerations in nutrition support: home and alternate site care. Nutr Clin Pract.
patients with postpyloric enteral feeding tubes. Am J Health-System 2014;29(5):542-555.
Pharm. 2014;71(7):549-556. 88. Ganzini L. Artificial nutrition and hydration at the end of life: ethics
83. Institute for Safe Medication Practices. Oral dosage forms that should and evidence. Palliat Support Care. 2006;4(2):135-143.
not be crushed 2016. http://www.ismp.org/tools/DoNotCrush.pdf. Ac- 89. Barrocas A, Geppert C, Durfee SM, et al. A.S.P.E.N. ethics position
cessed March 8, 2018. paper. Nutr Clin Pract. 2010;25(6):672-679.
84. McClave SA, DiBaise JK, Mullin GE, Martindale RG. ACG clinical 90. Schwartz DB, Barrocas A, Wesley JR, et al. Gastrostomy tube place-
guideline: nutrition therapy in the adult hospitalized patient. Am J ment in patients with advanced dementia or near end of life life. Nutr
Gastroenterol. 2016;111(3):315-334. Clin Pract. 2014;29(6):829-840.
Special Report

Nutrition in Clinical Practice


Volume 33 Number 6
Pediatric Nasogastric Tube Placement and Verification: Best December 2018 921–927

C 2018 American Society for

Practice Recommendations From the NOVEL Project Parenteral and Enteral Nutrition
DOI: 10.1002/ncp.10189
wileyonlinelibrary.com

Sharon Y. Irving, PhD, CRNP, FCCM, FAAN1,2 ; Gina Rempel, MD, FRCPC3,4 ;
Beth Lyman, RN, MSN, CNSC, FASPEN5 ; Wednesday Marie A. Sevilla, MD, MPH,
CNSC6 ; LaDonna Northington, DNS, RN, BC7 ; Peggi Guenter, PhD, RN, FAAN,
FASPEN8 ; and The American Society for Parenteral and Enteral Nutrition

Abstract
The placement of a nasogastric tube (NGT) in a pediatric patient is a common practice that is generally perceived as a benign bedside
procedure. There is potential risk for NGT misplacement with each insertion. A misplaced NGT compromises patient safety,
increasing the risk for serious and even fatal complications. There is no standardized method for verification of the initial NGT
placement or reverification assessment of NGT location prior to use. Measurement of the acidity or pH of the gastric aspirate is the
most frequently used evidence-based method to verify NGT placement. The radiograph, when properly obtained and interpreted,
is considered the gold standard to verify NGT location. However, the uncertainty regarding cumulative radiation exposure related
to radiographs in pediatric patients is a concern. To minimize risk and improve patient safety, there is a need to identify best practice
and to standardize care for initial and ongoing NGT location verification. This article provides consensus recommendations for best
practice related to NGT location verification in pediatric patients. These consensus recommendations are not intended as absolute
policy statements; instead, they are intended to supplement but not replace professional training and judgment. These consensus
recommendations have been approved by the American Society for Parental and Enteral Nutrition (ASPEN) Board of Directors.
(Nutr Clin Pract. 2018;33:921–927)

