Вы находитесь на странице: 1из 129

PEDIATRIC

SELF-ASSESSMENT PROGRAM

2 0 1 6 B O OK 2

PEDIATRIC
CRITICAL CARE
Series Editors
Marcia L. Buck, Pharm.D., BCPPS, FCCP, FPPAG
Kalen B. Manasco, Pharm.D., BCPS

AMERICAN COLLEGE OF CLINICAL PHARMACY


IMPORTANT INFORMATION ON THE RELEASE OF
PedSAP 2016 BOOK 2 Pediatric Critical Care
TESTING

BCPPS test deadline: 11:59 p.m. (Central) on January 17, 2017.


ACPE test deadline: 11:59 p.m. (Central) on September 14, 2019.

Online Errata: Follow this link to check for any changes or updates to this Pediatric Self-Assessment Program release. Be sure to
check the online errata before submitting a posttest.

You may complete one or all modules for credit. Tests may not be submitted more than one time. For information on passing lev-
els, assignment of credits, and credit reporting, see Continuing Pharmacy Education and Recertification Instructions page for
each module.

Important Notice on BCPPS Recertification: Submitting a required posttest for BCPPS recertification attests that you have com-
pleted the test as an individual effort and not in collaboration with any other individual or group. Failure to complete this test as
an individual effort may jeopardize your ability to use PedSAP for BCPPS recertification.

BOOK FORMAT AND CONTENT

E-Media Format: All purchasers of this PedSAP book also have access to the e-media version. Follow these instructions to load
the text and self-assessment questions in this book onto your e-reader, tablet, or Android phone.

Electronic annotation: The online format of this book (PDF) can be saved to the desktop or printed. The latest version of Adobe
Reader (available free) offers functionality such as highlighting or adding sticky notes to the text.

Hyperlinks: This book contains both internal and external hypertext links (visible as underlined text in the print book). These
links are active in the Online and e-Media versions of the book.

NOTE: To facilitate further learning and research, this publication incorporates live hyperlinks to websites administered by other
organizations. The URLs provided are those of third parties not affiliated in any way with ACCP. ACCP assumes no liability for
material downloaded from or accessed on these websites. It is the responsibility of the reader to examine the copyright and
licensing restrictions of linked pages and to secure all necessary permissions.

Abbreviations, Laboratory Values: This table, which is also reached by links at the beginning of each chapter, lists selected
abbreviations and reference ranges for common laboratory tests that can be used as a resource in completing the self-assess-
ment questions.

NOTE: The editors and publisher of PedSAP recognize that the development of this volume of material offers many opportunities
for error. Despite our best efforts, some errors may persist into publication. Drug dosage schedules are, we believe, accurate and
in accordance with current standards. Readers are advised, however, to check package inserts for the recommended dosages
and contraindications. This is especially important for new, infrequently used, and highly toxic drugs.
Director of Professional Development: Nancy M. Perrin, M.A., CAE
Associate Director of Professional Development: Wafa Y. Dahdal, Pharm.D., BCPS
Recertification Project Manager: Edward Alderman, B.S., B.A.
Medical Editor: Kimma Sheldon, Ph.D., M.A.
Information Technology Project Manager: Brent Paloutzian, A.A.S.

For ordering information or questions, write or call:


Pediatric Self-Assessment Program
American College of Clinical Pharmacy
13000 W. 87th St. Parkway
Lenexa, KS 66215-4530
Telephone: (913) 492-3311
Fax: (913) 492-4922
E-mail: accp@accp.com

Library of Congress Control Number: 2016947101


ISBN-13: 978-1-939862-35-8 (PedSAP 2016 Book 2, Pediatric Critical Care)

Copyright 2016 by the American College of Clinical Pharmacy. All rights reserved. This book is protected by copyright. No part
of this publication may be reproduced, stored in a retrieval system, or transmitted, in any form or by any means, electronic or
mechanical, including photocopy, without prior written permission of the American College of Clinical Pharmacy.

To cite PedSAP properly:

Authors. Chapter name. In: Buck ML, Manasco KB, eds. Pediatric Self-Assessment Program, 2016 Book 2. Pediatric Critical Care.
Lenexa, KS: American College of Clinical Pharmacy, 2016:page range.

PedSAP is a registered trademark of the American College of Clinical Pharmacy.

Pediatric
Self-Assessment
Program
TABLE OF CONTENTS
Pediatric Critical Care I. . . . . . . . . . . . . . . . . . . . . . . . . . 1 Trauma
Pediatric Critical Care I Panel. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
By Joseph M. LaRochelle, Pharm.D., BCPPS
Introduction to Traumatic Brain Injury . . . . . . . . . . . . . . . . . . . . 81
ARDS and Ventilator-Associated Events Cerebral Perfusion Pressure . . . . . . . . . . . . . . . . . . . . . . . . . . . . 81
Sedation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
By Jennifer L. Morris, Pharm.D., FCCM, BCPS, BCPPS Other Therapies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
LeMechanical Ventilation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7 Emerging Therapies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
Acute Lung Injury and PARDS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9 Introduction to Burns . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
Toward Evidence-Based Pharmacotherapy Management Initial Management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
of ALI and ARDS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11 Hypermetabolic State . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
Ventilator-Associated Events . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20 Infections . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
Current Quality of Care Surveillance . . . . . . . . . . . . . . . . . . . . . . 22 Pain Management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
Antibiotic Stewardship for IVAC Complications and VAP . . . . . 25 Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
Strategies for Prevention of VAEs . . . . . . . . . . . . . . . . . . . . . . . . . 26 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28 Self-Assessment Questions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
Self-Assessment Questions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
Clinical and Practice Updates I. . . . . . . . . . . . . . 95
Hemodynamic Instability Clinical and Practice Updates I Panel. . . . . . . . . . . . . . . . . . . . . 97

By Regine L. Caruthers, Pharm.D.


Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37 Interactive Patient Case: Severe Acute
Hypovolemic Shock . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38 Asthma (Status Asthmaticus)
Obstructive Shock . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
By Laura Lo Castro Bio, Pharm.D., BCPS
Cardiogenic Shock . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
Distributive Shock . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40 Interactive Case . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 101
Hypertensive Urgency References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102
and Emergency . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42 Self-Assessment Questions. . . . . . . . . . . . . . . . . . . . . . . . . . . . 104
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
Guideline Review: Diabetic Ketoacidosis and
Self-Assessment Questions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
Hyperglycemic Hyperosmolar State
By Elizabeth S. Goswami, Pharm.D., BCPS, BCPPS; and Susan Warrington,
Pediatric Critical Care II. . . . . . . . . . . . . . . . . . . . . . . 49
Pharm.D., BCPPS
Pediatric Critical Care II Panel. . . . . . . . . . . . . . . . . . . . . . . . . . . 51
Guideline Review . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
Extracorporeal Circuits in Children Self-Assessment Questions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112

By Elizabeth J. Beckman, Pharm.D., BCPS, BCPPS, BCCCP


Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55 Interactive Case: Status Epilepticus
Extracorporeal Membrane Oxygenation . . . . . . . . . . . . . . . . . . 56 By Christina Cox, Pharm.D., BCPS, BCPPS
Continuous Renal Replacement Therapy . . . . . . . . . . . . . . . . . 63
Interactive Case . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117
Extracorporeal Liver Support . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117
Plasmapheresis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
Self-Assessment Questions. . . . . . . . . . . . . . . . . . . . . . . . . . . . 120
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73
Self-Assessment Questions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77

PedSAP 2016 Book 2 Pediatric Critical Care iii Table of Contents


A MESSAGE FROM THE EDITORS
Welcome to the Pediatric Self-Assessment Program (Ped- from ACCPs other self-assessment programs, PedSAP mod-
SAP), a new recertification component for the Board Certified ules come in two formats: (1) the traditional review chapters;
Pediatric Pharmacy Specialist (BCPPS). ACCP has a long tra- and (2) Clinical and Practice Update modules that incorpo-
dition of offering the best products for continuing pharmacy rate new learning formats such as recorded webinars and
education and pharmacotherapy specialist certification. HTML pages. These new learning formats are hoped to speed
PedSAP continues that tradition by providing the latest in the dissemination of new clinical information and satisfy the
evidence-based information for the practitioner or clinician learning preferences of a wider audience.
working with this special population. Some things have not changed: inside this PedSAP book
In designing this series, the primary goal was to provide you will find user-friendly formatting as well as graphic ele-
updates that would improve clinical pharmacy practice and ments such as patient-care scenarios (demonstrating the
patient outcomes. The process began with a careful review application of concepts), treatment algorithms, descriptions
of the content outline developed by the Board of Pharmacy of pivotal studies that may change practice, and summative
Specialties for the Pediatric Pharmacy Specialty Certification practice points. All releases in this series are available elec-
Examination. The 20162018 PedSAP chapters will therefore tronically, enhancing the portability of this product. Prominent
cover the domains of patient management; practice manage- in each chapter are hyperlinks to reference sources, assess-
ment; information management and education; and public ment tools, guidelines and resources, data compilers such as
health and patient advocacy. Specific content for individual PubMed, and even informational videos. Our hope is that this
releases in this series was organized on the basis of the sys- depth of information, ease of access, and emphasis on clinical
tems and patient-care problems that might be expected of the application will have an immediate and positive impact on the
BCPPS. Finally, calls went out to recruit faculty panel chairs, care of pediatric patients.
authors, and reviewers committed to this new specialty and to We very much appreciate the efforts of all the faculty panel
the board certification process. chairs, authors, and reviewers who lent their time, energy, and
The presentation of information, and its incorporation into expertise to this new series.
practice, was also given careful consideration. In a departure
Marcia L. Buck and Kalen B. Manasco, series editors

PedSAP 2016 Book 2 Pediatric Critical Care iv Table of Contents


Pediatric Critical Care I
Pediatric Critical Care I Panel

Series Editors: Clinical Pharmacist


Department of Pharmacy
Marcia L. Buck, Pharm.D., BCPPS, FCCP, FPPAG
UC Health Northern Colorado-Poudre Valley Hospital
Clinical Coordinator
Fort Collins, Colorado
University of Virginia Childrens Hospital
Professor
Department of Pediatrics Hemodynamic Instability
University of Virginia School of Medicine
Clinical Professor Author
Virginia Commonwealth University School of Pharmacy
Regine L. Caruthers, Pharm.D.
Department of Pharmacy Services
Clinical Pharmacy Specialist, Pediatric Cardiology
University of Virginia Health System
Department of Pharmacy
Charlottesville, Virginia
Adjunct Clinical Assistant Professor of Pharmacy
Kalen B. Manasco, Pharm.D., BCPS College of Pharmacy
Clinical Associate Professor University of Michigan Hospital and Health Centers
Department of Clinical and Administrative Pharmacy Ann Arbor, Michigan
University of Georgia College of Pharmacy
Augusta, Georgia Reviewers
Ashley R. Schappell, Pharm.D., BCPS
Faculty Panel Chair:
Clinical & Unit Based Pharmacist
Joseph M. LaRochelle, Pharm. D., BCPPS Department of Pharmacy
Clinical Associate Professor Nemours/Alfred I. DuPont Hospital for Children
College of Pharmacy Wilmington, Delaware
Xavier University of Louisiana
Michael A. Maccia, Pharm.D, BCPS
Clinical Associate Professor of Pediatrics
Clinical Pharmacist
Louisiana State University Health Sciences
Pharmacy Department
Center School of Medicine
Moses H. Cone Memorial Hospital
New Orleans, Louisiana
Greensboro, North Carolina

ARDS and Ventilator-Associated Events The American College of Clinical Pharmacy and the authors
thank the following individuals for their careful review of the
Author Pediatric Critical Care I chapters:
Jennifer L. Morris, Pharm.D., FCCM, BCPS, BCPPS Ralph H. Raasch, Pharm.D., BCPS
Clinical Pharmacy Specialist, Pediatric Critical Care Associate Professor of Pharmacy (retired)
Department of Pharmacy Division of Practice Advancement and Clinical Education
Texas Childrens Hospital Eshelman School of Pharmacy
Houston, Texas The University of North Carolina at Chapel Hill
Chapel Hill, North Carolina
Reviewers
Jeffrey T. Sherer, Pharm.D., MPH, BCPS
Kristine A. Parbuoni, Pharm.D., BCPPS Clinical Associate Professor
Assistant Professor Department of Clinical Sciences and Administration
Department of Pharmacy Practice University of Houston College of Pharmacy
Loma Linda University School of Pharmacy Houston, Texas
Loma Linda, California
Brandi L. Strauser, Pharm.D., BCPS, BCCCP
DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST
Consultancies: Elizabeth J. Beckman (Lexi-Comp/Wolters/Kluwer Health); Regine L Caruthers (Lexi-Comp/Wolters Kluwer
Health); Joseph M. LaRochelle (ACCP); Lois F. Parker (ASHP/MSHP)

Stock Ownership:

Royalties:

Grants: Elizabeth J. Beckman (ASHP Foundation); Christina Cox (University of South Carolina);

Honoraria: Christina Cox (ACCP, Palmetto Health Richland Childrens Hospital); Jennifer L. Morris (ACCP, Society of Critical Care
Medicine, Pediatric Pharmacy Advocacy Group, Wolters Kluwer)

Other:

Nothing to disclose: Laura Lo Castro Bio, Joshua Caballero, Brooke Clark, Kyle Davis, Ifeyinwa D. Eboh, Elizabeth S. Goswami,
Aaron A. Harthan, Maha O. Kebir, Allison Lardieri, Michael A. Maccia, Kristine Parbuoni, Monica A. Puebla, Sara P. Rooney, Emily
L. Rowe, Ashley R. Schappell, Julie Pingel, Brandi L. Strauser, Andrea Joan Stumpe, Susan Warrington

ROLE OF BPS: The Board of Pharmacy Specialties (BPS) is an autonomous division of the American Pharmacists Association
(APhA). BPS is totally separate and distinct from ACCP. The Board, through its specialty councils, is responsible for specialty
examination content, administration, scoring, and all other aspects of its certification programs. PedSAP has been approved by
BPS for use in BCPPS recertification. Information about the BPS recertification process is available online.

Other questions regarding recertification should be directed to:

Board of Pharmacy Specialties


2215 Constitution Avenue NW
Washington, DC 20037
(202) 429-7591
CONTINUING PHARMACY EDUCATION AND
RECERTIFICATION INSTRUCTIONS
Continuing Pharmacy Education Credit: The American College of Clinical Pharmacy is accredited by the Accreditation
Council for Pharmacy Education (ACPE) as a provider of continuing pharmacy education (CPE).

PedSAP: Target Audience: The target audience for PedSAP 2016 Book 2(Pediatric Critical Care) is pediatric pharmacotherapy spe-
cialists and advanced-level clinical pharmacists caring for pediatric patients with several acute conditions and complications of
the pulmonary, cardiovascular, endocrine, and neurologic systems.

Available CPE credits: Purchasers who successfully complete all posttests for PedSAP 2016 Book 2 (Pediatric Critical Care) can
earn 13.0 contact hours of continuing pharmacy education credit. The universal activity numbers are as follows: Pediatric Critical
Care I 0217-0000-16-021-H01-P, 3.5 contact hours; Pediatric Critical Care II 0217-0000-16-022-H01-P, 4.5 contact hours; and
Clinical and Practice Updates 0217-0000-16-170-H01-P, 5.0 contact hours. You may complete one or all available modules for
credit. Tests may not be submitted more than once.

BCPPS test deadline: 11:59 p.m. (Central) on January 17, 2017.


ACPE test deadline: 11:59 p.m. (Central) on September 14, 2019.

Posttest access: Go to www.accp.com and sign in with your e-mail address and password. Technical support is available from
8 a.m. to 5 p.m. (Central) weekdays by calling (913) 492-3311. PedSAP products are listed under My Online Products on your My
Account page.

BCPPS Recertification Credit: To receive BCPPS recertification CPE credit, a PedSAP posttest must be submitted within the
4-month period after the books release. The first page of each print and online book lists the deadline to submit a required
posttest for BCPPS recertification credit. Only completed tests are eligible for credit; no partial or incomplete tests will be pro-
cessed. Tests may not be submitted more than once. The passing point for BCPPS recertification is based on an expert analysis
of the items in each posttest module.

ACPE CPE Credit: To receive ACPE CPE credit for a PedSAP module, a posttest must be submitted within the 3-year period after
the books release. The appropriate CPE credit will be awarded for test scores of 50% and greater.

Credit Assignment and Reporting: All required posttests that meet the 50% score standard will be immediately awarded the
appropriate ACPE CPE credit. Earned credits will be transmitted within 24 hours to www.mycpemonitor.net and should appear on
statements of credit within 3 business days.

Required posttests that are submitted before the BCPPS test deadline and that meet the passing point set by statistical analysis
will earn BCPPS recertification credits. These credits will be posted within 30 days after the BCPPS test deadline. For statements
of CPE credit, visit www.mycpemonitor.net.

All BCPPS recertification credits are forwarded by ACCP to the Board of Pharmacy Specialties (BPS). Questions regarding the
number of hours required for BCPPS recertification should be directed to BPS at (202) 429-7591 or www.bpsweb.org. The ACCP
Recertification Dashboard is a free online tool that can track recertification credits as they are earned through ACCP and schedule
new opportunities for credits from upcoming ACCP professional development programs.

Posttest Answers: The explained answers with rationale and supporting references will be posted 1 week after the BCPPS
test deadline and will be available to anyone who has either submitted a posttest or waived his or her right to receive credit (see
below) from a posttest. Go to www.accp.com and sign in with your e-mail address and password. Click the PedSAP book on your
My Account page and you will see a link to the explained answers.

Test Waivers: To access the explained answers without submitting a posttest, sign in to your My Account page, select the
PedSAP book, and click on the waiver link for that module. By completing the waiver form for a module, you waive the opportu-
nity to receive CPE credit for that module. After you submit a waiver, you will see a link to the PDF file that contains the answers
for the module you waived. Answers will be available starting 1 week after the BCPPS test deadline.

ACSAP 2016 Book 2 Pediatric Critical Care 6 Program Instructions


ARDS and Ventilator-Associated
Events
By Jennifer L. Morris, Pharm.D., FCCM, BCPS, BCPPS

Reviewed by Kristine A. Parbuoni, Pharm.D., BCPPS; and Brandi L. Strauser, Pharm.D., BCPS, BCCCP

LEARNING OBJECTIVES

1. Using the definitions for acute respiratory distress syndrome (ARDS), classify a patients pulmonary morbidity.
2. Analyze available data to design an appropriate pharmacotherapy plan, including exogenous pulmonary surfactant or the
use of corticosteroids, for pediatric patients with ARDS.
3. Analyze the current definition of ventilator-associated pneumonia to determine the need for surveillance, and measure
quality of care in pediatric ICU patients.
4. Evaluate deficiencies in current surveillance definitions and justify expanded surveillance in pediatric units to include
other ventilator-associated conditions.
5. For ventilator-associated infections in the pediatric ICU, design antimicrobial therapy plan that uses the narrowest-spec-
trum antibacterial agents for a duration that minimizes antibacterial exposure.
6. Justify a prevention strategy for ventilator-associated events in the pediatric ICU including, but not limited to, implemen-
tation of a ventilator-associated pneumonia bundle.

MECHANICAL VENTILATION
ABBREVIATIONS IN THIS CHAPTER
Invasive positive pressure ventilation is used in the treatment of crit-
ALI Acute lung injury
ically ill pediatric patients for a variety of indications that may arise
ARDS Acute respiratory distress syndrome
from a pulmonary, cardiac, neurologic, or neuromuscular condition.
IVAC Infection-related ventilator-associ-
ated condition Respiratory failure, either from inadequate oxygenation or inade-
MDRGN Multidrug-resistant gram-negative quate ventilation, may be caused by pulmonary or extrapulmonary
organisms conditions. Patients with significant cardiovascular dysfunction may
OI Oxygenation index benefit from mechanical ventilation, which may decrease both the
PALICC Pediatric Acute Lung Injury work of breathing and the myocardial oxygen demand. Neurologic
Consensus Conference and neuromuscular disorders can result in loss of ventilatory drive,
PARDS Pediatric acute respiratory dis- ventilatory muscle weakness, loss of protective reflexes, and need
tress syndrome for induced hyperventilation, all of which may be supported by posi-
PF ratio Ratio of Pao2 to Fio2 tive pressure ventilation (Venkataraman 2011).
PVAP Possible ventilator-associated
pneumonia Treatment of the Mechanically Ventilated Patient
SF ratio Ratio of saturation of arterial oxy- One pharmacist role in the treatment of mechanically ventilated
gen to fraction of inspired oxygen
patients is the management of pharmacotherapy used to facilitate
SRMD Stress-related mucosal disease
patient-ventilator synchrony and optimize patient comfort. Pediatric
VAC Ventilator-associated condition
patients requiring mechanical ventilation generally need some degree
VAE Ventilator-associated event
of pain and sedation management. Agents such as benzodiazepines,
VAP Ventilator-associated pneumonia
opioids, and dexmedetomidine may facilitate comfort, but their
use must be balanced with minimization of the associated adverse
events. This is usually achieved by identifying patient-specific goals

PedSAP 2016 Book 2 Pediatric Critical Care 7 ARDS and Ventilator-Associated Events
In addition to facilitating patient comfort and the phar-
VAT Ventilator-associated tracheobronchitis
macologic management of the primary disorder resulting
VTE Venous thromboembolism
in the need for positive pressure ventilation, pharmacists
have a role in preventing health careassociated conditions.
Table of other common abbreviations.
This includes infection prevention as well as prevention of
stress-related mucosal disease (SRMD) and venous thrombo-
embolism (VTE).
and using a standardized approach to drug therapy that is
Stress-related mucosal disease may develop in acutely ill
based on the predefined goal. Although the only randomized
patients secondary to an inflammatory, erosive insult to the
controlled trial of protocolized sedation in mechanically ven-
upper GI tract. Of most concern is the development of overt,
tilated, critical care pediatric patients found no difference in
clinically significant GI bleeding, which may worsen patient
duration of mechanical ventilation, several secondary out-
outcomes. Box 1-1 lists risk factors for SRMD identified in
comes were improved with this approach, including fewer
adult and pediatric hospitalized patients.
episodes of skin breakdown, fewer days of opioid exposure,
fewer sedative class exposures, and more study days awake
and calm (Curley 2015). Although no guidelines exist for treat-
ing the pain, agitation, and delirium of pediatric patients, the Box 1-1. Risk Factors Associated with
tenets of the adult guidelines can be generally applied to the Upper GI bleeding
care of critically ill pediatric patients to optimize therapy with Independent Risk Factors
these agents (Barr 2013). Pediatric:
Coagulopathy a

PRISM score 10
Respiratory failureb

In mechanically ventilated pediatric patients:


High pressure ventilation (positive inspiratory
BASELINE KNOWLEDGE STATEMENTS pressure 25 cm H2O)
Organ failure
Readers of this chapter are presumed to be familiar
with the following: Adult:
Basic ventilator terminology, including settings and Coagulopathy
monitoring parameters
Respiratory failure
Shock
Concepts and management of pain, agitation, and Multiple trauma
delirium in the critically ill patient
Factors Contributing to Cumulative Riskc
Knowledge of the pharmacology of pulmonary
Acute hepatic failure
surfactants and corticosteroids Acute renal failure
Knowledge of the organisms commonly associated Anticoagulation
with health careassociated pneumonia and Burn injury (> 35% total body surface area)
ventilator-associated pneumonia and appropriate High-dose corticosteroids (> 250 mg/day of hydrocor-
empiric therapy tisone)
Knowledge of antibiotic therapy including spec- History of GI bleed
trum of activity and dose-optimization strategies Hypotension
Low intrinsic gastric pH
Knowledge of prophylactic strategies for both
Major surgery (> 4 hr)
stress-related mucosal bleeding and venous
Sepsis
thromboembolism
Severe head or spinal cord injury
Table of common pediatric laboratory reference values. a
Studies have used several definitions of coagulopathy, includ-
ing prolonged PTT, prolonged PT, INR > 1.5, and Plt < 100,000/
mm3.
ADDITIONAL READINGS b
Need for mechanical ventilation.
The following free resources have additional back-
c
Identified in adult and/or pediatric studies.
ground information on this topic: Information from: ASHP Commission on Therapeutics. ASHP
therapeutic guidelines on stress ulcer prophylaxis. Am J
CDC. National Healthcare Safety Network [homep- Health Syst Pharm 1999;56:347-79; Chaibou M, Tucci M, Duga
age on the Internet]. MA, et al. Clinically significant upper gastrointestinal bleed-
ing acquired in pediatric ICU: a prospective study. Pediatrics
Institute for Healthcare Improvement. How-to
1998;102:933-8; and Reveiz L, Guerrero-Lozano R, Camacho
Guide: Prevent Ventilator-Associated Pneumonia A, et al. Stress-ulcers, gastritis, and gastrointestinal bleed-
[homepage on the Internet]. ing prophylaxis in critically ill pediatric patients: a systematic
review. Pediatr Crit Care Med 2010;11:124-32.

PedSAP 2016 Book 2 Pediatric Critical Care 8 ARDS and Ventilator-Associated Events
Guidelines for which pediatric ICU (PICU) patients should ACUTE LUNG INJURY AND PARDS
receive pharmacologic prophylaxis are unavailable, and adult Pathophysiology
guidelines were last published in 1999, although an update is
Acute respiratory distress syndrome (ARDS) is the clini-
expected in the near future. Given the available data and the
cal syndrome that occurs secondary to the development of
adult guidelines, it is reasonable to recommend pharmaco-
noncardiogenic pulmonary edema. Pulmonary edema devel-
logic prophylaxis to prevent upper GI bleeding from SRMD
ops secondary to breakdown of the permeability barrier in
in PICU patients who have one independent risk factor or at
the alveoli, in the setting of inflammatory and coagulation
least two cumulative risk factors (ASHP 1999). Prophylactic
dysregulation. When functioning properly, the permeability
strategies, including histamine-2 receptor antagonists and
barrier maintains separation between the fluid (capillaries)
proton pump inhibitors, have proved useful in maintaining a
and the air (alveoli) compartments within the lung and allows
gastric pH greater than 4 when compared with placebo or no
for appropriate gas exchange. With dysfunction, patients
treatment. However, no effect on mortality or decreased risk
develop the clinical symptoms of ARDS including hypoxemia,
of clinically significant bleeding was seen in pediatric stud-
changes on chest radiography, increase in physiologic dead
ies (Reveiz 2010). Therefore, it is important to balance the
space, and decrease in functional residual capacity and lung
benefits of SRMD prophylaxis with the potential risks (e.g.,
compliance. This pathophysiology usually results in the need
increased risk of infection, decreased bone mineral density).
for respiratory support to improve both gas exchange and
The incidence of VTE, including deep venous thrombosis
work of breathing (Sapura 2015).
and pulmonary embolism, has increased in hospitalized pedi-
Two phases of the syndrome have been described, given
atric patients (Sharathkumar 2012). This increase is postulated
the pathologic findings during clinical progression and the
to be caused by enhanced surveillance, as well as advanced
changes in pulmonary pathophysiology. The exudative phase,
management of complex medical conditions leading to
or early phase, of the ARDS is a state of overwhelming expres-
improved mortality. There appears to be a bimodal distribu-
sion of proinflammatory cytokines, neutrophil activation, and
tion of pediatric age groups (i.e., infants and adolescents) at
coagulation dysregulation. The irreversible fibroproliferative
higher risk of developing VTE. Other risk factors identified
phase, or late phase, occurs around day 7 of illness in those
in a hospitalized pediatric cohort include higher BMI, direct
who do not clinically improve. This phase is characterized by
admission to the ICU, bacteremia, central venous access,
fibrosis and loss of alveolar structure (Deal 2008).
immobilization, oral contraceptive exposure, length of stay
longer than 7 days, and mechanical ventilation. According to
a multiple regression analysis, the factors that continued to Prevalence in Pediatric Patients
be significantly associated with risk were direct admission to Although less prevalent than in adults, ARDS and its asso-
the ICU, bacteremia, central venous access, immobilization, ciated morbidity and mortality are regularly encountered in
oral contraceptive exposure, and length of stay longer than the pediatric critical care population. The reported incidence
7 days (Sharathkumar 2012). of ARDS in pediatric patients is 212.8 cases per 100,000
In adult inpatients, use of pharmacologic thrombo- person-years compared with the 17.981 cases per 100,000
prophylaxis with the low-molecular-weight heparins and person-years reported in adults. From 21% to 74% of pediatric
unfractionated heparin minimizes the risk of VTE and its asso- patients are reported to have preexisting illness (commonly
ciated poor outcomes, and thromboprophylaxis is routinely immunodeficiency) before developing ARDS. Mortality rates
used. However, the same supporting data do not exist for pedi- appears to be lower in pediatric patients (18%27%), although
atric patients. Although broad-scale, systematic investigation some studies report mortality closer to the adult rate (27%
of these prophylactic strategies is needed, it is clinically appro- 45%). Pediatric patient factors associated with an increased
priate to implement a risk assessment to identify high-risk risk of mortality from ARDS include immunodeficiency and
patients (Reiter 2012; Sharathkumar 2012). In most pediat- African American ethnicity. Although the risk of developing
ric patients identified as high risk, mechanical prophylaxis of ARDS appears to be higher in boys, the mortality risk is no
sequential compression devices can be implemented safely. higher than in girls (Khemani 2015). The patients age may
Pharmacologic prophylactic strategies can be implemented alter long-term morbidity because of the potential impact of
according to institution-specific protocols for high-risk patients ARDS on lung development in children (Sapura 2015).
with favorable risk-benefit for prophylactic anticoagulation. Underlying causes for developing this syndrome may arise
As noted previously, the need for mechanical ventilation from either direct injury to the alveolar epithelium or injury to the
may arise from either pulmonary or extrapulmonary condi- capillary endothelium from conditions such as sepsis, pneumo-
tions. Pulmonary morbidities in critically ill pediatric patients nia, trauma, and submersion injury. In adult patients with ARDS,
may result in the need for invasive mechanical ventilation or the most commonly associated clinical condition is sepsis; in
may be caused by this invasive supportive therapy. Two such pediatric patients it is respiratory infection. Respiratory viruses
conditions are pediatric acute respiratory distress syndrome are much more likely to result in ARDS in pediatric patients than
(PARDS) and ventilator-associated pneumonia (VAP). in adult; ARDS arising from this cause may have a histologic

PedSAP 2016 Book 2 Pediatric Critical Care 9 ARDS and Ventilator-Associated Events
pattern of injury different from other causes. Recent evidence parameters on the PF ratio by including positive end-expira-
suggests that the associated clinical cause alters outcomes tory pressure (PEEP) or continuous positive airway pressure in
(Sapura 2015). There are few controlled trials in patients with the definition. Impaired oxygenation was further delineated as
ARDS, and these are generally underpowered to be conclusive; mild, moderate, or severe. The term ALI was removed from the
however, epidemiologic studies have shown improvement in definition and replaced by the mild category. In addition, in the
ARDS mortality rates (Bembea 2015; Randolph 2009). absence of common risk factors for ARDS, the Berlin definition
excludes hydrostatic pulmonary edema from causes such as
Definition and Classification of PARDS heart failure and fluid management.
Initially reported in 1967, characterization of the adult respi- Risk factors for ARDS include direct lung injury such as
ratory distress syndrome and subsequently renamed ARDS pneumonia, aspiration, pulmonary contusions, inhalation
has undergone several revisions. The American-European injury, pulmonary vasculitis, and submersion injury, as well as
Consensus Conference (AECC) published the first consen- indirect lung injury such as sepsis, major trauma, pancreati-
sus definition in 1994; it delineated requirements for defining tis, transfusion-related, severe burn injury, and drug overdose
hypoxemia by the ratio of partial pressure of arterial oxygen to (ESICM 2012; Ferguson 2012). Table 1-1 details the AECC and
fraction of inspired oxygen (Pao2/Fio2, or PF ratio), the onset Berlin definitions of ARDS.
of hypoxemia, and chest radiographic findings. This definition Although using the Berlin definition may be reasonable,
also established the umbrella category of acute lung injury given the lack of pediatric-specific definitions, it may fail
(ALI). Establishing a consensus definition was highly useful to consider the pulmonary changes that occur throughout
in advancing ARDS clinical care and research; however, there development or how management strategies may be altered
were areas for optimization within this definition. (Sapura 2015; Yehya 2015; Cheifetz 2011). Because of the
The subsequent definition, called the Berlin definition, tried to potential issues with the adult-focused definition, the Pediatric
address these areas. In 2012, the European Society of Intensive Acute Lung Injury Consensus Conference (PALICC) sought to
Care Medicine convened a task force to develop a subsequent develop a pediatric-specific definition for PARDS (Figure 1-1).
consensus definition. The Berlin definition expanded the ARDS This definition incorporates measures of hypoxemia including
diagnostic criteria by clarifying the time interval for the onset oxygenation index (OI), oxygenation saturation index (OSI), and
of hypoxemia and minimizing the effect of other ventilator ratio of saturation of arterial oxygen to fraction of inspired oxy-
gen (SF ratio) as markers of oxygenation impairment.

Table 1-1. ARDS Definition Criteria

Parameter AECC 1994 Berlin 2012

Timing Acute Onset within 1 wk of:


Known clinical insult OR
New/worsening respiratory symptoms
Chest imaging Bilateral infiltrates seen on frontal Bilateral opacities that are not fully explained by effusions, lobar/
view lung collapse, or nodules
Pulmonary artery wedge 18 mm Hg Removed from definition
pressure If not measured, no clinical evidence
of left atrial hypertension
Presence of risk factors Not addressed Use to determine need to rule out hydrostatic edema
Origin of edema Not addressed In the absence of risk factors, hydrostatic edema should be excluded
by objective assessment (e.g., echocardiography)
ALI category PF ratio < 300 ALI term removed
Oxygenation PF ratio 200 Mild: PF ratio 201300 AND PEEP or CPAP 5 cm H2O
Moderate: PF ratio 101200 AND PEEP 5 cm H2O
Severe: PF ratio 100 AND PEEP 5 cm H2O

ALI = acute lung injury; ARDS = acute respiratory distress syndrome; CPAP = continuous positive airway pressure; PEEP = positive
end-expiratory pressure; PF ratio = Pao2/Fio2.
Information from Ferguson ND, Fan E, Camporota L, et al. The Berlin definition of ARDS: an expanded rationale, justification, and sup-
plementary material. Intensive Care Med 2012;38:1573-82; and ESICM. ARDS Definition Task Force. Acute respiratory distress
syndrome: the Berlin definition. JAMA 2012;307:2526-33.

PedSAP 2016 Book 2 Pediatric Critical Care 10 ARDS and Ventilator-Associated Events
Findings consistent with a new infiltrate(s)
secondary to acute parenchymal lung disease
Chest imaging
criteria
Patient with CHD, CLD, or LV dysfunction:
- New infiltrate from baseline

Non-invasive ventilation:
Acute onset within
Full face-mask BiPAP or CPAP 5 cm H2O with
7 days and
a:
pulmonary edema
PF ratio 300 OR
not fully explained
SF ratio 264
by cardiac failure
fluid overloada
Invasive ventilation:
Mild OI 4 to < 8 OR OSI 5 to < 7.5
Moderate OI 8 to < 16 OR OSI 7.5 to < 12.3
Severe OI 16 OR OSI 12.3

Patient with CHD:


Oxygentation Acute deterioration that meets the oxygenation
criteria criteria not explained by underlying
cardiac disease

Patient with CLD:


Acute deterioration that meets the oxygenation
criteria that is not consistent with baseline
oxygenation requirement

Patient with LV Dysfunction:


Acute deterioration that meets the oxygenation
criteria not explained by left ventricular
dysfunction

Figure 1-1. Pediatric ARDS definition.


a
Excludes patients with lung disease unique to the perinatal period such as surfactant deficiency, meconium aspiration, and
congenital diaphragmatic hernia.
BiPAP = bilevel positive airway pressure; CPAP = continuous positive airway pressure; CHD = cyanotic heart disease; CLD = chronic
lung disease; LV = left ventricular; OI = oxygenation index [(FiO2 mean airway pressure 100)/PaO2]; OSI = oxygen saturation index
([FiO2 mean airway pressure (Paw) 100)/SpO2]; PARDS = pediatric ARDS; PEEP = positive end-expiratory pressure; PF ratio =
PaO2/FiO2; SF ratio = SaO2/FiO2.
Information from Khemani RG, Smith LS, Zimmerman JJ, et al. Pediatric acute respiratory distress syndrome: definition, incidence,
and epidemiology: proceedings from the pediatric acute lung injury consensus conference. Pediatr Crit Care Med 2015;16:S23-S40.

There are several reasons to use OI and OSI rather than applied, but neither definition should be applied to patients
PEEP in the definition of PARDS. First, there is known vari- with perinatal-related respiratory failure (Khemani 2015).
ability in clinical ventilator management. Second, pediatric
intensivists may be less likely to use higher PEEP than their
adult colleagues. Third, PEEP is unavailable in high-fre-
TOWARD EVIDENCE-BASED
quency oscillatory ventilation, which is commonly used in PHARMACOTHERAPY MANAGEMENT
PARDS. In addition, the OI may better delineate the mortal- OF ALI AND ARDS
ity risk in pediatric patients than the PF ratio. The OSI and SF Using an evidence-based approach to managing ARDS can be
ratio are offered as alternatives to the OI and PF ratio, respec- difficult. Contradictory results among studies with small sam-
tively, when pulse oximetry is available but an arterial blood ple sizes for this heterogeneous syndrome make it difficult to
gas is not. There is no clearly defined upper age limit for when discern which pharmacologic therapies, if any, should be used
the PARDS definition versus the Berlin definition should be in these critically ill patients. Although the pathophysiology

PedSAP 2016 Book 2 Pediatric Critical Care 11 ARDS and Ventilator-Associated Events
of ARDS is likely similar between adults and children, there investigations. Subgroups with decreased mortality included
are also likely subtle differences because of the developmen- those with direct lung injury, including lung injury secondary
tal changes that occur in the lung parenchyma and immune to pneumonia or aspiration, and those with severe direct lung
system. These differences may be behind the variability in injury, defined as having a mean baseline PF ratio of 100
treatment outcomes between these two patient populations. 29.4 (Taut 2008). Subsequent prospective clinical trials of
Many pulmonary-specific and nonpulmonary-specific ther- both adult and pediatric patients have failed to achieve mor-
apies for managing ARDS have been evaluated; this chapter tality benefit in patients with direct lung injury using either
focuses on exogenous surfactant and corticosteroids. Much rSP-C or calfactant, though the calfactant study was dis-
clinical controversy exists concerning the use of these ther- continued secondary to slow enrollment before reaching the
apies (Tamburro 2015). The proceedings from the PALICC, predetermined sample size (Willson 2015, 2013; Spragg 2011).
convened in October 2012, address in further detail additional A review of clinicaltrials.gov shows no actively enrolling or
pulmonary-specific and nonpulmonary-specific pharmacother- ongoing investigations into the use of pulmonary surfactant in
apy, as well as ventilator management strategies. A complete ARDS. One pediatric investigation of calfactant evaluated all-
review of the PALICC is beyond the scope of this chapter. For cause mortality, duration of mechanical ventilation, PICU and
further details regarding the nonpharmacologic and pharma- hospital length of stay, and effect on oxygenation in patients
cologic management options (e.g., inhaled nitric oxide, inhaled with hematopoietic stem cell transplantation and acute leu-
prostacyclins, bronchodilators, diuretics, neuromuscular block- kemia and lymphoma with ALI; this study is complete, but
ing agents) see the published proceedings (PALICC 2015). results have not yet been reported (PSU 2015). Pending these
data, and using results from previous investigations, patients
Exogenous Surfactant with immunocompromise may be a reasonable group in
Direct injury to the alveoli, together with the intrusion of fluid which to attempt pulmonary surfactant.
into the alveolar airspace, is likely to result in pulmonary sur-
factant dysfunction or washout. The success of exogenous Pharmacoeconomic Considerations
pulmonary surfactant in respiratory distress syndrome of the To date, no pharmacoeconomic evaluations of surfactant
perinatal period has fueled interest in its use in ARDS. Many therapy in ARDS have been published, but cost must be con-
studies have attempted to delineate surfactants benefit in sidered when contemplating the use of this therapy for this
ARDS, with underwhelming results. Despite some positive indication. Of the surfactants investigated for this use, only
outcomes reported in the literature (Table 1-2; Table 1-3), sur- beractant, calfactant, and poractant alfa are commercially
factant investigations have varied greatly in surfactant product, available in the United States. However, the calfactant prod-
dose, routes of administration, population studied, and ARDS uct used in the most recent adult and pediatric study is not
definition used. In addition, although most studies have shown the commercially available product (Willson 2015, 2013).
improvements in oxygenation, improvements in clinical end Table 1-4 estimates the cost of this therapy using doses from
points have been inconsistent. Pharmacologic differences in the published literature and a PICU patient of moderate size.
phospholipid and surfactant protein composition of the differ- Even in the absence of a positive effect on mortality, the
ent surfactant products make comparing the results between additional drug cost may be justifiable if surfactant therapy
studies difficult. The pulmonary-specific therapies group at minimizes mechanical ventilation or length of stay. However,
the PALICC reached strong agreement that routine use of sur- because data to support clinical benefit are currently lacking,
factant therapy cannot be recommended (Tamburro 2015). it is difficult to justify the increased cost of therapy.

Future Directions and Ongoing Research Corticosteroids


Much investigative effort has been placed into evaluating Inflammation, endothelial damage, and altered coagulation
exogenous surfactant in ARDS, with little identified benefit. caused by an uncontrolled innate defense response result in
Potential reasons for failure to produce positive outcomes the potentially reversible pathology of ARDS. However, in some
include surfactant product, nonstandardized doses, prepara- instances, this initial injury is followed by an irreversible fibro-
tion techniques, administration methods, and heterogeneity of proliferative phase. Interest in corticosteroids arises from the
the ARDS population. Despite this extensive effort, the pulmo- ability to protect patients from the uncontrolled inflammatory
nary-specific therapy subgroup of the PALICC recommended response leading to ARDS and potentially irreversible fibrotic
that future investigations focus on the populations most changes. In addition, relative adrenal insufficiency can occur
likely to benefit from surfactant therapy, although the specific in patients with ARDS (MacLaren 2002).
groups this might include were not described (Tamburro 2015).
Secondary to the lack of identified survival benefit, a Evidence for Use, Dosing Regimen, and Timing
pooled analysis of five multicenter investigations of recombi- for Initiation
nant surfactant protein C (rSP-C) in adults sought to identify Investigations of corticosteroid use in adults with ARDS
which patient populations with ARDS to focus on in future span almost 30 years, a broad range of dosing strategies,

PedSAP 2016 Book 2 Pediatric Critical Care 12 ARDS and Ventilator-Associated Events
Table 1-2. Pediatric Evidence for Surfactant Use in Acute Hypoxemic Respiratory Failure

Reference Study Details End Point/Intervention Results Comments

Willson 1996 Open-label, multicenter, 25% improvement in OI 25% improvement in OI Ventilator management algorithm used
observational Calfactant 80 mL/m2; First dose (n=29): 83% Improvement in oxygenation and ventilator
n=29 endotracheal instillation in Second dose (n=17): 59% parameters in the 24 hr after surfactant
Mean age 4.7 yr four equal aliquots Third dose (n=10): 60% administration
Acute hypoxemic Up to four doses, depending Fourth dose (n=4): 50% Development of air leaks should be
respiratory failure on initial improvement, 4 of 29 patients died monitored for after administration
OI 7 followed by deterioration Complications: Effect on outcome cannot be assessed
Bronchospasm (1) because of lack of a control group
Pneumothoraces (2)
Pneumopericardium (1)

PedSAP 2016 Book 2 Pediatric Critical Care


Unrelenting hypoxia (2)
Multiorgan failure (1)
Pneumopericardium (1)
Luchetti 1998 Randomized, single center Surfactant treatment effect No difference in baseline characteristics Viral etiology only known in four patients
n=10 on gas exchange, PIP, or MV parameters (all RSV)
PPV + surfactant duration of PPV and PICU Surfactant vs. control: Gas exchange, pulmonary mechanics, and
Mean age 10.4 mo stay PF ratio improved significantly at 1, 3, 12, clinical outcomes improved in surfactant

13
n=10 Poractant alfa 0.625 mL/kg and 24 hr after surfactant group
PPV only administered in two or three Paco2 improved significantly at 12 and Other medications used in both groups
Mean age 11.2 mo aliquots 24 hr after surfactant including bronchodilators, corticosteroids,
Severe bronchiolitis PIP improved significantly at 3, 12, and and aminophylline, none of which
requiring: 24 hr after surfactant is considered standard of care in
PPV Duration of PPV and ICU length of stay bronchiolitis
PF ratio < 158 mm Hg decreased significantly
No adverse effects noted
Willson 1999 Multicenter, prospective, Surfactant treatment effect No difference in baseline characteristics Ventilator management algorithm used
randomized, unblinded on OI and clinical outcomes Surfactant vs. control Criteria for retreatment changed during
n=21 surfactant group Calfactant 80 mL/m2; Acute sustained improvement of a 50% the study. Subsequent criterion was
n=21 control group endotracheal instillation in decrease in OI from baseline at 24 hr retreatment in all patients unless a
Acute hypoxemic four equal aliquots after surfactant significant adverse event during previous
respiratory failure Up to four doses Duration of PPV and ICU length of stay administration or OI improved to < 7
OI 7 retreatment criteria varied decreased significantly With the exception of bronchospasm,
No significant difference in mortality complications in the surfactant group
Complications with surfactant were noted in a similar number of children
administration: in the control group
Bronchospasm (1)
Air leaks (1)
Pulmonary interstitial emphysema (1)

ARDS and Ventilator-Associated Events


(continued)
Table 1-2. Pediatric Evidence for Surfactant Use in Acute Hypoxemic Respiratory Failure (continued)

Reference Study Details End Point/Intervention Results Comments

Lopez-Herce Open-label, single-center, > 20% improvement in No significant difference in PF ratio Uncontrolled study
1999 observational PF ratio within 4 hr of improvement based on dose Heterogeneous population
n=20 administration Pulmonary or systemic pathology (n=13) High overall mortality
1 mo 16 yr Poractant alfa 0.625 mL/kg > 20% improvement in PF ratio in 10
Acute pulmonary (n=14) and 2.5 mL/kg (n=6) patients
disease with severe given in two equal aliquots 4 of 13 patients died
hypoxemia Number of repeat doses CCHD pathology (n=7)
PF ratio < 100 mm Hg (05) varied > 20% improvement in PF ratio in two
patients
Six of seven patients died

PedSAP 2016 Book 2 Pediatric Critical Care


Herting 2002 Retrospective report Poractant alfa given in two Significant improvement in PF ratio Rescue therapy
n=8 equal aliquots. Dose not within 1 hr of first administration iNO or prostacyclin used in two patients
1 mo 13 yr standardized (median dose Repeat dose in seven of eight patients All patients had severe impairment of
PF ratio < 100 mm Hg 1.625 mL/kg) Significant, but less pronounced oxygenation
Pneumonia Repeat doses given, improvement in PF ratio with second High overall mortality
depending on favorable administration
response Four of eight patients died

14
Pneumothorax (2)
Luchetti 2002 Multicenter, randomized Surfactant treatment No difference in baseline characteristics Nasotracheal intubation used in all
controlled effect on gas exchange, or MV parameters patients associated with increased risk
n=20 respiratory mechanics, need Surfactant vs. control of VAP
CMV + surfactant for retreatment, duration PF ratio improved significantly at 1, 3, 6, Ventilator management algorithm used
Mean age 8.7 mo of CMV, PICU stay, and 12, 24, and 48 hr Prior treatment with 2-agonist,
n=20 mortality Paco2 improved significantly at 1, 3, 6, corticosteroids, and epinephrine in all
CMV only Poractant alfa 0.625 mL/kg 12, 24, and 48 hr patients
Mean age 7.4 mo administered in two aliquots PIP improved significantly at 1, 3, 6, 12, Unusually low mortality rate
RSV-induced severe 24, and 48 hr
respiratory failure: Duration of PPV and ICU length of stay
PF ratio < 150 mm Hg decreased significantly
No adverse effects noted
All patients survived

ARDS and Ventilator-Associated Events


Willson 2005 Multicenter, randomized, Surfactant treatment effect No difference in baseline characteristics Ventilator guidelines
blinded on clinical outcomes Surfactant vs. control Eight patients (four in each group) were
Infants, children, and Calfactant administered in OI significantly improved at 12 hr protocol violations six did not meet
adolescents four equal aliquots Significantly lower CMV failure the OI criteria, and two received a non-
n=77 < 10 kg: 3 mL/kg Significantly lower mortality protocol surfactant
Surfactant 10 kg: 80 mL/m2 No difference in ventilator-free days High overall mortality likely because
Mean age 7.2 yr Two doses at 12-hr intervals Complications with surfactant of the number of immunocompromised
n=75 administration: patients
Placebo Significantly greater hypotension More immunocompromised patients in the
Mean age 6.7 yr Significantly greater transient hypoxia placebo group
Acute respiratory failure No difference in the development of air Mortality with placebo did not reach
OI < 7 leaks statistical significance when controlling
Enrollment within 48 hr for immune status

PedSAP 2016 Book 2 Pediatric Critical Care


of MV

Thomas 2012 Multinational, phase II, Duration of MV Lower baseline PEEP and tidal volume in Synthetic surfactant
double-blind, placebo- Lucinactant 5.8 mL/kg in the placebo group No longer commercially available
controlled, randomized four equal aliquots No other differences in baseline Ventilator management protocol
Children 38 wk PMA to Repeat doses 1224 hr after characteristics Subset of patients with ALI shorter
2 yr initial allowed if patient still Surfactant vs. control duration of MV with lucinactant than with

15
n=84 met inclusion criteria No difference in duration of MV placebo
Surfactant No difference in improvement in PF ratio
Mean age 23.1 wk PNA Fewer repeat doses in the lucinactant
n=81 group
Placebo Complications with surfactant
Mean age 23.4 wk PNA administration:
Acute respiratory failure Transient bradycardia
Two consecutive PF Transient hypoxia
ratios 300 mm Hg (or
SF ratio 250)
MV 6 hr but 48 hr
on inclusion

(continued)

ARDS and Ventilator-Associated Events


Table 1-2. Pediatric Evidence for Surfactant Use in Acute Hypoxemic Respiratory Failure (continued)

Reference Study Details End Point/Intervention Results Comments

Willson 2013 Multinational, randomized, Surfactant treatment effect No difference in baseline characteristics Combined adult and pediatric study (table
placebo-controlled, on clinical outcomes Surfactant vs. control 3 in: Willson 2015)
masked Calfactant in two equal No difference in mortality, ventilator-free Study terminated at planned interim
Infants, children, and aliquots days, or PICU-free days analysis for futility
adolescents < 10 kg: 1.7 mL/kg No improvement in oxygenation More homogeneous population direct
n=56 10 kg: 0.5 mL/cm Hospital-free days significantly less lung injury only
Surfactant Up to two repeat doses with Complications with surfactant Primarily infectious lung injury
Mean age 6.3 yr a 25% improvement in PF administration: Independent randomization of those
n=53 ratio (or SF ratio) within 12 Transient hypoxia with a PF ratio < 100 mm Hg, OI > 30, or
Placebo hr of the previous dose Transient bradycardia immunocompromised status to ensure

PedSAP 2016 Book 2 Pediatric Critical Care


Mean age 6 yr Pneumothorax equal distribution
ALI/ARDS secondary to Pneumomediastinum Calfactant product containing 60 mg/mL
direct lung injury of phospholipid
PF ratio 300 mm Hg
(or SF ratio 250)
Enrollment within 48 hr
of MV

16
CMV = conventional mechanical ventilation; CCHD = complex congenital heart disease; iNO = inhaled nitric oxide; MV = mechanical ventilation; PIP = positive inspiratory pressure;
PMA = postmenstrual age; PNA = postnatal age; PPV = positive pressure ventilations; RSV = respiratory syncytial virus; VAP = ventilator-associated pneumonia.
Information from: Willson DF, Jiao JH, Bauman LA, et al. Calfs lung surfactant extract in acute hypoxemic respiratory failure in children. Crit Care Med 1996;24:1316-22; Luchetti
M, Casiraghi G, Calsecchi R, et al. Porcine-derived surfactant treatment of severe bronchiolitis. Acta Anaesthesiol Scan 1998;42:805-10; Willson DF, Zartsky A, Bauman LA, et al.
Instillation of calf lung surfactant (calfactant) is beneficial in pediatric acute hypoxemic respiratory failure. Crit Care Med 1999;27:188-95; Lopez-Herce J, de Lucas N, Carrillo A,
et al. Surfactant treatment for acute respiratory distress syndrome. Ach Dis Child 1999;80:248-52; Herting E, Moller O, Schiffmann JH, et al. Surfactant improves oxygenation in
infants and children with pneumonia and acute respiratory distress syndrome. Acta Paediatr 2002;91:1174-8; Luchetti M, Ferrero F, Gallini C, et al. Multicenter, randomized, con-
trolled study of porcine surfactant in severe respiratory syncytial virus-induced respiratory failure. Pediatr Crit Care Med 2002;3:261-8; Willson WF, Thomas NJ, Markovitz BP, et al.
Effect of exogenous surfactant (calfactant) in pediatric acute lung injury a randomized controlled trial. JAMA 2005;293:470-6; Thomas NJ, Guardia CG, Moya FR, et al. A pilot, ran-
domized, controlled trial of lucinactant, a peptide-containing synthetic surfactant, in infants with acute hypoxemic respiratory failure. Pediatr Crit Care Med 2012;13:646-53; Willson
WF, Thomas NJ, Tamburro R, et al. Pediatric calfactant in acute respiratory distress syndrome trial. Pediatr Crit Care Med 2013;14:657-65; and Willson DF, Truwit JD, Conaway MR,
et al. The adult calfactant in acute respiratory distress syndrome trial. Chest 2015;148:356-64.

