Handbook of
Pediatric Hematology
and Oncology
Children’s Hospital & Research Center Oakland
SECOND EDITION
Caroline A. Hastings
Joseph C. Torkildson
Anurag K. Agrawal
Handbook of Pediatric Hematology
and Oncology
Handbook
of Pediatric
Hematology
and Oncology
Children’s Hospital & Research
Center Oakland
Caroline A. Hastings, MD
Pediatric Hematology and Oncology
Children’s Hospital & Research Center Oakland
Oakland, CA, USA
Joseph C. Torkildson, MD, MBA
Pediatric Hematology and Oncology
Children’s Hospital & Research Center Oakland
Oakland, CA, USA
Anurag K. Agrawal, MD
Pediatric Hematology and Oncology
Children’s Hospital & Research Center Oakland
Oakland, CA, USA
Second edition
This edition first published 2012
2012 by John Wiley & Sons, Limited.
Wiley-Blackwell is an imprint of John Wiley & Sons, formed by the merger of Wileys global Scientific, Technical,
and Medical business with Blackwell Publishing.
Registered office: John Wiley & Sons, Ltd, The Atrium, Southern Gate, Chichester, West Sussex, PO19 8SQ, UK
Editorial offices:
9600 Garsington Road, Oxford, OX4 2DQ, UK
The Atrium, Southern Gate, Chichester, West Sussex, PO19 8SQ, UK
111 River Street, Hoboken, NJ 07030-5774, USA
For details of our global editorial offices, customer services, and for information about how to apply for permission
to reuse the copyright material in this book please see our Website at www.wiley.com/wiley-blackwell
The right of the author to be identified as the author of this work has been asserted in accordance with the UK
Copyright, Designs and Patents Act 1988.
All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted, in
any form or by any means, electronic, mechanical, photocopying, recording or otherwise, except as permitted by
the UK Copyright, Designs and Patents Act 1988, without the prior permission of the publisher.
Designations used by companies to distinguish their products are often claimed as trademarks. All brand names and
product names used in this book are trade names, service marks, trademarks or registered trademarks of their
respective owners. The publisher is not associated with any product or vendor mentioned in this book. This
publication is designed to provide accurate and authoritative information in regard to the subject matter covered. It
is sold on the understanding that the publisher is not engaged in rendering professional services. If professional
advice or other expert assistance is required, the services of a competent professional should be sought.
The contents of this work are intended to further general scientific research, understanding, and discussion only
and are not intended and should not be relied upon as recommending or promoting a specific method,
diagnosis, or treatment by physicians for any particular patient. The publisher and the author make no
representations or warranties with respect to the accuracy or completeness of the contents of this work and
specifically disclaim all warranties, including without limitation any implied warranties of fitness for a particular
purpose. In view of ongoing research, equipment modifications, changes in governmental regulations, and the
constant flow of information relating to the use of medicines, equipment, and devices, the reader is urged to
review and evaluate the information provided in the package insert or instructions for each medicine,
equipment, or device for, among other things, any changes in the instructions or indication of usage and for
added warnings and precautions. Readers should consult with a specialist where appropriate. The fact that an
organization or Website is referred to in this work as a citation and/or a potential source of further information
does not mean that the author or the publisher endorses the information the organization or Website may
provide or recommendations it may make. Further, readers should be aware that Internet Websites listed in this
work may have changed or disappeared between when this work was written and when it is read. No warranty
may be created or extended by any promotional statements for this work. Neither the publisher nor the author
shall be liable for any damages arising herefrom.
Library of Congress Cataloging-in-Publication Data
Hastings, Caroline, 1960-
Handbook of pediatric hematology and oncology : Children’s Hospital &
Research Center Oakland / Caroline A. Hastings, Joseph Torkildson, Anurag
Kishor Agrawal. - 2nd ed.
p. ; cm.
Rev. ed. of: The Children’s Hospital Oakland hematology/oncology handbook /
Caroline Hastings. 1st ed. c2002.
Includes bibliographical references and index.
ISBN 978-0-470-67088-0 (pbk. : alk. paper)
I.
Torkildson, Joseph. II. Agrawal, Anurag Kishor. III. Hastings, Caroline,
1960- Children’s Hospital Oakland hematology/oncology handbook. IV. Children’s
Hospital Medical Center (Oakland, Calif.) V. Title.
[DNLM: 1. Child. 2. Hematologic Diseases-Handbooks.
3. Neoplasms-Handbooks. WS 39]
618.9706994-dc23
2011048154
A catalog record for this book is available from the British Library.
Set in 10/12.5 pt Minon by Thomson Digital, Noida, India
1
2011
Contents
Preface, vii
Acknowledgments, ix
1
Approach to the Anemic Child, 1
2
Hemolytic Anemia, 10
3
Sickle Cell Disease, 18
4
Thalassemia, 36
5
Transfusion Medicine, 44
6
Chelation Therapy, 57
7
Approach to the Bleeding Child, 62
8
Von Willebrand Disease, 71
9
Hemophilia, 79
10
The Child with Thrombosis, 85
11
The Neutropenic Child, 92
12
Thrombocytopenia, 103
13
Evaluation of the Child with a Suspected Malignancy, 122
14
Oncologic Emergencies, 133
15
Acute Leukemias, 144
16
Central Nervous System Tumors, 157
17
Hodgkin and Non-Hodgkin Lymphoma, 166
18
Wilms Tumor, 174
19
Neuroblastoma, 178
20
Sarcomas of the Soft Tissues and Bone, 183
vi Contents
21 Germ Cell Tumors, 193
22 Rare Tumors of Childhood, 200
23 Histiocytic Disorders, 207
24 Hematopoietic Stem Cell Transplantation, 212
25 Supportive Care of the Child with Cancer, 226
26 Central Venous Catheters, 235
27 Management of Fever in the Child with Cancer, 244
28 Acute Pain Management in the Inpatient Setting, 256
29 Palliative Care, 264
30 Chemotherapy Basics, 281
31 Guide to Procedures, 291
32 Treatment of Chemotherapy Extravasations, 300
Formulary, 305
Index, 365
Preface
The pace of change in the field of pediatric
never mentioned in the large studies or
hematology and oncology is staggering.
case reports.
Molecular biology, genomics, and bio-
This handbook represents the work of my
chemistry have accelerated the accumula-
colleagues at Children’s Hospital & Research
tion of knowledge and understanding of
Center Oakland toward this endeavor. The
disease states. Yet the application of this
guidelines offered here have been used to
new knowledge to the individual child
train medical students, pediatric residents,
before you, the work of the physician, is
and pediatric hematology/oncology fellows
often overwhelming, even for the most
for over 20 years. This handbook will give you
experienced practitioner. The course and
clinical approaches for common problems in
prognosis for the child is often determined
pediatric hematology and oncology, the
by the rapidity of disease onset, diagnosis,
knowledge to organize and to evaluate the
and initial treatment. What is needed is
care of your patients, and a framework to
a practical, tested approach to these
incorporate ever-expanding psychosocial
problems that ensures timely evaluation,
needs, clinical studies, medical treatments,
competent early care, and avoidance of
and science. All of these are essential com-
pitfalls that might prejudice future treat-
ponents that make up the care of the child
ment options. This practical approach is
with cancer or a blood disease.
clearly brought by spending time with
the patients and their families, and
Caroline Hastings, M.D.
observing the myriad variations that are
March 2012
Acknowledgments
We could not be here without the long-
improved outcomes. Our experiences have
standing love and support of our families.
taught us the magnitude of remembering
On a day to day basis, the patients and their
our roles: “to cure sometimes, to relieve
families continue to show us how to live
often, to comfort always.”
(Anonymous,
gracefully in even the hardest of times and
15thcentury)
inspire us to continue to endeavor for
Approach to the
1
Anemic Child
Anemia is the condition in which the con-
age (Table 1.1). The blood smear and red
centration of hemoglobin or the red cell
cell indices are very helpful in the diagnosis
mass is reduced below normal. Anemia
and classification of anemia. It allows for
results in a physiological decrease in the
classification by the cell size (MCV, mean
oxygen-carrying capacity of the blood and
corpuscular volume), gives the distribution
reduced oxygen supply to the tissues. Causes
of cell size
(RDW, red cell distribution
of anemia are increased loss or destruction
width), and may give important diagnostic
of red blood cells (RBCs) or a significant
clues if specific morphological abnormali-
decreased rate of production. When evalu-
ties are present (e.g., sickle cells, target cells,
ating a child with anemia, it is important to
and spherocytes). The MCV, RDW, and
determine if the problem is isolated to one
reticulocyte count are helpful in the differ-
cell line (e.g., RBCs) or multiple cell lines
ential diagnosis of anemia. A high RDW, or
(i.e., RBCs, white blood cells [WBCs], or
anisocytosis, is seen in stress erythropoiesis
platelets). When two or three cell lines are
and is often suggestive of iron deficiency or
affected, it may indicate bone marrow
hemolysis. A normal or low reticulocyte
involvement (leukemia, metastatic disease,
count is an inappropriate response to ane-
and aplastic anemia), sequestration
(i.e.,
mia and suggests impaired red cell produc-
hypersplenism), immune deficiency, or an
tion. An elevated reticulocyte count suggests
immune-mediated process (e.g., hemolytic
blood loss, hemolysis, or sequestration.
anemia and immune thrombocytopenic
The investigation of anemia requires
purpura).
the following steps:
1. The medical history of the anemic child
(Table 1.2), as certain historical points may
Evaluation of anemia
provide clues as to the etiology of the
anemia.
The evaluation of anemia includes a com-
2. Detailed physical examination (Table 1.3),
plete medical history, family history, physical
with particular attention to acute and chronic
examination, and laboratory assessment. See
effects of anemia.
Figure 1.1.
3. Evaluation of the complete blood count
The diagnosis of anemia is made after
(CBC), RBC indices, and peripheral blood
reference to established normal controls for
smear, with classification by MCV,
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
2
Chapter 1
History
Physical examination
Complete blood count
Reticulocyte count
Peripheral blood smear examination
Microcytic*
Normocytic*
Macrocytic*
Iron deficiency
Early iron deficiency
Normal newborn
Dietary
Acute blood loss
Reticulocytosis
Chronic blood loss
Hemolysis
Post splenectomy
Thalassemia, α or β
Red cell enzyme deficiency
Liver disease
Hemoglobin E
Red cell membrane defects
Aplastic anemia
Lead toxicity
Red cell aplasia
Bone marrow failure
Chronic disease/Infection
Aplastic anemia
syndromes
Severe malnutrition
DBA, TEC
Hypothyroidism
Sideroblastic anemia
Malignancy
Down syndrome
Infection
Preleukemia
Syndromes with elevated Hgb F
Renal failure
Megaloblastic anemia
Hypersplenism
Folic acid deficiency
Drugs
Malabsorption, drugs
Vitamin B12 deficiency
Evaluations
Dietary
To Consider
Pernicious anemia
Iron Studies
Red cell enzyme panel
Liver function tests
RDW, FEP, ferritin,
G6PD, Pyruvate kinase
Thyroid function tests
transferrin saturation, TIBC
Osmotic fragility/Ektacytometry
Hemoglobin electrophoresis
Hemoglobin electrophoresis
Coombs test (DAT)
Folic acid level
Lead level
Hemoglobin electrophoresis
Red cell
Family studies
Bone marrow aspirate
Serum
Check newborn screen
Vitamin B12 level
Oral iron challenge
Bone marrow aspirate
Figure 1.1 Diagnostic approach to the child with anemia. (Abbreviations: DBA, Diamond-Blackfan
anemia; TEC, transient erythroblastopenia of childhood; RDW, red cell distribution width; FEP, free
erythrocyte protoporphyrin; TIBC, total iron binding capacity; G6PD, glucose-6-phosphate dehy-
drogenase deficiency; DAT, direct antiglobulin test).
Refer to Table 1.1 for age-based normal values.
reticulocyte count, and RBC morphology.
Interventions
Consideration should also be given to the
WBC and platelet counts as well as their
Oral iron challenge
respective morphology.
An oral iron challenge may be indicated
4. Determination of an etiology of the
in the patient with significant iron deple-
anemia by additional studies as needed
tion, as documented by moderate-to-severe
(see Figures 1.1, 1.2, and 1.3).
anemia and deficiencies in circulating
Approach to the Anemic Child
3
Table 1.1 Red blood cell values at various ages.*
Age
Hemoglobin (g/dL)
MCV (fL)
Mean
2 SD
Mean
2 SD
Birth (cord blood)
16.5
13.5
108
98
1-3 d (capillary)
18.5
14.5
108
95
1 wk
17.5
13.5
107
88
2 wk
16.5
12.5
105
86
1 mo
14.0
10.0
104
85
2 mo
11.5
9.0
96
77
3-6 m
11.5
9.5
91
74
0.5-2 y
12.0
11.0
78
70
2-6 y
12.5
11.5
81
75
6-12 y
13.5
11.5
86
77
12-18 y female
14.0
12.0
90
78
12-18 y male
14.5
13.0
88
78
18-49 y female
14.0
12.0
90
80
18-49 y male
15.5
13.5
90
80
Compiled from the following sources: Dutcher TF. Lab Med 2:32-35, 1971; Koerper
MA, et al. J Pediatr 89:580-583, 1976; Marner T. Acta Paediatr Scand 58:363-368, 1969; Matoth Y,
et al. Acta Paediatr Scand 60:317-323, 1971; Moe PJ. Acta Paediatr Scand 54:69-80, 1965; Okuno
T. J Clin Pathol 2:599-602, 1972; Oski F, Naiman J. Hematological Problems in the Newborn, 2nd
ed., Philadelphia: WB Saunders, 1972, p. 11; Penttila I, et al. Suomen Laakarilehti 26:2173, 1973;
and Viteri FE, et al. Br J Haematol 23:189-204, 1972. Cited in: Rudolph AM (ed). Rudolph’s
Pediatrics, 16th ed., Norwalk, CT: Appleton & Lange, 1977.
Abbreviation: MCV, mean corpuscular volume.
and storage iron forms
(such as total
elemental iron) orally; third, draw another
iron-binding capacity [TIBC], serum iron,
serum iron level 30 to 60 minutes later.
transferrin saturation, and ferritin). Iron
The serum level is expected to increase by
absorption is impaired in certain chronic
at least
100 mcg/dL if absorption is ade-
disorders
(autoimmune diseases such as
quate. The oral iron challenge is a quick
lupus, peptic ulcer disease, ulcerative
and easy method to assess appropriateness
colitis, and Crohn’s disease), by certain
of oral iron to treat iron deficiency—a safer,
medications
(antacids and histamine-2
cheaper yet equally efficacious method of
blockers), and by environmental factors
treatment as parenteral iron.
such as lead toxicity.
Indications for an oral iron challenge
Parenteral iron therapy
include any condition in which a poor
Due to the potential risks of older parenteral
response to oral iron is being questioned,
iron preparations (specifically high molec-
such as in: noncompliance, severe anemia
ular weight iron dextran), a reluctance
secondary to dietary insufficiency (exces-
remains to use the newer and much safer
sive milk intake), and ongoing blood loss.
formulations. The majority of safety data
Administration of an oral iron challenge
exists with low molecular weight (LMW)
is quite simple: first, draw a serum iron level;
iron dextran although many practitioners
second, administer a dose of iron (3 mg/kg
have moved to newer (and perceived safer)
4
Chapter 1
Table 1.2 The medical history of the anemic child.
History of
Consider
Prematurity
Anemia of prematurity (EPO responsive)
Perinatal risk factors
Maternal illness (autoimmune)
Hemolytic anemia
Drug ingestion
Impaired production
Infections (TORCH [e.g., rubella, CMV],
hepatitis)
Perinatal problems
Acute blood loss
Fetal-maternal hemorrhage
Iron deficiency due to above or maternal iron
deficiency
Ethnicity
African-American
Hgb S, C; a- and b-thalassemia; G6PD deficiency
Mediterranean
a- and b-thalassemia; G6PD deficiency
Southeast Asian
a- and b-thalassemia; Hgb E
Family history
Gallstones, cholecystectomy
Inherited hemolytic anemia, spherocytosis,
elliptocytosis
Splenectomy, jaundice at birth or
Inherited enzymopathy, G6PD, pyruvate kinase
with illness
deficiencies
Isoimmunization (Rh or ABO)
Hemolytic disease of newborn (predisposed to
iron deficiency)
Sex
Male
X-linked enzymopathies (G6PD deficiency)
Early jaundice (<24 h of age)
Isoimmune, infectious
Persistent jaundice
Suggests hemolytic anemia
Diet (Usually > 6 mo)
Pica (ice, dirt)
Lead toxicity, iron deficiency
Excessive milk intake
Iron deficiency
Macrobiotic diets
Vitamin B12 deficiency
Goat’s milk
Folic acid deficiency
Drugs
Sulfa drugs, anticonvulsants
Hemolytic anemia (G6PD deficiency)
Chloramphenicol
Hypoplastic anemia
Low socioeconomic status
Pica
Lead toxicity, iron deficiency
Malnutrition
Malabsorption
Anemia of chronic disease
Environmental
Iron, vitamin B12, or folate deficiency, vitamin E
or K deficiency
Liver disease
Shortened red cell survival
Heinz bodies
Renal disease
Shortened red cell survival
Decreased red cell production (#EPO)
Infectious diseases
Mild viral infection (acute gastroenteritis,
Transient mild decreased Hgb
otitis media, pharyngitis)
Sepsis (bacterial, viral, mycoplasma)
Hemolytic anemia
Parvovirus
Anemia with reticulocytopenia (TEC)
Abbreviations: EPO, erythropoietin; TORCH, toxoplasmosis, other, rubella, cytomegalovirus,
herpes simplex virus; G6PD, glucose-6-phosphate dehydrogenase deficiency; TEC, transient
erythroblastopenia of childhood.
Approach to the Anemic Child
5
Table 1.3
Physical examination of the anemic child.
System
Clinical sign or symptom
Potential underlying disorder
Skin
Pallor
Severe anemia
Jaundice
Hemolytic anemia, acute and chronic
hepatitis, aplastic anemia
Petechiae, purpura
Autoimmune hemolytic anemia with
thrombocytopenia, hemolytic uremic
syndrome, bone marrow aplasia or
infiltration
Cavernous hemangioma
Microangiopathic hemolytic anemia
HEENT
Frontal bossing, prominent
Extramedullary hematopoiesis (thalassemia
malar and maxillary bones
major, congenital hemolytic anemia)
Icteric sclerae
Congenital hemolytic anemia and hyper-
hemolytic crises associated with infection
(red cell enzyme deficiencies, red cell
membrane defects, thalassemias,
hemoglobinopathies)
Angular stomatitis
Iron deficiency
Glossitis
Vitamin B12 or iron deficiency
Chest
Rales, gallop rhythm,
Congestive heart failure, acute or severe
tachycardia
anemia
Spleen
Splenomegaly
Congenital hemolytic anemia, infection,
hematological malignancies, portal
hypertension, resultant hypersplenism
Extremities
Radial limb dysplasia
Fanconi anemia
Spoon nails
Iron deficiency
Triphalangeal thumbs
Red cell aplasia
formulations including ferric gluconate and
Dosage ðmLÞ ¼ 0:0442
LBW ðkgÞ
iron sucrose. Three additional compounds
ðHgbn HgboÞ
have been approved recently, 2 in Europe
þ ½0:26
LBW ðkgÞ ;
(ferric carboxymaltose and iron isomalto-
where
side) and 1 in the United States (ferumox-
LBW ¼ lean body weight
ytol). These newer agents have the potential
benefit of total dose replacement in a very
males: 50 kg þ 2:3 kg for every
short and single infusion as compared to
inch over 5 ft in height
ferric gluconate and iron sucrose which
females: 45:5 kg þ 2:3 kg for
require multiple doses. LMW iron dextran
every inch over 5 ft in height
is approved as a total dose infusion for
Hgbn ¼ desired hemoglobin ðg=dLÞ ¼ 12
adults in Europe but not the United States.
Due to the smaller dose generally required
if
< 15 kg or 14:8 if > 15 kg
in pediatric patients, total iron replacement
Hgbo ¼ measured hemoglobin ðg=dLÞ
is feasible in
1 to 2 doses of LMW iron
dextran. Calculation of the necessary dose is
The maximum adult dose is 2 mL and
as follows:
each milliliter of iron dextran contains 50 mg
6
Chapter 1
Assess degree of anemia
Mild
Moderate
Severe
(Hemoglobin > 10 g/dL)
(Hemoglobin 7-10 g/dL)
(Hemoglobin < 7 g/dL)
History and physical exam
compatible with iron deficiency
History, physical,
iron studies, consider
or
hemoglobin electrophoresis
and family studies
Trial of oral iron
Iron Studies
4-6 mg/kg/day
FEP
Consider
Dietary counseling
Iron, TIBC, % iron
Reticulocyte count
Deficient
saturation
Review smear
Hospitalization
at 1 week
Reticulocyte count
Transfusion
Stool guaiac
IV or oral iron
(if indicated)
Not Deficient
Continue oral iron
Hemoglobinopathy/Thalassemia
3-6 months
Consider
Hgb electrophoresis
Family studies
Blood Loss
Lead toxicity
Consider
Urinalysis
Stool guaiac
Meckel’s scan
Severe iron deficiency
Hemolysis
Red cell aplasia
Coombs test (DAT)
Malignancies
Peripheral smear
Infections
Hemoglobinopathy/Thalassemia
Hemolytic anemia with
Hemoglobin electrophoresis
illness/infection
Family studies
Thalassemia
Lead poisoning
Hgb H disease
Iron malabsorption
β-thalassemia major
Iron studies
Hemoglobinopathy
Oral iron challenge
Sickle cell disease
Consider parenteral iron
Bowel disease (Crohn’s, IBD)
Inflammatory disorder (lupus)
Figure 1.2 Evaluation of the child with microcytic anemia. (Abbreviations: FEP, free erythrocyte
protoporphyrin; TIBC, total iron binding capacity; DAT, direct antiglobulin test; IBD, inflammatory
bowel disease).
of elemental iron. Add 10 mg elemental iron/
utilized. A test dose (10 to 25mg) should be
kg to replenish iron stores (chronic anemia
given prior to the first dose with observation
states). Replacement may be given in a single
of the patient for 30 to 60 minutes prior to
dose, depending on the dose required. See
administering the remainder of the dose. A
the formulary for further information.
common side effect is mild to moderate
Severe allergic reactions can occur
arthralgias the day after drug administration,
with iron dextran and the low molecular
especially in patients with autoimmune
weight product should be preferentially
disease.
Acetaminophen
frequently
Approach to the Anemic Child
7
Low hemoglobin concentration
Low
Elevated or normal
reticulocyte
reticulocyte count
count
Infection
TORCH
Congenital hypoplastic anemia
Coombs test (DAT)
Transcobalamin II deficiency
Immune hemolytic anemia
Positive
ABO incompatibility
Rh incompatibility
Minor blood group incompatibility
Negative
Microcytic
α-thalassemia syndrome
Normocytic
Chronic intrauterine blood loss
Macrocytic
Iron deficiency
Blood loss
Peripheral smear
Iatrogenic
Traumatic delivery
Internal hemorrhage
Normal
Twin-twin transfusion
Fetal-maternal transfusion
Abnormal
Infection
TORCH
Membrane defect
Hereditary spherocytosis
Hereditary elliptocytosis
Red cell enzyme deficiency
G6PD
Pyruvate kinase
Figure 1.3 Approach to the full-term newborn with anemia. (Abbreviations: DAT, direct antiglobulin
test; G6PD, glucose-6-phosphatase deficiency; TORCH, toxoplasmosis, other, rubella, cytomegalo-
virus, herpes simplex virus).
alleviates the arthralgias. Iron dextran is
doses are more convenient and feasible than
contraindicated in patients with rheuma-
the outpatient setting. With continued
toid arthritis.
usage and safety data, ferumoxytol will
Iron sucrose or ferric gluconate can be
likely replace the currently used products
considered in inpatients in which multiple due to the much larger maximum dose that
8
Chapter 1
can be given, lack of need for a test dose, and
transfusions to children with cardiovascular
excellent side effect profile.
compromise (i.e., gallop rhythm, pulmo-
nary edema, excessive tachycardia, and poor
Erythropoietin
perfusion) while being monitored in an ICU
Recombinant human erythropoietin
setting. Transfusions are given in multiple
(EPO) stimulates proliferation and differ-
small volumes, sometimes separated by sev-
entiation of erythroid precursors, with an
eral hours, with careful monitoring of the
increase in heme synthesis. This increased
vitals and fluid balance. For those children
proliferation creates an increased demand
who have gradual onset of severe anemia,
in iron availability and can result in a
without cardiovascular compromise, con-
functional iron deficiency if not given with
tinuous transfusion of 2 mL/kg/h has been
iron therapy.
shown to be safe and result in an increase in
Indications for EPO include end-stage
the hematocrit of 1% for each 1 mL/kg of
renal disease, anemia of prematurity,
transfused packed RBCs (based on RBC
anemia of chronic disease, anemia associ-
storage method). The hemoglobin should
ated with treatment for AIDS, and autolo-
be increased to a normal value to avoid
gous blood donation. EPO use for the
further cardiac compromise
(i.e., Hgb
8
treatment of chemotherapy-induced ane-
to 12 g/dL). Again, the final endpoint may
mia remains controversial and is not rou-
be dependent on several factors including
tinely recommended in pediatric patients
nature of anemia, ongoing blood loss or lack
(see Chapter 25).
of production, baseline hemoglobin, and
The most common side effect of EPO
volume to be transfused. Care should be
administration is hypertension, which may
taken to avoid unnecessary exposure to
be somewhat alleviated with changes in the
multiple blood donors by maximal use of
dose and duration of administration.
the unit of blood, proper division of units in
Typical starting dose of EPO is 150 U/kg
the blood bank, and avoidance of opening
three times a week (IV) or subcutaneous
extra units for small quantities to meet a
(SC). CBCs and reticulocyte counts are
total volume. See Chapter 5 for product
checked weekly. Higher doses, and more
preparation, ordering, and premedication.
frequent dosing, may be necessary. Response
A posttransfusion hemoglobin can be
is usually seen within 1 to 2 weeks. Adequate
checked if necessary at any point after the
iron intake (3 mg/kg/d orally or intermittent
transfusion has been completed. Waiting
parenteral therapy) should be provided to
for “reequilibration” is anecdotal and un-
optimize effectiveness and prevent iron
necessary.
deficiency.
Transfusion therapy
Case study for review
Children with very severe anemia (Hgb < 5
g/dL) may require treatment with red cell
You are seeing a one year old for their well
transfusion, depending on the underlying
child check in clinic. As part of routine
disease and baseline hemoglobin status,
screening, a fingerstick hemoglobin is
duration of anemia, rapidity of onset, and
recommended.
hemodynamic stability. The pediatric liter-
ature is scarce as to the best method of
1. What questions in the history might help
transfusing such patients. However, it
screen for anemia?
appears to be common practice to give slow
2. What about the physical examination?
Approach to the Anemic Child
9
Multiple questions in the history can be
disease, or a suggestive newborn screen, an
helpful. Dietary screening for excessive milk
empiric trial of oral iron supplementation
intake is important in addition to asking
should not be performed. Similarly, if there
about intake of iron-rich foods such as
are signs that are consistent with a hemolytic
green leafy vegetables and red meat. One
process or a significant underlying disorder,
should also ask about pica behavior such as
further workup should be done. In these
eating dirt or ice and include questions
cases, it would be correct to next perform
regarding the age of the house to help screen
a CBC. If there are concerns for sickle cell
for lead paint exposure and ingestion. Any
disease or thalassemia, it would be reason-
sources of blood loss should also be
able to also perform hemoglobin electro-
explored including blood in the urine or
phoresis. If there are concerns for hemoly-
stool as well as frequent gum or nose bleed-
sis, labs including reticulocyte count, total
ing (more likely in an older child). Finally,
bilirubin, lactate dehydrogenase, and a
family history should be explored regarding
direct Coombs should be performed.
anemia during pregnancy, previous history
Finally, if there is concern for a systemic
of iron deficiency in siblings, and history of
illness such as leukemia, a manual differ-
hemoglobinopathies.
ential should be requested. Further workup
Physical examination to search for ane-
for iron deficiency
(ferritin and TIBC)
mia should be focused. Pallor, especially
as well as lead toxicity could be included
subconjunctival, perioral, and periungual
or deferred until the anemia is better
should be checked. Tachycardia, if present,
characterized utilizing the MCV and RDW
would be more consistent with acute anemia
on the CBC.
rather than well-compensated chronic ane-
mia. Splenomegaly, sclera icterus, and jaun-
dice may point to an acute or chronic
Suggested Reading
hemolytic picture.
Auerbach M, Ballard H. Clinical use of intrave-
You do the fingerstick hemoglobin in
nous iron: administration, efficacy, and safety.
clinic and it is 10.2 g/dL. The history is not
Hematology Am Soc Hematol Educ Program
suggestive of iron deficiency and the exam is
338-347, 2010.
unremarkable.
Bizzarro MJ, Colson E, Ehrenkranz RA. Differ-
ential diagnosis and management of anemia
3. What are the reasonable next steps?
in the newborn. Pediatr Clin North Am
Depending on the prevalence of iron
51:1087-1107, 2004.
deficiency in your population, it would be
Hermiston ML, Mentzer WC. A practical
reasonable at this point to give a 1 month
approach to the evaluation of the anemic
child. Pediatr Clin North Am 49:877-891,
trial of oral iron therapy. The family should
2002.
be counseled that oral iron tastes bad and
Janus J, Moerschel SK. Evaluation of anemia in
should be given with vitamin C (i.e., orange
children. Am Fam Physician 81:1462-1471,
juice) and not milk to improve absorption.
2010.
If there is a low likelihood of iron deficiency,
Richardon M. Microcytic anemia. Pediatr Rev
a family history of thalassemia or sickle cell
28:5-14, 2007.
Hemolytic Anemia
2
Red blood cells (RBCs) normally live for
has intermittent jaundice, and hemolytic or
about 100 to 120 days in the circulation.
red cell aplastic episodes associated with
Hemolytic anemia results from a reduced red
viral infection, splenomegaly, and choleli-
cell survival due to increased destruction. To
thiasis. However, the clinical presentation is
compensate for a reduced RBC life span, the
quite variable, with most severe cases pre-
bone marrow increases its output of red cells,
senting in the newborn period or early
a response mediated by erythropoietin.
childhood and milder cases presenting in
Destruction of red cells can be intravascular
adulthood.
(within the circulation) or extravascular (by
Several membrane protein defects are
phagocytic cells of the bone marrow, liver,
responsible for HS. Most result in the insta-
or spleen). Red cell injury or destruction is
bility of spectrin, one of the major red cell
associated with a transformation to a rigid
skeletal membrane proteins. Structural
or abnormal form. Altered cell deformability
changes that result as a consequence of pro-
then leads to decreased survival. Hemolytic
tein deficiency lead to membrane instability,
anemia may be inherited
(thalassemias,
loss of surface area, abnormal membrane
hemoglobinopathies, red cell enzyme defi-
permeability, and decreased red cell deform-
ciencies, or membrane defects) or acquired
ability. Metabolic depletion accentuates the
(immune-mediated, associated with infec-
defect in HS cells, which accounts for an
tion, or medication related). It can be chronic
increase in osmotic fragility after a 24-hour
or acute. Some types of low-grade chronic
incubation of whole blood at 37
C. The
hemolytic anemias can have acute exacerba-
splenic sinusoids prevent passage of nonde-
tions, such as a child with glucose-6-
formable spherocytic red cells. This explains
phosphate dehydrogenase (G6PD) deficiency
the occurrence of splenomegaly in HS and
with an exposure to fava beans or
the therapeutic effect of splenectomy.
naphthalene.
Patients with HS have a mild-to-moderate
chronic hemolytic anemia. Red cell indices
reveal a normal to low mean corpuscular
Red cell membrane disorders
volume (MCV) depending on the number
of microspherocytes. Cellular dehydration
Hereditary spherocytosis (HS) is the most
increases the mean corpuscular hemoglobin
common congenital red blood cell mem-
concentration
(characteristically
>36%).
brane disorder. The usual patient with HS
The red cell distribution width (RDW) is
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Hemolytic Anemia
11
elevated because of the variable presence
peripheral blood after splenectomy and
of microspherocytes and reticulocytes in
the osmotic fragility is more abnormal, the
proportion to the degree of hemolysis. The
hemoglobin value is normal. Platelet
peripheral blood smear can be diagnostic
counts frequently increase to more than
with the presence of spherocytes, although
1000
109/L immediately after splenec-
this can be a normal finding in the patient
tomy but return to normal levels over
with severe anemia and a resultant reticulo-
several weeks. No therapeutic interventions
cytosis. Osmotic fragility tests and ektacyto-
are required for postsplenectomy thrombo-
metry studies are characteristic for HS,
cytosis in patients with HS.
with increased fragility in hypotonic
To minimize the risk of sepsis due to
environments.
Haemophilus influenza and Streptococcus
As with other hemolytic anemias,
pneumoniae, the splenectomy procedure
affected individuals are susceptible to hypo-
(when necessary) is often postponed until
plastic crises during viral infections. Human
after the child’s fifth or sixth birthday.
parvovirus B19, a frequent pathogen and the
Patients should be immunized against these
organism responsible for erythema infectio-
organisms in addition to Neisseria meningi-
sum (fifth disease), selectively invades ery-
ditis prior to splenectomy and receive pen-
throid progenitor cells and may result in a
icillin prophylaxis following the procedure.
transient arrest in red cell proliferation.
The increase in penicillin-resistant strains of
Recovery begins within 7 to 10 days after
S. pneumoniae has raised questions regard-
infection and is usually completed by 4 to 6
ing the use of prophylactic penicillin. No
weeks. If the initial presentation of a patient
studies have determined the frequency of
with HS is during an aplastic crisis, a diag-
this problem in children receiving prophy-
nosis of HS might not be considered because
lactic penicillin after splenectomy.
the reticulocyte count will be low and the
peripheral blood smear may be nondiagnos-
tic. The family history of HS should be
Red cell enzyme deficiencies
explored; if it is positive, the patient should
be evaluated for HS after recovery from the
Glucose is the primary metabolic substrate
aplastic episode.
for the red cell. Because the mature red cell
Splenectomy is often considered for
does not contain mitochondria, it can
patients who have had severe hemolysis
metabolize glucose only by anaerobic
requiring transfusions or repeated hospital-
mechanisms. The two major metabolic
ization. In patients with mild hemolysis, the
pathways within the red cell are the
decision to perform splenectomy should be
Embden-Meyerhof pathway (EMP) and the
delayed; in many cases, it is not required.
hexose monophosphate shunt.
For pediatric patients who have excessive
Red cell morphological changes are
splenic size, an additional consideration for
minimal in patients with red cell enzyme
splenectomy is to diminish the risk of trau-
deficiency involving the EMP. Red cell
matic splenic rupture. The risks of splenec-
indices are usually normocytic and normo-
tomy must be considered before any clinical
chromic. The reticulocyte count is elevated
decision is made regarding the procedure.
in proportion to the extent of hemolysis.
Red cell survival returns to normal
Because many enzyme activities are
values after splenectomy unless an accessory
normally increased in young red cells, a
spleen develops. Although an increased
mild deficiency in one of the enzymes may
number of spherocytes can be seen in the
be obscured by the reticulocytosis.
12
Chapter 2
Pyruvate kinase (PK) deficiency is the
Mediterraneans, American Indians, South-
most common enzyme deficiency in the
east Asians, and Sephardic Jews are also
EMP. The inheritance pattern of this disor-
affected. In African-Americans, 12% of the
der is autosomal recessive. Homozygotes
male population has the deficiency, 18% of
usually have hemolytic anemia with spleno-
the female population is heterozygous, and
megaly, whereas heterozygotes are usually
2% of the female population is homozy-
asymptomatic. The disorder is found world-
gous. In Southeast Asians, G6PD deficiency
wide, although it is most common in Cau-
is found in approximately 6% of the male
casians of northern European descent. The
population. Most likely, the prevalence of
range of clinical expression is variable, from
this enzyme abnormality confers resistance
severe neonatal jaundice to a fully compen-
to malaria, thus its geographic distribution.
sated hemolytic anemia. Anemia is usually
Many variants of G6PD deficiency are
normochromic and normocytic, but macro-
known and have been characterized at the
cytes may be present shortly after a hemo-
biochemical and molecular levels. A variant
lytic crisis, reflecting erythroid hyperplasia
found in Mediterraneans is associated with
and early release of immature red cells. The
chronic hemolytic anemia. Other variants
osmotic fragility of red cells is normal to
are associated with an unstable enzyme that
slightly reduced. Diagnosis is confirmed by a
has normal levels in young red cells. These
quantitative assay for pyruvate kinase, by
result in hemolysis only in association with
the measurement of enzyme kinetics and
an oxidant challenge (as found in African-
glycolytic intermediates, and by family
Americans). In some cases of G6PD defi-
studies.
ciency, hemolysis may be triggered by the
Splenectomy is a therapeutic option for
oxidant intermediates generated during
PK-deficient patients. As with HS, the deci-
viral or bacterial infections or after ingestion
sion should be made on the basis of the
of oxidant compounds. Shortly after expo-
patient’s clinical course. Unlike HS patients,
sure to the oxidant, hemoglobin is oxidized
PK-deficient patients, although they
to methemoglobin and eventually dena-
improve after splenectomy, do not have
tured, forming intracellular inclusions
complete correction of their hemolytic ane-
called Heinz bodies that attach to the red
mia. As with all hemolytic anemias, these
cell membrane. This portion of the mem-
patients should have dietary supplementa-
brane may be removed by reticuloendothe-
tion with folic acid (1 mg/day) to prevent
lial cells resulting in a “bite” cell that has a
megaloblastic complications associated with
shortened survival owing to its loss of mem-
relative folate deficiency. Immunization
brane components. To compensate for
against H. influenza, S. pneumonia, and N.
hemolysis, red cell production is increased
meningiditis should be given, as well as
and thus the reticulocyte count is increased.
lifelong penicillin prophylaxis in the sple-
Individuals with the Mediterranean or
nectomized patient.
Asian forms of G6PD deficiency, in addition
Glucose-6-phosphate dehydrogenase
to being sensitive to infections and certain
(G6PD) deficiency is the most common
drugs, often have a chronic, moderately
X-linked red cell enzyme deficiency, with
severe anemia, with nonspherocytic red
partial expression in the female population
cells and jaundice. Hemolysis usually starts
and full expression in the affected male
in early childhood. Reticulocytosis is pres-
population. The distribution of G6PD defi-
ent and can increase the MCV.
ciency is worldwide, with the highest inci-
When a hemolytic crisis occurs in G6PD
dence in Africans and African-Americans.
deficiency (or favism), pallor, scleral icterus,
Hemolytic Anemia
13
hemoglobinemia, hemoglobinuria, and
jaundice, scleral icterus, elevated bilirubin,
splenomegaly may be noted. Plasma hapto-
and anemia with reticulocytosis). Fortu-
globin and hemopexin concentrations are
nately, the majority of pediatric cases of
low with a concomitant rise in plasma-free
autoimmune hemolytic anemia (AIHA) are
hemoglobin. The peripheral smear shows
acute and self-limited.
the fragmented bite cells and polychromato-
In the DAT, the patient’s erythrocytes
philic cells. Red cell indices may be normal.
are washed and then incubated with specific
Special stains can detect Heinz bodies in the
antiglobulin antisera (usually anti-IgG and
cells during the first few days of hemolysis.
anti-C3d). Agglutination indicates a posi-
A diagnosis of G6PD deficiency should be
tive test. In patients with severe immune-
based on family history, ethnicity, laboratory
mediated hemolytic anemia, the DAT is
features, physical findings, and clinical sus-
often strongly positive, although the
picion suggested by a recent exposure to
strength of the reaction does not always
oxidants with resultant acute hemolysis. The
correlate to the severity of the disease. Sim-
diagnosis is confirmed by a quantitative
ilarly, up to 80% of patients will have anti-
enzyme assay or by molecular analysis of the
bodies in the serum as well, measured by the
gene. Since reticulocytes may have a normal
indirect Coombs (indirect antiglobulin test,
level of G6PD enzyme activity, screening tests
IAT). In the IAT, donor erythrocytes are
during acute hemolysis may be falsely ele-
incubated with test serum, washed, and then
vated; therefore, it is important to test once
incubated with specific antiglobulin anti-
the hemolytic crisis has ended and the patient
sera. Agglutination again indicates a posi-
again has mature red blood cells. Treatment is
tive test. Of note, patients without symp-
directed toward supportive care during the
toms of hemolysis may have a positive DAT
acute event and counseling regarding pre-
and/or IAT; therefore, screening is only
vention of future hemolytic crises. In patients
recommended in the setting of clinical and
with chronic hemolysis, dietary supplemen-
laboratory signs of hemolysis. In approxi-
tation with folic acid (1 mg/day) is recom-
mately 5% to 10% of cases, patients may
mended. Use of vitamin E, 500 mg/day, may
have an AIHA with a negative DAT.
improve red cell survival in patients with
The initiation of autoimmunity is poorly
chronic hemolysis.
understood. Viral syndromes are often pro-
posed as a culprit, although causation has
been hard to prove. A majority of cases of
Autoimmune hemolytic anemia
AIHA in pediatrics are due to “warm” anti-
bodies, so named because they react at
In addition to intrinsic causes of hemolytic
37 C. These are often secondary to a viral
anemia, patients may also develop an auto-
syndrome, although patients with an under-
antibody and/or alloantibody toward their
lying autoimmune disease or oncological
red blood cells. The underlying cause for
process can also present with a warm AIHA.
this antibody formation is often idiopathic
The formation of IgG antibodies leads
or due to a secondary condition including
to extravascular hemolysis in which pieces
drugs, infectious syndromes, autoimmune
of the red cell membrane are sequentially
diseases, or an oncological process. A pos-
removed during passages through the
itive direct antiglobulin test (DAT, direct
spleen. Patients may also develop direct
Coombs) is pathognomonic for immune-
Coombs positive hemolytic anemia and
mediated hemolysis with the appropriate
immune-mediated
thrombocytopenia
clinical and laboratory findings
(i.e.,
(Evans syndrome).
14
Chapter 2
Hemolytic disease of the
damaged by the formation of an intravas-
newborn
cular fibrin mesh due to hypercoagulability,
leading to fragmentation (e.g., schistocytes)
Intrinsic causes of hemolytic anemia can
and intravascular hemolysis.
present as jaundice in the newborn period.
These syndromes must be differentiated
Evaluation
from hemolytic disease of the newborn in
which alloimmunization in the mother
The evaluation of hemolytic anemia includes
occurs due to foreign RBC antigens from
a thorough history assessing for evidence of
the fetus. RBC antigens can either be major
chronic hemolytic anemia and possible pre-
(ABO) or minor (Rh, Kell, Duffy, etc.). For
cipitants of an acute event (see Figure 2.1).
ABO hemolytic disease, typically the mother
The family history is equally important
is type O and the fetus is type A; anti-A
and questions to ask include:
antibodies subsequently produced by the
mother then traverse the placenta leading
History of newborn jaundice
to hemolytic anemia in the fetus.
Gallstones
RhoGAM (Rho[D] Immune Globulin) has
Splenomegaly or splenectomy
virtually eliminated hemolytic disease in the
Episodes of dark urine and/or yellow skin/
Rh-negative
(D-negative) mother with a
sclerae
Rh-positive (D-positive) fetus although is
Anemia unresponsive to iron
still possible in the mother not receiving
supplementation
prenatal care. Many cases of hemolytic dis-
Medications
ease of the newborn are now due to other
Environmental exposures
minor RBC antigens with varying levels of
Ethnicity
clinical severity. AIHA in the mother can
Dietary history
also lead to hemolytic disease of the new-
born. In this case, maternal antibodies tra-
The physical exam should be complete, but
verse the placenta and are transferred to the
focused on:
fetus. If the mother is DAT positive but does
Skin color
(pallor, jaundice, and icteric
not have clinical signs of hemolytic anemia,
sclerae)
there is usually no risk to the fetus.
Facial bone changes
(extramedullary
hematopoiesis)
Abdominal fullness and splenomegaly
Microangiopathic hemolytic
anemia
The laboratory evaluation includes:
Complete blood count, RBC indices, and
Microangiopathic hemolytic anemias are
reticulocyte count (")
due to extracorpuscular abnormalities and
Peripheral blood smear (assess for fragmen-
are not associated with antibody formation.
ted forms or evidence of inherited ane-
Causes include disseminated intravascular
mia with specific morphological
coagulation, thrombotic thrombocytopenic
abnormalities)
purpura/hemolytic uremic syndrome, pre-
Bilirubin (")
eclampsia, malignant hypertension, valvular
Coombs test, direct and indirect (to exclude
abnormalities, and march hemoglobinuria.
antibody-mediated red cell destruction)
In these cases, red blood cells travel through
Urinalysis (for heme, bili)
damaged blood vessels or heart valves or are
Free plasma hemoglobin (")
Hemolytic Anemia
15
History
Physical examination
Low hemoglobin
Increased reticulocyte count
Peripheral smear
Morphologic characteristics
Coombs test (DAT)
Immune mediated
Coagulation studies
ANA
Identification of antibody
Red cell
Spherocytes
Target cells
Normal
fragmentation
Elliptocytes
Sickle cells
Nonspecific
abnormalities
Consider
RBC enzyme studies
Ceruloplasmin
DIC
Coagulation
Family studies
Hemoglobin
Enzyme deficiency
screen
Osmotic fragility or
electrophoresis
PK deficiency
(fibrinogen,
ektacytometry
PT, PTT, FDP)
G6PD deficiency
Platelet count
Wilson disease
Blood cultures
Antibiotics
HUS
Blood pressure
Hereditary spherocytosis
Hgb SS
BUN, Cr
Hereditary elliptocytosis
Hgb SC
Urinalysis
Hypersplenism
Hgb CC
Platelet count
Coombs (DAT) negative
Hgb S/ β-thalassemia
Cardiac prosthesis
autoimmune hemolysis
Figure 2.1 Diagnostic approach to the child with hemolytic anemia. (Abbreviations: DAT, direct
antiglobulin test; ANA, anti-nuclear antibody; DIC, disseminated intravascular coagulation;
PT, prothrombin time; PTT, partial thromboplastin time; FDP, fibrin degradation products;
HUS, hemolytic uremic syndrome; BUN, blood urea nitrogen; Cr, creatinine; PK, pyruvate kinase;
G6PD, glucose-6-phosphate dehydrogenase).
Specific tests for diagnosis may include:
cells is increased when the surface area to
Osmotic fragility
volume ratio of the red cells is decreased, as
Ektacytometry
in hereditary spherocytosis, in which mem-
Red cell enzyme defects (G6PD and PK)
brane instability results in membrane loss
Red cell membrane defects (HS)
and decreased surface area. Conversely,
osmotic fragility is decreased in liver disease
The osmotic fragility test is used to measure
as the ratio of the red cell surface area to
the osmotic resistance of red cells. Red cells
volume is increased. Ektacytometry mea-
are incubated under hypotonic conditions,
sures the deformability of red cells subjected
and their ability to swell before lysis is
simultaneously to shear stress and osmotic
determined. The osmotic fragility of red
stress.
16
Chapter 2
Treatment
fatigued over the last few days with a red
color to the urine. Fingerstick hemoglobin
Therapy depends on the underlying cause
at the pediatrician’s office reveals a hemo-
of the anemia and the degree of acute
globin of 5 g/dL prior to transfer to the ED.
hemolysis. In chronic hemolysis, such as
On the basis of this history and hemoglobin,
that associated with hereditary spherocyto-
it appears that the child is suffering from a
sis, splenectomy is often recommended to
hemolytic anemia.
decrease the degree of splenic destruction
1. What initial lab studies will help confirm
and level of anemia and to decrease the
the diagnosis and also help with the initial
incidence of bilirubin gallstones. This
treatment plan?
therapy must be now weighed against the
potential long-term complications of sple-
Initial lab studies should include a com-
nectomy including risk for infection,
plete blood count with reticulocyte count.
thrombosis, and pulmonary hypertension.
The reticulocyte count is an important first
In other forms of inherited anemias in
step to confirm that the patient is undergoing
which the hemolysis is more significant and
hemolysis, which should present with a low
even life-threatening, such as thalassemia or
hemoglobin and a resultant increase in the
some forms of enzymopathies, chronic
reticulocyte count. A low reticulocyte count
transfusion therapy is recommended. Other
in this setting should lead to consideration of
general measures include folic acid replace-
alternative diagnoses such as viral suppres-
ment due to high cell turnover, avoidance of
sion (although one would not expect hemo-
oxidant chemicals and drugs, and iron che-
lysis). A complete metabolic panel as well as
lation therapy as indicated for transfusion-
lactate dehydrogenase (LDH) should be done
related iron overload.
to ensure that the patient is actually suffering
Immune hemolytic anemias can require
from jaundice (elevated total bilirubin) and
more immediate and aggressive therapy.
hemolysis (elevated LDH and AST). A direct
The underlying disease, if present and
and indirect Coombs
(DAT/IAT) is an
identifiable, warrants treatment. Addition-
important first step to determine if the
ally, the use of corticosteroids in high doses
is frequently necessary. Splenectomy and
patient is undergoing an immune or nonim-
immunosuppressive drugs have also
mune hemolytic anemia.
been successful. Microangiopathic hemo-
The patient is noted to have a hemoglobin
lytic anemias can also be severe and life-
of 4.6 g/dL with 12.6% reticulocytes. One
threatening. Treatment should again first be
should first determine if the patient is having
directed toward the primary disorder to
an appropriate bone marrow response to
remove the cause of trauma, if possible.
anemia by calculating the reticulocyte index
Transfusions are frequently necessary and
(RI):
splenectomy may be needed in some
patients with severe hypersplenism.
RI ¼ Reticulocyte count ð%Þ
current hemoglobin
expected hemoglobin
Case study for review
In this case, the RI is 12.6%
(4.6/13) ¼ 4.5.
You are seeing a 6-year-old child in the
An RI
3.0 is consistent with an appro-
emergency department. The family notes
priate bone marrow response to anemia, and
that the child has been jaundiced and
therefore helps to rule out bone marrow
Hemolytic Anemia
17
dysfunction in this case. Modern blood cell
of symptomatic anemia and congestive heart
analyzers have the ability to calculate the
failure.
absolute reticulocyte count and the fraction
4. How should your treatment change at
of “immature” reticulocytes directly. Patients
this point?
who are demonstrating an appropriate
response to hemolysis will have an elevated
Since the patient has a falling hemoglobin
absolute reticulocyte count and immature
with clinical signs of cardiac instability and
reticulocyte fraction; these will be low or
volume overload, the patient should be
normal in patients with an inadequate
transfused. The term
“least incompatible
response.
unit” has been used in the past but is a
Other labs include a total bilirubin of
misnomer if phenotypically matched blood
6.7 mg/dL, LDH of 936 U/L (reference range
is given. The patient may not have a normal
313 to 618 U/L), and AST of 161 U/L. DAT is
increase in hemoglobin with transfusion
noted to be positive for IgG and C3d.
due to continued hemolysis and the poten-
2. What is the likely diagnosis?
tial for increased, bystander hemolysis with
transfusion. Because of the cardiac instabil-
With the positive DAT to IgG and com-
ity, it is advisable to give the transfusion
plement and clinical and laboratory signs of
slowly and monitor for worsening cardiac
hemolysis, warm antibody-mediated AIHA
function. Finally, a change in therapy would
is the likely diagnosis. It should be noted
be advisable at this point with either intra-
that a positive DAT without clinical and
venous immunoglobulin or a different
laboratory signs of hemolysis is not suffi-
immunosuppressant such as cyclosporine,
cient for the diagnosis of AIHA.
cyclophosphamide, or rituximab (monoclo-
3. What should be the initial treatment plan?
nal antibody to CD20).
The patient is started on steroid therapy,
IV methylprednisolone 1 mg/kg BID. After a
couple of days, the hemoglobin has contin-
Suggested Reading
ued to decrease to 3 g/dL even though the
methylprednisolone has been increased to
Garratty G. Immune hemolytic anemia—a
4 mg/kg BID and the patient is showing signs
primer. Semin Hematol 42:119-121, 2005.
Sickle Cell Disease
3
Sickle cell disease refers to a group of genetic
it is not possible to predict the severity of
disorders that share a common feature:
disease in advance of severe complications.
hemoglobin S (Hgb S) alone or in combi-
Generally, children who have vaso-occlusion
nation with another abnormal hemoglobin.
and other complications have a more severe
The sickle cell diseases are inherited in
course. Increased leukocyte count, decreased
an autosomal codominant manner. The
hemoglobin with concomitant increased
molecular defect in Hgb S is due to the
reticulocytosis, as well as frequency and
substitution of valine for glutamic acid at
severity of vaso-occlusive episodes (VOEs)
the sixth position of the b-globin chain.
are associated with increased morbidity and
The change of location of this substitution
mortality.
results in polymerization of the hemoglobin
Alpha-thalassemia (frequency 1% to 3%
and causes the red cells to transform from
in African-Americans) may be coinherited
deformable biconcave discs into rigid,
with sickle cell trait or disease. Individuals
sickle-shape cells. Hypoxia, acidosis, and
who have both a-thalassemia and sickle cell
hypertonicity facilitate polymer formation.
anemia are less anemic than those who have
The most common combinations of
sickle cell anemia alone due to a more
abnormal hemoglobins are
(1) Hgb SS,
similar concentration of a- and b-globulins.
(2) Hgb SC, and (3) Hgb S with a beta-
However, a-thalassemia trait does not
thalassemia, either Sbþ or Sb0. The most
appear to prevent frequency or severity of
severely affected individuals have either Hgb
vaso-occlusive complications, resulting in
SS or Sb0 (no normal beta-globin produc-
eventual end-organ damage.
tion). Individuals with Hgb Sbþ have
Sickle cell disease is not uncommon and
decreased beta-globin production and less
has developed due to protection from
severe disease, whereas children who have
malaria in those with sickle cell trait. In
Hgb SC have intermediate severity of dis-
African-Americans, the frequency of genetic
ease. There is phenotypic overlap between
alteration is quite high: 8% have the Hgb S
Hgb SS and Hgb SC; some children with
gene,
4% the Hgb C gene, and 1% the
Hgb SC are more symptomatic than chil-
b-thalassemia gene. Approximately
1
in
dren with Hgb SS. There are many variables
600 African-American infants has sickle cell
to expression of this hemoglobinopathy
anemia. Sickle cell disease also occurs in
including haplotypes, Hgb F concentration,
children from the Middle East, India, Cen-
and other yet to be delineated factors. As yet,
tral and South America, and the Caribbean.
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Sickle Cell Disease
19
All children who have sickle cell hemo-
Most children with sickle cell disease are
globinopathies have a variable degree of
identified at birth, started on prophylactic
hemolytic anemia and vaso-occlusive tis-
penicillin by age 2 months, and aggressively
sue ischemia resulting in numerous clinical
monitored and treated for signs of infection.
complications. Organs most sensitive to
However, with the increasing concern for
the ischemic-hypoxic injury of red cell
bacterial antibiotic resistance, health care
sickling are the lungs, spleen, kidneys,
providers need to be vigilant when con-
bone marrow, eyes, brain, and the heads
fronted with an infant or a child who has
of the humeri and femurs. Sickling has
fever (
38.3
C) and/or appears ill. Overall,
both acute and long-term implications for
Streptococcus pneumoniae is responsible for
organ function. Cerebral vascular disease
>80% of the morbidity of infection. In
can be subtle, causing only abnormal
some areas of the United States, up to
neuropsychological testing or it can be
50% of pneumococcal isolates are penicillin
catastrophic, resulting in hemiparesis,
resistant. Infections can precipitate vaso-
coma, or death; acute pulmonary sickling
occlusive episodes and other complications
causes lung injury leading to restrictive
of sickle cell anemia and, in this population,
lung disease and eventually pulmonary
can quickly become fulminant. Although
hypertension; osteonecrosis of the femoral
the American Academy of Pediatrics guide-
head can be debilitating, resulting in the
lines recommend the discontinuation of
need for hip replacement; untreated reti-
penicillin prophylaxis after 6 years of age,
nopathy can lead to blindness; and, sickle
our institutional practice is to continue it
cell nephropathy can cause asymptomatic
as long as possible
(until compliance
proteinuria, an early sign of the risk of
becomes an issue) due to the high risk of
eventual renal failure.
S. pneumoniae infection.
Now that newborn hemoglobinopathy
Additional bacteria that cause morbidity
testing is mandatory in most states, chil-
and mortality include Haemophilus influen-
dren are diagnosed early and receive
zae, Neisseria meningitidis, Mycoplasma
appropriate care before they are at risk for
pneumoniae, Staphylococcus aureus, Salmo-
complications. All infants who have an
nella species, Escherichia coli, and Strepto-
electrophoretic pattern of Hgb FS at
coccus pyogenes. The S. pneumoniae and
birth will have some form of sickle cell
H. influenzae vaccines have importantly
disease.
resulted in a lowered case rate of sepsis from
these organisms. Viral infections, particu-
larly parvovirus B19, can cause severe aplas-
Fever and infection in sickle cell
tic crises as well as acute chest syndrome
disease
(ACS).
Infants, young children, and any patient
Susceptibility to infection is increased not
who has a central venous catheter with a
only because of loss of splenic function due
fever
(
38.3
C) should have a complete
to infarction but also because of other
evaluation and laboratories including CBC
acquired immunologic abnormalities.
with differential, reticulocyte count, blood
This can result in life-threatening episodes
culture, urinalysis, and urine culture (see
of sepsis. Recognition of this susceptibility
Figure 3.1). A chest radiograph should also
and aggressive medical management have
be obtained. Meningitis can occur in chil-
resulted in an increased life span for most
dren with sickle cell disease but routine
patients.
lumbar puncture without physical signs of
20
Chapter 3
Rapid assessment
history & physical
Infant, child less than
Septic appearing child
2-3 years of age:
nontoxic appearing
Rapid assessment
History (last dose of penicillin):
for degree of respiratory and
Cardiovascular and respiratory status,
cardiovascular compromise;
splenomegaly, jaundice, pain;
meningismus;
neurologic screen for weakness and
cardiovascular monitoring and
altered mental status
pulse oximetry
IV access
* Respiratory distress or
IV access
Blood culture, CBC with
pulse oximetry saturation
Blood culture, CBC with
differential and reticulocyte count
< 90%
differential and reticulocyte count
Catherized urine culture
Oxygen
Catherized urine culture
Consider LP
Chest X-ray
Consider LP
Type and cross match
ABG
Chest X-ray
Chest X-ray
Antibiotics
Fluid bolus to
Antibiotics
Ceftriaxone 50 mg/kg IV push
maintain blood pressure
Ceftriaxone 50 mg/kg
or rapid infusion
Note: fluid overload can lead
(plus azithromycin if CXR findings
to ACS
concerning for atypical
pneumonia)
Evaluate intervention;
if stable complete physical exam
*Respiratory distress: It is preferred to
and history and
obtain an ABG on room air; in a critically ill
review labs
child oxygen should be given first, and
oxygenation status assessed after stabilization
Figure 3.1 Fever in a child with sickle cell disease.
(Abbreviations: IV, intravenous; CBC, complete
blood count; LP, lumbar puncture; ABG, arterial blood gas; CXR, chest x-ray; ACS, acute chest
syndrome).
meningitis is not warranted. Urosepsis is
practitioners would recommend complete
common in sickle cell patients of all ages.
evaluation in all patients with a fever.
All infants (up to 2 to 3 years of age) should
Children with a documented fever
be admitted and cultures followed for 48 to
should have the same workup as infants,
72 hours. These patients should not be
although admission is not required if they
discharged during this time even if they
are well appearing, with reassuring labs and
appear well and are afebrile. Some
chest radiograph, and follow-up can be
Sickle Cell Disease
21
reasonably guaranteed. Acute chest syn-
occur and eventually lead to collapse of the
drome should be high on the differential
vertebral plates and compression. The clas-
in all children with fever. While physical
sic radiographic appearance is of
“fish
examination is essential, 60% of pulmonary
mouth” disc spaces and the “step” sign (a
infiltrates in children with sickle cell disease
depression in the central part of the verte-
and fever will be missed on exam alone,
bral body). Back pain is a common symp-
therefore chest radiography is essential. If
tom in sickle cell disease, likely as a result of
there is another possible source of infection,
recurrent infarction and vertebral compres-
appropriate cultures should be obtained. If a
sion. Infarction in the long bones can cause
child appears to be ill, has a temperature of
swelling and edema in the overlying soft
38.3
C or higher, or has an elevated WBC
tissues. It may be difficult to differentiate
with a left shift, treatment should be imme-
VOE from acute osteomyelitis. Although
diate with parenteral antibiotics and close
uncommon, infection should be consid-
monitoring, preferably in a hospital setting.
ered. Osteomyelitis may be ruled out by
Of note, studies have shown that the higher
close clinical observation, blood cultures,
the temperature, the more likely the risk for
and occasionally, aspiration of the affected
bacterial sepsis. If there is difficulty with
area. Plain radiographs are not helpful in
intravenous
(IV) access, ceftriaxone can
the early stages of infection and bone
be given intramuscular (IM) while access
scans may not differentiate a simple infarct
is being obtained.
from osteomyelitis.
Children who do not appear septic, in
Dactylitis, or hand-foot syndrome,
whom there is a low index of suspicion, and
refers to painful swelling of the hands and
who are older than 2 to 3 years of age (per
feet. This is seen exclusively in infants and
institutional preference), can be treated as
children (<5 years of age). It presents with
outpatients with IV/IM ceftriaxone while
pain, low-grade fever, and diffuse nonpit-
awaiting culture results, only if close mon-
ting edema of the dorsum of the hands and
itoring can be assured (parents can be con-
feet, which extends to the fingers. One or
tacted by telephone and daily evaluation is
more extremities may be affected at one
easily done while still febrile). If all cultures
time. Radiographic changes (periostitis and
are negative after 2 to 3 days and the child is
subperiosteal new bone formation with
afebrile without clinical symptoms, the anti-
periosteal elevation) may appear
1 week
biotics and follow-up can be discontinued.
or more after the clinical presentation.
All patients who appear ill should be
Therapy is analgesics, hydration, and paren-
hospitalized and treated for presumed infec-
tal reassurance. Although the swelling or
tion. Many children will develop increasing
radiographic changes may persist for weeks,
symptoms after being evaluated and hospi-
the syndrome is almost always self-limited.
talized. Acute chest syndrome commonly
Transfusions and antibiotics are not neces-
occurs after hydration and is frequently pre-
sary, unless there is concern for infection or
cipitated by a vaso-occlusive pain episode.
the medical condition worsens.
Occasionally, there is a precipitating
event such as hypoxia (obstructive sleep
Vaso-occlusive episodes
apnea), fever, viral illness, or dehydration
that leads to the vaso-occlusive episode. Few
The bones and joints are the major sites
children have frequent pain episodes; it
of pain in sickle cell disease. In trabecular
should not be taken for granted that each
bones, such as the vertebrae, infarction can
of these episodes is similar, and a source for
22
Chapter 3
each painful event should be sought. If the
High-risk factors for complications
pain changes or is very persistent, the child
other than VOE in children with pain:
should be reevaluated. If a young child has a
* Fever
101 F
(38.3 C) and signs of
symptom that is interpreted as pain, such as
infection
refusal to walk or a limp, also consider that
* Acute pulmonary symptoms (chest pain,
the cause may be due to an acute CNS event.
hypoxia, abnormal ausculatory exam)
The most common type of pain is mus-
* Persistent vomiting
culoskeletal pain. The pain may be of any
* Pain that is unusual for the patient
configuration: isolated, multifocal, symmet-
* Severe abdominal pain
ric, migratory, or associated with erythema
* Extremity weakness or loss of function
or swelling. There can be low-grade fevers,
* Any neurological symptoms
possibly associated with other clinical symp-
* Severe headache
toms. It can sometimes be difficult to dis-
* Acute joint swelling
tinguish pain from an infection, synovitis,
or other pathological process.
Children are frequently seen with
Pain management
abdominal pain. A surgical abdominal
problem needs to be considered. Children
Following are the recommendations for the
who have sickle cell anemia have a high
management of vaso-occlusive events. How-
incidence of cholecystitis. Also consider
ever, many children and adolescents need
pancreatitis, urinary tract infection, pelvic
individualized care plans for their routine
inflammatory disease, and pneumonia
treatment.
presenting as abdominal pain. Ileus and
acute chest syndrome are frequent com-
Outpatient
plications of abdominal vaso-occlusive
Mild pain
episodes.
* Increase fluids to 1 to 1.25
maintenance.
Vaso-occlusive episodes can last for
Water, fruit juice, and fruit drinks as well as
many days. Do not assume that the episode
decaffeinated soda are recommended.
is controlled when the acute administration of
* Acetaminophen without codeine (15 mg/
analgesia is effective. Children with these
kg q4 to 6 hours) orally should be tried prior
symptoms may have a medical problem that
to using acetaminophen with codeine
needs aggressive treatment beyond therapy
(1.0 mg codeine/kg q4
to
6 hours). For
for their pain. A vaso-occlusive episode is a
many children, acetaminophen alone is not
diagnosis of exclusion. The pain needs to be
sufficient for vaso-occlusive pain.
treated while there is a workup for potential
* Ibuprofen (10 mg/kg q6 to 8 hours) orally
other causes of pain beyond the vaso-occlu-
should be used routinely, every 8 hours even
sive episode itself.
if pain is temporarily controlled.
A reliable tool should be used to assess a
* Rest and heat to the area of pain.
child’s level of pain. The assessment should
* Relaxation and biofeedback exercises,
be modified depending on the age and
such as positive visualization and meditation.
developmental level of the child, and the
pain tool should be able to rate both sub-
Severe pain
jective and objective aspects of the child’s
* IV hydration: bolus to correct fluid losses,
pain. Physicians should be familiar with
then IV with D5W 1/2 NS at 1 to 1.25
their use and refer to them when assessing
maintenance (IV þ PO). Use caution not
children and adolescents with pain.
to overhydrate, especially if there are any
Sickle Cell Disease
23
pulmonary symptoms (overhydration can
days (10 to
15 doses). There should be
lead to pulmonary edema and precipitate
follow-up within 72 hours. If this regimen
acute chest syndrome). Note that if the
is not successful, if other symptoms develop
patient does not appear volume depleted,
with hydration, or a fever develops, the
an IV fluid bolus is not routinely required.
patient should be admitted and observed.
* Analgesics
(IV): Morphine:
0.1
to
0.15 mg/kg/dose q3 to 4 hours. In children
Inpatient
<2 years who have not been exposed to
As noted above, the administration of nar-
narcotic analgesics, 0.05 to 0.1 mg/kg should
cotics to acutely ill patients requires careful
be used. The pharmacokinetics of morphine
monitoring. A plan for pain management is
differs between individual children and
defined on admission and followed for the
doses must be titrated for individual
duration of hospitalization. In addition to
patients. Both underdosing and overdosing
whirlpool therapy, warm packs, and bio-
need to be avoided. Careful management
feedback therapy (as available), routine care
is required! Previous medication history is
includes:
often a good starting place.
* Ketorolac: (child) IV: 1 mg/kg/load, then
Management
0.5 mg/kg q6 hours (max 30 mg); (adult) IV:
* Correct acute losses due to dehydration.
30 to 60 mg/load, then 15 to 30 mg/kg q6
Do not overhydrate children who have pul-
hours. GI bleeding has been reported, and
monary symptoms.
an H2 blocker such as ranitidine is required
* Total fluid intake (IV þ PO) should be 1
if ketorolac is given IV. Morphine and
to 1.25
daily maintenance fluids; do not
ketorolac can be given simultaneously or
overhydrate.
in succession for the initial treatment of
* Monitor daily weight, record strict intake
pain in children. Added to narcotic therapy,
and output (l&O), and observe for signs of
ketorolac increases analgesia and has a nar-
fluid overload.
cotic sparing effect. Ketorolac is very useful
* Encourage ambulation, sitting up in bed,
in pain control, but should not be used for
and taking deep breaths.
more than 5 days. There are numerous
* Incentive spirometry, 10 times an hour,
other analgesic medications that have been
every hour while awake.
used to treat sickle cell pain.
Analgesics
Upon presentation to the clinic or emer-
* Pain medication should be administered
gency room, the child or adolescent should
on a fixed-time schedule, with an interval
be rapidly assessed and treated for pain.
that does not extend beyond the duration of
Initially, both morphine and ketorolac
the analgesic effect. Do not give narcotics on
should be given at the maximum recom-
a PRN basis except for when the patient is
mended doses. If the pain returns, repeat the
ready for discharge.
parenteral narcotic dose; it is unlikely that
* Titration schedules
(requires a written
oral analgesics will be successful if this reg-
plan, close observation, and a flow sheet
imen is not effective. If pain relief can be
to monitor effectiveness; sedation and
achieved and maintained for
3
hours,
hypoventilation can lead to hypoxia and
administer an oral narcotic and observe for
increased sickling and pain).
1 hour. If this regimen is successful, give a
* Patient-controlled analgesia (PCA). This
prescription of narcotic and nonsteroidal
is the method of choice for controlling pain
anti-inflammatory medications for several
in children who are
7 years of age or older.
24
Chapter 3
PCA protocols may be hospital specific.
Drug addiction is extremely rare and
Generally, there is an initial bolus
should not be a primary concern. The goal
(0.1 mg/kg morphine), and an on-demand
should be to provide patients with ade-
dose (0.01 to 0.04 mg/kg morphine) with a
quate relief by understanding the pharma-
10 to 20 minute lock out, and a 1 hour
cology of the medications, drug tolerance,
maximum of 0.1 to 0.15 mg/kg of mor-
and physical dependence. Drug tolerance
phine. At night, there may be a need for a
is common, and withdrawal symptoms
continuous low-dose infusion
(0.01
to
after hospitalization are probably under-
0.02 mg/kg/h morphine) without changing
reported by the patients. All patients
the 1 hour lock out.
should start a narcotic taper while in the
hospital and complete this as an outpa-
Side effects of narcotics include respiratory
tient. On average, patients admitted for the
depression, nausea, vomiting, pruritus,
management of VOE require 3 to 7 days of
hypotension, constipation, inappropriate
inpatient care.
secretion of anti-diuretic hormone, and
change in seizure threshold. Low-dose nal-
oxone drip has been used for severe pruritus
Acute chest syndrome/
and urinary retention due to morphine. The
pneumonia
most common side effects of nausea, vomit-
ing, and pruritus resolve over time. IV or
Acute chest syndrome (ACS) is defined as
PO diphenhydramine may be used safely in
the development of a new pulmonary infil-
this setting and can have a synergistic pos-
trate in the presence of fever or respiratory
itive effect with morphine. Meperidine
symptoms. As chest radiograph changes may
should not be substituted for morphine.
be delayed, the diagnosis may not be appar-
The metabolites of meperidine can accumu-
ent at presentation. Approximately 50% of
late and cause seizures, especially if used
children with sickle cell disease will have
over a longer period at high doses. A plan
acute chest syndrome. ACS is frequently
for withdrawal should be a part of discharge
caused by community-acquired pathogens
planning; clonidine hydrochloride has been
such as chlamydia, mycoplasma, and other
used successfully in this setting, but should
bacterial or viral organisms. In older chil-
be carefully monitored.
dren, adolescents, and young adults, ACS
All patients who are hospitalized for pain
may be more commonly associated with
should have an incentive spirometer at the
vaso-occlusion, infarction, fat embolism, or
bedside. Ensure that the patient has been
in situ sickling. These episodes are charac-
instructed on its usage and can demonstrate
terized by chest pain, fever, hypoxia, and
it appropriately.
pulmonary infiltrates. Although ACS usually
Alternative methods of pain control
improves with medical management, it can
(behavior modification, relaxation, visuali-
present with or progress rapidly to respira-
zation, self-hypnosis, and transcutaneous
tory failure (acute respiratory distress syn-
electrical nerve stimulation) are helpful
drome), requiring mechanical ventilation
adjuvants in the outpatient and inpatient
and emergent exchange transfusion.
setting but should not replace standard
Sixty percent of children who have pneu-
therapy. Children should have access to a
monia on X-ray will be missed by physical
psychologist who is experienced in the man-
exam alone. In addition, many children with
agement of sickle cell pain.
a negative chest film on admission will
Sickle Cell Disease
25
Child presents with respiratory
distress, cough, and/or chest pain,
with or without fever
Physical exam:
Evidence of respiratory distress: abnormal or absent breath sounds,
egophony, tachypnea, splinting
(60% of children with infiltrate will have a normal examination)
Pulse oximetry and chest radiography
CBC, reticulocyte count, blood culture
Dyspnea and tachypnea
Extensive or progressive infiltrate
Infiltrate without
ABG PaO2 < 70 mmHg on room air
respiratory distress and
Evidence of right heart failure or
stable pulse oximetry > 89%
pulmonary hypertension
Antibiotics:
Ceftriaxone and azithromycin
Intensive care unit
Incentive spirometry
Emergent transfusion:
Maintain fluid balance
straight or exchange
Lasix for fluid retention (I > O)
(exchange if severe ACS,
Analgesics per pain protocol
high hematocrit, or clinically
Continuous pluse oximetry
worsens or remains hypoxic
Albuterol aerosols
after straight transfusion)
Frequent re-evaluation
Children who have an episode of life threatening acute chest syndrome will need to be on
hydroxyurea therapy or chronically transfused. In addition, they should be followed for
evidence of pulmonary hypertension with cardiac echocardiography
Figure 3.2 Sickle-cell disease with acute pulmonary infiltrate. (Abbreviations: CBC, complete blood
count; ABG, arterial blood gas; ACS, acute chest syndrome).
develop findings after hydration. All chil-
incentive spirometry are helpful adjuncts
dren who have symptoms of pulmonary
in treatment and prevention of worsening
disease, such as fever, shortness of breath,
ACS. If there is an infiltrate, the patient
tachypnea, chest pain, cough, wheezing,
should be closely observed for hypoxia
rales, or dullness to percussion, should
and progression of pulmonary infiltrates,
have an assessment of oxygen saturation,
with repeat chest radiographs. Overhydra-
a chest radiograph, receive parenteral anti-
tion can lead to pulmonary edema and
biotics, and be admitted to the hospital
exacerbate acute chest syndrome. Over-
(see Figure
3.2). Bronchodilators and sedation with narcotics can also lead to
26
Chapter 3
hypoventilation, increasing the risk for ACS.
Hgb SC disease have relatively more fat
Narcotics should be used with caution in
in their marrow and resultantly can have
this setting.
more severe pulmonary fat emboli when
Acute chest syndrome can develop in a
their course is complicated by acute chest
matter of hours and is associated with
syndrome.
a high rate of morbidity and mortality.
Stroke and other central nervous sys-
Children should have oxygen saturation
tem events are more common in children
monitored and, if indicated, arterial
with ACS in the 2 week period after the
blood gases also. Oxygen therapy should
event.
be used only for significant hypoxia (O2
saturation
92%). Supplemental oxygen
can decrease erythropoietin production
Laboratory evaluation of acute
and lead to more severe anemia. Pulmo-
chest syndrome
nary infections should be treated aggres-
* CBC with differential and reticulocyte
sively, and these children watched closely.
count.
If there is no improvement, and/or wors-
* Type and hold for possible blood trans-
ening anemia, a red blood cell transfusion
fusion (phenotypically matched, sickledex
(straight or exchange, dependent upon
negative).
the severity of the hypoxia, anemia, and
* Chest radiograph, as often as clinically
clinical status of the patient) may help to
indicated to monitor progression of
correct the anemia, decrease the percent
disease.
Hgb S, and improve oxygen-carrying
* Continuous pulse oximetry and baseline
capacity to aid in reversing the pulmo-
arterial blood gas (on room air if possible).
nary sickling and improve the clinical
* Blood culture for fever.
course. Transfusions are more effective
when administered early in the course
of ACS, rather than as a life-saving mea-
Management
sure in a critically ill child.
All patients with evidence of acute pulmo-
There is a distinct difference between
nary pathology should be admitted to the
the etiology of acute chest syndrome in
hospital. If fever is present or if an infec-
children and that of adolescents and
tious process is suspected, antibiotic ther-
adults. In children, the incidence of acute
apy should be started immediately. See
chest syndrome is seasonal, lower in the
Fever and Infection section, and Figures 3.1
summer months with increasing rates in
and 3.2.
the winter when viral infections are prev-
* Oxygen
alent. In adults and adolescents, ACS is
Hypoxemic patients
(PaO2
70
to
frequently a complication of an episode of
80 mmHg, O2 saturation
92%): 2 L/min
vaso-occlusion
(without fever) due to
per nasal cannula.
pulmonary fat embolism. This event will
Reevaluate arterial blood gas on oxygen.
progress to include chest pain, fever, and a
* Antibiotics
pulmonary infiltrate (usually basilar with
Initiate broad spectrum antibiotic: cef-
pleural effusion). Adults and adolescents
triaxone
50 mg/kg/d IV every
24
hours
more frequently need transfusions and
after blood cultures obtained
(maximum
less frequently have a viral or bacterial
2 g/dose).
infection associated with their episode
Due to the frequency of atypical organ-
of acute chest syndrome. Individuals with
isms (e.g., chlamydia and mycoplasma), a
Sickle Cell Disease
27
macrolide or quinolone antibiotic should
Priapism
be included: azithromycin 10 mg/kg on day
1, followed by
5 mg/kg on days 2 to 5
Forty percent of men who are homozygous
(maximum 500 mg on day 1, 250 mg on
for Hgb S report having priapism in their
days 2 to 5).
adolescence and early adulthood. Priapism
* Analgesics
has a second peak at 21 to 29 years. Priapism
As indicated for vaso-occlusive pain man-
is defined as a painful erection that lasts for
agement, administration must be moni-
more than 30 minutes. It frequently results
tored to provide the maximum pain control
in interference with the urinary stream.
and prevent hypoventilation or atelectasis
Priapism can be precipitated by prolonged
from splinting or narcotization.
intercourse or masturbation, frequently
* Hydration
occurs at night, and can be differentiated
PO and IV hydration (D5W 1/2 NS) at a
from a nocturnal erection by its duration
maximum of 1.25
maintenance. Caution
and pain. Children will occasionally com-
should be used in patients with potential
plain of dysuria as the first complaint of
acute chest syndrome to avoid fluid over-
priapism. Prognosis is poorer in adolescents
load. Monitor Is & Os, daily weight.
and adults who have recurrent prolonged
* Other supportive measures
priapism, and if aggressive treatment has
Continuous pulse oximetry.
not been successful, impotence may result.
Incentive spirometry, 10 times an hour,
Children who have priapism generally have
every hour while awake
(prevention of
a better prognosis than adolescents and
hypoventilation and atelectasis).
adults and usually do not have prolonged
Albuterol aerosols q4
hours
(airway
episodes requiring aggressive therapy. Pria-
hyperreactivity common in ACS).
pism can occur with vaso-occlusive episodes
* Physical therapy
as well as with fever and sepsis.
Warm packs.
Priapism can last for hours, days, or
Ambulation as tolerated, sitting up in bed.
weeks with moderate-to-severe pain, or it
* Transfusion
can occur as a pattern of painful erections
Straight transfusion.
that recur over a period of days to several
Exchange transfusion (for patients with
weeks (stuttering priapism). Chronic non-
hematocrit > 30% or moderate-to-severe
painful priapism can also occur.
ACS to more efficiently reduce Hgb S%).
Since priapism can lead to impotence,
early medical management is indicated. A
Transfusion decreases the proportion of
urologist familiar with priapism in sickle cell
sickling red cells and increases blood oxygen
disease should be consulted. Treatment
affinity. The main indication is worsening
includes hydration, pain management, intra-
respiratory function, as documented by
cavernous injection of alpha-adrenergic
hypoxemia (paO2 <70 mmHg on room air),
agents, exchange transfusion, and shunting
worsening chest pain, evolving clinical
surgery. Oral alpha-adrenergics (e.g., pseu-
examination, or worsening infiltrates on
doephedrine hydrochloride 30 mg BID) have
chest radiography. For patients with chronic
been used successfully to treat stuttering
hypoxemia, a drop of >10% from baseline
priapism. Chronic transfusion therapy has
is a reasonable level at which to transfuse.
been used in patients with recurrent priapism
Delays in instituting transfusion therapy,
(maintenance of Hgb S below 30% to 40%),
particularly in rapidly deteriorating patients,
although no controlled trials have been per-
should be avoided.
formed proving the efficacy of this therapy.
28
Chapter 3
Stroke (cerebrovascular
during an exchange transfusion realizing
accident)
that, relative to the normal population,
children with sickle cell disease are hypo-
Stroke is a common event in children
tensive. The diagnosis of stroke in children
homozygous for Hgb S. Between 10% and
should initially be made based on clinical
15% of children with sickle cell disease
findings and treated emergently. Children
suffer from overt strokes. In children,
should be monitored until they are stable
strokes are more frequently the result of
and the hemoglobin electrophoresis is
cerebral vascular stenosis and infarction.
documented. Those who have had a stroke
The mean age of occurrence of clinically
require chronic transfusion therapy for an
evident stroke is 7 to 8 years with the highest
undetermined length of time.
risk occurring between 2 and 9 years. Hem-
Children with sickle cell disease have
orrhage and infarct may occur together.
more frequent and more severe headaches
Strokes in children who have sickle cell
than other children; these may be manifes-
disease involve stenosis and occlusion of the
tations of cerebral hypoxia and vasodilation.
major anterior arteries of the brain, includ-
Children who have severe headaches accom-
ing the carotids. The presenting symptoms
panied by vomiting need extensive evalua-
of stroke can be dramatic and acute, such as
tion with imaging as well as neurologic and
coma, seizure, hemiparesis, hemianesthesia,
neuropsychological testing.
visual field deficits, aphasia, or cranial nerve
Transcranial Doppler (TCD) has been
palsies. Subtle limb weakness (without pain)
shown to predict increased risk of stroke in
is often mistaken for an acute vaso-occlusive
children who have increased flow velocity in
episode but can be due to stroke. By defi-
major cerebral arteries. Unsuspected cere-
nition, the symptoms must persist for at
bral vascular damage was detected by CT
least 24 hours to be classified as a stroke. The
and MRI in 25% to 30% of children in the
presentation of severe headache and vomit-
Cooperative Study of Sickle Cell Disease. No
ing with no other neurological findings can
clinical factors, with the exception of tran-
be symptoms of an intracranial hemorrhage.
scranial Doppler, predicted the occurrence
Initially, a CT scan without contrast will
of stroke. In this study, there was a corre-
be normal for up to 6 hours after a stroke
lation between first stroke and transient
but will rule out intracerebral hemorrhage,
ischemic attack, acute chest syndrome, ele-
abscess, tumor, or other pathology that
vated systolic blood pressure, and severe
would explain the neurological symptoms.
anemia. All children who can cooperate
Magnetic resonance imaging
(MRI) and
with transcranial Doppler (usually by 2 years
magnetic resonance angiography (MRA) are
of age) should have an annual evaluation
much more sensitive methods to determine
if the study is available locally. Normal
intracranial infarct but can remain normal
middle cerebral artery velocity in children
for 2 to 4 hours after an event. All cata-
with sickle cell disease is approximately
strophic neurological events should first be
120 cm/s. Children who have velocities of
evaluated with CT since it is generally more
170 to 199 cm/s are considered at high risk
available and will rule out acute pathology.
and those with velocities
200 cm/s are at
With clinical concern prior to definitive
highest risk. The STOP trial
(Stroke
diagnosis by MRI/MRA imaging, children
Prevention Trial in Sickle Cell Anemia)
are exchange transfused to achieve a con-
showed that transfusion greatly reduces the
centration of Hgb S of less than 30%. Blood
risk of first stroke in children who have
pressure should be continually monitored
abnormally high TCD velocity (
200 cm/s).
Sickle Cell Disease
29
Prophylactic chronic transfusion should be
6 months to 3 years of age when the spleen
strongly considered if there are two studies
becomes fibrotic due to multiple infarc-
at least 2 weeks apart with velocities
200
tions. These crises can occur in older chil-
cm/s. Compliance issues and risks of
dren who have Hgb SC or Sbþ thalassemia
chronic transfusions must also be consid-
but are usually not as severe; still, fatal
ered when deciding on this therapy.
events have been recorded for these chil-
Transient ischemic attacks are defined
dren. The incidence is between 10% and
as focal neurological deficits with a vascular
30% and the recurrence rate is 50%. Mild
distribution persisting for less than
24
events can indicate the possibility of life-
hours, although they typically last less than
threatening sequestration events. Chronic
an hour. Patients with transient ischemic
sequestration can also be a sequelae with
attacks are treated in a similar manner to
chronic anemia and thrombocytopenia.
those who have an infarct. The diagnosis is
The clinical presentation is usually rapid
made in retrospect when the follow-up MRI
in onset; the child will typically have sudden
is negative for a persistent lesion that would
weakness, pallor of the lips and mucous
explain the neurological symptoms.
membranes, breathlessness, rapid pulse,
Intracerebral hemorrhage or subarach-
faintness, and abdominal fullness. Evalua-
noid hemorrhage may present without
tion of the CBC will frequently show a
focal neurologic symptoms. Exchange
precipitous drop from the baseline
transfusion should be carried out immedi-
hemoglobin.
ately. Arteriography is used to identify the
The treatment for splenic sequestration
arterial bleed and the patient may need
is transfusion to restore circulating blood
emergent surgical intervention. Mortality
volume, usually with phenotypically mat-
is very high during the acute event (
50%).
ched blood (unless there is a life-threatening
situation). As the shock is reversed and
the transfused blood decreases the percentage
Acute anemia
of Hgb S, the splenomegaly regresses and
much of the blood is remobilized with a
There are two common causes of acute life-
rapid rise in the child’s hemoglobin.
threatening anemia in sickle cell disease:
Splenectomy should be strongly consid-
splenic sequestration and aplastic crisis.
ered in all children who have a splenic
sequestration crisis. Transfusions can
Splenic sequestration
prevent recurrence until surgery can be
Infants and young children who have Hgb
arranged. Some children who have Hgb
SS and Sb0 thalassemia and older children
SS and older children who have other S
(over 10 years) who have Hgb SC or Sbþ
hemoglobinopathies can have massive
thalassemia syndromes can have intrasple-
splenomegaly with hypersplenism after an
nic pooling of large amounts of blood and
episode of splenic sequestration. If hypers-
platelets. This can lead to anemia, throm-
plenism (with resultant anemia, neutrope-
bocytopenia, hypovolemia, cardiovascular
nia, or thrombocytopenia) is persistent
collapse, and sudden death within hours
and severe, splenectomy is indicated.
of the onset of sequestration. The syndrome
Prior to splenectomy, younger children
has been reported in infants as young as 2
should have an evaluation of splenic func-
months of age. Classically, however, it
tion with a pit count and a spleen scan. In
occurs in children with Hgb SS after the
normally eusplenic persons, fewer than 1%
disappearance of Hgb F from approximately
of the circulating red cells are pitted; values
30
Chapter 3
of
2% to 12% may represent decreased
last for weeks. Avascular necrosis (osteone-
splenic function. If they still have a func-
crosis) causes pain that can sometimes be
tional spleen, usually under 2 years of age, a
confused with a bone infarct. Symptoms of
partial splenectomy can be performed. Chil-
avascular necrosis, such as limping, can
dren should have appropriate immuniza-
sometimes be confused with stroke. Limp-
tions, including pneumococcal and menin-
ing with weakness but without pain is
gococcal vaccinations, prior to splenectomy.
stroke not avascular necrosis.
Penicillin prophylaxis is continued indefi-
Avascular necrosis is common in all age
nitely in all children who have been
groups but is more frequently diagnosed in
splenectomized.
the adolescent and usually involves the
humeral and femoral heads. It is more
Aplastic crisis
common in individuals who have Hgb
Severe anemia can develop over several days
Sb0 thalassemia and Hgb SS with alpha-
due to shortened red cell survival without
thalassemia. It is also seen, though with a
compensatory reticulocytosis. Reticulocyto-
lower frequency, in those who have Hgb
penia can last 7 to 10 days. The primary
Sbþ thalassemia. Individuals who have Hgb
cause of transient red cell aplasia is parvo-
SS are more frequently affected than those
virus B19, though it can follow other viral
with Hgb SC.
infections as well. Parvovirus B19, the most
Avascular necrosis is a result of an infarct
common cause of aplasia in children with
in the cancellous trabeculae in the head of
hemoglobinopathies, can also cause neutro-
either the femur or the humerus. The pro-
penia. While many patients recover spon-
cess of necrosis and repair can be progres-
taneously, the anemia can be profound. Red
sive, leading to collapse of the head or arrest
cell transfusion is indicated for those who
with varying degrees of disability and scle-
become symptomatic from anemia or if the
rosis. Below is a method of describing the
hemoglobin falls 2 g/dL or more from base-
progression of avascular necrosis:
line. Symptoms occurring with aplastic cri-
Stage 0: no evidence of disease
sis include nausea and vomiting, myalgias,
Stage 1: X-ray normal, bone scan abnormal,
and arthralgias. Splenic and hepatic seques-
MRI abnormal
tration have also been reported with aplastic
Stage 2: X-ray: sclerosis and cystic changes
crises. If the ability to follow-up is of con-
without collapse
cern, the child should be hospitalized for
Stage
3: Subchondral collapse
(crescent
observation. The patient should be isolated
sign) without collapse
from other patients with chronic hemolytic
Stage 4: Collapse and flattening of the fem-
anemias (sickle cell disease or thalassemia)
oral head without acetabular involve-
or red cell aplasia and should not have
ment, normal joint space
pregnant caregivers. If the child is mildly
Stage 5: Joint narrowing and/or acetabular
anemic and asymptomatic, outpatient mon-
involvement
itoring is reasonable.
Stage 6: Increased joint narrowing and/or
acetabular involvement
Avascular necrosis
Treatment for avascular necrosis of the
femur includes bed rest with crutch walking
Bone pain is common in sickle cell disease.
for 6 weeks, nonsteroidal anti-inflammatory
Marrow infarcts can cause pain that can
drugs (NSAIDs), and core decompression
Sickle Cell Disease
31
surgery for stages 1 or 2. Extensive physical
occurs most commonly in individuals who
therapy has been found to be beneficial but
have Hgb SC.
is often difficult to perform. Decompression
Treatment of sickle cell retinopathy
surgery is relatively simple; a core is
requires recognition. The damaged periph-
removed from the head of the femur.
eral vessels are not generally appreciated by
The coring begins approximately
2 cm
direct ophthalmic examination and require
below the trochanteric ridge extending
the use of indirect binocular stereoscopic
through the neck and into the head of the
ophthalmoscopy. Sea fanning, vitreous
femur. Acutely this procedure provides
hemorrhage, and retinal detachment can
relief of pain and is thought to arrest the
be observed by direct ophthalmoscopy.
progression of avascular necrosis; however,
Annual ophthalmic evaluations should
this has not been confirmed in a prospective
begin at age 10 for all children with sickle
trial. Higher stage avascular necrosis
cell disease. Treatment of early neovascular-
requires joint replacement.
ization requires laser photocoagulation.
Surgical approaches are required for
advanced lesions.
Retinopathy/Hyphema
Hyphema is a medical emergency in
sickle cell disease. It requires immediate
The eye is particularly sensitive to hypoxia.
ophthalmic evaluation and transfusion
Vaso-occlusion of retinal vessels and hyp-
(exchange or straight) to reduce sickling
oxia of the retina cause permanent retinal
and improve oxygenation. If a conservative
damage. Blood in the anterior chamber of
approach is not successful, anterior cham-
the eye (hyphema) becomes rapidly deoxy-
ber paracentesis is performed to relieve
genated and permanently sickled, obstruct-
intraocular pressure and remove the
ing the outflow from the aqueous humor.
hyphema.
The accumulation of aqueous humor in the
anterior chamber increases intraocular pres-
sure leading to decreased blood flow to the
Hyperbilirubinemia /Gallstones
retina until the perfusion pressure of the
globe is reached. This leads to sudden vas-
Bilirubin gallstones can eventually be
cular stasis and blindness. The events occur-
detected in most patients with chronic
ring in hyphema can also occur in children
hemolytic anemia. In sickle cell disease,
who have sickle cell trait.
gallstones occur in children as young as 3
Retinal vascular occlusion initially occurs
to 4 years of age, and are eventually found in
in peripheral retinal vessels without signifi-
approximately 70% of patients. It may be
cant sequelae. It eventually leads to neovas-
difficult to differentiate between gallbladder
cularization from the retina into the vitreous
disease and abdominal vaso-occlusive crisis
(sea fans). These abnormal vessels are fragile
in patients with recurrent abdominal pain.
and can bleed into the vitreous causing
Cholecystectomy may be necessary for
“floaters” or blindness if the hemorrhage is
patients with fat intolerance, presence of
sufficient. If bleeds do not obscure vision and
gallstones, and recurrent abdominal pain.
are unnoticed, they can cause collapse of the
Hepatomegaly and liver dysfunction
vitreous, traction on the retina, and eventu-
may be caused by a combination of intra-
ally cause retinal detachment. Retinal disease
hepatic trapping of sickled cells,
of the eye is common in sickle cell disease. It
transfusion-acquired
infection,
or
32
Chapter 3
transfusional hemosiderosis. The combina-
study obtained prior to any transfusion.
tion of hemolysis, liver dysfunction, and
Sickle cell patients should receive pheno-
renal tubular defects can result in very high
typically matched blood to decrease the
bilirubin levels. Benign cholestasis of sickle
risk of alloimmunization. In addition, all
cell disease results in severe asymptomatic
blood should be sickledex negative and
hyperbilirubinemia without fever, pain, leu-
leukodepleted. The following transfusion
kocytosis, or hepatic failure.
guidelines are recommended. See also
Chapter 5.
a. Patients with Hgb SS or Sb0 disease
who are undergoing any procedure other
Perioperative management of
than myringotomy tube placement
sickle cell patients
should have a simple transfusion to
achieve a hemoglobin of 10 to 12 g/dL.
Following are guidelines for the manage-
Do not exceed 12 g/dL due to the risk of
ment of sickle cell patients who have sur-
stasis/sludging.
gical procedures or require anesthesia for
b. Patients with Hgb SC disease or Sbþ
other purposes. General anesthesia results
who have baseline hemoglobin below
in atelectasis and hypoxia, which is poorly
10 g/dL may need transfusions to achieve
tolerated by the patient with sickle cell
a hemoglobin of 10 to 12 g/dL. However,
disease; therefore, special precautions must
do not transfuse these patients prior to
be taken perioperatively to decrease mor-
consulting the hematologist. Many fac-
bidity and mortality associated with even
tors may affect this decision including a
minor surgical procedures. Other risk fac-
history of significant complications of
tors include the presence of chronic organ
their sickle cell disease (i.e., pneumonia,
damage in some patients, the effects of asple-
vaso-occlusive episodes, priapism, and
nia, and the propensity for sickling and
aseptic necrosis).
obstruction of microvasculature with even
c. Consult the hematologist if there is
mild hypoxia. The guidelines suggested
confusion regarding individual patients.
below are extrapolated from a multicenter
7. Patients with central venous catheters or
randomized trial of transfusion (exchange vs
those undergoing dental procedures should
straight) in the perioperative period.
receive antibiotic prophylaxis prior to
surgery.
Preoperative care
1. Admission to the hospital the afternoon
prior to scheduled surgery.
Postoperative care
2. Hydration at 1.25
maintenance (IV þ
1. Adequate pain management to prevent
PO) the evening and night prior to surgery.
splinting and atelectasis, with care to pre-
3. Pulse oximetry on room air
(spot
vent narcosis.
checks).
2. Pulse oximetry for at least the first 12 to
4. Incentive spirometry at least 10 times per
24 hours postoperatively.
hour while awake.
3. Incentive spirometry at least 10 times per
5. Lab work: CBC with differential and
hour while awake. Be aggressive!
reticulocyte count, urinalysis, type and
4. Ambulation as early as possible, taking
cross, chemistry panel.
into account the specific surgical procedure.
6. Transfusion: All sickle cell patients
5. Lab work: CBC with reticulocyte count
should have a red cell phenotype
daily.
Sickle Cell Disease
33
6. Follow-up in clinic approximately
cell pheresis is recommended. Studies sug-
1 week post-hospital discharge or sooner
gest early transfusion may prevent progres-
if there are ongoing problems.
sion of acute pulmonary disease.
The efficacy of transfusion in the man-
Transfusion therapy
agement of acute stroke has not been well
studied, though anecdotal reports indicate
Red blood cell transfusion increases oxygen-
that early exchange transfusion may limit
carrying capacity and improves microvas-
neuronal damage by improving local oxy-
cular perfusion by decreasing the propor-
genation and perfusion. Chronic transfu-
tion of sickle red cells (Hgb S%). Transfu-
sion therapy reduces the rate of recurrence
sions are given to patients with sickle cell
and is indicated in all patients after a first
disease to stabilize or reverse an acute med-
stroke.
ical complication, or as part of chronic
therapy in certain situations to prevent
Perioperative transfusion
future complications.
Patients with sickle cell anemia undergoing
major surgery should be prepared in
Indications
advance by transfusion to a hemoglobin
Acute illnesses
of approximately 10 g/dL and a decrease
Splenic sequestration
in Hgb S% to approximately 60%. While
Transient red cell aplasia
standard practice guidelines have not
Hyperhemolysis (infection, ACS)
been developed for Hgb SS patients under-
going minor procedures or those with Hgb
Patients should be transfused if there is
SC, it is generally acceptable to not trans-
evidence of cardiovascular compromise
fuse these patients, assuming they are med-
(heart failure, dyspnea, hypertension, or
ically stable.
marked fatigue). Generally, transfusion is
indicated if the hemoglobin falls below 5 g/
Chronic transfusion therapy
dL or drops greater than 2 g/dL from the
Chronic transfusions are indicated in
steady state.
several conditions in which the potential
medical complications warrant the risks
Sudden severe illness
of alloimmunization, infection, and trans-
Acute chest syndrome
fusional iron overload. Transfusions are
Stroke
given every 3 to 4 weeks, with a goal to
Sepsis
maintain the Hgb S% at 30% to 50%. While
Acute multiorgan failure
straight transfusions are acceptable, red cell
pheresis or exchange transfusions may be
These life-threatening illnesses are often
preferred to decrease the rate of iron
accompanied by a falling hemoglobin.
overload.
Transfusion therapy has become standard
medical practice in the management of
Indications
these illnesses. When ACS is associated with
1. Primary stroke prevention
hypoxia and a falling hemoglobin, transfu-
2. Pulmonary hypertension/chronic lung
sion is indicated. Many patients can be
disease
treated with straight transfusion, though
3. Frequent ACS
in severe cases, exchange transfusion or red
4. Chronic debilitating pain
34
Chapter 3
Other indications for transfusions:
prophylaxis in more patients. Some would
Transfusions are sometimes suggested for
argue that all patients with hemoglobin SS
a number of conditions in which efficacy is
and Sb0 thalassemia should be on HU ther-
unproven,butmaybeconsideredundersevere
apy; this may become the standard of care in
circumstances. Such circumstances include:
the near future.
Acute priapism
HU therapy begins at 20 mg/kg/d and
Pregnancy (frequent complications)
should be titrated based on beneficial incre-
“Silent” cerebral infarcts
ment in Hgb F% and resultant increase in
Leg ulcers
hemoglobin and MCV. Potential toxicities
with HU therapy that should be monitored
closely include neutropenia, leukopenia, and
Hydroxyurea therapy
anemia. Elevation in ALT and thrombocyto-
penia can also occur, though less commonly.
Hydroxyurea (HU) was originally utilized as
an antineoplastic agent, but due to its effects
on increasing fetal hemoglobin (Hgb F), it
Hematopoietic stem cell trans-
has gained favor in the treatment of patients
plantion in sickle cell disease
with sickle cell disease. Hydroxyurea’s
mechanism of action is not clearly under-
Hematopoietic stem cell transplantion is
stood; however, it is thought that inhibition
curative for patients with sickle cell disease
of ribonucleotide reductase leads to down-
and should be considered in young patients
stream cytotoxicity of erythroid progenitors
with hemoglobin SS and Sb0 thalassemia
leading to upregulation of fetal hemoglobin
and a matched sibling donor.
production. In addition, hydroxyurea’s
Patients with severe manifestations of
cytotoxic effects lead to leukopenia and
sickle cell disease including multiple pain
neutropenia, beneficial in sickle cell disease
episodes, recurrent ACS, stroke, and end-
secondary to the viscous properties of these
organ damage should be considered for
cells during vaso-occlusion.
unrelated bone marrow or umbilical cord
Long-term adult studies have shown no
blood transplantation when a matched sib-
significant complications of HU usage and it
ling donor is not available.
is being used more commonly in pediatric
patients with sickle cell disease. Current
Case study for review
recommendations for HU usage include the
following:
You are a primary care physician taking care
* Frequent episodes of VOE including
of a 2-year-old with known sickle cell disease
dactylitis
(Hgb SS). You are counseling the family on
* Acute chest syndrome
* Chronic hypoxia
particular risks for the patient at this age.
* Low hemoglobin and Hgb F
1. What are some of the main areas of
* Signs of increased hemolysis (increased
discussion?
LDH and total bilirubin)
* Abnormal TCD or history of stroke with
In evaluating the patient as a whole, one
refusal for chronic transfusion therapy
must first discuss growth and development
and the risks of low hemoglobin on brain
As experience grows with HU therapy,
development. Assuming the patient has had
practitioners are recommending HU as
previous laboratory studies, past hemoglobin
Sickle Cell Disease
35
values can help objectively guide this discus-
to walk, decreased appetite, as well as local-
sion as phenotypic disease is related to level
ized swelling and/or tenderness. A pain plan
of anemia and concomitant reticulocytosis,
should be established with the family which
amount of leukocytosis, as well as evidence
should include the availability of oral med-
of hemolysis (total bilirubin and LDH levels).
ications at home (e.g., acetaminophen, ibu-
In this discussion, newer treatment modali-
profen, Tylenol with codeine, and/or Lortab
ties including hydroxyurea therapy and the
elixir) as well as adjuvant strategies such as
potential for hematopoietic stem cell trans-
warm bath and heating pads.
plantation should be discussed.
Finally, the practitioner should discuss
Fortunately, this child has had a relatively
other potential acute complications such as
benign course to date.
splenic sequestration. If the child has a pal-
pable spleen, the family should be advised on
2. What are some of the potential acute
the current size and to monitor for changes
risks that should again be discussed with
in size if the patient appears to be pale or
the family?
weak. In addition, if the child does appear
The major risks to the child include severe
to have these symptoms without a change
infection, pain including dactylitis, and
in the size of the spleen or does not have a
splenic sequestration. These risks should all
palpable spleen, the family should be advised
be discussed in detail with the family.
that these symptoms should again lead to
In terms of severe infection, the family
prompt evaluation as they could be signs of
should be aware that they should monitor
splenic sequestration or an aplastic crisis.
the child closely if the child appears ill and
check the temperature before administering
Suggested Reading
antipyretics, even if given for pain alone. In
addition, they should be aware that if the
Adams RJ, McKie VC, Hsu L, et al. Prevention of
child appears ill, they should immediately call
a first stroke by transfusion in children with
the on-call physician and will likely require
sickle cell anemia and abnormal results on
immediate evaluation including blood work,
transcranial Doppler ultrasonography. N Engl
chest radiography, parenteral antibiotics, and
J Med 339:5-11, 1998.
Ballas SK. Current issues in sickle cell pain and its
hospital admission and observation. You
management. Hematology Am Soc Hematol
should again reinforce the need for the child
Educ Program 97-105, 2007.
to be on penicillin prophylaxis. Finally, one
Claster S, Vichinsky EP. Managing sickle cell
should review the child’s vaccinations to
disease. BMJ 327:1151-1155, 2003.
ensure they are up to date, especially for
Rees DC, Olujohungbe AD, Parker NE, et al.
encapsulated bacteria including S. pneumo-
Guidelines for the management of the
niae, H. influenzae, and N. meningitidis.
acute painful crisis in sickle cell disease. Br
J Haematol 120:744-752, 2003.
Regarding episodes of pain, although it is
Vichinsky EP, Neumayr LD, Earles AN, et al.
reassuring the child has had a relatively unre-
Causes and outcomes of the acute chest syn-
markable course to date, VOE can start at any
drome in sickle cell disease. National Acute
age. At 2 years of age, the child is still at risk
Chest Syndrome Group. N Engl J Med
for developing episodes of dactylitis. The
342:1855-1865, 2000.
family should be advised on the signs and
Ware RE. How I use hydroxyurea to treat young
symptoms to monitor for pain in a young
patients with sickle cell anemia. Blood
115:5300-5311, 2010.
child which may present as irritability, refusal
Thalassemia
4
The thalassemias are a diverse group of
An excess of alpha-globin chains (beta-
genetic diseases characterized by absent or
thalassemia) leads to the formation of
decreased production of normal hemoglo-
alpha-globin tetramers that accumulate in
bin, resulting in a microcytic anemia. The
the erythroblast. These aggregates are very
alpha-thalassemias are concentrated in
insoluble and precipitation of these aggre-
Southeast Asia, Malaysia, and southern
gates interferes with erythropoiesis, cell
China. The beta-thalassemias are seen pri-
maturation, and cell membrane function
marily in the Mediterranean, Africa, and in
leading to ineffective erythropoiesis and
Southeast Asia. Due to global migration
anemia.
patterns, there is an increase in the incidence
An excess of beta-globin chains (alpha-
of thalassemia in North America, primarily
thalassemia) leads to tetramers of beta-
because of immigration from Southeast
globin, Hgb H. These tetramers are more
Asia. Like with sickle cell anemia, develop-
stable and soluble, but as the red cell ages in
ment of thalassemia is directly related to
the circulation and, under conditions of
evolutionary pressure secondary to malaria.
oxidant stress, Hgb H precipitates and inter-
Normally,
95% of adult hemoglobin
feres with cell membrane function leading
found on electrophoresis is Hgb A (a2b2).
to an increase in hemolysis.
Two minor hemoglobins occur: 2% to 3.5%
is Hgb A2 (a2d2) and
2% is Hgb F (a2g2).
A mutation affecting globin chain produc-
Alpha-thalassemia
tion or deletion of one of the globin chains
leads to a decreased production of that
The alpha-thalassemias are caused by a
chain and an abnormal globin ratio. The
decrease in the production of alpha-globin
globin that is produced in normal amounts
due to a deletion or mutation of one or more
is in excess and forms aggregates or inclu-
of the four alpha-globin genes located on
sions within the red cells. These aggregates
chromosome
16. Alpha-gene mapping
become oxidized and damage the cell mem-
can be obtained to determine the specific
brane leading to ineffective erythropoiesis,
mutation. The alpha-thalassemias are cate-
hemolysis, or both. The quantity and prop-
gorized as: silent carrier, alpha-thalassemia
erties of these globin chain aggregates deter-
trait, hemoglobin H disease, hemoglobin
mine the phenotypic characteristics of the
H-constant spring, and alpha-thalassemia
thalassemia.
major. Frequently, the diagnosis of
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Thalassemia
37
alpha-thalassemia trait in a parent is discov-
Hemoglobin H (
/ a) should be con-
ered after the birth of an affected child.
sidered in the case of a neonate in whom all
Silent carrier status is characterized by
of the red blood cells are very hypochromic.
three functional genes for a-globin:
Neonates who have Hgb H will also have a
( a/aa). Outside the newborn period, it
high percentage of Hgb Bart’s on their
is not possible to make this diagnosis by
newborn screen (
20%). In children, this
conventional methods. There is overlap
hemoglobinopathy is characterized by mod-
between the red blood cell indices of these
erate anemia with a hemoglobin in the 8 to
individuals and normals, although the MCV
10 g/dL range, hypochromia, microcytosis,
may be slightly lower. This state is deduced
red cell fragmentation, and a fast migrating
when a “normal” individual has a child with
hemoglobin (Hgb H) on electrophoresis.
Hgb H disease or with microcytic anemia
Hemoglobin H does not function as a
consistent with alpha-thalassemia trait. An
normal hemoglobin, and has a high oxygen
unusual case of the silent carrier state is the
affinity, so the measured hemoglobin in these
individual who carries the hemoglobin con-
children is misleading. Individuals who have
stant spring mutation (acsa/aa or aacs/aa).
Hgb H generally have a persistent stable
This is an elongated a-globin due to a
state of anemia that may be accentuated by
termination codon mutation. Individuals
increased hemolysis during viral infections
who have this mutation have normal red
and by exposure to oxidant medications,
blood cell indices, but can have children
chemicals, and foods such as sulfa-drugs,
who have Hgb H-constant spring if the
benzene, and Fava beans, similar to indivi-
other parent has alpha-thalassemia trait
duals who have G6PD deficiency. As the red
(aa/
). Generally, these children are more
cells mature, they lose their ability to with-
affected clinically than other children who
stand oxidant stress and Hgb H precipitates,
have Hgb H.
leading to hemolysis. Therapy for individuals
Individuals who have alpha-thalassemia
who have Hgb H disease includes folate,
trait ( a/-a) or (aa/
) are identified by
avoidance of oxidant drugs and foods,
microcytosis, erythrocytosis, hypochromia,
genetic counseling, education, and frequent
and mild anemia. The diagnosis is made
medical care. Uncommon occurrences in a
by genetic studies, ruling out both iron-
child with Hgb H would be severe anemia,
deficiency anemia and beta-thalassemia
cholelithiasis, skin ulceration, and spleno-
trait. In the neonatal period, when hemo-
megaly requiring splenectomy. Unlike indi-
globin Bart’s
(g4) is present
(
5% on
viduals who have beta-thalassemia, hemosi-
newborn screen), the diagnosis can be sus-
derosis is rare in Hgb H disease.
pected. In children, there are no markers
Children with hemoglobin H-constant
such as elevated Hgb A2 and Hgb F (as seen
spring (
/acsa or acs-/a-) have a more
in beta-thalassemia trait) to make the diag-
severe course than children who have Hgb
nosis. The diagnosis is one of exclusion and
H. They have a more severe anemia, with a
is often mistaken for iron-deficiency anemia
steady-state hemoglobin ranging between 7
secondary to the microcytosis. Clues for the
and
8 g/dL. They more frequently have
diagnosis include a normal RDW and an
splenomegaly and severe anemia with febrile
increase in red blood cells for the level of
illnesses and viral infections, often requiring
hemoglobin. During pregnancy, the micro-
transfusion. If anemia is chronically severe
cytic anemia can be mistaken for anemia of
and the child has splenomegaly, a splenec-
pregnancy, and may be a clue for a family
tomy may be performed. If splenectomy is
history of alpha-thalassemia trait.
anticipated, a complication can be severe
38
Chapter 4
postsplenectomy thrombocytosis with
unique mutations. Individuals with beta-
hypercoagulability leading to thrombosis
thalassemia major are usually homozygous
of the splenic vein or hepatic veins. This
for one of the common mutations (as well as
complication has also been reported as
having one of the geographically unique
recurrent pulmonary emboli and clotting
mutations) that lead to the absence of
diathesis. Children who are scheduled to
beta-chain production.
have surgery are treated with low-molecular-
The beta-thalassemia syndromes are
weight heparin, followed by low-dose aspi-
much more diverse than the alpha-thalassemia
rin, continued indefinitely.
syndromes due to the diversity of the
The most severe form of alpha-thalasse-
mutations that produce the defects in the
mia is alpha-thalassemia major
(
/
).
beta-globin gene. Unlike the deletions that
This diagnosis is frequently made in the last
constitute most of the alpha-thalassemia
months of pregnancy when fetal ultrasound
syndromes, beta-thalassemias are caused
indicates a hydropic fetus. The mother fre-
by mutations on chromosome 11 that affect
quently exhibits toxemia and can develop
all aspects of beta-globin production: tran-
severe postpartum hemorrhage. These
scription, translation, and the stability of the
infants are usually stillborn. There can be
beta-globin product. Most hematologists
other congenital anomalies, though none
feel there are three general categories of
are pathognomonic for alpha-thalassemia
beta-thalassemia: beta-thalassemia trait,
major. Because of in utero hypoxia, the
beta-thalassemia intermedia, and beta-
hemoglobins found in these infants are Hgb
thalassemia major. However, with the lack
Portland (z2g2), Hgb H (b4), and Hgb Bart’s
of genotypic differentiation, there remains
(g4), and no Hgb A or A2. These babies can
phenotypic overlap between these three
have other complications associated with
general categories.
hydrops such as heart failure and pulmo-
Splice site mutations also occur and are
nary edema.
of clinical consequence when combined
If the diagnosis is made early, intrauter-
with a thalassemia mutation. Three splice
ine transfusions can be performed. There
site mutations occur in exon 1 of the beta-
are reports of survival with chronic trans-
globin gene. These mutations result in three
fusion in these infants, now more recently
different abnormal hemoglobins: Malay, E,
replaced by curative hematopoietic stem cell
and Knossos. Hemoglobin E is a very com-
transplantation. Undoubtedly, more of
mon abnormal hemoglobin in the Southeast
these infants could be saved if the diagnosis
Asian population and when paired with a
was anticipated by prenatal diagnosis and
b0-thalassemia, mutation can produce
the treatment provided.
severe transfusion-dependent thalassemia.
Hemoglobin E is described in the section
on newborn screening.