Keywords
enteral nutrition; misplacement; NG feeding tube; pediatric

Introduction when initially placed or for reverification of NGT location


before use in pediatric patients. There is a need to identify
In 2014, a report on 255,140 hospital discharges in the best practice and standardize care for initial and ongoing
United States revealed that 25% of all patients receiving verification of NGT location to decrease the risk of a
enteral nutrition (EN) were children. Of those, 6% of the misplaced NGT. Therefore, the objective of this article is to
total number of patients were under 12 months of age.1 This
report indicates that a significant number of hospitalized From the 1 University of Pennsylvania School of Nursing,
children require feeding tube placement for the adminis- Philadelphia, Pennsylvania, USA; 2 Department of Critical Care
tration of EN. A 2016 study reported that approximately Nursing, The Children’s Hospital of Philadelphia, Philadelphia,
25% of hospitalized pediatric patients require a temporary Pennsylvania, USA; 3 Rady Faculty of Health Sciences, Max Rady
feeding tube.2 The most common enteral feeding tube College of Medicine, University of Manitoba, Winnipeg, Canada;
4 Children’s Hospital Winnipeg, Winnipeg, Manitoba, Canada;
used in hospitalized children is a nasogastric tube (NGT).2 5 Nutrition Support Team, Children’s Mercy Hospital, Kansas City,
Insertion of an NGT is a high-volume practice commonly Missouri, USA; 6 Division of Pediatric Gastroenterology, Children’s
performed by nurses as a blind procedure, without the use Hospital of Pittsburgh, University of Pittsburgh Medical Center,
of technology to guide or visualize the internal path of the Pittsburgh, Pennsylvania, USA; 7 University of Mississippi Medical
tube. Although the vast majority of blind NGT insertions Center School of Nursing, Jackson, Mississippi, USA; and 8 Clinical
Practice, Quality, and Advocacy, American Society for Parenteral and
result in successful placement in the intended location— Enteral Nutrition (ASPEN), Silver Spring, Maryland, USA.
the stomach—each tube can potentially be misplaced, even
This article originally appeared online on September 6, 2018.
when the procedure is performed by a healthcare provider
experienced in NGT placement. A misplaced NGT compro- Corresponding Author:
Sharon Y. Irving, PhD, CRNP, FCCM, FAAN, Assistant Professor,
mises patient safety, increasing the risk for severe and even University of Pennsylvania School of Nursing, Claire M. Fagin Hall,
fatal complications.3-6 Clinicians practicing in the United 418 Curie Blvd., RM 427, Philadelphia, PA 19104.
States lack guidance for best practice to verify NGT location Email: ysha@nursing.upenn.edu
922 Nutrition in Clinical Practice 33(6)

develop and disseminate recommendations for best practice misplacements in pediatric patients in the United States, it
related to NGT location verification in pediatric patients is well recognized that even 1 undetected, misplaced NGT
based on the available literature. may have major implications for the individual patient, the
The consensus recommendations presented here are not family, and the entire healthcare system.21
intended as absolute policy statements. Use of these prac- Although the reporting of NGT misplacements is incon-
tice recommendations does not in any way guarantee any sistent in the United States, the National Health Service
specific benefit in outcome or survival. The professional (NHS) in the United Kingdom has issued a series of Patient
judgment of the attending health professional is the primary Safety Alert (PSA) reports, the most recent in 2016.22
component of quality medical care delivery. Because con- In that 2016 PSA report spanning 5 years (2011–2016),
sensus recommendations cannot account for every variation there were 95 occurrences of NGT misplacement, with 32
in circumstances, practitioners must always exercise pro- deaths from the over 3 million NGT (or orogastric tube)
fessional judgment when applying these recommendations placements recorded.22 The most common error cited was
to individual patients. These consensus recommendations the inaccurate interpretation of a radiograph used to verify
are intended to supplement, but not replace, professional NGT position. Other errors cited include the use of unap-
training and judgment. proved methods for NGT placement verification, nursing
error in performing pH testing, and communication failures
for which the NGT location was not checked before use.
Background Failure at the organizational level to implement previously
The literature is replete with case reports in both pediatric identified processes to ensure correct NGT placement was
and adult patients describing NGT misplacements.5 An determined to be a primary cause of the misplacements.
early account of submucosal placement of an NGT that Thus, a mandate was issued by the NHS to declare NGT
occurred in an adult patient under anesthesia was reported misplacement a never event, and a PSA was directed to
by Daly in 1953 and again by Lind et al. in 1978.7,8 Compli- organizations emphasizing the seriousness with which NGT
cations in adult patients related to NGT placement can be placement is to be regarded, holding both the provider and
found from the 1970s through the present day.8-11 Similar the organization accountable for patient safety related to
reports of misplaced NGTs in children describe insertions NGT location verification.22,23
into the esophagus,4,12,13 pylorus,4 pharyngeal mucosa,14,15
intracranium,16 and most commonly the respiratory tract.17
Both gastric and bladder perforation related to NGT
Current Practice
misplacement have been described in children.18,19 These Correct NGT placement begins with accurate measurement
reports of misplaced NGTs in both adult and pediatric of the length of the tube to be inserted into the patient to
patients demonstrate the risks associated with insertion, reach the stomach. Two methods commonly used by nurses
emphasizing that NGT placement is not always a benign to determine NGT depth are the Nose→Earlobe→Xiphoid
procedure, as is often perceived. process→Midline of the Umbilicus (NEMU) method and
Although NGT misplacements have frequently been the Nose→Earlobe→Xiphoid (NEX) method of measure-
described in the United States, it is difficult to quantify ment. When the 2 methods were evaluated, the NEMU
an actual number of misplacements, particularly in chil- method demonstrated superior accuracy (97% vs 59%) over
dren. There are limitations to state reporting mechanisms, the NEX method for placement in the stomach.24 Upon
a lack of a national reporting system, and a presumed closer evaluation, use of the NEX method often resulted in
hesitancy among institutions to provide public access to high esophageal NGT misplacement, increasing the risk of
such information. Without a denominator for the number of aspiration and jeopardizing patient safety.24,25
NGTs placed and a definitive number of misplacements, it Current practice to verify NGT location is variable
is virtually impossible to accurately quantify the number of among institutions, patient care units, and providers.2,11,26
NGT misplacements that occur. Pennsylvania is one of few Commonly used methods for NGT location verification
states where reporting of NGT misplacements is required. A include: auscultation, aspiration with visual inspection
2017 Pennsylvania Patient Safety Authority report describes of gastric fluids, pH testing of gastric secretions, and
166 enteral tube misplacements documented from 2011– radiography.27 Safety and practice alerts warn against the
2016.20 In this report, 10.2% of the misplacements occurred use of auscultation28 and visual inspection of gastric
in pediatric patients, with many of these misplacements aspirate29 as the means of NGT location verification be-
associated with adverse events.20 This report is one of cause neither method is confirmatory, and either can give
few references that quantifies the number of enteral tube false affirmation of correct NGT placement. Despite these
misplacements in a defined time period, thus depicting the warnings and practice alerts, recent studies found that these
scope of the problem for verification of NGT location. methods are still widely used by nurses caring for both
Despite the lack of robust prevalence data describing NGT pediatric and adult patients.2,30
Irving et al 923