ARDS and Ventilator-Associated Events


Table 1-3. Adult Evidence for Surfactant Use in ARDS

Reference Study Details End Point/Intervention Results Comments

Weg 1994 Multicenter, double-blind, Safety of nebulized surfactant Numerically more females in the placebo group Synthetic surfactant, contains
placebo-controlled, Surfactant treatment effect No other differences in baseline characteristics no surfactant proteins
randomized, parallel on oxygenation and clinical Surfactant vs. placebo Product not manufactured
n=17 outcomes No safety issues for nebulization, intended for
12-hr nebulized Exosurf nebulized No significant differences in oxygenation endotracheal administration
surfactant 12-hr group: ~1.6 mL/kg per day improvements No longer commercially
n=17 24-hr group: ~3.2 mL/kg per day No significant differences in mortality or available
24-hr nebulized Administered for up to 5 days hospital days
surfactant
n=17

PedSAP 2016 Book 2 Pediatric Critical Care


12- or 24-hr nebulized
saline
Sepsis-induced ARDS
Within in 18 hr of onset

Anzueto 1996 Multicenter, double-blind, Surfactant treatment effect No differences in baseline characteristics Synthetic surfactant, contains
placebo-controlled, on oxygenation and clinical Surfactant vs. placebo no surfactant proteins
randomized outcomes Similar number completed 5 days of treatment Product not manufactured

17
n=364 Exosurf ~8.3 mL/kg per day No significant differences in oxygenation for nebulization, intended for
Nebulized surfactant Administered for up to 5 days improvements endotracheal administration
n=361 No significant differences in mortality or No longer commercially
Nebulized saline hospital days available
Sepsis-induced ARDS Complications with surfactant administration: Mean PF ratio for both groups
ARDS developed in the Worsening hypoxia < 150 mm Hg
preceding 48 hr Respiratory arrest
Increased PIP
Increased secretions
Hypotension (both groups)
Barotrauma (both groups)

(continued)

ARDS and Ventilator-Associated Events


Table 1-3. Adult Evidence for Surfactant Use in ARDS (continued)

Reference Study Details End Point/Intervention Results Comments

Gregory 1997 Multicenter, open-label, Surfactant treatment effect Significant differences in baseline Included adolescent patients
randomized, controlled on oxygenation and clinical characteristics include: Large volumes of surfactant
n=43 outcomes Increased weight in the 4 mL/kg of LBW for up generally well tolerated
Surfactant Safety of endotracheally to four doses compared with control patients
n=16 administered surfactant More patients with risk factor for multiple

Control group Beractant in four equal aliquots transfusions in the 2 mL/kg of LBW for up
ARDS: 2 mL/kg of LBW for up to eight to eight doses and 4 mL/kg of LBW for up to
From trauma, doses (n=8) eight doses compared with control patients
multiple transfusions, 4 mL/kg of LBW for up to four Surfactant vs. control
aspiration of gastric doses (n=16) Significant reduction in Fio2 at 120 min in the 4

PedSAP 2016 Book 2 Pediatric Critical Care


contents, or sepsis 4 mL/kg of LBW for up to eight mL/kg of LBW for up to four doses group
Age 1570 yr doses (n=19) Significant improvement in Fio2 and PEEP at 7
AECC definition Assessment for retreatment days 4 mL/kg of LBW for up to four doses group
PEEP 5 cm H2O every 6 hr up to 96 hr Significantly increased length of stay in the 4
Retreatment criteria: mL/kg of LBW for up to eight doses group
PF ratio > 60 to < 200 mm Hg No difference in mortality
PEEP > 10 cm H O Complications with surfactant administration:
2
PF ratio 200 but 250 mm Oxygen desaturation

18
Hg AND PEEP 5 but 10 cm Air leak
H2 O Bronchospasm
Hypotension

Spragg 2004 Two clinical trials Surfactant treatment effect Percentage of patients with baseline APACHE Orphan drug status
Multicenter, randomized, on oxygenation and clinical II score > 23 was significantly higher in the Synthetic surfactant product
double-blind, controlled outcomes surfactant group Heterogeneous population of
n=224 rSP-C 1 mL/kg of LBW in All other baseline characteristics similar patients with ARDS
Surfactant + standard two aliquots intratracheal Surfactant vs. control Multivariate analysis showed:
therapy administration 91% received all four doses of surfactant Baseline PEEP and PIP

n=224 Up to three repeat doses Significant improvement in PF ratio at 424 hr associated with increased
Standard therapy only Assessment of retreatment from the first treatment relative risk of MV thus,
ARDS every 4 hr for 24 hr No difference in PF ratio at 48 hr from the first compromised lung function
AECC definition Retreatment criteria: treatment associated with fewer
PEEP 5 cm H2O PF ratio > 60 to < 200 mm Hg No difference in mortality or ventilator-free days ventilator-free days
Enrollment 48 hr AND Complications with surfactant administration: Enrollment in the European/

(North American sites) PEEP 5 cm H O


2
Significantly more adverse events in the South African arm, higher
to 72 hr (European/ surfactant group APACHE II, and older
South African sites) No significant difference in the number of patients had increased risk
from diagnosis serious adverse events of mortality
Most common events: hypoxemia, hypotension,
bradycardia

ARDS and Ventilator-Associated Events


Spragg 2011 Multicenter, randomized, Surfactant treatment effect No differences in baseline characteristics Orphan drug status
controlled, blinded on oxygenation and clinical Surfactant vs. control Synthetic surfactant product
n=419 outcomes No difference in mortality, gas exchange Fairly homogeneous patient
Surfactant + standard rSP-C 1 mL/kg of LBW in parameters, need for mechanical ventilation, or population
therapy two aliquots intratracheal the number of non-pulmonary organ failure Because of alteration in
n=424 administration free days the surfactant suspending
Standard therapy only Up to seven repeat doses at 6, Complications with surfactant administration: process and lack of expected
ARDS 12, 24, 36, 48, 72, and 96 hr after Significantly more serious adverse events in the outcome, surfactant activity
Severe impairment the first dose surfactant group was performed post hoc
of gas exchange Retreatment criteria: Adding shearing to the

from pneumonia or PF ratio > 60 to 170 mm Hg surfactant preparation


aspiration AND process may have
PF ratio 170 mm Hg PEEP 5 cm H O
2
impaired the function of

PedSAP 2016 Book 2 Pediatric Critical Care


Age 1285 yr the surfactant, which may
explain the deviation from
results of previous studies

Willson 2015 Multicenter, randomized, Surfactant treatment effect No differences in baseline characteristics Combined adult and pediatric
controlled, masked on oxygenation and clinical Surfactant vs. control: trial (table 3 in: Willson 2013)
n=151 outcomes No significant difference in mortality, ventilator- Study terminated at planned
Surfactant Calfactant 0.5 mL/cm in two free days, ICU-free days, or hospital-free days interim analysis for futility

19
n=157 aliquots No significant improvement in measures of More homogeneous
Placebo Up to two repeat doses oxygenation population direct lung injury
ARDS Retreatment criteria Complications with surfactant administration: only
AECC definition 25% improvement in PF ratio 116 adverse events possibly related to the Calfactant product containing
because of direct lung (or SF ratio) within 12 hr of the treatment intervention 60 mg/mL of phospholipid
injury previous dose Six events in the surfactant group were graded Ventilator algorithm used
Within 48 hr of as severe
initiating MV Most common adverse events were hypoxia and
Age 1885 hypotension

APACHE = Acute Physiology and Health Evaluation; LBW = lean body weight; rSP-C = recombinant surfactant protein C.
Information from: Weg JG, Balk, RA, Tharratt RS, et al. Safety and potential efficacy of an aerosolized surfactant in human sepsis-induced adult respiratory distress. JAMA
1994;272:1433-8; Anzueto A, Baughman RP, Guntupalli KK, et al. Aerosolized surfactant in adults with sepsis-induced adult respiratory distress. N Engl J Med 1996;334:1417-21;
Gregory TJ, Steinberg KP, Spragg R, et al. Bovine surfactant therapy for patients with acute respiratory distress syndrome. Am J Respir Crit Care Med 1997;155:1309-15.
Spragg RG, Lewis JF, Walmrath HD, et al. Effect of recombinant surfactant protein C-based surfactant on the acute respiratory distress syndrome. N Engl J Med 2004;351:884-92;
Spragg RG, Taut FJH, Lewis JF, et al. Recombinant surfactant protein C-based surfactant for patients with severe direct lung injury. Am J Respir Crit Care Med 2011;183:1055-61;
Willson WF, Thomas NJ, Tamburro R, et al. Pediatric calfactant in acute respiratory distress syndrome trial. Pediatr Crit Care Med 2013;14:657-65; and Willson DF, Truwit JD,
Conaway MR, et al. The adult calfactant in acute respiratory distress syndrome trial. Chest 2015;148:356-64.

ARDS and Ventilator-Associated Events


Table 1-4. Cost of Beractant, Calfactant, and Poractant Therapya

Beractant Calfactantb Poractant Alfa

WAC per vial 8-mL vial = $677.88 6-mL vial = $671.11 3-mL vial = $764.40

Cost of single dose 4 mL/kg 32 kg = 128 mL ~0.85 mL/cm 139 cm = 118 0.6252.5 mL/kg = 2080 mL
Rounded to whole vial = 16 mL Rounded to whole vial = 7 27
vials Rounded to whole vial = 20 vials 3 mL
Cost per dose = $10,846.08 vials 6 mL Cost per dose =
Cost per dose = $13,422.20 $5350.8$20,638.80

Cost of studied 128 mL 4 doses = 512 mL 118 mL 3 doses = 354 mL Cost not calculated secondary to
treatment course Rounded to whole vial = 64 Rounded to whole vial = 60 repeat doses varied greatly
vials 8 mL vials 6 mL
Cost per treatment course = Cost per treatment course =
$43,384.32 $40,266.60

a
Calculated for a 10-yr-old boy with height 55 inches, weight 32 kg.
b
Equivalent dose based on phospholipid component; calfactant product studied contains 60 mg/mL of phospholipid; Infasurf con-
tains 35 mg/mL of phospholipid.
WAC = wholesale acquisition cost.
Information from: Willson WF, Thomas NJ, Tamburro R, et al. Pediatric calfactant in acute respiratory distress syndrome trial. Pediatr
Crit Care Med 2013;14:657-65; and Willson DF, Truwit JD, Conaway MR, et al. The adult calfactant in acute respiratory distress syn-
drome trial. Chest 2015;148:356-64.

and heterogeneous patient populations. Initial corticoste- using pediatric data (Guglani 2006; Haselton 2000; Goh 1999;
roid studies focused on prevention and used short courses of Martinot 1997).
high-dose corticosteroids (30 mg/kg of methylprednisolone Because of an overwhelming lack of evidence in pediat-
every 6 hours). These data did not support corticosteroid use ric patients and inconsistent evidence in adult ARDS studies,
in the prevention of ARDS; rather, this treatment strategy may similar to surfactant therapy, the pulmonary-specific therapy
increase the risk of developing ARDS (Deal 2008). subgroup of the PALICC strongly agreed that corticosteroids
Subsequent investigations have used lower doses for more cannot be recommended as part of routine care in PARDS
prolonged courses, generally using methylprednisolone. (Tamburro 2015). According to adult data, if corticosteroids
These investigations have sought to delineate the role of cor- are used in PARDS, moderate- to low-dose prolonged therapy
ticosteroids in both the early and late phases of the disease; courses should be used. In addition, initiating this therapy is
however, responses in clinical outcomes have been inconsis- likely most useful in the late phase, but initiation should occur
tent (Meduri 2007, 1998; Steinberg 2006). Table 1-5 compares on or before day 13 of onset of the syndrome. Pending addi-
the treatment regimens, phase of ARDS, and outcomes. tional data, corticosteroids for ARDS should be avoided in
Meta-analyses and systematic reviews have also had incon- patients with influenza-related ARDS.
sistent or inconclusive results with respect to treatment effect
Consequences of Corticosteroid Use in the
on mortality or resource use (Ruan 2014; Tang 2009; Peter
Critically Ill Patient
2008). Of interest, when evaluating the effect of steroids on
mortality in ARDS given the underlying etiology, there is an Concerns with corticosteroid use in critically ill patients with
increased risk of mortality with steroid administration in those ARDS include risk of infection, hyperglycemia, and neuromus-
with influenza-related ARDS, but mortality is decreased in post- cular weakness. In randomized controlled trials, the risk of
operative patients with ARDS who receive steroids (Ruan 2014). infection has not been identified more commonly in the corti-
Data for pediatric patients are lacking. Published reports of costeroid treatment group than in the control groups (Meduri
corticosteroid use in pediatric ARDS are limited to three case 2007; Steinberg 2006; Meduri 1998). Both hyperglycemia and
reports and one case series of six patients. Similar to the data neuromyopathy have been identified as occurring more com-
available in adult patients, dosing regimens, time since onset monly in methylprednisolone-treated patients (Steinberg 2006).
of the syndrome to initiation of corticosteroids, and underly-
ing cause of ARDS vary greatly. The combination of the lack VENTILATOR-ASSOCIATED EVENTS
of data and variability prevents the ability to form conclusions Enhancing the quality of medical care through prevent-
on the use of corticosteroids in pediatric patients with ARDS ing health careassociated infections is imperative in

PedSAP 2016 Book 2 Pediatric Critical Care 20 ARDS and Ventilator-Associated Events
Table 1-5. Randomized Controlled Trials of Methylprednisolone in Adult ARDS

Meduri 1998 Steinberg 2006 Meduri 2007

Number of subjects 24 180 91


Methylprednisolone (n=16) Methylprednisolone (n=89) Methylprednisolone (n=63)

Phase of ARDS Late Late Early

Time since onset of 7 days 728 days Within 72 hr


ARDS

Dosing regimen Day 1: 2 mg/kg loading Day 1: 2 mg/kg loading Day 1: 1 mg/kg loading dose x 1
dose 1 dose 1 Days 114: 1 mg/kg
(mg/kg PER dose) Days 114: 0.5 mg/kg q6hr Days 114: 0.5 mg/kg q6hr Days 1521: 0.5 mg/kg
Days 1521: 0.25 mg/kg q6hr Days 1521: 0.5 mg/kg q12hr Days 2225: 0.25 mg/kg
Days 2228: 0.125 mg/kg q6hr Note: Those still intubated Days 2628: 0.125 mg/kg
Days 2930: 0.063 mg/kg q6hr at day 21 were tapered over Note: Patients extubated before day 14
Days 3132: 0.031 mg/kg q6hr 4 days. If extubated by day were advanced to day 15 and weaned
Note: Patients extubated 21, or developed infection Administered as continuous infusion,
before day 14 were advanced weaned over 2 days transitioned to daily oral dose, once
to day 15 and weaned possible

Oxygenation Significant improvement in PF Significant improvement in NR


ratio by day 5 PF ratio at days 3 and 14

LIS Significant improvement by NR Significantly more likely to have


day 5 improvement at day 7

Mortality Significantly higher in the No difference No difference


placebo group Higher risk of mortality in
methylprednisolone group
for those enrolled at > 14
days after ARDS onset

ICU length of stay NR Significantly shorter Significantly shorter

MV Significantly shorter duration Significantly shorter Significantly more likely to be extubated


duration by day 7

LIS = lung injury score; NR = not reported; q = every.


Information from: Meduri GU, Golden A, Freire AX, et al. Methylprednisolone infusion in early severe ARDS: results of a randomized
controlled trial. Chest 2007;131:954-63; Steinberg KP, Hudson LD, Goodman RB, et al. Efficacy and safety of corticosteroids for persis-
tent acute respiratory distress syndrome. N Engl J Med 2006;354:1671-84; and Meduri GU, Headley AS, Golden E, et al. Effect of
prolonged methylprednisolone therapy in unresolving acute respiratory distress syndrome: a randomized controlled trial. JAMA
1998;280:159-65.

optimizing care for patients and minimizing resource use. vary depending on the patients age. Figure 1-2 delineates
Ventilator-associated pneumonia is one such health care the current pediatric surveillance definition for the pneumo-
associated infection that must be considered for surveillance nia-reporting module (CDC 2016a). Applying this definition is
and prevention. At this time in PICUs, VAP is the only ventila- contingent on meeting the criteria for a health careassoci-
tor-associated event (VAE) that is considered for surveillance. ated infection, which states that the date of occurrence must
However, this current state of quality surveillance is being be on or after the third calendar day of an inpatient admis-
evaluated to ensure that an objective and outcomes-focused sion. In patients receiving mechanical ventilation on the date
definition is used. of the occurrence, the event must occur more than 2 calen-
dar days from the date of tracheal intubation to be considered
Defining VAP in Pediatric Patients ventilator associated. In addition to these criteria, further lab-
Ventilator-associated pneumonia is a device-related, report- oratory assessments primarily concerning lower respiratory
able, health careassociated infection. Definitions of VAP tract specimens can be used in determining VAP. Concerns

PedSAP 2016 Book 2 Pediatric Critical Care 21 ARDS and Ventilator-Associated Events
Patients 1 yr Patients > 1 yr to 12 yr

Chest imaging with one of the following: Chest imaging with one of the following:
1. New or progressive and persistent infiltrate 1. New or progressive and persistent infiltrate
2. Consolidation 2. Consolidation
3. Cavitation 3. Cavitation
4. Pneumatoceles

Clinical signs and symptoms including: Clinical signs and symptoms including at
Worsening gas exchange least three of the following:
PLUS three of the following: 1. Fever or hypothermia
1. Temperature instability 2. Leukopenia or leukocytosis
2. Leukocytosis, leukopenia, or left shift 3. Respiratory secretion findings such as:
3. Respiratory secretion findings: New-onset New-onset purulent sputum, change in
purulent sputum, change in sputum sputum character, increase in respiratory
character, increase in respiratory secretions, secretions, or increase in suctioning
or increase in suctioning requirements requirements
4. Apnea, tachypnea, nasal flaring with 4. New-onset worsening cough OR dyspnea,
retractions, or grunting apnea, or tachypnea
5. Wheezing, rales, or rhonchi 5. Rales or bronchial breath sounds
6. Cough 6. Worsening gas exchange
7. Bradycardia or tachycardia

Figure 1-2. Hospital-acquired/ventilator-associated pneumonia clinical criteria for infants and children.
Information from: CDC. CDC/NHSN Surveillance Device-Associated Module: Pneumonia (ventilator-associated and non-ventilator-
associate pneumonia) Event. 2016.

with the complexity and subjectivity of this definition are dis- at the time of data collection, identified a VAP prevalence in the
cussed later in this section. PICU of 7.1 cases per 1000 ventilator-days (Gupta 2015).
Health careacquired infections, including VAP, can be
Prevalence, Outcomes, and Associated Cost of associated with worse outcomes and increased health care
VAP in Pediatric Patients costs. Ventilator-associated pneumonia in pediatric patients
Reported rates of VAP vary depending on the reference, likely has been associated with a significant increase in PICU length
because of inter-reporter variability in data collection and of stay, duration of mechanical ventilation, and mortality
interpretation (Phongjitsiri 2015). Regardless, VAP is a com- (Gupta 2015; Bigham 2009). Even when controlling for sever-
mon cause of health careassociated infections in pediatric ity of illness, PICU patients who develop VAP are 3 times more
patients requiring mechanical ventilation. The most current likely to die than are those who did not develop the infection
report of the device-associated module, which includes VAP, (Gupta 2015). In addition to these worse outcomes, increased
from the National Healthcare Safety Network summarizes sur- health care use results in increased health care cost. The
veillance data from January to December 2013. This report VAP-attributable cost in pediatric patients has been reported
provides VAP data from 134 pediatric locations including 14 at about $50,000 per incident (Brilli 2008).
cardiothoracic critical care units (CCUs), 10 medical CCUs, and
110-combined medical-surgical CCUs. The overall ventilator
use ratio, defined as the total number of ventilator-days to total CURRENT QUALITY OF CARE
patient-days, was 0.37, with a VAP rate of 0.67 cases per 1000 SURVEILLANCE
ventilator-days (Dudeck 2015). This rate is less than the rates Because of concerns with the complexity and subjectivity of
reported in the literature. A recent multicenter, prospective the surveillance definitions for VAP, surveillance in adult insti-
evaluation, which used the CDC surveillance definition current tutions was redirected in 2013. The goal was to develop a more

PedSAP 2016 Book 2 Pediatric Critical Care 22 ARDS and Ventilator-Associated Events
objective and reliable approach, resulting in a refocus on VAEs least 2 calendar days of respiratory improvement or stability.
when the surveillance is prompted by worsening oxygenation Thus, the adult approach focuses on a change in respiratory
as opposed to chest radiograph changes (CDC VAE 2016). clinical status as opposed to less objective changes in chest
Currently, pediatric units continue to report device-associ- radiography. Streamlining and enhancing the objectivity of
ated events according to the VAP definition (see Figure 1-2), the surveillance definition in pediatric patients will hopefully
whereas adult units report VAEs. Pediatric reporting is only for allow for automation of surveillance in the future. In compari-
PICU areas. Neonatal ICUs can track VAP internally, but VAP is son, the pediatric VAP definition is far less objective. Table1-6
no longer part of the surveillance plan option to be analyzed compares the current pediatric and adult unit surveillance
by the National Healthcare Safety Network. The surveillance modules. For completeness, the table includes criteria that
definition used for reporting is determined by the location, were used to assess ventilator-associated tracheobronchitis
not the age, of the patient undergoing surveillance. Thus, a (VAT) in studies discussed later in the chapter.
patient 18 years or younger in an adult unit would report by the
Classification of VAEs
VAE module, and a patient 18 years or older in a pediatric unit
would report by the pneumonia module (CDC 2016a, 2016b). Ventilator-associated events are further delineated into
ventilator-associated conditions (VACs), infection-related
Differences in Adult vs. Pediatric Surveillance ventilator-associated conditions (IVACs), and possible ventila-
The pediatric pneumonia surveillance definition is a single-tier tor-associated pneumonia (PVAP). After initiating mechanical
approach, initiated by changes in chest radiography. The adult ventilation, calendar day 3 is the earliest an event may occur
VAE surveillance definition is a three-tier approach that is initi- that can meet the criteria of a VAE on day 4 of mechanical
ated secondary to indicators of worsening oxygenation after at ventilation.

Table 1-6. Comparison of Ventilator-Associated Event Criteria

PedVAP VAT VAC IVACa PVAP

Worsening after Not considered Not considered Required Required Required


stability or
improvement

Chest imaging Change in chest No change in chest Not Not considered Not considered
imaging required imaging considered

Change in Fio2 No specific criteria Not considered Specific for Specific for Specific for worsening
but worsening gas worsening worsening oxygenation
exchange oxygenation oxygenation
1 yr: required
112 yr: optional

Change in PEEP No specific criteria Not considered Specific for Specific for Specific for worsening
but worsening gas worsening worsening oxygenation
exchange oxygenation oxygenation
1 yr: required
112 yr: optional

WBC Leukocytosis, 12 yr: 12 103 cells/ 12 103 cells/mm3 OR


leukopenia, or left- 15 103 cells/ mm3 OR < 4 103 cells/mm3
shift ( 1 yr) mm3 OR < 4 103 cells/
< 4 103 cells/mm3 mm3
> 12 yr:
12 103 cells/
mm3 OR
< 4 103 cells/mm3

Temperature 1 yr: instability > 38C OR > 38C OR > 38C OR


112 yr: fever or < 36C < 36C < 36C
hypothermia

(continued)

PedSAP 2016 Book 2 Pediatric Critical Care 23 ARDS and Ventilator-Associated Events
Table 1-6. Comparison of Ventilator-Associated Event Criteria (continued)

PedVAP VAT VAC IVACa PVAP

New antimicrobial Not considered Not considered Started and Started and continued
continued for for 4 days
4 days

Sputum change New, changed, or Not considered Not considered


increased

Respiratory BAL: 100,000 CFU 10,000 CFU EA: 1,000,000 CFU


culture Lung tissue: BAL: 100,000 CFU
quantitationb 100,000 CFU Lung tissue: 100,000
PBS: 10,000 CFU CFU
PBS: 10,000 CFU

Sputum WBCc Not considered At least moderate 25 neutrophils AND


10 epithelial cells per
low-power field

Other positive Organism from N/A Organism from pleural


infectious pleural fluid fluid
testingd Lung histopathology Lung histopathology
showing: abscess showing: abscess
formation OR fungal formation OR fungal
infiltration infiltration
Positive Legionella Positive Legionella
Positive respiratory Positive respiratory
virus virus

Clinical symptoms Required N/A Not considered


of respiratory
distress

a
Require only that either WBC or temperature criteria be met.
b
Considered in criteria 1 and 2 for PVAP.
c
Considered in criterion 2 for PVAP.
d
Considered in criterion 3 for PVAP.
BAL = bronchoalveolar lavage; EA = endotracheal aspirate; IVAC = infection-related ventilator-associated condition; N/A = not appli-
cable; PBS = protected brush specimen; PedVAP = pneumonia criteria for pediatrics; PVAP = possible VAP; VAC = ventilator-associated
condition; VAT = ventilator-associated tracheobronchitis.
Information from: Beardsley AL, Nitu ME, Cox EG, et al. An evaluation of various ventilator-associated infection criteria in a PICU.
Pediatr Crit Care Med 2016;17:73-80; CDC. CDC/NHSN Surveillance Device-Associated Module: Pneumonia (ventilator-associated and
non-ventilator-associate pneumonia) Event; and CDC. CDC/NHSN Surveillance Device-Associated Module: Ventilator-Associated
Events. 2016.

Ventilator-Associated Conditions temperature or WBC count abnormalities plus initiation of a


After a period of stability or improvement on the ventilator, new antimicrobial agent that is continued for at least 4 days.
an increase in the daily minimum Fio2 of at least 0.2 or an
increase in the daily minimum PEEP of at least 3 mm Hg is Possible Ventilator-Associated Pneumonia
defined as a VAC. These changes must be sustained for 2 or Categorization as PVAP occurs if one of three criteria is met
more calendar days. within 2 days before or after the onset of the VAC. The criteria
for PVAP incorporate information that may suggest an infec-
IVAC Complications tious pathology. Concerning microbiologic data, one of three
Patients presenting within 2 days before or after the onset criteria may be used to meet the definition of PVAP. Criterion
of the VAC are considered to have IVACs if they have either 1 is a report of quantitative or semiquantitative growth from

PedSAP 2016 Book 2 Pediatric Critical Care 24 ARDS and Ventilator-Associated Events
endotracheal aspirates, bronchoalveolar lavage (BAL), lung A multicenter, retrospective, matched cohort study of
tissue, or protected brush specimen. Criterion 2 is purulent PICU, cardiac ICU, and neonatal ICU was conducted to find
respiratory secretions evaluated in combination with quanti- a pediatric VAC definition that would identify patients at risk
tative or semiquantitative respiratory culture results that do of worse outcomes. Worsening of oxygenation for at least 2
not meet specified quantitation thresholds. Criterion 3 are days after 2 days of stability or improvement was defined by
identification of pathologic organisms from positive pleural Fio2 daily increases of 0.2, 0.25, and 0.3 and mean airway pres-
fluid culture, lung histology consistent with abscess or paren- sure (MAP) daily increases of 4, 5, 6, and 7 cm H2O. Patients
chymal invasion by fungi, Legionella, or viral testing positive with a VAC were matched with patients without a VAC. Mean
for respiratory viruses. airway pressure was used for this investigation because it
better reflects changes in lung compliance and allows for an
Utility of Surveillance Definitions assessment of patients receiving high-frequency oscillatory
The paradigm shift in the need for a new surveillance method ventilation. The rate of VACs, according to these definitions,
in adult patients arose secondary to the fact that the cur- including MAP in a multicenter approach, resulted in a sig-
rent definition used for surveillance of pneumonia is neither nificantly lower rate of VACs of 2.93.2 cases per 1000
sensitive nor specific. In a recent retrospective analysis, the ventilator-days than in the 2015 single-center cohort study.
current pediatric VAP, IVAC, VAT, and lower respiratory tract Each assessed increase in Fio2 and MAP was associated
definitions were compared in 300 episodes of mechani- with a greater risk of death and, among survivors, prolonged
cal ventilation in which there were 30 ventilator-associated hospitalization, time in the ICU, and duration of mechani-
infections. Of the 30 infections, 9 met more than one of the cal ventilation. Outcomes assessment was consistent with
ventilator-associated infection criteria. Despite only moder- the earlier single-center cohort study (Cocoros 2016). Given
ate overlap in the definitions with respect to diagnosis, the these results, the advisory committee recommended the use
risk factors for developing a ventilator-associated infection of a daily minimum increase in Fio2 of 0.25 or a daily minimum
and outcomes were similar, suggesting that each definition increase in MAP of 4 cm H2O for a pediatric VAC. This defini-
has some validity (Beardsley 2016). tion should be used in further studies to assess risk factors
In adults, significant concern surrounded the use of a def- and preventive measures.
inition in which surveillance was prompted by radiographic Given these data and the overwhelming concern with the
findings, which may not accurately identify VAP. In addition, current surveillance definition, it seems probable that there
the clinical signs and symptoms, although potentially useful will soon be a shift in the surveillance of pediatric VAP to
in diagnosis, may not consistently be documented, making something consistent with the VAE surveillance now used
them poor surveillance features for quality improvement by adult institutions. The utility of a more objective surveil-
endeavors. Optimizing surveillance definitions is important lance definition that identifies more than just VAP may help
in facilitating the development of optimal prevention strate- enhance prevention strategies and better anticipate out-
gies (CDC 2016b). comes in these patients.

Potential Transition of Pediatric Surveillance


Understanding that a new approach to surveillance would be ANTIBIOTIC STEWARDSHIP FOR IVAC
required for pediatric patients, a workgroup was convened to COMPLICATIONS AND VAP
explore the possibility of a new surveillance definition. This While optimizing the surveillance and diagnostic criteria for
workgroup agreed that the current surveillance definition had IVAC and VAP, pharmacists can focus on antimicrobial stew-
room for optimization, but it was cautious about adopting the ardship for these health careassociated infections. The
adult definitions for VAE without further data. primary goal is to enhance clinical outcomes while minimizing
A single-center, retrospective study applied the current any undue consequences (Dellit 2007). Antimicrobial stew-
adult VAE definitions to a pediatric cohort. Rates of VACs ardship in IVAC and VAP can be achieved through several of
and IVACs per 1000 ventilator-days were 20.9 and 12.9 the standard strategies, including implementing institutional
cases, respectively. Most of the patients classified as hav- guidelines or clinical pathways, minimizing combination ther-
ing an IVAC were then further classified as having either apy when appropriate, de-escalating therapy, and optimizing
possible or probable pneumonia. Compared with those therapy duration, depending on the diagnosis and culture data.
without a VAC, patients with a VAC had a significantly lon- Institutional guidelines and clinical pathways considering the
ger duration of mechanical ventilation and duration of both diagnostic criteria for IVAC and VAP, and an understanding of
hospitalization and ICU length of stay, as well as a higher institution and local bacterial resistance patterns, can help
risk of mortality. Immunocompromised status, tracheos- facilitate the use of appropriate but not overly broad empiric
tomy dependence, and chronic respiratory failure were antimicrobial therapy, pending culture results.
identified as independent risk factors for developing a VAC Use of combination therapy for either empiric or targeted
(Phongjitsiri 2015). therapy is a management strategy with more questions

PedSAP 2016 Book 2 Pediatric Critical Care 25 ARDS and Ventilator-Associated Events
than answers. Use of combination therapy has long been data support that courses of therapy less than 7 days may
recommended as a method to prevent the emergence of be appropriate in patients with signs and symptoms consis-
resistance, but evidence to support the routine use of this tent with VAT.
strategy is lacking (Dellit 2007). However, use of combina- Pharmacists can intervene to optimize duration of ther-
tion therapy for empiric coverage may be appropriate in apy in patients with VAP. No pediatric data comparing
certain instances. durations of antibiotics for VAP are available; however, one
At the time of publication, current guidelines for adults with randomized, prospective, double-blind study compared
VAP recommend empiric combination therapy for patients at treatment durations for VAP in adult patients. Patients were
risk of multidrug-resistant bacterial pathogens; however, this randomized to receive 8 or 15 days of an antibiotic regi-
set of guidelines is now more than 10 years old (ATS 2005). men chosen by the treating physician. No difference was
Recently, two studies evaluated the outcomes for pediatric found in mortality, recurrent infections, ventilator-free days,
patients with gram-negative bacteremia treated with combi- organ failurefree days, or length of ICU stay between the
nation antibiotics versus monotherapy. Neither study found two treatment groups. However, the 8-day group had signif-
improved time to bacteremia resolution or mortality with icantly more antibiotic-free days, as would be expected. In
empiric combination therapy compared with monotherapy patients with nonfermenting gram-negative organisms (e.g.,
(Berkowitz 2015; Sick 2014). Pseudomonas, Acinetobacter), the risk of pulmonary infec-
Survival benefit was identified in one study of patients tion recurrence was higher in the 8-day group; thus, for this
with multidrug-resistant gram-negative organisms (MDRGN), group, the noninferiority of 8 days could not be confirmed
leading the authors to conclude that in patients with risk (Chastre 2003).
factors for MDRGN, empiric combination therapy may be Despite these data, it is reasonable to reassess patients
beneficial. Risk factors for MDRGN were a history of coloni- with nonfermenting gram-negative organisms at 8 days
zation or infection with MDRGN, exposure to broad-spectrum of therapy to determine whether, given the patients clin-
antibiotic therapy in the past 30 days, prolonged current hos- ical improvement and status, the treatment duration is
pitalization, or a high prevalence of MDRGN in the community appropriate. For all other patients with VAP having clinical
(Sick 2014). This list is much less extensive than that pro- improvement, 8 days is a reasonable therapy duration.
vided in the adult VAP guidelines.
An assessment of antibiotic duration in VAT in pediatric
patients identified an increased risk of subsequent develop- STRATEGIES FOR PREVENTION
ment of colonization or infection with a multidrug-resistant OF VAES
organism in patients receiving combination therapy (Tamma As with all health careassociated conditions, prevention is
2011). Given these data, as well as the lack of pediatric VAP key. To this point, prevention strategies for VAE are focused on
guidelines, the outdated nature of the adult VAP guidelines, preventing infectious-related complications. As surveillance
and the risk of common combination agents (particularly ami- shifts to include noninfectious VAE, expanded surveillance
noglycosides), it seems prudent for pharmacists and other will allow for the implementation of prevention strategies
health care providers to judiciously consider which patients for VAC. Improved surveillance may also help with determin-
should receive empiric combination therapy. Furthermore, ing methods to prevent progression to IVAC and VAP/PVAP.
therapy de-escalation to the narrowest-spectrum agent(s) Prevention includes a combination of reducing modifiable
should occur once culture and sensitivity data are available. risk factors (Box 1-2), when possible, and implementing pre-
Therapy de-escalation is often hindered by the rate-limiting vention bundles (Liu 2013; Foglia 2007).
step of culture speciation and susceptibility data. Progress Of the potentially modifiable risk factors that have been
in developing rapid diagnostic testing that will provide both identified, only the need for reintubation or self-extubation,
organism and susceptibility for respiratory specimens will steroid administration, bloodstream infection, prior antibiotic
facilitate earlier optimization of therapy. therapy, bronchoscopy, and presence of a genetic syndrome
Clarifying the IVAC being treated, especially when consid- were risk factors in a meta-analysis of VAP in the PICU (Lui
ering therapy for VAT versus VAP, can optimize the therapy 2013). Increased risk with the administration of acid-sup-
duration provided. A retrospective, cohort study of pedi- pressive therapy has been somewhat controversial. Although
atric patients with clinically diagnosed VAT compared the there is a known increased risk of community-acquired
outcome of progression with hospital-acquired pneumonia pneumonia with proton pump inhibitor therapy, data for
(HAP) or VAP for prolonged ( 7 days) versus short (< 7 days) increased risk of VAP in adult patients have been inconsis-
courses of antibiotic therapy. Prolonged courses of antibiot- tent (Plummer 2014). It is theorized that when the potentially
ics did not provide a protective effect for progression to HAP immune protective effects of gastric acid are neutralized
or VAP. Patients who received prolonged courses of antibiot- with acid-suppressive therapy, patients are at an increased
ics were more likely to develop an infection or colonization risk of bacterial growth in their gastric contents, which may
with a multidrug-resistant organism (Tamma 2011). These heighten the risk of pneumonia during periods of aspiration.

PedSAP 2016 Book 2 Pediatric Critical Care 26 ARDS and Ventilator-Associated Events
Patient Care Scenario Box 1-2. Potentially Modifiable Factors
A 13-month-old boy born at 28 weeks gestation has That Increase VAP Riska
a medical history that includes chronic lung disease of Acid-suppressive therapy
prematurity and pulmonary hypertension. He presents Bloodstream infectionb
to the PICU with respiratory failure secondary to respi- Bronchoscopyb
ratory syncytial virus requiring mechanical ventilation. Continuous enteral nutrition
He was discharged from the neonatal ICU at 3 months of Mechanical ventilation and duration > 3 days
age and has not been readmitted; he has no recent anti- Need for reintubation or self-extubationb
biotic exposure. During the first 48 hours of admission, Neuromuscular blockade
he develops worsening respiratory status, and he is Prior antibiotic therapyb
given a diagnosis of severe ARDS. On day 6 of mechani- Steroid administrationb
cal ventilation, his temperature is 101.7F (38.7C), and Supine positioning
chest radiography reveals a new infiltrate. Because of Transfusion
concern for developing VAP, a mini-BAL is obtained. Transport out of PICU
Considering the risk of potential drug-resistant organ- Trauma
isms and the tenets of antimicrobial stewardship, what Uncuffed endotracheal tube
is the best empiric antibiotic coverage to initiate?
a
As identified in both adult and pediatric studies.
b
Identified as a risk factor in a meta-analysis of pediatric VAP
ANSWER studies.
The best empiric antibiotic coverage in this patient
would be an antipseudomonal -lactam such as
cefepime or piperacillin/tazobactam plus vancomycin.
Given the duration of hospitalization, the patient would
potentially be at risk of resistant organisms. Although
duration of hospitalization may place the patient at
risk of resistant organisms, data do not support the
routine use of combination empiric therapy for gram- Box 1-3. Example of a Pediatric VAP
negative organisms, most commonly the addition of Prevention Bundle
an aminoglycoside. Outcomes data in pediatric bacte- Prevent colonization of the oropharynx, stomach, and
remia support consideration of combination therapy in sinuses
patients with a history of colonization or infection with
MDRGN, exposure to broad-spectrum antibiotic therapy
Change ventilator circuit and in-line suction only when
visibly soiled
in the past 30 days, prolonged current hospitalization,
or a high prevalence of MDRGN in the community. This
Drain condensation from the ventilator circuit every
24 hr
patient does not have these risk factors. As well, data do
not support that combination therapy is a strategy that
Rinse oral suction device after use; then store in a
nonsealed plastic bag
prevents the development of resistance.
Use hand hygiene before and after ventilator circuit
1. Berkowitz NM, Spaeder MC, DeBiasi RL, et al. contact
Empiric monotherapy versus combination ther- Wear gown when caring for patient if soiling from respi-
apy for Enterobacteriaceae bacteremia in children. ratory secretions is expected
Pediatr Infect Dis J 2015;34:1203-6. Provide mouth care every 24 hr
Prevent aspiration of contaminated secretions
2. Sick AC, Tschudin-Sutter S, Turnbull AE, et al.
Empiric combination therapy for gram-negative
Elevate head of bed 3045 degrees
bacteremia. Pediatrics 2014;133:1-8.
Drain ventilator circuit before repositioning
In patients > 12 yr, use endotracheal tube with dorsal
3. Dellit TH, Owens RC, McGowan JE, et al. Infectious lumen, when possible
Diseases Society of America and the Society of
Healthcare Epidemiology of America guidelines Adapted from: Bigham MT, Amato R, Bondurrant P, et al.
for developing an institutional program to enhance Ventilator-associated pneumonia in the pediatric ICU: charac-
terizing the problem and implementing a sustainable solution.
antimicrobial stewardship. CID 2007;44:159-77.
J Pediatr 2009;154:582-7.

Administration of histamine-2 receptor antagonists or proton prophylaxis against SRMD, VTE prophylaxis, and oral hygiene
pump inhibitors was not associated with an increased risk of with chlorhexidine. Despite the potential increased risk of
VAP in the aforementioned meta-analysis (Lui 2013). VAP with acid-suppressive therapy, the IHI ventilator bun-
The Institute for Healthcare Improvement (IHI) offers ven- dle recommends that prophylaxis against SRMD be used in
tilator bundle recommendations, including semirecumbent mechanically ventilated patients when the benefits outweigh
positioning (elevating head of bed 3045 degrees), daily the risks (IHI 2012). Ventilator-associated pneumonia bundles
sedation holidays and assessment of extubation readiness, decrease the risk of VAP in pediatric patients, and one such

PedSAP 2016 Book 2 Pediatric Critical Care 27 ARDS and Ventilator-Associated Events
bundle reduced hospital cost by about $2.5 million over 2 fis- REFERENCES
cal years (Brilli 2008). One evaluated bundle (Box 1-3) reduced ASHP Commission on Therapeutics. ASHP therapeutic
the rate of VAP from 5.6 cases to 0.3 cases per 1000 ventila- guidelines on stress ulcer prophylaxis. ASHP Commission
tor-days (Bigham 2009). on Therapeutics and approved by the ASHP Board of
Directors on November 14, 1998. Am J Health Syst Pharm
As with all prevention strategies, sustaining the use of the
1999;56:347-79.
prevention bundle is of utmost importance. Compliance with
the various components of the bundle described in the text that American Thoracic Society (ATS), Infectious Diseases
follows waned to 78%100% 2 years after implementation, thus Society of America. Guidelines for the management of
adults with hospital-acquired, ventilator-associated, and
delineating the need for continued follow-up and assessment of
healthcare-associated pneumonia. Am J Respir Crit Care
compliance to ensure persistence with prevention strategies.
Med 2005;171:388-416.

CONCLUSION Barr J, Fraser GL, Puntillo K, et al. Clinical practice guide-


lines for the management of pain, agitation, and delirium
Pulmonary morbidities in pediatric patients are significant in adult patients in the intensive care unit. Crit Care Med
and can lead to poor outcomes and death. Unfortunately, in 2013;41:236-306.
ARDS, pulmonary-specific pharmacotherapies have proven
Beardsley AL, Nitu ME, Cox EG, et al. An evaluation of various
less than successful at altering clinical outcomes. For cor-
ventilator-associated infection criteria in a PICU. Pediatr
ticosteroids, pediatric data are lacking, and the pediatric Crit Care Med 2016;17:73-80.
pharmacist must have a working knowledge of adult data
to help discern corticosteroid use. Similarly, with VAE and Bembea MM, Jouvet P, Willson D, et al. Methodology of
the Pediatric Acute Lung Injury Consensus Conference.
the shift in the surveillance process, which is likely to affect
Pediatr Crit Care Med 2015;16:S1-S5.
pediatric patients in the near future, pharmacists must under-
stand the reasons for this shift as well as the new conditions Berkowitz NM, Spaeder MC, DeBiasi RL, et al. Empiric
to be considered. Pharmacists should be actively involved monotherapy versus combination therapy for
Enterobacteriaceae bacteremia in children. Pediatr Infect
in VAP prevention strategies and in optimizing treatment in
Dis J 2015;34:1203-6.
patients who develop IVAC or VAP.
Bigham MT, Amato R, Bondurrant P, et al. Ventilator-
associated pneumonia in the pediatric intensive care unit:
Practice Points characterizing the problem and implementing a sustain-
able solution. J Pediatr 2009;154:582-7.
When designing evidence-based pharmacotherapy
regimens for ARDS, pharmacy practitioners should con- Brilli RJ, Sparkling KW, Lake MR, et al. The business case for
sider the following: preventing ventilator-associated pneumonia in pediatric
Intensive assessment of exogenous surfactant products intensive care unit patients. Jt Comm J Qual Patient Saf
has not resulted in significant improvements in patient 2008;34:629-38.
outcomes.
An ongoing study is assessing the effect of exogenous CDC. CDC/NHSN Surveillance Device-Associated Module:
surfactant on outcomes in the leukemia and stem cell Pneumonia (ventilator-associated and non-ventilator-
transplant populations. Previous studies have alluded to associate pneumonia) Event. 2016a.
the potential benefit in this population; thus, cautious con-
CDC. CDC/NHSN Surveillance Device-Associated Module:
sideration and risk-benefit analysis may be considered.
Ventilator-Associated Events. 2016b.
In the absence of pediatric data, cautious consideration of
glucocorticoid use in both early and late phases of ARDS Chastre J, Wolff M, Fagon JY, et al. Comparison of 8 vs. 15
may be evaluated on a patient-by-patient basis. days of antibiotic therapy for ventilator-associated pneu-
Pending further data, glucocorticoids should be avoided for monia in adults. JAMA 2003;290:2588-98.
ARDS secondary to influenza or in those whose onset of
disease was more than 13 days before initiation of therapy. Cheifetz IM. Pediatric acute respiratory distress syndrome.
The pharmacists role in the care of PICU patients Respir Care 2011;56:1589-99.
results in the need to:
Cocoros NM, Kleinman K, Priebe GP, et al. Ventilator-
Have an understanding of health careassociated infection
associated events in neonates and children a new
surveillance, including the concerns surrounding the cur-
paradigm. Crit Care Med 2016;44:14-22.
rent VAP surveillance module
Use understanding of antibiotic pharmacotherapy, geo- Curley MA, Wypij D, Watson RS, et al. Protocolized sedation
graphic resistance patterns, and tenets of antimicrobial vs usual care in pediatric patients mechanically ventilated
stewardship to apply the principles of judicious empiric and for acute respiratory failure: a randomized clinical trial.
targeted antibiotic use for VAP and VAT in this population JAMA 2015;313:379-89.
Participate in developing prevention strategies and pro-
grams for health careassociated conditions, including Deal EN, Hollands J, Schramm GE, et al. Role of corticoste-
VAP roids in the management of acute respiratory distress
syndrome. Clin Ther 2008;30:787-99.

PedSAP 2016 Book 2 Pediatric Critical Care 28 ARDS and Ventilator-Associated Events
Dellit TH, Owens RC, McGowan JE, et al. Infectious Diseases Meduri GU, Golden A, Freire AX, et al. Methylprednisolone
Society of America and the Society of Healthcare infusion in early severe ARDS: results of a randomized
Epidemiology of America guidelines for developing an controlled trial. Chest 2007;131:954-63.
institutional program to enhance antimicrobial steward-
ship. CID 2007;44:159-77. Meduri GU, Headley AS, Golden E, et al. Effect of prolonged
methylprednisolone therapy in unresolving acute respi-
Dudeck MA, Edwards JR, Allen-Bridson K, et al. National ratory distress syndrome: a randomized controlled trial.
Healthcare Safety Network report, data summary for JAMA 1998;280:159-65.
2013, device-associated module. Am J Infect Control
2015;43:206-21. Pediatric Acute Lung Injury Consensus Conference (PALICC),
Khemani RG, Smith LS, et al. Pediatric acute respiratory
ESICM. ARDS Definition Task Force, Ranieri VM, Rubenfeld distress syndrome: definition, incidence, and epide-
GD, et al. Acute respiratory distress syndrome: the Berlin miology: proceedings from the Pediatric Acute Lung
definition. JAMA 2012;307:2526-33. Injury Consensus Conference. Pediatr Crit Care Med
2015;16:S1-S131.
Ferguson ND, Fan E, Camporota L, et al. The Berlin definition
of ARDS: an expanded rationale, justification, and supple- Penn State University (PSU). CALIPSO: calfactant for pediat-
mentary material. Intensive Care Med 2012;38:1573-82. ric acute lung injury in pediatric stem cell transplantation
and oncology patients. In: ClinicalTrials.gov [Internet].
Foglia E, Meier MD, Elward A. Ventilator-associated pneumo- Bethesda, MD: U.S. National Library of Medicine, 2015.
nia in neonatal and pediatric intensive care unit patients.
Clin Microbiol Rev 2007;20:409-25. Peter JV, John P, Graham PL, et al. Corticosteroids in the
prevention and treatment of acute respiratory distress syn-
Goh AYT, Sekaran D, Roziah M. Corticosteroid rescue in drome (ARDS) in adults: meta-analysis. BMJ 2008;336:1006.
late paediatric acute respiratory distress syndrome.
Respirology 1999;4:295-7. Phongjitsiri S, Coss-Bu J, Kennedy C, et al. The Centers
for Disease Control and Preventions new definitions for
Guglani L, Jain S, Lodha R. Methylprednisolone ther- complications of mechanical ventilation shift the focus
apy in a child with unresolving ARDS. Indian Pediatr of quality surveillance and predict clinical outcome in a
2006;43:639-42. PICU. Crit Care Med 2015;43:2446-51.
Gupta S, Boville BM, Blanton R, et al. A multicentered pro- Plummer MP, Blaser AR, Deane AM. Stress ulceration: preva-
spective analysis of diagnosis, risk factors, and outcomes lence, pathology, and association with adverse outcomes.
associated with pediatric ventilator-associated pneumo- Crit Care 2014;18:213-9.
nia. Pediatr Crit Care Med 2015;16:e65-73.
Randolph AG. Management of acute lung injury and acute
Haselton DJ, Kiekamp JG, Christman BW, et al. Use of respiratory distress syndrome in children. Crit Care Med
high-dose corticosteroids and high-frequency oscil- 2009;37:2448-54.
latory ventilation for treatment of a child with diffuse
alveolar hemorrhage after bone marrow transplantation: Reiter PD, Wathen B, Valuck RJ, et al. Thrombosis risk factor
case report and review of the literature. Crit Care Med assessment and implications for prevention in critically ill
2000;28:245-8. children. Pediatr Crit Care Med 2012;13:381-6.

Institute for Healthcare Improvement (IHI). How-to Guide: Reveiz L, Guerrero-Lozano R, Camacho A, et al. Stress-
Prevent Ventilator-Associated Pneumonia. Cambridge, ulcers, gastritis, and gastrointestinal bleeding prophylaxis
MA: IHI, 2012. in critically ill pediatric patients: a systematic review.
Pediatr Crit Care Med 2010;11:124-32.
Khemani RG, Smith LS, Zimmerman JJ, et al. Pediatric acute
respiratory distress syndrome: definition, incidence, and Ruan AY, Lin HH, Huang CT, et al. Exploring the heterogeneity
epidemiology: proceedings from the Pediatric Acute of the effects of corticosteroids on the acute respiratory
Lung Injury Consensus Conference. Pediatr Crit Care Med distress syndrome: a systematic review and meta-analy-
2015;16:S23-S40. sis. Crit Care 2014;18:R63.

Liu B, Li S, Zhang S, et al. Risk factors of ventilator-asso- Sapru A, Flori H, Quasney MW, et al. Pathobiology of acute
ciated pneumonia in pediatric intensive care unit: a respiratory distress syndrome. Pediatr Crit Care Med
systematic review and meta-analysis. J Thorac Dis 2015;16:S6-S22.
2013;5:525-31.
Sharathkumar AA, Mahajerin A, Heidt L, et al. Risk-prediction
MacLaren R, Jung R. Stress-dose corticosteroid therapy tool for identifying hospitalized children with a predis-
for sepsis and acute lung injury or acute respiratory dis- position for development of venous thromboembolism:
tress syndrome in critically ill adults. Pharmacotherapy Peds-Clot clinical decision rule. J Thromb Haemost
2002;22:1140-56. 2012;10:1326-34.

Martinot A, Fourier C, Cremer R, et al. Short-course high- Sick AC, Tschudin-Sutter S, Turnbull AE, et al. Empiric com-
dose corticosteroid treatment in six children with ARDS. bination therapy for gram-negative bacteremia. Pediatrics
Pediatr Pulmonol 1997;23:314-6. 2014;133:1-8.

PedSAP 2016 Book 2 Pediatric Critical Care 29 ARDS and Ventilator-Associated Events
Spragg RG, Taut FJH, Lewis JF, et al. Recombinant sur- Taut FJH, Rippin G, Schenk P, et al. A search for subgroups
factant protein C-based surfactant for patients with of patients with ARDS who may benefit from surfactant
severe direct lung injury. Am J Respir Crit Care Med therapy: a pooled analysis of five studies with recombi-
2011;183:1055-61. nant surfactant protein-C surfactant (Venticute). Chest
2008;134:724-32.
Steinberg KP, Hudson LD, Goodman RB, et al. Efficacy and
safety of corticosteroids for persistent acute respiratory Venkataraman ST. Mechanical ventilation and respira-
distress syndrome. N Engl J Med 2006;354:1671-84. tor care. In: Fuhrman BP, Zimmerman JJ, eds. Pediatric
Critical Care, 4th ed. Philadelphia: Elsevier, 2011.
Tamburro RF, Kneyber MCJ. Pulmonary specific ancil-
lary treatment for pediatric acute respiratory distress Willson DF, Truwit JD, Conaway MR, et al. The adult calfac-
syndrome: proceedings from the Pediatric Acute Lung tant in acute respiratory distress syndrome trial. Chest
Injury Consensus Conference. Pediatr Crit Care Med 2015;148:356-64.
2015;16:S61-S72.
Willson WF, Thomas NJ, Tamburro R, et al. Pediatric calfac-
Tamma PD, Turnbull AE, Milstone AM, et al. Ventilator- tant in acute respiratory distress syndrome trial. Pediatr
associated tracheitis in children: does antibiotic duration Crit Care Med 2013;14:657-65.
matter? CID 2011;52:1324-31.
Yehya N, Servaes S, Thomas HJ. Characterizing the degree
Tang BMP, Craig JC, Eslick GD, et al. Use of corticosteroids in of lung injury in pediatric acute respiratory distress syn-
acute lung injury and acute respiratory distress syndrome: drome. Crit Care Med 2015;43:937-46.
a systematic review and meta-analysis. Crit Care Med
2009;37:1594-603.