Beta-thalassemia
Individuals who have beta-thalassemia
trait have microcytosis and hypochromia;
Beta-thalassemia is caused by mutations in
there may be targeting and elliptocytosis,
the beta-globin gene. Although there have
though some individuals have an almost
been hundreds of mutations identified
normal smear. These hematologic features
within the beta-globin gene locus, about
can be accentuated in women with trait who
20 different alleles make up about 80% of
are pregnant and individuals who are folate
the mutations found worldwide. Within
or iron deficient. If iron deficiency is con-
each geographic population, there are
current with beta-thalassemia trait, there
Thalassemia
39
may be a normal Hgb A2. Iron deficiency
Problems can still arise if both alpha- and
causes decreased hemoglobin production
beta-thalassemia coexist since the changes in
and folate or vitamin B12 deficiency can
Hgb A2 and F will not be apparent as noted
lead to megaloblastic anemia with increased
above. Family studies and DNA analysis can
Hgb A2. Both of these deficiencies need to be
be used to make a definitive diagnosis.
treated prior to evaluation for thalassemia
Children who are diagnosed with thalas-
trait. In iron-, B12-, and folate-replete indi-
semia intermedia have a homozygous or
viduals, the Hgb A2 can be as high as 3.5% to
heterozygous beta-globin mutation that
8% and the Hgb F as high as 2% to 5%.
causes a greater decrease in beta-chain pro-
Generally, beta-thalassemia trait is milder in
duction than seen in thalassemia minor, but
African-Americans (who frequently have a
not to the degree for which chronic transfu-
promoter gene mutation) but has a similar
sion therapy is required. The phenotype can
presentation in individuals of Chinese,
also occur in children who have a mutation
Southeast Asian, Greek, Italian, and Middle
that increases production of a-globin, in
Eastern heritage.
children who have coinherited alpha- and
Infants born in most states in the United
beta-thalassemia, and in other rarer muta-
States are screened for hemoglobinopathies.
tions. Children who have thalassemia
In states without newborn screening for
intermedia are able to maintain a hemoglo-
hemoglobinopathies and in recent immi-
bin of 7 g/dL or slightly higher with a greatly
grants to this country, affected children are
expanded erythron and may manifest bony
frequently found later than the newborn
deformities, pathological fractures, and
period, and the evaluation of their micro-
growth retardation. Children who have thal-
cytic anemia includes differentiation bet-
assemia intermedia can also have delayed
ween iron deficiency and beta-thalassemia
pubescence, exercise intolerance, leg ulcers,
trait. The red blood cell indices can be
inflammatory arthritis, and extramedullary
helpful in this differentiation as the hemo-
hematopoiesis causing spinal cord compres-
globin concentration and the red cell count
sion—a medical emergency requiring radia-
will generally be lower in iron deficiency.
tion therapy and transfusion. They can also
The distinguishing finding in beta-thalasse-
have iron overload due to increased absorp-
mia is a hemoglobin electrophoresis with an
tion of iron from the gastrointestinal tract
elevated Hgb A2
and F. Both will be
and intermittent transfusion. They are at risk
increased in beta-thalassemia trait without
for the cardiac and endocrine complications
iron deficiency, and will be normal or
of hemosiderosis, but usually at an older age
decreased in alpha-thalassemia and isolated
than chronically transfused children. Chela-
iron-deficiency anemia. There are several
tion therapy is indicated for increasing fer-
formulas to help in office screening, but
ritin and elevated liver iron.
they are also based on the assumption that
Children who cannot maintain a hemo-
the child is not iron deficient. Usually, iron
globin between 6 and 7 g/dL should have an
deficiency can be ruled out using free
alternative diagnosis considered. If thalas-
erythrocyte protoporphyrin, transferrin sat-
semia is the cause of the anemia, transfusion
uration, or ferritin as a screening test in
and/or splenectomy should be considered.
children who have a hypochromic microcytic
Frequently, adolescents and adults are
anemia. The least expensive test is a trial of
unable to tolerate the degree of anemia
iron and a repeat hemogram after a month. A
that is seen in thalassemia intermedia.
lead level should be obtained if there is an
Hypersplenism, splenic pain, congestive
index of suspicion for lead toxicity.
heart failure, severe exercise intolerance,
40
Chapter 4
thrombocytopenia, and leukopenia should
ratio as g, rather than b, globin is present in
be considered indications for transfusion
Hgb F). These children have little or no Hgb
and splenectomy.
A2 and a low reticulocyte count. The diag-
Appropriate clinical management of
nosis can be made with certainty by dem-
thalassemia intermedia patients may be
onstrating thalassemia trait in both parents,
more difficult than patients with thalasse-
by globin biosynthetic ratios, or by
mia major requiring chronic transfusion
beta-gene screening. Beta-gene screening
due to phenotypic heterogeneity. Patients
identifies the most common and some
with clinically mild disease still may have
uncommon mutations, but not all muta-
serious long-term complications as
tions. An electrophoresis showing only Hgb
described above due to ineffective erythro-
F, a complete blood count, and a peripheral
poiesis as well as chronic hemolysis, leading
blood smear will generally be diagnostic. In
to pulmonary hypertension with resultant
most states, these children will be discovered
congestive heart failure and thrombosis in
by newborn screening or occasionally by the
addition to cholelithiasis. Currently, trans-
obstetrician who makes a diagnosis of thal-
fusion therapy is limited to patients with
assemia trait in the mother and obtains a
symptomatic anemia or in children with
family history of thalassemia or anemia in
delayed growth and development and is
both parents prior to the birth of the baby.
generally tailored to the individual patient.
Children who have untreated thalasse-
Hydroxyurea has been utilized as in sickle
mia die in the first decade of life from
cell anemia, increasing Hgb F and obviating
anemia, septicemia, and pathologic frac-
some of the need for transfusion. Splenec-
tures. When palliative transfusions are
tomy has been utilized but now is thought to
introduced, children live into their late teens
be at least partly responsible for the devel-
eventually succumbing to heart failure due
opment of pulmonary hypertension and
to iron overload.
thrombosis secondary to chronic hemolysis.
With the introduction of frequent
Some argue that transfusion therapy is
chronic transfusion therapy with regular
underutilized in thalassemia intermedia and
iron chelation, children are now surviving
improved risk stratification is needed.
into adulthood, adding to the complexity of
Thalassemia major was first described
the disease. The longevity of patients who
by a Detroit pediatrician, Thomas Cooley,
are compliant with their chelation therapy,
in 1925. The clinical picture he described is
or in those who have received bone marrow
prevalent today in countries without the
transplantation, is not yet known.
necessary resources to provide patients with
chronic transfusions and iron chelation
therapy. Children who have untreated thal-
Neonatal screening for
assemia major have ineffective erythropoi-
hemoglobinopathies
esis, decreased red cell deformability, and
enhanced clearance of defective red cells by
Newborn screening identifies patients with
macrophages. The result is a very hypermet-
beta-thalassemia major, hemoglobin H, and
abolic bone marrow with thrombocytosis,
sickle cell disease. Patients with beta-
leukocytosis, and microcytic anemia in the
thalassemia major and sickle cell disease will
young child prior to the enlargement of
be asymptomatic at birth due to high fetal
their spleen. At presentation, they have
hemoglobin levels but will become symp-
almost 100% Hgb F (these cells have a
tomatic over the next 2 to 3 months as
longer life span due to a balanced globin
normal production of Hgb F wanes. Patients
Thalassemia
41
Table 4.1 Classification of the alpha-thalassemias.
Diagnosis
Genetic finding
Symptoms
Barts (%)
Silent carrier
a-/aa
Hematologically normal
1-3
a-thalassemia trait
a-/a- or aa/—
Mild anemia with microcytosis
3-6
and hypochromia
Hemoglobin H disease
a-/-
Moderately severe hemolytic
5-20
OR Hemoglobin
aaCS/— or a-/aCS-
anemia
H-constant spring
Icterus and splenomegaly
a-thalassemia major
-/-
Severe anemia not compatible
100
with life; hydrops fetalis
without intrauterine
transfusion
with significant Bart’s hemoglobin
(see
hemoglobin A and A2 in the normal patient.
Table 4.1) should be considered to have
The pattern FA is normal and should not be
hemoglobin H and require alpha-gene
interpreted as beta-thalassemia trait. This is
screening to detect the possibility of Hgb
a diagnosis made by the practitioner when
H-constant spring.
microcytic anemia is seen during routine
The presence of Hgb F only on the
childhood screening and an investigation
newborn screening will be interpreted as
for thalassemia confirms the diagnosis.
beta-thalassemia major (see Table 4.2). This
Hgb FE will be presumed to be Hgb E-
diagnosis must be confirmed with parental
beta-thalassemia until that diagnosis is inves-
electrophoresis and repeat testing of the
tigated and confirmed, or ruled out by repeat
infant at 2 to 3 months after fetal hemoglo-
electrophoresis and family studies. Hemoglo-
bin wanes with concomitant increase in
bin E is the most common abnormal
Table 4.2 Interpretation of common newborn screening results.
Result
Diagnostic possibilities
Action required
FA
Normal
None
b-thalassemia trait
Genetic counseling
F
b-thalassemia major
Referral to hematologist
FAS
Sickle cell trait
Genetic counseling
FS
Sickle cell disease (Hgb SS)
Repeat testing at 2-3 months
Hgb S/b0-thalassemia
Family studies
Referral to hematologist
FSA
Hgb S/bþ -thalassemia
Referral to hematologist
FSC
Sickle cell disease (Hgb SC)
Referral to hematologist
FAC
Hemoglobin C trait
Genetic counseling
FE
Hgb E/b-thalassemia
Repeat testing at 2-3 months
Hgb EE
Family studies
FAE
Hgb E trait
Genetic counseling
Order of results is listed from highest to lowest frequency (i.e., in FA, higher percentage of
Hgb F than Hgb A).
42
Chapter 4
hemoglobin discovered in the state of Cali-
MCV, and reticulocyte count would be helpful
fornia on newborn screening. It is common in
to narrow the differential. Iron deficiency is
Laos, Cambodia, and Thailand. Hemoglobin
unlikely before 6 months of age due to mater-
E results from a mutation in an exon (exon 1,
nal iron stores unless the baby was born
codon 26: GAG to AAG) that creates an
prematurely (and would have been routinely
alternate splice site competing with the nor-
put on iron therapy) or if the mother had
mal splice site. This results in abnormal
severe anemia during pregnancy or if there
hemoglobin production and mild microcytic
was acute or occult blood loss. Viral suppres-
anemia (Hgb
10 g/dL) in the homozygous
sion or aplasia from infections such as par-
state. Electrophoresis reveals about 90% Hgb
vovirus is possible, but less likely without
E with varying amounts of Hgb F. The het-
any history of recent infection or fever.
erozygote has a hemoglobin of about 12 g/dL
Beta-thalassemia as well as sickle cell anemia
with microcytosis and an electrophoretic
should be high in the differential as fetal
pattern consistent with Hgb E plus Hgb A.
hemoglobin lives 60 to 90 days and therefore
When combined with other more severe
by 4 months of age the infant will be produc-
beta-thalassemias, Hgb E-beta-thalassemia can
ing little fetal hemoglobin (a2g2) and should
produce an anemia that is profound requiring
have moved to almost solely hemoglobin A
chronic transfusion therapy. All children who
(a2b2) unless there is a problem with the
have Hgb E and Hgb F on their state screen
b-globin chain. Additional possibilities
require scrutiny for the emergence of a severe
include a hemolytic anemia, transient ery-
thalassemia syndrome. Individuals who are
throblastopenia of childhood (though rarely
homozygous for Hgb E should not have a
seen at this young of an age), infant leuke-
significant anemia and do not require special
mia, and folate and vitamin B12 deficiency if
care. Like patients with alpha-thalassemia, they
the baby is being fed goat’s milk or the
should not be treated with iron for their micro-
mother is vegan, respectively.
cytic anemia unless they are proven to have
concomitant iron deficiency.
2. What additional piece of information
may be helpful in this young child?
Case study for review
In addition to asking about a family history
of sickle cell anemia and thalassemias, the
You are seeing an infant that has been fol-
provider should look for the results of new-
lowing in your clinic since the newborn
born screen which could be very helpful in
period. The baby was born full-term without
this case. If the patient has a newborn screen
any significant issues. You are now seeing the
that is FA, sickle cell anemia and b-thalas-
baby back at 4 months for the well-child
semia major can be ruled out. More con-
check and their secondary vaccinations. The
cerning newborn screening results would
parents note that the baby has been more
include F (b-thalassemia major), FS (Hgb
listless, not eating as well, and looking paler.
SS disease or Hgb S/b0-thalassemia), and
There are no recent infections or fevers.
FSA
(Hgb S/bþ -thalassemia). b0- and
bþ -thalassemia are differentiated based
1. Assuming this baby is anemic, what would
on the presence (bþ ) or absence (b0) of
your differential diagnosis be at this point?
hemoglobin A, although phenotypically
the patient can have an extremely vari-
Causes of anemia are quite broad at this point.
able clinical presentation, in part due to
Complete blood count with differential,
the persistence of fetal hemoglobin. If the
Thalassemia
43
patient had sickle cell trait, their newborn
would be FA. As adults, after fetal hemo-
screen result would be FAS as the percentage
globin production has decreased, they
of hemoglobin A should be greater than
would have increased amounts of both A2
the percentage of hemoglobin S. If neces-
(>3.5%) and F (>2%) in addition to hemo-
sary, results can be confirmed at
4
to
globin A.
6 months of age after fetal hemoglobin
levels should be significantly decreased or,
5. What other clinical signs might be appar-
if possible, the parents can be tested. In
ent in the infant?
this case, the newborn screen results are F
only.
One would expect to see increasing signs of
extramedullary hematopoiesis with increas-
3. How might repeat hemoglobin electro-
ing age and decreasing fetal hemoglobin
phoresis be different at this age?
production. Some of the early signs could
be frontal bossing as well as maxillary and
The patient that has no hemoglobin A will
mandibular prominence. The infant may
be on the extreme end of the clinical spec-
have failure to thrive as well as the devel-
trum with definitive b-thalassemia major.
opment of hepatosplenomegaly.
For those patients with some b-
globin production, the clinical severity of
Suggested Reading
disease at this point cannot be determined
on a molecular level. Patients who are solely
Aessopos A, Kati M, Meletis J. Thalassemia inter-
fetal hemoglobin (a2g2) on newborn screen
media today: should patients regularly receive
transfusions? Transfusion 47:792-800, 2007.
will likely continue to make some amount of
Olivieri NF. The beta-thalassemias. N Engl J Med
fetal hemoglobin in addition to hemoglobin
341:99-109, 1999.
A2 (a2d2). Their repeat hemoglobin electro-
Singer ST, Kim HY, Olivieri NF, et al. Hemoglo-
phoresis will represent this with variable
bin H-constant spring in North America: an
amounts of hemoglobin A2 and F and no
alpha-thalassemia with frequent complica-
hemoglobin A.
tions. Am J Hematol 84:759-761, 2009.
Taher AT, Musallam KM, Karimi M, et al. Over-
4. What would you expect the parents’
view on practices in thalassemia intermedia
hemoglobin electrophoresis to show?
management aiming for lowering compli-
cation rates across a region of endemicity:
the OPTIMAL CARE study. Blood
115:
The parents likely both have b-thalassemia
1886-1892, 2010.
trait. If they were tested as newborns, they
Transfusion Medicine
5
Transfusion of blood products continues
patients to eliminate the risk of clerical
to be an important and necessary part of
error.
therapy in children with hematologic and
Directed donation from first-degree
oncologic diagnoses. Many complications
relatives
(especially maternal) should be
can result from transfusion, both infectious
discouraged for patients who may be can-
and noninfectious. With continued
didates for an allogeneic bone marrow
improvements in donor screening, and
transplant due to the possibility of antigen
better testing techniques, infections from
sensitization. If it cannot be avoided, the
known entities have become a rarity. How-
blood should be irradiated to prevent graft-
ever, as long as blood component therapy is
versus-host disease (GVHD). Studies have
derived from human blood donation, the
shown that blood from directed donation is
risk will persist. Donor selection criteria are
not safer than the general donation pool due
designed to screen out potential donors with
to current sophisticated screening and
increased risk of infection with HIV-1/2,
testing methods.
HTLV-I/II, and hepatitis B and C, as well
These risks must be carefully considered
as other infectious pathogens. Despite rig-
and weighed against expected benefit
orous screening and testing for these infec-
each time a transfusion is contemplated.
tions, the risk of transmitting these viruses is
Informed consent should be obtained prior
not totally eliminated.
to every nonemergent transfusion. In
Clerical errors and misidentification are
California, the Gann Act must be renewed
major risks for transfusion and can result in
on a yearly basis for those patients under-
serious consequences, including death. It is
going frequent transfusion due to an
essential that all blood samples drawn for a
underlying hematologic or oncologic
type and cross-match be clearly labeled with
condition.
the patient’s identification. Before adminis-
tration of the blood product, the order
should be checked, patient identification
Packed red blood cell transfusion
reviewed
(patient’s identification band),
and blood type verified. Many institutions
Transfusion decisions are made on the basis
have moved toward testing two separate
of clinical context, rather than the level of
blood type specimens for nonemergent
hemoglobin alone (see Figure 5.1). Prior to
transfusion in previously untransfused
transfusion, it is necessary to assess the
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Transfusion Medicine
45
46
Chapter 5
mechanism responsible for anemia (bone
related risks including risk of infection,
marrow infiltration, ineffective erythropoi-
febrile nonhemolytic transfusion reactions
esis of chronic disease, occult or obvious
(FNHTRs), and alloimmunization from
blood loss, transfusion or drug-related sup-
PRBC transfusion. Although not universal
pression of normal hematopoiesis, and
as yet in the United States, leukoreduction is
nutritional deficiency), the severity of the
becoming standard of care and is universal
signs and symptoms, and the likelihood of
in Canada and much of Europe.
resumption of normal hematopoiesis.
Irradiation of blood products should be
Whole blood is rarely, if ever, utilized in
utilized for all patients who are immuno com-
standard practice, though may occasionally
promised in order to prevent transfusion-
be utilized for massive transfusion after
associated graft-versus-host disease
(TA-
military trauma. Packed red blood cells
GVHD). This should include all oncology
(PRBCs) are depleted of the majority of
patients as well as neonates.
platelets and white blood cells and need
Cytomegalovirus (CMV)-negative blood
to be ABO and RhD compatible, at the
should be considered in patients in the
minimum. Phenotypically, and now more
peritransplant period who are noted to be
recently genotypically, matched blood for
CMV seronegative in addition to neonates
minor antigens should be utilized for
born to CMV-negative mothers and patients
patients undergoing chronic transfusion
with immunodeficiencies. Institutional
(e.g., sickle cell disease). One unit of PRBCs
guidelines vary and should be consulted in
is approximately
250
to
350 mL with a
determining the need for CMV-negative
hematocrit of 55% to 60% when prepared
blood secondary to the limited supply of
with adsol. Based on the hematocrit of the
this product.
stored PRBCs, one can calculate the pre-
Finally, washing of PRBCs is rarely indi-
sumed rate of rise from blood transfusion.
cated and decreases the red cell mass by
For adsol-preserved units, the transfusion
approximately 20%. Washing is indicated
factor is approximately
5
(assuming no
for severe allergic transfusion reactions,
hemolysis of transfused blood or blood
hyperkalemia, large-volume transfusions,
loss). The estimated hemoglobin
(Hgb)
and selective IgA deficiency.
increase can be derived utilizing this trans-
fusion factor:
Indications for PRBC transfusion
Estimated hemoglobin rise ðg=dLÞ
Neonates
Guidelines for transfusion in neonates and
¼VolumeofPRBCtransfusionðmL=kgÞ
Transfusion factor
infants vary widely in the literature. Clinical
correlation is required in addition to the
For example, for a transfusion of 15 mL/kg,
following basic guidelines:
the estimated hemoglobin increase should
* Transfusion of leukopoor, irradiated
be
3 g/dL. Of note, waiting a certain
blood in premature infants and low-birth
period of time for a posttransfusion
weight neonates
(CMV-negative if CMV
“reequilibration” prior to rechecking the
status of mother is negative or unknown)
hemoglobin is not necessary.
* High oxygen requirement or severe car-
Removal of the vast majority of remain-
diac disease, keep Hgb 11 to 15 g/dL
ing white blood cells, or leukoreduction, is
* Mild-to-moderate respiratory distress or
an important step in reducing transfusion-
perioperative care, keep Hgb 10 g/dL
Transfusion Medicine
47
Stable infants who become symptomatic
Oncology patients
from anemia, keep Hgb > 7 to 8 g/dL (based
In general, a hemoglobin of 7 g/dL is used as
on degree of symptoms)
the threshold for transfusion in pediatric
oncology patients. This may be altered in
Acute blood loss and nonimmune
cases where the patient is asymptomatic
hemolytic anemia
with imminent recovery of red blood cells
Unlike chronic conditions that result in a
(often heralded by increase in platelet
slow drop in hemoglobin and time for
count). Infants with effects on growth and
compensation, patients with acute blood
development due to anemia should be
loss or nonimmune hemolytic anemia may
maintained at higher hemoglobins; simi-
be symptomatic at much higher hemoglo-
larly, adolescents may complain of headache
bin levels and should be transfused accord-
and fatigue and be less symptomatic with
ingly. In situations of continuing blood loss,
hemoglobins in the
8 to
10 g/dL range.
patients should be transfused to achieve an
Those with cardiopulmonary dysfunction
expected hemoglobin level in addition to
and those requiring procedural sedation
receiving blood to compensate for ongoing
with anticipated blood loss should be kept
losses. In an emergent situation, transfusion
in the 8 to 10 g/dL range as well.
consent is not required, and if there is no
time for a cross-match, O-negative blood
Autoimmune hemolytic anemia
should be given.
In patients with an autoimmune hemolytic
anemia, immunosuppressive therapy is usu-
Severe chronic anemia
ally sufficient to abate the underlying pro-
In pediatric patients, iron-deficiency ane-
cess. In those patients who are symptomatic
mia is the most likely cause of severe
with a continuing drop in hemoglobin and/
chronic anemia
(i.e., Hgb < 5 g/dL), in
or resultant significant cardiopulmonary
addition to viral suppression, transient
dysfunction, phenotypically matched blood
erythroblastopenia of childhood, aplastic
may be given with close observation know-
anemia, and newly diagnosed leukemia.
ing that hemolysis is likely to continue and
Pediatric patients can present with an
may even be augmented.
extremely low hemoglobin (e.g., 2 to 3 g/
dL) and yet be relatively well compensated.
Chronically transfused patients
Patients should be assessed closely for sub-
Patients with b-thalassemia major as well as
tle signs of congestive heart failure (cardi-
some with b-thalassemia intermedia, Hgb
omegaly on chest radiograph, increased
SS, and Hgb S/b0-thalassemia require
diastolic blood pressures, hepatomegaly,
chronic transfusion therapy, as outlined in
oliguria, and periorbital edema) prior to
previous chapters. These patients should
transfusion. Slow transfusion (e.g.,
1 mL/
have a red cell phenotype prior to initiating
kg/h) has been recommended in the past,
transfusions and based on institutional
although studies have shown that
2
to
practice will usually receive phenotypically
3 mL/kg/h is safe in patients with normal
matched blood to a small panel of antigens
underlying cardiopulmonary function.
(at our institution, other Rh antigens
Those with signs of cardiopulmonary dys-
including C/c and E/e as well as Kell). If
function should be transfused slowly with
the patient develops antibodies to other
judicious usage of diuretics, and exchange
minor red cell antigens (e.g., MNS, Duffy
transfusion should be considered in those
[Fya/Fyb], Kidd [Jka/Jkb], and Lewis [Lea/
with heart failure.
Leb]), more extensive phenotype matching
48
Chapter 5
is performed in order to decrease the con-
sickle cell events, hyperbilirubinemia, or in
tinued risk for alloimmunization. Genotyp-
an anemic patient treated with severe fluid
ing for a wider range of antigens is becoming
restriction (increased ICP). In sickle cell dis-
more universally available and will likely
ease, exchange transfusion quickly reduces
become the future standard of care. Sickle
the concentration of sickle cells without
cell patients in addition should receive sick-
increasing the hematocrit or whole blood
ledex-negative blood as blood donors are
viscosity. In addition, red cell exchange
not screened for sickle cell trait. Transfusion
transfusion reduces iron accumulation since
is given on a regular basis (i.e., every 3 to 4
an equal volume of red cells and iron are
weeks) in order to suppress ineffective
removed as infused. Therefore, for sickle cell
erythropoiesis.
disease, erythrocytopheresis is preferred over
straight transfusion in patients requiring
chronic transfusion in addition to those
Dosing of PRBC transfusion
patients with acute events. Automated ery-
throcytopheresis can be done rapidly and
The desired incremental increase in hemo-
safely in most situations. Limitations of this
globin should be considered when deter-
technique include increased red cell utiliza-
mining the amount of blood to be trans-
tion, venous access, and increased cost.
fused. In general, 10 to 20 mL/kg over 4
hours is given in order to increase the
hemoglobin by 2 to 4 g/dL. Care should
Platelets
be taken to not waste blood; therefore,
orders should be rounded to the nearest
Platelet transfusions are indicated for throm-
unit or half unit, as feasible. Remaining
bocytopenic patients with bleeding due to
blood may be sterilely aliquoted and used
severely decreased platelet production or for
later in the same patient in order to decrease
patients with bleeding secondary to function-
exposure to multiple donors. Slow transfu-
ally abnormal platelets. Transfusion is not
sion should be given in patients with signs of
indicated for those with rapid destruction
volume overload or cardiopulmonary dys-
(e.g., immune thrombocytopenic purpura
function. Patients with frank heart failure
and neonatal alloimmune thrombocytope-
should be exchange transfused. Care should
nia) unless there is life-threatening hemor-
be taken to not increase the hemoglobin
rhage; however, transfusion may be useful in
above 12 g/dL in patients with sickle cell
the bleeding patient with rapid consumption
disease secondary to hyperviscosity and
(e.g., disseminated intravascular coagula-
increased risk of CNS events. Similarly, in
tion [DIC]) or dilutional thrombocytopenia
leukemic patients with hyperleukocytosis at
(massive transfusion or exchange). Platelets
diagnosis(whitebloodcellcount>100
109/
are frequently needed in the patient receiving
L), the hemoglobin should preferably be kept
chemotherapy or one thrombocytopenic
below 10 g/dL and transfusion avoided if
secondary to a marrow infiltrative process.
possible due to the same risks of increasing
viscosity and the propensity for leukostasis.
Indications for platelet
Exchange transfusion
transfusion
Exchange transfusion or erythrocytopher-
1. Premature or sick infants
esis may be indicated in the severely anemic
a. Stable infant with platelet count
patient with congestive heart failure, acute
<50
109/L
Transfusion Medicine
49
b. Distressed infant with platelet count
g. Intramuscular injection
(i.e., PEG-
<100
109/L
asparaginase) with platelet count
2. Children
<20
109/L
a. Platelet count <10
109/L; higher if
Dosing of platelet transfusion
febrile, septic, or with active bleeding
One unit of random platelets (
40 mL) per
b. Platelet count <20 to 50
109/L with
10 kg will increase the platelet count by 40 to
a minor invasive procedure such as:
50
109/L if there is no active consumptive
i. Lumbar puncture
process (e.g., fever, immune thrombocyto-
ii. ECMO or CV bypass
penic purpura, sepsis, alloimmunization, or
iii. Other minor procedures such as
DIC) or sequestration. This is equivalent to
central line placement
an increment in platelet count of 10
109/L
c. Invasive procedure in a patient with a
per mL/kg of transfused platelets (i.e., in a
qualitative platelet defect
10 kg child, 10 mL/kg or 100 mL of trans-
d. More invasive procedures will require
fused plates should raise the platelet count
discussion with the surgical team regarding
by 100
109/L). The platelet count should
platelet transfusion threshold, although
be checked 1 to 2 hours after infusion to
recommendations are generally not evi-
identify refractory patients. A patient is
dence based
refractory if
1 hour after transfusion the
3. Patients
undergoing therapy for
platelet increment is less than
5
to
malignancy
10
109/L per unit transfused for two sep-
a. Platelet count <10
109/L; higher if
arate transfusions.
febrile, septic, or with active bleeding
A patient may have platelet refractori-
b. Induction chemotherapy:
ness secondary to alloantibodies (immune
i. ALL <10
109/L
mediated). Nonimmune causes of platelet
ii. AML <10 to 20
109/L
refractoriness are common and include
c. Children undergoing intensive ther-
splenomegaly, fever, infection, DIC, and use
apy with active mucositis should have
of amphotericin B.
their platelet count maintained at 30 to
For refractory patients, a trial of cross-
50
109/L (i.e., Head Start protocol for
matched platelets should be given. Other
brain tumors)
possibilities for treatment should this fail
d. Lumbar puncture with platelet count
include leukocyte-depleted, human leuko-
<20 to 50
109/L; potentially <50 to 100
cyte antigen
(HLA)-matched platelets,
109/L with diagnostic lumbar puncture
intravenous immunoglobulin (IVIG) with
e. Bleeding patient with normal coagu-
HLA-matched platelets, or massive trans-
lation studies, platelets
<50
109/L;
fusion with random donor platelets
(to
with abnormal coagulation studies,
overwhelm the antibody).
platelet count <100
109/L
Most institutions now utilize pheresed
f. Patient requiring a moderately inva-
platelets that are harvested from a single
sive surgical procedure, with a platelet
donor and generally contain greater than
count
<50 to 100
109/L
(requiring
30
109/L of platelets, equivalent to
discussion with surgical team secondary
approximately 6 to 8 units of random plate-
to lack of evidence-based guidelines).
lets. The volume is usually 250 to 350 mL.
Most surgeons would like platelet count
Secondary to the apheresis, these platelet
to remain >50 to 75
109/L for 48 to
products are considered to be leukoreduced.
72 hours after the surgical procedure
In addition, although controversial, most
50
Chapter 5
believe that apheresis is sufficient to reduce
concentration of plasma antibodies, FFP is
the risk of CMV transmission. Plasma ABO
often the cause in cases of transfusion-asso-
compatibility should generally be utilized,
ciated acute lung injury (TRALI) and there-
especially with increasing volume of trans-
fore should be used judiciously. The direct
fusion. Type-specific platelets can be given
antiglobulin test (DAT; Coombs test) may
in cases of refractoriness and should be
also be positive for this reason.
given to patients in the peritransplant
period to eliminate the production of any
Indications for FFP
potential red blood cell antibodies. As with
* Bleeding or invasive procedure with
PRBC transfusion, platelet irradiation is
documented clotting factor deficiency and
required in oncology patients, premature
appropriate factor not available
or low birth weight newborns, and patients
* Treatment of protein C or S deficiency,
with immunodeficiencies.
factor XI deficiency (hemophilia C)
Dosage of platelet transfusion is gener-
* Bleeding during massive transfusion, not
ally 10 to 15 mL/kg and can be given over 30
from thrombocytopenia
minutes to 1 hour. Due to the expected
increase in platelet count and short life-
Dosing of FFP
span of platelets, 1 unit of pheresed platelets
The usual dosage of FFP is 10 to 15 mL/kg
is usually sufficient for transfusion in
over 1 hour, which is expected to increase
patients over 30 kg. As with PRBCs, pher-
the concentration of coagulation factors by
esed platelet units should be rounded to
25% to
50%. Due to the presence of
the nearest half and full units as feasible
isohemagglutinins, FFP should be ABO
to decrease waste and may be sterilely
matched. White blood cells are killed or
aliquoted as needed.
made nonfunctional during the freezing
process; therefore, leukoreduction and irra-
diation are unnecessary.
Fresh frozen plasma
Fresh frozen plasma (FFP) is a source of
Cryoprecipitate
plasma proteins, including nonlabile clotting
factors, such as fibrinogen. It is used for the
Cryoprecipitate is prepared by thawing FFP
treatment of stable clotting factor deficiencies
and recovering the cold precipitate. Each
in which no concentrate is available (not for
bag contains >80 U factor VIII coagulant
factor VIII or IX). By definition, each milliliter
activity and >150 mg fibrinogen in approx-
of undiluted plasma contains 1 international
imately 15 mL of plasma. In addition, cryo-
unit (IU) of each coagulation factor.
precipitate contains factor XIII and von
Plasma consists of the anticoagulated
Willebrand factor. Bags must usually be
clear portion of blood separated by centri-
pooled to achieve an adequate dose.
fugation. FFP is collected from single
donors, with each unit being removed from
Indications for cryoprecipitate
a unit of whole blood and frozen within 6 to
* Bleeding or invasive procedure with
8 hours of collection. FFP should not be
factor VIII deficiency or von Willebrand
used when the coagulopathy can be cor-
disease and factor concentrate not available
rected more effectively with specific treat-
* Bleeding or invasive procedure with
ment such as vitamin K, cryoprecipitate, or
hypofibrinogenemia or factor XIII
factor concentrate. Due to the high
deficiency
Transfusion Medicine
51
Dosing of cryoprecipitate
increased granulocyte yields, and therefore
The usual dosage for cryoprecipitate is
the therapeutic benefit of transfusion, as
1 unit/5 kg. Obtain a fibrinogen level at
the dose of granulocytes is the most impor-
30 minutes postinfusion. For hypofibrino-
tant factor in success of this treatment
genemia with coagulopathy, the goal is to
modality.
maintain fibrinogen >100 mg/dL Specific
Granulocytes migrate toward, phago-
factor or coagulation protein levels need
cytize, and kill bacteria. When given a gran-
to be determined in addition to assessing
ulocyte transfusion, the cells migrate to the
clinical status to decide on frequency of
foci of infection, though there is rarely a
transfusion. As with FFP, cryoprecipitate
measurable increase in the peripheral
is preferably ABO compatible and leukor-
granulocyte count. This is likely from
eduction and irradiation are not required.
sequestration at the site of infection, prior
immunization to leukocyte antigens, or a
consumptive process secondary to the infec-
Antithrombin III
tion. Side effects of granulocyte transfusion
include the risk of CMV infection, TA-
Antithrombin III
(ATIII) concentrate is
GVHD, respiratory distress with pulmonary
available for use in patients with inherited
infiltrates (TRALI, concurrent administra-
or acquired ATIII deficiency
(sepsis,
tion of amphotericin B, or secondary to
thrombosis, and medication induced). It
granulocyte
sequestration),
and
may also be needed in patients receiving
alloimmunization.
heparin therapy who have a low ATIII
level. Patients with thrombosis after aspar-
Preparation of granulocyte
aginase therapy should have an ATIII
concentrates
level checked and repletion as necessary.
Donor mobilization is recommended with
ATIII replacement should also be consid-
G-CSF at 5 mcg/kg (maximum 300 mcg IV/
ered in patients with veno-occlusive disease
SC) and dexamethasone 12 hours prior to
(sinusoidal obstructive syndrome) after
col-lection. Granulocytes are then collected
hematopoietic stem cell transplantation.
by apheresis and ideally should be transfused
Dosing of ATIII is based on the baseline
within 8 to 12 hours of collection. RBC
and desired level.
contamination requires ABO compatibility.
Due to risks of TA-GVHD and CMV infec-
tion, irradiation and CMV seronegativity
Granulocyte transfusion
are required
(in those that are CMV-
negative), respectively. Amphotericin B and
Patients with profound and prolonged neu-
concomitant granulocyte transfusion is
tropenia are at increased risk for serious life-
potentially associated with severe pulmo-
threatening fungal and bacterial infections.
nary reactions, although the evidence is
The beneficial effect of granulocyte trans-
controversial; however, it is reasonable to
fusion in this population, with known
space these therapies apart by at least
persistent infection, especially with Gram-
4
hours. HLA-matched granulocytes
negative organisms and fungus, is yet to be
should be given in patients with known
proven by randomized controlled trials,
alloimmunization. Since granulocyte half-
although observational studies are available.
life is only
7 hours, daily collection and
Donor mobilization with granulocyte
transfusion for several days are likely
colony-stimulating factor
(G-CSF) has
required for benefit.
52
Chapter 5
Indications for granulocyte
clerical error resulting in the transfusion of
transfusion
an ABO incompatible unit. AHTRs classi-
With a lack of randomized controlled trials,
cally present with fever, chills, nausea, and
there are no evidence-based guidelines for
vomiting in addition to dyspnea, hypo-
granulocyte transfusion. Limited data in
tension, shock, hemoglobinuria, and DIC.
neonates with sepsis has failed to show a
Bacterial contamination must be considered
significant benefit. After weighing the
in the differential for an AHTR. Delayed
potential risks and benefits, as well as deter-
hemolytic transfusion reactions (DHTRs)
mining the daily availability of an eligible
present 2 to 14 days after transfusion with
donor and collection site, granulocyte trans-
milder symptoms including low-grade
fusion can be considered in severely neu-
fever, jaundice, and a posttransfusion
tropenic patients with a refractory or pro-
hemoglobin increment less than expected
gressive bacterial or fungal infection on
due to minor antigen incompatibility
appropriate, aggressive therapy with neu-
(alloimmunization) from prior transfusion.
tropenia that is expected to continue for, at
Evaluation of a potential AHTR should
the least, several days.
include work-up of all recently transfused
blood products (see Figure 5.2). A bedside
check of labeling should occur followed by
Transfusion reactions
laboratory evaluation including repeat
cross-matching and a DAT (Coombs test).
Approximately 4% of transfusions are asso-
The DAT may not always be positive if all
ciated with some form of adverse reaction,
the antibodies have been destroyed during
ranging from brief episodes of fever to life-
the hemolytic crisis. Other labs that should
threatening episodes of hemolysis and
be sent to rule out intravascular hemolysis
shock. Fortunately, the majority of reactions
include indirect bilirubin, LDH, plasma-
are short-term, specifically FNHTRs and
free hemoglobin (and/or haptoglobin), and
allergic reactions, and easily managed.
urinalysis for hemoglobinuria.
Life-threatening reactions are nearly always
DHTRs lead to extravascular hemolysis
due to clerical error resulting in transfusion
and should be evaluated in a patient with
of an ABO incompatible unit. The challenge
clinical or laboratory symptoms after the
for the clinician is to promptly recognize
first
24 hours. Labs to follow include a
potential serious complications that may
posttransfusion hemoglobin level and retic-
present with common symptoms such as
ulocyte count, indirect bilirubin, LDH, and
fever. For any transfusion reaction, a bed-
DAT.
side check of all labels, forms, and patient
If an AHTR is suspected, emergent man-
identification should be done, in addition to
agement is vital. The transfusion should be
notifying the blood bank. Transfusion reac-
immediately stopped and IV fluid resusci-
tions are summarized below and include
tation commenced. Inotropic support may
FNHTRs, allergic reactions, immune-
be required for hypotension and shock.
mediated hemolysis, TRALI, transfusion-
Renal perfusion and urine output should
associated circulatory overload
(TACO),
be followed closely and additional blood
TA-GVHD, and acute infection.
product support may be required. DHTRs
usually do not require intervention
Hemolytic transfusion reactions
although may rarely cause profound
Hemolytic transfusion reactions can either
anemia.
be acute or delayed. Acute hemolytic trans-
FNHTRs were significantly more com-
fusion reactions (AHTRs) are usually due to
mon prior to near universal leukoreduction
Transfusion Medicine
53
Fever (38.0ºC or 100.4ºF)
during blood transfusion
Any concerning clinical signs including:
Signs of sepsis (hypotension, delayed capillary refill, mental status changes)
Signs of hemolysis (hemoglobinuria, dyspnea, hypotension)
Yes
No
Stop transfusion
1ºC (1.8ºF) in temperature
Initiate supportive care (IVF,
over last 24 hours?
antibiotics)
Contact Intensive Care
Yes
No
Continue transfusion
Stop transfusion
Monitor clinical exam
closely
Contact Blood Bank
Bedside check of all forms, labels, and patient identification
Blood culture from patient and remaining blood product
Repeat crossmatch on remaining blood product
STAT labs: CBC, indirect bili, LDH, UA, DAT, plasma free hgb
Consider CXR if respiratory symptoms or new hypoxia
Positive work-up?
Yes
No
Provide appropriate care based on
Restart transfusion
likely diagnosis and clinical symptoms
Monitor clinical exam closely
*See text for more detail
Complete in allotted time as possible
Figure 5.2 Approach to fever during blood transfusion. (Abbreviations: CBC, complete blood
count; LDH, lactate dehydrogenase; UA, urinalysis; DAT, direct antiglobulin test,
(Coombs);
CXR, chest X-ray).
and occur due to pyrogenic cytokines
discontinued until an AHTR can be ruled
released by leukocytes during blood com-
out (see Figure 5.2). Fever due to FNHTR is
ponent storage. FNHTR is defined as a
usually self-limited and resolves with anti-
temperature increase of 1
C (1.8
F) with-
pyretic usage and stopping the transfusion.
out any other attributable cause. More seri-
Chills, rigors, and discomfort can occur,
ous causes of fever including AHTR and
mimicking an AHTR. Transfusion can
bacterial contamination must be ruled out
potentially be restarted after AHTR has been
prior to the diagnosis of FNHTR being
ruled out based on the clinical status of the
made. Transfusion must therefore be
patient and urgency of the transfusion.
54
Chapter 5
Although patients with a history of FNHTR
transfusion with plasma-containing blood
oftenreceive premedicationwith acetamin-
components. Signs and symptoms include
ophen with future transfusions, this prac-
hypoxia, chest infiltrates (without volume
tice is not evidence based. Antipyretics
overload), dyspnea/tachypnea, fever, and
should be considered for the patient with
hypotension. TRALI generally should
multiple FNHTRs; if acetaminophen is not
occur within 6 hours of transfusion com-
effective, a trial of washed blood compo-
pletion. A majority of patients with TRALI
nents can be considered. Of note, transfu-
will recover in a
2- to
4-day period,
sion can be given to the febrile patient if
although respiratory support, and in some
urgently necessary, as the criteria for work-
cases mechanical ventilation, is often
up (beyond a close and continued clinical
required.
assessment) is an increase of 1
C in fever
compared to the fever curve from the
Transfusion-associated
previous 24 hours.
circulatory overload
TACO also is becoming more widely rec-
Allergic transfusion reactions
ognized but is likely rare in the pediatric
Allergic reactions are typically type I hyper-
population. TACO occurs due to cardio-
sensitivity reactions to plasma in blood
genic edema from too rapid or too large a
components. Allergic reactions are com-
volume of transfusion. Clinical signs such as
mon, occurring in 1% to 5% of transfusions,
dyspnea/tachypnea and hypoxia may be
and usually with mild symptoms such as
confused with TRALI. Differentiating fea-
urticaria, although anaphylaxis can occur.
tures include hypertension rather than
For the majority of allergic reactions, an
hypotension due to pulmonary overcircula-
antihistamine such as diphenhydramine is
tion with a positive fluid balance. Chest
sufficient to alleviate symptoms and the
radiograph should be more consistent with
transfusion can likely be completed in the
pulmonary edema/effusion rather than
allotted time. For more severe symptoms,
infiltrates. Aggressive diuresis should be
the transfusion should be stopped. Steroids,
utilized.
an H2 blocker (e.g., ranitidine), epineph-
rine, volume expansion, b2 agonists (e.g.,
albuterol), and oxygen may be required.
Transfusion-associated
Patients with a severe allergic reaction
graft-versus-host disease
should be tested for IgA deficiency. As with
TA-GVHD can occur in immunocompro-
FNHTRs, premedication (with an antihis-
mised patients who receive nonirradiated
tamine) remains controversial in patients
blood or platelet transfusion due to donor
with a history of a reaction. Patients with
T-lymphocytes that cannot be rejected by
multiple episodes may benefit from preme-
the host. TA-GVHD has also been reported
dication with an antihistamine and, if not
in immunologically normal patients with an
effective, potentially corticosteroids. If aller-
HLA-compatible donor such as in homo-
gic reactions continue, washed blood pro-
geneous populations or in cases where the
ducts should be utilized.
donor is a close relative (directed donation).
Clinical symptoms are equivalent to those
Transfusion related acute lung
with transplant GVHD including fever,
injury
anorexia, vomiting/diarrhea, and skin rash.
TRALI is becoming increasingly recognized
Hepatic dysfunction and pancytopenia can
as an important cause of morbidity after
similarly manifest.
Transfusion Medicine
55
Bacterial infections
2. What amount of PRBCs should be given?
Blood components may be contaminated
3. Are there any special considerations for
with bacteria at the time of collection or
the type of blood product required?
during processing. Infection rates are higher
4. What are some specific risks with this
after platelet transfusion since platelets are
patient presentation?
stored at room temperature, although clin-
First, the patient should be given PRBCs,
ical signs and symptoms are often more
pheresed platelets
(if possible), and FFP.
severe after transfusion of contaminated
Cryoprecipitate may or may not be required
PRBCs. Infection is associated with a rapid
dependent on the increase in fibrinogen
onset of symptoms and a high rate of
with FFP. If the patient is clinically wors-
morbidity and mortality, especially from
ening (e.g., problems oxygenating, drop-
Gram-negative organisms. The patient may
ping blood pressures), consideration should
present acutely septic with fever/chills,
be given to transfusing O-negative PRBCs
hypotension, tachycardia, and shock. If
while awaiting cross-matching.
bacterial contamination is suspected, the
Second, one can utilize the transfusion
transfusion should be stopped immediately.
Aggressive supportive treatment including
factor to estimate the increment in hemo-
globin with transfusion. In this case, the
IV fluid resuscitation, initiation of broad-
spectrum antibiotics, and potential therapy
transfusion factor will likely overestimate
the bump as the patient is actively septic
for renal failure, shock, and/or DIC should
and therefore hemolyzing PRBCs and con-
be started emergently.
suming platelets. Still, estimating will help
determine a reasonable plan. Assuming the
Case study for review
decision is made to give 15 mL/kg (3 units),
one would expect at most an increase in
You are following a 12-year-old, 60-kg boy
hemoglobin of 3 g/dL:
admitted to the pediatric intensive care unit
Transfusion amount
(mL/kg)/transfusion
with septic shock. He is secondarily found to
factor ¼ Hgb increase (g/dL)
be anemic and thrombocytopenic with coa-
Therefore, in this case (15 mL/kg)/5 ¼ 3 g/
gulopathy and hypofibrinogenemia in DIC.
dL
After initial stabilization of the patient
with IVF, antibiotics, mechanical ventila-
In general, platelet and FFP transfusion will
tion, and pressors, the PICU team is deter-
also be 10 to 15 mL/kg. Platelet transfusion
mining their transfusion plan. Current
should initially be a maximum of one pher-
hemoglobin is
7.0 g/dL, platelets are
esed unit (approximately 250 to 350 mL).
34
109/L, PT is
27.2 seconds, PTT is
Posttransfusion hemoglobin, platelets, and
74.6 seconds, and fibrinogen is 76 mg/dL.
coagulation studies can help determine the
There is no noted active bleeding. 1:1 mix-
need for additional transfusion as well as the
ing studies correct both the PT and PTT,
likely frequency of necessary transfusion
implying a factor deficiency rather than an
(directly dependent on the clinical status
antibody. The patient does appear to have
of the patient). Time for
“equilibration”
some amount of hemolysis with an elevated
after the completion of transfusion is
indirect bilirubin and LDH but has a neg-
unnecessary.
ative direct Coombs test.
Third, in this case, no significant special
1. What blood products should be
arrangements need to initially be made in
transfused?
regards to the transfusions. The patient does
56
Chapter 5
not need sickledex-negative blood unless
transfusion-associated
microchimerism.
they have concomitant sickle cell disease.
Microchimerism is the presence of donor
Also, phenotypically matched blood is
lymphocytes in the recipient’s circulation.
unnecessary. As the patient does not
The significance of transfusion-associated
have a known underlying reason to be
microchimerism is unknown. Patients
immunocompromised, irradiated blood
receiving large amounts of plasma are at
and platelets are not required. Similarly,
risk for developing TRALI that must be
CMV-negative blood is unnecessary.
considered with worsening respiratory
Finally, specific risks exist for this patient
symptoms.
and must be considered during transfusion.
Due to the likelihood of continued transfu-
sions as well as the presence of sepsis, the
Suggested Reading
patient may develop volume overload and
potentially TACO; therefore, the judicious
Eder AF, Chambers LA. Noninfectious complica-
use of diuretics with transfusion should be
tions of blood transfusion. Arch Pathol Lab
Med 131:708-718, 2007.
considered. In addition, a significant vol-
Fasano R, Luban NL. Blood component therapy.
ume of PRBC transfusion can have a dilu-
Pediatr Clin N Am 55:421-445, 2008.
tional effect on platelets and coagulation
Klein HG, Spahn DR, Carson JL. Red blood cell
factors, thus increasing transfusion require-
transfusion in clinical practice. Lancet
ments for these blood products. The patient
370:415-426, 2007.
is also at risk for hyperkalemia with increas-
Roseff SD, Luban NL, Manno CS. Guidelines for
ing PRBC transfusion. If hyperkalemia
assessing appropriateness of pediatric trans-
begins to occur, washed PRBCs should
fusion. Transfusion 42:1398-1413, 2002.
be given. Massive PRBC or whole blood
Stroncek DF, Rebulla P. Platelet transfusions.
transfusion has been shown to induce
Lancet 370:427-438, 2007.
Chelation Therapy
6
Transfusional iron overload
routine use. Currently, R2
MRI (proton
magnetic resonance imaging) is becoming
Iron overload may occur in any patient
more widely utilized as an accurate and
receiving intermittent transfusions for acute
noninvasive measure of organ iron content,
illness
(e.g., sickle cell disease), chronic
and T2
MRI is becoming the new standard
transfusion therapy (thalassemia, sickle cell
for measuring cardiac iron stores. Further
disease, red cell aplasia, bone marrow failure
study is required for these new techniques to
syndromes), or in those having received
become uniformly calibrated methods.
an intensive period of frequent transfusion
Institution of chelation therapy relative to
(myelosuppressive chemotherapy/radiation
transfusion frequency is based on institu-
for treatment of malignancy and after
tional practice. Patients that will require
hematopoietic stem cell transplant). Each
chronic transfusion therapy should be che-
milliliter of blood contains 1 mg of iron and
lated before their liver iron reaches a high
normal iron stores are approximately 3 g,
level, measured by liver biopsy, MRI quan-
with 2 g in the blood and 1 g in the liver.
tification, or based on the number of trans-
Therefore, patients receiving frequent trans-
fusions received (i.e., chelation initiated after
fusion likely will have received a large
receiving 10 to 20 transfusions, or approxi-
amount of transfused iron and should be
mately 3 to 6 g of transfusional iron in a
monitored for evidence of iron accumulation.
30 kg patient). Thalassemia patients tend to
There is no simple test for quantifying
have a higher iron burden than sickle cell
total iron burden. Serial serum ferritin levels
patients as the chronic inflammatory state in
are helpful in determining hepatic iron
sickle cell disease limits gastrointestinal iron
stores, but values are altered in states of
absorption. Thalassemia patients should be
inflammation. Liver biopsy remains the
counseled to be on a low-iron diet.
gold standard for quantification of total
Patients receiving intermittent transfu-
body iron but is an invasive procedure. Liver
sion or those receiving frequent transfusion
iron levels
7 mg/g dry weight liver are
therapy in a short period of time (e.g., with
indicative of iron overload. The supercon-
intensive leukemia therapies) may have a
ducting quantum interference device
more insidious development of iron overload.
(SQUID) is accepted as a noninvasive
Patients with leukemia should have serum
method to quantitate total body iron, but
ferritin levels checked at the end of therapy.
its limited availability does not allow for
Those with ferritin levels
> 1000 ng/mL
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
58
Chapter 6
should have serial checks every 3 months; if
effective in all patients, which must be
ferritin remains above this threshold after 6 to
weighed against the risks of noncompliance
12 months, the patient should undergo liver
with Desferal. Deferiprone
(Ferriprox or
biopsy or R2 MRI for more accurate iron
Kelfer) was approved by the Food and Drug
quantification. These patients, especially
Administration
(FDA) in October
2011.
male adolescents, may benefit from monthly
Usage of two agents concomitantly is cur-
phlebotomy which can be discontinued when
rently undergoing clinical trials and may have
the ferritin and liver iron concentration reach
particular benefit in chelation of cardiac iron.
normal levels (i.e., 1.6 mg/g dry liver weight).
Concomitant administration of ascorbic
Younger children and menstruating females
acid (vitamin C) increases the excretion of
have been shown to have reversible iron
iron when given with chelation. Iron che-
overload in most such cases.
lators should not be administered when
Current treatment strategies for iron
there is concern for a bacterial infection.
overload and potential side effects of iron
By mobilizing free iron, chelators promote
chelators are summarized in Table 6.1. Des-
bacterial growth, in particular Yersinia
feroxamine (Desferal) has the longest treat-
enterocolitica. For febrile patients on chela-
ment record but requires parenteral admin-
tor therapy, chelation should be stopped
istration and has been widely replaced by
until blood cultures are definitively nega-
deferasirox (Exjade) due to the convenience
tive. Broad-spectrum antibiotic prophylaxis
of oral intake and therefore presumed
should be initiated and include coverage for
increased compliance. Exjade may not be
this unusual organism.
Table 6.1 Iron chelators.
Name
Desferoxamine (Desferal)
Deferasirox
Deferiprone (Ferriprox/
(Exjade)
Kelfer/L1)
Dose (mg/kg/d)
25-50
20-40
75-100
Administration
SC/IV, given as continuous
PO, daily
PO, TID
infusion over 8-24 hours,
5-7 d/wk
Side effects
Irritation at infusion site,
GI disturbance,
Agranulocytosis, neutro-
ototoxicity (tinnitus,
rash, renal and
penia, GI disturbance,
transient hearing loss),
hepatic
transaminitis,
ocular disturbance
impairment,
arthropathy, progres-
(decreased night vision),
GI hemorrhage
sion of hepatic fibrosis
allergic reactions, growth
failure, skeletal distur-
bance, pulmonary
hypersensitivity
Potential thera-
Highly effective but compli-
Long-term data
Recently approved for
peutic issues
ance may be an issue due
lacking; may
use by the FDA; long-
to route of administration
not be effective
term data lacking
in all patients
Abbreviations: SC, subcutaneous; IV, intravenous; GI, gastrointestinal; FDA, Food and Drug
Administration.
Chelation Therapy
59
Lead toxicity
minimal amounts
(3%) detected in the
plasma. The half-life in the blood is approx-
Lead poisoning is an environmental disease
imately 21 to 30 days. Excretion is primarily
that has undergone a major evolution in the
through the kidneys, with small amounts
past few decades. Recognition of the devas-
deposited in the hair, nails, and bile. The
tating neurologic effects of high lead levels
lead that remains in the body accumulates
and knowledge of the causes have led to
mostly in the bone (65% to 90%). Lead can
universal efforts to decrease environmental
enter any cell and toxicity may occur in any
lead contamination, with a resultant
tissue or organ. Classically, in severe lead
decrease in measured blood lead levels in
intoxication, gastrointestinal and central
children over the past
2
to
3
decades.
nervous system toxicities are the most clin-
Sources of lead have included gasoline addi-
ically apparent. Gastrointestinal symptoms
tives, food can soldering, lead-based paints,
include anorexia, nausea, vomiting, abdom-
ceramic glazes, certain toys, drinking water
inal pain, and constipation. The blood lead
systems (lead pipes), and folk remedies. The
level is typically 50 mcg/dL or greater when
use of these products has been markedly
these symptoms are present. Lead poisoning
reduced as the result of federal guidelines
was a lethal disease in the United States
and the development of cost-effective alter-
decades ago, primarily related to neurotoxic
natives. Lead in gasoline and paint is at
effects. At levels above 100 mcg/dL, children
extremely low levels and has been elimi-
may show evidence of encephalopathy,
nated altogether from food can soldering.
including a marked change in mentation
Housing built prior to 1960 is likely to have
or activity, ataxia, seizures, and coma.
been painted with high-content lead-based
Increased intracranial pressure may be pres-
paint, and since lead isotopes are very stable,
ent on examination. These effects are usu-
environmental exposure presents an on-
ally permanent with long-term sequelae of
going risk. High risk populations have a
retardation, palsies, and growth failure.
greater likelihood of living in older housing,
Most children who have elevated blood
which has not had lead abatement. Risk
lead levels have subclinical disease. Fewer
factors for excessive lead exposure include
than
5% of children present with overt
poverty, age younger than 6 years, African-
symptoms of lead toxicity. An elevated
American ethnicity, and urban housing.
blood lead level, suggesting excessive envi-
Lead may enter the body through direct
ronmental exposure, is defined as 10 mcg/dL.
ingestion, inhalation, or via skin absorption.
Many studies have shown associations
The most common pathway among young
between blood lead levels and impaired neu-
children is through the mouth. Lead absorp-
rocognitive function. These results have pro-
tion is enhanced in the presence of other
vided the primary impetus for current public
dietary mineral deficiencies, such as calcium
health efforts. Of note, no lead level is normal
and iron, due to competitive biochemical
and even lead levels below 10 mcg/dL are
pathways. Pica behavior enhances the like-
thought to lead to subtle neurocognitive
lihood of direct ingestion. Toddlers in par-
dysfunction.
ticular are at risk due to normal develop-
mental behaviors such as putting objects
Screening for lead toxicity
and toys in the mouth and chewing on
Prevention and treatment of lead toxicity
unusual surfaces (e.g., window sills).
remains a major public health concern, and
Lead entering the intravascular space
efforts for screening have primarily focused
rapidly attaches to the red blood cell, with
on high risk populations. All children
60
Chapter 6
Table 6.2 Recommendations for the treatment of lead toxicity.
Blood lead
Recommendations
level (mcg/dL)
<10
Environmental assessment, risk reduction, nutritional guidance, retest in 3 mo if
concern for exposure
10-14.9
Environmental assessment, risk reduction, nutritional guidance, report to public
health department, confirm result with venous sample
15-19.9
As above, if no improvement on retest, aggressive environmental assessment,
abdominal radiographs if ingestion suspected
20-44.9
Aggressive environmental intervention, abdominal radiographs if ingestion is
suspected. If blood lead levels persist on retest, chelation with oral succimer
(DMSA) should be considered although does not have proven efficacy in
reducing blood lead levels at these concentrations and therefore impacting
neurocognitive outcomes
45-69.9
Chelation therapy with oral succimer (DMSA) 10 mg/kg TID
5 d followed by
10 mg/kg BID
14 d. Abdominal radiograph to assess for enteral lead. If with
CNS symptoms, should treat as if lead level >70 mcg/dL. May require
hospitalization to monitor for adverse effects, institute environmental
abatement, and ensure compliance. Consider alternative regimen of CaNa2
EDTA 25 mg/kg/d for 5 d as IV infusion (continuous or intermittent) with
required inpatient administration for hydration and monitoring electrolytes.
Ensure calcium salt is given
>70
Dimercaprol (BAL) 25 mg/kg/d IM, divided Q4 hours for minimum 72 h; after
the second dose of BAL, immediately follow with CaNa2 EDTA 50 mg/kg/d
continuous IV for 5 d. Urine should be alkalinized with BAL therapy. In
addition, can cause hemolysis with G6PD deficiency and is dissolved in
peanut oil. Multiple potential side effects. BAL and EDTA cause renal
dysfunction, EDTA may also cause hypokalemia. Must give adequate
hydration and monitor electrolytes. After the initial treatment, subsequent
courses may be BAL and CaNa2 EDTA or CaNa2 EDTA alone based on repeat
lead level. Ensure calcium salt is given
Abbreviations:
CNS, central nervous system; EDTA, ethylenediaminetetraacetic acid; IV,
intravenous; IM, intramuscular.
should have a screen of potential environ-
years of age. Venous blood samples should
mental exposures by their primary health
be used to assess blood lead levels as capil-
care provider starting at 1 year of age, and
lary samples may give falsely low results. If
repeated when the child is mobile and
the screening test confirms an elevated
attains hand-to-mouth behavior. The Cen-
blood lead level, specific management
ters for Disease Control
(CDC) and the
guidelines have been developed by the CDC
American Academy of Pediatrics
(AAP)
and AAP and are summarized in Table 6.2.
recommend venous blood lead sampling for
The vast majority of children with elevated
children identified to be at high risk (living
lead levels are not candidates for chelation
in housing built prior to 1960, indigent,
therapy with currently available drugs. Chil-
urban, and minority children) at 1 and 2
dren with low levels
(<20 mcg/dL) are
Chelation Therapy
61
asymptomatic and unlikely to have signifi-
3. Ensuring adequate nutrition, especially
cant increase in lead excretion with chela-
minerals, to limit lead absorption, including
tion. These children benefit primarily from
evaluation for concomitant iron deficiency.
decreased exposure.
4. Administering medications
(chelators)
Blood lead levels measure the blood con-
in children with very high lead levels to
centration at a point in time and may not be
increase lead excretion.
able to accurately predict bone stores. Bone
lead content may be assessed noninvasively
After chelation therapy, a period of reequili-
using a radiographic technique called X-ray
bration for 10 to 14 days should be allowed,
fluorescence. Measurement of the heme pre-
prior to repeat assessment of the blood lead
cursor, free erythrocyte protoporphyrin
concentration.
Subsequent treatments
(FEP), may also provide a useful clue about
should be based on these levels, using the
the duration of exposure and the degree of
same criteria specified in Table 6.2. Ongoing
lead accumulation. Excessively high FEP
efforts should be made to provide the family
levels classically are seen with severe lead
with education in order to prevent exposure
toxicity. Other causes of elevated FEP levels
in the child’s environment in addition to
include iron deficiency, inflammatory disor-
assuring adequate nutrition. Family mem-
ders
(due to decreased iron absorption),
bers and siblings should be screened as well.
increased fetal hemoglobin, and rarely,
porphyria.
Suggested Reading
Management of lead toxicity
Bellinger DC. Very low lead exposures and chil-
The primary aims of management are pre-
dren’s neurodevelopment. Curr Opin Pediatr
20:172-177, 2008.
vention of future lead exposure and resul-
Brittenham GM. Iron-chelating therapy for
tant absorption as well as enhancement of
transfusional iron overload. N Engl J Med
excretion. The steps to accomplish these
364:146-156, 2011.
goals include:
Chandra L, Cataldo R. Lead poisoning: basics and
1. Assessment of the environment to elimi-
new developments. Pediatr Rev 31:399-406,
nate the sources of exposure or removal of the
2010.
child from the contaminated environment.
Neufeld EJ. Update on iron chelators in thalas-
2. Modifying the child’s behavior to decrease
semia. Hematology Am Soc Hematol Educ
hand-to-mouth activity.
Program. 451-455, 2010.
Approach to the
7
Bleeding Child
Hemostasis is a critical protective response
ecchymoses. Typical manifestations are pro-
of the body to reverse a loss of vascular
longed oozing from minor wounds or abra-
integrity and prevent excessive blood loss.
sions, or abnormal intraoperative bleeding.
It requires a coordinated interaction
Aberrations in secondary hemostasis are
between platelets, vascular endothelial cells,
characterized by bleeding from large vessels
and plasma clotting factors. The first and
with subcutaneous, palpable hematomas,
primary stage of hemostasis is the formation
hemarthroses, or intramuscular hemato-
of a platelet plug that involves a complex
mas. The hemophilias are examples of dis-
interaction between circulating platelets and
orders in this category.
the exposed vascular subendothelial layer.
The steps in the process include platelet
adhesion, mediated by an interaction
Evaluation of the bleeding child
between von Willebrand factor (VWF) and
platelet surface glycoprotein (Gp) Ib, and
There are three critical questions that must
platelet activation, mediated by platelet sur-
be addressed when faced with a child who is
face Gp IIb/IIIa and leading to release of
actively bleeding or has experienced a major
platelet contents. Gp IIb/IIIa interacts with
hemorrhage in the past. The first two ques-
VWF and fibrinogen, leading to platelet
tions are,
“is the patient continuing to
aggregation and enlargement of the platelet
bleed?” and “is the patient hemodynami-
plug. This lays the foundation for the for-
cally stable?” These questions should be
mation of a fibrin clot, the secondary stage
answered quickly and simultaneously.
of hemostasis caused by activation of the
Patients who are actively bleeding but are
coagulation cascade.
hemodynamically stable should have efforts
Clinical disorders associated with abnor-
directed at controlling the bleeding. While
malities of primary hemostasis include vas-
therapies that are specific for a particular
cular abnormalities, qualitative platelet
bleeding disorder should not be provided
abnormalities, quantitative platelet abnor-
before a diagnosis is made, more general
malities, and von Willebrand disease. These
strategies such as ice, pressure, and elevation
aberrations in primary hemostasis are
can be used. Patients who do not appear to
characterized by bleeding of the mucous
be actively bleeding but are hemodynami-
membranes, epistaxis, and superficial
cally unstable require rapid initiation of
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Approach to the Bleeding Child
63
vascular reexpansion and a search for occult
A family history and pedigree are crucial,
bleeding.
as many bleeding disorders are hereditary.
Once the patient is stabilized and bleed-
The history should also address the use of
ing is controlled, the third critical question
over-the-counter and prescription drugs
is whether the child presenting with bleed-
that can induce bleeding. The most com-
ing warrants an evaluation for a bleeding
mon offenders are aspirin, ibuprofen, and
disorder. Examples of excessive bleeding
naproxen. It is important to ask the patient
include epistaxis lasting more than 15 min-
specifically about the use of medications
utes despite the appropriate application of
for colds, sinus trouble, muscle aches, or
pressure to the side of the nose, significant
headaches, which may contain these medi-
blood loss following a dental procedure
cations. Some antibiotics, penicillins in
lasting more than 24 hours or requiring
particular, can affect platelet function or
blood transfusion, bruising that seems
be associated with specific inhibitors of
excessive following trauma, or menorrhagia
clotting. Anticonvulsants can cause thrombo-
(heavy menstrual bleeding lasting for 7 days
cytopenia, and procainamide has been asso-
or more, or loss of more than 80 mL of
ciated with an acquired lupus anticoagulant.
blood per cycle). When evaluating a child
for
“excessive bruising,” it is essential to
Physical examination
determine whether the bruising could be the
In addition to the routine examination, the
result of nonaccidental trauma. Bruising
skin should be scrutinized carefully for pete-
involving the scalp, back, or chest, or having
chiae, purpura, and venous telangiectasias.
a pattern suggestive of common instruments
The joints should be examined for swelling
of abuse such as belts, cords, or wire should
or chronic changes such as contractures or
be reported. Excessive bruising caused by
distorted appearance with asymmetry rela-
underlying bleeding disorders occurs on
ted to repeated bleeding episodes. Mucosal
areas more commonly involved in falls or
surfaces such as the gingiva and nares
trauma such as shins and bony prominences.
should be examined for bleeding.
History
Initial laboratory evaluation
Assessment of the child with a suspected or
The purpose of the initial laboratory eval-
known bleeding diathesis begins with a
uation is to screen for the presence of a
complete history. The nature of the bleeding
bleeding disorder, hopefully categorize the
should be explored with particular attention
disorder as primary or secondary, and direct
to location, duration, frequency, and the
further evaluation. Appropriate screening
measures necessary to stop it. The time of
tests include a complete blood count (CBC),
the patient’s first episode of bleeding should
peripheral blood smear, prothrombin time
be documented and a careful history of
(PT), and partial thromboplastin time
bruising during the toddler age is impor-
(PTT). In certain circumstances, this list
tant. A previous history of bleeding asso-
will also include a fibrinogen and a throm-
ciated with trauma or surgery, dental
bin time (TT).
extraction, circumcision, or tonsillectomy
The CBC provides several pieces of use-
is also important, as is a history of petechial
ful information. It provides the platelet
rash, arthritis with hemarthroses, or blood
count, identifying in most cases the presence
transfusion. In females, the duration and
or absence of thrombocytopenia. However,
severity of menstrual bleeding should be
it is important to review the peripheral
documented.
blood smear, as a small number of patients
64
Chapter 7
with a low measured platelet count will have
some coagulation proteins. A normal value
pseudothrombocytopenia, a condition
is 1.00 to 1.10. The INR is used to evaluate
caused by platelet clumping in the presence
the adequacy of oral anticoagulant therapy;
of the anticoagulant EDTA. It provides the
the PT should be used to look for the
hemoglobin and mean corpuscular volume
presence of a clotting factor deficiency.
(MCV), which can provide clues regarding
The PTT assesses the integrity of the
the duration and severity of the patient’s
intrinsic system of coagulation. Depending
bleeding. The presence of anemia indicates a
on the laboratory methods, the PTT will be
clinically significant bleeding disorder; the
normal when the activity of all measured
concomitant presence of microcytosis sug-
coagulation factors is at least 30% of nor-
gests the bleeding has been prolonged and
mal. It is prolonged in patients with hemo-
has led to iron deficiency. A normocytic
philia, and in some patients with VWD due
anemia suggests that the bleeding has been
to the decreased concentration of factor VIII
more recent and likely more severe, as the
in the plasma. However, the PTT is much
blood loss has likely been more significant.
more susceptible than the PT to spurious
Abnormalities in more than one cell line
abnormalities caused by errors in collection
(anemia, thrombocytopenia, and/or neutro-
or processing of the specimen. These are
penia) suggest the presence of a bone marrow
outlined in Table 7.1.
failure state such as aplastic anemia or
The most commonly encountered inhi-
marrow infiltration as is seen in leukemia.
bitors of coagulation detected in children
Many analyzers provide a measure of the
are so-called lupus anticoagulants. These are
mean platelet volume which can be useful
IgG antibodies directed against phospholi-
in evaluating the thrombocytopenic patient.
pids, and while they are commonly identi-
Large platelets are often seen in consumptive
fied in adults with autoimmune disease,
thrombocytopenia such as immune throm-
they occur frequently in children as a post-
bocytopenic purpura
(ITP), normal-sized
infectious phenomenon that is short-lived.
platelets in hypoproliferative thrombocyto-
As phospholipid is an essential cofactor in the
penia, and small platelets in inherited con-
PTT assay, antiphospholipid antibodies will
ditions such as Wiskott-Aldrich syndrome.
commonly cause prolongation of the test.
The PT evaluates the extrinsic system of
Addition of normal plasma (with additional
coagulation. Factor VII is the only coagu-
phospholipid) as is done in mixing studies
lation factor measured by the PT that is not
will often at least partially normalize the test,
measured by the PTT. Thus, in isolated
but it will most often remain abnormal.
factor VII deficiency, the PT is prolonged
These antibodies have no effect on clotting
with a normal PTT. Factor VII is vitamin K
in vivo, and patients with lupus anticoagu-
dependent and has a very short half-life, so
lants do not have clinical bleeding; in fact,
the PT is one of the most sensitive measures
their most common coagulation problem is
of oral anticoagulant therapy with vitamin
thrombophilia rather than bleeding.
K antagonists such as warfarin. Most labo-
The TT is useful in evaluating the ter-
ratories now report an international nor-
minal steps of coagulation and identifying
malized ratio (INR) along with the PT. The
anticoagulants present in plasma. The TT is
INR is calculated as the patient PT/control
abnormal when the plasma concentration of
PT to the power of the international sensi-
fibrinogen is decreased
(hypofibrino-
tivity index. This index corrects for the large
genemia or afibrinogenemia), when the
variation in the sensitivity of thromboplas-
fibrinogen present is dysfunctional (dysfi-
tin reagents to low plasma concentrations of
brinogenemia), or when there are
Approach to the Bleeding Child
65
Table 7.1 Factors affecting the validity of the partial thromboplastin time (PTT) test.
Aspect
Problem
Remarks
Sample
Clots
Clots caused by slow blood flow or delay in transferring
collection
sample will cause abnormal results
Plasma volume
Volume of anticoagulant must be corrected for plasma
volume. If tube is not completely filled, or patient is
extremely anemic or polycythemic, sample will not
be appropriately anticoagulated
Heparin contamination
Very small amounts of heparin will cause the PTT to be
abnormal
Interpretation
Age-dependent normal
Mild prolongation is normal in the newborn period
values
(immaturity of the coagulation system)
Inhibitor vs. factor
A number of agents can interfere with the PTT assay
deficiency
and cause a factitious prolongation of the test. If the
concentration is high they can lead to a mild pro-
longation of the PT as well. These are diagnosed by
mixing the patient’s plasma and normal plasma in a
1:1 ratio and repeating the test. If the PTT is
prolonged due to factor deficiency, this maneuver
will increase the level of all coagulation factors to at
least 50% and normalize the test. If the prolongation
is due to an inhibitor, it may improve but will
remain prolonged. See the discussion of inhibitors
Abbreviations: PTT, partial thromboplatin time; PT, prothrombin time.
circulating anticoagulants
(heparin) or
chapters. Two exceptions are management
fibrin degradation products. As dysfibrino-
of epistaxis and menorrhagia. These types
genemia is most often asymptomatic, a
of bleeding can be caused by a variety of
prolonged TT in the face of a normal fibrin-
different conditions; yet in most situations,
ogen level is almost always associated with
the following management plans will be
heparin or an inhibitor.
effective. See also Figure 7.1.
Abnormalities in one or more of the
above screening tests will be noted with
Management of epistaxis
most bleeding disorders. An approach to
1. Place patient in a sitting position to
identifying the most likely coagulation dis-
decrease venous pressure, or if the patient
orders given the results of these tests is
is recumbent in bed, turn head to the side.
presented in Table 7.2.
Keep the head higher than the level of the
heart. Do not allow patient to lie flat.
2. Flex neck anteriorly, with the chin touch-
Management
ing the chest.
3. With the thumb and index finger, pinch
Once the diagnosis is made, specific treat-
the soft parts of the nose. Hold pressure
ment can be provided based on the recom-
firmly over the lower half of the nose. Avoid
mendations outlined in the following
compressing the upper half of the nose.
66
Chapter 7
Table 7.2 Interpretation of screening coagulation tests.
Test results
Differential diagnosis
Follow-up laboratory studies
PT normal
Von Willebrand Disease
PFA-100
PTT normal
Platelet function disorder
Von Willebrand studies
Platelet count
Factor XIII deficiency
Platelet aggregation studies
normal
Fibrinolytic defect
Urea clot lysis test
Euglobulin clot lysis
Factor XIII assay
Alpha-2-antiplasmin, PAI-1, and TPA
PT normal
PTT inhibitor
PTT mixing study
PTT prolonged
Von Willebrand disease
Factor assays (VIII, IX, and XI)
Platelet count
Hemophilia A or
Von Willebrand studies
normal
Hemophilia B
TT/reptilase time
Heparin contamination
PT prolonged
PT inhibitor
PT mixing study
PTT normal
Vitamin K deficiency
Factor assays (II, VII, IX, and X)
Platelet count
Warfarin
normal
Factor VII deficiency
PT prolonged
Circulating inhibitor
PT/PTT mixing studies
PTT prolonged
Liver dysfunction
TT/reptilase time
Platelet count
Vitamin K deficiency
Fibrinogen
normal
Factor deficiency (II, V, X,
Factor assays
or fibrinogen)
Dysfibrinogenemia
PT prolonged
DIC
TT
PTT prolonged
Liver disease
Fibrinogen
Platelet count low
Kasabach-Merritt syndrome
Factor assays
D-dimers
PT normal
Acute ITP
None
PTT normal
Chronic ITP
Antinuclear antibodies
Platelet count low
Collagen vascular disease
Anticardiolipin antibodies
Early bone marrow failure
Direct antiglobulin test (Coombs)
syndrome
Serum immunoglobulin levels
Serum complement levels
HIV and hepatitis C antibody testing
Bone marrow aspirate
Marrow chromosomal analysis
Abbreviations: PT,
prothrombin time; PTT, partial
thromboplastin time; PFA-100, platelet
function analyzer-100; PAI-1, plasminogen activator
inibitor-1; TPA, tissue plasminogen
activator; TT, thrombin time; DIC, disseminated intravascular coagulation; ITP, immune
thrombocytopenic purpura.
Approach to the Bleeding Child
67
Menorrhagia : >80 mL blood loss/cycle or heavy, regular periods >7 days duration with pad/tampon use
>1 per hour or passing >1 inch wide clots
and
Diagnosis or suspicion of von Willebrand disease or other bleeding disorder: type and severity may influence
nature of bleeding and intervention
Assessment:
1. History: categorize bleeding pattern by duration, regularity, quantification of bleeding
2. Medication History: evaluate for drugs associated with bleeding including warfarin, heparin,
salicylates, non-steroidal anti-inflammatory drugs, phenytoin, antipsychotics (SSRIs) and tricyclic anti-
depressants, prolonged antibiotic use (low vitamin K), herbal supplements
3. Physical: evidence of bleeding disorder or gynecologic problem
4. Laboratory: assess for degree of anemia and red cell indices with CBC, urine βHCG for pregnancy
screen, consider STI (gonorrhea, chlamydia with bleeding and use of OCPs)
5. Imaging: assess for anatomic etiology with pelvic ultrasound to look for fibroids, polyps,
endometriosis, pregnancy complications, ovarian cysts, endometrial cancer
Management
Active, heavy bleeding and anemia
Routine Management (listed in order of preference,
1. Avoid D&C, may worsen bleeding
patients may require 1 or more interventions):
2. Treat with high dose estrogen PO or IV,
1. OCP*: combined oral contraceptive, minimum
e.g., estrogen 30 mcg/norgestrel 0.3 mg
3 month trial (synthetic estrogen + progestin);
(LoOvral):
1 pill QID x 4 days, then
consider no withdrawal week for 1-3 months;
TID x 3 days, then BID x 2 days, then
consider increase in dose if breakthrough
daily for 3 weeks
bleeding or persistence of menorrhagia after
3. Consider desmopressin or factor
3 months
concentrate
2. Desmopressin: intranasal at initiation of
4. If controlled, follow routine
menses, could give second dose at 24 hours
management
3. Antifibrinolytic therapy with initiation of
menses: tranexamic acid or aminocaproic acid
4. Intrauterine device impregnated with
progestin (Mirena), may cause bleeding with
*Estrogen containing therapies may not be safe
insertion and require preventive intervention
for women who smoke or have known
5. Avoid medications that may cause or
thrombophilia; may increase coagulation proteins
exacerbate bleeding
Figure 7.1 Menorrhagia in young women with bleeding diathesis: clinical assessment and manage-
ment strategies. Abbreviations: SSRI, selective serotonin reuptake inhibitors; CBC, complete blood
count; STI, sexually transmitted infection; OCP, oral contraceptive pill, D&C, dilatation and curettage.
4. Hold pressure for 20 minutes with the
clot(s) can be visualized, remove clot(s) and
head in a flexed position. If manual pressure
reapply digital pressure for 20 minutes.
is stopped momentarily to examine or
5. Advise patient not to blow nose for at
change dressings, the
20
minute digital
least 12 hours to avoid dislodging the clot.
pressure will likely need to start again. Pres-
6. Nasal packing may be indicated if the
sure and time allow for clot formation to
source of bleeding is not well visualized or
occur. If bleeding continues, reassess loca-
bleeding is profuse. Types of packing include
tion of digital pressure and reapply. If bleed-
compressed sponge, Vaseline gauze packing,
ing stops and recurs, repeat manual pressure
gelfoam, or topical thrombin packing. Com-
for 20 minutes. If bleeding continues and
pressed sponges are compressed when dry and
68
Chapter 7
expand when wet. The expansion produces
predicting menstrual blood loss of >80 mL
active and passive absorption and places gen-
include the passing of clots >1 inch in diam-
tle pressure on the mucosa. The nasal sponge
eter, a low serum ferritin, or changing a pad
should fit snugly through the nare and be
or tampon more often than hourly.
placed along the floor of the nasal cavity.
2. The most effective management of men-
Sponges are easy to insert and can be removed
orrhagia is prevention. This can often be
with little discomfort. Neosporin can be
accomplished with hormonal therapy. The
applied to the sponge for ease of insertion
most common approach is the use of com-
and to act as an antimicrobial agent. Topical
bination estrogen-progestin oral contracep-
thrombin powder (a vasoconstrictor) applied
tive pills (COCP), both cyclic and extended
to the inserted end of the sponge can provide
cycle use. Regular COCPs can been prescribed
additional hemostasis. Anterior packs may be
in an extended cycle pattern, with the patient
left in place for 1 to 5 days, though should be
taking 63 to 84 days of active pills before
removed within 24 hours in an immunocom-
stopping for
7 days to allow withdrawal
promised patient due to the risk of infection.
bleeding. Young women often prefer this
Humidification and nasal saline spray can help
approach despite the occasional breakthrough
prevent drying and crusting of the oral
bleeding that may occur. Table 7.3 lists COCP
mucous membranes as a result of mouth
preparations that have been used successfully
bleeding. Broad-spectrum antibiotics, to
in managing menorrhagia in women with
cover skin flora as well, should be considered
bleeding disorders. There are no data that
in patients who are immunosuppressed with a
indicate that any of these agents are superior
nasal pack in place. Vaseline gauze packing,
to the others in controlling bleeding.
when placed correctly and snugly, is a reliable
3. Other hormonal approaches to prevent
means of packing. Packing may be soaked in
menorrhagia include the levonorgestrel
4% topical cocaine or a solution of 4% lido-
intrauterine system (Mirena), oral progesto-
caine and topical epinephrine (1:1000) to
gens, and injected progestogens. Mirena has
provide local anesthesia and vasoconstriction.
been shown to be as efficacious as endome-
trial ablation in decreasing the incidence and
Management of menorrhagia
severity of menorrhagia. It has also been
1. The American College of Obstetricians
shown in one small study to be well tolerated
and Gynecologists and the American Acad-
by nulliparous adolescent females. Oral
emy of Pediatrics issued a committee con-
progestin-only OCPs (Micronor, Nor-QD)
sensus report in 2006 in which they defined
contain norethindrone as the active agent.
normal menstruation. This report stated that
The progestin implant (Implanon) contains
normal menstruation begins at 11 to 14 years
etonogestrel. Depo-Provera is an injectable
of age, the normal cycle interval is 21 to 45
progestin only contraceptive containing
days, and the normal length of menstrual
medroxyprogesterone as the active agent.
flow is 7 days or less with product use no
4. Young women presenting with the acute
more than three to six pads or tampons per
complaint of menorrhagia should have a
day. Based on this, menorrhagia has been
pregnancy test performed to ensure that
defined as heavy menstrual bleeding lasting
they are not having a miscarriage. They
for more than 7 days or resulting in the loss of
should also be carefully evaluated to rule
more than 80 mL per menstrual cycle. How-
out the presence of severe anemia or hypo-
ever, attempts to quantify menstrual blood
volemia. If they are hemodynamically stable,
loss in clinical practice can be quite difficult.
have no prior personal or family history of
Variables that have been identified as
abnormal bleeding, and are not allergic to
Approach to the Bleeding Child
69
Table 7.3 Preparations of combination oral contraceptive pills used to treat menorrhagia in women
with bleeding disorders.
Hormone content
Brand name
150 mcg desorgestrel/30 mcg ethinyl estradiol
Apri
Cyclessa
Desogen
Ortho-Cept
Reclipsen
Velievet
150 mcg levonorgestrel/30 mcg ethinyl estradiol
Enpresse
Jolessa
Levora
Lutera
Nordette
Portia
Quansense
250 mcg norgestimate/35 mcg ethinyl estradiol
MonoNessa
Ortho-Cyclen
Previfem
Sprintec
3 mg drospirenone/30 mcg ethinyl estradiol
Ocella
Yasmin
nonsteroidal anti-inflammatory drugs
treat menorrhagia in women both with and
(NSAIDs), ibuprofen 200 mg every 4 to 6
without coagulopathies.
hours can be tried. NSAIDs have been
7. Women with inherited coagulopathies
shown to decrease the amount of menstrual
(Type 1 VWD, hemophilia carriers, platelet
flow by 25% to 30% by altering the endo-
function disorders) should ideally be tested for
metrial prostaglandin balance. However,
responsiveness to DDAVP prior to menarche.
they are contraindicated in women with
If they are responsive, this should be used as
known or suspected coagulopathy.
first-line therapy for menorrhagia. It can be
5.
“Double-dose” COCPs can be used to
given IV at a dose of 0.3 mcg/kg in 50 mL
treat young women presenting with acute
normal saline over 30 minutes. A concentrated
menorrhagia as well. Any of the COCPs in
nasal preparation is also available (Stimate)
Table 7.3 containing 35 mcg of ethinyl estra-
containing 150 mcg/spray; it is given at a dose
diol/tablet can be prescribed twice a day for
of one spray in each nostril once daily. Either
up to 7 days until the bleeding stops, after
preparation can be repeated daily for up to 3 to
which the remainder of the pack is com-
4 days during the heaviest menstrual flow. It
pleted on a once daily schedule.
should be used in conjunction with tranexa-
6. If these steps are not effective or the
mic acid if the bleeding is heavy. Side effects
bleeding is more significant, tranexamic
include flushing, headache, nausea, fluid
acid can be added to any of these treatments
retention, and rarely, hyponatremia.
at a dose of 1300 mg (two tablets) three
8. Women with persistent bleeding despite
times daily for up to 5 days. This antifibri-
these therapies should be hospitalized for
nolytic agent has been successfully used to
more aggressive therapy, including
70
Chapter 7
intravenous estrogen. Consultation with a
The presence of petechiae and increased
gynecologist or adolescent medicine physi-
superficial bruising suggests a disorder of
cian well versed in the management of this
primary hemostasis. The lack of a previous
disorder is recommended.
history and a negative family history suggest
this is more likely to be an acute problem
rather than a congenital one, although this
Case study for review
is not certain. Acquired problems of primary
hemostasis include acquired (often immune-
A 4-year-old girl presents to the emergency
mediated) thrombocytopenia or acquired
department with a 3 day history of easy
(often drug-induced) platelet function
bruising and “rash.” She denies any other
abnormalities. This presentation could also
bleeding including epistaxis, oral bleeding,
result from a vasculitis, although most
hematuria, or hematochezia. She has oth-
patients with such a problem would likely
erwise been well except for an upper respi-
appear more ill. The best next steps would be
ratory infection 2 weeks ago.
to obtain a CBC and coagulation studies to
see if you can identify where an abnormality
1. What other information would be help-
might be. The CBC reveals a hemoglobin of
ful in evaluating this child?
12.3 g/dL, a WBC count of 6.5
109/L with
The girl’s parents deny any previous history
a normal differential, and a platelet count
of easy bruising, hemarthroses, or muscle
of 6
109/L. The PT is 12.2 seconds (refer-
bleeding in the past. The family history is
ence range, 12.6 to 13.5 s) and the PTT is
significant for two older brothers who have
30 seconds (range, 26 to 33 s).
never had any bleeding abnormalities. There
4. What do you think now, and what is
is no history of epistaxis, gingival bleeding,
your next step?
menorrhagia, or excessive bleeding with sur-
gery or trauma in any other family members.
The patient has isolated thrombocytopenia
There is no family history of malignancy or
with a normal hemoglobin and platelet
autoimmune disorders. The patient has not
count, and no evidence of a clotting factor
been taking any medications recently.
deficiency. The recent history of a viral
illness is very consistent with ITP. The
2. What will you be looking for on physical
absence of any clinically significant bleeding
examination?
at this point means that no immediate inter-
The vital signs are normal, and the patient is
vention is necessary. The management of
at the 50th percentile for height and weight.
ITP is discussed more fully in Chapter 12.
She appears completely healthy. There are
no signs of oral bleeding or purpura. Her
Suggested Reading
heart and lung exam are normal, and she has
no tenderness, masses, or hepatosplenome-
Allen GA, Glader B. Approach to the bleeding
galy on abdominal exam. You note a diffuse
child. Pediatr Clin N Am 49:1239-1256, 2002.
petechial rash over her entire body, with
Hayward CP. Diagnostic approach to platelet
several ecchymoses on her shins and arms,
function disorders. Trans Apheresis Sci
with a few on her back. Her neurological
38:65-76, 2008
exam is completely normal.
Ahuja SP, Hertweck SP. Overview of bleeding
disorders in adolescent females with menor-
3. What are your first thoughts, and what
rhagia. J Pediatr Adolesc Gynecol 23:S15-S21,
would you like to do next?
2010.
Von Willebrand
8
Disease
Von Willebrand disease (VWD) is the most
plasma. This accounts for the prolonged
common inherited bleeding disorder,
partial thromboplastin time (PTT) seen in
affecting as much as
1% of the general
some patients.
population, and equally affects both genders
VWD is classified into three types:
as well as all races and ethnicities. The actual
Type 1, found in 70% to 80% of cases, is
prevalence is difficult to determine, as many
characterized by partial deficiency of nor-
affected individuals are either asymptomatic
mally functioning VWF. Type 2 is charac-
or have such mild symptoms that they do
terized by functional defects in VWF. It is
not seek medical attention. While most
divided into four subtypes. Type 2A is char-
patients with VWD have the inherited form,
acterized by a decreased number of high-
an acquired form also occurs. The three
molecular-weight multimers and a concom-
characteristic clinical features of the inher-
itant decrease in platelet adhesion. The
ited form are excessive mucocutaneous
measured amount of VWF:Ag (antigen) is
bleeding, abnormal von Willebrand factor
normal or only slightly decreased, but the
(VWF) laboratory studies, and a family
VWF:RCo (ristocetin cofactor), a measure
history of abnormal bleeding.
of VWF activity, is much lower. Type 2B is
VWF is a large multimeric glycoprotein
caused by mutations that pathologically
that is synthesized in megakaryocytes and
increase platelet-VWF binding, which leads
endothelial cells. VWD is usually inherited
to the proteolytic degradation and depletion
in an autosomal dominant manner, but a
of large, functional VWF multimers. Circu-
rare autosomal recessive form and an X-
lating platelets are also coated with mutant
linked recessive form have also been
VWF, which may prevent the platelets from
described. VWD results from either a defi-
adhering at sites of injury. The VWF-coated
ciency or a defect in the VWF protein. This
platelets often become sequestered in the
protein plays two critical roles in hemosta-
microcirculation, leading to thrombo-
sis: the large multimers bind to platelet
cytopenia. Type
2M is characterized by
glycoprotein Ib (GPIb) causing platelet acti-
decreased interaction between VWF and
vation and adherence to damaged endothe-
platelet GPIb on connective tissue. The ratio
lium, and it is the carrier protein for factor
of VWF:RCo to VWF:Ag is low as in Type 2A,
VIII (FVIII), stabilizing it and protecting
but the multimer panel appears normal.
it from degradation and clearance from
Type 2N is marked by decreased binding
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
72
Chapter 8
between VWF and FVIII, resulting in a phe-
themselves with a safety razor) can be more
notype of autosomal recessive hemophilia.
informative. Often, the child is diagnosed
Type 3, a rare condition, is characterized by
with VWD and then a parent or sibling is
the almost complete absence of VWF.
found to have the disorder after screening.
The majority of affected patients expe-
In patients with mild VWD, the stress
rience mucocutaneous bleeding such as epi-
associated with serious operations or child-
staxis, easy bruising, and menorrhagia in
birth may prevent symptoms. VWF is an
women. They may also experience posttrau-
acute phase reactant and the level will fre-
matic or postsurgical bleeding. Patients with
quently increase into the normal range fol-
severe VWD may experience bleeding into
lowing surgery, in pregnancy, or in patients
muscles and joints as well. VWD should be
with active liver disease or collagen vascular
suspected in the patient with platelet-type
disease. It can be reduced in hypothyroid-
bleeding and a family history of a bleeding
ism. Even the stress of phlebotomy can
diathesis.
increase the level of VWF, which can
make it difficult to confirm the diagnosis
even with repeat testing. Neonates have
Clinical presentation
elevated VWF levels following vaginal deliv-
ery, making it difficult to diagnose in the
The clinical features of VWD can be quite
neonatal period. VWF levels can be up to
variable. In most cases of VWD, the bleed-
25% lower in individuals who are blood
ing is mild. Mucosal bleeding, such as epi-
type O, increasing the difficulty in distin-
staxis, gingival bleeding with tooth brush-
guishing between VWD and normal indivi-
ing, ecchymoses, or menorrhagia, are clas-
duals with a low VWF level.
sic. The initial presentation, however, may
be postoperative bleeding, such as following
a tonsillectomy and adenoidectomy or den-
Diagnosis
tal extraction. In retrospect, the child who
was thought to have normal childhood
Screening tests are of little value in making
complaints (bruising and epistaxis) is real-
the diagnosis of VWD. The complete blood
ized to actually have a bleeding disorder.
count (CBC) may demonstrate iron defi-
Recurrent or severe epistaxis, particularly in
ciency anemia if the patient’s blood loss has
the older child or adult, is unusual and
been significant, but it is nonspecific. The
warrants investigation. It is important to
rare patient with Type 2B VWD may have
obtain a thorough medical history of bleed-
mild thrombocytopenia. If the level of VWF
ing from the family as this may provide
is sufficiently low, the PTT may be prolonged
clues. Since other affected family members
due to decreased concentration of FVIII, but
are often unaware that they have the condi-
this is neither sensitive nor specific. The
tion, simply inquiring about a history of
PFA-100 platelet function analyzer is a more
bleeding problems often yields negative
useful screening test for VWD. It measures
results. Specific questions such as “Have any
the time required for blood, drawn through a
women in the family had hysterectomies
fine capillary, to block a membrane coated
during their child-bearing years because of
with collagen and epinephrine or collagen
intractable uterine bleeding?” or “Do any
and ADP. Its sensitivity in identifying
men in the family insist on shaving with
patients with VWD is fairly high, but its
an electric razor?” (because of the prolonged
specificity is low. Four specific tests are used
oozing they experience if they nick
to diagnose VWD:
Von Willebrand Disease
73
1. Factor VIII coagulant (FVIIIc): func-
VWD. The National Heart, Lung, and Blood
tional measurement of FVIII coagulant
Institute (NHLBI) of the National Institutes
activity, which is carried in the circulation
of Health published an algorithm, presented
by VWF.
in Figure 8.1, to assist in identifying affected
2. Von Willebrand factor antigen (VWF:
individuals while at the same time avoiding
Ag): VWF protein as measured by protein
overdiagnosis. Except for patients with
assays; does not imply functional ability.
clearly abnormal VWF levels (<30%), the
Immunologic quantitation of VWF by
diagnosis should be made after consultation
either the quantitative immunoelectropho-
with a hematologist.
retic assay (Laurell assay) or the enzyme-
linked immunosorbent assay.
3. Von Willebrand factor activity (VWF:
Treatment
RCo): ristocetin induces the binding of
VWF to the GPIb receptor on formalin-
The goal of treatment in VWD is to control
fixed platelets. The slope of the platelet
or prevent serious or life-threatening bleed-
agglutination curve correlates with the level
ing. There are no effective therapies to limit
of plasma VWF.
the chronic bruising that many active chil-
4. Multimeric analysis: an agarose gel elec-
dren experience, and parents need to be
trophoretic study used to identify quantita-
given assistance to accept that reality. Many
tive or qualitative multimer abnormalities.
children and adults with Type
1 VWD,
The most common type of VWD (Type 1)
especially males, never require therapy.
has a normal multimeric analysis.
Individuals with Type 2 or Type 3 VWD
are much more likely to need treatment at
The classification of patients with VWD
various times throughout life.
based on the results of these tests is pre-
Three strategies are used to treat patients
sented in Table 8.1.
with VWD: increase the plasma concentra-
Even with these test results, it can be
tion of VWF by releasing endogenous VWF
difficult to determine whether a child has
stores through stimulation of endothelial
VWD or not, especially in mild cases. Unlike
cells with desmopressin, replace VWF by
hemophilia, there are not clearly defined
using human plasma-derived, viral-inacti-
levels of VWF antigen or activity that sep-
vated concentrates, and use agents that pro-
arate normal patients from those with
mote hemostasis and wound healing but do
Table 8.1 Classification of von Willebrand disease.
VWD subtype VWF:Ag VWF:RCo FVIII:C RCo:Ag ratio (%) Multimer pattern
1
#
#
# or N
>60
Normal
2A
#
##
# or N
<60
Abnormal
2B
#
##
# or N
<60
Abnormal
2M
#
##
# or N
<60
Normal
2N
# or N
# or N
10-40%
>60
Normal
3
###
###
<10%
-
-
Abbreviations: VWD, von Willebrand disease; VWF:Ag, von Willebrand factor antigen; VWF:
RCo, von Willebrand factor ristocetin cofactor activity; FVIII:C, Factor VIII coagulant; N, normal.
74
Chapter 8
Von Willebrand Disease
75
not substantially alter the plasma concen-
other measures. Given its high cost and
tration of VWF. These strategies are not
relatively short shelf life (6 months after
mutually exclusive; different bleeding epi-
the bottle is opened), it is less suitable for
sodes may require a combination of two or
individuals with very infrequent bleeding
even all three of these strategies to be
episodes. Although effective, there is some
effective.
variability in response due to inconsistent
Desmopressin (DDAVP) is the treat-
absorption. Therefore, patients should be
ment of choice in mild Type
1 disease
tested for responsiveness prior to prescrib-
(65% to 80% of all cases) and may be of
ing. The dose for children weighing less
benefit with some of the other variants. It
than 50 kg is one spray (150 mcg) once
stimulates endogenous release of VWF from
daily. The effective dose in adolescents and
endothelial cells, with most patients
adults is one spray in each nostril (total
experiencing a two- to fourfold increase
dose of
300 mcg) once daily. The peak
in their plasma VWF and FVIII levels within
effect is observed 60 to 90 minutes after
15
to
30
minutes of the infusion. The
administration.
approximate half-life of these released fac-
Patients undergoing surgical procedures
tors is 8 to 10 hours. Occasionally, patients
such as tonsillectomy or dental extractions
do not respond to this therapy (especially
may need extended therapy to maintain
children younger than 18 months of age).
increased levels of VWF and FVIII until
The peak effect is observed in 30 to 60
healing has occurred. Studies have demon-
minutes. A therapeutic trial of DDAVP
strated that DDAVP can be repeated daily
should be given to determine individual
for up to 4 days with little loss of efficacy as
responsiveness to this therapy with mea-
measured by VWF levels or PFA-100. Pre-
surement of FVIII and VWF levels before
and postinfusion measurements of VWF:Ag
and after administration. After baseline
or VWF:RCo can be done to establish con-
levels are obtained, a standard dose of
tinued clinical responsiveness. However, it
0.3 mcg/kg is administered intravenously
may be difficult to obtain the results of these
in 30 to 50 mL of normal saline over 15
tests quickly.
to 30 minutes, and levels are measured again
Patients with Type 1 VWD who have
60 minutes after completion of the infusion.
severe bleeding manifestations unrespon-
A positive trial is defined as at least a twofold
sive to DDAVP or those patients with
increase in the VWF:Ag and VWF:RCo
desmopressin-unresponsive Type
2
or
3
activity. Ninety percent of patients with
disease require treatment with exogenous
Type
1
VWD demonstrate a positive
VWF. Unlike FVIII replacement products,
response to intravenous (IV) DDAVP.
there are no recombinant VWF products yet
A unique intranasal form of DDAVP
in clinical use, although one product is being
(Stimate, 1500 mcg/mL) is also available to
evaluated in animal models. The FDA has
treat bleeding in VWD. A more dilute form
approved three plasma-derived FVIII/VWF
of DDAVP (100 mcg/mL) is approved for
products, Humate-P, Wilate, and Alphanate
use in patients with diabetes insipidus but
for the treatment of VWD. A fourth plasma-
should not be prescribed for VWD, as it is
derived product, Koate DVI, is licensed in
impossible to deliver an adequate dose with
the United States for the treatment of hemo-
this preparation. Stimate is most useful for
philia and is used off-label to treat VWD.
individuals with more frequent episodes of
These agents are not identical, and have
bleeding requiring therapy, such as women
differing ratios of FVIII to VWF, so should
with menorrhagia not controlled using
not be considered interchangeable.
76
Chapter 8
Humate-P contains 50 to 100 IU/mL VWF:
amount of endometrial development that
RCo activity and 20 to
40 IU/mL FVIII
takes place. Minor surgical procedures such
activity. It has the highest ratio of VWF:
as laceration repairs or dental extractions
RCo to FVIII activity and the highest con-
can frequently be managed with a single
centration of high-molecular-weight multi-
treatment of desmopressin or factor con-
mers. Alphanate contains 40 to 180 IU/mL
centrate followed by antifibrinolytic therapy
FVIII activity and at least 16 IU/mL VWF:
for 7 to 10 days. Two antifibrinolytic agents
RCo activity. The FDA approved Wilate, the
are available, epsilon aminocaproic acid
most recently licensed product, in 2010. It
(Amicar) and tranexamic acid; Amicar is
was approved specifically for use in VWD,
the more widely used agent. The oral dose of
and has essentially equal amounts of VWF:
aminocaproic acid is
100
to
200 mg/kg
RCo and FVIII activity. It has demonstrated
(maximum dose, 10 g) as the initial dose,
both safety and efficacy in patients with
followed by
100 mg/kg (maximum dose,
VWD. There are no head-to-head studies
5 g) every 6 hours. To maintain effective-
comparing the efficacy of any of these pro-
ness, families must be instructed to adhere
ducts. Wilate’s packaging promotes more
to the every 6 hour schedule. The dose of
frequent dosing with lower doses of prod-
tranexamic acid is 25 mg/kg/dose every 6 to
uct, which the company states is more
8 hours. Both drugs are available in oral and
physiological and more cost-effective.
intravenous forms. These drugs are partic-
The goal of factor replacement therapy is
ularly effective in minimizing bleeding from
to reach a therapeutic level of VWF:RCo
mucosal surfaces due to the increased level
of
100 IU/mL and nadir of
>50 IU/mL.
of fibrinolytic activity present in these areas.
Recommended starting doses are
40
to
Patients with Type 3 VWD have plasma
60 IU/kg of VWF:RCo activity, followed
VWF and FVIII levels that are extremely
by 20 to 40 IU/kg every 8 to 24 hours as
low (typically less than 5 IU/dL). Desmo-
needed to maintain desired levels. Major
pressin is usually ineffective in these cases,
surgical procedures should only be per-
but a therapeutic trial of desmopressin may
formed at centers that have the capability
be given, as occasionally patients will
of measuring VWF activity levels in-house
respondappropriately.These patients often
on a daily basis. To decrease the risk of
have significant bleeding including pro-
thrombotic events, VWF:RCo levels should
found epistaxis or hemarthroses similar
be <200 IU/mL, and FVIII levels should be
to that seen in patients with hemophilia
<250 to 300 IU/mL.
due to their low levels of both FVIII and
Other adjuvant therapies are available
VWF. With bleeding, such patients should
that are often helpful in managing bleeding
be managed similarly to patients with
in patients with VWD. Direct pressure is
hemophilia, using the previously men-
very effective in managing epistaxis and
tioned FVIII concentrates that contain
bleeding from tooth sockets or lacerations.
adequate levels of VWF and FVIII.
Family members must be taught to main-
Patients with Type 2A VWD have abnor-
tain continuous pressure “without peeking”
mally small VWF multimers, and desmo-
for at least 20 minutes for best results. If the
pressin, though frequently effective, often
bleeding remains uncontrolled, a single dose
has only a transient effect. Serious bleeding
of desmopressin is usually effective to stop
should be treated with plasma-derived
bleeding in patients who have shown
FVIII/VWF concentrates. These patients
responsiveness. Low-estrogen oral contra-
may do well with desmopressin for the
ceptives have been very helpful in control-
treatment of minor bleeds or dental
ling menorrhagia as they minimize the
extractions.
Von Willebrand Disease
77
Type 2B VWD is often associated with
thrombocytopenic purpura (ITP), lympho-
mild thrombocytopenia due to excessive
proliferative diseases, systemic lupus erythe-
platelet binding to an abnormal VWF
matosus, other autoimmune disorders, and
molecule and rapid clearance. Stress can
some cancers, most commonly Wilms
exacerbate the thrombocytopenia. Desmo-
tumor. Pathologic increases in fluid shear
pressin is usually contraindicated due to
stress can occur with cardiovascular lesions
the potential for worsening thrombocyto-
such as ventricular septal defect and aortic
penia and failure to demonstrate a thera-
stenosis, or with primary pulmonary hyper-
peutic response. However, in mild bleeding,
tension. This leads to increased destruction
desmopressin may be effective. For internal
of VWF, particularly the larger multimers;
bleeding or surgery, patients should receive
the multimer panel often resembles that seen
FVIII/VWF concentrate. If profound throm-
in Type 2A VWD. Its occurrence in Wilms
bocytopenia exists, concomitant administra-
tumor and other malignancies is believed to
tion of platelets may be necessary.
be due to expression of platelet GPIb on the
Patients with 2M VWD will demonstrate
tumor cell surface, leading to binding of
an increase in VWF:Ag but not in VWF:RCo
VWF to the tumor with subsequent degra-
in response to DDAVP, due to the defective
dation. Hyaluronic acid secretion by nephro-
binding of their VWF to GPIb. These
blastoma cells is another potential mecha-
patients routinely require replacement with
nism in Wilms tumor patients leading to
FVIII/VWF concentrates for bleeding epi-
decreased efficacy of VWF. Patients may
sodes or for surgery. Patients with Type 2N
present with classic signs and symptoms of
VWD similarly require factor replacement;
VWD and their levels of VWF, VWF:Ag, and
although they produce normal amounts of
FVIII are typically quite low.
FVIII, it is destroyed prematurely due to the
Desmopressin may induce a transient rise
inability of their VWF to bind to the FVIII
in VWF in AVWS. One small study demon-
molecule and protect it. Replacement with
strated that intravenous immunoglobulin
functional VWF will typically normalize their
(IVIG) at a dose of 1 g/kg/d for 2 days was
FVIII levels even if the relative concentration
beneficial in improving VWF levels and
of FVIII in the product is low since they are
decreasing clinical bleeding, but this is only
able to produce normal amounts.
beneficial in immune-mediated AVWS. Plas-
mapheresis, corticosteroids, and immuno-
suppressive agents have also been successful
Acquired von Willebrand
in these patients. For serious bleeding, treat-
syndrome
ment with factor concentrate (high VWF
content) should be given. The antibodies
Acquired von Willebrand syndrome
typically disappear with control or resolution
(AVWS) refers to defects in VWF concen-
of the underlying disease.
tration, structure, or function that are not
inherited directly but are consequences of
other medical disorders. It is usually caused
Other considerations
by one of three mechanisms: autoimmune
clearance or inhibition of VWF, increased
As a general precaution, all children with
shear-induced proteolysis of VWF, or
bleeding disorders should be immunized with
increased binding of VWF to platelets or
the hepatitis B vaccine, with boosters given at
other cell surfaces. Autoimmune causes of
appropriate time intervals, since these chil-
AVWS in children include postviral anti-
dren have a lifelong potential for receiving
body production similar to immune
blood products. Patients and families should
78
Chapter 8
be counseled to avoid aspirin, NSAIDs, and
Suggested Reading
other platelet-inhibiting drugs. Also, it is
generally recommended that children wear
Nichols WL, Hultin MB, James AH, et al. Von
MedicAlert bracelets and have information
Willebrand disease: evidence based diagnosis
and management guidelines, the National
readily available (at school, home, and doc-
Heart, Lung, and Blood Institute Expert Panel
tor’s office) specifying the diagnosis and
report. Haemophilia 14:171-232, 2008.
treatment for bleeding.
Hemophilia
9
Hemophilia is defined as an inherited bleed-
50%). The frequency of bleeding symptoms
ing disorder caused by low levels, or
generally correlates well with the measured
absence, of a blood protein that is essential
residual factor activity. Boys with severe
for clotting. The congenital hemophilias are
hemophilia bleed with minimal trauma and
uncommon disorders, with a total incidence
bleeding symptoms are usually apparent by
of 10 to 20 per 100,000 births. The most
the time the infant begins to crawl or walk,
common form is factor VIII (FVIII) defi-
whereas those with mild hemophilia usually
ciency
(hemophilia A or classic hemo-
bleed only with significant trauma or surgery
philia), with an estimated incidence of 1
and may have a delayed diagnosis.
in 5000 males. The second most common
Several other congenital factor deficien-
form is factor IX (FIX) deficiency (hemo-
cies have been described as well, but are all
philia B), with an incidence of approxi-
very uncommon. They are described in
mately 1 in 25,000 males. Both the FVIII
Table 9.1.
and FIX genes are carried on the X chro-
Afibrinogenemia is associated with fre-
mosome; thus, both hemophilia A and B are
quent and serious bleeding. Spontaneous
X-linked recessive disorders. Approximately
bleeding is rare if the fibrinogen level is
80% of hemophilia patients have hemo-
>50 mg/dL. The thrombin time (TT) is the
philia A and 20% have hemophilia B. Both
most sensitive test to diagnose this disorder.
disorders are found throughout the world
Hypoprothrombinemia is an autosomal
and do not appear to have any ethnic or
recessive disorder associated with mild bleed-
racial predisposition.
ing. All patients have some prothrombin
Hemophilia A and B are clinically indis-
activity. Prothrombin deficiency also occurs
tinguishable since both FVIII and FIX are
in patients with vitamin K deficiency, liver
essential factors in the intrinsic clotting
disease, and warfarin use. Factor V (FV)
pathway for activating factor X (FX). Factor
deficiency is often characterized by bleeding
levels are measured in comparison to a
from mucous membranes including epi-
reference standard that is assumed to have
staxis, menorrhagia, and bleeding after dental
a factor level of 100% (1.0 IU/mL). In the
procedures. Congenital deficiency of both FV
normal individual, FVIII and FIX levels range
and FVIII can occur as a result of a mutation
from 50% to 200% (0.50 to 2.0 IU/mL).
in the ERGIC-53 gene, which acts as a chap-
Disease severity is defined as severe (<1%),
erone for intracellular transport of these
moderate (1% to 5%), and mild (6% to
factors through the endoplasmic reticulum.
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
80
Chapter 9
Table 9.1 Rare coagulation factor deficiencies.
Deficiency
Estimated
Source of
Biological
Gene on
prevalence
replacement
half-life
chromosome
Fibrinogen
1/1,000,000
Cryoprecipitate
90 h
4
Prothrombin
1/2,000,000
Activated PCCs
60 h
11
Factor V
1/1,000,000
FFP
12 h
1
Factor VII
1/500,000
rFVIIa
2-6 h
13
Factor X
1/1,000,000
Activated PCCs
24 h
13
Factor XI
1/1,000,000
FFP
40 h
4
Factor XIII
1/2,000,000
FFP
3-5 days
1 and 6
Abbreviations: PCC, prothrombin complex concentrate; FFP, fresh frozen plasma; rFVIIa,
recombinant factor VII.
Patients with factor VII deficiency who have
effectively inhibit plasmin, and therefore lyse
levels less than 1% have bleeding symptoms
clots abnormally quickly leading to abnormal
similar to that of classic hemophilia with
bleeding. As clots are formed normally, all the
hemarthrosis, intramuscular hemorrhage,
usual screening coagulation tests are normal
and intracranial hemorrhage
(ICH).
except for the euglobulin and urea clot lysis
Heterozygotes are typically asymptomatic.
tests. Factor XII (FXII) deficiency, prekal-
Two-thirds of patients with factor X (FX)
likrein deficiency, and high-molecular-
deficiency have hemarthroses and hemato-
weight kininogen deficiency all cause pro-
mas. The proportion of patients with FX
longation of the PTT but are not associated
deficiency who require treatment is higher
with clinical hemorrhage.
than that of the other rare coagulation
deficiencies. Factor XI (FXI) deficiency is
characterized by mucocutaneous and geni-
Clinical presentation
tourinary tract bleeding; the likelihood of
bleeding does not correlate well with mea-
Boys with hemophilia may develop symp-
sured FXI levels. Only individuals with factor
toms very early in life, experiencing a
XIII
(FXIII) deficiency whose levels are
hemorrhage after circumcision or with sep-
<1% are symptomatic. Bleeding manifesta-
aration of the umbilical cord. However, up
tions include soft-tissue hemorrhages,
to 50% of affected male infants have no
hemarthroses, and hematomas, and patients
difficulty during the neonatal period, and
undergoing surgery or trauma may experi-
a negative history of bleeding after circum-
ence poor wound healing.
cision does not rule out the diagnosis of
Alpha2-antiplasmin deficiency, factor XII
hemophilia. Mild hemophilia may go
deficiency, prekallikrein
(Fletcher’s factor)
unsuspected for years until the patient
deficiency, and high-molecular-weight kini-
experiences trauma or has a surgical proce-
nogen (Fitzgerald’s factor) deficiency have
dure. Conversely, patients with severe
also been identified using appropriate clot-
hemophilia often experience spontaneous
ting measurements. Alpha2-antiplasmin is
hemorrhages, both externally and inter-
the primary inhibitor of plasmin, a key com-
nally, into the head, joints, muscles, and
ponent of the fibrinolytic pathway. Patients
retroperitoneum. Bleeding from the mouth
with a2-antiplasmin deficiency are unable to
or frenulum often occurs during infancy,
Hemophilia
81
and minimal lacerations may cause very
treatment of an acute hemarthrosis is
prolonged bleeding. Intramuscular injec-
recommended to relieve symptoms and
tions (as with immunizations) often result
hopefully prevent recurrent bleeding into
in large hematomas. Joint hematomas
the same joint, setting up the situation of
(hemarthroses) can result in secondary joint
a “target” joint.
degeneration. Life-threatening blood loss
Bleeding into muscle is characterized by
can occur with intramuscular bleeds. ICH
pain and limitation of mobility. They can be
occurs in 2% to 14% of hemophilia patients
difficult to evaluate, particularly if they
and is associated with an 18% mortality.
occur in deep muscles such as the iliopsoas
Although ICH is much more common in
muscle (see below). They can result in long-
severe hemophilia, it has been reported in
term complications such as permanent
patients with mild hemophilia as well.
muscle contractures. It is important to
ICH is the most serious complication in
exclude any potential neurovascular com-
hemophilia. A history of trauma is elicited
promise that can result from compression of
in only 20% to 25% of patients with central
adjacent nerves.
nervous system (CNS) hemorrhages, and
An iliopsoas hemorrhage can be quite
there may be a delay of days or weeks before
devastating due both to the long-term con-
symptoms develop. Bleeding can be intra-
sequences and to the large volume of blood
cerebral, subdural, subarachnoid, or epidu-
loss that can occur acutely into this large
ral. Patients who survive these episodes
muscle bed and into the retroperitoneal
frequently experience complications such
space. The presenting signs of groin or lower
as mental retardation, seizure disorders,
abdominal/upper thigh pain can be difficult
or motor impairment. Any history of head
to differentiate from a hip bleed. With an
trauma should be considered emergent. A
iliopsoas bleed, examination reveals inabil-
nonfocal neurologic examination does not
ity to extend the hip, with normal internal
exclude a diagnosis of an intracranial bleed.
and external hip rotation. Radiologic con-
Any suspicion of a significant intracranial
firmation by CT scan is recommended.
injury should prompt a head CT and close
Soft tissue hemorrhages occur very
neurological observation. Immediate treat-
commonly and do not require therapy
ment to achieve a factor level of 80% to
unless their location is near vital structures.