Use of Radiographs to Verify NGT Placement less than 12 months of age having the highest prescription
rate for H2 RA medications.40
The current gold standard to verify NGT placement is a A true gastric aspirate is necessary to obtain an accurate
properly obtained and interpreted radiograph. However, pH measurement; therefore, a second issue to consider
uncertainty regarding cumulative radiation exposure related concerns reverification of correct NGT location in those
to radiograph frequency,31 as well as concerns over the ac- patients receiving continuous enteral feeding. These patients
curate and consistent interpretation and reporting of NGT may require a period of cessation of formula infusion and
location by both radiologists32,33 and nonradiologists,34 water or air flush of the NGT to ensure accuracy when
raise questions regarding the use of radiography for NGT obtaining a gastric aspirate to measure pH, although there
location verification as the gold standard in pediatric pa- are no data available to support or refute this practice.
tients. Accurate NGT location by radiographic verification A limitation to using gastric pH to assess NGT location
depends on clearness of the image, interpretation, and arises if the tube is misplaced into the esophagus, where
the accuracy and clarity of the radiographic report. The it is difficult to withdraw secretions. More importantly, if
report should contain information on the path of the NGT esophageal placement occurs, the pH of any aspirate may
and the exact location of the tube tip that indicates its mimic that of gastric secretions due to aspiration or reflux
readiness for use.32 Concerns of radiation exposure, the at the time of pH testing, which carries a high risk of
variability of technique, and the lack of standardization false representation of NGT placement. Similarly, distal
for the amount of radiation used has led many institutions migration of the NGT to the level of the pylorus or beyond
to use the as low as reasonably achievable35 concept for can also result in difficulty obtaining a gastric sample to test
pediatric imaging. Given these factors, radiograph is often pH for accurate gastric placement. Although the use of pH
not the first-line method used to verify NGT placement in measurement for tube tip verification is the best evidence-
many children’s hospitals.2 However, it is the standard by based method, there are issues—as outlined above—that
which all other methods of verification are compared for need to be considered.
accuracy in establishing NGT location.