PedSAP 2016 Book 2 Pediatric Critical Care 30 ARDS and Ventilator-Associated Events
Self-Assessment Questions
Questions 17 pertain to the following case. C. PF ratio less than 200 and PEEP of 5 cm H2O or
E.S. is a 5-year-old boy (height 42 inches, weight 31 kg) who greater, moderate PARDS
received a stem cell transplant for hemophagocytic lym- D. PF ratio less than 200 and PEEP of 5 cm H2O or
phohistiocytosis. However, despite initial engraftment, E.S. greater, severe PARDS
experienced graft rejection. He. is admitted to the PICU with
2. On day 2 of intubation and mechanical ventilation, E.S.s
signs and symptoms consistent with septic shock. His blood
oxygenation continues to worsen; his OI is now 18, and
cultures are positive for gram-negative rods, and he is initiated
the team is considering a transition to high-frequency
on meropenem and amikacin. On PICU day 1, despite bilevel
oscillatory ventilation (HFOV). Because of the patients
positive airway pressure (BiPAP), his respiratory status contin-
worsening clinical status, the PICU attending would like
ues to deteriorate. E.S. is intubated for impending respiratory
to discuss administering calfactant. Which one of the
failure. He is initiated on continuous infusion fentanyl and mid-
following best justifies administering a single dose of
azolam for pain and sedation management during mechanical
calfactant by endotracheal tube for E.S.?
ventilation. He is maintained NPO (nothing by mouth) and is ini-
tiated on intravenous pantoprazole for stress-related mucosal A. His oxygenation status is worsening, and surfactant
disease (SRMD) prophylaxis. Over the first 24 hours in the PICU, has consistently shown sustained improvements in
his oxygenation continues to worsen. Morning chest radiogra- oxygenation.
phy is consistent with new bilateral infiltrates. His ventilator B. With an immunocompromised host, surfactant has
settings and arterial blood gas are as follows: shown a potential improvement in clinical outcomes.
C. Because he developed acute respiratory distress
syndrome (ARDS) from an indirect lung injury,
Ventilator settings Arterial blood gas
surfactant is most likely to be beneficial.
SIMV-VC pH = 7.26 D. With the impending need for HFOV, surfactant may
Rate = 18 Pco2 = 61 be used as a strategy to prevent the need to escalate
Fio2 = 0.55 Pao2 = 99 mechanical ventilation support.
Ti = 0.8 Bicarbonate (calcu-
3. On day 8 of mechanical ventilation, E.S.s oxygenation
Positive inspiratory pressure (PIP) = lated) = 27
has not improved clinically, and his PARDS is still cate-
41 cm H2O Sao2 = 97
gorized as severe. Which one of the following drugs is
Set Vt = 200 mL
best to recommend for E.S.?
Positive end-expiratory pressure
(PEEP) = 10 cm H2O A. Calfactant 80 mL/m2 by endotracheal tube once
PS = 10 cm H2O B. Methylprednisolone 60 mg intravenously every 12
Paw = 18 cm H2O hours
C. Inhaled nitric oxide 20 ppm continuous
Blood culture data: Three of three sets of cultures are posi- D. Albuterol 2.5 mg nebulized every 4 hours
tive for Klebsiella pneumonia susceptibility data as follows:
4. Which one of the following strategies would best prevent
ventilator-associated pneumonia (VAP) in E.S.?
Susceptible Intermediate Resistant
A. Discontinue pantoprazole, assess safety of daily
Amikacin ( [MIC] Imipenem (MIC Cefepime (MIC 64) sedation awakening, elevate head of the bed to 30
= 16) = 2) Gentamicin (MIC 16) degrees, change ventilator circuit every 24 hours,
Tigecycline Levofloxacin (MIC 8) and provide routine oral care with chlorhexidine.
(MIC = 4) Meropenem (MIC = 8) B. Continue pantoprazole, assess safety of daily
Piperacillin/tazobac- sedation awakening, elevate head of the bed to 15
tam (MIC 128) degrees, change ventilator circuit every 24 hours,
Tobramycin (MIC 16) and provide routine oral care with chlorhexidine.
C. Continue pantoprazole, assess safety of daily
1. Given his clinical signs and symptoms and his clinical sedation awakening, elevate head of the bed to 30
and laboratory data, which one of the following best degrees, change ventilator circuit when visibly soiled,
describes E.S.s diagnosis? and provide routine oral care with chlorhexidine.
D. Discontinue pantoprazole, assess safety of daily
A. Oxygenation index (OI) = 10, moderate pediatric
sedation awakening, elevate head of the bed to 15
acute respiratory distress syndrome (PARDS)
degrees, change ventilator circuit when visibly soiled,
B. OI = 10, severe PARDS
and provide routine oral care with chlorhexidine.

PedSAP 2016 Book 2 Pediatric Critical Care 31 ARDS and Ventilator-Associated Events
5. On day 14 of mechanical ventilation, E.S. develops a Questions 810 pertain to the following case.

new consolidation on chest radiography. Despite previ- K.M. is a 2-year-old girl (weight 15 kg) with a medical his-
ous improvements in oxygenation, his Fio2 requirement tory positive for trisomy 21, complete atrioventricular canal
is increasing this morning, and he is febrile (tempera- after repair at 6 months, and mild pulmonary hypertension.
ture 102.2F [39C]). His WBC is 1.57 x 103 cells/mm3. A She presents to the PICU with respiratory distress requiring
mini-BAL is obtained, and Gram stain shows few WBCs full face mask BiPAP, for which she is receiving dexmedeto-
and rare gram-negative rods. Which one of the following midine infusion to enhance tolerance of therapy. Her BiPAP
best describes E.S.s clinical criteria for a diagnosis of settings are 10 cm H2O/6 cm H2O with an Fio2 of 60%, and her
VAP? current Sao2 is 85%. K.M.s respiratory viral diagnostics are
positive for influenza A.
A. New consolidation on chest radiography, worsening
gas exchange, febrile 8. Which one of the following best describes K.M.s oxygen-
B. Worsening gas exchange, febrile, leukopenia ation impairment?
C. New consolidation on chest radiograph, worsening A. PF ratio = 140
gas exchange, mini-BAL Gram stain positive for B. SF ratio = 140
gram-negative rods C. PF ratio = 7
D. Worsening gas exchange, febrile, mini-BAL Gram D. SF ratio = 7
stain positive for gram-negative rods
9. K.M.s condition continues to worsen on BiPAP, and she
6. Given his historical data and the new information, which requires intubation for mechanical ventilation. Given her
one of the following regimens is most appropriate to rec- OI, she is classified as having severe PARDS. The attend-
ommend for E.S.? ing physician requests beractant administration for
A. Cefepime and amikacin PARDS secondary to pneumonia. Which one of the fol-
B. Imipenem/cilastatin and amikacin lowing responses to this request would be best for K.M.?
C. Cefepime and tobramycin A. Beractant is not FDA labeled for this indication;
D. Imipenem/cilastatin and tobramycin the medical team should submit an emergency
7. On hospitalization day 16 for E.S., the culture results Investigational New Drug application before use.
from the mini-BAL done 2 days earlier return: B. Exogenous surfactant does not improve clinical
outcomes and is not recommended for use in
Mini-BAL: Many WBCs, no epithelial cells > 100,000 CFU/
PARDS.
mL Klebsiella pneumoniae Susceptibilities:
C. Calfactant is a better choice of surfactant product
Susceptible: Amikacin (MIC = 2), cefepime (MIC = 1),
than beractant to treat ARDS.
ciprofloxacin (MIC = 1), levofloxacin (MIC = 1), merope-
D. Exogenous surfactant cannot be used beyond the
nem (MIC 0.25), piperacillin/tazobactam (MIC = 16)
neonatal period because of the excessive instillation
Resistant: Ceftriaxone, gentamicin (MIC 16), tobramy-
volume.
cin (MIC 16)
During this period, E.S.s oxygenation has improved clin- 10. On intubation day 14, K.M. continues to require signifi-
ically. Given his clinical status and culture data, which cant ventilator support, and the team would like to initiate
one of the following is best to recommend for E.S.? methylprednisolone by the Meduri protocol. Which one
of the following is best to recommend for K.M.?
A. Ciprofloxacin, complete a total antibiotic course of 8
days A. Do not use corticosteroids in this patient with ARDS.
B. Ciprofloxacin, complete a total antibiotic course of B. Use an alternative dosing protocol that has been
15 days reported in pediatric patients.
C. Cefepime, complete a total antibiotic course of 8 C. Use hydrocortisone at doses equivalent to the
days methylprednisolone dosing protocol.
D. Cefepime, complete a total antibiotic course of 15 D. Provide frequent blood glucose monitoring.
days 11. Which one of the following statements best describes
the risk and benefits of using corticosteroids in adult
patients with ARDS?
A. High-dose methylprednisolone may be beneficial
in the prevention of ARDS, but patients should
be closely monitored for the development of new
infections.

PedSAP 2016 Book 2 Pediatric Critical Care 32 ARDS and Ventilator-Associated Events
B. Low- to moderate-dose methylprednisolone is tracheostomy. Now, on day 16 of mechanical ventilation, 2
associated with a higher risk of death when used days before his scheduled tracheostomy, K.N. is febrile (tem-
in the late phase of ARDS and places patients at an perature 101.3F [38.5C]). The volume and thickness of his
increased risk of infections. secretions have not changed. A mini-BAL is sent. His chest
C. High-dose methylprednisolone may be beneficial radiography is stable with no signs of new infiltrate. K.N.s
in the prevention of ARDS, but patients should be WBC is 10.5 103 cells/mm3. His ventilator parameters remain
closely monitored for neuromuscular weakness. stable, and K.N. continues to oxygenate well.
D. Low- to moderate-dose methylprednisolone may
14. Given his clinical signs and symptoms, which one of the
beneficial in the late phase of ARDS when initiated
following diagnoses is K.N. most likely to receive?
before day 14 of illness, but patients should be
monitored closely for neuromuscular weakness. A. Ventilator-associated tracheobronchitis
B. Ventilator-associated condition (VAC)
12. A 15-year-old girl is admitted to an adult ICU after a coil-
C. VAP
ing arteriovenous malformation; she remains intubated.
D. Infection-related VAC (IVAC)
Which one of the following is the best quality reporting
approach for this patient? 15. The Gram stain from K.N.s mini-BAL specimen shows
moderate WBC and gram-negative rods. The team wishes
A. Given her age, reporting should occur by the device-
to initiate antibiotic therapy, pending culture results. K.N.
related pneumonia module. Surveillance reporting
has not received any antimicrobial therapy during this
begins on day 1 of mechanical ventilation.
hospitalization. Which one of the following is the most
B. Given her age, reporting should occur by the device-
appropriate empiric antibiotic regimen for K.N?
related pneumonia module. Surveillance reporting
begins on day 3 of mechanical ventilation. A. Ceftriaxone plus gentamicin
C. Given her location, reporting should occur by the B. Ceftriaxone
device-related ventilator-associated event (VAE) C. Cefepime plus gentamicin
module. Surveillance reporting begins on day 1 of D. Cefepime
mechanical ventilation. 16. On day 2 of K.N.s antimicrobial therapy, the following
D. Given her location, reporting should occur by the culture data become available:
device-related VAE module. Surveillance reporting
Mini-BAL: Moderate WBCs, no epithelial cells
begins on day 3 of mechanical ventilation.
> 10,000 CFU/mL Pseudomonas aeruginosa
13. Which one of the following statements best supports Pan-susceptible
reevaluating the current reporting structure for the K.N.s chest radiography continues to be without signs
device-related pneumonia module used in pediatric of a pneumonia, and his oxygenation has not worsened.
units? Which one of the following therapy durations (in days) is
A. Clinical criteria used for surveillance reporting are best to recommend for K.N.?
consistently documented in the medical record. A. 5
B. Change in oxygenation after a period of stability is B. 10
an objective measure of clinical deterioration. C. 15
C. Chest radiography is likely to accurately identify D. 21
VAP.
17. Empiric combination therapy for gram-negative organ-
D. Clinical criteria used for surveillance reporting are
isms may not be needed in all patients initiated on
objective.
antibiotic therapy for IVACs or VAP. Assuming a low risk
of MDRGN (multidrug-resistant gram-negative organ-
Questions 1416 pertain to the following case.
isms) from the community, in which one of the following
K.N. is a 19-month-old boy (weight 8.3 kg) with a medical patients would combination empiric coverage best be
history of very long-chain acyl-coenzyme A dehydrogenase initiated?
deficiency. He was admitted to the PICU for respiratory failure
A. A previously healthy 7-month-old boy, mechanically
secondary to respiratory syncytial virus (RSV) bronchiolitis.
ventilated 3 days for respiratory failure secondary to
He was subsequently intubated and placed on continuous
RSV bronchiolitis
infusions of fentanyl and midazolam for pain and sedation
B. A previously healthy 14-year-old boy, mechanically
management. Before this hospitalization, K.N. had been rel-
ventilated for 6 days secondary to a traumatic brain
atively healthy. His home acid suppression of ranitidine was
injury
continued. He remained intubated for a prolonged course
because of weakness, and the care team planned to place a

PedSAP 2016 Book 2 Pediatric Critical Care 33 ARDS and Ventilator-Associated Events
C. A 6-year-old girl receiving induction chemotherapy Second, you are asked to develop a plan to identify which
for acute lymphocytic leukemia, mechanically pediatric patients should receive venous thromboembolism
ventilated for 8 days secondary to septic shock (VTE) prophylaxis.
D. A 12-year-old girl with asthma, mechanically
19. In which one of the following Homefield patients would
ventilated for 2 days secondary to an acute asthma
it be most appropriate to discontinue SRMD prophylaxis
exacerbation
while the patient is receiving mechanical ventilation?
18. The new model of device-related event reporting for ven-
A. A patient who sustained multiple traumatic injuries,
tilated adult patients is a multi-tier approach for which
including a severe traumatic brain injury, who is
surveillance is initiated on the basis of changes in oxy-
receiving continuous jejunal feeding
genation versus change in chest radiography. A similar
B. A patient with chronic respiratory failure, receiving
set of surveillance definitions will likely be developed
gastric feeding, who has not received acid-
for pediatric patients, with the recommendation to use
suppressive therapy chronically
mean airway pressures (MAPs) instead of PEEP. Which
C. A patient with Guillain-Barr syndrome receiving
one of the following best justifies this change?
pulse dose steroids (methylprednisolone 1000 mg x
A. MAP allows the surveillance definition to be used in 3 doses) and gastric feeding
the evaluation of patients receiving HFOV. D. A patient with acute-on-chronic hepatic failure who
B. PEEP is a better marker of lung compliance. has now developed acute kidney injury, receiving
C. MAP is more affected by worsening gas exchange in jejunal feeding
pediatric patients.
20. Which one of the following responses is best to give the
D. PEEP use is inconsistent among pediatric
Homefield Hospital working group regarding the request
intensivists.
to identify patients who should receive VTE prophylaxis?
A. No patients; pediatric patients are not at an
Questions 19 and 20 pertain to the following case.
increased risk of VTE.
Homefield Childrens Hospital is part of a large health system.
B. No patients; although some might be at an
As the PICU pharmacist, you serve on a quality committee
increased risk of VTE, no data support that
responsible for developing a VAP prevention bundle. The
prophylaxis mitigates this risk in pediatric patients.
working groups plan is to incorporate bundle recommenda-
C. Adolescent patients with known risk factors for
tions from both IHI and the literature. You have been tasked
developing VTE and no contraindications: should
with two areas of the bundle. First, as a means of mitigat-
receive at least mechanical prophylaxis.
ing modifiable risk factors, you are asked to identify patients
D. All immobilized pediatric patients as long as they
in whom discontinuing SRMD prophylaxis is acceptable.
have a normal platelet count.

PedSAP 2016 Book 2 Pediatric Critical Care 34 ARDS and Ventilator-Associated Events
Learner Chapter Evaluation: ARDS and Ventilator-Associated Events.

As you take the posttest for this chapter, also evaluate the Use the 5-point scale to indicate whether this chapter pre-
materials quality and usefulness, as well as the achievement pared you to accomplish the following learning objectives:
of learning objectives. Rate each item using this 5-point scale:
12. Using the definitions for acute respiratory distress syn-
drome (ARDS), classify a patients pulmonary morbidity.
Strongly agree
13. Analyze available data to design an appropriate phar-
Agree macotherapy plan, including exogenous pulmonary
Neutral surfactant or the use of corticosteroids, for pediatric
Disagree patients with ARDS.
Strongly disagree 14. Analyze the current definition of ventilator-associated
pneumonia to determine the need for surveillance, and
1. The content of the chapter met my educational needs. measure quality of care in pediatric ICU patients.
2. The content of the chapter satisfied my expectations. 15. Evaluate deficiencies in current surveillance definitions
3. The author presented the chapter content effectively. and justify expanded surveillance in pediatric units to
include other ventilator-associated conditions.
4. The content of the chapter was relevant to my practice
and presented at the appropriate depth and scope. 16. For ventilator-associated infections in the pediatric ICU,
design antimicrobial therapy plan that uses the narrow-
5. The content of the chapter was objective and balanced.
est-spectrum antibacterial agents for a duration that
6. The content of the chapter is free of bias, promotion, or minimizes antibacterial exposure.
advertisement of commercial products.
17. Justify a prevention strategy for ventilator-associated
7. The content of the chapter was useful to me. events in the pediatric ICU including, but not limited to,
8. The teaching and learning methods used in the chapter implementation of a ventilator-associated pneumonia
were effective. bundle.
9. The active learning methods used in the chapter were 18. Please provide any specific comments related to any
effective. perceptions of bias, promotion, or advertisement of
commercial products.
10. The learning assessment activities used in the chapter
were effective. 19. Please expand on any of your above responses, and/or
provide any additional comments regarding this chapter:
11. The chapter was effective overall.

PedSAP 2016 Book 2 Pediatric Critical Care 35 ARDS and Ventilator-Associated Events
Hemodynamic Instability

By Regine L. Caruthers, Pharm.D.

Reviewed by Ashley R. Schappell, Pharm.D., BCPS; and Michael A. Maccia, Pharm.D., BCPS

LEARNING OBJECTIVES

1. Distinguish between the four types of shock using physical examination findings and laboratory measurements.
2. Classify the types of fluid used in resuscitating the patient with hypovolemic shock.
3. Justify the use of an inotrope or vasopressor in the treatment of circulatory shock.
4. Account for the mechanisms by which circulatory shock occurs.
5. Distinguish between hypertensive urgency and hypertensive emergency.

INTRODUCTION
ABBREVIATIONS IN THIS CHAPTER
Hemodynamic instability, also known as shock, reflects a state of
CO Cardiac output
inadequate perfusion and oxygenation (Kleinman 2010; Mitrovic
PE Pulmonary embolism
2010; Weil 2005). Although there are many causes of shock, they can
VAD Ventricular assist device
all be categorized under one of the following types: hypovolemic,
Table of other common abbreviations. obstructive, cardiogenic, or distributive (Weil 2005). Hypovolemic
shock is the most common type reported in children and is the lead-
ing cause of mortality in pediatric patients in the United States and
the world (Pasman 2015; Kleinman 2010). Early recognition and diag-
nosis of shock can significantly decrease morbidity and mortality
(Pasman 2015).
Signs and symptoms depend on the phase of shock, which can be
classified as compensated, decompensated, and/or irreversible. In
compensated shock, the body tries to maintain perfusion and oxy-
genation (i.e., cardiac output [CO] and perfusion pressure) and is
thus associated with tachycardia, increased systemic vascular resis-
tance, cool and pale extremities, prolonged capillary refill (greater
than 2 seconds), weak peripheral pulses, and normal blood pressures
(Kleinman 2010). Decompensated shock results when compensatory
mechanisms fail to maintain adequate CO and end-organ perfu-
sion, which ultimately leads to profound hypotension and the need
for pharmacologic treatment (Table 2-1) (Kleinman 2010). Signs of
this decompensated state include altered mental status, decreased
urine output, metabolic acidosis, tachypnea, weak central pulses,
weak-to-absent peripheral pulses, and skin mottling (Mtaweh 2013;
Kleinman 2010). Irreversible shock is a state beyond decompensated
shock in which death is inevitable, despite appropriate resuscitation
(McKiernan 2005).

PedSAP 2016 Book 2 Pediatric Critical Care 37 Hemodynamic Instability


HYPOVOLEMIC SHOCK renin, and aldosterone (Mitronvic 2005). Hypernatremia, hyper-
Hypovolemic shock results from a deficiency in intravascu- chloremia, increased BUN/SCr ratio, and an increase in base
lar blood volume and thus, an inability to maintain CO and deficit and lactate are also characteristics of hypovolemic
end-organ perfusion (Mitrovic 2010). Disease states associ- shock. Symptoms on presentation vary with the etiology and
ated with this specific type of shock include trauma, surgery, degree of hypovolemic shock but may include increased thirst,
GI bleeding, gastroenteritis, burns, diabetes insipidus, heat nausea, weakness, light-headedness, and dizziness.
stroke, and capillary leak with third spacing. The mainstay of treatment for hypovolemic shock is fluid
Signs of hypovolemic shock include hypotension, tachycar- resuscitation with crystalloid solutions, with sodium con-
dia, increased systemic vascular resistance (vasoconstriction), centrations approximating normal sodium values (i.e., 0.9%
tachypnea, and decreased urine output; the specific compen- sodium chloride) and lactated Ringer solution (DiPiro 2011).
satory mechanisms in the hypovolemic state can be attributed Selection of a sodium-containing intravenous fluid is based
to an increase in the secretion of catecholamines (epinephrine, solely on clinician preference. Because of the large amount
norepinephrine), vasopressin, angiotensin II, glucocorticoids, of fluid needed to correct the intravascular volume deficit,
smaller boluses repeated over time achieve hemodynamic
stability more safely and efficiently; this is therefore the rec-
ommended practice (McKiernan 2005). This recommendation
is especially pertinent when caring for patients with heart fail-
BASELINE KNOWLEDGE STATEMENTS
ure and those at risk of pulmonary edema. The clinician should
Readers of this chapter are presumed to be familiar consider administering several fluid boluses at 1020 mL/kg.
with the following: Colloid solutions (e.g., 5% and 25% albumin) and hydroxyethyl
Knowledge of normal ranges of vital signs in starch are safe and effective treatment options but have not
children been shown to be more advantageous than crystalloid solutions
Baseline knowledge of inotropes and vasopressors in the treatment of hypovolemic shock. Colloid solutions are not
Basic knowledge of anticoagulant and antiplatelet recommended as the preferred treatment option because of
therapies their higher cost and lack of additional benefits. Resuscitation
Understanding of prehypertension, stage 1 with blood products is no more effective than other colloid solu-
hypertension, and stage 2 hypertension for age, tions and, in fact, poses issues including an increase in blood
sex, and height percentile in pediatric patients
viscosity, immunomodulatory concerns, and infection risk.
Table of common pediatric laboratory reference values.
OBSTRUCTIVE SHOCK
ADDITIONAL READINGS Major impediments to pulmonary and systemic blood flow
ultimately impair CO and end-organ perfusion, resulting in
The following free resources are available for readers obstructive shock (Mitronic 2010; Weil 2005). Common causes
wishing additional background information on this
topic.
McKiernan CA, Lieberman SA. Circulatory shock in
Table 2-1. General Dosing of Vasopressor and
children: an overview. Pediatr Rev 2005;26:451-60.
Inotropic Agents
Monagle P, Chan AKC, Goldenberg NA, et al.
Antithrombotic therapy in neonates and children:
Initial Dose Dosing Range
antithrombotic therapy and prevention of thrombo-
sis: American College of Chest Physicians Dopamine 25 mcg/kg/min 220 mcg/
evidence-based clinical practice guidelines. Chest kg/min
2012;141(suppl 2):e737S-e801S.
Epinephrine 0.010.05 mcg/ 0.012 mcg/
National High Blood Pressure Education Program kg/min kg/min
(NHBPEP) Working Group on High Blood Pressure
in Children and Adolescents. The fourth report on Milrinone 0.25-0.3 mcg/ 0.25-0.75 mcg/
the diagnosis, evaluation, and treatment of high kg/min kg/min
blood pressure in children and adolescents.
Norepinephrine 0.020.05 mcg/ 0.022 mcg/
Pediatrics 2004;114(suppl 2):1-22.
kg/min kg/min
Overgaard CB, Dzavik V. Inotropes and vasopres-
sors: review of physiology and clinical use in Phenylephrine 0.050.1 mcg/ 0.050.5 mcg/
cardiovascular disease. Circulation kg/min kg/min
2008;118:1047-56.
Vasopressin 0.00010.0002 0.00010.002
unit/kg/min unit/kg/min

PedSAP 2016 Book 2 Pediatric Critical Care 38 Hemodynamic Instability


of obstructive shock include acute pulmonary embolism (PE), anticoagulants continue to have a place in treatment because
tension pneumothorax, and cardiac tamponade (Pasman 2015; of the risks associated with thrombolytic therapy and embo-
Weil 2005). Signs and symptoms of obstructive shock depend lectomy. Unfractionated heparin is preferred because of
on the cause of obstruction. The following discussion focuses its pharmacokinetic profile (i.e., shorter half-life); however,
on acute PE and the development of obstructive shock. low-molecular-weight heparins can be used to stabilize
Pulmonary embolism is a rare but potentially fatal disease the thrombus and prevent enlargement, of which enoxa-
in children (Van Ommen 2006). The nonspecific and often parin has the most pediatric clinical data (Monagle 2012).
masked symptoms of PE (e.g., shortness of breath, pleuritic Unfractionated heparin should be titrated to a goal activated
chest pain, hemoptysis, persistent tachypnea) make the diag- PTT correlating with an anti-factor Xa (anti-Xa) of 0.350.7
nosis difficult and the disease easy to overlook (Patocka 2012; unit/mL. Subcutaneous enoxaparin doses should be adjusted
Biss 2008; Van Ommen 2006). The definitive diagnosis of PE to obtain an anti-Xa of 0.51 unit/mL achieved 46 hours
can be made using several imaging modalities, including pul- after a dose is administered. Table 2-2 lists initial dose rec-
monary angiography, ventilation-perfusion lung scanning, ommendations for heparin and enoxaparin.
helical CT, MRI, and echocardiography (Patocka 2012).
Clinical manifestations of PE with hemodynamic instability
can occur when an acute thrombus occludes more than 50% Table 2-2. Initial Dosing Recommendation for
of the pulmonary circulation (Van Ommen 2006). An acute PE Heparin and Enoxaparin in the Treatment of Pulmonary
negatively affects the integrity and function of the right and left Embolism
ventricles of the heart. Impaired ventricular function is a con-
sequence of the following cascade of events (Figure 2-1). Left Age Initial Dose

ventricular dysfunction alters CO and leads to systemic hypo- Heparin < 1 yr Bolus: 75 units/kg over 10 min
tension and inadequate perfusion to the periphery. Altered Continuous infusion: 28 units/kg/hr
coronary perfusion with subsequent myocardial ischemia
> 1 yr Bolus: 75 units/kg over 10 min
can occur in the setting of right-sided heart failure and hypo- Continuous infusion: 20 units/kg/hr
tension (Van Ommen 2006; Smulders 2000). Other signs of
Enoxaparin < 2 mo 1.5 mg/kg/dose SC q12hr
hemodynamic instability during an acute PE include cyanosis,
dysrhythmia, pallor, and syncope (Patocka 2012). > 2 mo 1 mg/kg/dose SC q12hr
Prompt dissolution using thrombolysis or removal by
embolectomy is the treatment of choice for PE with hemo- q = every; SC = subcutaneously.
dynamic instability (Van Ommen 2006). Despite this goal,

Acute increase in Increase in right


Dilation of the
right ventricular ventricular and
right ventricle
afterload pulmonary artery
systolic pressures

Abnormal filling of Leftward shift of Right-sided heart


the left ventricle the intraventricular failure and tricuspid
during diastole septum regurgitation

Hypotension and
Altered cardiac
inadequate
output
systemic perfusion

Figure 2-1. Pulmonary embolism and hemodynamic instability.


Information from Smulders YM. Pathophysiology and treatment of haemodynamic instability in acute pulmonary embolism: the
pivotal role of pulmonary vasoconstriction.Cardiovasc Res 2000;48:23-33; Van Ommen CH, Peters M. Acute pulmonary embolism in
childhood.Thromb Res2006;118:13-25; and Wood KE. Major pulmonary embolism: review of a pathophysiologic approach to the
golden hour of hemodynamically significant pulmonary embolism.CHEST 2002;121:877-905.

PedSAP 2016 Book 2 Pediatric Critical Care 39 Hemodynamic Instability


When thrombolytic therapy is considered, alteplase is the adult patients with cardiogenic shock compared with norepi-
agent of choice for pediatric patients (Monagle 2012). Other nephrine (De Backer 2010). In the absence of pediatric data,
thrombolytic agents (i.e., urokinase and streptokinase) are clinicians have begun to consider the negative findings in the
effective for clot resolution. However, previous concerns adult population and to preferentially use epinephrine as the
about urokinases efficacy, as well as concerns about fibrin first-line inotropic agent.
specificity and immunogenicity of urokinase and streptoki- When drugs fail to stabilize blood pressure and restore
nase, have made them second-line agents (Monagle 2012; perfusion to the vital organs, mechanical circulatory support
Gupta 2001; PHS 1999). Alteplase is the drug of choice in with extracorporeal membrane oxygenation or an extracor-
pediatric patients; although optimal dosing of intravenous poreal ventricular assist device (VAD) may be needed (Kirk
alteplase has not been explained, 0.5 mg/kg/hour is the dose 2014; Mitaweh 2013). These modalities help unload the ven-
most commonly reported (Monagle 2012; Wang 2003). tricles, decrease myocardial demand, improve coronary
Data comparing low-dose alteplase (0.010.06 mg/kg/ perfusion, and stabilize hemodynamics to reestablish sys-
hour) with standard-dose alteplase (0.10.5 mg/kg/hour) temic perfusion (Kirk 2014; Subramaniam 2012). Mechanical
are limited to small cohort studies and single-center expe- circulatory support can be used as a bridge to transplanta-
riences. One of these studies evaluated and compared the tion, candidacy for transplantation, or recovery. It can also be
efficacy and safety of low- and standard-dose alteplase in the destination therapy (i.e., long-term support on a VAD when
treatment of arterial and venous thrombi. Thirty-five patients candidacy for transplantation is limited or nonexistent); this
were enrolled, and complete clot resolution occurred in 97% is becoming increasingly possible with improved VAD tech-
of patients with acute thrombi (17 of 17 patients in the low- nologyis center-dependent and not routinely recommended
dose group and 11 of 12 patients in the standard-dose group); thearpy ng strategies (high and low dose). The previous state-
all six patients with chronic thrombi had partial resolution. ment includes thi.
One patient had a major bleed (Wang 2003). The decision to The advances in VAD technology and the use of VAD
use low-dose versus standard-dose alteplase is based on in pediatric patients do not parallel the trend in adults
patient characteristics (e.g., age, symptoms, clot size), risks, (VanderPluym 2014). Even so, six VADs are currently avail-
and benefits. Adult dosing (100 mg over 2 hours) should be able for use in pediatric patients (Table 2-3). Anticoagulation
considered in larger pediatric patients. and antiplatelet therapies pose the most significant chal-
lenges for pharmacists caring for patients on a VAD. There is
a fine balance between clot formation and bleeding complica-
CARDIOGENIC SHOCK tions in this patient population, and the optimal approach has
Cardiogenic shock occurs when impaired myocardial con- yet to be explained. Anticoagulants of choice include hepa-
tractility results in decreased stroke volume, CO, and systemic rin, low-molecular-weight heparins, and warfarin. Heparin is
hypoperfusion (Pasman 2015; Subramaniam 2012; Tobias most often used in the immediate postoperative period, with
1996). Common causes of cardiogenic shock include arrhyth- eventual transition to warfarin for chronic anticoagulation.
mias, cardiomyopathies, myocarditis, and congenital heart The target for unfractionated heparin is an anti-Xa activity of
disease (Pasman 2015; McKiernan 2005). Compensatory 0.350.7 unit/mL and an INR of 23 for most VADs. The role of
mechanisms include activation of the renin-angiotensin- low-molecular-weight heparins varies and is limited by renal
aldosterone system to increase fluid retention, sympathetic clearance and long half-life in the immediate postoperative
activation to increase contractility, and peripheral vaso- period. Scant pediatric data limit the use of other anticoag-
constriction to perfuse only the vital organs (Long 2015). ulants, including fondaparinux, apixaban, and rivaroxaban.
Symptoms of compensated cardiogenic shock are tachycar- Dual antiplatelet therapy with aspirin and dipyridamole or
dia, cool skin, decreased peripheral pulses, and deceased clopidogrel is the standard of care. Monitoring with throm-
urine output (Long 2015; Subramaniam 2015). boelastography studies is center-dependent and has not yet
Treatment options include drugs to acutely improve con- been incorporated into the clinical practice guidelines and
tractility of the heart, CO, and blood pressure. A combination recommendations.
of milrinone and a vasopressor agent is the recommended
treatment. Milrinone increases myocardial contractility and
provides afterload reduction, improving CO without increas- DISTRIBUTIVE SHOCK
ing oxygen consumption (Kirk 2014; Bailey 1999; Chang Distributive shock is caused by inappropriate relaxation
1995). The potential for hypotension exists but usually does of peripheral vascular tone and profound vasodilation that
not outweigh the benefits of milrinone. Despite the paucity results in increased venous capacitance, relative hypovole-
of data in pediatric patients, dopamine and epinephrine are mia with no actual fluid loss, and decreased preload (Pasman
routinely used to improve cardiac contractility and increase 2015; Mitrovic 2010). A decrease in preload (i.e., filling vol-
systemic vascular resistance. In a subgroup analysis, dopa- umes) leads to a decrease in stroke volume, which ultimately
mine was associated with increased mortality at 28 days in decreases CO and end-organ perfusion. Common causes

PedSAP 2016 Book 2 Pediatric Critical Care 40 Hemodynamic Instability


Table 2-3. VADs Used in Pediatric Patients

Ventricular Assist Device Population for Use Uses

Berlin Heart (EXCOR) Infants LVAD, RVAD, BiVADa


Smaller children

Thoratec VAD (IVAD/PVAD)b Children with BSA > 1.5 m2 LVAD, RVAD, BiVAD

HeartMate II Children with BSA 1.4 m2 LVAD

HeartWare HVAD Children with BSA 1.1 m2 LVAD

Total artificial heart Children with BSA 1.7 m2 Replaces both ventricles

HeartAssist 5 pediatric VAD (formerly the Children 516 yr with BSA 0.71.5 m 2
LVAD
DeBakey VAD Child)

a
Can support one or two ventricles in any anatomic arrangement
b
VAD not commonly used because of improved technology.
BiVAD = biventricular assist device; BSA = body surface area; LVAD = left ventricular assist device; RVAD = right ventricular assist
device; VAD = ventricular assist device.
Information from: Kirk R, Dipchand AI, Rosenthal DN. The International Society for Heart and Lung Transplantation Guidelines for
the management of pediatric heart failure. J Heart Lung Transplant 2014;33:888-909; and VanderPluym C, Urschel S, Buchholz H.
Advanced therapies for congenital heart disease: ventricular assist devices and heart transplantation. Can J Cardiol
2013;29:796-802.

include sepsis, drug intoxications, anaphylaxis, and neuro- of continuous infusion epinephrine are more beneficial than
logic injury, including head and spinal injuries (Tobias 1996). its vasopressor effects in anaphylactic shock.
Sepsis and septic shock are the most common cause and Although the mechanism and pathophysiology of cardio-
form of distributive shock and are associated with significant vascular collapse are poorly understood, cardiac support and
morbidity and mortality. stabilization with improvement in heart contractility and CO are
Anaphylaxis describes a state in which the bodys pivotal in minimizing lethal outcomes (Brown 2007). Aggressive
response to an allergic reaction is excessive. During anaphy- volume resuscitation with 20 mL/kg of normal saline increases
laxis, the mast cells and basophils become unstable and leak intravascular volume, preload, stroke volume, and ultimately
vasoactive mediators, including histamine, heparin, tryptase, CO. On rare occasions, other agents have been needed to
chymase, and tumor necrosis factor , into the smooth mus- completely normalize venous capacitance. In case reports,
cles of the lung, heart, and systemic vasculature (Kirkpatrick phenylephrine and norepinephrine are second-line options
2008; Brown 2007; Fisher 1992). This can lead to wheezing, that have been used with variable outcomes. Vasopressin and
shortness of breath, decreased tone of the blood vessels, metaraminol (unavailable in the United States) have also been
decreased blood pressure, skin flushing, hives, vomiting, diar- effective in case reports (Heytman 2004; Kill 2004; Schummer
rhea, decreased myocardial contractility, bradycardia, and 2004). Atropine can be used in sustained bradycardia, despite
third spacing (Brown 2007; Heytman 2004; Adams 2001). Any treatment with inotropic and vasopressor agents.
combination of these symptoms can decrease CO and oxy- Neurogenic shock is defined as the presence of hypoten-
genation; however, the changes in vascular tone and cardiac sion and relative bradycardia after head and high-level spinal
function are most often associated with the development of cord injury (often at the cervical or high thoracic level); com-
anaphylactic shock (Brown 2007). pensatory tachycardia is not present in neurogenic shock
Treatment of anaphylactic shock differs from that of ana- (Breslin 2012; Guly 2008; Cocchi 2007). Neurogenic shock
phylaxis (i.e., antihistamine, corticosteroids, intramuscular or is a diagnosis of exclusion that results from the sudden loss
subcutaneous epinephrine). Treatment of anaphylactic shock of sympathetic autonomic activity followed by vasodilation,
focuses on the symptomatic treatment of myocardial dys- decreased venous capacitance, altered CO, hypotension,
function and the hypovolemic state secondary to the change and bradycardia (Summers 2013; Mitrovic 2010; ACS 2008).
in venous capacitance and third spacing. First-line treat- Treatment options include intravenous fluids to improve CO
ment options include continuous infusion epinephrine and and vasopressors and inotropes to improve vascular tone
aggressive fluid resuscitation; second-line options include (Kirkpatrick 2008; Cocchi 2007). Use of vasopressors and
norepinephrine or phenylephrine and atropine (Hussain 2008; inotropes should be withheld until fluid resuscitation is com-
Brown 2007; Brown 2004; Fisher 1986). The inotropic effects plete (Kirkpatrick 2008).

PedSAP 2016 Book 2 Pediatric Critical Care 41 Hemodynamic Instability


Table 2-4. Intravenous Antihypertensive Agents

Bolus Dose Initial Dosing Dosing Range Additional Information

Enalaprilat None 0.05 mg/kg/dose 0.050.1 mg/kg q6hr Can cause acute renal failure and
as an IV bolus Max 1.25 mg/dose hyperkalemia

Esmolol 100500 mcg/kg 25-100 mcg/kg/min 100500 mcg/kg/min Monitor to avoid profound
Max 80 mg/dose bradycardia

Fenoldopam None 0.2 mcg/kg/min 0.20.8 mcg/kg/min Increases intracranial pressure

Hydralazine None 0.1 mg/kg/dose q46hr 0.10.2 mg/kg/dose


Max 20 mg/dose q46hr
Max 20 mg/dose

Labetalol 0.11.0 mg/kg 0.251 mg/kg/hr 0.253 mg/kg/hr Intermittent doses at 0.31 mg/kg/
Max 40 mg/dose dose can also be used

Nicardipine None 0.5-1 mcg/kg/min 13 mcg/kg/min Increases intracranial pressure

Nitroprusside None 0.3-0.5 mcg/kg/min 0.510 mcg/kg/min Monitor cyanide and thiocyanate
concentrations with doses greater
than 1.8 mcg/kg/min, prolonged
use or in renal failure to minimize
toxicity

Phentolamine None 0.1 mg/kg/dose 0.10.2 mg/kg/dose


Max 5 mg/dose

Information from: Lexicomp; Chandar J, Zilleruelo G. Hypertensive crisis in children. Pediatr Nephrol 2012;27:741-51; and National
High Blood Pressure Education Program (NHBPEP) Working Group on High Blood Pressure in Children and Adolescents. The fourth
report on the diagnosis, evaluation, and treatment of high blood pressure in children and adolescents. Pediatrics 2004;114:1-22.

HYPERTENSIVE URGENCY hydralazine, labetalol, nicardipine, and sodium nitroprusside


AND EMERGENCY (Chandar 2012; Patel 2012). Other treatment options include
Pediatric hypertensive urgency and emergency are defined phentolamine, fenoldopam, and enalaprilat (Chandar 2012).
by an acute change in blood pressure well above the 99th Selection depends on the underlying cause of hypertension
percentile for age, sex, and height. Hypertensive urgency and the clinicians comfort and preference (Table 2-4). Goals
is usually associated with nausea and vomiting but poses of therapy include no more than a 25% reduction in blood
no risk to end-organ function (Chandar 2012). In contrast, pressure within 8 hours of initiating treatment, followed by a
a hypertensive emergency is usually associated with more slower decrease in blood pressure over the next 2448 hours
severe symptoms, including encephalopathy and seizures, (Chandar 2012; NHBPEP 2004).
and can lead to rapid end-organ damage. End-organ dam- Hypertensive urgency and emergency rarely occur in pedi-
age can result in congestive heart failure, encephalopathy, or atric patients. Prompt treatment with appropriate monitoring
renal failure (Patel 2012; NHBPEP 2004). Although pediatric is needed to minimize end-organ damage.
hypertension usually develops secondary to renal or cardiac
etiologies, a pediatric hypertensive emergency is angioten-
sin-dependent and has been associated with high vascular
CONCLUSION
reactivity, elevated concentrations of norepinephrine and Hemodynamic instability reflects a complex of decreased
vasopressin, and decreased concentrations of vasodilating or inadequate perfusion and oxygenation. Causes of hemo-
hormones (Chandar 2012). dynamic instability include hypovolemia, primary cardiac
Only intravenous drugs should be used in the treatment of dysfunction, trauma with spinal cord injury, hyperacute
a hypertensive emergency, unlike hypertensive urgencies, in response to anaphylaxis, obstruction, and acute rise in blood
which intravenous or oral medications can be used (NHBPEP pressure. Early recognition and prompt treatment of the
2004). The most effective and commonly used treatment underlying cause and supportive care are key in minimizing
options for a hypertensive emergency include esmolol, morbidity and mortality in all forms of shock.

PedSAP 2016 Book 2 Pediatric Critical Care 42 Hemodynamic Instability


Fisher M. Treating anaphylaxis with sympathomimetic
Practice Points drugs. BMJ 1992;305:1107-8.
Hemodynamic instability is caused by a cascade of events
that ultimately result in decreased or inadequate perfusion Fisher MM. Clinical observations on the pathophysiology
and oxygenation. and treatment of anaphylactic cardiovascular collapse.
The four types of shock hypovolemic, cardiogenic, dis- Anaesth Intensive Care 1986;14:17-21.
tributive, and obstructive have unique causes that result
Guly HR, Bouamra O, Lecky FE, et al. The incidence of neuro-
in hemodynamic instability, namely hypotension.
genic shock in patients with isolated spinal cord injury in
Approaches to treatment differ for each type of shock; the emergency department. Resuscitation 2008;76:57-62.
prompt and appropriate treatment is key to minimizing
morbidity and mortality. Gupta AA, Leaker M, Andrew M, et al. Safety and outcomes
Hypertensive emergency is another form of hemodynamic of thrombolysis with tissue plasminogen activator for
instability that requires immediate treatment with an intra- treatment of intravascular thrombosis in children. J
venous antihypertensive agent. Pediatr 2001;139:682-8.

Heytman M, Rainbird A. Use of alphaagonists for manage-


ment of anaphylaxis occurring under anaesthesia: case
studies and review. Anaesthesia 2004;59:1210-5.
REFERENCES
Adams HA, Baumann G, Gnsslen A, et al. Definition of shock Hussain AM, Yousuf B, Khan MA, et al. Vasopressin for the
types. Anasthesiol Intensivmed Notfallmed Schmerzther management of catecholamine-resistant anaphylactic
2001;36:S140-3. shock. Singapore Med J 2008;49:E225-8.

American College of Surgeons (ACS); Committee on Trauma. Kill C, Wranze E, Wulf H. Successful treatment of severe
Shock: Advanced Trauma Life Support for Doctors, anaphylactic shock with vasopressin. Int Arch Allergy
Student Course Manual, 8th ed. Chicago: ACS, 2008. Immunol 2004;134:260-1.

Bailey JM, Miller BE, Lu W, et al. The pharmacokinetics Kirk R, Dipchand AI, Rosenthal DN. The International Society
of milrinone in pediatric patients after cardiac surgery. for Heart and Lung Transplantation Guidelines for the
Anesthesiology 1999;90:1012-8. management of pediatric heart failure. J Heart Lung
Transplant 2014;33:888-909.
Biss TT, Brendao LR, Kahr KW, et al. Clinical features
and outcome of pulmonary embolism in children. Br J Kirkpatrick AW, Ball CG, DAmours SK, et al. Acute resus-
Haematol 2008;142:808-18. citation of the unstable adult trauma patient: bedside
diagnosis and therapy. Can J Surg 2008;51:57-69.
Breslin K, Dewesh A. The use of methylprednisolone in
acute spinal cord injury: a review of the evidence, con- Kleinman ME, Chameides L, Schexnayder SM, et al. Pediatric
troversies, and recommendations. Pediatr Emerg Care advanced life support: 2010 American Heart Association
2012;28:1238-45. guidelines for cardiopulmonary resuscitation and emer-
gency cardiovascular care. Pediatrics 2010;126:e1361-e99.
Brown SG. The pathophysiology of shock in anaphylaxis.
Immunol Allergy Clin North Am 2007;27:165-75. Long JD, Fairbrother HE. Pediatric Updates: Pediatric
Cardiogenic Shock. December 5, 2015.
Brown SG, Blackman KE, Stenlake V, et al. Insect sting
anaphylaxis; prospective evaluation of treatment with McKiernan CA, Lieberman SA. Circulatory shock in children:
intravenous adrenaline and volume resuscitation. Emerg an overview. Pediatr Rev 2005;26:451-60.
Med J 2004;21:149-54. Mitrovic I. Cardiovascular disorders: vascular disease. In:
Chandar J, Zilleruelo G. Hypertensive crisis in children. McPhee SJ, Hammer GD, eds. Pathophysiology of Disease,
Pediatr Nephrol 2012;27:741-51. 6th ed. New York: McGraw-Hill, 2010:chap 11.

Chang AC, Atz AM, Wernovsky G, et al. Milrinone: systemic Monagle P, Chan AKC, Goldenberg NA, et al. Antithrombotic
and pulmonary hemodynamic effects in neonates after therapy in neonates and children: antithrombotic therapy
cardiac surgery. Crit Care Med 1995;23:1907-14. and prevention of thrombosis: American College of Chest
Physicians evidence-based clinical practice guidelines.
Cocchi MN, Kimlin E, Walsh M, et al. Identification and resus- Chest 2012;141:e737S-e801S.
citation of the trauma patient in shock. Emerg Med Clin
North Am 2007;25:623-42. Mtaweh H, Trakas EV, Su E, et al. Advances in monitor-
ing and management of shock. Pediatr Clin North Am
De Backer D, Biston P, Devriendt J, et al. Comparison of 2013;60:641-54.
dopamine and norepinephrine in the treatment of shock. N
Engl J Med 2010;362:779-89. National High Blood Pressure Education Program (NHBPEP)
Working Group on High Blood Pressure in Children and
DiPiro JT, Talbert RL, Yee GC, et al. Hypovolemic shock. In: Adolescents. The fourth report on the diagnosis, evalua-
Pharmacotherapy: A Pathophysiologic Approach, 8th ed. tion, and treatment of high blood pressure in children and
New York: McGraw-Hill, 2011:chap 31. adolescents. Pediatrics 2004;114(suppl 2):1-22.

PedSAP 2016 Book 2 Pediatric Critical Care 43 Hemodynamic Instability


Pasman E. Shock in Pediatrics. 2015. Subramaniam S, Rutman M. Cardiogenic shock. Pediatr Rev
2015;36:225-6.
Patel NH, Romero SK, Kaelber DC. Evaluation and man-
agement of pediatric hypertensive crises: hypertensive Summers RL, Baker SD, Sterling SA, et al. Characterization of
urgency and hypertensive emergencies. Open Access the spectrum of hemodynamic profiles in trauma patients
Emerg Med 2012;4:85-92. with acute neurogenic shock. J Crit Care 2013;28:531.e1-5.

Patocka C, Nemeth J. Pulmonary embolism in pediatrics. J Tobias JD. Shock in children: the first 60 minutes. Pediatr
Emerg Med 2012;42:105-16. Ann 1996;25:330-8.

Public Health Service (PHS), FDA. Important Drug Warning: VanderPluym C, Urschel S, Buchholz H. Advanced therapies
Safety Information Regarding the Use of Abbokinase for congenital heart disease: ventricular assist devices
(Urokinase). Rockville, MD: FDA, 1999. and heart transplantation. Can J Cardiol 2013;29:796-802.

Schummer W, Schummer C, Wipperman J. Anaphylactic Van Ommen CH, Peters M. Acute pulmonary embolism in
shock: is vasopressin the drug of choice? Anesthesiology childhood. Thromb Res 2006;118:13-25.
2004;101:1025-7.
Weil MH. Defining hemodynamic instability. In: Pinsky MR,
Smulders YM. Pathophysiology and treatment of haemody- Payen D, eds. Functional Hemodynamic Monitoring.
namic instability in acute pulmonary embolism: the pivotal Berlin: Springer-Verlag, 2005:9-17.
role of pulmonary vasoconstriction. Cardiovasc Res
2000;48:23-33.

Subramaniam K, Boisen M, Shah PR, et al. Mechanical circu-


latory support for cardiogenic shock. Best Pract Res Clin
Anaesthesiol 2012;26:131-46.

PedSAP 2016 Book 2 Pediatric Critical Care 44 Hemodynamic Instability


Self-Assessment Questions
Questions 2123 pertain to the following case. Questions 24 and 25 pertain to the following case.

E.B. is a 17-year-old female adolescent who has an atrial S.S. is an 8-month-old boy referred to the ED by his pedia-
septal defect after an Amplatzer device closure (6 months trician for further evaluation of his poor color and serum
ago). She was transferred to the ICU after having a cardiac sodium of 166 mEq/L. Three days earlier, his nasojejunal was
arrest at a friends house. The friend reports hearing E.B. dislodged and found to be nasogastric (NG). As a result, S.S.
breathing abnormally but thought she was snoring. The received NG feeds and had increased diarrhea. In addition,
friend later found E.B. unresponsive and called paramedics, S.S. did not receive any formula on the day of presentation to
who began CPR (cardiopulmonary resuscitation) and admin- the hospital. No recent history of fever, respiratory distress,
istered four shocks, four doses of epinephrine, and 2 g of upper respiratory infection symptoms, vomiting, or edema
magnesium sulfate for torsades de pointes. E.B. was also was noted. Vital signs for S.S. are Tmax 36.6C, heart rate 180
given one dose of naloxone. She had return of spontaneous beats/minute, blood pressure 54/30 mm Hg, and Spo2 85%.
circulation and was intubated and transported to Childrens P.G.s laboratory test results include Na 164 mEq/L, K 5.1
Hospital. On arrival at the hospital, a non-contrast head mEq/L, Cl 131 mEq/L, CO2 24 mEq/L, SCr 0.73 mg/dL (base-
CT was negative. The cooling protocol was initiated. Chest line 0.3 mg/dL), serum glucose 81 mg/dL, calcium 7.2 mg/
radiography revealed asymmetric opacities. Blood cul- dL, albumin 4.4, magnesium 2.3 mEq/L, WBC 17.6 103 cells/
tures were obtained, and empiric antibiotics were initiated. mm3, Hgb 14.8 g/dL, Hct 53.2%, and Plt 276,000/mm3.
Toxicology screening was negative. An electroencephalo-
24. Which one of the following best categorizes the type of
gram showed diffuse slowing. Echocardiography showed a
shock S.S. is experiencing?
large thrombus versus vegetation on the right atrial side of
the Amplatzer device as well as two small thrombi versus A. Hypovolemic
vegetations in the right ventricle. An ECG showed QTc 540 B. Cardiogenic
milliseconds. E.B. was not ill before the cardiac arrest and C. Obstructive
had made no recent complaints of chest pain or shortness D. Neurogenic
of breath. Her vital signs on admission were blood pressure 25. Which one of the following is best to recommend for S.S.?
70/45 mm Hg, heart rate 124 beats/minute, temperature
A. Reinitiate his feeds at 1.5 times his normal rate
36.7C, respiratory rate 18 breaths/minute, and peripheral
B. Fluid resuscitation with 5% albumin
capillary oxygen saturation (Sp O 2) 90%.
C. Fluid resuscitation with 0.9% normal saline
21. Which one of the following is the most likely cause of D. Initiate dopamine
E.B.s cardiac arrest?
A. Sepsis with progression to septic shock Questions 26 and 27 pertain to the following case.

B. Acute pulmonary embolism with progression to J.C., a 12-year-old boy, sustains a head and neck injury after
obstructive shock diving into the shallow end of a swimming pool. He is taken by
C. Heart failure with progression to cardiogenic shock emergency services to the nearest hospital.
D. Arrhythmia with progression to cardiogenic shock 26. Which one of the following forms of shock should the ED
22. On presentation to the ICU, which one of the following is staff be most concerned about in J.C.?
best to immediately initiate to treat E.B.s hemodynamic A. Neurogenic
instability? B. Hypovolemic
A. Alteplase C. Cardiogenic
B. Milrinone D. Obstructive
C. Amiodarone infusion 27. On presentation, J.C.s vital signs are as follows: heart
D. Norepinephrine rate 50 beats/minute, blood pressure 70/40 mm Hg,
23. Which one of the following is most important to monitor respiratory rate 25 breaths/minute, and Sao2 95%. Which
in E.B.? one of the following is best to avoid in J.C.?

A. Urine output and renal function A. Fluid resuscitation


B. Heart rate and rhythm B. Phenylephrine
C. Lactates C. Norepinephrine
D. Cerebral perfusion pressure D. Milrinone

PedSAP 2016 Book 2 Pediatric Critical Care 45 Hemodynamic Instability


Questions 2830 pertain to the following case. minute. On physical examination, Q.T. is peripherally vaso-
P.J. is a 9-year-old boy who is recovering from aortic sur- dilated, and wheezes can be heard on lung examination.
gery. His blood pressure readings have consistently been Which one of the following is best to recommend for Q.T.?
greater than 180/90 mm Hg. P.J. is no longer responding to
A. Dopamine
commands.
B. Phenylephrine
28. Which one of the following best categorizes P.J.s blood C. Intravenous fluids
pressure? D. Atropine
A. Stage 1 hypertension
B. Stage 2 hypertension Questions 3436 pertain to the following case.