100% should be provided and maintained
For instance, a retropharyngeal bleed with
until an intracranial bleed can be fully
potential airway compromise should be
excluded. An intraspinal hemorrhage
aggressively treated. Vigorous examination
should be excluded with any back trauma
of the oropharynx with a tongue blade or
or symptoms of a peripheral neuropathy. A
other manipulations should be avoided to
lumbar puncture should never be per-
prevent injury with subsequent hemor-
formed in a hemophilia patient without
rhage. A “straddle” injury resulting in a
factor therapy to avoid this complication.
hematoma of the perineal/perirectal area
Hemarthroses appear in young children
should be closely evaluated for neurologic
as they become mobile. The elbows are often
compromise as evidenced by either bladder
first affected as the infant begins crawling,
or bowel dysfunction.
followed by shoulders, wrists, hips, knees,
About 70% of hemophilia patients will
and ankles. The hands and spine are rarely
have at least one episode of hematuria.
involved. The initial signs of a hemarthrosis
Usually, these episodes are painless, thus
are vague and difficult to detect, especially
if there is severe pain associated with hema-
in the nonverbal infant. Early, aggressive
turia, a thrombosis in the renal pelvis
82
Chapter 9
should be excluded. Increased fluid intake
Newborn males with a possible or con-
and bed rest is recommended. Factor
firmed diagnosis of hemophilia should be
replacement may be warranted with per-
carefully evaluated for an intracranial hem-
sistent symptoms. Treatment with antifi-
orrhage, especially if delivered via assisted
brinolytic drugs is contraindicated given
vaginal delivery
(vacuum extraction or
the risk of upper urinary tract obstruction
forceps). However, infants delivered via
caused by clot.
cesarean section are also at risk for severe
Epistaxis occurs in hemophilia patients,
bleeding. The initial PTT can be performed
most commonly in those with severe dis-
on cord blood, but it must be remembered
ease. Gastrointestinal bleeding can also
that the upper limit of normal for newborns
occur but is atypical; an underlying gastro-
is longer than for adults. The normal range
intestinal lesion should be excluded. Intra-
for FVIII levels in the newborn is the same as
mural bleeding into the bowel wall can
for adults, but FIX levels are lower until the
occur, with signs of severe pain and possible
concentrations of factors produced by the
intestinal obstruction. Appropriate therapy
liver mature to adult levels (approximately 6
may avoid an unnecessary laparotomy.
months of age). Therefore, the diagnosis of
Prolonged gingival oozing is common
hemophilia B may be more difficult initially.
after shedding of deciduous teeth, eruption
Confirmatory studies should be performed
of new teeth, or instrumentation. Treatment
on a sample obtained by venipuncture if a
with an antifibrinolytic medication is
cord blood sample was initially obtained.
recommended for prolonged symptoms.
Arterial as well as femoral or jugular venous
Routine dental care is particularly important
sites to obtain plasma samples should be
in the patient with a bleeding disorder to
avoided if possible. Prenatal testing for both
prevent the need for restorative care in the
conditions is available to pregnant women
future where extensive factor replacement
who are known carriers.
therapy is often indicated. Regional block
anesthesia is discouraged, even with factor
coverage, given the risk of hemorrhage that
Treatment
can lead to nerve damage and extension into
the neck causing airway compromise.
After the newborn period, affected infants
rarely require treatment until they become
more mobile with increasing age. Treatment
Diagnosis
recommendations for the hemophilia
patient depend on several factors: the type
The diagnosis of FVIII deficiency should be
and severity of disease (mild, moderate, or
suspected in males presenting with a char-
severe; FVIII or FIX deficiency); the site of
acteristic bleeding history, especially if there
the bleed
(proximity to vital structures)
is a family history of males with abnormal
with consideration of long-term debilitating
bleeding. However, 30% of patients with
consequences; and therapy response (factor
hemophilia A and B have spontaneous
recovery/survival and inhibitor status).
mutations and no family history of abnor-
Treatment of patients with hemophilia
mal bleeding. A prolonged PTT in conjunc-
and bleeding is by replacement of factor in a
tion with a normal PT and platelet count is
concentrated form. Many high-purity
suggestive of the diagnosis; this can be
plasma-derived preparations are currently
confirmed by specifically measuring levels
available, as are recombinant FVIII and FIX
of FVIII and FIX.
products. Most experts feel that hemophilia
Hemophilia
83
patients who have not been previously trea-
the calculated target dose. Doses are calcu-
ted with plasma-derived products should be
lated using the following formula:
treated with only recombinant products.
There are two treatment strategies cur-
Dose ðunits of FVIIIÞ ¼ Weight ðkgÞ
rently used for patients with hemophilia.
desired % rise of FVIII level
0:5
Episodic or on-demand treatment involves
providing treatment at the time a bleeding
For FIX, multiply result by 2, for recom-
episode occurs. Prophylactic therapy is the
binant FIX, multiply by 2.4.
periodic infusion of factor replacement to
Round up to the whole vial since factor is
prevent such bleeding episodes. A number
expensive and not uniformly dispersed in
of single institutional case series have dem-
suspension. Table 9.2 outlines the dose and
onstrated the superiority of prophylaxis
duration of therapy for commonly encoun-
regimens in the prevention of joint disease,
tered bleeding episodes in hemophilia
but widespread adoption of this strategy for
patients. More serious bleeding episodes
all patients has been limited by the high cost
should be treated using doses at the higher
of factor replacement therapy and the chal-
end of the range.
lenge of adequate vascular access, particu-
Other important interventions for a
larly in young patients.
hemarthrosis include initial immobilization
When providing on-demand therapy,
and application of ice to the area or use of a
the duration, frequency, and doses of factor
cryocuff. For particularly critical joints such
provided depend on the severity of the
as the shoulder or hip, where long-term
hemophilia and bleeding episode. Factor
consequences of pressure-induced avascular
vials vary in the number of units of
necrosis can be debilitating, a longer treat-
factor activity they contain. The required
ment course is recommended. Iliopsoas
factor dose should be calculated, and vials
bleeds should be treated as retroperitoneal
should then be selected that come closest to
rather than as muscle bleeds.
Table 9.2 Guidelines for factor replacement in hemophilia A and B.
Site of hemorrhage
Optimal factor
Dose (units/kg body
Minimum duration of
level (%)
weight)
treatment (days)
Factor VIII Factor IX
Muscle
30-50
20-30
30-40
1-2
Joint
50-80
25-40
50-80
1-2
Gastrointestinal tract
40-60
30-40
40-60
10-14
Oral mucosa
30-50
20-30
30-40
2-3
Epistaxis
30-50
20-30
30-50
2-3
Hematuria
30-100
25-50
70-100
1-2
Retroperitoneal
80-100
50
100
7-10
Central nervous system
80-100
50
100
14
Trauma or surgery
80-100
50
100
14
Recombinant FIX should be dosed 1.2 times higher.
84
Chapter 9
Inhibitors
Other considerations
The development of an inhibitor, an anti-
In addition to aggressive factor replacement
body that inactivates the coagulant func-
when bleeding has occurred and the use of
tion of replacement factor, is one of the
physical therapy programs to prevent or min-
most serious complications of hemophilia
imize chronic joint disease, comprehensive
treatment. FVIII inhibitors develop in 20%
medical care must be provided to all patients
to 30% of boys with severe hemophilia A,
with hemophilia. All immunizations includ-
whereas FIX inhibitors develop in 3% to
ing hepatitis B vaccine should be given on
5% with severe hemophilia B. These usu-
schedule. Regular dental care is essential to
ally develop within the first 50 exposures to
prevent excessive decay and gingival disease
factor replacement. Risk factors for inhib-
that may predispose to complications. Psy-
itor development include the nature of the
chological support is essential to assist patients
underlying mutation, with those leading
and families cope with the emotional and
to substantial loss of coding information
social burden imposed by the disease. A com-
representing greater risk, high-intensity
prehensive program of patient and family
product exposure, CNS bleeding, and Afri-
education will prevent complications caused
can-American race. Inhibitors are quanti-
by failure to recognize or understand warning
tated by Bethesda titers: one Bethesda unit
signs of hemorrhage as well as empower
(BU) is the amount of inhibitor that inac-
family members and eventually the patient
tivates 50% of FVIII function in a one-stage
to take an active role in the management of the
clotting assay. Low-titer inhibitors
(<5
disease. Education in home treatment is vital,
BU) can be managed by using larger doses
and should be used whenever possible, as
of factor replacement; higher titer inhibi-
speed in the treatment of bleeding, especially
tors require the use of alternative hemostatic
hemarthroses, is directly related to minimizing
agents. Alternative agents include prothrom-
long-term complications of the disease.
bin complex concentrates (PCCs), activated
Suggested Reading
PCCs (aPCCs), and recombinant factor VIIa
(rFVIIa). Immune tolerance induction with
Kessler CM. New perspectives in hemophilia
high, frequent doses of FVIII or FIX has also
treatment. Hematology Am Soc Hematol
been successful. Management of patients
Educ Prog 429-435, 2005.
with inhibitors is difficult and complex and
Manco-Johnson MJ. Advances in the care and
should be performed only by experienced
treatment of children with hemophilia. Adv
hematologists.
Pediatr 57:287-294, 2010.
The Child with
10
Thrombosis
Virchow’s triad describes risk factors for
Evaluation of thrombosis
thrombus (blood clot) formation: (1) venous
stasis, (2) endothelial injury, and (3) hyper-
Pediatric patients may have both congenital
coagulability. Hypercoagulability, also called
and acquired risk factors underlying throm-
thrombophilia or a prothrombotic state, is
bus formation. The single most common
defined as a propensity to develop throm-
acquired risk factor for venous thromboem-
bosis secondary to aberrations in the coag-
bolism is the presence of a central venous
ulation system. Thrombophilia is becoming
catheter
(e.g., endothelial injury). Other
increasingly recognized in pediatric patients
acquired risk factors include trauma, surgery,
and can refer to multiple potential condi-
infection, nephrotic syndrome, inflammatory
tions: (1) spontaneous thrombus formation,
syndromes, diabetes, complex congenital
(2) recurrent thrombosis, (3) thrombosis out
heart disease, liver disease, and malignancy
of proportion to the underlying risk factor,
(e.g., acute lymphoblastic leukemia in asso-
and (4) thrombosis at a young age.
ciation with the use of asparaginase or solid
Optimal prevention and treatment in
tumors with tumor thrombus). Asymptom-
pediatric patients with thrombosis differs
atic patients may be found to have an
from adult patients for several reasons.
incidental clot on imaging, or the patient
These include physiologic age-dependent
may be symptomatic, most often with swell-
differences in the hemostatic system that
ing, pain, or erythema in an extremity.
influence the risk for venous thromboem-
Venous ultrasonography with Doppler flow
bolism, differing underlying etiologies and
remains the cornerstone of evaluation for
location of clots, and differing responses to
thrombosis, as in adults. Lack of venous
antithrombotic agents. Little evidence exists
compression or Doppler flow is diagnostic
as how best to manage pediatric patients
for thrombus. This imaging modality is
with thrombosis, both from a diagnostic
limited to the extremities, neck, distal sub-
and from a treatment standpoint. Much
clavian veins, and potentially, distal iliac
of the currently utilized data is based on
veins. Echocardiography can be utilized to
adult studies, and here we summarize the
evaluate for atrial clots and the proximal
generally accepted recommendations in lieu
superior and inferior vena cava. Imaging of
of evidence-based pediatric guidelines (see
the abdomen and pelvis as well as the upper
Figure 10.1).
venous system requires CT or magnetic
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
86
Chapter 10
Initial evaluation
Physical examination
History (medications, family history)
Laboratory (PT, PTT, fibrinogen, CBC)
Diagnostic imaging confirming VTE
Assessment of risk
Unusual site without underlying risk factor (e.g., portal vein, renal vein)
Life threatening without underlying risk factor (pulmonary embolus,
CNS event)
Recurrent
Laboratory assessment of primary
Family history of VTE
thrombophilia traits
Yes
Adolescent on OCPs
Factor V Leiden mutation
Neonatal stroke
Prothrombin gene mutation
ATIII deficiency
Protein C deficiency
Protein S deficiency
No
Hyperhomocysteinuria
Elevated lipoprotein (a)
Antiphopholipid antibodies
Further assessment of risk factors:
Elevated factor VIII
Venous catheter
Trauma, surgery, immobilization
Prematurity
Congenital heart disease
Nephrotic syndrome
Inflammatory bowel disease
No multitrait
Malignancy (ALL)
thrombophilia
Collagen vascular disease
Multitrait ( 3)
Yes
thrombophilia
No
High risk for recurrence
Moderate risk for recurrence
Minimal risk for recurrence
Long-term anticoagulation
Consideration for long-term
Long-term anticoagulation
anticoagulation while with
not required
continued risk factors
Figure 10.1 Evaluation of the child with venous thromboembolism (VTE). (Abbreviations: PT,
prothrombin time; PTT, partial thromboplastin time; CBC, complete blood count; CNS, central
nervous system; OCP, oral contraceptive pill; ATIII, antithrombin III; ALL, acute lymphoblastic
leukemia).
resonance venography
(MRV). Similarly,
underlying hypercoagulable state. In the
sinus venous thrombosis must be eva-
patient with a known acquired risk factor
luated by magnetic resonance imaging/
and secondary thrombosis, it is unclear that
arteriography
(MRI/MRA). Pulmonary
testing for thrombophilia will aid in the
emboli or thrombosis can be identified with
management of the patient (e.g., asymptom-
a helical CT scan.
atic catheter-related thrombosis). Patients
might have a single-trait thrombophilia on
Testing for thrombophilia
testing, but this alone may not contribute
Although often undertaken, clear guidelines
significantly to thrombus development.
are lacking as to the utility of testing for an
Patients with a consumptive process (e.g.,
The Child with Thrombosis
87
disseminated intravascular coagulation and
asymptomatic patient with a positive family
sepsis) and secondary thrombosis such as
history, the decision to test for thrombophi-
with deficiencies in protein C and S and
lia must be made on an individual basis after
antithrombin III could benefit from testing
appropriate family counseling. The family
and factor replacement if found to be defi-
must be advised that a positive screen for a
cient. For patients with spontaneous throm-
single thrombophilia trait will not result in a
bosis, often found in adolescents
(e.g.,
change in management and may lead to
adolescent female starting oral contracep-
unnecessary anxiety. Similarly, negative test-
tives) and in neonates (secondary to stroke
ing may give false reassurance and does not
or sinus venous thrombosis), testing should
completely abrogate the potential for throm-
be considered. For patients with a symptom-
bosis. The primary thrombophilia traits are
atic catheter-related thrombosis or for an
listed in Table 10.1.
Table 10.1 Primary thrombophilia traits.
Thrombophilia trait
Description
Factor V Leiden
G1691A polymorphism, present in
5% of Caucasian popula-
tion, leads to inherited activated protein C resistance
Prothrombin gene mutation
PT G20210A polymorphism, present in
2% of Caucasian
population, leads to elevated prothrombin levels
Antithrombin III deficiency
Can be seen in consumptive process (sepsis, DIC, active clot),
liver disease, heparin therapy, complex congenital heart dis-
ease, asparaginase use in acute lymphoblastic leukemia
Protein C deficiency
Can be seen in consumptive process (sepsis, DIC, active clot),
liver disease, warfarin therapy, nephrotic syndrome, complex
congenital heart disease
Protein S deficiency
Can be seen in consumptive process (sepsis, DIC, active clot),
liver disease, warfarin therapy, nephrotic syndrome, complex
congenital heart disease
Hyperhomocysteinemia
Check fasting homocysteine, can be secondary to underlying
MTHFR mutation
Elevated lipoprotein (a)
Poorly understood, competes with plasminogen, thus decreasing
fibrinolysis, also activates PAI-1 decreasing fibrinolysis and
potentially leading to increased thrombogenesis
Antiphospholipid antibodies
Antibodies against cell membrane phospholipid, called lupus
anticoagulant, paradoxically leads to elevated PTT without
correction on 1:1 mix. Secondary confirmatory testing
includes anticardiolipin antibodies and b2 glycoprotein, must
be confirmed on two separate occasions 12 weeks apart as can
be seen transiently with infection
Elevated factor VIII
An acute phase reactant, can be constitutively elevated
Abbreviations: DIC, disseminated intravascular coagulation; MTHFR, methylenetetrahydrofolate
reductase; PAI-1, plasminogen activator inhibitor-1; PT, prothrombin time; PTT, partial
thromboplastin time.
88
Chapter 10
Management of thrombosis
first-pass liver metabolism, although this is
mitigated with the use of systemic TPA as it is
Management of pediatric thrombosis is again
expected that much of the TPA will bypass
based on adult studies and expert guidelines.
the obstruction through collateral circulation.
Removal of the precipitating agent should be
Patients receiving systemic low-dose TPA
done as possible in cases with an acquired risk
therapy should have close monitoring of
factor. Treatment of thrombosis is under-
coagulation studies
(prothrombin time
taken to prevent clot propagation, pulmo-
[PT], partial thromboplastin time [PTT], and
nary embolus, and the postthrombotic syn-
fibrinogen), platelets, and plasminogen to
drome (PTS). Although reported frequently
ensure hemostatic and fibrinolytic potential
in adults, PTS is poorly understood in chil-
and the need for potential repletion with fresh
dren but can potentially lead to chronic pain,
frozen plasma (FFP).
swelling, skin changes, and collateral venous
formation. The decision to initially utilize a
Anticoagulation
thrombolytic (tissue plasminogen activator,
If TPA is thought to pose significant risk, or
TPA) versus an anticoagulant (unfractio-
if the patient has a nonocclusive thrombus
nated heparin, UH or low-molecular-weight
or transient risk factors, UH or LMWH may
heparin, LMWH) must be made in consul-
be started. UH is a natural anticoagulant
tation with a pediatric hematologist. In some
and works by complexing to the physiologic
cases, thrombectomy or thrombolysis by
inhibitor antithrombin III (ATIII), acceler-
intervention radiology should be considered.
ating the inhibition of thrombin and other
coagulant proteins. It is therefore important
Thrombolysis
to ensure adequate levels of ATIII when
In general, a thrombolytic should be con-
administering heparin. Many potential
sidered in patients with an occlusive throm-
issues are present with UH usage in chil-
bus without significant risk factors for
dren:
(1) rapid clearance,
(2) low ATIII
bleeding
(recent surgery, central nervous
levels in the first few months of life,
system hemorrhage). Patients with a long-
(3) greater variability in dosing compared
standing occlusive thrombus (i.e., >14 days)
to adults, and (4) lack of evidence assessing
or without evidence of improvement on
the optimal target PTT range for the preven-
imaging studies after 24 to 48 hours of
tion and treatment of venous thromboem-
TPA therapy may benefit from thrombect-
bolism. The patient must also be monitored
omy or thrombolysis by interventional
for the development of heparin antibodies
radiology. Although not evidence based,
that can most notably lead to heparin-
we typically treat with low-dose systemic
induced thrombocytopenia
(HIT). Long-
TPA as suggested by Manco-Johnson
term usage can also lead to osteoporosis.
(see suggested further reading) at a dose
LMWH is more widely utilized now due
of 0.03 to 0.06 mg/kg/h with a maximum
to a more narrow dosing range and wide
of 2 mg/h for 24 to 48 hours to decrease
therapeutic window secondary to direct
the risk of significant hemorrhage that is
targeting of Factor Xa as well as reduced
seen with bolus TPA, unless there is a life-
incidence of HIT and osteoporosis.
threatening clot (e.g., massive pulmonary
In general, we utilize LMWH unless
embolism). Arterial clots may benefit from
there is potential need for immediate rever-
a higher systemic dose for a shorter time
sal of anticoagulation. Although protamine
period. Also if possible, TPA should be
can be utilized with LMWH, the reversal
instilled distal to the clot in order to decrease
effect is not complete (thought to be about
The Child with Thrombosis
89
two-thirds effective) and therefore a risk of
marker elevation may be confounded by
bleeding can still be present if, for instance,
continuing inflammation.
surgical intervention is required emergently.
In these cases, UH should be utilized due to
Duration of therapy
its short half-life and ability to be fully
Duration of therapy depends on multiple
reversed. UH is generally given at a loading
factors including the time to removal of
dose of 75 U/kg followed by a maintenance
inciting agents such as central venous cathe-
dose of 20 U/kg/h with a goal PTT of 2 to
ters, presence of thrombophilia traits,
3
the upper limit of the reference range or,
and time to clot resolution. Patients initially
preferably, anti-Xa levels of 0.3 to 0.7 U/mL,
treated with TPA or UH therapy should be
if available in a timely manner. Currently,
transitioned to LMWH or warfarin therapy
the only LMWH approved by the FDA for
for long-term maintenance. Because of the
pediatric patients is enoxaparin, with a half-
time required to reach an appropriate ther-
life of 4 hours and subcutaneous dosing. For
apeutic level, warfarin therapy should begin
treatment of thrombosis, enoxaparin must
at least 48 hours prior to discontinuation of
be given every 12 hours, usually starting at
heparin therapy. Maintenance with LMWH
1.0
to
1.25 mg/kg
(see Formulary and
is frequently used due to the difficulty in
Manco-Johnson for more specific dosing
maintaining a therapeutic international nor-
guidelines). Treatment effectiveness is fol-
malized ratio (INR) with warfarin therapy.
lowed by anti-Xa levels, with goal levels of
For patients with clot resolution, antic-
0.5 to 1.0 U/mL (checked 4 hours after the
oagulation should be continued at prophy-
second or subsequent dose). For prophy-
lactic dosing for 6 weeks to 3 months after
laxis, 0.5 mg/kg is typically given twice daily.
removal of the inciting agent (e.g., central
Anti-Xa levels do not need to be checked
venous catheter) due to the likelihood of
with prophylactic dosing. In practice, some
continued endothelial injury. For patients
hematologists utilize 1 mg/kg once daily for
without clot resolution (potentially after inef-
prophylaxis, although available data suggest
fective interventional thrombectomy and/or
this may undertreat a group of patients,
thrombolysis), anticoagulation should be
especially those <5 years of age.
continued for 6 to 12 months to potentially
Concomitant use of TPA and UH or
decrease or resolve the clot and prevent the
LMWH may also be beneficial in clot lysis
development of PTS. Patients that had a
without significantly increasing hemorrhagic
spontaneous or recurrent thrombus and were
risk, although again this recommendation is
found to have multitrait thrombophilia (
3
not evidence-based in pediatric patients and
traits) or antiphospholipid syndrome should
must be determined on a per patient basis in
continue life-long anticoagulation.
consultation with a pediatric hematologist,
weighing the potential risks of hemorrhage
New agents
and potential benefits of clot lysis. Therapeu-
Multiple new agents will be available in the
tic efficacy can simply be followed with repeat
near future for pediatric patients and are
imaging, although this can be difficult with
summarized in Table 10.2.
central venous clots that may require multi-
ple CT or MRI with resultant increased radi-
ation exposure and cost, respectively. Other
Case study for review
potential markers to follow for clot lysis
include d-dimers or fibrin-split products
You are seeing a 17-year-old female in clinic
which should increase with lysis, although
who is deciding on whether to start oral
90
Chapter 10
Table 10.2 Novel anticoagulants.
Name
Comments
Factor Xa inhibitors
Fondaparinux
Selective factor Xa inhibitor, mediates its effects through ATIII,
decreased HIT, half-life 17 hours, once daily SC dosing
Idraparinux
Hypermethylated analogue of fondaparinux, 80-hour half-life
(once weekly SC dosing)
Rivaroxaban
Oral factor Xa inhibitor, once daily dosing in adults for
thromboprophylaxis, has shown noninferiority to warfarin
Direct thrombin inhibitors
Argatroban
Approved in adult patients with HIT, narrow therapeutic
window and short-acting IV formulation
Dabigatran
Oral formulation, approved in adult patients, has shown
noninferiority to warfarin, currently approved in the United
States for stroke prevention in patients with atrial fibrilla-
tion, in the United Kingdom as an alternative to warfarin for
thromboprophylaxis
Abbreviations: ATIII, antithrombin III; HIT, heparin-induced thrombocytopenia; SC,
subcutaneous; IV, intravenous.
contraceptive pills (OCPs). She has an aunt
incidence is also highest in the first year of
that had a “blood clot” when she was in her
OCP usage.
fifties. Your patient does not know any
Thrombophilia testing can be considered
further details about the incident but does
on an individual basis for the patient wish-
know about the potential increased risk of
ing to start OCPs who has a family member
blood clots with oral contraceptive pills.
with a known thrombophilia. Additionally,
the patient should be offered alternative
1. What advice would you give the patient?
lower risk forms of contraception. In this
2. She asks about testing for clotting risk
case, the practitioner should try to get
factors, what do you advise her?
more information regarding the episode
of thromboembolism in the aunt. Were
All patients who are starting OCPs should
there acquired risk factors such as trauma,
be made aware of the increased risk of blood
surgery, or immobility? Was the clot
clots with estrogen-containing contracep-
thought to be spontaneous? If so, was the
tives (about a three times increased risk).
aunt tested for thrombophilia?
This risk is thought to be about 2 to 3 per
Current recommendations would not
10,000 per year on OCPs, or about 0.02% to
advise testing your patient without further
0.03% incidence per year
(baseline risk
information. If the aunt had a spontaneous
being approximately
0.008% per year).
thrombus, one could advise that she
Single-trait thrombophilias, most com-
should be tested for thrombophilia, and if
monly Factor V Leiden mutation, increase
found to be positive your patient could be
this risk significantly. Specifically for Factor
tested as well. If it was determined that
V Leiden, the risk for a heterozygous carrier
there was an underlying risk factor in the
increases the risk approximately 30 times to
aunt’s thromboembolism, testing her and/
a yearly incidence of 0.6% to 0.9%. This
or your patient would not be advised. In the
The Child with Thrombosis
91
situation where thrombophilia testing of
Goldenberg NA. Thrombophilia states and mar-
your patient is not advisable (the most likely
kers of coagulation activation in the predic-
tion of pediatric venous thromboembolic
scenario in general), you should counsel
outcomes: a comparative analysis with respect
your patient closely on the risks and benefits
to adult evidence. Am Soc Hematol Educ Prog
of OCPs and the potential lower risk alter-
236-244, 2008.
native therapies such as progesterone-only
Manco-Johnson MJ. How I treat venous throm-
preparations (e.g., Depo-Provera).
boembolism in children. Blood 107:21-29,
2006.
Raffini L. Thrombophilia in children: who to test,
Suggested Reading
how, when, and why? Am Soc Hematol Educ
Prog 228-235, 2008.
Bounameaux H, Perrier A. Duration of anticoa-
Rosendaal FR. Venous thrombosis: the role of
gulation therapy for venous thromboembolism.
genes, environment, and behavior. Am Soc
Am Soc Hematol Educ Prog 252-258, 2008.
Hematol Educ Prog 1-12, 2005.
The Neutropenic Child
11
Neutrophils are a key component in the
determining the individual’s risk for infec-
defense against infection. They contain toxic
tion and the urgency of medical interven-
cytoplasmic granules that, following inges-
tion. However, this definition fails to take
tion of infecting bacteria and fungi, are
into account important variations in nor-
released into the phagocytic vacuole and
mal neutrophil number related to age and
destroy them. Once released from the bone
ethnicity. Up to
25% of young African-
marrow, they circulate in the blood for a
American children will have an ANC
brief time (4 to 6 hours) before leaving the
between 1.0 and 1.5
109/L, and this level
circulation and entering the tissue where, in
should be considered normal in this group.
response to the presence of infection, they
Also, the normal range for ANC extends
act to control the infection while sending
down to 1.0
109/L in infants between 2
chemotactic signals to recruit additional
weeks and 6 months of age, and should not
neutrophils to the area and stimulate the
be considered abnormal if less than this.
accelerated production of neutrophils in the
bone marrow. While the presence of ade-
quate neutrophil numbers in the tissue is the
Risk assessment
best predictor of the patient’s ability to fight
infection, there is no easy clinical method to
The first question when evaluating a child
determine the number of tissue neutrophils,
with neutropenia is:
“What is the risk
so the number present in the blood is used
that this patient has or will develop a life-
as a surrogate marker.
threatening infection?” This is particularly
Neutropenia has traditionally been de-
pertinent if the patient presents with fever.
fined as a decrease in the absolute neutro-
The susceptibility to bacterial infection in
phil count
(ANC) to <1.5
109/L. The
neutropenic patients is quite variable and
ANC is calculated by multiplying the white
depends on a number of factors. The first
blood cell count by the total percentage of
is the severity of neutropenia, as described
segmented
(mature) neutrophils plus
above. Patients with mild neutropenia have
bands. Mild neutropenia is defined as an
minimal to no increased risk of infection,
ANC of 1.0 to 1.5
109/L, moderate neu-
those with moderate neutropenia have a
tropenia is an ANC of 0.5 to 1.0
109/L,
mildly increased risk of frequent or severe
and severe neutropenia is an ANC below
infections, and those with severe neutropenia
0.5
109/L. This division is useful for
are highly susceptible to bacterial infection.
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
The Neutropenic Child
93
However, patients who are neutropenic as a
Etiology of neutropenia
result of cytotoxic therapy (e.g., chemother-
apy and radiation) may be at an increased
A large variety of conditions can lead to
risk of infection because of the rate of decline
neutropenia in childhood. They can be clas-
of the neutrophil count or other immuno-
sified as being either acquired causes or
suppressive effects of chemotherapy, even
inherited causes. Of these, acquired causes
before the ANC falls below
0.5
109/L.
are much more frequent. These are pre-
Another factor is the cause of the neutrope-
sented in Table 11.1.
nia, if known. Patients with neutropenia
Viral infection is the most common
caused by intrinsic bone marrow failure or
cause of mild-to-moderate neutropenia.
hypoplasia have a greater risk of infection
Transient marrow suppression is seen in
than those whose neutropenia is due to
children with Epstein-Barr virus
(EBV),
factors extrinsic to the bone marrow such
respiratory syncytial virus, influenza A and
as excessive neutrophil destruction or
B, hepatitis, human herpesvirus-6 (HHV6),
sequestration
(immune neutropenia and
varicella, rubella, and rubeola. This typically
splenomegaly, respectively). While patients
lasts for 3 to 8 days. Neutropenia can also be
with extrinsic neutropenia may have a low
seen in the setting of overwhelming bacte-
circulating neutrophil count, their tissue
rial infection as well as with typhoid fever,
counts are near normal and they are more
Rocky Mountain spotted fever, and tuber-
protected from infection. A third factor is the
culosis. Phagocytosis of microbes leads to
presence or absence of other phagocytic cells.
release of toxic metabolites, which then
Many patients with chronic neutropenia
activate the complement system, inducing
have an elevated circulating monocyte count,
neutrophil aggregation and adherence of
which provides some additional protection
leukocytes to the pulmonary capillary bed.
against pyogenic organisms. Conversely,
Tumor necrosis factor and interleukin-1,
patients being treated with steroids or other
released by macrophages, likely accelerate
immunosuppressive therapy will have treat-
this process. Activated granulocytes seques-
ment-related abnormalities in cell-mediated,
tered in the lungs may cause acute cardio-
humoral, and macrophage-monocyte immu-
pulmonary complications. Neonates have a
nity and may be at increased risk of invasive
limited granulocyte pool in their bone mar-
infection even with a normal neutrophil
row, which can be exhausted rapidly during
count. Patients with a history of splenectomy
overwhelming bacterial sepsis.
have an increased risk of overwhelming infec-
Replacement of the bone marrow (as
tion caused by encapsulated bacteria regard-
occurs with hematologic malignancies, gly-
less of their neutrophil count.
cogen storage diseases, granulomas associated
The age at presentation with neutropenia
with infection, and fibrosis related to chemical
is an important factor in risk assessment.
or radiation injury or osteoporosis) results in
The younger the child is at the time of
neutropenia. Frequently, the erythroid and
diagnosis, the greater the concern that the
megakaryocytic lines are also affected.
patient could have a severe form of chronic
Medications are a common cause of neu-
neutropenia, whereas children who are
tropenia. Neutropenia is a common and
found to be neutropenic later in life and
expected side effect of anticancer therapy.
who do not have a history of frequent
Many chemotherapeutic agents have a direct
bacterial infections or chronic illness are
toxic effect on the early marrow stem cells.
more likely to have a more benign form
The severity and duration of the neutropenia
of neutropenia.
depend on the particular medication and
94
Chapter 11
Table 11.1 Acquired causes of neutropenia.
Condition
Pathogenesis
Occurrence
Associated findings
Infection
Viral marrow suppression,
Common
EBV, parvovirus, HHV6,
viral-induced immune
other viruses
neutropenia
Bacterial sepsis-endotoxin
Less common
Severe infection
suppression
Bone marrow
Infiltration of marrow with
Uncommon
Vary depending on
replacement
malignant cells, fibrosis,
underlying cause
granulomas
Drug-induced
Direct marrow suppression
Common
Underlying condition
Immune destruction
Less common
Autoimmune
Primary (molecular
Common
Monocytosis common
mimicry)
Secondary (SLE, Evans
syndrome)
Newborn
Alloimmune-maternal
Rare
Antigen difference in
immune
sensitization
newborn and mother
Due to maternal autoim-
Maternal neutropenia
mune neutropenia
Chronic
Ineffective or decreased
Common
Consider also familial
idiopathic
production
benign neutropenia
Often asymptomatic
Sequestration
Hypersplenism
Common if
Mild neutropenia
spleen is
Enlarged spleen—
enlarged
many causes
Nutritional
Vitamin B12 or folate
Rare in children
Hypersegmented
deficiency
neutrophils
Copper deficiency
Zinc excess
Impaired DNA processing
Abbreviations:
EBV, Epstein-Barr virus; HHV6, human herpesvirus-6; SLE, systemic lupus
erythematosus.
dosage as well as the patient’s underlying
immune-mediated destruction, toxic effect
disease, state of nutrition, and general health.
of the drug or metabolites on the marrow
Many other medications can induce neutro-
stem cells, and toxic effects on the marrow
penia, including antibiotics (chlorampheni-
microenvironment. After withdrawal of the
col, cephalosporins, penicillins, and sulfona-
drug, the marrow can repopulate with early
mides), anticonvulsants
(phenytoin and
myeloid forms within 3 to 4 days and appear
valproic acid), anti-inflammatory agents,
morphologically normal by 1 to 2 weeks.
cardiovascular agents, tranquilizers, and
The duration of neutropenia is likely related
hypoglycemic agents. The severity and dura-
to the underlying mechanism; some chronic
tion of drug-induced neutropenia are vari-
idiosyncratic drug reactions can last from
able. The underlying mechanism is not
months to years.
known, although studies with certain drugs
Autoimmune neutropenia is another
have led to various hypotheses including common cause of neutropenia following a
The Neutropenic Child
95
viral infection, and may be difficult to dis-
neutrophils caused by impaired neutrophil
tinguish from virus-associated marrow sup-
nuclear maturation is a clue to this condition.
pression due to the difficulty in identifying
Similarly, copper deficiency is a rare cause of
antineutrophil antibodies. Unlike viral sup-
neutropenia often secondary to an underlying
pression, autoimmune neutropenia can per-
malabsorptive process or due to zinc excess.
sist for 7 to 24 months, and affected children
These patients usually present with concom-
occasionally suffer from infections of the
itant anemia and thrombocytopenia.
ear, lung, skin, or other sites.
A number of inherited conditions have
Neonatal alloimmune neutropenia is
neutropenia as one of their characteristic
analogous to Rh-related hemolytic disease
features. The more common of these con-
of the newborn; mothers generate IgG anti-
ditions are presented in Table 11.2.
bodies against paternal neutrophil antigens
Children with severe congenital neutro-
expressed on fetal neutrophils. These anti-
penia (SCN) often present in early infancy
bodies cross the placenta and cause neutro-
with umbilical infection, skin infections, oral
penia in the fetus and newborn.
ulcers, pneumonia, or perineal infections.
“Chronic idiopathic” neutropenia, also
There are two forms: an autosomal recessive
known as chronic benign neutropenia of
form (Kostmann syndrome) involving muta-
childhood, is likely caused by a variety of
tions in the HAX1 gene that is involved in
disorders without a unifying etiology. It is
signal transduction and an autosomal dom-
likely that many of these children in fact
inant form involving mutations in the neu-
have autoimmune neutropenia or familial
trophil elastase gene (ELA2) or, more rarely,
benign neutropenia. They often have mild-
in the GFI1 gene that targets ELA2. Bone
to-moderate neutropenia; the susceptibility
marrow aspiration reveals a maturational
to infection is roughly proportionate to the
arrest at the promyelocyte stage. In addition
degree of neutropenia. The blood neutro-
to the risk of death due to overwhelming
phil count remains stable over years, and
infection, patients with either of these con-
becomes elevated in response to an infection
ditions have an increased risk of developing
in a subset of children. Spontaneous remis-
myelodysplastic syndrome (MDS) or acute
sions at
2 to
4 years of age have been
myelogenous leukemia (AML).
reported. Affected individuals have normal
Cyclic neutropenia is characterized by
life expectancies. Evaluation of the bone
approximately 21-day cycles of changing
marrow shows decreased myelopoiesis
neutrophil counts, with frank neutropenia
(often with monocytosis), and there is con-
developing at regular intervals and lasting 3
siderable variability in the stage at which
to 6 days. Fever and oral ulcerations usually
maturation is arrested. These patients are at
are seen during the nadir, as well as gingi-
low risk for the development of serious
vitis, pharyngitis, and skin infections. Diag-
infections, and no treatment is required
nosis is often delayed because the neutrophil
except during infectious episodes.
count may have improved by the time
Splenomegaly from any cause including
the patient seeks medical attention. These
chronic hemolytic anemia, portal hyperten-
patients also demonstrate mutations in the
sion, liver disease, and storage disorders can
ELA2 gene, but at different locations than in
cause mild neutropenia, as well as anemia
those with SCN. They do not have the same
and thrombocytopenia. If severe, splenec-
risk of developing MDS or AML. Diagnosis
tomy may be necessary.
is made by obtaining serial complete blood
Deficiencies of folate and B12 are rare in
counts (CBCs) 2 to 3 times/week over 4 to
children; the presence of hypersegmented
6 weeks to demonstrate the periodicity of
96
Chapter 11
The Neutropenic Child
97
98
Chapter 11
the cycle. Granulocyte colony-stimulating
have very short germ line telomeres, and
factor (G-CSF) has been used in patients
approximately half have mutations in one
who develop recurrent infections during
of six genes encoding proteins that maintain
their nadir.
telomere function. Affected patients rarely
Shwachman-Diamond syndrome in-
present with hematologic abnormalities dur-
cludes exocrine pancreatic insufficiency,
ing childhood.
short stature, metaphyseal dysplasia, and
Diamond-Blackfan anemia is a congen-
bone marrow failure with typically mild-to-
ital anemia that presents in infancy. There
moderate neutropenia. Affected patients
is typically a failure of erythropoiesis with
also have an increased risk of MDS and
preservation of myeloid and platelet
AML. Intestinal malabsorption with failure
production. Only 20% to 25% of cases are
to thrive commonly occurs, especially in
inherited; the rest are sporadic. Approxi-
infancy and early childhood.
mately 50% of affected persons have devel-
Benign familial neutropenia is charac-
opmental abnormalities including growth
terized by moderate neutropenia with min-
retardation and craniofacial, upper limb/
imal risk of invasive bacterial infections. It
hand, cardiac, and genitourinary malforma-
has been postulated that the underlying
tions. Neutropenia is seen in 25% to 40%
cause is a defect in neutrophil mobilization
of affected children. Patients also have an
from the bone marrow, but the etiology is as
increased risk of MDS, AML, and osteogenic
yet unknown. It occurs more commonly in
sarcoma. Genetic studies have identified
individuals of African, Arabic, and Yeme-
heterozygous mutations in at least one of
nite Jewish descent. Periodontal disease is
eight ribosomal protein genes in up to 50%
the most frequent complication.
of cases. Mutations in ribosomal protein L5
Fanconi anemia is characterized by a
(RPL5) are associated with multiple physi-
defect in DNA repair leading to extensive
cal abnormalities including cleft lip/palate,
chromosomal breakage. Affected patients
thumb, and heart anomalies. The diagnosis
have mutations within the FANC family of
is often difficult due to incomplete pheno-
genes, including FANC A, C, and G. It pre-
types and a wide variability of clinical
sents most commonly during the early school
expression. Up to 80% of patients respond
age years; patients with the characteristic
to a course of steroids with improvement in
physical findings may be diagnosed sooner.
their cytopenias.
Thrombocytopenia is often the presenting
The etiology of neutropenia in glycogen
hematologic abnormality, with anemia and
storage disease 1b is not definitively known,
neutropenia developing later. Up to 10% of
but appears to be related to increased
patients ultimately develop MDS or AML.
neutrophil apoptosis caused by an excess
Bone marrow transplantation is curative, but
of reactive oxygen species due to a defect in
challenging to complete successfully given
neutrophil energy metabolism.
the underlying chromosomal fragility.
Almost one-third of patients with selec-
Dyskeratosis congenita (DKC) is char-
tive IgA deficiency have evidence of
acterized clinically by the triad of abnormal
autoimmune disease; neutropenia in this
nails, reticular skin pigmentation, and oral
condition is felt most likely related to the
leukoplakia. Bone marrow failure occurs
presence of antineutrophil antibodies.
during early adulthood and is associated with
Patients with a mild form of Wiskott-
a high risk of developing aplastic anemia,
Aldrich syndrome
(WAS) known as
MDS, leukemia, and solid tumors. Patients
X-linked neutropenia have a unique “gain
The Neutropenic Child
99
of function” mutation in the GTPase-binding
include antineutrophil antibodies, assess-
domain of the WAS protein (WASp) that
ment of cellular and serum immune status,
leads to increased actin polymerization in
and careful review of the peripheral smear
neutrophils, causing defective mitosis and cell
for morphologic abnormalities of the white
movement and ultimately severe neutropenia.
cells. If severe congenital neutropenia or
Kostmann syndrome is suspected, assess-
ment for ELA2
and HAX1
mutations
Initial evaluation of the child with
should be made. A bone marrow aspirate
neutropenia
and biopsy may be necessary to identify
granulocyte precursors and to search for
Evaluation of the child with neutropenia
defects in myeloid maturation. In addition,
begins with a thorough history and physical
the bone marrow aspirate and biopsy can be
examination. It is critical to know whether
used to exclude hematologic malignancies,
the child has had recent or recurrent bac-
marrow infiltration, or fibrosis.
terial infections, and whether there is a
family history of neutropenia or recurrent
bacterial infections. On examination, atten-
Management of the child with
tion should be paid to any phenotypic
neutropenia and fever
abnormalities, adenopathy, splenomegaly,
evidence of a chronic or underlying disease,
Children identified with neutropenia but who
and meticulous evaluation of the skin and
are free of fever or other signs of infection
mucous membranes (particularly in the oral
should be evaluated as outlined above, but
and perirectal areas). A CBC must be done
require no other therapy at that time. Man-
to determine if the patient has isolated
aging the child with neutropenia and fever is
neutropenia or neutropenia associated with
more complex and depends on many factors
anemia and/or thrombocytopenia. The
including the nature of the neutropenia
approach to a child with bi- or trilineage
(acute or chronic), its severity, and the asso-
abnormalities is different than that in a child
ciation with immune defects, underlying
with isolated neutropenia; multiple abnor-
illnesses, or malignancies. Patients with
mal lineages increase the likelihood of a
acquired neutropenia arising from malig-
more generalized marrow failure state such
nancy or chemotherapeutic drugs have a
as aplastic anemia or a marrow infiltrative
diminished inflammatory capability and have
process such as leukemia. It is valuable to
a greater susceptibility to sepsis. Fever may be
repeat the CBC at least once, especially if the
the earliest and only warning sign. Sepsis
child appears well, to avoid proceeding with
related to chemotherapy-induced neutrope-
a major evaluation due to a laboratory error.
nia remains a leading cause of mortality in
Well-appearing children with mild-to-
these patients. Aggressive management of the
moderate neutropenia can typically be
febrile, neutropenic cancer patient in the
observed over the next 2 to 3 weeks with
hospital has markedly reduced morbidity and
serial CBCs. Children with persistent or
mortality due to infection. For management
worsening neutropenia require further eval-
of oncology patients with fever and neutro-
uation. Patients with recurrent neutropenia
penia, see Chapter 27.
should have blood counts checked 2 to 3
The management of the febrile child who
times per week for
6 weeks to evaluate
is discovered to be neutropenic depends on
for cyclic neutropenia. Additional studies
several factors. Patients with fever and mild
100
Chapter 11
or moderate neutropenia and signs and
antibiotic choices are appropriate and
symptoms suggestive of a viral illness that
depend on local practice.
otherwise look well may be managed sup-
* Careful physical evaluation, paying
portively without antibiotics, although in
meticulous attention to potential sites of
most cases they will receive a long-acting
occult infection (e.g., the oral cavity and
cephalosporin such as ceftriaxone prior to
perineum).
discharge from the clinic or emergency
* Laboratory studies: CBC with differential,
department. Their parents should be
blood cultures, urinalysis and urine culture,
instructed to bring them back in 24 hours
and cultures from sites of suspected infec-
for further evaluation, or sooner if they
tion such as the skin, throat, and stool.
develop additional symptoms or begin to
* Chest radiograph if any pulmonary
appear unwell. If they become afebrile, their
symptoms are present.
neutropenia should be evaluated as outlined
* Daily blood cultures and a CBC with
above. Patients with fever and mild or mod-
differential every 24 hours in the persistently
erate neutropenia who have evidence of
febrile patient to help determine further
localized bacterial disease
(otitis media,
management.
sinusitis, or local skin infections) may be
* CT scan of the chest (þ / sinuses, abdo-
treated with appropriate oral antibiotics in
men, and pelvis) looking for evidence of
the outpatient setting. Patients with mild
occult infection in any patient remaining
neutropenia and evidence of bacterial pneu-
febrile for
>96 hours without a specific
monia, GU tract infections, lymphadenitis,
cause being identified. They should be
or systemic symptoms should have cultures
started on empiric antifungal coverage due
of the infection site and of the blood. They
to the increased risk of invasive fungal
may be treated with appropriate oral anti-
infection in this patient population.
biotics if their appearance is not concerning,
but should be seen back for reevaluation
Prophylactic antibiotics have been used in
within 24 hours. Similar patients with mod-
the past in an attempt to decrease the fre-
erate neutropenia can also be treated in the
quency of serious infections in patients with
outpatient setting if they appear totally well.
severe neutropenia. However, this has gen-
Febrile patients with moderate or severe
erally fallen out of favor. Exceptions include
neutropenia who appear in any way unwell
the use of prophylactic penicillin or amox-
require emergent medical assessment and
icillin in patients following splenectomy and
initiation of therapy, as do children who
the use of trimethoprim-sulfamethoxazole
are known to have severe forms of neutro-
prophylaxis to prevent Pneumocystis jiroveci
penia such as severe congenital neutrope-
(previously known as Pneumocystis carinii)
nia or Kostmann syndrome. These children
pneumonia in patients with T-cell dysfunc-
require:
tion in addition to neutropenia.
* Hospital admission.
The use of cytokines, particularly G-CSF,
* Coverage with broad-spectrum antibio-
has become increasingly popular in the
tics due to their increased risk of mortality
management of symptomatic neutropenia.
due to sepsis. These should provide cover-
It is particularly useful in patients with
age for Gram-negative and Gram-positive
symptomatic cyclic neutropenia or in the
organisms. A combination of an aminogly-
neutropenia-accompanying disorders such
coside and a b-lactam antibiotic provides
as glycogen storage disease
1b, as these
initial broad coverage and is synergistic for
patients typically respond well to low dose
Pseudomonas species, but several different
therapy (1 to 2 mcg/kg/dose) on a daily or
The Neutropenic Child
101
alternate day schedule with a goal of main-
she was discharged from the nursery at
taining an ANC between
1.0
and 1.5
24 hours of life. She is an only child. There
109/L. It is also used frequently in patients
is no family history of recurrent fevers, or
with severe chronic neutropenia. However,
severe or unusual infections. Her parents are
the doses required to raise the ANC into the
unaware of any “blood problems” in other
desired range vary significantly between
family members.
patients. A typical starting dose is
3
to
5 mcg/kg/day, and the dose is increased
2. What additional studies would you like
slowly until the desired ANC is achieved.
to obtain?
Patients with severe neutropenia who fail
to respond to doses as high as
50
to
Given her age and degree of fever, a sepsis
100 mcg/kg/day are considered poor respon-
workup is indicated. A CBC reveals a
ders. These patients are candidates for bone
WBC count of 12.4
109/L, a hemoglobin
marrow transplantation given their high like-
of
11.7 g/dL, and a platelet count of
lihood of mortality due to infection as well as
423
109/L. The differential reveals
0%
an up to 30% chance of developing myeloid
neutrophils,
73% lymphocytes, and 27%
malignancy.
monocytes. A urinalysis is normal. Because
of her severe neutropenia she is admitted to
the hospital and started on broad-spectrum
Case study for review
antibiotics. A blood culture is performed
and is negative. The vulvar lesion is cultured
A 7-week-old girl presents to your clinic for
and grows Pseudomonas aeruginosa. The
evaluation of a fever to 38.6
C. She had
lesion resolves with intravenous antibiotics.
been seen 4 weeks earlier for a well child visit
A bone marrow aspiration is performed
and was noted to have a small ulcer on her
and reveals an adequately cellular specimen
vulva. This was treated with oral antibiotics
with normal megakaryocytes and erythroid
and resolved. Her parents report that the
precursors. The myeloid lineage shows a
vulvar lesion had recurred 24 hours before
maturational arrest at the myelocyte stage,
she developed the fever. She had no blood
however.
work performed at the previous visit. Her
physical examination reveals her to be alert
3. What are you thinking now? What would
and nontoxic appearing. A 6-mm shallow
you do next?
ulceration is present on her vulva with
minimal surrounding erythema, drainage,
The isolated neutropenia in the face of a
or induration. It is mildly tender, however.
normal hemoglobin and platelet count
Her physical examination is otherwise
makes congenital aplastic anemia unlikely.
normal.
The young age at presentation, the signifi-
cant degree of neutropenia, and the unusual
1. What additional information would you
organism identified raise concern that this
like?
child may have a severe form of congenital
neutropenia. Once her active infection has
The patient was born at term via spontaneous
resolved and she is afebrile she can be dis-
vaginal delivery after an uncomplicated preg-
charged from the hospital, but should have
nancy. Her weight, height, and head circum-
regular follow-up to determine what her
ference were appropriate for gestational age.
neutrophil count does over time. More
Her nursery course was unremarkable, and
importantly, her parents need to understand
102
Chapter 11
the importance of bringing her immediately
family history, the autosomal recessive form
to you or to the emergency room if she
(Kostmann syndrome) would be more likely,
develops another fever.
although she may have a new ELA2 muta-
Twice weekly CBCs are performed over
tion. Genetic testing for HAX1 and ELA2
the next 6 weeks and reveal a consistent
mutations can be sent for confirmation.
pattern of severe neutropenia, with ANCs
A trial of G-CSF can also be initiated to see
ranging from 0 to 170. Serum immunoglo-
if it will result in an increase in the ANC.
bulins are measured and are normal, as are
T-cell subset studies. Over that time period,
Suggested Reading
she has several episodes of oral mucosal
ulcers.
Berliner N, Horwitz M, Loughran TP. Congenital
and acquired neutropenia. Hematol Am Soc
4. What would be your next step?
Hematol Educ Prog 63-79, 2004.
James RM, Kinsey SE. The investigation and
The persistent, noncyclic nature of her neu-
management of chronic neutropenia in chil-
tropenia provides further evidence that this
dren. Arch Dis Child 91:852-858, 2006.
infant is suffering from a form of severe
Klein C. Congenital neutropenia. Hematol Am
congenital neutropenia. Given the lack of a
Soc Hematol Educ Prog 344-350, 2009.
Thrombocytopenia
12
Thrombocytopenia, or low platelets, can
low platelet count and symptoms of muco-
occur as an isolated finding or in conjunc-
cutaneous bleeding. Most cases are consid-
tion with a multitude of underlying clinical
ered idiopathic whereas others are second-
conditions. Normal platelet counts range
ary to coexisting conditions. The diagnosis
from 150 to 400
109/L and thrombocyto-
of ITP is one of exclusion (see Table 12.1,
penia is generally defined as a platelet count
Figures 12.1, 12.2, and 12.3).
of less than 100
109/L. Thrombocytopenia
The most common cause of destructive
may further be defined as mild
(50
to
thrombocytopenia is autoimmunity. Short-
100
109/L), moderate (20 to 50
109/L),
ened platelet survival is due to platelet auto-
and severe
(<20
109/L). Other sources
antibody production (usually IgG), often
define thrombocytopenia as a platelet
stimulated by infection or drug exposure.
count of <150
109/L or in patients with
IgM antibodies and complement activation
a drop of >50% from their baseline platelet
are less frequently found but can also be seen
counts (if known). Thrombocytopenia is
in childhood ITP. It is an acute, self-limited
typically subdivided into immune and
disease of isolated thrombocytopenia that
nonimmune causes. Immune causes gener-
usually occurs in children aged 2 to 5 years
ally cause increased platelet destruction.
(though may occur at any age from infancy
Nonimmune causes may cause increased
to adolescence), typically resolving in more
destruction or decreased bone marrow
than 80% of children within 6 weeks to 6
production. In patients with splenomegaly,
months. When ITP occurs in the child over
platelet sequestration may also lead to
10 years of age, especially females, the course
thrombocytopenia.
may be chronic and associated with an
autoimmune disorder. The otherwise
healthy child presents with sudden onset
Acute immune thrombocytope-
of severe thrombocytopenia (usually < 20
nic purpura
109/L), manifested by petechiae and
purpura and less frequently mucosal bleed-
Immune thrombocytopenic purpura (ITP;
ing such as epistaxis, menorrhagia, hema-
also referred to as immune thrombocyto-
turia, and hematochezia. There is a history
penia or idiopathic thrombocytopenic pur-
of an antecedent viral illness within the past
pura) is an acquired, isolated disorder in
1 to 3 weeks in up to 60% to 70% of
which the patient typically presents with a
children. It may also follow vaccination with
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
104
Chapter 12
Table 12.1 Differential diagnosis of thrombocytopenia.
Destructive thrombocytopenias
Immunologic
ITP
Drug-induced (valproic acid, amphotericin B, digoxin)
Infection-induced (HIV, CMV, EBV, parvovirus B19)
Postvaccination (MMR, Varivax)
Posttransfusion purpura
Autoimmune disease (SLE, JIA)
Evans syndrome (AIHA/ITP)
Posttransplant
Hyperthyroidism
Lymphoproliferative disorders (ALPS)
Nonimmunologic
Microangiopathic disease
Cyclic thrombocytopenia
Hemolytic uremic syndrome
Thrombotic thrombocytopenic purpura
Platelet consumption/
Sepsis/DIC
destruction
Giant hemangioma (Kasabach-Merritt syndrome)
Cardiac (prosthetic heart valves, repair of
intracardiac defects)
Malignant hypertension
Neonatal problems
Neonatal alloimmune (NAIT)
Neonatal autoimmune (maternal ITP)
Pulmonary hypertension
Polycythemia
RDS
Infection (viral, bacterial, protozoal, spirochetal)
Sepsis/DIC
Prematurity
Meconium aspiration
Erythroblastosis fetalis (Rh incompatibility)
Maternal hypothyroidism
Impaired production
Congenital and
TAR syndrome
hereditary disorders
Fanconi anemia
Bernard-Soulier syndrome
Wiskott-Aldrich syndrome
Glanzmann thrombasthenia
MYH9 disorders (May-Hegglin anomaly)
CAMT
Rubella syndrome
Von Willebrand disease, type II
ATRUS
Dyskeratosis congenita
Agenesis of the corpus callosum
Associated with
Trisomy 13 or 18
chromosomal defect
Thrombocytopenia
105
Table 12.1 (Continued)
Metabolic disorders
Marrow infiltration
Malignancies (leukemia, neuroblastoma, metastatic
solid tumors)
Storage disease
Myelofibrosis
Acquired processes
Aplastic anemia
Liver disease/failure
Drug-induced
Radiation-induced
Severe iron deficiency
Sequestration
Hypersplenism (portal hypertension, neoplastic,
infectious, glycogen storage disease, cyanotic
heart disease)
Hypothermia
Abbreviations: ITP, immune thrombocytopenic purpura; SLE, systemic lupus erythematosis; JIA,
juvenile idiopathic arthritis; AIHA, autoimmune hemolytic anemia; ALPS, autoimmune
lymphoproliferative syndrome; DIC, disseminated intravascular coagulation; NAIT, neonatal
alloimmune thrombocytopenia; CAMT, congenital amegakaryocytic thrombocytopenia; TAR,
thrombocytopenia absent radii; ATRUS, amegakaryocytic thrombocytopenia radio-ulnar
synostosis; RDS, respiratory distress syndrome.
MMR or Varivax (live virus vaccines).
be associated with trauma. Bleeding into
Severe bleeding such as protracted epistaxis,
joints
(hemarthroses) should lead one to
hemoptysis, or gastrointestinal bleeding
consider an alternative bleeding disorder
leading to severe anemia and need for trans-
(e.g., congenital or acquired factor defi-
fusion is rare. Intracranial hemorrhage
ciency such as hemophilia). There should
(ICH) is the most feared complication with
be no history of unexplained fevers, bone
an estimated incidence of 0.1% to 0.5%;
pain, or weight loss which would be con-
more than 50% of patients with ICH present
cerning for an underlying malignancy or
with this finding at diagnosis or within
chronic infection. A medication history is
1 week of diagnosis. More than
75% of
critical as many drugs have been implicated
patients with ITP and ICH survive. It has
in inducing drug-mediated immune throm-
yet to be defined what risk factors predis-
bocytopenia. Implicated drugs include hep-
pose children with ITP to sustain this rare
arin, aspirin, aspirin-containing cold med-
but extremely serious complication.
ications, nonsteroidal anti-inflammatory
drugs (NSAIDs), and seizure medications
Evaluation
such as valproic acid. Chronic infections
The initial evaluation of the child with
with HIV, CMV, and hepatitis C may cause
suspected ITP begins with a complete his-
a low platelet count and appropriate screen-
tory and physical examination. Other than a
ing should be done. Helicobacter pylori has
possible antecedent illness and the acute
been implicated in adults with ITP and there
onset of minor bleeding and bruising, the
are some reports of this association in chil-
child should be otherwise clinically well
dren as well. Family history should include
appearing. More significant bleeding may
an assessment for autoimmune diseases,
106
Chapter 12
History, physical examination,
CBC, differential, platelet count,
peripheral smear evaluation
Thrombocytopenia with
neutropenia or pancytopenia
Consider
Marrow infiltration
Marrow failure
Hypersplenism
Assess
Bone marrow aspirate
Imaging for splenomegaly
(ultrasound/CT/MRI)
DDx:
Aplastic anemia
Fanconi anemia
Malignancy
Storage disease
Hypersplenism
Figure 12.1 Approach to the child with thrombocytopenia and additional cytopenias. Abbreviations:
CBC, complete blood count; DDx, differential diagnosis.
immune deficiency, or congenital syndromes
neutrophil count may be altered due to
associated with thrombocytopenia
(see
a recent infection, and the hemoglobin
Table 12.1). The physical exam should assess
may be low due to bleeding. Coagulation
for lymphadenopathy, hepatosplenomegaly,
screening tests including PT, PTT, and
short stature, dysmorphic features, or the
fibrinogen and the complete metabolic
presence of congenital anomalies
(e.g.,
panel should be normal. Therefore, no
radius, thumb, and fingers).
studies other than a CBC truly need to
The laboratory features of ITP include
be done if the diagnosis of ITP is strongly
a low platelet count in the face of an
suspected. The platelet count should be
otherwise normal complete blood count
confirmed to be low on a second evalua-
(CBC). However, the hemoglobin, white
tion if the physical findings do not corre-
blood cell count, and/or absolute
late with the laboratory value.
Thrombocytopenia
107
History, physical examination,
CBC, differential, platelet count,
peripheral smear evaluation
Isolated thrombocytopenia
Assess platelet morphology
Normal
Abnormal
Large platelets
Assess for splenomegaly
Negative
Congenital
and/or lymphadenopathy
anomalies
Bernard-Soulier syndrome
or
MYH9 disorders (May-Hegglin)
Jacobsen syndrome
Positive
Platelet-type von Willebrand
disease
Benign Mediterranean
macrothrombocytopenia
Bone marrow aspirate
Normal exam
Gray platelet syndrome
Small platelets
Wiskott-Aldrich syndrome
DDx:
ITP (see Table 12.1)
Viral-induced
DDx:
Drug-induced
Malignancy
HIV
Skeletal
Infection
Autoimmune disease
Fanconi anemia
HIV, EBV, CMV
Infant
TAR, ATRUS
Storage disease
Alloimmune (NAIT)
Cyanotic heart disease
Gaucher disease
Eczema
Autoimmune (maternal Ab transfer)
Hypersplenism
Hemangioma (may be visceral)
Wiskott-Aldrich syndrome
Aplastic anemia
Hemangiomas
Kasabach-Merritt syndrome
Fanconi anemia
Congenital amegakaryocytic
thrombocytopenia (CAMT)
Figure 12.2 Approach to the child with isolated thrombocytopenia. Abbreviations: CBC, complete
blood count; DDx, differential diagnosis; ITP, immune thrombocytopenia purpura; HIV, human
immunodeficiency virus; EBV, Epstein-Barr virus; CMV, cytomegalovirus; NAIT, neonatal alloim-
mune thrombocytopenia; Ab, antibody; TAR, thrombocytopenia absent radius; ATRUS, amegakar-
yocytic thrombocytopenia radio-ulnar synostosis.
If two cytopenias are present with any
megakaryocytes in normal or increased num-
findings on the history or physical suggestive
bers with normal erythroid and myeloid
of another diagnosis, consideration should
lineages. By convention, a BMA is done on
be given to performing a bone marrow aspi-
individuals with suspected ITP only if ster-
ration
(BMA). However, a bone marrow
oids are to be part of the treatment plan due
evaluation is rarely necessary to confirm
to the small chance the thrombocytopenia
the diagnosis. BMA in ITP typically demon-
may be an early manifestation of leukemia.
strates
the presence of immature However, even this practice is controversial.
108
Chapter 12
History, physical examination,
CBC, differential, platelet count,
peripheral smear evaluation
Thrombocytopenia with anemia
DAT test
no
Assess for intravascular
yes
(Coombs)
Microangiopathic
hemolysis, red cell
process
fragments
Consumption
negative
positive
DDx:
Reticulocyte
Evans syndrome
count
Autoimmune disease
Drug-induced
DDx:
DIC, sepsis
HUS
TTP
high
normal or low
CHD
Assess physical:
Bone marrow aspirate:
see Figure 12.2
see Figure 12.1
Figure 12.3 Approach to the child with thrombocytopenia and anemia. Abbreviations: CBC, complete
blood count; DAT, direct antiglobin test (Coombs); DDx, differential diagnosis; DIC, disseminated
intravascular coagulation; HUS, hemolytic uremic syndrome; TTP, thrombotic thrombocytopenic
purpura; CHD, congenital heart disease.
Additional laboratory evaluations may
Review of the peripheral blood smear
be required for specific clinical indications
confirms a low platelet count, and the few
(e.g., screening for autoimmune or thyroid
remaining platelets typically are large. If
disease, viral testing for HIV, hepatitis C,
platelet size is determined by an automated
CMV, or parvovirus B19; direct antiglobu-
cell counter (Coulter), it is usually elevated,
lin testing [DAT; Coombs] and reticulocyte
consistent with young platelet age (due to
count if concern for autoimmune hemolytic
early marrow release associated with rapid
anemia; and immunoglobulin levels and T/
peripheral platelet destruction). Oftentimes
B cell numbers if concern for underlying
the platelet count is underestimated as very
immunodeficiency syndrome). Antiplatelet
large immature platelets
(megakaryocytic
antibody tests are not sensitive or specific
fragments) are not counted by the Coulter.
and not required for diagnosis or for routine
This may in part explain the relative lack of
clinical management.
bleeding symptoms in children with very
Thrombocytopenia
109
low platelet counts. The erythrocyte and
and general anticipatory guidance such as
leukocyte counts and morphology should
the use of protective gear (helmets) and seat
be normal and there should be no evidence
belts. Intramuscular injections should be
of hemolysis or microangiopathic disease
withheld until platelet counts increase. Spe-
(e.g., schistocytes and/or burr cells).
cific medications that interfere with platelet
False values for platelet counts can result
function (such as NSAIDs, SSRIs [selective
from aggregation of platelets in the syringe
serotonin reuptake inhibitors], anticonvul-
or collection tube, counting of small non-
sants, and aspirin) should not be given.
platelet particles (fragmented red or white
The use of a bleeding score as the frame-
cells) by automated cell counters, and pseu-
work for a consistent treatment strategy is
dothrombocytopenia due to in vitro platelet
gaining in popularity and several scoring
agglutination by anticoagulant-dependent
systems have been published to date.
ethylenediaminetetraacetic acid
(EDTA)
Patients are categorized by the presence of
antibodies. Review of the blood film should
clinical signs and symptoms of bleeding;
assess for clumps of agglutinated platelets at
more than 80% present with mild clinical
the periphery of the slide.
bleeding (manifested by bruising, petechiae,
mild epistaxis, and no anemia). Moderate
Treatment
clinical bleeding is characterized by more
The natural history of acute ITP is com-
significant mucosal bleeding such as more
plete resolution in the majority of children.
severe epistaxis or menorrhagia and severe
Despite the relatively benign course experi-
bleeding
(usually
< 5% of patients) is
enced by most children with ITP, the sud-
marked by unusual or severe bleeding
den onset, concern for a possible serious
requiring hospitalization for control and/
condition such as leukemia or aplastic ane-
or blood transfusion support with symp-
mia, and concern for life-threatening bleed-
toms seriously interfering with quality of
ing often guide the initial treatment decision
life. All patients may present with very low
making. Parents often bring their child to
platelet counts (e.g., 1 to 20
109/L) though
see the primary care or emergency physi-
those with severe symptoms tend to have
cian, many of whom are not accustomed to
initial platelet counts < 10
109/L.
encountering such severe thrombocytope-
Utilizing the bleeding score, many
nia. Risk factors for the rare life-threatening
patients with clinically mild or moderate
hemorrhage have yet to be elucidated and
bleeding can be effectively managed with
dogma has been to treat the patient according
observation. The anxiety of patients,
to platelet count rather than being guided by
families, and other caregivers can often be
the degree of clinical symptoms. This clinical
alleviated with education on the pathophys-
scenario has led to significant controversy
iology and natural history of ITP in addition
as to whether intervention is warranted,
to general preventive measures and periodic
when it is warranted, and with what treat-
clinical and laboratory assessments. Platelet
ment. Contemporary therapies have not been
counts do not need to be checked more
shown to alter the course or outcome of
often than every 1 to 2 weeks (or even less
children with acute ITP. As such, the concept
often in the clinically stable child). Disrup-
of nontreatment in ITP is increasingly
tion of school or work is not necessary,
becoming the standard of care in most pedi-
though participation in contact sports or
atric hematology/oncology centers.
other situations that could lead to head or
All patients and families should be coun-
abdominal injury should be avoided until
seled regarding rough play, contact sports,
the platelet count is known to have returned
110
Chapter 12
to a safe levels (typically >50
109/L). With
premedication with acetaminophen is
observation, the majority of patients with
advisable. Rare side effects include anaphy-
ITP will have resolution of symptoms and
laxis in an IgA-deficient patient, aseptic
the underlying thrombocytopenia while
meningitis, acute renal failure, pulmonary
avoiding unnecessary costs and side effects
insufficiency, and thrombosis. Although
of medications and hospitalization.
IVIG is a pooled blood product, it is thought
Pharmacological treatments to raise
to be safe with regard to viral transmission
the platelet count in children with moder-
and does not require consent as with blood
ate-to-severe bleeding include two first-line
product administration. Subsequent dosing
therapies: intravenous immune globulin
with IVIG may be indicated depending on
(IVIG) and RhoGAM (anti-Rh [anti-D]
clinical symptomatology and continued
immune globulin, also Rhophylac). IVIG
thrombocytopenia.
is thought to function by saturating the
RhoGAM is a less-expensive alternative
Fc receptors on the reticuloendothelial cells
therapy to IVIG. The dose is 75 mg/kg IV
in the spleen and liver, thereby decreasing
over 20 to 30 minutes. Response rate and
the clearance of opsonized platelets. These
time to onset are similar to IVIG. Anti-D
treatments typically provide a quick yet
works by binding the RhD antigen expressed
transient increase in the platelet count, often
on the surface of red blood cells, leading to
sustained 2 to 4 weeks. More than 80% of
their recognition by Fc receptors on cells of
patients respond with an increase in the
the reticuloendothelial system. The coated
platelet count, which is sufficient to decrease
red cells are thought to compete with the
or eliminate bleeding symptoms. Patients
antiplatelet-antibody coated platelets for the
and families should be counseled as to the
activated Fc receptor sites, thereby slowing
transitory effect of IVIG treatment due to
platelet clearance. Side effects include fever
the misconception that a subsequent
and chills, intravascular hemolysis, head-
decrease in platelet count is indicative of
ache, emesis, and rarely anaphylaxis. Preme-
increased disease severity. The dose of IVIG
dication with acetaminophen and diphen-
is 0.8 to 1 g/kg over 4 to 6 hours. If neces-
hydramine may prevent these adverse effects
sary, IVIG may be repeated two to three
with subsequent infusions. A response is
times for a total dose of 2 to 3 g/kg; doses
typically seen within 24 to 48 hours. Patients
should be given at least 24 hours apart to
must be Rh positive, have a functional spleen
allow sufficient time to determine response.
(at least not known to be splenectomized
A rise in the platelet count is usually seen
or asplenic), and not be anemic (i.e., > 2 to
within 24 to 72 hours and peaks at approx-
4 g/dL below expected hemoglobin for age).
imately 9 days. A good initial response is an
RhoGAM does cause transient hemolysis
increase in platelet count by 20 to 50
109/L.
and a decrease in hemoglobin of
1
to
Side effects of IVIG are usually immediate,
2 g/dL over the ensuing 3 to 4 days should
relate to the rate of infusion, and include
be expected. The hemoglobin should subse-
nausea, lightheadedness, and headache. At
quently recover within 10 days. Though rare,
times, onset of a severe headache can
the U.S. Food and Drug Administration
prompt an emergent head CT to assess for
currently mandates that patients be moni-
potential intracranial hemorrhage. These
tored for 8 hours with serial urinalysis after
symptoms can be alleviated by slowing the
administration of anti-D due to cases of
rate of infusion and premedicating with
severe, life-threatening hemolysis.
diphenhydramine if further doses are
Amicar (e-aminocaproic acid) may be
needed. Fever may also occur, and
utilized in patients with prolonged
Thrombocytopenia
111
thrombocytopenia and persistent mucosal
Dexamethasone has also been used with
bleeding without anemia (mild bleeding).
efficacy in refractory patients.
Reports of efficacy with minimal side effects
make low-dose therapy a good option
Emergency therapy
(10 mg/kg/dose every 6 to 8 hours). Another
Patients with known or suspected life-
option is low-dose prednisone which may
threatening hemorrhage such as central
benefit the patient with a long course of ITP
nervous system hemorrhage from ITP (usu-
who secondarily develops alteration in vas-
ally with concomitant platelet count < 10
cular integrity. Low-dose steroids would not
109/L) should be treated emergently with
be expected to raise the platelet count; how-
combination therapy including IVIG 1 g/kg
ever, they would have minimal side effects.
daily for 2 to 3 days and intravenous high-
Corticosteroids may be used in the ini-
dose methylprednisolone 30 mg/kg/day (up
tial medical management of ITP, though
to
1 g/day) for
1 to 3 days. Active life-
typically are considered a second-line ther-
threatening hemorrhage with refractory
apy in pediatrics. Response is slower than
thrombocytopenia is one of the only current
with IVIG or RhoGAM and is usually within
indications for emergency splenectomy.
3 to 4 days. The first steroid prescribed is
Continuous infusion of platelets should be
prednisone,
1
to
2 mg/kg/day for
2
to
considered in an emergent situation and
4
weeks, tapering the dose post-platelet
during surgery or childbirth in a severely
response
(i.e., >100
109/L) over several
thrombocytopenic patient, in conjunction
weeks. The initial response rate is 70% to
with other therapies. Off-label recombinant
90%. An immune rebound may occur with a
factor VIIa (rFVIIa) may also be considered.
taper that is too rapid and patients will often
require prolonged steroid therapy to main-
Chronic immune thrombocytope-
tain the desired platelet count. There are
nic purpura
several mechanisms by which steroids affect
Acute and chronic ITP are defined based on
the platelet count: inhibition of phagocyto-
disease duration, with chronic ITP defined
sis of the antibody-coated platelets leading
as persisting for > 6 months. Approximately
to prolonged platelet survival; inhibition of
10% to 20% of children with acute ITP that
antibody production by B-lymphocytes;
persists will go on to have chronic ITP. The
improvement of capillary integrity reducing
child with chronic ITP is still likely to have
clinical bleeding; and an increase in platelet
complete resolution within
6 months to
production. Many patients respond to this
2 years or may have an associated auto-
treatment, although side effects may occur
immune disease or underlying immunod-
with repeated treatment or chronic use such
eficiency state. Some practitioners define
as gastritis, fluid retention, weight gain,
chronic ITP as that lasting beyond a 1 year
mood lability, acne, striae, high blood pres-
period. Most patients with chronic ITP do
sure, and elevated serum glucose. An alter-
not need treatment as the platelet count is
native to oral steroids is high-dose pulse
often above 20
109/L and bleeding is min-
infusion of intravenous methylpredniso-
imal. Platelet count alone does not correlate
lone. Some patients become thrombo-
with the risk of hemorrhage as platelets are
cytopenic again after therapy and require
large due to a healthy marrow response to
retreatment. A relapse may be managed
peripheral thrombocytopenia and, as a con-
safely by observation and restriction of
sequence, have greater than normal procoa-
activity and medications, or with inter-
gulant activity. In the rare patient with
mittent IVIG and/or pulse steroids.
chronic, refractory ITP who has clinical
112
Chapter 12
hemorrhage or cannot tolerate the living
decrement in humoral response, although
restrictions imposed by the thrombocytope-
decreased total IgG immunoglobulin levels
nia, intervention with medical or surgical
and increased infections have not been fre-
therapies should be considered.
quently reported. For the treatment of ITP
ITP may be the initial manifestation of
without an underlying immunodeficiency,
aplastic anemia including Fanconi anemia
it remains unclear if increased infection is a
before progression to pancytopenia, Evans
risk in pediatric patients after rituximab.
syndrome (association of ITP with autoim-
Although considered a T-cell-dependent
mune hemolytic anemia), systemic lupus
infection, B-cell depletion may lead to an
erythematosis (SLE), or autoimmune lym-
increased risk of PCP (Pneumocystis jiroveci
phoproliferative syndrome. HIV infection
pneumonia) and cotrixomazole prophylaxis
should be considered in all patients with
should be considered. Hepatitis B reactiva-
isolated thrombocytopenia as this is fre-
tion and progressive multifocal leukoence-
quently the first manifestation of disease
phalopathy secondary to JC virus are rare
in children. ITP may also be present in
considerations in this population.
children years before the diagnosis of
Splenectomy may be considered for
immunodeficiency (i.e., common variable
patients with chronic ITP with bleeding
immunodeficiency) is evident.
and/or limitation in activities negatively
Therapies utilized in the treatment of
impacting quality of life. Splenectomy is suc-
acute ITP may be utilized for chronic ITP,
cessful in 60% to 85% of patients; however,
though continued administration, cost, and
relapse of ITP may occur due to immune-
side effects frequently limit their use. Other
mediated platelet destruction in other organs,
considerations include additional medical
especially the liver. Patients should ideally
and surgical approaches.
receive appropriate immunization several
Rituximab is a chimeric human-mouse
weeks prior to surgery due to the risk of
monoclonal antibody directed against the
infection with encapsulated organisms
transmembrane CD20 antigen present on B
(pneumococcus, haemophilus influenza, or
cells, leading to apoptosis and antibody-
meningococcus). Lifelong penicillin prophy-
dependent cellular cytotoxicity. Children
laxis is recommended postsplenectomy.
with chronic ITP may have a 30% to 50%
Vinca alkaloids (vincristine and vinblas-
response rate with rituximab. The standard
tine), danazol (a virilizing androgen), and
dosage is
375 mg/m2 IV weekly infusion
immunosuppressive agents such as azathio-
(over
4 to
6 hours) for
4 weeks. Most
prine and cyclophosphamide have also been
patients start to show a response by the
used with some success. Ascorbic acid, cyclo-
second week, though delayed responses sev-
sporine, and interferon a-2b are other agents
eral weeks later may occur. Responses may
that have also been investigated for use in
be short-lived or last for years. Acute tox-
chronic ITP. Long-term management of
icity is minimal though patients may expe-
chronic ITP includes periodic assessments
rience fever and chills (often abates with
for development of other manifestations of
subsequent infusions), serum sickness,
immunodeficiency or autoimmune disease,
headache, nausea, emesis, and mucocutane-
counseling regarding activities and medica-
ous reactions (continuum from hives and
tions, and periodic assessments of the platelet
dermatitis to rarely reported Stevens-John-
count to assess for continuation or resolution
son syndrome and toxic epidermal necro-
of the underlying process.
lysis). B-cell depletion lasts for approxi-
The thrombopoietin
(TPO) mimetics
mately
6
months with a significant
are currently under investigation for the
Thrombocytopenia
113
treatment of childhood chronic ITP, follow-
with an HPA-1a-positive fetus). HPA-1a
ing successful clinical trials in adults. As ITP
resides on the GP IIb/IIIa complex that is
is likely the result of impaired production in
responsible for fibrinogen receptor activity
addition to peripheral immune-mediated
and thus important in aggregation and
destruction, these drugs may have some role
platelet-plug formation. Therefore, devel-
in treatment to stimulate increased produc-
opment of anti-HPA-1a antibodies may
tion, even in patients with normal throm-
interfere with normal platelet aggregation
bopoietin levels. Drugs currently under
in addition to decreasing platelet number,
investigation are romiplostim
(Nplate
)
resulting in a qualitative platelet defect as
and eltrombopag (Promacta
). Concerning
well as in addition to a quantitative one.
toxicities include marrow reticulin fibrosis
This may in part explain the high incidence
and thromboembolism. It is likely that
of serious bleeding in these infants as com-
these drugs or subsequent generations will
pared with those born to mothers with ITP
provide potential new therapeutic options
with alloantibodies directed against alter-
in the future.
nate antigens. HPA-1a is common in the
Caucasian population in addition to HPA-
5b. HPA-4 incompatibility is more common
Neonatal alloimmune
in Asian populations.
thrombocytopenia
The infant with NAIT typically presents
with moderate-to-severe thrombocytopenia
Neonatal alloimmune thrombocytopenia
(<20
to
50
109/L) and may rapidly
(NAIT) is caused by the transplacental pas-
develop petechiae, purpura, and bleeding.
sage of maternal alloantibodies directed
ICH is common, occurring in 10% to 20%
against fetal platelets, similar in pathophys-
of neonates and may occur prenatally or
iology to hemolytic disease of the newborn.
antenatally. Therefore, head ultrasound
Unlike hemolytic disease of the newborn,
should always be part of the initial diagnos-
NAIT often affects the first-born offspring.
tic workup. Complications of early central
Although rare, NAIT is the most common
nervous system hemorrhage include hydro-
cause of severe thrombocytopenia in the
cephalus, porencephaly, seizures, and fetal
first few days of life. Alloantibodies are
loss. Early jaundice occurs in 20% of cases
secondary to human leukocyte antigen
owing to resolution of intracranial or
incompatibility as fetal platelet antigens are
intraorgan hemorrhage. The infant should
inherited from the father. Immunization in
otherwise be generally healthy without other
the mother against fetal platelet antigens can
perinatal complications and the mother
occur during the current or a previous
should have an unremarkable hematologic
pregnancy or secondary to a previous plate-
history. For a mother with a previous low
let transfusion and may lead to severe
platelet count, the differential diagnosis
thrombocytopenia in the fetal-newborn
should include maternal idiopathic, auto-
period, with a high risk of fatal hemorrhage.
immune, and drug-dependent thrombocy-
Thrombocytopenia leading to fetal loss and
topenias, infection, and preeclampsia.
hemorrhage has been reported as early as 16
Infants may also be thrombocytopenic
to 24 weeks of gestation. Although several
secondary to birth asphyxia, infection,
platelet antigens have been implicated, the
congenital bone marrow hypoplasia, or
large majority of cases can be related to
from prematurity. The presence of hepatos-
human platelet antigen
1a
(HPA-1a)
plenomegaly, intrauterine growth retarda-
incompatibility
(HPA-1a-negative mother
tion, and/or intracranial calcifications with
114
Chapter 12
thrombocytopenia should suggest a congen-
every 6 to 8 hours, with IVIG is frequently
ital viral infection. However, if a secondary
effective. Regular head ultrasounds should
diagnosis is not clearly delineated, it is
be done while the infant remains severely
important to exclude alloimmune throm-
thrombocytopenic (<50
109/L). If intra-
bocytopenia by appropriate immunologic
cranial bleeding is present, platelets should
testing as future-affected pregnancies tend
be kept at
100
109/L. Head MRI is nec-
to be more severe and require close ante-
essary to further define the hemorrhage.
natal monitoring.
Ultrasound should be repeated at 1 month
Early platelet alloantigen evaluation of
in children with ICH to identify early
the newborn and parents is important, both
hydrocephalus. Resolution of thrombocyto-
in offering the affected infant appropriate
penia typically occurs within 2 to 6 weeks.
treatment and in order to minimize the risk
Mothers with an affected infant should
of devastating complications with future
be counseled on the need for aggressive
pregnancies. Evaluation includes platelet
monitoring with future pregnancies due
typing on the mother and father, specifically
to the risk of increasingly severe NAIT. Fetal
looking for antigens responsible for alloim-
platelet counts should be obtained starting
munization such as HPA-1a. Serum from
around 20 weeks of gestation, with ultra-
the mother and infant is screened for anti-
sound monitoring for hemorrhage. Mater-
platelet antibodies against either the infant’s
nal antiplatelet titers cannot be used to
or the father’s platelets. We send blood for
accurately predict affected fetuses. IVIG
the NAIT workup to the Blood Center of
1 g/kg/week given to the mother from
Wisconsin
mid-gestation until near term has been
Several treatment options are available
shown to effectively increase fetal platelet
for the infant with neonatal alloimmuniza-
counts in the majority of cases. Delivery
tion. Transfusion with antigen-negative pla-
should be planned near term with an elec-
telets has been the mainstay of treatment,
tive cesarean section or planned induced
when available. Since HPA-1a-negative pla-
vaginal delivery after documented increase
telets are present in only 2% of the Cauca-
in fetal platelet count following administra-
sian population, the most available source
tion of maternal IVIG. Antigen-negative
of platelets is from the mother. Random
platelets, which can be obtained from the
platelet transfusions (10 to 20 mL/kg) may
mother by apheresis, should be obtained
provide a transient increase, lasting 1 to 2
and prepared prior to delivery in the event
days, and should be used in cases of serious
of extreme thrombocytopenia or hemor-
hemorrhage, while antigen-negative plate-
rhage. The infant’s platelet count should
lets are being obtained and prepared. If
be checked at birth and every 6 to 12 hours
random donor platelets are ineffective, then
for 1 to 2 days and be kept
50
109/L.
cross-matched donor platelets or washed
Frequency of monitoring can be decreased
maternal platelets may be used if available.
depending on the stability of the platelet
An excellent alternative treatment is IVIG.
count and the clinical status of the infant.
The recommended dose is 1 g/kg/24 hours
for 1 to 2 doses (until the platelet count is
50
109/L). Platelet transfusion may still
Neonatal autoimmune
be necessary with IVIG if immediate cor-
thrombocytopenia
rection of the thrombocytopenia is needed.
Corticosteroids can be effective in reducing
Neonatal autoimmune thrombocytopenia
platelet destruction and increasing vascular
occurs due to the passive transfer of auto-
integrity. Methylprednisolone, 1 mg/kg IV
antibodies from mothers with ITP and may
Thrombocytopenia
115
be idiopathic, caused by other disorders
illness, organomegaly and sequestration,
including autoimmune diseases such as sys-
development of alloimmunization, sepsis,
temic lupus erythematosis, hypothyroidism,
and other medications. Other potential
and lymphoproliferative states, or second-
offending drugs include anticonvulsants
ary to medications. Unlike NAIT, these
such as valproic acid, chlorothiazides, estro-
antibodies recognize both maternal and
genic hormones, ethanol, ristocetin, and
fetal platelet antigens. Maternal ITP should
protamine sulfate.
be distinguished from gestational thrombo-
Heparin-induced thrombocytopenia
cytopenia that tends to occur late in preg-
(HIT) is caused by antibody formation to
nancy and leads to mild thrombocytopenia
complexes of heparin and platelet factor 4
in the mother
(e.g.,
70
to
100
109/L)
leading to platelet activation, often resulting
without the development of antibodies.
in severely low counts as well as risk for
Therefore, the infant is not at risk for
thrombosis. It is much less common in
thrombocytopenia. Maternal platelet counts
children than adults. HIT should be sus-
return to normal shortly after birth. The
pected in the child receiving heparin (usu-
degree of thrombocytopenia seen in neo-
ally secondary to unfractionated but can
nates born to mothers with ITP is less severe
also occur with low molecular weight) who
than with NAIT, with only 10% to 15%
develops unexplained thrombocytopenia of
having platelet counts <50
109/L. Bleed-
any degree within 5 to 10 days of exposure,
ing is minimal and ICH is rare (<1% to
often defined as a decrease of
50% from
2%). The platelet count may be normal at
baseline (even if still in the normal range).
birth but falls within 1 to 3 days of delivery.
Testing for heparin-induced antibodies is
Platelet counts should be monitored closely
not very specific as many patients without
and a head ultrasound obtained to exclude
HIT will have circulating antibody. If sus-
ICH. Treatment should be initiated if the
pected, based on clinical scoring that defines
platelet count falls below 30 to 50
109/L
likelihood, functional studies of platelet
with the same treatment regimen as for
activation under the presence of heparin
NAIT: IVIG, IV methylprednisolone, and
can be done by specialized laboratories.
random donor platelet transfusion if neces-
These tests include the serotonin release
sary for hemorrhage. The duration of
assay and heparin-induced platelet aggrega-
thrombocytopenia is usually 3 to 6 weeks.
tion assay. Heparin should be discontinued
and replaced by an anticoagulant that does
not lead to antibody formation such as
Drug-induced thrombocytopenia
argatroban. Warfarin should not be utilized
immediately due to associated protein C
In addition to immune-mediated mechan-
deficiency with risk of microthrombosis
isms induced by drugs, many bone marrow
leading to skin necrosis and gangrene.
suppressive agents such as chemotherapy
cause thrombocytopenia, though usually
in the face of pancytopenia. Management
Nonimmune thrombocytopenia
is usually with platelet transfusion, to pre-
vent or treat bleeding. The bone marrow
Many nonimmune-mediated processes lead
effects of these agents often define their
to increased platelet consumption. Gener-
dose-limiting toxicity, and thrombocytope-
alized platelet activation with trapping of
nia is a common effect. Patients usually
microaggregates in the small vasculature
respond well to platelet transfusion but can
contributes to microangiopathic hemolytic
become refractory because of the underlying
anemia (MAHA) occurring in congenital
116
Chapter 12
heart disease, hemolytic uremic syndrome
in the microvasculature that can also be seen
(HUS), and thrombotic thrombocytopenic
in the lymph nodes and spleen. Classic signs
purpura (TTP). HUS is primarily associ-
and symptoms include fever, malaise,
ated with a prothombotic state induced by
nausea and vomiting, abdominal and chest
exposure to shiga-toxin producing Escher-
pain, arthralgia and myalgia, pallor, jaun-
ichia coli
(usually O157:H7), particularly
dice, purpura, progressive renal failure, and
affecting the renal vasculature and leading
fluctuating neurologic signs and symptoms.
to the triad of MAHA, platelet consump-
Laboratory features include thrombocyto-
tion, and renal failure. HUS is the most
penia and MAHA. The peripheral blood
common cause of acute renal failure in
smear will show polychromasia, basophilic
children. Patients may present with abdom-
stippling, schistocytes, microspherocytes,
inal pain and bloody diarrhea and are trea-
and nucleated red cells. The DAT (Coombs)
ted with supportive care (i.e., red cell trans-
should be negative as TTP is not an auto-
fusions, dialysis as necessary). Platelet trans-
immune hemolytic anemia. The LDH and
fusion may worsen the clinical status and
unconjugated bilirubin will be elevated and
should be used with caution.
haptoglobin reduced due to the MAHA,
Idiopathic TTP is a rare disease in chil-
with associated hemoglobinuria. Without
dren, characterized by the pentad of throm-
treatment, mortality is >90%. Plasmaphe-
bocytopenia, hemolytic anemia, renal
resis is the mainstay of therapy in the
impairment, neurologic symptoms, and
acquired form. Fresh frozen plasma is usu-
fever, although few patients present with
ally sufficient to treat the congenital form.
the full gamut of symptoms. Idiopathic TTP
Patients may also benefit from rituximab
is often clinically indistinguishable from
and other immunosuppressive drugs
diarrhea-negative HUS. There is also a rare
including steroids, cyclosporine, cyclophos-
congenital form in which affected neonates
phamide, vincristine, and azathioprine.
present with jaundice and thrombocytope-
Increased utilization of platelets may
nia, although patients may not have an
occur in active bleeding, infection, or sepsis.
episode of overt TTP for years until triggered
In disseminated intravascular coagulation
by infection, pregnancy, or stress. Patients
(DIC), there is an imbalance between intra-
with congenital TTP have low levels of
vascular thrombosis and fibrinolysis, with
ADAMTS13, a protein that cleaves unusually
increased platelet consumption, depletion
large multimers of von Willebrand factor
of plasma clotting factors, and formation
into a biologically less-active form. Absence
of fibrin. DIC can be initiated by many
of ADAMTS13 inhibits cleavage of these
events, including sepsis due to bacteria,
large multimers allowing spontaneous plate-
viruses, or fungi; malignancy, particularly
let adhesion and aggregation.
acute promyelocytic leukemia and neuro-
Patients with acquired TTP, which is
blastoma; hemolytic transfusion reactions;
often idiopathic, commonly demonstrate
and trauma. Therapy is aimed at treating
antibodies to the protein, unlike the con-
the underlying etiologic process. Supportive
genital form in which there is a constitutive
care consists of platelet transfusion to
deficiency. Affected individuals are more
maintain platelet counts >50
109/L and
commonly female, of African descent, and
plasma protein replenishment to correct
diagnosis can be associated with pregnancy,
coagulopathies and maintain fibrinogen
autoimmune disease, infection, or trans-
>100 mg/dL.
plantation. The hallmark of disease is the
Thrombocytopenia can occur in the sick
presence of segmental hyalin microthrombi
newborn for many reasons, most
Thrombocytopenia
117
commonly with infection, prematurity,
Interferon a-2a has been shown to be ben-
asphyxia, respiratory distress syndrome,
eficial in correcting the platelet count and
pulmonary hypertension, or meconium
shrinking the lesion. Recent fortuitous dis-
aspiration. These infants appear to have
covery of propanolol as a treatment for
normal to increased platelet production, but
hemangiomas has now made it first-line
a decreased platelet life span for reasons that
therapy; although not clearly delineated, the
are unclear. Thrombocytopenia is a fre-
mechanism of action is likely related to the
quent occurrence in congenital cyanotic
inhibition of angiogenesis. Supportive
heart disease associated with compensatory
transfusion therapy is indicated with active
polycythemia. Therapeutic phlebotomy
bleeding due to thrombocytopenia. Platelet
may lessen the thrombocytopenia.
transfusion as well as infusion of coagula-
The association of thrombocytopenia
tion factors (fresh frozen plasma, cryopre-
and giant hemangiomas occurs in the
cipitate, and antifibrinolytic drugs) may be
infant with Kasabach-Merritt syndrome
helpful but usually are of only transitory
and represents a form of localized intravas-
benefit. Antiplatelet medications
(aspirin
cular coagulation. The hemangiomas may
and dipyridamole) have been used in the
be multiple and may involve only viscera.
past to interfere with platelet trapping
Therefore, in an infant with unexplained
within the hemangioma but carry the risk
thrombocytopenia, imaging studies should
of causing platelet dysfunction in addition
be done to look for a vascular anomaly.
to the existing thrombocytopenia.
Hemangiomas are proliferative lesions that
A variety of conditions that result in
grow rapidly for several months and then
splenomegaly are associated with throm-
regress spontaneously. Platelet thrombi may
bocytopenia. The large spleen sequesters
develop in these lesions and platelet life span
and destroys circulating platelets. Anemia,
may be decreased. These infants may also
leukopenia, and neutropenia may also be
have a consumptive coagulopathy with low
present. Megakaryocytic production in the
fibrinogen levels and elevated concentra-
marrow is normal, and may be accelerated
tions of fibrin degradation products. The
in response to a decrease in the circulation.
lesions are also prone to necrosis and infec-
Storage diseases, early portal hyperten-
tion. A particular hemangioma’s size or
sion, hemolytic conditions (red cell mem-
location cannot predict whether it will lead
brane defects), infections such as with
to platelet trapping and thrombocytopenia.
HIV, EBV, and CMV, and malignancies
These infants should be managed by close
are frequently associated with splenomeg-
observation and hematologic monitoring
aly and may result in hypersplenism
while waiting for regression to occur. How-
(increased splenic activity and resultant
ever, the lesions may become large enough
red cell destruction).
to compromise the infant by impinging on
the airway or vital organs, leading to com-
partment syndrome, and resulting in serious
Decreased platelet production
illness or death. External compression of
hemangiomas by firm bandaging, when
Thrombocytopenia due to decreased produc-
possible due to location, may reduce blood
tion may be a result of an acquired or inher-
flow and platelet trapping. Corticosteroid
ited disease process. Decreased production
treatment at a dose of 1 to 2 mg/kg/day may
may be a direct effect of marrow crowding
bring about regression of the lesion and
due to malignancy (leukemia or metastatic
normalization of the platelet count.
solid
tumors such as lymphoma,
118
Chapter 12
neuroblastoma, medulloblastoma, and
difficult to appreciate small platelets in the
rhabdomyosarcoma) or storage diseases
newborn and the mean platelet volume
(Gaucher, Neimann-Pick, etc.). Drugs may
reported on the CBC is unreliable in the
be implicated in decreased production. The
face of thrombocytopenia.
liver is the site of TPO production and liver
Patients with inherited thrombocytope-
disease is associated with chronic severe
nia may have normally sized platelets in
thrombocytopenia. Severe iron deficiency
certain conditions including congenital ame-
can result in decreased production, though
gakaryocytic thrombocytopenia
(CAMT),
early iron deficiency states are associated with
thrombocytopenia with absent radii (TAR),
an elevated platelet count likely due to mar-
familial platelet disorder and predisposition
row stress. Diseases affecting the marrow
to AML (acute myelogenous leukemia), ame-
matrix (aplastic anemia and myelofibrosis)
gakaryocytic thrombocytopenia with radio-
cannot support stem cell growth and matu-
ulnar synostosis (ATRUS), and autosomal
ration with resultant thrombocytopenia.
dominant thrombocytopenia. CAMT, a rare
Thrombocytopenia related to an inher-
cause of neonatal thrombocytopenia, is a
ited condition is frequently distinguished
bone marrow failure syndrome inherited in
by characteristic clinical features, early
an autosomal recessive manner due to defi-
presentation and chronic course, family
ciency in the TPO receptor c-mpl. Bleeding
history, platelet morphology, and lack of
symptoms lead to diagnosis in infancy
response to classic treatments for ITP. A
although CAMT is often initially confused
number of these conditions are associated
with other more common neonatal causes of
with macrothrombocytes and mild-to-
thrombocytopenia such as alloimmune- and
moderate thrombocytopenia including
autoimmune-mediated processes. However,
Bernard-Soulier syndrome, MYH9-related
unlike these other conditions, the thrombo-
disorders
(May-Hegglin anomaly with
cytopenia does not resolve with time. There
bluish cytoplasmic inclusions including
are no classic physical features. Diagnosis is
Sebastian, Fechtner, Epstein, and Alport-
based on markedly reduced megakaryocytic
like syndromes), platelet-type von Willeb-
precursors in the bone marrow with normal
rand disease, gray platelet syndrome (stor-
erythroid and myeloid lineages with elevated
age pool disease), benign Mediterranean
TPO levels. Current treatment is supportive
macrothrombocytopenia, Paris-Trousseau
care, platelet transfusion as needed, and con-
type thrombocytopenia, and more poorly
sideration for curative hematopoietic stem
defined syndromes such as Montreal plate-
cell transplantation. Gene therapy is being
let syndrome. Microthrombocytes are seen
developed.
in Wiskott-Aldrich syndrome, an X-linked
Children with ATRUS present similarly
disorder resulting from a mutation on the
to CAMT with severe thrombocytopenia at
WAS gene. Wiskott-Aldrich syndrome is
birth, but with the addition of associated
characterized by thrombocytopenia, recur-
skeletal anomalies and sensorineural hear-
rent bacterial and viral infections secondary
ing loss. Skeletal abnormalities are fusion of
to T-cell dysfunction, chronic eczema, and a
the radius and ulna at the elbow, often
propensity to develop autoimmune disor-
associated with minor clinodactyly. The
ders. Patients with defects in the WAS gene
disease may progress to aplastic anemia
may also have a milder syndrome called
or leukemia. TAR syndrome is also thought
X-linked thrombocytopenia with small
to be secondary to a defective response to
platelets and immune dysregulation which
TPO with variable thrombocytopenia and
may develop over time. Of note, it is
normal erythroid and myeloid lineages.
Thrombocytopenia
119
Mostcasesarediagnosedatbirthorinutero
3. What findings do you look for on exam-
due to bilateral absence of the radii man-
ination to either confirm or dispute your
ifested as a shortening of the forearms
suspected diagnosis?
and flexion at the elbows. The thumbs are
present, which distinguish TAR from the
Obviously, the first concern is to try to stop
skeletal anomaly associated with Fanconi
the bleeding and treat him for acute, symp-
anemia. Patients may have additional plate-
tomatic hemorrhage. You suspect a bleeding
let dysfunction and are at risk for significant
disorder and see this is an otherwise healthy
bleeding episodes. Typically, the thrombocy-
child with no known inherited bleeding
topenia improves through childhood. Asso-
diathesis. Although ITP presents with acute
ciated clinical features include additional
massive bleeding in fewer than 5% of cases,
skeletal limb defects, renal and cardiac anom-
you are appropriately concerned that he has
alies, facial capillary hemangiomas, and cow’s
mucosal type bleeding associated with either
milk intolerance. Fanconi anemia is an in-
an acquired or inherited platelet problem,
herited disorder characterized by chromo-
which may be qualitative or quantitative.
somal instability with skeletal anomalies
You order a STAT CBC to determine both
and hypoproductive thrombocytopenia,
platelet and hemoglobin levels and initiate
although other cell lines are eventually
fluid resuscitation awaiting the results.
affected. These patients have an increased
Additional historical points to elicit
susceptibility to develop leukemia and may
include recent illness, medication exposure,
benefit from early hematopoietic stem cell
underlying disease, and prior symptoms.
transplantation if a suitable donor is available.
You are contemplating other diagnoses such
as von Willebrand disease and acute leuke-
mia and ask questions related to these pos-
sibilities. In this case, the mother states that
Case study for review
he has been healthy, with no prior medica-
tions, illnesses, hospitalizations, or other
A 9-year-old previously well Hispanic boy
comorbidities. There is no family history
presents to the emergency department
of a diagnosed bleeding disorder (though
with a 2-hour history of profuse epistaxis.
you make a mental note to pursue these
On presentation he is noted to be well-
questions later as family members may not
developed and well-nourished, frightened,
have been evaluated for symptoms sugges-
fatigued, and pale, with numerous ecchy-
tive of von Willebrand disease). The mother
moses and blood pouring from both
states that he on awakening this morning
nares. He is emergently triaged and has
had unexplained bruises over his torso and
bilateral nasal packing placed. He con-
extremities and blood blisters in his mouth.
tinues to have massive bleeding and has
He complained of feeling sick to his stom-
repeated emesis of digested and bright red
ach and began to have simultaneous emesis
blood. His vitals are T 37.2 C, HR 156, RR
and nose bleeding, which led to her calling
30, and BP 62/38.
an ambulance as she could not stop it.
1. What are your immediate thoughts
On examination, you note he is alert and
regarding possible diagnosis? How do you
oriented, though frightened to see so much
confirm this and what steps do you take in
blood and expresses to you he thinks he is
treatment and stabilization of your patient?
going to die. You provide reassurance and
2. What pertinent history should you
quickly note his oral and skin findings of
obtain?
diffuse petechiae and purpura. Purpuric
120
Chapter 12
lesions on the torso are very unusual and
(1 pheresed unit every 2 to 3 hours), FFP
frequently not associated with trauma, reflect-
infusion, further doses of methylpredniso-
ing more spontaneous bleeding. He has no
lone, and a second dose of IVIG. On day 3,
adenopathy, organomegaly, other skin rashes,
the hemoglobin drops to 4.6 g/dL and the
and no pain on palpation of the extremities
decision is made to perform an emergent
and abdomen with full range of motion of all
splenectomy. The family is counseled that he
joints, and in general appears well developed
may not survive this procedure. Fortunately,
and appropriate for age. All these findings are
however, he does very well and is supported
supportive of a diagnosis of ITP.
with transfusions without a worsened clinical
Your patient’s CBC comes back with a
course. Unfortunately, there is no immediate
platelet count of 2
109/L, hemoglobin 6.2 g/
response to splenectomy. Typically, in
dL, and white blood cell count of 7.4
109/L
patients that do respond there is an imme-
with a normal differential. You explain the
diate increase in postoperative platelet count,
anemia as secondary to massive epistaxis.
often to above normal levels due to an exag-
Your patient now has evidence of gross
gerated marrow response that persists after
hematuria and melena, with a further drop
the source of peripheral destruction or
in blood pressure despite having received a
sequestration has been removed. Splenec-
unit of packed red blood cells and volume
tomy is not successful in
30% of cases and
support. The complete metabolic panel and
it cannot be predicted who will respond. Our
coagulation studies are normal.
patient continues to receive massive blood
product support for one week, then starts to
4. What is your next thought for treatment?
stabilize with an increase in hemoglobin to
5. How do you counsel the family?
8 g/dL and platelet count of 54
109/L. He
continues on a course of steroids, eventually
As you are now certain that this is acute ITP
transitioning to oral prednisone, and taper-
and your patient is experiencing life-threat-
ing off by 6 weeks. By this time his platelet
ening hemorrhage, you initiate therapy
count normalizes, and several years later he
with IVIG 1 g/kg IV over 4 hours, methyl-
continues to have a normal platelet count.
prednisolone 1 mg/kg IV every 8 hours,
Of interest, the patient re-presents with
continuous infusion of packed red blood
moderate epistaxis, no anemia, and a nor-
cells, and fluid support with Lactated
mal platelet count 1 year later. Further diag-
Ringer’s or normal saline. Consideration
nostic workup reveals a diagnosis of type 1
should also be given to continuous infusion
von Willebrand disease.
of platelets. Consultation with an ENT
surgeon should be obtained for nasal pack-
6. How would the knowledge of this
ing and control of the bleeding in the
comorbidity have influenced your manage-
posterior pharynx.
ment of the patient with the initial presen-
The prognosis of ITP, even in the face of
tation of ITP and massive epistaxis?
such a dramatic presentation, is excellent.
However, this situation is tenuous due to
Knowledge of this disease may have led
lack of therapies that could cause an imme-
to the use of additional therapies such
diate rise in the platelet count. At this time,
as DDAVP to stimulate release of von
his prognosis is guarded.
Willebrand factor, and Humate-P, a
Your patient is hospitalized in the PICU
Factor VIII concentrate that also contains
and continues to received red cell trans-
von Willebrand factor and is used for von
fusions, continuous platelet transfusion
Willebrand factor replacement. It is
Thrombocytopenia
121
unknown whether such additional therapies
Israels SJ. Diagnostic evaluation of platelet func-
may have impacted the clinical course.
tion disorders in neonates and children: an
update. Semin Thromb Hemost 35:181-188,
2009.
Neunert CE, Buchanan GR, Imbach P, et al.
Suggested Reading
Severe hemorrhage in children with newly
diagnosed immune thrombocytopenic pur-
Buchanan GR. Bleeding signs in children with
pura. Blood 112:4003-4008, 2008.
idiopathic thrombocytopenic purpura. J
Neunert C, Lim W, Crowther M, et al. The
Pediatr Hematol Oncol 25:S42-S46, 2003.
American Society of Hematology 2011 evi-
Bussel JB, Sola-Visner M. Current approaches to
dence-based practice guidelines for immune
the evaluation and management of the fetus
thrombocytopenia. Blood
117:4190-4207,
and neonate with immune thrombocytope-
2011.
nia. Semin Perinatol 301:35-42, 2009.
Segel GB, Feig SA. Controversies in the diagnosis
Cines DB, Bussel JB, Liebman HA, Luning Prak
and management of childhood acute immune
ET. The ITP syndrome: pathogenic and clin-
thrombocytopenic purpura. Pediatr Blood
ical diversity. Blood 113:6511-6521, 2009.
Cancer 53:318-324, 2009.
Evaluation of the Child
13
with a Suspected
Malignancy
Each year approximately 15,000 children
The history is the first step in the diag-
and adolescents under 20 years of age are
nostic process, with the chief complaint
diagnosed with cancer in the United States.
providing the most important clue. Most
The likelihood of a young adult having a
of the symptoms of childhood cancer are
history of childhood cancer is approxi-
either due to a mass and its effect on the
mately 1 in 300. Although cancer remains
surrounding tissues, invasion of the mar-
the leading cause of death in children except
row, or secretion of a substance by the
for accidents, survival continues to steadily
tumor that disturbs normal function. A
increase. In adolescents, cancer deaths are
careful family history should be elicited and
less common than those caused by acci-
include familial cancers. Certain conditions
dents, homicide, and suicide. More than
can predispose to malignancy such as
80% of children diagnosed with cancer are
genetic diseases
(e.g., Down syndrome,
now expected to be cured of their disease.
Beckwith-Wiedemann syndrome, neurofi-
That being said, cancer remains a devastat-
bromatosis), prior history of a malignancy,
ing diagnosis. The initial approach to the
or radiation therapy. Environmental and
child and family must be with a heightened
genetic factors have been associated with
sensitivity to the emotional impact. Once a
the development of malignancy; genetic
diagnosis of cancer is suspected, an imme-
factors are known to play a significant role
diate and thorough evaluation should
in the development of pediatric cancer,
proceed.
whereas environmental factors are postu-
Initial symptoms of cancer may be some-
lated to play a role in the increasing inci-
what elusive, given the subtle and overlap-
dence of certain cancers.
ping symptoms and signs that may be pres-
Timely diagnosis is critical though can be
ent in both malignant and nonmalignant
difficult due to the nonspecific symptoms
disease. Many pediatricians and clinicians
and rarity of the diseases. Though highly
may only see a new case of childhood cancer
curable in many circumstances, earlier diag-
every 5 to 7 years, and each case may be so
nosis may play a factor in improved prog-
unique as to not allow for the increased
nosis. Consideration should be given to the
awareness of this possibility.
nature of the complaint by the child and
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Evaluation of the Child with a Suspected Malignancy
123
Table 13.1 Presenting signs and symptoms of some common pediatric cancers and their
differential diagnoses.
Presenting signs or
Common diagnoses
Potential malignancy
symptoms
(nonmalignant conditions)
Headache, morning
Migraine, sinusitis
Brain tumor
vomiting
Lymphadenopathy
Infection
Lymphoma, leukemia
Bone pain/limping
Infection, trauma, growing pains
Bone tumor, leukemia,
neuroblastoma
Abdominal mass
Constipation, kidney cyst, full
Wilms tumor, neuroblastoma
bladder
Extremity mass
Cyst, infection, trauma
Bone tumor, soft tissue sarcoma
Mediastinal mass
Infection, cyst
Lymphoma, leukemia
Pancytopenia
Infection
Leukemia
Bleeding
Coagulation disorders, platelet
Leukemia (APL), neuroblastoma
disorders, ITP
Back pain
Trauma
Leukemia, lymphoma, CNS
tumor, extension of abdomi-
nal tumor into spinal cord
Chronic ear drainage
Otitis media/externa
Langerhans cell histiocytosis
Feminizing/masculinizing
Precocious puberty
Adrenocortical carcinoma, brain
symptoms
tumor, germ cell tumor
Abbreviations: APL, acute promyelocytic leukemia (AML M3); ITP, immune
thrombocytopenic purpura; CNS, central nervous system.
family, potential explanations for the com-
changes in the quality, frequency, and pat-
plaint (or lack thereof), persistence of symp-
tern of the headaches. Associated neurologic
toms, and lack of response to common
symptoms or signs should warrant an emer-
interventions (see Table 13.1).
gent evaluation for a tumor in the central
Headache is one of the most common
nervous system. These include, but are not
complaints in the pediatric population, the
limited to, localizing symptoms such as
majority of which are attributable to non-
focal weakness, cranial nerve palsies, ataxia,
malignant conditions. Although few head-
failure to thrive, and developmental delays
aches are caused by intracranial masses,
or regression. A thorough neurologic
primary brain tumors or metastases must
examination, including evaluation for
be ruled out when dealing with a patient
papilledema, should precede radiographic
with repeated or persistent headaches.
imaging. Other important historical points
Headaches are often the first symptom of
include an assessment of changes in visual
a brain tumor and, dependent on location,
acuity, behavior, and academic perfor-
are frequently accompanied by more subtle
mance. MRI is recommended to evaluate
findings on neurologic examination. His-
for the presence of an intracranial mass in
tory should include questions to character-
children with symptoms suggestive of a
ize the headache including worrisome signs
brain tumor. CT may be appropriate as
such as recurrent morning headache, head-
an initial test in certain settings depending
ache that awakens the child, intense inca-
on the availability of MRI. CT is a good,
pacitating headache, associated vomiting, or
rapid tool for the assessment of acute
124
Chapter 13
Table 13.2 Conditions suggesting need for
concerning for malignancy as is a unilateral
brain imaging in children with headache.
location. Nodes that are hard, nonmobile,
and nontender are worrisome for malig-
Presence or onset of neurologic abnormality
nancy. Nodes associated with fluctuance,
Ocular findings such as papilledema,
tenderness, warmth, or overlying erythema
decreased visual acuity, or loss of vision
are more consistent with infection. How-
Vomiting that is persistent, increasing in
frequency, or preceded by recurrent
ever, these are only generalizations as rap-
headaches
idly growing malignant nodes may be ten-
Change in character of headache such as
der and infectious nodes may be quite firm
increased severity and frequency
and nonmobile.
Recurrent morning headaches or headaches
Important historical points to elicit
that repeatedly awaken child from sleep
include:
Short stature or deceleration of linear growth
* Duration of lymphadenopathy or local-
Precocious puberty
ized mass
Diabetes insipidus
* Presence of a recent infectious illness
Age 3 years or less
* Skin lesions, cuts, or abrasions (and rela-
Neurofibromatosis
History of acute lymphoblastic leukemia with
tionship to nodal drainage patterns)
irradiation of central nervous system
* Recent immunizations
* Medications
* Animal contact (e.g., cat scratch, rodent
hemorrhage or increased intracranial pres-
bite, tick bite)
sure (see Table 13.2).