Use of Gastric pH to Verify NGT Placement Other Methods Used for NGT Verification
Measurement of the acidity of the gastric aspirate is an The use of an electromagnetic sensor-guided device for
evidence-based method used to verify NGT placement. NGT placement verification in pediatric patients is contro-
Commercial products to measure pH from gastric aspi- versial. A PSA issued in the United Kingdom described 2
rate show variance in measurement increments of 0.5–1.0. patient deaths associated with the use of an electromag-
Studies have demonstrated that obtaining a pH ࣘ 5.5 from netic sensor-guided device.41 The PSA mandates gastric pH
gastric aspirate obtained from an NGT is a reliable indicator measurement to accompany the use of the electromagnetic
that the tube is properly placed in the stomach.4,23,29,36-38 sensor-guided device to verify NGT placement. A limitation
Standard practice in the United Kingdom is to obtain a gas- associated with this device is tube size. Currently, the
tric pH measurement as the primary method to determine smallest tube that accompanies the device is 8 French, which
NGT location; a pH value of 1–5.5 is considered indicative is often too large for many pediatric patients. Also, the
of correct gastric placement for an NGT.22 external sensor necessary for use with the device may be too
An issue to consider when using gastric aspirate pH large and too heavy to be placed on the chest of smaller
measurement to determine NGT location is the use of infants and children. Safety concerns with the use of the
histamine-2 receptor antagonists (H2 RAs) and/or proton electromagnetic sensor-guided device include injury to the
pump inhibitors (PPIs). These classes of medications lower intestinal intima42 and inconsistency between the actual
gastric acidity and can result in a pH measurement > 5.5, tube location and the image projected by the device.43,44 A
causing concern about tube misplacement.4 However, a US Food and Drug Administration (FDA) alert issued in
discrepancy of accurate gastric aspirate pH measurements January 2018, regarding use of an electromagnetic sensor-
when these medications are used has not been validated in guided device for NGT placement, recommends user train-
the literature. A recently completed retrospective study of ing from the manufacturer with mandated competency in
neonates demonstrated that 97% of 6979 pH measurements device operation, and use of an additional method of
obtained in 1024 infants were ࣘ5.39 Whereas not many of NGT location verification.45 Studies in the United States
the infants received a PPI or H2 RA, 95% and 92% of those have demonstrated the use of an electromagnetic sensor-
who did had pH ࣘ 5, respectively. Additionally, a 2016 guided device to be helpful for the placement of transpyloric
retrospective study from a large medical record database feeding tubes in adult and pediatric patients46,47 ; however,
reports a significant increase in H2 RA and PPI prescriptions definitive data for successful NGT placement in pediatric
in children over a 6-year (2005–2011) period, with children patients is limited.
924 Nutrition in Clinical Practice 33(6)

Figure 1. NGT placement and verification decision tree.


NG, nasogastric.

The use of ultrasound technology as a noninvasive is needed. Its portability, absence of radiation, and
substitute for radiologic imaging to verify NGT placement noninvasive properties make ultrasound a potentially
shows promise. A recent study in a pediatric intensive useful method for verifying NGT location. An additional
care unit demonstrated 100% sensitivity with the use of method for verification of NGT placement, capnography,
ultrasound for correct placement of NGTs at the bedside has demonstrated enteral placement with 98% accuracy
when operated by a radiologist.48 Further investigation in one study49 ; however, it is not currently recommended
into the feasibility and applicability of ultrasound to verify to be used as an independent method to verify NGT
NGT location in children at the bedside by nonradiologists placement.
Irving et al 925