C. Hypertensive urgency R.L., a 3-month-old girl, presents to the ED with projectile


D. Hypertensive emergency vomiting. A workup for pyloric stenosis is in progress when
tachycardia is noted. Two doses of adenosine are given
29. Which one of the following is the best antihypertensive
without a response. An ECG and echocardiography reveal
agent to recommend for P.J.?
fascicular ventricular tachycardia and a dilated left ventri-
A. Isradipine cle with severely depressed left ventricular function. A dose
B. Esmolol of intravenous verapamil is administered with conversion to
C. Nicardipine sinus tachycardia. R.L.s vital signs immediately afterward
D. Fenoldopam are blood pressure 80/54 mm Hg, heart rate 207 beats/min-
30. Which one of the following best describes the goal of ute, temperature 36.5C, respiratory rate 52 breaths/minute,
therapy for P.J.? and Sao2 100%.

A. Immediate blood pressure reduction to the normal 34. Which one of the following is best to initiate as a continu-
range ous infusion for R.L.?
B. A 25% blood pressure reduction within 8 hours after A. Milrinone
therapy initiation B. Enalaprilat
C. Stabilization of blood pressure at the current level C. Dopamine
D. Improvement in neurologic status D. Epinephrine

Questions 3133 pertain to the following case.


35. Despite treating the arrhythmia and the echocardio-
graphic findings, R.L. continues to decompensate with
Q.T. is a 4-year-old girl who presents to the ED after consum-
worsening heart function and more frequent runs of
ing a peanut butter and jelly sandwich at her day care center.
fascicular ventricular tachycardia. Which one of the fol-
She has a known peanut allergy and has already received
lowing best justifies placement of a ventricular assist
two doses of intramuscular epinephrine. She is experiencing
device for R.L.?
respiratory distress and gasping for air; her lips are swollen,
and her body is covered with a rash. On admission, her vital A. Bridge to recovery
signs are Tmax 37.2C, heart rate 160 beats/minute, blood B. Destination therapy
pressure 55/45 mm Hg, and Spo2 80%. C. Bridge to transplantation
D. Bridge to determining the optimal treatment regimen
31. Which one of the following best describes the type of
for arrhythmia
shock Q.T. is experiencing?
36. Which one of the following poses the greatest barrier for
A. Cardiogenic
dosing warfarin in R.L.?
B. Anaphylactic
C. Obstructive A. Maturation of the CYP enzymes
D. Neurogenic B. Risk of bleeding
C. Formula diet
32. Which one of the following is best to recommend for
D. Lack of oral liquid formulation
immediate initiation in Q.T.?
A. Continuous infusion epinephrine Questions 37 and 38 pertain to the following case.
B. Dopamine
C.W. is a 15-year-old girl (weight 90 kg) who presents to the
C. Intramuscular epinephrine
pediatricians office with a 3-day history of headache and not
D. Norepinephrine
feeling herself. Her vital signs on presentation are tempera-
33. After receiving the recommended agent for 15 minutes, ture 37.8C, blood pressure 145/90 mm Hg, respiratory rate 20
Q.T.s vital signs are heart rate 180 beats/minute, blood breaths/minute, and heart rate 85 beats/minute. C.W.s medi-
pressure 70/50 mm Hg, and respiratory rate 60 breaths/ cal history is significant for obesity.

PedSAP 2016 Book 2 Pediatric Critical Care 46 Hemodynamic Instability


37. Which one of the following is the most likely cause of beats/minute, respiratory rate 25 breaths/minute, and Sao2
C.W.s headache? 98%. Laboratory test results are significant for the following:
Na 153 mEq/L, Cl 116 mEq/L, CO2 18 mEq/L, SCr 1.5 mg/dL
A. Tension migraine
(baseline 0.4 mg/dL), WBC 19 x 103 cells/mm3, Hct 45.2%, and
B. Elevated blood pressure
Plt 150,000/mm3.
C. Obesity
D. Viral infection 39. Which one of the following best describes the type of
shock T.C. is experiencing?
38. Which one of the following is best to recommend in the
pediatricians office to treat C.W.s headache? A. Hypovolemic
B. Cardiogenic
A. Isradipine
C. Distributive
B. Ibuprofen
E. Obstructive
C. Nicardipine
D. Diet and exercise 40. Which one of the following is best to recommend for T.C.?
A. Dopamine
Questions 39 and 40 pertain to the following case. B. Norepinephrine
T.C. is a 4-year-old boy with a medical history significant for C. Fluid resuscitation with a crystalloid solution
kidney transplantation. He presents to the ED and reports a D. Fluid resuscitation with a colloid solution
5-day history of bloody diarrhea. His vital signs are temper-
ature 39.1C, blood pressure 80/50 mm Hg, heart rate 150

PedSAP 2016 Book 2 Pediatric Critical Care 47 Hemodynamic Instability


Learner Chapter Evaluation: Hemodynamic Instability.

As you take the posttest for this chapter, also evaluate the Use the 5-point scale to indicate whether this chapter pre-
materials quality and usefulness, as well as the achievement pared you to accomplish the following learning objectives:
of learning objectives. Rate each item using this 5-point scale: 31. D
 istinguish between the four types of shock using physi-
cal examination findings and laboratory measurements.
Strongly agree 32. C
 lassify the types of fluid used in resuscitating the patient
Agree with hypovolemic shock.
Neutral 33. J
 ustify the use of an inotrope or vasopressor in the treat-
Disagree ment of circulatory shock.
Strongly disagree 34. A
 ccount for the mechanisms by which circulatory shock
occurs.
20. The content of the chapter met my educational needs. 35. D
 istinguish between hypertensive urgency and hyperten-
21. The content of the chapter satisfied my expectations. sive emergency.
22. The author presented the chapter content effectively. 36. P
 lease provide any specific comments related to any
perceptions of bias, promotion, or advertisement of com-
23. T
 he content of the chapter was relevant to my practice
mercial products.
and presented at the appropriate depth and scope.
37. P
 lease expand on any of your above responses, and/or
24. The content of the chapter was objective and balanced.
provide any additional comments regarding this chapter:
25. T
 he content of the chapter is free of bias, promotion, or
advertisement of commercial products.
Questions 3840 apply to the entire learning module.
26. The content of the chapter was useful to me.
38. How long did it take you to read the instructional materi-
27. T
 he teaching and learning methods used in the chapter
als in this module?
were effective.
39. How long did it take you to read and answer the assess-
28. T
 he active learning methods used in the chapter were
ment questions in this module?
effective.
40. P
 lease provide any additional comments you may have
29. T
 he learning assessment activities used in the chapter
regarding this module:
were effective.
30. The chapter was effective overall.

PedSAP 2016 Book 2 Pediatric Critical Care 48 Hemodynamic Instability


Pediatric Critical Care II
Pediatric Critical Care II Panel

Series Editors: Joshua Caballero, Pharm.D., BCPP


Marcia L. Buck, Pharm.D., BCPPS, FCCP, FPPAG Associate Professor
Department of Pharmacy Practice
Clinical Coordinator
Nova Southeastern University, College of Pharmacy
University of Virginia Childrens Hospital
Fort Lauderdale, Florida
Professor
Department of Pediatrics Allison Lardieri, Pharm.D., BCPPS
University of Virginia School of Medicine Assistant Professor
Clinical Professor Department of Pharmacy Practice and Science
Virginia Commonwealth University School of Pharmacy University of Maryland School of Pharmacy
Department of Pharmacy Services Baltimore, Maryland
University of Virginia Health System
Charlottesville, Virginia
Kalen B. Manasco, Pharm.D., BCPS
Trauma
Clinical Associate Professor Author
Department of Clinical and Administrative Pharmacy
Joseph M. LaRochelle, Pharm. D., BCPPS
University of Georgia College of Pharmacy
Augusta, Georgia Clinical Associate Professor
College of Pharmacy
Faculty Panel Chair: Xavier University of Louisiana
Clinical Associate Professor of Pediatrics
Joseph M. LaRochelle, Pharm. D., BCPPS
Louisiana State University Health Sciences
Clinical Associate Professor Center School of Medicine
College of Pharmacy New Orleans, Louisiana
Xavier University of Louisiana
Clinical Associate Professor of Pediatrics Reviewers
Louisiana State University Health Sciences
Sara P. Rooney, Pharm.D., BCPS, BCPPS
Center School of Medicine
New Orleans, Louisiana Pediatric Intensive Care (PICU) Clinical Pharmacy Specialist
Department of Pharmacy
Childrens National Health System
Extracorporeal Circuits in Children Washington, District of Columbia
Andrea Joan Stumpe, Pharm.D., BCPS, BCPPS
Author
Clinical Pharmacist
Elizabeth J. Beckman, Pharm.D., BCPS, BCPPS, BCCCP Pharmacy Department
Clinical Pharmacy Specialist, Pediatric Critical Care Methodist Germantown Hospital
Department of Pharmacy Germantown, Tennessee
Riley Hospital for Children at Indiana University Health
Indianapolis, Indiana The American College of Clinical Pharmacy and the authors
thank the following individuals for their careful review of the
Reviewers Pediatric Critical Care II chapters:
Aaron A. Harthan, Pharm.D., BCPPS
Nicola G. Dahl, Pharm.D.
PGY-1 Residency Program Coordinator
Pediatric Critical Care Pharmacist Medical Writer
Department of Pharmacy Kanab, Utah
OSF Saint Francis Medical Center/
Childrens Hospital of Illinois
Peoria, Illinois
DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST
Consultancies: Elizabeth J. Beckman (Lexi-Comp/Wolters/Kluwer Health); Regine L Caruthers (Lexi-Comp/Wolters Kluwer
Health); Joseph M. LaRochelle (ACCP); Lois F. Parker (ASHP/MSHP)

Stock Ownership:

Royalties:

Grants: Elizabeth J. Beckman (ASHP Foundation); Christina Cox (University of South Carolina);

Honoraria: Christina Cox (ACCP, Palmetto Health Richland Childrens Hospital); Jennifer L. Morris (ACCP, Society of Critical Care
Medicine, Pediatric Pharmacy Advocacy Group, Wolters Kluwer)

Other:

Nothing to disclose: Laura Lo Castro Bio, Joshua Caballero, Brooke Clark, Kyle Davis, Ifeyinwa D. Eboh, Elizabeth S. Goswami,
Aaron A. Harthan, Maha O. Kebir, Allison Lardieri, Michael A. Maccia, Kristine Parbuoni, Monica A. Puebla, Sara P. Rooney, Emily
L. Rowe, Ashley R. Schappell, Julie Pingel, Brandi L. Strauser, Andrea Joan Stumpe, Susan Warrington

ROLE OF BPS: The Board of Pharmacy Specialties (BPS) is an autonomous division of the American Pharmacists Association
(APhA). BPS is totally separate and distinct from ACCP. The Board, through its specialty councils, is responsible for specialty
examination content, administration, scoring, and all other aspects of its certification programs. PedSAP has been approved by
BPS for use in BCPPS recertification. Information about the BPS recertification process is available online.

Other questions regarding recertification should be directed to:

Board of Pharmacy Specialties


2215 Constitution Avenue NW
Washington, DC 20037
(202) 429-7591
CONTINUING PHARMACY EDUCATION AND
RECERTIFICATION INSTRUCTIONS
Continuing Pharmacy Education Credit: The American College of Clinical Pharmacy is accredited by the Accreditation
Council for Pharmacy Education (ACPE) as a provider of continuing pharmacy education (CPE).

PedSAP: Target Audience: The target audience for PedSAP 2016 Book 2(Pediatric Critical Care) is pediatric pharmacotherapy spe-
cialists and advanced-level clinical pharmacists caring for pediatric patients with several acute conditions and complications of
the pulmonary, cardiovascular, endocrine, and neurologic systems.

Available CPE credits: Purchasers who successfully complete all posttests for PedSAP 2016 Book 2 (Pediatric Critical Care)
can earn 13.0 contact hours of continuing pharmacy education credit. The universal activity numbers are as follows: Pediatric
Critical Care I 0217-0000-16-021-H01-P, 3.5 contact hours; Pediatric Critical Care II 0217-0000-16-022-H01-P, 4.5 contact
hours; and Clinical and Practice Updates 0217-0000-16-170-H01-P, 5.0 contact hours. You may complete one or all available mod-
ules for credit. Tests may not be submitted more than once.

BCPPS test deadline: 11:59 p.m. (Central) on January 17, 2017.


ACPE test deadline: 11:59 p.m. (Central) on September 14, 2019.

Posttest access: Go to www.accp.com and sign in with your e-mail address and password. Technical support is available from
8 a.m. to 5 p.m. (Central) weekdays by calling (913) 492-3311. PedSAP products are listed under My Online Products on your My
Account page.

BCPPS Recertification Credit: To receive BCPPS recertification CPE credit, a PedSAP posttest must be submitted within the
4-month period after the books release. The first page of each print and online book lists the deadline to submit a required
posttest for BCPPS recertification credit. Only completed tests are eligible for credit; no partial or incomplete tests will be pro-
cessed. Tests may not be submitted more than once. The passing point for BCPPS recertification is based on an expert analysis
of the items in each posttest module.

ACPE CPE Credit: To receive ACPE CPE credit for a PedSAP module, a posttest must be submitted within the 3-year period after
the books release. The appropriate CPE credit will be awarded for test scores of 50% and greater.

Credit Assignment and Reporting: All required posttests that meet the 50% score standard will be immediately awarded the
appropriate ACPE CPE credit. Earned credits will be transmitted within 24 hours to www.mycpemonitor.net and should appear on
statements of credit within 3 business days.

Required posttests that are submitted before the BCPPS test deadline and that meet the passing point set by statistical analysis
will earn BCPPS recertification credits. These credits will be posted within 30 days after the BCPPS test deadline. For statements
of CPE credit, visit www.mycpemonitor.net.

All BCPPS recertification credits are forwarded by ACCP to the Board of Pharmacy Specialties (BPS). Questions regarding the
number of hours required for BCPPS recertification should be directed to BPS at (202) 429-7591 or www.bpsweb.org. The ACCP
Recertification Dashboard is a free online tool that can track recertification credits as they are earned through ACCP and schedule
new opportunities for credits from upcoming ACCP professional development programs.

Posttest Answers: The explained answers with rationale and supporting references will be posted 1 week after the BCPPS
test deadline and will be available to anyone who has either submitted a posttest or waived his or her right to receive credit (see
below) from a posttest. Go to www.accp.com and sign in with your e-mail address and password. Click the PedSAP book on your
My Account page and you will see a link to the explained answers.

Test Waivers: To access the explained answers without submitting a posttest, sign in to your My Account page, select the
PedSAP book, and click on the waiver link for that module. By completing the waiver form for a module, you waive the opportu-
nity to receive CPE credit for that module. After you submit a waiver, you will see a link to the PDF file that contains the answers
for the module you waived. Answers will be available starting 1 week after the BCPPS test deadline.

ACSAP 2016 Book 2 Pediatric Critical Care 54 Program Instructions


Extracorporeal Circuits in Children
By Elizabeth J. Beckman, Pharm.D., BCPS, BCPPS, BCCCP

Reviewed by Aaron A. Harthan, Pharm.D., BCPPS; Joshua Caballero, Pharm.D., BCPP; and Allison Lardieri, Pharm.D., BCPPS

LEARNING OBJECTIVES

1. Account for the basic mechanics and components of extracorporeal therapies.


2. Develop a drug therapy regimen based on pharmacokinetic changes in extracorporeal therapies.
3. Design a therapeutic monitoring plan for patients receiving extracorporeal therapies.
4. Evaluate an anticoagulation strategy for patients receiving extracorporeal therapies.
5. Assess the attainment of therapeutic end points for patients receiving extracorporeal therapies.
6. Design a plan to treat common complications associated with extracorporeal therapies.

INTRODUCTION
ABBREVIATIONS IN THIS CHAPTER
Critically ill patients have pharmacokinetic and pharmacodynamic
ACT Activated clotting time
alterations that necessitate changes in medication regimens. These
AT Antithrombin
alterations include variations in medication absorption, increased
CRRT Continuous renal replacement
therapy volumes of distribution from fluid resuscitation or capillary leak, and
ECMO Extracorporeal membrane a decline in renal and hepatic function attributable to compromise of
oxygenation end-organ perfusion in shock states. Critically ill pediatric patients
FRF Filter replacement fluid require further pharmacotherapy modifications as they mature.
PMP Polymethylpentene Various extracorporeal technologies introduce an additional layer of
PVC Polyvinylchloride complexity to support critically ill children. There is often a lack of
SPAD Single-pass albumin dialysis or delay in pharmacokinetic studies using these technologies, and
TAMOF Thrombocytopenia-associated even less evidence with extracorporeal therapies in children. These
multiple organ failure supportive technologies are being modified at a rapid pace, antiquat-
TPE Therapeutic plasmapheresis ing the few pharmacokinetic data available and their application.
VA Veno-arterial The clinician must have an understanding of the pharmacokinetic
VV Veno-venous and pharmacodynamic changes in critically ill children on these sup-
portive modalities in order to provide safe, effective, and optimal
Table of other common abbreviations. medication management.
This chapter presents the basic mechanics and components
of extracorporeal membrane oxygenation (ECMO), continuous
renal replacement therapy (CRRT), and therapeutic plasmaphere-
sis (TPE) and discusses the pharmacokinetic changes that occur
with the introduction of this technology in children. In addition, liver
assist devices will be reviewed, although current clinical application
in pediatrics is absent because of the paucity of data surrounding
this novel therapy.

PedSAP 2016 Book 2 Pediatric Critical Care 55 Extracorporeal Circuits in Children


EXTRACORPOREAL MEMBRANE ECMO. One application of ECMO that is gaining traction is
OXYGENATION extracorporeal cardiopulmonary resuscitation for patients
Prevalence with cardiopulmonary arrest without return of spontane-
ous circulation for greater than 20 minutes. In addition, the
Extracorporeal membrane oxygenation is a life-sustaining
Institute of Medicine recommended in 2006 that ECMO be
modality that provides gas exchange and cardiac output
used to increase the number of viable organs for donation
in patients with cardiorespiratory failure. This device origi-
after cardiac death (Annich 2012).
nated in the 1930s with the work of Dr. John H. Gibbon, who
The continued expansion of technology and familiarity
used it in response to a patient who died of a pulmonary
with ECMO will decrease the number of relative contrain-
embolus. The modern-day ECMO circuit was pioneered by
dications to therapy. Absolute contraindications generally
adult surgeon Dr. Robert Bartlett, who was the first to use
consist of irreversible processes that evoke medical futility
ECMO support for neonates and children in 1972. Since
(e.g., lethal chromosomal abnormalities, severe neurologic
then, this technologys reach has continued to expand both
compromise, incurable malignancy). Other relative con-
domestically and internationally. As of January 2013, the
traindications (e.g., recent intracranial hemorrhage) and
Extracorporeal Life Support Organization registry listed
high-risk patients (e.g., severe multiorgan failure, coagulop-
53,000 ECMO cases with a hospital discharge survival rate of
athy) have been identified in the literature; the use of ECMO
61%. Almost 45,000 of these ECMO cases involved neonates
in these patients is often institution-dependent. Although
and children (Stang 2015).
the concern for medical futility will prohibit the use of ECMO,
exclusions will wane as the scope of extracorporeal support
Indications
expands.
Patients who receive ECMO often have a reversible cause or
a specific therapeutic destination that will alleviate the need
Circuit Characteristics
for further support. Acute respiratory failure (e.g., meconium
The ECMO circuit comprises a membrane oxygenator (arti-
aspiration syndrome, acute respiratory distress syndrome),
ficial lung), blood pump (artificial heart), heat exchanger,
acute cardiac failure (e.g., cardiomyopathy, inability to wean
and bladder/reservoir, all connected by a network of tubing
from cardiopulmonary bypass in the operating room), and
to the patient. Most of the circuit tubing consists of poly-
bridge to other destination therapies (i.e., ventricular-assist
vinylchloride (PVC) mixed with a plasticizer, di(2-ethylhexyl)
devices or solid organ transplantation) are common uses of
phthalate (DEHP), to provide flexibility. Leaching of DEHP
has been found in ECMO circuits but has been decreased
with biocompatible polymer-coated circuit elements
BASIC KNOWLEDGE STATEMENTS (Burkhart 2007). This system contains several access
points for blood product and drug infusions, as well many
Readers of the chapter are presumed to be familiar safety sensors that detect blood flow rates, pressure, and
with the following:
air in the system (Figure 1-1).
Pharmacokinetic and pharmacodynamic changes
in children
Vascular Access Points
Normal vital signs, weight and height for age, and
Extracorporeal membrane oxygenation support can be deliv-
laboratory values for children
ered using veno-arterial (VA) or veno-venous (VV) access.
Coagulation cascade and fibrinolytic system
Full cardiorespiratory support is provided by VA-ECMO,
constituents
whereas VV-ECMO provides respiratory support and allows
Table of common pediatric laboratory reference values. native cardiac output to participate. Veno-arterial access can
be transthoracic or extrathoracic. Transthoracic VA-ECMO
ADDITIONAL READINGS involves venous cannulation of the right atrium and arterial
cannulation of the ascending aorta. This type of cannula-
The following free resources have additional back- tion is used most commonly in postoperative cardiovascular
ground information on this topic:
surgery patients with sternotomy. Extrathoracic VA-ECMO
Extracorporeal Life Support Organization. ELSO involves venous cannulation through the femoral or internal
Guidelines [homepage on the Internet]
jugular vein and arterial cannulation through the femoral or
Kidney Disease Improving Global Outcomes. KDIGO
carotid artery. Veno-venous cannulation uses the internal jug-
Guidelines for Acute Kidney Injury. [homepage on
ular, femoral, or saphenous veins (Annich 2012).
the Internet]
University of Louisville Kidney Disease Program.
Circuit Priming
Drug Prescribing in Renal Failure. [homepage on
the Internet] The volume needed to prime the ECMO circuit is significant;
the neonatal circuits may need up 400500 mL to prime

PedSAP 2016 Book 2 Pediatric Critical Care 56 Extracorporeal Circuits in Children


Oxygenated Patient Blood Drainage
Blood Return (Venous)
(Arterial or Venous)

Heat Venous
Exchanger Saturation
Bridge Sensor
Pressure
Monitor

Membrane Bladder/
Oxygenator Reservoir
Blood Pump

Pressure
Heparin Venous Monitor
Infusion Blood
Port Sample
Port

Figure 1-1. Diagram of an extracorporeal membrane oxygenation (ECMO) circuit.

the tubing, membrane oxygenator, and blood pump. The the plastic surfaces (Annich 2012). The recommended
native circulating blood volume in neonates and infants is priming solution is institution-specific and depends on the
7090mL/kg. Therefore, adding an ECMO circuit can dou- type and size of circuit used. Neonates and smaller chil-
ble or triple the total circulating volume in children weighing dren require a blood prime before ECMO initiation because
less than 5 kg and increase the intravascular volume of of the potential for hemodilution with the wet (bloodless)
distribution. However, the ratio of ECMO circuit volume to prime solution.
patient native blood volume generally decreases as patient
size increases. Membrane Oxygenator
The ECMO circuit can be primed on demand or wet pre- The membrane oxygenator functions as an extempo-
primed and stored until needed. Wet pre-primed circuits are raneous lung by adding oxygen and removing CO 2 from
typically primed first with CO 2 gas to displace the bubble- blood through diffusive gas exchange. Oxygen is blown
inducing nitrogen, followed by normal saline to displace into the device and across the membrane to create a dif-
CO 2 (Annich 2012). These sterile, wet-primed circuits can fusion gradient, in which CO 2 is passively removed across
be stored for up to 30 days (Walczak 2005). In general, all the membrane. This oxygen gas delivery is termed sweep
ECMO circuits are initially primed with a balanced elec- and is titrated to maintain the desired Pco2. To increase CO2
trolyte, isotonic solution that mimics extracellular fluid clearance, the gas flow rate or sweep is increased, whereas
composition and pH. Additional components may be added oxygen delivery is optimized by altering the blood flow rate
to the circuit before ECMO initiation. For example, packed through the membrane oxygenator and hemoglobin concen-
red blood cells (PRBCs) may be added to create a circuit tration (Annich 2012).
hematocrit of 35%40%. Priming the circuit with blood The ideal membrane is made of hydrophobic polymers
products requires adding heparin for anticoagulation and to prevent plasma leak and prolong the life of the mem-
calcium because of the citrated blood product. Albumin brane. Initial membrane oxygenators were made of silicone
can be added to create an oncotic load as well as to coat rubber, which afforded adequate gas exchange but had

PedSAP 2016 Book 2 Pediatric Critical Care 57 Extracorporeal Circuits in Children


several disadvantages. These membrane oxygenators had initiated on cannulation and continued throughout ECMO.
low mechanical strength and developed holes or cracks with Unfractionated heparin is the standard of practice in most
increased pressure or force. In addition, silicone rubber mem- ECMO centers because of its short half-life, nonrenal elim-
brane oxygenators had a large surface area, requiring large ination, reversibility with protamine, and established use
priming volumes that caused an inflammatory response from in cardiopulmonary bypass (Annich 2015). Heparin binds
blood contact on the large surface area. Modern membrane with antithrombin (AT) to form a complex that potenti-
oxygenators are composed of polymethylpentene (PMP) hol- ates the inhibition of activated factors IX, X, XI, XII, and II.
low fibers coated with antithrombotic and anti-inflammatory Antithrombin is produced in the liver and inhibits factor
surfaces, which contribute to less plasma leak and improved Xa and thrombin. Deficiency in AT decreases unfraction-
durability and flexibility (Stang 2015). ated heparin sensitivity, necessitating an escalation of
unfractionated heparin doses to meet therapeutic end
Blood Pump points. Infants younger than 1 year have decreased AT pro-
The goal of the blood pump is to facilitate the movement of duction, which is likely the cause of the relative heparin
blood throughout the extracorporeal system and the patient. resistance reported (ELSO 2014). Antithrombin is found in
The circuit blood flow delivers oxygenated blood to the fresh frozen plasma (1 unit/mL) but is usually insufficient
patient and, depending on the cannulation access, may pro- to significantly change the AT activity level at standard
vide full cardiac output. doses. Antithrombin concentrate products are available,
Two types of blood pumps are available: roller and cen- but expensive.
trifugal pumps. Classically, the roller pump has been the Exogenous AT supplementation has gained momentum for
primary pump used to propel blood through the ECMO cir- the treatment of low AT activity in patients receiving ECMO.
cuit. The roller ball is housed inside a curved raceway, which One retrospective, multicenter study of children younger
passes over and compresses the blood tubing. This causes than 18 years described a 5-fold increase in off-label AT use
an occlusion, pushes blood forward, and creates negative (i.e., from 5% to 50%) over a 10-year period (Wong 2013).
pressure behind the occlusion. Most roller pump systems Despite increasing use in pediatric ECMO, the results of AT
use synchronized double rollers so that one roller is always supplementation on clinical outcomes are mixed. One study
compressing the tubing, creating relatively nonpulsatile reported the AT supplementation experiences in 28 pediatric
flow. The roller pump is a reusable pumping mechanism patients receiving ECMO. This cohort had a mean age of 0.3
that can provide lower blood flow rates than centrifugal years and was a mixed group of primary cardiac failure and
pumps. The main disadvantage is the potential for tubing primary respiratory failure. A statistically significant increase
fracture from the repeated compression, which can allow in AT activity was noted at hours 8 and 24 after AT concen-
air into the circuit. trate supplementation (baseline: 61.5 units/mL; hour 8, 96.8
The centrifugal pump is a newer alternative to the roller units/mL; hour 24, 92 units/mL). However, there was no dif-
pump and uses a spinning motor to create the rotation of the ference in the unfractionated heparin rate, activated clotting
pump with subsequent propulsion of blood. Like the roller time (ACT), chest tube output, or total volume of PRBC trans-
pump, it generates negative pressure entering the pump fusions (Niebler 2011).
and positive pressure exiting the pump. Historically, cen- Another center retrospectively investigated AT use in 64
trifugal pumps have been avoided because of concern for children receiving ECMO. The AT supplementation group had
a higher risk of hemolysis. Recent literature suggests that a mean AT activity level of 80%, but only 8% achieved the ther-
newer centrifugal pumps have similar rates of hemolysis apeutic target activity of 120%. The AT cohort did show an
while decreasing the risk of air emboli compared with roller unfractionated heparin infusion rate drop by 10 units/kg/hour
pumps (Meyer 2012). However, a major downside of centrif- for 12 hours after AT administration. However, both groups
ugal pumps is the cost because they are not reusable. Both had similar clot burden in the ECMO circuit and no identified
pumps create nonpulsatile flow in the ECMO circuit. This is an in vivo thrombosis. In addition, there was no difference in the
important consideration for end-organ perfusion as well as number of circuit changes, bleeding events, transfusions, or
for the effects on drug absorption, metabolism, and elimina- ICU or hospital length of stay (Wong 2015).
tion in the patient who lacks adequate native cardiac output, In a similarly designed retrospective study of 40 pediat-
as seen in VA-ECMO (Annich 2012). ric ECMO cases, no change in unfractionated heparin rate
with AT supplementation occurred, but the study detected
Anticoagulation a decrease in unfractionated heparin anti-factor Xa (anti-
The interaction of whole blood with the foreign surfaces Xa) concentrations in the supplemented group (0.19 unit/
of the ECMO circuit stimulates an acute inflammatory mL vs. 0.25 unit/mL; p<0.05). This center also reported a
response, activation of platelets, and activation of the clot- greater frequency of ECMO circuit changes in the AT sup-
ting cascade with clot formation in the circuit and patient. plementation cohort (HR 3.15; 95% CI, 1.218.16; p<0.05),
To prevent clot formation, anticoagulation therapy is but the authors attributed the diminished circuit life to the

PedSAP 2016 Book 2 Pediatric Critical Care 58 Extracorporeal Circuits in Children


subtherapeutic unfractionated heparin anti-Xa concentra- had statistically significant less mean blood loss (16 mL/
tions (Byrnes 2014). kg/day vs. 51 mL/kg/day) and fewer platelet transfusions
Instead of using a previously published bolus AT strat- (3 mL/kg/day vs. 33 mL/kg/day) and plasma transfusions
egy, one group of investigators reported their experience (5.9 mL/kg/day vs. 12 mL/kg/day). Patients in the bivaliru-
with using continuous infusion AT. This observational study din arm had a nonsignificant decline in PRBC transfusions
compared AT dosing strategies in 11 postoperative, pediat- (15 mL/kg/day vs. 25 mL/kg/day; p=0.067). In addition, the
ric cardiac patients receiving ECMO. Five patients received cost of anticoagulation (e.g., drug, blood product use, and
intermittent boluses of AT to maintain an activity level greater AT) favored bivalirudin, with an almost 50% cost reduc-
than 60%, and six patients received continuous infusion AT tion in both the adult and pediatric cohorts (Ranucci 2011).
to achieve an AT activity level above 100%. There were more Although this study had a small sample size and a hetero-
significant bleeding events in the AT bolus group than in the geneous age group and lacked generalizability, the results
continuous infusion group (3 of 5 vs. 0 of 6) (Agati 2006). are intriguing.
Controversy still exists regarding the role of AT supplemen- Argatroban has not been well studied in pediatric ECMO.
tation in pediatric ECMO as the optimal target for AT activity, Case reports and small case series report relative success
and the ideal supplementation strategy to provide positive with this agent in children receiving ECMO with confirmed or
clinical outcomes has not been described. suspected heparin-induced thrombocytopenia. The argatro-
The ECMO experience with alternative parenteral antico- ban dosing reported in these instances is wide and variable
agulants is not as well established as with unfractionated (0.124 mcg/kg/minute) (Hursting 2006). A 2011 open-label
heparin, but there are some theoretical advantages of using pediatric pharmacokinetic study reported a starting dose of
direct thrombin inhibitors such as bivalirudin or argatroban. 0.75 mcg/kg/minute in children younger than 16 years, with
These agents, without the need for AT, bind directly to both a hepatic failure adjustment to 0.2 mcg/kg/minute (Young
free thrombin and clot-bound thrombin to inhibit further 2011). Adult literature suggests that doses even lower than
clot formation. Unlike unfractionated heparin, these agents FDA-approved starting doses are necessary to avoid over-
are unaffected by fluctuations in other plasma proteins and anticoagulation in critically ill patients with multiorgan
blood cells (ELSO 2014). dysfunction (Smythe 2009; Beiderlinden 2007).
A comparative unfractionated heparin and bivaliru- Much is still unknown about the safety, efficacy, and ben-
din study retrospectively investigated adult and pediatric efits of using direct thrombin inhibitors for anticoagulation
patients cannulated in a transthoracic fashion after cardiac in pediatric ECMO. With the lack of a direct reversal antidote
surgery and subsequently placed on VA-ECMO. Data points and the high cost associated with these therapies, unfrac-
were captured during the first 4 days of ECMO, even though tionated heparin will likely remain the preferred anticoagulant
most had a duration beyond 96 hours (11 of 21 patients). in pediatric ECMO for the immediate future. Direct thrombin
The patients in the bivalirudin group were older (bivalirudin inhibitors will remain the drug of choice in heparin-induced
mean 36.5 19 years vs. heparin mean 13.9 29 years) but thrombocytopenia in ECMO (Table 1-1).

Table 1-1. Anticoagulation Options for ECMO

Starting
Drug Initial Bolus Infusion Monitoring Special Considerations

Unfractionated heparin 50100 units/kg 7.528.0 units/ ACT, aPTT, anti-Xa Platelet administration, increased
kg/hr urine output, and CRRT may
increase need; consider AT for
infusions > 35 units/kg/hr
Argatroban 100200 mcg/kg 0.20.75 mcg/ ACT, aPTT Consider lower starting dose in
kg/min critically ill patients; lower doses
needed with hepatic dysfunction
Bivalirudin 0.040.14 mg/kg 0.050.3 mg/ ACT, aPTT Metabolized by blood proteases;
kg/hr lower doses needed with renal
dysfunction

ACT = activated clotting time; aPTT = activated PTT; AT = antithrombin; CRRT = continuous renal replacement therapy; ECMO =
extracorporeal membrane oxygenation.

PedSAP 2016 Book 2 Pediatric Critical Care 59 Extracorporeal Circuits in Children


Several therapeutic monitoring avenues for ECMO anti- has not accepted any one therapeutic monitoring strat-
coagulation are available. The ACT is a functional test of egy as the standard of practice, but rather, leaves this to
hemostasis that measures the time in seconds needed for the discretion of each ECMO program. No single monitor-
whole blood to clot on exposure to an activator of the intrin- ing strategy is error free; thus, a combination monitoring
sic pathway. This test can be performed at the bedside, with strategy should be considered and tailored to the individual
the results provided within minutes. In addition, ACT testing patient receiving ECMO.
remains relatively inexpensive, providing an attractive option
for monitoring the various ECMO anticoagulation therapies. Antimicrobial Prophylaxis
Target ACT values of 180220 seconds are typical among The infection rate in neonatal and pediatric ECMO is 10.1
ECMO centers. infections and 20.8 infections per 1000 ECMO-days, respec-
The anti-Xa concentration is a marker of anticoag- tively. Coagulase-negative Staphylococcus is the most
ulation effect on factor Xa, which limits this test to commonly identified bacterial organism (15.9%), followed by
monitoring unfractionated heparin. Anti-Xa concentrations Pseudomonas aeruginosa (10.5%) and Staphylococcus aureus
for unfractionated heparin require a specific unfractionated (9.4%). Candida infections were the most common fungal
heparincalibrated assay, which is not synonymous with infections reported (12.7%). Infection rates increased after 14
the anti-Xa assay used with therapeutic drug monitoring of days of ECMO (Bizzarro 2011). The most common infection
low-molecular-weight heparins. The unfractionated hepa- sites are the bloodstream, followed by surgical sites, urinary
rin anti-Xa assay is not influenced by other coagulopathies, tract, and respiratory tract (OHoro 2016).
hemodilution, or thrombocytopenia like ACTs. The down- The risk of infection with percutaneous ECMO cannulation
side to the anti-Xa assay is that it can be falsely lowered by is comparable with the risk associated with central intrave-
high plasma free hemoglobin, a marker of hemolysis that is nous catheter placement. After sterile intravenous catheter
often present in ECMO. Hyperbilirubinemia, hyperlipidemia, placement procedures, using central lineassociated blood-
and AT deficiency can also affect the assay results. Goal stream infection tool kits and care bundles is recommended.
anti-Xa concentrations are typically 0.30.7 unit/mL for If surgical cutdown for cannula placement is needed, pre-
unfractionated heparin. operative antimicrobial prophylaxis with a first-generation
Activated PTT (aPTT) is a plasma test that measures the cephalosporin to cover staphylococci and streptococci skin
time to fibrin formation. It represents the activity of coagu- flora is warranted. Patients who are cannulated by open ster-
lation factors XII, XI, X, IX, VIII, V, and II (prothrombin) and I notomy may benefit from skin flora prophylaxis to decrease
(fibrinogen). Because of alterations in the development of the the risk of mediastinitis (Annich 2012).
hemostatic system, infants often have a prolonged aPTT at At this time, the literature does not support routine anti-
baseline. Therefore, the utility of using aPTT exclusively for biotic prophylaxis as a means to reduce the transmission of
monitoring anticoagulation in infants receiving ECMO may be nosocomial infections during ECMO (OHoro 2016). Similarly,
less than ideal. data are lacking to support infectious surveillance by daily
Thromboelastography (TEG) and thromboelastometry blood and respiratory cultures in patients receiving ECMO
(ROTEM) point-of-care testing are other tools that can help (Annich 2012). Standards of care for infection control should
characterize a patients bleeding and thrombosis risk. These be used to prevent catheter-, ventilator-, and health care
tests can measure platelet aggregation, clot strength and associated infections.
stability, fibrin cross-linking time, and fibrinolysis time, all in
the presence of anticoagulants and procoagulants (Whiting Complications
2014). This global look at a patients hemostatic system can As previously discussed, a profound inflammatory response,
provide useful information, allowing the clinician to make hemolysis, thrombosis, and infections are all potential com-
patient-specific therapy and monitoring plans. The out- plications of ECMO. In addition, the systemic anticoagulation
put is a series of tracings and numeric values that must be necessary in ECMO may lead to bleeding events, such as
interpreted in order to comprehend the coagulation and fibri- surgical site, intracranial, GI, or mucous membrane bleed-
nolysis process, which can be difficult for the novice user. ing. Using subcutaneous and intramuscular administration
Another downside to this technology is that patient factors routes is not recommended for risk of injection-site bleeding
(e.g., age, sex) or technical factors (e.g., sample hemodilution, and hematoma.
blood collection site) have been reported to influence results Administration of blood products to maintain an insti-
(Ganter 2008; Luddington 2005). As clinicians become more tution-specific threshold concentration of hemoglobin,
familiar with this technology, TEG and ROTEM use will likely hematocrit, platelets, and fibrinogen is commonplace in
continue to expand outside its historical use within the oper- ECMO. The antifibrinolytic agents aminocaproic acid and
ating room. tranexamic acid have been used to manage surgical site
Anticoagulation monitoring in ECMO is complex and ever- bleeding and oozing in ECMO (ELSO 2014). These agents
changing. The Extracorporeal Life Support Organization work by inhibiting fibrin degradation through activity on

PedSAP 2016 Book 2 Pediatric Critical Care 60 Extracorporeal Circuits in Children


plasminogen. Aminocaproic acid decreased surgical site and heterogeneity of this patient population. A critically ill
bleeding (from 12% to 7%) in patients receiving ECMO, with child has alterations in organ function, disturbances in end-
the most profound drop in bleeding in the pediatric car- organ perfusion, and fluctuation in fluid status. Adding ECMO
diac surgery population (from 30% to 10%) (Downard 2003). support to a critically ill child creates a new pharmacokinetic
However, no benefit was found in using aminocaproic acid challenge to provide optimal yet safe drug therapies.
to decrease the rate of intracranial hemorrhage in neonates
receiving ECMO (Horwitz 1998). Adsorption
In another study, aminocaproic acid was associated with Drug loss and sequestration in the ECMO circuit are well
thrombotic complications and an increase in the number documented and can contribute to significantly lower drug
of ECMO circuit changes in the treatment group (p=0.02) exposures than expected. Most data on drug adsorption in the
(Downard 2003). Tranexamic acid has shown benefit in ECMO circuit are from ex vivo studies and pharmacokinetic
decreasing blood transfusion (tranexamic acid group: 1.13 modeling. To truly understand the impact of drug sequestra-
mL/kg/hour; no antifibrinolytic group: 2.96 mL/kg/hour; tion in the ECMO circuit, the circuit contribution and the drug
p=0.03) and surgical site bleeding (tranexamic acid group: properties must be appreciated.
0.49 mL/kg/hour; no antifibrinolytic: 2.26 mL/kg/hour; Most adsorption data are from silicone membrane oxy-
p=0.02) in neonates with congenital diaphragmatic hernia genators and non-coated PVC tubing, which are assumed
repair while receiving ECMO. In addition, thrombotic com- to sequester drug to a greater extent than present-day
plications occurred at the same rate in both groups (van der materials. The presence of PVC and other plastics in the
Staak 1997). extracorporeal circuit provides a hydrophobic site for the
Use of activated recombinant factor VII to control refrac- migration of water-insoluble drugs. In a comparative study
tory bleeding in ECMO remains controversial. It has been of a centrifugal pump with PMP membrane oxygenator ver-
found to reduce chest tube output and decrease blood prod- sus a roller pump with silicone membrane oxygenator, drug
uct transfusions in children receiving ECMO (Niebler 2010). recovery of fentanyl and midazolam was significantly lower
Conversely, both non-catastrophic and fatal thrombotic com- in the roller pump/silicone oxygenator circuit (fentanyl 24%
plications have been reported in children and adults receiving vs. 0.4% midazolam; 63% vs. 0.6%; p<0.05) (Wildschut 2010).
ECMO after activated recombinant factor VII administra- The authors did not explain this difference, but given other
tion (Long 2014; Syburra 2010; Chalwin 2008). The decision published experiences, the silicone membrane oxygenator
to administer activated recombinant factor VII should be may be a contributor.
entered into cautiously, weighing the risks versus benefits. In 2007, investigators reported that the PMP membrane
Prothrombin complex concentrates may be a viable option oxygenator did not significantly contribute to the loss of
to alleviate active bleeding and decrease the need for blood fentanyl (around 5%); rather, the PVC tubing alone was asso-
transfusions. These products contain inactivated vitamin ciated with almost 80% of fentanyl sequestration. Morphine
Kdependent factors II, VII, IX, and X and protein C and S at had similar loss in both of the circuit models (with and with-
various concentrations, depending on the product chosen. out PMP membrane oxygenator) in this study, suggesting
However, these agents are contraindicated in disseminated that the membrane oxygenator is not the site of drug seques-
intravascular coagulation and have been associated with one tration (Preston 2007). Coated PVC tubing tested for drug
adult ECMO death (Bui 2002). adsorption showed no difference in the amount of mor-
Aside from hematologic complications in ECMO, envi- phine and fentanyl lost compared with uncoated PVC tubing.
ronmental complications may be prevalent. Because of the However, the rate of drug loss was different. Morphine was
size of the ECMO circuit and the amount of blood circulating sequestered to its saturation point within 5 minutes of admin-
outside the patient in an ambient temperature environment, istration, whereas fentanyl never reached a saturation point
heat loss can be substantial and lead to hypothermia. for the duration of the 6-hour study (Preston 2010).
Active warming of the ECMO circuit blood almost always As the ECMO circuit ages, the surfaces may become sat-
takes place in neonatal and pediatric patients via a heat urated, and drug loss may not be as extreme. This principle
exchanger (see Figure 1-1). Topical warming strategies (e.g., was assessed in a newly primed circuit and a circuit that was
heating blankets) can be effective in smaller patients, but used on a patient for at least 48 hours. In this study, recovery
larger patients often require a heat exchanger that warms of fentanyl, morphine, and midazolam did not differ signifi-
the circuit blood before returning it to the patient. The goal cantly between the new and used circuit after a 180-minute
temperature in ECMO is normothermia (36C37.5C), unless run in each (Wildschut 2010).
otherwise indicated. Drugs that are basic, nonpolar, and lipophilic tend to bind
to the ECMO circuit components and contribute to drug loss.
Pharmacokinetic Changes The logP represents the octanol-water coefficient in which
Clinical pharmacokinetic studies of ECMO are limited by small higher values denote lipophilicity and lower and negative val-
sample sizes, variations in ECMO circuits, and complexity ues indicate hydrophilicity (Table 1-2).

PedSAP 2016 Book 2 Pediatric Critical Care 61 Extracorporeal Circuits in Children


Table 1-2. Drug Characteristics and Adsorption in ECMO

Drug LogPa Protein Binding (%) Drug Loss (%)

Sedatives/Analgesics
Dexmedetomidine 2.8 94 1173
Diazepam 2.82 98 88
Fentanyl 4.05 8085 70100
Hydromorphone 0.9 20 NT
Lorazepam 2.39 8591 3040
Midazolam 3.89 97 6887
Morphine 0.89 3040 2040
Propofol 3.79 9599 98
Antibiotics
Ampicillin 1.35 1018 1538
Cefazolin -0.58 7486 1548
Ceftriaxone -1.7 95 20
Ciprofloxacin 0.28 2040 4
Gentamicin -3.1 < 30 10
Meropenem -0.6 2 20
Piperacillin/tazobactam 0.3/-1.8 2633/3132 NT
Tobramycin -5.8 < 30 0
Vancomycin -3.1 55 936

a
LogP is the octanol-water coefficient. The higher the number, the higher the lipophilicity; negative logP implies hydrophilicity.
NT = not tested, no published information available for circuit adsorption of drug.

A drug with a high logP value and thus a high lipophilicity significantly sequestered by the ECMO circuits compared
has been associated with greater adsorption into the ECMO with the PVC controls (recovered fentanyl: 3% vs. 82%; recov-
circuit (Shekar 2015b). Some commonly used lipophilic drugs ered midazolam: 13% vs. 100%; p<0.05). Morphine recovery
in critically ill children receiving ECMO include fentanyl, ben- in the circuit and control samples was similar, which is likely
zodiazepines, and other sedatives. Literature has described because of its more hydrophilic properties (Shekar 2012b).
fentanyl sequestration of 70%100% in ECMO circuits Of the antimicrobials tested ceftriaxone, caspofungin,
(Wildschut 2010; Mehta 2007; Preston 2007). Similarly, there ciprofloxacin, fluconazole, linezolid, meropenem, and van-
are reports of significant sequestration in the ECMO circuit of comycin only meropenem showed a difference in drug
sedatives such as lorazepam, midazolam, diazepam, and pro- recovery between the control and circuit samples (42% vs.
pofol (Mulla 2000). The obstacle with applying these data to 20%; p<0.05) (Shekar 2015b). The authors concluded that
present-day practice is that many of these studies were not meropenem was not stable at the physiologic temperature
conducted on contemporary ECMO circuits with PMP hollow- of the ECMO circuit and thus was degraded and sequestered,
fiber membrane oxygenators and coated PVC tubing. leading to drug loss (Shekar 2012b). To combat adsorption
Some investigators have been funded to a complete a drug loss, higher doses should be considered to achieve
multiphase pharmacokinetic study of antibiotics, seda- desired results. In addition, infusing the drug into the patient
tives, and analgesics in a series of modern ECMO models: rather than the circuit exposes the patient to the drug first
in vitro, in animals, and the final phase in adults. The first before it can be sequestered (Hoie 1993).
phase, which tested 80 samples for each drug, was con- One additional factor to consider is the effect of ECMO on
ducted in four contemporary ECMO circuits and four control total parenteral nutrition, specifically the lipid component. Fat
PVC jars over a 24-hour sampling period. As expected, the emulsions may cause issues within the ECMO circuit because
lipophilic drugs such as fentanyl and midazolam were of their hydrophobic properties, including layering out of the

PedSAP 2016 Book 2 Pediatric Critical Care 62 Extracorporeal Circuits in Children


emulsion, agglutination, and clot formation. Toavoid these receiving ECMO, leading to recommendations to extend the
complications, fat emulsions or a 3:1 total parenteral nutrition administration interval (Mulla 2005; Buck 2003). The stan-
should be administered to the patient instead of through an dard practice of routine serum creatinine and urine output
ECMO circuit access point (Buck 2005). monitoring can provide the information necessary to guide
renal dosing adjustments in children receiving ECMO.
Distribution
Management of medication therapy in patients receiving
Volume of distribution of medication is increased in ECMO
ECMO is complex. Balancing the pharmacokinetic changes
for several reasons. Drug sequestration, as described pre-
in the critically ill child together with the effects of ECMO on
viously, can result in increased distribution of lipophilic
the patient and drug therapy is not easy. Specific drug dosing
compounds (Shekar 2012a). In a series of ovine ECMO mod-
recommendations for patients receiving ECMO are scarce.
els, lipophilic drugs were associated with an increased
Monitoring serum drug concentrations to avoid organ toxicity
volume of distribution at steady state (p<0.05) (Shekar
and optimize therapeutic end points is prudent. When serum
2015a). Temperature or pH fluctuations within the ECMO
drug concentrations are unavailable, titrating the drug to the
system may affect the binding affinities for protein-bound
desired effect while avoiding adverse effects is a reasonable
medications. An association with higher protein binding
approach.
and increased volume of distribution has occurred in ovine
models (p=0.05) (Shekar 2015a). The additional volume Future Direction
of the ECMO circuit and the administration of blood prod- The technology allowing for ECMO support continues to
ucts and other volume expanders can lead to hemodilution, evolve and bring new and exciting discoveries that will
thus increasing the volume of distribution of certain drugs, improve outcomes. As technologies are introduced into prac-
especially hydrophilic medications (Shekar 2012a). This tice, the pharmacist will be challenged to provide optimal
expanded volume for drug distribution can be significant for medication therapy with the paucity of data. An understand-
achieving the desired pharmacodynamic end points. Special ing of how ECMO technologies interact and influence drug
attention should be paid to these medications because they adsorption, distribution, metabolism, and elimination within
may need dosage adjustments to meet the pharmacody- the circuit and within the patient will be pivotal to providing
namic target (Buck 2003). exceptional pharmaceutical care.

Metabolism
CONTINUOUS RENAL REPLACEMENT
The liver receives pulsatile blood flow from the left side of the
THERAPY
heart, which produces perfusion pressure. In VA-ECMO, blood
Indication
flow is altered, causing changes in end-organ perfusion pres-
sure, which can affect the function and capacity of the organ. Historically, the indications for hemodialysis were sum-
Hepatic metabolism may be altered by the nonpulsatile flow marized using the acronym AEIOU acidosis, electrolyte
to the liver that occurs during ECMO; however, this effect has imbalances (e.g., hyperkalemia, hyperphosphatemia,
not been quantified. Moreover, the liver end-organ perfusion hypercalcemia), intoxications, fluid overload, and uremia.
status may change if the liver was injured in the events lead- Continuous renal replacement therapy has been used in
ing up to cannulation. acute kidney injury and in patients with chronic kidney dis-
ease who cannot tolerate intermittent hemodialysis because
Elimination of hemodynamic compromise. Treatment of fluid overload
Changes in renal elimination during ECMO are likely multi- has become a topic of discussion, given that the adult lit-
factorial. Renal hypoxia and hemodynamic instability with erature suggests increased mortality with fluid overload in
alterations in renal blood flow, injury mediated by poor per- septic shock (Boyd 2011). Pediatric literature states that fluid
fusion, and inflammation can reduce renal clearance of overload greater than 10% leads to increased mortality, and
medications. The impact of nonpulsatile blood flow to the the addition of hemofiltration may increase survival (Gillespie
kidney with VA-ECMO has been theorized to contribute to 2004; Goldstein 2001).
decreased renal clearance (Buck 2003). Renal dysfunction In recent years, there has been a shift toward using
has also been described with VV-ECMO, which allows the CRRT for nonrenal indications such as systemic inflamma-
native cardiac output to provide end-organ perfusion pres- tory response syndrome and sepsis. Profound systemic
sure (Buck 2003). Therefore, the presence of pulsatile versus inflammatory responses create capillary leak and endothe-
nonpulsatile blood flow may not be as contributory as previ- lial damage from many cytokines and complement proteins
ously thought. Because of the many inciting factors stated and can lead to death without adequate support. Continuous
earlier, renal clearance of medications in patients receiving renal replacement therapy has been evaluated for filtration
ECMO can be delayed from the patients baseline. Delayed of the blood of these pro- and anti-inflammatory mediators
clearance of drugs removed by the kidney (e.g., gentami- (Honore 2013). Mixed results have been documented with
cin, vancomycin) has been described in neonatal patients conventional use of CRRT, and research has moved toward

PedSAP 2016 Book 2 Pediatric Critical Care 63 Extracorporeal Circuits in Children


creating more adsorbent filters and columns to remove patients (Hackbarth 2005; Pasko 2003). Other marketed fil-
these mediators (Honore 2013; Schetz 1999). Therefore, an ters are composed of polyarylethersulfone or polysulfone
increase in CRRT use earlier in the treatment course for ill- derivatives. Although these products boast lower bradyki-
nesses such as septic shock or acute respiratory distress nin release on CRRT initiation, they are unavailable in the
syndrome may become more common (Schetz 1999). range of sizes necessary to meet the needs of all pediatric
patients.
Circuit Characteristics
Dialysate and Replacement Fluids
The advantages of CRRT are providing continuous solute
Dialysate and replacement fluids typically contain sodium,
and/or fluid removal in hemodynamically unstable patients.
potassium, chloride, bicarbonate, dextrose, calcium, mag-
This stems from the slower solute and fluid removal rate,
nesium, phosphate, and lactate. Only small amounts of
which allows CRRT use in patients who cannot tolerate fluid
lactate are available in the solutions, given that previous
shifts or the high blood flow rate of intermittent hemodialysis.
studies have shown that lactate onlybuffered fluids con-
The standard of care for CRRT circuits in the United States
tribute to lactic acidosis and hemodynamic instability and
includes an integrated machine with pumps that control the
are inferior to bicarbonate-buffered solutions (Sutherland
blood flow rate, fluid rates, and safety mechanisms (e.g., air
2012). Calcium-containing fluids do not include phosphorus
detectors, pressure monitors) and a balancing system that
because of precipitation, and similarly, calcium-contain-
manages fluid removal. The continuous and complex nature
ing fluids are not usually used with citrate anticoagulation.
of this modality requires close monitoring and nursing care to
The dialysate composition may require several adjustments
support, thus necessitating ICU-level care.
throughout the CRRT run to maintain desired electrolyte con-
Vascular Access centrations in the patient. Many commercially available fluids
All CRRTs discussed in this chapter are VV systems. A are in varying electrolyte concentrations (e.g., potassium, cal-
mechanical pump moves the patients blood forward through cium, bicarbonate), which can meet the needs of the patient
the VV system. Often, the limiting step to initiating CRRT is the with little to no additional electrolyte manipulation. Fluids
availability and placement of the appropriate-sized venous can be compounded in the pharmacy if alternative electrolyte
cannula. Infants weighing less than 6 kg generally have the concentrations are required (e.g., greater than 140 mEq/L of
most difficulty with placement and maintenance of venous sodium). Patient-specific dialysis fluids introduce many ave-
access at low blood flow rates (Sutherland 2012). The site nues for error, from prescribing to compounding. Because of
of placement may also influence the longevity of the circuit the risks involved, the Institute for Safe Medication Practices
and success of providing CRRT. Femoral venous lines have has placed dialysis solution on the high-alert medication list
the most complications related to patient movement and for acute care centers.
also carry the highest infection risk. Subclavian and internal Dialysate runs countercurrent to the patients blood and
jugular venous cannula placement is associated with longer remains separated by the semipermeable membrane in the
circuit survival because these catheters are not as sensitive hemofilter. The goal of the dialysate is to create a diffusion
to patient movement (Sutherland 2012). gradient across the membrane in order to return the blood
to normal physiologic electrolyte composition. The counter-
Circuit Filter current flow optimizes the concentration gradient across the
Many hemofilters and biocompatible membranes are avail- length of the filter. Titration of blood flow rate is more effec-
able for use with CRRT. These filters come in a variety of tive at optimizing diffusion than titration of dialysate rate.
pore sizes, but most of the high-flux membranes used in After passage through the hemofilter, the dialysate fluid is
CRRT have pore sizes of 20,00030,000 Da (Bhler 1999). routed to an effluent bag, which will be wasted.
Pore size is an important attribute when considering the Filter replacement fluid (FRF) is deposited into the hemofil-
passage of drugs across the dialysis membrane. Similar to ter either before or after the patients blood enters the chamber
ECMO, the CRRT circuit requires blood priming for infants if (Figure 1-2). Pre-FRF is used to dilute the blood entering the
the circuit volume exceeds 15% of the patients estimated filter to decrease hemofilter clotting and enhance convection.
blood volume (Sutherland 2012). This can be particularly Post-FRF is added to reverse hemoconcentration after solute
problematic when using an acrylonitrile-69 (AN-69) filter, and plasma water removal that occurred within the hemofil-
which is associated with bradykinin release syndrome. In ter. Whether pre- or post-FRF, the solution will be infused into
bradykinin release syndrome, blood encounters the AN-69 the bloodstream. Therefore, the electrolyte composition of
filter, ultimately leading to the release of bradykinin, a potent the FRF should be conducive to the ongoing treatment of the
vasodilator, which results in profound hypotension shortly patient. When concurrent dialysate and FRF are used, using
after initiation. Strategies have been published to mitigate the same solution composition for both greatly decreases the
this reaction and provide a safer initiation process for these risk of medication errors (Sutherland 2012).