* Recent transfusion
Lymphadenopathy is a common finding
* Travel
on examination in children. Enlarged lymph
* Possible sexually transmitted infection
nodes are a frequent presenting sign in
* Presence of symptoms such as arthralgias,
association with malignancies or infection,
weight loss, and night sweats
and differentiation requires a meticulous
and systematic approach. Lymph nodes
The differential diagnosis of lymphadenop-
become large in response to infection or
athy includes infectious etiologies (viral, bac-
infiltration. Generally, a lymph node is con-
terial, spirochetal, and protozoan), connec-
sidered enlarged if it measures greater than
tive tissue disease, hypersensitivity states,
10 mm, although inguinal nodes may be
lymphoproliferative disorders, immunodefi-
15 mm or greater before being considered
ciency states, storage diseases, and malig-
worrisome and epitrochlear nodes are con-
nancy. In infants, the differential diagnosis
cerning once greater than 5 mm. In addition
of head and neck lumps includes lymphade-
to size, other factors to consider in establish-
nopathy and congenital malformations such
ing a differential between infection and
as cystic hygroma, thyroglossal duct cyst,
malignancy include persistence or rate of
branchial cleft cyst, epidermoid cyst, and
growth, quality of the node, location and
neonatal torticollis. Localized adenopathy
distribution
(e.g., adenopathy that is
may be infectious in origin including bacte-
symmetric, localized, regional, or dissemi-
rial causes such as Staphylococcus aureus,
nated), and presence of other signs or symp-
beta-hemolytic strep, cat-scratch disease
toms of infection. Adenopathy in the supra-
(Bartonella henselae), and tuberculous and
clavicular, axillary, or epitrochlear areas is
nontuberculous mycobacteria, in addition to
considered an abnormal finding. Adenopa-
nonbacterial causes such as HIV, EBV, cyto-
thy that persists longer than
6 weeks is
megalovirus (CMV), and toxoplasmosis.
Evaluation of the Child with a Suspected Malignancy
125
The physical examination of the child
as indicated by history and examination. A
with lymphadenopathy includes an assess-
biopsy should be done on enlarging or
ment of the size, location, and quality of the
persistently large nodes or if adenopathy
node(s). A full examination should include
is also seen on chest radiography. It is
assessment of the skin and mucocutaneous
recommended that excisional biopsies
tissues draining to the particular node in
(i.e., removal of intact node) be performed
question, and evidence of other signs related
when malignancy is suspected to evaluate
to an underlying disease process such as
the architecture of the node in addition to
hepatomegaly and splenomegaly. Nodes
the cellular infiltrate. The largest node
should be measured in largest diameter, the
should be biopsied when possible, with
quality should be assessed as to mobility,
avoidance of the upper cervical and ingui-
firmness, tenderness, and overlying skin
nal areas. Studies to be performed on the
changes, and these findings should be docu-
tissue include Gram stain and culture (bac-
mented. Children with mildly enlarged
terial, mycobacterial, viral, and/or fungal);
nodes should be monitored with frequent
histology and immunohistochemistry
examinations, and when not associated with
for suspected malignancy; and if malig-
malignancy, most of these nodes will revert
nancy is confirmed, flow cytometry and
to normal size.
specific cytogenetic testing for further
If infection is considered the cause of
classification.
the adenopathy, as in localized cervical
Splenomegaly is the finding of a palpa-
adenitis, it is reasonable to treat the patient
ble spleen edge on examination. A 1 to 2 cm
with a 2 week course of antibiotics. Depen-
splenic tip is found in 30% of full-term
dent on the history, physical, and clinical
neonates and in as many as 10% of healthy
suspicion, a particular antibiotic regimen
children. Approximately
3% of healthy
can be chosen in addition to sending serol-
college students have palpable spleens.
ogy or further testing for likely organisms.
Therefore, this finding on a routine physical
A baseline complete blood count (CBC)
examination of an otherwise healthy child
with differential and peripheral blood
should not create great concern. Children
smear should be done if there is any con-
with other signs of systemic disease,
cern for a diagnosis other than infection.
however, should have their splenomegaly
The patient should be seen again following
evaluated.
completion of the antibiotics and if the
Splenomegaly is associated with many
node has not changed or has increased in
disease states, both congenital and acquired:
size, consideration should then be given to
* Hemolytic anemia: hereditary spherocy-
biopsy. Many cases of infection-related
tosis, thalassemia, splenic sequestration in
adenopathy resolve spontaneously in 2 to
sickle cell disease, and autoimmunity
6 weeks or as a result of antibiotic treat-
* Immunological disease: common variable
ment (see Figure 13.1).
immune deficiency, connective tissue dis-
For the large node (i.e., >2.5 cm in size)
orders, and autoimmune lymphoprolifera-
or in the patient without response to anti-
tive disease
biotics, further evaluation is required.
* Infection: viral (EBV, CMV, HIV, hepa-
Additional studies should include a PPD
titis), bacterial (tularemia, abscesses, tuber-
(or quantiferon gold), chest radiograph,
culosis, infective endocarditis), spirochetal,
CBC with differential (if not already done),
protozoan, and fungal
chemistries including lactate dehydroge-
* Storage disease: Gaucher, Neimann-Pick,
nase (LDH) and uric acid, and serologies
and mucopolysaccharidoses
126
Chapter 13
Initial evaluation
Physical examination
Size, location, character of nodes
Presence of organomegaly
Other signs/symptoms concerning for malignancy
Evaluation for source of infection
History
Duration
Concurrent symptoms, recent skin abrasions/lesions
Animal contact
Travel
Laboratory evaluation
CBC, differential, peripheral blood smear assessment
ESR or CRP
LDH, uric acid for suspected malignancy
CMV, EBV, and other titers as indicated (HIV, B. henselae, toxoplasmosis, etc.)
Place PPD or send quantiferon gold
Consider CXR for large, unexplained nodes
Cytopenias
Hilar adenopathy
Suspicious cells on smear
Consider Hodgkin lymphoma, tuberculosis
Lymph node excisional biopsy
Imaging with CT chest/abdomen/pelvis
Uncertain diagnosis
Nodes in suspicious locations
Bone marrow aspirate
(axillary, supraclavicular, epitrochlear) or
Duration 6 weeks or longer or
Infection confirmed or likely
Growing in size or
Node 2.5 cm or
Node < 2.5 cm
Empiric antibiotic therapy
Hard, nonmobile or
No infectious signs/symptoms
Consider other titers (STI,
non-tuberculous mycobacterium,
histoplasmosis, etc.) as necessary
No diagnosis
Excisional node biopsy
No resolution with antibiotics, within 2-6 weeks
Figure 13.1 Evaluation of the child with adenopathy. (Abbreviations: CBC, complete blood count;
ESR, erythrocyte sedimentation rate; CRP, C-reactive protein; LDH, lactate dehydrogenase; CMV,
cytomegalovirus; EBV, Epstein-Barr virus; CXR, chest X-ray; CT, computed tomography; STI, sexually
transmitted infection).
Malignancy: leukemias as well as Hodgkin
bleeding or bruising (malignancy), travel
and non-Hodgkin lymphoma
to endemic areas (malaria), trauma (splenic
hematoma), and family history (hemoglo-
A detailed history should be obtained to
binopathies, thalassemia, and hereditary
gain clues as to the etiology of the enlarged
spherocytosis with prior splenectomy or
spleen. The patient should be questioned
cholescystectomy). The physical examina-
regarding current or recent infectious symp-
tion should include a measurement of the
toms, fevers or rigors
(e.g., in subacute
spleen size (centimeters below the midcostal
bacterial endocarditis, infectious mononu-
margin), consistency, and presence of ten-
cleosis, and malaria), jaundice (with hemo-
derness (which suggests rapid increase in
lytic anemia or liver disease), abnormal
size) in addition to the presence of
Evaluation of the Child with a Suspected Malignancy
127
adenopathy or hepatomegaly. The vitals
* Connective tissue disease: ESR, comple-
should be reviewed for evidence of fever
ment (C3, C4, CH50), antinuclear antibody,
or hypotension and the skin assessed for
and rheumatoid factor
cutaneous bleeding. Other considerations
* Infiltrative diseases: bone marrow aspirate
include evaluation for stigmata of specific
and biopsy
(looking for blasts, storage
disease states including jaundice, cardiac
cells) and enzyme study for Gaucher
murmurs, arthritis, as well as specific find-
(glucocerebrosidase)
ings of endocarditis including Roth spots
* Lymph node biopsy: can be done if per-
(retinal hemorrhages), Janeway lesions
formed with coexistent lymphadenopathy
(nontender hemorrhagic lesions on the
* Imaging: volumetric and heterogeneity
palms/soles), and Osler nodes
(tender
assessment with ultrasound, CT, or MRI;
microemboli on the fingers and toes).
liver-spleen scan with99mTc-sulfur colloid
The laboratory and imaging assessment
for functional analysis; can also send pit
is determined based on suspicion of the
count for pitted red blood cells.
underlying etiology of splenomegaly.
Consideration should also be given to a
Bone pain is unusual in children and ado-
secondary effect of hypersplenism in which
lescents except when associated with trauma.
the child may have cytopenias due to a large
Growing pains are a common complaint and
spleen. Persistent splenomegaly should be
consist of bilateral
(80% of cases) lower
fully investigated as follows:
extremity pain occurring in the afternoon,
* CBC: evaluate red cell indices, reticulo-
evening, or night, affect children between 3
cyte count, platelet count, white blood
and 14 years of age, occur most commonly on
cell count, and review peripheral blood
a weekly or monthly basis and much less
smear (all to help rule out hematologic
frequently daily, and are relieved with mas-
disorders such as hemolytic anemia;
sage in the majority of cases. Children pre-
membrane disorders with spherocytes or
senting with this complaint have an excellent
elliptocytes; increased red cell indices and
prognosis. Pain is otherwise usually due to
anemia in thalassemia; atypical lympho-
bone, bone marrow, or nerve infiltration.
cytes as in EBV; leukemic blasts; and
Back pain in young people is pathologic and
cytopenias secondary to leukemia or
may be due to a tumor in the spinal cord or
hypersplenism)
one causing external compression such as
* Infection: blood culture, viral studies,
neuroblastoma, rhabdomyosarcoma, or a
PPD (or quantiferon gold), thick and thin
leukemic chloroma. This finding should
smear, and serologies to rule out EBV,
prompt an MRI of the spine, as these con-
CMV, HIV, histoplasmosis, tuberculosis,
ditions typically result in no abnormalities on
and malaria
plain film.
* Hemolytic evaluation: CBC, reticulocyte
Patients with primary bone tumors
count, direct antiglobulin test (Coombs),
often present with localized pain, fre-
haptoglobin, serum bilirubin, LDH, red cell
quently in association with a growing mass.
enzyme assays (G6PD, pyruvate kinase defi-
Pain may be attributed to recent mild
ciency), osmotic fragility testing or ektacy-
trauma or growing pains. In some cases,
tometry, and urinalysis
patients sustain a pathological fracture after
* Liver disease: complete metabolic panel,
seemingly mild trauma due to infiltration
coagulation studies, hepatitis panel, a-1
of the periosteum and weakening of the
antitrypsin, ceruloplasmin
(Wilson dis-
bone. Bone pain is typically a presenting
ease), and 24-hour urine copper
symptom in patients with osteogenic
128
Chapter 13
sarcoma and Ewing sarcoma. Langerhans
for a marrow infiltrative process, primarily
cell histiocytosis very often involves bone
leukemia. Isolated anemia, leukopenia, or
and may present with localized bone pain
thrombocytopenia can occur in leukemia
anywhere in the body, often with overlying
but much more commonly the patient will
soft tissue swelling.
present with bi- or pancytopenia. Pancyto-
Diffuse or multifocal bone pain is a
penia may also indicate the lack of blood cell
common presenting symptom in acute
production as in aplastic anemia, or is
leukemia. It is reported in more than
caused by the proliferation of malignant
25% of patients diagnosed with acute lym-
cells in the marrow with resultant crowding.
phoblastic leukemia (ALL), but is seen less
The anemia is frequently characterized as
commonly in acute myelogenous leukemia
one of chronic diseases (i.e., normochromic
(AML). It is due to marrow crowding with
and low reticulocyte count). Leukocyte
leukemic cells. Patients will often com-
counts are variable at presentation in acute
plain of back or leg pain that is persistent
leukemia and may be normal, decreased, or
and increasing in intensity and very young
elevated. The cell differential, however, is
children may become irritable and refuse
likely to show neutropenia and the periph-
to walk or participate in normal activities.
eral blood smear will likely, though not
Musculoskeletal pain in children is often
always, demonstrate blasts or immature
diagnosed as arthritis or bone or joint
cells. Unless metastasis to the marrow has
infection. Pain may be asymmetric and
occurred, leukopenia and thrombocytope-
often children present with a limp with
nia are rarely associated with extramedul-
pain seemingly disproportionate to the
lary malignancies (see Table 13.3).
findings on examination. In contrast to
Patients with bi- and pancytopenia
children with arthritis, children with leuke-
require bone marrow aspiration and possi-
mia will have worsened pain at night, severe
bly biopsy for suspected diagnosis of a
pain that may shift to other locations, no
marrow infiltrative process, most likely
morning stiffness or swelling, and may have
acute leukemia. The situation can be more
associated constitutional symptoms such as
complex in the case of a single depressed
weight loss and night sweats. Associated
lineage. Children with persistent or wors-
laboratory findings that may suggest malig-
ening normocytic, normochromic anemia
nancy include an elevated erythrocyte sed-
without manifestations of hemolysis, clini-
imentation rate, elevated serum LDH, ane-
cal suspicion of transient erythroblastopenia
mia, thrombocytopenia, neutropenia, and/
of childhood, or renal disease with low
or leukopenia. Any of these findings should
erythropoietin production should have
prompt an examination of the bone
diagnostic bone marrow studies performed.
marrow.
Those with isolated severe thrombocytope-
Bone pain may also be a direct result
nia suspected of having immune thrombo-
of bony metastatic disease or marrow
cytopenic purpura (ITP) with no clinical
infiltration secondary to other tumors
signs or symptoms of malignancy should be
including neuroblastoma, rhabdomyosar-
followed closely and, if with the develop-
coma, Ewing sarcoma, and non-Hodgkin
ment of additional cytopenias or the need
lymphoma. Localized bone pain warrants
to start corticosteroids as part of their treat-
radiographic evaluation
(two-view plain
ment for ITP, should also have a bone
radiographs) for the assessment of a lesion
marrow examination. The necessity of bone
or leukemic changes.
marrow examination prior to the initiation
Cytopenias and/or abnormalities on
of steroids for the treatment of ITP is
the peripheral blood smear are suspicious
controversial.
Evaluation of the Child with a Suspected Malignancy
129
Table 13.3 Evaluation of the child with suspected leukemia.
History and physical examination
Assess life-threatening conditions including severe anemia, thrombocytopenia, DIC, infection,
compression of vital organs, hyperleukocytosis, and metabolic derangements
Laboratory studies
CBC with manual differential, reticulocyte count, examination of peripheral blood smear
Metabolic panel with electrolytes, BUN, creatinine, uric acid, LDH, AST, ALT, alkaline phos-
phatase, total bilirubin, magnesium, calcium, and phosphorus
Serologies: varicella, CMV, HSV, hepatitis A, B, and C (obtain prior to transfusion if possible)
Coagulation studies (PT, PTT, fibrinogen, FDP, or D-dimers) in suspected AML (especially APL)
Type and screen for red cell transfusion, if necessary
If febrile or ill-appearing: blood and urine cultures
Radiographic studies
Chest radiograph (assess for mediastinal mass)
Plain bone films of sites of bone pain (assess for pathological fractures)
Diagnostic studies
Bone marrow aspiration
Specimens for morphology, immunophenotyping, and karyotype
Extra “pulls” as per protocol for biological studies (Children’s Oncology Group or local
institutional studies)
For “dry” tap, obtain bone marrow biopsy for diagnostic studies
Lumbar puncture (platelet count
50-100
109/L per institutional protocol)
Cytology, chamber count (WBC, RBC, protein, and glucose), and CSF culture if patient is febrile
Initial procedure should be done by an experienced clinician, after careful evaluation for elevated
ICP
Abbreviations: DIC, disseminated intravascular coagulation; CBC, complete blood count; BUN,
blood urea nitrogen; LDH, lactate dehydrogenase; PT, prothrombin time; PTT, partial throm-
boplastin time; AML, acute myelogenous leukemia; APL, acute promyelocytic leukemia (AML
M3); WBC, white blood cell; RBC, red blood cell; CSF, cerebrospinal fluid; ICP, intracranial
pressure; FDP, fibrin degradation products; CMV, cytomegalovirus; HSV, herpes simpex virus.
Bone marrow examination with aspi-
leukemia. Manifestations typically involve
rate and/or biopsy is indicated in the fol-
mucocutaneous tissues with clinical signs
lowing situations:
including petechiae, purpura, epistaxis, and
* Significant depression of one or more
menorrhagia. Patients with acute promye-
peripheral blood lineages (white cells, red
locytic leukemia (APL; AML M3) are at risk
cells, and platelets) without an obvious
for severe bleeding at presentation due to
explanation
underlying coagulation abnormalities and
* Presence of circulating blasts on the
disseminated intravascular coagulation
peripheral blood smear
(DIC), whereas Wilms tumor patients may
* Any cytopenias associated with unex-
have acquired von Willebrand disease at
plained lymphadenopathy, splenomegaly,
presentation increasing bleeding risk.
hepatomegaly, anterior mediastinal mass,
Patients with extensive marrow involvement
or bone pain.
such as advanced stage neuroblastoma may
also present with significant purpura.
Bleeding as a presenting sign in cancer is
Mediastinal masses may lead to com-
usually related to severe thrombocytopenia
pression of respiratory, vascular, or other
and occurs commonly in children with acute
structures and can range from an
130
Chapter 13
asymptomatic incidental finding to signifi-
stool, intussusception, abdominal aorta, a
cant compromise and an emergent situa-
distended bladder, hydronephrotic kidneys,
tion. Most mediastinal masses in children
and pregnancy.
are malignant. Imaging with chest radio-
The age of the child can provide a clue to
graphy and chest CT yields information
diagnosis. In the newborn, an abdominal
with respect to location in the mediastinum
mass is most likely to be a congenital abnor-
and potential for compromise.
mality of renal origin. The most common
Anterior mediastinal masses are more
malignant tumors in young children are
typically seen in older children and adoles-
neuroblastoma and Wilms tumor. Children
cents and frequently are associated with lym-
with Wilms tumor most often present well
phomas, T-cell leukemia, thymic tumors,
appearing and the mass is an incidental
thyroid tumors, and some benign tumors
finding by a family member or during a
(teratomas, lipomas, and angiomas). Malig-
well-child check. Unlike Wilms tumor, neu-
nant tumors, especially lymphomas and
roblastoma will often present with evidence
leukemia, may have rapid growth rates and
of spread and systemic symptoms including
quickly lead to compromise with presenta-
weight loss, fever, and bone pain. In older
tion of superior vena cava syndrome, airway
patients, the mass may be related to leuke-
obstruction, dysphagia, and symptoms of
mia or lymphoma with enlargement of the
increased intracranial pressure due to
spleen and liver. The most common lym-
decreased cerebral venous return. The mass
phoma in children is Burkitt lymphoma
may cause a pericardial effusion or directly
that may present as a rapidly enlarging
obstruct cardiac outflow leading to cardiac
abdominal mass leading to pain and
compromise. See Chapter 14 for assessment
obstructive symptoms (gastrointestinal and
and management.
urinary tracts) in association with metabolic
Middle mediastinal masses are also
derangements from tumor lysis. Burkitt and
more likely malignant. Infections should
other lymphomas, as well as primary gas-
be in the differential diagnosis and one
trointestinal tumors, may also occur in the
should consider tuberculosis or histoplas-
ileocecal area and serve as a lead point for
mosis in addition to pericardial cysts, bron-
intussusception.
chogenic cysts, esophageal lesions, or direct
The history is important to determine if
extension of an abdominal mass. Malignant
thesymptomsarerelatedtothemass.Acareful
tumors common in this location include
genitourinary history should be obtained to
Hodgkin lymphoma and nodal masses of
determine if the mass may be of renal origin.
neuroblastoma, rhabdomyosarcoma, and
Historical points may provide suspicion of
germ cell tumors.
catecholamine production such as flushing,
Posterior mediastinal masses are gen-
palpitations, diarrhea, and sweating
(very
erally neurogenic in origin and include
rare). Constitutional symptoms such as fail-
benign and malignant tumors. These
ure to thrive, fever, night sweats, and sudden
include ganglioneuroma, neurofibroma,
weight loss should lead one to suspect a
and neuroblastoma. Most of these lesions
disseminated process such as neuroblastoma
are asymptomatic, but may present with
in young children or lymphoma in older
symptoms of spinal cord compression such
children and adolescents.
as pain or focal neurological signs.
A meticulous and careful physical exam-
A palpable abdominal mass is one of
ination of the child should be done by first
the most common presenting findings of a
attempting to have the child relax. When
malignant solid tumor in children. Non-
examining the abdomen, keep in mind the
malignant etiologies include impacted
normal structures that may be present such
Evaluation of the Child with a Suspected Malignancy
131
as the liver or spleen edges, kidneys, aorta,
and genitourinary abnormalities have been
sigmoid colon, stool, or spine. A rectal exam-
reported in association with Wilms tumor.
ination and pelvic/vaginal examination may
Subcutaneous nodules
(typically bluish),
be indicated but should be performed only
periorbital ecchymoses, opsoclonus-myoc-
by an experienced practitioner and after
lonus, and presence of organomegaly are
obtaining laboratory studies
(should not
seen with neuroblastoma. Signs of preco-
be neutropenic for a rectal examination).
cious puberty may be seen with tumors
Care should be taken to palpate the mass
involving the liver, adrenal glands, or
gently and limit the number of examiners.
gonads (i.e., germ cell tumors). The neu-
Imaging will also help determine size and
rological examination may show evidence
location. A careful general physical exami-
of a Horner’s syndrome with apical tumors
nation is vital as many tumors may have
(often neuroblastoma) or spine compres-
associated signs and symptoms or underly-
sion with large abdominal masses. See
ing syndromes. Aniridia, hemihypertrophy,
Table 13.4 for the workup of a child with
Table 13.4 Evaluation of the child with an abdominal mass.
History and physical examination
Radiological studies
First steps
Flat plate and upright views of the abdomen
Abdominal ultrasound
Further assessment as needed
Abdominal/pelvis CT or MRI
Chest CT if with abdominal primary to assess extent of local disease and presence of metastatic
disease
Bone scan in suspected neuroblastoma, rhabdomyosarcoma, clear cell or rhabdoid tumor of the
kidney
MRI and plain radiography of the spine in tumors with neurologic impairment or other
radiographic suggestion of spinal invasion
MIBG scan for neuroblastoma
Laboratory studies
First steps
CBC with differential, reticulocyte count (bleeding into mass may cause iron deficiency anemia;
tumor may also involve the bone marrow and cause cytopenias), and review of peripheral
smear
Electrolytes, calcium, phosphorus, uric acid, LDH, BUN, creatinine, and liver transaminases
Urinalysis
As indicated by history/examination/imaging:
Urine for tumor markers: catecholamines (VMA and HVA)
Serum markers: neuron-specific enolase, a-fetoprotein, b-HCG, ESR, copper, and ferritin
Bone marrow aspirate/biopsy; if neuroblastoma, lymphoma, or rhabdomyosarcoma suspected
or confirmed
Abbreviations: CT, computed tomography; MRI, magnetic resonance imaging; MIBG, metaio-
dobenzylguanidine; CBC, complete blood count; LDH, lactate dehydrogenase; BUN, blood urea
nitrogen; VMA, vanillylmandelic acid; HVA, homovanillic acid; HCG, human chorionic
gonadotrophin; ESR, erythrocyte sedimentation rate.
132
Chapter 13
an abdominal mass. A surgical consultation
Suggested Reading
should be obtained and the decision made
whether to obtain a biopsy or perform a
Neville KA, Steuber CP. Clinical assessment of the
resection of the mass. Surgical staging is done
child with suspected cancer. In Basow DS (ed),
UpToDate. Waltham, MA, 2011.
by assessing tumor margins, nodal involve-
Wilne S, Koller K, Collier J, et al. The diagnosis of
ment, presence of locally invasive or distant
brain tumours in children: a guideline to assist
disease, and for possible tumor spillage (see
healthcare professionals in the assessment of
Chapters 18 and 19).
children who may have a brain tumour. Arch
Dis Child 95:534-539, 2010.
Oncologic
14
Emergencies
The prognosis for children diagnosed with
Valsalva maneuver. The physical examina-
cancer has steadily improved over the past
tion often reveals a plethoric, edematous
50 years, and it is now estimated that more
face and neck, jugular venous distension,
than 80% of children newly diagnosed with
papilledema, and pulsus paradoxus. Blood
cancer will ultimately be cured of their
pressure changes, pallor, and even cardiac
disease. Given this positive outlook, it
arrest can result from postural changes.
becomes even more important that life-
Superior mediastinal syndrome is the com-
threatening complications arising either as
bination of SVCS and tracheal compression
a result of the patient’s cancer diagnosis or
that leads to symptoms of cough, dyspnea,
the treatment being provided be promptly
air hunger, and wheezing. Examination
recognized and appropriately treated.
often reveals decreased breath sounds,
Oncologic emergencies can be catego-
wheezing, stridor, or cyanosis. Affected chil-
rized based on their pathogenesis, including
dren are often incredibly anxious as well.
emergencies caused by space-occupying
Malignant tumors are the most common
lesions, those caused by abnormalities of
primary cause of SVCS. The most common
blood and blood vessels, and metabolic
cause is non-Hodgkin lymphoma (NHL;
emergencies.
usually lymphoblastic lymphoma or diffuse
large B-cell lymphoma). Other malignant
causes include Hodgkin lymphoma, T-cell
Emergencies caused by
acute lymphoblastic leukemia
(T-ALL),
space-occupying lesions
malignant teratoma, thymoma, neuroblas-
toma, rhabdomyosarcoma, or Ewing sar-
Superior vena cava syndrome and
coma. A secondary cause can be thrombosis
superior mediastinal syndrome
of the major vessels caused by the presence
Superior vena cava syndrome (SVCS) refers
of a central venous catheter. Nonmalignant
to the signs and symptoms resulting from
causes include mediastinal granulomas
the compression or obstruction of the SVC
(histoplasmosis), aortic aneurysms, vascular
caused by an anterior mediastinal mass.
thrombosis complicating cardiovascular
These include orthopnea, headache, facial
surgery for congenital heart disease, shunting
swelling, dizziness or fainting, sudden pal-
for hydrocephalus, catheterizations, and
lor, and exacerbation of symptoms with the
infections (e.g., tuberculosis and syphilis).
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
134
Chapter 14
The SVC is a thin-walled vessel with low
General anesthesia may cause cardio-
intraluminal pressure, prone to thrombosis,
vascular and/or respiratory compromise
and surrounded by thymus and nodes that
by increasing abdominal tone and decreas-
drain the right side and lower left side of
ing respiratory muscle tone and lung vol-
the chest. Part of the SVC is also in the
ume. Sedatives can also decrease venous
pericardial reflection. Lymph nodes, thy-
return and should be used cautiously.
mus, and the pericardium may become
5. Bone marrow aspiration under local anes-
enlarged from infection or tumor involve-
thesia. If the bone marrow is involved, this
ment and compress the SVC. Adjacent cor-
test will allow the diagnosis to be made
onary and collateral vessels can become clot-
without a biopsy of the mass itself.
ted. Compression, clotting, and edema lead
6. Examination of pleural fluid/pericardial
to diminished air flow and blood flow. Addi-
fluid/ascitic fluid. In addition to relieving
tionally, the trachea and right mainstem
distress, removal of fluid and subsequent
bronchus are less rigid in children as com-
cytological examination may allow a defin-
pared to adults.
itive diagnosis to be made, especially in the
case of NHL.
Evaluation
7. Echocardiography. This is necessary to
1. History and physical examination. The
look for pericardial effusion and cardiac tam-
history is nonspecific, with a typically short
ponade and to evaluate myocardial function.
period of progressively worsening symptoms.
8. Pulmonary function tests. The patient’s
Patients with ALL may have a short history of
peak expiratory flow rate and the shape of
fever, bone pain, bruising, petechiae, or other
the flow volume loop can reliably predict
signs of marrow dysfunction. Presenting
the patient’s ability to tolerate various diag-
symptoms and signs include cough and dys-
nostic and therapeutic procedures.
pnea (commonest), closely followed by dys-
9. Measurement of b-HCG (b-subunit of
phagia, orthopnea, hoarseness, chest pain,
human chorionic gonadotrophin) and AFP
facial edema, wheezing, pleural effusion,
(a-fetoprotein). Elevations in these markers
pericardial effusion, features of carbon diox-
are diagnostic of a malignant germ cell
ide retention like anxiety, confusion, leth-
tumor, but they are generally not available
argy, headache, distorted vision, and syn-
urgently.
cope, and occasionally other symptoms like
conjunctival suffusion and cyanosis.
Management
2. Chest radiography including a lateral
1. Children should be under close observa-
view. This usually shows mediastinal wid-
tion in the intensive care unit with elevation
ening, tracheal deviation or compression,
of the head, continuous cardiovascular and
and possibly pleural effusions.
respiratory monitoring, and pulse oximetry.
3. CT scan of the chest. This helps delineate
2. Children with impending or actual air-
distortion of normal anatomy, identify tra-
way obstruction should receive emergent
cheal compression, and allow assessment of
radiation therapy, given in 1 to 2 Gy frac-
mediastinal mass width. This may be done
tions for 1 to 4 days. A small area of the
in the prone position if the patient is unable
tumor should be shielded to prevent radi-
to lie supine.
ation-induced changes if a biopsy still needs
4. Tissue diagnosis with least invasive proce-
to be performed to establish the diagnosis.
dures, with no sedation, and local anesthesia
The surgeon and radiation oncologist
only. A biopsy is essential for diagnosis,
should communicate to ensure that the
but must be able to be performed safely.
most easily accessible area is shielded.
Oncologic Emergencies
135
Tracheal swelling and further airway com-
compression, more commonly from an intra-
promise can occur as a result of the
medullary location.
radiation.
Evaluation
3. Empiric therapy for suspected malig-
1. Localized back pain or radicular pain
nancy may need to be initiated due to the
extending down the leg occurs in up to
life-threatening situation. Steroids, cyclo-
80% of children with cord compression.
phosphamide, vincristine, or anthracyclines
The duration is variable but generally short;
have been given in this situation to children
one series reported a range of 5 days to
with suspected leukemia or lymphoma.
4 weeks. Characteristically, straight leg rais-
Prednisone is commonly used as most of
ing, neck flexion, or the Valsalva maneuver
these children are ultimately diagnosed with
aggravates the pain. Pain is almost always
lymphoma or leukemia, especially if other
the first presenting symptom, with weak-
clues to the diagnosis are apparent (e.g.,
ness and bowel or bladder dysfunction
organomegaly, generalized adenopathy,
occurring later. Sensory loss may occur but
evidence of tumor lysis syndrome [TLS],
is often difficult to identify, especially in
elevated WBC
[white blood cell] counts).
young children. Any objective neurological
Intravenous methylprednisolone is started
deficit requires further evaluation.
at a dose of
50 mg/m2/day divided BD.
2. Plain radiographs are generally not help-
Hydration should be given and the child
ful in identifying abnormalities in children,
monitored for tumor lysis syndrome that
as cord compression typically occurs via
may be exacerbated by the initiation of
extension of tumor through the interverte-
therapy and is discussed later.
bral foramina without bony erosion or
injury. MRI is the best study for demon-
strating intraspinous extension, but is not
Spinal cord compression
always immediate available. While CT is less
Acute spinal cord compression occurs in
sensitive in identifying extension of disease
about 5% of children with cancer. Prolonged
into the spinal canal, it will effectively iden-
cord compression leads to irreversible neu-
tify paraspinous disease.
rological injury with paralysis, sensory loss,
3. Examination of the cerebrospinal fluid
and loss of bowel and bladder control. Once a
(CSF) is rarely helpful, and patients may
neurological deficit occurs, it often progresses
experience rapid neurological deterioration
to paraplegia within days or even hours. The
following a lumbar puncture (spinal coning).
most frequent cause of cord compression is
external compression caused by extension of a
Management
paravertebral tumor through an interverte-
1. Patients with rapidly progressing spinal
bral foramen into the epidural space. The
cord dysfunction require immediate inter-
tumor compresses the vertebral venous
vention. Dexamethasone 1 mg/kg should be
plexus leading to cord edema, venous hem-
given IV even before imaging is performed,
orrhage, myelopathy, and ischemia. The most
and an MRI should be obtained immedi-
common tumors leading to spinal cord com-
ately. Patients with suspicious findings who
pression include neuroblastoma, Ewing
are stable should be started on a lower dose
sarcoma, NHL, and Hodgkin lymphoma.
of oral dexamethasone (0.25 to 0.5 mg/kg
Intraspinous chloromas in patients with acute
every
6 hours) with imaging performed
myelogenous leukemia (AML) are a less com-
within 24 hours.
mon cause. Spinal cord astrocytomas and
2. If imaging reveals a tumor with spinal
ependymomas can also cause cord
cord compression, this must be promptly
136
Chapter 14
relieved. Surgical decompression with a
at first, and then becomes more frequent
laminectomy quickly relieves pressure and
and severe. It characteristically occurs in
allows tumor to be removed for diagnosis. It
the morning, and is often but not always
should be performed if the diagnosis is
accompanied by vomiting without diar-
unknown. If the diagnosis is known and
rhea. Other signs and symptoms include
the tumor is radiosensitive, emergent radi-
diplopia, ataxia, hemiparesis, speech dis-
ation therapy should be promptly initiated.
turbance, neck stiffness, dizziness, lethargy,
Chemotherapy is an alternative for chemo-
and coma.
therapy-responsive tumors such as neuro-
Some tumors may also cause focal neu-
blastoma, Ewing sarcoma, Hodgkin lym-
rological changes. Cerebellar astrocytomas
phoma, and NHL; the onset of action in
may lead to ipsilateral weakness, hypotonia,
these tumors is similar to radiation therapy.
and ataxia. Herniation of a cerebellar tonsil
often causes head tilt and neck stiffness.
The prognosis for recovery of function is
Tumors near the third ventricle (cranio-
directly related to the degree of disability at
pharyngiomas, germ cell tumors, optic gli-
diagnosis. This is associated with the dura-
omas, and hypothalamic and pituitary
tion of symptoms and the length of time
tumors) may cause visual loss, increased
required to make the diagnosis. Patients
intracranial pressure, and hydrocephalus.
who are ambulatory at diagnosis typically
A pineal tumor may lead to Parinaud’s
remain ambulatory; approximately 50% of
syndrome
(impairment of upward gaze,
children who are nonambulatory at diagno-
convergence nystagmus, and alterations in
sis regain the ability to ambulate.
pupillary response).
Increased intracranial pressure
Evaluation
and brain herniation
1. Assessment of vital signs is critical.
Brain tumors are the most common solid
Increased ICP will often result in Cushing’s
tumor in childhood, and the majority of
triad: bradycardia, hypertension, and apnea.
patients present with signs and symptoms
2. A careful physical examination looking
of increased intracranial pressure (ICP). For-
for signs of increased ICP or impending
tunately, few of these patients progress to
herniation must be done quickly. In addition
actual brain herniation. Most pediatric brain
to the findings mentioned above, impending
tumors are infratentorial and cause increased
brain herniation may cause changes in respi-
ICP by obstructing the third or fourth ven-
ratory pattern, pupil size and reactivity,
tricle with resultant obstructive hydroceph-
extraocular movements, spontaneous motor
alus. They can also cause increased ICP sim-
function, and responsiveness to verbal and
ply by mass effect. This most commonly
physical stimuli.
occurs with astrocytomas, as well as with
primitive neuroectodermal tumors includ-
Management
ing medulloblastoma.
If increased ICP or impending brain herni-
The presentation can vary significantly
ation is suspected, appropriate management
depending on patient age. Common symp-
must be initiated prior to more definitive
toms in infants include vomiting, lethargy,
testing to hopefully avoid death or perma-
personality changes, loss of developmental
nent neurological injury. The following
milestones, and increased head circumfer-
steps should be initiated:
ence. Older children commonly complain
1. Fluid intake should be limited to no
of headache. This is frequently intermittent
more than 75% maintenance.
Oncologic Emergencies
137
2. A loading dose of dexamethasone 0.5 to
urine output and azotemia), or dissemi-
1 mg/kg should be given IV followed by 0.25
nated intravascular coagulation.
to 0.5 mg/kg every 6 hours.
2. A biopsy and staging evaluation including
3. An emergent CT scan of the head should
CT scan of the chest, abdomen, and pelvis,
be performed. A noncontrast scan will effec-
bone scan, bilateral bone marrow aspiration
tively identify bleeding, most tumors, cere-
and biopsy, and other imaging such as MRI
bral edema, and hydrocephalus.
of the brain and spine should be performed
4. If the diagnosis of increased ICP is con-
quickly if the patient is able to tolerate these
firmed, the patient should be admitted to
procedures. This will allow prompt initiation
the ICU. Neurosurgical consultation should
of appropriate therapy.
be immediately obtained if a tumor is iden-
tified or the patient has hydrocephalus.
Treatment
5. Acetazolamide 5 mg/kg/dose every 6 hours
1. If the patient is stable and does not exhibit
can be given to decrease CSF production.
any of the findings above, careful observation
6. Intubation and hyperventilation to
and supportive care should be provided.
decrease the pCO2 to 20 to 25 mmHg will
2. If the patient is unstable, treatment with
decrease cerebral perfusion. Care must be
chemotherapy or radiation therapy should
taken not to cause cerebral ischemia.
be considered. Cyclophosphamide 5 mg/kg/
7. Surgical options
(external ventricular
day for 5 days has been shown to be effec-
drainage, third ventriculostomy) should
tive, as has radiation therapy at a dose of
be considered.
150 cGy/day
3 days.
8. Prophylaxis with antiseizure medication
should also be considered.
9. Intracranial pressure monitoring may be
Emergencies caused by
beneficial.
abnormalities of blood and
10. An MRI should be obtained as soon as
blood vessels
possible to look for or confirm the presence
of a tumor.
Hyperleukocytosis
Hyperleukocytosis is defined as a peripheral
WBC count exceeding 100
109/L. It is
Massive hepatomegaly
most commonly seen at presentation or
Massive hepatomegaly is the most emergent
relapse of AML, ALL, or chronic myeloge-
complication of stage 4S neuroblastoma in
nous leukemia (CML). Using this defini-
infants, especially those under 4 weeks of
tion, the incidence of hyperleukocytosis at
age. While 4S neuroblastoma often regresses
presentation is 9% to 13% of children with
spontaneously, when it occurs during early
ALL, 5% to 22% with AML, and almost all
infancy the resulting hepatomegaly can
children in chronic phase of CML. How-
cause death due to respiratory insufficiency
ever, clinically significant hyperleukocytosis
or hepatic failure.
occurs with WBC counts >200
109/L in
AML and >300
109/L in ALL and CML.
Evaluation
Using these criteria, the incidence is lower.
1. The patient should be examined for evi-
The most common complication of hyper-
dence of gastrointestinal dysfunction, respi-
leukocytosis in AML and CML is stroke,
ratory compromise (respiratory rate >60/
whereas in ALL it is TLS. It is more common
min or oxygen requirement), poor venous
in infantile ALL and AML, T-cell ALL, and
return (leg edema), renal dysfunction (poor
in any phase of CML.
138
Chapter 14
Hyperleukocytosis leads to occlusion of
magnesium, liver function studies, and
small veins in the brain, lung, and other
LDH at presentation, and then follow the
organs through the formation of WBC
electrolytes, BUN, creatinine, calcium,
aggregates (white thrombi). The excessive
phosphorus, and uric acid frequently to
leukocytes also cause local hypoxia by com-
assess response to therapy.
peting for available oxygen, leading to blood
vessel damage and bleeding. The degree of
Treatment
obstruction is related to the whole blood
1. Therapeutic intervention should
be
viscosity, which is related to the deform-
immediate.
ability of the cells present and the sum of the
2. If the child is being transported, these
packed erythrocyte and packed leukocyte
simple measures should be initiated prior
volumes. As myeloblasts and monoblasts
to transport. The mainstay of therapy is
are less deformable than lymphoblasts or
aggressive hydration, typically two to three
granulocytes, leukostasis is more likely to
times maintenance intravenous fluids. The
occur in AML than in ALL or CML. Other
fluids should be alkalinized to promote
factors that can increase the risk of leukos-
excretion of uric acid by converting it to a
tasis include dehydration and an elevated
more soluble urate salt. A fluid such as D5W
hemoglobin level.
1/4NS with 40 mEq/L NaHCO3 is quite suit-
Poor perfusion and anaerobic metabo-
able. Allopurinol or urate oxidase (rasburi-
lism in the microcirculation lead to lactic
case) should also be started to prevent or
acidosis. When the WBC count is >300
decrease the severity of hyperuricemia. If
109/L, local proliferation of cells occurs
urate oxidase is given, alkalinization is not
within the cerebral vasculature and brain and
required as the uric acid level falls precipi-
vessel damage occurs, leading to secondary
tously and uric acid solubility is no longer an
hemorrhage. Vessel damage can occur any-
issue. This topic is discussed more fully in the
where in the body, though the most clinically
section on TLS.
significant is in the brain and lungs.
3. If aggressive hydration does not bring the
WBC count down quickly to
100
to
Evaluation
200
109/L or the WBC count is rapidly
1. Signs and symptoms of leukemia are
rising due to rapid tumor growth, cytore-
usually present (pallor, fatigue, fever, bleed-
duction may be necessary. Two techniques
ing, bone pain, adenopathy, organomegaly,
that have been used previously to acutely
anemia, thrombocytopenia).
decrease the number of circulating WBCs
2. Possible signs and symptoms of leukos-
are leukapheresis and exchange transfu-
tasis in the lungs or brain include hypoxia,
sion. Because of the infectious disease
acidosis, dyspnea, cyanosis, blurred vision,
concerns with exchange transfusion, leuka-
papilledema, stupor, coma, or ataxia. Pria-
pheresis is used almost exclusively where it
pism may also occur in this setting.
is available and technically feasible. The
3. The laboratory assessment should
purpose of leukapheresis is to decrease
include frequent monitoring (q6 to 8 hours)
tumor bulk, decrease the risk of TLS, and
of the WBC count to follow the rate of rise
correct the anemia and hyperviscosity (goal
or the response to therapy. Metabolic stud-
of >30% leukocyte reduction with leuka-
ies also need close monitoring as the child
pheresis). If used, single volume exchange
may have evidence of tumor lysis and renal
transfusion should be performed with
dysfunction. Evaluate electrolytes, BUN,
repeat laboratory analysis to determine
creatinine, uric acid, calcium, phosphorus,
effectiveness. Further exchanges can be
Oncologic Emergencies
139
performed to reach the goal of a total WBC
2. Several authors of textbooks on pediatric
count <200
109/L.
oncology recommend that children with
4. Therapy for the underlying malignancy
profound anemia
(hemoglobin
<5 g/dL)
should begin as soon as possible and is the
that has developed over an extended period
only permanent therapy for hyperleukocy-
of time be transfused more slowly (1 vs.
tosis. In general, it is safer from a metabolic
3 mL/kg/h) to avoid causing congestive
viewpoint to initiate chemotherapy with a
heart failure. This concern is based on an
lower WBC.
extrapolation of the Starling law of cardiac
5. Emergency cranial radiation therapy has
physiology, but there are few data support-
not been demonstrated to improve outcome
ing this concern. Two published studies as
and is not recommended.
well as experience in our own institution
6. Platelet transfusions can be given safely
suggest that this practice is unnecessary,
in small volumes, as platelets have not been
especially if these children are being cared
shown to increase viscosity or worsen leu-
for in the ICU where more careful moni-
kostasis. Platelets should be given if the child
toring is being performed. This can avoid
is bleeding or the platelet count is
<20
potentially exposing patients to additional
109/L. Red cell transfusions should be
donor units unnecessarily.
given with extreme caution due to the
3. All blood products given to known or
potential to increase total blood viscosity
potential oncology patients should be
and worsen leukostasis. The only indication
irradiated to prevent transfusion-associated
is cardiovascular compromise caused by
graft-versus-host disease (TA-GVHD). They
anemia. The hemoglobin level should be
should also be leukoreduced to decrease
kept below 10 g/dL.
the risk of CMV transmission.
4. Erythropoietin is not used in children at
this time to increase the hemoglobin and
Anemia
decrease the need for transfusion due to data
Anemia is a common finding in children
from adult studies that suggest patients
presenting with malignancy. Up to 80% of
receiving erythropoietin have inferior out-
children presenting with ALL will be anemic
comes compared to those who do not.
at diagnosis. This can result from bleeding,
inflammation, or marrow infiltration by
tumor. It is rarely an emergency; children
Leukopenia
can tolerate a hemoglobin level as low as 2 to
Leukopenia is commonly found in newly
3 g/dL if it develops slowly from decreased
diagnosed oncology patients as well as during
production. Conversely, levels as low as 5 g/
therapy. This predisposes them to a variety of
dL can be life threatening if they occur as a
infections with bacteria, viruses, fungi, and
result of sudden hemorrhage.
protozoa. The evaluation and management
of this is discussed in Chapter 27.
Treatment
1. Anemia in this situation is treated with
Coagulopathy
packed red blood cell (PRBC) transfusions.
Coagulopathy in children with cancer
As a general rule, 10 mL/kg of PRBCs will
results from a variety of mechanisms. The
increase the hemoglobin level by 2 to 3 g/dL.
most common is thrombocytopenia, which
In practice, children with acute anemia due
can result from decreased platelet produc-
to blood loss may need 15 to 20 mL/kg to
tion (marrow infiltration or adverse effect
reach this level.
of therapy) or from increased consumption
140
Chapter 14
(infection/DIC, splenomegaly, and sinu-
count is less predictable than the increase
soidal obstruction syndrome). Abnormal
in hemoglobin following PRBC transfusion.
platelet function is seen in patients with
3. As with PRBCs, platelets should be
uremia. Reduced levels of coagulation fac-
irradiated and leukoreduced to decrease the
tors are seen in patients with liver disease or
risk of TA-GVHD and CMV transmission,
in DIC when coagulation factors are abnor-
respectively.
mally consumed. It is important to remem-
4. Patients with bleeding and an abnormal
ber that consumption can deplete anticoag-
PT/PTT should receive fresh frozen plasma
ulant proteins as well, potentially making
(FFP) at a dose of
10 mL/kg. This will
the patient hypercoagulable.
correct most clotting factor deficiencies
with the exception of Factor VIII and fib-
Evaluation
rinogen. Patients with a fibrinogen level
1. The CBC will reveal thrombocytopenia,
<100 mg/dL or a persistently prolonged
and the prothrombin time
(PT), partial
PTT despite FFP replacement should
thromboplastin time (PTT), fibrinogen, and
receive cryoprecipitate at a dose of 5 mL/
D-dimers will provide evidence of a clotting
kg; a typical unit of cryoprecipitate has a
disorder.
volume of 20 to 50 mL. FFP and cryopre-
2. Patients presenting with a stroke-like
cipitate do not need to be irradiated or
syndrome should have measurements of
leukoreduced as they are acellular products
protein C, protein S, and antithrombin III,
and have no risk of causing TA-GVHD or
especially if they have recently received
transmitting CMV.
asparaginase therapy.
3. A patient with an alteration in con-
sciousness or neurological abnormalities
Metabolic emergencies
associated with symptoms or signs sugges-
tive of a coagulopathy should have a non-
Tumor lysis syndrome
contrast CT scan of the brain performed
TLS is a pattern of specific metabolic abnor-
emergently looking for evidence of bleeding
malities that occurs as a result of extremely
or thrombosis.
rapid cellular turnover. It is almost always
encountered in the context of malignant
Management
tumors that either have a rapid rate of
1. The short-term therapy for thrombocyto-
apoptosis or after initiation of therapy in
penia is platelet transfusion. Patients who are
bulky tumors that are exquisitely sensitive
ill or unstable should be transfused to keep
to the therapy being provided. The rapid
their platelet count
>20
109/L. Patients
lysis of tumor cells in either situation causes
who are actively bleeding should be trans-
large amounts of potassium, phosphate,
fused if their platelet count is <50
109/L in
and nucleic acids to be released into the
case they have a component of platelet dys-
circulation, resulting in hyperuricemia,
function. Patients undergoing invasive pro-
hyperphosphatemia, and hyperkalemia.
cedures (intubation, central venous catheter
When the uric acid concentration in the
placement, or biopsy) should be transfused
renal tubule exceeds its solubility coeffi-
to a platelet count of
50 to
100
109/L
cient, urate crystals form in the tubule
depending on the procedure.
causing obstruction. Lactic acidosis, sec-
2. One pheresed unit of platelets/m2 will
ondary to poor tissue oxygenation in
typically increase the platelet count by 50 to
patients with high WBC counts, may con-
60
109/L, although the increase in platelet
tribute to uric acid deposition. In addition,
Oncologic Emergencies
141
phosphates are released when tumor cells
3. Carefully monitor vital signs, intake and
lyse. When the calcium- phosphorus prod-
output for evidence of renal insufficiency,
uct exceeds 60, precipitation occurs in the
and possible symptoms of hypocalcemia
microvasculature, particularly in an alka-
(anorexia, vomiting, cramps, spasms, tet-
line environment, further obstructing the
any, and seizures). A renal ultrasound may
renal tubules and resulting in hypocalce-
be indicated in suspected renal dysfunction.
mia. Lymphoblasts are particularly rich in
phosphate, containing up to four times the
Therapy
amount found in normal lymphocytes.
1. Aggressive hydration should be provided
Potassium is also released from tumor cells,
at a rate of two to three times maintenance.
is secondarily elevated in poor renal func-
If alkalinization of serum and urine is
tion, and can lead to fatal arrhythmia. The
desired, an appropriate starting fluid is
complete syndrome includes all these com-
D5W 1/4NSþ
40 mEq/L NaHCO3. The
ponents, with progressively worsening
bicarbonate should then be titrated to keep
renal failure. Tumor infiltration of the kid-
the urine pH between 7 and 7.5, which will
ney and dehydration can further impair
optimize the excretion of both uric acid and
renal function and accelerate the develop-
calcium phosphate.
ment of TLS.
2. An agent to prevent or reverse hyper-
In children, TLS develops most
uricemia should also be initiated immedi-
frequently in association with Burkitt
ately. Allopurinol is the most commonly
lymphoma or T-ALL. These malignancies
used agent for this purpose, and it can be
typically present with a large tumor
given at presentation and as needed during
mass, short doubling time
(38
to
116
the early phase of therapy for the preven-
hours in Burkitt lymphoma), poor urine
tion or treatment of TLS. The urine is
output, and elevated uric acid and LDH.
typically alkalinized when allopurinol is
TLS has also been reported in children
used, as it often does not cause a significant
with hepatoblastoma and advanced stage
decrease in the uric acid level until the rate
neuroblastoma.
of tumor lysis slows. It is given at a dose of
300 mg/m2/day divided every
8
hours
Evaluation
(800 mg/day maximum). Another agent,
1. Evaluate for signs and symptoms of
rasburicase (urate oxidase), is a recombi-
metabolic abnormalities. These include
nant enzyme that acts as a catalyst in the
lethargy, nausea, vomiting, hypotension,
degradation of uric acid into allantoin, a
muscle spasm, and cardiac dysrhythmia.
highly soluble product compared with uric
2. Perform a metabolic panel in all chil-
acid. It works quickly (within 30 minutes
dren with suspected malignancy to assess
of IV infusion) and keeps the uric acid level
for TLS. Frequent reassessments should be
low for 12 to 24 hours or longer, depend-
performed early in therapy as well, par-
ing on the underlying rate of cell turnover.
ticularly in patients with high tumor
Rasburicase has been studied in children
burdens and those expected to respond
with cancer who have hyperuricemia as a
rapidly to treatment. The timing of this
result of tumor lysis and has been found to
will depend on the underlying disease,
be safe and effective. However, it is quite
evidence of preexisting renal dysfunction,
expensive compared to allopurinol. Many
timing of initiation of therapy, and the
hospital pharmacies have practice guide-
risk of TLS.
lines that specify the conditions where
142
Chapter 14
rasburicase should be prescribed to avoid
phosphate from the circulation. Persistent
overuse. It is given at a dose of 0.15 to
hyperphosphatemia may require dialysis.
0.2 mg/kg/day in 50 mL preservative-free
normal saline IV over 30 minutes. While
Hypercalcemia
the package insert states that this dose
Malignancy-associated
hypercalcemia
should be repeated daily for up to 5 days,
(MAH) is a rare complication of childhood
in practice a single dose will often keep the
cancer, occurring in 0.4% to 1.3% of newly
uric acid level low for 48 hours or more,
diagnosed cases. It is seen most frequently in
and may be sufficient to prevent TLS for
ALL and alveolar rhabdomyosarcoma, but has
the entire course. Patients treated with
also been reported in children diagnosed with
rasburicase do not require alkalinization,
rhabdoid tumor, hepatoblastoma, Hodgkin
which is an advantage when managing
lymphoma, NHL, AML, brain tumors, and
other electrolyte abnormalities. It will
neuroblastoma. The cause of MAH can be
precipitate hemolysis in patients with
increasedboneresorptionfromskeletalmetas-
G6PD deficiency and has caused methe-
tases or production of calcium-mobilizing
moglobinemia in susceptible individuals.
substances such as parathyroid hormone
Routine screening for G6PD deficiency is
(PTH)-related peptide or osteoclast-activat-
not recommended before use, but families
ing factor by the tumor. MAH in turn
should be asked about this possibility. It
adversely affects the ability of the kidney to
also has rarely caused anaphylaxis.
concentrate the urine, leading to dehydration,
3. Potassium should not be added to IV
decreased glomerular filtration rate (GFR), a
fluids until it is clear that the potassium is
further reduction in calcium excretion, and
stable and not increasing due to TLS or renal
worsening hypercalcemia.
insufficiency.
4. The patient’s intake and output must be
Evaluation
closely monitored to ensure it remains
1. Symptoms of mild hypercalcemia (12 to
balanced, taking into account insensible
15 mg/dL) include generalized weakness,
losses.
poor appetite, nausea, vomiting, constipa-
5. Careful metabolic monitoring should be
tion, abdominal or back pain, polyuria,
performed.
and drowsiness. More severe hypercalcemia
6. An ECG should be obtained if the
(>15 mg/dL) results in profound muscle
potassium is 7 mEq/L or higher. This may
weakness, severe nausea and vomiting, coma,
show widened QRS complexes and peaked
and bradydysrhythmias with broad T waves
T waves in hyperkalemia. Symptomatic
and prolonged PR interval on the ECG.
hyperkalemia requires immediate therapy
2. Patients exhibiting any of these symp-
with calcium gluconate, albuterol, and
toms should have a serum Ca2þ level
glucose and insulin, along with kayexalate
measured immediately. Measurement of
to remove excess potassium from the body.
an ionized Ca2þ should be done if there
If this is ineffective, continuous renal
is any question about the validity of the total
replacement therapy or dialysis may be
calcium measurement or to rule out
required.
primary hyperparathyroidism or pseudohy-
7. If hyperphosphatemia is present, treat-
percalcemia due to increased plasma pro-
ment is forced saline diuresis with furo-
tein binding capacity.
semide and dietary phosphate restriction.
3. Measurement of the intact parathy-
Oral phosphate binders are typically not
roid hormone
(PTH) level should be
effective, as they do nothing to remove
performed to exclude concomitant
Oncologic Emergencies
143
hyperparathyroidism. PTH levels are usu-
3. If the patient is hypophosphatemic,
ally low, normal, or suppressed in MAH.
phosphate replacement at a dose of
The serum concentration of calcitriol
10 mg/kg/dose 2 to 3 times/day may inhibit
should be measured when the hypercalce-
osteoclastic activity and promote calcium
mia is suspected to be due to Hodgkin
deposition into bone.
lymphoma or NHL, as it has been impli-
4. Bisphosphonates have become the
cated as a key mediator of hypercalcemia in
treatment of choice for adults with MAH,
almost all cases of Hodgkin lymphoma and
but these have not been widely studied in
30% to 40% of cases of NHL.
children. The most published experience is
with pamidronate at a dose of 0.5 to 2 mg/
kg given IV over 2 to 24 hours. This has
Treatment
been highly successful in correcting MAH
1. The goals of treatment are to increase
within 1 to 4 days of treatment. When
renal clearance of calcium and decrease
MAH is associated with ALL, rapid initi-
bone resorption.
ation of induction therapy has been shown
2. Aggressive hydration will help increase
to be very important in reversing the
renal excretion of calcium. Forced diuresis
hypercalcemia.
with normal saline at two to three times
maintenance and furosemide 2 to 3 mg/kg
every 2 hours was previously recommended.
Suggested Reading
Furosemide blocks calcium reabsorption by
the kidney, and can decrease the calcium
Coiffier B, Altman A, Pui CH, et al. Guidelines for
level in 24 to 48 hours. However, a recent
the management of pediatric and adult tumor
meta-analysis suggests the data to support
lysis syndrome: an evidence based review.
this approach are limited, and it can also
J Clin Oncol 26:2767-2778, 2008.
cause severe loss of sodium, potassium, and
Kelly KM, Lang B. Oncologic emergencies.
magnesium. At present, the use of furose-
Pediatr Clin N Am 44:809-830, 1997.
mide is recommended only for patients
Seth R, Bhat AS. Management of common
experiencing fluid overload following
oncologic emergencies. Indian J Pediatr 78:
709-717, 2011.
aggressive saline diuresis.
Acute Leukemias
15
Acute leukemia is the most common type
developing leukemia (e.g., Down syndrome
of malignancy in children, accounting for
[DS], Fanconi anemia, Bloom syndrome,
approximately
25% of newly diagnosed
ataxia telangiectasia). Siblings, in particular
cancers in patients less than 15 years of age.
identical twins, have an increased risk of
Approximately 2500 to 3000 new cases are
developing leukemia as children. Most cases
diagnosed each year in the United States.
of leukemia stem from somatic genetic
The peak incidence is between 2 and 5 years
alterations, as opposed to an inherited
of age. Acute lymphoblastic leukemia (ALL)
genetic predisposition. Though unclear at
accounts for
75% of cases of leukemia,
this time, inciting events in the development
followed by acute myelogenous leukemia
of leukemia in children and adolescents
(AML) in 20%, with the remainder being
involve complex relationships between host
other, rarer forms. The acute leukemias are
genetic polymorphisms, environmental
biologically classified by blast morphology,
exposures, and infections.
surface proteins, cytogenetic abnormalities,
and cytochemical staining. This informa-
tion, in addition to clinical features, is uti-
Acute lymphoblastic leukemia
lized for risk stratification into treatment
groups. Risk-based therapy optimizes cura-
The majority of ALL cases arise from B-cell
tive potential while minimizing risks and
committed progenitors. T-cell ALL repre-
side effects.
sents approximately
15% of ALL cases.
A number of hypotheses regarding the
Cytogenetic abnormalities are common and
etiology and pathogenesis of acute leukemia
provide important prognostic information.
have been described, including Knudson’s
Hyperdiploidy (an excess of chromosomes;
two-hit hypothesis that entails an initial
e.g.,
>50) is seen in approximately one-
early event (possibly prior to birth), fol-
third of children with B-precursor ALL, and
lowed by a second, environmentally-medi-
the TEL-AML translocation t(12;21)(p13;
ated
(possibly infectious) event. Ionizing
q22) is present in another 25% of cases.
radiation and exposure to benzene have
Both of these cytogenetic findings are asso-
been associated with an increased risk of
ciated with a favorable outcome, whereas
developing leukemia. Certain syndromes
hypodiploidy (
44 chromosomes), and the
and chromosomal abnormalities have also
presence of certain translocations such
been associated with a high incidence of
as the Philadelphia chromosome t(9;22)
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Acute Leukemias
145
and the mixed lineage leukemia
(MLL)
such as fatigue, irritability, and anorexia
rearrangement t(4;11) or t(1;19) have tra-
as well as clinical signs such as pallor, tachy-
ditionally been associated with poorer out-
cardia, and more rarely evidence of conges-
comes. The addition of tyrosine kinase inhi-
tive heart failure. Low-grade fever of
bitors in therapy may be significantly alter-
unknown etiology may be a presenting
ing the outcome of patients with the Phila-
manifestation due to presumed cytokine
delphia chromosome translocation. It is
release or may be related to infection
important to note that conventional cyto-
secondary to neutropenia and immunosup-
genetics may not detect these common
pression. Bleeding secondary to thrombo-
translocations seen in ALL and therefore
cytopenia is usually mild and manifests as
FISH (fluorescent in situ hybridization) is
petechiae, bruising, gingival oozing, and/or
necessary for detection.
epistaxis. Life-threatening hemorrhage may
occur, but is very rare. Bone pain (typically
Clinical presentation
long bones) is common and may result in
The clinical manifestations of leukemia are a
refusal to walk or irritability in young chil-
direct result of marrow invasion with resul-
dren. This may result from direct leukemic
tant cytopenias
(anemia, thrombocytope-
infiltration of the periosteum or expansion
nia, leukopenia, and/or neutropenia) and
of the marrow cavity by leukemic cells.
rarely extramedullary involvement
(see
Pathologic fractures may also be present
Table 15.1). Children typically present with
at diagnosis and cause significant pain (see
nonspecific symptoms related to anemia
Table 15.2). Patients with T-cell ALL present
Table 15.1 Common clinical and laboratory features of acute lymphoblastic
leukemia (ALL) at presentation.
Finding
Percentage of patients (%)
Fever
60
Pallor
40
Bleeding
50
Bone pain
25
Lymphadenopathy
50
Splenomegaly
60
Hepatosplenomegaly
70
White blood cell count (
109/L)
<10
50
10-49
30
50
20
Hemoglobin (g/dL)
<7.0
40
7.0-11.0
45
>11.0
15
Platelet count (
109/L)
<20
30
20-99
45
100
25
146
Chapter 15
Table 15.2 Radiographic changes of the
spinal cord or CNS hemorrhage (related to
bones in leukemia.
leukostasis and coagulopathy) are rare and
more likely to be associated with AML.
Osteolytic lesions involving the medullary
Leukemic involvement of the testes
cavity or cortex
occurs in 2% to 5% of boys at diagnosis
Subperiosteal new bone formation;
and presents as painless enlargement, either
pathologic fracture
Transverse metaphyseal radiolucent bands
unilaterally or bilaterally. Early testicular
Transverse metaphyseal lines of increased
involvement is also associated with T-cell
density (growth arrest lines)
disease, elevated WBC count, and lympho-
matous features.
Diagnostic evaluation
with some distinctive features. It more fre-
Children presenting with more than one
quently occurs in males, and the incidence of
cytopenia and clinical symptomatology sug-
central nervous system (CNS) leukemia at
gestive of bone marrow infiltration should
diagnosis is higher than for B-cell ALL (10%
undergo a diagnostic evaluation to investi-
to 15% vs. 2% to 5%). Patients are more
gate for the possibility of acute leukemia.
likely to present with a mediastinal mass
Laboratory and imaging studies indi-
(nearly 50%) or a white blood cell (WBC)
cated are:
count above 100
109/L (30% to 50%). The
* Complete blood count (CBC), differen-
presence of an anterior mediastinal mass
tial, review of peripheral blood smear by an
may cause airway or cardiovascular compro-
experienced individual
mise (see Chapter 14).
* Bone marrow aspirate (consider biopsy
Frequently seen laboratory abnormali-
for dry tap or inadequate specimen) for
ties include elevated liver enzymes and lac-
morphology, blast count, immunopheno-
tate dehydrogenase
(LDH). Evidence of
typing, cytogenetics, cytochemistry
tumor lysis related to a large tumor burden
* Metabolic panel, to include liver function
and rapid cell turnover may result in hyper-
studies, electrolytes, lactate dehydrogenase
uricemia, hyperkalemia, and hyperpho-
(LDH), uric acid, phosphorus, calcium,
sphatemia (with resultant hypocalcemia due
blood urea nitrogen, and creatinine
to precipitation of calcium phosphate).
* Blood culture if febrile; cultures of other
CNS involvement occurs in less than 5%
suspected sites of infection
of children at presentation, but is more
* Chest radiography to evaluate for the
common in children with T-cell disease. It
presence of a mediastinal mass
is usually detected in an asymptomatic child
* Plain films of the long bones for patients
with analysis of the cerebrospinal fluid
presenting with pain to evaluate for patho-
(CSF). Rarely, a patient may present with
logic fracture or evidence of leukemic
signs or symptoms of increased intracranial
changes
pressure (e.g., cranial nerve VI palsy with
resultant esotropia and diplopia, papille-
Typically the bone marrow is 80% to 100%
dema, visual changes, morning headache,
replaced with lymphoblasts at diagnosis. At
vomiting, lethargy, irritability, possible sei-
times, due to the packed condition of the
zures) or evidence of parenchymal involve-
marrow, it may be difficult to aspirate a
ment, hypothalamic syndrome, or diabetes
sample (dry tap) and a bone marrow biopsy
insipidus. Other rare complications such
may provide diagnostic samples. The diag-
as chloromas causing compression of the
nosis of acute leukemia requires the
Acute Leukemias
147
presence of 25% or more blasts (M3 bone
puncture is done to assess possible involve-
marrow); however, the diagnosis is sus-
ment of the CSF and to assist with
pected when the marrow contains 5% to
therapeutic decision-making and risk strat-
25% blasts (M2 bone marrow with M1
ification. Many patients require a platelet
being a normal bone marrow with <5%
transfusion prior to the initial lumbar punc-
blasts) (Table 15.3). Lymphoblasts have a
ture, which should be performed by an
typical morphology with a high nuclear to
experienced clinician to decrease the chance
cytoplasmic ratio, fine nuclear chromatin,
of a traumatic tap. There is some evidence
and the presence of nucleoli. In addition,
that a platelet count of
100
109/L is
the cells tend to have a uniform size and
desired to decrease this risk. In addition,
appearance. Immunophenotyping and
evidence exists that a traumatic tap at diag-
cytochemistry are used to differentiate ALL
nosis (with peripheral blasts) can theoreti-
and AML and identify T- and B-cell ALL.
cally seed the CSF space increasing the risk
Lymphoid malignancies should lack mye-
of later CNS relapse. Subsequent procedures
loid cell surface markers (CD13 and CD33;
may be done with platelet counts of 20 to
CD [cluster of differentiation]). B-cell ALL
50
109/L, per institutional and provider
should have B-cell surface markers such as
preference. A cell count and cytocentrifuge
CD10, CD19, kappa, and lambda, whereas
examination for cell morphology is done on
T-cell ALL should have T-cell surface mar-
the fresh CSF. The diagnosis of CNS leuke-
kers including CD2, CD5, and CD7. T-cell
mia requires the presence of 5 or more
ALL cells should also stain for terminal
WBCs/mL and identification of blasts on the
deoxynucleotidyl transferase, an immature
CSF cytocentrifuge examination. CNS leu-
lymphoid marker.
kemia is classified as follows:
After the diagnosis is confirmed, evalu-
* CNS1: no detectable blasts
ation of the CSF should be done prior to
* CNS2: <5 WBCs/mL, blasts present
therapy (this is frequently done at the time
* CNS3:
5 WBCs/mL and blasts present or
of the diagnostic bone marrow aspirate if
signs of CNS leukemia (i.e., facial nerve
the clinical suspicion is high). A lumbar
palsy, hypothalamic syndrome, or brain/eye
involvement)
Table 15.3 Differential diagnosis of acute
lymphoblastic leukemia (ALL).
A traumatic lumbar puncture is defined as
the presence of
10 red blood cells/mL.
Nonmalignant disorders
Formulas are available to assist with inter-
Aplastic anemia
pretation of CNS status in the event of
Immune thrombocytopenia
traumatic initial taps with blasts on cytospin
Infections: EBV, CMV, pertussis,
(Steinherz/Bleyer algorithm).
parapertussis
Malignant disorders (round blue cell
Other assessments prior to
tumors with marrow involvement)
Lymphoma
initiation of therapy
Neuroblastoma
An echocardiogram and electrocardiogram
Retinoblastoma
are done as baseline studies in all patients
Medulloblastoma
who will be receiving anthracyclines. These
Rhabdomyosarcoma
drugs are frequently given in induction for
high risk patients and in the delayed inten-
Abbreviations: EBV, Epstein-Barr virus;
sification phase for all patients. Acute and
CMV, cytomegalovirus.
delayed cardiotoxicity may occur, though
148
Chapter 15
late complications are more often related to
approximately two-thirds of pre-B and
high cumulative doses (i.e., >300 mg/m2)
one-third of T-cell ALL patients are classified
given at a young age (<4 years) and asso-
as SR. Better outcomes are observed in youn-
ciated with other risk factors for cardiac
ger children, B-cell phenotype, those with
disease such as mediastinal radiation, obe-
more favorable cytogenetic features, and
sity, family history of early cardiovascular
females.
disease, and abnormal lipid profiles.
Further stratification into treatment
Viral serologies for hepatitis B and C,
groups is done within the first few weeks
cytomegalovirus
(CMV), herpes simplex
of therapy based on early morphologic
virus (HSV), and varicella zoster virus (VZV)
response to therapy (as measured by day
are obtained as baseline information (see
7and/or day 14 bone marrow aspirate),
Chapter 25). This aids in determining if later
cytogenetic abnormalities identified on the
infection with one of these viral agents could
diagnostic marrow, and remission status
be related to transfusion (potentially in the
at the end of induction (morphologic and
case of hepatitis B and C), and whether
as measured by minimal residual disease
the infection is primary or reactivation (in
[MRD] detection). Other considerations
the case of CMV, HSV, and VZV). Baseline
include presence of extramedullary disease
knowledge of previous exposure may impact
such as testicular or CNS disease. Infants
later treatment choices.
have a special category as they are a partic-
ularly high risk group, especially the very
Risk group classification
young.
The concept of risk stratification allows
patients at comparatively high risk of
Treatment
relapse to be treated with more intensive
Treatment of childhood leukemia is one of
and potentially more toxic therapies,
the success stories in pediatric oncology,
whereas patients at lower risk of relapse are
though much remains to be accomplished.
given lower intensity regimens that main-
Remarkable advances have occurred by the
tain the same low risk of recurrence while
identification of more effective drug com-
minimizing exposure to unnecessary agents
binations, recognition of drug sanctuary
and decreasing toxicity. Classification into
sites and routine presymptomatic CNS
risk groups is generally based on clinically
directed therapy, intensification of therapy
apparent prognostic factors identified by
in early phases, and identification of clinical
retrospective analysis and then verified
and biologic variables predictive of outcome
by prospective studies. Many risk factors
for use in risk stratification.
have been utilized in risk classification for
Most children with cancer in the United
acute leukemia, but only age, WBC, and
States are treated at pediatric oncology cen-
immunophenotype (i.e., AML vs. ALL, T-
ters and have access to participation in clin-
vs. B-cell ALL) at diagnosis continue to be
ical trials. These studies have accelerated the
the most significant factors for initial
advances made in the diagnosis and treat-
classification.
ment of pediatric cancer and clearly are a
The commonly used National Cancer
major reason for many of the successes. The
Institute criteria classify standard risk (SR)
Children’s Oncology Group is a national
ALL as children 1 to 10 years of age with a
collaborative group of professionals involved
WBC <50
109/L and high risk (HR) ALL
in the treatment of children with cancer.
as children with age
10 years or WBC
Most pediatric cancer centers register
50
109/L. Utilizing these criteria,
patients on these trials. Families are asked
Acute Leukemias
149
to give informed consent to participate in
cytarabine
(Ara-C) and methotrexate.
these trials, which may include clinical and
Patients with HR ALL also receive daunoru-
biologic questions. For families not wishing
bicin. Consolidation for HR patients incor-
to participate, the standard of care is offered
porates cyclophosphamide, cytarabine,
(best published clinical regimen). Many
asparaginase, 6-mercaptopurine, and inten-
centers may also participate in smaller pilot
sified therapy directed toward the CNS (either
studies or have their own clinical protocols.
intrathecal methotrexate alone or with radi-
The purpose of therapy in ALL is to
ation for patients with CNS disease). SR
induce a permanent biologic and clinical
patients receive intensified CNS therapy with
remission (no evidence of disease on labo-
minimal systemic chemotherapy. The delayed
ratory and physical assessment). Overall,
intensification (DI) phase has led to signifi-
approximately 85% of children with ALL will
cantly improved outcomes as has further
be cured of their disease. Treatment regimens
intensification of the interim maintenance
are classically divided into phases of therapy:
phase (intensified vincristine and methotrex-
* Induction: refers to the first 28 to 35 days
ate compared to standard maintenance). DI
of therapy, after which the child should be in
is a concept in which reintensification of
a morphologic remission. Remission induc-
therapy following attainment of remission
tion rate in SR ALL is 98%.
can be achieved with some alteration in drugs
* Consolidation: further systemic chemo-
utilized previously in the induction and con-
therapy is given, in addition to a focus on
solidation phases. Maintenance classically
CNS prophylaxis.
consists of pulses of steroid (prednisone or
* Interim Maintenance: similar drugs as
dexamethasone) and vincristine in addition
maintenance, but in a more intensified
to oral methotrexate and 6-mercaptopurine.
manner.
Individualized drug dosing is required to
* Delayed Intensification: refers to rein-
maintain certain hematologic levels and dif-
duction and reconsolidation; may be given
ferences in metabolism and tolerability are
once or twice, depending on the protocol.
likely related to individual polymorphisms.
* Maintenance: continuation of therapy;
This aspect of management in addition to
this phase lasts 2 to 3 years (longer in males
the length of maintenance makes it a unique
due to theoretical increased risk for testic-
cancer therapy for children.
ular relapse).
CNS prophylaxis continues with peri-
* CNS directed therapy: intrathecal meth-
odic intrathecal methotrexate throughout
otrexate; cranial radiation in select groups
therapy. Children with CNS3 disease receive
(CNS3, T-cell).
augmented therapy with 1800 cGy cranial
radiation, in addition to intrathecal meth-
Open clinical trials will likely alter some
otrexate. Treatment of children with CNS2
aspect of therapy for certain risk groups
disease is controversial although recent
in an attempt to improve outcomes and
Children’s Oncology Group studies have
diminish acute and late toxicity. Some
showed no difference between CNS1 and
examples may include adding a new agent
CNS2 patients receiving equivalent therapy.
in certain phases, altering chemotherapy
This is due to the addition of medications
dosing or timing in specific phases, or pos-
with improved penetration through the
sibly asking a radiation-related question.
blood-brain barrier, including dexametha-
Conventional induction therapy includes
sone instead of prednisone and high-dose
prednisone or dexamethasone, asparaginase,
methotrexate. Without CNS prophylaxis, it
and vincristine in addition to intrathecal
is estimated that 60% to 70% of children
150
Chapter 15
would relapse in the CNS (based on histor-
on to have a long-term remission with inten-
ical data). T-cell patients are at higher risk
sified therapy. Due to the frequency of ALL as
for CNS relapse and augmented CNS-
compared to other pediatric malignancies,
directed therapy is given to all patients (even
relapsed ALL is the fourth most common
without overt CNS involvement) with pro-
oncologic diagnosis in children. Timing and
phylactic 1200 cGy cranial radiation.
site of relapse are prognostic and important
determinants for future therapy. Sites of
Complications of therapy
relapse include the bone marrow or an extra-
Because of the myelosuppressive nature of
medullary site, such as the CNS or testes, or
therapy and the underlying disease state,
some combination of sites. Early relapse
children undergoing chemotherapy and/or
(<18 months since therapy initiation) is
radiation may expect to experience compli-
associated with a particularly grim prognosis.
cations during the course of their treatment.
Concepts of therapy include systemic retreat-
Many of these complications can be antici-
ment and radiation therapy to sites of extra-
pated and prevented (see Chapter 25). Most
medullary relapse. Allogeneic hematopoietic
patients will require transfusion support with
stem cell transplantation
(HSCT) may
packed red blood cells and/or platelets (see
improve the outcome of patients with mar-
Chapter 5). Children with neutropenia are
row or combined relapse. Overall survival
instructed to take special precautions to
after relapse is also impacted by cumulative
decrease the risk of infection exposure. Any
toxicity from prior therapy and high infec-
child receiving chemotherapy who develops a
tion rates due to the increased intensity of
fever should have emergent medical atten-
therapy. In general, children with late, iso-
tion, especially during phases of anticipated
lated extramedullary relapse fare better.
neutropenia (i.e., induction, consolidation,
delayed intensification; see Chapter
27).
New agents
Certain therapies are associated with specific
New agents are currently under investigation
toxicities: anthracyclines with cardiotoxicity,
to improve outcomes for high-risk and
vincristine with peripheral neurotoxicity,
relapsed patients. Such agents include the
intrathecal methotrexate with central neuro-
tyrosine kinase inhibitors (imatinib, dasati-
toxicity, steroids (especially dexamethasone)
nib) in Philadelphia chromosome positive
with avascular necrosis of bone, and radiation
ALL and nelarabine in T-cell disease. Finding
with acute and late effects (see Chapter 30).
agents with favorable toxicity profiles for use
These anticipated side effects should also be
in highly treated relapse patients is a
discussed in detail with the patient and family
challenge. Monoclonal antibodies such as
to help with earlier recognition and support-
inotuzumab and epratuzumab (anti-CD22
ive care should be initiated as needed. Poten-
antibodies) may be part of the solution.
tial late toxicities are monitored throughout
Clofarabine, a new generation nucleoside
the course of treatment and in the off-therapy
analog that inhibits DNA synthesis, is
phase of follow-up.
currently being investigated in relapsed and
refractory ALL. Other drugs are being
Relapse
explored utilizing newly acquired knowledge
Despite the dramatic improvements in our
of novel signaling pathways such as mTOR
knowledge about the biology of ALL, prog-
inhibitors. It is anticipated that the next
nostic factors, and more tailored therapy,
generation of clinical trials will be utilizing
20% of children with ALL will relapse, with
a combination of conventional chemother-
only about one-third of those patients going
apy and immunomodulator therapy.
Acute Leukemias
151
Acute myelogenous leukemia
by 4 to 6 weeks of age. However, these
children have a 20% to 30% risk of devel-
Approximately 20% of acute childhood leu-
oping AML before 3 years of age. Children
kemia is myelogenous. Unlike ALL, AML
with transient myeloproliferative disorder
does not have a peak incidence in children
or presumed congenital AML who do not
as there are only subtle increases in inci-
display phenotypic features of DS should
dence from infancy to adolescence. On the
have a karyotype performed to look for
other hand, there appears to be a steady
possible trisomy 21 mosaicism.
increase with age in adults, in whom the
Patients with inherited bone marrow
incidence of AML is approximately four
failure syndromes such as Fanconi anemia,
times that of ALL. AML is associated with
Kostmann syndrome, Bloom syndrome,
a poorer prognosis than ALL, with long-
dyskeratosis
congenita,
Shwachman-
term survival expected to be 50% to 60%.
Diamond syndrome, congenital amegakar-
Additionally, therapy for AML, which
yocytic thrombocytopenia, and Diamond-
includes multiagent chemotherapy and
Blackfan anemia have a predisposition to
HSCT, is one of the most toxic therapies
malignancy, including AML. Other condi-
in childhood cancer. Hispanic children are
tions such as Li-Fraumeni syndrome,
noted to have a higher incidence of AML,
Noonan syndrome, acquired paroxysmal
mostly accounted for by the acute promye-
nocturnal hemoglobinuria, and myelodys-
locytic leukemia subtype (APL; M3). The
plastic syndrome confer an increased risk.
incidence of secondary AML following
Twins and siblings of AML patients have an
treatment of childhood cancer is rising, due
increased risk of AML, though this is much
in part to increased survival after primary
more significant in monozygotic twins
malignancy as well as increased use of agents
within 6 years of initial diagnosis. Exposure
that cause direct DNA damage (e.g., alka-
to ionizing radiation has also been convinc-
lytors
[cyclophosphamide, ifosfamide],
ingly implicated, especially following the
anthracyclines
[doxorubicin, daunorubi-
atomic bombs in World War II.
cin], radiation therapy), and those that
inhibit topoisomerase II (e.g., epidophyllo-
Clinical presentation
toxins [etoposide] and anthracyclines).
AML and ALL are indistinguishable at pre-
The etiology of AML is likely sporadic
sentation, with the majority of signs and
although several inherited and acquired risk
symptoms being related to infiltration of the
factors may predispose to this disease. The
marrow and other body organs. Dependent
most common inherited predisposition to
on the number of circulating myeloblasts
develop leukemia is DS. Children with DS
and total WBC, some of the presenting
have a
10- to
20-fold increased risk of
manifestations may be life threatening.
developing acute leukemia compared to
Children with extreme leukocytosis (WBC
other children. In early life, children with
> 200
109/L) may present with metabolic
DS have a particularly high risk of the
abnormalities and tumor lysis syndrome
megakaryoblastic subtype of AML (AMKL;
(see Chapter 14). Clinically, they are at risk
M7). Overall, children with DS constitute
for hypoxia in the small vessels of the brain
approximately 10% of children with AML.
and lungs as a result of increased blood
In addition, approximately 10% of neonates
viscosity related to the rheology of the mye-
with DS may develop a transient myelopro-
loblasts (large, adherent, and nondeform-
liferative disorder that mimics congenital
able cells). This may translate to sludging
AML but typically improves spontaneously
in the small vasculature of the retinal,
152
Chapter 15
pulmonary, and central nervous systems
thromboplastin from cytoplasmic granules
with resultant decreased oxygen delivery,
in the promyelocytic blasts.
hypoxia, and stroke.
Children with AML and hyperleukocyto-
Patients typically present with signs and
sis (WBC >100
109/L) are at risk for tumor
symptoms related to pancytopenia with pal-
lysis syndrome at presentation and with the
lor, fatigue, anorexia, and bleeding. Fever is
initiation of therapy. Risk factors include the
very common at presentation, often with no
presence of bulky disease with lymphadenop-
apparent infectious etiology. Bone pain is
athy and/or hepatosplenomegaly as well as a
also common, though not typically associ-
rapidly escalating WBC count suggesting a
ated with fractures. Some unique and sug-
high mitotic index (see Table 15.4).
gestive features of AML at presentation
include leukemia cutis in infants (blueberry
Diagnostic evaluation
muffin rash thought secondary to extrame-
The same studies suggested for the evalua-
dullary hematopoiesis) and chloromas
tion of ALL should be obtained in the
(extramedullary collections of myeloblasts).
evaluation of the child with suspected AML.
Chloromas may occur in up to 10% of
It is common to see severe anemia and
children with AML and present most com-
thrombocytopenia in association with an
monly in the bones, skin, soft tissues, and
elevated total WBC count. The definitive
lymph nodes, but may occur anywhere in
diagnosis is made with the evaluation of the
the body. Gingival hypertrophy is a unique
bone marrow, with at least 25% to 30% of
finding in AML secondary to chloroma of
the cellular elements identified as myelo-
the gums. A particularly rare yet important
blasts based on immunophenotyping and
presentation is spinal cord compression
cytochemistry. Morphologic review may
from a chloroma in the paraspinal/epidural
also reveal Auer rods (clumps of azurophilic
region.
granules shaped into elongated needles and
Life-threatening bleeding is another
located in the cytoplasm), classically only
complication somewhat unique to AML.
seen in some subtypes of AML (i.e., M2 and
Although the majority of patients will be
M3). A lumbar puncture should be done as
thrombocytopenic secondary to marrow
up to 5% of children will have overt CNS
infiltration with myeloblasts, patients with
involvement at presentation. A coagulation
concomitant coagulopathy due to dissemi-
evaluation should be performed in these
nated intravascular coagulation are at partic-
patients including PT, INR, PTT, fibrino-
ularly high risk, a finding seen most com-
gen, and D-dimers to evaluate for the
monly in APL (M3 AML). This association is
possibility of disseminated intravascular
thought to be secondary to the release of
coagulation (most typically seen in M3).
Table 15.4 Clinical manifestations of hyperleukocytosis in acute myelogenous leukemia (AML).
Organ at risk
Consequence
Lungs
Hypoxia, respiratory distress, pulmonary infiltrates
CNS
Alteration of mental status, stroke, intracranial bleeding
Eyes
Decreased visual acuity
Kidneys
Renal insufficiency (exacerbated by tumor lysis syndrome)
Abbreviation: CNS, central nervous system.
Acute Leukemias
153
Several morphologic classification sys-
prognosis. Finally, chromosomal aberra-
tems are available for AML, including the
tions including monosomy 7 or 7q deletion,
classic French-American-British (FAB) clas-
monosomy 5 or 5q deletion, and complex
sification of M0
to M7 and the World
abnormalities
(
3
or more unrelated
Health Organization (WHO) categorization
changes) all impart a worse prognosis.
into four major subgroups (Table 15.5).
Monosomy 5 and 7 are also highly associ-
Additionally, chromosome analyses are per-
ated with cases of secondary myeloprolifer-
formed to obtain important diagnostic and
ative disorders and AML that develop after
prognostic markers. Approximately 60% of
chemotherapy for a primary malignancy.
children with primary AML demonstrate
See Other assessments prior to initiation of
clonal abnormalities in the blast lineage
therapy in the ALL section for further
prior to therapy initiation. The t(8;21)
information.
translocation is the most common chromo-
some translocation and is associated with
Risk group classification
the M2 subgroup and a more favorable
Historically AML has been classified utiliz-
prognosis. The t(15;17) translocation is
ing the FAB morphologic classification sys-
classically seen in APL (AML M3). Inversion
tem (Table 15.5). The WHO classification
of chromosome 16 (inv(16)) is associated
system was subsequently developed to also
with acute myelomonoblastic leukemia
include cytogenetics, disease biology, and
(AML M4) and also portends a favorable
clinical history. Host factors such as age,
Table 15.5 Morphologic classification for acute myelogenous leukemia (AML).
AML Subtype
Clinical/biologic features
M0
Undifferentiated leukemia
Similar to ALL, requires CD13, CD33, CD117,
and absence of lymphoid markers
M1
Acute myeloblastic leukemia
MPO þ by special stains/flow cytometry
without differentiation
M2
Acute myeloblastic leukemia with
Auer rods, t(8;21) common, chloromas,
differentiation
good prognosis
M3
Acute promyeloblastic leukemia
Auer rods, DIC/bleeding, t(15;17), good
prognosis with ATRA therapy
M4
Acute myelomonoblastic leukemia
Mixed myeloblasts (20%) and monoblasts,
peripheral monocytosis
M5
Acute monocytic leukemia
80% marrow nonerythroid cells are
monocytic, MLL (11q23) rearrangements
in infants, CNS disease more common,
chloromas (e.g., gingival hyperplasia)
M6
Erythroleukemia
Rare in children
M7
Acute megakaryoblastic leukemia
Classically seen in Down syndrome with
GATA1 mutation, good prognosis; rare
in non-DS with t(1;22), poor prognosis
Abbreviations: ALL, acute lymphoblastic leukemia; MPO, myeloperoxidase; DIC, disseminated
intravascular coagulation; ATRA, all-trans retinoic acid; MLL, mixed lineage leukemia; CNS,
central nervous system; DS, Down syndrome.
154
Chapter 15
gender, race, and constitutional abnormal-
have shown no added benefit for a mainte-
ities all have some prognostic influence on
nance phase following intensification. Allo-
outcome. Females do slightly better than
geneic HSCT is recommended for patients
males, older children do better than infants
with high risk cytogenetics (i.e., monosomy
(though outcomes in infants are improving
7, FLT3/ITD [internal tandem duplication],
with current therapies), and Caucasians fare
monosomy 5, or 5q-) or those who have not
better than non-white patents. Children
attained a remission following two courses
with DS, particularly those under the age
of induction therapy. Patients with favor-
of 4 years at diagnosis, have an improved
able risk cytogenetics and good early
outcome with less intensive therapy (overall
responses to induction therapy receive allo-
survival 80%) but also show increased tox-
geneic HSCT only in the case of relapse and
icity with therapy. Children with an initial
if a second remission has been achieved. It
WBC < 20
109/L have an improved out-
remains unclear if HSCT has benefit for
come and those with hyperleukocytosis
those AML patients with intermediate risk
(WBC >100
109/L) have a worse out-
features (usually defined as neither high risk
come. A number of recurring cytogenetic
features nor low risk features) and decisions
abnormalities are associated with AML and
must be made on an individual basis asses-
confer prognostic significance. Overall,
sing the clinical status of the patient, avail-
poor risk features include WBC count >100
ability of an appropriate match, and family
109/L, monosomy 7, and secondary AML.
preference.
Good risk features include certain FAB sub-
The morphologic subtypes of AML with
types (M1/M2 with Auer rods, M3, M4 with
highest involvement of the CNS are FAB M4
eosinophilia), rapid response to initial ther-
and M5. Overall, the frequency of CNS
apy, and favorable chromosomal features
involvement is between 15% and 20% at
such as t(8;21), t(15;17), and inversion 16.
diagnosis. All patients receive CNS conso-
As with ALL, assessment of MRD is becom-
lidative therapy with intrathecal cytarabine
ing an important marker of outcome with
in addition to high-dose cytarabine, which
chemotherapy and transplant.
has good CSF penetration. AML patients
with myeloblasts in the CNS can usually be
Treatment
cleared with intrathecal cytarabine. Cranial
As with ALL, the goal of induction therapy is
irradiation is reserved for those children
to induce a clinical and biologic remission
with refractory CNS involvement. Children
by rapidly reducing the number of malig-
with Down syndrome rarely have CNS
nant cells. Approximately 80% of children
involvement, and due to their increased
achieve remission with induction therapy.
risk of toxicity, CNS prophylaxis has been
The most effective induction regimens use a
greatly reduced without negative conse-
combination of cytarabine and an anthra-
quence in these patients.
cycline, typically daunorubicin. Individual
APL is distinctly different from the other
trials have built upon this backbone with
AML subtypes and has a particularly good
changes in dosing and timing, alternative
prognosis due to the development of tar-
anthracyclines, or introduction of new sup-
geted therapies. Approximately
3% of
portive care strategies to better manage
patients with APL die in the early phases
potential adverse effects of therapy. Other
of therapy as a result of the unique hemor-
agents utilized in continuation therapy
rhagic complications of this disease.
include etoposide, dexamethasone, 6-thio-
Patients receive treatment with differentiat-
guanine, and asparaginase. Prior studies
ing agents such as all-trans retinoic acid
Acute Leukemias
155
(ATRA) and arsenic trioxide (ATO). The
Complications of therapy
t(15;17) translocation is present in the
Given the intensity of therapy for AML,
majority of cases and leads to the produc-
children can expect to experience a number
tion of a fusion protein involving the reti-
of complications that include cytopenias
noic acid receptor alpha and promyelocyte
requiring frequent blood product support,
gene leading to arrest of differentiation at
mucositis, liver toxicity, need for parenteral
the promyelocyte stage. ATRA induces the
hyperalimentation, and severe, prolonged
differentiation of immature promyelocytes
marrow suppression. Infection is common
into mature granulocytes, followed by res-
and should be anticipated. Once neutrope-
toration of normal hematopoiesis. ATRA
nic, these children are at particularly high
does not cross the blood-brain barrier
risk for the development of sepsis with
and is therefore ineffective in treating CNS
Gram-negative organisms in addition to
involvement with APL. A WBC
10
109/L
invasive fungal disease. Routine antifungal
is associated with a higher risk of relapse.
prophylaxis with fluconazole is recom-
Higher WBC count is also associated with
mended in addition to PCP prophylaxis.
the microgranular variant (M3v), presence
Streptococcus viridans sepsis is associated
of the FLT3/ITD mutation, and the bcr3
with the use of high-dose cytarabine and
isoform. Treatment includes induction with
resultant altered integrity of the gastroin-
ATRA and anthracycline therapy (i.e., idar-
testinal mucosa. Sepsis with this organism
ubicin or daunorubicin) followed by anthra-
can be rapidly fatal. Empiric therapy for
cycline consolidation and maintenance with
fever and neutropenia should include cov-
intermittent ATRA and low dose 6-mercap-
erage for S. viridans and be started imme-
topurine. The use of arsenic trioxide in the
diately with the first fever. Prolonged hos-
initial treatment of patients with APL
pitalization is expected during the course of
remains controversial and is currently under
treatment for AML and these patients
investigation. Similarly, the length and use
should be in protective environments.
of a consolidation phase as well as the need
Coagulopathy at diagnosis is not uncom-
for intrathecal chemotherapy are yet to be
mon, especially in the child with APL. Coag-
answered questions. Patients can expect an
ulation studies should be done routinely. In
event-free overall survival of 70% to 80%.
general, platelet counts should be maintained
Toxicities with ATRA may be significant;
above 10
109/L (>50
109/L for the APL
specifically, ATRA may lead to a differenti-
patient at diagnosis). The use of fresh frozen
ation syndrome in 2% to 6% of patients and
plasma, cryoprecipitate, and platelets are
present with fever, respiratory distress,
important supportive care measures due to
weight gain, edema, and, importantly,
the frequently observed coagulopathy, hypo-
hyperleukocytosis. The incidence of ATRA
fibrinogenemia (fibrinogen <150 mg/dL),
syndrome has decreased due to the addition
and thrombocytopenia, respectively. This
of anthracycline therapy in induction and
will help prevent bleeding complications,
can be treated with IV dexamethasone.
especially in the APL patient (in addition
ATRA may cause other side effects including
to the early initiation of ATRA therapy).
dryness of the lips and mucosa, fever, trans-
aminitis, and headache associated with
Relapse
pseudotumor cerebri. Due to the rarity of
Even with current intensive therapies, only
APL in pediatric patients, the role of arsenic
about half of children diagnosed with AML
trioxide and transplant after relapse is still
are expected to be long-term survivors. As
poorly defined.
mentioned, patients with DS and APL have
156
Chapter 15
an improved prognosis. Some patients suc-
nucleoside analogues (clofarabine), farnesyl-
cumb to early mortality related to progressive
transferase inhibitors
(tipifarnib), histone
disease, infection, bleeding, and other com-
deacetylase inhibitors
(vorinostat), protea-
plications of therapy. After relapse, chemo-
some inhibitors (bortezomib), and demethy-
therapy alone results in <10% 1-year disease-
lating agents (decitabine and azacitidine).
free survival (DFS). For those patients that
can again obtain a morphologic remission
after reinduction chemotherapy, HSCT
Suggested Reading
improves survival to 30% to 50%. MRD after
reinduction therapy for AML relapse has
Pui CH, Carroll WL, Meshinchi S, Arceci RJ.
been shown to be a vital prognostic factor
Biology, risk stratification, and treatment of
in DFS after HSCT. Newer modalities of
pediatric acute leukemias: an update. J Clin
therapy are needed and are under investiga-
Oncol 29:551-565, 2011.
tion. These include targeted immunotherapy
Seibel NL. Treatment of acute lymphoblastic
(gemtuzumab targeted against CD33), tyro-
leukemia in children and adolescents: peaks
sine kinase inhibitors (lestaurtinib and sor-
and pitfalls. Am Soc Hematol Educ Prog
afenib for patients with FLT3 mutations),
374-380, 2008.
Central Nervous
16
System Tumors
Central nervous system (CNS) tumors are the
criteria useful in grading of tumors include
most common cancer diagnosed in children
cellular pleomorphism, mitotic rate, degree
after leukemia, accounting for 20% of all
of anaplasia, and presence or absence of
pediatric malignancies. The most common
necrosis. A growing number of chromo-
location is infratentorial in children up to 14
somal abnormalities and specific genomic
years of age, while supratentorial tumors are
signatures have been identified in pediatric
more common in adolescents. Spinal cord
brain tumors, and the use of these is becom-
tumors are also more common in adolescents
ing increasingly important for accurate
than in younger children (9% vs. 3%).
pathologic classification.
The cause of most pediatric CNS tumors
is unknown. The majority of them are
sporadic, although a small percentage is
Clinical presentation
associated with genetic disorders such as
neurofibromatosis, tuberous sclerosis, von
It is often difficult to make the diagnosis of
Hippel-Lindau syndrome, and Li-Fraumeni
a CNS tumor in a child. The presenting
syndrome (germ line mutation of p53, a
symptoms of CNS tumors can be quite
suppressor oncogene). Radiation therapy
variable, depending on the location of the
is the most frequently identified cause of
tumor, its rate of growth, and the age of the
pediatric CNS tumors; these occur as second
child. Additionally, most of the symptoms
malignancies in children who have previ-
are nonspecific, and occur more commonly
ously received radiation therapy for the
in children with a variety of benign condi-
treatment of leukemia or primary CNS
tions. Prompt diagnosis of a CNS tumor
tumors.
requires an appropriate index of suspicion
In general, brain tumor classification is
and a good understanding of the duration of
based on embryonic derivation and histo-
symptoms that is expected with other dis-
logic cell of origin, and occasionally on
orders. Due to the frequent association of
tumor location. The World Health Organi-
hydrocephalus with infratentorial tumors,
zation Classification System for brain
50% of children with brain tumors will
tumors contains more than
100 entries,
present with signs of increased intracranial
making it difficult for pathologists to agree
pressure (ICP), including lethargy, vomit-
on a diagnosis in specific cases. Microscopic
ing, and irritability. These symptoms may
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
158
Chapter 16
be less prominent in infants because the
Seizures are relatively rare in younger
cranial sutures remain open and the skull
children but occur twice as often in adoles-
can accommodate the increase in ventric-
cents (13% vs. 25%). They can be focal or
ular size. For this reason, accurate mea-
generalized. When focal they may provide a
surement of the head size should be part of
clue as to the location of the tumor. An
every well-baby examination. Headache is
often-overlooked symptom in school-age
a presenting symptom in 70% of children
children is deterioration in school perfor-
with posterior fossa tumors and 50% of
mance and behavioral changes. It is not
central tumors. Features that are helpful in
uncommon for these children to be referred
distinguishing tumor-related headache
by a psychologist or psychiatrist after many
from a primary headache syndrome
months of unsuccessful therapy for these
include nausea and vomiting (particularly
symptoms.
in the morning), increasing frequency and/
Symptoms of spinal cord tumors are
or severity over a few weeks, and abnor-
caused by compression of the spinal cord
malities on neurologic examination.
or nerve roots by the mass. The most com-
Neurologic deficits such as ataxia, visual
mon symptom in children with spinal cord
changes, and hemiparesis occur in about
tumors is back pain. Unlike adults, the
25% of cases. Weakness is sometimes sub-
complaint of back pain is very rare in child-
tle, but parents often relate a loss of phys-
hood. “Benign” causes of back pain include
ical function or developmental milestones.
trauma, medical conditions such as sickle
Visual changes include diplopia caused by
cell disease, or involvement in aggressive
cranial nerve dysfunction and loss of visual
sports such as football, gymnastics, skate-
acuity caused by optic nerve involvement
boarding, or motocross bicycling. In the
or papilledema.
absence of such a history, there should be
Two specific clinical syndromes are fre-
concern that a tumor might be the cause.
quently associated with childhood brain
Tumor-related pain is often described as
tumors. Parinaud’s syndrome consists of
being worse in the supine position or with
paralysis of upward gaze, accommodative
Valsalva’s maneuver. Other symptoms asso-
paresis, nystagmus during attempts at
ciated with spinal cord tumors include sci-
upward gaze, eyelid retraction
(Collier’s
atica, weakness, numbness, problems with
sign), and
“setting-sun sign”
(conjugate
or loss of bowel and/or bladder control, and
downward gaze). It is most commonly asso-
spinal deformity.
ciated with pineal region tumors, but can also
Tumors within the spinal canal are either
be caused by obstructive hydrocephalus from
intramedullary (within the spinal cord or
any cause, posterior fossa tumors, or a num-
nerve roots) or extramedullary. Intramedul-
ber of infectious or vascular abnormalities
lary spinal cord tumors are almost always glial
leading to ischemia or damage to the dorsal
in origin, the most common histologies being
midbrain. Diencephalic syndrome is a rare
astrocytoma, myxopapillary ependymoma,
cause of failure to thrive in infants and young
and oligodendroglioma. Extramedullary tu-
children. Characteristic features include pro-
mors are either intradural or extradural.
found emaciation with normal caloric intake,
Extramedullary intradural tumors are most
absence of subcutaneous fat, locomotor
commonly neurofibromas, Schwannomas, or
hyperactivity, euphoria, and alertness. It is
more rarely meningiomas. Extradural tumors
almost always associated with hypothalamic/
are usually not CNS tumors at all, but
suprasellar tumors, most commonly optic
metastatic tumors from other locations such
pathway gliomas.
as neuroblastoma, lymphoma, or Ewing
Central Nervous System Tumors
159
sarcoma. Tumors within the vertebral bodies
for identification of tumor within areas of
such as primary bone tumors, tumors met-
surrounding edema, detects focal areas of
astatic to bone, and histiocytosis can also
blood-brain barrier (BBB) breakdown, and
extend into the spinal canal and cause the
improves delineation of cysts from solid
above symptoms.
tumor.
Recent advances include MR spectros-
copy, tractography, and functional MRI
scanning. MR spectroscopy allows the
Diagnostic evaluation
determination of the chemical makeup of
tissue within a predefined region of interest.
Imaging studies
Different types of tumors have characteristic
The first step in evaluating a child with a
spectroscopic signatures, allowing the iden-
suspected CNS tumor is neuroimaging. A
tification of the most likely tumor type
variety of imaging techniques can be useful
before surgery takes place. Tractography is
in this assessment.
a software technique that allows identifica-
tion of the location of white matter tracts
Computerized tomography
and their associated nuclei. This has become
Computerized tomography (CT) was the
very useful as a preoperative planning tool
first imaging modality to revolutionize the
that allows the neurosurgeon to avoid dis-
diagnosis of CNS tumors both in children
ruption of eloquent nuclei and fiber tracts
and in adults. It is often still the first imaging
during surgery. Functional MRI is another
study obtained when evaluating patients
technique that precisely identifies specific
suspected of having a tumor. However,
motor regions, speech and language centers,
magnetic resonance imaging
(MRI) has
and other eloquent areas for preoperative
largely replaced it as the definitive neuro-
planning purposes.
imaging study. Advantages of CT include
widespread availability, rapid acquisition
Positron emission tomography
time, superior resolution of bone and vas-
Positron emission tomography (PET) imag-
cular structures including lesions of the
ing is a relatively new modality for evalu-
skull base, and utility in patients with con-
ating a variety of tumors including those of
traindications to MRI (e.g., ferrous metal
the CNS. It detects the relatively higher rate
implants). Its limitations include inferior
of metabolism present in many neoplasms
resolution of the brain itself, particularly
by measuring the rate of metabolism of
within the posterior fossa, and significant
one of a growing number of radioactively
radiation exposure, especially with regular
labeled compounds. It has been used to
monitoring.
distinguish between recurrent tumor and
necrotic tissue, scar, or edema, each of
Magnetic resonance imaging
which can develop following the treatment
An MRI with and without gadolinium
of a malignant brain tumor. It is used to
enhancement is the single best study for
determine the most appropriate site for a
evaluating the majority of tumors within
biopsy. Its utility in young children is lim-
the brain and spine. It provides greater
ited due to the need for a 30- to 60-minute
sensitivity in areas that may be obscured
period of limited mental activity between
by proximity to bone, such as the temporal
injection and scanning to optimize the dis-
lobe and posterior fossa. It also provides
tinction between normal and abnormal
multiplanar images. Contrast MRI allows
structures.
160
Chapter 16
Single-photon emission C T
Bone marrow aspiration
Single-photon emission CT is a three-
In the past it was recommended that patients
dimensional imaging technique introduced
with medulloblastoma undergo a bone mar-
in the early 1980s for the investigation of
row aspiration at the time of diagnosis to rule
regional cerebral blood flow that often cor-
out metastatic disease prior to starting ther-
relates with tumor metabolic activity. In this
apy. However, more recent studies have
technique, radioactive thallium and radiola-
indicated that fewer than 3% of patients
beled tyrosine have been used to localize
diagnosed with medulloblastoma have extra-
brain tumors as well as distinguish viable
neural metastases at diagnosis, with fewer
tumor from radiation necrosis. It is much
than 25% of these having disease in the
less expensive than PET scanning, but the
marrow. Therefore, recent clinical trials have
resolution is less and the 20- to 30-minute
no longer recommended routine examina-
acquisition times needed for high-quality
tion of the bone marrow, but have instead
pictures again limit the utility in young
recommended this only for patients with
children.
unexplained cytopenias. Extraneural metas-
tases are almost unheard of in other types of
pediatric CNS tumors, making bone marrow
Other studies
aspiration at diagnosis unnecessary.
Lumbar puncture
Bone scan
Certain primary CNS tumors such as
While 90% of extraneural metastases in
medulloblastoma, primitive neuroectoder-
patients with medulloblastoma are to bone,
mal tumor (PNET), ependymoma, malig-
this remains an unlikely presenting finding
nant germ cell tumors (GCTs), and, to a
since, as mentioned above, fewer than 3%
lesser extent, malignant gliomas have a pro-
of patients have extraneural metastases at
pensity to metastasize throughout the sub-
diagnosis. Therefore, bone scan should be
arachnoid space. As with other malignant
reserved for brain tumor patients who pres-
tumors, the presence of metastatic disease
ent with bone pain with the underlying
alters both treatment and prognosis. For
diagnosis of medulloblastoma.
this reason, a lumbar puncture (LP) is a
routine part of the diagnostic workup of
these patients. Spinal fluid is collected and
Treatment
sent for cell count and differential, glucose,
protein, and cytological examination to
Children with CNS tumors are typically
look for malignant cells. Measurements of
a-fetoprotein (AFP) and the beta subunit of
treated with some combination of neuro-
surgery, radiation therapy, and chemother-
human chorionic gonadotropin (b-HCG)
should also be performed on patients with
apy. The specific treatment chosen is depen-
suprasellar and pineal region tumors to
dent on the histology, location, resectability,
identify malignant germ cell tumors. These
and prognosis of the patient’s tumor.
markers can, if present, also be used to
measure response to therapy. The diagnostic
Neurosurgery
LP should be delayed for at least 7 days
Initial neurosurgical intervention is indi-
following tumor resection to allow any
cated in the majority of patients. Exceptions
nonviable tumor cells present in the CSF
include diffuse intrinsic pontine gliomas
as a result of the surgery itself to be cleared.
due to the infiltrative nature of the tumor
Central Nervous System Tumors
161
throughout the pons and tumors that are
6 years of age can be expected to suffer
radiographically consistent with germ cell
significant impairment of intellectual and
tumors and present with elevation of both
physical development as a result of this
AFP and b-HCG. This combination con-
therapy. A group of young Canadian chil-
firms the diagnosis of nongermanomatous
dren with medulloblastoma who received
germ cell tumor
(NGGCT) without a
“curative” doses of radiation therapy were
biopsy, and aggressive surgery at diagnosis
followed longitudinally into adulthood;
has not been shown to improve survival.
none of the survivors were capable of inde-
The role of surgery in children with func-
pendent living. Another concern is the
tioning pituitary adenomas is also limited.
development of secondary malignancies in
The goals of neurosurgical intervention
the radiation field. Children receiving pro-
are to achieve the maximum tumor removal
phylactic craniospinal radiation for leuke-
possible while minimizing morbidity and
mia have been shown to have a >20-fold
mortality and to obtain tissue for a histo-
increase in the risk of secondary CNS
logic diagnosis. In the case of medulloblas-
tumors; more than 80% of these tumors
toma, PNET, ependymoma, and high-grade
occurred in children who were radiated
gliomas, extent of surgical resection is one of
before 5 years of age. Children with standard
the major predictors of outcome.
risk medulloblastoma who survive >5 years
Children with increased ICP caused by
following conventional treatment including
obstructive hydrocephalus will likely have a
radiation without a relapse are more likely
CSF diversion procedure performed as well,
to die from a secondary radiation-induced
either before their definitive surgery if they
cancer than from a recurrence of their pri-
are unstable or at the time of resection. An
mary tumor.
external ventricular drain will often be placed
A variety of innovations in radiation
initially and will be clamped at some point
therapy delivery have been developed
after surgery to determine if normal CSF flow
recently to minimize the side effects of
has been restored. Children who redevelop
radiation. These include
3D conformal
symptoms of increased ICP will likely require
planning techniques and intensity-modu-
placement of a permanent shunt.
lated radiation therapy, which minimize the
dose of radiation delivered to adjacent nor-
Radiation therapy
mal tissue. Another innovation is proton
Radiation therapy has played a pivotal role
beam therapy, which takes advantage of the
in the management of children with malig-
shorter decay path of high-energy protons
nant brain tumors for decades. It has been
compared with gamma rays (photons). This
used to destroy remaining tumor in cases of
minimizes the exit dose delivered beyond
incomplete surgical resection and to treat
the site of the tumor. These innovations
microscopic residual disease and metastatic
have proven very valuable in decreasing the
disease in children who have malignant
radiation exposure to heart, lung, bowel,
tumors with a high likelihood of tumor
and reproductive organs in children receiv-
regrowth. The dose of radiation used is
ing whole spine radiation. Nevertheless, the
dependent on the type of tumor and the
use of radiation therapy continues to carry
location and volume to be irradiated.
the risk of significant morbidity. Work con-
Unfortunately, the use of radiation ther-
tinues to develop novel strategies that will
apy is associated with significant adverse late
allow the avoidance of radiation therapy
effects, particularly in young children.
completely in children with CNS tumors,
Those children receiving radiation before
or at least to delay it in very young children.
162
Chapter 16
Chemotherapy
Medulloblastoma is the most common
The utility of chemotherapy in the treat-
malignant CNS tumor in children, account-
ment of CNS tumors was recognized more
ing for up to 20% of all childhood brain
recently than in other tumors. It was initially
tumors. Supratentorial PNET is much less
felt that the BBB would prevent the delivery
common, comprising about 2.5% of cases
of effective concentrations of chemothera-
when pinealoblastoma is included. It has a
peutic agents to the tumor. However, it
propensity to metastasize early; in up to
became apparent that many drugs do ade-
40% of cases, widespread seeding of the
quately cross the BBB, or that this barrier is
subarachnoid space is present. Extraneural
abnormally permeable in many malignant
spread is unusual at diagnosis, but may
brain tumors. Other factors such as tumor
develop later if tumor recurrence occurs.
heterogeneity, cell kinetics, drug distribu-
The initial mode of therapy is surgery,
tion, and drug excretion may also have a
and extent of surgical resection directly
significant impact on effectiveness of
affects prognosis and treatment. Patients
chemotherapy. Lower grade tumors are
with <1.5 cm3 of residual tumor after sur-
characterized by a low mitotic index and
gery and no evidence of metastatic disease
relatively slow growth rate; it was long felt
are defined as standard risk, while patients
that these tumors would be less sensitive to
with >1.5 cm3 of residual disease or meta-
chemotherapy, while more malignant
static disease are considered high risk. Once
tumors would be more sensitive. However,
the diagnosis is confirmed, patients should
several studies over the last two decades
be staged with an MRI of the spine and an
have demonstrated that many low-grade
LP for cell count, glucose, protein, and
tumors are sensitive to chemotherapy.
cytology. Additional studies looking for
Commonly used drugs for the treatment
extraneural metastases are no longer con-
of brain tumors include vincristine, meth-
sidered necessary.
otrexate, temozolomide, procarbazine,
PNETs including medulloblastoma are
lomustine (CCNU), cisplatin, carboplatin,
very sensitive to radiation therapy, and as a
cyclophosphamide, and etoposide.
result this has traditionally been considered
the mainstay of postsurgical therapy. Due to
the high likelihood of metastases through-
Specific tumor types
out the CNS, it has been considered neces-
sary to deliver a dose of radiation to the
Primitive neuroectodermal tumor
entire brain and spine (craniospinal irradi-
PNETs are the most common malignant
ation; CSI) to these patients, with a higher
brain tumors in children. They are comprised
(boost) dose delivered to the region of their
primarily of small cells with hypochromatic
tumor. As the long-term consequences of
nuclei and can occur throughout the CNS.
radiation have become more apparent,
There has been controversy regarding
especially for young children, strategies to
whether these tumors should be classified
delay or avoid radiation in young children
as a single entity or divided by location.
have become more widely used and
The WHO has chosen the latter approach,
attempts have been made to decrease the
and divided these tumors into medullo-
doses of radiation given to older children.
blastoma
(occurring in the cerebellum),
The value of chemotherapy both in
pinealoblastoma
(occurring in the pineal
improving survival among patients with
gland), and supratentorial PNET (occurring
PNETs and in allowing a reduction in the
elsewhere in the supratentorial region).
dose of radiation therapy required for cure
Central Nervous System Tumors
163
has become increasingly evident over the
multimodality therapy is >80%. Children
past three decades. For the past several years
with high risk medulloblastoma and supra-
a combination of cisplatin, vincristine,
tentorial PNET do less well; the most
CCNU, and cyclophosphamide has been
recently published
5-year EFS rates are
used following completion of radiation
50% to 60%.
therapy with good effect. Vincristine is also
used during radiation therapy, and the
Gliomas
Children’s Oncology Group is evaluating
Gliomas account for just over 50% of all
whether the addition of carboplatin during
CNS tumors in childhood. They are a het-
radiation therapy will improve outcomes
erogeneous collection of tumors including
further in children with high risk disease.
astrocytoma (60%), ependymoma (15%),
Due to the devastating effects that CSI
oligodendroglioma (15%), mixed gliomas
can have in children less than 6 years of age
(5%), and other unspecified glial tumors
at the time of treatment, an alternative
(5%). They can occur anywhere in the CNS,
strategy was developed in the early 1990s
although certain subtypes have a propensity
that substituted very intensive chemother-
to occur in specific locations.
apy followed by consolidative myeloablative
Gliomas are classified histologically into
chemotherapy and hematopoietic stem cell
grades 1 to 4 based on cellular pleomor-
rescue without radiation therapy for these
phism, cell density, mitotic index, and
young children. Known as the Head Start
necrosis. Grade 1 and 2 tumors are defined
regimen, this strategy proved fairly effective
as low grade gliomas (LGG); they are con-
for children with standard risk embryonal
sidered by some to be “benign,” but many
tumors, but results were suboptimal for
neuro-oncologists disagree with this defini-
patients with high risk medulloblastoma
tion. Grade 3 and 4 gliomas are defined as
or supratentorial PNET, including pinealo-
high grade gliomas (HGG) and are clearly
blastoma. In 1995, the second Head Start
malignant. For most gliomas, the histologic
study added high-dose methotrexate to the
classification is the most important factor in
existing chemotherapy; results from this
choosing therapy and predicting long-term
modification were very encouraging.
outcome.