Recommendations - Difficulty placing the NGT


- NGT placement in any patient at high risk
Based on the available evidence and as outlined in Figure of misplacement. This includes those with
1, the following are recommendations for best practice known history of facial fractures, neurologic
standards to verify NGT location in pediatric patients: injury/insult/baseline abnormality, respiratory
r Provide education
concerns, decreased or absent gag reflex, and
those who are critically ill.
- Education should be provided for all clinicians - In any patient whose condition deteriorates
placing NGTs within institutions and across shortly after NGT placement
care settings.
- Education should include competency valida-
r Improve interpretation and communication about
tion for placement, pH measurement, decision the radiograph.
making to determine need for radiographic eval-
uation, documentation of tube placement, and - The radiograph requisition should clearly re-
patient tolerance of the procedure. quest “NGT placement verification” or similar
- Competency-based education should be in place language.
for providers interpreting radiographs to verify - The radiograph report should contain a state-
NGT placement. ment of the tube path, the location of the tube
tip, and confirmation that the tube is positioned
r Use appropriate NGT placement and securing in the desired location and is appropriate for use.
methods.
Future Considerations
- Use the Nose→Earlobe→Xiphoid process→
Midline of the Umbilicus (NEMU) method for Challenges persist surrounding the placement and location
determination of NGT insertion length. verification of NGTs in pediatric patients. The following are
- Document the centimeter marking on the tube, potential solutions to be considered:
where it exits the nose or mouth, once cor-
rect tube placement is confirmed periodically
r Adoption of these recommendations with units and
depending on the policy of the healthcare set- institutions implementing them to meet their specific
ting. needs
- In an NGT with a stylet in place prior to inser-
r Partnering with key stakeholders for technology and
tion, if the NGT has been flushed with sterile product development to allow for placement verifica-
water to facilitate stylet lubrication and removal tion and reverification in real time for the duration of
after insertion, aspirate the entire fill volume of NGT use
sterile water and discard. A second aspiration
r Research on best practice for ongoing reverification
is necessary to obtain gastric secretions for pH of correct NGT placement, including data on fre-
testing. quency of assessment for reverification of correct
NGT location for patients on intermittent feedings
r Measure gastric pH. and/or continuous feedings and/or NGT medication
administration
- Use gastric pH testing as the first-line method r Standardized, mandated, state and federal reporting
for NGT location verification. mechanisms for NGT misplacements, followed by a
- A gastric pH value of 1–5.5 without a change root cause analysis to evaluate the misplacement and
in the patient’s clinical status is indicative of to ascertain opportunities for improved education
gastric placement. and practice
- When used intermittently for enteral feedings
and/or medication administration, establish a Conclusions
schedule for frequency of NGT location confir-
mation. Placement of NGTs in pediatric patients to facilitate provi-
- When used continuously for enteral feeding, de- sion of EN, medication administration, and fluid infusion
termine the need for frequency of confirmation is a common practice performed by nurses without the use
with documentation of NGT location. of technology. Despite the frequency of NGT placement, it
carries the potential of patient harm if the tube is misplaced.
r Consider a radiograph for any patient in whom there Various methods for determining location verification exist.
is any concern for correct NGT placement, such as: Radiography is the gold standard and measurement of
926 Nutrition in Clinical Practice 33(6)