PedSAP 2016 Book 2 Pediatric Critical Care 64 Extracorporeal Circuits in Children


Venous outlet
(to patient)

Calcium infusion Hemofilter


(to patient)

Venous inlet
(from patient)

Blood
Blood
Pump
Pump

Effluent Infusiona Dialysate FRF

Figure 1-2. Continuous renal replacement therapy (CRRT) circuit arrangement.


a
Infusion can be pre-blood pump anticoagulation, such as trisodium citrate.
FRF = filter replacement fluid (can be pre- and/or post-hemofilter).

CRRT Comparison transmembrane pressure, or convection. Diffusion allows the


Continuous renal replacement therapy can be continuous passage of smaller molecules, whereas convection moves
hemodialysis, hemofiltration, or both (Table 1-3). Hemodialysis both small- and middle-weight molecules across the mem-
is accomplished through diffusion across a semipermeable brane (Sutherland 2012).
membrane from an area of high concentration to low con- The prescription for CRRT depends on the modality used. The
centration, whereas hemofiltration is the movement of solute blood flow rate is ultimately determined by the size of the cath-
with fluid across the semipermeable membrane because of eter but is usually 215 mL/kg/minute (50200 mL/minute).

Table 1-3. Differences Between Continuous Renal Replacement Modalities

Abbreviation Continuous Modality Clearance Mechanism FRF Needed? Influences Drug Removal

CVVHD Hemodialysis Diffusion No Dialysis flow rate


CVVH Hemofiltration Convection Yes Ultrafiltration rate
SCUF Hemofiltration Convection No Ultrafiltration rate
CVVHDF Hemodiafiltration Diffusion, convection Yes Dialysis flow rate, ultrafiltration rate

CVVH = continuous veno-venous filtration; CVVHD = continuous veno-venous hemodialysis; CVVHDF = continuous veno-venous
hemodiafiltration; FRF = filter replacement fluid; SCUF = slow continuous ultrafiltration.

PedSAP 2016 Book 2 Pediatric Critical Care 65 Extracorporeal Circuits in Children


ionized calcium of 12 mg/dL (0.250.5 mmol/L). Because
Box 1-1. Example of a CRRT Prescription
of the risk of hypocalcemia, patients should receive a con-
CRRT Parametersa: tinuous calcium chloride or calcium gluconate infusion to
1. Type: CVVHDF
correct their ionized calcium to normal physiologic con-
Filter: M60
centrations (4.45.2 mg/dL or 1.11.3 mmol/L) (Brophy
Catheter: Central, right internal jugular
Initiation procedure: Blood bypass 2005). The calcium infusion is administered to the patient
2. Blood flow: 50 mL/min to replete the ionized calcium deficit hence, the term
Post-FRF: 50 mL/hr regional anticoagulation. Citrate is metabolized to bicarbon-
Pre-FRF: 0 mL/hr ate in the liver and skeletal muscle, which can contribute
Dialysis rate: 800 mL/hr
to elevated bicarbonate concentrations and a subsequent
Fluid balance: 0 mL/hr
3. Anticoagulation: Anticoagulant Citrate Dextrose metabolic alkalosis. Citrate clearance may be impaired
Pre-pump citrate: 40 mL/hr in patients with liver dysfunction, resulting in an accu-
mulation of citrate, which potentiates hypocalcemia
a
Typically, a CRRT prescription has three parts (1) circuit/ (ionized) and hypotension (Sutherland 2012; Fleming 2011).
access information and mode of CRRT, (2) dialysis and ultrafil-
tration information, and (3) anticoagulation information. Monitoring ionized calcium in the circuit and ionized and
CRRT = continuous renal replacement therapy; CVVHDF = con- total calcium concentration in the patient is imperative for
tinuous veno-venous hemodialysis; FRF = filter replacement successful CRRT regional anticoagulation. Rapid bedside
fluid.
point-of-care devices that perform ionized calcium con-
centrations have made regional citrate anticoagulation a
feasible option.
Dialysis rates range from 20 mL/kg/hour to a maximum of 40 Trisodium citrate and unfractionated heparin have been
mL/kg/hour (Sutherland 2012). The FRF rate is determined by compared as anticoagulation strategies in pediatric CRRT.
the intended use. Ultrafiltration is set as an hourly net loss or net Both agents have equal efficacy with respect to circuit anti-
gain or to remain net even, according to total hourly input that coagulation, but unfractionated heparin is associated with
includes the FRF rate as well as intravenous medications given. an increased risk of bleeding. This is not surprising because
Accumulated excess extracellular fluid is not directly removed unfractionated heparin produces systemic anticoagulation in
through CRRT, and hemodynamic instability may occur with over- the patient in addition to the circuit (Brophy 2005).
zealous intravascular fluid removal via ultrafiltration (Box 1-1).
Complications
Anticoagulation Technical complications of the CRRT circuit include clot for-
Much like the ECMO circuit, the CRRT circuit stimulates an mation and obstructed vascular access, resulting in inlet or
acute inflammatory response, the activation of platelets, and outlet pressure concerns. Patient-specific complications
the clotting cascade, leading to clot formation because of the such as bleeding from heparinization, metabolic alkalo-
interaction of whole blood with foreign surfaces. Similar to sis from citrate overload, electrolyte abnormalities from
ECMO, systemic anticoagulation with unfractionated heparin aggressive dialysis, or hypotension from ultrafiltration have
has been used for anticoagulation of CRRT circuit; however, previously been discussed.
regional anticoagulation with trisodium citrate, also known as With aggressive dialysis and ultrafiltration, the loss of
anticoagulant citrate dextrose, is an alternative. amino acids and water-soluble vitamins, with the exception
Anticoagulation practices vary by institution and modal- of B12, can be significant. Critically ill patients including chil-
ity. Given the intermittent hemodialysis experience, dren are often hypermetabolic and hypercatabolic because
unfractionated heparin has been the mainstay for CRRT of the increased energy expenditures of the stressed state.
anticoagulation for many years. The unfractionated hepa- Furthermore, the clinician must consider the energy needed
rin infusion is administered into the CRRT circuit pre-filter for normal growth and development or at least maintenance
and titrated to achieve a post-filter aPTT of 1.52 times nor- of the nutritional status during critical illness in a child.
mal values or an ACT of 180220 seconds (Sutherland 2012). Therefore, a positive nitrogen balance should be maintained,
Other parenteral anticoagulants such as argatroban and even in the presence of renal failure.
bivalirudin do not have much utility, even in heparin-induced Adult studies suggest that 5%21% of amino acids admin-
thrombocytopenia, since the introduction of citrate regional istered in the CRRT circuit can be lost (Btiache 2008). Amino
anticoagulation. acid loss in children receiving CRRT has been cited as up to
Citrate binds to ionized calcium, which is a cofactor for 12% (Maxvold 2000). Therefore, giving more than 2 g/kg/day
activation of the intrinsic pathway of the clotting cascade. of protein is recommended (Maursetter 2011; Btiache 2008).
Removing available calcium halts the forward progression Water-soluble vitamins can be replaced with a standard
of the cascade. Trisodium citrate is delivered pre-filter to enteral childrens multivitamin or an intravenous multivita-
chelate calcium within the filter to achieve an intra-circuit min added to total parental nutrition. Patients receiving high

PedSAP 2016 Book 2 Pediatric Critical Care 66 Extracorporeal Circuits in Children


ultrafiltration rates may need additional thiamine because supplemental dosing, as used in intermittent hemodialysis.
stores of this water-soluble vitamin can be exhausted in However, predicting the elimination rate and magnitude at
as quickly as 1 week (Berger 2004). The trace elements which a drug is removed is difficult. Much of the drug loss
selenium, copper, manganese, chromium, and zinc are not depends on the physiochemical properties of the medication,
extensively removed by CRRT in children (Pasko 2009). the type and rate of CRRT modality (hemodialysis or hemo-
Daily supplementation of these trace elements above nor- filtration), and even the residual renal function of the patient.
mal recommended daily values is not recommended unless Physiochemical properties that can influence medica-
a deficiency is identified. tion elimination by CRRT include molecular size, charge,
Environmental complications are possible with CRRT hydrophilicity, volume of distribution, and protein binding.
like with ECMO. Administration of large volumes of fluid Smaller molecules are more likely to cross than larger com-
at ambient temperature can result in hypothermia in these pounds. Small molecules (less than 500 Da) typically move
patients. A blood warmer attached to the return line of the across the dialysis membrane by diffusion, whereas middle
patient may alleviate this issue, but using warming strate- molecules (5005000 Da) usually cross with the convec-
gies directly to the patient may also be of benefit. Warming tive forces of ultrafiltration (Veltri 2004). Larger compounds
the dialysis fluid or FRF is not recommended because above 5000 Da such as albumin seldom pass across the
uniform heating of these fluid bags is difficult and may com- membrane (Box 1-2).
promise stability. Molecular charge will limit the passage of molecules
across the semipermeable membrane. As with most mem-
Pharmacokinetic Changes brane scenarios, polar molecules become trapped on one
The pharmacokinetic properties most influenced by the side of a membrane, whereas neutral molecules pass more
addition of a CRRT circuit are drug distribution and elim- readily. However, this can be complicated by the surface
ination. Metabolism is not likely affected by CRRT, but it is charge of the semipermeable membrane, making it difficult
assumed altered by the critical illness of the child. Few stud- to predict molecular movement.
ies describe the extent of drug adsorption in CRRT. The filter Medication solubility also affects movement across the
and tubing of the CRRT circuit are made of plastic polymers membrane. Water-soluble drugs tend to have smaller vol-
that are assumed to adsorb drugs much like in ECMO, but the umes of distribution than their lipophilic counterparts.
clinical significance at this time is unknown. Drugs with smaller volumes of distribution are concentrated

Distribution
Critical illness and acute renal failure can increase the volume
of distribution, regardless of the addition of dialysis modali- Box 1-2. Molecular Size of
ties. The same volume of distribution increase seen with the Common Drugs
ECMO circuit occurs with CRRT because of the additional vol- Small (< 500 Da)
ume necessary to prime and run the circuit. The CRRT circuit Amino acids
is a network of tubing and a filter that requires a fluid volume Ampicillin
of 40200 mL, depending on the filter set chosen. Therefore, Barbiturates
drugs that distribute predominantly in the extracellular space
Benzodiazepines
Carbapenems
will have an increase in volume of distribution. Volume of dis- Catecholamines
tribution also plays a role in the ability of the CRRT circuit Electrolytes
to remove the medication. Medication with a volume distri- Fluoroquinolones
bution over 0.7 L/kg likely will not be significantly removed Levocarnitine
(Veltri 2004). Because of the continuous nature of the modal-
Linezolid
Nafcillin
ity, it is possible to have removal of drugs with larger volumes Opioids
of distribution, but the rate of this removal depends on the Thiamine
intercompartment equilibration rate as well as other phys- Zinc
iochemical properties discussed in the text that follows Medium (5005000 Da)
(Veltri 2004). Protein binding is unlikely to be affected by the Aminoglycosides
addition of the CRRT circuit, but protein binding does influ- Cephalosporins
ence drug elimination across the filter membrane.
Daptomycin
Piperacillin/tazobactam
Elimination
Vancomycin
Large (> 5000 Da)
The most significant role of CRRT is clearance of solute and Albumin
removal of fluid, but often at the expense of drug removal. Immune globulin
The continuous nature of CRRT eliminates the need for Monoclonal antibodies

PedSAP 2016 Book 2 Pediatric Critical Care 67 Extracorporeal Circuits in Children


in the extracellular compartment and available in the circula-
Box 1-3. Equations for Estimating
tion for removal. Larger volume of distribution medications
Clearance in CRRT
(e.g., digoxin, amiodarone) are widely distributed in tissues,
allowing for minimal impact of dialysis on drug removal. Estimating Specific Medication Clearance
Sieving coefficient
Drugs that are 80% or greater protein bound are unlikely
to be removed by hemodialysis and hemofiltration (Veltri
SC = 1 fraction bound of drug
CVVH
2004). Unbound or free drug is more efficiently removed by Qr SC
a

either mechanism of CRRT (Joy 1998). The nature of criti- If pre-FRF prescribed: Qr SC (QL/[QL + Qrf])
cally illness and variability in protein production throughout If post-FRF prescribed: Qr SC
CVVHD
normal growth and development of children provides addi-
tional complexity in predicting pharmacokinetic changes
Qd Sd or Qd SC
Sd is not always available and has been clinically ap-
with protein binding (Veltri 2004). plied as SC
Drug clearance may be enhanced if the native kidney CVVHDF
maintains some residual function or if other nonrenal elimi- (Qr + Qd) SC
nation pathways are present. If the native kidney has some Adjustment for the pediatric patient:
residual filtration and secretion capacity, it will contribute Clearance (mL/min)/BSA (m ) 2

to drug clearance. It is difficult to quantify residual renal


Clearance (mL/min)/kg
Estimating CrCl for any CRRT modality
function on CRRT because the modality removes urea and
([Qd + Qr] 1.73 )/(BSA 60 )
b c

creatinine. Therefore, using serum creatinine as a marker


for calculating creatinine clearance is unreliable. Urine a
The ultrafiltration rate is the sum of pre- and post-FRF rate
less the hourly net loss or net gain.
output may be an indicator of residual renal function, but b
This factor (1.73) reflects the standard adult BSA that the
it is difficult to interpret and quantify the urine clearance childs BSA is normalized against.
of creatinine without additional testing. Nonrenal routes c
This factor reflects changing the units from milliliters per hour
of elimination may arise in the setting of renal failure. For to milliliters per minute.
example, imipenem clearance in patients with acute kidney BSA = body surface area; CVVH = continuous veno-venous fil-
tration; CVVHDF = continuous veno-venous hemodiafiltration;
failure is higher than in patients with chronic renal failure Qd = dialysate rate (milliliters per hour); QL = blood flow rate
(95 mL/minute vs. 10.5 mL/minute, p<0.02), likely because (milliliters per minute); Qr = ultrafiltration rate (milliliters per
hour); Qrf = pre-replacement fluid rate (milliliters per hour); SC
of hepatic elimination compensation for the acute change
= sieving coefficient; Sd = saturation coefficient.
in renal clearance (Mueller 1993).
Many strategies are used to estimate drug clearance in
CRRT. Some methods are more difficult because they require
the use of information that is difficult to retrieve such as potential underdosing of -lactam antibiotics have been
the sieving coefficient, saturation coefficient, and drug con- reported in adult patients receiving CRRT (Roberts 2015;
centrations in effluent fluids (Joy 1998). It is estimated that Ulldemolins 2014; Seyler 2011). For example, the recom-
continuous veno-venous filtration (CVVH) corresponds to mended dosing of meropenem in children receiving CRRT
a creatinine clearance rate of 1030 mL/minute/1.73 m2, is 2040 mg/kg every 12 hours; however, a pediatric phar-
whereas continuous veno-venous hemodialysis (CVVHD) macokinetic modeling study suggested a dosing interval
estimates a clearance of 1520 mL/minute/1.73 m2. When of 8 hours in children younger than 5 years (Nehus 2014).
convection and diffusion clearance are combined in continu- To further complicate matters, an in vivo study of children
ous veno-venous hemodiafiltration (CVVHDF), the estimated older than 5 years confirmed the dosing of meropenem 20
drug clearance rate is 1050 mL/minute/1.73 m2. When CRRT mg/kg every 12 hours in CRRT (Nehus 2016). It is import-
dosing information is unavailable, using these creatinine ant to evaluate the literature that addresses individual drug
clearance estimations is a reasonable starting point in mak- therapy behavior within the CRRT circuit. Data from the
ing drug dosing adjustments in CRRT. Because CRRT doses primary literature, coupled with the clinical scenario and
in children vary greatly with patient size and CRRT modality, knowledge of the interactions with the extracorporeal cir-
creatinine clearance estimations should be normalized for an cuit, should form the basis to provide optimal drug therapy
adult body surface area (1.73 m2) (Aronoff 2007) (Box 1-3). for these patients. Monitoring drug concentrations when
An optimal medication therapy regimen must be created clinically feasible is recommended to ensure attainment of
for a patient receiving CRRT. Reports suggest that cur- therapeutic targets.
rent recommendations for dosing in CRRT are inadequate
in meeting therapeutic end points. Variable clearance and

PedSAP 2016 Book 2 Pediatric Critical Care 68 Extracorporeal Circuits in Children


Patient Care Scenario
J.R. is a 4-year-old boy (height 39 inches [100 cm],
weight 15 kg) with febrile neutropenia admitted to the Blood flow: 80 mL/min
pediatric ICU for gram-negative septic shock. He was ini- Dialysis rate: 1000 mL/hr
tiated on CVVHDF for fluid overload and an acute kidney
Post-FRF: 200 mL/hr
injury that was precipitated by the infection. He remains
Ultrafiltration: Net loss 10 mL/hr
anuric. His current BUN is 12 mg/dL and SCr is 0.3 mg/dL.
His CVVHDF prescription is written as follows:
What is this patients estimated CrCl on CVVHDF?
ANSWER
This patients CrCl on CVVHDF is estimated as 54 mL/
min/1.73 m2.

Body surface area (BSA) = (weight in kilograms


height in centimeters/3600) Using SCr to estimate the CrCl would not be accurate
(15 kg 100 cm/3600) = 0.645 m2 because most of the SCr is being eliminated by the CRRT
circuit. It is unlikely that J.R. has any residual renal func-
Qr = Pre-FRF + Post-FRF (fluid balance)
tion that would contribute to CrCl, as suggested by his
Qr = 0 + 200 (-10) = 210 mL/hr anuria. Therefore, it is assumed that CrCl is performed by
the CRRT circuit, and this calculation can be used as a
CrCl = ([Qd + Qr] 1.73)/(BSA 60) surrogate to help dosing medication in this scenario.
([1000 + 210)] 1.73)/(0.645 60) = 54 mL/min/1.73 m2

1. Bhler, J, Donauer J, Keller F. Pharmacokinetic principles during continuous renal replacement therapy: drugs and doses. Kidney
International. 1999;56:S24-8.
2. Veltri MA, Neu AM, Fivush BA, et al. Drug dosing during intermittent hemodialysis and continuous renal replacement therapy: special
considerations in pediatric patients. Paediatr Drugs 2004;6:45-65.

Future Direction coagulopathy, hepatic encephalopathy, cerebral edema,


As with ECMO, renal replacement therapies continue to hypotension, and renal failure. Because of the nature of
evolve. Currently, a CRRT circuit is used in Europe for pre- and this disease and the lack of therapeutic options, interest in
full-term neonates and infants weighing 210 kg. This circuit, extracorporeal liver support has grown. The first artificial liv-
the Cardio-Renal Pediatric Dialysis Emergency Machine or ers were constructed in Italy and Japan in the 1950s. Over
CARPEDIEM, touts efficiency with smaller vascular access the next several decades, machines were designed with
points (4 French), a smaller priming volume (30 mL), and more advances in membrane technology and improved compre-
sensitive scales (within 1 g) to allow for lower blood flow, dial- hension of the functionality of the liver.
ysis rates, and ultrafiltration rates (Ronco 2014). This fills a Both biologic and non-biologic liver support systems
necessary gap in current CRRT management for the small- have been explored. Biologic liver support systems integrate
est patients. a mechanical device with human or porcine hepatocytes
More attention on conducting quality pharmacokinetic that have synthetic, metabolic, and excretory functions
studies in CRRT is needed. Current literature features dif- (Hassanein 2011). These biologic extracorporeal liver circuits
ferent modes of CRRT, differing doses of hemodialysis and are only available in the investigational setting at this time.
hemofiltration, and differing medication doses tested both in Non-biologic systems mimic hemodialysis with albumin and
vitro and in vivo. A standardized approach may help clarify other artificial membranes to detoxify the blood, similar to a
the magnitude of drug clearance in differing CRRT modalities functional liver. Historically, non-biologic systems have been
(Vaara 2012). used as a bridge to liver transplantation or until organ recov-
ery. Only one system (MARS; Gambro, Stockholm, Sweden)
has FDA approval for hepatic encephalopathy, drug overdose,
EXTRACORPOREAL LIVER SUPPORT
or poisoning.
Indication Current evidence is limited, and improvement in sur-
Fulminant hepatic failure is an egregious and relentless dis- vival has not been established. However, small studies with
ease process with few therapeutic modalities to reverse it, liver assist devices have reported improvement in hepatic
and subsequently, a high mortality rate. Acute liver fail- encephalopathy (p=0.003) and decreases in bilirubin concen-
ure manifests as life-threatening complications such as trations (p=0.005) (Tsipotis 2015). In addition, improvements

PedSAP 2016 Book 2 Pediatric Critical Care 69 Extracorporeal Circuits in Children


in the hemodynamic dysregulation seen in liver failure have system (Prometheus; Fresenius Medical Care, Bad Homburg,
occurred with albumin dialysis devices (Stange 2011). In chil- Germany), features a high-flux hemofilter as well as direct
dren, a single-pass albumin dialysis (SPAD) study bridged albumin adsorption. First, the venous blood flows through
7 of the 9 enrolled patients to liver recovery or transplanta- a specialized filter that extracts albumin-rich plasma; this
tion, reduced hepatic encephalopathy scores in 6 patients, is then is transferred to a neutral resin and then to an anion
improved bilirubin concentrations (baseline: 22.6 mg/dL; 24 exchanger. The filtered plasma passes through an absor-
hours: 8.2 mg/dL), and resulted in a 50% decline in ammonia bent circuit and is reunited with the patients blood; finally,
concentrations (Ringe 2011). it is passed through a high-flux dialyzer before returning
to the patient. The first step in this process removes the
Circuit Characteristics toxin-carrying albumin from the blood, and the second step
All non-biologic extracorporeal liver support systems use the removes ammonia and urea, as expected in hemodialysis.
principles of adsorption and filtration. These systems may be This entire process takes place within one circuit specific to
run continuously like CRRT or intermittently for 48 hours a the Prometheus system. This modality is prescribed intermit-
day. The circuits have a configuration similar to a standard tently with 4- to 8-hour run times once daily, as used in recent
dialysis circuit. Venous blood is removed from the patient, studies (Tsipotis 2015).
passes through a special membrane filter with countercur- The final and most complex artificial liver system cur-
rent albumin, and is then returned. The main variance within rently available is the molecular adsorbent recirculating
non-biologic artificial liver support systems is the manage- system (MARS). It is composed of a blood circuit and an albu-
ment of the toxin-bound albumin. min circuit. Venous blood is pumped into the biocompatible
The SPAD system is the simplest extracorporeal liver sys- high-flux dialyzer, where blood is dialyzed against albumin
tem. This circuit can be likened to continuous veno-venous dialysate. This high-flux dialysis filter features a pore size of
hemodiafiltration (CVVHDF) with an albumin-containing dialy- 50,000 Da to allow the passage of protein-bound molecules
sate solution. The high-flux filter in this circuit features much across the semipermeable membrane. The albumin dialy-
larger pores (50,000 Da) than the typical CRRT filter. This larger sate is regenerated in a closed-loop circuit. It passes through
pore size is necessary to move albumin-bound and other large a low-flux dialysis filter against standard dialysis fluid to
toxins across the membrane and out of the blood, but it does remove water-soluble toxins and then travels to two differ-
not remove immunoglobulins, complement, or clotting pro- ent adsorption columns to remove the protein-bound toxins.
teins. The albumin and protein-bound toxins empty into an One adsorptive column contains charcoal, and the other con-
effluent bag and are discarded after one pass through the tains an anion-exchange resin. After passing through the
hemofilter. The disadvantage of this modality is that it does columns, the albumin is restored and travels through the
not recycle the albumin, which increases the cost. However, high-flux dialyzer again. The filtered blood is then returned
the ability to use this modality on a conventional CRRT to the patient. This complex system can be executed with
machine with albumin dialysate and the appropriate filter pro- a standard CRRT machine for the blood separation and an
vides a distinct advantage. In addition, SPAD is a continuous additional closed-loop circuit for the albumin detoxification.
removal system like CRRT and has lower blood flow rate, which Like the Prometheus system, MARS is typically administered
is advantageous in the hemodynamically unstable patient. intermittently each day for 68 hours. Because of this device
Another extracorporeal support system, the fraction- setup, it is an easier transition back to CRRT when the albu-
ated plasma separation and adsorption hemodialysis min dialysis run is complete (Table 1-4).

Table 1-4. Extracorporeal Liver Assist Device Differences

Liver Assist High-Flux Low-Flux Adsorbent Albumin Prescribed


Device Dialyzer Dialyzer Columns Recycled Interval Machine Used

SPAD Yes No No No Continuous CRRT


MARS Yes Yes Yes (2) Yes Over 48 hr, CRRT +
daily proprietary
circuit
Prometheus Yes No Yes (2) Yes Over 68 hr, Proprietary
daily circuit

MARS = molecular adsorbent recirculating system; SPAD = single-pass adsorption dialysis.

PedSAP 2016 Book 2 Pediatric Critical Care 70 Extracorporeal Circuits in Children


Anticoagulation highly ionic metals and minerals such as sodium, iron, lead,
Use of anticoagulation in these systems is controversial. and iodine are not extensively bound. The anion-exchange
Inherently, patients with fulminant liver failure are coagulo- resin compound binds bile acids and other organic anions.
pathic because of the decreased production of coagulation Drug loss would vary, depending on the placement of these
factors and platelet dysfunction. As a further complication, adsorbent columns and the protein binding of the drug. For
critically ill patients may present with systemic inflamma- example, in the MARS system, the drug would have to pass
tion or disseminated intravascular coagulation. Therefore, across the first semipermeable membrane and into the albu-
providing therapy that blocks the coagulation cascade may min circuit to come in contact with the adsorbent columns.
be excessive and increase the risk of bleeding. Low-dose Highly protein-bound drugs are more likely to be sequestered
unfractionated heparin and regional citrate anticoagulation in the albumin dialysis systems. Even with protein-bound drug
have been used successfully in the MARS and Prometheus sequestration in these extracorporeal devices, the overall
systems (Tsipotis 2015). The SPAD circuit has success- systemic exposure may not be influenced, especially if there
fully used heparin (Ringe 2011). With the short-course, is a high volume of distribution into the tissues. No literature
intermittent daily use of some of these devices (MARS and exists currently to quantify the amount of drug adsorption
Prometheus), the exposure to anticoagulation is limited with these modalities.
(Stange 2011). It is unknown whether distribution and metabolism are
directly affected by the artificial liver assist devices. The
Complications SPAD system, which is similar to CVVHDF, could be postu-
Most published complications consist of catheter-related lated to have effects on distribution similar to that mode of
bleeding, hematomas, and infections. One adult MARS CRRT. Hepatic failure will induce changes in drug distribution
study found no significant difference in bleeding rate in the from fluid overload, ascites, and low protein stores, as well as
study population compared with the standard medical ther- decreased hepatic metabolism and clearance of drugs.
apy group (17.8% vs. 9.5%; p=0.14) (Baares 2013). Similarly, With the high-flux dialysis filter featured in each of these
a large Prometheus randomized controlled trial reported non-biologic liver support devices, drug elimination is likely
a bleeding rate of 43% in the study group and 49% in the affected. The pore size on these filters is quite large; thus,
standard medical therapy group (p=0.79) (Kribben 2012). many drugs used in the critical care setting are likely to
Bleeding rates were not routinely reported in the SPAD trials, move across that membrane. The MARS or Prometheus
although a nonsignificant decline in platelets was reported circuit may be used intermittently with corresponding
compared with standard medical therapy (mean difference alterations in elimination during these periods. The SPAD
15,840 per microliter [95% CI, 24,000 to 55,960]; p=0.44) circuit should behave similarly to CVVHDF drug elimination,
(Tsipotis 2015). The pediatric SPAD study reported a decline given the similarities in configuration with the addition of a
in platelets within the first 24 hours before returning to or larger-pore filter.
exceeding baseline values (Ringe 2011). In a comparative
trial of MARS therapy versus SPAD in adult patients, a sig-
nificant platelet drop occurred in the MARS group compared PLASMAPHERESIS
with the SPAD group (8000 vs. 2000, p=0.032) (Sponholz Plasmapheresis and its intention, in some form, have been
2016). Although the infection rates reached 50% or above around for centuries. The ancient humors of blood, phlegm,
in both groups, there was no difference between standard black bile, and yellow bile were proposed by Hippocrates,
therapy and the MARS or the Prometheus systems (Baares and Claudius Galen later emphasized the need to keep these
2013; Kribben 2012). humors in balance (Fortenberry 2006). The humor imbal-
ances described in ancient medicine are now recognized as
Pharmacokinetic Changes the autoantibodies seen in autoimmune diseases such as
Although development of artificial liver support systems Guillain-Barr syndrome, antigen-antibody complexes, allo-
started over 50 years ago, literature pertaining to pharmaco- proteins responsible for transplant rejection and transfusion
kinetic and pharmacodynamic alterations is lacking. Biologic reactions, endogenous toxins seen in liver failure, and anti- and
liver support systems may have intrinsic metabolic and excre- proinflammatory mediators seen in sepsis. Plasmapheresis,
tory functions that would affect drug dosing. Understanding or TPE, is the practice of separating plasma from whole blood
the technology will facilitate comprehension of how medica- and returning to the body plasma-poor blood with exoge-
tions interact with the liver support devices. nously supplemented fresh frozen plasma, albumin, and other
Because the MARS and Prometheus circuits contain physiologic fluids. Historically, whole blood was manually
adsorbent columns (charcoal and anion-exchange resin), a removed from the patient in aliquots and spun down by a cen-
drug may be lost through adsorption. Charcoal is a porous, trifuge to separate the plasma from the blood cells, and blood
high-surface-area carbon resin that binds well to many sub- cells were returned to the patient. This process is now mod-
stances. However, polar molecules such as alcohols or ernized and administered by mobile machines.

PedSAP 2016 Book 2 Pediatric Critical Care 71 Extracorporeal Circuits in Children


Indication plasma by densities. Plasmapheresis is typically prescribed
Plasmapheresis indications have grown lengthy over the for 34 hours a day, often for several days in a row (35 days).
decades. The American Society for Apheresis maintains an
exhaustive list of diseases that may benefit from TPE, which Anticoagulation
are also graded according to potential benefit. The indications Trisodium citrate regional anticoagulation has been a suc-
are extensive: from autoimmune to infectious to inflammatory cessful anticoagulant in centrifugal TPE circuits. Heparin
processes (Schwartz 2013). Plasmapheresis has gained inter- can be used for either exchange modality and is preferred for
est for managing sepsis with multiorgan failure, especially plasma filtration. The risks and benefits of heparin or triso-
with the scarcity of options to remove excessive anti- and pro- dium citrate anticoagulation are similar to those discussed in
inflammatory mediators to restore homeostasis (Fortenberry previous sections.
2006). The massive systemic inflammatory response is
caused by cytokines such as tumor necrosis factor alpha, Complications
interleukins, prostaglandins, and other mediators. An imbal- Loss of plasma components can create unnecessary com-
ance in pro- and anti-inflammatory systems leads to tissue plications. Proteins such as immune globulins, albumin, and
injury, multiorgan dysfunction syndrome, disseminated intra- coagulation factors are removed, with significant losses
vascular coagulation, and relative immunosuppression. occurring with repeated exchanges. After a single pass of
The goal of TPE in fulminant sepsis is to remove cytotoxins plasma volume exchange, patient immunoglobulin G (IgG)
and return to the body anti-inflammatory mediators, immu- concentrations may drop by 28%. If the exchange volume is
noglobulins, and pro- and anti-inflammatory proteins (Kawai increased to 1.5 times the total body plasma volume, the loss
2015). Both adults and children have reported benefit with this increases to 35% of total body IgG (Annich 2012). Albumin is
intervention in sepsis and thrombocytopenia-associated mul- typically monitored before and after each exchange to help
tiple organ failure (TAMOF). In 1999, one of the first groups to guide the replacement fluid plan. Monitoring of IgG after
report that TPE was associated with a slight improvement in completion of the multiday therapy is warranted. Exogenous
the number of organs that failed in sepsis (plasmapheresis replacement of IgG, albumin, and fresh frozen plasma can
2.57 vs. standard therapy 2.94; p=0.37) found no difference in combat deficiencies and potential complications.
mortality (57% vs. 50%; p=0.73) (Reeves 1999). In a larger ran- With unfractionated heparin anticoagulation, there is
domized controlled study of adults with septic shock, 28-day always a remote risk of bleeding. Paresthesias caused by
all-cause mortality was decreased in the TPE group compared hypocalcemia from citrate exposure have also been reported
with the standard therapy group (33% vs. 53%; p=0.05) (Busund (Winters 2012). Because of its intermittent nature and use
2002). A retrospective pediatric study found similar results, in hemodynamically unstable patients, hypotension may
with 28-day mortality favoring the TPE group over the stan- manifest from fluid shifts. Heat loss from the circuit and
dard therapy group in children with TAMOF (26.6% vs. 70.3%; the addition of large volumes of colloids and crystalloids in
p=0.006) (Sevketoglu 2014). Plasmapheresis has even been replacement fluids stored at ambient temperature contribute
investigated in children with TAMOF receiving other extracor- to hypothermia (Annich 2012).
poreal therapies (e.g., ECMO with or without CRRT), where it
was found to have a 5-fold lower vasopressor infusion score Pharmacokinetic Changes
and improved organ function after treatment (Kawai 2015). The pharmacokinetic changes associated with TPE are not
well described. Many case reports describe the magnitude of
Circuit Characteristics drug removal but do not address changes in adsorption, dis-
The configuration for TPE is similar to that for other extra- tribution, and metabolism.
corporeal therapies. Plasmapheresis can be administered Drug sequestration possibly occurs in the TPE circuit
through two large-bore peripheral intravenous lines; a central because of adsorption to the filter and tubing similar to the other
venous line is not necessary for every patient. Whole blood extracorporeal circuits previously described. Drug adsorption
is extracted from the venous system of the patient and then has not yet been quantified in the published literature for this
separated by plasma filtration or continuous flow centrifuga- modality. Distribution may be intermittently affected with the
tion. The cells are returned to the patient through a venous removal of plasma and protein binding sources and the addi-
access port with a replacement fluid of 4%5% albumin, fresh tion of replacement fluids that would dilute the serum; however,
frozen plasma, normal saline, or a combination, depending on this change is likely transient. Alterations in hepatic metabo-
the clinical scenario. Plasma filtration takes place in a large- lism have not been studied, but plasma esterase metabolism
bore polymer membrane plasma separator accomplished may decrease because of the depletion of pseudocholin-
using modern-day CRRT circuits with the appropriate-sized esterases by TPE, which could ultimately lead to prolonged
filter and necessary replacement fluids. Centrifugal plasma neuromuscular blockade (Naik 2002).
exchange requires a dedicated machine with a spinning cham- Drug elimination is the best-described pharmacokinetic
ber that relies on gravity to separate blood components and alteration in TPE. Typically, TPE is prescribed for 11.5 times

PedSAP 2016 Book 2 Pediatric Critical Care 72 Extracorporeal Circuits in Children


Table 1-5. Pharmacokinetic Changes in Extracorporeal Therapies

Modality Adsorption Distribution Metabolism Elimination

ECMO NC to

CRRT NC or NC or NC or a
Liver assist devices Unknown NC or b c
TPE Unknown NC or Unknown

a
Elimination rate depends on dialysis and ultrafiltration rates.
Liver failure contributes to changes and likely a decrease in hepatic metabolism.
b

Increased elimination of protein-bound drugs only.


c

NC = no change; TPE = therapeutic plasmapheresis.

the patients circulating plasma volume, which can remove


up to 63%72% of circulating plasma components (Ibrahim Practice Points
2012). Plasmapheresis is the most effective modality of drug ECMO
removal in intoxications in highly protein-bound (greater than Because of the large network of polymer tubing, drug
adsorption and sequestration may be significant, neces-
80%) medications or those with a small distribution (volume
sitating higher-than-usual doses of highly adsorbed
of distribution 0.2 L/kg or less) (Ibrahim 2007). Drugs with
medications.
higher free fraction available are not likely removed by TPE. End-organ perfusion changes caused by an inciting
Removal also depends on the serum concentration at the event may adversely affect hepatic and renal clearance.
time of the exchange. Medication administration may need Recommend serum concentration monitoring, when
to be held until after TPE because removal increases with possible; otherwise, follow clinically and titrate to
desired effect.
higher concentrations such as those during peak concen-
CRRT
trations (Ibrahim 2012). Therapeutic drug monitoring should
The different CRRT modalities may use diffusion, con-
be delayed for at least 2 hours after termination of the daily vection, or both with subsequent changes in clearance
exchange to account for redistribution and equilibration. properties.
Drug removal by the CRRT circuit depends on drug phys-
CONCLUSION iochemical properties (e.g., ionization, molecular size,
volume of distribution) and CRRT flow rates.
Caring for the critically ill child is becomingly increasingly Residual renal function can contribute to additional drug
complicated with the addition of various extracorporeal ther- clearance, but this is difficult to quantify.
apies. Alterations in pharmacokinetics and the interaction of Estimating circuit clearance by dialysate and replace-
medications within the extracorporeal systems constantly ment fluid rates can be helpful for designing medication
therapy plans.
challenge the pediatric pharmacist because of the lack of
Extracorporeal Liver Support
clinical data to guide recommendations. When extracorpo-
Large filter pores allow passage of components that are
real therapies are used in combination, a greater degree of 50,000 Da or less, which includes many protein-bound
complexity is added. Even fewer data exist on the interactions drugs.
of medications within concurrent extracorporeal circuits. The magnitude of drug removal likely depends on
Additive therapies create a challenge in designing safe medi- protein binding and therapy duration, although no stud-
ies or case reports have quantified the extent of drug
cation regimens as well as in optimizing pharmacodynamic
removal with these therapies.
targets. The pharmacists recommendations regarding
Plasmapheresis
extracorporeal circuits should account for known drug prop-
Medications with high protein binding (80%) and small
erties and any available literature. Apractical monitoring plan volumes of distribution (0.2 L/kg or less) are effectively
should be in place to ensure the attainment of therapeutic removed by TPE.
end points and avoid toxicities in all cases (Table 1-5). If possible, schedule drug administration after TPE to
decrease drug extraction.

REFERENCES
Agati S, Ciccarello G, Salvo D, et al. Use of a novel antico- Annich GM. Extracorporeal life support: the precarious bal-
agulation strategy during ECMO in a pediatric population: ance of hemostasis. J Thromb Haemost 2015;13(suppl
single-center experience. ASAIO J 2006;52:513-6. 5):336-42.

PedSAP 2016 Book 2 Pediatric Critical Care 73 Extracorporeal Circuits in Children


Annich GM, Lynch WR, MacLaren G, et al, eds. ECMO Byrnes JW, Swearingen CJ, Prodhan P, et al. Antithrombin
Extracorporeal Cardiopulmonary Support in Critical Care, III supplementation on extracorporeal membrane oxygen-
4th ed. Ann Arbor, MI: ELSO, 2012. ation: impact on heparin dose and circuit life. ASAIO J
2014;60:57-62.
Aronoff GR, Bennett WM, Berns JS, et al., eds. American
College of Physicians. Drug Prescribing in Renal Failure: Chalwin RP, Tiruvoipati R, Peek GJ. Fatal thrombosis with
Dosing Guidelines for Adults and Children, 5th ed. activated factor VII in a paediatric patient on extracor-
Philadelphia: Versa Press, 2007. poreal membrane oxygenation. Eur J Cardiothorac Surg
2008;34:685-6.
Baares R, Nevens F, Larsen FS, et al; RELIEF study group.
Extracorporeal albumin dialysis with the molecular adsor- Downard CD, Betit P, Chang RW, et al. Impact of AMICAR on
bent recirculating system in acute-on-chronic liver failure: hemorrhagic complications of ECMO: a ten-year review.
the RELIEF trial. Hepatology 2013;57:1153-62. JPediatr Surg 2003;38:1212-6.

Beiderlinden M, Treschan T, Grlinger K, et al. Argatroban Extracorporeal Life Support Organization (ELSO). ELSO
in extracorporeal membrane oxygenation. Artif Organs Anticoagulation Guidelines. 2014.
2007;31:461-5.
Fleming GM. Renal replacement therapy review.
Berger MM, Shenkin A, Revelly JP, et al. Copper, selenium, Organogenesis 2011;7:2-12.
zinc, and thiamine balances during continuous venove-
Fortenberry JD, Paden ML. Extracorporeal therapies in
nous hemodiafiltration in critically ill patients. Am J Clin
the treatment of sepsis: experience and promise. Semin
Nutr 2004;80:410-6.
Pediatr Infect Dis 2006;17:72-9.
Bizzarro MJ, Conrad SA, Kaufman DA, et al. Extracorporeal
Ganter MT, Hofer CK. Coagulation monitoring: current tech-
Life Support Organization Task Force on Infections,
niques and clinical use of viscoelastic point-of-care
Extracorporeal Membrane Oxygenation. Infections acquired
coagulation devices. Anesth Analg 2008;106:1366-75.
during extracorporeal membrane oxygenation in neonates,
children, and adults. Pediatr Crit Care Med 2011;12:277-81. Gillespie RS, Seidel K, Symons JM. Effect of fluid overload
and dose of replacement fluid on survival in hemofiltra-
Bhler J, Donauer J, Keller F. Pharmacokinetic principles
tion. Pediatr Nephrol 2004;19:1394-9.
during continuous renal replacement therapy: drugs and
dosage. Kidney Int 1999;56:S24-8. Goldstein SL, Currier H, Graf JM. Outcome in children receiv-
ing venovenous hemofiltration. Pediatrics 2001;107:1309-12.
Boyd JH, Forbes J, Nakada TA, et al. Fluid resuscitation in
septic shock: a positive fluid balance and elevated central Hackbarth RM, Eding D, Gianoli C, et al. Zero balance ultra-
venous pressure are associated with increased mortality. filtration (Z-BUF) in blood-primed CRRT circuits achieves
Crit Care Med 2011;39:259-65. electrolyte and acid-base homeostasis prior to patient
connection. Pediatr Nephrol 2005;20:1328-33.
Brophy PD, Somers MJ, Baum MA, et al. Multi-centre evalu-
ation of anticoagulation in patients receiving continuous Hassanein TI, Schade RR, Hepburn IS. Acute-on-chronic liver
renal replacement therapy. Nephrol Dial Transplant failure: extracorporeal liver assist devices. Curr Opin Crit
2005;20:1416-21. Care 2011;17:195-203.
Buck ML. Pharmacokinetic changes during extracorporeal Hoie EB, Hall MC, Schaaf LJ. Effects of injection site and
membrane oxygenation: implications for drug therapy of flow rate on the distribution of injected solutions in an
neonates. Clin Pharmacokinet 2003;42:403-17. extracorporeal membrane oxygenation circuit. Am J Hosp
Pharm 1993;50:1902-6.
Buck ML, Wooldridge P, Ksenich RA. Comparison of methods
for intravenous infusion of fat emulsion during extra- Honore PM, Jacobs R, Joannes-Boyau O, et al. Newly
corporeal membrane oxygenation. Pharmacotherapy designed CRRT membranes for sepsis and SIRS a prag-
2005;25:1536-40. matic approach for bedside intensivists summarizing the
more recent advances: a systematic structured review.
Bui JD, Despotis GD, Trulock EP, et al. Fatal thrombosis after
ASAIO J 2013;59:99-106.
administration of activated prothrombin complex concen-
trates in a patient supported by extracorporeal membrane Horwitz JR, Cofer BR, Warner BW, et al. A multicenter trial of
oxygenation who had received activated recombinant fac- 6-aminocaproic acid (Amicar) in the prevention of bleed-
tor VII. J Thorac Cardiovasc Surg 2002;124:852-4. ing in infants on ECMO. J Pediatr Surg 1998;33:1610-3.
Burkhart HM, Joyner N, Niles S, et al. Presence of plasticizer Hursting MJ, Dubb J, Verme-Gibboney CN. Argatroban anti-
di-2(ethylhexyl)phthalate in primed extracorporeal circula- coagulation in pediatric patients: a literature analysis.
tion circuits. ASAIO J 2007;53:365-7. JPediatr Hematol Oncol 2006;28:4-10.
Busund R, Koukline V, Utrobin U, et al. Plasmapheresis in Ibrahim RB, Balogun RA. Medications in patients treated
severe sepsis and septic shock: a prospective, randomised, with therapeutic plasma exchange: prescription dosage,
controlled trial. Intensive Care Med 2002;28:1434-9. timing, and drug overdose. Semin Dial 2012;25:176-89.

PedSAP 2016 Book 2 Pediatric Critical Care 74 Extracorporeal Circuits in Children


Ibrahim RB, Liu C, Cronin SM, et al. Drug removal by plasma- Nehus EJ, Mizuno T, Cox S, et al. Pharmacokinetics of
pheresis: an evidence-based review. Pharmacotherapy meropenem in children receiving continuous renal
2007;27:1529-49. replacement therapy: validation of clinical trial simula-
tions. J Clin Pharmacol 2016;56:291-7.
Joy MS, Matzke GR, Armstrong DK, et al. A primer on con-
tinuous renal replacement therapy for critically ill patients. Nehus EJ, Mouksassi S, Vinks AA, et al. Meropenem in chil-
Ann Pharmacother 1998;32:362-75. dren receiving continuous renal replacement therapy:
clinical trial simulations using realistic covariates. J Clin
Kawai Y, Cornell TT, Cooley EG, et al. Therapeutic plasma Pharmacol 2014;54:1421-8.
exchange may improve hemodynamics and organ failure
among children with sepsis-induced multiple organ dys- Niebler RA, Christensen M, Berens R, et al. Antithrombin
function syndrome receiving extracorporeal life support. replacement during extracorporeal membrane oxygen-
Pediatr Crit Care Med 2015;16:366-74. ation. Artif Organs 2011;35:1024-8.

Kribben A, Gerken G, Haag S, et al; HELIOS Study Group. Niebler RA, Punzalan RC, Marchan M, et al. Activated
Effects of fractionated plasma separation and adsorption recombinant factor VII for refractory bleeding during
on survival in patients with acute-on-chronic liver failure. extracorporeal membrane oxygenation. Pediatr Crit Care
Gastroenterology 2012;142:782-9. Med 2010;11:98-102.

Long MT, Wagner D, Maslach-Hubbard A, et al. Safety and OHoro JC, Cawcutt KA, De Moraes AG, et al. The evidence
efficacy of recombinant activated factor VII for refrac- base for prophylactic antibiotics in patients receiving extra-
tory hemorrhage in pediatric patients on extracorporeal corporeal membrane oxygenation. ASAIO J 2016;62:6-10.
membrane oxygenation: a single center review. Perfusion
2014;29:163-70. Pasko DA, Churchwell IF, Jain JC, et al. Continuous venove-
nous hemodiafiltration trace element clearance in pediatric
Luddington RJ. Thrombelastography/thromboelastometry. patients: a case series. Pediatr Nephrol 2009;24:807-13.
Clin Lab Haematol 2005;27:81-90.
Pasko DA, Mottes TA, Mueller BA. Pre-dialysis of blood prime
Maursetter L, Kight CE, Mennig J, et al. Review of the in continuous hemodialysis normalizes pH and electro-
mechanism and nutrition recommendations for patients lytes. Pediatr Nephrol 2003;18:1177-83.
undergoing continuous renal replacement therapy. Nutr
Clin Pract 2011;26:382-90. Preston TJ, Hodge AB, Riley JB, et al. In vitro drug adsorp-
tion and plasma free hemoglobin levels associated with
Maxvold NJ, Smoyer WE, Custer JR, et al. Amino acid loss hollow fiber oxygenators in the extracorporeal life support
and nitrogen balance in critically ill children with acute (ECLS) circuit. J Extra Corpor Technol 2007;39:234-7.
renal failure: a prospective comparison between classic
hemofiltration and hemofiltration with dialysis. Crit Care Preston TJ, Ratliff TM, Gomez D, et al. Modified surface
Med 2000;28:1161-5. coatings and their effect on drug adsorption within the
extracorporeal life support circuit. J Extra Corpor Technol
Mehta NM, Halwick DR, Dodson BL, et al. Potential drug 2010;42:199-202.
sequestration during extracorporeal membrane oxygen-
ation: results from an ex vivo experiment. Intensive Care Ranucci M, Ballotta A, Kandil H, et al; Surgical and Clinical
Med 2007;33:1018-24. Outcome Research Group. Bivalirudin-based versus
conventional heparin anticoagulation for postcardiot-
Meyer AD, Wiles AA, Riveria O, et al. Hemolytic and throm- omy extracorporeal membrane oxygenation. Crit Care
bocytopathic characteristics of extracorporeal membrane 2011;15:R275.
oxygenation systems at simulated flow rates for neonates.
Pediatr Crit Care Med 2012;13:e255-61. Reeves JH, Butt WW, Shann F, et al. Continuous plasmafiltra-
tion in sepsis syndrome. Plasmafiltration in Sepsis Study
Mueller BA, Scarim SK, Macias ML. Comparison of imipe- Group. Crit Care Med 1999;27:2096-104.
nem pharmacokinetics in patients with acute or chronic
renal failure treated with continuous hemofiltration. Am J Ringe H, Varnholt V, Zimmering M, et al. Continuous
Kidney Dis 1993;21:172-9. veno-venous single-pass albumin hemodiafiltration in
children with acute liver failure. Pediatr Crit Care Med
Mulla H, Lawson G, von Anrep C, et al. In vitro evaluation of 2011;12:257-64.
sedative drug losses during extracorporeal membrane
oxygenation. Perfusion 2000;15:21-6. Roberts DM, Liu X, Roberts JA, et al; RENAL Replacement
Therapy Study Investigators. A multicenter study on the
Mulla H, Pooboni S. Population pharmacokinetics of van- effect of continuous hemodiafiltration intensity on anti-
comycin in patients receiving extracorporeal membrane biotic pharmacokinetics. Crit Care 2015;19:84.
oxygenation. Br J Clin Pharmacol 2005;60:265-75.
Ronco C, Garzotto F, Brendolan A, et al. Continuous renal
Naik B, Hirshhorn S, Dharnidharka VR. Prolonged neu- replacement therapy in neonates and small infants: devel-
romuscular block due to cholinesterase depletion by opment and first-in-human use of a miniaturised machine
plasmapheresis. J Clin Anesth 2002;14:381-4. (CARPEDIEM). Lancet 2014;383:1807-13.

PedSAP 2016 Book 2 Pediatric Critical Care 75 Extracorporeal Circuits in Children


Schetz M. Non-renal indications for continuous renal Sutherland SM, Alexander SR. Continuous renal replacement
replacement therapy. Kidney Int 1999;56(suppl 72):S88-94. therapy in children. Pediatr Nephrol 2012;27:2007-16.