Patients with standard risk medulloblas-
Surgical resection is a key component of
toma had 5-year event-free survival (EFS)
therapy for most gliomas that are amenable
rates of 50% and overall survival rates of
to this approach. This includes cerebellar
75%. Children who relapsed following che-
gliomas and supratentorial gliomas that are
motherapy alone were effectively salvaged
not centrally located or within the optic
with reoperation and radiation therapy in
pathway. Gliomas occurring in the tectal
half of the cases. This strategy led to more
region of the midbrain are an exception
than 70% of children with standard risk
to this rule; these are very indolent tumors
disease treated on Head Start II being cured
that typically require no intervention. LGG
without radiation therapy. The Children’s
that are completely resected do not require
Oncology Group is investigating a similar
adjuvant therapy and are simply observed. If
strategy for children <3 years of age with
they recur, re-resection is generally curative.
high risk medulloblastoma and supraten-
Surgery should be used judiciously, how-
torial PNET.
ever. Most LGG can be controlled success-
The outlook for children >3 years of
fully with chemotherapy, so surgery that
age with standard risk medulloblastoma is
leaves the patient with significant postop-
quite good;
5-year EFS with current
erative morbidity should be avoided. HGG
164
Chapter 16
always require adjuvant therapy, but the
therapy has been estimated to prolong sur-
likelihood of cure is related to the degree
vival for a median of 2 months; since the
of surgical resection.
therapy takes 6 weeks to deliver, the impact
Radiation therapy was a key part of the
of this treatment on improving quality of
treatment of all gliomas for many years, but
life may be minimal, especially for children
it is used very rarely now to treat children
requiring anesthesia for their radiation.
with LGG due to the frequency of long-term
Many trials have been conducted over the
consequences of this therapy and the real-
past 25 years trying to improve the survival
ization that these tumors can be managed
of children with this tumor, all with disap-
successfully with chemotherapy. It remains
pointing results.
an essential component of therapy for HGG,
Although ependymomas are traditionally
however. Since these tumors metastasize
classified with other glial tumors, their behav-
much less frequently than PNETs, children
ior and management is quite different. They
do not require CSI, but they do require
represent about 5% to 10% of childhood
doses of 50 to 60 Gy to their tumor sites
primary brain tumors. About 60% of intra-
to achieve local control.
cranial ependymomas occur in the posterior
The value of chemotherapy in the man-
fossa and 40% are supratentorial. Grade 1
agement of children with LGG has become
ependymomas are rare and almost never
increasingly apparent over the past ten years.
occur in the brain, although Grade 1 (myx-
This is especially true for children with neu-
opapillary) ependymomas of the spine com-
rofibromatosis-associated LGG, as the fre-
prise about 10% of childhood ependymomas.
quency of secondary malignant tumors in
Grade 4 ependymoma does not exist.
these children after treatment with radiation
The extent of surgical resection is the
therapy is alarmingly high. The goal of ther-
most important prognostic factor in chil-
apy in this situation is different than with
dren with ependymoma. The 5-year EFS
other tumors. Chemotherapy rarely leads to
for patients having a complete resection is
total disappearance of the tumor, but rather
80%, versus 40% for those patients having a
produces a state of stable disease that is
near-total or subtotal resection. Unfortu-
hopefully maintained after therapy is discon-
nately, ependymomas are often difficult to
tinued. In this regard, management of
completely resect, especially in the posterior
incompletely resected LGG is similar to man-
fossa due to their predilection to surround
aging a chronic illness like asthma, with
cranial nerves and other vital structures. It
periods of stability, occasional flares of dis-
remains unclear that postoperative chemo-
ease, and a hope that the child will eventually
therapy improves survival in patients with
“grow out” of their condition.
ependymoma, but it may improve outcome
Gliomas arising in the brain stem deserve
when given before second look surgery to
special mention. These comprise about 15%
improve the likelihood that a complete
of brain tumors in children. About 70% of
resection can be achieved. Radiation ther-
these arise in the pons and are diffusely
apy remains an important component of
infiltrative, and are termed diffuse intrinsic
therapy for this tumor; the doses required
pontine gliomas. These are the most deadly
are similar to those needed to achieve local
of all childhood brain tumors; median sur-
control of other glial tumors. Ependymo-
vival of children with this diagnosis is 8 to 9
mas are more likely to metastasize within
months, and 90% of affected children are
the CNS than other gliomas, especially if
dead within 2 years of diagnosis regardless
they originate in the posterior fossa or are
of the treatment provided. Radiation
Grade 3. Assessment of the spinal fluid
Central Nervous System Tumors
165
postoperatively is important, although neg-
management quite challenging. The best
ative spinal fluid cytology does not reliably
outcomes for these children occur when
identify patients less likely to recur away
they are diagnosed early and treated at a
from their primary tumor site.
center with a multidisciplinary neuro-
oncology program. Therefore, every effort
Germ cell tumors
should be made to ensure that potential
CNS GCTs are relatively uncommon. Their
signs of a brain tumor are not missed and
management shares many similarities with
that early referral to such a multidisciplinary
GCTs occurring outside the CNS. They are
center occurs.
discussed in Chapter 21.
Suggested Reading
Summary
Robertson PL. Advances in treatment of pediatric
In summary, although brain tumors occur
brain tumors. NeuroRx 3:276-291, 2006.
relatively commonly in children, the varia-
Wen PY, Schiff D, Kesari S, et al. Medical man-
tions in histologic appearance, presentation,
agement of patients with brain tumors. J
treatment, and prognosis make their
Neurooncol 80:313-332, 2006.
Hodgkin and Non-
17
Hodgkin Lymphoma
Hodgkin and non-Hodgkin lymphoma
B-cells. In both forms of the disease, the
together account for approximately 10% to
tumor cells comprise only 0.1% to 10% of
12% of malignancies in children; they are
the cells within the tumor.
third in relative frequency after acute leuke-
Epidemiologic studies suggest that two
mias and brain tumors. Non-Hodgkin lym-
different forms of pediatric HL exist: child-
phoma
(NHL) comprises approximately
hood and adolescent/young adult (AYA).
60% of all lymphomas.
The childhood form is defined as occurring
in those
14 years of age, and is character-
ized by a male predominance and a histo-
Hodgkin lymphoma
logic subtype that is more likely to be mixed
cellularity (30% to 35%) or nodular lym-
Hodgkin lymphoma (HL) is rare among
phocyte predominant (10% to 20%). An
children <5 years of age and relatively rare
increased proportion of childhood HL is
in the adult population, but is the most
associated with Epstein-Barr virus (EBV)
commonly diagnosed cancer among adoles-
infection, and studies suggest that early
cents aged 15 to 19. HL is classified into two
exposure to EBV is a risk factor for devel-
general groups: classical HL (95% of cases)
oping HL in childhood. AYA HL occurs in
and nodular lymphocyte predominant HL
patients between 15 and 35 years of age.
(NLPHL, 5% of cases). Classical HL is further
There is no gender predilection. The most
subdivided into nodular sclerosis, mixed
common histologic subtype is nodular
cellularity, lymphocyte-depleted, and lym-
sclerosis
(70% to
80%). EBV infection
phocyte-rich forms. The tumor cells in clas-
appears to play a role in AYA HL also. Other
sical HL are designated Hodgkin and Reed-
infectious agents that have been associated
Sternberg (HRS) cells, whereas in NLPHL
with HL include human herpesvirus 6 and
they are designated lymphocyte-predomi-
cytomegalovirus. There is clearly an associ-
nant (LP) cells. HRS cells are now felt to
ation between HL and alterations in immune
derive from germinal center B-cells that have
function. HL is seen more commonly in
acquired unfavorable immunoglobulin V
patients with HIV disease, acquired immu-
gene mutations and that normally would
nodeficiency (e.g., hematopoietic stem cell or
have undergone apoptosis, whereas LP cells
solid organ transplant), and autoimmune
derive from antigen-selected germinal center
disease.
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Hodgkin and Non-Hodgkin Lymphoma
167
Clinical presentation
should be identified, as patients with HL
The most common presentation of HL is
can present with a variety of paraneoplastic
painless cervical or supraclavicular adeno-
syndromes. All superficial nodal groups
pathy. The nodes are typically described as
should be carefully examined, and the size
rubbery or firm and nontender, although
of any enlarged nodes and their character
may be sensitive to touch with rapid growth.
should be documented.
As the disease progresses, the nodes enlarge
2. The laboratory evaluation should
and often aggregate into larger masses that
include a complete blood count (CBC) with
may become fixed to the underlying tissue.
differential, erythrocyte sedimentation rate
As children often have reactive or infectious
(ESR), C-reactive protein (CRP), complete
adenopathy, it is common to have received
metabolic panel (including transaminases,
treatment with several courses of antibiotics
lactate dehydrogenase [LDH], and alkaline
before ultimately coming to biopsy. This is
phosphatase), serum copper, and serum
more common with cervical than with
ferritin. One or more of these nonspecific
supraclavicular adenopathy, as the latter is
inflammatory markers may be elevated and
less frequently associated with infection or
thus be useful initially to monitor response
inflammation and leads more quickly to
to therapy and later to identify possible
referral for possible malignancy. At least
recurrence.
two-thirds of patients with cervical or
3. A posterior-anterior
(PA) and lateral
supraclavicular adenopathy have mediasti-
chest radiograph will identify bulky medi-
nal involvement as well. This is frequently
astinal disease, if present. This is important,
asymptomatic, but may be associated with
as it will influence the approach used to
cough (usually nonproductive), shortness of
obtain diagnostic material since patients
breath, chest pain, or superior vena cava
with bulky mediastinal disease are more
syndrome. HL limited to infradiaphrag-
likely to experience complications with gen-
matic sites occurs in <5% of pediatric cases,
eral anesthesia. Other important imaging
occurring most commonly in femoral,
studies include a CT scan of the neck, chest,
superficial iliac, or inguinal nodes.
abdomen, and pelvis. The radiologist should
Constitutional symptoms commonly
be reminded to include the Waldeyer ring in
occur in patients with HL. They are caused
the scan as this nodal area can be involved
by cytokines produced by the HRS cells
with disease as well, especially in patients
within the tumor. Certain specific symp-
with upper cervical node involvement. Oral
toms known as “B symptoms” have been
and intravenous contrast are required for
demonstrated to have a negative prognostic
accurate identification of intra-abdominal
significance. These are unplanned weight
adenopathy. Splenic involvement occurs in
loss of >10% of body weight over the 6
30% to
40% of children at diagnosis.
months preceding diagnosis, drenching
Positron emission tomography
(PET)-CT
night sweats, or fever >38 C for at least
is now commonly used at diagnosis as a very
three consecutive days. Other common con-
sensitive tool for identifying sites of involve-
stitutional symptoms include generalized
ment, when it is available. A gallium scan
pruritus and alcohol-induced pain.
should be performed if PET is not available.
4. Bone marrow involvement at initial pre-
Evaluation
sentation of pediatric HL is uncommon and
1. A careful history and physical examina-
rarely occurs as an isolated site of extranodal
tion is imperative. The presence of B symp-
disease. However, it is seen more commonly
toms and any other unusual symptoms
in patients with advanced stage (stage III
168
Chapter 17
or IV) disease or patients with B symptoms.
therapy (RT). While this approach was fairly
These patients should undergo bone mar-
effective in curing the disease in low risk
row aspiration and biopsy from at least two
patients, it resulted in unacceptable muscu-
sites as part of their diagnostic evaluation.
loskeletal hypoplasia, cardiovascular and
All patients experiencing a recurrence of
pulmonary dysfunction, and the develop-
their disease should have their bone marrow
ment of subsequent secondary cancers. This
examined.
led to the development of combined modal-
5. Skeletal metastases are also rare at diag-
ity therapy with the goal of improving
nosis, and bone scans have typically been
event-free and overall survival, especially
performed only in patients with bone pain,
in higher risk patients, and decreasing the
unexplained elevations in the alkaline phos-
long-term adverse effects of a radiation
phatase, or other sites of extranodal disease.
only approach. Current research efforts in
PET-CT is a sensitive test for identifying
HL treatment seek to maintain the excellent
bone metastases and ultimately may replace
survival rates presently achieved while con-
bone scanning all together. It is important
tinuing to decrease the frequency of later
to remember that typical PET-CT imaging
adverse events. Today, patients with favor-
stops at the knee; patients with distal lower
able disease presentations receive fewer
extremity pain will need to have the exam
cycles of multiagent chemotherapy than
carried further or have alternative imaging
those with advanced and unfavorable clin-
of these areas.
ical presentations, either alone or combined
with low-dose, involved-field radiation.
Staging
With current therapy, more than 80% of
The most widely used staging system for
children diagnosed with classical HL are
pediatric HL is the Ann Arbor staging
expected to be long-term disease-free survi-
system. This system divides lymph node
vors, regardless of stage at diagnosis.
regions into nodal sites (e.g., neck, axilla,
For treatment purposes, patients with
and mediastinum), and the number of
HL are typically divided into low, inter-
involved sites then determines the stage.
mediate, and high risk disease groups.
Stage I disease involves only a single nodal
These groups are defined by clinical stage,
site, whereas stage II involves two or more
presence of bulky disease, and presence
sites that lie together on one side of the
of B symptoms. The current approach to
diaphragm. Stage III disease spans the dia-
therapy also includes the adaptation of
phragm and stage IV is disseminated sys-
treatment based on the patient’s response
temic disease. Additional modifiers include
to early therapy, a so-called response-based
A or B, to describe the absence or presence
approach.
of systemic symptoms respectively, and E,
A variety of chemotherapy agents have
involvement of a single extranodal site that
been shown to be effective in treating
is contiguous or proximal to a known nodal
HL. However, many early regimens such
site. The presence of bulky mediastinal
as nitrogen mustard, vincristine, procar-
disease, described as greater than one-third
bazine, and prednisone (MOPP) resulted
of the intrathoracic diameter, is also
in a very high rate of secondary malig-
considered.
nancy. Newer regimens have replaced
nitrogen mustard with cyclophosphamide
Treatment
(COPP) and added other agents such as
The first successful treatment strategy for
doxorubicin, bleomycin, vinblastine, and
HL was based solely on the use of radiation
etoposide.
Hodgkin and Non-Hodgkin Lymphoma
169
Classical HL—low risk disease
node can be successfully treated with surgery
A number of studies have attempted to elim-
alone. Stage I patients with more than one
inate the use of RT completely in children
involved node and stage II patients, which
with low risk disease. This approach has
represent more than 80% of patients with
resulted in an increased rate of relapse in
NLPHL, are successfully treated with similar
patients not receiving RT, but many of these
protocols used in low risk classical HL
patients were successfully salvaged with com-
patients. Current studies are investigating
bination chemotherapy and RT, thus long-
whether therapy can be reduced even further
term survival was not affected. The most
in this group of very low risk patients.
recent Children’s Oncology Group study
treated patients with three cycles of doxoru-
Complications of treatment
bicin, vincristine, prednisone, and cyclophos-
Short term
phamide. Those having a complete response
The chemotherapy regimens used for the
by PET-CT or gallium scan were observed;
treatment of HL are moderately emetogenic.
those having a partial response received
The nausea and vomiting can usually be
involved-field radiation therapy (IFRT).
controlled with serotonin receptor antago-
nist antiemetics such as ondansetron. It is
Classical HL—intermediate and
not uncommon for patients to develop
high risk disease
anticipatory nausea and vomiting with
The majority of patients with intermediate
repeated cycles; this can be managed with
risk disease either have stage I/II disease with
benzodiazepines such as lorazepam. Myelo-
one or more unfavorable features (B symp-
suppression occurs very commonly, espe-
toms, lymph node bulk, hilar lymph node
cially with the more intensive regimens, and
involvement, involvement of three or more
patients occasionally require hospitalization
lymph node regions, and extranodal exten-
for fever and neutropenia. Varicella zoster
sion to contiguous structures) or have
occurs frequently both during and after ther-
stage IIIA disease. High risk patients typically
apy, with a frequency that correlates with the
include those with stage IIIB or IV disease.
intensity of therapy. Appropriate preventive
Due to their more advanced stage, these
and therapeutic steps should be taken
patients are treated with more aggressive
quickly in the event of exposure or infection
chemotherapy followed by IFRT. Current
(see Chapter 25). Peripheral neuropathy can
therapy includes doxorubicin, bleomycin,
result from vincristine and vinblastine ther-
vincristine, etoposide, prednisone, and
apy, as can constipation. Pulmonary toxicity
cyclophosphamide (ABVE-PC); slow respon-
can result from bleomycin, with develop-
ders may also receive additional chemother-
ment of restrictive lung disease and reduc-
apy. The role of IFRT continues to be
tions in diffusion capacity. Anthracycline-
investigated in the treatment of intermediate
induced cardiac toxicity is unusual with the
risk patients, whereas high risk patients all
decreased doses used in current regimens,
receive consolidation with IFRT.
but is more common when combined with
mediastinal RT.
Nodular lymphocyte
predominant HL
Long term
Several studies have demonstrated that
The most significant long-term complica-
patients with NLPHL who present with
tion of HL therapy is the development of a
stage I disease and a single involved lymph
second malignant neoplasm. The frequency
170
Chapter 17
has decreased with recent modifications in
and has been linked to several subtypes of
chemotherapy and reductions in RT dose
NHL in the developed world. Other identi-
and volume, but it remains a concern dur-
fied risk factors include Helicobacter pylori
ing long-term follow-up. One specific issue
infection, tobacco exposure, and chemical or
is the risk of development of early breast
other environmental exposures. It is likely
cancer in female patients treated during
that there is not a single etiology responsible
adolescence, especially those treated with
for the development of all cases of NHL.
mediastinal radiation. Other potential
The histologic classification of NHL
long-term consequences of therapy include
has changed frequently over time and has
sterility/infertility (most common in males
become more precise as our understand-
treated with alkylating agents), secondary
ing of the development of lymphoma has
acute myelogenous leukemia
(etoposide,
improved and better diagnostic tools
alkylating agents), pulmonary fibrosis
(immunophenotyping, cytogenetics, molec-
(bleomycin), and atherosclerotic heart dis-
ular biology, and gene profiling) have been
ease (anthracyclines þ RT). Comprehen-
developed. The current World Health Orga-
sive long-term follow-up is essential for
nization classification is now widely used.
survivors of HL due to their increased risk
There are four major histologic subtypes of
of late complications of therapy.
NHL that occur commonly in children (in
order of frequency): diffuse, large B-cell
lymphoma (DLBCL), lymphoblastic lym-
Non-Hodgkin lymphoma
phoma (LL), BL, and anaplastic large cell
lymphoma (ALCL).
NHL is about 1.5 times as common as
DLBCL and ALCL tend to be heteroge-
Hodgkin lymphoma. The incidence is low
neous immunologically. Most are B-cell
in children less than 5 year of age, but from
derived, although some are T-cell derived
that point it increases steadily with age
or arise from the macrophage-histiocyte
throughout life. In all age groups, there is
lineage. Burkitt and Burkitt-like subtypes
a significant male predominance, particu-
are virtually all B-cell tumors, distinguished
larly among patients with Burkitt lym-
only by the amount of cellular heterogene-
phoma (BL). During childhood, the disease
ity. The most common site of occurrence of
is more common among non-Hispanic
LL is the mediastinum, and these tumors are
whites and Asians/Pacific Islanders; after
virtually all of T-cell origin. Less commonly
age twenty, it occurs more commonly in
LL occurs in bone or subcutaneously; these
African-Americans. The incidence of differ-
tumors are typically of B-cell origin. All of
ent histologic subtypes is very different in
these subtypes can invade the bone marrow
children and adults.
and undergo leukemic transformation.
Children with congenital or acquired
immune system dysfunction have a high risk
Clinical presentation
of developing lymphoma. Congenital condi-
The clinical presentation of NHL varies and
tions include ataxia-telangiectasia, as well
depends on the primary site of disease,
as Bloom, Wiskott-Aldrich, and Chediak-
histologic subtype, and extent of disease.
Higashi syndromes; acquired conditions
NHL can arise anywhere in the body, but
include HIV and prolonged immunosup-
occurs most frequently in the lymph nodes,
pressive therapy following bone marrow or
thymus, Waldeyer’s ring, Peyer’s patches,
solid organ transplant. EBV is clearly asso-
and bone marrow. In the United States,
ciated with BL in underdeveloped countries,
BL typically presents in the intestine,
Hodgkin and Non-Hodgkin Lymphoma
171
resulting in obstruction. These children
* A CBC with differential and review of the
usually present with nausea, vomiting, and
peripheral smear to assess for possible bone
abdominal distention. It is not unusual for
marrow involvement or leukemia.
NHL to be confused with a surgical abdo-
* Serum chemistries to include liver trans-
men, such as appendicitis. BL also presents
aminases, blood urea nitrogen, creatinine,
in Waldeyer’s ring or in the facial bones.
LDH, uric acid, electrolytes, calcium, and
T-cell LL most commonly arises from the
phosphorus. Lymphomas, particularly LL
thymus; these children typically present
and BL, can present with overt tumor lysis
with respiratory symptoms, cervical or
syndrome (TLS) and/or renal dysfunction.
supraclavicular adenopathy, or superior
The LDH can be useful prognostically, espe-
vena cava syndrome. Children may have
cially in patients with advanced stage
very limited disease affecting the tonsils,
disease.
nasopharynx, or Waldeyer’s ring, and the
* Bilateral bone marrow aspirations and
diagnosis has resulted from the pathologic
biopsies as the child may actually have leu-
examination following a tonsillectomy and
kemia, defined as a marrow with >25%
adenoidectomy. Lymphomas arising in less
replacement by lymphoblasts.
common sites such as superficial lymph
* A lumbar puncture to assess the cerebro-
nodes, bone, skin, thyroid, orbit, eyelid,
spinal fluid for involvement by NHL.
kidney, and the epidural space can be
caused by any subtype of lymphoma.
Radiologic procedures that should be per-
formed will vary depending on the location
Diagnostic evaluation
of the mass and associated symptoms, but
It is not unusual to begin with a surgical
should include:
procedure for excision or biopsy of a
* ApromptPAandlateralchestradiograph
node or laparotomy in the case of an acute
to evaluate for mediastinal involvement.
abdominal presentation. Many times the
* CT scan of the neck, chest, abdomen, and
diagnosis is not suspected prior to these
pelvis if the patient is sufficiently stable.
procedures. In any event, histologic confir-
* MRI of the brain for patients with immu-
mation is necessary for diagnosis. The diag-
nodeficiency-related NHL as these patients
nosis of NHL is usually made using biopsy
more commonly present with parenchymal
material but it can also be confirmed by
brain disease.
cytological examination of effusion fluids or
* MRI of the brain and spine for patients
review of bone marrow smears. In this way,
with focal neurologic symptoms.
major surgical procedures can often be
* PET-CT to include all identified sites of
avoided in patients presenting with BL and
disease. Ideally, this should be performed
LL, who are frequently unstable at presen-
following the diagnostic procedure, as these
tation. If possible, sufficient material should
patients frequently have residual abnormal-
be obtained for immunologic and cyto-
ities in their surgical bed on plain CT that
genetic or molecular biology studies. Many
are discovered on PET-CT to not represent
therapeutic trials now have requirements for
active disease.
submission of material for central pathology
review or biology studies; these require-
ments should be taken into consideration
Staging
when planning a diagnostic procedure.
NHL is staged using the St. Jude (Murphy)
The laboratory studies that should be
staging system that is presented in
obtained include:
Table 17.1.
172
Chapter 17
Table 17.1 St. Jude staging system for non-Hodgkin lymphoma.
Stage
Description
I
A single tumor (extranodal) or single anatomic area (nodal), excluding
mediastinum or abdomen
II
A single tumor (extranodal) with regional node involvement
On same side of diaphragm:
* Two or more nodal areas
* Two single (extranodal) tumors regional node involvement
* A primary gastrointestinal tract tumor (usually ileocecal) with or without
associated mesenteric node involvement, grossly completely resected
III
On both sides of the diaphragm:
* Two single tumors (extranodal)
* Two or more nodal areas
All primary intrathoracic tumors (mediastinal, pleural, thymic)
All extensive primary intra-abdominal disease; unresectable
All primary paraspinal or epidural tumors regardless of other sites
IV
Any of the above with central nervous system or bone marrow involvement
(<25%) at diagnosis
Treatment
regimens for patients with B-cell lym-
Therapy is based on clinical staging, local-
phoma, although clear evidence that it sig-
ized versus disseminated disease, and histo-
nificantly improves pediatric outcomes is
logic subtype. In general, children with
lacking. Patients with LL have superior out-
localized NHL have an excellent prognosis,
comes with longer acute lymphoblastic
with a 90% long-term survival. Therapy
leukemia-type therapy. CNS prophylaxis
may begin with surgical resection in the
in either type is indicated in advanced stages
case of primary gastrointestinal tumors that
and in those patients with parameningeal or
are amenable to complete resection, but this
overt CNS disease at diagnosis. The optimal
is not a necessary part of therapy for treat-
treatment of ALCL is not yet well defined.
ment of other sites (unless a life-threatening
Patients with stage I and II disease do well
complication is present).
with brief, multiagent intensive chemother-
These tumors are exquisitely sensitive to
apy and CNS prophylaxis. Patients with
chemotherapy, and multidrug regimens,
advanced stage disease who express the
similar to those used for leukemia, are fre-
fusion protein generated by the anaplastic
quently recommended. Radiotherapy is
lymphoma kinase (ALK) gene have superior
generally reserved for some patients with
outcomes compared to patients who are
CNSþ lymphoblastic lymphoma, relapsed
ALK negative. The latter patients typically
patients, or oncologic emergencies due to
require a more intensive, anthracycline-
tumor compression. In general, B-cell lym-
containing regimen.
phomas require intensive short-course ther-
With the initiation of therapy, the
apy ranging from 2 to 8 months in duration
child must be closely monitored for the
depending on initial tumor burden. Ritux-
development of TLS and resultant meta-
imab is now frequently added to treatment
bolic abnormalities. Some therapeutic
Hodgkin and Non-Hodgkin Lymphoma
173
regimens begin with a “reduction phase” of
Adolescents and Young Adults. New York:
treatment, using minimal therapy until the
Springer, 2007, pp 35-66.
Patte C, Bleyer A, Cairo M. Non-Hodgkin lym-
time of massive lysis has passed, then pro-
phoma. In: Bleyer WA, Barr RD (eds), Cancer
ceeding to the induction phase with insti-
in Adolescents and Young Adults. New York:
tution of multidrug therapy.
Springer, 2007, pp 127-149.
Punnett A, Tsang RW, Hodgson DC. Hodgkin
lymphoma across the age spectrum: epidemi-
Suggested Reading
ology, therapy, and late effects. Semin Radiat
Oncol 20:30-44, 2010.
Hudson MM, Schwartz C, Constine LS. Treat-
Shukla NN, Trippet TN. Non-Hodgkin’s lym-
ment of pediatric Hodgkin lymphoma. In:
phoma in children and adolescents. Curr
Bleyer WA, Barr RD
(eds), Cancer in
Oncol Rep 8: 387-394, 2006.
Wilms Tumor
18
Wilms tumor is the second most common
identified in two syndromes associated with
retroperitoneal tumor in children, account-
Wilms tumor: WAGR syndrome (Wilms
ing for approximately
6% of childhood
tumor, Aniridia, GU anomalies, and mental
malignancies. A tumor of the developing
Retardation) and Denys-Drash syndrome
kidney, it typically occurs in young children
(Wilms tumor, nephropathy, and GU
between the ages of 1 and 5 years, with equal
anomalies including pseudohermaphrodit-
incidence among boys and girls (though
ism). Beckwith-Wiedemann syndrome
interestingly occurs at earlier ages in boys).
(macroglossia, omphalocele, viscerome-
Most cases of Wilms tumor are sporadic,
galy, with or without hemihypertrophy)
with approximately 1% being familial and
is associated with imprinting defects at
2% to 4% associated with rare congenital
several genes at chromosome 11p15.5, a
syndromes. Familial cases are more likely to
locus referred to as the WT2 gene. Patients
present with bilateral tumors and occur at a
with this syndrome have a genetic predis-
younger age.
position to develop Wilms tumor, with an
incidence as high as 5% to 10%.
Genetics
Clinical presentation
Congenital anomalies occur in 12% to 15%
of cases, the most common being hemihy-
Most children with Wilms tumor are gen-
pertrophy, aniridia, and genitourinary tract
erally in good health at presentation and
(GU) anomalies such as cryptorchidism,
come to medical attention due to abdom-
hypospadias, horseshoe kidney, ureteral
inal enlargement or when a family member
duplication, and polycystic kidney. Most
has felt a mass. Associated signs and symp-
children with Wilms tumor have a normal
toms include abdominal pain, malaise,
karyotype; however, a chromosomal dele-
fever, hypertension, and microscopic
tion in the short arm of chromosome 11
hematuria. Bleeding within the tumor may
(11p13 deletion) is seen in association with
occur and result in anemia, pallor, and
congenital aniridia. The Wilms tumor 1
fatigue. Tumor thrombus may extend into
(WT1) gene encodes a transcription factor
the vena cava, causing partial obstruction,
important in normal kidney development.
hypertension, and distention of abdominal
Mutations within the WT1 gene have been
veins. Polycythemia may be seen in Wilms
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Wilms Tumor
175
tumor and may or may not be related to
patients with Wilms tumor with lung or
elevated erythropoietin levels. Acquired
liver metastatic disease and bony
von Willebrand disease with reduced von
symptoms
Willebrand factor (VWF), factor VIII coag-
* MRI of the brain for metastases if the
ulant, and ristocetin cofactor levels has
tumor is a rhabdoid tumor or clear cell
been reported in approximately
8% of
sarcoma
patients newly diagnosed with Wilms
* Echocardiogram for detecting tumor
tumor and typically resolves with therapy
extension from the IVC to the right
initiation. Its occurrence in Wilms tumor is
atrium and also indicated in the child
thought secondary to binding of VWF to
receiving anthracycline chemotherapy
the tumor with subsequent degradation as
well as hyaluronic acid secretion by nephro-
Pathologic diagnosis
blastoma cells leading to decreased efficacy
Children with suspected Wilms tumor
of VWF.
should have an immediate consultation
with a pediatric surgeon. Surgical removal
Evaluation of suspected Wilms
of the involved kidney and intact tumor is
tumor
indicated at diagnosis if possible. Cases
* History: include family history of malig-
with tumor in the renal pelvis and IVC
nancies, congenital anomalies, or syndromes
require a detailed imaging assessment to
* Physical examination: assess gently for
guide the surgeon. In some cases, complete
abdominal/flank mass (usually distinct and
removal of the primary tumor at diagnosis
not crossing the midline), congenital anom-
may not be possible. In these cases, open
alies (hemihypertrophy, GU malformations,
biopsy is indicated for pathologic confir-
aniridia), hypertension, abdominal venous
mation and determination of histology.
distention, and liver enlargement
Tumor pathology is critical in diagnosis
* Laboratory studies: urinalysis with
as well as helping to determine the staging
microscopic evaluation, complete blood
and therefore subsequent treatment.
count (polycythemia and anemia), serum
Classic Wilms tumor presents with three
chemistries
(hypercalcemia in rhabdoid
types of nephroblastoma tissue elements:
tumor or congenital mesoblastic nephroma),
blastema, stroma (mesenchyme), and epi-
and coagulation studies including von Will-
thelium. Additional assessment includes
ebrand panel
determination of histology as either favor-
* Diagnostic imaging studies:
able
(well-differentiated components) or
* Doppler abdominal ultrasound to eval-
unfavorable
(presence of anaplasia with
uate the tumor, possible vena cava
poor differentiation and a worse prognosis).
involvement, blood flow, and contralat-
Other malignant tumors of the kidney need
eral kidney
to be considered and ruled out including
* Abdominal CT or MRI with special
rhabdoid tumor of the kidney, clear cell
attention to evidence of bilateral involve-
sarcoma of the kidney, and renal cell carci-
ment, vessel involvement or extension to
noma. Chromosome analysis is performed
the inferior vena cava (IVC), lymph node
on the tumor and may prove to be especially
involvement, and liver metastases
useful in association with congenital anom-
* Chest radiography and chest CT to assess
alies and familial cases. Loss of heterozygos-
for lung metastases (most common site)
ity (LOH) of chromosomes 1p and 16q has
* Bone scan to assess for metastases if the
also recently been confirmed to be a nega-
tumor is a clear cell sarcoma and for all
tive prognostic indicator.
176
Chapter 18
Staging
Treatment
Staging is done postoperatively and is deter-
The prognosis of Wilms tumor is excellent,
mined based on the extent of disease, ability
with more than 85% of children being cured
to perform a total resection, and the path-
of their disease. Histology is more prognos-
ologic findings. Patients unable to undergo
tic for outcome than staging; for example,
a radical nephrectomy at diagnosis due to
patients with stage IV disease but with
excessive risk receive preliminary staging
favorable histology still have a 4-year sur-
and preoperative chemotherapy with
vival of more than 85%, compared to chil-
delayed resection. A careful pathologic eval-
dren with diffuse anaplasia and advanced
uation at the time of excision should be
disease who have very poor outcomes. Chil-
performed regardless of whether this occurs
dren with stage I and II favorable histology
at diagnosis or later following preoperative
tumors have an event-free survival rate
chemotherapy, as the histology may be dif-
exceeding 95%.
ferent than that seen on initial biopsy. This
Many tumors are low stage at presenta-
evaluation includes identifying tumor his-
tion and are able to be fully resected.
tology and looking for extension of disease
Additionally, these tumors are very sensitive
outside the kidney, including penetrance of
to chemotherapy and radiation. Based on
the tumor through the renal capsule into the
the exceedingly good outcomes, contempo-
renal pelvis and vessels and presence of
rary studies have looked to decrease therapy
nodal involvement.
in the lower risk stratum. This includes the
Clinicopathologic staging is as follows:
elimination of chemotherapy in patients
* Stage I: tumor limited to the kidney and
younger than 2 years with stage I disease
completed excised without tumor rupture
and tumor mass of <550 g without LOH at
or biopsy prior to resection
1p and 16q. For patients receiving chemo-
* Stage II: tumor extends through the renal
therapy, the backbone of therapy is based on
capsule, has been biopsied, or ruptured
previous studies by the National Wilms
prior to excision with spillage confined to
Tumor Study Group and includes vincris-
the flank; tumor may involve the perirenal
tine, doxorubicin, and dactinomycin.
soft tissue and/or infiltrate vessels outside
Higher stage patients may require radiation
the kidney but is completely excised
therapy in addition to this chemotherapeu-
* Stage III: residual nonhematogenous dis-
tic backbone. Patients with stage IV disease
semination of tumor confined to the abdo-
or patients with advanced stage and LOH at
men; tumor may extend beyond surgical
1p and 16q receive more aggressive therapy
margin at resection, may involve local
with the addition of cyclophosphamide and
lymph nodes, or have tumor spillage with
etoposide. In general, patients with pulmo-
peritoneal implants
nary metastasis will require lung irradiation
* Stage IV: hematogenous dissemination
although newer protocols are studying
of tumor to lungs, liver, bone, brain, or
whether this can be eliminated in patients
distant lymph nodes
with a rapid response to chemotherapy and
* Stage V: bilateral renal involvement
no additional risk factors. Finally, patients
with higher stage anaplastic disease as well
Patients with bilateral involvement of tumor
as patients with rhabdoid tumor of the
(stage V) should have each side indepen-
kidney or metastatic clear cell sarcoma
dently staged with treatment based on the
receive a similar regimen with the addition
side with the highest stage.
of carboplatin.
Wilms Tumor
177
Relapse is uncommon in Wilms tumor;
patients with right-sided tumors who
patients may be salvaged with aggressive
receive flank irradiation and those receiving
multiagent chemotherapy, radiation if pre-
whole abdomen irradiation have an
viously not given, and possibly may benefit
increased risk of developing a secondary
from high-dose chemotherapy with autol-
liver cancer. These potential issues must all
ogous stem cell rescue. As survival is excel-
be taken into account in the patient’s long-
lent, much attention has focused on late
term follow-up plan.
effects related to therapy and limiting
potential toxicities. Patients are at risk for
Suggested Reading
hypertension and renal insufficiency, as
most children will have a solitary remaining
Davidoff AM. Wilms tumor. Curr Opin Pediatr
kidney. Patients with bilateral disease,
21:357-364, 2009.
partial resection, and radiation exposure are
Nakamura L, Ritchey M. Current management of
particularly at risk for developing protein-
Wilms tumor. Curr Urol Rep 11:58-65, 2010.
uria and renal compromise in addition to
Pritchard-Jones K. Nephrectomy only for Wilms
hypertension. Cardiac complications may
tumor: negotiating the tangled web requires
be seen in those patients who receive anthra-
multiprofessional input. Pediatr Blood Can-
cyclines and lung irradiation. Additionally,
cer 54:865-866, 2010.
Neuroblastoma
19
Neuroblastoma is a neoplasm of the sym-
commonly, amplification of the oncogene
pathetic nervous system and the most com-
N-myc in tumor tissue has been found to
mon solid tumor of early childhood, with a
have a profound negative impact on prog-
peak incidence around 2 years of age. Neu-
nosis. Similarly, a near diploid (low DNA
roblastoma is a disease of developing neural
index, DNA index ¼ 1) chromosome num-
crest tissue and has an extremely heteroge-
ber in the tumor has also been found to be a
neous prognosis based on multiple factors
negative prognostic indicator as compared
such as the age of the patient, differentiation
to patients with a DNA index >1. Based on
and biology of the tumor, and extent of
these factors, in addition to age, stage, and
disease spread. Screening in the Japanese
histology (favorable or unfavorable based
infant population found a high incidence
on mitosis-karyorrhexis index and maturity
of neuroblastoma, although the majority of
of the tumor), a complicated risk-stratified
these cases matured or involuted spontane-
treatment regimen has been created.
ously, again implying the heterogeneous
prognosis based on patient age. Although
survival rates for neuroblastoma have
Clinical presentation
improved over the last 30 years, the majority
of these gains have been observed in patients
As neuroblastoma originates from primor-
with lower risk disease. Recent break-
dial neural crest cells that normally give rise
throughs hold promise to increase survival
to the adrenal medulla and the sympathetic
in those with high risk disease as well.
ganglia, tumors present in the abdomen and
Familial cases of neuroblastoma are
along the sympathetic neural pathway. The
quite rare. However, genetic risk factors for
most common presentation is an abdomi-
neuroblastoma have been identified includ-
nal mass with primary tumor in the adrenal
ing the anaplastic lymphoma kinase 1 and
gland, often with metastatic disease via lym-
PHOX2B homeobox genes. Patients with
phatic and hematogenous spread. Tumors
Hirschsprung’s disease, neurofibromatosis
are often seen along the paraspinal ganglion
type I, and congenital hypoventilation syn-
and may be found in the neck, thorax, and
drome may have genetic alterations in
pelvis. Infants are more likely to have tho-
PHOX2B and an increased risk of develop-
racic and cervical tumors. Common sites of
ing neuroblastoma, although clear genetic
metastasis are the lymph nodes, bone mar-
links have not as yet been identified. More
row, bone, liver, and skin.
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Neuroblastoma
179
The signs and symptoms at presentation
be carefully evaluated for the presence of
depend on the site of the tumor, size, and
an occult neuroblastoma. Children with
the degree of spread. Patients with locore-
OMAS tend to have a low-stage, highly
gional disease may be relatively asymptom-
curable neuroblastoma. Unfortunately, this
atic, whereas patients with widely metastatic
cure often has little impact on the OMAS,
disease are often ill-appearing with fever,
and these children are frequently left with
pain, weight loss, and irritability. Abdomi-
severe, chronic neurologic deficits. The
nal tumors are usually palpable, hard, fixed
pathophysiology of this syndrome is not
masses. The liver may be enlarged, leading
well defined but is likely due to formation
to respiratory compromise, especially in the
of an antibody directed against the neuro-
infant. There may be evidence of anemia
blastoma cells also targeting cerebellar neu-
(pallor, weakness, and fatigue), coagulopa-
rons. In addition to treatment of the neu-
thy (bruising and bleeding), and bone pain
roblastoma, these children may benefit
or limping with bone or bone marrow
from immunosuppressive therapy with
involvement. Thoracic masses are usually
agents such as dexamethasone, cyclophos-
picked up in the posterior mediastinum
phamide, intravenous immune globulin
incidentally by imaging studies done for
(IVIG), and adrenocorticotropic hormone
other reasons. Cervical masses may initially
(ACTH). Neuroblastoma patients may also
be treated as cervical adenopathy related to
present with hypersecretory diarrhea sec-
infection. The presence of Horner’s syn-
ondary to hypersecretion of vasoactive
drome (miosis [contracted pupil], ptosis,
intestinal peptide
(VIP) from the neuro-
enophthalmos
[posterior eye displace-
blastoma cells. In most cases, treatment of
ment], and anhidrosis) or heterochromia
the underlying disease will eliminate the
iridis should prompt an evaluation for cer-
VIP hypersecretion.
vicothoracic neuroblastoma. Pelvic masses
may cause bowel or bladder symptoms and
tumors along the sympathetic ganglia may
Diagnostic evaluation
cause spinal cord compression. Skin lesions
tend to be limited to infants and appear as
History
bluish, nontender subcutaneous nodules.
Assess for constitutional symptoms, abdom-
Sphenoid or retro-orbital bone involvement
inal pain, bowel or bladder control problems,
may occur and appear clinically as “raccoon
bleeding, bone pain, and limping.
eyes” secondary to periorbital hemorrhage.
In addition to Horner’s syndrome, the
Physical examination
clinician should be aware of potential para-
Assess vital signs (fever and hypertension)
neoplastic syndromes with neuroblastoma.
and evaluate for abdominal mass, spinal
One unusual presentation is opsoclonus-
cord compression, unusual signs/symptoms
myoclonus-ataxia syndrome (OMAS), also
associated with neuroblastoma
(hetero-
referred to as “dancing eyes/dancing feet.”
chromia, raccoon eyes, Horner’s syn-
These children have cerebellar and truncal
drome), subcutaneous nodules in infants,
ataxia as well as myoclonus (muscle jerks)
enlarged liver or lymph nodes, and evidence
and opsoclonus (rapid, involuntary, unco-
of anemia or coagulopathy.
ordinated eye movements). Developmental
delay, language deficits, and behavioral
Laboratory studies
abnormalities
(e.g., irritability) may also
Obtain complete blood count with differ-
be present. Children with OMAS should
ential, serum chemistries
(liver function
180
Chapter 19
studies and lactate dehydrogenase), ferritin
* Bone scan (technetium-99m).
(may be a tumor marker and elevated in
* MIBG (metaiodobenzylguanidine) scan
neuroblastoma, though also an acute-phase
(123I-MIBG scan preferred over131I-MIBG
reactant), and urine for catecholamines
due to decreased risk to thyroid function).
including homovanillic acid
(HVA) and
Prior to MIBG scanning, the patient must
vanillylmandelic acid (VMA). These urine
receive thyroid protection with potassium
metabolites are elevated in more than 90%
iodide
(SSKI) drops and should have
of children with neuroblastoma, especially
baseline thyroid function
(FT4/TSH)
in the higher stages of disease. In patients
checked. MIBG, a functional analog of
with clinical suspicion for neuroblastoma
norepinephrine, is taken up by sympathetic
and negative urine markers, urine dopa-
neurons and is a sensitive test for neuro-
mine can be measured in addition to
blastoma cells. It is also positive in
24-hour urine collections for HVA and
pheochromocytoma.
VMA. Neuron-specific enolase
(NSE), a
* Due to the high radiation doses with both
serum marker specific to the sympathetic
bone scan and MIBG scanning, patients can
nervous system, is relatively specific to neu-
be followed solely with MIBG if results of
roblastoma although it can be elevated after
baseline imaging are concordant.
brain injury and seizures.
Localized tumors should be surgically
Diagnostic studies
removed if it deemed safe and feasible.
Patients with confirmed neuroblastoma
Many tumors are initially unresectable.
should have bilateral bone marrow aspira-
These tumors should be surgically biopsied,
tion and biopsy performed to assess for
with sampling of local lymph nodes. Diag-
bone marrow involvement. Special stains
nosis is generally established by pathologic
(S100, synaptophysin, and NSE) are used
assessment of the resected or biopsied mass
to differentiate this tumor from other small,
and can be confirmed by diagnostic features
round blue cell tumors of childhood such as
such as bone marrow involvement with
rhabdomyosarcoma, Ewing sarcoma, lym-
appropriate immunohistochemical mar-
phoma, and primitive neuroectodermal
kers, MIBG avidity, and positive urine cate-
tumor. Lumbar puncture should be avoided
cholamines and serum NSE.
as this procedure may increase the incidence
of central nervous system metastases.
Imaging studies should include:
Staging
* CT or MRI of the primary site (MRI is
preferred due to the radiation exposure
The International Neuroblastoma Staging
associated with repeated CT imaging). Cal-
System as shown in Table 19.1 was most
cifications and hemorrhage are commonly
recently revised in 1993 and provides a basic
seen, especially in large abdominal masses.
prognostic model that has been more
The tumors tend to be large and displace
recently complemented by biologic and
adjacent organs; however, the tumor can
pathologic assessments including N-myc
wrap around major structures, causing
amplication, DNA ploidy, and histology.
obstruction and dysfunction. If there is
A new risk-stratified staging system by the
suspicion of paraspinal or intraspinal
International Neuroblastoma Risk Group is
involvement, MRI and plain films of the
currently being prospectively evaluated in
spine should be done.
clinical trials.
Neuroblastoma
181
Table 19.1 International Neuroblastoma Staging System.
Stage I
Localized tumor confined to the area of origin; complete gross resection, with or
without microscopic residual disease; negative lymph nodes
Stage IIA
Unilateral tumor with incomplete gross resection; negative lymph nodes
Stage IIB
Unilateral tumor with complete or incomplete gross resection; positive ipsilateral
lymph nodes, negative contralateral lymph nodes
Stage III
Tumor infiltrating across midline with or without regional lymph node involve-
ment, unilateral tumor with contralateral lymph node involvement, or midline
tumor with bilateral regional lymph node involvement
Stage IV
Tumor disseminated to distant lymph nodes, bone, bone marrow, liver, or other
organs (except as defined by stage IVS)
Stage IVS
Localized primary tumor as defined for stage I or II with dissemination limited to
liver, skin, and/or bone marrow (under 1 year of age and <10% bone marrow
involvement)
Treatment
of age with unfavorable histology and/or
DNA ploidy ¼ 1
and all patients
>18
Risk stratification divides patients into mul-
months of age
tiple potential treatment categories that are
4. Stage IVS patients with N-myc nonam-
continually evaluated through clinical stud-
plification who have unfavorable histology
ies. For simplicity, we divide treatment
and/or DNA ploidy ¼ 1
strategies into three main groups: (1) obser-
vation, (2) chemotherapy, and (3) high-dose
High-dose chemotherapy with
chemotherapy with autologous stem cell res-
autologous transplant
cue and adjuvant therapy with immunomo-
* Stage III, IV, and IVS patients not
dulators (described below).
included above and all patients >12 years
of age with stage III or greater disease
Observation
1. All Stage I patients
(after surgical
As mentioned, clinical studies are ongoing
resection)
to refine treatment groups. Specifically,
2. Stage IIA/B patients with >60% tumor
attempts are being made to reduce treat-
resection and N-myc nonamplification
ment in some of the lower risk subgroups
3. Stage IVS patients with N-myc nonam-
that still require therapy but have an excel-
plification, favorable histology, and DNA
lent overall prognosis
(>90% survival).
ploidy >1
Novel therapeutic agents are being utilized
in addition to the current backbone of
Chemotherapy
therapy that includes drugs such as carbo-
1. Stage IIA/B patients with
<60%
platin, cyclophosphamide, doxorubicin,
resection
and etoposide. The majority of work is
2. Stage III patients with N-myc nonampli-
centered on high risk patients who continue
fication, not including patients >18 months
to have an extremely poor prognosis (30%
of age with unfavorable histology
to 40% survival). Multiple efforts are cur-
3. Stage IV patients with N-myc nonampli-
rently being evaluated in clinical trials
fication, not including patients >12 months
including the benefit of myeloablative
182
Chapter 19
chemotherapy with autologous stem cell
patients, potentially increasing survival in
rescue as well as tandem transplantation.
this population to as high as 60%.
Therapy with isotretinoin
(cis-retinoic
acid; cis-RA) has been shown beneficial as
amaturing agent in neuroblastoma patients
Suggested Reading
after cytotoxic therapy with minimal
residual disease and is being evaluated in
Maris JM. Recent advances in neuroblastoma.
combination with monoclonal antibodies
N Engl J Med 362:2202-2211, 2010.
directed at neuroblastoma-specific anti-
Park JR, Eggert A, Caron H. Neuroblastoma:
gens, specifically gangliosidase
(GD2).
biology, prognosis, and treatment. Hematol
Pilot studies with anti-GD2 antibodies and
Oncol Clin N Am 24:65-86, 2010.
concomitant use of cis-RA and cytokine
Yu AL, Gilman AL, Ozkaynak MF, et al. Anti-
therapy (interleukin-2 and GM-CSF) have
GD2 antibody with GM-CSF, interleukin-2,
shown promising results in the high risk,
and isotretinoin for neuroblastoma. N Engl J
poor prognosis subgroup of neuroblastoma
Med 363:1324-1334, 2010.
Sarcomas of the Soft
20
Tissues and Bone
Sarcomas comprise a heterogeneous group
Osteosarcoma is believed to arise from
of malignant tumors that arise in the soft
osteoblasts and has a bimodal age distribu-
tissues or bone. Soft tissue sarcomas (STS)
tion with an initial peak in the second
are derived from primitive mesenchymal
decade of life and a second peak in older
cells such as muscle, connective tissue (ten-
adulthood. Ewing sarcoma is thought to
dons and synovial tissue), supportive tissue
derive from neural crest and is also most
(fat and nerves), and vascular tissue (lymph
frequently diagnosed in the second decade
and blood vessels). Rhabdomyosarcoma
of life.
(RMS) is the most common STS in younger
children, comprising more than half of these
tumors in children up to 9 years of age.
Genetics
Other nonrhabdomyomatous soft tissue
sarcomas
(NRSTS) include synovial sar-
The majority of soft tissue and bone sarco-
coma, liposarcoma, fibrosarcoma, alveolar
mas develop without a predisposing genetic
soft part sarcoma, leiomyosarcoma, and
risk factor, although there have been mul-
malignant peripheral nerve sheath tumor.
tiple reported associations: RMS in patients
STS account for approximately 6% of can-
with neurofibromatosis type 1, Beckwith-
cer cases in those less than 19 years of age
Wiedemann syndrome, Costello syndrome,
(annual incidence 11 cases per million in the
Li-Fraumeni familial cancer syndrome, and
United States). The relative incidence of
cardio-facial-cutaneous syndrome. There
various STS varies by age, gender, and race.
has also been an association with maternal
For instance, NRSTS such as fibrosarcoma
use of marijuana and cocaine as well as first-
and malignant hemangiopericytoma are
trimester exposure to radiation, including
more common in infants (less than 1 year
diagnostic radiographs. Children with
of age). NRSTS also account for the major-
hereditary retinoblastoma (who harbor the
ity of STS in patients over 10 years of age.
Rb germ line mutation) are at increased risk
The most common bone sarcomas in
of developing sarcoma. Alveolar RMS
children are osteosarcoma and Ewing sar-
(ARMS) has a characteristic translocation
coma (annual incidence 8.7 per million).
of the FKHR gene at 13q14 with PAX3 at
Approximately two-thirds of cases are oste-
2q35 or less commonly PAX7 at 1p36, lead-
osarcoma and one-third Ewing sarcoma.
ing to a fusion transcription factor that is
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
184
Chapter 20
thought to inappropriately activate gene
bone turnover. Soft tissue and bone sarco-
transcription. Embryonal RMS (ERMS) dis-
mas also occur as secondary malignancies.
plays a loss of heterozygosity in the short
arm of chromosome 11, especially at 11p15.
The allelic loss of 11p15 has also been seen in
Soft tissue sarcoma
translocation negative, histologic ARMS. In
general, patients with ERMS have a better
Clinical presentation
prognosis than patients with ARMS, in large
STS may occur anywhere in the body and
part due to the increased risk of metastatic
are not limited to muscle or connective
disease at presentation with ARMS. DNA
tissues. The most common sites for RMS
content or ploidy has also been implicated
are head and neck (35%), genitourinary
in prognosis with hyperdiploid tumors
system
(26%), and extremities
(20%).
(
51 chromosomes) having a better out-
Younger children less than 10 years of age
come. NRSTS can occur in sites of prior
tend to develop ERMS in head and neck or
radiation, and children with human immu-
genitourinary locations, whereas adoles-
nodeficiency virus may develop leiomyosar-
cents tend to develop extremity, truncal,
coma (often in association with Epstein-
or paratesticular ARMS. The majority of
Barr virus). Interestingly, family members
RMS are localized, and, of the
15% to
of children affected with Ewing sarcoma
20% of cases with metastatic disease at
family of tumors (ESFT; also referred to
presentation, most involve the lungs, bone
as primitive neuroectodermal tumors) have
marrow, lymph nodes, or bone.
an increased risk of neuroectodermal and
As RMS can arise from anywhere in the
stomach malignancies. The translocation
body, the presenting symptoms are highly
t(11;22)(q24;q12) occurs in greater than
variable. Typically, RMS presents as a pain-
95% of ESFT and some consider this finding
less growing mass. Depending on location,
to be pathognomonic and sufficient to con-
adjacent structures may be compressed and
firm the diagnosis. This translocation results
lead to symptoms such as airway obstruc-
in the EWS-FLI1 fusion transcript that acts
tion or cranial nerve findings with nasopha-
as an aberrant transcription factor altering
ryngeal tumors, proptosis with orbital
tumor suppressor gene pathways.
tumors, and urinary obstruction with blad-
Osteosarcoma is characterized by com-
der or prostate tumors.
plex unbalanced karyotypes with alterations
in the Rb and p53 tumor suppressor path-
Diagnostic evaluation
ways, linking this tumor to retinoblastoma
Patients presenting with an enlarging mass
and Li-Fraumeni syndrome, respectively.
suspected to be malignancy should have an
Osteosarcoma has long been thought to
open surgical biopsy for confirmation of
be associated with adolescent growth spurts
diagnosis. Sufficient tissue should be
and the higher incidence in large dog breeds
obtained for pathologic evaluation and
and taller people support this correlation.
staining in addition to cytogenetic and
Girls have a peak incidence younger than
molecular biologic studies. Patients may
boys (12 vs. 16 years) correlating with the
be eligible to participate in a clinical ther-
different average age for pubertal develop-
apeutic or biologic trial that also mandates
ment. Well-documented risk factors include
submission of tissue. A complete assessment
radiation exposure, Paget’s disease, and
of extent of disease with meticulous atten-
other disorders associated with increased
tion to sites of metastatic disease is
Sarcomas of the Soft Tissues and Bone
185
mandated prior to initiating definitive ther-
essential to tailoring of therapy to maximize
apy. Some protocols also mandate biopsy of
outcome. A unique aspect of the RMS stag-
regional nodes or a sentinel node in patients
ing is the postoperative clinical grouping
with primary extremity tumors. These eva-
system that is based on extent of surgical
luations include:
resection and results of the lymph node
* Complete history and physical examina-
evaluation. Risk group classification is then
tion with careful assessment of the mass,
based on pretreatment stage, postoperative
adjacent structures, and regional lymph
clinical group, and histology. Favorable sites
nodes.
include the orbit, genitourinary (nonbladder
* Laboratory studies including complete
nonprostate), and nonparameningeal head
blood count, complete metabolic panel with
and neck locations. Unfavorable sites are
renal and liver function studies, coagulation
bladder/prostate, extremity, cranial parame-
testing, and urinalysis.
ningeal, trunk, and retroperitoneum.
* Radiographic studies including mag-
Low risk group: patients with nonmeta-
netic resonance imaging
(MRI) and/or
static favorable site ERMS (stage I, regard-
computed tomography (CT) scan of the
less of degree of initial resection) and
primary lesion, depending on the location.
completely resected (groups I and II) non-
Chest CT and radioisotope bone scan
metastatic unfavorable site ERMS
should also be conducted to evaluate for
Intermediate risk group: patients with
potential lung metastases and bone metas-
any nonmetastatic ARMS and all ERMS
tases, respectively. Site-dependent studies
with unfavorable sites (stages 2 or 3) that
may include: brain MRI for parameningeal
have been incompletely resected (group III)
head and neck tumors, ultrasound and
High risk group: patients with meta-
cystourethroscopy in bladder or prostate
static tumors, both ARMS and ERMS
tumors, and spine MRI in patients with
NRSTS are generally staged using the
evidence of medullary compression. Fluo-
same criteria above. The major determi-
rine-18-fluorodeoxyglucose positron emis-
nants of outcome include tumor grade, size,
sion tomography (FDG-PET) imaging may
extent of initial resection, and presence of
be helpful to determine initial extent of
metastatic disease. Extent of resection is
disease and to monitor response to therapy.
strongly correlated with treatment stratifi-
* Bilateral bone marrow aspirate and
cation and prognosis.
biopsy should be performed, especially in
patients with metastatic disease or alveolar
histologic subtype of RMS.
Pathologic diagnosis and
* Lumbar puncture in patients with para-
treatment
meningeal head and neck primary lesions.
RMS is classified pathologically as embryo-
nal (ERMS) with botryoid and spindle cell
Staging and classification
variants or alveolar (ARMS). Investigation
RMS are stratified into groups based on
for particular translocations (see Genetics
prognostic factors. The principal determi-
section) should be done to assist with defin-
nants of prognosis are primary site, tumor
itive tissue diagnosis in combination with
size, histologic subtype, degree of regional
histochemical studies.
spread and nodal involvement, distant met-
A multidisciplinary approach to the
astatic disease, and extent of prechemother-
treatment of STS is crucial. Basic principles
apy tumor resection. Risk stratification is
of therapy include systemic treatment of
186
Chapter 20
micrometastatic disease with adjuvant che-
Recent trials have not been able to show
motherapy and aggressive local control with
improvement in survival in patients with
definitive surgery and addition of radiation
metastatic rhabdomyosarcoma and new
as necessary.
drug therapies are being investigated. Such
Local control is achieved with either
therapies include irinotecan, temozolomide,
complete surgical resection (with negative
bevacizumab
(anti-vascular endothelial
margins) or surgery and radiation (if micro-
growth factor monoclonal antibody), tem-
scopic or gross residual disease remains after
sirolimus
(mTOR inhibitor), vinorelbine
surgical intervention). Primary re-excision
(vinca alkaloid), and cixutumumab (mono-
may be indicated prior to initiation of che-
clonal antibody to the insulin-like growth
motherapy in certain situations such as: if
factor-1 receptor). High-dose chemother-
the diagnosis was not suspected and only a
apy with hematopoietic stem cell rescue has
biopsy was performed; gross or microscopic
not shown benefit in patients with high risk
residual disease is amenable to wide exci-
and metastatic disease.
sion; or if there is uncertainty about residual
disease or margins. Aggressive surgical
Prognosis
debulking that would result in significant
Overall, survival without relapse is greater
loss of function is not recommended. RMS
than 70% in children and adolescents 5 years
often arise in sites in which complete or
from diagnosis. Low risk groups, represent-
wide resection is not feasible (e.g., orbit,
ing approximately 30% of RMS, can be
most genitourinary and parameningeal
expected to have an excellent outcome with
sites). STS are moderately sensitive to radi-
long-term survival greater than
90%. Of
ation therapy, and this treatment modality
the 55% of patients with intermediate risk
is critical for tumors that cannot be fully
disease, 5-year overall survival is approxi-
excised surgically with negative margins.
mately 55% to 65%. In patients with met-
Radiation is typically delayed until the com-
astatic alveolar disease (15% to 20% of the
pletion of 10 to 20 weeks of systemic che-
population), survival is poor, at less than
motherapy, but may be indicated earlier in
30%. Several caveats are present that help
patients with intracranial primary tumors
determine prognosis:
or in those with compromise of function
* Site of the primary tumor has a major
that may be debilitating or life threatening.
impact on survival and is associated with
Current studies are evaluating the optimal
pathologic subtypes, ease of surgical resect-
timing of irradiation.
ability, timing to presentation, and involve-
Chemotherapy provides the backbone
ment of regional nodes.
of treatment for patients with RMS and
* Extent of disease is the most significant
is initiated following the initial surgical
predictive factor for survival. Children with
procedure. After definitive local control
localized, completely resected disease do
surgery and/or radiation, chemotherapy
better than those with widespread or dis-
continues due to the risk of microscopic
seminated disease.
residual or metastatic disease. The most
* Patients with smaller tumors
(<5 cm)
active drugs in the treatment of RMS are
have improved survival compared to chil-
vincristine, dactinomycin, and cyclo-
dren with tumors >5 cm.
phosphamide
(VAC), doxorubicin, and
* The alveolar subtype of RMS has an
ifosfamide and etoposide
(IE). VAC is
adverse prognosis and is often associated
considered the standard of therapy in all
with an aggressive clinical course and met-
risk groups.
astatic disease at diagnosis and relapse.
Sarcomas of the Soft Tissues and Bone
187
Patients between 1 and 9 years of age
this group of patients. Patients with meta-
have a better prognosis compared to infants
static NRSTS have a dismal long-term prog-
and older children.
nosis and fewer than 20% survive 5 years
from diagnosis. Neoadjuvant chemora-
Children with recurrent RMS have a dismal
diotherapy is being explored in this group
prognosis with long-term survival of less
but there continues to be a paucity of new
than 15%, particularly if the disease recurs
potential therapeutic agents.
in a metastatic site or area of prior irradi-
ation. The majority of relapses occur within
3 years of therapy completion. Metastatic
Bone sarcomas
recurrence is essentially incurable, though
treatment may offer palliation. Treatment
Primary malignant bone tumors in children
with surgical resection and adjuvant mul-
are the sixth most common malignant neo-
tiagent chemotherapy is recommended with
plasm in children and the third most com-
new drug combinations such as ifosfamide/
mon malignancy in adolescents and young
carboplatin/etoposide, docetaxel/gemcita-
adults. They constitute approximately 6% of
bine, and irinotecan in combination with
all childhood malignancies. The two most
temozolomide or vinorelbine. Durable
common bone tumors are osteosarcoma
remissions for several years may be obtained
and Ewing sarcoma. Other malignant bone
with aggressive local retreatment and sys-
tumors include chondrosarcoma and non-
temic therapy. High-dose chemotherapy
Hodgkin lymphoma of bone. Bone lesions
followed by hematopoietic stem cell rescue
may also represent Langerhans cell histio-
has not been shown to be advantageous in
cytosis, benign tumors, or metastatic dis-
this group.
ease. Osteosarcoma is classified into three
major subtypes: osteoblastic, chondroblas-
tic, and fibroblastic, reflecting the predom-
Nonrhabdomyomatous soft
inant type of matrix in the tumor. Histo-
tissue sarcomas
logic variants include telangiectatic, small
cell, periosteal, and parosteal.
Few clinical trials and prospective studies
have been conducted in this population of
Clinical presentation
children. Surgery is the mainstay of effective
Patients typically present with a mass in the
therapy and every effort should be made to
involved area and pain, with symptoms pre-
completely excise the tumor with clear
ceding diagnosis often by several months.
tumor margins. Patients with completely
Patients frequently attribute the pain to a
excised low-grade tumors or high-grade
minor trauma and indeed may present with a
tumors <5 cm have a favorable outcome
pathologic fracture in the affected bone. A
with survival exceeding 85%. Patients with
palpable mass or swelling of the involved site
high-grade NRSTS larger than 5 cm and
typically arises after the onset of pain. Sys-
those with unresectable, localized disease
temic symptoms such as weight loss or short-
have an intermediate prognosis with
ness of breath may be late sequelae and
approximately 50% survival. Patients with
secondary to metastatic disease. Fever is a
high-grade tumors and positive tumor
common symptom of ESFT and may lead to
resection margins typically receive adjuvant
confusion with osteomyelitis and a longer
irradiation. It is not clear if adjuvant che-
period of time from development of symp-
motherapy confers a survival advantage in
toms to diagnosis.
188
Chapter 20
Diagnostic evaluation
and confirm suspicion of a malignant
Osteosarcoma may occur in any bone but
tumor. Osteosarcoma usually produces
primarily occurs in the metaphyses of the
dense sclerosis in the metaphyses of long
most rapidly growing bones. The most com-
bones with soft tissue extension seen in 75%
mon primary sites are the distal femur,
of tumors, radiating calcifications (sunburst
proximal tibia, and proximal humerus with
pattern) in 60% of tumors, osteosclerotic
approximately 50% of tumors originating
lesions in 45% of cases, lytic lesions in 30%
around the knee. Approximately 10% of
of cases, and mixed lesions in 25% of cases.
patients have primary tumors in the axial
A triangular area of periosteal calcification
skeleton including the pelvis, and 15% to
in the border region of the tumor and
20% present with metastatic disease (lung,
healthy tissue is known as a Codman trian-
bone, lymph nodes, and rarely brain).
gle. ESFT are described as lytic with an
Most ESFT occur in bones and their loca-
“onion peel” periosteal reaction and typi-
tions tend to differ from that of osteosarcoma.
cally occur in the diaphysis. Patients with
Flat bones of the axial skeleton are more
soft tissue ESFT only may have normal
commonly affected; in long bones, the diaphy-
radiographs. Additional assessment of the
seal portion is usually involved. The most
primary tumor site with MRI should be
common primary locations include the pelvic
undertaken to view the soft tissue compo-
bones, the long bones of the lower extremities,
nent, surrounding structures, vessels and
and the bones of the chest wall. Metastatic
nerves, and the intramedullary extension
disease is present in 25% of patients at diag-
to assist with surgical planning. MRI should
nosis and is primarily located in the lungs,
include the entire involved bone and neigh-
bones, or bone marrow. Site of primary disease
boring joints to evaluate for skip lesions.
is related to the incidence of metastases at
Metastatic disease is typically in the lungs or
diagnosis; central primaries are associated
bone and therefore imaging with chest CT
more frequently with distant disease (40%),
and technetium-99m bone scan is essential
whereas distal primary lesions have the lowest
in the initial workup for staging. FDG-PET
incidence (15%).
imaging is a sensitive screening tool for the
The requisite elements of evaluation
detection of bone metastases in ESFT but its
include:
role in evaluation of osteosarcoma has yet to
* A complete history and physical
be determined.
examination.
* Bilateral bone marrow aspirate and biopsy
* Routine laboratory studies include a com-
should be performed in patients with ESFT.
plete blood count, complete metabolic panel
* Baseline audiogram and echocardiogram
with renal and liver function tests, serum
should be obtained prior to initiation of
creatinine, creatinine clearance, and urinal-
ototoxic and cardiotoxic chemotherapy,
ysis. A measured or calculated glomerular
respectively.
filtration rate should be obtained prior to
initiation of nephrotoxic chemotherapy. The
Pathologic diagnosis
alkaline phosphatase may be elevated and has
Patients presenting with a bone mass sus-
been associated with an inferior outcome in
picious for malignancy should undergo a
osteosarcoma. The serum lactate dehydroge-
diagnostic incisional or core biopsy. It is
nase may also be elevated and may correlate
strongly recommended that an experienced
with tumor burden.
oncologic orthopedic surgeon who will also
* Imaging studies should begin with plain
be performing the definitive procedure per-
radiographs to visualize osseous changes
form the biopsy so that the biopsy tract can
Sarcomas of the Soft Tissues and Bone
189
be excised en bloc with the planned surgical
techniques in younger children. Tumors of
resection. Sufficient tissue should be
the pelvis or axial skeleton present a difficult
obtained for diagnostic histopathology as
situation, as resection with adequate mar-
well as for any submissions required for
gins may not be possible. Outcome is related
participation in a clinical trial. Biopsy may
to degree of resection and these patients as a
be taken from an extraosseous component,
group have inferior outcomes compared to
if present, to prevent pathologic fracture.
patients with extremity tumors.
As the histologic and immunophenoty-
Radiation therapy has limited utility in
pic features of ESFT overlap to some degree
patients with osteosarcoma, but is indicated
with many other small, round blue cell
for those unable to undergo complete resec-
tumors of childhood, an expanded immu-
tion. The most active chemotherapeutic
nohistochemical panel should include
agents are cisplatin, doxorubicin, high-dose
assessment for CD99 (a cell surface glyco-
methotrexate, ifosfamide, and etoposide.
protein with strong expression in ESFT) as
Patients can expect to be treated for approx-
well as markers for neuroblastoma (neuron-
imately
1 year, with a definitive surgical
specific enolase and S100), synovial sarcoma
procedure performed at approximately
(Leu-7), and rhabdomyosarcoma (vimen-
week 12, followed by continuation of mul-
tin, desmin, and myogenin). Molecular
tiagent chemotherapy.
genetic studies using fluorescent in situ
Patients presenting with metastatic dis-
hybridization or reverse transcriptase-
ease undergo the same therapeutic approach
polymerase chain reaction are valuable in
with systemic chemotherapy and definitive
diagnosis, specifically for the detection of
surgery of the primary site in addition to
characteristic translocations that allow for
surgery of metastatic sites, as feasible.
definitive diagnosis of ESFT, RMS, and
Approximately one-third of osteosarcoma
synovial cell sarcoma.
patients with pulmonary metastases will
have additional microscopic pulmonary
Treatment
nodules not visible on the current, highest
Systemic multiagent chemotherapy prior to
resolution CT imaging. It is recommended
and following definitive radical surgery is
that pulmonary nodules identified on CT
the standard of care for treatment of oste-
imaging be surgically resected by open tho-
osarcoma. Complete resection of all disease
racotomy that will allow intraoperative lung
sites including metastatic lesions is critical
palpation to potentially identify additional
to long-term survival. An assessment of
sites of metastatic disease. Patients with
histologic response to initial induction che-
unilateral lung metastases on imaging
motherapy at the time of definitive surgery
should undergo bilateral thoracotomies for
may guide subsequent treatment and helps
this reason.
define prognosis. The orthopedic oncologic
As with osteosarcoma, cure in ESFT
surgeon determines the type of surgical
can be achieved only with a multimodal
procedure performed and factors used in
approach, using surgery and/or radiation
this determination include the location and
therapy for local control of the primary
size of the tumor, patient age and skeletal
lesion and chemotherapy for eradication
maturity, presence of metastatic disease,
of subclinical micrometastases. Definitive
and patient lifestyle choices. Limb sparing
surgery follows a period of induction
procedures are feasible in the majority of
chemotherapy and many patients are
patients and the development of expandable
candidates for complete surgical resection
endoprostheses has allowed the use of these
with limb salvage procedures. Induction
190
Chapter 20
chemotherapy may also render initially
estimated at 2 years as 10% to 30%. The
unresectable tumors resectable. ESFT are
degree of tumor necrosis following neoad-
radiation responsive, although typically this
juvant
(presurgery) chemotherapy is an
therapeutic modality is reserved for surgi-
independent predictor of event-free and
cally unresectable tumors or in tumors with
overall survival, presumably reflecting
positive margins after resection. Patients
tumor sensitivity to chemotherapy. Specif-
who receive radiation therapy as the only
ically, osteosarcoma patients with a good
local control modality have an inferior
response, defined as greater than
95%
outcome. Frequently, these patients have
tumor necrosis at the time of surgery, have
other adverse features including large tumor
a superior survival. A similar cutoff is yet to
size, unfavorable locations (e.g., vertebral
be defined for ESFT. To date, intensified
tumors), or both. Radiation is also utilized
treatment for those with less favorable
in the treatment of metastatic disease.
tumor necrosis has not improved outcome.
Debulking procedures have not been shown
The prognosis for patients with recur-
to improve outcome and should not be part
rence is quite poor. Most local recurrence is
of local control therapy.
associated with concomitant distant disease.
Active chemotherapy agents in ESFT
Time to relapse and tumor burden correlate
include vincristine, dactinomycin, cyclo-
with postrelapse outcome. Late recurrence
phosphamide, doxorubicin, ifosfamide, and
(e.g., >5 to 10 years) has been reported in
etoposide (similar to RMS). As with osteo-
bone sarcoma. Surgical resection remains
sarcoma, the length of systemic therapy is
the mainstay of therapy for curative second-
approximately one year. The current stan-
line therapy. Patients with osteosarcoma
dard of care for localized pediatric ESFT is
who develop pulmonary lesions after com-
alternating cycles of vincristine, doxorubi-
pletion of therapy can be cured with surgery
cin, and cyclophosphamide
(VDC) with
alone. The role of adjuvant chemotherapy in
ifosfamide and etoposide (IE) given in a
relapse is controversial, though patients are
compressed, every 2-week schedule. This
often offered such treatments including
increased time-dose intensity schedule has
cyclophosphamide/topotecan, irinotecan/
shown benefit in pediatric patients though
temozolomide, and gemcitabine/taxotere.
not for adults. Although still being investi-
Radiation has a role in the treatment of
gated, high-dose chemotherapy with autol-
patients with ESFT who develop new met-
ogous stem cell rescue has not as yet shown
astatic sites and for palliation and pain
benefit in the treatment of bone sarcoma.
control in either osteosarcoma or ESFT.
Targeted and biologically based thera-
Prognosis
pies are being investigated in patients with
Most patients with bone sarcomas have
poor response to initial chemotherapy,
micrometastatic disease at diagnosis since,
those with metastatic disease, and those
in the past, 80% to 90% developed lung
with recurrent disease. Novel agents with
metastases following local control therapy
potential therapeutic benefit include mono-
(i.e., surgery and/or radiation) alone. There-
clonal antibodies to insulin growth factor-1
fore, prior to the use of systemic chemo-
receptor, tyrosine kinase inhibitors, mTOR
therapy, survival rates at 2 years were 15%
inhibitors, and monoclonal antibodies to
to 20%. Currently, patients with localized
vascular endothelial growth factor. Specific
disease can expect a long-term survival of
novel therapies for osteosarcoma include
greater than
70%. Survival in patients
bisphosphonates (e.g., zoledronic acid) and
with metastatic disease remains dismal,
RANK inhibitors
(e.g., denosumab); for
Sarcomas of the Soft Tissues and Bone
191
ESFT, transcription inhibition of the EWS-
Case study for review
FLI1 translocation is being studied.
A 9-year-old boy presents with a nontender
Late effects
3
4 cm mass in the right inguinal region.
Site and extent of disease, as well as thera-
He denies fever, weight loss, or pain else-
peutic interventions with chemotherapy,
where. The mass has been present and
radiation, and surgery all contribute to late
growing for several weeks.
sequelae in sarcoma patients. A majority of
first-line protocols utilize high cumulative
1. What key elements should you obtain in
doses of anthracyclines (i.e., doxorubicin)
the history?
resulting in a lifelong risk of developing
2. What are you looking for on the physical
cardiomyopathy. Thus, lifelong cardiac
examination?
monitoring with serial echocardiograms
3. What is your differential diagnosis for
and periodic functional assessments is
this boy?
recommended. Alkylator therapy (e.g., ifos-
4. How do you approach a diagnostic eval-
famide) can lead to gonadal toxicity (pri-
uation and staging?
marily in males) and increased risk of
5. What do you tell the child and his family?
secondary malignancy. Second malignant
6. Assuming you find out this is a localized
neoplasms occur in approximately 3% of
undifferentiated sarcoma, what is the child’s
patients within 10 years of therapy comple-
prognosis and what is the general treatment
tion, likely related to both a genetic predis-
plan?
position and treatment with mutagenic
therapies. Platinum therapy (e.g., cisplatin)
Key elements in the history include length of
can lead to long-term oto- and nephrotox-
symptoms, possible relationship to infec-
icity. High-frequency hearing loss is com-
tion (could this be a lymph node?), animal
mon and some patients will require hearing
exposures and bites, travel, cuts or injuries
aids. Renal dysfunction is rare but could
to the affected leg, systemic symptoms such
result in chronic electrolyte wasting due to
as malaise, fevers, unexplained weight loss,
proximal tubular damage (see Chapter 30
or discomfort that limits activities or neces-
for more details).
sitates medication.
For osteosarcoma patients, contempo-
The physical examination should be
rary orthopedic surgery techniques are
complete, head to toe. The mass should
aimed at preservation of function and
be measured and characterized for mobility,
improved limb salvage without compromis-
tenderness, warmth, and erythema. The
ing survival. Amputation may result in
mass may be hard and nontender if malig-
superior limb function in some cases. In a
nant disease is present. If the mass is sus-
skeletally immature child, expandable pros-
pected to be a node, it may be enlarged as a
theses may require subsequent revisions
result of disease spread and therefore the
though some may benefit from newer
distal extremity must be carefully examined.
self-expanding prostheses. Late prosthetic
All other lymph node chains as well as the
failures or infection can occur, requiring
liver and spleen should also be carefully
surgical revision or delayed amputation.
assessed.
Radiation therapy can lead to local effects
The differential diagnosis of a mass in the
including growth impairment, muscle
inguinal area consists of lymphadenopathy
fibrosis, and increased risk of secondary
secondary to an infectious or malignant
malignancy, in a dose-dependent manner.
etiology. Infections may be bacterial,
192
Chapter 20
protozoal, fungal, or viral and may be the
aggressive local control (complete surgical
result of introduction by a cut (which may
excision and focal radiation) can expect to
have since healed), animal bite, or scratch.
have approximately a 70% event-free sur-
A mass that is hard, nontender, and non-
vival. This is in stark contrast to patients
mobile, without signs and symptoms of
with metastatic disease in which case sur-
infection, is concerning for malignancy.
vival is less than
20% despite aggressive
Malignancies that can lead to regional
therapy. Those patients with regional
spread in an extremity include sarcomas
spread, similar to this case, have an inter-
(bone and soft tissue).
mediate prognosis. Such cases would be
Assuming you could find no lump or
treated with aggressive multiagent chemo-
tender area on the extremity exam, it is
therapy in addition to aggressive local con-
prudent to begin a detailed investigation
trol. Future therapies may include novel
with imaging and biopsy. Imaging should
biologic or targeted agents. In this child,
consist of MRI of the inguinal area and leg
local control would involve treatment of the
(to the tips of the toes), bone scan, and plain
toe and the inguinal mass. One should also
films. An excisional biopsy should be per-
consider treatment (i.e., radiation therapy)
formed as well given the concern for malig-
of the lymph nodes in between these regions
nancy
(and to prevent potential tumor
in addition to nearby pelvic nodes.
tracking which can occur with an incisional
biopsy). Once the mass is a biopsy-proven
Suggested Reading
malignancy, further staging should evaluate
for distant disease and include CT of the
Caudill JS, Arndt CA. Diagnosis and manage-
chest, abdomen, and pelvis.
ment of bone malignancy in adolescence.
Prior to initiation of the biopsy and
Adolesc Med State Art Rev 18:62-78, 2007.
imaging, it is best to be straightforward with
Hayes-Jordan A, Andrassy R. Rhabdomyosar-
the family and let them know your concern
coma in children. Curr Opin Pediatr
that this mass likely represents a malignancy.
21:373-378, 2009.
As the mass is in the inguinal area, it could
Loeb DM, Thornton K, Shokek O. Pediatric soft
represent regional spread of a distant tumor.
tissue sarcomas. Surg Clin North Am
In this case, the child had a nonpalpable
88:615-627, 2008.
mass in the fifth distal toe found on MRI.
McCarville MB. The child with bone pain: malig-
nancies and mimickers. Cancer Imaging 9:
The pathologic diagnosis of undifferenti-
S115-S121, 2009.
ated sarcoma was made on excisional biopsy
Spunt SL, Skapek SX, Coffin CM. Pediatric non-
of the inguinal mass. No other evidence of
rhabdomyosarcoma soft tissue sarcomas.
distant tumor was found on further imag-
Oncologist 13:668-678, 2008.
ing. Prognosis for undifferentiated sarcoma
Subbiah V, Anderson P, Lazar AJ, et al. Ewing’s
is generally similar to alveolar rhabdomyo-
sarcoma: standard and experimental treat-
sarcoma, and patients with localized disease
ment options. Curr Treat Options Oncol
who receive systemic chemotherapy and
10:126-140, 2009.
Germ Cell Tumors
21
Germ cell tumors (GCT) arise from primor-
more frequent in males. Testicular GCT
dial cells involved in gametogenesis and
have several known risk factors including
occur primarily in the testes and ovaries.
infertility/testicular atrophy as well as crypt-
These tumors represent approximately 4%
orchidism. Males with cryptorchidism have
of pediatric cancers. Primordial germ cells
a higher incidence of testicular cancer in
(PGCs) are thought to originate in the yolk
both the undescended testis and the nor-
sac endoderm and migrate along the genital
mally descended contralateral testis despite
ridge and thus may also present in midline
surgical correction, hormonal therapy, or
extragonadal sites such as the sacrococcygeal
spontaneous descent. However, early cor-
region, midline of the brain, mediastinum,
rection may partially ameliorate this risk.
and retroperitoneum. Due to the pluripoten-
The incidence of testicular GCT in the
tiality of PGCs, GCT are an array of different
Caucasian population is increasing at a rate
histologic subtypes, adding to the complexity
of 3% to 6% per year. Perinatal and envi-
of the disease. In addition, GCT can occur
ronmental risk factors are thought to play a
due to tumorigenesis or as part of normal
role although these are yet to be clearly
embryonal development, in part explaining
elucidated. Young parental age at birth, low
the pediatric bimodal age of distribution (2
birth order, low birth weight, and breech
and 20 years). In addition, age of presenta-
birth have all been noted to be statistically
tion and histologic subtype varies signifi-
significant risk factors in multiple studies.
cantly between males and females. This is
Maternal exposure to pesticides or hor-
thought to be due in part to the differential
mones, parental smoking, and alcohol con-
timing of male and female development;
sumption may also be factors. Genetic risk
female germ cells enter meiosis at 11 to 12
factors also play a role, specifically in tes-
weeks of gestation while male germ cells
ticular GCT, as a family history of cancer,
begin meiosis with the onset of puberty.
especially at a young age, has been associ-
ated with increased risk in pediatric
patients. Additionally, there is a higher inci-
Epidemiology
dence in monozygotic twins and some
familial clusters have been reported. A num-
Due to the differential timing of germ cell
ber of congenital genitourinary anomalies
maturation, GCT are more common in
such as retrocaval ureter, bladder divertic-
females until adolescence and then become
ulum, and inguinal hernia have been
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
194
Chapter 21
associated with increased risk, emphasizing
develop malignant components (i.e., squa-
that abnormalities in development likely
mous cell carcinoma) and therefore are
play a role. Similarly, disorders of sexual
treated with complete surgical resection.
development, especially in cases with excess
Immature teratomas are tumors of inter-
Y chromosome material (e.g., gonadal dys-
mediate malignant potential with less dif-
genesis), pose an increased risk in both
ferentiated tissues.
males and females. Children with Down
Germinomas may be of pure histology or
syndrome, who generally have a decreased
mixed. Syncytiotrophoblastic cells occur in
risk of solid tumors, are at a predisposition
about 5% of germinomas and secrete the
to develop testicular GCT, whereas patients
beta subunit of human chorionic gonado-
with Klinefelter syndrome have an increased
tropin (b-HCG). Embryonal carcinoma is
risk of mediastinal GCT. Gain of chromo-
most often found as a component of testic-
some 12p is also seen in the majority of
ular mixed GCT and is thought to be the
testicular and malignant ovarian GCT.
driver for the development of other histo-
Approximately half of all pediatric GCT are
logic subtypes including teratoma, yolk sac
extragonadal in origin, with about 15% of
tumor, and choriocarcinoma. Yolk sac
these being malignant.
tumor is generally found as a part of testicular
mixed GCT but can also be seen as a pure
histologic variant in young children. Yolk sac
Pathology and serum tumor
tumor secretes a-fetoprotein (AFP).
markers
Choriocarcinoma contains syncytiotro-
phoblastic cells and therefore also secretes
The main histologic variants of GCT include
b-HCG. Ovarian cases should be distin-
germinomas (dysgerminoma in the ovary
guished from metastatic gestational
and seminoma in the testis), embryonal
choriocarcinoma.
carcinoma, yolk sac tumor
(endodermal
In the ovary and testis, GCT should be
sinus tumor), choriocarcinoma, and tera-
distinguished from sex cord stromal
toma (mature and immature). Mixed GCT
tumors, which arise from the nongermina-
are composed of two or more histologies.
tive components of gonadal tissue and
Teratomas contain tissues from all three
include tumors involving the Sertoli-Leydig
germ layers
(ectoderm, endoderm, and
cells of the testis and granulosa cells of the
mesoderm). Mature teratomas contain no
ovary. Sarcoma as well as gonadal infiltra-
malignant germ cell elements and are most
tion by leukemia and non-Hodgkin lym-
commonly located in the ovary or sacro-
phoma should also be considered in the
coccygeal region. The vast majority of
differential diagnosis of gonadal tumors.
female GCT are teratomas. Teratomas in
Due to the specific histologies that
the prepubertal male are benign, whereas
secrete AFP and b-HCG, these tumor mar-
those occurring during or after the onset of
kers can be quite helpful in distinguishing
puberty are always malignant, as they arise
tumor type as well as following disease.
from other forms of testicular germ cell
Mature teratomas secrete neither marker;
tumor (specifically embryonal carcinoma).
therefore, if a serum marker is present in the
Therefore, teratomatous elements in the
patient diagnosed with a mature teratoma,
testis largely occur as a component of a
one must consider that the tumor contains
mixed GCT. In the female or prepubertal
one or more foci of yolk sac tumor if the
male, these tumors are thought to derive
AFP is positive, choriocarcinoma or germi-
from a benign germ cell but may rarely
noma if the b-HCG is positive, and that the
Germ Cell Tumors
195
patient in fact has a mixed malignant germ
pain due to testicular torsion. The differen-
cell tumor. Due to physiologic changes in the
tial diagnosis of a testicular mass includes
secretion of fetal AFP, this marker can be
hydrocele, hematocele, varicocele, inguinal
difficult to utilize in young infants. AFP is
hernia, and torsion of a normal testis. Girls
made in the fetal liver and does not decline to
most frequently present with abdominal
adult levels until approximately
6
to
8
pain, distention, and weight gain. Torsion
months of age. Age appropriate levels can
of an involved ovary, hemorrhage, or tumor
be obtained from published tables. AFP can
rupture may occur and present as an acute
also be secreted by regenerating liver post-
surgical abdomen. Secretion of b-HCG may
injury or by certain liver tumors such as
lead to isosexual precocity. Occasionally, the
hepatoblastoma. Degree of AFP elevation
increasing abdominal girth and elevated
may be related to tumor volume. b-HCG
urine and serum b-HCG have been con-
is elevated in pregnancy and can also be seen
fused with pregnancy.
in association with childhood hepatic tumors
The most common site of extragonadal
and other cancers involving the breast, stom-
tumors is the sacrococcygeal region.
ach, and pancreas in adults. Nonspecific
Although usually benign, patients can have
markers of GCT include lactate dehydroge-
malignant tumors of higher stage. Older age
nase isoenzyme 1, neuron-specific enolase,
at diagnosis is associated with a greater risk
and placental alkaline phosphatase.
of malignancy. Sacrococcygeal tumors are
classified as four anatomic variants: type I
(predominantly outside the body), type II
Clinical presentation
(equally inside and outside the body),
type III
(largely intra-abdominal), and
GCT should be considered in children who
type IV (completely within the body). More
present with a pelvic, ovarian, or testicular
than 90% of tumors present with some
mass, or a midline mass in the sacrococcy-
component of an externally visible mass.
geal region, retroperitoneum, mediastinum,
These tumors are often detected in utero
or pineal or suprasellar regions of the brain.
with prenatal ultrasonography. They may
GCT present with signs and symptoms
also present later in children with constipa-
related to the site and size of the primary
tion, urinary obstruction, urgency or fre-
tumor. Compression of internal structures
quency, lower back or pelvic pain, lower
can lead to pain, constipation, and urinary
extremity weakness, or sensory changes, all
obstruction. Large ovarian tumors can addi-
signs and symptoms due to compression of
tionally lead to decreased lung volumes and
normal structures by tumor.
respiratory distress. GCT can metastasize,
Approximately
20% of children with
most commonly to the lungs, but patients
GCT will present with metastatic disease,
rarely present with clinical symptoms refer-
most commonly to the lung, liver, or lymph
able to these metastases. Occasionally,
nodes, and rarely to the central nervous
patients may present with hormonal man-
system, bone, or bone marrow. Regional
ifestations secondary to secretion of
spread resulting in surgically unresectable
b-HCG. Specifically, peripheral conversion
tumors occurs in approximately 25% of
of androgen to estrogen can lead to gyneco-
patients at presentation. Tumor dissemina-
mastia or uterine bleeding.
tion occurs by local extension, intracavitary
Boys with gonadal GCT typically present
seeding, or hematogenous spread. Intraca-
with a slowly enlarging testicular mass,
vitary seeding may involve the omentum,
occasionally complicated by acute severe
bowel, spleen, diaphragm, or pelvic organs.
196
Chapter 21
Rarely, bone involvement can occur by
sperm banking should be discussed with the
direct extension.
patient and family prior to radical inguinal
orchiectomy.
Diagnostic evaluation and risk
Treatment and prognosis
stratification
Staging is done postoperatively and is based
The following studies should be performed
on pathologic degree of tumor invasion,
in a child with a suspected or confirmed
presence of lymph node involvement, con-
GCT:
tinued positivity of tumor markers, positive
* Complete history
peritoneal washings (in ovarian tumors),
* Physical examination
and presence of residual or metastatic dis-
* Baseline laboratory studies consisting of a
ease. Factors known to affect prognosis
complete blood count, transaminases, blood
include extreme tumor marker elevation
urea nitrogen, creatinine, and electrolytes
and failure of these markers to appropriately
* Baseline tumor markers including AFP,
decline following tumor resection, based on
b-HCG, and lactate dehydrogenase (LDH)
the appropriate half-life for each marker. Site
* Imaging studies to evaluate the extent of
of tumor is prognostic, with mediastinal
the mass and potential metastatic sites:
tumors fairing more poorly. Histology is also
ultrasound, CT, or MRI of the primary mass
important as pure germinomas are generally
(testicular ultrasound in males, abdominal
very chemotherapy sensitive and have a high
and pelvic ultrasound, CT, or MRI in
response rate. Patients with higher stage,
females); abdominal and pelvic MRI or
metastatic disease also fair more poorly.
CT as well as baseline chest radiography
In the patient with suspected GCT,
with or without CT of the chest to evaluate
every effort should be made to perform a
for metastatic disease (all CTs should be
complete resection with negative margins.
done with contrast)
In situations where the patient presents
emergently with torsion, there may not
Determination of the extent of disease at
be time to wait for a definitive diagnosis
diagnosis allows proper decision making
prior to surgery; every effort should be
regarding the safest and most reasonable
made in these cases to perform a gross
place to biopsy. Localized tumors of the
total resection (GTR), assuming there is
ovary or testis should be completely
low operative risk, after sending the appro-
removed. Resection of ovarian tumors
priate tumor markers. In the case of a large,
should include abdominal exploration and
invasive tumor, biopsy should be done
peritoneal washings for cytologic assess-
followed by administration of chemother-
ment. Risks and benefits of lymph node
apy and delayed surgical resection. Due to
sampling and biopsy of the contralateral
the location of some extragonadal tumors,
testis or ovary should be discussed with the
especially those in the mediastinum and
surgeon, patient, and family.
retroperitoneum, neoadjuvant chemother-
GCT are known to grow rapidly and
apy is often required prior to surgery in
metastasize frequently. Diagnostic studies
order to help facilitate the chance of a
should be done expeditiously to allow ini-
safe GTR.
tiation of therapy. Due to the potential
Depending on tumor location and his-
underlying risk of contralateral testicular
tology, some GCT can be observed after
atrophy, infertility, or carcinoma in situ,
GTR without additional therapy. Young
Germ Cell Tumors
197
patients with extragonadal tumors and a
retroperitoneal lymph node dissection may
GTR can generally be observed after surgery,
also be recommended in patients with exten-
as these tumors rarely have malignant ele-
sive disease at presentation. Patients with a
ments. Sacrococcygeal tumors must be
poor response to therapy or recurrent disease
removed with the coccyx in order to
may benefit from second-line therapies such
decrease the risk of local recurrence. Young
as paclitaxel
(Taxol), ifosfamide, cisplatin
patients are more likely to present with pure
(TIP); vinblastine, ifosfamide, cisplatin
yolk sac tumors, and these patients can also
(VeIP); or gemcitabine/oxaliplatin. GCT are
be observed after GTR. Gonadal tumors that
also quite radiosensitive, although the high
are histologically consistent with mature
cure rates seen with chemotherapy alone
teratoma similarly require only observation
have obviated the need for radiation in most
after GTR. Localized testicular seminoma
cases. Radiation may be considered in refrac-
can be observed, although patients with
tory or recurrent disease, or for palliation.
continued elevation of tumor markers
Additionally, high-dose chemotherapy with
should undergo adjuvant radiotherapy.
autologous stem cell rescue may be benefi-
Patients with this pure histology are very
cial in a subset of refractory patients.
chemotherapy sensitive and easily salvaged
Patients treated with high-dose platinum
in the event of relapse. In general, patients
agents may develop high-frequency hearing
with localized testicular mixed GCT can also
loss and require hearing assistive devices.
be observed assuming that tumor markers
Additionally, these drugs may lead to tran-
normalize after surgery, again due to the
sient proximal tubule renal dysfunction and
high rate of salvage with potential relapse.
resultant electrolyte wasting. Patients who
Due to this risk of relapse, which is directly
receive alkylator therapy (e.g., ifosfamide)
related to the amount of vascular tumor
are at risk for secondary malignancy and
invasion, size of the primary tumor, and
infertility (especially in males). Topoisom-
percent embryonal carcinoma, patients
erase II inhibitors (etoposide) also increase
should be presented the options of obser-
risk for secondary neoplasms. Bleomycin
vation versus chemotherapy. Due to the
may lead to pulmonary fibrosis, although
rarity of malignant ovarian GCT, recom-
the risk is poorly quantified.
mendations for chemotherapy versus obser-
vation generally follow those presented for
testicular tumors.
Germ cell tumors of the central
Higher stage, nonlocalized GCT are trea-
nervous system
ted with chemotherapy and are generally
very chemotherapy sensitive. Patients are
Intracranial GCT comprise approximately
treated with bleomycin, etoposide, and
3% of primary childhood brain tumors with
cisplatin (BEP) regimens for 3 to 4 cycles.
a peak incidence between the second and
These tumors exhibit a steep dose-response
third decade of life. The majority of these
curve to platinum compounds, though the
tumors are germinomas, which account for
risk of toxicity (oto- and nephrotoxicity)
50% to 70% of cases. Nongerminomatous
must be considered. Due to the potential
germ cell tumors (NGGCT) account for the
risk of long-term pulmonary fibrosis from
remaining third and consist of multiple
bleomycin, etoposide/cisplatin
(EP) and
histologies including endodermal sinus
etoposide (VP-16), ifosfamide, and cisplatin
(yolk sac) tumors, choriocarcinoma, mature
(VIP) regimens have been developed as
and immature teratoma, and embryonal
alternative therapies to BEP. Bilateral
carcinoma. Most NGGCT are of mixed
198
Chapter 21
histology and may contain germinomatous
surgical GTR, as safely feasible. In other
elements but will not have this as a pure
tumor types, the value of a surgical resection
histology. Mature and immature teratomas
has not been clearly delineated and is
predominate in the neonatal period. Patients
recommended only in specific situations
typically present with symptoms depending
(i.e., persistence of tumor following induc-
on the location and size of the tumor,
tion chemotherapy). Pure germinomatous
further influenced by the extent of pituitary
tumors are highly sensitive to both radio-
dysfunction and the presence or absence of
and chemotherapy. In the past, radiother-
hydrocephalus. Midline pineal tumors are
apy was the modality of choice for these
often associated with symptoms related to
tumors due to the high cure rate
increased intracranial pressure due to
(5-year survival >80%). Due to the recog-
obstruction of the cerebral aqueduct and
nized long-term risks from this therapy
Parinaud’s syndrome (paralysis of upward
(neuropsychological and endocrine), adju-
gaze) due to involvement of the tectal plate.
vant chemotherapy is now being used to
Tumors in the suprasellar area often lead to
allow a reduction in the dose and extent of
endocrinopathies such as diabetes insipidus
radiotherapy without increasing relapse
(DI) and visual field defects. Occasionally
rate. Chemotherapeutic agents are similar
DI is related to occult involvement of the
to those used for systemic GCT and include
infundibulum. In children with clinical
platinum agents (cisplatin and carboplatin),
symptoms of DI and consistent MRI find-
alkylators
(cyclophosphamide and ifosfa-
ings (i.e., absence of a posterior pituitary
mide), and topoisomerase II inhibitors (eto-
bright spot, possibly in association with
poside). Of note, patients with DI should be
thickening of the pituitary stalk), the differ-
carefully monitored in an ICU setting while
ential diagnosis should include GCT and
receiving alkylators or platinum therapy due
Langerhans cell histiocytosis. The presence
to the increased risk of nephrotoxicity.
or absence of CSF tumor markers may assist
Management during this therapy can be
with this diagnostic dilemma.
simplified by the use of IV vasopressin
Children with midline brain tumors
administered as a continuous infusion.
should be assessed for a GCT. CSF should
Patients with NGGCT have had a poorer
be obtained for routine studies (glucose,
outcome in the past as these tumors are
protein, chamber count, and cytology) in
generally less radiosensitive than pure
addition to assessment of tumor markers
germinoma. Outcome is related to tumor
(AFP and b-HCG). Patients with classic
histology as those patients with pure embry-
findings on MRI and elevated AFP with
onal carcinoma, endodermal sinus tumor,
or without a significantly elevated b-HCG
and choriocarcinoma fair much more
do not require a biopsy for diagnosis as they
poorly than those with mixed histology,
have chemical evidence of NGGCT. Patients
especially those with a high proportion of
with negative markers or a modest elevation
teratoma or germinoma. Neoadjuvant plat-
in b-HCG only should have a biopsy per-
inum-based chemotherapeutic regimens
formed. Central nervous system GCT often
have shown benefit in patients with NGGCT
spread along the CSF pathways; thus, MRI
prior to radiation therapy. Second-look sur-
of the spine should be obtained in addition
gery is beneficial in those patients with an
to head imaging for diagnostic and staging
incomplete radiologic response or persis-
purposes.
tently elevated tumor markers following
Patients with benign tumors such as
induction chemotherapy, prior to radiation
mature teratoma should have a curative
therapy. High-dose chemotherapy with
Germ Cell Tumors
199
autologous stem cell rescue has been shown
though may benefit from additional chemo-
to be of benefit in patients with residual
therapy, surgery, radiation, and/or high-
malignant disease after chemotherapy and
dose chemotherapy with stem cell rescue.
surgery.
Patients with recurrent disease may be
successfully treated, depending on initial
Suggested Reading
therapy. Patients with pure germinoma
treated solely with chemotherapy may ben-
Echevarria ME, Fangusaro J, Goldman S. Pedi-
efit from additional chemotherapy followed
atric central nervous system germ cell tumors:
by radiation therapy. Those that have pre-
a review. Oncologist; 13:690-699, 2008.
viously received radiation may benefit from
Horton Z, Schlatter M, Schultz S. Pediatric germ
high-dose chemotherapy with autologous
cell tumors. Surg Oncol 16:205-213, 2007.
stem cell rescue. Patients with recurrent
Horwich A, Shipley J, Huddart R. Testicular
NGGCT have a much more dismal outcome
germ-cell cancer. Lancet 367:754-765, 2006.
Rare Tumors of
22
Childhood
The rarer tumors of childhood collectively
utmost importance in affected families due
comprise fewer than 20% of all pediatric
to the risk of tumors in siblings as well as
cancers. The more frequently seen of these
offspring of survivors, especially in those with
include retinoblastoma (2% to 3%), liver
bilateral retinoblastoma.
tumors (1%), and epithelial tumors of the
The RB1 gene is a tumor suppressor gene
adrenal or thyroid gland (2% to 3%), which
and individuals with loss of function of both
are discussed herein. Many other rare pedi-
allelic copies are predisposed to malignancy.
atric malignancies also exist that are beyond
Germline de novo mutations of the RB1 gene
the scope of this chapter.
largely occur during spermatogenesis. Sec-
ondary mutations may then be germline or
somatic depending on the timing of events.
Retinoblastoma
Those mutations that are germline are her-
itable for future generations.
Retinoblastoma is the most common
Retinoblastoma arises from the photo-
malignant ocular tumor in childhood,
receptor cells of the innermost layer of the
affecting 200 to 300 children per year in
retina. The tumor frequently extends into
the United States. Retinoblastoma is rarely
the vitreous cavity, presenting with a fleshy
diagnosed after 5 years of age. This unique
nodular mass visible on ophthalmologic
neoplasm has a strong genetic component
examination. Large tumors may occupy
related to mutation in the RB1 gene located
most of the posterior chamber. Less fre-
on chromosome 13. It was the model for the
quently, tumors may extend externally
development of Knudson’s two-hit hypoth-
resulting in retinal detachment. The tumor
esis after observation that children with
tends to outgrow its blood supply and may
bilateral retinoblastoma developed disease
develop areas of necrosis and calcification.
at an earlier age compared to those with
Due to the large mass or retinal detach-
unilateral disease. The genetic form of the
ment, the normal papillary red reflex is
disease is inherited in an autosomal domi-
replaced by leukocoria (white papillary dis-
nant manner yet only 15% to 25% of children
coloration) and is often first noticed by the
have a family history, suggesting the acqui-
family. Strabismus is the second most com-
sition of a new germ line or somatic mutation
mon presenting sign and is due to visual
in the majority. Genetic counseling is of
impairment from the tumor. Other less
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Rare Tumors of Childhood
201
common presenting signs include hetero-
to radiation that increases the likelihood of
chromia, inflammatory changes, hyphema,
secondary malignancy. MRI is most useful
and glaucoma. Familial cases are often
for identification of extraocular extension
detected early when the family is appropri-
and trilateral retinoblastoma. MRI of the
ately advised on the need for frequent and
spine and lumbar puncture for cerebrospi-
careful screening with ophthalmologic
nal fluid cytology should be done in patients
examinations under anesthesia. The differ-
with intracranial spread, involvement of the
ential diagnosis of leukocoria includes a
optic nerve, or evidence of tumor invasion
number of rare nonmalignant conditions
beyond the lamina cribrosa on pathology
such as Coats disease, congenital cataracts,
after eye enucleation. In patients with delay
toxocariasis, retinopathy of prematurity,
in diagnosis and concern for metastatic
and persistent hyperplastic primary vitre-
disease, bone marrow studies and bone scan
ous. Careful retinal examination by an
should be performed.
ocular oncologist can rule out these other
Treatment for retinoblastoma is individ-
diseases and obviate the need for tissue
ualized and dependent on multiple factors
biopsy. Patients with advanced disease may
including unilaterality or bilaterality of dis-
present with proptosis and swelling depend-
ease, potential for preserving vision, and
ing on the degree of extraocular invasion.
extent of intraocular and extraocular dis-
Metastatic spread can occur through infil-
ease. Enucleation performed by an experi-
tration of the optic nerve, dissemination
enced ophthalmologist is indicated for
into the subarachnoid space, invasion into
large tumors filling the vitreous and in
the choroid plexus, and anteriorly into the
those with little or no likelihood of vision
conjunctiva. Invasion through the optic
preservation. Enucleation is often indicated
nerve and subarachnoid can lead to involve-
for unilateral localized retinoblastoma, and
ment of the brain and spinal cord, invasion
alone is effective therapy in these children.
of the vascular choroid plexus can lead to
The eye must be removed intact to avoid
vascular spread, and anterior invasion can
seeding and care must be taken to resect
lead to regional lymph node involvement.
enough optic nerve to ensure negative mar-
Patients with bilateral retinoblastoma
gins. After enucleation, patients are fitted
may present with a concurrent intracranial
with a prosthetic implant.
neuroblastic tumor, referred to as trilateral
Focal treatments such as laser photoco-
retinoblastoma. These patients tend to have
agulation, cryotherapy, and thermotherapy
a poor prognosis and usually present with a
may be utilized for very small tumors. Che-
pinealoblastoma, although they may have a
motherapy (usually vincristine, etoposide,
neuroblastic tumor in the suprasellar region
and carboplatin) can be utilized for local-
as well. The intracranial tumor may present
ized tumors in order to facilitate focal treat-
at the same time as the retinoblastomas, or
ment for tumor eradication and prevent the
after the identification and treatment of the
need for enucleation.
original tumors, emphasizing the need for
Patients with bilateral disease may
close follow up after initial treatment.
require enucleation of the more affected
Ultrasound, CT, and MRI are typically
eye, but fortunately bilateral enucleation is
used in addition to ophthalmologic exam-
rarely indicated. As with localized unilateral
ination to further characterize the tumor.
retinoblastoma, chemotherapy may be uti-
CT is the most useful test as it effectively
lized prior to focal therapy to avoid enucle-
demonstrates intraocular calcifications that
ation in either eye or in the less-affected eye.
confirm the diagnosis, but it creates risk due
External beam radiation therapy is generally
202
Chapter 22
reserved for patients with refractory or
Hepatoblastoma is the most common,
recurrent disease after these other treatment
representing more than
60% of hepatic
modalities have been exhausted due to the
tumors, and occurs almost exclusively
high incidence of side effects including risk
during infancy and young childhood. The
of secondary malignancy.
incidence of hepatoblastoma is increasing,
Patients with regional extraocular disease
possibly due to increased survival in very
and metastatic disease have historically faired
low birth weight infants. Hepatocellular
very poorly. Newer studies have shown the
carcinoma (HCC), approximately 20% to
potential benefit of chemotherapy (or high-
25% of childhood primary liver tumors, is
dose chemotherapy with autologous stem
seen most often in children above 10 years of
cell rescue in those with metastatic disease)
age. Although chronic hepatitis B infection
with external beam radiation therapy.
is the leading cause of HCC in Asia, few
Children treated for retinoblastoma are
American children with HCC have an
at high risk for cosmetic and functional
apparent etiology. Benign hepatic tumors,
impairment related to either enucleation
such as hemangioendothelioma, hamartoma,
or radiation. Patients with a heritable muta-
mature teratoma, angiolipoma, and ade-
tion in the RB1 gene have an especially high
noma, also occur in childhood and
rate of secondary malignancy after radiation
account for up to 20% of liver tumors in this
therapy. Reported second malignancies
age group. Other extremely rare malignant
include head and neck cancers, osteosar-
hepatic tumors include cholangiocarcinoma,
coma, soft tissue sarcoma, and melanoma.
rhabdoid tumor, yolk sac tumor, rhabdomyo-
Adult tumors also have an increased inci-
sarcoma, undifferentiated sarcoma, angiosar-
dence in those with germline RB1 muta-
coma, leiomyosarcoma, and lymphoma.
tions, especially epithelial tumors such as
Malignant hepatic tumors of childhood
lung cancer. Radiation therapy also may
have been associated with a number of
lead to decreased visual acuity, eye irrita-
genetic syndromes and environmental risk
tion, and dry eye as well as growth abnor-
factors. Hepatoblastoma has been reported
malities in the orbital bones. After therapy,
in children with Beckwith-Wiedemann syn-
patients should be followed closely due to
drome, familial adenomatous polyposis
the risk of recurrent disease. Patients with
(which includes Gardner syndrome), and
bilateral retinoblastoma and those diag-
isolated hemihypertrophy. The strongest
nosed with unilateral retinoblastoma at a
association has been reported in infants
young age (i.e., <1 year of age) should be
born at less than 1500 g. Additional associa-
observed for the development of an intra-
tions may include maternal smoking (inde-
cranial mass and, in the case of unilateral
pendent of birth weight), use of infertility
disease, a contralateral retinal mass. Recur-
treatment, fetal alcohol syndrome, use of
rence or new disease rarely occurs after 5
oral contraceptives during pregnancy, and
years of age, although an ophthalmologist
occupational exposure to metals, petro-
should examine these patients annually.
leum, paints, and pigments. HCC is seen
in association with hereditary tyrosinemia,
biliary cirrhosis, glycogen storage diseases,
Liver tumors
hemochromatosis, ataxia telangiectasia, and
a1-antitrypsin deficiency. Infection with
Primary hepatic tumors in childhood and
hepatitis B or C has been identified as a
adolescence account for just more than 1%
risk factor for HCC as have exposure to
of malignancies in these age groups.
alcohol, anabolic steroids, aflatoxin, and
Rare Tumors of Childhood
203
certain carcinogens such as pesticides and
more accurate reporting. Those with HCC
vinyl chloride.
may also have elevation in AFP but not to
the same magnitude as hepatoblastoma.
Clinical presentation
b-HCG may be elevated in patients with
The most common presenting signs of a
carcinoma of the biliary tract. Of note,
malignant hepatic tumor in childhood are
carcinoembryonic antigen (CEA) and vita-
generalized abdominal enlargement or an
min B12 binding capacity may be elevated in
asymptomatic abdominal mass palpated by
some cases of HCC. Imaging studies should
the family or physician. Patients with HCC
begin with abdominal and specifically liver
may present with constitutional symptoms
ultrasound with Doppler flow to character-
including weight loss, anorexia, emesis, and
ize the tumor as well as the relationship
abdominal pain. Rarely, children may pres-
between tumor and hepatic vessels. CT or
ent with jaundice in HCC or in cases with
MRI should also be obtained prior to sur-
biliary tree involvement. Occasionally, males
gical intervention. Angiography may be
with hepatoblastoma present with signs of
required to help with surgical planning, but
isosexual precocious puberty due to excessive
is rarely needed now due to the excellent
production of the beta subunit of human
resolution of current generation CT and
chorionic gonadotropin
(b-HCG) that is
MRI. Evaluation for metastatic disease
converted to testosterone. Lung metastases
should include chest CT.