gastric pH is a validated method; however, there is no 19. Mattar MS, al-Alfy AA, Dahniya MH, al-Marzouk NF. Urinary
standardization in the United States on best practice for bladder perforation: an unusual complication of neonatal nasogastric
tube feeding. Pediatr Radiol. 1997;27:858–859.
NGT location verification. The recommendations presented
20. Wallace SC. Data Snapshot: Complications Linked to Iatrogenic En-
here are a necessary first step in establishing best prac- teral Feeding Tube Misplacements. PA Patient Saf Advis. 2017;14:1-60.
tice related to NGT placement verification in the pedi- 21. Kemper C, Northington L, Wilder K, Visscher D. A call to action:
atric patient. The future considerations present numerous the development of enteral access safety teams. Nutr Clin Pract.
opportunities for collaboration between nurse scientists, 2014;29:264–266.
22. National Health Service. Nasogastric tube misplacement: continuing
clinicians, researchers, institutional leaders, policy makers,
risk of death and severe harm. Patient Safety Alert. 2016. NHS/PSA/
and industry partners to improve patient safety related to RE/2016/006. Available at: https://improvement.nhs.uk/news-alerts/na
NGT placement and verification in children. sogastric-tube-misplacement-continuing-risk-of-death-severe-harm/.
Accessed November 7, 2017.
23. National Health Service. Placement devices for nasogastric tube inser-
References tion do not replace initial position checks. Patient Safety Alert. 2013.
1. Healthcare Cost and Utilization Project (HCUP). Overview of NHS/PSA/W/2013/001. Available at: https://www.england.nhs.uk/
the National (Nationwide) Inpatient Sample (NIS). Agency for wp-content/uploads/2013/12/psa-ng-tube.pdf.
Healthcare Research and Quality. 2014. Available at: www.hcup-us. 24. Ellett ML, Cohen MD, Perkins SM, Croffie JM, Lane KA, Austin JK.
ahrq.gov/nisoverview.jsp. Accessed August 24, 2017. Comparing methods of determining insertion length for placing gastric
2. Lyman B, Kemper C, Northington L, et al. Use of temporary tubes in children 1 month to 17 years of age. J Spec Pediatr Nurs.
enteral access devices in hospitalized neonatal and pediatric pa- 2012;17:19–32.
tients in the United States. JPEN J Parenter Enteral Nutr. 2016;40: 25. Klasner AE, Luke DA, Scalzo AJ. Pediatric orogastric and nasogastric
574–580. tubes: a new formula evaluated. Ann Emerg Med. 2002;39:268–272.
3. Cirgin Ellett ML, Cohen MD, Perkins SM, Smith CE, Lane KA, 26. Simons SR, Abdallah LM. Bedside assessment of enteral tube place-
Austin JK. Predicting the insertion length for gastric tube placement ment: aligning practice with evidence. Am J Nurs. 2012;112:40–46; quiz
in neonates. J Obstet Gynecol Neonatal Nurs. 2011;40:412–421. 48, 47.
4. Ellett ML, Croffie JM, Cohen MD, Perkins SM. Gastric tube place- 27. Irving SY, Lyman B, Northington L, Bartlett JA, Kemper C. Nasogas-
ment in young children. Clin Nurs Res. 2005;14:238–252. tric tube placement and verification in children: review of the current
5. Baskin WN. Acute complications associated with bedside placement of literature. Nutr Clin Pract. 2014;29:267–276.
feeding tubes. Nutr Clin Pract. 2006;21:40–55. 28. (NACH) NACH. Blind Pediatric NG Tube Placements Continue to
6. Yardley IE, Donaldson LJ. Patient safety matters: reducing the risks of Cause Harm. Overland Park, KS: Child Health Patient Safety Orga-
nasogastric tubes. Clin Med (Lond). 2010;10:228–230. nization, Inc.; 2012.
7. Daly W. Unusual complication of nasal intubation: report of case. 29. Metheny N. AACN Practice Alert. Initial and ongoing verification of
Anesthesiology. 1953;14:96. feeding tube placement in adults (applies to blind insertions and place-
8. Lind LJ, Wallace DH. Submucosal passage of a nasogastric tube ments with an electromagnetic device). Critl Care Nurse. 2016;36(2):
complicating attempted intubation during anesthesia. Anesthesiology. e8–e13.
1978;49:145–147. 30. Metheny NA, Stewart BJ, Mills AC. Blind insertion of feeding tubes
9. Aronchick JM, Epstein DM, Gefter WB, Miller WT. Pneumotho- in intensive care units: a national survey. Am J Crit Care. 2012;21:
rax as a complication of placement of a nasoenteric tube. JAMA. 352–360.
1984;252:3287–3288. 31. Kleinerman RA. Cancer risks following diagnostic and therapeu-
10. Bankier AA, Wiesmayr MN, Henk C, et al. Radiographic detection tic radiation exposure in children. Pediatr Radiol. 2006;36 Suppl 2:
of intrabronchial malpositions of nasogastric tubes and subsequent 121–125.
complications in intensive care unit patients. Intensive Care Med. 32. Cohen MD, Ellett M. Quality of communication: different patterns of
1997;23:406–410. reporting the location of the tip of a nasogastric tube. Acad Radiol.
11. Metheny NA, Meert KL, Clouse RE. Complications related to feeding 2012;19:651–653.
tube placement. Curr Opin Gastroenterol. 2007;23:178–182. 33. Rollins H, Arnold-Jellis J, Taylor A. How accurate are X-rays to check
12. Crisp CL. Esophageal nasogastric tube misplacement in an infant NG tube positioning? Nurs Times. 2012;108:14–16.
following laser supraglottoplasty. J Pediatr Nurs. 2006;21:454–455. 34. Lee KH, Cho HJ, Kim EY, et al. Variation between residents and
13. Ellett ML, Cohen MD, Croffie JM, Lane KA, Austin JK, Perkins SM. attending staff interpreting radiographs to verify placement of nutri-
Comparing bedside methods of determining placement of gastric tubes tion access devices in the neonatal intensive care unit. Nutr Clin Pract.
in children. J Spec Pediatr Nurs. 2014;19:68–79. 2015;30:398–401.
14. De Espinosa H, Garcı́a de Paredes C. Traumatic perforation of the 35. Don S. Pediatric digital radiography summit overview: state of confu-
pharynx in a newborn baby. J Pediatr Surg. 1974;9:247–248. sion. Pediatr Radiol. 2011;41:567–572.
15. Wynne DM, Borg HK, Geddes NK, Fredericks B. Nasogastric tube 36. Gilbertson HR, Rogers EJ, Ukoumunne OC. Determination of a
misplacement into Eustachian tube. Int J Pediatr Otorhinolaryngol. practical pH cutoff level for reliable confirmation of nasogastric tube
2003;67:185–187. placement. JPEN J Parenter Enteral Nutr. 2011;35:540–544.
16. Arslantas A, Durmaz R, Cosan E, Tel E. Inadvertent insertion of 37. Stock A, Gilbertson H, Babl FE. Confirming nasogastric tube position
a nasogastric tube in a patient with head trauma. Childs Nerv Syst. in the emergency department. Pediatr Emerg Care. 2008;24:805–809.
2001;17:112–114. 38. Metheny NA, Pawluszka A, Lulic M, Hinyard LJ, Meert KL. Test-
17. Creel AM, Winkler MK. Oral and nasal enteral tube placement errors ing placement of gastric feeding tubes in infants. Am J Crit Care.
and complications in a pediatric intensive care unit. Pediatr Crit Care 2017;26:466–473.
Med. 2007;8:161–164. 39. Kemper C, Haney B, Oschman A, et al. Use of pH testing to verify
18. Glüer S, Schmidt AI, Jesch NK, Ure BM. Laparoscopic repair of enteral feeding tube placement in neonates. Submitted to Advances in
neonatal gastric perforation. J Pediatr Surg. 2006;41:e57–58. Neonatal Care, 2018.
Irving et al 927