Schwartz J, Winters JL, Padmanabhan A, et al. Guidelines on Syburra T, Lachat M, Genoni M, et al. Fatal outcome of
the use of therapeutic apheresis in clinical practice-evi- recombinant factor VIIa in heart transplantation with
dence-based approach from the Writing Committee of the extracorporeal membrane oxygenation. Ann Thorac Surg
American Society for Apheresis: the sixth special issue. J 2010;89:1643-5.
Clin Apher 2013;28:145-284.
Tsipotis E, Shuja A, Jaber BL. Albumin dialysis for liver
Sevketoglu E, Yildizdas D, Horoz OO, et al. Use of therapeutic failure: a systematic review. Adv Chronic Kidney Dis
plasma exchange in children with thrombocytope- 2015;22:382-90.
nia-associated multiple organ failure in the Turkish
thrombocytopenia-associated multiple organ failure net- Ulldemolins M, Vaquer S, Llaurad-Serra M, et al. Beta-
work. Pediatr Crit Care Med 2014;15:e354-9. lactam dosing in critically ill patients with septic shock
and continuous renal replacement therapy. Crit Care
Seyler L, Cotton F, Taccone FS, et al. Recommended -lactam 2014;18:227.
regimens are inadequate in septic patients treated with con-
tinuous renal replacement therapy. Crit Care 2011;15:R137. Vaara S, Pettila V, Kaukonen KM. Quality of pharmacoki-
netic studies in critically ill patients receiving continuous
Shekar K, Fraser JF, Smith MT, et al. Pharmacokinetic renal replacement therapy. Acta Anaesthesiol Scand
changes in patients receiving extracorporeal membrane 2012;56:147-57.
oxygenation. J Crit Care 2012a;27:741.e9-18.
van der Staak FH, de Haan AF, Geven WB, et al. Surgical
Shekar K, Roberts JA, Barnett AG, et al. Can physicochemi- repair of congenital diaphragmatic hernia during
cal properties of antimicrobials be used to predict their extracorporeal membrane oxygenation: hemorrhagic com-
pharmacokinetics during extracorporeal membrane oxy- plications and the effect of tranexamic acid. J Pediatr
genation? Illustrative data from ovine models. Crit Care Surg 1997;32:594-9.
2015a;19:437.
Veltri MA, Neu AM, Fivush BA, et al. Drug dosing during
Shekar K, Roberts JA, Mcdonald CI, et al. Protein-bound intermittent hemodialysis and continuous renal replace-
drugs are prone to sequestration in the extracorporeal ment therapy: special considerations in pediatric patients.
membrane oxygenation circuit: results from an ex vivo Paediatr Drugs 2004;6:45-65.
study. Crit Care 2015b;19:164.
Walczak R, Lawson D, Kaemmer D, et al. Evaluation of a
Shekar K, Roberts JA, Mcdonald CI, et al. Sequestration of preprimed microporous hollow-fiber membrane for rapid
drugs in the circuit may lead to therapeutic failure dur- response neonatal extracorporeal membrane oxygen-
ing extracorporeal membrane oxygenation. Crit Care ation. Perfusion 2005;20:269-75.
2012b;16:R194.
Whiting D, Dinardo J. TEG and ROTEM: technology and clini-
Shekar K, Roberts JA, Smith MT, et al. The ECMO PK Project: cal applications. Am J Hematol 2014;89:228-32.
an incremental research approach to advance understand-
ing of the pharmacokinetic alterations and improve patient Wildschut ED, Ahsman MJ, Allegaert K, et al. Determinants
outcomes during extracorporeal membrane oxygenation. of drug absorption in different ECMO circuits. Intensive
BMC Anesthesiol 2013;13:7. Care Med 2010;36:2109-16.

Shekar K, Roberts JA, Welch S, et al. ASAP ECMO: Antibiotic, Winters JL. Plasma exchange: concepts, mechanisms,
Sedative and Analgesic Pharmacokinetics during and an overview of the American Society for Apheresis
Extracorporeal Membrane Oxygenation: a multi-cen- guidelines. Hematology Am Soc Hematol Educ Program
tre study to optimise drug therapy during ECMO. BMC 2012;2012:7-12.
Anesthesiol 2012c;12:29. Wong TE, Delaney M, Gernsheimer T, et al. Antithrombin con-
Smythe MA, Koerber JM, Forsyth LL, et al. Argatroban centrates use in children on extracorporeal membrane
dosage requirements and outcomes in intensive care oxygenation: a retrospective cohort study. Pediatr Crit
versus non-intensive care patients. Pharmacotherapy Care Med 2015;16:264-9.
2009;29:1073-81. Wong TE, Huang YS, Weiser J, et al. Antithrombin concen-
Sponholz C, Matthes K, Rupp D, et al. Molecular adsorbent trate use in children: a multicenter cohort study. J Pediatr
recirculating system and single-pass albumin dialysis in 2013;163:1329-34.
liver failure a prospective, randomised crossover study. Young G, Boshkov LK, Sullivan JE, et al. Argatroban therapy
Crit Care 2016;20:2. in pediatric patients requiring nonheparin anticoagulation:
Stange J. Extracorporeal liver support. Organogenesis an open-label, safety, efficacy, and pharmacokinetic study.
2011;7:64-73. Pediatr Blood Cancer 2011;56:1103-9.

PedSAP 2016 Book 2 Pediatric Critical Care 76 Extracorporeal Circuits in Children


Self-Assessment Questions
Questions 1 and 2 pertain to the following case. pneumonia. He has been receiving VV-ECMO for 2 days and
T.M. is an 18-month-old girl (weight 15 kg) admitted to the is anticoagulated with unfractionated heparin. G.V.s current
pediatric ICU for acute respiratory failure caused by influ- infusion rate is 50 units/kg/hour, and the most recent acti-
enza A infection and complex parapneumonic effusions from vated clotting time is 140 seconds. His urine output is 1.2 mL/
methicillin-resistant S. aureus. T.M. is placed on VV-ECMO kg/hour without diuretics.
support. She is currently sedated with fentanyl at 5 mcg/
4. Which one of the following best explains G.V.s clinical
kg/hour and midazolam 0.3 mg/kg/hour, which have been
picture in regard to anticoagulation?
aggressively titrated over the past 24 hours.
A. Increase in hepatic metabolism of heparin
1. Which one of the following best justifies rapidly escalat-
B. Increase in heparin volume of distribution
ing infusions for T.M.?
C. Decreased antithrombin (AT) levels
A. Enhanced renal elimination of fentanyl and D. Decreased clotting factor production
midazolam
5. G.V.s left chest tube starts to ooze blood around the site.
B. Elevated CYP metabolism of fentanyl and
Which one of the following is best to recommend for G.V.?
midazolam
C. Decreased volume of distribution of fentanyl and A. Decrease heparin infusion
midazolam B. Change to bivalirudin infusion
D. Increased adsorption to the circuit of fentanyl and C. Consider AT administration
midazolam D. Add aminocaproic acid infusion

2. Which one of the following would best optimize sedation 6. G.V.s chest tube continues to ooze and is requiring daily
in T.M.? blood transfusions to manage his declining hemoglobin
and hematocrit. No other sources of bleeding have been
A. Discontinue fentanyl and change to morphine
identified (e.g., intracranial, mucosal), but the ECMO
infusion.
circuit has visible fibrin strands developing post-mem-
B. Discontinue midazolam and change to pentobarbital
brane oxygenator. The team is considering administering
infusion.
recombinant activated factor VII. Which one of the fol-
C. Give intravenous bolus fentanyl dose and increase
lowing is best to recommend for G.V.?
fentanyl infusion by 20%.
D. Give intravenous bolus of midazolam and increase A. Consider administration because the patient is not
midazolam infusion by 20%. bleeding from any other source.
B. Consider administration because the blood product
3. A 4-year-old boy (weight 16 kg) is on day 9 of VA-ECMO
requirement is rising.
because of viral myocarditis. He has been on a con-
C. Avoid administration because the optimal dose has
tinuous infusion of opioid and benzodiazepine for the
not been established.
duration and is experiencing profound constipation,
D. Avoid administration because thrombosis risk
despite best efforts with enteral stimulants and osmotic
outweighs the benefit.
agents. The medical team is asking whether subcutane-
ous methylnaltrexone would be a good option for this
Questions 7 and 8 pertain to the following case.
patient to promote a bowel movement. Which one of the
following best justifies avoiding subcutaneous methyln- R.W. is a 16-day-old, full-term girl (weight 3.3 kg) now immedi-
altrexone administration in this patient? ately postoperative from a Norwood procedure. She returned
with her sternum open. R.W. decompensated overnight and
A. Increased volume of distribution in the patient
was placed on VA-ECMO support.
B. Decreased renal elimination of methylnaltrexone
C. Bleeding risk due to systemic anticoagulation 7. The team caring for R.W. wants to continue antibiotic pro-
D. High lipophilicity of methylnaltrexone phylaxis for the open sternotomy cannulation site. Which
one of the following is best to recommend for R.W.?
Questions 46 pertain to the following case. A. Ampicillin
G.V. is a 4-month-old boy (weight 5.5 kg) with known bron- B. Cefazolin
chomalacia, now admitted with respiratory syncytial virus C. Cefepime
bronchiolitis and complicated Streptococcus pneumoniae D. Gentamicin

PedSAP 2016 Book 2 Pediatric Critical Care 77 Extracorporeal Circuits in Children


8. R.W. is now on day 4 of VA-ECMO and continues to have 12. The oncology group wants to initiate R.S.s weekly eto-
intermittent hemodynamic instability. Her WBC rises poside therapy as soon as possible. Which one of the
significantly. Which one of the following is best to rec- following pharmacokinetic concepts is most important
ommend for R.W.? to consider in designing a dosing recommendation for
R.S.?
A. Add fluconazole to current skin flora prophylaxis.
B. Add vancomycin to current skin flora prophylaxis. A. Adsorption of etoposide to CRRT circuit
C. Expand to piperacillin/tazobactam and vancomycin. B. Distribution of etoposide in a critically ill infant
D. Expand to cefuroxime and clindamycin. C. Metabolism of etoposide in a critically ill infant
D. Elimination of etoposide through CRRT and residual
Questions 911 pertain to the following case. renal function
H.L. is a 17-year-old female adolescent (height 65 inches,
13. R.S. is having issues with hypothermia and temperature
weight 102 kg) now 20 days after a bone marrow transplant
control while receiving both ECMO and CRRT. The heat
for refractory acute lymphoblastic leukemia. She is receiving
exchanger on the ECMO circuit is at maximal capacity.
CVVH for acute kidney injury and fluid overload. Her SCr is 0.5
Which one of the following would be best to keep R.S.
mg/dL, and she remains anuric. H.L.s CVVH prescription is
warm?
pre-filter replacement rate 1000 mL/hour, post-filter replace-
ment rate 3000 mL/hour, and ultrafiltration rate 0 mL/hour A. Add external heating blanket.
(net even). The team wants to initiate piperacillin/tazobactam B. Place dialysis bags in warmer.
and vancomycin to rule out sepsis. C. Increase the room temperature.
D. Add blood warmer to CRRT return line.
9. Which one of the following piperacillin/tazobactam regi-
mens is best to recommend for H.L.? 14. R.S. has been stable on vancomycin 90 mg intravenously
every 6 hours for several days for the treatment of coag-
A. 4.5 g intravenously every 8 hours
ulase-negative Staphylococcus bacteremia, with troughs
B. 4.5 g intravenously every 12 hours
within the goal range. This morning, the nephrology team
C. 3.375 g intravenously every 8 hours
is adjusting his CRRT ultrafiltration rate to a net loss of
D. 3.375 g intravenously every 12 hours
10 mL/hour. Which one of the following is best to recom-
10. Which one of the following vancomycin doses, adminis- mend for R.S.s vancomycin therapy?
tered intravenously, is best to recommend for H.L.?
A. Decrease vancomycin interval to every 8 hours.
A. 500 mg B. Increase dose to vancomycin 120 mg intravenously
B. 750 mg every 6 hours.
C. 1000 mg C. Recheck a vancomycin trough before the next
D. 2000 mg dose.
11. Which one of the following is the best time after the first D. No change is necessary.
dose to obtain a vancomycin serum concentration in 15. A 4-year-old boy with acute on chronic renal failure
order to redose vancomycin for H.L.? has been receiving CVVHDF for solute clearance and
A. 6 hours optimization of nutrition for 17 days. Which one of the
B. 12 hours following laboratory values is best to measure in this
C. 24 hours patient?
D. 48 hours A. Copper
B. Thiamine
Questions 1214 pertain to the following case. C. Manganese
R.S. is a 2-month-old boy (length 22 inches [57 cm], weight D. IgG
6kg) with a new diagnosis of hemophagocytic lymphohistio-
cytosis. He is receiving VV-ECMO for acute respiratory failure Questions 16 and 17 pertain to the following case.

and CVVHDF for acute kidney injury and fluid overload. His V.T. is a 16-year-old girl (weight 45 kg) given a diagnosis of
laboratory results include SCr 0.2 mg/dL, BUN 20 mg/dL, total myasthenia gravis 2 months ago. She presented for admission
bilirubin 1.2 mg/dL, and direct bilirubin 0.8 mg/dL. R.S.s urine to the pediatric ICU 5 days ago with progressing weakness
output is 220 mL in 24 hours. His CVVHDF prescription is as that necessitated mechanical ventilation support. Yesterday,
follows: dialysis rate 800 mL/hour, pre-filter fluid replacement V.T. completed her last dose of intravenous immunoglobulin.
100 mL/hour, post-filter fluid replacement 50 mL/hour, and She has made little progress, so V.T.s team is considering ini-
ultra-filtration rate 0 mL/hour. tiating plasmapheresis today.

PedSAP 2016 Book 2 Pediatric Critical Care 78 Extracorporeal Circuits in Children


16. The neurology team would like to know whether V.T.s pyr- Which one of the following is best to recommend for
idostigmine therapy will be affected by plasmapheresis. B.T.s dosing adjustment?
Which one of the following best answers this question
A. Make no change in recommended dose.
from V.T.s team?
B. Decrease by 25% of recommended dose.
A. No protein binding will allow for elimination by C. Decrease by 50% of recommended dose.
plasmapheresis. D. Avoid giving rituximab while patient is receiving
B. Hepatic metabolism precludes plasmapheresis CRRT.
elimination.
19. B.T.s serum calcium has increased to 13.4 mg/dL, but his
C. Volume of distribution of 0.5 L/kg is too large for
ionized serum calcium is low at 3.4 mg/dL. He remains
elimination.
on trisodium citrate regional anticoagulation and is
D. Poor oral absorption inhibits plasmapheresis
receiving a calcium chloride infusion. Which one of the
elimination.
following best explains the reason for these abnormali-
17. Which one of the following laboratory values is best to ties in B.T.s calcium?
obtain after V.T. completes her multiday plasmapheresis
A. Excessive infusion rate of calcium chloride
course?
B. Underdosing of trisodium citrate infusion
A. Platelets C. Delayed citrate clearance by the liver
B. IgG D. Underdosing of calcium chloride infusion
C. Albumin
20. A 6-month-old girl with hypoplastic left heart syndrome
D. Hepatic enzymes
presents after hemi-Fontan surgery. She remains on VA-
ECMO postoperatively, cannulated through her sternum.
Questions 18 and 19 pertain to the following case. Nutrition is to be initiated in the form of 3-in-1 total par-
B.T. is a 12-year-old boy (height 63 inches [160 cm], weight enteral nutrition. Which one of the following is the best
61 kg) with newly diagnosed Burkitt lymphoma. He is now access point for this patients administration of intrave-
receiving CVVHDF for tumor lysis syndrome. B.T. is receiving nous nutrition?
regional citrate anticoagulation. He remains anuric, and his
A. Internal jugular venous catheter
SCr is 0.4 mg/dL. Current CRRT orders are as follows: dialy-
B. Peripheral venous catheter
sis rate 2000 mL/hour, pre-filter rate 2000 mL/hour, post-filter
C. Post-roller pump stopcock
rate 1000 mL/hour, and ultrafiltration rate 0 mL/hour.
D. Pre-membrane oxygenator stopcock
18. The oncology team wants to continue with B.T.s treat-
ment and scheduled chemotherapy next with rituximab.
Learner Chapter Evaluation: Extracorporeal Circuits in Children.

As you take the posttest for this chapter, also evaluate the Use the 5-point scale to indicate whether this chapter pre-
materials quality and usefulness, as well as the achievement pared you to accomplish the following learning objectives:
of learning objectives. Rate each item using this 5-point scale:
12. Account for the basic mechanics and components of
Strongly agree extracorporeal therapies.
Agree 13. Develop a drug therapy regimen based on pharmacoki-
Neutral netic changes in extracorporeal therapies.
Disagree 14. Design a therapeutic monitoring plan for patients receiv-
Strongly disagree ing extracorporeal therapies.
15. Evaluate an anticoagulation strategy for patients receiv-
1. The content of the chapter met my educational needs. ing extracorporeal therapies.
2. The content of the chapter satisfied my expectations. 16. Assess the attainment of therapeutic end points for
3. The author presented the chapter content effectively. patients receiving extracorporeal therapies.

4. The content of the chapter was relevant to my practice 17. Design a plan to treat common complications associ-
and presented at the appropriate depth and scope. ated with extracorporeal therapies.

5. The content of the chapter was objective and balanced. 18. Please provide any specific comments related to any
perceptions of bias, promotion, or advertisement of
6. The content of the chapter is free of bias, promotion, or commercial products.
advertisement of commercial products.
19. Please expand on any of your above responses, and/or
7. The content of the chapter was useful to me. provide any additional comments regarding this chapter:
8. The teaching and learning methods used in the chapter
were effective.
9. The active learning methods used in the chapter were
effective.
10. The learning assessment activities used in the chapter
were effective.
11. The chapter was effective overall.

PedSAP 2016 Book 2 Pediatric Critical Care 80 Extracorporeal Circuits in Children


Trauma
By Joseph M. LaRochelle, Pharm.D., BCPPS

Reviewed by Sara P. Rooney, Pharm.D., BCPS, BCPPS; and Andrea Joan Stumpe, Pharm.D., BCPS, BCPPS

LEARNING OBJECTIVES

1. Develop strategies to optimize a patients mean arterial pressure.


2. Assess strategies to decrease or prevent increased intracranial pressure.
3. Demonstrate appropriate sedation management strategies, as well as appropriate glucose control and seizure
prophylaxis, for the patient with traumatic brain injury.
4. Design appropriate fluid management and treatment approach to increase lean body mass in a patient with burns.
5. Compose strategies to optimize pain control and treat infections in a patient with burns.

INTRODUCTION TO TRAUMATIC
ABBREVIATIONS IN THIS CHAPTER
BRAIN INJURY
CPP Cerebral perfusion pressure
One of four pediatric patients will have a traumatic injury, which is the
GCS Glasgow Coma Scale
leading cause of death in children (Osterman 2015). Traumatic brain
ICP Intracranial pressure
injury (TBI) accounts for 500,000 incidents, 35,000 hospitalizations,
MAP Mean arterial pressure
and 2000 deaths each year (Faul 2010). Severe TBI is usually defined
TBI Traumatic brain injury
as a Glasgow Coma Scale (GCS) score less than 8 or the need for
TBSA Total body surface area
mechanical ventilation (Piatt 2012; Stanley 2012); this type of injury
Table of other common abbreviations. is the focus of this chapter.
In a retrospective study of 236 pediatric patients with severe TBI,
the average age was 8 years, most patients were boys, and abu-
sive head trauma was the most common injury (Vavilala 2014). The
pathophysiology of TBI is complex and includes both primary and
secondary injuries. In the primary injury, rapid acceleration, decelera-
tion, and/or rotational forces move the brain against the cranium, or
a direct insult to the brain (e.g., penetrating trauma) can produce the
injury. This causes hemorrhage, hematoma, or diffuse axonal injury
(Greve 2009). Diffuse axonal injury is a global phenomenon that can
lead to poor neurologic outcomes (Bhalla 2012). Secondary injury
occurs because of an increase in tumor necrosis factor alpha (TNF-)
and interleukin (IL)-1, causing inflammation, neuronal death, and
edema. Glutamate and other excitatory neurotransmitters increase
calcium influx into the cell through N-methyl-d-aspartate (NMDA)
receptors, releasing oxygen-free radicals and nitric oxide, leading to
lipid peroxidation and cell death (Greve 2009).

CEREBRAL PERFUSION PRESSURE


In TBI, the focus of treatment should be on increasing cerebral perfu-
sion pressure (CPP). Cerebral perfusion pressure is the mean arterial
pressure (MAP) minus the intracranial pressure (ICP). Therefore, the

PedSAP 2016 Book 2 Pediatric Critical Care 81 Trauma


perfusion into the brain depends on blood pressure and the Hyperosmolar Agents
force that the head is pushing back on the blood flow. Hypertonic Saline
Increases in ICP cause a decrease in the CPP, resulting in fur-
Mean Arterial Pressure ther injury to brain tissue as a result of secondary injury. An
Much controversy exists regarding the optimal MAP for pedi- ICP of 20 mm Hg or greater for 5 minutes or more will require
atric patients with TBI. The consensus is to maintain a MAP active interventions to decrease the pressure and secondary
at a level about normal for age, with a minimum goal of 4050 injury. A cutoff of 15 mm Hg or greater can be used for infants
mm Hg. Adult studies avoid MAP goals greater than 70 mm and small children for ICP intervention because of their lower
Hg. Hypotension should be avoided because poor neuro- physiologic MAP (Kochanek 2012).
logic outcomes are associated with hypotensive episodes. Hypertonic saline (3%) is the preferred agent in pediatric
As in other aspects of critical care, hypotension should ini- TBI to lower ICP; it changes the osmolar gradient, cell volume,
tially be managed with fluid resuscitation. The fluid of choice and cell membrane resting potential, leading to its positive
for fluid resuscitation in pediatric TBI is 0.9% sodium chlo- therapeutic benefit of lowering ICP. In addition, hypertonic
ride because other fluids are less osmolar or lack the needed saline may release atrial natriuretic peptide, enhance car-
sodium content (Kochanek 2012). diac output, and decrease inflammation (Kochanek 2012). A
After appropriate fluid resuscitation, vasoactive agents bolus dose of 6.510 mL/kg, followed by a continuous infu-
may be needed to increase MAP. Although no specific vaso- sion of 0.11 mL/kg/hour, should be initiated and titrated to
pressor has proved superior in pediatric TBI, dopamine, a maximum serum osmolarity of 360 mOsm/L and a sodium
phenylephrine, and norepinephrine have been studied. concentration of 170180 mEq/L (LaRochelle 2015; Kochanek
Although all positively affect MAP, phenylephrine and nor- 2012). Central pontine myelinolysis, hyperchloremic meta-
epinephrine have larger and more sustained effects because bolic acidosis, and renal insufficiency can occur with sodium
of their greater selectivity for 1-adrenergic receptors chloride therapy.
(Sookplung 2011). In some animal models of TBI, vasopressin Although 3% saline is the mainstay of TBI therapy, evi-
can alter the blood-brain barrier and increase cerebral edema dence is emerging to support higher concentrations in
by activating vasopressin receptor 1a (Zink 2010). However, the management of increased ICP. A 2012 case report
because clinical data are lacking to support its use and alter- describes two cases of successful use of 23.4% saline. In
native agents are available, vasopressin should be avoided in the first case, a 23-month-old child (weight 12 kg) had an
pediatric TBI. initial GCS score of 3 after sustaining a closed-head injury.
A transthoracic echocardiogram revealed left systolic dys-
function with an estimated ejection fraction of 10%15%.
Because of elevated ICP (44 mm Hg), the patient was given
six boluses of 3% saline 50 mL, a norepinephrine contin-
BASELINE KNOWLEDGE STATEMENTS uous infusion to maintain MAP, two boluses of mannitol,
sedation, paralytics, phenobarbital, CSF drain, and bilateral
Readers of this chapter are presumed to be familiar
decompressive craniectomies. Because of the concern for
with the following:
fluid overload together with decreased myocardial func-
General knowledge of general critical care manage-
tion, the 3% saline was changed to 23.4% saline; the patient
ment of patients
received 15 boluses of 23.4% saline (10 mL each) over 7
Basic drug knowledge of sedation strategies
days. After each dose of 23.4% saline, the ICP decreased
Pharmacology of vasopressor agents
more (7.9 mm Hg average) than with the 3% doses. No
Table of common pediatric laboratory reference values. adverse effects were observed with the 23.4% saline, and
the maximal serum sodium reached was 170 mEq/L. In the
second case, a 4-year-old child (weight 14 kg) had a two-
ADDITIONAL READINGS
story fall. Initially, her ICP was managed by sedation and a
The following free resources have additional back- craniotomy for a skull fracture. The patients ICP increased
ground information on this topic: to 44 mm Hg, and she was given 12 boluses of 23.4% saline
Kochanek PM, Carney N, Ashwal S, et al. Guidelines (810 mL) over 9 days. The only strength of hypertonic
for the acute medical management of severe saline used was 23.4%. Similar to the first case, significant
traumatic brain injury in infants, children, and decreases in ICP occurred (up to 19 mm Hg), with a maxi-
adolescents-second edition. Pediatr Crit Care Med
mum sodium concentration of 146 mEq/L, with no adverse
2012;13:S1-S82.
effects detected (Nakagawa 2012). The 23.4% saline solu-
Jeschke MG, Herndon DN. Burns in children:
tion used in the two cases was administered through a
standard and new treatments. Lancet
2014;383:1168-78. central venous catheter; if 23.4% saline is used, peripheral
intravenous administration is not advised.

PedSAP 2016 Book 2 Pediatric Critical Care 82 Trauma


A retrospective cohort study in Australia evaluated 23.4% proper sedation will be required, as in most intubated criti-
saline administration in 32 pediatric patients with severe TBI cal care patients. The effect of the sedative agents used can
over 8 years (20032011). The standard hospital protocol was decrease ICP through overall metabolic suppression, blunt
0.5 mL/kg to a maximum of 30 mL given over 10 minutes. The the response to painful stimuli and procedures, and have anti-
hospital protocol for increased ICP used similar treatment epileptic properties (Kochanek 2012). Sedation in critical care
modalities of standard care, including sedation, neuromuscu- is overall a controversial topic, given that a consensus on the
lar blocking agents, normothermia, and inotropic support for optimal agents and protocols has yet to be shown. In gen-
hypotension. Three patients received only one dose of 23.4% eral, patients with TBI will require pain control with an opioid
saline; however, most received several doses (up to 28 in one analgesic such as a fentanyl continuous infusion starting at 1
patient) because of the inability to maintain a hypernatremic mcg/kg/hour and an anxiolytic such as a midazolam continu-
state. The average ICP decrease was 10.2 mm Hg, and mean ous infusion starting at 0.1 mg/kg/hour (Taketomo 2015). All
sodium was 139 mEq/L with a maximum of 161 mEq/L. No of the standard agents used for sedation can cause hypoten-
major adverse effects, including renal failure, were noted. sion, which will decrease CPP. In hemodynamically unstable
Seventy-four percent of the patients had outcomes that patients, adequate blood pressure should be ensured before
would allow them to have independent functioning, and the initiating these agents, either using a fluid bolus or initiating
mortality rate was low (6%) (Piper 2015). a vasopressor agent until the MAP increases.
Although data for using 23.4% saline are limited to retro- Use of sedatives such as ketamine, dexmedetomidine,
spective studies and case reports, it may be considered in and propofol as adjunctive agents has been described in TBI.
pediatric patients with TBI. This modality may be most ben- Ketamine works by blocking glutamate, one of the excitatory
eficial in patients with fluid overload or decreased cardiac neurotransmitters released in TBI, through antagonizing the
function. Further randomized prospective trials are needed to NMDA receptor, which leads to a decrease in ICP (Bar-Joseph
establish its role in pediatric patients. 2009). In addition, ketamine releases catecholamines, which
can help increase MAP, a critical determinant in CPP. Use
Mannitol
of ketamine in TBI has been met with resistance because
Mannitol has been used for years to reduce ICP in TBI. It of 1970s reports indicating its potential to increase ICP.
decreases blood viscosity and maintains blood flow in low blood The mechanisms behind this concern are that an increase
volume states. This mechanism of action takes effect rapidly in Pco2 in nonventilated patients leads to vasodilation and
and lasts about 1 hour (Kochanek 2012). Its second mechanism ketamines ability to block nitric oxide synthase, which can
of action is that of an osmolar agent, which changes the gradi- increase oxygen extraction in the brain. In a recent review,
ent decreasing ICP. This mechanism of action has an onset of seven articles evaluated ketamine in severe TBI in 55 pediatric
about 15 minutes but lasts for up to 6 hours. The dose is 0.25 patients and 101 adult patients. Four studies used continu-
0.5 g/kg intravenously every 6 hours; however, doses of 1 g/kg ous infusion ketamine as sedation, with no increases in ICP
can be used clinically (Taketomo 2015). In light of increasing in these patients. In the three studies that used bolus dosing
use of hypertonic saline, the use of mannitol is falling out of (two pediatric studies), none showed increases in ICP, and
favor because of its propensity to accumulate in injured sec- there were even some decreases in ICP (Zeiler 2014). From
tions of the brain. This causes a reverse osmotic gradient that the limited data, the initial concern surrounding ketamine
can lead to an increase in ICP (Kochanek 2012). use in TBI appears not to apply to mechanically ventilated
patients. In pediatric patients, ketamine in severe TBI could
Barbiturates be considered for procedural sedation as a bolus dose. Until
Barbiturates lower ICP by changing vascular tone, causing met- reports of continuous infusion ketamine in pediatric patients
abolic suppression and decreasing oxygen free radicals, which are available, it should be avoided for continuous sedation.
will decrease lipid peroxidation. Therapy, usually limited by Randomized control trials are needed to establish ketamines
hypotension, should only be used in hemodynamically stable role in pediatric severe TBI.
patients after other treatment modalities have failed (Kochanek Propofol is a general anesthetic agent that works through
2012). Although thiopental has been studied, pentobarbital is -aminobutyric acid activation and blocking NMDA recep-
used most commonly. Bolus dosing of 510 mg/kg followed by tors (Taketomo 2015). Because of its short half-life, propofol
a continuous infusion of 12 mg/kg/hour should be used to tar- is used in TBI to assist in the neurologic examination. Its
get burst suppression on electroencephalogram and not dosed use, however, is limited to less than 24 hours because of
to a particular drug concentration (Taketomo 2015). the increased risk of propofol-related infusion syndrome
(Sabsovich 2007).
SEDATION Dexmedetomidine is a centrally acting 2-agonist used for
Because of the extensive injuries and cerebral dysfunc- sedation in various critical care patients, including those with
tion associated with severe TBI, the patient will probably TBI. It also activates the extracellular signal-regulated protein
be intubated and placed on mechanical ventilation. Thus, kinase 1 and 2 pathways, which may have neuroprotective

PedSAP 2016 Book 2 Pediatric Critical Care 83 Trauma


effects (Schoeler 2012). Dexmedetomidine can cause hypo- potentially leading to poor neurologic outcomes (Kochanek
tension through decreased conduction in the AV node and 2012). Hypoglycemia in children is detrimental because of
should be used with caution because of the potential decrease the risk of decreased neurologic functioning (Wayne 1990).
in MAP (Buck 2010). This agent is dosed as a bolus of 0.51 In a study of 57 children, hyperglycemia 48 hours after injury
mcg/kg, followed by a continuous infusion of 0.20.7 mcg/ was associated with poor outcomes, especially when blood
kg/hour, with higher doses used clinically (Taketomo 2015). glucose concentrations were greater than 200 mg/dL (Smith
Etomidate is a short-acting benzylimidazole with anes- 2012). In a retrospective study of 271 mechanically ven-
thetic properties (Taketomo 2015). It has been used in tilated patients with TBI in which glucose measurements
single-dose administration in patients with TBI with a reduc- were obtained, patients were stratified into mild, moderate,
tion in ICP (Bramwell 2006). The TBI guidelines recommend and severe hyperglycemia categories. Patients in the severe
against its global use for sedation in TBI; however, it may be hyperglycemia group (greater than 200 mg/dL) had a signif-
considered an option (Kochanek 2012). Etomidate causes icantly greater risk of death and poorer outcomes than the
adrenal suppression, so caution must be used. In addition, other two blood glucose groups. The severe hyperglycemia
it contains propylene glycol, which could lead to toxicity with group had a 3.5-fold increase in poor outcomes over the mild
prolonged infusion. hyperglycemia group (110160 mg/dL) (Elkon 2014). From the
After effective and adequate sedation has been achieved, limited available data, it appears that glucose should be kept
another option to help decrease ICP is the use of neuromus- in a normal range of 80180 mg/dL while avoiding both hypo-
cular blocking agents. They decrease intrathoracic pressure, glycemia (less than 60 mg/dL) and hyperglycemia (greater
which is likely increased secondary to mechanical ventila- than 200 mg/dL).
tion. This decrease in pressure allows for an increase in brain
venous outflow and a decrease in ICP. These agents paralyze Seizure Prophylaxis
the skeletal muscle, which can decrease metabolic oxygen About 10%20% of pediatric patients with TBI have an early
demand, decreasing ICP (Kochanek 2012). Succinylcholine post-TBI seizure (Arango 2012; Liesemer 2011). Loss of
is the only neuromuscular blocking that can increase ICP; it consciousness at the scene, focal neurologic deficits, hema-
should be used only if other agents are not available. toma, bone or metal fragments, penetrating injury, GSC less
than 10, and age younger than 2 years are risk factors for sei-
OTHER THERAPIES zures in TBI (Liesemer 2011). Routine antiseizure prophylaxis
in patients with TBI is under debate. In a multicenter study
Hypothermia
of 477 children, those who received an antiseizure drug had
Hypothermia decreases metabolic oxygen demand, excitotox- decreased mortality (Tilford 2001). In contrast, another study
icity, inflammation, cell death, and seizures (Kochanek 2012). of 300 patients showed no difference (Liesemer 2011).
Although studies of pediatric patients with TBI have been According to the available evidence, it seems prudent to
somewhat inconclusive, most show no improved outcomes use prophylactic agents for seizures if the patient has risk
when therapeutic hypothermia strategies are used. A retro- factors because of the injury. Phenytoin is most commonly
spective study of 190 pediatric patients showed an increased used for seizure prophylaxis. A loading dose of 1520 mg/
mortality when hypothermia was present at admission kg, followed by 5 mg/kg/day divided every 812 hours, of phe-
(Sundberg 2011). In a phase III multicenter trial of hypo- nytoin or fosphenytoin is recommended (Taketomo 2015).
thermia in TBI, an interim analysis showed futility, and the Pharmacokinetic parameters and especially albumin bind-
study was terminated early (Adelson 2012). In a multicenter ing are altered in critical care and especially in patients with
study conducted in Australia, New Zealand, and Canada, 50 TBI. It is recommended to obtain free phenytoin concentra-
patients with severe TBI were randomized to hypothermia tions when initiating therapy and every 23 days thereafter,
(32C33C) or normothermia. This study, which followed the with a goal of 12 mcg/mL. During a hypothermia trial, inves-
children 12 months postinjury, showed no difference in length tigators found that phenytoin elimination was decreased by
of stay, mortality, or pediatric cerebral performance category about half during cooling protocol procedures, necessitating
score (Beca 2015). This further adds to the evidence that a phenytoin dose reduction (Empey 2013).
therapeutic hypothermia should be avoided in pediatric TBI. Another, newer agent for TBI seizure prophylaxis that
Hyperthermia, however, should also be avoided and treated has gained increased use in pediatric patients is levetirace-
aggressively because of its potential to increase ICP. tam (Jones 2008). A pilot phase II study evaluated the use
of levetiracetam in preventing late posttraumatic epilepsy.
Glycemic Control The patients (618 years of age) were administered 55 mg/
The debate over optimal glucose control in the adult and pedi- kg/day divided twice daily orally (intravenously if NPO [noth-
atric critical care populations is ongoing (Brunkhorst 2008; ing by mouth]) within the first 8 hours of injury and until day
Yung 2008; van den Berghe 2001). Hyperglycemia can pro- 30 postinjury. Twenty patients were included in the treatment
duce oxygen free radicals and metabolic acidosis in the brain, group and were matched to 20 control patients. Headache,

PedSAP 2016 Book 2 Pediatric Critical Care 84 Trauma


fatigue, and somnolence were common adverse effects in Cytokines, histamine, prostaglandins, substance P, and oxy-
the levetiracetam-treated patients. A patient in the levetirace- gen free radicals are released, causing systemic vasodilation
tam group had a seizure, the only patient in the 40-sample and regional vasoconstriction (Pham 2008; Scott 2005). In
size. Although this study included patients with a GCS score severe burns (greater than 20% TBSA), catecholamines and
greater than 6, levetiracetam appears to be safe to use in cortisol are released, resulting in an increased metabolic
pediatric patients with TBI (Pearl 2013). state. This can subsequently cause hyperglycemia and lipol-
ysis in addition to muscle wasting (Jeschke 2008).
EMERGING THERAPIES Treatment occurs in three phases. The first phase, initial
care, occurs at the scene and hospital and consists of rapid
Bone marrow mononuclear cells can change regional inflam-
assessment, resuscitation, and admission to a burn center.
mation, enhance angiogenesis, and reduce growth factor
The second phase occurs while the child is in the hospital and
(Walker 2010; Qu 2007). In a phase I trial of 10 children, organ
consists of infection control, surgical procedures, and main-
function returned after autologous bone marrow cells were
taining adequate organ function. The last phase occurs after
administered (Cox 2011). In a follow-up study, these 10 patients
discharge and consists of reconstruction and rehabilitation.
were compared with an age- and severity-matched control
group. The treated patients had a significantly decreased
intensity level of therapy score, logistic organ dysfunction
INITIAL MANAGEMENT
score, and duration of ICP monitoring (Liao 2015). Although Initial therapy goals focus on salvaging limbs, organs, and life.
this was done in only 10 patients, the results seem promising Initial fluid resuscitation should be initiated according to the
and may be a treatment option for patients with severe TBI. Parkland formula, which is about 4 mL/kg/%TBSA burned of
Progesterone can reduce cerebral edema, inflammation, crystalloid over the first 24 hours (Jeschke 2014b). However,
oxidative damage, excitation, and apoptosis. It up-regulates in children, this may be an underestimate, and up to 6 mL/
P-glycoprotein, which increases the efflux pumps in the blood- kg/%TBSA burned can be used to prevent burn shock (Pham
brain barrier, thus decreasing cerebral edema. Aquaporin 4 2008). The general strategy follows that of typical dehydration
is a water membrane channel that becomes up-regulated management, in which half of the fluid is given over the first
in injured sections of the brain. Progesterone can decrease 8 hours and the second half is given over the next 16 hours.
the amount of Aquaporin 4, which leads to decreased cere- Consequences of fluid under resuscitation include death and
bral edema. Progesterone decreases inflammation through multiorgan system dysfunction. Administration of excessive
decreasing proinflammatory cytokines (TNF, IL-1, and IL-6), fluids increases the risk of respiratory distress syndrome,
an important aspect in the neuroinflammation that occurs pneumonia, multiple organ dysfunction, and death, neces-
after TBI. In addition, it inhibits nitric oxide synthase, which sitating an adequate but not excessive fluid administration.
reduces the production of nitric oxide. It also up-regulates Urine output and MAP should be monitored for successful
superoxide dismutase, which reduces lipid peroxidation. In resuscitation with goals of greater than 0.51 mL/kg/hour
studies of adult patients, decreased mortality and less dis- and greater than 65 mm Hg, respectively (Jeschke 2014b). No
ability 30 days postinjury occurred in patients who received large trials assessing the optimal fluid in these populations
progesterone. One large trial of adults with TBI who were have been conducted, but lactated Ringer solution is used
treated with progesterone was terminated because of a lack most often (Jeschke 2014b). In patients with 10%20% TBSA
of better outcomes. Other large multicenter trials evaluat- burned (moderate burns), lower fluid resuscitation volumes
ing the role of progesterone in adults with TBI are under way. may be considered. A study of 88 patients showed that 2 mL/
Because there are developmental changes in the brain and its kg/%TBSA resulted in a shorter length of stay (Walker 2014).
response to progesterone, this may become an option in pedi- Until further data are available, aggressive and adequate
atric TBI (Robertson 2015). fluid resuscitation up to 6 mL/kg/%TBSA should be used in
severely burned (greater than 20% TBSA) pediatric patients.
Inhalation injury from fires confers significant mortality
INTRODUCTION TO BURNS
because of pulmonary edema and ventilation-perfusion mis-
Burns account for 400,000 injuries each year in the United match with subsequent respiratory failure (Jeschke 2014b).
States, with 4000 deaths (WHO 2015; Jeschke 2014b). Burns If needed, mechanical ventilation should be used to support
over a greater than 30% total body surface area (TBSA) con- oxygen delivery, as well as standard ventilator strategies and
stitute a risk factor for mortality in children. Causes of death medications.
from burn injury are usually sepsis, multiorgan system failure,
and anoxic brain injury (Jeschke 2014b).
HYPERMETABOLIC STATE
The pathophysiology of burns involves a complex cas-
cade that occurs not only in the burned tissue but also in the Glycemic Control
healthy tissue. Changes in ATPase activity increase sodium The hypermetabolic state that occurs because of burn injury
inside the cell. This leads to cellular edema and hypervolemia. results in hyperglycemia and the inability to use lipids and

PedSAP 2016 Book 2 Pediatric Critical Care 85 Trauma


amino acids. This extensive catabolism leads to protein have reported increased overall muscle mass, increased
breakdown within the body and can lead to poor outcomes, wound healing, and decreased hospital stay (Jeschke 2014b).
including death. The hyperglycemia that results is an inde- A study of 12 severe burn patients showed increased muscle
pendent risk factor for sepsis and mortality. proteins in those who received oxandrolone 0.1 mg/kg twice
In a small study of 29 patients, those who maintained a daily for 6 months (Tuvdendorj 2011). In a larger and longer
tight glucose control (80110 mg/dL) had improved muscle study of 70 patients who were given oxandrolone for 1 year,
oxidative capacity and less resting energy expenditure than there was a significant decrease in the hypermetabolic state
those whose glucose was maintained at less than 215 mg/dL compared with controls (Porro 2012). Although the data are
(Fram 2010). In a larger study of 239 patients, those whose limited and the long-term survival effects are unknown, use of
serum glucose was maintained in the 80110 mg/dL range oxandrolone may help increase lean muscle mass.
had lower mortality than those whose serum glucose was Studies have shown that growth hormone decreases
maintained in the 140180 mg/dL range (Jeschke 2010). In catabolism and increases regeneration of the skin through
a study of 106 severely burned children, investigators found cellular mechanisms (Branski 2009; Pereira 2005). In a
that children with glucose concentrations greater than 150 study of 86 children treated with growth hormone, a slightly
mg/dL had a higher incidence of sepsis and longer duration shorter healing time in skin grafts occurred, but no difference
of mechanical ventilation because of bacterial pneumonia in major outcomes of hospital stay or mortality was found
than those with serum glucose concentrations below 150 (Breederveld 2012). Other studies have shown no difference;
mg/dL (Kraft 2014). thus, growth hormone is usually not used in the treatment of
A study of 660 severely burned patients investigated burn patients (Jeschke 2014b).
episodes of hypoglycemia on several clinical end points.
Of these, 568 patients were never hypoglycemic, and 198 Bisphosphonates
patients had hypoglycemia (less than 60 mg/dL). A signifi- Pediatric burn patients can be afflicted with resorptive bone
cantly higher percentage of patients in the hypoglycemia loss because of the increase in glucocorticoids second-
group died, developed sepsis, and had longer hospital stays ary to the stress response and overall inflammatory process
(Jeschke 2014a). The ideal glucose value in critically ill (Borsheim 2014). Pamidronate is a bisphosphonate that
burned patients appears to be elusive, but both hypoglycemia inhibits bone resorption and decreases osteoclast activity
and hyperglycemia should be avoided. Euglycemia should be (Taketomo 2015). In a study of 17 patients, 10 received pami-
targeted with a serum glucose of 80130 mg/dL. dronate and 7 received placebo. The authors found that the
control group had a greater muscle breakdown rate and a nega-
Nutritional Support tive nitrogen balance, both of which were significantly different
Adequate and early nutritional support is essential for the from the pamidronate group (Borsheim 2014). Further studies
critical care patient (Mehta 2009). In a study of 688 pediat- are needed to determine their true role in pediatric burns, but
ric burn patients, those who received early enteral therapy bisphosphonates may be a future option in these patients.
had decreased mortality (Khorasani 2010). In pediatric burn
patients, no specific enteral or parenteral formulation has -Blockers

proved more successful than another. Experts recommend During the hypermetabolic response, catecholamines are
high-protein, high-glucose, and low-fat formulations (Jeschke released; this, in turn, causes increases in heart rate and blood
2014b). In a trial of 27 severely burned pediatric patients using pressure. Propranolol, a nonselective -blocker, decreases
amino acid supplementations with Travasol and phenylala- both the hypermetabolic and the hyperglycemic response
nine, patients were followed to discharge, 6 months postburn, and increases lean body mass. In a study of 174 patients who
and 1 year postburn. No difference occurred in skeletal mus- received 4 mg/kg/day for 1 year, patients had lower heart rate,
cle fractional synthesis rate, showing that additional amino lower resting energy expenditure, and increased lean muscle
acid supplementation did not improve muscle protein syn- mass (Herndon 2012). Another larger study of 340 patients
thesis (Porter 2013). In addition, trace elements and vitamins had similar findings with propranolol doses up to 6 mg/kg/
are usually decreased in burn patients and should be supple- day (Williams 2011). Propranolol can be considered in pediat-
mented (Jeschke 2014b). ric burn patients, especially if there is an extensive metabolic
response. Further trials should help determine the long-term
Hormones outcomes of propranolol use in pediatric burn patients.
The overall catabolic state that results from severe burns
causes protein and muscle wasting. Oxandrolone is a hormone INFECTIONS
medication with a structure similar to testosterone; taken Because of the extent of injuries, the severely burned pediat-
orally, it can enhance protein synthesis in skeletal muscle ric patient is at increased risk of infections, including sepsis.
through its action as a synthetic androgen receptor hormone Prompt recognition and adequate treatment are essential for
(Hart 2011; Tuvdendorj 2011). Studies using oxandrolone patient survival. Infection is generally a late complication in

PedSAP 2016 Book 2 Pediatric Critical Care 86 Trauma


burn patients, usually occurring after 72 hours postinjury. benzodiazepines with adjunctive use of ketamine, NSAIDs,
Fever in burn patients can occur because of the inflamma- dexmedetomidine, and gabapentin if neuropathic pain
tory processes associated with the burn or because of an persists.
infection. In a retrospective analysis of 353 patients admit- In one study of 500 burn patients; almost all patients
ted for a burn injury (all severity levels), only 30.6% had a received an opioid, 75% received a benzodiazepine; and
fever. In general, patients with a fever had higher TBSA burns, about 50% received propofol or ketamine (Stoddard 2011).
more hospital days, and higher mortality. In the patients with A retrospective study of 147 patients observed the effects
a fever, almost half had a bacterial infection. The common of opioids administered early in burn treatment. Opioid dos-
organisms included Escherichia coli, Staphylococcus aureus, ing within the first 7 days of injury correlated with decreased
and Enterococcus faecium. All patients with a fever should be Child Stress Disorders Checklist scores, signifying less post-
evaluated, but it appears that greater TBSA burned, as well as traumatic stress disorder. The authors also found that the
the presence of indwelling catheters, are risks for bacterial effect was long lasting (up to 4 years after discharge) and
infections, including sepsis (Vyles 2014). dose related (Sheridan 2014). In a study that evaluated dex-
In another retrospective analysis of 92 ventilated burn medetomidine (average dose 0.57 mcg/kg/hour for 40 hours)
patients admitted to a single burn center, about 40% had a as a sedative in 11 mechanically ventilated severely burned
ventilator-associated pneumonia, which correlated with a patients, the patients had better sedation and fewer episodes
statistically significantly longer hospital stay. Organisms of hypotension (Lin 2011).
responsible for the pneumonia included Streptococcus pneu- In a study of 8 severely burned patients, the children were
moniae, S. aureus, Pseudomonas aeruginosa, and Acinetobacter given escalating doses of morphine (average 0.45 mg/kg/
baumannii. Half of the deaths in the cohort were attributed hour) and midazolam (average 0.44 mg/kg/hour) until dexme-
to pneumonia (Rogers 2014). Similar to other critically ill detomidine was added at a bolus of 1 mcg/kg over 10 minutes
patients, early recognition and treatment of infections are followed by a continuous infusion of 0.752.5 mcg/kg/hour.
important for ventilated burn patients. Three of the patients had a drop in MAP below 50 mm Hg;
Secondary to their hypermetabolic state and need for thus, the continuous infusion was held. In general, the aver-
aggressive fluid management, burn patients may have age decrease in MAP was 37%, and heart rate decreased
altered pharmacokinetics. In 70 patients administered ami- by almost 20%. During burn injury, there is a hypermeta-
kacin together with piperacillin/tazobactam and vancomycin bolic state with increased catecholamines, which can lead
for presumed or actual sepsis, a pharmacokinetic model was to hypertension. Administration of dexmedetomidine can
used to evaluate standard dosing of amikacin applied to the decrease this, but a significant drop in blood pressure also
burn patients. Clearance of amikacin increased, but peak can be observed (Shank 2013). According to this small study,
concentrations remained similar to those of other critically ill bolus dosing likely should not be used or given over 30 min-
children. Given the mechanism of action of aminoglycosides, utes in burn patients.
this increased clearance may not affect the efficacy of the Studies evaluating the proper pain management and
antibiotic in burn patients (Sherwin 2014). sedation strategies in burn patients have been inadequate,
In a small study evaluating vancomycin pharmacokinetics but opioids and benzodiazepines should be the mainstay of
in 13 severe burn patients, patients were administered a 10- to therapy, with opioids administered early to these children.
15-mg/kg/dose four times a day infused over 1 hour. On ini- Dexmedetomidine can be used as an adjunct or agent for
tial dosing, a target vancomycin trough plasma concentration sedation in patients with severe burns, using normal hospital
greater than 10 mcg/mL was achieved in only 15% of patients. sedation protocols.
An increase in the daily dose to an average of 98 mg/kg/day
produced an average trough concentration of 13 mcg/mL. CONCLUSION
With this vancomycin dose, an AUC/MIC ratio greater than Treatment of TBI in the pediatric patient is complex and con-
400 was achieved in 93% of patients, assuming an S. aureus sists of various treatment modalities. Initial management of
MIC of 0.5 mg/L (Gomez 2013). Given these results from this MAP includes aggressive fluid resuscitation with 0.9% saline
small number of patients, together with the dosage strate- and vasopressors. Management of elevated ICP includes
gies used in critical care in general, doses of 80100 mg/kg/ hypertonic saline, adequate sedation with benzodiazepines,
day divided every 6 hours should be used for vancomycin in and opioids with adjunctive sedative agents as needed.
pediatric burn patients to achieve optimal vancomycin trough Neuromuscular blockage, seizure prophylaxis, normothermia,
targets of 1520 mcg/mL. and euglycemia should also be used in the patient with TBI.
Burn pathophysiology is complex with a generalized hyper-
PAIN MANAGEMENT metabolic state. Early and adequate fluid resuscitation with
The pain associated with burns and the procedures asso- lactated Ringer solution at up to 6 mL/kg/%TBSA should
ciated with treatment must be treated adequately and be used. Strategies such as euglycemia, early nutritional
aggressively. General treatment strategies involve opioids and support, oxandrolone, and -blockers potentially improve

PedSAP 2016 Book 2 Pediatric Critical Care 87 Trauma


outcomes. Burn patients are at an increased risk of infec- in pediatric patients with severe traumatic brain injury.
tions, including sepsis and ventilator-associated pneumonia. Pediatr Emerg Care 2006;22:90-3.
Early and aggressive antibiotic management should be used Branski LK, Herndon DN, Barrow RE, et al. Randomized con-
to decrease infectious complications. Pain, a significant trolled trial to determine the efficacy of long-term growth
comorbidity of burn injuries, should be managed with opioids hormone treatment in severely burned children. Ann Surg
and adjunctive agents as needed. 2009;250:514-23.

Breederveld RS, Tuinebreijer WE. Recombinant human


growth hormone for treating burns and donor sites.
Practice Points Cochrane Database Syst Rev 2012;12:CD008990.
Traumatic Brain Injury: Brunkhorst FM, Engel C, Bloos F, et al. Intensive insulin ther-
CPP = MAP ICP apy and pentastarch resuscitation in severe sepsis. N Engl
Optimize MAP using 0.9% saline and vasopressors J Med 2008;358:125-39.
Decrease ICP:
Sedation (watching for hypotension) Buck ML. Dexmedetomidine use in pediatric intensive
3% saline care and procedural sedation. J Pediatr Pharmacol Ther
23.4% saline in certain situations (e.g., fluid over- 2010;15:17-29.
load or cardiac dysfunction)
Cox CS, Baumgartner JE, Harting MT, et al. Autologous bone
Barbiturates marrow mononuclear cell therapy for severe traumatic
Mannitol (but falling out of favor) brain injury in children. Neurosurgery 2011;68:588-600.
Avoid hypothermia and hyperthermia
Keep patient euglycemic Elkon B, Riva Cambrin J, Hirshberg E, et al. Hyperglycemia:
Seizure prophylaxis with fosphenytoin if focal neurologic an independent risk factor for poor outcome in chil-
deficits, bone or metal fragments, young age, penetrating dren with traumatic brain injury. Pediatr Crit Care Med
injury 2014;15:623-31.
Burns
Empey PE, Velez de Mendizabal N, Bell MJ, et al. Therapeutic
Initial fluid management with lactated Ringer at up to
hypothermia decreases phenytoin elimination in children
6mL/kg/%TBSA
with traumatic brain injury. Crit Care Med 2013;41:2379-87.
Keep patient euglycemic
Start nutrition early Faul M, Xu L, Wald MM, et al. Traumatic brain injury in the
Oxandrolone and -blockers for hypermetabolic state United States: emergency department visits, hospitaliza-
Watch for infections and treat aggressively tions and deaths 2002-2006. Atlanta: CDC, National Center
Adequate pain control for Injury Prevention and Control, 2010.