(typically asymptomatic) may occur at
AFP is the most sensitive marker for
presentation in up to 20% to 30% of children
disease, especially in hepatoblastoma.
with hepatoblastoma and HCC. Extrahepatic
Patients with small-cell undifferentiated his-
extension may include peritoneal implants as
tology have a poorer prognosis and tend to
well as spread to regional and distant lymph
have low serum AFP, which is a poor
nodes, bone, bone marrow, and rarely the
prognostic marker. Decline in AFP levels with
central nervous system. Liver tumors may
courses of treatment is prognostic and AFP
also invade into adjacent intra-abdominal
should be followed for normalization after
structures.
surgical resection and as a marker for poten-
tial disease recurrence. Of note, serum AFP
Diagnosis and staging
levels vary with age in infancy, declining to
Work up of a suspected hepatic mass should
adult levels by around 6 to 8 months of age.
include laboratory measurement of liver
In addition, AFP has a long serum half-life
function with coagulation studies in addi-
of 5 to 6 days and therefore may take several
tion to transaminases, total bilirubin, and
weeks to normalize in cases with an extremely
alkaline phosphatase. Patients should have a
high level at presentation.
baseline complete blood count (CBC) in
In cases where clinical presentation, imag-
addition to metabolic studies as patients
ing, and tumor markers are highly suggestive
may present with thrombocytosis and/or
of hepatoblastoma, patients should undergo
leukocytosis. Patients with suspected hepa-
a primary resection if it is deemed safe and
toblastoma should have elevation of a-feto-
feasible. In cases where a complete resection
protein
(AFP) and, less commonly, of
is not feasible or the diagnosis is uncertain,
b-HCG. In certain cases with an extremely
the patient should first undergo biopsy. A
high AFP, the value reported out may be
current Children’s Oncology Group clinical
falsely low due to overwhelming of the assay
trial hopes to determine whether patients
(hook effect). When the index of suspicion
who undergo a complete resection with a
is high, serial dilution of serum can allow for
favorable histology (i.e., pure fetal histology)
204
Chapter 22
can be safely observed without adjuvant che-
hepatic artery chemoembolization, per-
motherapy. Patients who cannot obtain a
cutaneous radiofrequency ablation, and
complete resection or have a less favorable
percutaneous ethanol injection may be
histology will receive chemotherapy. In the
beneficial in patients with localized yet unre-
past, chemotherapy for hepatoblastoma has
sectable HCC.
consisted of cisplatin, 5-FU (fluorouracil),
and vincristine
(C5V). For patients with
higher risk disease (i.e., small-cell undiffer-
Adrenocortical carcinoma
entiated histology or higher stage), the addi-
tion of other chemotherapeutic agents such
Adrenocortical carcinoma (ACC) is a rare
as doxorubicin and irinotecan is being
malignancy in childhood and adolescence
explored. Patients should receive chemother-
with peaks in both the first and fourth dec-
apy in order to facilitate total resection when
ades of life. The incidence varies worldwide
feasible, and if not feasible, prior to ortho-
with an interestingly high incidence in south-
topic liver transplantation. Consolidative
ern Brazil. ACC has been reported in asso-
chemotherapy is given after resection to treat
ciation with specific genetic syndromes such
any residual microscopic metastatic disease.
as Beckwith-Wiedemann syndrome, isolated
Patients with gross metastatic disease at pre-
hemihypertrophy, Li-Fraumeni syndrome,
sentation have a poor prognosis, as do
and in association with congenital adrenal
patients with recurrent disease.
hyperplasia and multiple endocrine neopla-
In general, HCC has a poor prognosis
sia type
1
(MEN-1 syndrome). Germline
and often presents with multifocal liver and
mutations in the p53 tumor suppressor gene
metastatic disease. Previous studies treated
have been implicated in the majority of pedi-
pediatric patients with a similar chemother-
atric cases in the United States and Brazil.
apeutic regimen as hepatoblastoma
(i.e.,
ACC may be functionally inactive or
C5V) with disappointing results. Gross total
secrete hormones such as glucocorticoids,
surgical resection remains the mainstay of
sex steroids and their precursors, and miner-
cure for HCC. In cases where resection is
alocorticoids. Benign adrenocortical tumors
not safe or feasible at presentation, neoad-
are more common and must be carefully
juvant chemotherapy may be of benefit and
differentiated from ACC. Patients frequently
allow for a later total resection or for ortho-
present with clinical signs and symptoms of
topic liver transplantation.
excessive cortisol production
(Cushing’s
A distinct pathologic variant, fibrolamel-
syndrome) or excessive androgen produc-
lar HCC, has been associated with a higher
tion (virilization). Other less common symp-
resection rate and improved survival com-
toms include abdominal pain and weight
pared to other forms of HCC. Novel tar-
loss. The diagnosis is made based on the
geted agents have shown a modest benefit at
presence of elevated concentrations of adre-
best, but may have promise when used as
nocortical hormones and their intermediates
part of multimodal therapy.
in the serum or urine. Baseline studies
Radiation therapy has not been proven to
should include a 24-hour free cortisol, dexa-
be effective in the treatment of hepatoblas-
methasone suppression test, basal cortisol,
toma and HCC, but it may have a role in
and adrenocorticotropic hormone (ACTH)
palliation of lung metastases. Similarly,
to measure glucocorticoid secretion; testos-
high-dose chemotherapy with autologous
terone, estradiol, androstenedione, dehydro-
stem cell rescue has not as yet been
epiandrosterone sulfate
(DHEA-S), and
found beneficial. Local therapies including
17-OH-progesterone to measure sex steroids
Rare Tumors of Childhood
205
and their precursors; and aldosterone and
a subset of patients with invasive, meta-
renin in patients with hypertension or hypo-
static, or recurrent disease. Mitotane may
kalemia to rule out mineralocorticoid excess.
have benefit in patients with localized, fully
Pheochromocytoma can be ruled out by
resected disease, although this question has
checking catecholamines and metane-
not been clearly answered. Mitotane levels
phrines. Imaging with CT or MRI of the
greater than 14 mg/L seem to be most ben-
abdomen confirms the presence of a supra-
eficial. As mitotane is an adrenocorticolytic
renal mass. FDG-PET may be utilized in
agent, replacement with hydrocortisone and
equivocal cases or if CT/MRI are negative
fludrocortisone is necessary to prevent adre-
with positive hormone markers. FDG-PET is
nal insufficiency. For patients that progress
negative in benign adrenocortical tumors.
on mitotane or have side effects that limit its
Many patients present with large primary
usage, chemotherapy with cisplatin, etopo-
tumors and evidence of metastatic disease
side, and doxorubicin may be beneficial.
involving the lungs, liver, or bones. CT of the
ACC is considered radioresistant, so this
chest, abdomen, and pelvis in addition to
is not a therapeutic option.
bone scan should be done for staging
purposes prior to surgical resection. ACC
are locally aggressive tumors with a thin
Thyroid tumors
pseudocapsule; an open adrenalectomy
should be performed to prevent extensive
The incidence of malignant thyroid carci-
hemorrhage or tumor rupture. Surgical
noma in children is very low, comprising
resection is the only known curative therapy
less than 2% of pediatric cancers. The peak
for ACC, both for primary and metastatic
incidence is in the older adolescent age
sites. Surgery is also indicated in recurrent
group, 15 to 19 years of age, with a prepon-
disease to prolong survival.
derance in females. Childhood thyroid
Due to the poor response to chemother-
carcinomas are clinically and biologically
apy, patients with advanced disease have a
distinct from those seen in adults. The
very poor prognosis. A tumor weight <
majority of thyroid carcinomas are papillary
200 g (<5 cm in diameter in adults) is a
or follicular variants of papillary carcinoma.
good prognostic factor. Patients with local-
Exposure to head and neck irradiation is
ized disease should undergo aggressive gross
associated with an increased risk of thyroid
total surgical resection. Patients with large
carcinoma, particularly the papillary vari-
tumors (>200 g) without metastatic disease
ant. An increased incidence has been seen in
may also benefit from radical retroperito-
children receiving therapeutic radiation
neal lymph node dissection
(RPLND).
with a median latency of 13 years and has
Patients with advanced disease
(invasive
also been observed in survivors following the
or metastatic) should be treated with aggres-
Chernobyl nuclear accident. Genetics plays
sive surgical resection of the primary tumor,
a major role in patients with medullary
RPLND, and resection of metastatic lesions
carcinoma with many children having a
in addition to chemotherapy. If resection
positive family history. Inherited medullary
of metastases is not feasible, resection of
carcinoma is also seen in association with
the primary tumor may not be of benefit.
MEN types 2A and 2B or as part of familal
Mitotane, a synthetic derivative of the insec-
medullary carcinoma. MEN types 2A and 2B
ticide
dichlorodiphenyltrichloroethane
are caused by germline mutations in the RET
(DDT), has a specific cytotoxic effect on
(rearranged during transfection) proto-
adrenocortical cells. It has shown benefit in
oncogene. Patients with MEN type
2A
206
Chapter 22
develop medullary thyroid carcinoma, pheo-
obtain tissue diagnosis. Surgical removal
chromocytoma, and parathyroid tumors.
may be considered in higher risk patients
Patients with MEN type 2B may develop
with suspicious features on imaging, posi-
medullary carcinoma or pheochromocytoma
tive family history, or past history of irra-
and are associated with a marfanoid body
diation. When a thyroid carcinoma is diag-
habitus, mucosal neuromas, and ganglio-
nosed, staging is completed with a neck
neuromatosis. A high incidence of papillary
ultrasound, CT, or MRI to evaluate lymph
carcinoma of the thyroid is seen in familial
nodes, chest CT to evaluate for metastasis,
adenomatous polyposis coli and Cowden
and possibly a neck MRI to assess for degree
disease.
of local invasion.
Patients typically present with a painless
Surgical resection is key to curative ther-
solitary thyroid nodule (approximately 70%
apy in thyroid carcinoma. Patients should
to 75%) and up to 35% to 40% present with
undergo a total or subtotal thyroidectomy.
palpable cervical adenopathy. If the tumor
No specific recommendations are currently
has invaded locally, patients may present with
available in children with regard to lymph
dysphagia or dysphonia. Children are more
node dissection. When lymph node involve-
likely than adults to present with advanced or
ment is confirmed on imaging or by biopsy,
metastatic disease (primarily lungs). Pulmo-
a modified lateral neck dissection is recom-
nary metastases classically appear as miliary
mended. Following surgery, a diagnostic
lesions. Staging in children is based on the
whole body scan with I123 or I131 is recom-
presence or absence of metastases. Younger
mended to define areas of residual disease.
children may have an increased risk of recur-
Radioiodine ablation is then recommended
rence. Presence of metastatic disease, age, size
and has been shown to reduce the incidence
of tumor, and degree of extrathyroid invasion
of locoregional recurrence. Lifetime thyroid
are predictive of outcome.
replacement is required after surgery and
The initial evaluation of the child with a
radioablation.
thyroid nodule should include thyroid
ultrasound as well as measurement of free
Suggested Reading
thyroxine
(FT4) and thyroid-stimulating
hormone (TSH). Up to 20% of thyroid
Khara L, Silverman A, Bethel C, et al. Thyroid
nodules in children will be malignant, and
papillary carcinoma in a 3-year-old American
younger age, past history of irradiation, and
boy with a family history of thyroid cancer: a
family history are all highly predictive of
case report and literature review. J Pediatr
malignant disease. Ultrasound characteris-
Hematol Oncol 23:e118-e121, 2010.
tics may guide the clinician in the diagnosis
Litten JB, Tomlinson GE. Liver tumors in chil-
as malignant tumors are more likely to have
dren. Oncologist 13:812-820, 2008.
indistinct margins, vascularity, an absence
Lohmann D. Retinoblastoma. Adv Exp Med Biol
685:220-227, 2010.
of an echogenic halo around the nodule,
Ribeiro RC, Pinto EM, Zambetti GP. Familial
and presence of calcifications. Nuclear
predisposition to adrenocortical tumors: clin-
medicine thyroid scintigraphy
(usually
ical and biologic features and management
with I123) can be utilized but is not diag-
strategies. Best Pract Res Clin Endocrinol
nostic and delivers quite a bit of radiation.
Metab 24:477-490, 2010.
The majority of thyroid nodules are cold on
Sinnott B, Ron E, Schneider AB. Exposing the
thyroid scan but most of these are benign
thyroid to radiation: a review of its current
follicular adenomas. Image-guided fine
extent, risks, and implications. Endocr Rev
needle aspiration is recommended to
31:756-773, 2010.
Histiocytic Disorders
23
Histiocytes
(composed of dendritic cells
Langerhans cell histiocytosis
and monocytes/macrophages; also called
tissue macrophages) are cells of the mono-
LCH is now the accepted umbrella term
nuclear phagocytic system that fight infec-
for a wide variety of other conditions pre-
tion and clear debris. Histiocytic disorders
viously described, including histiocytosis X,
occur due to abnormal proliferation or
eosinophilic granuloma, Hand-Schuller-
activity of these cells and have a wide variety
Christian syndrome, Letterer-Siwe disease,
of clinical presentations, from the localized
Hashimoto-Pritzker syndrome, self-healing
and mild to the generalized and severe.
histiocytosis, pure cutaneous histiocytosis,
The classification of these conditions can
Langerhans cell granulomatosis, Langerhans
be as confusing as the conditions themselves.
cell
(eosinophilic) granulomatosis, type II
The most recent World Health Organization
histiocytosis, and nonlipid reticuloendothe-
(WHO) classification was developed in 1997
liosis. The pathogenesis of LCH is poorly
and is presented in Table 23.1. While most of
understood. Immune dysregulation likely
the histiocytic disorders are considered non-
plays a role and may be reactive, occurring
malignant conditions, many are treated sim-
secondary to trauma and infection, especially
ilarly to malignant disease, and the mortality
in those patients with remitting disease. A
of several conditions is quite high. The eti-
neoplastic process may also be possible in
ology of most types is unknown, but appears
cases that are systemic and widely dissemi-
secondary to poorly understood pathophys-
nated. LCH is a misnomer as the Langerhans
iological mechanisms. This chapter focuses
cell, an epidermal dendritic cell, is not the
on the more common presentations of dis-
sole culprit; rather, like other histiocytic
ease, specifically Langerhans cell histiocytosis
disorders, LCH is due to abnormalities in
(LCH) and hemophagocytic lymphohistio-
the mononuclear phagocytic system. Diag-
cytosis
(HLH). Less common disorders
nosis is based on histologic features of these
include juvenile xanthogranuloma (includ-
Langerhans cells (characterized by Birbeck
ing Erdheim-Chester disease) and sinus his-
granules on electron microscopy) in addition
tiocytosis with massive lymphadenopathy
to specific immunophenotypic markers
(Rosai-Dorfman disease).
(CD1a and CD207).
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
208
Chapter 23
Table 23.1 Classification of histiocytic disorders.
Group
Cell of origin
Condition
Disorders of varied
Dendritic cell
Langerhans cell histiocytosis
biological behavior
Secondary dendritic cell processes
“Excludes disorders
Juvenile xanthogranuloma and related disorders
that are considered
Solitary histiocytomas of various dendritic cell
to be malignant
phenotypes
while recognizing a
Macrophage
Hemophagocytic lymphohistiocytosis
wide scope of
(familial and sporadic; commonly elicited by
severity ranging
viral infections)
from self-limited to
Secondary hemophagocytic syndromes
lethal disease”
Infection-associated
Malignancy-associated
Other
Rosai-Dorfman disease (sinus histiocytosis with
massive lymphadenopathy)
Solitary histiocytoma with macrophage phenotype
Malignant disorders
Monocyte
Leukemias (FAB and revised FAB classifications)
Monocytic leukemia (M5A and B)
Acute myelomonocytic leukemia (M4)
Chronic myelomonocytic leukemia
Extramedullary monocytic tumor or sarcoma
(monocytic counterpart of granulocytic
sarcoma)
Dendritic cell
Dendritic cell-related histiocytic sarcoma (localized
or disseminated)
Specify phenotype; follicular dendritic cell,
interdigitating dendritic cell, etc.
Macrophage
Macrophage-related histiocytic sarcoma (localized
or disseminated)
Abbreviations: FAB, French-American-British.
LCH most often presents in the bone as a
underlying immune reaction. Observation is
solitary or as multifocal lesions and may also
generally favored in this situation, with certain
be localized to the skin or lymph nodes. Bone
caveats:
lesions may be painful or painless. On plain
1. Vertebral lesions have an inherent risk of
film, lesions appear lytic, often with a
causing vertebral compression
(vertebra
“punched out,” beveled appearance and may
plana) or spinal cord compression if signif-
have an associated periosteal reaction with
icant soft tissue swelling exists, therefore
soft tissue swelling. Skin lesions are scaly,
radiation therapy should be considered.
erythematous papules often involving the
2. Large or symptomatic solitary lesions
scalp. Localized disease in these areas portends
may benefit from surgical curettage or resec-
a good prognosis with chance for spontane-
tion, intralesional steroid injection, or radi-
ous remission due to “burning out” of the
ation therapy.
Histiocytic Disorders
209
3. For solitary bone lesions, the recom-
This condition presents with prolonged
mendation for observation excludes
and excessive activation of macrophages,
sites that put the patient at increased
histiocytes, and cytotoxic T-lymphocytes
risk for the development of diabetes insi-
(CTL). Natural killer (NK) cells, vital to
pidus
(facial bones and anterior/middle
antigen recognition and killing as well
cranial fossa). For these
“special sites,”
as contraction of the immune response
and for multifocal bone, skin, or lymph
from T-cells and histiocytes, often have
node involvement, chemotherapy with
decreased activity, in part allowing for this
12 months of prednisone and vinblastine
hyperproliferative immune response. Viral
is recommended.
infection by Epstein-Barr virus is the most
common known acquired cause, although
Systemic disease, which involves multiple
multiple other viruses have been implicated,
organs including the bone marrow, liver,
and often the pathogen is not identified.
spleen, or lungs, has a much worse progno-
Familial HLH can occur secondary to mul-
sis, especially in those patients without an
tiple known genetic causes that lead to NK
early response to therapy. Poor responders
and CTL dysfunction. The familial causes
likely will require more intensive therapy
are outlined in Table 23.2. An underlying
including, potentially, hematopoietic stem
genetic predisposition may still require a
cell transplantation.
“second hit,” such as a viral infection, to
develop HLH.
Clinical findings are secondary to the
Hemophagocytic
chronic inflammatory state and most
lymphohistiocytosis
notably include fever, splenomegaly,
cytopenias, and hepatitis. Diagnostic cri-
Like other histiocytic disorders, immune
teria for HLH are summarized in
dysregulation is the key feature in HLH.
Table
23.3. For the patient with high
Table 23.2 Genetic mutations associated with HLH.
Gene
Pathophysiology
Perforin 1 (PRF1)
Synthesized in natural killer and cytotoxic
T-cells, permeabilizes target membrane
allowing granzyme B to initiate apoptotic
pathways
MUNC 13-4 (UNC13D)
Important for cytolytic granule fusion to
target membrane
RAB27A (Griscelli syndrome)
Docking of secretory granules
Syntaxin (STX11/STXBP2)
Failure of degranulation when encountering
susceptible targets
LYST (Chediak-Higashi syndrome)
Involved in maturation of cytolytic enzyme
granules
AP3B1 (Hermansky-Pudlak syndrome type II)
Polarization/intracellular movement of
cytolytic granules
SH2D1A (X-linked lymphoproliferative
Polarization/intracellular movement of
syndrome)
cytolytic granules
210
Chapter 23
Table 23.3 Diagnostic criteria for hemophagocytic lymphohistiocytosis.
Diagnosis by molecular criteria (summarized in Table 23.2)
OR
Five of the following eight criteria:
Fever
Splenomegaly
Bicytopenia (i.e., hemoglobin <9 g/dL, platelets < 100
109/L, neutrophils < 1
109/L)
Hemophagocytosis in bone marrow, spleen, lymph nodes, or central nervous system
Fasting hypertriglyceridemia (
265 mg/dL) and/or hypofibrinogenemia (
150 mg/dL)
Low or absent natural killer activity
Ferritin
500 mcg/L
CD25 (soluble IL-2 receptor) >2400 U/mL (based on reference level)
clinical suspicion for HLH, treatment
to the underlying inflammatory state, but
should be initiated promptly even while
these levels will trend down with the
pending pertinent laboratory evaluation.
development of MAS. Fevers may become
Current therapy includes dexamethasone,
nonremitting, and due to worsening liver
etoposide, and cyclosporine. For patients
dysfunction, there may be a paradoxical
that have a complete response after
8
improvement in inflammatory markers
weeks, treatment can be stopped. For
such as the erythrocyte sedimentation rate
those that have not responded completely
(ESR) and fibrinogen with a resultant
or relapse after stopping therapy, hemato-
improvement in inflammatory symptoms
poietic stem cell transplantation is the
such as arthritis. Treatment for MAS
treatment of choice.
includes pulse steroids and cyclosporine.
Excessive activation of macrophages and
Duration of therapy is dependent on clinical
T-lymphocytes may also lead to a secondary
response; patients that do not respond or
or reactive HLH known as macrophage
relapse may benefit from primary HLH
activation syndrome (MAS). Clinical crite-
therapy although outcomes are poor.
ria for MAS are similar to HLH and the
underlying pathogenesis is due to an auto-
immune condition, most commonly sys-
Case study for review
temic onset juvenile idiopathic arthritis
(adult Still’s disease). Multiple other auto-
You are seeing a 6-year-old child that pre-
immune diseases have been reported to
sents to the emergency department with 3
cause MAS. NK cells are vital in the pre-
weeks of an erythematous and painful dif-
vention of autoimmunity; decreased NK
fuse and spreading rash. Fever began soon
activity could result in both the develop-
after the rash with associated weakness,
ment of an autoimmune disease and sec-
malaise, and anorexia. Initial laboratory
ondary HLH. Often it is difficult to separate
tests include the following:
the symptoms of MAS from the underlying
Complete blood count:
autoimmune disease or primary HLH.
12.5
Patients may have leukocytosis, thrombo-
19.8
150
cytosis, and hyperfibrinogenemia secondary
Histiocytic Disorders
211
Differential: 35% segs, 44% bands, 6%
A ferritin is sent and is very elevated
lymphs, and 12% monos
at
68,300 mcg/L. Fasting triglycerides are
253 mg/dL.
ESR: 47 mm/h
CRP: 6.3 mg/dL
Complete metabolic panel:
2. How do these lab values help with the
131
103
12
differential?
222
3.7
28
0.7
Total protein 5.2 g/dL
The patient continues to have signs of an
Albumin 2.1 g/dL
underlying inflammatory picture and
AST 146 U/L
hypercytokinemia. Sepsis should still be
ALT 44 U/L
considered on the differential with the
Total bili 0.3 mg/dL
continued elevated ESR, CRP, and fibrin-
ogen. The worsening bicytopenia though
On examination, the patient is alert and
should make HLH and MAS higher on the
interactive, but seems somewhat uncom-
differential. The extremely elevated ferri-
fortable from the rash. There is mild spleno-
tin is relatively specific for HLH and MAS,
megaly and no hepatomegaly. The patient is
and a level this high is concerning for
started on antibiotics for presumed sepsis/
MAS. The decreasing ESR and fibrinogen
toxic shock with initial resolution of fever.
also raise the concern that the patient is
Cultures are all normal. Due to worsening
moving for symptoms of an underlying
labs and rash, as well as no source of infec-
autoimmune disorder to frank MAS.
tion, the differential diagnosis is revisited.
Finally, the elevated white blood cell count
The patient again becomes febrile.
with neutrophilia makes MAS much more
likely than HLH.
1. What findings in the initial laboratory
tests suggest sepsis as well as HLH and MAS?
The patient has elevated inflammatory markers
Suggested Reading
(CRP and ESR). Hyponatremia and hypoalbu-
minemia are often seen with an underlying
Arceci RJ. When T cell and macrophages do not
talk: the hemophagocytic syndromes. Curr
inflammatory condition. There are no signifi-
Opin Hematol 15:359-367, 2008.
cant cytopenias, making HLH less likely. In
Filipovich AH. Hemophagocytic lymphohistio-
addition, the high CRP makes HLH less likely.
cytosis (HLH) and related disorders. Hema-
You review the lab trends:
tol Am Soc Hematol Educ Prog 127-131,
2009.
Hemoglobin: 12.5 ! 10.3 ! 8.9
Filipovich A, McClain K, Grom A. Histio-
Platelets: 150 ! 98 ! 67
cytic disorders: recent insights into patho-
White blood cell count: 18.4 ! 28.7 ! 39.7
physiology and practical guidelines. Biol
(continued left shift)
Blood Marrow Transplant
16:S82-S89,
ESR: 50 ! 47
2010.
CRP 6.3 ! 12.9 ! 18.1
Fibrinogen: 441 ! 387 mg/dL
Hematopoietic Stem
24
Cell Transplantation
Hematopoietic stem cell transplantation
allogeneic transplantation is generally
(HSCT) has become increasingly accepted
recommended for hematologic malignan-
as a therapeutic modality for a variety of
cies due to the underlying bone marrow
malignant and nonmalignant conditions.
involvement (Table 24.1). In addition to
HSCT involves the ablation of the reci-
the effectiveness of high-dose chemotherapy
pient’s bone marrow with high doses of
in tumor killing, there is benefit of a “graft-
chemotherapy as well as, in some cases,
versus-leukemia” (or lymphoma) effect in
radiation therapy in order to allow engraft-
allogeneic transplant in which the immu-
ment of the donor’s stem cells. Collected
nosensitized donor T-cells can theoretically
stem cells can either be allogeneic (from a
kill residual malignant cells. In contrast,
separate donor) or autologous (from the
autologous transplant is generally reserved
patient). The rationale for HSCT is based
for patients with solid tumors and no evi-
on the logarithmic dose-response curve
dence of bone marrow involvement after
for many chemotherapeutic agents: a much
failure of standard chemotherapeutic regi-
higher dose effectively increases tumor kill-
mens (Table 24.2). Recommendations for
ing at the cost of profound myelosuppres-
allogeneic HSCT in nonmalignant condi-
sion. Secondarily, multidrug therapy is
tions are summarized in Table 24.3.
required to overcome resistance and
heterogeneity within the malignant cell
population.
Types of transplantation
Stem cells for allogeneic transplantation are
Transplantable conditions
collected from bone marrow, peripheral
blood, or umbilical cord blood
(UCB),
Diseases treated using HSCT can be divided
whereas stem cells for autologous transplan-
into malignant and nonmalignant condi-
tation are collected most commonly from
tions. Recommendations for malignant
peripheral blood. Collection of stem cells
conditions are continually being updated
from peripheral blood occurs after mobili-
due to changes in the effectiveness of
zation with granulocyte colony-stimulating
standard chemotherapy and the risks and
factor (G-CSF). The usual volume of bone
benefits of HSCT. In pediatric patients,
marrow required to ensure successful
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Hematopoietic Stem Cell Transplantation
213
Table 24.1 Malignant conditions potentially
transplantation and highest after peripheral
benefiting from allogeneic transplant.
blood stem cell transplant (PBSCT). En-
graftment of donor cells occurs earliest after
Acute lymphoblastic leukemia (ALL) in first
PBSCT. Availability of allogeneic stem cells
remission and high risk for relapse
is often limited by the lack of an eligible
Relapsed ALL in second remission
donor.
Acute myelogenous leukemia (AML)
Juvenile myelomonocytic leukemia (JMML)
Hodgkin or non-Hodgkin lymphoma in
second or subsequent partial or complete
Donor matching in allogeneic
remission
transplantation
Myelodysplastic syndromes (MDS)
Relapsed, refractory, or familial hemopha-
The major histocompatibility complex
gocytic lymphohistiocytosis
(MHC) is a large genomic region on chro-
mosome 6 that encodes the human leuko-
cyte antigen (HLA) system. MHC is vital for
engraftment of donor cells is 10 to 20 mL/kg
the immune system to recognize self versus
of recipient body weight. Younger donors
nonself and varies greatly between indivi-
have a higher proportion of marrow
duals. MHC is divided into two major
repopulating cells. Peripheral blood stem
classes, class I and class II. MHC class I
cells are collected via apheresis and identi-
molecules are found on nucleated cells and
fied by the presence of a cell surface marker,
present the MHC to cytotoxic T-cells and
CD34 (cluster of differentiation). In general,
natural killer cells. MHC class I includes
5
106
CD34þ cells/kg are required to
HLA-A, HLA-B, and HLA-C. MHC class II
ensure engraftment. For allogeneic trans-
molecules are located on antigen-presenting
plant, the physician must balance the risks,
cells (B-cells and macrophages) and present
benefits, and availability of cells derived
the MHC to T helper cells. MHC class II
from bone marrow, peripheral blood, and
includes HLA-DP, HLA-DQ, and HLA-DR.
UCB. Peripheral blood has the highest yield
Many other minor histocompatibility
of CD34þ cells (with G-CSF mobilization),
complexes are important in the immune
whereas UCB has the lowest (due to the
response; however, current donor matching
volume of UCB). The risk of graft-versus-
for HSCT is limited to class I and II
host disease (GVHD) is lowest after UCB
molecules.
Identification of a suitable donor
Table 24.2 Malignant conditions potentially
requires matching the recipient’s MHC
benefiting from autologous transplant.
class I and II antigens with those of the
donor. Greater disparity between donor and
High risk neuroblastoma
recipient leads to increasing risk of rejection
High risk brain tumors (medulloblastoma/
of the donor cells and, if engraftment
PNET)
Metastatic retinoblastoma
occurs, GVHD. Matched family member
Recurrent high risk germ cell tumors
donor grafts have been found to be the least
Relapsed Hodgkin or non-Hodgkin
immunogenic. Unfortunately, 60% to 70%
lymphoma
of patients will not have a matched family
Relapsed Wilms tumor
donor. In these cases, unrelated volunteer
donors can be found through bone marrow
Abbreviation: PNET, primitive neuroecto-
donor registries, such as the National Mar-
dermal tumor.
row Donor Program in the United States
214
Chapter 24
Table 24.3 Nonmalignant conditions benefiting from allogeneic transplant.
Congenital syndromes
Immunodeficiency syndromes
SCID, congenital agammaglobulinemia (Bruton’s),
DiGeorge syndrome, Wiskott-Aldrich syndrome,
chronic mucocutaneous candidiasis,
lymphoproliferative syndromes
Hematologic disorders
Sickle cell disease, b-thalassemia, Fanconi anemia,
Shwachman-Diamond syndrome, Diamond-Blackfan
anemia, dyskeratosis congenita, chronic granulomatous
disease, Chediak-Higashi syndrome, leukocyte
adhesion deficiency
Metabolic disorders
Storage diseases, lysosomal diseases, mucolipidosis,
mucopolysaccharidoses
Acquired syndromes
Severe aplastic anemia
Paroxysmal nocturnal hemoglobinuria
Abbreviation: SCID, severe combined immunodeficiency syndrome.
and Bone Marrow Donors Worldwide that
alloreactivity of these immature cells. Ulti-
coordinates multiple worldwide registries.
mately, the physician must balance the avail-
Due to the unavailability of a matched
ability of a matched donor with other factors
donor in many cases, partially matched
such as the patient’s disease stage, remission
donors must be used with increasing risk
status, and general condition. In most cases,
of immune reactions.
disease stage has a greater impact on survival
Currently, HLA matching is limited to
than the level of HLA mismatch.
HLA-A, HLA-B, HLA-C, HLA-DR, and
HLA-DQ. National Marrow Donor
Pretransplant preparative
Program retrospective reviews on HLA
regimens
matching in unrelated donor bone marrow
transplantation have shown that mismatches
High-dose chemotherapy, with or without
at MHC class I (HLA-A, HLA -B, and HLA
radiation, is administered in order to max-
-C) and MHC class II DR (specifically HLA-
imize tumor killing and, in patients under-
DRB1) each had a separate, significant effect
going allogeneic transplant, to affect a suf-
on survival and risk of GVHD. Additionally,
ficient amount of immunosuppression to
an increasing number of mismatches led to
overcome recipient rejection of the HSCT.
decreasing survival. HLA-DQB1 was also
Commonly used conditioning agents, their
found to have an additive negative effect in
mode of action, and potential side effects are
patients with other mismatches. UCB trans-
summarized in Table 24.4.
plantation has been shown to not require the
same level of HLA matching, and matching is
Engraftment and graft failure
limited to HLA-A, HLA-B, and HLA-DR.
UCB transplantation is thought to be less
Evidence of donor replacement of the reci-
immunogenic secondary to the decreased
pient’s bone marrow begins with increasing
Hematopoietic Stem Cell Transplantation
215
Table 24.4 Common agents used in pretransplant preparative regimens.
Agent
Mode of action
Common potential risks
Antithymocyte globulin
Alteration of function and
Fever and chills, pruritus, anaphy-
elimination of T-cells
laxis, serum sickness
BCNU (Carmustine)
Alkylation leading to DNA
Nausea and vomiting, pulmonary
damage
infiltrates and fibrosis, transami-
nitis, nephrotoxicity
Busulfan
Alkylating agent
Nausea and vomiting, electrolyte
abnormalities, seizures, mucosi-
tis, alopecia, hyperpigmentation,
sterility
Carboplatin
Inhibits DNA synthesis by
Nausea and vomiting, type I
forming DNA cross-links
hypersensitivity, renal
impairment and electrolyte
wasting, ototoxicity
Cyclophosphamide
Alkylating agent, elimination
Fluid retention (SIADH), hemor-
of T regulatory cells,
rhagic cystitis, nausea, vomiting,
immunosuppressant
and anorexia, cardiomyopathy,
sterility
Etoposide
Inhibits topoisomerase II
Hypotension, nausea, skin blisters or
causing DNA strand
erythema, nephropathy, hemor-
breakage
rhagic cystitis, alopecia, stomati-
tis, transaminitis
Fludarabine
Purine analog inhibiting
Nausea, vomiting, and anorexia,
DNA synthesis,
mucositis, hemolytic anemia
immunosuppressant
Melphalan
Alkylating agent
Nausea and vomiting, mucositis
Thiotepa
Alkylating agent
Nausea, vomiting, and anorexia,
sterility, excretion through the
skin
Topotecan
Inhibits topoisomerase I
Nausea, vomiting, and anorexia,
causing DNA strand
diarrhea, mucositis, peripheral
breakage
neuropathy
Total body irradiation
Antitumor activity,
Fever, myelosuppression, mucositis,
immunosuppressant
alopecia, diarrhea, skin reactions
and hyperpigmentation, parotitis,
pancreatitis, multiple late effects
including risk of secondary
malignancy
Abbreviation: SIADH, syndrome of inappropriate antidiuretic hormone. See Chapter 30
and Formulary for more information.
white blood cell counts and decreasing trans-
neutrophil count (ANC) > 0.5
109/L. Due
fusion dependency. Engraftment is defined to
to the volume of CD34þ stem cells, engraft-
occur when the transplant recipient achieves
ment of donor cells occurs earliest after
three consecutive days of an absolute
PBSCT and latest after UCB transplantation.
216
Chapter 24
Engraftment syndrome, characterized by
patients at high risk for infection with a
fever, rash, as well as pulmonary symptoms
variety of organisms. Finally, immunogenic
and weight gain (secondary to capillary leak),
donor T-cells can lead to GVHD due to the
may take place during the initial rapid rise in
recognition of host alloantigens as foreign.
the white blood cell count. After infection has
Multiple late sequelae of HSCT must also be
been ruled out, a short course of intravenous
considered.
steroid therapy can alleviate symptoms.
Primary graft failure is defined as a lack
Infections
of engraftment within 6 weeks after trans-
Practitioners must be cognizant of the many
plant. Chimerism studies, either of the
potential infectious complications in the
peripheral blood or bone marrow, measure
HSCT patient. Infection prophylaxis is an
percentage of donor and recipient cells and
important aspect of supportive care and is
thus also help determine marrow engraft-
outlined in more detail below
(text and
ment or primary graft failure. Factors in
Table 24.12). The duration of severe neutro-
graft failure include: a nonmyeloablative
penia
(ANC < 0.5
109/L) and time to
preparative regimen, insufficient volume
engraftment are significant risk factors, as the
of stem cells
(i.e., UCB transplant),
large majority of patients will have a docu-
increased immunogenicity due to mis-
mented infection after 4 to 5 weeks of neu-
matched HSCT, and the use of myelosup-
tropenia. Even though the white blood cell
pressive agents
(i.e., medications for
count recovers in weeks after transplant,
GVHD). Secondary graft failure (graft rejec-
immune reconstitution takes months to
tion) occurs after the initial wave of engraft-
years depending on the type of transplant
ment secondary to the continued presence
(autologous vs. allogeneic) as well as
of recipient cytotoxic T-cells. Chimerism
the continued use of immunosuppressive
studies must again be utilized to show that
agents to prevent or treat GVHD. The
the majority of cells are of host origin. Other
presence of a central venous catheter is a
causes of graft failure including infection
secondary risk factor. Infections tend to
and recurrence of an underlying hemato-
occur at different times after transplant as
logical malignancy should be ruled out.
outlined in Table 24.5. Work up of fever in
the posttransplant period is outlined in
Table 24.6. Engraftment syndrome, GVHD,
Complications of hematopoietic
and medications such as G-CSF can all be
stem cell transplantation
causes of fever in the posttransplant period
but are diagnoses of exclusion. Clostridium
HSCT recipients have a unique set of poten-
difficile should be considered in the patient
tial complications that must be well under-
with associated symptoms such as fever,
stood when caring for these patients. The
abdominal pain, and diarrhea.
pretransplant preparative regimen can lead
to a number of posttransplant complica-
Veno-occlusive disease/sinusoidal
tions as outlined in Table 24.4. A common
obstructive syndrome
complication is veno-occlusive disease
Veno-occlusive disease (VOD), also called
(VOD) of the liver, also known as sinusoidal
sinusoidal obstructive syndrome
(SOS),
obstructive syndrome (SOS). The profound
results from hepatic injury caused by
immunosuppression that occurs both
high-dose chemotherapy or total body irra-
before and after engraftment puts HSCT
diation leading to fibrosis of small hepatic
Hematopoietic Stem Cell Transplantation
217
Table 24.5 Common infections seen at different time points following hematopoietic stem cell
transplantation.
First 30 days
Bacterial
Gram-negative aerobes and anaerobes
Staphylococcus epidermidis
Fungal
Aspergillus species
Candida
Viral
Herpes simplex type I reactivation
30-120 days
Fungal
Candida albicans and C. tropicalis
Aspergillus
Other Candida sp., Trichosporon sp., Fusarium sp.
Pneumocystis jiroveci
Viral
Cytomegalovirus (CMV)
Adenovirus
Epstein-Barr virus (EBV)
Human herpesvirus 6 (HHV-6)
Protozoal
Toxoplasma sp.
Formerly Pneumocystis carinii, generally considered to be a fungal organism.
vessels. VOD/SOS typically occurs in the
Supportive care in transplant
first
30
days after allogeneic transplant
patients
and presents with worsening weight gain,
jaundice, and hepatomegaly. Treatment of
Routine care for the HSCT patient is a
VOD/SOS is supportive; general guidelines
unique and vital aspect of preventing
are outlined in Table 24.7.
infections and a multitude of other
potential complications including bleeding,
Graft-versus-host disease
transfusion-associated GVHD, and CMV
GVHD occurs due to the proliferation of
reactivation, as well as ABO incompatibility
donor T-cells that recognize host antigen
between recipient and donor.
as foreign. Direct effects of the T-lympho-
cytes and cytokine response lead to the signs
Infection prophylaxis
and symptoms of GVHD. Acute GVHD can
Due to profound immunosuppression,
begin within weeks of transplantation;
HSCT patients are at risk for reactivation
chronic GVHD is defined as GVHD lasting
of latent viral infections in addition to fun-
beyond 100 days after transplant. Signs and
gal and bacterial infection. Guidelines for
symptoms of acute and chronic GVHD are
infection prophylaxis and surveillance are
summarized in Table 24.8, grading of GVHD
outlined in Table 24.12.
is outlined in Table 24.9, and prophylaxis and
treatment are described in Table
24.10.
Diet
Patients should be checked daily for signs
Due to the pretransplant preparative regi-
and symptoms of acute GVHD while hospi-
men, patients will have an extended period
talized following HSCT.
of anorexia requiring total parenteral
nutrition
(TPN). Triglycerides should be
Late sequelae
followed weekly while on TPN in addition
Multiple late effects must be considered and
to routine monitoring of electrolytes. Trace
are summarized in Table 24.11.
elements should be included in the TPN.
218
Chapter 24
Table 24.6 Evaluation and empiric treatment of fever in the posttransplant period.
Rule out bacterial infection
Daily blood cultures from central catheter (aerobic and anaerobic)
while febrile
Consider noncatheterized urinalysis and culture
Rule out pneumonia
Chest radiography at first fever and then as clinically indicated
Rule out fungal infection
Daily fungal cultures from central catheter while febrile
Careful skin exam; if concern for infection perform skin biopsy
CT of the chest and/or sinuses if with localizing signs or symptoms
Rule out occult infection
Consider CT of the chest ( sinuses, abdomen, pelvis) if
asymptomatic with prolonged fevers (i.e.,
5-7 days) without
a source
Consider galactomannan antigen testing if with persistent fever
Rule out viral infection
CMV serology; consider studies for adenovirus, HHV-6, EBV, BK
virus (urine) if prolonged fever (i.e.,
5-7 days) and consistent
clinical symptoms
Rule out pneumonia or
CT of the chest if with consistent symptoms or CXR equivocal
interstitial pneumonitis
Treatment
If not on empiric antibiotic therapy (see Table 24.12), begin
antipseudomonal cephalosporin (i.e., ceftazidime or cefepime)
aminoglycoside (i.e., tobramycin) or carbapenem (i.e.,
meropenem)
If with prolonged fevers (i.e.,
5-7 days), consider switching
empiric cephalosporin to carbapenem for broader (anaerobic)
coverage. Addition of an echinocandin (i.e., micafungin) for
broader fungal coverage with prolonged fevers (i.e.,
3-7 days)
Consider vancomycin for staphylococcal and streptococcal cov-
erage if patient worsening clinically
Directed antimicrobial coverage or further studies based on
positive work-up
Based on institutional preference.
Abbreviations: CT, computed tomography; CMV, cytomegalovirus; HHV-6, human herpesvirus 6;
EBV, Epstein-Barr virus; CXR, chest radiography.
Once the patient is eating, it is vital that the
packed red blood cell (PRBC) transfusions
patient’s family be aware of the bone
should be CMV negative for HSCT recipi-
marrow transplant low microbial diet of
ents who are identified as CMV seronega-
acceptable foods and appropriate methods
tive during the pretransplant period. CMV
of handling, preparation, and storage in
seronegative platelets are not routinely
order to prevent infection from food-borne
required as leukofiltered platelets are con-
pathogens.
sidered sufficiently leukoreduced to pre-
vent transmission of CMV. In order to
Transfusion guidelines
diminish the risk of spontaneous bleeding,
Due to immunosuppression, all HSCT
platelet level is kept above 20
109/L after
patients should receive irradiated blood
HSCT. This threshold is increased to above
products in order to eliminate the risk of
50
109/L in certain cases, including
transfusion-associated GVHD. In addition,
patients with sickle cell disease, brain tumor
Hematopoietic Stem Cell Transplantation
219
Table 24.7 Treatments for veno-occlusive disease/sinusoidal obstructive syndrome.
Supportive care
Fluid and sodium restriction
Spironolactone therapy (in lieu of loop diuretics) for sodium and water
diuresis (e.g., aldactone 1-3 mg/kg/day div BID PO)
Antioxidant therapy
N-acetylcysteine. Loading dose of 150 mg/kg IV over 15 min, followed by
50 mg/kg over 4 hours. Maintenance dose of 100-150 mg/kg/day
Vitamin C, vitamin E, selenium (can be added to parenteral nutrition)
Thrombolytic therapy TPA. 0.1 mg/kg/h IV for 4 hours over 4 consecutive days; maximum dose
5.0 mg/h
Heparin. Initial bolus of 20 units/kg IV (max 1000 units), concomitantly
with TPA infusion, then 150 units/kg/day. Dose adjusted to keep PTT
just at or slightly above ULN
Antithrombin III. Loading dose of 50 U/kg IV every 8 h for the initial 24 h
followed by 50 U/kg daily
Defibrotide is an experimental agent with fibrinolytic and antithrombotic
properties that has shown potential benefit for prophylaxis of VOD/
SOS and possible therapeutic benefit in severe disease. Patients must be
off all other thrombolytic therapies. In experimental study given at
25 mg/kg/day div Q6 h IV for at least 21 days
Abbreviations: TPA, tissue plasminogen activator; PTT, partial thromboplastin time; ULN, upper
limit of normal; VOD/SOS, veno-occlusive disease/sinusoidal obstructive syndrome.
Table 24.8 Signs and symptoms of acute and chronic graft-versus-host disease.
Acute GVHD
Skin
Mild maculopapular rash to generalized erythroderma;
can be nonspecific
GI
Secretory diarrhea, nausea, vomiting, anorexia, stomatitis,
hepatic dysfunction
Hematologic
Anemia, thrombocytopenia
Ocular
Photophobia, hemorrhagic conjunctivitis
Pulmonary
Interstitial pneumonitis, alveolar hemorrhage
Chronic GVHD
Skin
Sclerodermatous changes, contractures, alopecia
GI
Xerostomia, oral atrophy with depapillation of tongue,
oral erythema and/or lichenoid lesions, esophagitis,
cholestasis, malabsorption, hepatic dysfunction
Hematologic
Thrombocytopenia
Ocular
Dry eyes, keratoconjunctivitis sicca (conjunctival and/or
corneal inflammation caused by dryness)
Pulmonary
Interstitial pneumonitis, bronchiolitis obliterans
Other
Arthritis, immunologic abnormalities
Abbreviations: GVHD, graft-versus-host disease; GI, gastrointestinal.
220
Chapter 24
Table 24.9 Grading of graft-versus-host disease.
Clinical Skin
Liver (TB,
Diarrhea (children;
Diarrhea (adults;
grade
mg/dL)
<70 kg) (mL/kg/day)
70 kg) (mL/day)
I
<25% involvement,
1.5-3.0
10-15
500-1000, nausea/
maculopapular
vomiting
II
25-50% involvement,
3.0-6.0
16-20
1000-1500, nausea/
maculopapular
vomiting
III
>50% involvement,
6.0-15.0
21-25
>1500, nausea/
maculopapular or
vomiting
generalized
erythroderma
IV
Desquamation/bullae
>15
>25, pain/ileus
>2500, pain/ileus
Abbreviation: TB, total bilirubin; adapted from Lanzkowsky P. Hematopoietic stem cell
transplantation. In: Manual of Pediatric Hematology and Oncology, 5th ed. New York:
Elsevier, 2011.
Table 24.10 Agents used for prophylaxis and treatment of graft-versus-host disease.
Agent
Mode of action
Common potential risks
Cyclosporine
Inhibits T-cell activation
Hypomagnesemia, bicarbonate wasting,
renal insufficiency, nausea, vomiting,
hyperglycemia, gingival hypertrophy,
hirsutism, hypertension, seizures,
paresthesias, tremors
Methotrexate
Cell cycle specific; inhibits
Transaminitis, mucositis, renal insuffi-
T-cells as they divide
ciency, effusions, nausea, vomiting,
anorexia, bone marrow suppression
Tacrolimus
Inhibits T-cell activation
Hypomagnesemia, hyperkalemia, renal
insufficiency, hypertension, seizures,
paresthesias, nausea, vomiting,
hyperglycemia
Methylprednisolone
Immunosuppressant;
Hypertension, increased blood sugars,
mechanism not well
increased appetite, insomnia, mood
understood
swings, acne, truncal obesity
Mycophenolate mofetil
Inhibits T-cell proliferative
Nausea, vomiting, anorexia, bone
(MMF)
response
marrow suppression, multiple others
Antithymocyte
Alteration of function and
Fever and chills, pruritus, anaphylaxis,
globulin (ATG)
elimination of T-cells
serum sickness
Etanercept/infliximab
TNF-a inhibitors;
Severe immunosuppression
consideration in
refractory GVHD
Abbreviations: TNF, tumor necrosis factor; GVHD, graft-versus-host disease.
Hematopoietic Stem Cell Transplantation
221
Table 24.11 Late effects of hematopoietic stem cell transplantion.
Late effect
Underlying cause
Endocrine disorders (gonadal failure,
TBI
delayed pubescence, growth hormone
deficiency, hypothyroidism)
Sterility
TBI, busulfan, cyclophosphamide, thiotepa
Secondary malignancy
TBI, busulfan, ATG, cyclophosphamide, etoposide,
genetic factors
Cataracts
TBI
Renal insufficiency
Cyclosporine, other nephrotoxic drugs
Pulmonary disease
Chronic GVHD, BCNU
Cardiomyopathy
Anthracycline therapy, TBI, chronic GVHD
Avascular necrosis
Steroid therapy
Leukoencephalopathy
IT methotrexate
Immunological dysfunction
Chronic GVHD, immunosuppressive therapy
Posttransplant lymphoproliferative
Immunosuppressive therapy
disorder
Poor dentogenesis (in young children)
TBI
Decreased bone mineral density
Multiple factors
Abbreviations: GVHD, graft-versus-host disease; TBI, total body irradiation; ATG, antithymocyte
globulin; IT, intrathecal; adapted from Lanzkowsky P. Hematopoietic stem cell transplantation.
In: Manual of Pediatric Hematology and Oncology, 5th ed. New York: Elsevier, 2011.
patients, and patients with severe mucositis,
recover immune function. Loss of memory
VOD/SOS, or problems with hemostasis (e.
B-lymphocytes due to the pretransplant
g., epistaxis and GI bleeding). A lower thresh-
preparative regimen leads to loss of anti-
old of 10
109/L can be utilized in stable
body secondary to vaccination and lifetime
patients who are out of the immediate post-
environmental exposures. Immune recov-
transplant period. Hemoglobin levels are
ery is variable but generally occurs once the
usually kept above 8 to 9 mg/dL depending
patient is off immunosuppressant therapy,
on institutional practice. Platelet transfu-
and therefore is often earlier in patients
sions should be limited to 1 pheresed unit
undergoing autologous transplantation.
(see Chapter 5). ABO blood types of both the
Once there is sufficient evidence of immune
donor and recipient must be considered
reconstitution, HSCT patients will proceed
when transfusing platelets and PRBCs.
with revaccination. Studies of immune
Appropriate transfusion in ABO-incompat-
reconstitution, which begin once the abso-
ible donor and recipient is summarized in
lute lymphocyte count is >1
109/L and
Table 24.13.
the patient is off immunosuppressive ther-
apy, are summarized in Table 24.14.
Immune reconstitution
Case study for review
Even with the recovery of white blood cell
counts and specifically lymphocyte counts,
A 14-year-old boy is on the bone marrow
HSCT patients take months to years to
transplant unit after receiving an HSCT for
222
Chapter 24
Table 24.12 Routine infection prophylaxis and surveillance in transplant patients.
Prophylaxis for
PCP
TMP-SMX until 2 days prior to transplant, then restarted once ANC
>0.75
109/L for 2-3 consecutive days; for at least six months (until
PRP >1 mcg/mL or per institutional guidelines, see Table 24.14)
HSV
If seropositive, at least 30 days of acyclovir (until off immunosup-
pressive therapy; until HSV blastogenesis present if with recurrent
infection)
VZV
If seropositive, at least 180 days of acyclovir (until VZV blastogenesis
present)
Candida albicans
Fluconazole until immune reconstitution to Candida; hold if
transaminases increased to >2-3
ULN
Bacterial infection
Anti-pseudomonal cephalosporin (i.e., ceftazidime or cefepime)
aminoglycoside (i.e., tobramycin) or carbapenem (i.e., meropenem)*;
start with first fever or once ANC drops to <0.5
109/L if afebrile,
continue until afebrile and ANC > 0.5
109/L for 2 days
Routine surveillance
CMV
Weekly serology
Occult bacterial infection
Weekly to three times weekly blood culture if on steroids and afebrile
(controversial)
Pneumonia
Weekly chest radiograph (controversial)
Deficient IgG
Monthly quantitative IgG; replace with IVIG if IgG < 400 mg/dL
Based on institutional preference.
Abbreviations: PCP, Pneumocystis jiroveci pneumonia; TMP-SMX, trimethoprim-sulfamethoxazole;
PRP, polyribose phosphate; ANC, absolute neutrophil count; HSV, herpes simplex virus; VZV,
varicella zoster virus; ULN, upper limit of normal; CMV, cytomegalovirus; IgG, immunoglobulin
G; IVIG, intravenous immune globulin.
acute myelogenous leukemia
(AML) in
otherwise unremarkable. His medications
remission. His stem cell reinfusion took
include ceftazidime, tobramycin, flucona-
place 14 days ago. He has been having daily
zole, and acyclovir.
fevers up to 39.5
C for 3 days with negative
blood cultures to date. He is not in any
1. What is the differential for fever at this
respiratory distress and his physical exam is
point after HSCT?
unremarkable except for a mild rash. His
laboratory tests are remarkable for a WBC
Differential diagnosis:
count that has been increasing, most
Bacterial infection (Gram-negative rods,
recently
0.2
109/L. He has a double
Staphylococcus epidermidis, Streptococ-
lumen central catheter and is on total
cus viridans species)
parenteral nutrition. He has moderate-
Viral infection (CMV, herpes simplex I)
to-severe mucositis that is improving. His
Fungal infection (Candida, Aspergillus)
weight has been stable and his tests are
Engraftment syndrome
Hematopoietic Stem Cell Transplantation
223
Table 24.13 Transfusion in ABO-incompatible transplantation.
Recipient Donor Transplant
Choice for
Choice for First-choice Second-
type
type
incompatibility PRBC
plasma
platelets
choice
transfusion
platelets
A
O
Minor
O
A, AB
A
AB, B, O
A
B
Major
O
AB
AB
A, B, O
A
AB
Major
A, O
A, AB
AB
A, B, O
B
O
Minor
O
B, AB
B
AB, A, O
B
A
Major
O
AB
AB
B, A, O
B
AB
Major
B, O
B, AB
AB
B, A, O
O
A
Major
O
A, AB
A
AB, B, O
O
B
Major
O
AB
B
AB, A, O
O
AB
Major
O
AB
AB
A, B, O
AB
O
Minor
O
AB
AB
A, B, O
AB
A
Minor
A, O
AB
AB
A, B, O
AB
B
Minor
B, O
AB
AB
B, A, O
Abbreviation: PRBC, packed red blood cell.
Table 24.14 Immune reconstitution studies following bone marrow transplantation.
Test
Interpretation
Blastogenesis
Viral panel (CMV, HSV, VZV)
Measures response to CMV, HSV, and
VZV and defines length of acyclovir
prophylaxis
Mitogens (Concanavalin A, poke-
Concanavalin A measures T- and B-cell
weed, phytohemagglutinin)
function
Pokeweed measures B-cell function
Phytohemagglutinin measures T-cell
function, must be reactive before
PCP prophylaxis is discontinued
Antigens (tetanus toxoid, Candida
Lack of response to tetanus toxoid
albicans)
indicates need for revaccination
Response to Candida defines length of
fluconazole prophylaxis
PRP
(If done by institution)
Measurement of T- and B-cell function;
indicates need for PCP prophylaxis as
well as ability to respond to conju-
gated vaccines
Abbreviations: CMV, cytomegalovirus; HSV, herpes simplex virus; VZV, varicella zoster virus; PCP,
Pneumocystis jiroveci pneumonia; PRP, polyribose phosphate.
224
Chapter 24
2. What medications would you consider
Viral infection
adding based on the above differential?
Engraftment syndrome
Heart failure
(secondary to infection,
The patient is well covered for Gram-neg-
fluid overload, and previous anthracy-
ative infections with ceftazidime and
cline therapy)
tobramycin. Gram-positive infections
Graft-versus-host disease
usually have a positive blood culture,
although vancomycin should be consid-
At this time point after transplant, with a
ered if the patient is clinically worsening
negative infectious work up, VOD/SOS is
(i.e., respiratory distress or hypotension).
the most likely diagnosis. If the patient is
The patient is at risk for S. epidermidis
having increasing WBC counts with rash
secondary to the central catheter and
and diarrhea, engraftment syndrome is
S. viridans secondary to mucositis, neither
possible but should be a diagnosis of exclu-
of which is covered with ceftazidime and
sion. Although GVHD is a possible cause of
tobramycin. Fungal infection must be
hyperbilirubinemia and weight gain, it is
seriously considered. Fluconazole will
less likely this soon after HSCT. Heart
cover Candida albicans but not other can-
failure is a possibility secondary to toxicity
didal species or Aspergillus. You should
from therapy or an underlying infection
consider discontinuing fluconazole and
and should be ruled out with
starting an antifungal agent with a broader
echocardiogram.
spectrum of activity such as an echinocan-
din (i.e., micafungin). Micafungin has a
4. What would be changes in therapy that
broader spectrum of activity than flucon-
you should institute?
azole, but only has moderate coverage
against Aspergillus and does not cover most
Most patients with VOD/SOS ultimately
mold species. Finally, the addition of
recover with supportive care alone. Fluid and
steroids for engraftment syndrome with
sodium restriction should be initiated in
fever and rash should be a consideration
addition to starting spironolactone therapy.
if infection is relatively well ruled out
An abdominal ultrasound with Doppler flow
after negative blood cultures and chest
should be undertaken to look for hepatic
radiography.
fibrosis and changes in portal venous flow.
After a few days the patient defervesces
Antioxidants including vitamin C, E, and
but has developed an increase in his
selenium should be added to the TPN. If the
bilirubin. The infectious work up has been
patient develops increasing weight gain and
negative to date. On looking at his fluid
liver dysfunction, more aggressive interven-
status for the last 48 hours, you note that he
tions such as antithrombin III therapy or
is 2 L positive and his weight has increased
defibrotide should be instituted.
by 2 kg from the time of his transplant.
3. What is the differential diagnosis for
Suggested Reading
hyperbilirubinemia and weight gain at this
point after transplant?
Copelan EA. Hematopoietic stem-cell trans-
plantation. N Engl J Med
354:1813-1826,
Differential diagnosis:
2006.
Veno-occlusive
disease/sinusoidal
Coppell JA, Richardson PG, Soiffer R, et al.
obstructive syndrome
Hepatic veno-occlusive disease following
Hematopoietic Stem Cell Transplantation
225
stem cell transplantation: incidence, clinical
Lanzkowsky P. Hematopoietic stem cell transplan-
course, and outcome. Biol Blood Marrow
tation. In: Manual of Pediatric Hematology and
Transplant 16:157-168, 2010.
Oncology, 5th ed. New York: Elsevier, 2011.
Ferrara JL, Levine JE, Reddy P, Holler E. Graft-
Wayne AS, Baird K, Egeler RM. Hematopoietic
versus-host disease. Lancet
373:1550-1561,
stem cell transplantation for leukemia. Pediatr
2009.
Clin N Am 57:1-25, 2010.
Supportive Care of the
25
Child with Cancer
Much of the dramatic improvement in
they have an increased exposure to nosoco-
outcomes in pediatric oncology over the
mial organisms as well. Many of these chil-
last 50 years can be attributed to the devel-
dren have central venous catheters and may
opment of novel chemotherapeutic agents
intermittently have mucosal breakdown
and an increase in therapeutic intensity.
secondary to chemotherapy, both factors
With this increased intensity, supportive
disrupting the integrity of the body’s phys-
care has become a vital component in
ical defense barriers. Poor nutrition also
preventing, recognizing, and treating the
plays a significant role in host susceptibility.
potential side effects of this therapy.
Certain standards of care are indicated to
The practitioner should be familiar with
minimize the risk of acquiring infection in
the potential risks of chemotherapy as out-
these children. Infection prophylaxis
lined in Chapter 30, guidelines for trans-
remains the cornerstone of supportive care
fusional supportive care as described in
in children with malignancies, decreasing
Chapter 5, and treatment for febrile neu-
morbidity and mortality.
tropenia as discussed in Chapter 27. Here
General measures for the prevention of
we discuss infection prophylaxis, anti-
infection include avoidance of crowded
emetic therapy, and the use of hematopoi-
environments, wearing a mask in public when
etic growth factors. Specific supportive care
severely neutropenic (i.e., absolute neutro-
information for patients after hematopoi-
phil count [ANC] <0.5
109/L), and careful
etic stem cell transplantation
(HSCT) is
hand washing (by the patient and all those
discussed in Chapter 24.
who have direct contact). Good nutrition and
proper dental hygiene cannot be overempha-
sized. Oral hygiene should include daily
Infection prophylaxis
brushing (with a soft brush) and oral rinsing
with tap water or saline solution. Cleanliness
Children receiving chemotherapy for treat-
of the perianal area is important, especially in
ment of their malignancies are susceptible to
the neutropenic state. Constipation should be
acquiring infection from bacterial, viral,
avoided with an age-appropriate diet, and, if
fungal, and protozoal organisms. They are
necessary, a stool softener
(e.g., docusate
chronically immunosuppressed as a direct
sodium) or laxative (e.g., MiraLax). Rectal
result of chemotherapy, and at times
suppositories and rectal temperatures should
severely myelosuppressed. While inpatient
be avoided to decrease the possibility of a
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Supportive Care of the Child with Cancer
227
mucosal tear and infection with enteric
Fungal prophylaxis is indicated in
organisms.
severely myelosuppressed patients. Mouth
Trimethoprim/sulfamethoxazole (TMP/
care with brushing and oral rinsing is
SMX) prophylaxis is indicated in immuno-
routinely recommended for the prevention
suppressed children to reduce the risk of
or treatment of mouth sores and yeast. Flu-
acquiring Pneumocystis jiroveci pneumonia
conazole (3 mg/kg/day PO or IV) is indicated
(PCP) due to T-lymphocyte dysfunction
in patients that are at very high risk of
secondary to chemotherapy. Current pro-
developing infection (i.e., acute myelogenous
phylactic dosing is 5 mg/kg/day of the TMP
leukemia [AML], relapsed acute lymphoblas-
component in two divided doses on two to
tic leukemia [ALL] patients). The most com-
three successive days per week. Evidence
mon fungal infections in patients receiving
indicates that this regimen also provides
intensive chemotherapy include candidiasis
good general antibacterial prophylaxis,
and aspergillosis. Efforts to prevent invasive
although probably more effectively in
fungal disease (especially Aspergillus sp., an
patients given daily prophylaxis, which
airborne organism) include respiratory iso-
must be balanced with potential allergy or
lation, laminar air flow rooms, and high-
intolerance to TMP/SMX and especially
efficiency particulate air filters. Patients
secondary to myelosuppression from the
should not have live plants in the room, play
therapy. For patients unable to take TMP/
in dirt or gardens, or be in proximity to
SMX, inhaled pentamidine or oral dapsone
construction work. Some patients may ben-
may be given. Pentamidine is preferred but
efit from the addition of an echinocandin (e.
due to the need for a cooperative patient
g., micafungin
1.5-2 mg/kg IV daily) for
who can appropriately inhale the medica-
broader prophylactic fungal coverage while
tion, is usually limited to children above the
inpatient with prolonged fevers and severe
age of 7 years. The dose is 300 mg inhaled on
neutropenia (see Chapter 27).
a monthly basis. Dapsone is given once
Prophylaxis for subacute bacterial
weekly at 4 mg/kg, with a maximum dose
endocarditis is recommended for patients
of 200 mg. It is dispensed as 25 and 100 mg
with central venous catheters undergoing
tablets. Patients should commence prophy-
invasive procedures that could cause tran-
laxis at the time of diagnosis and continue
sient bacteremia and seeding of the catheter
until 3 months after completion of therapy
or heart valves. Such interventions include
to ensure T-lymphocyte immune reconsti-
dental cleaning or procedures and poten-
tution. Prophylaxis with TMP/SMX should
tially surgery involving the gastrointestinal
be discontinued one day prior to the admin-
or genitourinary tract (controversial). The
istration of high-dose IV methotrexate
standard regimen is the same as recom-
(doses >1 g/m2) and restarted after the
mended by the American Heart Association
serum methotrexate level has fallen to below
for children with congenital heart disease:
1
10-7 M due to competitive excretion
amoxicillin 50 mg/kg (maximum 2 g) orally
between TMP/SMX and methotrexate and
1 hour prior to the procedure. Azithromy-
the risk for delayed methotrexate clearance.
cin may be given to penicillin-allergic
Temporary interruption of TMP/SMX or a
patients
(15 mg/kg, max 500 mg, 1 hour
change to an alternative therapy may be
prior to the procedure).
necessary for prolonged marrow suppres-
sion or transaminitis rather than decreasing
Viral prophylaxis and treatment
the dose of maintenance chemotherapy
Common viral infections may be particu-
for children with acute lymphoblastic
larly virulent in immunocompromised chil-
leukemia (ALL).
dren. These viruses include varicella zoster
228
Chapter 25
(VZV), herpes simplex (HSV), cytomegalo-
chemotherapy during the incubation period
virus (CMV), Epstein-Barr virus, hepatitis
should be based on the intensity of expo-
types A and B, respiratory syncytial virus,
sure, condition of the patient, and intensity
and rubeola (measles). Infection with these
of the chemotherapy. If >96 hours have
viruses may result in prolonged viral excre-
passed since exposure, the patient should
tion, increased morbidity, or death. At the
be given acyclovir at 80 mg/kg/day (max
time of diagnosis, an immunization and
dose 800 mg/dose) divided QID PO for 7
infection history should be determined. In
days. In the event of varicella or zoster
addition, serologies for VZV, HSV, and
infection, chemotherapy should be stopped
CMV should be obtained as potential future
and IV acyclovir should be given, 30 mg/kg/
exposure to and infection with these agents
day divided TID for 7 to 10 days, until all
will result in different treatment recommen-
lesions are crusted and no new lesions have
dations depending on the potential for pri-
appeared for 24 to 48 hours. Monitor renal
mary infection versus reactivation. CMV
function and fluid status daily due to poten-
serology should also be known in determin-
tial nephrotoxicity from acyclovir. Of note,
ing whether a potential HSCT patient needs
it is safe for household contacts to receive
CMV-negative blood (see Chapter 5).
varicella vaccination as transmission from
The child exposed to varicella or zoster
healthy recipients rarely occurs. If skin
with known negative varicella immune sta-
lesions occur after vaccination, exposure
tus (negative titers [IgG] and no history of
should be avoided until all lesions have
varicella infection) should receive Varicella
crusted over.
Zoster Immune Globulin (VariZIG) within
Children with a history of recurrent
96 hours of exposure (of note, VariZIG
herpes simplex infections are at an
remains an investigational agent in the
increased risk of reactivation with subse-
United States and requires institutional
quent courses of chemotherapy or during
review board approval and completion of
and after HSCT. Acyclovir administered
an investigational new drug form). Parents
prophylactically can prevent or decrease
should be counseled on the risk of exposure
the severity of recurrent herpes infection;
for the child with negative immune status
the recommended oral dose is 200 mg TID-
and the need for immediate evaluation and
QID for children above 2 years of age.
treatment if exposed. The dosage is one vial
Immunocompromised patients with active
(125 units) per 10 kg, with a minimum dose
HSV infection should receive IV acyclovir
of 125 units and a maximum of 625 units (5
(30 mg/kg/day or 1500 mg/m2/day divided
vials), intramuscularly. Administration of
q8h). HSCT guidelines for HSV and
VariZIG extends the incubation period
VZV prophylaxis are summarized in
from 14 to 28 days and decreases, but does
Chapter 24.
not eliminate, the possibility of clinical
infection with VZV (the incubation period
generally is shortened in the immunocom-
Immunization during
promised patient who does not receive Var-
chemotherapy
iZIG). If VariZIG is not available then
intravenous immune globulin can be given
Patients receiving chemotherapy, or who are
at 400 mg/kg. Myelosuppressive chemother-
otherwise immunocompromised, should not
apy may need to be stopped 7 days after the
receive live virus vaccines (MMR, live atten-
exposure and held until the end of the
uated influenza vaccine [LAIV; FluMist
],
incubation period. The decision to hold
varicella, rotavirus, and oral polio). Siblings
Supportive Care of the Child with Cancer
229
or household contacts should not receive oral
Catch-up immunizations should occur
polio although MMR, varicella, and rotavirus
after the completion of therapy. Most
are safe as transmission is rare. Limited
sources feel that waiting 3 to 6 months after
information is available regarding risk of
therapy completion will produce sufficient
transmission with LAIV; since an inactivated
immune reconstitution to allow an appro-
form of influenza vaccination is available,
priate response to inactivated vaccines.
this should be the preferred immunization
Patients who had not previously completed
in family members as well as health care
their primary vaccination series should
providers. Although varicella vaccination has
restart from the beginning. Patients who
been shown to be safe in patients with ALL in
previously completed the primary series
maintenance, the benefits and risks of this
can have antibody titers drawn to deter-
immunization should be weighed carefully.
mine what protection they have lost, if
Consideration should be given to the likeli-
any. Children above 5 years of age (not
hood of developing a protective response
including patients with HL) have a small
while still on chemotherapy and the poten-
risk of developing significant disease
tial to develop active infection with admin-
from pneumococcus or Hemophilus in-
istration of a live attenuated vaccine. This
fluenza, but revaccination against these
must be weighed with the significant risks
pathogens should be considered in all
from natural varicella infection in the immu-
patients after therapy. Postvaccination
nocompromised, although the chance of
titers can be considered although the
infection has decreased significantly with
likelihood of an insufficient response is
herd immunity.
minimal in patients who are vaccinated
Inactivated vaccines (e.g., DTaP, Tdap,
6 months after chemotherapy has been
Hepatitis A, Hepatitis B, pneumococcal,
completed. Little evidence exists as to
Hib, IPV, meningococcal, and influenza)
the appropriate and safe timeframe to
can be safely given during therapy, although
receive live vaccinations after chemo-
response will be significantly attenuated
therapy. In general, we recommend waiting
based on the level of immunosuppression.
12 months after therapy completion before
Yearly influenza vaccination is reasonable
reimmunizing with MMR and varicella.
during therapy as the potential benefit of
HSCT guidelines are briefly described in
immunization outweighs the risks, espe-
Chapter 24.
cially for children with ALL in maintenance.
In order to increase the chance of an appro-
priate antibody response, vaccination
Prevention of chemotherapy-
should be spaced out from chemotherapy
induced nausea and vomiting
as much as feasible. Due to potential risks of
pneumococcal and Hib infection in patients
Antiemetics are a vital component of the
with Hodgkin lymphoma
(HL) some
supportive care regimen for patients
experts recommend vaccination against
receiving chemotherapy or radiation.
these pathogens prior to starting therapy.
There are three types of chemotherapy-
Although not immunocompromised, un-
induced nausea and vomiting (CINV): (1)
treated patients with HL have an underlying
anticipatory, (2) acute, and (3) delayed.
B-lymphocyte dysfunction and the benefits
Anticipatory emesis occurs before chemo-
of immunization prior to therapy are often
therapy is administered and may be a
significantly diminished. Evidence is lacking
result of nausea and vomiting experienced
to make firm recommendations.
during previous cycles of therapy. Acute
230
Chapter 25
emesis occurs within the first 24 hours of
based on limited evidence in pediatric
therapy; delayed emesis occurs at least 24
patients and are generally derived from
hours after therapy has been completed.
adult data. Here we summarize general
Poor control of nausea and vomiting may
recommendations for the use of these
prolong, or result in, hospitalization and
agents in pediatric oncology.
lead to dehydration and electrolyte abnor-
malities. The single most important factor
Granulocyte colony-stimulating
in CINV is the emetogenic potential of a
factor
particular chemotherapeutic agent, which,
G-CSF is a lineage specific cytokine that
in pediatric patients, is also dose depen-
stimulates the proliferation of neutrophils
dent for some drugs (Table 25.1).
(granulocytes). As patients may have pro-
Multiple agents are useful for the pre-
longed periods of myelosuppression after
vention and treatment of CINV. Recogni-
chemotherapy, G-CSF is recommended
tion of the chemoreceptor trigger zone and
for adult patients with: (1) the expectation
the importance of the 5-HT3 receptor have
for prolonged myelosuppression to pre-
been instrumental in better controlling
vent episodes of febrile neutropenia and
CINV. Cytotoxic chemotherapy appears
infection, (2) a history of febrile neutro-
to be associated with release of local
penia, (3) the history of a delay between
mediators such as 5-HT and substance P
chemotherapy cycles, and (4) a diagnosis
from the enterochromaffin cells of the
of febrile neutropenia. Pediatric evidence
small intestine. The release of these local
is minimal compared to adult studies but
mediators subsequently stimulates vagal
supports the use of G-CSF for patients with
afferents that initiate vomiting.
5-HT3
two of these four scenarios: (1) prevention
receptors and substance P receptors (called
of febrile neutropenia and infection (pri-
neurokinin-1) are located both peripherally
mary prophylaxis), and (2) treatment of
(vagal nerve terminals) and centrally in the
febrile neutropenia. Although infection-
chemoreceptor trigger zone; thus,
5-HT3
related morbidity has not been shown
and substance P antagonists may have both
to decrease with G-CSF usage in these
peripheral and central effects in inhibiting
scenarios, risk of infection and length of
vomiting.
hospitalization have been shown to be sig-
The major pediatric antiemetics, and
nificantly reduced in meta-analyses, with
their specific mechanisms of action and
potential benefits for quality of life and
dose, are summarized in Table 25.2.
decreased cost from shorter hospitalization.
GM-CSF has shown similar results to G-CSF
but has not been shown to be superior in
Hematopoietic growth factors in
clinical studies. In general, G-CSF is the
children with cancer
colony-stimulating factor of choice in
pediatric patients. Clinical studies have
Several hematopoietic growth factors have
also established that the appropriate dose
been approved for clinical use in children
of G-CSF and GM-CSF are 5 mcg/kg/day
including granulocyte colony-stimulating
and 250 mcg/m2/day respectively, given SC
factor
(G-CSF, filgrastim) granulocyte-
or IV. It should be noted that the IV dose
macrophage colony-stimulating factor
may be less effective than the SC dose. A
(GM-CSF, sargramostim), and erythro-
longer lasting pegylated form of G-CSF
poietin (EPO, epoetin alfa). The indica-
(pegfilgrastim) has been shown to be
tions for each of these growth factors are
equally efficacious in adult studies with
Supportive Care of the Child with Cancer
231
232
Chapter 25
Table 25.2 Common pediatric antiemetic agents.
Agent
Mechanism of action
Dose
Comments
Ondansetron
5-HT3 receptor
0.15 mg/kg q8h IV/PO
Well tolerated; common
antagonist
(max 32 mg/day)
side effects include
headache, fatigue, con-
stipation, diarrhea. Also
available as orally disin-
tegrating tablet
Lorazepam
Interaction with
0.25-0.5 mg q4-6 h IV/
Used as adjunctive; can be
GABA receptor;
PO, max dose 2 mg
utilized for anticipatory
poorly understood
nausea. At higher doses
antiemetic effects
has more sedation/anxi-
olytic effect than anti-
emetic effect
Diphenhydramine
H1 histamine recep-
0.5-1 mg/kg q6 h IV/
Used as adjunctive; can be
tor antagonist
PO, max dose 50 mg
utilized for anticipatory
nausea. Higher dose used
for prevention of dys-
tonic reaction with
metoclopramide
Metoclopramide
Dopamine antagonist
1 mg/kg q4-6 h IV/PO,
Used as adjunctive; higher
max dose 50 mg
dose required for anti-
emetic effect as com-
pared to prokinetic.
Must be given with
diphenhydramine at
higher dose
Decadron
Poorly understood
5 mg/m2 q6 h IV/PO
Used as adjunctive; cannot
be used in malignancies
where steroids are part of
the treatment regimen
Scopolamine
Anticholinergic
Transdermal patch for
Must be changed q72 h;
adolescents/adults
patient must be advised
to not touch patch and
then rub eyes as this will
lead to mydriasis
Dronabinol
Cannabinoid; agonist
5 mg/m2 q2-4 h, max
No established pediatric
antiemetic effect
dose 15 mg/m2 in
dosing. Teens and young
adults
adults should be advised
to not smoke cannabi-
noids that can contain
impurities or increase the
risk of fungal infection
Aprepitant
Neurokinin-1 recep-
80-125 mg daily PO in
Pediatric dosing not estab-
tor antagonist
adults
lished; insufficient stud-
ies in pediatric patients
to date
Supportive Care of the Child with Cancer
233
less frequent dosing; pediatric randomized
LDH, or alkaline phosphatase. Occasionally
controlled studies are lacking.
G-CSF has been reported to cause fever,
nausea and vomiting, diarrhea, splenomeg-
aly, and erythema at the injection site.
Primary prophylaxis
G-CSF should be given to patients receiving
Recombinant human
multiagent chemotherapy who are expected
erythropoietin
to experience a high incidence of febrile
Erythropoietin induces proliferation and dif-
neutropenia or severe, prolonged neutrope-
ferentiation of red blood cell progenitors. The
nia (i.e., ANC <0.5
109/L for 7 or more
recombinant product erythropoietin alfa
days). G-CSF should not be given on the
(rHuEPO) has been approved in pediatric
same days patients are given myelosuppres-
patients although administration of rHuEPO
sive chemotherapy or radiation therapy.
to oncology patients with chemotherapy-
G-CSF is generally not administered to
induced anemia remains controversial. A sec-
patients with myeloid leukemia due to the
ond recombinant, darbopoietin alfa, which
theoretical risk of stimulating proliferation
has a two- to threefold longer half-life, has
of the leukemic clone.
been approved in adult patients only.
Meta-analysis of adult data has shown
that although rHuEPO decreases transfu-
Treatment of febrile neutropenia
sion requirements in cancer patients, there
Pediatric guidelines are not clear but treat-
is a significant increase in venous throm-
ment should be considered in patients with
boembolism and, potentially, mortality.
profound neutropenia (ANC <0.1
109/L),
Pediatric data are limited but have not to
uncontrolled primary disease, pneumonia,
date shown a significant difference in sur-
hypotension, multiorgan failure, and inva-
vival with the use of rHuEPO, although
sive fungal infection.
quality of life may be improved. In addi-
tion, venous thromboembolism has not
Duration
been seen in pediatric patients. Concern
G-CSF should be started between 1 to 5
remains that the decrease in adult survival
days after the last dose of myelosuppressive
rates with rHuEPO may be secondary to
chemotherapy or radiation. G-CSF should
the ubiquitous expression of the EPO-
be continued until the ANC is greater than
receptor on tumor cells and therefore
1.5
109/L for 1 to 2 days following the
tumor upregulation with rHuEPO usage.
expected neutrophil nadir from chemo-
Conclusive in vivo data are lacking.
therapy. Specific protocols may call for
Pediatric guidelines for rHuEPO therefore
different ANC thresholds.
include the following: (1) use of rHuEPO is
not recommended in pediatric oncology
patients, and (2) rHuEPO should be consid-
Monitoring
ered on a case-by-case basis in special popu-
Once G-CSF is initiated, a complete blood
lations where blood product usage is rela-
count
(CBC) and differential should be
tively contraindicated. Specifically, Jehovah’s
monitored at least weekly.
Witnesses forbid blood product transfusion
and therefore the potential risks and benefits
Adverse effects
of rHuEPO should be discussed with the
The most common side effects of G-CSF
practicing patient and their family. A candid
are bone pain and elevation of uric acid,
discussion should occur between the
234
Chapter 25
provider, patient, family, and potentially
Suggested Reading
patient advocate, to discuss this complex
situation. Although rHuEPO may ameliorate
Feusner J. Guidelines for Epo use in children
some transfusion need, it may not be able to
with cancer. Pediatr Blood Cancer 53:7-12,
2009.
eliminate this need completely. Additionally,
Hesketh PJ. Chemotherapy-induced nausea
the patient and family should be made aware
and vomiting. N Engl J Med
358:2482-
of the theoretical risks regarding survival
2494, 2008.
with rHuEPO usage.
Central Venous
26
Catheters
Indwelling central venous catheters (CVCs)
These catheters are typically made of plastics
have revolutionized the care of children
with flexible silicone rubber tubing. They
with cancer by simplifying the administra-
are surgically placed
(and removed) by
tion of therapy, increasing safety, decreasing
experienced practitioners via a percutane-
pain, and improving quality of life by reduc-
ous or cut down technique. The patient
ing physical and psychological stress.
should be assessed for bleeding risk by
Peripheral venous access can become
having a platelet count and coagulation
increasingly difficult in young children.
studies checked preoperatively. External
Additionally, peripheral administration of
right atrial catheters are typically inserted
certain antineoplastic drugs may cause
into the subclavian, internal jugular, or
injury due to extravasation. CVCs provide
external jugular vein and are tunneled to
a safe, accessible route for infusion of che-
an exit site on the anterior or lateral chest.
motherapy, total parenteral nutrition, blood
Placement in other locations may be neces-
products, antibiotics, pain medications,
sary for patients with a history of throm-
hematopoietic stem cells, and other medi-
bosis, multiple previous CVCs, or in the
cations and infusions. These devices also
patient with compression of the upper
allow for frequent and convenient blood
venous system due to tumor. The location
sampling, which can be taught to home care
of the catheter tip should be confirmed by
providers. CVCs are now an integral aspect
fluoroscopy or chest radiography to be at
of management of cancer or chronic illness
the junction of the superior or inferior vena
in children.
cava and the right atrium prior to the
The optimal type and timing of place-
catheter being used. Ultrasonography or
ment of a CVC are not standardized. Many
angiography may assist the surgeon in
types of CVCs are available and are divided
determining the most accessible vein for
between temporary (peripherally inserted
CVC placement. External CVCs have a
central catheters) and more permanent,
Dacron cuff attached to the catheter wall
tunneled catheters. Tunneled catheters can
that is positioned 1-2 cm from the skin exit
either be entirely under the skin or have an
site. This allows for the growth of fibrous
external portion that allows access. This
tissue (scar) into the cuff, preventing migra-
chapter focuses on the permanent catheters,
tion and loss of the line. Surgeons will
placed to provide access for months to years.
typically place sutures externally as well that
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
236
Chapter 26
may be removed after healing. Totally
collection early in their course of therapy
implanted catheters (ports) are secured to
may benefit from the placement of a phere-
the deep fascia on the anterior chest wall
sible catheter, such as a Medcomp. These
with sutures. Ports are made of nonimmu-
lines require meticulous maintenance and
nogenic materials and some now allow for
are not repairable, unlike the standard
high-pressure infusion of contrast for radio-
external tunneled catheters. Due to the
graphic imaging. The silicone membrane of
larger lumen size required for a pheresible
the reservoir is accessed by a special non-
catheter and associated risk of thrombosis,
coring needle (Huber) and is self-sealing
these lines are not always feasible for long-
with needle removal. Topical anesthesia is
term access in smaller children. The Bard
typically applied over the reservoir prior to
PowerLine is becoming a popular double
internal catheter access.
lumen pheresible catheter due to the higher
The most frequently used tunneled
flow rates that are possible through them. In
external catheters are Broviac, Hickman,
addition, they can frequently be placed in
and Leonard. Totally implantable catheters
children who are too small to safely receive a
include Medi-Port, Port-a-Cath, BardPort,
conventional double lumen catheter (see
and PowerPort. Single lumen external
Table 26.1).
catheters and ports are used most com-
The decision about the type of CVC to be
monly in children, but double and triple
placed is based on multiple factors including
lumen catheters and ports are available.
length and intensity of therapy; frequency of
Patients that require pheresis for stem cell
lab draws; the need for multiple lumens to
Table 26.1 Common tunneled central venous catheters and related care.
Catheter type Saline flush
Heparin
Site care
Hickman
10 kg: 5 mL/lumen
10 units/mL
Dressing change weekly
Broviac
>10 kg: 10 mL/lumen
2 mL q24 hours
or per institution policy
after access
PRN after catheter
(or when wet, dirty)
access or blood
Type of dressing per
draw
institution (tegaderm,
Mepore, etc.)
Pheresible
10 mL each lumen
100 units/mL
Dressing change weekly or
catheter
q24 hours
>30 kg: 2 mL/lumen
per institution policy
15-30 kg: 1.5 mL/lumen
(or when wet, dirty)
<15 kg: 1 mL/lumen
Do not use iodine or
q24 hours and after
iodine-based disinfectants
access
Mediport
10 mL q24 hours and
10 units/mL
Huber needle changed
PRN when accessed
2 mL q24 hours and
weekly if continuously
Flush 20 mL after
PRN while accessed
accessed; dress securely
blood draw
100 units/mL
when accessed; apply
2 mL with deaccess
topical anesthetic prior to
and monthly
port access
<10 kg, give 2 mL of 10 units/mL.
Central Venous Catheters
237
Table 26.2 Type of central venous catheter to be placed by diagnosis.
CVC type
Number of lumens
Diagnosis
External
Single
ALL, high risk
Solid tumors, not metastatic
Neuroblastoma, low and intermediate risk
Non-Hodgkin lymphoma
External
Double
AML
Neuroblastoma, advanced stage
Brain tumors requiring Head Start/HSCT
Solid tumors, advanced stage
Internal
Single
ALL, standard and high risk
Wilms tumor
Hodgkin and non-Hodgkin lymphoma
Malignant glioma
Abbreviations: ALL, acute lymphoblastic leukemia; AML, acute myelogenous leukemia; CVC,
central venous catheter; HSCT, hematopoietic stem cell transplantation.
support simultaneous administration of
potential for introduction of infection. Ulti-
chemotherapy, hydration, pain medication,
mately, in cases where an external catheter is
parenteral nutrition, antibiotics, and other
not essential, the decision on the type of
agents; age; and lifestyle. In general, external
catheter to utilize must include a discussion
and multilumen devices are associated with
with the patient and the family reviewing
greater risk, primarily due to an increased
the pros and cons of each type of device.
incidence of infection and thrombosis.
Table 26.2 summarizes the type of CVC to
Ports are becoming increasingly more pop-
consider based on diagnosis and subsequent
ular, especially with lower intensity thera-
therapy intensity.
pies and in older children and adolescents.
Ports are cosmetically more satisfactory, do
not require routine care, and allow for easy
Maintenance
bathing and swimming. However, ports
may be difficult to place and access in obese
External catheters require daily heparin
or very thin patients. Additionally, the use
flushes and periodic dressing changes
of ports for routine blood draws is limited
(see Table 26.1). Families require detailed
due to their need to be accessed prior to use.
education on the care and potential com-
An external CVC should be used in patients
plications of CVCs. They should be taught
requiring frequent laboratory monitoring.
sterile technique to access the CVC for
External CVCs and ports may be used
blood draws or flushes and to provide exter-
immediately after placement. In the case
nal care. Frequency of dressing changes may
of a port, the surgeon should be asked to
be dependent on institution policy, type of
access the device when it is placed, prior
CVC, and local factors such as a wet, dirty,
to development of postoperative edema.
or loose dressing. At all times, the CVC exit
This allows immediate use, decreasing
site should be kept dry and clean. Patients
patient discomfort and reducing the
are instructed to protect it during bathing
238
Chapter 26
and not immerse the dressing or exit site in a
colonization and consequent seeding of the
bath, hot tub, or pool. Should this occur, the
pathogen into the circulation (see Chapter 27
dressing should be changed immediately
for common pathogens and management of
with careful cleansing of the exit site. Ports
fever in patients with CVCs). Patients with
require less care, and heparin flushes, which
CVCs require constant attention for pre-
are required only monthly, are typically per-
vention and early recognition of infection.
formed by the health care team. No dressing
Despite the meticulous techniques used by
is needed for implantable devices when they
health care providers and caregivers to main-
are not accessed. Each institution has a stan-
tain sterility, many children with CVCs will
dardized approach to the care and mainte-
develop a line infection or bacteremia. Infec-
nance of CVCs that is followed by staff, family
tion is often related to organisms that
members, and home health care agencies.
are ubiquitous but can lead to true infection
in the immunosuppressed patient (such as
gut and skin flora), or be due to organisms
Complications: mechanical
introduced with venous access or with blood
product or medication/fluid administration.
The use of CVCs can result in a number of
Coagulase-negative staphylococci, Staphylo-
short- and long-term complications. Imme-
coccus aureus, aerobic Gram-negative bacilli,
diate complications related to CVC placement
and Candida albicans most commonly cause
include malposition, hemorrhage, pneumo-
catheter-related bloodstream infection. Addi-
thorax, chylothorax, arterial cannulation, car-
tionally, development of a thrombus in the
diac dysrhythmia, and failure to place the line.
catheter may serve as a nidus for infection.
Rarely, complications may occur due to the
Occasionally, despite best efforts and appro-
anesthetic required for surgical placement.
priate antibiotic therapy, the line may be
Long-term complications include mechanical
colonized and need to be removed. Recent
failure, catheter breakage, leakage, and port
techniques for catheter sterilization include
extrusion. Mechanical failures have been
antibiotic and ethanol locks. Additionally,
reported in up to 10% of patients and may
many manufacturers have created antibacte-
result in removal of the CVC. Some types of
rial substances bonded to the catheters.
external CVCs may be repaired if the break is
Risk factors for CVC infection include
distal to the catheter exit site. Children with
the placement of an external catheter, young
implanted ports are at risk for the Huber
patient age, multilumen catheter, early
needle becoming dislodged during infusion,
placement (i.e., within 2 weeks of diagnosis
leading to extravasation with resultant skin
of acute lymphoblastic leukemia
[ALL]),
irritation or breakdown. These require careful
stem cell transplant, solid tumor, lack of
periodic assessment.
perioperative prophylactic antibiotics, high-
intensity protocol, and placement during
periods of neutropenia. These factors must
Complications: infectious
be weighed against the benefits of early line
placement, type of line placed, and number
Infection is the most common complication of
of catheter lumens. The relatively higher
CVCs. Most catheter-related infectionsarise by
infection rate seen with external catheters
one of two mechanisms: (1) infection at the
favors the use of implanted catheters when
exit site with migration of the pathogen along
possible.
the external catheter surface, and (2) contam-
Approximately 50% of catheter-related
ination of the hub, leading to intraluminal
infections occur locally at the exit site, along
Central Venous Catheters
239
the tunneled catheter or as a “pocket” infec-
review meticulous local CVC management
tion around the implanted reservoir. Skin
with the caregivers.
irritation may occur with external CVCs
In addition to site infections, coloniza-
and requires alteration of the prescribed exit
tion of the catheter may occur. In this case,
site care and dressing to avoid repeated
the patient will have repeatedly positive
irritation, skin breakdown, infection, and
cultures with the same organism, often
pain. Patients with local catheter infections
without systemic symptoms of infection.
typically have local signs and symptoms
In cases of colonization, cultures may
with tenderness on palpation, erythema,
become quickly positive and may persist
purulent drainage, lack of healing, swelling,
through appropriate antibiotic therapy. At
and pain. Severely neutropenic patients
times, catheters may be reseeded from dis-
may only have pain and tenderness as these
tant sites of occult infection (e.g., cardiac
symptoms do not require the presence of
vegetations, osteomyelitis, deep abscesses).
neutrophils. If drainage is present, the site
The CVC should always be considered
should be cultured. Tunnel infections are
the source of infection in a patient who
characterized by a spreading cellulitis along
presents with fever until proven otherwise.
the subcutaneous tract of long-term cathe-
Fever, chills, or hypotension temporally
ters. The catheter should be removed if the
related to line flushing increase the likeli-
administration of appropriate parenteral
hood of a line infection. Broad-spectrum
antibiotics does not result in resolution of
empiric antibiotics should be administered
the signs and symptoms within 24 to 48
after drawing cultures from each lumen of
hours. When evaluating a child with fever
the CVC. Persistent bacteremia despite
and a CVC, palpate along the tunnel to
appropriate antibiotic coverage
(>72
assess for tenderness or to express drainage.
hours), signs of sepsis (hypotension, chills,
Pocket abscesses may need to be surgically
persistent fever, cool extremities, delayed
drained and packed and almost always
capillary refill), or infection with fungus
will require removal of the reservoir and
or water-borne bacteria (e.g., pseudomonas,
catheter. Site infections with minor exit site
stenotrophomonas) warrant CVC removal.
erythema and tenderness are treated with
If placement of a new CVC is deemed
topical antibiotics and monitored with care-
necessary, the patient should first complete
ful, daily assessment; systemic antibiotics
an appropriate course of antibiotic therapy
should be added for clinical worsening, lack
allowing for sufficient healing time and
of improvement, or a positive blood culture
decreasing the risk of subsequent reinfection
from the line. Tunnel and pocket infections
of the new CVC.
should be treated with both topical and
CVC infection may be overdiagnosed,
systemic antibiotics, as well as a daily sterile
resulting in either prolonged antibiotic
dressing change. The patient should be
administration or unnecessary removal of
treated systemically with broad-spectrum
the catheter. Differential time to positivity
antibiotics providing good Gram-positive
is a reliable diagnostic technique to differ-
coverage and be monitored for the devel-
entiate a true line infection from bacter-
opment of an abscess requiring surgical
emia. Paired blood samples (aerobic and
drainage. The antibiotic regimen may be
anaerobic) from a peripheral vein and the
altered with culture results, but if the patient
central catheter are obtained and compared
is neutropenic it should include both broad-
with respect to time to positivity. If the
spectrum and directed antibiotic therapy.
culture from the catheter turns positive
Site infections provide an opportunity to
2 or more hours before the peripheral
240
Chapter 26
culture, this is diagnostic of a catheter-
is not currently recommended as standard
related infection. A positive differential
practice; however, it may be warranted in
time to positivity may not change manage-
certain situations with documented
ment, although consideration should be
increased risk.
given to the type and duration of systemic
Thrombosis should be suspected in
antibiotic therapy, utility of an antimicro-
patients whose catheter does not flush or
bial or ethanol lock, and the need for cath-
draw easily or in the event of suggestive
eter removal. Additional factors include
clinical findings (pain or swelling in extrem-
the type of catheter, prior history of infec-
ity, prominent vasculature or swelling on
tion, current neutrophil count, likelihood
side of neck/chest with CVC, inability to
of short term recovery, and current che-
successfully access an implanted port). It
motherapeutic regimen. When a specific
should be noted that a line that does not
pathogen has been identified, antibiotic
draw or flush might be occluded by the vessel
therapy should be targeted, with additional
wall or a valve. Patients suspected of having
broad empiric coverage in the neutropenic
a thrombosis should undergo imaging to
patient (see Chapter 27). The possibility
confirm this and determine the extent
of an infected catheter-related thrombus
(see Figure 26.1). Based on the timing of
should be considered, and treatment may
line dysfunction or symptom development,
also involve thrombolytic agents or
the age of the clot can be approximated.
anticoagulation.
Additionally, pertinent family history of
thrombosis should be sought out to help
guide the necessity of genetic evaluation for
Assessment and management of
thrombophilia
(see Chapter
10). Finally,
catheter-related thrombosis
environmental and medication prothrombo-
tic factors should be considered (e.g., immo-
Venous thromboembolism is another
bilization, steroids, and other medications).
common complication of long-term CVCs.
Treatment of thrombosis may require
The extent of thrombosis may include the
thrombolytic therapy, anticoagulation, or
catheter tip (ball-valve clot), the length of
catheter removal. Below are guidelines for
the catheter (fibrin sheath), or the cathe-
the assessment and treatment of a suspected
terized vessel (e.g., the upper limb with or
CVC-associated thrombus:
without the central vasculature of the
* Examine the catheter for any kinks in the
neck or mediastinum). Catheter-related
tubing.
venous thromboembolism can result in
* Reposition the patient; flush the CVC
significant morbidity, and subclinical
with normal saline (if possible) and again
thrombosis may occur in up to 50% of
attempt to withdraw blood. Have the
patients with CVCs. The risk of thrombosis
patient hold their hands above their head,
is influenced by catheter location, insertion
turn, cough, or hold their breath.
technique, catheter kinking or compres-
* Attempt a tissue plasminogen activator
sion by a mass or other structure, ratio
(TPA) dwell if the catheter can be flushed:
of the catheter size to the intraluminal
* Infants
<3 months receive
0.25 mg
vessel diameter, blood flow rheology,
(0.5 mL) per lumen; older children receive
genetic predisposition, and administration
0.5 mg (1 mL) per lumen. Dwell should be
of prothrombotic medications or infusions
administered for 30 minutes.
(e.g., asparaginase and total parenteral
* After the first dwell, attach an empty 3 mL
nutrition). Primary thromboprophylaxis
syringe and attempt to withdraw blood. If
Central Venous Catheters
241
Ini al evalua on
Physical examina on, including assessment of malfunc oning CVC
History (prothrombo c medica ons, family history)
Laboratory assessment (fibrinogen, PT, PTT, CBC)
Diagnos c imaging for confirma on of TE:
Ultrasound with Doppler and/or
CT with venography and/or
Catheter dye study
Is CVC essen al for care?
no
yes
Thrombophilia risk assessment (see
Occlusive: ini ate thrombolysis, then
Chapter 10)
an coagula on
Nonocclusive: an coagula on
Occlusive: ini ate thrombolysis up to 48-72 hours; an coagula on
Nonocclusive: consider thrombolysis up to 48-72 hours;
an coagula on
Func onal CVC?
no
yes
Successful lysis
Residual thrombus
of TE
Con nue therapeu c
Remove CVC
an coagula on
Con nue therapeu c an coagula on
Consider CVC removal
if s ll with residual clot un l clot
dissolu on
Con nue prophylac c an coagula on
Therapeu c an coagula on for 3 months
for 6 weeks to 3 months a er clot
followed by prophylaxis un l CVC
resolu on
removed
Figure 26.1 Evaluation of suspected thromboembolism in patients with tunneled central venous
catheters. Abbreviations: CVC, central venous catheter; PT, prothrombin time; PTT, partial throm-
boplastin time; CBC, complete blood count; TE, thromboembolism; CT, computed tomography.
successful, obtain blood work and flush line
* Obtain further imaging studies
per protocol. If unsuccessful, instill a second
such as a dye study of the line, ultra-
TPA dwell for an additional 30 minutes.
sound with Doppler flow, or CT with
* If the TPA dwell is unsuccessful, further
venography to evaluate for thrombosis
workup is required to determine the extent
location and extent. Assess for sleeve
of thrombus.
thrombus, mechanical failure of the
* Obtain a chest radiograph to confirm
CVC, or migration of the catheter out-
appropriate line position.
side the vessel.
242
Chapter 26
If a thrombus is identified and the time
genetic predisposition to thrombosis based
from symptom onset is <14 days, throm-
on family history or a personal history of
bolytic therapy with low-dose systemic TPA
previous thromboembolism, a thrombophi-
should be attempted if the catheter can be
lia trait workup should be sent (see Chapter
flushed
10 for details). If the patient has multitrait
* Infants < 3 months receive 0.06 mg/kg/
thrombophilia and therefore a higher risk of
h for 6 to 24 hours; older children receive
rethrombosis, every effort should be made to
0.03 mg/kg/h. If no clinical improvement
remove the CVC once the risk of having an
in 24 hours, double the dose to 0.06-
indwelling line outweighs the benefit (i.e.,
0.12 mg/kg/h (max dose 2 mg/h).
maintenance therapy in the ALL patient).
* Systemic TPA can be given for up to
If the patient on prophylactic anticoagula-
96 hours.
tion develops rethrombosis, management
* Monitor the fibrinogen level and main-
should be the same as with the initial clot with
tain above 100 mg/dL with cryoprecipitate.
systemic low-dose TPA followed by therapeu-
* Monitor the patient for signs of sepsis as
tic anticoagulation with LMWH until clot
bacteria can be released into the blood-
dissolution. If the patient is on therapeutic
stream with dissolution of the thrombus.
anticoagulation with clot extension, the CVC
* If the catheter becomes functional after
will need to be removed. In cases where the clot
administration of systemic TPA, obtain a
cannot be dissolved or stabilized and the line
follow-up radiographic study to assess for
needs to be removed, 3 to 5 days of antic-
clot resolution.
oagulation is recommended for clot stabiliza-
* Remove the catheter if the thrombus
tion prior to catheter removal. Special circum-
cannot be cleared after 24 to 48 hours.
stances will warrant disruption in anticoagula-
tion, such as lumbar punctures to administer
If the thrombus dissolves, or is stabilized
intrathecal chemotherapy or other surgical
with normal CVC function, determination
procedures. In general, LMWH should be held
should be made as to the current necessity of
for 24 hours in advance of theseprocedures and
the line and risk for recurrent thrombosis.
resumed 12 to 24 hours after procedure com-
If the CVC can be safely removed, some
pletion, depending on the nature of the pro-
practitioners recommend short-term pro-
cedure and risk of postoperative bleeding.
phylactic anticoagulation to mitigate the
Additionally, LMWH should be held during
risk for rethrombosis (usually 6 weeks to 3
periods of moderate thrombocytopenia (i.e.,
months of low-molecular-weight heparin
<50
109/L) to minimize additional risk
[LMWH]). If the line needs to be maintained
of bleeding. If the CVC is removed and it is
and the clot has resolved, the patient should
necessarytoplaceanewcatheter,consideration
continue on therapeutic LMWH for
3
should be given to anticoagulation prophylaxis
months and then switch over to prophylactic
to prevent recurrence of thrombosis.
LMWH until the line is removed. For the
patient with a stable clot and a functional
Suggested Reading
CVC, therapeutic anticoagulation with
LMWH should be continued until approx-
Gallieni M, Pittiruti M, Biffi R. Vascular access in
imately 3 months after clot dissolution and
oncology patients. CA Cancer J Clin
then switched to prophylactic anticoagula-
58:323-346, 2008.
tion until line removal (see Chapter 10 and
Hirsh J, Guyatt G, Albers GW, et al. Antithrom-
Formulary for dosing guidelines). Addition-
botic and thrombolytic therapy: American
ally, if the patient is determined to have a
College of Chest Physicians evidence-based
Central Venous Catheters
243
clinical practice guidelines
(8th edition).
optimal type and timing of placement. J Clin
Chest 133:71S-109S, 2008.
Oncol 23:3024-3029, 2005.
McLean TW, Fisher CJ, Snively BM, Chauvenet
Shivakumar SP, Anderson DR, Couban S. Catheter-
AR. Central venous catheters in children with
related thrombosis in patients with malignancy.
lesser risk acute lymphoblastic leukemia:
J Clin Oncol 27:4858-4864, 2009.
Management
27
of Fever in the Child
with Cancer
Children with cancer are at an increased
* Defects in cellular immunity such as seen
risk for serious bacterial, viral, and fungal
in patients with T-cell malignancies or those
infections. Many factors contribute to this
receiving steroids or radiation; persistence
susceptibility in immunocompromised
of lymphopenia due to chemotherapy.
children. The two most important deter-
* Colonization from endogenous micro-
minants of susceptibility to bacterial and
flora.
fungal infection are the number of circu-
* Presence of indwelling catheters or shunts.
lating neutrophils
(absolute neutrophil
The ANC is calculated by multiplying the
count; ANC) and the duration of severe
percentage of neutrophils (segmented neu-
neutropenia. Other factors include:
trophils þ bands) by the total white blood
* Underlying disease: leukemia, especially
cell (WBC) count.
acute myelogenous leukemia
(AML),
Example: WBC ¼ 1.0
109/L, segmented
advanced stage lymphoma, or other meta-
neutrophils ¼ 10%, bands ¼ 10%
static solid tumors are at higher risk due to
ANC ¼ 20%
1.0
109/L ¼ 0.2
109/L
increased therapy intensity.
* Remission status: patients not in remis-
ANC<1.5
109/L is defined as neutropenia
sion are at higher risk.
ANC < 1.0
109/L is defined as moderate
* Type of therapy with dose- or time-inten-
neutropenia
sive therapies: administration of high-dose
ANC < 0.5
109/L represents severe neu-
cytarabine and stem cell transplant confer
tropenia
the highest risk.
ANC < 0.1 to 0.2
109/L represents pro-
* Nutritional status: malnutrition adversely
found neutropenia
affects immune function.
* Disruption of protective barriers such as
In general, patients with an ANC of less than
skin, mucocutaneous tissues, gastrointesti-
0.5
109/L or those with dropping counts
nal tract, and exit sites of catheters.
after chemotherapy are considered severely
* Defects in humoral immunity related to
neutropenic. The ANC at time of presenta-
therapy, impaired splenic function, or B-cell
tion with respect to the most recent chemo-
malignancies.
therapy gives important information. For
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Management of Fever in the Child with Cancer
245
instance, if a child is neutropenic without
may have fever as their only symptom of
recent myelosuppressive therapy, infection
infection. It should be noted that neutro-
(and less likely relapse of a hematologic
penia will not prevent the patient from
malignancy) should be considered as the
having a temperature spike with infection;
most likely etiology due to marrow suppres-
one exception is the patient on steroids
sion. In general, the expected nadir in ANC
(such as in acute lymphoblastic leukemia
occurs 7 to 14 days from the start of mye-
[ALL] induction) who may not mount a
losuppressive chemotherapy. This timing
fever with an underlying infection. A careful
should be considered when assessing the
clinical examination is critical as the
patient and determining the management
patient’s status may change quickly and
plan. Practitioners may also utilize the abso-
dramatically.
lute phagocyte count (APC), which is the
Septic shock results from overwhelming
summation of the ANC and absolute mono-
infection with microorganisms in the
cyte count, in determining the risk of bac-
blood leading to central vascular dilation
terial infection as monocytes also possess
with resultant circulatory failure and inad-
bacteria-fighting ability. An increasing
equate tissue perfusion. Symptoms of septic
monocyte count is often seen prior to and
shock include hypotension, tachycardia,
often heralds neutrophil recovery.
tachypnea, clammy extremities (although
Fever is defined as a single temperature
can be warm with initial sepsis), decreased
of 38.3 C (101.0 F) or greater, two tem-
urine output, and deterioration of mental
peratures of
38.0
to
38.2 C
(100.4
to
status. Although septic shock can be seen
100.9 F) within a
24-hour period, or a
with any organism, the usual culprits with
temperature of 38.0 to 38.2 C (100.4 to
acute and overwhelming sepsis are Gram-
100.9 F) persistently for 1 hour, taken in
negative rods.
the axilla, orally, or by tympanic probe.
The most common organisms causing
Generally, oral and tympanic temperatures
bacteremia and sepsis are:
are preferred since the axillary temperature
* Gram-positive: Staphylococci (coagulase-
can be affected by outside temperature. Oral
negative, Staphylococcus aureus including
temperatures preferably are taken without
methicillin-resistant [MRSA]), streptococci
any recent hot or cold oral intake. Once a
(a-hemolytic) including Streptococcus viri-
fever is documented, it should be consid-
dans and S. mitis
ered as real no matter what the underlying
* Gram-negative: Enterobacteriaceae (E. coli,
circumstances are, even if the patient defer-
Enterobacter, Klebsiella, Serratia), Pseudomo-
vesces without any intervention. Patients
nas aeruginosa, Stenotrophomonas maltophi-
who develop a low-grade fever (38.0 C to
lia, Acinetobacter sp.
38.2 C) should be monitored without anti-
* Anaerobic: Clostridium difficile, Bacter-
pyretics to determine if a fever spike will
oides sp., Proprionobacterium acnes
occur. Rectal temperatures should never be
Other pathogens infecting cancer
taken in potentially neutropenic children.
patients include:
* Fungi: Candida sp., Aspergillus sp., zygo-
mycetes, cryptococci
Fever and neutropenia
* Viruses: Herpes simplex virus
(HSV),
varicella zoster virus (VZV), cytomegalovi-
Every oncology patient who presents with
rus (CMV), Epstein-Barr virus, respiratory
febrile neutropenia (FN) is considered sep-
syncytial virus, adenovirus, influenza, para-
tic until proven otherwise. These children
influenza, human herpesvirus 6
246
Chapter 27
Other: Pneumocystis jiroveci, Toxoplasma
a. Determination of vitals and evalua-
gondii, Strongyloides stercoralis, cryptospo-
tion for shock.
ridium, Bacillus sp., atypical mycobacterium
b. Meticulous physical examination with
particular attention to sites of occult
infection such as the skin, exit sites of
Initial evaluation of febrile
catheters, sites of recent bone marrow
neutropenia
aspiration, oral cavity, and perianal areas.
See Figure 27.1.
Even subtle evidence of inflammation
1. History and physical examination
(faint erythema, tenderness, or minimal
Fever: Temperature 38.3°C x 1 or 38.0°C x 2 at least 1 hour apart
Neutropenia: ANC <0.5 x 109/L or <1.0 x 109/L and falling
Ini al Evalua on
History: Chills, rigors, fever within 1 hour of CVC flush
Recent immunosuppressive therapy
Symptoms of AGE, URI, pain
Physical examina on: Vitals, CVC exit site(s), skin/mucosa, perineum
Laboratory assessment: CBC with differen al
Blood culture from each CVC lumen (peripheral, see text)
Culture from all suspicious sites (throat, skin, stool, catheter site)
Urinalysis and urine culture
Stool culture, C. difficile toxin, rotavirus for diarrhea/abdominal pain
Electrolytes, BUN, crea nine, liver transaminases
Imaging: CXR if signs/symptoms of pulmonary process
CT as necessary per physical findings (sinuses, chest, abdomen, pelvis)
Risk group assessment
High risk
Low risk
Any malignancy not controlled (i.e., relapse, refractory, induc on)
Diagnosis: ALL, NHL, solid tumors in remission
AML, high risk ALL in consolida on or delayed intensifica on
Neutropenia: Dura on <7 days
High dose cytarabine
Nontoxic appearance
Prolonged neutropenia an cipated ≥7 days
No focal infec on, mucosi s, diarrhea
Profound neutropenia <0.1 x 109/L
Toxic appearance: Hypotensive, rigors, shock, tachypnea, hypoxia
Evidence of infec on: Pneumonia, celluli s, abdominal pain and
diarrhea, neurologic (mental status) changes
Known coloniza on with MRSA
Prior history of bacteremia/sepsis
Empiric an bio c therapy; monotherapy
Mucosi s following chemotherapy
Consider outpa ent management
Empiric therapy, broad spectrum, ini ated promptly
Hospitaliza on
Treatment of co-morbidi es as appropriate
Pain management as appropriate
Directed therapy as appropriate:
Vancomycin for recent high dose cytarabine, new fever in AML pa ent
Clindamycin or meropenem for diarrhea/abdominal pain
Figure 27.1
Evaluation and initial management of febrile neutropenia in the child with cancer.
(Abbreviations: CVC, central venous catheter; AGE, acute gastroenteritis; URI, upper respiratory
infection; CBC, complete blood count; BUN, blood urea nitrogen; CXR, chest radiography; CT,
computed tomography; ALL, acute lymphoblastic leukemia; NHL, non-Hodgkin lymphoma; AML,
acute myelogenous leukemia; MRSA, methicillin-resistant Staphylococcus aureus).
Management of Fever in the Child with Cancer
247
discharge) may represent a source of
i. If diarrhea is present, send a stool
infection as neutropenia diminishes clas-
sample for stool culture, rotavirus, and
sic inflammatory changes. Pain, however,
C. difficile antigen.
is always a concerning finding and should
j. In contrast to nononcology patients, a
be considered a potential clue to site and
lumbar puncture (LP) is not routinely
source of infection.
done as part of a serious bacterial infec-
2.
Laboratory evaluation
tion evaluation in patients with FN and
a. Complete blood count (CBC) with
an oncologic diagnosis. If an LP appears
manual differential to determine ANC.
clinically indicated, evaluation for
b. Blood cultures from each lumen of the
increased intracranial pressure with
central venous catheter (CVC); peripheral
appropriate imaging
(i.e., head CT)
culture is necessary if the child does not
should be done and the pediatric oncol-
have a central line or it is unobtainable
ogy attending notified prior to the pro-
from the central line. Controversy still
cedure. An adequate platelet count and
exists as to utility of a peripheral blood
at least near-normal coagulation studies
culture when cultures are taken from the
are also required in advance of the LP.
line as delayed time to positivity between
k. If another implanted device is present
these two sources can help determine the
(e.g., ventriculoperitoneal shunt or
existence of a line infection versus bac-
Ommaya reservoir), do not attempt to
teremia (see Chapter 26). Do not flush
obtain a culture without speaking to
the catheter after labs are obtained in
the appropriate attending physicians.
the patient with a recent history of high
Intervention by a neurosurgeon may
fever, hypotension, chills, or rigors within
be required. It is uncommon for these
1 hour of flushing the catheter, suggestive
devices to be the source of infection if
of a line infection. See below under Initial
it has been more than 2 to 4 weeks since
management.
they were inserted.
c. Serum chemistries to include electro-
lytes, liver, and renal function studies.
Initial management
d. Urinalysis and urine culture (no cath-
Risk stratification models for pediatric
eter sample for neutropenic patients).
oncology patients with FN to determine
e. Chest radiograph only if the patient
which patients are low risk and could poten-
has ausculatory signs or symptoms of
tially be managed as outpatients and which
infection or respiratory compromise.
are higher risk and should be monitored as
f. If the child has tenderness over the
inpatients have yet to be validated and
sinuses, perform diagnostic imaging to
widely accepted. Adult studies suggest that
evaluate for sinusitis (plain radiographs
patients with predicted time of severe neu-
and/or CT).
tropenia of
7 days and no concerning
g. Patients with symptoms of esophagi-
clinical signs including focal infection such
tis should be evaluated for viral or fungal
as pneumonia, new abdominal pain, or
causes with serologies and culture, if
hypotension can be safely managed as out-
possible.
patients. With the lack of data in pediatric
h. Cultures and bacterial Gram stain or
patients, we continue to recommend that
fungal stains from suspicious sites such
parenteral broad-spectrum antibiotics be
as the oropharynx, skin breakdown sites,
started immediately and the patient admit-
and catheter sites.
ted to the hospital. FN is considered an
248
Chapter 27
oncologic emergency and all measures
combination with broad-spectrum coverage
should be taken to ensure prompt evaluation
due to the high risk of S. viridans infec-
and treatment, with a goal of
60 minutes
tion with its accompanying septic shock
between presentation and the initiation of
and acute respiratory distress syndrome.
antibiotics. Antibiotics should be chosen
Cytarabine is known to alter mucosal integ-
based on microbial prevalence and antibi-
rity and allow entry of this organism.
otic sensitivity patterns at each institution.
Patients with S. viridans sepsis have a high
In general, due to the more acute risk from
rate of morbidity and mortality. They
Gram-negative organisms, broad coverage
typically require prolonged hospitalization
for these organisms (including Pseudomo-
following chemotherapy until neutrophil
nas) should be initiated. Many combina-
recovery occurs to allow emergent initiation
tions of antibiotics are effective and accept-
of treatment in the event of fever or other
able regimens include:
signs of impending shock or sepsis. Other
1. Monotherapy:
indications for vancomycin in the empiric
Fourth generation cephalosporin (anti-
regimen include:
pseudomonal b-lactam)
* Presentation with hypotension or other
Cefepime 150 mg/kg/day intravenous
evidence of shock
(IV) divided q8h
* Mucositis due to chemotherapy
Carbapenem
* Patients with prior history of a-hemolytic
Imipenem 50 mg/kg/day IV divided
streptococcus bacteremia
q6-8h
* Catheter site infection or other skin
Meropenem 60 to 120 mg/kg/day IV
breakdown
divided q8h
* Patients colonized with resistant organ-
Piperacillin/Tazobactam
isms treatable only with vancomycin
Zosyn
240
to
300 mg/kg/day IV
* Vegetations on echocardiogram
divided q8h
2. Dual therapy (antipseudomonal b-lac-
Anaerobic therapy should be considered in
patients who have significant mucosal
tam plus an aminoglycoside):
breakdown, perianal skin breakdown, peri-
Ceftazidime
100 to 150 mg/kg/day IV
toneal signs, or in cases of typhilitis with
divided q8h and
presumed or proven C. difficile infection.