40. Faden HS, Ma C. Trends in oral antibiotic, proton pump inhibitor, 45. Drug USFa, (FDA) A, Division of Industry and Consumer Ed-
and histamine 2 receptor blocker prescription patterns for children ucation. Feeding Tube Placement Systems, Letter to Health Care
compared with adults: implications for clostridium difficile infection in Providers. Silver Spring, MD: U.S. Food and Drug Administration
the community. Clinical Pediatrics. 2016;55: 712–716. 2018. Available at: https://www.fda.gov/MedicalDevices/Safety/Lett
41. National Health Service. Nasogastric tube misplacement: continuing erstoHealthCareProviders/ucm591838.htm.
risk of death and severe harm. Patient Safety Alert. 2016. NHS/PSA/ 46. Kline AM, Sorce L, Sullivan C, Weishaar J, Steinhorn DM. Use
RE/2016/006. Available at: https://improvement.nhs.uk/news-alerts/n of a noninvasive electromagnetic device to place transpyloric feeding
asogastric-tube-misplacement-continuing-risk-of-death-severe-harm/. tubes in critically ill children. Am J Crit Care. 2011;20:453–459;
Accessed March 20, 2018. quiz 460.
42. U.S. Food and Drug Administration. MAUDE – Manufacturer and 47. Brown AM, Perebzak C, Handwork C, Gothard MD, Nagy K. Use of
user facility device experience. us department of health and hu- electromagnetic device to insert postpyloric feeding tubes in a pediatric
man services. 2017. Available at: https://www.accessdata.fda.gov/scripts intensive care unit. Am J Crit Care. 2017;26:248–254.
/cdrh/cfdocs/cfmaude/search.cfm. Accessed August 22, 2017. 48. Atalay YO, Aydin R, Ertugrul O, Gul SB, Polat AV, Paksu MS. Does
43. Bryant V, Phang J, Abrams K. Verifying placement of small-bore feed- bedside sonography effectively identify nasogastric tube placements in
ing tubes: electromagnetic device images versus abdominal radiographs. pediatric critical care patients? Nutr Clin Pract. 2016.
Am J Crit Care. 2015;24:525–530. 49. Chau JP, Lo SH, Thompson DR, Fernandez R, Griffiths R. Use
44. Metheny NA, Meert KL. Update on effectiveness of an electromagnetic of end-tidal carbon dioxide detection to determine correct place-
feeding tube-placement device in detecting respiratory placements. Am ment of nasogastric tube: a meta-analysis. Int J Nurs Stud. 2011;48:
J Crit Care. 2017;26:157–161. 513–521.

Potrebbero piacerti anche