Fram RY, Cree MG, Wolfe RR, et al. Intensive insulin therapy
improves insulin sensitivity and mitochondrial function in
REFERENCES severely burned children. Crit Care Med 2010;38:1475-83.
Adelson PD. Hypothermia in Children After Trauma. 2012.
Clinical Trials.gov:NCT00222742. Gomez DS, Campos EV, de Azevedo RP, et al. Individualised
vancomycin doses for paediatric burn patients to achieve
Arango JI, Deibert CP, Brown D, et al. Posttraumatic seizures PK/PD targets. Burns 2013;39:445-50.
in children with severe traumatic brain injury. Childs Nerv
Syst 2012;28:1925-9. Greve MW, Zink BJ. Pathophysiology of traumatic brain
injury. Mt Sinai J Med 2009;76:97-104.
Bar-Joseph G, Guilburd Y, Tamir A, et al. Effectiveness of
ketamine in decreasing intracranial pressure in chil- Hart DW, Wolf SE, Ramzy PI, et al. Anabolic effects of oxan-
dren with intracranial hypertension. J Neurosurg Pediatr drolone after severe burn. Ann Surg 2001;233:556-64.
2009;4:40-6.
Herndon DN, Rodriguez NA, Diaz EC, et al. Long-term pro-
Beca J, McSharry B, Erickson S, et al. Hypothermia for trau- pranolol use in severely burned pediatric patients: a
matic brain injury in children a phase II randomized randomized controlled study. Ann Surg 2012;256:402-11.
controlled trial. Crit Care Med 2015;43:1458-66.
Jeschke MG, Chinkes DL, Finnerty CC, et al.
Bhalla T, Dewhirst E, Sawardekar A, et al. Perioperative man- Pathophysiologic response to severe burn injury. Ann Surg
agement of the pediatric patient with traumatic brain 2008;248:387-401.
injury. Pediatr Anaesth 2012;22:627-40.
Jeschke MG, Kulp GA, Kraft R, et al. Intensive insulin therapy
Borsheim E, Herndon DN, Hawkins HK, et al. Pamidronate in severely burned pediatric patients: a prospective ran-
attenuates muscle loss following pediatric burn injury. J domized trial. Am J Respir Crit Care Med 2010;182:351-59.
Bone Miner Res 2014;29:1369-72.
Jeschke MG, Pinto R, Herndon DN, et al. Hypoglycemia is
Bramwell KJ, Haizlip J, Pribble C, et al. The effect of etomi- associated with increased postburn morbidity and mortal-
date on intracranial pressure and systemic blood pressure ity in pediatric patients. Crit Care Med 2014a;42:1221-31.

PedSAP 2016 Book 2 Pediatric Critical Care 88 Trauma


Jeschke MG, Herndon DN. Burns in children: standard and Piper BJ, Harrigan PW. Hypertonic saline in paediatric trau-
new treatments. Lancet 2014b;383:1168-78. matic brain injury: a review of nine years experience with
23.4% hypertonic saline as standard hyperosmolar ther-
Jones KE, Puccio AM, Harshman KJ, et al. Levetiracetam ver- apy. Anaesth Intensive Care 2015;43:204-10.
sus phenytoin for seizure prophylaxis in severe traumatic
brain injury. Neurosurg Focus 2008;25:E3-E3. Porro LJ, Herndon DN, Rodriguez NA, et al. Five-year out-
comes after oxandrolone administration in severely
Khorasani EN, Mansouri F. Effect of early enteral nutrition burned children: a randomized clinical trial of safety and
on morbidity and mortality in children with burns. Burns efficacy. J Am Coll Surg 2012;214:489-502.
2010;36:1067-71.
Porter C, Cotter M, Diaz EC, et al. Amino acid infusion fails to
Kochanek PM, Carney N, Ashwal S, et al. Guidelines for the stimulate skeletal muscle protein synthesis up to 1 year
acute medical management of severe traumatic brain after injury in children with severe burns. J Trauma Acute
injury in infants, children, and adolescents-second edition. Care Surg 2013;74:1480-84.
Pediatr Crit Care Med 2012;13:S1-S82.
Qu R, Li Y, Gao Q, et al. Neurotrophic and growth factor gene
Kraft R, Herndon DN, Mlcak RP, et al. Bacterial respiratory expression profiling of mouse bone marrow stromal cells
tract infections are promoted by systemic hyperglyce- induced by ischemic brain extracts. Neuropathology
mia after severe burn injury in pediatric patients. Burns 2007;27:355-63.
2014;40:428-35.
Robertson CL, Fidan E, Stanley RM, et al. Progesterone for
LaRochelle JM. Trauma. In: Eiland LS, Todd TJ, eds. neuroprotection in pediatric traumatic brain injury. Pediatr
Advanced Pediatric Therapeutics. Memphis: Pediatric Crit Care Med 2015;16:236-44.
Pharmacy Advocacy Group, 2015:chap 23.
Rogers DA, Deal C, Argent AC, et al. Ventilator associ-
Liao GP, Harting MT, Hetz RA, et al. Autologous bone marrow ated pneumonia in major paediatric burns. Burns
mononuclear cells reduce therapeutic intensity for severe 2014;40:1141-8.
traumatic brain injury in children. Pediatr Crit Care Med
2015;16:245-55. Sabsovich I, Rehman Z, Yunen J, et al. Propofol infusion syn-
drome: a case of increasing morbidity with traumatic brain
Liesemer K, Bratton SL, Zebrack CM, et al. Early post-trau- injury. Am J Crit Care 2007;16:82-5.
matic seizures in moderate to severe pediatric traumatic
brain injury: rates, risk factors, and clinical features. J Schoeler M, Loetscher PD, Rossaint R, et al.
Neurotrauma 2011;28:755-62. Dexmedetomidine is neuroprotective in an in vitro model
for traumatic brain injury. BMC Neurol 2012;12:20.
Lin H, Faraklas I, Sampson C, et al. Use of dexmedetomidine
for sedation in critically ill mechanically ventilated pediat- Scott JR, Muangman PR, Tamura RN, et al. Substance P
ric burn patients. J Burn Care Res 2011;32:98-103. levels and neutral endopeptidase activity in acute burn
wounds and hypertrophic scar. Plast Reconst Surg
Mehta NM, Compher C; ASPEN Board of Directors. ASPEN 2005;115:1095-102.
clinical guidelines: nutrition support of the critically ill
child. JPEN J Parenter Enteral Nutr 2009;33:260-76. Shank ES, Sheridan RL, Ryan CM, et al. Hemodynamix
responses to dexmedetomidine in critically injured intu-
Nakagawa K, Chang CWJ, Koenig MA, et al. Treatment of bated pediatric burned patients: a preliminary study. J
refractory intracranial hypertension with 23.4% saline Burn Care Res 2013;34:311-7.
in children with traumatic brain injury. J Clin Anesth
2012;24:318-23. Sheridan RL, Stoddard FJ, Kazis LE, et al. Long-term post-
traumatic stress symptoms vary inversely with early
Osterman MJK, Kochanek KD, MacDorman MF. Annual opiate dosing in children recovering from serious burns:
summary of vital statistics: 2012-2013. Pediatr effects durable at 4 years. J Trauma Acute Care Surg
2015;135:1115-25. 2014;76:828-32.
Pearl PL, McCarter R, McGavin CL, et al. Results of phase Sherwin CMT, Wead S, Stockmann C, et al. Amikacin popu-
II levetiracetam trial following acute head injury in lation pharmacokinetics among paediatric burn patients.
children at risk for posttraumatic epilepsy. Epilepsia Burns 2014;40:311-8.
2013;54:e135-37.
Smith RL, Lin JC, Adelson PD, et al. Relationship between
Pereira CT, Murphy KD, Herndon DN. Altering metabolism. J hyperglycemia and outcome in children with severe trau-
Burn Care Rehabil 2005;26:194-9. matic brain injury. Pediatr Crit Care Med 2012;13:85-91.
Pham TN, Cancio LC, Gibran NS. American Burn Association Sookplung P, Siriussawakul A, Malakouti A, et al.
practice guidelines burn shock resuscitation. J Burn Care Vasopressor use and effect on blood pressure after severe
Res 2008;29:257-66. adult traumatic brain injury. Neurocrit Care 2011;15:46-54.

Piatt JH, Neff DA. Hospital care of childhood traumatic brain Stanley RM, Bonsu BK, Zhao W, et al. US estimates of hos-
injury in the United States, 1997-2009: a neurosurgical per- pitalized children with severe traumatic brain injury:
spective. J Neurosurg Pediatr 2012;10:257-67. implications for clinical trials. Pediatrics 2012;129:e24-30.

PedSAP 2016 Book 2 Pediatric Critical Care 89 Trauma


Stoddard FJ, White GW, Kazis LE, et al. Patterns of medica- Walker PA, Harting MT, Jimenez F, et al. Direct intrathe-
tion administration from 2001 to 2009 in the treatment of cal implantation of mesenchymal stromal cells leads to
children with acute burn injuries: a multicenter study. J enhanced neuroprotection via an NFkappaB-mediated
Burn Care Res 2011;32:519-28. increase in interleukin-6 production. Stem Cells Dev
2010;19:867-76.
Sundberg J, Estrada C, Jenkins C, et al. Hypothermia is asso-
ciated with poor outcome in pediatric trauma patients. Am Walker TLJ, Urriza Rodriguez D, Coy K. Impact of reduced
J Emerg Med 2011;29:1019-22. resuscitation fluid on outcomes of children with 10-20%
body surface area scalds. Burns 2014;40:1581-86.
Taketomo CK, Hodding JH, Kraus DM. Pediatric and
Neonatal Dosage Handbook, 22nd ed. Hudson, OH: Lexi- Wayne EA, Dean HJ, Booth F, et al. Focal neurologic deficits
Comp, 2015. associated with hypoglycemia in children with diabetes. J
Pediatr 1990;117:575-7.
Tilford JM, Simpson PM, Yeh TS, et al. Variation in therapy
and outcome for pediatric head trauma patients. Crit Care Williams FN, Herndon DN, Kulp GA, et al. Propranolol
Med 2001;29:1056-61. decreases cardiac work in a dose-dependent manner in
severely burned children. Surgery 2011;149:231-9.
Tuvdendorj D, Chinkes DL, Zhang XJ, et al. Long-term oxan-
drolone treatment increases muscle protein net deposition WHO. Media Center. Burns. 2016.
via improving amino acid utilization in pediatric patients 6
months after burn injury. Surgery 2011;149:645-53. Yung M, Wilkins B, Norton L, et al. Glucose control, organ fail-
ure, and mortality in pediatric intensive care. Pediatr Crit
van den Berghe G, Wouters P, Weekers F, et al. Intensive Care Med 2008;9:147-52.
insulin therapy in critically ill patients. N Engl J Med
2001;345:1359-67. Zeiler FA, Teitelbaum J, West M, et al. The ketamine
effect on ICP in traumatic brain injury. Neurocrit Care
Vavilala MS, Kernic MA, Wang J, et al. Acute care clinical 2014;21:163-73.
indicators associated with discharge outcomes in chil-
dren with severe traumatic brain injury. Crit Care Med Zink BJ, Szmydynger-Chodobska J, Chodobski A. Emerging
2014;42:2258-66. concepts in the pathophysiology of traumatic brain injury.
Psychiatr Clin North Am 2010;33:741-56.
Vyles D, Sinha M, Rosenberg DI, et al. Predictors of serious
bacterial infections in pediatric burn patients with fever. J
Burn Care Res 2014;35:291-95.

PedSAP 2016 Book 2 Pediatric Critical Care 90 Trauma


Self-Assessment Questions
Questions 2124 pertain to the following case. C. Cultures and broad-spectrum antibiotics
L.T. is a 10-year-old boy (weight 30 kg) who was an unre- D. No treatment needed
strained passenger in a motor vehicle crash. At the scene, his 26. A 9-year-old girl was admitted to the ICU after a severe
Glasgow Coma Scale (GSC) score is 4, and he is intubated. TBI. She has been stabilized and has normal blood pres-
Initial vital signs include blood pressure 40/20 mm Hg and sure and adequate sedation. The patients ICP has been
heart rate 140 beats/minute. 10 mm Hg for the past several hours. Laboratory val-
21. Which one of the following is best to recommend for ues are as follows: Na 160 mEq/L, K 4.1 mEq/L, Cl 100
L.T.s initial treatment? mEq/L, HCO3 25 mEq/L, BUN 20 mg/dL, and SCr 0.6 mg/
dL. The patients osmolarity is 340 mOsm/L. Her perti-
A. Normal saline 20 mL/kg bolus
nent drugs include fentanyl 1 mcg/kg/hour, midazolam
B. Albumin 5% 20 mL/kg bolus
0.1 mg/kg/hour, 3% saline at 0.3 mL/kg/hour, and man-
C. Dextrose 5% 20 mL/kg/bolus
nitol 0.25 g/kg every 6 hours. Her ICP suddenly increases
D. 3% normal saline 10 mL/kg
to 35 mm Hg for 6 minutes, with no changes to her vital
22. L.T. begins to have rales on physical examination; how- signs. Which one of the following is most appropriate in
ever, his blood pressure is 50/25 mm Hg. Which one of this patients ICP management?
the following is best to recommend for L.T.?
A. Increase mannitol to 0.5 g/kg every 6 hours.
A. Albumin 5% 20 mL/kg bolus B. Increase fentanyl and midazolam.
B. Norepinephrine 0.1 mcg/kg/minute C. Administer 3% saline bolus 7 mL/kg.
C. Milrinone 0.5 mcg/kg/minute D. Administer pentobarbital 5 mg/kg.
D. Vasopressin 1 milliunit/kg/minute
23. On transfer to your hospital, L.T. still has a GCS score of 7 Questions 27 and 28 pertain to the following case.

and is becoming hypertensive. The interventions in the pre- V.R. is a 6-year-old girl who sustains a TBI from a motor vehi-
vious questions are discontinued, and L.T.s blood pressure cle crash. She is intubated for a GCS score of 6. All of her
is now stable at 120/80 mm Hg. Rales are now absent from laboratory values are normal except for Na 170 mEq/L and
the lung fields. No other drugs have been given. The neu- serum osmolarity 355 mOsm/L. V.R.s heart rate is 150 beats/
rosurgery team has begun placing an intracranial pressure minute, and her blood pressure is 170/100 mm Hg. Her per-
(ICP) monitor and drain when L.T.s blood pressure becomes tinent drugs include fentanyl 1 mcg/kg/hour, midazolam
150/100 mm Hg with a heart rate of 150 beats/minute. 0.1 mg/kg/hour, 3% saline at 0.3 mL/kg/hour, and mannitol
Which one of the following is the best intervention for L.T.? 0.25 g/kg every 6 hours. Her ICP is 30 mm Hg.
A. Therapeutic hypothermia 27. Which one of the following is best to recommend for V.R.?
B. 3% hypertonic saline
A. Increase mannitol to 0.5 g/kg every 6 hours.
C. Sedation with fentanyl and midazolam bolus
B. Give fentanyl and midazolam bolus.
D. A neuromuscular blocking agent
C. Administer 3% saline bolus 7 mL/kg.
24. L.T.s ICP monitor is placed without incident, and his D. Administer pentobarbital 5 mg/kg.
blood pressure and heart rate are normal and stable.
28. V.R. receives the intervention described earlier, and her
Laboratory values are normal. Initial ICP is 35 mm Hg.
vital signs remain normal. The team will insert a central
Which one of the following is best to initiate for L.T.?
and arterial line on the patient. Which one of the follow-
A. Fosphenytoin ing would be best to prevent ICP spikes in V.R.?
B. Normal saline bolus
A. 23.4% saline bolus
C. Pentobarbital
B. 3% saline bolus
D. 3% saline
C. Mannitol 0.25 g/kg intravenous bolus
25. A 3-year-old child who sustains a traumatic brain injury D. Propofol bolus
(TBI) after a motor vehicle collision is admitted to the
29. A 4-year-old girl sustains a TBI and is intubated for a
pediatric ICU. Five 5 hours after the injury, her temper-
GCS score of 5. All of her laboratory values and vital
ature is 101.9F. Which one of the following is the best
signs are normal except for Na 170 mEq/L and serum
course of action for this patient?
osmolarity 355 mOsm/L. Her pertinent drugs include
A. Acetaminophen fentanyl 2 mcg/kg/hour, midazolam 0.2 mg/kg/hour,
B. Therapeutic hypothermia vecuronium 0.1 mg/kg/hour, 3% saline at 0.3 mL/kg/hour,

PedSAP 2016 Book 2 Pediatric Critical Care 91 Trauma


and mannitol 0.25 mg/kg every 6 hours. Sedation and Questions 3340 pertain to the following case.

paralysis are adequate. The patients ICP is now 30 mm Q.V. is a 3-year-old boy (weight 10 kg) who sustained a 50%
Hg. Which one of the following is best for this patients total body surface area burn in a house fire. Q.V. is intubated
treatment? and brought to your burn center. On admission, a central line
is placed.
A. Increase fentanyl to 3 mcg/kg/hour.
B. Increase 3% saline to 0.4 mL/kg/hour. 33. Which one of the following would best provide fluid
C. Give mannitol 0.5 mg/kg intravenous bolus. resuscitation in Q.V.?
D. Give pentobarbital 5 mg/kg bolus, followed by A. 5% albumin
continuous infusion. B. Lactated Ringer
C. 0.9% saline
Questions 3032 pertain to the following case.
D. 3% saline

T.F. is a 1-year-old girl who arrives to the pediatric ICU intu- 34. Which one of the following represents the optimal
bated after a TBI. On CT, there is diffuse axonal injury (DAI) administration of intravenous fluids for resuscitation
as well as bone fragments in the brain. Echocardiography inQ.V.?
reveals an ejection fraction of 10% after the injury. On A. 188 mL/hour for 8 hours; then 93 mL/hour for 16
day 2 of admission, T.F.s drug therapy includes fentanyl, hours
midazolam, mannitol, vecuronium, pentobarbital, and B. 125 mL/hour for 8 hours; then 63 mL/hour for 16
3% saline. Her vital signs and laboratory values are nor- hours
mal. Her serum sodium is 145 mEq/L, and osmolarity is C. 125 mL/hour for 24 hours
300mOsm/L. Sedation is adequate. Rales are auscultated D. 83 mL/hour for 24 hours
bilaterally in both lungs. T.F.s ICP rises to 40 mm Hg for
35. Which one of the following glycemic ranges would be
7minutes.
optimal for Q.V.?
30. Which one of the following is best for T.F.?
A. 6080 mg/dL
A. Give 23.4% saline 0.5 mL/kg. B. 80130 mg/dL
B. Increase fentanyl and midazolam. C. 140180 mg/dL
C. Give 3% saline bolus 10 mL/kg. D. 170215 mg/dL
D. Give mannitol 0.5 g/kg intravenous bolus.
36. Q.V. is intubated and requires sedative agents. Which one
31. T.F.s attending physician asks your opinion about sei- of the following is best to recommend for Q.V.?
zure prophylaxis. Which one of the following is best to
A. Midazolam
recommend for T.F.?
B. Dexmedetomidine
A. Seizure prophylaxis is discouraged in the TBI C. Ketamine and clonidine
guidelines. D. Fentanyl and midazolam
B. The patient has risk factors of DAI, young age,
37. On day 3 of admission, Q.V. spikes a fever (temperature
andbone fragments; thus, prophylaxis should be
102.5F). Which one of the following is best to recom-
given.
mend for Q.V.?
C. The patient has risk factors of mannitol
administration, a decreased ejection fraction, and A. This is a post-burn fever, and antibiotics are
rales; thus, prophylaxis should be given. unnecessary.
D. The patient has no risk factors for seizures; thus, B. This is viral related, and supportive care is
prophylaxis is not indicated. indicated.
C. Give piperacillin/tazobactam and vancomycin.
32. T.F.s care team decides to initiate seizure prophylaxis.
D. Give ampicillin and gentamicin.
Which one of the following is the best agent and monitor-
ing to recommend for T.F.? 38. Q.V.s team decided to initiate vancomycin, independent
of your suggestion. Which one of the following is the best
A. Fosphenytoin and total phenytoin concentration
dosing strategy for Q.V.?
B. Fosphenytoin and free phenytoin concentration
C. Phenobarbital and phenobarbital trough A. 150 mg intravenously every 8 hours
concentration B. 150 mg intravenously every 6 hours
D. Valproic acid and valproic acid trough C. 200 mg intravenously every 8 hours
concentration D. 200 mg intravenously every 6 hours

PedSAP 2016 Book 2 Pediatric Critical Care 92 Trauma


39. After 1 week, Q.V. has signs of a hypermetabolic state.
Which one of the following is best for Q.V.?
A. Oxandrolone
B. Growth hormone
C. Pamidronate
D. Clonidine
40. The intervention you recommended for Q.V. was partly
successful. However, he still has signs of tachycardia
and hypertension. Which one of the following is best to
recommend for Q.V.?
A. Esmolol
B. Lisinopril
C. Verapamil
D. Propranolol

PedSAP 2016 Book 2 Pediatric Critical Care 93 Trauma


Learner Chapter Evaluation: Trauma.

As you take the posttest for this chapter, also evaluate the Use the 5-point scale to indicate whether this chapter pre-
materials quality and usefulness, as well as the achievement pared you to accomplish the following learning objectives:
of learning objectives. Rate each item using this 5-point scale:
31. Develop strategies to optimize a patients mean arterial
Strongly agree pressure.
Agree 32. Assess strategies to decrease or prevent increased intra-
Neutral cranial pressure.
Disagree 33. Demonstrate appropriate sedation management strate-
Strongly disagree gies, as well as appropriate glucose control and seizure
prophylaxis, for the patient with traumatic brain injury.
20. The content of the chapter met my educational needs. 34. Design appropriate fluid management and treatment
21. The content of the chapter satisfied my expectations. approach to increase lean body mass in a patient with burns.

22. The author presented the chapter content effectively. 35. Compose strategies to optimize pain control and treat
infections in a patient with burns.
23. The content of the chapter was relevant to my practice
and presented at the appropriate depth and scope. 36. Please provide any specific comments related to any
perceptions of bias, promotion, or advertisement of
24. The content of the chapter was objective and balanced. commercial products.
25. The content of the chapter is free of bias, promotion, or 37. Please expand on any of your above responses, and/or
advertisement of commercial products. provide any additional comments regarding this chapter:
26. The content of the chapter was useful to me.
27. The teaching and learning methods used in the chapter
Questions 3840 apply to the entire learning module.
were effective.
38. How long did it take you to read the instructional materi-
28. The active learning methods used in the chapter were als in this module?
effective.
39. How long did it take you to read and answer the assess-
29. The learning assessment activities used in the chapter ment questions in this module?
were effective.
4 0. Please provide any additional comments you may have
30. The chapter was effective overall. regarding this module:

PedSAP 2016 Book 2 Pediatric Critical Care 94 Trauma


Clinical and Practice Updates I
Clinical and Practice Updates I Panel

Series Editors: Kyle Davis, Pharm.D., BCPS


Marcia L. Buck, Pharm.D., BCPPS, FCCP, FPPAG Clinical Pharmacy Specialist-Internal Medicine
Department of Pharmacy and Internal Medicine
Clinical Coordinator
Ochsner Medical Center
University of Virginia Childrens Hospital
New Orleans, Louisiana
Professor
Department of Pediatrics
University of Virginia School of Medicine
Guideline Review: Diabetic
Clinical Professor
Virginia Commonwealth University School of Pharmacy Ketoacidosis and Hyperglycemic
Department of Pharmacy Services Hyperosmolar State
University of Virginia Health System
Authors
Charlottesville, Virginia
Elizabeth S. Goswami, Pharm.D., BCPS, BCPPS
Kalen B. Manasco, Pharm.D., BCPS
Clinical Pharmacy Specialist
Clinical Associate Professor
Department of Pharmacy
Department of Clinical and Administrative Pharmacy
The Johns Hopkins Hospital
University of Georgia College of Pharmacy
Baltimore, Maryland
Augusta, Georgia
Susan Warrington, Pharm.D., BCPPS
Faculty Panel Chair: Clinical Pharmacist, Pediatric Critical Care
Joseph M. LaRochelle, Pharm. D., BCPPS Department of Pharmacy
The Childrens Hospital of Philadelphia
Clinical Associate Professor
Philadelphia, Pennsylvania
College of Pharmacy
Xavier University of Louisiana
Reviewers
Clinical Associate Professor of Pediatrics
Louisiana State University Health Sciences Julie Pingel, Pharm.D.
Center School of Medicine Pediatric Critical Care Clinical Pharmacist
New Orleans, Louisiana Pediatric Pharmacy
Monroe Carell Jr. Childrens Hospital at Vanderbilt
Nashville, Tennessee
Interactive Case: Severe Acute Asthma Ifeyinwa D. Eboh, Pharm.D., MPH, BCPS
(Status Asthmaticus) Clinical Pharmacist
Pharmacy Department
Author
Texas Health Presbyterian Hospital
Laura Lo Castro Bio, Pharm.D., BCPS Allen, Texas
Clinical Pharmacist
Monica A. Puebla, Pharm.D., MBA, MHA, BCPS
Department of Pharmacy
Medication Safety-Healthcare Professional Reviewer
Lucile Packard Childrens Hospital Stanford
Med-ERRS, Inc.
Palo Alto, California
Horsham, Pennsylvania
Reviewers
Lois F. Parker, BSPharm, FASHP Interactive Case: Status Epilepticus
Clinical Pharmacist
Department of Pharmacy Author
Massachusetts Eye and Ear Christina Cox, Pharm.D., BCPS, BCPPS
Boston, Massachusetts
Assistant Professor
Brooke Clark, Pharm.D., BCPS Clinical Pharmacy and Outcomes Sciences
Clinical Pharmacy Program Manager South Carolina College of Pharmacy,
Banner Health University of South Carolina
Phoenix, Arizona Columbia, South Carolina
Reviewers The American College of Clinical Pharmacy and the authors
Emily L. Rowe, Pharm.D., BCPS, BCPPS, MS-PREP thank the following individuals for their careful review of the
Senior Clinical Pharmacy Specialist, Pediatrics Clinical and Practice Updates I features:
Department of Pharmacy
Nicola G. Dahl, Pharm.D.
The Floating Hospital for Children at Tufts Medical Center
Medical Writer
Boston, Massachusetts
Kanab, Utah
Maha O. Kebir, Pharm.D., MS, BCPS, CPHQ
Clinical Pharmacist
Department of Pharmacy
Christiana Care Health System
Newark, Delaware
DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST
Consultancies: Elizabeth J. Beckman (Lexi-Comp/Wolters/Kluwer Health); Regine L Caruthers (Lexi-Comp/Wolters Kluwer
Health); Joseph M. LaRochelle (ACCP); Lois F. Parker (ASHP/MSHP)

Stock Ownership:

Royalties:

Grants: Elizabeth J. Beckman (ASHP Foundation); Christina Cox (University of South Carolina);

Honoraria: Christina Cox (ACCP, Palmetto Health Richland Childrens Hospital); Jennifer L. Morris (ACCP, Society of Critical Care
Medicine, Pediatric Pharmacy Advocacy Group, Wolters Kluwer)

Other:

Nothing to disclose: Laura Lo Castro Bio, Joshua Caballero, Brooke Clark, Kyle Davis, Ifeyinwa D. Eboh, Elizabeth S. Goswami,
Aaron A. Harthan, Maha O. Kebir, Allison Lardieri, Michael A. Maccia, Kristine Parbuoni, Monica A. Puebla, Sara P. Rooney, Emily
L. Rowe, Ashley R. Schappell, Julie Pingel, Brandi L. Strauser, Andrea Joan Stumpe, Susan Warrington

ROLE OF BPS: The Board of Pharmacy Specialties (BPS) is an autonomous division of the American Pharmacists Association
(APhA). BPS is totally separate and distinct from ACCP. The Board, through its specialty councils, is responsible for specialty
examination content, administration, scoring, and all other aspects of its certification programs. PedSAP has been approved by
BPS for use in BCPPS recertification. Information about the BPS recertification process is available online.

Other questions regarding recertification should be directed to:

Board of Pharmacy Specialties


2215 Constitution Avenue NW
Washington, DC 20037
(202) 429-7591
CONTINUING PHARMACY EDUCATION AND
RECERTIFICATION INSTRUCTIONS
Continuing Pharmacy Education Credit: The American College of Clinical Pharmacy is accredited by the Accreditation
Council for Pharmacy Education (ACPE) as a provider of continuing pharmacy education (CPE).

PedSAP: Target Audience: The target audience for PedSAP 2016 Book 2(Pediatric Critical Care) is pediatric pharmacotherapy spe-
cialists and advanced-level clinical pharmacists caring for pediatric patients with several acute conditions and complications of
the pulmonary, cardiovascular, endocrine, and neurologic systems.

Available CPE credits: Purchasers who successfully complete all posttests for PedSAP 2016 Book 2 (Pediatric Critical Care)
can earn 13.0 contact hours of continuing pharmacy education credit. The universal activity numbers are as follows: Pediatric
Critical Care I 0217-0000-16-021-H01-P, 3.5 contact hours; Pediatric Critical Care II 0217-0000-16-022-H01-P, 4.5 contact
hours; and Clinical and Practice Updates 0217-0000-16-170-H01-P, 5.0 contact hours. You may complete one or all available mod-
ules for credit. Tests may not be submitted more than once.

BCPPS test deadline: 11:59 p.m. (Central) on January 17, 2017.


ACPE test deadline: 11:59 p.m. (Central) on September 14, 2019.

Posttest access: Go to www.accp.com and sign in with your e-mail address and password. Technical support is available from
8 a.m. to 5 p.m. (Central) weekdays by calling (913) 492-3311. PedSAP products are listed under My Online Products on your My
Account page.

BCPPS Recertification Credit: To receive BCPPS recertification CPE credit, a PedSAP posttest must be submitted within the
4-month period after the books release. The first page of each print and online book lists the deadline to submit a required
posttest for BCPPS recertification credit. Only completed tests are eligible for credit; no partial or incomplete tests will be pro-
cessed. Tests may not be submitted more than once. The passing point for BCPPS recertification is based on an expert analysis of
the items in each posttest module.

ACPE CPE Credit: To receive ACPE CPE credit for a PedSAP module, a posttest must be submitted within the 3-year period after
the books release. The appropriate CPE credit will be awarded for test scores of 50% and greater.

Credit Assignment and Reporting: All required posttests that meet the 50% score standard will be immediately awarded the
appropriate ACPE CPE credit. Earned credits will be transmitted within 24 hours to www.mycpemonitor.net and should appear on
statements of credit within 3 business days.

Required posttests that are submitted before the BCPPS test deadline and that meet the passing point set by statistical analysis
will earn BCPPS recertification credits. These credits will be posted within 30 days after the BCPPS test deadline. For statements
of CPE credit, visit www.mycpemonitor.net.

All BCPPS recertification credits are forwarded by ACCP to the Board of Pharmacy Specialties (BPS). Questions regarding the
number of hours required for BCPPS recertification should be directed to BPS at (202) 429-7591 or www.bpsweb.org. The ACCP
Recertification Dashboard is a free online tool that can track recertification credits as they are earned through ACCP and schedule
new opportunities for credits from upcoming ACCP professional development programs.

Posttest Answers: The explained answers with rationale and supporting references will be posted 1 week after the BCPPS
test deadline and will be available to anyone who has either submitted a posttest or waived his or her right to receive credit (see
below) from a posttest. Go to www.accp.com and sign in with your e-mail address and password. Click the PedSAP book on your
My Account page and you will see a link to the explained answers.

Test Waivers: To access the explained answers without submitting a posttest, sign in to your My Account page, select the
PedSAP book, and click on the waiver link for that module. By completing the waiver form for a module, you waive the opportu-
nity to receive CPE credit for that module. After you submit a waiver, you will see a link to the PDF file that contains the answers
for the module you waived. Answers will be available starting 1 week after the BCPPS test deadline.

ACSAP 2016 Book 2 Pediatric Critical Care 100 Program Instructions


Interactive Patient Case: Severe
Acute Asthma (Status Asthmaticus)
By Laura Lo Castro Bio, Pharm.D., BCPS

Reviewed by Lois F. Parker, BSPharm, FASHP; Brooke Clark, Pharm.D., BCPS; and Kyle Davis, Pharm.D., BCPS

LEARNING OBJECTIVES

1. Devise an appropriate -agonist regimen based on presentation of a pediatric patient with severe acute asthma.
2. Justify the use of ipratropium as adjunct therapy for pediatric patients with severe acute asthma.
3. Identify the most appropriate magnesium sulfate regimen for severe acute asthma in pediatric patients.
4. Evaluate the benefits and limitations of various treatment modalities for severe acute asthma management in pediatric patients.
5. Design a stepwise approach to escalate therapy for a child with severe acute asthma refractory to standard of care.

INTERACTIVE CASE
ABBREVIATIONS IN THIS CHAPTER
Click here to begin this PedSAP activity.
CRS Clinical Respiratory Score
DBP Diastolic blood pressure
ICS Inhaled corticosteroids
IQR Interquartile range BASELINE KNOWLEDGE STATEMENTS
IVF Intravenous fluids
LOS Length of stay Readers of this chapter are presumed to be familiar
with the following:
MAP Mean arterial pressure
Physiology of the respiratory tract and immune
PAS Pediatric asthma score
system
PASS Pediatric asthma severity score
Drug delivery systems including metered-dose
PRAM Pediatric respiratory assessment
inhalers, nebulizers, and injectables
measure
Weight-based dose calculations
PS Pulmonary score
Fluid and electrolyte management for non-critically
RAD Respiratory rate, accessory mus-
cle use, decreased breath sounds ill patients
SABA Short-acting -agonist Normal vital signs for children
SCS Systemic corticosteroids
ADDITIONAL READINGS

The following free resources have additional back-


ground information on this topic:
AboutKidsHealth. Appropriate administration
technique for an MDI with spacer [homepage on
the Internet].
Web MD. Asthma Health Center. Patient education
on severe acute asthma [homepage on the
Internet].

PedSAP 2016 Book 2 Pediatric Critical Care 101 Interactive Patient Case: Severe Acute Asthma
REFERENCES Dalabih AR, Bondi SA, Harris ZL, et al. Aminophylline infu-
Allen JY, Macias CG. The efficacy of ketamine in pediatric sion for status asthmaticus in the pediatric critical care
emergency department patients who present with acute unit setting is independently associated with increased
severe asthma. Ann Emerg Med 2005;46:43-50. length of stay and time for symptom improvement. Pulm
Pharmacol Ther 2014;27:57-61.
FDA Over-The-Counter Drugs Advisory Committee. Panel
recommends limits on alcohol content of nonprescription Denmark TK, Crane HA, Brown L. Ketamine to avoid mechan-
products. Am J Hosp Pharm 1993;50:400. ical ventilation in severe pediatric asthma. J Emerg Med
2006;30:163-6.
Arnold DH, Gebretsadik T, Abramo TJ, et al. The RAD score:
a simple acute asthma severity score compares favorably DiPiro CV. Electrolyte homeostasis. In: Wells BG, DiPiro
to more complex scores. Ann Allergy Asthma Immunol JT, Schwinghammer TL, et al., eds. Pharmacotherapy
2011;107:22-8. Handbook. New York: McGraw-Hill, 2015:798-812.

Barnes PJ, Adcock I. Anti-inflammatory actions of ste- Doymaz A, Schneider J, Sagy M. Early administration of ter-
roids: molecular mechanisms. Trends Pharmacol Sci butaline in severe pediatric asthma may reduce incidence
1993;14:436-41. of acute respiratory failure. Ann Allergy Asthma Immunol
2014;112:207-10.
Berlinski A, Willis JR, Leisenring T. In-vitro comparison of 4
large-volume nebulizers in 8 hours of continuous nebuliza- Ducharme FM, Chalut D, Plotnick L, et al. The Pediatric
tion. Respir Care 2010;55:1671-9. Respiratory Assessment Measure: a valid clinical score for
assessing acute asthma severity from toddlers to teenag-
Bhogal SK, McGillivray D, Bourbeau J, et al. Early admin- ers. J Pediatr 2008;152:476-80.
istration of systemic corticosteroids reduces hospital
admission rates for children with moderate and severe Egelund TA, Wassil SK, Edwards EM, et al. High-dose mag-
asthma exacerbation. Ann Emerg Med 2012;60:84-91. nesium sulfate infusion protocol for status asthmaticus: a
safety and pharmacokinetics cohort study. Intensive Care
Bigham MT, Jacobs BR, Monaco MA, et al. Helium/oxy- Med 2013;39:117-22.
gen-driven albuterol nebulization in the management
of children with status asthmaticus: a random- Gorelick MH, Stevens MW, Schultz T, et al. Difficulty in
ized, placebo-controlled trial. Pediatr Crit Care Med obtaining peak expiratory flow measurements in children
2010;11:356-61. with acute asthma. Pediatr Emerg Care 2004;20:22-6.

Bio LL, Willey VJ, Poon CY. Comparison of levalbuterol and Green SM, Roback MG, Kennedy RM, et al. Clinical practice
racemic albuterol based on cardiac adverse effects. J guideline for emergency department ketamine disso-
Pediatr Pharmacol Ther 2011;16:191-8. ciative sedation: 2011 update. Ann Emerg Med. 2011
May;57(5):449-61.
Bogie AL, Towne D, Luckett PM, et al. Comparison of intrave-
nous terbutaline versus normal saline in pediatric patients Griffiths B, Ducharme FM. Combined inhaled anticholiner-
on continuous high-dose nebulized albuterol for status gics and short-acting beta2-agonists for initial treatment
asthmaticus. Pediatr Emerg Care 2007;23:355-61. of acute asthma in children. Cochrane Database Syst Rev
2013;8:CD000060.
Browne GJ, Lam LT. Single-dose intravenous salbutamol
bolus for managing children with acute severe asthma in Hames H, Seabrook JA, Matsui D, et al. A palatability study
the emergency department: reanalysis of data. Pediatr Crit of a flavored dexamethasone preparation versus predniso-
Care Med 2002;3:117-23. lone liquid in children. Can J Clin Pharmacol 2008;15:e95-8.

Carrie S, Anderson TA. Volatile anesthetics for status asth- Hendeles L. Selecting a systemic corticosteroid for acute
maticus in pediatric patients: a comprehensive review and asthma in young children. J Pediatr 2003;142:S40-4.
case series. Paediatr Anaesth 2015;25:460-7.
Institute for Safe Medication Practices (ISMP). Safety Brief:
Castro-Rodriguez JA, J Rodrigo G, E Rodrguez-Martnez C. Questionable Safety with Continuous Inhalation Albuterol
Principal findings of systematic reviews of acute asthma Infusion Set-up. ISMP Medication Safety Alert! Acute Care
treatment in childhood. J Asthma 2015;52:1038-45. Newsletter. June 3, 2010.

Charmandari E, Kino T, Chrousos GP. Glucocorticoids. Keeney GE, Gray MP, Morrison AK, et al. Dexamethasone for
In: Yaffe SJ, Aranda JV, eds. Neonatal and Pediatric acute asthma exacerbations in children: a meta-analysis.
Pharmacology. Philadelphia: Wolters Kluwer, Pediatrics 2014;133:493-9.
2011:760-72.
Kelly CS, Littleman CA, Pestian JP, et al. Improved outcomes
Childrens Hospital of Philadelphia (CHOP). Acute Asthma for hospitalized asthmatic children using a clinical path-
Emergent Care. CHOP Clinical Pathway. Updated way. Ann Allergy Asthma Immunol 2000;84:509-16.
September 2015.
Kenyon CC, Fieldston ES, Luan X, et al. Safety and effec-
Dahmani S, Delivet H, Hilly J. Emergence delirium in children: tiveness of continuous aerosolized albuterol in the
an update. Curr Opin Anaesthesiol 2014;27:309-15. non-intensive care setting. Pediatrics 2014;134:e976-82.

PedSAP 2016 Book 2 Pediatric Critical Care 102 Interactive Patient Case: Severe Acute Asthma
Kim MK, Yen K, Redman RL, et al. Vomiting of liquid corti- Powell C, Kolamunnage-Dona R, Lowe J, et al. Magnesium
costeroids in children with asthma. Pediatr Emerg Care sulphate in acute severe asthma in children (MAGNETIC):
2006;22:397-401. a randomised, placebo-controlled trial. Lancet Respir Med
2013;1:301-8.
Koninckx M, Buysse C, de Hoog M. Management of status
asthmaticus in children. Paediatr Respir Rev 2013;14:78-85. Raissy HH, Kelly HW, Szefler AJ. Antiasthmatics. In: Yaffe
SJ, Aranda JV, eds. Neonatal and Pediatric Pharmacology.
Koumbourlis AC, Mastropietro C. Continuous inhalation Philadelphia: Wolters Kluwer, 2011:574-97.
of ipratropium bromide for acute asthma refractory
to 2-agonist treatment. J Pediatr Pharmacol Ther Rampa S, Allareddy V, Asad R, et al. Outcomes of invasive
2015;20:66-9. mechanical ventilation in children and adolescents hos-
pitalized due to status asthmaticus in United States: a
Liu LL, Gallaher MM, Davis RL, et al. Use of a respiratory clini- population based study. J Asthma 2015;52:423-30.
cal score among different providers. Pediatr Pulmonol
2004;37:243-8. Shankar V, Churchwell KB, Deshpande JK. Isoflurane ther-
apy for severe refractory status asthmaticus in children.
McMahon AW, Levenson MS, McEvoy BW, et al. Age and risks Intensive Care Med 2006;32:927-33.
of FDA-approved long-acting beta2-adrenergic receptor
agonists. Pediatrics 2011;128:e1147-54. Smith SR, Baty JD, Hodge D. Validation of the pulmonary
score: an asthma severity score for children. Acad Emerg
Miller AG, Breslin ME, Pineda LC, et al. An asthma proto- Med 2002;9:99-104.
col improved adherence to evidence-based guidelines for
pediatric subjects with status asthmaticus in the emer- Soltsz S, Mencke T, Stunz M, et al. Attenuation of a
gency department. Respir Care 2015;60:1759-64. rocuronium-induced neuromuscular block in patients
receiving prednisolone. Acta Anaesthesiol Scand
National Asthma Education and Prevention Program 2009;53:443-8.
(NAEPP). Expert Panel Report 3 (EPR-3): Guidelines for the
Diagnosis and Management of Asthma. Clinical Practice Stoicea N, Versteeg G, Florescu D, et al. Ketamine-based
Guidelines. National Institutes of Health, National Health, anesthetic protocols and evoked potential monitoring: a
Lung, and Blood Institute. NIH Publication No. 08-4051, risk/benefit overview. Front Neurosci 2016;10:37.
2007.
Tobias JD. Inhalational anesthesia: basic pharmacology, end
Nievas IF, Anand KJ. Severe acute asthma exacerbation in organ effects, and applications in the treatment of status
children: a stepwise approach for escalating therapy in asthmaticus. J Intensive Care Med 2009;24:361-71.
a pediatric intensive care unit. J Pediatr Pharmacol Ther
2013;18:88-104. Torres S, Sticco N, Bosch JJ, et al. Effectiveness of magne-
sium sulfate as initial treatment of acute severe asthma
Nuorti JP, Whitney CG. Prevention of pneumococcal disease in children. A randomized, controlled trial. Arch Argent
among infants and children use of 13-valent pneumo- Pediatr 2012;110:291-7.
coccal conjugate vaccine and 23-valent pneumococcal
polysaccharide vaccine: recommendations of the advisory Wheeler DS, Jacobs BR, Kenreigh CA, et al. Theophylline ver-
committee on immunization practices (ACIP). Morb Mortal sus terbutaline in treating critically ill children with status
Wkly Rep 2010;59(RR11):1-18. asthmaticus: a prospective, randomized, controlled trial.
Pediatr Crit Care Med 2005;6:142-7.
Papiris SA, Manali ED, Kolilekas L, et al. Acute severe
asthma: new approaches to assessment and treatment. Wong JJ, Lee JH, Turner DA, et al. A review of the use of
Drugs 2009;69:2363-91. adjunctive therapies in severe acute asthma exacerbation in
critically ill children. Expert Rev Respir Med 2014;8:423-41.
Phumeetham S, Bahk TJ, Abd-Allah S, et al. Effect of high-
dose continuous albuterol nebulization on clinical Wyatt EL, Borland ML, Doyle SK, et al. Metered-dose inhaler
variables in children with status asthmaticus. Pediatr Crit ipratropium bromide in moderate acute asthma in chil-
Care Med 2015;16:e41-6. dren: a single-blinded randomised controlled trial. J
Paediatr Child Health 2015;51:192-8.
Powell C, Dwan K, Milan SJ, et al. Inhaled magnesium sulfate
in the treatment of acute asthma. Cochrane Database Syst
Rev 2012;12:CD003898.

PedSAP 2016 Book 2 Pediatric Critical Care 103 Interactive Patient Case: Severe Acute Asthma
Self-Assessment Questions
1. A 5-year-old boy (weight 16 kg) with a 1-year history air). She has received three doses of albuterol and ipratro-
of mild intermittent asthma 1 presents to his pediatri- pium by nebulization without clinical response. Which one
cians clinic with his mother because of increased work of the following is best to recommend for this patient?
of breathing and lack of response to three doses of alb-
A. Repeat her albuterol nebulization.
uterol nebulizations administered at home. Physical
B. Initiate magnesium sulfate intravenous intermittent
examination reveals expiratory wheezing, accessory
dose.
muscle use, decreased breath sounds at the bases bilat-
C. Initiate continuous albuterol nebulization.
erally, and SaO2 91% on room air. The pediatrician is
D. Initiate terbutaline intravenous infusion.
concerned about the severity of this acute asthma epi-
sode and arranges transport to the hospital for further 5. A 26-month old boy (weight 14.2 kg) requires continuous
evaluation and management. As the clinic pharmacist, albuterol nebulization. Which one of the following doses
which one of the following is best to recommend as this is best to recommend for this patient?
patient awaits transport? A. 5 mg/hour
A. Continuous albuterol nebulization 5 mg/hour B. 7.5 mg/hour
B. Intubation C. 15 mg/hour
C. Ipratropium nebulization 2.5 mg/3 mL once D. 20 mg/hour
D. Prednisolone 16 mg by mouth once 6. Although ipratropium nebulization is commonly used
2. In the pediatric ED triage, a 3-year-old girl is evaluated in the treatment of children with acute asthma present-
for severe acute asthma. Vital signs and physical exam- ing in the ED, which one of the following degrees of
ination findings include heart rate 90 beats/minute, asthma exacerbation severity is most supported by the
respiratory rate 55 breaths/minute, positive intercos- evidence?
tal retractions, inspiratory and expiratory wheezing,
A. Mild persistent
decreased breath sounds bilaterally, and positive nasal
B. Moderate persistent
congestion. The patient received three doses of nebulized
C. Severe persistent
albuterol/ipratropium inhalation separated by 20 minutes,
D. Mild intermittent
but her symptoms persist. Prednisone 1 mg/kg/dose for
oral administration is ordered immediately. Which one of 7. A 6-year-old girl (weight 29.6 kg) with a medical history
the following is best to recommend for this patient? of moderate persistent asthma is being admitted to the
pediatric unit from the ED after her severe acute asthma
A. Continuous albuterol nebulization
failed to respond to standard therapy. She received alb-
B. Intubation
uterol with ipratropium nebulization for three doses and
C. Magnesium sulfate intravenously
dexamethasone 1 mg/kg/dose by mouth, and continu-
D. Methylprednisolone intravenously
ous albuterol 15 mg/hr was initiated. Laboratory values
3. A 32-month-old boy is admitted to the inpatient pediatric are within normal limits. Her home drugs include alb-
unit for the management of a moderate asthma exacer- uterol metered-dose inhaler (MDI) 2 puffs as needed for
bation. On hospital day 2, he is receiving the following wheezing, fluticasone/salmeterol dry powder inhaler 250
drugs: continuous albuterol nebulization, prednisone mcg/50 mcg inhaled twice daily, and a multivitamin. She
1 mg/kg/dose by mouth twice daily, and dextrose 5% and has no known drug allergies. Her asthma scores con-
sodium chloride 0.45% at maintenance rate. He is NPO tinue to indicate severe work of breathing, according
status (nothing by mouth). Which one of the following is to the examination. Which one of the following orders
best to monitor for potential adverse effects from this would be best to place for this patient before the admis-
patients therapy? sion order?
A. WBC A. Magnesium 1 g intravenously over 30 minutes
B. Serum potassium concentration before transfer
C. Platelet count B. Dextrose 5% and sodium chloride 0.9% with
D. Serum sodium concentration potassium chloride 20 mEq/L at 70 mL/hour
4. A 4-year-old girl (weight 14.7 kg) with a medical history of C. Increase continuous albuterol nebulization to
mild persistent asthma presents to the ED in acute respi- 30 mg/hour
ratory distress (increased work of breathing, shortness of D. Methylprednisolone 30 mg intravenously every
breath, respirations 48 breaths/minute, Sao2 93% on room 12 hours

PedSAP 2016 Book 2 Pediatric Critical Care 104 Interactive Patient Case: Severe Acute Asthma
Questions 8 and 9 pertain to the following case. C. Children with severe acute asthma who were treated
T.R. is an 8-year-old boy (weight 36 kg) with a medical his- with intravenous magnesium sulfate in the ED had
tory of mild persistent asthma. He is admitted for lack of reduced admission rates but did not have clinical
response to severe acute asthma standard therapy in the asthma scores in the inpatient setting consistently.
ED. He received albuterol with ipratropium nebulization D. The dose was below the recommended weight-
for three doses, prednisone 30 mg by mouth once, contin- based dose and should be repeated regardless of
uous albuterol nebulization at a dose of 15 mg/hour, and the treatment setting to improve clinical asthma
magnesium sulfate intravenously 1 g over 30 minutes. He scores.
has no known drug allergies. T.R.s home regimen includes
10. Which one of the following best describes the role of neb-
albuterol MDI 24 puffs as needed for wheezing and beclo-
ulized magnesium therapy in severe acute asthma?
methasone 40-mcg inhalation twice daily. Results of the
laboratory tests obtained on his arrival to the inpatient unit A. Nebulized magnesium can replace nebulization of
are as follows: albuterol and ipratropium.
B. A nebulized dosage form of magnesium is
145 | 102 | 12 / 145
commercially available.
2.9 | 18 | 0.67 \ calcium 8.9 mg/dL, magnesium 2.3 mg/dL
C. Clinical asthma score may improve with nebulized
T.R.s physical examination on admission revealed a heart magnesium when administered with standard
rate of 128 beats/minute, blood pressure 98/68 mm Hg, respi- therapy.
ratory rate 36 breaths/minute, temperature 98.2F (36.8C), D. Nebulized magnesium reduces hospital admission
and Sao2 91% on 100% fraction of inspired oxygen (Fio2) rates when used in children with severe acute
delivered at 2 L/minute with albuterol by a large-volume neb- asthma.
ulizer. Wheezing persisted during the prolonged expiratory
11. A 7-year-old girl (weight 34 kg) with a medical history
phase, and accessory muscles are in use. Breath sounds are
of mild persistent asthma, seasonal allergies, eczema,
decreased at the bases, and he speaks in phrases when he
and gastroesophageal disease was hospitalized 1 year
takes off the mask. T.R.s asthma score placed him in mod-
ago for an asthma exacerbation caused by viral pneu-
erate severity.
monia. She was admitted to the pediatric ICU for the
8. On arrival at the inpatient unit, which one of the follow- management of severe acute asthma after an inade-
ing is best to recommend for T.R.s fluid and electrolyte quate response to standard therapy and magnesium
management? intravenously 1 g in the ED. She received two doses of
A. Dextrose 5% and sodium chloride 0.9% with terbutaline subcutaneously and a continuous albuterol
potassium chloride 20 mEq/L continuous infusion at nebulization and was switched to terbutaline intravenous
76 mL/hour continuous infusion at 1 mcg/kg/minute. The patient is
B. Potassium chloride 20 mEq/100 mL intravenous now receiving BiPAP (bilevel positive airway pressure)
rider over 2 hours 100% Fio2 delivered by a non-rebreather mask. Her most
C. Sodium chloride 0.9% continuous infusion at 76 mL/ recent evaluation classifies her asthma as severe. The
hour attending physician is considering intubation and is will-
D. Dextrose 5% with potassium chloride 20 mEq/L ing to discuss options to avoid intubation. Which one of
continuous infusion at 76 mL/hour the following is best to share with this patients attend-
ing physician?
9. T.R.s attending physician asks whether the magnesium
sulfate intravenous dose should be repeated because A. Isoflurane has been used with success in a few
the serum concentration after the initial infusion is 2.3 children with severe refractory asthma and is an
mg/dL and symptoms persist. Which one of the following option for this patient.
is the best response to give T.R.s physician? B. Ketamine has been used to avoid intubation in a few
case reports and is an option for this patient.
A. Continuous magnesium intravenous infusion can
C. Heliox has improved the delivery of albuterol
achieve higher serum concentrations and has
nebulization and is an option for this patient.
proved more effective than intermittent magnesium
D. Proceed with intubation because no other
dosing in improving clinical asthma scores.
intervention is beneficial at this time.
B. Use of intravenous magnesium sulfate for severe
acute asthma management improves clinical 12. Which one of the following best describes the role of ami-
asthma scores in all health care settings. nophylline in the treatment of severe acute asthma?

PedSAP 2016 Book 2 Pediatric Critical Care 105 Interactive Patient Case: Severe Acute Asthma
A. Recent literature concluded aminophylline provides
no benefit in the treatment of severe acute asthma
in children.
B. When initiating aminophylline intravenously in a
patient receiving theophylline, the dose should be
80% of the oral theophylline dose.
C. The Expert Panel Review 3 recommends
aminophylline for severe acute asthma but only
in the ICU setting and only in intubated pediatric
patients.
D. Aminophylline delays improvement in clinical
asthma scores and lengthens hospital admission
when used for severe acute asthma in children.

PedSAP 2016 Book 2 Pediatric Critical Care 106 Interactive Patient Case: Severe Acute Asthma
Learner Evaluation: Interactive Case: Severe Acute Asthma (Status Asthmaticus).

As you take the posttest for this chapter, also evaluate the 10. The learning assessment activities used in the chapter
materials quality and usefulness, as well as the achievement were effective.
of learning objectives. Rate each item using this 5-point scale: 11. The chapter was effective overall.

Strongly agree
Use the 5-point scale to indicate whether this chapter pre-
Agree pared you to accomplish the following learning objectives:
Neutral
12. Devise an appropriate -agonist regimen based on pre-
Disagree sentation of a pediatric patient with severe acute asthma.
Strongly disagree
13. Justify the use of ipratropium as adjunct therapy for pedi-
atric patients with severe acute asthma.
1. The content of the chapter met my educational needs.
14. Identify the most appropriate magnesium sulfate regimen
2. The content of the chapter satisfied my expectations. for severe acute asthma in pediatric patients.
3. The author presented the chapter content effectively. 15. Evaluate the benefits and limitations of various treatment
4. The content of the chapter was relevant to my practice and modalities for severe acute asthma management in pedi-
presented at the appropriate depth and scope. atric patients.
5. The content of the chapter was objective and balanced. 16. Design a stepwise approach to escalate therapy for a child
with severe acute asthma refractory to standard of care.
6. The content of the chapter is free of bias, promotion, or
advertisement of commercial products. 17. Please provide any specific comments related to any
perceptions of bias, promotion, or advertisement of com-
7. The content of the chapter was useful to me.
mercial products.
8. The teaching and learning methods used in the chapter
18. Please expand on any of your above responses, and/or
were effective.
provide any additional comments regarding this chapter:
9. 
The active learning methods used in the chapter were
effective.