Tobramycin 7.5 mg/kg/day IV divided
For patients on dual therapy with an
q8h or 7-9 mg/kg/dose IV daily
aminoglycoside, trough levels should be
Other antibiotics used in specific circum-
monitored weekly due to the risks of
stances (see below) are:
nephro- and ototoxicity, especially in those
1. Anaerobic drugs:
receiving other nephro- or ototoxic drugs
(e.g., vancomycin, furosemide). For patients
Clindamycin 40 mg/kg/day IV divided
on therapy with vancomycin, trough levels
q6-8h
should be monitored for toxicity as well as
Metronidazole
30 mg/kg/day
(loading
medication effectiveness, with goal trough
dose 15 mg/kg) IV divided q6h
levels of 10 to 15 mcg/mL. Dose increases
2. Vancomycin
60 mg/kg/day IV divided
are often required in pediatric patients to
q8h
reach these levels. Monitor daily electro-
Patients with AML receiving high-dose
lytes, fluid balance, renal function studies,
cytarabine (Ara-C) should have vancomy-
and ensure adequate hydration. Patients
cin as part of their empiric regimen in
with renal dysfunction should receive renal
Management of Fever in the Child with Cancer
249
dosing with more frequent trough levels to
the fever curve, presence of new symptoms or
prevent toxicity.
signs of infection with a meticulous physical
Any patient that presents with hypoten-
examination, and results of prior cultures
sion, chills, rigors, or fever immediately
and diagnostic imaging performed
(see
following, or within
1
hour, of central
Figure 27.2). The status of catheters should
venous catheter flushing should be seen
be assessed (site, function, and concern for
emergently due to the risk of bacteremia
thrombosis). Daily blood cultures and a CBC
and septic shock. In our practice, we elect
with differential should be done on persis-
not to use the central venous line for IV
tently febrile patients. Determination of the
fluids or medication administration initially
ANC is also important for patients who have
in this situation due to the risk of infusing
defervesced and have negative cultures to
a large amount of bacteria into the blood-
determine the length of appropriate therapy.
stream and worsening the signs of clinical
When the neutropenic child becomes afebrile
sepsis. Patients should have central catheter
(no fever for 24 hours) and cultures are
cultures, placement of a peripheral IV
negative
(obtained daily while febrile),
with peripheral cultures, administration
broad-spectrum coverage should be contin-
of broad-spectrum antibiotics, and a fluid
ued until there is evidence of adequate mar-
bolus (e.g., NS/LR 20 mL/kg), if necessary.
row recovery and an appropriate period of
The central catheter should not be flushed
therapy for any initially positive cultures has
or utilized until the patient improves clin-
been provided. Resolution of neutropenia is
ically with defervescence, resolution of
defined as an ANC of >0.5
109/L and
hypotension, and negative follow-up blood
evidence of bone marrow recovery is an ANC
cultures. If the line cannot be flushed, a TPA
of >0.2
109/L and rising on 2 consecutive
dwell can be attempted; if unsuccessful, the
days. Some practitioners also use an APC of
line will need to be removed (see Chap-
>0.5
109/L as sufficient evidence of recov-
ter 26). It is advisable to give an IV fluid
ery from neutropenia. When the patient no
bolus prior to reusing the catheter in the
longer has fever, has negative blood cultures,
event the patient again develops symptoms
and has signs of neutrophil recovery, the
of bacteremia with hypotension following
antibiotics may be safely discontinued and
the flush. In the event the initial blood
the child discharged, assuming an appropri-
culture from the catheter is positive, the
ate treatment length for any positive cultures
clinician will need to decide whether to
has been provided. Should the child have any
remove the catheter or attempt to reuse it
local evidence of infection such as diarrhea or
depending on the patient’s clinical condi-
skin breakdown, appropriate therapy should
tion and the type of organism that is grow-
continue, but in the outpatient setting.
ing. Patients that have persistent fever and
Modification should be made to the
Gram-negative organisms may do best with
initial broad-spectrum antibiotic regimen
removal of the line; patients that have defer-
if a source of infection is found to explain
vesced and are looking clinically well can
the initial fever, or the fever persists for
generally have their line reused and ulti-
more than 3 to 7 days. If the source of the
mately salvaged.
infection is a catheter site with accompa-
nying signs or symptoms of inflammation
Modification of initial antibiotic
(discharge, palpable tenderness, local
treatment
abscess), vancomycin should be added for
Patients should be assessed daily for changes
Gram-positive coverage. If the source is
in vital signs, persistence of fever or change in
perianal, gingival, or intra-abdominal,
250
Chapter 27
Empiric therapy
Nega ve cultures 48-72 hours, unknown
site of infec on
Blood or site
cultures posi ve or site
organism suspected
Persistent
Afebrile ≥24 hours
fever/neutropenia
Evidence of neutrophil
Broaden coverage:
Con nue empiric
recovery (ANC ≥0.5 or ≥
Staphylococcal species: add vancomycin
therapy:
0.2 x 109/L and rising on
Fungus: add echinocandin and/or triazole
Reassess every
2 consecu ve days)
Mucosi s/perineal breakdown: add clindamycin or
24 hours with PE
Discon nue an bio cs
meropenem
and cultures
Esophagi s: add acyclovir, consider an fungal
Pulmonary infiltrates: add triazole and/or echinocandin
a er BAL/bronchoscopy
Persistent neutropenia:
Catheter associated bacteremia >72 hours or fungemia:
Con nue an bio cs, un l neutrophil
remove CVC
recovery
If fever recurs, reassess with PE and
cultures, broaden an bio cs, and
consider an fungal therapy
Persistent fever 3-5 days with neutropenia:
Imaging for infec on if with expected
con nua on of neutropenia: CT chest, other
areas based on symptoms
Urine cytospin for hyphae, fungal culture
Con nue broad spectrum an bio cs,
broaden as indicated clinically
Ini ate empiric an fungal therapy with
echinocandin or triazole
Figure
27.2
Ongoing management of fever and neutropenia.
(Abbreviations: ANC, absolute
neutrophil count; PE, physical examination; BAL, bronchoalveolar lavage; CVC, central venous
catheter; CT, computed tomography).
anaerobic coverage should be added to the
immunocompromised patient. Finally,
regimen (clindamycin, metronidazole, or
though rarely seen, MRSA should be a
meropenem).
consideration in the very sick patient.
Vancomycin is the most effective anti-
Vancomycin should be used judiciously due
biotic against skin flora such as coagulase
to emerging resistance patterns such as van-
negative staphylococci (i.e., S. epidermidis)
comycin-resistant enterococcus
(VRE).
as well as mouth flora, most commonly
Vancomycin should not be part of the
streptococcal species (S. viridans, Peptostrep-
empiric antibiotic regimen unless the insti-
tococcus). Vancomycin is also effective
tutional experience and susceptibility pat-
against bacillus nonanthracis species that
terns require it or there are special circum-
can be true pathogens in the
stances (see above).
Management of Fever in the Child with Cancer
251
Protracted fever may signify the presence
* In multilumen devices, the antibiotic
of a second or previously untreated infec-
infusion should be rotated among the
tion. Patients should be reassessed daily for
lumens as infection may not be limited to
new signs or symptoms of infection and
one lumen. If one particular lumen has the
appropriate interventions and studies per-
positive blood culture, it is reasonable to
formed. Antibiotic-induced colitis may
run the majority of the antibiotic through
occur after any antibiotic. C. difficile over-
this lumen.
growth may be asymptomatic, lead to mild
* Daily blood cultures while febrile or if
diarrhea, or may have moderate-to-severe
growth is present
(from each lumen of
diarrhea and abdominal pain. The patient
CVC), until afebrile and negative cultures
may also develop pseudomembranous coli-
for 3 days.
tis with peritoneal signs, mucosal erosions,
* Assess cardiac valves for vegetations with
and bloody diarrhea. Since C. difficile is a
an echocardiogram for repeatedly positive
normal bowel inhabitant, the toxin must
cultures with Gram-positive organisms.
be documented to diagnose this condition.
* If bacteremia persists for more than 3
If oral therapy is possible, vancomycin
days of appropriate therapy, the catheter
(40 mg/kg divided q8h) or metronidazole
should be removed.
(30 mg/kg divided q6h) may be given; in the
patient unable to tolerate oral medications,
For patients with a history of multiple
IV metronidazole (same as PO dose) should
positive cultures or a difficult to eradicate
be given. Abdominal pain may also be due
organism, consideration should be made
to infection with aerobic Gram-negative
for the use of ethanol or vancomycin lock
bacteria, enterococcus (which no cephalo-
therapy to possibly prevent the need for line
sporin covers in vivo), or anaerobes.
removal. Ethanol locks should not be used
If the patient is determined to have a
with polyurethane catheters
(PowerLine,
catheter-associated bacteremia with a spe-
MedComp) as ethanol will damage the
cific organism, broad-spectrum antibiotics
catheter material.
should be continued with addition of anti-
biotics as needed to ensure adequate
Duration of therapy
coverage of the identified organism until
When a pathogen has been identified, its
sensitivity patterns are reported. If daily
antibiotic susceptibility pattern must be
cultures are persistently positive with the
determined and appropriate antibiotic cov-
same organism for 72 hours after initiation
erage provided along with broad-spectrum
of appropriate antibiotics, it will be neces-
coverage until the patient is afebrile and has
sary to remove the catheter. In addition,
evidence of bone marrow recovery. Once
several organisms including S. aureus, Pseu-
the child is afebrile and the ANC recovers,
domonas, B. cereus, Corynebacterium, Myco-
antibiotics can be tailored to the specific
bacterium sp., Stenotrophomonas, and fungi
organism for a 10- to 14-day course, with
usually require line removal. General care
day 1 being the day of the first negative
measures are as follows:
culture.
* Hand washing with clean or sterile gloves
Patients that defervesce but do not have
prior to manipulation of CVC.
recovery of the ANC should continue on
* Monitor CVC site on a daily basis; change
broad-spectrum antibiotics to prevent sec-
CVC dressing weekly.
ondary infection. Some practitioners may
* Assess site for skin infection (including
discontinue antibiotics after 14 days if the
tunnel infection) with any fever.
patient continues to be neutropenic but
252
Chapter 27
afebrile, although our general practice is to
fungal nodules. Fungal cultures should be
continue until count recovery.
sent from the blood and urine as well as
any suspicious skin site. In addition, the
Empiric antifungal therapy
patient should have a serum galactoman-
When FN persists for 4 to 7 days, empiric
nan performed to screen for Aspergillus
treatment should be broadened to include
infection. CT imaging of the chest, as well
fungal prophylaxis. The risk of fungal
as imaging of other symptomatic areas,
infection is directly related to the duration
should be performed. It should be noted
of neutropenia, which is secondary to the
that in the severely neutropenic patient,
severity of chemotherapy or radiation
areas of infection may not be appreciated
cytotoxicity. Thus, patients undergoing
on imaging until neutrophil recovery has
more intensive therapy such as for AML,
occurred. In these cases, the patient will
relapsed ALL, and hematopoietic stem cell
generally have continued or new fevers with
transplantation are at highest risk of an
count recovery that should alert the clini-
invasive fungal infection. The major caus-
cian to the need for repeat CT imaging
ative fungi are Aspergillus and Candida,
which should include the sinuses, chest,
with mortality as high as 30% to 60% with
and abdomen.
documented invasive disease. Fungal infec-
Commonly used antifungal agents for
tions can be difficult to detect due to the
empiric therapy in prolonged FN include:
frequent absence of localizing signs or
1. Echinocandins
symptoms and lack of means of detection
Micafungin
by culture. Prompt diagnostic assessment
and initiation of antifungal empiric therapy
Prophylaxis 1 to 2 mg/kg IV daily
Treatment 3 to 4 mg/kg IV daily
are paramount for improving outcomes in
these patients.
Caspofungin
Anidulafungin
Empiric antifungal therapy prevents
fungal overgrowth in patients with pro-
2. Triazoles
longed neutropenia. It also provides early
Voriconazole
treatment of clinically occult infection.
Posaconazole
Clinical trials have found that antifungal
3. Amphotericin B
treatment of children with persistent or
4. Lipid formulations of amphotericin B
recurring fever reduced morbidity and
mortality from invasive fungal disease. This
The echinocandins have replaced ampho-
is especially true in patients with profound
tericin B as the empiric antifungal agent of
neutropenia not receiving antifungal pro-
choice due to their once daily dosing, sig-
phylaxis. The current recommendation is
nificantly decreased side effect profile, and
that high risk groups (i.e., those expected to
broad coverage. Echinocandins are fungi-
have neutropenia >7 days) should receive
cidal to most Candida species and at least
antifungal prophylaxis during episodes of
fungistatic against Aspergillus species. They
prolonged FN.
also have limited activity against Fusarium
A meticulous physical examination,
species and Zygomycetes. The triazoles are
laboratory assessments, and radiographic
derivatives of fluconazole with broad-spec-
studies in search of deep-seated infections
trum antifungal activity and limited side
are also warranted after this 4 to 7 day time
effects. Of note, the triazoles are fungicidal
frame. Particular attention should be paid
against Aspergillus species. In general, we
to the skin examination as it can reveal
utilize the echinocandins as first-line
Management of Fever in the Child with Cancer
253
empiric therapy and consider adjustment in
and mortality. In transplant patients, pri-
patients that are worsening clinically or have
mary infection or reactivation with cyto-
signs consistent with Aspergillus infection.
megalovirus (CMV) is an important cause
Patients on either an echinocandin or tria-
of interstitial pneumonitis, marrow aplasia,
zole should be monitored closely for hepatic
and other infections. Antiviral drugs are
toxicity; voriconazole may also cause visual
available for a limited number of infections
disturbance that is more common in adult
including herpes simplex virus (HSV), var-
patients. Posaconazole may provide better
icella zoster virus (VZV), CMV, and influ-
coverage against Aspergillus species but is
enza and should be utilized in the treatment
only available as an oral medication and
of immunocompromised patients. Other
clinical trial data are limited in pediatric
patients may also be candidates for prophy-
patients.
laxis
(e.g., solid organ transplant patients
Amphotericin B remains the most widely
and patients with a prior history of infection
active antifungal agent but has generally
on therapy).
been replaced due to its significant toxicity
Patients with documented infection
profile, most notably nephrotoxicity and
with HSV should be treated with IV acy-
infusional toxicity. Fever, chills, nausea,
clovir (30 mg/kg/day if <12 years; 15 mg/
metabolic abnormalities, hypokalemia and
kg/day for
12 years divided TID for 7 to 14
other electrolyte disturbances, and hepatic
days). Infection with VZV is potentially life
toxicity can all occur. In order to treat, and
threatening and therefore chemotherapy
possibly prevent, some of these complica-
should be stopped while treating with IV
tions, several measures are taken. Patients
acyclovir (30 mg/kg/day divided TID for 7
with a history of nephrotoxicity may benefit
to 10 days or until all lesions are crusted and
from receiving pre- and postinfusion NS
no new lesions have appeared for 24 to 48
boluses and diuretics. Premedications
hours). It is important to monitor renal
including acetaminophen, diphenhydra-
function and ensure adequate fluid intake
mine, and meperidine may be given to treat
due to potential nephrotoxicity from
or prevent acute symptoms related to the
acyclovir.
infusion
(fever, chills, rigors, nausea).
Patients with symptomatic primary
Hydrocortisone can also be added to
CMV infection or reactivation can be trea-
address infusion-related toxicity. Use of
ted with ganciclovir
(10 mg/kg/day IV
liposomal formulations of amphotericin B
divided q12h for 2 to 4 weeks until resolu-
(Ambisome and Abelcet) results in signifi-
tion of viremia by polymerase chain reac-
cantly fewer adverse events, but they are
tion). Patients with persistent infection
much more expensive than amphotericin B.
may require longer therapy or may have
resistant disease and require alternate
therapy with foscarnet or cidofovir. Of
Viral infection
note, ganciclovir can be myelosuppressive,
whereas foscarnet and cidofovir are quite
Immunosuppressed children are able to tol-
nephrotoxic. Concomitant therapy with
erate many viral infections without diffi-
intravenous immune globulin (IVIG) may
culty. However, defects in cellular immunity
be beneficial especially in the setting of
predispose them to unusually severe infec-
CMV pneumonitis. Data regarding pro-
tions with certain viruses, particularly the
phylactic therapy in this setting is lacking,
herpes family of viruses. Primary varicella
although once daily dosing can be consid-
infection is associated with high morbidity
ered in patients that complete a course of
254
Chapter 27
antiviral therapy and remain neutropenic
component of
15 mg/kg/day IV divided
or severely immunocompromised.
q8h for 14 to 21 days. If the patient fails
Patients who develop influenza while on
to respond to TMP/SMX within 72 hours,
therapy should be treated with neuramini-
or develops this infection while on prior
dase inhibitors (i.e., oseltamivir for children
TMP/SMX prophylaxis with good compli-
1 year of age, twice daily for 5 days) to
ance, initiate therapy with pentamidine,
reduce symptom duration and severity of
4 mg/kg IV daily. In patients with moderate
disease. Oseltamivir may also be given as
or severe proven infection, concomitant
daily prophylaxis for 5 days to patients that
administration of corticosteroids may be
have been exposed to the virus. If given as
beneficial with IV methylprednisolone or
treatment, it is most effective when given
prednisone given 2 to 4 times per day for
within 48 hours of symptom onset.
5 to 7 days, followed by a taper over 1 to 2
weeks.
Pneumocystis jiroveci
pneumonia
New sites of infection
Pneumocystis jiroveci is now considered an
New or persistent fever and neutropenia
atypical fungus and is a ubiquitous organ-
may be associated with the development
ism that may cause severe or fatal pneumo-
of new sites of infection due to continued
nitis in immunocompromised patients
severe immunosuppression. Particular
(most notably patients with HIV and those
symptoms may provide clues to site and
being treated for cancer). Patients typically
potential pathogen.
develop a rapid onset of fever in association
1. Burning retrosternal pain may be esoph-
with tachypnea and other symptoms of
agitis from Candida sp. or HSV. Cytotoxic
respiratory compromise such as nasal flar-
treatment may also cause severe pain due to
ing and intercostal retractions. Lung sounds
mucosal erosions. Empiric micafungin or
may be clear and typically patients do not
acyclovir, or both, may be needed. Esoph-
have rales. Hypoxemia is evident by pulse
agitis may also be bacterial, especially as a
oximetry and arterial blood gas. Chest radi-
result of Gram-positive aerobes.
ography or CT shows an interstitial pattern.
2. Pulmonary infiltrates may be due to
Without appropriate therapy, patients rap-
resistant bacteria, Pneumocystis, fungi, or
idly progress to respiratory failure.
viruses. Neutropenic patients may not show
Diagnosis is made by demonstrating the
evidence of pulmonary infiltrates due to
organism by Gomori methanamine silver
lack of an appropriate local inflammatory
stain on a sample obtained by induced spu-
response. However, with the recovery of the
tum, bronchoalveolar lavage, percutaneous
ANC, these sites of infection may become
needle biopsy, or open lung biopsy. Patients
evident. If FN has been less than 7 days,
who are at risk and present with the classic
pulmonary infiltrates are very likely to be of
signs and symptoms should be treated while
bacterial origin. If the neutropenia persists
awaiting definitive diagnosis, given the
for greater than 7 days, the patient is at
rapidity of clinical deterioration.
particularly high risk for fungal infection.
For documented or highly suspected
Bronchoalveolar lavage or induced sputum
infection with P. jiroveci, trimethoprim/sul-
may identify Pneumocystis, viruses, or fungi.
famethoxazole (TMP/SMX) is the treatment
The risks and benefits of open lung biopsy
of choice, at a dose based on the TMP
and pursuing an aggressive diagnostic
Management of Fever in the Child with Cancer
255
evaluation must be considered. It is impor-
ings should have appropriate diagnostic
tant to remember that the child may rapidly
procedures. Clinical judgment must be uti-
become very ill and this decision must be
lized to determine the risk for the patient
made expeditiously after discussion between
without an obvious source of infection.
the oncologist, surgeon, radiologist, pulmo-
Issues in the social history such as family
nologist, as well as the patient and family.
reliability, compliance, and ability to travel
to the outpatient center must be factored
Other supportive measures
into a decision plan. If the patient has
recently received chemotherapy and is
Granulocyte colony-stimulating factor
expected to reach a nadir in the next few
(G-CSF) may be indicated in the setting
days, consideration should be given to more
of FN; however, there are no specific stan-
frequent assessment or hospital admission.
dards for its use. Therapy with 5 mcg/kg/day
As with the neutropenic patient, blood
subcutaneously is recommended under cer-
cultures should be drawn and empiric anti-
tain conditions in which the clinical course
biotics initiated. The risk for Pseudomonas
is likely to worsen before the anticipated
is significantly decreased in the patient
marrow recovery (see Chapter 25). Patients
without severe neutropenia and therefore
who remain severely neutropenic with life-
ceftriaxone IV/IM at 50 mg/kg (maximum
threatening infections should receive G-
2 g) can be given daily while febrile in the
CSF. Granulocyte transfusions may be con-
outpatient setting in the otherwise well-
sidered in the setting of profound neutro-
appearing child for at least the first
48
penia and sepsis, especially an invasive bac-
hours, while awaiting results of the blood
terial or fungal infection that is not improv-
culture. Hospitalization may be required
ing on appropriate therapy with expected
in patients who are persistently febrile with
continuation of profound neutropenia (see
a dropping ANC and in those with a pos-
Chapter
5). However, such transfusions
itive blood culture or the development of
are becoming increasingly more difficult
localizing signs.
to obtain.
Suggested Reading
Fever in the nonneutropenic
oncology patient
Donowitz GR, Maki DG, Crnich CJ, et al. Infec-
tions in the neutropenic patient—new views
of an old problems. Hematol Am Soc Hema-
The febrile, nonneutropenic patient
tol Educ Prog 113-139, 2001.
remains susceptible to infection secondary
Freifeld AG, Bow EJ, Sepkowitz KA, et al. Clinical
to immune dysfunction and often second-
practice guideline for the use of antimicrobial
ary to the presence of a CVC. These patients
agents in neutropenic patients with cancer:
should have a careful assessment with his-
2010 update by the Infectious Disease Society
tory, physical examination, and pertinent
of America. Clin Infect Dis 52:e56-e93, 2011.
laboratory studies as in the febrile neutro-
Pizzo PA. Fever in immunocompromised
penic patient. Patients with localizing find-
patients. N Engl J Med 341:893-900, 1999.
Acute Pain
28
Management in the
Inpatient Setting
Pain is a common problem for children with
framework that physicians could use when
hematologic or oncologic conditions, as it
developing treatment plans for cancer
can result both from the condition itself and
pain, paving the way for significant im-
from its management. A number of studies
provements in its management. Over the
have demonstrated that effective pain man-
subsequent 25 years, it has undergone much
agement not only increases a patient’s com-
scrutiny and several modifications have
fort level but can also effect long-term
been proposed. A recent adaptation is pre-
changes in a patient’s pain threshold, and,
sented in Figure 28.1.
in critically ill patients, it has been demon-
The cornerstone of the ladder rests on
strated to improve morbidity and mortality.
five simple recommendations that remain
Despite this, other studies have demon-
valid today:
strated that pediatric pain management is
1. The oral form of medication should be
often suboptimal. This results from percep-
chosen whenever possible.
tions on the parts of both parents and care
2. Analgesics should be given at regular
providers that pain is less frequent or less
intervals. It is necessary to respect the dura-
severe than is the case, or from concerns of
tion of the medication’s efficacy and to
causing dependence or addiction.
prescribe doses at specific intervals in accor-
The management of chronic pain is a
dance with the patient’s level of pain. The
frequent challenge for pediatric hematologists
dose should be adjusted until the patient is
and oncologists, but is outside the scope of this
comfortable.
chapter. For pain management at the end of
3. Analgesics should be prescribed accord-
life, see Chapter 29. For pain management in
ing to pain intensity as evaluated by a scale
patients with sickle cell disease, see Chapter 3.
of intensity of pain. The prescription must
be given according to the level of the
The World Health Organization
patient’s pain and not according to the
analgesic ladder
medical staff’s perception of the pain.
4. Dosing of pain medication should be
In
1986 the World Health Organization
adapted to the individual. The correct dos-
presented the analgesic ladder as a
age is one that will allow adequate relief of
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Acute Pain Management in the Inpatient Setting
257
STEP 4
Neurosurgical
procedures
Nerve block
Epidurals
STEP 3
PCA pump
Acute pain
Neuroly c block therapy
Chronic pain without control
Strong opioids
Spinal s mulators
Acute crises of chronic pain
Methadone
STEP 2
Oral administra on
Transdermal patch
Weak opioids
STEP 1
Chronic pain
Non-malignant pain
Nonopioid
Cancer pain
analgesics
NSAIDs
NSAIDs
(with or without adjuvants
at each step)
Figure 28.1 Adaptation of the World Health Organization analgesic ladder. Abbreviations: NSAID,
non-steroidal anti-inflammatory drug; PCA, patient-controlled analgesia.
pain. The dosing should be adapted to
on the best method of assessment have
achieve the best balance between the anal-
made its measurement a challenge. As a
gesic effect and the side effects.
result, a variety of pediatric pain assessment
5. Analgesics should be prescribed with a
scales have been developed to assist in this
constant concern for detail. The regularity
process.
of analgesic administration is crucial for the
adequate treatment of pain.
Neonates and infants
The Neonatal Infant Pain Scale
(NIPS),
It is important to remember that these are
developed at the Children’s Hospital of
guidelines and that there are situations
Eastern Ontario, is a widely used method
where a stepwise approach to pain is not
of assessing pain in this patient population.
the best choice for management. For exam-
It evaluates a series of six parameters, with a
ple, management of sickle cell vaso-occlusive
range of scores between 0 and 7. Scores
crises typically involves early introduction of
above 3 are considered indicative of a pain
step 3 medications without waiting for the
level sufficient to require intervention. The
patient to fail step 1 and 2 therapies.
scale is presented in Table 28.1. Its major
limitation is that it may underestimate the
level of pain in infants who are too ill to
Assessment of pain
respond appropriately or are receiving a
paralyzing agent.
The accurate and reproducible assessment
of pain is essential to effective management,
Preverbal/Nonverbal children
allowing the provider to determine when
Pain assessment for children who are unable
intervention is necessary and to more
to describe their pain or use an interactive
objectively measure the response to inter-
pain assessment scale due to young age,
vention. However, the subjective nature of
cognitive dysfunction, injury, or medical
pain and the impact of developmental stage
interventions such as intubation requires
258
Chapter 28
Table 28.1 The Neonatal Infant Pain Scale (NIPS).
Parameter
0 points
1 point
2 points
Facial expression
Relaxed
Contracted/grimacing
Cry
Absent
Mumbling
Vigorous
Breathing patterns
Relaxed
Different than basal
Arms
Relaxed
Flexed/stretched
Legs
Relaxed
Flexed/stretched
State of arousal
Sleeping/calm
Uncomfortable
an alternative similar to the NIPS that allows
School aged and older children
the subjective evaluation of pain-related
Children 5 years of age and older have been
behaviors by another individual. One of the
shown to have the ability to accurately rate
more commonly used assessment tools is
their level of pain using a numerical rating
the Face, Legs, Activity, Cry, Consolability
scale. A commonly used tool is the Wong-
(FLACC) Behavior Scale. Developed at the
Baker FACES scale, presented in Figure 28.2.
University of Michigan as a tool to assess
Developed in the early 1980s, it remains one
postoperative pain, it has been shown to
of the most widely used tools for assessing
reliably measure pain in a variety of clinical
pain in both children and adults.
situations. It is presented in Table 28.2. The
range of possible scores is from 0 to 10,
When pain assessment scales are used
consistent with other commonly used
appropriately they can be very valuable in
numerical rating scales used for older
helping the care team know whether their
children.
pain management interventions are
Table 28.2 The Face, Legs, Activity, Cry, Consolability (FLACC) Scale.
Parameter
0 Points
1 Point
2 Points
Face
No particular expression Occasional grimace, or
Frequent to constant
or smile
frown, withdrawn or
frown, clenched jaw,
disinterested
quivering chin
Legs
Normal position or
Uneasy, restless, or tense Kicking or legs drawn up
relaxed
Activity
Lying quietly, normal
Squirming, shifting back Arched, rigid, or jerking
position, moves easily
and forth, or tense
Cry
No cry
Moans, whimpers, or
Crying steadily, screams
occasional complaint
or sobs, frequent
complaints
Consolability Content, relaxed
Reassured by occasional
Difficult to console or
touching, hugging, or
comfort
being talked to;
distractible
Acute Pain Management in the Inpatient Setting
259
0
2
4
6
8
10
No pain
Li le pain
Mild pain
Moderate pain Severe pain
Worst pain
Figure 28.2 The Wong-Baker FACES Pain Rating Scale.
appropriate or need to be adjusted. How-
age. Hepatic enzymes are initially immature,
ever, there is no “magic number” that indi-
but these mature quickly. Children 2 to 6
cates when interventions need to be initi-
years of age develop a larger relative hepatic
ated. Studies consistently show that there is
size for weight and often metabolize drugs
little correlation between a child’s pain score
more quickly than younger or older chil-
and their perception as to whether or not
dren; this can translate to a need for larger
they need pain medication. Therefore, in
doses of drug given more frequently to
situations where patients are likely to have
achieve adequate analgesia. Variations in
pain, it is essential to question them regu-
age, weight, blood flow, and organ function
larly regarding their need for medication.
can all affect effective drug dosing.
Step 1 therapy: nonopioid
Pain pharmacology
analgesics
Drugs that are commonly used for step 1
Pain can be subdivided into two categories,
therapy of pain in children are presented in
nociceptive and neuropathic. Acute pain in
Table 28.3. These should be first-line therapy
children is most often nociceptive, and a
for pain unless the clinical situation dictates
variety of drugs can be used to treat this pain.
that therapy should begin at a higher step.
Much of the information regarding this ther-
The use of NSAIDs in children with can-
apy is empiric, as there are few formal drug
cer is generally discouraged due to the com-
studies for many of these agents, especially in
mon occurrence of thrombocytopenia in this
children less than
12. Nonsteroidal anti-
population and concern for an increased risk
inflammatory drugs (NSAIDs), opiates, mus-
of bleeding due to an inhibition of platelet
cle relaxants, local anesthetics, and tramadol
function. However, there have been no large,
can be used. Some other adjuvant therapies
controlled trials demonstrating the validity of
can also be utilized, such as psychological
this concern. Several clinical trials have dem-
intervention, distraction, and biofeedback.
onstrated that ibuprofen can safely be used
When treating pain in neonates it is
following dental surgery, tonsillectomy, and
important to remember that several factors
neurosurgery in healthy children and in vitro
can modify their response and clearance of
data suggest that ibuprofen causes no signif-
drugs. They have increased body water con-
icant prolongation of the platelet function
tent and decreased fat, which can alter drug
assay PFA-100.
distribution. A relative increase in blood
flow to the brain and a somewhat “leaky”
Step 2 therapy: weak opioids
blood-brain barrier can cause a prolonga-
Patients whose pain is inadequately con-
tion of drug effect. Renal clearance of drugs
trolled with step 1 therapy or who are being
can be relatively decreased up to 1 year of
weaned from step
3
therapy require
260
Chapter 28
Table 28.3 Step 1 pain therapy medications.
Drug
Dose
Comments
Ibuprofen
1-3 months: 5 mg/kg 3-4 times daily
* Block conversion of arachi-
3 months-1 year: 50 mg 3 times daily
donic acid into prostaglan-
1-4 years: 100 mg 3 times daily
dins and thromboxanes
4-7 years: 150 mg 3 times daily
* Nonselective; variably impair
7-10 years: 200 mg 3 times daily
platelet function
10-12 years: 300 mg 3 times daily
* Ketorolac can affect renal
12-18 years 300-400 mg 3 times daily
function or bone growth with
Maximum dose 30 mg/kg/day; 2.4 g/day
prolonged use
Naproxen
5 mg/kg/dose 2 times daily
Maximum dose 15 mg/kg/day
Ketorolac
0.5 mg/kg/dose IV/IM every 6 hours
Maximum dose 30 mg
Maximum 20 doses
Acetaminophen
15 mg/kg/dose every 4-6 hours
* Has no antiplatelet effect
Maximum 5 doses/day; maximum 4 g/day
* Can be hepatotoxic
* Avoid rectal dosing in neu-
tropenic patients
* Parenteral form recently
licensed in United States
Abbreviations: IV, intravenous, IM, intramuscular.
treatment with step 2 agents. In the United
Step 3 therapy: strong opioids
States, the most common agents used are
Patients not responding to step 2 therapy or
those that combine acetaminophen with
whose condition indicates the need for
a weak opioid. These include codeine or
stronger pain therapy at the outset are can-
hydrocodone, although there is some dis-
didates for step 3 therapy. All of the med-
agreement about the inclusion of hydroco-
ications in this category are strong opioids.
done in the weak opioid category.
They act by binding to m-receptors in the
Commonly used agents in this category are
spinal cord and central nervous system
presented in Table 28.4.
(CNS). These receptors are also found
It is important to remember that the
throughout the body, and binding to
designation “weak opioid” does not translate
peripheral sites accounts for many of the
to “without side effects.” The use of codeine
side effects seen with opiate therapy.
and hydrocodone can cause the same side
Opioids vary both by their duration of
effects seen with stronger opioids: respiratory
action and by the emotional effect they
depression, hypoxia, nausea, vomiting, pru-
produce. Meperidine and oxycodone typi-
ritus, constipation, physical tolerance, and
cally cause euphoria; conversely, morphine
dependence. Although the Food and Drug
more commonly causes dysphoria. Equiva-
Administration
(FDA) classifies them as
lent doses of the strong opioids and the
Schedule III medications, the risks of abuse
pharmacokinetics of their oral formulations
and diversion still exist.
are presented in Table 28.5.
Acute Pain Management in the Inpatient Setting
261
Table 28.4 Step 2 pain therapy medications.
Drug
Dose
Comments
Codeine
0.5-1 mg/kg q4-6 hour
* Comes as 30 and 60 mg tablets
Max: 60 mg/dose
* Up to 35% of children are
inefficient metabolizers of
codeine to morphine; they
will achieve minimal benefit
from this product
Acetaminophen with
0.5-1.0 mg/kg/dose of codeine
* Tablet: 300 mg/15 mg, 300 mg/
codeine
q4-6 hour
30 mg
Max: 2 tablets/dose; 15 mL/dose
* Liquid: 120 mg/12 mg per 5 mL
* Up to 35% of children are
inefficient metabolizers of
codeine to morphine; they
will achieve minimal benefit
from this product
Acetaminophen with
>2 years: 0.135 mg/kg/dose hydro-
* Tablet: 5 mg/500 mg
hydrocodone
codone
* Liquid: 7.5 mg/500 mg per
<40 kg: do not exceed 5 mg hydro-
15 mL
codone per dose
* Elixir contains 7% alcohol
>40 kg: do not exceed 7.5 mg
hydrocodone per dose
Oral starting doses of step 3 medications
to deal with postchemotherapy mucositis
are presented in Table 28.6. It is important
or sickle cell vaso-occlusive crisis. Rather, it
to remember that patients with severe
refers to long-term, ambulatory PCA pump
chronic pain will often be on doses much
use for severe chronic pain. With the
larger than this due to tolerance. Tactics
increased availability of highly concen-
such as narcotic substitution can be used to
trated oral narcotics and novel delivery
deal with tolerance or with side effects
systems, the use of home PCA therapy has
resulting from large doses. However, these
declined significantly. Additional step
4
steps should be performed under the guid-
therapies include nerve blocks, epidural
ance of individuals who are well versed in
injections, and other alternative interven-
such measures. Transdermal fentanyl has
tions routinely provided by a specialized
not been included; practitioners familiar
pain service.
with its usage should transition patients
to this agent when deemed necessary.
Nonpharmacologic
Step 4 therapy
approaches to pain
As can be seen in Figure 28.1, step 4 therapy
does not introduce new medications.
The effectiveness of psychological interven-
Patient-controlled analgesia (PCA) pumps
tions to decrease the perception of pain
are listed; this does not refer to short-term
and the anxiety associated with it, and to
PCA pump use such as following surgery or
improve the quality of life in patients
262
Chapter 28
Acute Pain Management in the Inpatient Setting
263
Table 28.6 Step 3 oral pain therapy medications.
Drug
Oral dose
Comments
Oxycodone
* Instant release: 0.05-0.15 mg/kg/
dose up to 5 mg/dose q4-6 hour
* Sustained release: for patient
taking >20 mg/day of oxycodone
can administer 10 mg q12 hour
Morphine
* 0.3-0.6 mg/kg/dose every 12 hours
* Injection (mg/mL): 1, 2, 4, 5, 10
for sustained release
* Injection, preservative free
* 0.2-0.5 mg/kg/dose q4-6 hour prn
(mg/mL): 1, 5
for immediate release tablets or
* Oral solution (mg/mL): 2, 4, 20
solution
* Tablet (IR) (mg): 10, 15, 30
* Tablet (ER) (mg): 15, 30, 60,
100, 200
Hydromorphone
0.03-0.08 mg/kg/dose PO q4-6
hour; max: 5 mg/dose
Methadone
0.03-0.08 mg/kg/dose PO q4-6
hour; max: 5 mg/dose
Abbreviations: IR, immediate release; ER, extended release.
with chronic pain, has been clearly demon-
quality of life in patients with sickle cell
strated in a number of clinical settings.
disease.
These include:
* Minimizing postoperative pain and anx-
It is vital that child-life workers become
iety through the use of preoperative inter-
involved with children admitted with a pos-
ventions before painful procedures. These
sible diagnosis of cancer very shortly after
include the explanation of the event in
admission. Data indicate that psychological
developmentally appropriate terms, mini-
interventions are most effective in decreas-
mizing wait times once the patient arrives in
ing anxiety related to painful procedures
the operating room, child-friendly waiting
when initiated before the first painful expe-
spaces, and cognitive-behavioral therapies
rience. It is also important to obtain input
(CBT) such as directed imagery, deep
from a psychologist early on for children
breathing, and role playing. These interven-
with conditions causing chronic pain so that
tions are typically provided and coordinated
appropriate interventions can be started.
by the child-life program.
* The use of CBT, relaxation therapy, and
biofeedback to decrease the perception of
Suggested Reading
pain among children suffering from chronic
pain caused by cancer, headache, and
Friedrichsdorf SJ, Kang TI. The management of
abdominal pain.
pain in children with life-limited illnesses.
* The use of CBT, relaxation, self-hypno-
Pediatr Clin N Am 54:645-672, 2007.
sis, biofeedback, and social support groups
Verghese ST, Hannallah RS. Acute pain manage-
to decrease painful episodes and improve
ment in children. J Pain Res 3:105-123, 2010.
Palliative Care
29
The World Health Organization defines
There is a role for palliative care for all
pediatric palliative care as care that “aims
children diagnosed with cancer, even those
to improve the quality of life of patients
whose cancers have an excellent prognosis
facing life-threatening illnesses, and their
with appropriate therapy. It also applies to
families, through the prevention and relief
the large number of children with life-
of suffering by early identification and treat-
threatening nononcologic conditions. The
ment of pain and other problems, whether
definition stresses the importance of “early
physical, psychosocial, or spiritual.” It is the
identification and treatment of pain and
active total care of the child’s body, mind,
other problems.” Palliative care is not syn-
and spirit, and also involves giving support
onymous with “end-of-life” care; palliative
to the family. It begins when illness is diag-
care must begin at the time of diagnosis and
nosed, and continues irrespective of
continue on throughout the period of dis-
whether or not a child receives disease-
ease-directed therapy. It also must include
directed treatment. It requires that health
many resources from throughout the
providers evaluate and alleviate a child’s
patient’s and family’s range of experience:
physical, psychological, and social distress
physicians, nurses, social workers, psychol-
at all times during their treatment. To be
ogists, spiritual counselors, and a variety of
effective, it requires a broad multidiscip-
other support personnel chosen because of
linary approach that includes the family and
each child’s unique situation. During any
makes use of available community
hospitalization, an important responsibility
resources. However, it can be successfully
of the medical team is to ensure that
implemented even if community resources
each child has access to all needed services
are limited. Palliative care can and should be
and is being supported to the greatest
provided in tertiary-care facilities, commu-
extent possible.
nity health centers, and at home.
This definition contains several concepts
that deserve consideration. It contains the
Individualized care planning
broadly defined term “life-threatening” that
and coordination
includes situations in which a cure is pos-
sible, instead of the more limited “life-limit-
At the time of diagnosis, parents and care-
ing” that includes only those conditions for
givers of children with life-threatening ill-
which there is no realistic hope of cure.
ness have two goals: a care-directed goal of
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Palliative Care
265
cure and a comfort-directed goal of decreas-
undergoing bone marrow transplantation,
ing suffering. Introducing palliative care
the model is useful in helping to define what
principles early on in the care process is
care options are acceptable to the patient and
respectful and supportive of these goals.
family given the prognosis and goals of treat-
A model developed by the Palliative and
ment. Key concepts in this model include the
End-of-Life Care Task Force at St. Jude
establishment of a close and caring relation-
Children’s Hospital, the Individualized Care
ship with the patient and family, clarifying
Planning and Coordination model is
patient and family values and priorities,
designed to facilitate this introduction
determining the goals of care based on the
(Figure 29.1). Although developed to assist
patient’s and family’s understanding of prog-
in making care decisions for children
nosis, and allowing them to choose from
Eligibility
Step 1 : Rela onship
Understanding
Sharing
illness
Relevant
Experience
Informa on
Needs assessment
Step 2 : Nego a on
Prognosis
Goals
Treatment
op ons
Step 3 : Plan
Medical plan
Life plan
Comperhensive
Care Plan
Ac on
(Care Coordina on)
Figure 29.1 The Individualized Care Planning and Coordination Model.
266
Chapter 29
available goal-directed treatment options.
totally clear. Studies have also demonstrated
From this foundation, the members of the
that significant levels of parental dissatisfac-
care team can coordinate the provision of
tion with hospital staff can arise from
care to ensure the social, psychological, and
confusing, inadequate, or uncaring commu-
emotional needs of the patient and family are
nication regarding treatment and prognosis
addressed at the same time that appropriate
as well as discrepancies between parents
medical care is provided.
and care providers in understanding the
terminal condition. When a language bar-
rier exists between family and staff, these
End of life care
problems are often exacerbated.
Despite the tremendous advances made in
the care of children with cancer over the past
Common symptoms at the end
50 years, at the present time one in five
of life
children diagnosed with cancer will ulti-
mately die of their disease. The death of a
Pain
child affects the physical and psychological
Parents and caregivers report that their
well-being of family members for the rest of
child’s pain is the symptom they fear most.
their lives. Events occurring around the time
A large survey of parents whose children
of death, both positive and negative, play a
died of cancer revealed that 75% of them felt
critical role in defining how family members
their child suffered pain at the end of life,
grieve and ultimately come to terms with
but only 37% felt the pain was successfully
the event. Families who have lost a child
managed.
have identified several needs regarding end-
The principles of pediatric pain manage-
of-life care. These include the need to have
ment are presented in Chapter 28 and pain
complete information honestly communi-
management at the end of life follows these
cated, to have easy access to essential staff
same principles. However, it becomes par-
members who will be supportive, to have
ticularly important to clarify and under-
assistance in coordinating necessary ser-
stand the goals of treatment as elucidated
vices, to have their relationship with their
by the patient and parents. The desire to
child maintained as much as possible, and to
maintain alertness and interaction early in
be allowed to feel that their child’s life and
treatment may give way to a desire to keep
death has meaning.
the patient comfortable, even if it means the
Research studies involving pediatric pal-
patient will be more sedated and less aware.
liative care are limited, but they have iden-
Acceding to the family’s wishes that the
tified several important shortcomings in the
patient not be hooked up to monitors might
level of palliative care provided to children.
outweigh concern that the doses of pain
Several studies have demonstrated a lack of
medications being used could result in
successful management of pain and other
respiratory depression. These decisions
distressing symptoms. Other studies have
often run counter to what physicians see
demonstrated that the majority of children
as their primary goal of care, to prolong life
dying of life-threatening diseases die in the
as long as possible. Working through some
hospital, often in the intensive care unit.
of these difficult philosophical issues with
The impact of this outcome on the appro-
the help of a mentor can be essential to
priateness of care provided and its effect on
working comfortably and effectively in this
the family’s bereavement process are not
type of setting.
Palliative Care
267
Nausea and vomiting
A stepwise approach to the management
Nausea and vomiting at the end of life can be
of nausea and vomiting in the palliative care
triggered by many different stimuli. A variety
setting is outlined in Table
29.1. A key
of toxins such as medications and their
feature of this approach is to ensure that
metabolites, urea resulting from renal failure,
the interventions selected are in keeping
or metabolic byproducts of hepatic failure
with the patient’s and family’s goals of care.
can all stimulate the chemoreceptor trigger
A number of medications can be tried to
zone and cause nausea. Increased intracranial
control nausea and vomiting. The ideal
pressure and a variety of emotional and
agent is one that targets the specific cause
sensory stimuli can mediate nausea and
of the patient’s distress if such an agent can
vomiting through cortical pathways. The
be identified. A list of useful medications is
gastrointestinal (GI) tract can stimulate the
outlined in Table 29.2.
vomiting center in response to local receptor
Opioids frequently cause nausea and
stimulation caused by obstruction, stasis,
vomiting due to a direct effect on the che-
toxins, or drugs. In addition, pharyngeal
motherapy trigger zone or secondarily to
stimulation by mucous or mucosal break-
their effect on GI motility leading to gastro-
down can also trigger the vomiting center.
paresis and constipation.
Table 29.1 Management of nausea and vomiting.
Step
Objective
Intervention
1
Attempt to identify causes
History (timing, quality, severity of nausea;
description of vomiting)
Physical exam focused on neurological, abdominal
symptoms
Medication history (opioids, chemotherapy, anti-
biotics, NSAIDs, other drugs)
2
Initiate environmental changes
Small meals
Minimize strong smells
Create a comfortable environment
Provide support for emotional distress
3
Clarify scope of interventions
Discuss goals of care with patient and family
that will be acceptable to
patient and family
4
Choose treatment based on
Chemically induced: stop medications if possible,
etiology and goals of care
opioid rotation if felt to be cause; antiemetics
Increased ICP: surgical treatment if possible (shunt
insertion, shunt revision), medical therapy
(steroids, antiemetics)
Obstruction/ileus: surgical management if consis-
tent with goals of care; medical management
(steroids, octreotide, opioids with scopolamine,
haloperidol)
Other/unknown cause: trial of antiemetics
Abbreviations: NSAIDs, nonsteroidal anti-inflammatory drugs; ICP, intracranial pressure.
268
Chapter 29
Table 29.2 Medications for treatment of nausea and vomiting.
Drug
Dose
Metoclopramide (prokinetic/
Prokinetic dose: 0.1 mg/kg/dose IV or orally every 6 hours
dopamine antagonist)
Dopamine antagonist (antiemetic) dose: 0.5-1 mg/kg/dose
IV or orally every 6 hours; use with diphenhydramine to
prevent extrapyramidal symptoms
Ondansetron (serotonin receptor
0.45 mg/kg/day IV or orally; may be dosed once daily or
antagonist)
divided every 8 hours
Scopolamine (anticholinergic)
For children >40 kg: 1.5 mg patch behind ear every
72 hours
Meclizine (antivertigo agent)
For children >12 years of age: 25-75 mg/day orally in up to
3 divided doses
Dexamethasone (anti-
Antiemetic: 10 mg/m2 IV or orally daily; maximum dose
inflammatory)
20 mg daily
Increased ICP: increase dosing frequency to 2-4 times
daily, with a maximum dose of 40 mg/day
Octreotide (somatostatin analogue)
5-10 mg/kg/day (continuous 24-hour IV infusion or
divided twice daily subcutaneously)
Dronabinol (cannabinoid)
For children aged 6 years and older; 2.5-5 mg/m2/dose
every 4-6 hours. Not recommended for children who
have clinical depression
Lorazepam (benzodiazepine)
0.025 mg/kg/dose IV or orally every 6 hours
Abbreviations: IV, intravenous; ICP, intracranial pressure.
Constipation
intake of fluids and food, decreased physical
Constipation often causes pain and distress
activity, neurological dysfunction, and intes-
for children at the end of life, both physically
tinal obstruction due to presence of tumor.
and psychologically. Prevention of consti-
Assessment
pation should therefore be a primary goal of
History
end-of-life care.
One of the most common causes of
* Stool frequency, consistency, and associ-
constipation are opioids, which decrease
ated discomfort
GI motility and fluid secretion, leading to
* Abdominal pain, nausea, vomiting, and
hard, dry stools. Unlike other narcotic side
anorexia
* Neurological symptoms: urinary pro-
effects, constipation does not seem to
decrease with ongoing use. Patients and
blems, lower extremity numbness, weak-
ness, and paresthesias
families should be informed ahead of time
about this side effect, as children in severe
* Diarrhea
(sign of encopresis or
pain may not consider regular bowel move-
obstipation?)
ments a priority. Once well established,
* Prior episodes, previously effective
constipation may require quite aggressive
therapies
and unpleasant therapies to correct it.
Other factors that contribute to consti-
Physical Examination
pation in terminal patients include poor oral
* Abdominal palpation
Palliative Care
269
External rectal exam (digital exam rarely
Table 29.3 Appetite stimulants for cancer-
needed and may be dangerous)
associated anorexia.
* Neurologic exam
Drug
Dose
Medications
Cyproheptadine
2-6 year: 2 mg bid
* Stool softeners (docusate): rarely effective
(Periactin)
>6 year: 4mg bid
alone
Megestrol acetate
7.5-10 mg/kg/day in
* Osmotic agents (Miralax
, magnesium
(Megace)
1-4 divided doses
sulfate, and lactulose): effective for long-
Maximum 800 mg/day
or 15 mg/kg/day
term use; Miralax requires 4 to 8 oz. of fluid
Dronabinol
2.5 mg/m2/dose every
which can be a limitation
(Marinol)
4-6 hours
* Stimulants (senna, bisacodyl): often use-
Use with caution in
ful and necessary in the end-of-life setting
children with
when less concern exists regarding develop-
depression; avoid
ment of dependence
in children with
sensitivity to
Anorexia and weight loss
sesame oil
Anorexia or loss of appetite is a familiar
symptom in children with cancer. It can occur
at any point in treatment; institutional studies
Support for emotional distress
indicate that it affects as many as 40% of
Discontinue medications that might be
children with cancer at diagnosis and the pre-
causing anorexia
valence increases to as high as 70% in children
with advanced stage disease. Early in treat-
Step 3: more aggressive therapies
ment it is most often a side effect of chemo-
(if consistent with plan of care)
therapy or radiation, or a result of stomatitis,
Medications
(see Table 29.3)
dysphagia, constipation, pain, or depression.
As the child’s cancer becomes progressive,
Nutritional supplementation
If acceptable to patient and family, enteral
it is often a component of the anorexia/
feeding with a nasogastrostomy (NG) or
cachexia syndrome, a complex set of central
gastrostomy tube is often reasonably well
nervous system (CNS) and metabolic abnor-
tolerated unless the patient has a GI
malities caused by a combination of tumor
obstruction or is chronically nauseated.
byproducts and host cytokine release.
Total parenteral nutrition
(TPN) is
Step 1: assessment
rarely useful in the end-of-life setting except
State of primary disease
for short-term use in the patient with GI
Diet and fluid history
obstruction.
Nausea/vomiting/constipation
Oral lesions (including candidiasis)
Fatigue
The National Comprehensive Cancer Net-
Medication history
work defines cancer-related fatigue as “a dis-
History of prior eating disorder
tressing persistent, subjective sense of physi-
Sense of smell
cal, emotional and/or cognitive tiredness or
Step 2: environmental changes
exhaustion related to cancer or cancer treat-
Small meals
ment that is not proportional to recent activ-
Regular mouth care
ity and interferes with usual functioning.”
Comfortable environment
It is pervasive, multidimensional, and
270
Chapter 29
incapacitating; as such it significantly de-
associate their feelings of fatigue to sleep
creases health-related quality of life in chil-
disturbance or side effects of therapy
dren. While it can exist throughout the entire
whereas parents are more likely to include
period of cancer treatment, fatigue is most
emotions and nutritional status as potential
often seen as significant as the child’s disease
causes. A recent study suggested that pain
progresses. In several studies, parents have
and dyspnea were frequently associated with
identified fatigue as the most significant
fatigue; the authors postulated that opioids
symptom that moderately or severely im-
and benzodiazepines, common therapies for
pacted their child’s well-being toward the end
pain and dyspnea, might have significantly
of life. These same studies have also demon-
contributed to their patients’ fatigue.
strated that it is the symptom most likely to be
In a recent review on the assessment and
missed by care providers; fewer than 50% of
management of fatigue and dyspnea in pedi-
children whose parents described them as
atric palliative care, Dr. Christina Ulrich
suffering highly from fatigue had this docu-
defined a conceptual model for understand-
mented in their medical record.
ing fatigue in this population. This frame-
Cancer-related fatigue is more common
work is outlined in Figure 29.2.
and more severe in children with brain
Data are very limited regarding effective
tumors than in those with other cancers.
treatments for cancer-related fatigue in chil-
Pediatric patients tend more often to
dren, although a number of interventions
Psychosocial factors
Depression
Familial stressors
Anxiety
Spiritual concerns
Fear
Caregiver status
Boredom
Prac cal concerns
Sleep disturbance
Unrelieved symptoms
Change in environment
Fa gue experience
Circadian rhythm altera on
Medica on effect
Physical factors
Underlying illness
Metabolic abnormali es
Type, site, stage
Nutri onal impairment
Disease-directed treatment
Cachexia
Unrelieved symptoms
Dehydra on
Side effects of symptom treatment
Comorbidi es
Changes in muscle
Anemia
Decondi oning
Infec on
Abnormal muscle structure, func on
Hormone imbalance
Organ dysfunc on
Figure 29.2 A model of fatigue in life-threatening illness.
Palliative Care
271
Table 29.4 Treatments for cancer-related fatigue.
Nonpharmacologic interventions
Pharmacologic interventions
Psychosocial
Stimulants
* Education
* Methylphenidate
* Support groups
* Modafinil
* Individual counseling
Antidepressants
* Coping strategies
* SSRIs
* Stress management training
Paroxetine
* Individualized behavioral intervention
Sertraline
Exercise
* Other antidepressants
Sleep therapy
Bupropion
* Behavioral therapy
Steroids
* Stimulus control
* Sleep restriction
* Sleep hygiene
Acupuncture
Abbreviation: SSRI, selective serotonin reuptake inhibitor.
have been shown to be effective in adults.
treatment is a key component of end-of-
These are outlined in Table 29.4.
life care.
Psychosocial and activity-based inter-
Dyspnea can result from either an
ventions have been shown to be quite effec-
increased metabolic demand or an inability
tive in adults with cancer-related fatigue;
to maintain a normal minute ventilation.
data in children are lacking. Exercise has the
The former can be caused by complications
strongest data regarding its value, but this is
of the disease or its treatment, such as
largely in adult cancer survivors suffering
infection, metabolic acidosis, or increased
from fatigue after the completion of curative
metabolic demands caused by extensive
therapy. Its utility in ameliorating fatigue in
tumor burden. The latter can result from
patients near the end of life is less well
conditions such as decreased lung compli-
established.
ance due to tumor, infection, or fluid, or by
other conditions such as interstitial lung
Dyspnea
disease, anemia, or fatigue.
Dyspnea is the term used to describe the
It follows that treatment of dyspnea
feeling of breathlessness that occurs when
would involve steps to decrease metabolic
the respiratory system fails to meet the
demand or improve the efficiency of respi-
body’s need for oxygen uptake or carbon
ration. However, this can prove to be quite
dioxide removal. It occurs under normal
challenging in practice. It is often difficult to
circumstances such as after brisk exercise,
determine whether the cause of dyspnea is
but in pathological conditions it can cause
in fact respiratory or metabolic, and once
severe distress. It occurs very frequently at
discovered, the unwillingness of the patient
the end of life, as respiratory failure occurs
or family to accept certain interventions can
very commonly as a terminal event. There-
create additional challenges for the treat-
fore, recognition of dyspnea and its effective
ment team.
272
Chapter 29
Appropriate therapies to decrease meta-
Support of respiration
bolic demand depend on the cause of the
When intubation and mechanical ventilation
increased demand. For the patient who
are not appropriate or acceptable to the
becomes dyspneic with activity, it may be
patient and family, noninvasive positive pres-
as simple as recommending that the patient
sure ventilation (NIPPV) can be useful in
use a wheelchair instead of walking or
supporting respiration. However, the use of
decrease physical activity in other ways.
NIPPV in adult patients who have elected to
Increased metabolic demand caused by
forgo intubation or other aggressive respira-
infection or inflammation can be treated
tory support is controversial. Its use in chil-
with aggressive use of antibiotics or anti-
dren with cancer is unstudied; there have been
inflammatory agents, as long as such ther-
a handful of papers published regarding its use
apies are in keeping with the patient’s and
in children with neuromuscular disorders.
family’s wishes.
From the adult literature, it appears clear
Respiratory failure can result from two
that the most important issue when deciding
general causes: lung parenchymal dysfunc-
on the use of NIPPV to support respiration is
tion and respiratory muscle fatigue or fail-
a clear understanding of the goals of therapy
ure. It is important to distinguish between
among all involved. In 2007, the Society of
these, as the treatments are different. An
Critical Care Medicine Palliative Noninva-
algorithm for distinguishing between these
sive Positive Pressure Ventilation Task Force
causes is presented in Table
29.5. It is
published its recommendations on the use
important to note that hypercarbia can be
of NIPPV in the palliative care setting.
a late finding in parenchymal dysfunction
While intended for adults, the concepts can
as well.
be applied to the pediatric care setting as well.
The Task Force proposed a three-category
Treatment
approach to assist in decision making regard-
Treatment for dyspnea can be divided into
ing this technology. An overview of this
three general categories: mechanical sup-
approach is presented in Table 29.6.
port of respiration, support of gas exchange,
The key areas of distinction in this model
and therapies to decrease the sensation of
are goals of care, determination of success,
dyspnea.
appropriate endpoints, and response to
Table 29.5 Classification and features of respiratory failure.
Parenchymal dysfunction
Muscle failure/fatigue
Causes
Airway obstruction
Neuromuscular disease
* Interstitial, bronchial inflammation
* Weak musculature
* Excessive secretions
* Lowered fatigue threshold
External compression
* Prolonged recovery time
* Pleural fluid
Abnormal load on respiratory system
* Pleural-based nodules
Weakness caused by systemic disease
* Infiltration of interstitium by
metastases
Blood gas
Hypoxemia
Hypoxemia, hypercarbia
findings
Palliative Care
273
274
Chapter 29
Palliative Care
275
failure. It is imperative that each member of
as well. The dose that is required is lower
the care team agrees on each of these com-
than what is commonly used for pain; a
ponents to avoid confusion and conflict
morphine dose of 0.025 mg/kg, or an equiv-
later in the course.
alent dose of fentanyl or Dilaudid, is often
effective in the narcotic-naıve child, and an
Support of gas exchange
increase in the dose by 25% will often be
NIPPV can also be used to augment gas
effective in the child already receiving mor-
exchange in patients with low lung volumes
phine. The dose should then be titrated to
in whom the surface area for gas exchange is
achieve symptom relief, with no set upper
reduced and the caliber of the airways is
limit. Benzodiazepines can help with the
decreased, increasing the likelihood of
anxiety associated with dyspnea; lorazepam
obstruction. Continuous positive airway
0.05 mg/kg (maximum 2 mg) can be given
pressure and bilevel positive airway pressure
every 4 to 8 hours for this purpose.
are useful in situations in which the pressure
A variety of nonpharmacologic interven-
necessary to maintain airway patency is too
tions have been shown to decrease the dis-
high to allow for efficient exhalation.
tress associated with dyspnea. These
In the setting of respiratory failure with-
include:
out hypercapnia, supplemental oxygen is
1. Cool air blown on the face
the most appropriate intervention to facil-
2. Chest wall percussion
itate gas exchange. It is often better tolerated
3. Relaxation therapy and counseling
than NIPPV devices, especially by young
4. Proper positioning
children, facilitating pulmonary vasculature
5. Limitation of activities
dilatation and inhibiting cerebral artery
6. Cooler room temperature
dilatation, a frequent cause of headaches.
7. Avoidance of respiratory irritants
It is important to remember, however, that
(tobacco smoke and cleaning fluids)
patients with hypercapnic respiratory fail-
ure frequently rely on a “hypoxic drive” to
Most experts agree that these work best as an
maintain respiration. Provision of supple-
adjunct to pharmacologic therapy; they are
mental oxygen to these patients can lead to
rarely effective alone.
hypopnea or apnea.
Therapies to decrease the sensation of
Conclusion
dyspnea
In end-of-life settings respiratory failure is
Unfortunately, many questions remain
often the ultimate cause of death and the
about how to best care for children who are
main goal of therapy is minimizing the
destined to die before they reach adulthood.
distress resulting from the sensation of dys-
These include practical issues such as how to
pnea as this occurs. This goal can be
best manage distressing symptoms such as
achieved with a combination of pharmaco-
those listed above, how to best communicate
logic and nonpharmacologic therapies.
with children about the dying process and
The two most effective classes of drugs
involve them in decisions regarding their
for minimizing the perception of dyspnea
care, and how to avoid aggressive end-of-life
are opioids and benzodiazepines. Morphine
therapies that only delay death and prolong
is the opioid used most often for this pur-
suffering instead of extending life. After the
pose, but fentanyl and hydromorphone
child’s death, families must continue to move
(Dilaudid) have been shown to be effective
forward, raising questions about how best to
276
Chapter 29
help them deal with the bereavement that
Questions
inevitably accompanies such an event. It is
* As Julie’s physician, are you comfortable
fortunate that research into these questions
with her parents’ approach?
has expanded greatly over the past few years,
* Should Julie’s parents prevent her from
and it is likely that answers to these questions
knowing information relevant to her
will soon be available in the medical litera-
disease?
ture. The children we care for, but cannot
* How do you reconcile your responsibility
save, deserve no less.
to be forthright with Julie while simulta-
neously being respectful of her parents’
request?
Case study for review
* If you believe that her parents’ approach
is not in Julie’s best interest, how might you
Julie, a 13-year-old girl with widely meta-
proceed?
static alveolar rhabdomyosarcoma diag-
* Should children with life-limiting ill-
nosed 6 months earlier continues to have
nesses be involved in end-of-life decisions?
extensive bony disease, including possible
new metastatic lung lesions. Julie’s treat-
Understanding familial adaptation toward
ment has been complicated by chronic
the dying process is imperative. As health-
pain and a challenging family situation.
care providers, our goal is to anticipate and
Although Julie lives with her mother, her
identify problems and to assist dying chil-
father oversees her care. Her parents do not
dren and their families as they adjust to and
get along with one another. They frequently
cope with the emotional responses, which
argue and scream in her presence, which
are part of the dying process.
clearly upsets her.
Many children, parents, and physicians
Since diagnosis, her father has adamantly
are hesitant to discuss death and dying so
refused to let caregivers have one-on-one
that opportunities for planning how to cope
conversations with Julie. He insists on
are often missed. Failure to prepare ade-
remaining the only one to relate to Julie
quately for death can deprive families of the
any disease-related information, as he
chance to enjoy what time they have left
knows her best and can gauge how she will
with one another.
react. He has stated that Julie need not know
Each child and family grieve in their own
that she will likely die of her disease and he
unique way, determined by personal experi-
has forbidden the medical team from dis-
ences with death, religious and cultural
cussing issues related to prognosis.
backgrounds, and individual makeup.
Julie is to undergo a CT-guided lung
Dying is a process and all parties need to
biopsy to determine the nature of the lung
learn to live with dying or, as stated by the
lesions. Her parents state that she is already
American Academy of Pediatrics, “the goal
anxious enough and do not want the med-
[of palliative care] is to add life to the
ical team to tell her about the biopsy or the
child’s years, not simply years to the child’s
possibility that her disease has progressed.
life” [1].
Instead, they ask that Julie be told that she is
Parents’ natural tendency to protect
to be sedated only for a “scan.” Moreover,
their children influences the amount of
should she prove to have new disease, they
information children receive and the degree
do not want Julie to be informed and ask
to which they are involved in decision
that her caregivers not discuss end-of-life
making about their care [2]. Some parents
matters with her.
feel that by giving their child a choice their
Palliative Care
277
responsibilities are being usurped, while
important to follow the child’s lead [11].
others perceive including their sick child
In other words, answer what children ask
in decisions as overly burdensome. For
and on their terms. Often, rather than being
many parents, the sicker their child, the
direct, a child’s question or statement may
more they assume decisional priority and
be suggestive of something they are uncom-
attempt to minimize, veto, or even preclude
fortable asking and therefore it is vital to
their child from having a role in decisions at
determine what their underlying intention
all. Seemingly, such an approach is under-
is and not to give too much or too little
standable; however, parents need to be
information or answer the wrong question.
aware of the consequences of not preparing
Finally, failure to inform and involve chil-
their children for medical outcomes that are
dren can lead to feelings of isolation and
inevitable, such as anxiety, stress, and
distress [12].
confusion.
Ideally, discussions should occur early
Children frequently know more about
and routinely and physicians should reassess
their disease than for which they are credited.
how the child and parents understand the
Even when kept in the dark by parents
plan and goals of care. This helps the dying
and providers, children often know that
child and parents make appropriate deci-
the endpoint of a life-threatening illness is
sions. Discussions should be in language
death [3-5]. As children appreciate certain
that parents and children can understand
facts about their diseases and know that they
and be presented gently, accurately, and
are dying, it stands to reason that they should
repeatedly. Doing so will help minimize
be involved in decisions about their care so
misunderstandings and defuse conflicts.
that they can voice their preferences [6,7].
The challenge is to do so in a way that is
Therefore, the question we ask ourselves
both sensitive and respectful of the child’s,
should not be “should I tell,” rather it should
parents’, and providers’ needs, needs that
be “how do I tell?” Increasingly, research
are often in conflict with one another.
supports the need for direct communication
According to the Institute of Medicine
between parents, physicians, and children
(IOM), “conflicts may . . . be productive or
with life-limiting illnesses. This includes dis-
beneficial”
[11]. Confronted with a dis-
cussions relating to prognosis and even
agreement, parties tend to engage in a more
death [1,2,8,9]. Hinds et al. [10] found that
in-depth discussion allowing for a greater
children with life-limiting cancer between
appreciation of each other’s position, and,
ages 10 to 20 years were capable of partici-
in the process, each side may become more
pating in end-of-life decision making.
sensitive to the other’s values and concerns.
Physicians must be aware of the unique
Mack et al. [13] surveyed parents of
barriers to a child’s ability to participate in
children with cancer and found that par-
decision making as it relates to the dying
ents rated the quality of care provided by
process. The first question we must ask is,
physicians more favorably when physicians
“does this child have the ability to understand
communicated directly with children
that he/she is dying and what dying entails.”
(when appropriate). Similarly, in a survey
As children mature, their intellectual and
of more than 400 Swedish parents of chil-
emotional understanding of serious illness
dren who died of cancer, Kreicbergs
and the prospect of death matures too.
et al. [14] found that none of the parents
Discussions about death should be honest
who spoke with their child about death
and take into consideration the child’s
regretted doing so, whereas more than
emotional and developmental level. It is
one-quarter of parents who did not speak
278
Chapter 29
with their child regretted not doing so. The
consider temporary steps that accommo-
latter parent group had higher levels of
date each party’s goals, and (3) periodically
anxiety and depression than parents who
reevaluate each party’s views concerning
did speak to their children.
the goals of care and the options to achieve
Parents are not alone in valuing the
those goals [11].
importance of direct communication
between children and physicians. Recent
research has shown that children with can-
Case resolution
cer consider direct communication
Since Julie’s diagnosis, her father has peri-
between doctors and children more impor-
odically sought the guidance of the unit’s
tant than any other aspect [15]. Nearly 40
social worker. Prior to the lung biopsy,
years ago, following parental approval,
Julie’s father agreed to meet with her
Nitschke et al. [4] began including children
oncologist and the social worker in the
aged 5 years and older who were near death
latter’s office, a place where he is comfort-
from cancer in end-of-life discussions.
able speaking. Following a brief update by
They found that the majority of children
the oncologist of Julie’s current medical
and parents found the child’s inclusion a
status, the social worker asked her father
positive experience. They also reported that
to share his concerns about Julie not being
some children from whom information
included in disease-related discussions. Her
was withheld experienced fear and isolation
father stated that Julie is “just a kid,” and
prior to dying.
that it was “his job to protect, and to decide
what is best for her.” He recounted his
Conclusions and practical
limited parental role in Julie’s life prior to
suggestions
her cancer diagnosis and blamed himself for
Situations like Julie’s do not lend themselves
not being more present in her life, admitting
to easy solutions. Dishonesty, evading the
that Julie’s illness and likely death “terrify”
question at hand, inconsistencies, and white
him and that he is resolute not to abandon
lies are to be avoided at all costs. Deception
her again.
is difficult to maintain. When the truth is
The social worker asked Julie’s father
ultimately discovered, the patient-parent-
what he believes she knows of her disease
physician relationship is often irrevocably
and its prognosis. He replied that Julie
damaged, an unfortunate circumstance in
knows she has cancer and that it is
any situation, but particularly for a child like
“treatable,” and nothing more. She then
Julie who is nearing the end of life. Such
asked if he would allow Julie to join them,
action may lead Julie to withdraw and even
which would permit the oncologist (in his
to experience feelings of isolation and fear,
presence) to answer any questions she may
let alone profoundly disrespect her devel-
have about the “scan” and to clarify what
oping autonomy.
she indeed knows. Hesitant at first, he
By helping to facilitate, clarify, and
ultimately agreed. After greeting and
resolve areas of contention, pediatricians
thanking her for joining them, the oncolo-
can be extremely helpful. As appreciated by
gist asked Julie if she has any questions
the IOM, the following steps may be useful:
about the upcoming CT scan. Julie asked
(1) postpone decisions to allow for time to
why she has to have another CT when
think about concerns and to discuss goals,
reevaluation scans, including a chest CT,
(2) rather than making definitive decisions
were just completed 1 week ago. Directing
that offer no room for compromise,
her words to the oncologist, Julie asked,
Palliative Care
279
“is something wrong?” Before the oncolo-
Freyer DR. Care of the dying adolescent: special
gist or her father could reply, Julie stated,
considerations. Pediatrics 113:381-388, 2004.
Himelstein BP, Hilden JM, Boldt AM, Weissman
“I’ve been online talking to other kids with
D. Pediatric palliative care. N Engl J Med
cancer. A few of them said that the only
350:1752-1762, 2004.
reason to repeat a scan so soon is to see
Kazak AE, Rourke MT, Alderfer MA, et al.
if the disease is back.” Without hesitation
Evidence-based assessment, intervention and
and still speaking to her oncologist, Julie
psychosocial care in pediatric oncology: a
said, “I know that most kids with my kind of
blueprint for comprehensive services across
cancer die.”
treatment. J Pediatr Psychol 32:1099-1110,
The social worker encouraged Julie and
2007.
her father to speak openly and freely. Ten-
Liben S, Papadatou D, Wolfe J. Paediatric palli-
tatively, and with the support of the oncolo-
ative care: challenges and emerging ideas.
Lancet 371:852-864, 2008.
gist and social worker, they began to speak
Wolfe J, Grier HE, Klar N, et al. Symptoms and
to one another and share their respective
suffering at the end of life in children with
concerns and goals. Following their conver-
cancer. N Engl J Med 342:326-333, 2000.
sation, Julie’s father cautiously agreed to
include Julie in future discussions. The
oncologist assured both Julie and her father
References
that although he remains hopeful, her dis-
ease is difficult to cure and that she might
1.
American Academy of Pediatrics. Committee
indeed die of her cancer. However, the
on Bioethics and Committee on Hospital
medical team would continue to treat both
Care. Palliative care for children. Pediatrics
her cancer and her pain, all along adhering
106:351-357, 2000.
to her and her parents’ wishes.
2.
Bluebond-Langer M. The Private Worlds of
Successful care of the dying child
Dying Children. Princeton, NJ: Princeton
requires that healthcare providers acknowl-
University Press, 1978.
3.
Hilden JM, Watterson J, Chrastek J. Tell the
edge families’ emotions, assure families that
children. J Clin Oncol 18:3193-3195, 2000.
their responses are normal, provide a bal-
4.
Nitschke R, Meyer WH, Sexauer CL, et al.
anced and honest perspective, and commu-
Care of terminally ill children with cancer.
nicate frequently and consistently.
Med Pediatr Oncol 34:268-270, 2000.
We would like to acknowledge and thank
5.
McConnell Y, Frager G, Levetown M. Deci-
Yoram Unguru, MD, MS, MA, for writing
sion making in pediatric palliative care. In:
this case and instructor guide. Dr. Unguru is
Carter B, Levetown M (eds), Palliative Care
an attending physician in the Division of
for Infants, Children, and Adolescents: A
Pediatric Hematology/Oncology, The Her-
Practical Handbook. Baltimore: JHU Press,
man and Walter Samuelson Children’s Hos-
2004.
6.
Informed consent, parental permission, and
pital at Sinai, and Berman Institute of Bio-
assent in pediatric practice. Committee on
ethics, Johns Hopkins University.
Bioethics, American Academy of Pediatrics.
Pediatrics 95:314-317, 1995.
7.
Spinetta JJ, Masera G, Jankovic M, et al. Valid
Suggested Reading
informed consent and participative decision-
making in children with cancer and their
Feudtner C. Collaborative communication in
parents: a report of the SIOP working com-
pediatric palliative care: a foundation for
mittee on psychosocial issues in pediatric
problem-solving
and decision-making.
oncology. Med Pediatr Oncol 40:244-246,
Pediatr Clin N Am 54:583-607, 2007.
2003.
280
Chapter 29
8. Wolfe J, Hinds P, Sourkes B (eds). Textbook
12. Sourkes B. Armfuls of Time: The Psycholog-
of Interdisciplinary Pediatric Palliative Care.
ical Experience of the Child with a Life-
Philadelphia: Elsevier, 2011.
Threatening Illness. Pittsburgh: University
9. Himelstein BP, Hilden JM, Morstad Boldt A,
of Pittsburgh Press, 1995.
Weissman D. Pediatric palliative care. N Engl
13. Mack JW, Hilden JM, Watterson J, et al.
J Med 350:1752-1762, 2004.
Parent and physician perspectives on quality
10. Hinds PS, Drew D, Oakes LL, et al. End-of-
of care at the end of life in children with
life care preferences of pediatric patients with
cancer. J Clin Oncol 23:9155-9161, 2005.
cancer. J Clin Oncol 23:9146-9154, 2005.
14. Kreicbergs U, Valdimarsdottir U, Onelov E,
11. Field MJ, Behrman RE (eds). When children
et al. Talking about death with children who
die: improving palliative and end-of-life care
have severe malignant disease. N Engl J Med
for children and their families. Committee on
351:1175-1186, 2004.
Palliative and End-of-Life Care for Children
15. Unguru Y, Sill A, Kamani N. The experiences
and Their Families. Board on Health Sciences
of children enrolled in pediatric oncology
Policy, Institute of Medicine. Washington,
research: implications for assent. Pediatrics
DC: National Academies Press, 2003.
125:e876-e883, 2010.
Chemotherapy Basics
30
Chemotherapeutic agents have multiple
Erwinia (Erwinase
). Asparaginase is a nat-
mechanisms of action and many potential
urally occurring enzyme produced by many
side effects. In order to be prepared for the
microorganisms. In the L- and PEG-aspar-
potential complications, we discuss the most
aginase forms, the enzyme is produced by
common pediatric agents, their mechanisms
Escherichia coli, whereas the Erwinia form
of action, the types of malignancies for which
is produced by Erwinia chrysanthemi. PEG-
they are utilized, and finally, the most likely
asparaginase is a modified form of L-aspar-
side effects. We discuss how best to monitor
aginase with a much longer half-life resulting
for these adverse events, and how to manage
from its covalent binding to PEG, allowing
them should they occur. Note that side effects
the same therapeutic effect from fewer doses.
from chemotherapy are numerous and we
Intramuscular asparaginase is the current
detail some of the more common ones rather
standard of care due to concerns about severe
than provide a comprehensive list; further
anaphylaxis with intravenous (IV) use; how-
reading may be required in appropriately
ever, two current Children’s Oncology
diagnosing and managing a patient. Emeto-
Group (COG) protocols are investigating the
genic potential of common pediatric agents is
safety and efficacy of IV PEG-asparginase.
included in Chapter 25. The majority of
Less frequent dosing with PEG-asparaginase
agents are dosed depending on the patient’s
is important for patient comfort and con-
body surface area (BSA) rather than weight,
venience until intravenous asparaginase
especially for patients older than 1 year of
becomes more widely accepted. Since Erwi-
age. Multiple formulas exist for the calcula-
nia is produced by a different microorgan-
tion of BSA; we prefer the Mosteller formula:
ism, it is immunologically distinct and can be
utilized in patients that have had a hyper-
rffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffifi
HeightðcmÞ WeightðkgÞ
sensitivity reaction to the E. coli forms.
BSA ¼
3600
Mechanism of action
L-asparagine is a nonessential amino acid
Asparaginase
that cannot be synthesized by malignant cells
of lymphoid and myeloid origin. Asparagi-
Asparaginase comes in three forms: L-
nase depletes L-asparagine from leukemic
asparaginase (Elspar
), polyethylene glycol
cells by catalyzing the conversion of
(PEG)-asparaginase
(Oncaspar
),
and
L-asparagine to aspartic acid and ammonia.
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
282
Chapter 30
Utilized in
Utilized in
Acute lymphoblastic leukemia
Germ cell tumors
Acute myelogenous leukemia
Hodgkin lymphoma
Non-Hodgkin lymphoma
Side effects, monitoring, and
treatment
Side effects, monitoring, and
Bleomycin can cause infusional fever and
treatment
chills followed later by mucositis, as well as
The most immediate side effect from aspar-
pruritus and excoriation leading to hyper-
aginase compounds is an allergic reaction
pigmentation. Raynaud’s phenomenon can
that can range from local irritation to ana-
also commonly occur later, especially with
phylaxis. Patients should be monitored
combination chemotherapy. Patients may
closely for at least
1
hour after Elspar
occasionally complain of rash, dysgeusia,
injection, and 2 hours following Oncaspar
and anorexia. Due to the formation of
injection. Based on the level of allergic
oxygen-free radicals, patients are at risk for
symptoms, treatment can range from an
dose-dependent pneumonitis and rarely
antihistamine such as diphenhydramine to
pulmonary fibrosis and therefore should
treatment of anaphylaxis with steroids, an
be monitored with pulmonary function
antihistamine, an H2 blocker (i.e., raniti-
testing, especially carbon monoxide-diffus-
dine), and epinephrine. A common later
ing capacity (PFT/DLCO). An anaphylac-
side effect is coagulopathy secondary to
toid-type reaction may also rarely occur.
decreased synthesis of antithrombin III
(ATIII), fibrinogen, and other clotting fac-
tors. Currently, routine monitoring of ATIII
Cisplatin/Carboplatin
and fibrinogen and repletion of low levels
are not recommended without clinical
Cisplatin was the first of the platinum-based
symptoms. Rare side effects that should
chemotherapeutic agents. Carboplatin has
be considered include pancreatitis, throm-
less severe side effects, in particular decreased
bosis, and seizures.
nephrotoxicity and ototoxicity. It is more
myelosuppressive, however.
Bleomycin
Mechanism of action
The platinum agents, like the alkylators,
Bleomycin is obtained as a mixture of
cause interstrand DNA cross-linking. In
antibiotics isolated from Streptomyces
addition, they bind to replicating DNA
verticillus.
causing single-strand breaks.
Mechanism of action
Utilized in
Bleomycin leads to the formation of oxy-
Brain tumors
gen-free radicals that cause single-strand
Germ cell tumors
and double-strand DNA breaks. In addi-
Hepatoblastoma
tion, bleomycin leads to cellular degrada-
Hodgkin lymphoma
tion of cellular RNA. Bleomycin is cell cycle
Neuroblastoma
specific, targeting the G2 and M phases, thus
Osteogenic sarcoma
inhibiting cell growth and division, espe-
Soft tissue sarcomas
cially in rapidly dividing cells.
Wilms tumor
Chemotherapy Basics
283
Side effects, monitoring, and
measurements of the glomerular filtration
treatment
rate and likely platinum dose reduction.
Infusional nausea and vomiting are common
as platinum agents have an extremely high
emetogenic potential. This is followed later
Cyclophosphamide/Ifosfamide
by myelosuppression. Electrolyte abnormal-
ities including hypokalemia, hypomagnese-
Cyclophosphamide (Cytoxan) and ifosfamide
mia, hypocalcemia, and hyponatremia are
are alkylating agents and structural analogues.
common and the patient may develop Fan-
They are related to nitrogen mustard.
coni syndrome (diminished reabsorption of
solutes by the proximal tubule resulting in
hypophosphatemia, metabolic acidosis, and
Mechanism of action
secondary hypokalemia). Nephrotoxicity
Cyclophosphamide and ifosfamide require
and ototoxicity
(high-frequency sensori-
conversion by the hepatic P450 system to
neural hearing loss) occur occasionally with
their active form that ultimately leads to
carboplatin but are more common and
the intracellular release of two compounds,
severe with cisplatin.
acrolein and phosphoramide mustard.
Ototoxicity must be monitored with
Phosphoramide mustard causes interstrand
serial audiograms during therapy. Due to
DNA cross-linking.
the risk of electrolyte abnormalities, chem-
istry panels including magnesium should be
Utilized in
followed daily while receiving platinum-
Acute lymphoblastic leukemia
based therapy. In addition, due to the risk
Acute myelogenous leukemia
of nephrotoxicity, the patient’s intake and
Brain tumors
output should be followed closely and be
Ewing sarcoma
well-matched. If it appears the patient is
Germ cell tumors
developing a negative fluid balance, addi-
Hodgkin and non-Hodgkin lymphoma
tional fluids should be provided. If the
Neuroblastoma
patient develops a positive fluid balance,
Osteogenic sarcoma
mannitol should be used to increase urine
Soft tissue sarcomas
output. Renal function should be moni-
Wilms tumor
tored closely, as a reduction in the glomer-
ular filtration rate will necessitate a dose
reduction. In general, for pediatric patients
Side effects, monitoring, and
with no previous history of nephrotoxicity,
treatment
serum creatinine can serve as a measure of
Cyclophosphamide and ifosfamide com-
renal function utilizing the Cockcroft-Gault
monly cause nausea, vomiting, and anorexia
equation:
with drug infusion. Myelosuppression, im-
munosuppression, and alopecia are common
later adverse effects. Sterility is dose depen-
Estimated creatinine clearance
dent
(greatest with cumulative doses of
Age½years Þ MassðkgÞ
½0:85ðif femaleÞ
¼ð140
cyclophosphamide >7.5 g/m2) and occurs
72
serum creatinineðmg=dLÞ
more commonly in pubertal males (though
prepubertal males and females are also at
Decrease in the estimated creatinine
risk). Due to the risk of nephrotoxicity,
clearance should lead to more formal
particularly with ifosfamide, patients should
284
Chapter 30
be well-hydrated prior to and after infusion.
cycle specific, killing cells during synthesis
Though the syndrome of inappropriate
(S phase); it therefore specifically targets
diuretic hormone (SIADH) is rare, patients
rapidly dividing cells.
should be monitored for appropriate urine
output by following their intake and output
Utilized in
as well as urine specific gravity. Electrolytes
Acute lymphoblastic leukemia
are also routinely followed for hyponatremia.
Acute myelogenous leukemia
If there are signs of fluid retention, the patient
Hodgkin and non-Hodgkin lymphoma
should initially be given increased hydration
followed by diuresis with furosemide or
Side effects, monitoring, and
mannitol if hydration is ineffective.
treatment
Excretion of acrolein into the urine can
Cytarabine commonly causes nausea,
cause hemorrhagic cystitis. The addition of
vomiting, and anorexia immediately after
MESNA for bladder protection when the cyclo-
infusion. Later, myelosuppression, stomati-
phosphamide dose exceeds 1.0 g/m2/day has
tis, and alopecia are common. Although not
significantly decreased the frequency of this
common, the patient should be monitored
adverse event. MESNA specifically binds to
for Ara-C syndrome that includes fever,
acrolein and other toxic metabolites in the
myalgias, bone pain, malaise, conjunctivitis,
urine to detoxify them and protect the bladder
maculopapular rash, and occasionally chest
wall. The urine should be monitored for the
pain. High-dose IV cytarabine requires pro-
presence of occult blood during and after in-
phylaxis with dexamethasone eye drops to
fusion and, if positive, should be examined
prevent conjunctivitis. A flu-like syndrome
microscopically. If red blood cells are noted
with fever, chills, and rash occurs occasion-
on microscopy, hydration should be increased.
ally with cytarabine; infection must still be
Of note, MESNA can cause a false positive test
ruled out with bacterial cultures. Streptococ-
for ketones on urine dipstick. Finally, metab-
cus viridans sepsis and acute respiratory
olism of ifosfamide leads to formation of chlor-
distress syndrome are possible after high-
oacetaldehyde, a byproduct thought responsi-
dose cytarabine, therefore the addition of
ble for nephrotoxicity as well as the neurotox-
vancomycin with fever or clinical deterio-
icity seen occasionally with ifosfamide (som-
ration should be strongly considered. Intra-
nolence, depressive psychosis, and confusion).
thecal (IT) cytarabine similarly can cause
Thiamine prophylaxis may have benefit in a
fever, nausea, and vomiting in addition to
patient with prior neurotoxicity.
headache. More serious and immediate side
effects including arachnoiditis, somnolence,
meningismus, convulsions, and paresis are
Cytarabine (Ara-C)
rare but should be considered as clinically
indicated.
Cytarabine, also known as cytosine
arabinoside or Ara-C
(arabinofuranosyl
cytidine), is utilized in the treatment of
hematologic malignancies.
Dactinomycin (Actinomycin-D)
Mechanism of action
Dactinomycin is an antibiotic compound
Cytarabine is an antimetabolite that inhibits
isolated from Streptomyces parvullus, similar
DNA polymerase. In addition, it is cell
to the anthracycline class.
Chemotherapy Basics
285
Mechanism of action
rather than an alcohol. Idarubicin lacks a
Dactinomycin intercalates with DNA, inhi-
methoxy group which increases its lipophi-
biting RNA and DNA synthesis. In addition,
licity as compared with other anthracyclines.
dactinomycin interacts with topoisomerase,
which is required for DNA replication, and
Utilized in
leads to single-strand DNA breaks.
Acute lymphoblastic leukemia
Acute myelogenous leukemia
Utilized in
Hepatoblastoma
Soft tissue sarcomas
Hodgkin and non-Hodgkin lymphoma
Wilms tumor
Neuroblastoma
Osteogenic sarcoma
Soft tissue sarcomas
Side effects, monitoring, and
Wilms tumor
treatment
Infusional nausea and vomiting are com-
mon, followed later by alopecia and mye-
Side effects, monitoring, and
treatment
losuppression. Anorexia, fatigue, diarrhea,
Anthracyclines commonly cause nausea and
and mucositis also occur occasionally.
vomiting with treatment. Due to the color of
Radiation recall can occur in patients who
the infusion, the patient should be warned
previously received radiation therapy.
that urine, saliva, tears, and sweat may all
have a pink or red coloring. After treatment,
Daunorubicin/Doxorubicin/
myelosuppression, alopecia, and mucositis
Idarubicin
commonly occur. Due to the production
of free radicals during electron reduction
Daunorubicin and doxorubicin are both
of anthracyclines, patients are at risk for
anthracycline antibiotics isolated from
dose-dependent cardiotoxicity occurring as
Streptomyces species (S. coeruleorubidus and
a late finding. Echocardiographic monitor-
S. peucetius, respectively). Idarubicin is an
ing is required during and after treatment at
analogue of daunorubicin used less com-
regular intervals based on the total anthra-
monly in pediatric oncology.
cycline dose received as well as concomitant
radiation therapy to the chest. Cardioprotec-
Mechanism of action
tion with continuous versus rapid infusion as
Anthracyclines as a group are cytotoxic to
well as agents such as dexrazoxane remains
malignant cells due to nucleotide base inter-
controversial and is not yet routinely recom-
calation and cell membrane lipid-binding
mended. Dexrazoxane may have benefit
activity. Nucleotide intercalation inhibits
when patients receive high cumulative doses
replication as well as DNA and RNA poly-
of anthracyclines (i.e.,
300 mg/m2). Addi-
merases. The anthracyclines also interact
tionally, anthracyclines provide different
with topoisomerase II, which is vital for
cumulative toxicity, and dose conversion
DNA replication. Cell membrane binding
based on doxorubicin isotoxic dose equiva-
affects a variety of cellular functions. In addi-
lents must be performed as below:
tion, electron reduction of the anthracyclines
Doxorubicin: multiply total dose
1
produces free radicals leading to DNA dam-
Daunorubicin: multiply total dose
0.833
age and lipid peroxidation. Daunorubicin
Epirubicin: multiply total dose
0.67
differs from doxorubicin structurally as the
Idarubicin: multiply total dose
5
side chain terminates in a methyl group
Mitoxantrone: multiply total dose
4
286
Chapter 30
Radiation recall is also a potential rare
Mechanism of action
complication of anthracyclines when given
Imatinib is a selective inhibitor of the tyro-
after radiation therapy.
sine kinase activity of the BCR-ABL fusion
protein, a product of the Philadelphia chro-
mosome (reciprocal translocation of chro-
Etoposide
mosome 9 and 22) seen mainly in chronic
myeloid leukemia and rarely with acute
Etoposide is derived from podophyllotoxin,
lymphoblastic leukemia.
a toxin found in the American mayapple.
Utilized in
Acute lymphoblastic leukemia (Philadelphia
Mechanism of action
chromosome-positive)
Etoposide binds to topoisomerase II, which
Chronic myelogenous leukemia
is vital for DNA replication, leading to DNA
strand breakage. Etoposide is cell cycle spe-
Side effects, monitoring, and
cific and appears to act mainly on the G2
treatment
and S (synthesis) phases, thus targeting
Imatinib has multiple potential side effects.
rapidly dividing cells.
Common immediate effects include fluid
retention, nausea, and diarrhea. Later com-
Utilized in
mon effects include fatigue, muscle cramps,
Acute myelogenous leukemia
rash, arthralgias, and myelosuppression.
Brain tumors
Ewing sarcoma
Germ cell tumors
Irinotecan
Hodgkin and non-Hodgkin lymphoma
Hemophagocytic lymphohistiocytosis
Irinotecan is a semisynthetic analog isolated
Neuroblastoma
from the plant alkaloid Camptotheca
Osteogenic sarcoma
acuminata.
Soft tissue sarcomas
Wilms tumor
Mechanism of action
Irinotecan in its active form is a potent
Side effects, monitoring, and
inhibitor of topoisomerase I which is vital
treatment
for DNA replication. Inhibition of topo-
Infusional nausea and vomiting is common
isomerase inhibits replication and also leads
followed by myelosuppression and alopecia.
to DNA damage.
Although rare, the patient should be mon-
itored closely for hypotension and anaphy-
Utilized in
laxis during the infusion. In addition to
Brain tumors
fluid support, the infusional rate can be
Hepatoblastoma
slowed if the patient develops hypotension.
Soft tissue sarcomas
Side effects, monitoring, and
Imatinib (Gleevec )
treatment
Patients receiving irinotecan can suffer from
Imatinib was the first successful targeted
cholinergic symptoms including intestinal
drug therapy for oncology patients.
hyperperistalsis that can lead to abdominal
Chemotherapy Basics
287
cramping and early diarrhea. Patients should
leukemia and dietary deficiency of folic acid
be monitored closely for early diarrhea; if
could improve leukemia symptoms.
it occurs loperamide should be given prior
to an anticholinergic (e.g., atropine). Other
Mechanism of action
cholinergic symptoms that may occur with
Methotrexate inhibits folic acid by prevent-
drug administration include rhinitis, in-
ing the reduction of folic acid by the enzyme
creased salivation, miosis, lacrimation, dia-
dihydrofolate reductase. This inhibition
phoresis, and flushing. Common later side
subsequently limits the synthesis of purines
effects include diarrhea, alopecia, transami-
and DNA. Some of the methotrexate meta-
nitis,
neutropenia,
mucositis,
and
bolites also lead to DNA damage.
hyperbilirubinemia.
Utilized in
Acute lymphoblastic leukemia
Mercaptopurine (6-MP)
Acute myelogenous leukemia
Brain tumors
Mercaptopurine is a purine analog.
Non-Hodgkin lymphoma
Osteogenic sarcoma
Mechanism of action
Mercaptopurine is converted into several
Side effects, monitoring,
active metabolites that inhibit RNA and DNA
and treatment
synthesis. As a purine analog it can also
Because of significant enterohepatic circula-
interfere with purine biosynthesis. Mercap-
tion of methotrexate, transaminitis is com-
topurine is converted to nucleotide metabo-
mon. Nausea, vomiting, and anorexia may
lites, some of which can lead to DNA toxicity.
also occur; therefore, oral doses are generally
given at bedtime. Many side effects of meth-
Utilized in
otrexate are due to delayed clearance after
Acute lymphoblastic leukemia
high-dose IV treatment. Aggressive hydration
Non-Hodgkin lymphoma
and alkalinization of fluids can improve renal
clearance. Drugs including trimethoprim-
Side effects, monitoring, and
sulfamethoxazole, penicillin, nonsteroidal
treatment
Mercaptopurine is generally well tolerated,
anti-inflammatories (NSAIDs), and proton
pump inhibitors (PPIs) can competitively
although myelosuppression occurs com-
monly. Mercaptopurine is usually adminis-
inhibit renal clearance and must be held
during high-dose therapy. Leucovorin
tered at night as patients may occasionally
complain of anorexia, nausea, and vomit-
(folinic acid) is used to decrease many of the
toxic effects of folic acid antagonists such as
ing. Many foods can decrease absorption
and therefore administration should be
methotrexate. Leucovorin can participate in
metabolic reactions requiring folic acid with-
separated from meals. Diarrhea and an ery-
thematous rash may also occasionally occur.
out the necessity of reduction by dihydrofo-
late reductase which is inhibited by metho-
trexate. This mechanism of action can also
Methotrexate
counteract the therapeutic effect of metho-
trexate; therefore, leucovorin should not be
Methotrexate was the first successful chemo-
started until 18 to 24 hours after the meth-
therapeutic agent utilized in children after
otrexate infusion has been completed. For
the observation that folic acid worsened
this reason, patients should be advised to not
288
Chapter 30
take folic acid supplements during metho-
Mechanism of action
trexate therapy as this may similarly reduce
Steroids have a variety of actions on the
efficacy.
body. Although not completely understood,
Urine pH should be monitored closely
steroids are thought to destroy lympho-
and kept above 7.5 during high-dose ther-
blasts by binding to the cortisol receptor
apy to facilitate renal excretion of metho-
found on lymphoid cells and specifically in
trexate. The patient’s intake and output
large number on lymphoblasts. Steroids are
should also be followed. Methotrexate levels
immunosuppressive and target T-lympho-
are usually drawn starting 24 hours after the
cytes, monocytes, and eosinophils. Steroids
infusion is started. Leukemia protocols pro-
may also function by halting DNA synthesis.
vide a methotrexate nomogram that allows
Dexamethasone has better central nervous
the practitioner to determine if methotrex-
system (CNS) penetration than prednisone
ate levels are declining appropriately or
and has benefit in preventing CNS relapse
are in a toxic range requiring an increase
in ALL.
in hydration and leucovorin frequency or
dose. Rarely, renal failure occurs and can
be managed with carboxypeptidase G2.
Utilized in
Toxicity is typically worse after delayed
Acute lymphoblastic leukemia
clearance and includes severe mucositis and
Hemophagocytic lymphohistiocytosis
myelosuppression. IT methotrexate often
Hodgkin and non-Hodgkin lymphoma
causes nausea and headache. Arachnoiditis
Langerhans cell histiocytosis
occasionally occurs and the patient should
be monitored for symptoms including
fever, vomiting, and meningismus. Long-
Side effects, monitoring, and
term cognitive dysfunction and learning
treatment
disabilities occasionally occur. Leukoence-
Steroid treatment results in a multitude of
phalopathy and progressive cognitive dete-
side effects. Common ones include hyper-
rioration rarely occur, especially in adoles-
phagia, insomnia, personality changes,
cents and adults, with high-dose IV meth-
adrenal suppression, acne, immuno-
otrexate and accentuated by cranial radia-
suppression, and Cushing’s syndrome.
tion therapy. Management of patients with
Occasional side effects include gastritis,
neurotoxicity including seizures, confusion,
hyperglycemia, poor wound healing, facial
ataxia, cranial nerve palsies, speech disor-
erythema, striae, thinning of the skin, mus-
ders, and paraparesis is controversial. Prac-
cle weakness, osteopenia, and cataracts.
titioners may recommend leucovorin rescue
Avascular necrosis of various joints, most
after future IT doses or replacement with IT
commonly hips, knees, and ankles, occurs
cytarabine, with or without hydrocortisone.
rarely. It is more common in adolescents,
Conclusive evidence is lacking on the use of
and females are at higher risk. It appears to
dextromethorphan as a neuroprotectant.
occur more commonly with dexametha-
sone than with prednisone. Hypertension
occurs rarely; blood pressure should be
Steroids
monitored closely in addition to blood
glucose. Patients should be on an H2
Dexamethasone and prednisone are the
blocker
(i.e., ranitidine) while receiving
steroids used most commonly as chemo-
daily steroids to prevent gastritis and peptic
therapeutic agents.
ulcer disease.
Chemotherapy Basics
289
Temozolomide
Although rare, the patient should be moni-
tored for transaminitis as well as signs of veno-
Temozolomide (Temodar ) is an oral alky-
occlusive disease
(now termed sinusoidal
lating agent.
obstructive syndrome) and hepatic fibrosis.
Mechanism of action
As with other alkylators, temozolomide
Topotecan
leads to DNA interstrand cross-linking.
Like irinotecan, topotecan is a semisynthetic
Utilized in
analog isolated from the plant alkaloid
Brain tumors
C. acuminata.
Side effects, monitoring,
Mechanism of action
and treatment
Topotecan is a potent inhibitor of topo-
Temozolomide commonly causes anorexia,
isomerase I that is vital for DNA replication.
nausea, vomiting, and constipation fol-
Inhibition of topoisomerase inhibits repli-
lowed by myelosuppression. Occasionally,
cation and also leads to DNA damage.
temozolomide may cause abdominal pain,
diarrhea, headache, and mucositis. Contin-
Utilized in
uous low-dose (metronomic) therapy may
Brain tumors
be effective in some patients with a
Neuroblastoma
decreased side effect profile.
Side effects, monitoring,
Thioguanine (6-TG)
and treatment
Topotecan can commonly cause nausea,
Like mercaptopurine, thioguanine is an oral
vomiting, diarrhea or constipation, fever, pain
antimetabolite and purine analog.
and later myelosuppression, fatigue, and alo-
pecia. The patient should also be monitored
for the occasional findings of headache, rash,
Mechanism of action
hypotension, transaminitis, and mucositis.
Metabolites of thioguanine interfere with
purine synthesis and DNA replication. The
intercalation of nucleotide metabolites into
DNA also leads to DNA strand breaks.
Vincristine/Vinblastine
Utilized in
Vincristine and vinblastine are alkaloids
Acute lymphoblastic leukemia
isolated from the Madagascar periwinkle
Acute myelogenous leukemia
plant
(Vinca rosea, now Catharanthus
Non-Hodgkin lymphoma
roseus) and therefore often referred to as
vinca alkaloids. Vincristine and vinblastine
Side effects, monitoring,
are structurally identical except for a single
and treatment
substitution (a formyl group in vincristine is
Thioguanine commonly leads to myelosup-
replaced by a methyl group in vinblastine),
pression and occasionally causes fatigue,
which leads to significant differences in their
nausea, vomiting, diarrhea, and anorexia.
cytotoxic effects.
290
Chapter 30
Mechanism of action
commonly causes neurotoxicity including
The vinca alkaloids bind to microtubules
constipation and loss of deep tendon
especially in the mitotic spindle leading to
reflexes. Both can lead to alopecia. Jaw
metaphase arrest, thus targeting rapidly divid-
pain, peripheral paresthesias, wrist and
ing cells. They have other disruptive cellular
foot drop, and abnormal gait occasionally
functions that may or may not be related to
occur, especially with vincristine. Ptosis,
their effects on tubulin and microtubules.
vocal cord dysfunction, and damage to the
eighth cranial nerve (clinically with dizzi-
Utilized in
ness, nystagmus, vertigo, and hearing loss)
Acute lymphoblastic leukemia
are rare findings. Vinca alkaloids are vesi-
Brain tumors
cants; therefore, extravasation, if it occurs,
Ewing sarcoma
can lead to local ulceration (see Chapter 32
Hepatoblastoma
for management of extravasation).
Hodgkin and non-Hodgkin lymphoma
Langerhans cell histiocytosis
Neuroblastoma
Suggested Reading
Soft tissue sarcomas
Wilms tumor
Committee on Shortening the Time Line for New
Cancer Treatments. National Cancer Policy
Side effects, monitoring,
Board. In: Adamson PC, Weiner SL, Simone
and treatment
JV, Gelband H (eds), Making Better Drugs for
Vinblastine more commonly causes mye-
Children with Cancer. Washington, DC:
losuppression, whereas vincristine more
National Academies Press, 2005.
Guide to Procedures
31
Patients with suspected or known oncologic
cells, and white blood cells in addition to
or hematologic disease undergo procedures
the supporting matrix to allow for cell growth
to obtain valuable diagnostic information
and maturation. Aspirates are routinely
and receive certain therapies. Traditionally,
obtained for morphologic as well as immu-
tumor biopsies are performed under general
nohistochemical and flow cytometric evalu-
anesthesia by pediatric surgeons, pediatric
ation in patients with suspected leukemia.
orthopedic oncologists, and pediatric inter-
Those with suspected or known solid tumors,
ventional radiologists. Pediatric hematol-
storage diseases, aplastic anemia, or other
ogy/oncology physicians and their trained
marrow failure states require evaluation by
staff perform lumbar punctures (LPs) to
bone marrow biopsy as well. A core section of
look for involvement of the cerebral spinal
the marrow matrix is obtained in order to
fluid (CSF) by malignancy and to adminis-
assess the suitability of the marrow environ-
ter intrathecal (IT) chemotherapy. Intrathe-
ment for growth of normal cellular elements,
cal refers to the administration of a drug
the cellularity of the marrow, and the presence
directly into the subarachnoid space of the
of abnormal cells that may be adherent to the
spinal column. Drugs are administered in
trabeculae. Samples are typically taken from
this manner to bypass the blood-brain bar-
more than one site when looking for evidence
rier and therefore be more available for
of marrow involvement by solid tumors in
central nervous system
(CNS) directed
order to increase the sensitivity of the test.
therapy. Currently, there are only three
Basic principles for performing such
agents licensed for intrathecal chemother-
procedures include: (1) ensuring the proce-
apy: methotrexate, cytarabine (Ara-C), and
dure is indicated for diagnosis, assessment
hydrocortisone. Other common procedures
of response to therapy, or for possible
include bone marrow aspiration (BMA) and
relapse; (2) ensuring the proper medication
biopsy and administration of chemotherapy
is being administered at the proper time;
via a peripheral vein or Ommaya reservoir.
(3) providing a safe and sterile environment;
Bone marrow examination is required for
and, (4) obtaining informed consent with
the diagnosis of leukemia, lymphoma, bone
proper documentation. The patient’s med-
marrow failure states, evaluation of pancyto-
ical record should be carefully reviewed
penia of unknown etiology, suspected storage
prior to the procedure and the purpose and
diseases, and certain cases of anemia. The
nature of the procedure be reviewed with
bone marrow produces the cellular elements
the patient and family. A discussion should
of the blood including platelets, red blood
be held to review anticipated risks and
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
292
Chapter 31
benefits and documented in the medical
CNS lymphoma (treatment or assessment
record with the signed informed consent.
of relapse), CNS malignancy, and therapy
All procedures should be performed or
complications related to the CNS, specifi-
supervised by practitioners with technical
cally infection or neurotoxicity
(patients
expertise. For patients undergoing anesthe-
with suspected meningitis, encephalitis, or
sia, a skilled caregiver (i.e., anesthesiologist,
change in mental status without evidence of
nurse anesthetist, or critical care physician)
increased intracranial pressure).
should administer the sedation and monitor
the patient. In addition to the practitioners
Pretreatment evaluation
performing the procedure and sedation,
1. Review patient history for use of
another skilled caregiver (i.e., nurse or phy-
anticoagulants
(should be discontinued
sician) should be present to assist with
with appropriate time interval prior to pro-
positioning, sterile transfer of chemother-
cedure), seizures, or other CNS concerns
apy, and general patient care and monitor-
including prior complications with LPs or
ing. Many centers perform all LPs and
intrathecal chemotherapy. Review of sys-
BMAs under deep sedation or anesthesia;
tems to include history of headaches, altered
however, some patients prefer to have these
mental status, fevers, bleeding, back pain, or
procedures performed awake, or possibly
lower extremity weakness.
with mild sedation.
2. Physical examination should be per-
Prior to initiation of the procedure, a
formed prior to the procedure with special
time out should be done to properly identify
attention to a focused neurologic evaluation
the patient
(hospital wrist band, medical
with assessment for papilledema or other
record, and labels on medications) and
evidence of increased intracranial pressure
acknowledge the procedure and informed
such as high blood pressure, widened pulse
consent. All materials and medications
pressure, or altered level of consciousness.
should be brought into the room and
Evaluate vitals and assess for any evidence of
checked by at least two practitioners. If mul-
infection or metabolic abnormalities to
tiple medications are to be administered, care
ensure safety for anesthesia. Evaluate site
should be taken to avoid an error in admin-
of procedure to assure no localized infection
istration by bringing in one drug at a time.
or skin breakdown.
Standardized procedures should be followed
3. Laboratory parameters must be checked
to minimize error. Procedures may be per-
to confirm that the chemotherapy should be
formed in an outpatient or inpatient setting
given (platelet count, absolute neutrophil
by individuals who have been properly
count, and hemoglobin). A current meta-
trained and supervised. Many teaching hos-
bolic panel and coagulation tests should be
pitals will also provide a training opportunity
checked as indicated. Patients with moderate
for procedures and close supervision by an
thrombocytopenia
(platelet count
< 50
experienced practitioner is mandatory.
109/L) may need a platelet transfusion,
although this varies with the skill of the
practitioner and the purpose of the proce-
Lumbar puncture/intrathecal
dure (many practitioners prefer a platelet
chemotherapy
count
100
109/L for a diagnostic LP
in suspected or newly diagnosed leukemia).
Indications
4. The chemotherapy to be administered
Patients with suspected leukemia or lym-
should be checked on the label to ensure
phoma (staging), history of leukemia or
the correct drug, dose, mode of infusion,
Guide to Procedures
293
and patient identification. This should be
4. Prepare the skin surface in a sterile man-
compared to the patient’s medical record
ner with providone iodine solution begin-
and schema sheet to ensure appropriate
ning at the site of puncture, working out-
timing as well.
ward with friction. Repeat for a total of three
scrubs. The skin may then be cleansed with
Materials
alcohol. Place a fenestrated drape over the
1. Standard LP tray, 10 mL sterile syringe for
site and ensure an adequate sterile field with
chemotherapy transfer,
22 g spinal needle
additional drapes as desired.
(1.5 in. for infants, 2.5 in. for older children,
5. Have the assistant transfer the chemo-
longer needles available as needed for larger
therapy in a sterile manner to the sterile
patients), 25 gauge needle for lidocaine injec-
syringe on the tray. Open all CSF collection
tion if needed, providone iodine and alcohol,
tubes.
mask, and sterile gloves. A Quincke needle is
6. Administer local anesthetic with preser-
the standard needle used for an LP; however,
vative-free
1% lidocaine with a 25 gauge
the pencil point Whitacre needle may be
needle, if desired. Awake patients may prefer
indicated in patients with prior history of
application of a topical lidocaine anesthetic
severe spinal headache
(see below under
(LMX or EMLA) or ethyl chloride cold
Post-procedure monitoring).
spray. Local anesthesia is not necessary in
2. Chemotherapy agent(s) to be adminis-
the anesthetized patient.
tered. Errors have occurred in the past with
7. Insert the spinal needle into the midline of
vincristine being inadvertently administered
the interspace with the bevel up, directed at
intrathecally. This is almost uniformly fatal.
a slight (10-20 ) angle toward the umbilicus.
This drug should never be brought into the
Ensure the needle is perpendicular to the
room of a patient undergoing an LP with IT
surface. Advance the needle; if bony resis-
chemotherapy.
tance is noted, draw back and reposition. The
less experienced practitioner should check
Procedure
for CSF flow every 2 to 3 mm by withdrawing
1. Proper positioning is the key to a success-
the stylet and looking at the translucent
ful procedure. The lateral decubitus position
window hub for visualization of CSF. If
may be used for a sedated or small patient.
blood is returned, the needle should be
The patient should be placed on a firm bed,
removed and the procedure be reattempted
head flexed with chin to chest, and legs
one interspace higher. The most experienced
maximally flexed toward the head. Ensure
practitioner should assume responsibility for
the hips and shoulders are aligned and the
subsequent LPs.
back is straight. Alternatively, an awake
8. Once clear CSF return is established,
patient may prefer the sitting position, flexed
rotate the needle
90
counterclockwise
forward, and supported by the assistant.
(bevel in transverse plane) for patients in
2. Identify the landmark with palpation of
the lateral decubitus position to increase
the interspaces. The L4-5
interspace is
rapidity of CSF flow and allow for easier
located by a perpendicular line at the top
administration of the chemotherapy.
of the iliac crests; either this space or the one
Patients in the sitting forward position
above (L3-4) may be used.
do not usually need repositioning of the
3. Some centers require the person per-
needle. Obtain the desired amount of
forming the procedure to wear a mask to
CSF in 2 or 3 collection tubes. Tradition-
prevent infection. Put on sterile gloves and
ally the volume of CSF withdrawn should
set up the tray.
approximate the volume of medication
294
Chapter 31
administered. CSF should be collected in
Post-procedure monitoring
one tube for a chamber count (red blood
and complications
cell and white blood cell counts) and a
1. Observe the patient for 1 hour or until
second tube for cytology (assessment of
fully recovered from anesthesia with appro-
morphology). Additional tubes may be
priate postanesthesia monitoring. Most cen-
necessary for culture, immunohistochem-
ters direct that patients remain in a supine
istry, or any special studies such as myelin
position (without head pillow, mild Tren-
basic protein, tumor markers, or research
delenburg position) for 1 hour post-proce-
studies. Glucose and protein are checked
dure to minimize spinal headache and assist
initially for all patients, though not rou-
with dilution and flow of chemotherapy in
tinely for leukemia patients receiving
the CSF.
intrathecal chemotherapy.
2. Patients should be monitored for signs of
9. For intrathecal administration of che-
bleeding, pain at site, headache, nausea,
motherapy, attach the chemotherapy
vomiting, or change in neurologic status.
syringe to the spinal needle, ensuring no
In addition to monitoring for post-procedure
advancement or withdrawal of the spinal
complications, the patient’s cardiovascular
needle and a tight fit to avoid leakage. One
and respiratory status should be monitored
can attempt to withdraw CSF to assure
along with frequent vital signs until recovered
proper placement (will see a mix in the
from anesthesia.
syringe) but this may not be possible and
3. Headache occurs following lumbar
is not necessary. Slowly inject the chemo-
puncture in a small percentage of patients,
therapy over 1 to 2 minutes. There should
primarily adolescents and females. Typi-
be no resistance.
cally, onset of headache is within 12 hours
10. Chemotherapy should not be adminis-
to 5 days of the procedure and is due to a
tered when any of the following situations
slow leakage of CSF from the puncture site
exist: the spinal fluid is bloody indicating
(though may not be visible externally).
puncture of a vessel; the patient is moving
Other symptoms include nausea, vomiting,
not allowing for a safe procedure; the che-
dizziness, neck stiffness, light sensitivity,
motherapy does not advance easily and
and diminished hearing or vision. These
when the syringe is removed, the flow of
symptoms may be worse in a standing
CSF has stopped or greatly diminished; the
position.
chemotherapy drug or dose is incorrect; or
a. Patients are instructed to take fluids
the awake patient experiences pain with
liberally in addition to caffeine (highly
injection.
caffeinated sodas or coffee) with onset of
11. Remove the needle. Apply gentle pres-
symptoms and after subsequent LPs.
sure, cleanse as necessary with alcohol, and
b. Patients with severe headache or neu-
place a dry sterile dressing.
rologic symptoms should be evaluated
12. Assess patient for any adverse effect of
immediately. If a postdural puncture
the procedure. Have the patient in the
headache is suspected, treatment is ini-
supine position.
tiated with caffeine, fluids, and narcotics.
13. Label all CSF tubes and send to the lab.
If these steps are ineffective, a dural
14. Document the procedure in the medical
blood patch may be performed by an
record.
anesthesiologist to provide immediate
15. Follow up with the patient and family as
relief.
to the results of the procedure, including
c. Use of a pencil point spinal needle
interpretation of the CSF specimens.
(Whitacre) for subsequent LPs should be
Guide to Procedures
295
considered for these patients. These nee-
management of patients for whom it is dif-
dles are designed to spread the dural
ficult to perform LPs for any reason. They are
fibers and help reduce the frequency and
also used in patients who suffer a CNS relapse
severity of postdural headaches. They
of their leukemia and require frequent
can be more difficult to use and may
administration of intrathecal chemotherapy.
require an introducer to puncture the
Practitioners may opt to administer chemo-
skin and soft tissues.
therapy in reduced dosing compared to stan-
4. Fever may occur following administra-
dard intrathecal dosing (50% to 100% of IT
tion of intrathecal chemotherapy. Some
dosing) or give small daily dosing for up to
drugs (e.g., cytarabine) have been impli-
4 days (based on the concept of concentration
cated in causing fever; however, the patient
time to optimize therapeutic benefit).
should be assessed for presumed infection.