PedSAP 2016 Book 2 Pediatric Critical Care 107 Interactive Patient Case: Severe Acute Asthma
Guideline Review: Diabetic
Ketoacidosis and Hyperglycemic
Hyperosmolar State
By Elizabeth S. Goswami, Pharm.D., BCPS, BCPPS; and Susan Warrington, Pharm.D., BCPPS

Reviewed by Julie Pingel, Pharm.D.; Ifeyinwa D. Eboh, Pharm.D., MPH, BCPS; and Monica A. Puebla, Pharm.D., MBA, MHA, BCPS

LEARNING OBJECTIVES

1. Classify the severity of diabetic ketoacidosis (DKA) using patient-specific biochemical data.
2. Distinguish between DKA and hyperosmolar hyperglycemic state (HHS) with regard to pathophysiology and presentation.
3. Devise a strategy for fluid and electrolyte management in a pediatric patient with DKA.
4. Develop a plan for insulin therapy in a pediatric patient with DKA.
5. Evaluate a pediatric patient with DKA for risk factors and clinical signs or symptoms of cerebral edema and recommend
appropriate therapy.
6. Construct an argument against controversial, ineffective, or harmful therapies for DKA.

GUIDELINE REVIEW
ABBREVIATIONS IN THIS FEATURE
Topic Article
BOHB -hydroxybutyrate
CVC Central venous catheter Wolfsdorf JI, Allgrove J, Craig ME, et al. ISPAD Clinical Practice
Consensus Guidelines 2014 Compendium. Diabetic ketoacidosis
DKA Diabetic ketoacidosis
and hyperglycemic hyperosmolar state. Ped Diab 2014:15(Suppl.
HHS Hyperglycemic hyperosmolar 20):154-79.
state
ISPAD International Society for Pediatric
and Adolescent Diabetes On-demand Webcast
VTE Venous thromboembolism Click here to begin this PedSAP activity.
Click here to view a transcription of this recorded webcast.
Table of other common abbreviations.

BASELINE KNOWLEDGE STATEMENTS

Readers of this chapter are presumed to be familiar


with the following:
General knowledge of pathophysiology and treat-
ment of both type 1 and type 2 diabetes mellitus.
Normal-for-age laboratory reference ranges and
vital signs for pediatric patients
Pharmacokinetics and pharmacodynamics of
various insulin products.
Fluid and electrolyte management for pediatric patients.

Table of common pediatric laboratory reference values.

PedSAP 2016 Book 2 Pediatric Critical Care 109 Guideline Review: Diabetic Ketoacidosis
REFERENCES
ADDITIONAL READINGS Bagdure D, Rewers A, Campagna E, et al. Epidemiology of
hyperosmolar hyperglycemic syndrome in children hospi-
The following free resources have additional back- talized in USA. Pediatr Diabetes 2013;14:18-24.
ground information on this topic:
Carlotti AP, St George-Hyslop C, Bohn D, et al. Hypokalemia
American Diabetes Association. Standards of
during treatment of diabetic ketoacidosis: clinical evi-
medical care in diabetes 2016: 11. Children and
dence for an aldosterone-like action of insulin. J Pediatr
adolescents. Diabetes Care
2013;163:201-12.e1.
2016;39(suppl1):S86-S93.
Danne T, Bangstad H, Deeb L, et al. ISPAD Clinical Chua H, Venkatesh B, Stachowski E, et al. Plasma-Lyte 148
Practice Consensus Guidelines 2014. Insulin vs 0.9% saline for fluid resuscitation in diabetic ketoacido-
treatment in children and adolescents with sis. J Crit Care 2012;27:138-45.
diabetes. Pediatric Diabetes
Decourcey DD, Steil GM, Wypij D, et al. Increasing use of
2014;15(suppl20):115-34.
hypertonic saline over mannitol in the treatment of symp-
Springer SC, Silverstein J, Copeland K, et al. tomatic cerebral edema in pediatric diabetic ketoacidosis:
Management of type 2 diabetes mellitus in children an 11-year retrospective analysis of mortality. Pediatr Crit
and adolescents. Pediatrics 2013;131:e648-e664. Care Med 2013;14:694-700.
Chafe R, Albrechtsons D, Hagerty D, et al. Reducing
episodes of diabetic ketoacidosis within a youth Fiordalisi I, Novotny WE, Holbert D, et al. An 18-yr prospec-
population: a focus group study with patients and tive study of pediatric diabetic ketoacidosis: an approach
families. BMC Res Notes 2015;8:395. to minimizing the risk of brain herniation during treatment.
Pediatr Diabetes 2007;8:142-9.

Glaser NS, Ghetti S, Casper TC, et al. Pediatric diabetic keto-


acidosis, fluid therapy and cerebral injury: the design of
Practice Points a factorial randomized controlled trial. Pediatr Diabetes
DKA 2013;14:435-46.
DKA results from an absolute or relative insulin deficiency. Glaser NS, Wooton-Gorges WL, Buonocore MH, et al.
The severity of DKA varies, depending on the degree of Subclinical cerebral edema in children with diabetic keto-
acidosis.
acidosis randomized to 2 different rehydration protocols.
Patients with DKA have a 5%10% fluid deficit and require
Pediatrics 2013;131:e73-80.
fluid replacement.
Insulin at 0.050.1 unit/kg/hr is needed to reverse ketosis. Grimberg A, Cerri RW, Satin-Smith M, et al. The two bag
Patients will require replacement of potassium and may system for variable intravenous dextrose and fluid admin-
require phosphate replacement. istration: benefits in diabetic ketoacidosis management. J
Patients with DKA should not be administered these Pediatr 1999;134:376-8.
therapies: insulin boluses, rapid fluid boluses (except
in hemodynamic instability), and bicarbonate (except in Nallasamy K, Jayashree M, Singhi S, et al. Low-dose vs stan-
life-threatening hyperkalemia). dard-dose insulin in pediatric diabetic ketoacidosis. JAMA
HHS Pediatr 2014;168:999-1005.
HHS is rare in pediatric patients, but it is increasing in oc-
Rosenbloom AL. The management of diabetic ketoacidosis
currence.
in children. Diabetes Ther 2010;1:103-20.
HHS presents as extreme hyperglycemia and hyperosmo-
lality without ketosis. So T, Gruenwalder E. Evaluation of the two-bag system for
Patients with HHS are extremely dehydrated (1215%) fluid management in pediatric patients with diabetic keto-
from osmotic diuresis and require significant fluid and acidosis. J Pediatr Pharmacol Ther 2009;14:100-5.
electrolyte supplementation.
Insulin should not be administered to patients with HHS Sottosanti M, Morrison GC, Singh RN, et al. Dehydration in
until the rate of blood glucose decline becomes inadequate children with diabetic ketoacidosis: a prospective study.
with fluid administration alone. Arch Dis Child Ed 2012;97:96-100.

Ugale J, Mata A, Meert KL, et al. Measured degree of dehy-


dration in children and adolescents with type 1 diabetic
ketoacidosis. Pediatr Crit Care Med 2012;13:e103-7.

Watts W, Edge JA. How can cerebral edema during treatment


of diabetic ketoacidosis be avoided. Pediatr Diabetes
2014;15:271-6.

PedSAP 2016 Book 2 Pediatric Critical Care 110 Guideline Review: Diabetic Ketoacidosis
Wolfsdorf JI. The International Society of Pediatric and Wolfsdorf JI, Allgrove J, Craig ME, et al. ISPAD Clinical
Adolescent Diabetes guidelines for management of dia- Practice Consensus Guidelines 2014. Diabetic ketoac-
betic ketoacidosis: do the guidelines need to be modified? idosis and hyperglycemic hyperosmolar state. Pediatr
Pediatr Diabetes 2014;15:277-86. Diabetes 2014;15(suppl 20):154-79.

Wolfsdorf J, Glaser N, Sperling MA. Diabetic ketoacidosis in Zeitler J, Haqq A, Rosenbloom A, et al. Hyperglycemic
infants, children, and adolescents. A consensus statement hyperosmolar syndrome in children: pathophysiological
from the American Diabetes Association. Diabetes Care considerations and suggested guidelines for treatment. J
2006;29:1150-9. Pediatr 2011;158:9-14.

PedSAP 2016 Book 2 Pediatric Critical Care 111 Guideline Review: Diabetic Ketoacidosis
Self-Assessment Questions
Questions 1315 pertain to the following case. 17. You are the pediatric clinical pharmacist at a community
K.C. is an 11-year-old boy (weight 34 kg) with a history of hospital that cares for both adult and pediatric patients;
type 1 diabetes and asthma. He presents to the ED with it has 25 pediatric acute care beds and a level III neo-
a 2-day history of increased thirst and urination. K.C. has natal ICU. Your hospital is undergoing an expansion that
vomited five times today. He has no fever and no history will include an eight-bed pediatric ICU. Your hospital pre-
of recent illness. His insulin is administered by an insulin viously referred patients with DKA to a nearby tertiary
pump. At presentation, the following laboratory values are pediatric hospital in the same health system. However,
obtained: after the expansion, the hospital plans to accept these
patients into the pediatric ICU and will have pediatric
Venous pH Na K Cl Bicarbonate SCr Glucose endocrinologists available to consult. Which one of the
6.98 128 5.1 97 4 mEq/L 1.02 435 following would best ensure the safe and appropriate
mEq/L mEq/L mEq/L mg/dL mg/dL care of pediatric patients with DKA at your institution?
A. Assist in developing an educational inservice for the
Urinalysis shows 3+ ketones, and point-of-care -hydroxybu-
staff pharmacists verifying orders for the pediatric
tyrate (BOHB) is 7.5 mmol/L.
ICU.
13. Which one of the following is most likely the cause of B. Assist in developing institutional guidelines for the
K.C.s osmotic diuresis? care of children with DKA.
C. Ensure that intravenous fluids are administered by a
A. Glucosuria
two-bag system instead of a one-bag system.
B. Glucosuria and ketonuria
D. Reach out to the pediatric ICU pharmacist at
C. Ketonuria
the tertiary pediatric hospital as a resource for
D. Antidiuretic hormone deficiency
complicated clinical questions.
14. Given the available laboratory data, which one of the
18. A 6-year-old boy (weight 25 kg) presents to the ED with
following best classifies the severity of K.C.s diabetic
new diagnosis of type 1 diabetes and moderate DKA. He
ketoacidosis (DKA)?
is alert and oriented with vomiting and signs of dehydra-
A. Mild tion. His capillary refill is 3 seconds and blood pressure
B. Moderate is 96/52. Venous pH is 7.13. He was urinating frequently
C. Severe for the past 4 days, but has only urinated twice in the
D. Ultra-severe past 12 hours. Which one of the following is best to rec-
15. Which one of the following best describes K.C.s acid- ommend for initial fluid resuscitation in this patient?
base status and its most likely cause? A. Administer 20 mL/kg 0.9% NaCl as rapidly as
A. Anion gap metabolic acidosis; hyperglycemia possible and reassess hemodynamic status.
B. Anion gap metabolic acidosis; ketonemia B. Administer 40 mL/kg 0.9% NaCl over 2 hours and
C. Non-anion gap metabolic acidosis; ketonemia reassess hemodynamic status.
D. Anion gap respiratory acidosis; hyperglycemia C. Administer 10 mL/kg 0.9% NaCl as rapidly as
possible and reassess hemodynamic status.
16. A 3-year-old girl presents with mild DKA and new-onset
D. Administer 20 mL/kg 0.9% NaCl over 2 hours and
type 1 diabetes. On physical examination, her capillary
reassess hemodynamic status.
refill is 4 seconds, and she shows tenting when her skin
turgor is examined. Her respiratory rate is 57 breaths/ Questions 19 and 20 pertain to the following case.
minute. Both her central and peripheral pulses are pal-
T.E. is an 8-year-old girl (height 51 inches [130 cm], weight
pable, and her blood pressure is normal for age. She
27 kg) who is admitted to the pediatric ICU with moderate
has had frequent urination for the past 3 days, and she
DKA. The ED staff estimates her fluid deficit at 5% and orders
just had a 3-mL/kg urine void before the physical exam-
a 10-mL/kg bolus of NaCl 0.9% over 2 hours. T.E. is quickly
ination. Which one of the following best estimates this
transferred to the pediatric ICU. After the fluid bolus, she
patients fluid deficit?
is initiated on intravenous fluids using the two-bag system
A. 5% and continuous insulin infusion. At presentation, her blood
B. 10% glucose was 550 mg/dL; this decreased to 489 mg/dL after
C. 12% 1 hour of the fluid bolus, and is now 402 mg/dL at the end of
D. 15% the 2-hour fluid bolus.

PedSAP 2016 Book 2 Pediatric Critical Care 112 Guideline Review: Diabetic Ketoacidosis
19. Which one of the following best represents an appropri- which one of the following is the optimal fluid regimen
ate total hourly intravenous fluid rate for T.E.? adjustment for this patient?
A. 23 mL/hr A. Reduce both fluid A and fluid B to 31 mL/hr.
B. 67 mL/hr B. Reduce fluid A to 31 mL/hr, and keep fluid B at
C. 90 mL/hr 47 mL/hr.
D. 180 mL/hr C. Discontinue fluid A, and increase fluid B to 94 mL/hr.
D. Increase fluid A to 94 mL/hr, and discontinue fluid B.
20. Which one of the following maintenance intravenous
solutions is best for T.E. for the first 4 hours after fluid 23. Eighteen hours ago, a 7-year-old boy (weight 25 kg) with a
resuscitation? history of type 1 diabetes and asthma was admitted to the
pediatric ICU with DKA. His current medications include:
A. NaCl 0.45%
B. NaCl 0.9%
Dextrose 12.5% plus NaCl 0.45% plus 40 mEq/L of potas-
C. Dextrose 5%, NaCl 0.45%
sium acetate at 110 mL/hr; regular insulin infusion at 0.1
D. Dextrose 5%, NaCl 0.9%
unit/kg/hr; and fluticasone (100 mcg/puff) 2 puffs twice
21. You work in an ED at a small community hospital that daily. His most recent laboratory values are as follows:
admits both adult and pediatric patients. A 2-year-old
boy (weight 14 kg) presents with DKA and new-onset dia- Venous pH Na K Cl Bicarbonate Glucose BOHB
betes. Urinalysis is positive for large ketones, and initial
7.27 137 3.2 110 11 mEq/L 55 mg/ 4.2
laboratory results are as follows.
mEq/L mEq/L mEq/L dL mmol/L

Venous pH Na K Cl Bicarbonate SCr Glucose Which one of the following is best to recommend for this
7.04 134 3.2 97 4 mEq/L 0.5 700 patient?
mEq/L mEq/L mEq/L mg/dL mg/dL A. Change fluids to dextrose 15% plus NaCl 0.45% plus
40 mEq/L of potassium acetate at 110 mL/hr.
The physician orders a bolus of regular insulin 0.1 unit/ B. Keep the same fluids, but change the rate to
kg intravenously 1 after initial fluid resuscitation. 165 mL/hr.
Which one of the following is best to recommend for this C. Change the insulin rate to 0.05 unit/kg/hr.
patient? D. Transition to subcutaneous insulin and enteral diet.

A. Prepare a 10-unit/mL dilution of insulin so that 24. The clinical pharmacy manager at your institution has
the dose can be accurately measured in a 1-mL assigned you to provide clinical management of a potas-
intravenous syringe. sium phosphate drug shortage. Currently, the DKA order
B. Recommend 0.05 unit/kg as an appropriate set has a two-bag system with the following fluids:
intravenous insulin bolus for a patient with DKA, and
Fluid A: NaCl 0.9% plus 20 mEq/L of potassium chloride
ask the physician to change the order.
plus 15 mmol/L of potassium phosphate
C. Refuse to dispense the insulin dose because
intravenous insulin boluses increase the risk of Fluid B: dextrose 10% plus NaCl 0.9% plus 20 mEq/L
cerebral edema, and ask the physician to change the of potassium chloride plus 15 mmol/L of potassium
order. phosphate
D. Refuse to dispense the insulin dose until potassium
is above 3.5 mEq/L, and ask the physician to change Which one of the following would you recommend as the
the order. best option for conserving potassium phosphate?
22. You are the clinical pharmacist in the pediatric ICU at A. Make no changes, and prioritize potassium
an institution that uses the two-bag system. Your proto- phosphate supply to use for DKA intravenous fluids.
col includes lactated Ringer solution with 40 mEq/L of B. Remove potassium phosphate from intravenous
potassium chloride for fluid A and dextrose 10% lactated fluids on the DKA order set, and make no additional
Ringer solution with 40 mEq/L of potassium chloride changes.
for fluid B. You are caring for a 4-year-old girl (weight 23 C. Remove potassium phosphate from intravenous
kg) with a history of diabetes who was admitted over- fluids on the DKA order set, and replace with an
night with DKA. Her glucose is currently 90 mg/dL. Her additional 20 mEq/L of potassium chloride.
blood glucose 1 hour earlier was 210 mg/dL. She is cur- D. Remove potassium phosphate from intravenous
rently receiving fluid A at a rate of 47 mL/hr and fluid B fluids on the DKA order set, and replace with 20
at 47 mL/hr. Given her new blood glucose concentration, mEq/L of potassium acetate.

PedSAP 2016 Book 2 Pediatric Critical Care 113 Guideline Review: Diabetic Ketoacidosis
25. You are working in the ED at a community hospital that Questions 2831 pertain to the following case.

admits both pediatric and adult patients. An 8-year-old R.T. is a 15-year-old boy (weight 90 kg) with a history of type
girl (weight 46 kg) presents in severe DKA. Results of her 2 diabetes who presents to the ED with the following labora-
laboratory tests are as follows: tory data:

Venous pH Na K Cl Bicarbonate Glucose BOHB Venous pH Na K


6.99 132 2.1 98 4 mEq/L 609 8.2 7.31 150 mEq/L 1.3 mEq/L
mEq/L mEq/L mEq/L mg/dL mmol/L
Phosphorus Magnesium Serum
Lactate osmolality
2.1 mmol/L 1.2 mg/dL 1.1 mEq/L 361 mOsm/
kg
The physician asks for your recommendation regarding
Cl Bicarbonate Glucose BOHB
bicarbonate administration for this patient. Which one of
the following is best to recommend for this patient? 122 28 mEq/L 988 mg/ < 0.3
mEq/L dL mmol/L
A. Do not administer bicarbonate because of the risk of
cerebral edema. R.T. states that he has been urinating more over the past
B. Do not administer bicarbonate because of the risk of week or two. His blood pressure is 74/56 mm Hg.
hyperkalemia.
C. Administer bicarbonate to reverse severe 28. Which one of the following best represents R.T.s fluid
ketoacidosis. deficit?
D. Administer bicarbonate to reverse severe lactic
A. 5L
acidosis.
B. 9L
26. You are reviewing patients for inclusion in a retrospective C. 12 L
quality improvement project for patients with cere- D. 20 L
bral edema and DKA. The projects definition for DKA
29. After initial fluid resuscitation, R.T.s blood pressure has
matches the criteria for clinical diagnosis referenced by
improved to 98/68 mm Hg. He has ongoing osmotic
the DKA guidelines. Which one of the following patients
diuresis. Which one of the following is best to rec-
would be best to include in the study?
ommend regarding the management of R.T.s urinary
A. A 3-year-old girl with fluctuating mentation and replacement?
vomiting
A. Do not replace urinary losses
B. A 10-year-old boy with fluctuating mentation and
B. Replace urinary losses with NaCl 0.225%
lethargy
C. Replace urinary losses with NaCl 0.45%
C. An 8-year-old girl with fluctuating mentation and
D. Replace urinary losses with NaCl 0.9%
headache
D. A 6-year-old boy with fluctuating mentation and 30. Fluid administration is initiated for R.T. One hour later,
diastolic blood pressure 92 mm Hg you are asked to recommend a dose for R.T.s insulin infu-
sion. Which one of the following is best to recommend
27. Six hours after starting therapy for DKA, a 12-year-old
regarding insulin therapy for R.T.?
boy with known type 1 diabetes begins to have decorti-
cate posturing. There is immediate concern for cerebral A. Initiate insulin at 0.1 unit/kg/hr when blood glucose
edema. Which one of the following is best to recommend decline is less than 50 mg/dL/hr with fluid therapy.
for this patient? B. Initiate insulin at 0.05 unit/kg/hr when blood
glucose decline is less than 50 mg/dL/hr with fluid
A. Infuse 5 mL/kg of NaCl 3% IV immediately after head
therapy.
CT results return
C. Initiate insulin at 0.1 unit/kg/hr at this time.
B. Infuse 1 g/kg of mannitol IV immediately after
D. Initiate insulin at 0.05 unit/kg/hr at this time.
stopping continuous fluid replacement
C. Infuse 2.5 mL/kg of NaCl 3% IV immediately after 31. Which one of the following is best to recommend for the
continuous replacement fluids are decreased to 1 to treatment of R.T.s hypophosphatemia?
1.3x maintenance A. Administer 0.3 mmol/kg of potassium phosphate
D. No interventions are required at this time. Continue to intravenously now.
monitor the patient and treat only if enough major and B. Administer 0.3 mmol/kg of potassium phosphate
minor criteria for diagnosis of cerebral edema are met intravenously when R.T. becomes symptomatic from
hypophosphatemia.

PedSAP 2016 Book 2 Pediatric Critical Care 114 Guideline Review: Diabetic Ketoacidosis
C. Use replacement fluids containing 20 mEq/L of
potassium chloride and 15 mmol/L of potassium
phosphate now.
D. Use replacement fluids containing 20 mEq/L of
potassium chloride and 15 mmol/L of potassium
phosphate when R.T. becomes symptomatic from
hypophosphatemia.
32. A 13-year-old girl (weight 38 kg) has a history of type 1 dia-
betes diagnosed at age 5. She presents to the ED with a
1-day history of abdominal pain, increased urination, and
vomiting. She has no history of diarrhea or fever. Her labora-
tory values are consistent with moderate DKA. Her parents
state they have recently been concerned that she has devel-
oped an eating disorder because she is not gaining weight
and has been very selective in what she eats. They have
scheduled an appointment with her regular pediatrician
next week. The patients home insulin regimen includes
insulin glargine at bedtime and insulin lispro for correction
and at mealtimes. She administers insulin on her own with-
out supervision. This is her third DKA episode in 12 months.
The patients family is in a high socioeconomic class, and
she has not missed any scheduled appointments. The fam-
ily has private insurance. There are no concerns for abuse
or unstable home situation. Which one of the following is
most likely contributing to this patients recurrent DKA?
A. Insulin pump failure
B. Lack of access to medical care
C. Gastroenteritis
D. Insulin omission

PedSAP 2016 Book 2 Pediatric Critical Care 115 Guideline Review: Diabetic Ketoacidosis
Learner Evaluation: Guideline Review: Diabetic Ketoacidosis and Hyperglycemic
Hyperosmolar State.

As you take the posttest for this chapter, also evaluate the Use the 5-point scale to indicate whether this chapter pre-
materials quality and usefulness, as well as the achievement pared you to accomplish the following learning objectives:
of learning objectives. Rate each item using this 5-point scale: 30. Classify the severity of diabetic ketoacidosis (DKA) using
patient-specific biochemical data.
Strongly agree 31. 
Distinguish between DKA and hyperosmolar hypergly-
Agree cemic state (HHS) with regard to pathophysiology and
Neutral presentation.
Disagree 32. Devise a strategy for fluid and electrolyte management in
Strongly disagree a pediatric patient with DKA.
33. Develop a plan for insulin therapy in a pediatric patient
19. The content of the chapter met my educational needs. with DKA.
20. The content of the chapter satisfied my expectations. 34. Evaluate a pediatric patient with DKA for risk factors and
21. The author presented the chapter content effectively. clinical signs or symptoms of cerebral edema and recom-
mend appropriate therapy.
22. The content of the chapter was relevant to my practice
and presented at the appropriate depth and scope. 35. Construct an argument against controversial, ineffective,
or harmful therapies for DKA.
23. The content of the chapter was objective and balanced.
36. Please provide any specific comments related to any
24. The content of the chapter is free of bias, promotion, or
perceptions of bias, promotion, or advertisement of com-
advertisement of commercial products.
mercial products.
25. The content of the chapter was useful to me.
37. Please expand on any of your above responses, and/or
26. The teaching and learning methods used in the chapter provide any additional comments regarding this chapter:
were effective.
27. The active learning methods used in the chapter were
effective.
28. The learning assessment activities used in the chapter
were effective.
29. The chapter was effective overall.

PedSAP 2016 Book 2 Pediatric Critical Care 116 Guideline Review: Diabetic Ketoacidosis
Interactive Case: Status Epilepticus

By Christina Cox, Pharm.D., BCPS, BCPPS

Reviewed by Emily L. Rowe, Pharm.D., BCPS, BCPPS, MS-PREP; and Maha O. Kebir, Pharm.D., MS, BCPS, CPHQ

LEARNING OBJECTIVES

1. Distinguish the cause of status epilepticus (SE) in neonates, infants, children, and adolescents.
2. Design an appropriate treatment plan for the pediatric patient with SE or refractory SE.
3. Design a treatment strategy for the patient with complications of SE, including hypoxemia and acidosis.
4. Evaluate the patient with SE for the development of long-term sequelae of prolonged seizure activity and/or treatment
of SE.

INTERACTIVE CASE
ABBREVIATIONS IN THIS FEATURE
Click here to begin this PedSAP activity.
AES American Epilepsy Society
BMP Basic metabolic panel
CSE Convulsive status epilepticus
REFERENCES
EEG Electroencephalograph
Abend NS, Monk HM, Licht DJ, et al. Intravenous levetiracetam in
HSV Herpes simplex virus critically ill children with status epilepticus or acute repetitive sei-
ICP Intracranial pressure zures. Pediatr Crit Care Med. 2009;10(4):505-10.
LP Lumbar puncture
Abend NS, Dlugos DJ. Treatment of refractory status epilepti-
NCSE Non-convulsive status
cus: literature review and a proposed protocol. Pediatr Neurol
epilepticus
2008;38:377-90.
NMDA N-methyl-d-aspartate
RSE Refractory status epilepticus American Academy of Pediatrics Committee on Drugs. Inactive
SE Status epilepticus ingredients in pharmaceutical products: update (subject review).
American Academy of Pediatrics Committee on Drugs. Pediatrics
1997;99:268-78.
Table of other common abbreviations.
Appleton R, Macleod S, Martland T. Drug management for acute ton-
ic-clonic convulsions including convulsive status epilepticus in
children. Cochrane Database Syst Rev. 2008;16;(3): CD001905.

Argawal P, Kumar N, Chandra R, et al. Randomized study of intra-


venous valproate and phenytoin in status epilepticus. Seizure
2007;16:527-32.

Babl FE, Sheriff N, Borland M, et al. Emergency management of pae-


diatric status epilepticus in Australia and New Zealand: practice
patterns in the context of clinical practice guidelines. J Paediatr
Child Health. 2009;45:541-6.

Barberio M, Reiter PD, Kaufman J, et al. Continuous infusion pen-


tobarbital for refractory status epilepticus in children. J Child
Neurol 2012;27:721-6.

PedSAP 2016 Book 2 Pediatric Critical Care 117 Interactive Case: Status Epilepticus
Bleck TP. Management approaches to prolonged sei- Hanhan UA, Fiallos MR, Orlowski JP. Status epilepticus.
zures and status epilepticus. Epilepsia 1999;40(suppl Pediatr Clin North Am 2001;48:683-94.
1):S59-S63.
Harbord MG, Kyrkou NE, Kyrkou MR, et al. Use of intranasal
Brevoord JC, Joosten KF, Arts WF, et al. Status epilepticus: midazolam to treat acute seizures in paediatric commu-
clinical analysis of a treatment protocol based on midaz- nity settings. J Paediatr Child Health 2004;40:556-8.
olam and phenytoin. J Child Neurol 2005;20:476-81.
Harrison AM, Lugo RA, Schunk JE. Treatment of convul-
Brophy GM, Bell R, Claassen J, et al. Guidelines for the evalu- sive status epilepticus with propofol: case report. Pediatr
ation and management of status epilepticus. Neurocritical Emerg Care 1997;13:420-2.
Care 2012;17:3-23.
Haut SR, Shinnar S , Moshe SL, et al. The association
Chamberlain JM, Okada P, Holsti M, et al. Lorazepam vs between seizure clustering and convulsive status epilep-
diazepam for pediatric status epilepticus: a randomized ticus in patients with intractable complex partial seizures.
clinical trial. JAMA 2014;311:1652-60. Epilepsia 1999;40:1832-4.

Chin RF, Neville BG, Peckham C, et al. Treatment of com- Hayashi K, Osawa M, Aihara M, Research Committee
munity-onset, childhood convulsive status epilepticus: on Clinical Evidence of Medical Treatment for Status
a prospective, population-based study. Lancet Neurol Epilepticus in Childhood, et al. Efficacy of intravenous mid-
2008;7(8):696-703. azolam for status epilepticus in childhood. Pediatr Neurol
2007;36:366-72.
Chin RF, Neville BG, Peckham C, NLSTEPSS Collaborative
Group, et al. Incidence, cause, and short-term outcome of Hellstrm-Westas L, Boylan G, Agren J. Systematic review
convulsive status epilepticus in childhood: prospective of neonatal seizure management strategies provides
population-based study. Lancet 2006;368:222-9. guidance on anti-epileptic treatment. Acta Paediatr
2015;104:123-9.
Claassen J, Hirsch LJ, Emerson RG, et al. Treatment
of refractory status epilepticus with pentobarbital, Hofler J, Trinka E. Lacosamide as a new treatment option in
propofol, or midazolam: a systemic review. Epilepsia status epilepticus. Epilepsia 2013;54:393-404.
2001;43:146-53.
Holsti M, Sill BL, Firth SD, et al. Prehospital intranasal mid-
Claassen J, Hirsch LJ, Mayer SA. Treatment of status azolam for the treatment of pediatric seizures. Pediatr
epilepticus: a survey of neurologists. J Neurol Sci. Emerg Care 2007;23:148-53.
2003;211:37-41
Igartua J, Silver P, Maytal J, et al. Midazolam coma for
DeLorenzo R, Hauser WA, Towne AR, et al. A prospective, refractory status epilepticus in children. Crit Care Med
population-based epidemiologic study of status epilepti- 1999;27:1982-5.
cus in Richmond, Virginia. Neurology 1996;46:1029-35.
Ilvento L, Rosati A, Marini C, et al. Ketamine in refractory
Eriksson K, Metsaranta P, Huhtala H, et al. Treatment delay convulsive status epilepticus in children avoids endotra-
and the risk of prolonged status epilepticus. Neurology cheal intubation. Epilepsy Behav 2015;49:343-6.
2005;65:1316-8.
Kim JS, Lee JH, Ryu HW, et al. Effectiveness of intravenous
Fang Y, Wang X. Ketamine for the treatment of refractory sta- levetiracetam as an adjunctive treatment in pediat-
tus epilepticus. Seizure 2015;30:14-20. ric refractory status epilepticus. Pediatr Emerg Care
2014;30(8):525-528.
Gallagher RC, Van Hove JL, Scharer G, et al. Folinic acid-re-
sponsive seizures are identical to pyridoxine-dependent Kim SJ, Lee DY, Kim JS. Neurologic outcomes of pediatric
epilepsy. Ann Neurol 2009;65:550-6. epileptic patients with pentobarbital coma. Pedi Neurol
2001;25:217-20.
Gallentine WB, Hunnicutt AS, Husain AM. Levetiracetam
in children with refractory status epilepticus. Epilepsy Koul R, Chacko A, Javed H, et al. Eight-year study of child-
Behav. 2009;14(1):215-8. hood status epilepticus; midazolam infusion management
and outcome. J Child Neurol 2002;17:908-10.
Glass HC, Kan J, Bonifacio SL et al. Neonatal seizures: treat-
ment practices among term and preterm infants. Pediatr Koul RL, Raj Aithala G, Chacko A, et al. Continuous midaz-
Neurol 2012;46:111-5. olam infusion as treatment of status epilepticus. Arch Dis
Child 1997;76:445-8.
Glauser T, Shinnar S, Gloss D, et al. Evidence-Based
Guideline: Treatment of Convulsive Status Epilepticus in Kravljanac R, Jovic N, Djuric M, et al. Outcome of status epi-
Children and Adults: Report of the Guideline Committee lepticus in children treated in the intensive care unit: a
of the American Epilepsy Society. Epilepsy Currents 2016; study of 302 cases. Epilepsia 2011;52:358.
16(1):48-61.
Lacroix J, Deal C, Gauthier M, et al. Admissions to a pediatric
Grosso S, Zamponi N, Bartocci A, et al. Lacosamide in chil- intensive care unit for status epilepticus: a 10-year experi-
dren with refractory status epilepticus. A multicenter ence. Crit Care Med 1994;22:827-32.
Italian experience. Eur J Paediatr Neurol 2014;18:604-8.

PedSAP 2016 Book 2 Pediatric Critical Care 118 Interactive Case: Status Epilepticus
Lambrechtsen FA, Buchhalter JR. Aborted and refractory Rosati A, LErario M, Ilvento L, et al. Efficacy and safety of
status epilepticus in children: a comparative analysis. ketamine in refractory status epilepticus in children.
Epilepsia 2008;49:615-25. Neurology 2012;79:2355-8.
Maytal J, Shinnar S, Moshe SL, et al. Low morbidity and Sahin M, Menache CC, Holmes GL, et al. Outcome of severe refrac-
mortality of status epilepticus in children. Pediatrics tory status epilepticus in children. Epilepsia 2001;42:1461-7.
1989;83:323-31.
Saz EU, Karapinar B, Ozcetin M, et al. Convulsive status
Mehta V, Singhi P, Singhi S. Intravenous sodium valproate epilepticus in children: etiology, treatment protocol and
versus diazepam infusion for the control of refractory sta- outcome. Seizure 2011;20:115-8.
tus epilepticus in children: a randomized controlled trial. J
Child Neurol 2007;22:1191-7. Shehab N, Lewis CL, Streetman DD, et al. Exposure to the
pharmaceutical excipients benzyl alcohol and propylene
Morrison G, Gibbons E, Whitehouse WP. High-dose midaz- glycol among critically ill neonates. Pediatr Crit Care Med
olam therapy for refractory status epilepticus in children. 2009;20:115-8.
Intensive Care Med 2006;32:2070-6.
Shiloh-Malawsky Y, Fan Z, Greenwood R, et al. Successful
Nicolai J, van Kranen-Mastenbroek VH, Wevers RA, et al. treatment of childhood prolonged refractory status epilep-
Folinic acid-responsive seizures initially responsive to pyr- ticus with lacosamide. Seizure 2011;20:586-8.
idoxine. Pediatr Neurol 2006;34:164-7.
Shinnar S, Berg AT, Moshe SL, et al. The risk of seizure recur-
Novak G, Maytal J, Alshansky A, et al. Risk factors for sta- rence after a first unprovoked afebrile seizure in childhood:
tus epilepticus in children with symptomatic epilepsy. an extended follow-up. Pediatrics 1996;98(pt 1):216-25.
Neurology 1997;49:533-7.
Shinnar S, Pellock JM, Berg AT, et al. Short-term outcomes
Ozdemir D, Gulez P, Uran N, et al. Efficacy of continuous of children with febrile status epilepticus. Epilepsia
midazolam infusion and mortality in childhood refrac- 2001;42:47-53.
tory generalized convulsive status epilepticus. Seizure
2005;14:129-32. Shorvon S. Super-refractory status epilepticus: an approach
to therapy in this difficult clinical situation. Epilepsia
Patten W, Naqvi SZ, Raszynski A, et al. Complications during 2011;52(suppl 8):53-6.
the management of pediatric refractory status epilepticus
with benzodiazepine and pentobarbital infusions. Indian J Shorvon SD. Status Epilepticus: Its Clinical Features and
Crit Care Med 2015;19:275-7. Treatment in Children and Adults. Cambridge: Cambridge
University Press, 1987.
Prasad A, Worrall BB, Bertram EH, et al. Propofol and mid-
azolam in the treatment of refractory status epilepticus. Sillanpaa M, Shinnar S. Status epilepticus in a popula-
Epilepsia 2001;42:380-6. tion-based cohort with childhood-onset epilepsy in
Finland. Ann Neurol 2002;52:303-10.
Ram D, Martland T. Management of convulsive status epilep-
ticus in children. Paediatr Child Health 2015;25:522-7. Singh RK, Stephens S, Berl MM, et al. Prospective study of
new-onset seizures presenting as status epilepticus in
Raspall-Chaure M, Chin RF, Neville BG, et al. Outcome of pae- childhood. Neurology 2010;74:636-42.
diatric convulsive status epilepticus: a systematic review.
Lancet Neurol 2006;5:769-79. Singhi S, Murthy A, Singhi P, et al. Continuous midazolam
versus diazepam infusion for refractory convulsive status
Redecker J, Wittstock M, Benecke R, et al. Comparison of the epilepticus. J Child Neurol 2002;17:106-10.
effectiveness of four antiepileptic drugs in the treatment
of status epilepticus according to four different efficacy Sofou K, Kristjansdorrir R, Papachatzakis NE, et al. Management
criteria. Epilepsy Behav 2015;49:351-3. of prolonged seizures and status epilepticus in childhood: a
systematic review. J Child Neurol 2009;24:918-26.
Reiter PD, Huff AD, Knupp KG, et al. Intravenous levetirac-
etam in the management of acute seizures in children. Stockler S, Plecko B, Gospe SM, et al. Pyridoxine dependent
Pediatr Neurol. 2010 Aug;43(2):117-21. epilepsy and antiquitin deficiency: clinical and molecular
characteristics and recommendations for diagnosis, treat-
Rivera R, Segnini M, Baltodano A, et al. Midazolam in the ment and follow-up. Mol Genet Metab 2011;104:48-60.
treatment of status epilepticus in children. Crit Care Med
1993;21:991-4. Synowiec AS, Singh DS, Yenugadhati V, et al. Ketamine use
in the treatment of refractory status epilepticus. Epilepsy
Riviello JJ, Ashwal, S, Hirtz D, et al. Practice parameter: diag- Res 2013;105:183-8.
nostic assessment of the child with status epilepticus (an
evidence-based review): report of the Quality Standards Tobias JD. Tolerance, withdrawal, and physical dependency
Subcommittee of the American Academy of Neurology after long-term sedation and analgesia of children Tobias
and the Practice Committee of the Child Neurology JD. Tolerance, withdrawal, and physical dependency after
Society. Neurology 2006;67:1542-50. long-term sedation and analgesia of children in the pedi-
atric in the pediatric intensive care unit. Crit Care Med
2000;23:2122-32.

PedSAP 2016 Book 2 Pediatric Critical Care 119 Interactive Case: Status Epilepticus
Tobias JD. The use of propofol to treat status epilepticus White JR, Santos CS. Intravenous valproate associated with
in a nine-month-old female patient. Pediatr Emerg Care significant hypotension in the treatment of status epilepti-
1998;14:248-9. cus. J Child Neurol 1999;14:822-3.
Trinka E, Cock H, Hesdorffer D, et al. A definition and clas- Wilkes R, Tasker RC. Intensive care treatment of uncontrolled
sification of status epilepticus - report of the ILAE Task status epilepticus in children: systematic literature search
Force on Classification of Status Epilepticus. Epilepsia of midazolam and anesthetic therapies. Pediatr Crit Care
2015;56:1515-23. Med 2014;15:632-9.
Uberall MA, Trollmann R, Wunsiedler U, et al. Intravenous val- Wu YW, Shek DW, Garcia PA, et al. Incidence and mortality
proate in pediatric epilepsy patients with refractory status of generalized convulsive status epilepticus in California.
epilepticus. Neurology 2000;54:2188-9. Neurology 2002;58:1070-6.
Wada T, Takase M, Fukushima Y. Diamox acetazolamide in Yu KT, Mills S, Thompson N, et al. Safety and efficacy of
treatment of the epileptics with frequent seizures or sta- intravenous valproate in pediatric status epilepticus and
tus epilepticus. Folia Psychiatr Neurol Jpn 1961;15:98-109. acute repetitive seizures. Epilepsia 2003;44:724-6.

PedSAP 2016 Book 2 Pediatric Critical Care 120 Interactive Case: Status Epilepticus
Self-Assessment Questions
Questions 3335 pertain to the following case. A. Drug-drug interaction between levetiracetam and
L.N. is a 5-year-old girl (weight 18 kg) who is being transferred phenytoin
to the pediatric ICU from the ED for seizure activity at home B. Phenytoin binding to enteral tubing
(duration about 5 minutes per caregiver). While being trans- C. Not shaking the suspension
ferred to the unit, LN begins seizing again. D. Inappropriate home dose of phenytoin
33. Which one of the following best describes L.N.s seizure Questions 3638 pertain to the following case.
activity and need for pharmacologic therapy?
A.B. is a 10-day-old girl born at 29 weeks gestation (birth weight
A. She is in status epilepticus, defined as a seizure 1100 g). She has new-onset seizure activity. Maternal history is
duration longer than 5 minutes, and therapy should positive for preeclampsia and premature rupture of membranes.
be initiated. Since birth, A.B. has been weaned from continuous positive
B. She is in status epilepticus, defined as a seizure airway pressure (CPAP) and is currently receiving 1/2 L nasal
duration longer than 30 minutes, and therapy should cannula since seizure activity began. She has had increased
be initiated. apneic events requiring stimulation and decreased activity.
C. She is not in status epilepticus because she has Her current drugs include caffeine 5 mg/kg/dose intravenously
had multiple seizures with a duration of 5 minutes every 24 hours and parenteral nutrition with lipids (dextrose 10%,
with return to baseline function, and should not protein 4 g/kg, and lipids 2 g/kg). Results of physical examina-
receive therapy. tion and laboratory tests include the following: current weight
D. She is not in status epilepticus, defined as multiple 1160 g, sodium 131 mEq/L, urine output 3.4 mL/kg/hour, potas-
seizures with a duration of 30 minutes and return sium 5.5 mEq/L, chloride 99 mmol/L, CO2 25 mmol/L, WBC 2.4
to baseline function, and should not receive 103 cells/mm3, BUN 10 mg/dL, Hgb 8 g/dL, SCr 0.2 mg/dL, Hct
therapy. 40 g/dL, glucose 67 mg/dL, platelets 340,000/mm3, and calcium
34. L.N. receives two doses of lorazepam 0.1 mg/kg/dose, 9.7 mg/dL. Differential shows increased bands.
and the team is preparing to give a second-line agent at 36. Based on these data, which one of the following is the
the 15-minute period. On further investigation of L.N.s most likely etiology of A.B.s seizure activity?
home drugs and laboratory results, you confirm that she
A. Apnea of prematurity
is taking phenytoin suspension 8 mg/kg/day divided
B. Hypoglycemia
every 12 hours and levetiracetam solution 30 mg/kg/
C. Hyponatremia
day divided every 12 hours. Laboratory tests, including
D. Late-onset sepsis
levetiracetam and phenytoin concentrations, are sent for
evaluation. Which one of the following is best to recom- 37. A.B. has antibiotics initiated (ampicillin 100 mg/kg/dose
mend for L.N.? intravenously every 12 hours and gentamicin 4.5 mg/
kg/dose intravenously every 36 hours), but her seizure
A. Phenytoin 20 mg/kg intravenously 1 dose over 30
activity progresses from lasting 30 seconds to more pro-
minutes.
longed durations. The nurse alerts the team that A.B. has
B. Fosphenytoin 20 mg/PE/kg intravenously 1 dose
been seizing now for 3 minutes. After 5 minutes, which
over 10 minutes.
one of the following is the most appropriate pharmaco-
C. Levetiracetam 20 mg/kg intravenously 1 dose over
logic therapy to recommend for A.B.s seizures?
15 minutes.
D. Phenobarbital 20 mg/kg intravenously 1 dose over A. Levetiracetam 40 mg/kg intravenously 1 dose
20 minutes. B. Lorazepam 0.1 mg/kg intravenously 1 dose
C. Midazolam 0.05 mg/kg intravenously 1 dose
35. L.N.s laboratory values show a levetiracetam concentra-
D. Phenobarbital 20 mg/kg intravenously 1 dose
tion of 15 mcg/mL and a total phenytoin concentration of
21 mcg/mL. She was loaded with levetiracetam, and her 38. Which one of the following is best to recommend to the
seizure ceased. After observing the patient and ensur- care team to prevent long-term complications from phe-
ing return to baseline behavior and function, the patient nobarbital therapy in A.B.?
is to be discharged. The team is discussing potential A. Phenobarbital has not been associated with long-
causes of this episode. Which one of the following best term adverse effects in neonates.
describes the rationale for a supratherapeutic phenytoin B. Monitor for chronic lung disease from increased
concentration, assuming the patient had a normal albu- respiratory depression and advanced ventilation
min concentration? requirements.

PedSAP 2016 Book 2 Pediatric Critical Care 121 Interactive Case: Status Epilepticus
C. Monitor for vitamin D deficiency and decreased activity is present. His blood pressure has remained sta-
cognitive development. ble but lower since initiation of continuous infusion of
D. Monitor for development of necrotizing enterocolitis. midazolam 0.1 mg/kg/hour and fentanyl 1 mcg/kg/hour.
Fentanyl has been decreased to 0.5 mcg/kg/hour, and
Questions 3944 pertain to the following case. blood pressure monitoring continues. Which one of the
V.V. is a 3-year-old boy (weight 14 kg) who presents to the following is best to initiate for V.V.?
pediatric ICU with new-onset seizures of unknown etiology.
A. Lacosamide 1 mg/kg/day divided twice daily
Toxic exposure is not suspected, and the following laboratory
B. Pentobarbital 5-mg/kg bolus followed by 1 mg/kg/
results are available: pH on blood gas 7.3, sodium 139 mEq/L,
hour continuous infusion
lactate 5 mmol/L, potassium 3.5 mEq/L, AST 44 IU/L, chlo-
C. Propofol 3-mg/kg bolus followed by 100 mcg/kg/
ride 104 mmol/L, ALT 52 IU/L, CO2 15 mmol/L, BUN 30 mg/
minute continuous infusion
dL, SCr 0.7 mg/dL, glucose 134 mg/dL, magnesium 2 mEq/L,
D. Acetazolamide 10 mg/kg/day divided twice daily by
and calcium 10 mg/dL. V.V.s acetaminophen and salicylate
nasogastric tube
concentrations are undetectable, and the urine drug screen
is negative. He has no sick contacts. Although V.V. has had 42. Seven days after therapy initiation, burst suppression is
no previous seizure activity, he has a positive family history apparent on the EEG, but V.V. has had persistent hypo-
of seizures (paternal grandfather). The resident examines V.V. tension requiring 10 mcg/kg/minute of dopamine. His
while the boy is sleeping and notices seizure activity. After updated laboratory values, which have been consistent
about 3 minutes, V.V.s seizure activity has not subsided. He over the past few days, include the following: sodium
does not currently have intravenous access. 143 mEq/L, potassium 4.5 mEq/L, chloride 109 mmol/L,
CO2 15 mmol/L, BUN 32 mg/dL, SCr 0.5 mg/dL, glucose
39. Which one of the following is best to initiate for V.V.s
110 mg/dL, and calcium 10 mg/dL. Given V.V.s current
seizures?
clinical status, which one of the following best justifies
A. Intramuscular phenytoin 20 mg/kg 1 dose discontinuing V.V.s pentobarbital?
B. Intranasal midazolam 0.2 mg/kg 1 dose
A. The potential for hypotension
C. Oral lorazepam 0.1 mg/kg 1 dose
B. The potential for propylene glycol toxicity
D. Rectal diazepam 0.3 mg/kg 1 dose
C. The lack of correlation between burst suppression
40. Intravenous access has been obtained for V.V., and a and cessation of seizures in refractory SE
second dose of a benzodiazepine (lorazepam 0.1 mg/kg D. The lack of data on prolonged continuous infusions
intravenously 1 dose) is administered. The boy contin-
43. V.V. has now been receiving pentobarbital for 8 days, and
ues to seize for 10 minutes without cessation. The team
the neurologist wishes to discontinue this therapy. V.V. can
decides to administer fosphenytoin. Which one of the
tolerate a wean to 2 mg/kg/hour. Which one of the following
following best justifies the use of fosphenytoin versus
is the best strategy for discontinuing V.V.s pentobarbital?
phenytoin in V.V.?
A. Decrease pentobarbital by 10% every day until off.
A. Fosphenytoin can be administered faster and will
B. Convert to pentobarbital intermittent dosing by
have a quicker onset of action.
dividing the total daily dose into an every-6-hour
B. Fosphenytoin has a decreased extravasation risk
interval.
because of a lower pH.
C. Give a loading dose of phenobarbital, and initiate
C. Fosphenytoin contains more propylene glycol for
maintenance dosing at one-third the loading dose.
solubility and can be administered over a shorter
D. Do not wean the patient from therapy until additional
period.
agents are added for seizure control.
D. Fosphenytoin does not need therapeutic drug
monitoring. 44. Despite the above actions, V.V. continues to have sei-
zures. He is becoming hypotensive despite a fluid bolus
41. V.V. is still seizing after 15 minutes, despite fosphenyt-
of NS 20 mL/kg and decreasing his pain and sedation
oin administration. He is given levetiracetam 20 mg/
medications. The team initiates epinephrine continuous
kg intravenously 1 dose, and at 30 minutes, the deci-
infusion at 0.1 mcg/kg/min (titrate to goal systolic and
sion is made to intubate. He is intubated with fentanyl
diastolic blood pressure). Which of the following is best
1 mcg/kg intravenously 1 dose, midazolam 0.1 mg/kg
to recommend for V.V.s continued SE?
intravenously 1 dose, and vecuronium 0.1 mg/kg intra-
venously 1 dose. On intubation, he is continued on A. Acetazolamide 5 mg/kg/dose IV every 8 hours
fentanyl 1 mcg/kg/hour and midazolam 0.1 mg/kg/hour B. Ketamine 1 mg/kg/hr continuous infusion
continuous infusions for pain and sedation. A continu- C. Lacosamide 3 mg/kg/dose IV every 12 hours
ous EEG has been initiated on V.V., and continued seizure D. Propofol 10 mg/kg/hr continuous infusion

PedSAP 2016 Book 2 Pediatric Critical Care 122 Interactive Case: Status Epilepticus
Learner Evaluation: Interactive Case: Status Epilepticus.

As you take the posttest for this chapter, also evaluate the Use the 5-point scale to indicate whether this chapter pre-
materials quality and usefulness, as well as the achievement pared you to accomplish the following learning objectives:
of learning objectives. Rate each item using this 5-point scale: 49. Distinguish the cause of status epilepticus (SE) in neo-
nates, infants, children, and adolescents.
Strongly agree 50. Design an appropriate treatment plan for the pediatric
Agree patient with SE or refractory SE.
Neutral 51. Design a treatment strategy for the patient with complica-
Disagree tions of SE, including hypoxemia and acidosis.
Strongly disagree 52. 
Evaluate the patient with SE for the development of
long-term sequelae of prolonged seizure activity and/or
38. The content of the chapter met my educational needs. treatment of SE.
39. The content of the chapter satisfied my expectations. 53. P
 lease provide any specific comments related to any
40. The author presented the chapter content effectively. perceptions of bias, promotion, or advertisement of com-
mercial products.
41. The content of the chapter was relevant to my practice
and presented at the appropriate depth and scope. 54. Please expand on any of your above responses, and/or
provide any additional comments regarding this chapter:
42. The content of the chapter was objective and balanced.
43. The content of the chapter is free of bias, promotion, or Questions 5557 apply to the entire learning module.
advertisement of commercial products.
55. How long did it take you to read the instructional materi-
44. T
 he content of the chapter was useful to me. als in this module?
45. The teaching and learning methods used in the chapter 56. How long did it take you to read and answer the assess-
were effective. ment questions in this module?
46. The active learning methods used in the chapter were 57. Please provide any additional comments you may have
effective. regarding this module:
47. The learning assessment activities used in the chapter
were effective.
48. The chapter was effective overall.

PedSAP 2016 Book 2 Pediatric Critical Care 123 Interactive Case: Status Epilepticus
PedSAP 2016-2018 Releases

Title Release Date BCPPS Test Deadline ACPE Test Deadline

Immunology May 16, 2016 September 15, 2016 May 14, 2019

Pediatric Critical Care September 15, 2016 January 17, 2017 September 14, 2019

Research Ethics/
January 17, 2017 May 15, 2017 January 14, 2020
Study Design

Pediatric Emergencies May 15, 2017 September 15, 2017 May 14, 2020

Sedation and
September 15, 2017 January 16, 2018 September 14, 2020
Analgesia

Pediatric Oncology January 16, 2018 May 15, 2018 January 14, 2021

Fluids/Electrolytes/ May 15, 2018 September 17, 2018 May 14, 2021
Nutrition

Neonatal and Pediatric September 17, 2018 January 15, 2019 September 14, 2021
Sepsis

PedSAP Pricing ACCP Members Nonmembers

Single Book $65 $90

Any Three or More Books $50/book $70/book


Full Series
$260 $355
(eight books)

*Appropriate shipping charges will apply.

This book is one release from the American College of Clinical Pharmacys 2016-2018 Pediatric Self-Assessment Program (PedSAP).

Releases are available singly or as an eight-book series over 3 years. Topics for the series are designed to cover the content outline

for the Board Certified Pediatric Pharmacy Specialist examination administered by the Board of Pharmacy Specialties.

For specific faculty, chapter titles, and available continuing pharmacy education contact hours, go to www.accp.com/bookstore.

To order, contact the


American College of Clinical Pharmacy,
13000 W. 87th St. Parkway, Lenexa, KS 66215
Voice: (913) 492-3311 / Fax: (913) 492-0088
Online Bookstore: www.accp.com/bookstore
Pharmacotherapy
Pediatric
Self-Assessment
Self-Assessment
Program
Program

Вам также может понравиться