Frequently patients have indwelling central
Pretreatment evaluation
venous catheters and are receiving other
The same principles apply as in Lumbar
immunosuppressive therapy. Evaluation
puncture/intrathecal chemotherapy. Patients
should be comprehensive and may include
are not sedated for this procedure.
hospitalization for observation and admin-
istration of intravenous antibiotics.
Materials
5. Patients may experience pain or bleeding
1. Standard LP tray, two 5 mL sterile syrin-
at the LP site for several days. The patient
ges for chemotherapy transfer and CSF col-
should be evaluated and if experiencing
lection, 25 g butterfly needle, mask, razor,
minor bleeding, be treated with local therapy
antibacterial soap, 4
4 sterile gauze, pro-
(dry sterile bandaging) and pain medication
vidone iodine and alcohol, and two sets of
as needed. Prolonged or heavy bleeding
sterile gloves.
requires immediate evaluation including
2. Chemotherapy agent(s) to be adminis-
physical examination and imaging.
tered. Do not bring vincristine into the room.
6. Neurotoxicity may occur related to the
intrathecal medication or related to nerve
Procedure
damage secondary to the procedure.
1. The patient is placed in a supine or
Patients should be evaluated immediately
sitting position. If needed, the reservoir area
and intervention be taken as appropriate.
is shaved.
2. Topical anesthesia may be achieved with
lidocaine gels (EMLA or LMX) prior to the
Intra-Ommaya reservoir tap and
procedure.
injection of chemotherapy
3. While wearing a mask and sterile gloves,
the LP tray is set up.
Indications
4. Prepare the skin surface overlying the
See Lumbar puncture/intrathecal chemother-
reservoir site by cleansing with sterile gauze
apy. The Ommaya reservoir is an intraven-
moistened with an antibacterial soap in a
tricular catheter with a reservoir implanted
circular fashion, three times. Change gloves.
under the scalp that allows for administration
Continue with a routine sterile prep with
of chemotherapy or other medication directly
three providone iodine scrubs and alcohol
into the ventricular system, in addition to
wipes. Place a fenestrated drape over the site
facilitating sampling the CSF. Although they
and ensure an adequate sterile field with
are placed infrequently, they are useful in the
additional drapes as desired.
296
Chapter 31
5. Have the assistant transfer the chemo-
Bone marrow aspiration
therapy in a sterile manner to the sterile
and biopsy
syringe on the tray. Open all CSF collection
tubes.
Indications
6. Holding the reservoir firmly with one
The purpose of bone marrow aspiration
hand, puncture the reservoir site with a
or biopsy is to obtain tissue for diagnostic
25 g butterfly needle. The CSF is allowed
and staging evaluation of malignancies,
to drip (or is slowly withdrawn) from the
marrow infiltrative diseases, or marrow
butterfly into a sterile tube. The total
failure states.
volume collected should approximate the
total volume of chemotherapy and normal
Pretreatment evaluation
saline flush to be delivered. CSF should be
1. Review of systems including recent ill-
collected in one tube for a chamber count
nesses or back pain.
and a second tube for cytology. Additional
2. Physical examination should be per-
tubes may be necessary for culture, immu-
formed prior to the procedure, with vitals
nohistochemistry, or any special studies
and assessment for any evidence of infection
such as myelin basic protein, tumor mar-
or metabolic abnormalities to assure safety
kers, or research studies. Glucose and
for anesthesia. Evaluate site of procedure
protein are checked initially for all
to assure no localized infection or skin
patients, though not routinely for leuke-
breakdown.
mia patients receiving intra-Ommaya
3. Routine laboratory studies may include a
chemotherapy.
complete blood count, chemistries, and
7. The chemotherapy is injected over 2 to
coagulation studies. No specific platelet
3 minutes. Do not give the chemotherapy if
count is necessary for the procedure. The
the CSF is blood tinged.
anesthesiologist will want to ensure an ade-
8. The needle is removed and firm pressure
quate hemoglobin level and be aware of any
is applied to the site for several minutes. A
metabolic abnormalities.
spot bandage is applied.
4. The technician to assist with the marrow
9. Assess the patient for any adverse effects
slide preparation should be present at the
from the procedure.
start of the procedure with appropriate
10. Label all CSF tubes and send to the
materials.
lab.
11. Follow up with the patient and family as
to the results of the procedure, including
Materials
interpretation of the CSF specimens.
1. Biopsy tray with providone iodine,
alcohol,
4
4
gauze,
20 mL syringes
(2
Post-procedure monitoring
to
4 depending on samples to be col-
and complications
lected),
16 gauge bone marrow aspirate
1. Document in the patient’s chart details
needle, 11 gauge 4 in. or 13 gauge 3.5 in.
of the procedure, chemotherapy adminis-
Jamshidi biopsy needle
(or similar),
22
tered, specimens collected, and the patient’s
to
25 gauge needles, sterile gloves, and
status after the procedure.
Elastoplast adhesive or other pressure
2. See Post-procedure monitoring for LP
dressing.
regarding complications such as headache,
2. EDTA and heparin to anticoagulate sam-
fever, bleeding, or neurologic changes.
ples, lidocaine 1% for local anesthesia.
Guide to Procedures
297
Procedure
syringe, constant and strong pressure
1. Position patient in a prone or lateral
should be applied to draw up marrow into
decubitus position with a small lift under
the syringe. If awake, the patient may expe-
the hip to accentuate the posterior iliac
rience a shooting sensation down the legs at
crests. Identify the posterior superior iliac
the time of aspiration. Aspirate approxi-
spine. Have assistant secure patient’s posi-
mately 1 to 2 mL of marrow, detach the
tion. At times, other locations for bone
syringe carefully, and hand the specimen
marrow specimen collection are necessary
immediately to the technician for quick
(infants, children on ventilators, and so on)
visual inspection for marrow tissue
(fat,
and may include the anterior iliac spine,
spicules). Once this has been confirmed,
tibia, sternum, or other sites.
obtain additional marrow as needed with
2. Put on gloves and set up sterile tray.
the other primed syringes. Once complete,
3. Scrub the site with providone iodine,
remove the needle. If marrow is not visible
applying some friction, beginning at the
and the sample is thought to represent
site and moving outward, and repeating
peripheral blood, reposition the needle and
for a total of three scrubs. Repeat procedure
reattempt an aspirate.
with alcohol swabs. Allow to dry and apply
7. A bone marrow biopsy may be obtained
sterile drapes.
from the same side and skin puncture site as
4. Administer local anesthetic with a 22 to
the aspirate. The needle should be inserted
25 gauge needle. This is often done in the
into a fresh spot on the iliac spine (although
sedated patient to minimize post-procedure
should utilize the same skin puncture site).
discomfort. Local anesthesia is achieved
Holding the skin tight, insert the bone
with lidocaine 1% (2 to 3 mL depending
marrow (trephine) biopsy needle (with cut-
on size of the patient) injected down to and
ting trocar in place) holding at an angle
including the periosteum.
until through the skin, then placing perpen-
5. Prepare marrow aspirate and/or biopsy
dicular to the spine and inserting with
needles, ensuring the stylets are freely
strong, controlled pressure until the needle
removable. Prime syringes as per institu-
is firmly anchored into the cortex. Remove
tional protocol with heparin, EDTA, etc.
the trocar and holding the forefinger along
Ensure inner side of syringe is coated with
the needle at the desired depth of the core
anticoagulant, if being used.
biopsy (5 to 20 mm depending on size of the
6. Holding skin taught with outstretched
patient), insert the needle with a firm twist-
fingers, insert the bone marrow aspiration
ing pressure. The needle is then rocked in
needle (with stylet in place) initially at an
four angles (i.e., sideways as well as up and
angle to the skin and then perpendicular to
down) to break off the core marrow biopsy
the iliac spine once through the skin. With
sample at the base and then the needle is
gradual, controlled pressure and a gentle
removed. The provided push rod (no sharp
twisting motion, insert the needle into the
edge) is inserted into the sharp end of the
iliac spine. Once through the cortex, the
needle and the biopsy is pushed out gently
needle will “give” as it enters the marrow
onto sterile gauze. It is then examined to
space and a crunching sound or feeling may
ensure that adequate marrow tissue is pres-
be appreciated. The stylet should then be
ent. If so, it is given to the technician.
removed, and the needle should remain
8. Apply pressure for several minutes,
firmly in place. A 20 mL syringe is then
cleanse the site with alcohol swabs, and
firmly attached to the hub of the aspiration
apply a Band-Aid. If needed, a dry 4
4
needle. Holding onto both the needle and
folded into quarters may be applied over the
298
Chapter 31
Band-Aid with an elastic tape such as
practitioners, administration of push intra-
Elastoplast.
venous chemotherapy is a safe alternative to
9. Assess the patient for any adverse effect
the use of a CVC. Vincristine is frequently
of the procedure.
administered via peripheral vein in the out-
10. Document procedure(s) in the medical
patient setting during maintenance phases
record.
of treatment for acute lymphoblastic leuke-
11. Follow up with the patient and family to
mia. Peripheral access should not be used
ensure completion of procedure, explana-
routinely for infusional chemotherapy due
tion of any complications, review care of
to the difficulty in monitoring and ensuring
wound, and follow up on results of studies
continued patency of the IV during infu-
done on specimens obtained.
sion. Many chemotherapeutic drugs are
vesicants and may cause significant injury
Post-procedure monitoring
with extravasation (see Chapter 32).
and complications
1. Patients may be discharged from the
Pretreatment evaluation
recovery area after appropriate monitoring
1. Review the patient’s chemotherapy reg-
postanesthesia.
imen to determine what medications are to
2. The pressure dressing, if used, should be
be administered and at what dose. The
removed after 6 hours and the Band-Aid
patient’s height, weight, and total body
after 24 hours. The parents should inspect
surface area are verified with the dosage
the site for evidence of infection, bleeding,
calculations.
or other drainage. If noted, a practitioner
2. Ensure that required laboratory criteria
should be contacted and the site be evalu-
have been met (e.g., absolute neutrophil
ated. Pain medications including acetamin-
count, platelet count, and transaminases)
ophen, acetaminophen with codeine, or
as directed by the treatment protocol.
occasionally an intravenous narcotic may
3. Complete a physical examination on the
be administered for local pain (especially
patient and assess the adequacy of veni-
following biopsy). Patients may feel achy or
puncture sites.
bruised for several days following the pro-
4. Verify the labels on the syringe(s) of the
cedure. Discomfort can also be alleviated
drug(s) against the patient’s chart and
with a warm pack.
orders to ensure accuracy.
3. Patients may resume normal activities as
5. Prepare the patient for the procedure.
desired.
Explain the procedure, taking into account
the patient’s age, developmental status, and
Administration of peripheral
prior experience with the procedure. Elicit
chemotherapy
the patient’s help by encouraging him/her to
hold as still as possible. Enlist the assistance
Indications
of the parent(s) and staff (including child
Intravenous
(IV) chemotherapy may be
life) as needed. Explain each step as you go;
given into a central venous catheter (CVC)
be honest, thorough, and patient. Establish
or by peripheral vein administration in
a routine with each patient; many also like
patients without a CVC. Many patients may
to have the same practitioner if possible
have had their catheters removed due to
(confidence boosting).
infection, thrombosis, or electively in order
6. Patients desiring topical anesthesia
for the patient to resume more normal
should have a topical lidocaine gel (EMLA
activities. In the hands of experienced
or LMX) applied
30
to
60
minutes in
Guide to Procedures
299
advance of the procedure. This may result in
with clean gauze. Insert butterfly needle
vasoconstriction and increase difficulty of
with bevel pointed up. Advance needle until
access in some patients.
blood returns. When blood returns, remove
tourniquet, connect stopcock, and flush
Materials
line with 2 to 3 mL of saline.
1. Chemotherapy (premixed by pharmacy
4. If no sign of infiltration occurs (e.g., pain
in enclosed syringe).
or swelling) and blood return continues,
2.
25 g butterfly needle, stopcock, alcohol
administer chemotherapy slowly via IV
wipes, gauze
(2
2), tourniquet,
10 mL
push, checking intermittently every
5 to
saline flush, bandage, and gloves.
10 seconds for blood return or swelling.
When chemotherapy administration is
Procedure
completed, flush with remaining saline.
1. Assemble equipment; attach butterfly
5. Remove butterfly and apply pressure for
tubing to stopcock, attach chemotherapy
1 to 2 minutes with clean gauze. Apply
syringe to right side of stopcock, attach
bandage.
saline flush syringe to remaining junction
6. Document in the patient’s chart the
of stopcock, and flush stopcock and butter-
indication for and details of the proce-
fly with saline.
dure, medication, dosage, site, and any
2. Select an appropriate vein, preferably on
complications.
the dorsum of the hand or foot. Ask the
child to assume a comfortable position that
Post-procedure monitoring
also allows for easy access to the desired
and complications
vein. Avoid the antecubital fossae or joint
If infiltration has occurred, see Chapter 32.
spaces due to the possibility of deep extrav-
Areas that may have sustained tissue dam-
asation of chemotherapy with resultant
age should not be used for future adminis-
injury.
tration of chemotherapy. At times, subclin-
3. Clean the venipuncture site with alcohol
ical burns and scarring may occur and can
and apply tourniquet. Let air dry or wipe dry
appear as hyperpigmented areas.
Treatment of
32
Chemotherapy
Extravasations
Extravasation is the leakage of an intrave-
loss of the full thickness of the skin and,
nous (IV) drug into the surrounding tissues.
if severe, underlying structures.
Local reactions from extravasation of a ves-
icant chemotherapy agent can range from
The incidence of vesicant extravasation
mild pain and erythema to tissue necrosis,
injury ranges from 0.5% to 6% in peripheral
ulceration, and damage to tendons and
IV infusions and 0.3% to 4.7% in implanted
nerves. Cytotoxic drugs are classified as irri-
venous access port infusions, although real-
tants or vesicants (causing blisters), depend-
istically the incidence is likely higher as
ing on their potential for local toxicity.
many milder events may not be reported
* Extravasation of an irritant drug may
by patients. Treatment of an extravasation
cause an inflammatory reaction, with pain,
is determined by the particular chemother-
burning, tightness, or phlebitis at the nee-
apy agent involved, although the efficacy of
dle insertion site or along the vein. Clinical
such therapy may be modest. Prevention is
signs include warmth, erythema, and ten-
the key, and every possible measure should
derness in the area of extravasation, but
be taken to avoid such a complication. If it
there is no tissue sloughing or necrosis.
is anticipated that a patient will need fre-
Symptoms are typically of short duration
quent or prolonged infusions of vesicants, it
(days) and there are no long-lasting
is advisable to place a central venous cath-
sequelae. An irritant may cause a soft tissue
eter for safer drug administration. Rarely,
ulcer if a large amount of concentrated
extravasation may occur even with such
drug solution is inadvertently extravasated
a device.
causing an inflammatory reaction; again,
The causes of extravasation are multiple
this will not result in persistent tissue
and largely preventable. Factors that place
damage.
children at risk for peripheral IV extravasa-
* Extravasation of a vesicant drug may
tion include poor vein selection, multiple
cause tissue necrosis with a more severe
venous punctures to establish a patent
or lasting injury. Clinical signs and symp-
IV, obesity, dehydration, inability to report
toms may be similar to extravasated irri-
pain at the injection site, a moving
tants. Vesicant extravasation may result in
patient, and inexperience of the individual
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
Treatment of Chemotherapy Extravasations
301
administering the chemotherapy. Risk fac-
or cause pain. If this occurs, immediately
tors for extravasation from central venous
stop the infusion and attempt to withdraw
catheters include needle displacement, cath-
any medication in the needle/cannula.
eter migration, or fibrin sheath formation
* After infusing the medication, flush the
and thrombosis. Mechanical occlusions
vein with 3 to 10 mL of the carrier solution.
may be due to thrombus formation, drug
Do not continue to flush if resistance is met
precipitation, and positional catheter occlu-
or a local reaction suggesting a blown vein
sion or kinking of the line. Avoidance of
occurs.
extravasation depends on proper placement
and maintenance of IV access and frequent
Each incident of extravasation should be
monitoring. Recommendations to prevent
documented and reported.
extravasation are:
Although this complication is frequently
* Venipuncture and placement of the can-
encountered with antineoplastic agents, a
nula or other intravenous access performed
number of other drugs can also act as vesi-
by experienced personnel.
cants if extravasated into the surrounding
* Vesicants should be administered in
tissues. These noncytotoxic drugs include
accordance with the manufacturers’ recom-
alcohol, aminophylline, digoxin, nafcillin,
mendations (e.g., proper dilution and spec-
phenytoin, tetracycline, and total parenteral
ified administration time) with proper ver-
nutrition. Among cytotoxic drugs that cause
ification and identification
(patient and
extravasation injury, the anthracyclines are
protocol specific).
among the most important, both because of
* Avoid multiple venipunctures in the same
their widespread use in various chemother-
area. Do not use a vein distal to a recent
apeutic protocols and because of their abil-
venipuncture site as the vesicant may leak
ity to produce severe tissue damage and
from one of these proximal sites.
necrosis. The extent of tissue damage
* Instruct the patient to avoid movement.
depends on the chemotherapeutic agent’s
In young children, this may require addi-
binding capacity to DNA.
tional physical assistance for holding or
DNA-binding agents include anthracy-
coaxing with diversion techniques.
clines, antitumor antibiotics, platinum ana-
* Never use a previously placed IV access
logues, and some alkylating agents. These
device; a new IV cannula should be placed
drugs cause tissue damage by propagating
immediately prior to delivery of the
lethal DNA crosslinking or strand breaks
medication.
caused by free radicals, which lead to cell
* Choose a large, intact visible vein with
apoptosis. As the cells die, the drug is
good blood flow.
released and enters undamaged cells. This
* Use the smallest needle or cannula pos-
further increases the area of damage and
sible for venipuncture. Check the patency of
slows healing. Injuries from doxorubicin
the device by aspirating blood, as well as
may continue for weeks.
patency of the vein by flushing with the
Non-DNA-binding antineoplastics (vinca
carrier solution
(normal saline), before
alkaloids, taxanes, and topoisomerase inhibi-
administering the medication. Obtain a
tors) also function as vesicants by interfering
blood return prior to, and during, vesicant
with mitosis. These agents clear more easily
administration.
from extravasation sites and cause less dam-
* The intravenous infusion should flow
age than the DNA-binding agents. The tissue
freely without pressure. The local area
often resembles a chemical burn and heals
should not swell, become erythematous,
more quickly.
302
Chapter 32
Table 32.1 Extravasation treatment.
Drug
Local care
Pharmacologic treatment
DNA-binding vesicants
Anthracyclines
Cold pack, 20 minutes
Dexrazoxane 1000 mg/m2 IV within
Doxorubicin
4 times/day
48-72 hours
6 hour on day 1, repeat day 2;
Daunomycin
Elevate
500 mg/m2 day 3 or
DMSO 50% topical 1-2 mL within
10-25 minutes, then q8 hour
7-14 days; allow to dry, then
apply nonocclusive dressing
Do not use Dexrazoxane and DMSO
concurrently
Alkylating agents
Cold pack, 20 minutes
None
Nitrogen mustard
4 times/day
48-72 hours
Elevate
Other
Cold pack, 20 minutes
Sodium thiosulfate
Dactinomycin
4 times/day
48-72 hours
10% 2 mL in 6 mL sterile water for
Mitomycin C
Elevate
IV/SC injection
Dacarbazine
None
DMSO as above
Hydrocortisone 1% cream topically
Non-DNA binding vesicants
Vinca alkaloids
Warm pack, 20 minutes
None
Vinblastine
4 times/day
48-72 hours
Hyaluronidase 150 units in 1 mL
Vincristine
Elevate
injected SC in multiple sites with
Vendesine
small gauge needle
Taxanes
Cold pack, 20 minutes
Hyaluronidase as above
Docetaxel
4 times/day
48-72 hours
Paclitaxel
Elevate
Irritants
Alkylating agents
Carboplatin, cisplatin
Cold pack as above
Hydrocortisone 1% cream topically
Topotecan
Ifosfamide, cyclophosphamide
No local care
None
Melphalan
No local care
None
Antimetabolites
Cytarabine, fludarabine
None
None
Methotrexate, 5-
Cold pack as above
Hydrocortisone 1% cream topically
fluorouracil
Gemcitabine
None
None
Other
Bleomycin
None
None
Etoposide, irinotecan
Cold pack
Hydrocortisone 1% cream topically
Suggested as possible antidote in the literature; lack of prospective studies to currently advocate as
treatment. Abbreviation: SC, subcutaneous.
Treatment of Chemotherapy Extravasations
303
The signs and symptoms of extravasa-
Table 32.1) for which local heat is applied.
tion may be readily apparent with pain and
For other vesicant extravasations, including
erythema although it may take days for the
anthracyclines, initial treatment is geared
full extent of the epithelial damage to be
toward localizing and neutralizing the agent
evident. Discoloration and skin induration
with cold compresses, thus limiting its
may progress with the development of blis-
uptake into cells. The placement of extrav-
ters or necrosis and possibly ulceration or
asation kits that contain syringes and can-
deep tissue injury. Patients may develop
nulas, cold and hot packs, gauze pads, sterile
scarring or permanent hyperpigmentation
and chemoprotective gloves, and medica-
at the site of drug extravasation. These sites
tions to treat extravasation in locations
should not be utilized for subsequent
where chemotherapy is administered will
administration of chemotherapy or place-
facilitate early treatment (see Table 32.1).
ment of an IV device.
In addition to local care and possible phar-
Treatment should begin immediately
macologic antidotes, topical hydrocortisone
with discontinuation of the chemotherapy
1% and pain medications may alleviate local
and cooling or dilution of the site. Initial
discomfort.
treatment includes an attempt to aspirate
the vesicant with a 10 mL syringe. Clinicians
should work quickly to reduce morbidity
Suggested Reading
and avoid further patient harm. Manage-
ment of nonvesicant extravasation includes
Goolsby TV, Lombardo FA. Extravasation of
elevation and cooling and does not usually
chemotherapeutic agents: prevention and
include the use of pharmacologic therapy.
treatment. Semin Oncol 33:139-143, 2006.
An exception to the cooling technique is
Schulmeister L. Extravasation management.
extravasation with vinca alkaloids
(see
Semin Oncol Nurs 23:184-190, 2007.
Formulary
Sample entry:
drug in pregnant women may be acceptable
despite its potential risk.
Generic name
Trade and other names
X. Studies in animals or humans demon-
Drug category
strate fetal abnormalities or adverse reac-
How supplied
tion; reports indicate evidence of fetal risk.
Pregnancy category (see explanation below)
The risk of use in a pregnant woman clearly
Indications
outweighs any possible benefit.
Dosage
Notes, including adverse events, monitor-
ACYCLOVIR
ing, and dose modification
Zovirax
Pregnancy categories:
Antiviral
Capsules: 200 mg
A. Adequate studies in pregnant women
Tablets: 400, 800 mg
have not demonstrated a risk to the fetus
Suspension: 200 mg/5 mL
in the first trimester of pregnancy and there
Injection: 500 mg vial
is no evidence of risk in later trimesters.
B. Animal studies have not demonstrated a
Pregnancy category B
risk to the fetus, but there are no adequate
studies in pregnant women; or animal stud-
Indications:
ies have shown an adverse effect, but ade-
Treatment of initial, and prophylaxis for
quate studies in pregnant women have not
recurrent, mucosal and cutaneous herpes
demonstrated a risk to the fetus during the
simplex virus (HSV-1 and HSV-2) infections,
first trimester of pregnancy, and there is no
herpes simplex encephalitis, herpes zoster
evidence of risk in later trimesters.
infections, and varicella zoster infections.
C. Animal studies have shown an adverse
effect on the fetus, but there are no adequate
Dosage:
studies in humans; or there are no animal
reproduction studies and no adequate stud-
Children and neonates:
ies in humans.
Mucocutaneous HSV:
750 mg/m2/day IV
D. There is evidence of human fetal risk, but
divided q8h or
15
to
25 mg/kg/day IV
the potential benefits from the use of the
divided q8h for 5 to 10 days
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
306
Formulary
HSV encephalitis:
1500 mg/m2/day IV
m2/day IV q8-24h; maximum 600 mg/day,
divided q8h or
30
to
50 mg/kg/day IV
300 mg/dose
divided q8h for 10 days
Children >10 years of age to adult:
Neonatal HSV: 1500 mg/m2/day IV divided
600 to 800 mg/day PO q8-12h for the pre-
q8h or 30 to 50 mg/kg/day IV divided q8h
vention of acute uric acid nephropathy;
for 10 to 14 days
maximum 800 mg/day, 300 mg/dose or
200 to 400 mg/m2/day IV divided q8-24h;
Varicella zoster: 1500 mg/m2/day IV divided
maximum 600 mg/day, 300 mg/dose
q8h or 30 to 50 mg/kg/day IV divided q8h
for 7 to 10 days
Notes:
HSV prophylaxis: 750 mg/m2/day IV divided
Reduce dosage in renal impairment; discon-
q8h during risk period or 600 to 1000 mg/
tinue with rash (may be exacerbated with
day divided q6-8h during risk period
ampicillin or amoxicillin). Risk of hyper-
Notes:
sensitivity may be increased in patients
receiving thiazides/angiotensin-converting
Adjust dose in renal impairment. Adequate
enzyme inhibitors. May cause fever, neuri-
hydration and slow IV (1 hour) adminis-
tis, gastrointestinal disturbance, hepatotox-
tration are essential to prevent crystalliza-
icity, bone marrow suppression, and drows-
tion in the renal tubules. Oral absorption is
iness. Avoid concomitant use of amoxicillin,
unpredictable (15% to 30%). Use ideal body
ampicillin, mercaptopurine, cyclophospha-
weight for obese patients when calculating
mide, theophylline derivatives, and vitamin
dosage.
K antagonists.
ALLOPURINOL
ALTEPLASE
Zyloprim, Alloprim
Uric acid lowering agent, xanthine oxidase
Tissue plasminogen activator (TPA)
inhibitor, antigout agent
Activase, Cathflo Activase
Tablets: 100, 300 mg
Thrombolytic
Suspension: 20 mg/mL
Injection:
Injection (Alloprim): 500 mg vial
Cathflo Activase: 2 mg/2 mL vial
Activase: 50 mg (29 million units), 100 mg
Pregnancy category C
(58 million units)
Indications:
Pregnancy category C
Prevention of uric acid nephropathy in
myeloproliferative neoplastic disorders that
Indications:
may occur as a result of tumor lysis syn-
Treatment of recent severe or massive deep
drome (beginning 1 to 2 days prior to
vein thrombosis (DVT) or arterial throm-
initiation of chemotherapy); prevention of
bosis, pulmonary embolus, or occluded cen-
recurrent calcium oxalate calculi; preven-
tral venous catheter (CVC).
tion of gouty arthritis and nephropathy.
Dosage:
Dosage:
Pulmonary embolus, DVT, central venous
Children
10 years of age:
thrombosis, superior vena cava syndrome:
10 mg/kg/day PO divided q6-8h; maxi-
Systemic thrombolytic therapy should be
mum dose 800 mg/day or 200 to 300 mg/
given in consultation with a hematologist.
Formulary
307
Under 3 months of age: 0.06 mg/kg/hour for
AMIFOSTINE
6 to 24 hours
Ethyol
Over 3 months of age: 0.03 mg/kg/hour (max
Antidote for cisplatin, cytoprotective agent
2 mg/hour)
Injection: 500 mg vial
If no clinical improvement in 24 hours,
Pregnancy category C
double the dose to
0.06
to
0.12 mg/
kg/hour.
Indications:
Systemic TPA can be given for up to 96 hours.
A cytoprotective drug that scavenges free
In the case of a pulmonary embolus, bolus
radicals and binds to reactive drug deriva-
dosing of 1 mg/kg up to a maximum of
tives reducing toxicity of radiation and
50 mg may be given.
platinum-containing and alkylating agents
such as cisplatin, carboplatin, ifosfamide,
Maximum duration of therapy is 96 hours or
carmustine, melphalan, mechlorethamine,
based on the patient’s clinical course.
and cyclophosphamide.
Preferable to infuse thrombolytic agent dis-
tal to (and as close to) the site of thrombus
Dosage:
as possible.
Refer to individual protocol.
Occluded CVC:
Under 3 months of age:
0.25 mg IV (in
Usual dosage is 740 to 910 mg/m2 IV daily
0.5 mL)
over 15 minutes prior to the dose of a
Over 3 months of age: 0.5 mg IV (in 1 mL)
platinum or alkylating agent. Infusions over
Instill a dose in each lumen of the central
less than 15 minutes are associated with a
venous catheter; allow to dwell for 30 min-
higher incidence of adverse reactions.
utes. If unsuccessful, repeat dose. If the
catheter remains obstructed, begin systemic
Notes:
thrombolytic therapy as outlined above.
Avoid in hypotension or dehydration. Can
Purpura fulminans:
cause severe nausea and vomiting and usu-
0.5 mg/kg IV infusion over 1 hour, followed
ally requires antiemetic premedication.
by 0.25 mg/kg/hour over 3 hours (total dose
1.25 mg/kg over 4 hours)
AMINOCAPROIC ACID
Amicar
Notes:
Hemostatic agent
Tablet: 500, 1000 mg
Avoid central venous puncture and non-
Syrup: 250 mg/mL
compressible arterial sticks during infusion.
Injection: 250 mg/mL (20 mL)
Avoid use in excessive hypertension, within
10 days of a cerebral vascular accident, with
Pregnancy category C
gastrointestinal bleeding or trauma, within
1 week of surgery, in patients with a bleeding
Indications:
diathesis, or with suspicion of a subarach-
noid hemorrhage. Plasminogen levels
Treatment of bleeding resulting from exces-
should be followed daily and fresh frozen
sive activity of the fibrinolytic system.
plasma be given for replenishment if level
Typically used to treat mucosal-type bleed-
<50%.
ing in patients with bleeding diatheses (von
308
Formulary
Willebrand disease, mild hemophilia,
Rhizopus. Also used empirically to treat sus-
immune thrombocytopenia purpura).
pected invasive fungal disease in immuno-
compromised hosts with prolonged fever and
Dosage:
neutropenia. May be given intrathecally or
via bladder irrigation for localized therapy.
100 to 200 mg/kg IV/PO loading dose (max-
imum 10 g), followed by 50 to 100 mg/kg
Dosage:
q4-6h maintenance dose; maximum 30 g/24
Optional test dose: 0.1 mg/kg to a maximum
hour. Treat until symptoms resolve (1 to
of 1 mg IV over 20 to 60 minutes
14 days).
Initial dose: 0.5 to 1.0 mg/kg/day
Low doses (10 mg/kg) have been reported to
control bleeding in patients with thrombo-
The daily dose is increased by 0.25 mg/kg
cytopenia for long periods of time (weeks to
until the desired daily dose is reached. In
months).
critically ill patients, rapid escalation of
dosing may be needed.
Continuous IV infusion:
Empiric dose is 0.6 mg/kg/day
Loading dose of
100 mg/kg, then
10
to
33 mg/kg/hour in
5% dextrose in water
Therapeutic dose for confirmed invasive
(maximum 1.25 g/hour)
fungal infection is 0.6 to 1 mg/kg/day. Daily
infusion is over 2 to 6 hours, depending on
Notes:
the infusion tolerability. Salt loading with
10 to 15 mL/kg of normal saline prior to
May accumulate in patients with decreased
each infusion may prevent hypokalemia
renal function and require decreased dos-
and nephrotoxicity. Premedication is fre-
ing. Avoid in patients with disseminated
quently needed with acetaminophen and/
intravascular coagulation or hematuria.
or diphenhydramine, and if the patient
Increased risk of thrombosis with oral con-
experiences rigors, meperidine hydrochlo-
traceptives, estrogens, and factor IX or
ride may be given. Once therapy has been
prothrombin complex concentrates. May
established, alternate-day dosing may be
cause nausea, diarrhea, malaise, headache,
administered at a dose of 1 to 1.5 mg/kg/
decreased platelet function, and false
every other day.
increase in urinary amino acids.
AMPHOTERICIN B
Notes:
Fungizone, Amphocin
Because of the nephrotoxic potential of this
Polyene antifungal
drug, avoidance of other nephrotoxic med-
Injection: 50 mg vial
ications is advised, if possible. Monitor
daily electrolytes, renal and hepatic studies,
Pregnancy category B
and urine output. Common metabolic
abnormalities include hypokalemia, hypo-
Indications:
magnesemia, and hypocalcemia. Other pro-
Treatment of severe systemic infections and
blems that may occur are thrombocytope-
meningitis caused by susceptible fungi such
nia, hyperglycemia, diarrhea, dyspnea, back
as Candida species, Histoplasma capsulatum,
pain, and increases in transaminases and
Cryptococcus neoformans, Aspergillus species,
bilirubin. Common infusion-related toxici-
Blastomyces dermatitidis, Torulopsis glabrata,
ties are fever, chills, rigors, nausea, vomiting,
Coccidioides immitis, Mucormycoses, and
hypotension, and headache. Imidazole
Formulary
309
derivatives
(e.g., miconazole, fluconazole,
spleen, lung and liver, and therefore may be
and ketoconazole) may antagonize the
more beneficial in the treatment of hepa-
effect and induce fungal resistance to
tosplenic candidiasis. Cerebrospinal fluid
amphotericin.
(CSF) levels may be lower than with ampho-
tericin B or the liposomal compound.
AMPHOTERICIN B CHOLESTERYL
SULFATE
Dosage:
Amphotec
Usual dose is 2.5 to 5 mg/kg IV once daily
Polyene antifungal
over
2
hours. Rate should not exceed
Injection: 50,100 mg vial
2.5 mg/kg/hour.
Pregnancy category B
Notes:
Indications:
See Amphotericin B.
Treatment of invasive fungal disease in
patients who are refractory to or intolerant
AMPHOTERICIN B, LIPOSOMAL
of conventional amphotericin B.
AmBisome
Polyene antifungal
Dosage:
Injection: 50 mg vial
Start at
3 to 4 mg/kg/day and increase to
Pregnancy category B
6 mg/kg/day if necessary. A test dose of
10 mL of the diluted solution over 15 to
Indications:
30 minutes is recommended. Give first dose
Treatment of systemic or invasive fungal
at 1 mg/kg/hour; if well tolerated, the infusion
infection in patients refractory to or intoler-
time can be gradually decreased to 2 hours.
ant of conventional amphotericin B. Cere-
Notes:
brospinal fluid concentrations are higher
See Amphotericin B.
than with other amphotericin products.
Higher concentrations in the liver and spleen
AMPHOTERICIN B LIPID COMPLEX
than with conventional amphotericin B.
Abelcet
Polyene antifungal
Dosage:
Injection: 5 mg/mL (10, 20 mL)
Systemic fungal infections: 3 to 5 mg/kg/day
Pregnancy category B
IV over 2 hours. Doses as high as 10 mg/kg/
day have been used in patients with Asper-
Indications:
gillus species.
Treatment of aspergillosis or invasive fungal
Empiric therapy for fever and neutropenia:
infection in patients who are refractory to or
3 mg/kg/day
intolerant of conventional amphotericin B
Infusion may be shortened to 1 hour if well
therapy. Patients with acute or preexisting
tolerated.
renal toxicity (serum creatinine level double
that of baseline) should receive this product
Notes:
in lieu of conventional amphotericin B.
Higher concentrations are achieved in the
See Amphotericin B.
310
Formulary
ASPARAGINASE
Dose for Erwinia L-asparaginase is per
L-Asparaginase, (Elspar [native or Escheri-
protocol.
chia coli], Erwinase [Erwinia chrysanthemia])
Maximum 2 mL volume per injection site.
PEG-asparaginase
(polyethylene glycol
Many patients require multiple injections.
asparaginase; Oncaspar)
IM administration results in delayed peak
Antineoplastic, results in asparagine deple-
plasma concentration compared with IV
tion in malignant cells
administration.
Injection: 10,000 unit vial (Elspar)
Patients should be observed in a clinic or
10,000 unit vial (Erwinase)
hospital setting for at least 1 hour after admin-
3,750 unit vial (Oncaspar)
istration to monitor for a hypersensitivity
Pregnancy category C
reaction. Appropriate agents for treatment
of hypersensitivity should be readily available
(oxygen, epinephrine, antihistamines, intra-
Indications:
venous steroids) in addition to resuscitative
Treatment of acute lymphoblastic leukemia
equipment.
(ALL). Current and future trials for ALL treat-
Notes:
ment largely incorporate PEG-asparagase
during induction therapy due to an increased
Do not give to patients with prior significant
rapid early response compared to the native
pancreatitis associated with asparaginase.
form, prolonged half-life, and decreased like-
Hypersensitivity to E. coli or pegylated aspar-
lihood of developing neutralizing antibodies
aginase necessitates a switch to the Erwinia
during later phases of therapy. Due to the
form or omission of future doses. Patients
longer half-life of the pegylated form (5.8 days
who have had a significant hemorrhagic or
in children compared to 1.24 days with native
thrombotic event should have levels of fibrin-
and 0.65 days with Erwinase), longer intervals
ogen and antithrombin III checked and may
for therapy are suggested. Recent studies have
be rechallenged with future doses if with
established safety of the intravenous prepara-
improvement in their symptoms and nor-
tion, thereby decreasing the pain associated
malization of their laboratory parameters.
with intramuscular injection of the drug.
Use with caution in patients with hepatic
Erwinase has traditionally been utilized if
impairment or in those receiving other hep-
hypersensitivity develops to the native or
atotoxic drugs. Use with caution in patients
pegylated forms, although its availability may
receiving anticoagulation or nonsteroidal
be limited.
anti-inflammatory agents (NSAIDs). Aspar-
aginase may cause hyperglycemia, hyperuri-
cemia, hyperammonemia, hypofibrinogen-
Dosage:
emia, thrombosis, hemorrhage, anaphylaxis,
and hemorrhagic cystitis. Dexamethasone
Refer to individual protocol.
may alleviate allergic symptoms.
Usual dose in ALL of PEG-asparagase is
2500 IU/m2 IV/IM every
2
to
4
weeks
BEVACIZUMAB
(induction, delayed intensification, and
Avastin
interim maintenance phases).
Angiogenesis inhibitor; monoclonal anti-
Dose for native (E. coli) L-asparaginase is
body to vascular endothelial growth factor
6000 units/m2 IM three times a week for
Injection: 25 mg/mL
nine doses. High-dose protocols may give
15,000 to 20,000 IU/m2/dose.
Pregnancy category C
Formulary
311
Indications:
Indications:
Treatment of Hodgkin and non-Hodgkin
Treatment of metastatic colon cancer, lung
lymphoma, soft tissue sarcoma, renal cell car-
cancer, and glioblastoma multiforme. Cur-
cinoma, and germ cell tumor. Used as a scle-
rently being investigated in pediatric pro-
rosing agent to control malignant effusions.
tocols for the treatment of brain tumors,
refractory or recurrent solid tumors, and
neurofibromatosis.
Dosage:
Refer to individual protocol.
Dosage:
10 to 20 IU/m2 IV one to two times per week
Refer to individual protocol.
or once every 2 to 4 weeks.
15 mg/kg q2 weeks in 28-day cycles as a
Test dose recommended for lymphoma
single agent or
patients (1 to 2 units for the first 2 doses;
if well tolerated, give remainder of dose 1
5 to 10 mg/kg q2 weeks in combination
hour later). Administer over at least
10
regimens.
minutes, not to exceed 1 IU/minute.
May be given as continuous IV infusion 15
Notes:
to 20 IU/m2/day over 24 hours for 3 to 5
Avoid use in patients with recent surgery,
days in some protocols.
hemoptysis, gastrointestinal or central ner-
vous system bleeding, or any other serious
Notes:
bleeding. The interval required between
Do not give to patients with known hyper-
surgery and drug administration to avoid
sensitivity to bleomycin. Premedication
impairment in wound healing has not been
with acetaminophen, hydrocortisone, and
determined. General recommendation is to
antihistamine may decrease infusional tox-
allow 28 days prior to and following major
icity. Dose may need to be modified for
surgery. Use with caution in patients with
renal or pulmonary toxicity. Monitor renal
thrombocytopenia. An increased risk of
function studies and pulmonary function,
thromboembolic events have been reported
including forced expiratory volume in
1
in combination regimens. May impair fer-
minute, forced vital capacity, and carbon
tility and have adverse effects on fetal devel-
monoxide diffusion in lungs.
opment; adequate contraception must be
used during therapy. May cause infusional
toxicity (discontinue infusion until symp-
BUSULFAN
toms abate) or proteinuria. May potentiate
Myleran, Busulfex
cardiac effects of anthracyclines. Discon-
Antineoplastic
tinue therapy in patients who develop fis-
Tablet: 2 mg
tulas, hypertensive crisis, encephalopathy,
Injection: 6 mg/ml
or nephrotic syndrome.
Pregnancy category D
BLEOMYCIN SULFATE
Indications:
Blenoxane, generic
Treatment of chronic myelogenous leuke-
Antineoplastic antibiotic
mia (CML) and for marrow-ablative con-
Injection: 15 unit vial (1 unit ¼ 1 mg)
ditioning regimens prior to bone marrow
Pregnancy category D
transplant.
312
Formulary
Dosage:
Indications:
Treatment of pediatric brain tumors, neu-
Refer to individual protocol.
roblastoma, testicular tumors, relapsed leu-
For CML induction:
kemia, and solid tumors.
0.06 to 0.12 mg/kg once daily PO (or 1.8
to 4.6 mg/m2/day); titrate dose to maintain
Dosage:
a leukocyte count >40
109/L and discon-
tinue if the leukocyte count drops to
Refer to individual protocol. Many require
20
109/L.
dose calculation using the modified Calvert
formula to achieve the appropriate area
Hematopoietic stem cell transplant regimen:
under the curve for concentration over time
(dose based on actual body weight; adjust-
rather than dosing by body surface area. IV
ments based on therapeutic drug monitor-
administration over 15 minutes to 1 hour is
ing per protocol)
less toxic than bolus dosing.
12 kg: 1.1 mg/kg/dose q6h for 16 doses
Solid tumors:
over 4 consecutive days
400 to 560 mg/m2 every 3 to 4 weeks or per
>12 kg: 0.8 mg/kg/dose q6h for 16 doses
protocol
over 4 consecutive days
Brain tumor protocols:
Notes:
weekly for 4 weeks, then 2 week
175 mg/m2
Do not give to patients with known hyper-
recovery
sensitivity to busulfan; should not be used in
Dose is adjusted based on suppression of
pregnancy or while nursing. May cause
neutrophil and platelet counts or renal
severe bone marrow suppression. Use with
toxicity
caution with other myelosuppressive drugs
or radiation. May cause hemorrhagic cysti-
Bone marrow transplant preparative regimen:
tis. Use with thioguanine may increase
500 mg/m2/day for 3 days
hepatic toxicity. Patients with a known sei-
zure history or those at risk for seizures (e.g.,
Notes:
sickle cell disease) should be placed on a
Do not give to patients with known hyper-
prophylactic antiepileptic drug while receiv-
sensitivitytocarboplatin,cisplatin,orother
ing busulfan.
platinum-containing compounds. Ana-
Metabolism of busulfan may be inhib-
phylaxis may occur within minutes of
ited by antifungal agents (azoles), CYP3A4
administration. May cause hypotension,
inhibitors, dasatinib, and metronidazole
electrolyte abnormalities, nausea, hearing
leading to increased levels and effect.
loss, and peripheral neuropathy. Severe
Busulfan metabolism may be potenti-
marrow suppression or vomiting may
ated by CYP3A4 inducers, deferasirox, and
occur. Reduce dose in impaired renal func-
Echinacea, leading to decreased levels and
tion
(creatinine clearance
<60 mL/min)
effect.
utilizing the modified Calvert formula.
Aminoglycosides may increase serum
CARBOPLATIN
levels/effects of carboplatin and augment
Paraplatin-AQ, generic
ototoxicity and nephrotoxicity; nephro-
Antineoplastic, alkylating agent
toxic drugs may increase renal toxicity of
Injection: 50, 150, and 450 mg vials
carboplatin. Avoid concomitant adminis-
Pregnancy category D
tration of topotecan and taxanes.
Formulary
313
All patients should receive hydration
CASPOFUNGIN
prior to and following administration with
Cancidas
sodium chloride containing solution, with
Antifungal agent, echinocandin
or without mannitol and/or furosemide,
Injection: 50, 70 mg
to ensure good urine output and decrease
risk of nephrotoxicity. Reduce dose for
Pregnancy category C
young children
(<6 months) and with
Indications:
renal impairment. Electrolytes and magne-
sium should be monitored. Acute leukemia
Treatment of invasive aspergillosis in
has been reported as a second malignant
patients refractory or intolerant to ampho-
neoplasm.
tericin B (including lipid formulations) or
itraconzaole; candidemia, candidal intra-
CARMUSTINE
abdominal abscess, or esophageal candidi-
asis; empiric therapy of presumed fungal
BCNU
infection in febrile neutropenic patients.
BiCNU, Gliadel
Antineoplastic, alkylating agent
Dosage:
Injection: 100 mg vial
Preterm neonates to infants
<3 months:
Pregnancy category D
25 mg/m2/dose IV once daily
Indications:
Infants
3 months to adults: 70 mg/m2 load-
Treatment of brain tumors and Hodgkin
ing dose IV, followed by 50 mg/m2/dose IV
and non-Hodgkin lymphoma. Wafer
once daily
implant (Gliadel) may be an adjunct to
Patients may benefit from increased daily
surgery and radiation for glioblastoma mul-
dosing to 70 mg/m2 once daily dependent
tiforme and high-grade glioma.
on clinical status and response.
Empiric therapy should be given until neu-
Dosage:
tropenia resolves. In neutropenic patients,
Refer to individual protocol.
continue treatment for at least 7 days after
signs and symptoms of infection and neu-
Typical dose may be 200 to 250 mg/m2/dose
tropenia have resolved and at least 14 days
IV every 4 to 6 weeks.
following a positive culture in patients with
Bone marrow transplant conditioning regimen:
documented fungal infection.
300 to 600 mg/m2 divided into 1 to 6 doses,
infused over
2
hours and administered
Notes:
12 hours apart.
Modify dose in patients with hepatic
impairment. Drug interactions exist with
Notes:
cyclosporine, tacrolimus, and rifampin and
Do not give to patients with known hyper-
may require dose modification.
sensitivity tocarmustine.Severe, prolonged
(6 weeks) marrow suppression may neces-
CISPLATIN
sitate change in dosing for subsequent
Platinol, generic
cycles. Toxicity is cumulative and delayed
Antineoplastic, alkylating agent
pulmonary fibrosis may occur in patients
Injection: 50, 100, 150 mg vials
receiving 770 to 1800 mg/m2. Cimetidine
potentiates myelosuppressive effects.
Pregnancy category D
314
Formulary
Indications:
and decrease risk of nephrotoxicity. Reduce
Treatment of soft tissue sarcoma, osteosar-
dose for young children (<6 months) and
with renal impairment. Electrolytes and
coma, Hodgkin and non-Hodgkin lym-
phoma, brain tumors, germ cell tumor, and
magnesium should be monitored. Acute
leukemia has been reported as a second
neuroblastoma.
malignant neoplasm.
Dosage:
CLOFARABINE
Refer to individual protocol. Verify any
dosing schedule in which the cisplatin dose
Clolar
exceeds 120 mg/m2 per course.
Antineoplastic, antimetabolite
Injection: 1 mg/mL
Intermittent dosing schedule: 37.5 to 100 mg/
m2 every 2 to 3 weeks
Pregnancy category D
Daily dosing schedule: 15 to 20 mg/m2/day
for 5 days every 3 to 4 weeks
Indications:
Osteosarcoma and neuroblastoma:
60
to
Treatment of relapsed or refractory acute
100 mg/m2 once every 3 to 4 weeks
lymphoblastic leukemia, acute myelogenous
leukemia, myelodysplastic syndrome.
Bone marrow transplant: 55 mg/m2/day con-
tinuous infusion for 72 hours (total dose
Dosage:
165 mg/m2)
The rate of intravenous infusion is dose
Children (
1 year) to adults: 40 mg/m2/day
dependent and ranges from a 15- to 20-
for 5 days every 28 days or per protocol
minute infusion to a 6- to 8-hour infusion;
24-hour continuous infusions are also used.
Notes:
Cytokine release may develop into a sys-
Notes:
temic inflammatory response with resultant
Do not give to patients with known hyper-
capillary leak syndrome and organ failure. If
sensitivity to cisplatin or platinum-con-
this occurs, discontinue clofarabine and
taining compounds, preexisting renal
initiate therapy with diuretics, corticoster-
impairment, hearing impairment, and
oids, and albumin. If hypotension resolves
myelosuppression. Increased risk of neph-
without pharmacologic intervention, clo-
rotoxicity when given with other nephro-
farabine may be resumed. The risk for
toxic drugs (aminoglycosides and ampho-
hepatic toxicity and veno-occlusive disease
tericin B). Reduces renal elimination of
is increased in patients who have previously
methotrexate. Cisplatin may increase the
undergone hematopoietic stem cell trans-
levels/effects of aminoglycosides, taxane
plantion. Avoid concomitant use of neph-
derivatives, topotecan, and vinorelbine.
rotoxic and hepatotoxic drugs.
Effects or levels of cisplatin may be
increased with concomitant administration
CYCLOPHOSPHAMIDE
of loop diuretics.
Cytoxan, generic
All patients should receive hydration
Antineoplastic, alkylating agent
prior to and for 24 hours after administra-
Tablets: 25, 50 mg
tion with a sodium chloride containing
Injection: 500 mg, 1 g, 2 g vials
solution (with or without mannitol and/
or furosemide), to ensure good urine output
Pregnancy category D
Formulary
315
Indications:
Indications:
Treatment of Hodgkin and non-Hodgkin
Used with corticosteroids to prolong organ
lymphoma, acute leukemia, and neuroblas-
and patient survival in kidney, liver, heart,
toma. Conditioning therapy for bone mar-
and bone marrow transplants; treatment of
row transplant. Treatment of nephrotic syn-
aplastic anemia and other bone marrow
drome, systemic lupus erythematosus, and
failure syndromes.
other rheumatic diseases.
Dosage (Sandimmune):
Dosage:
Due to better absorption, lower doses of
Refer to individual protocol.
Neoral and Gengraf may be required com-
Acute lymphoblastic leukemia:
1000
to
pared to Sandimmune.
1200 mg/m2/dose, in consolidation and
Oral:
delayed intensification phases
Initial: 15 mg/kg single oral dose, beginning
Bone marrow transplant conditioning:
4 to 12 hours pretransplant
50 mg/kg/day IV for 3 to 4 days
Maintenance: 15 mg/kg/day PO for 1 to 2
Nephrotic syndrome: 2 to 3 mg/kg/day orally
weeks posttransplant, decrease by 5% per
for up to 12 weeks
week to 3 to 10 mg/kg/day
Systemic lupus erythematosus: 500 to 750 mg/
Intravenous:
m2 IV monthly; maximum dose 1g/m2
Initial: 5 to 6 mg/kg IV single dose, admin-
Juvenile idiopathic arthritis/vasculitis: 10 mg/
istered over 2 to 6 hours, beginning 4 to 12
kg IV every 2 weeks
hours pretransplant. Continue same IV dose
posttransplant until patient able to tolerate
Notes:
oral form.
Do not give to patients with known hyper-
Conversion from IV to PO dose (1:3 ratio):
sensitivity to cyclophosphamide. Dose
Multiply total daily IV dose by 3 and admin-
may need to be adjusted for myelosup-
ister in two divided oral doses per day.
pression or impaired renal function.
Mesna should be given with high-dose
Notes:
therapy (>1 g/m2/day) to reduce potential
Do not give to patients with known hyper-
of hemorrhagic cystitis. Allopurinol may
increase the myelotoxicity of cyclophos-
sensitivity to cyclosporine (castor oil is an
ingredient in the preparation). May cause
phamide by inhibiting its metabolism.
Ensure aggressive hydration with sodium
nephrotoxicity, hepatotoxicity, hypomag-
nesemia, hyperkalemia, hyperuricemia,
chloride containing fluids and frequent
bladder emptying.
hypertension, hursuitism, acne, gastrointes-
tinal symptoms, tremor, leukopenia, head-
CYCLOSPORINE
ache, and gingival hyperplasia. Use with
caution with concomitant administration
Sandimmune, Neoral, Gengraf
of other nephrotoxic drugs (e.g. amphoter-
Immunosuppressant
icin B, aminoglycosides, tacrolimus, acyclo-
Capsules: 25, 50, 100 mg
vir, NSAIDs). Requires close monitoring
Solution: 100 mg/mL
of renal and hepatic function and frequent
Injection: 50 mg/mL
determination of trough levels (drawn just
Pregnancy category C
prior to dose at steady state).
316
Formulary
Cyclosporine is a substrate for the
High-dose cytarabine:
(AML, relapsed or
cytochrome P450
3A4
oxidase system.
refractory ALL, non-Hodgkin lymphoma)
Drug interactions include ketoconazole,
3 g/m2/dose q12h for up to 12 doses
itraconazole, fluconazole, erythromycin,
For central nervous system (CNS) treatment
and methylprednisolone, which increase
and prophylaxis (intrathecal dose): Dosing
the cyclosporine concentration by inhibit-
per age
ing hepatic metabolism. Cimetidine may
Drug may be combined with other intra-
increase cyclosporine concentration. Refer
thecal agents such as hydrocortisone and
to the Physicians’ Desk Reference for more
methotrexate per protocol
extensive drug interaction information.
Children <1 year: 20 mg
CYTARABINE HYDROCHLORIDE
Children 1 to <2 years: 30 mg
Ara-C
Children 2 to <3 years: 50 mg
Cytosar-U, generic
Children
3 years and adults: 70 mg
Antineoplastic, antimetabolite
Injection: 100 and 500 mg, 1 and 2 g vials
Notes:
Pregnancy category D
Do not give to patients with known hyper-
sensitivity to cytarabine. May need to reduce
Indications:
dose with myelosuppression or hepatic dys-
function. Causes significant bone marrow
Treatment of acute lymphoblastic and
suppression. High doses have been associated
myelogenous leukemia (ALL and AML),
with gastrointestinal, CNS, pulmonary, and
refractory non-Hodgkin lymphoma, and
ocular toxicities as well as cardiomyopathy.
may be used in conditioning regimens for
May cause nausea, vomiting, mucositis, fever,
bone marrow transplantation. High doses
headache, somnolence, anorexia, alopecia,
penetrate the blood-brain barrier into the
conjunctivitis, ataxia, diarrhea, hepatic dys-
cerebrospinal fluid.
function, and peripheral neuropathy. Pro-
Dosage:
phylaxis with dexamethasone ophthalmic
drops may decrease effects of conjunctivitis.
Refer to individual protocol.
When prepared for intrathecal use,
Infants under 3 years:
added precautions should be taken when
3.3 mg/kg/day IV/SC for
4
days
(dose
other medications are also being delivered
reduced in children with Down syndrome)
to ensure appropriate labeling and handling
such that only medications intended for
Children and adults:
administration in the CNS are with the
Remission induction:
patient for the procedure.
200 mg/m2/day for 5 days at 2-week inter-
vals as a single agent; 100 to 200 mg/m2/
DACARBAZINE
day (or 2 to 6 mg/kg/day) for 5 to 10 days
DTIC
or every day until remission (as a part of
Antineoplastic
combination chemotherapy). Give IV con-
Injection: 100, 200 mg
tinuous infusion or every 12 hours.
Pregnancy category C
Maintenance:
1 to 1.5 mg/kg IM/SC as a single dose at 1- to
Indications:
4-week intervals or 70 to 200 mg/m2/day for
Treatment of Hodgkin lymphoma and solid
2 to 5 days at monthly intervals
tumors.
Formulary
317
Dosage:
Notes:
Do not give to patients with known hyper-
Refer to individual protocol.
sensitivity to dactinomycin. Avoid in infants
Solid tumors:
<6 months of age because of increased
200 to 470 mg/m2/day IV over 5 days every
adverse events. Use with caution in patients
3-4 weeks
with hepatobiliary dysfunction or who have
Neuroblastoma:
received radiation (radiation recall effect).
Reduce dosage in patients receiving concur-
800 to 900 mg/m2 IV as a single dose on day
rent radiation. Avoid extravasation. May
1 of the cycle every 3 to 4 weeks
cause myelosuppression, anorexia, vomit-
Hodgkin lymphoma:
ing, diarrhea, and stomatitis.
375 mg/m2 on days 1 and 15 of each course,
repeated every 28 days
DAPSONE
Generic
Notes:
Antibacterial
Do not give to patients with known hyper-
Tablet: 25, 100 mg
sensitivity to dacarbazine. Dosage reduc-
tion may be necessary in patients with
Pregnancy category C
renal or hepatic insufficiency. Drug extrav-
asation may result in tissue damage and
Indications:
severe pain.
Prevention and treatment of Pneumocystis
jiroveci pneumonia (PCP) in immunocom-
DACTINOMYCIN
promised hosts; treatment of Toxoplasma
Actinomycin D
gondii and Mycobacterium leprae.
Cosmegen
Antineoplastic antibiotic
Dosage:
Injection: 500 mg vial
Children 1 month to <12 years: 2 mg/kg/
Pregnancy category D
day or 4 mg/kg/dose once weekly (maximum
100 mg/dose daily or 200 mg/dose weekly)
Indications:
Children >12 years and adults: 100 mg/day
Treatment of Wilms tumor, rhabdomyosar-
or 200 mg once weekly
coma, Ewing sarcoma, ovarian germ cell
tumor, and gestational trophoblastic
Notes:
neoplasm.
Do not give to patients with known hyper-
sensitivity to dapsone. Dapsone is a strong
Dosage:
oxidizing agent and may cause hemolysis in
susceptible individuals. Screening for glu-
Refer to individual protocol. Note that
cose-6-phosphate dehydrogenase
(G6PD)
medication orders for dactinomycin may
deficiency is suggested in high-risk popula-
be written in micrograms (mcg) or milli-
tions (e.g., Mediterranean) and should not
grams (mg).
be used if positive. May be safe to use in
Children over 6 months to adult:
the African-American variant of G6PD defi-
15 mcg/kg/day or 400 to 600 mcg/m2/day once
ciency though some degree of hemolysis
daily for 5 days; repeat every 3 to 6 weeks.
may occur. May also cause marrow sup-
Higher doses are given in some protocols.
pression or methemoglobinemia. Drug
318
Formulary
interactions occur, primarily with rifampin.
Chapter 30). This may be an acute or late
May be given to immunosuppressed
effect and monitoring with ECHO or
patients who cannot tolerate or are allergic
MUGA and ECG is required per protocol.
to cotrimoxazole.
Secondary leukemia has been reported.
Extravasation may result in severe local
tissue necrosis and require intervention. A
DAUNORUBICIN HYDROCHLORIDE
transient red-orange discoloration of the
Cerubidine
urine, sweat, saliva, and tears may occur for
Antineoplastic antibiotic, anthracycline
up to 48 hours after a dose.
Injection: 20 mg vial
Pregnancy category D
DEFEROXAMINE MESYLATE
Desferal
Indications:
Chelating agent; antidote, iron toxicity
Treatment of acute lymphoblastic and acute
Injection: 500, 2000 mg
myelogenous leukemia (ALL and AML);
Pregnancy category C
used in combination with other chemother-
apeutic agents during ALL and AML induc-
Indications:
tion therapy.
Treatment of acute iron intoxication and
Dosage:
chronic transfusional iron overload.
Refer to individual protocol.
Dosage:
Infants <2 years or <0.5 m2should have dos-
Chronic iron overload:
ing based on body weight:
25 to 60 mg/kg/day SC/IV over 8 to 24
0.67 mg/kg/day IV with frequency depen-
hours; maximum 2 g/24 hours
dent on protocol
Children
2 years:
Notes:
25 to 60 mg/m2 IV with frequency depen-
Avoid in severe renal disease, anuria, or
dent on protocol
primary hemochromatosis. Prolonged use
can result in cataracts, decreased visual acu-
Notes:
ity, impaired peripheral, night, and color
Do not give to patients with known hyper-
vision, and neurotoxicity-related auditory
sensitivity to daunorubicin, congestive heart
abnormalities (i.e., high-frequency hearing
failure, arrhythmias, or preexisting bone
loss). Periodic hearing and vision exams
marrow suppression. Reduce dosage in
should be performed. High doses (>60 mg/
patients with hepatic, biliary, or renal
kg) especially in children
3 years have
impairment. May cause myelosuppression,
been associated with growth retardation;
nausea, vomiting, alopecia, stomatitis, and
reduction in dosage may increase growth
pigmentation of nail beds. Irreversible myo-
velocity.
cardial toxicity may occur as the cumulative
Discontinue use in febrile patients
dosage approaches 550 mg/m2 (or 400 mg/
because of increased susceptibility to infec-
m2 with chest irradiation or concomitant
tion with Yersinia enterocolitica. Avoid rapid
cyclophosphamide administration; based
IV administration as flushing, urticaria,
on anthracycline dose equivalents, see
hypotension, and shock have been reported.
Formulary
319
DEFERASIROX
caution in patients with hepatic disease
Exjade
as well as in patients with concomitant
anticoagulant, NSAID, or corticosteroid
Chelating agent, iron; antidote, iron toxicity
Tablet, for oral suspension: 125, 250, 500 mg
use. Monitor transaminases and liver
function. May cause ocular or auditory
Pregnancy category C
disturbances, skin rash, gastrointestinal
pain, headache, diarrhea, sore throat,
Indications:
nausea, and vomiting. Interruption or
Treatment of chronic iron overload due to
dose modification may be necessary for
blood transfusions.
evidence of renal, hepatic, or gastrointes-
tinal dysfunction.
Dosage:
Children
2 years to adults: 20 to 40 mg/kg
DESMOPRESSION ACETATE
daily (round to nearest whole tablet)
DDAVP, Stimate
Initiate therapy at
20 mg/kg/day. Adjust
Antihemophilic, hemostatic agent
dose every 3 to 6 months based on serum
Injection: 4 mg/mL (1 mL)
ferritin levels or other measurement of iron
Tablets: 0.1, 0.2 mg
overload; increase by
5 to
10 mg/kg/day
Solution: 1500 mcg/mL, 150 mcg/spray (25
(round to nearest whole tablet). Consider
sprays, 2.5 mL) (Stimate)
higher doses for ferritin >2500 ng/mL. Con-
100 mcg/mL, 10 mcg/spray (50 sprays, 5 mL,
sider holding dose or discontinuing for
with rhinal tube) (DDAVP)
ferritin
<500 ng/mL. Maintenance range:
Pregnancy category B
20 to 40 mg/kg/day.
Consider giving daily dose divided BID to
Indications:
patients experiencing abdominal discom-
Intranasal Stimate or IV DDAVP are indi-
fort or nausea and vomiting.
cated for maintenance of hemostasis in
Do not chew or swallow whole tablets. Dis-
patients with mild or moderate hemophilia
perse tablets in water, apple juice, or orange
A during surgery and postoperatively as well
juice
(use
3.5
ounces for doses
<1 g; 7
as treatment of mucosal bleeds in patients
ounces for doses
1 g); stir to form suspen-
with von Willebrand disease and mild
sion and drink entire contents. Rinse
hemophilia A. DDAVP is indicated for the
remaining residue in glass and drink.
treatment of central diabetes insipidus and
Administer at the same time every day, at
primary nocturnal enuresis.
least 30 minutes prior to food ingestion.
Dosage:
Notes:
Von Willebrand disease, hemophilia A, bleed-
Do not give to patients with known
ing diathesis:
hypersensitivity to deferasirox, low platelet
IV: 0.3 mcg/kg, dilute in normal saline (10 mL
counts (<50
109/L), creatinine clearance
for patients <10 kg and 50 mL for
10 kg),
<40 mL/min, or serum creatinine >2
infuse slowly over 15 to 30 minutes
age-appropriate upper limit of normal.
or
Assess patients for renal impairment and
concurrent nephrotoxic drugs. May cause
Intranasal spray
(Stimate): one puff
intermittent proteinuria. Monitor serum
(150 mcg) for children under 50 kg and 2
electrolytes and urinalysis. Use with
puffs (300 mcg) for children over 50 kg
320
Formulary
Peak effect is 1 to 5 hours with intranasal
A desmopressin challenge to document
route, 1.5 to 3 hours with IV route, and 2 to
responsiveness to these agents is indicated
7 hours with PO route. Duration of effect is
in patients with bleeding disorders to doc-
5 to 24 hours. Tachyphylaxis may occur
ument benefit prior to emergent use.
with repeated dosing within 72 hours.
Note: intranasal forms come in two concen-
Diabetes insipidus:
trations, use as specifically indicated.
Children
12 years: Start with 0.05 mg/dose
BID and titrate to effect (i.e., control of
DEXAMETHASONE
excessive thirst and urination); usual dose
Decadron, generic
range is 0.1 to 0.8 mg/24 hours.
Corticosteroid, anti-inflammatory, imm-
Children >12 years and adults: Start with
unosuppressant
0.05 mg/dose BID and titrate to effect; usual
Tablets: 0.25, 0.5, 0.75, 1.5, 2, 4, 6 mg
dose range is 0.1 to 1.2 mg/24 hours divided
Solution: 1 mg/mL
BID or TID or
Ophthalmic solution: 0.1% (5 mL)
Injection: 4 mg/mL
Children 3 months to 12 years: 5 to 30 mcg/24
hours intranasally divided BID.
Pregnancy category C
Children >12 years and adults: 10 to 40 mcg/
24 hours intranasally divided daily to TID;
Indications:
titrate dose to effect.
Used systemically for chronic inflamma-
Nocturnal enuresis (
6 years):
tion, allergic, hematologic, neoplastic (e.g.,
0.2 mg PO at bedtime, titrate to effect (max-
leukemia), autoimmune disease, cerebral
imum dose 0.6 mg) or
edema and increased intracranial pressure,
20 mcg intranasal at bedtime; divide and
and septic shock. Utilized as an antiemetic
for chemotherapy-induced nausea and
administer 10 mcg in each nostril.
vomiting and as an ophthalmic solution for
Notes:
control of chemical conjunctivitis from con-
Do not give to patients with known hyper-
comitant administrations of agents such as
sensitivity to desmopressin. Avoid in
high-dose cytarabine.
patients with severe type I, type IIB, or
platelet-type von Willebrand disease, hemo-
Dosage:
philia B, and severe hemophilia A (<1%
Antiemetic:
factor VIII activity). Patients with moderate
hemophilia A may not demonstrate an ade-
0.2 mg/kg/dose (5 mg/m2/dose) IV 30 min-
quate response. Use cautiously in patients
utes prior to and every
6
hours after
with a predisposition to thrombophilia,
chemotherapy
electrolyte imbalance, and hypertensive car-
Brain tumor associated cerebral edema:
diovascular disease. May cause headache,
Loading dose: 1 to 2 mg/kg IV as a single
nausea, emesis, seizures, blood pressure
dose; maintenance: 1 to 2 mg/kg/day IV in 4
changes, hyponatremia, nasal congestion,
to 6 divided doses for 1 to 5 days, or longer;
abdominal cramps, and hypertension.
maximum dose 16 mg/24 hours
Avoid in young children <2 years due to
Spinal cord compression with neurologic
hyponatremia and seizures. Not FDA
abnormalities:
approved in <6 years for treatment of noc-
turnal enuresis.
2 mg/kg/24 hours IV divided q6 hours
Formulary
321
Chemotherapy:
2000 mg; followed by 500 mg/m2/dose on
days 3; maximum dose 1000 mg.
Refer to individual protocol.
Do not use dimethylsulfoxide
(DMSO) in
Doses range from 6 to 20 mg/m2/day for 5 to
patients receiving dexrazoxane for anthracy-
7 days (may be longer in induction therapy
cline-induced extravasation.
for acute lymphoblastic leukemia).
Ophthalmic use:
Notes:
Instill 1 to 2 drops into the conjunctival sac
Limited experience in pediatrics (high risk
bilaterally. May use q1-2 hours as needed to
acute lymphoblastic leukemia). Use only in
prevent and control symptoms.
patients with a cumulative doxorubicin
dose of 300 mg/m2 who are continuing to
Notes:
receive anthracyclines or as directed by pro-
tocol. Dose-limiting toxicity is myelosup-
Do not administer during active, untreated
infections with viral, fungal, or bacterial
pression, which may be additive to chemo-
therapy. May chelate heavy metals, leading
organisms. Prolonged use may cause bone
pain, glucose intolerance, hypertension, fat
to increases in calcium, iron, and triglycer-
ides and decreases in sodium and zinc. May
redistribution and striae, and avascular
necrosis of bone.
have antitumor effects.
DIMETHYL SULFOXIDE
DEXRAZOXANE
DMSO
Zinecard, generic
Antidote (anthracycline extravasation)
Antidote, anthracycline; antidote (anthracy-
Topical: 50% solution
cline extravasation)
Injection: 250, 500 mg
Pregnancy category C
Pregnancy category C/D
Indications:
Used after cooling for local control of
Indications:
extravasation of anthracyclines. May also
Reduction of anthracycline-induced cardi-
be indicated for extravasation of ifosfamide,
otoxicity. Treatment for local tissue extrav-
cisplatin, fluorouracil, and carboplatin.
asation of anthracyclines. Currently, FDA
approved in adults only.
Dosage:
Dosage:
Apply 1 to 2 mL within 10 to 25 minutes
to affected area, then every 4 to 8 hours
Refer to individual protocol.
for 7 to 14 days, until resolution. Allow to
Children and adults: 10:1 dose ratio with
dry and apply clean, dry dressing. Vitamin
doxorubicin for acute lymphoblastic leuke-
E or aloe vera have been used topically
mia (ALL; i.e., 300 mg/m2 dexrazoxane for
following the first application to alleviate
30 mg/m2 doxorubicin). Complete anthra-
local burning or stinging (ensure DMSO
cycline administration within 30 minutes of
has dried).
start of dexrazoxane.
Treatment for extravasation: Adolescents
18
Notes:
years to adults: 1000 mg/m2/dose within 6
The role of DMSO in the treatment of
hours on days 1 and 2; maximum dose
anthracycline
extravasation
remains
322
Formulary
controversial. Do not use with dexrazoxane.
cadmium poisoning. Use with caution in
Use with local cooling of tissues.
patients with G6PD deficiency (may cause
hemolysis) and peanut sensitivity. Hydrate
DIMERCAPROL
and alkalinize urine to protect the kidneys.
May cause hypertension, tachycardia, gastro-
British anti-Lewisite (BAL)
intestinal disturbance, headache, fever,
Chelating agent (antidote for gold, mercury,
transient neutropenia, and nephrotoxicity.
lead, and arsenic toxicity)
Symptoms may be relieved by antihistamines.
Injection: 100 mg/mL
Ensure calcium salt is given if edetate calcium
Pregnancy category C
disodium is used in conjunction with BAL.
Indications:
DIPHENHYDRAMINE
Antidote to gold, mercury, and arsenic poi-
Benadryl, generic
soning; used in conjunction with edetate cal-
Antihistamine
cium disodium to treat severe lead poisoning.
Capsules/tablets (OTC): 25, 50 mg
Chewable tablets: 12.5 mg
Dosage:
Elixir: 12.5 mg/5 mL
Injection: 50 mg/mL
For severe lead poisoning (lead level
70 mcg/
dL) or encephalopathy:
Pregnancy category B
25 mg/kg/day divided q4h deep IM for a
minimum of 72 hours, may give up to 5
Indications:
days in severely symptomatic patients. Cal-
Treatment of allergic symptoms, anaphy-
cium sodium EDTA should be started at a
laxis, medication and transfusion reactions,
dose of 50 mg/kg/day continuous IV infu-
chemotherapy and other induced nausea
sion immediately after the second dimer-
and vomiting, and motion sickness; used
caprol dose. If symptoms of encephalopathy
as an antitussive or for mild sedation.
persist, a second course of treatment can
Prevents or treats metocloperamide- and
begin after a minimum of 2 days of rest
phenothiazine-induced dystonic reactions.
following the initial 5-day course. Therapy
should be continued until the patient is
Dosage:
clinically stable. Once this occurs, a 10- to
14-day period of equilibration should occur
Antiemetic and antivertigo:
before again measuring the lead level. If the
0.5 to 1 mg/kg/dose q6h PO, IM, or IV
level remains
70 mcg/dL, another course
Pruritus:
of double therapy should be provided.
0.5 to 1 mg/kg/dose q6h PO, IM, or IV
Less severe lead poisoning (45 to <70 mcg/dL):
Treatment with dimercaprol is not recom-
Notes:
mended in this situation due to toxicity;
Do not give to patients with known hyper-
patients should be treated with succimer or
sensitivity to diphenhydramine. Do not
calcium disodium EDTA instead.
use with concurrent MAO inhibitors, in
acute attacks of asthma, or in patients with
Notes:
GI or urinary obstruction. Use with caution
Do not give to patients with hepatic or renal
in patients with glaucoma, peptic ulcer
insufficiency. Do not use in iron, selenium, or
disease, urinary tract obstruction, and
Formulary
323
hyperthyroidism. Avoid alcohol. Note: many
ECHO or MUGA and ECG after every 75
preparations contain alcohol. May cause
to 100 mg/m2 of anthracycline given. Car-
sedation, nausea, vomiting, xerostomia,
diac toxicity may be acute or delayed and
blurred vision, and central nervous system
long-term monitoring is essential.
effects. May cause paradoxical activation in
Modify dosage in renal impairment.
children.
May cause severe bone marrow suppres-
sion, alopecia, mucositis, and transient
DOXORUBICIN HYDROCHLORIDE
(48 hour) red-orange discoloration of the
urine, sweat, saliva, and tears. Extravasa-
Adriamycin, Doxil (liposomal formulation)
tion may result in severe local tissue necro-
Antineoplastic antibiotic, anthracycline
sis. May cause photosensitivity reactions
Injection: 10, 50 mg vials (protect from light)
and patients should be instructed to avoid
Pregnancy category D
excessive exposure to sunlight and utilize
sunblock (SPF
15).
Indications:
Treatment of acute lymphoblastic leukemia,
DRONABINOL
acute myelogenous leukemia, lymphoma,
Marinol, tetrahydrocannabinol
Wilms tumor, and sarcoma.
Cannabinoid
Capsules: 2.5, 5, and 10 mg
Dosage:
Pregnancy category C
Refer to individual protocol.
20 to 90 mg/m2 IV in repeated doses (may be
Indications:
weekly or monthly or per phase of therapy).
Infusions may be over 15 minutes, several
Treatment of nausea and vomiting associ-
hours, or as a continuous infusion over 24
ated with chemotherapy in patients who
to 48 hours. Although controversial, longer
have failed to respond to conventional
infusions may be more cardioprotective, espe-
antiemetic therapy; treatment of anorexia
cially when high cumulative doses (i.e., >450
associated with weight loss in patients with
mg/m2) are anticipated. Children
<12 kg
human immunodeficiency virus
(HIV).
and infants are dosed based on weight rather
Evidence of benefit in chemotherapy-
than body surface area. The use of cardiopro-
induced anorexia is mixed; no well-con-
tective agents such as dexrazoxane is being
trolled studies have been performed in
studied in pediatric trials.
children.
Notes:
Dosage:
Do not give to patients with known hyper-
Antiemetic:
sensitivity to doxorubicin, severe congestive
5 mg/m2/dose 1 to 3 hours prior to chemo-
heart failure or cardiomyopathy, or preex-
therapy, then q2-4h; maximum 6 doses/24
isting myelosuppression. Use with caution
hour, 15 mg/m2/dose in adults. Titrate dose
in patients who have received very high
to effect.
cumulative doses of anthracyclines
(i.e.,
Appetite stimulant:
550 mg/m2 or
400 mg/m2 with concom-
itant use of cyclophosphamide or chest
Adult dosing: 2.5 mg BID 1 hour prior to
radiation; based on anthracycline dose
lunch/dinner; if not tolerated, reduce dose to
equivalents, see Chapter
30). Monitor
2.5 mg qhs; maximum dose 20 mg/24 hour.
324
Formulary
Notes:
and frequent monitoring of serum electro-
Do not give to patients with known hyper-
lytes including calcium and phosphorus,
renal function, and urinalyses. Establish
sensitivity to any cannabinoid or sesame oil.
Do not use in patients with history of sub-
urine flow prior to administration and
maintain hydration/urine output through-
stance abuse or mental illness. Use with
out course of therapy. Dose reduction is
caution in heart disease, seizures, and hepatic
recommended for mild renal disease. Mon-
disease. A dose-related “high” (easy laughing,
itor with continuous ECG due to risk for
elation, or heightened awareness) is reported
arrhythmias. Rapid IV infusion can result in
in one-quarter of patients using cannabi-
sudden increase in intracranial pressure in
noids as antiemetics. Other side effects
patients with cerebral edema. May cause
include dizziness, anxiety, difficulty concen-
zinc and copper deficiency. May be admin-
trating, hypotension, and increased appetite.
Psychological and physiologic dependence
istered IM; give with
0.5% prilocaine.
Ensure calcium salt is given.
may occur, but addiction is uncommon. It
is a controlled (Schedule III) substance.
ENOXAPARIN
EDETATE CALCIUM DISODIUM
Lovenox
Anticoagulant, low-molecular-weight hepa-
Calcium EDTA, CaNa2EDTA
rin (LMWH)
Calcium disodium versenate
Heavy metal antagonist; antidote, lead
Injection
(prefilled syringe):
30 mg/0.3 mL,
40 mg/0.4 mL, 60 mg/0.6 mL, 80 mg/0.8 mL,
toxicity
Injection: 200 mg/L
100 mg/mL
Approximate anti-factor Xa activity:
Pregnancy category B
100 IU/mg
Pregnancy category B
Indications:
Used as an adjunct in the treatment of acute
Indications:
and chronic lead poisoning.
Prophylaxis and treatment of throm-
Dosage:
boembolic disorders such as deep vein
thrombosis
(DVT) following surgery or
For severe lead poisoning (lead level
70 mcg/
trauma and pulmonary embolus.
dL) and/or encephalopathy:
50 mg/kg/day
(continuous IV infusion)
Dosage:
given in combination with dimercaprol
Treatment of DVT or thromboembolus (TE):
(BAL). Give at a separate site starting with
Infants <1 month: 1.625 mg/kg q12h SC
the second dimercaprol dose.
Less severe lead poisoning (45 to <70 mcg/dL):
Infants 1 to 12 months: 1.5 mg/kg q12h SC
Children 1 to 6 years: 1.375 mg/kg q12h SC
25 mg/kg/day as a continuous or intermit-
tent IV infusion
Children 6 to 21 years: 1.25 mg/kg q12h SC
Titrate dose to achieve peak anti-factor Xa
Notes:
levels of 0.5 to 1.0 U/mL. Peak anti-factor Xa
Do not give to patients with severe renal
levels should be drawn 4 hours after admin-
disease or anuria. Requires inpatient admis-
istration of a steady state dose (minimum 2
sion with aggressive intravenous hydration
to 3 prior doses).
Formulary
325
See Chapter 10 for further guidelines. Some
hypochromic anemia, thrombocytopenia,
centers advocate that TPA be given con-
and pain/erythema at the injection site.
comitantly with enoxaparin in high risk
In case of overdose, protamine sulfate
situations
(i.e., large central embolism).
can be given although the reversal is not
Prophylaxis or treatment with enoxaparin
complete as with unfractionated heparin.
may continue for prolonged periods depen-
1 mg of protamine sulfate neutralizes
dent on the underlying risks for thrombo-
approximately 0.6 to 0.7 mg of enoxaparin.
philia and response to therapy. For those
patients that will be maintained with oral
EPOETIN ALFA
anticoagulation (e.g., warfarin), 4 to 5 days
Recombinant human erythropoietin
of overlap with enoxaparin should be given
Epogen, Procrit
in order to achieve a therapeutic interna-
Blood formation
tional normalized ratio (INR).
Injection:
2000,
3000,
4000,
10,000,
DVT/TE prophylaxis:
20,000 U/mL (1 mL)
0.5 mg/kg q12h SC
Pregnancy category C
Anti-factor Xa levels do not need to be
followed in patients on prophylactic dosing.
Indications:
Data is limited but 1 mg/kg once daily dos-
Treatment of anemia associated with
ing can be considered in patients with com-
chronic renal failure, anemia related to
pliance issues and in those with difficulty
therapy with zidovudine (AZT) in HIV-
giving twice daily SC injections. An insuflon
infected patients, and anemia of prematu-
catheter should also be considered.
rity. Usage in cancer patients remains con-
Note: LMWH may be given IV at the same
troversial. May be considered as an adjunct
dose as SC.
for anemia treatment in patients with reli-
gious beliefs against utilization of blood
Notes:
transfusion.
Do not give to patients with known hyper-
sensitivity to enoxaparin or pork products
Dosage:
(derived from porcine intestinal mucosa),
Anemia in chronic renal failure:
active major bleeding, prosthetic heart valves,
acute heparin-induced (or LMWH-induced)
50 to 150 U/kg administered SC/IV three
thrombocytopenia, and recent major surgery
times a week; higher doses may be needed.
or cerebral hemorrhage. Use of the medica-
Dose is individualized to achieve and main-
tion should be discussed in patients with
tain the lowest hemoglobin sufficient to
religious beliefs which prohibit the consump-
avoid transfusion, not to exceed 12 g/dL.
tion of pork products. Do not give to patients
Anemia in AZT-treated HIV patients:
with drug-induced thrombocytopenia. Use
100 U/kg SC three times a week; dose range
with caution in patients with recurrent gas-
50 to 4000 U/kg two to three times per week
trointestinal ulcers, bleeding diathesis, and
severe renal dysfunction; lower initial doses
Anemia in cancer patients:
should be given to patients with renal insuf-
600 U/kg IV once weekly; dose titrated to
ficiency and failure. Do not use with concur-
effect (maximum 40,000 U). Do not initiate
rent spinal or epidural anesthesia or lumbar
therapy at Hgb
10 g/dL; adjust dose to
puncture for chemotherapy. May cause fever,
maintain the lowest Hgb level needed to
confusion, edema, nausea, hemorrhage,
avoid transfusions. Adult dose
150 U/kg
326
Formulary
SC three times a week or 40,000 U SC once
AML remission induction:
weekly.
150 mg/m2/day for 2 to 3 days for 2 to 3
Anemia of prematurity:
cycles in induction, followed by 250 mg/m2/
day for 3 days in consolidation and inten-
No FDA-approved dosing regimen, a vari-
sification courses. Dose per body weight in
ety of different doses and schedules have
infants.
been used. Utility in decreasing need for
transfusion in VLBW and ELBW infants is
Brain tumors:
not well-proven.
150 mg/m2/day for 2 days; may also be given
long term orally (using injection for oral
Notes:
administration); has good central nervous
Do not give to patients with known hyper-
system penetration
sensitivity to albumin, uncontrolled hyper-
Neuroblastoma:
tension, and in newborns with neutropenia.
100 mg/m2/day over 1 hour days 1 to 5 or
Use with caution in patients with porphyria
alternate day for 3 doses q3-4 weeks for 3 to
or a history of seizures. May cause headache
4 courses
and elevated blood pressure. Meta-analyses
have shown that usage leads to a significant
BMT conditioning regimen:
increased risk of death, thrombosis, and
160 mg/m2/day for 4 days
serious cardiovascular events in cancer
patients.
Notes:
Evaluate iron stores (ferritin and total
Do not give to patients with known hyper-
iron binding capacity [TIBC]) before ther-
sensitivity to etoposide. Use with caution and
apy initiation. Iron supplementation is
consider dose reduction in patients with
recommended unless iron stores are already
renal or hepatic impairment. May cause
in excess.
facial flushing, fever, fatigue, nausea, vomit-
ing, bone marrow suppression, and periph-
ETOPOSIDE
eral neuropathy. Associated with second
VP-16
malignant neoplasms, likely dose and fre-
VePesid, Toposar
quency related.
Antineoplastic, mitotic inhibitor
Give IV infusions over at least 1 hour
Capsule, softgel: 50 mg
due to associated hypotension with rapid
Injection: 20 mg/mL
infusion. Infusion should be stopped or
infusion rate be decreased with hypoten-
Pregnancy category D
sion. After fluid resuscitation, the infusion
may be restarted at a slower rate if stopped
Indications:
and the hypotension resolved.
Treatment of germ cell tumors, lymphoma,
brain tumors, acute myelogenous leukemia
EUTECTIC MIXTURE OF LIDOCAINE
(AML), neuroblastoma, rhabdomyosar-
AND PRILOCAINE
coma, histiocytosis, and conditioning for
bone marrow transplantion (BMT).
EMLA, LMX
Local anesthetic, topical anesthetic
Dosage:
Cream: 5, 30 g tubes
Refer to individual protocol.
Pregnancy category B
Formulary
327
Indications:
Prophylaxis:
Used as a topical anesthetic applied to nor-
Premature infant: 2 mg/kg/day, maximum
mal intact skin to provide local anesthesia
dose 15 mg/24 hour
for minor procedures such as venipuncture,
Term infant: 1 to 2 mg/kg/day, maximum
placement of peripheral venous access line,
dose 15 mg/24 hour
lumbar puncture, and minor dermatologic
procedures.
Notes:
Do not give to patients with known hyper-
Dosage:
sensitivity to iron salts, hemochromatosis,
Apply 2.5 g per site to normal intact skin
and transfusional iron overload. Absorption
and cover with occlusive dressing for 30 to
of iron is decreased when given with tetra-
60 minutes prior to procedure.
cycline, antacids, or milk. Less gastrointes-
tinal irritation when given with food. Con-
Notes:
current administration of 200 mg or more
of vitamin C per
30 mg elemental iron
Do not give to patients with known hyper-
increases absorption of oral iron; iron
sensitivity to lidocaine, prilocaine, or
replacement products should be given with
other local anesthetic or methemoglobine-
orange juice. May cause constipation, dark
mia. Do not use in neonates <37 weeks
stools, nausea, and epigastric pain.
gestation or in infants <12 months of age
who are receiving concurrent treatment
FERROUS SULFATE
with methemoglobin producing agents
(e.g., sulfa drugs, dapsone, phenobarbital,
Feosol, Fer-In-Sol
benzocaine).
Iron (20% elemental Fe)
Tablet OTC: 300 mg (60 mg Fe), 324 mg
FERROUS GLUCONATE
(65 mg Fe), 325 mg (65 mg Fe)
Drops (Fer-in-sol) OTC: 75 mg (15 mg Fe)/
Fergon
0.6 mL
Iron (12% elemental Fe)
Elixir OTC: 220 mg (44 mg Fe)/5 mL
Tablets (OTC): 240 mg (29 mg Fe), 300 mg
Oral liquid OTC: 300 mg (60 mg Fe)/5 mL
(36 mg Fe), 325 mg (39 mg Fe)
Pregnancy category A
Pregnancy category A
Indications:
Indications:
Prevention and treatment of iron deficiency
Prevention and treatment of iron deficiency
anemia.
anemia.
Dosage:
Dosage:
See Ferrous Gluconate.
Dose is expressed in terms of elemental iron
Severe iron deficiency anemia:
Notes:
4 to 6 mg/kg/day PO in two to three divided
See Ferrous Gluconate.
doses
Mild to moderate iron deficiency anemia:
FERROUS FUMARATE
3 mg/kg/day PO in 1 to 2 divided doses
Iron salt (33% elemental Fe)
328
Formulary
Tablet OTC: 90 mg (29.5 mg Fe), 200 mg
(ANC) is greater than
2
to
10
109/L
(66 mg Fe), 324 mg (106 mg Fe), 325 mg
(per protocol; given beyond nadir period
(106 mg Fe), 350 mg (115 mg Fe)
7 to 10 days after chemotherapy admini-
Chewable tablets: 100 mg (33 mg Fe)
stration).
Timed-release
tablet
OTC:
150 mg
Peripheral blood progenitor cell mobilization:
(50 mg Fe)
10 mcg/kg SC daily for 4 days before the first
Pregnancy category A
leukapheresis procedure and continued
until the last leukapheresis.
Indications:
Congenital neutropenia:
Prevention and treatment of iron deficiency
2.5
to
6 mcg/kg/day SC; titrate dose to
anemia.
desired ANC to prevent infection.
Dosage:
Idiopathic or cyclic neutropenia:
5 mcg/kg SC once daily; titrate dose to
See Ferrous Gluconate.
desired ANC to prevent infection.
Notes:
Elevation of ANC is usually within
24
See Ferrous Gluconate.
hours, though it may be delayed in severe
myelosuppression. Transient increase in
FILGRASTIM
ANC may occur when granulocyte col-
ony-stimulating factor (G-CSF) is begun
G-CSF
Neupogen
shortly after completion of chemotherapy;
avoid premature discontinuation.
Blood formation, colony-stimulating factor
Injection: 300 mcg/mL (1, 1.6 mL)
SC route of administration is preferred due
to prolonged serum levels over IV route.
Pregnancy category C
If used IV and the G-CSF concentration
>15 mcg/mL, add 2 mg albumin/1 mL of
Indications:
IV fluid to prevent drug absorption in the
Used to decrease the period of neutropenia
IV administration set.
and the associated risk of infection in
patients with malignancies receiving mye-
Notes:
losuppressive chemotherapy associated with
Avoid in patients with hypersensitivity to
a significant incidence (i.e., >20%) of severe
Escherichia coli derived proteins or G-CSF.
neutropenia. Has been used in zidovudine-
May cause bone pain and increases in uric
associated neutropenia in human immuno-
acid and lactate dehydrogenase. Do not
deficiency virus infected patients and in
administer 24 hours before or after admin-
patients with nonchemotherapy-induced neu-
istration of chemotherapy.
tropenia (acquired and congenital). Should
be considered in patients with complicated
infection with underlying neutropenia.
FLUCONAZOLE
Diflucan
Dosage:
Antifungal
Neonates: 5 to 10 mcg/kg IV/SC daily for
Tablet: 50, 100, 150, 200 mg
Injection: 2 mg/mL
3 to 5 days in neutropenia with sepsis
Suspension: 10, 40 mg/mL
Children and adults: 5 to 10 mcg/kg IV/SC
once daily until absolute neutrophil count
Pregnancy category C
Formulary
329
Indications:
fluconazole metabolism. Consult the
Prophylaxis and treatment of susceptible
Physicians’ Desk Reference or a pharmacist
when ordering fluconazole in a patient
fungal infections, including oropharyngeal,
esophageal, and vaginal candidiasis and sys-
receiving many medications.
temic fungal infections with Candida sp. as
well as treatment and suppression of cryp-
FOLIC ACID
tococcal meningitis. Species of Candida
Folate
with decreased in vitro susceptibility to flu-
Folvite
conazole are being isolated. Fluconazole is
Blood formation, water-soluble vitamin
more active against candidal species such as
Tablet OTC: 0.4, 0.8, 1 mg
C. albicans than species such as C. parapsi-
Solution: 50 mcg/mL
losis, C. glabrata, and C. tropicalis.
Injection: 5 mg/mL
Dosage:
Pregnancy category A/C
Oropharnygeal candidiasis:
Indications:
6 mg/kg loading dose PO/IV followed by
Treatment of megaloblastic anemia result-
3 mg/kg/day
(dependent on severity of
ing from folate deficiency; supplementa-
infection) for 2 to 4 weeks (dependent on
tion for patients with chronic hemolytic
clinical response and immune status of the
anemia (e.g., sickle cell disease, hereditary
patient). Neonates under 14 days of age are
spherocytosis).
dosed every 24 to 72 hours.
Esophageal candidiasis:
Dosage:
12 mg/kg loading dose PO/IV, followed by
Infants to children <12 months: 15 mcg/kg
6 mg/kg/day for 2 to 4 weeks; maximum
daily; maximum 50 mcg/day
dose 400 mg/day
Children 1 to 11 years: initial dose 1 mg/day;
Invasive systemic candidiasis and cryptococcal
maintenance dose 0.1 to 0.4 mg/day
meningitis:
Children >11 years and adults: initial dose
12 mg/kg loading dose PO/IV, followed by 6
1 mg/day; maintenance dose 0.5 mg/day
to 12 mg/kg/day for 2 to 4 weeks or longer
Prophylaxis in immunocompromised hosts:
Notes:
3 to 6 mg/kg/dose PO/IV; maximum dose
May mask hematologic effects of vitamin
400 mg/day
B12 deficiency but will not prevent progres-
sion of neurologic abnormalities.
Notes:
Do not give to patients with known hyper-
FOSCARNET
sensitivity to fluconazole or other azoles.
Foscavir, generic
May cause nausea, headache, rash, vomit-
Antiviral
ing, abdominal pain, hepatitis, cholestasis,
Injection: 24 mg/mL
and diarrhea. Antagonism may occur if
Pregnancy category C
amphotericin B and fluconazole are used
concurrently. Many drug interactions exist.
Indications:
May increase effects, toxicity, and/or levels
of cyclosporine, midazolam, tacrolimus,
Alternative to ganciclovir for treatment of
and many other drugs. Rifampin increases
CMV infection; treatment of acyclovir-
330
Formulary
resistant HSV infections in immunocom-
CMV Retinitis, children >3 years and adults:
promised hosts.
10 mg/kg/day divided q12h as a
1-hour
infusion for
14 to
21 days followed by
Dosage:
maintenance 5 mg/kg/day as a single daily
Children and adults:
dose for 7 days/week or 6 mg/kg/day for 5
days/week
CMV retinitis:
180 mg/kg/day IV divided
q8-12h for 14 to 21 days followed by daily
Treatment of CMV reactivation in trans-
IV maintenance of 90 mg/kg/day
plant recipients, children and adults:
10 mg/kg/day divided q12h for 1 to 2 weeks
Acyclovir-resistant HSV infection: 40 mg/kg/
followed by maintenance
5 mg/kg/day
dose IV q8-12h up to 3 weeks or until lesions
once daily for 7 days/week or 6 mg/kg/day
heal; repeat treatment may lead to resistance.
for
5
days/week until
100
days post-
Notes:
transplant
May cause renal impairment, adjust dose for
Other CMV infections, children and adults:
renal dysfunction. Assure adequate hydra-
10 mg/kg/day divided q12h for 14 to 21 days
tion. May adversely affect tooth and bone
or 7.5 mg/kg/day divided q8h followed by
growth in children, safety and efficacy not
maintenance 5 mg/kg/day once daily for 7
fully evaluated. May cause electrolyte imbal-
days/week or 6 mg/kg/day once daily for 5
ances and symptoms of hypocalcemia. May
days/week
cause seizures; risk factors include renal
Notes:
impairment, low serum calcium, and
underlying central nervous system condi-
Do not give to patients with known hyper-
tion. Foscarnet is a vesicant; only infuse into
sensitivity to ganciclovir, acyclovir, or
veins with adequate blood flow.
any component. Patients must use appro-
priate contraception due to teratogenic
GANCICLOVIR
effects for at least
90 days after therapy
Cytovene-IV
completion. Cytopenias may occur; do not
Antiviral
administer if ANC < 0.5
109/L or plate-
Injection: 500 mg (can be prepared into oral
lets
<25
109/L. Use with caution in
suspension)
patients with renal impairment.
Pregnancy category C
GEMTUZUMAB OZOGAMICIN
Indications:
Mylotarg
Treatment of CMV retinitis, pneumonitis,
Antineoplastic, monoclonal antibody
Injection: 5 mg
encephalitis, and gastrointestinal infection
in immunocompromised patients. Preven-
Pregnancy category D
tion of symptomatic CMV disease in trans-
plant patients with reactivation of latent
Indications:
disease. Has antiviral activity against
Treatment of acute myelogenous leukemia.
HSV-1 and HSV-2.
Dosage:
Dosage:
Children with body surface area (BSA) <0.6
Congenital CMV infections, neonates and
m2: 0.1 mg/kg/dose
infants:
12 mg/kg/day slow IV infusion
q12h for 6 weeks
Children with BSA
0.6 m2: 3 mg/m2/dose
Formulary
331
Notes:
Adults: 2 mg/24 hours PO divided q6-12h;
Do not give to patients with known hyper-
initiate first dose before chemotherapy.
sensitivity to gemtuzumab. Severe hyper-
Postoperative
nausea and vomiting
sensitivity reactions and infusion-related
prevention:
reactions may occur including pulmonary
Children
4 years to adults: 20 to 40 mcg/kg
edema, acute respiratory distress syndrome,
IV dosed prior to anesthesia or immediately
and anaphylaxis. Patients should be preme-
before anesthesia reversal; maximum dose
dicated with acetaminophen and diphenhy-
1 mg once
dramine. Administer by slow infusion over
Radiation-induced nausea and vomiting
2 hours. Severe hepatic toxicity including
prevention:
veno-occlusive disease (sinusoidal obstruc-
tive syndrome) has been reported. Use with
Adults: 2 mg PO daily administered 1 hour
caution in patients with renal or hepatic
prior to radiation
impairment or pulmonary disease. May
cause fetal harm; appropriate contraception
Notes:
is advised.
Inducers or inhibitors of the cytochrome
P450
drug-metabolizing enzymes may
GRANISETRON
increase or decrease, respectively, the drug’s
Kytril
clearance. Use with caution in liver disease.
Antiemetic agent, serotonin (5-HT3)
May cause hypertension, hypotension,
antagonist
arrhythmias, agitation, and insomnia.
Tablet: 1 mg
Solution: 0.2 mg/mL
HEPARIN SODIUM
Injection: 1 mg/mL
Lipo-Hepin, Hep-Lock
Anticoagulant
Pregnancy category B
Injection: many vial sizes, porcine based
origin; preservative free
Indications:
Lock flush solution: 1, 10, 100 U/mL (porcine
Prevention and treatment of nausea and
based, some products may be preservative
vomiting associated with chemotherapy.
free or contain benzyl alcohol)
Has also been utilized in prevention of
Injection for IV infusion:
(porcine based,
postoperative and radiation-induced nausea
D5W: 40 U/mL, 50 U/mL, 100 U/mL; 0.9%
and vomiting.
NaCl:
2 U/mL;
0.45% NaCl:
50 U/mL,
100 U/mL, contains EDTA)
Dosage:
120 U ¼ approximately 1 mg
Chemotherapy-induced
nausea
and
Pregnancy category C
vomiting:
Children
2 years to adults: 10 to 20 mcg/kg/
Indications:
dose IV 15 to 60 minutes before chemo-
Prophylaxis and treatment of thromboem-
therapy; may be repeated two to thee times
bolic disorders; thrombus prophylaxis for
following chemotherapy over
24
hours;
central venous access devices.
maximum dose 3 mg/dose or 9 mg/24 hour.
Alternatively, a single dose of 40 mcg/kg 15
Dosage:
to
60 minutes before chemotherapy has
been used.
Anticoagulation in infants and children:
332
Formulary
Initial: 75 U/kg IV bolus over 10 minutes
Pregnancy category C
Maintenance IV continuous infusion:
Indications:
<1 year: 28 U/kg/hour
Used to influence the dispersion and
1 year: 18 to 20 U/kg/hour
absorption of other drugs and increase rate
of absorption of parenteral fluids given by
or
hypodermoclysis
(subcutaneously); treat-
Maintenance intermittent dosing:
75
to
ment of IV extravasations.
100 U/kg/dose IV q4h
Adjust dose to keep APTT 1.5 to 2.5 times
Dosage:
upper limit of control reference range (best
Infants and children:
measure 6 to 8 hour after initiation or after
Dilute 150 unit vial in 10 mL normal saline
change in dosing).
(15 U/mL). Give 1 mL (15 U) by adminis-
Heparin flush:
tering five separate injections of
0.2 mL
Peripheral IV: 1 to 2 mL of 10 U/mL solution
(3 U) at borders of extravasation site SC
q4h
or intradermal using a
25- or
26-gauge
Central lines: 2 to 3 mL of 10 U/mL solution
needle. Administer as early as possible after
extravasation (minutes to 1 hour).
q24h and after access. Dosing often estab-
lished by institutional policy.
Notes:
Arterial lines and TPN lines (central lines):
Do not give to patients with known hypersen-
add heparin to make a final concentration of
sitivity to hyaluronidase or respective sources
0.5 to 1 U/mL.
(bovine, ovine). Do not inject in or around
Notes:
infected, inflamed, or cancerous lesions; do not
use with dopamine or alpha-agonist extrava-
Do not give to patients with known hyper-
sation. May cause urticaria.
sensitivity to heparin or pork products, severe
thrombocytopenia, suspected intracranial
HYDROCORTISONE
hemorrhage, shock, severe hypotension, and
Solu-Cortef, Cortef, and others
uncontrolled bleeding (unless secondary to
Corticosteroid
disseminated intravascular coagulation).
Tablet (Cortef): 5, 10, and 20 mg
Should discuss usage in patients with religious
Suspension: 2.5 mg/mL
beliefs which prohibit the consumption of
Injection, as sodium succinate (Solu-Cortef):
pork products. Use preservative free heparin
100, 250, 500, 1000 mg vial
in neonates and consider more dilute heparin.
Pregnancy category C
Antidote: protamine sulfate 1 mg per 100 U
heparin in previous 4 hour
Indications:
Treatment of inflammatory dermatoses and
HYALURONIDASE
adrenal insufficiency; also used as a chemother-
Amphdase, Hydase (bovine source, may
apeutic agent for intrathecal administration.
contain thimersol)
Hylenex (recombinant human source)
Dosage:
Vitrase (ovine source, preservative free)
Antidote (extravasation)
Physiologic replacement:
Injection: 150 U/mL (Vitrase 200 U/mL)
12 to 18 mg/m2/day PO divided q6-8h
Formulary
333
Stress dosing (consider for >2 weeks on glu-
pruritus than morphine. Approximate 6:1
cocorticoid therapy or patients in shock):
conversion from morphine (i.e., 6 mg mor-
phine is equivalent to 1 mg dilaudid).
Solu-Cortef, 25 to 100 mg/m2/day
Intrathecal chemotherapy with methotrexate
HYDROXYUREA
and/or cytarabine:
Hydrea, Droxia
Dose per protocol, range 15 to 30 mg
Antineoplastic
Notes:
Capsule: 500 mg (Hydrea); 200, 300, 400 mg
(Droxia)
Do not give to patients with known hyper-
sensitivity to hydrocortisone, polymyxin B
Pregnancy category D
sulfate, or neomycin sulfate. Avoid in
patients with infections from herpes sim-
Indications:
plex, vaccinia, and varicella.
Treatment of malignancies including
HYDROMORPHONE HYDROCHLORIDE
chronic myelogenous leukemia (CML), des-
moid tumors, and brain tumors; adjunct in
Dilaudid, Dilaudid-HP, generics
the management of sickle cell anemia to
Narcotic, analgesic
reduce pain events, acute chest syndrome,
Tablets: 2, 4, 8 mg
hospitalizations, transfusion needs, and
Solution: 1 mg/mL
mortality; utilized to treat hyperleukocyto-
Suppository: 3 mg
sis due to blast crisis in CML and in relapsed
Injection: 1, 2, 3, 4, 10 mg/mL
acute leukemias; treatment of hypereosino-
Pregnancy category C/D
philic syndrome.
Indications:
Dosage:
Management of moderate to severe pain.
Refer to individual protocol.
CML, hyperleukocytosis:
Dosage:
Initial dose 10 to 20 mg/kg PO once daily;
Children (not for use in neonates):
adjust dose according to hematologic
PO: 0.03
to
0.1 mg/kg/dose q4-6h PRN
response and symptoms.
(maximum 5 mg/dose)
Sickle cell anemia:
IV: 0.015 mg/kg/dose q4-6h PRN
Initial dose 15 mg/kg (range 10 to 20 mg/kg/
Adolescents to adults: 1 to 2 mg/dose IV,
day) once daily; increase dose in increments
IM, or SC q4-6h PRN; 1 to 4 mg PO
of 5 mg/kg/day every 12 weeks to a maxi-
q4-6h PRN
mum of 35 mg/kg/day. Discontinue for
bone marrow toxicity: ANC
<2 109/L,
Notes:
platelets
<80
109/L, reticulocyte count
<8 109/L, or hemoglobin <9 g/dL. Restart
Do not give to patients with known hyper-
therapy after recovery at 2.5 mg/kg/day less
sensitivity to hydromorphone. Dose reduc-
than dose that produced toxicity.
tion recommended in renal insufficiency or
severe hepatic impairment. Avoid use in
Notes:
neonates because of potential central ner-
vous system effects. Use with caution in
Do not give to patients with known hyper-
infants and young children. Causes less
sensitivity to hydroxyurea or those with
334
Formulary
severe anemia or severe bone marrow sup-
Injection: 1 g vial
pression. Use with caution in renal
Pregnancy category D
impairment.
Indications:
IDARUBICIN
Used in combination with certain other
Idamycin, generic
antineoplastics in the treatment of Hodgkin
Antineoplastic antibiotic, anthracycline
and non-Hodgkin lymphoma, acute lym-
Injection: 5, 10 mg
phoblastic leukemia, osteosarcoma, rhab-
Pregnancy category D
domyosarcoma, Ewing sarcoma, and
advanced Wilms tumor.
Indications:
Dosage:
Treatment of acute promyelocytic leukemia
(APL), other types of acute myelogenous
Refer to individual protocol.
leukemia, and relapsed lymphoblastic
Usual dose is 700 to 1800 mg/m2/day for 5
leukemia.
days every 3 to 4 weeks
Dosage:
Notes:
Refer to individual protocol.
Do not give to patients with known hyper-
APL induction:
sensitivity to ifosfamide. Avoid in severe
bone marrow suppression. Use with cau-
12 mg/m2/day IV as 4 doses on alternating
tion in impaired renal function; hydrate the
days for 8 days
patient prior to administration and ensure
good urine flow (i.e., urine specific gravity
Notes:
1.010). Ifosfamide may lead to syndrome
Do not give to patients with known hyper-
of inappropriate antidiuretic hormone
sensitivity to idarubicin, severe congestive
(SIADH) secretion. Mesna is used for uro-
heart failure, or cardiomyopathy. Avoid in
protection with higher doses to decrease
preexisting bone marrow suppression
risk of hemorrhagic cystitis. Urinalysis
unless the potential benefit warrants the
should be monitored closely for specific
risk. May need to reduce dosage with
gravity and heme. May cause central ner-
impaired renal or hepatic function. Irrevers-
vous system toxicity including hallucina-
ible myocardial toxicity may occur as the
tion, somnolence, confusion, coma, or
cumulative dosage approaches 550 mg/m2
encephalopathy.
(or
400 mg/m2 with chest irradiation or
concomitant cyclophosphamide adminis-
IMATINIB
tration; based on anthracycline dose equiva-
Gleevec
lents, see Chapter 30). This may be an acute
Antineoplastic, tyrosine kinase inhibitor
or late effect and monitoring with ECHO or
Tablet: 100, 400 mg
MUGA and ECG is required per protocol.
Secondary leukemia has been reported.
Pregnancy category D
IFOSFAMIDE
Indications:
Ifex
Treatment of newly diagnosed Philadelphia
Antineoplastic, alkylating agent
chromosome positive
(Phþ) chronic
Formulary
335
myelogenous leukemia (CML) in chronic
photosensitivity, puritus, rash, gastrointes-
phase ages
2 years to adult; recurrent Phþ
tinal disturbance, bone or joint pain, and
CML in chronic phase following stem cell
blurred vision.
transplantion; Phþ CML resistant to inter-
feron alpha therapy; Phþ acute lymphoblas-
IMMUNE GLOBULIN
tic leukemia (ALL) (relapsed or refractory);
Flebogamma 5% (50 mg/mL)
mastocytosis; desmoid tumor; gastrointesti-
Gamunex 10% (100 mg/mL)
nal stromal tumors (GIST); hypereosinophi-
Gammagard 10% (100 mg/mL)
lic syndrome or chronic eosinophilic leuke-
Octagam 5% (50 mg/mL)
mia; and myelodysplastic/myeloproliferative
Carimmune NF:
1,
3,
6,
12 g for
disease associated with platelet-derived
reconstitution
growth factor receptor
(PDGFR) gene
Polygam S/D: 2.5, 5, 10 g for reconstitution
rearrangements.
Immunoglobulins
Dosage:
Pregnancy category C
Refer to individual protocol.
Indications:
Children
2 years: Phþ CML, chronic phase,
Treatment of immunodeficiency states (e.g.,
new diagnosis:
340 mg/m2/day divided
human immunodeficiency virus, agamma-
q12-24h; maximum dose 600 mg/day
globulinemia),
secondary immunode-
Phþ CML, chronic phase and recurrent after
ficiencies
(e.g., bone marrow transplant),
stem cell transplantion, resistant to interferon:
immune thrombocytopenic purpura (ITP),
260 mg/m2/day
Kawasaki disease, and lymphoproliferative
May need to adjust dose with hepatic or
disorders.
hematologic toxicity.
The optimal duration of therapy for CML or
Dosage:
ALL is not determined.
Immunodeficiency:
400 mg/kg/dose IV every 3 to 4 weeks.
Notes:
Chronic lymphocytic leukemia:
Do not give to patients with known hyper-
400 mg/kg/dose IV every 3 weeks.
sensitivity to imatinib. May cause fluid
ITP:
retention, weight gain, edema, pleural effu-
800 to 1000 mg/kg IV for 1 to 2 consecutive
sion, pericardial effusion, and pulmonary
days, then every
3 to 4 weeks based on
edema. Use with caution in patients where
clinical response and platelet count
fluid retention may be poorly tolerated such
HIV infection:
as congestive heart failure or left ventricular
400 mg/kg IV every 4 weeks
dysfunction. May cause Stevens-Johnson
HIV-associated thrombocytopenia:
syndrome, hepatotoxicity, hemorrhage, and
500 to 1000 mg/kg/day IV for 1 to 5 days
hematologic toxicity. Use with caution in
Kawasaki disease:
patients with preexisting hepatic or renal
2 g/kg IV as a single dose
impairment. Use with caution in patients
Post-bone marrow transplant:
receiving concurrent therapy that alters
400 to 500 mg/kg q4 weeks (refer to pro-
cytochrome P450 activity or requires meta-
tocol; based on desired IgG level, usually
bolism by these isoenzymes. May cause
>400 mg/dL)
336
Formulary
Notes:
discontinuing. May cause severe, dose-
Avoid in patients with hypersensitivity to
limiting, and potentially fatal diarrhea.
Early diarrhea (during or shortly after infu-
immune globulin or blood products and in
those with immunoglobulin A deficiency
sion) may be accompanied by symptoms of
rhinitis, increased salivation, flushing, mio-
(except with the use of IgA-depleted pro-
sis, lacrimation, diaphoresis, and abdomi-
ducts such as Gammagard or Polygam).
nal cramping. Atropine
0.01 mg/kg IV
May cause infusion-related toxicity requir-
(maximum dose 0.4 mg) may be used to
ing slower IV rate and premedication with
prevent or treat symptoms.
acetaminophen and diphenhydramine. May
Late diarrhea occurs >24 hours after
lead to aseptic meningitis.
therapy and can be prolonged leading to
life-threatening dehydration and electro-
IRINOTECAN
lyte imbalance. Treat promptly with loper-
Camptostar, generic
amide until a normal pattern of bowel
Antineoplastic, topoisomerase inhibitor
movements returns. Antibiotic support
Injection: 20 mg/mL
(cefixime or cefpodoxime has been used
Pregnancy category D
in children) as a prophylactic and treatment
measure.
Indications:
If the patient develops persistent diar-
rhea, ileus, fever, or severe neutropenia,
Treatment of neuroblastoma, hepatoblas-
interrupt or reduce subsequent doses. If
toma, brain tumors, Ewing sarcoma, and
grade 3 (7 to 9 stools/day, incontinence,
rhabdomyosarcoma.
and/or severe cramping) or grade 4 (
10
stools/day, grossly bloody stool, and/or
Dosage:
need for parenteral support) diarrhea
Referral to individual protocol
occurs, interrupt treatment and reduce sub-
Children: refractory solid tumor, low dose,
sequent dosing.
protracted: 20 mg/m2/day for 2 consecutive
May cause severe myelosuppression; halt
weeks, followed by a week of rest; repeat
for neutropenic fever. Use with caution in
q3 weeks.
patients with renal or hepatic impairment.
Children: refractory solid tumor or brain
tumor: 50 mg/m2/day for 5 days, repeat cycle
IRON COMPLEX, POLYSACCHARIDE
q3 weeks.
Niferex-150, Fe-Tinic 150, Ferrex 150
Iron polysaccharide and ascorbic acid
Notes:
Tablet: 50 mg
Capsule: 150 mg
Do not give to patients with known hyper-
Elixir: 100 mg/5 mL
sensitivity to irinotecan. Avoid concomi-
tant administration with St. John’s wort or
Pregnancy category C
ketoconazole and in patients with severe
bone marrow failure. A new cycle of irino-
Indications:
tecan should not begin until serious treat-
Treatment of iron deficiency anemia.
ment-induced toxicity has recovered:
ANC
1.5
109/L and platelet count
Dosage:
>100
109/L. If the patient has not recov-
ered following a
2
week rest, consider
See Ferrous Gluconate.
Formulary
337
Notes:
Maximum daily dose, IM/IV (United States):
See Ferrous Gluconate.
Infants <5 kg: 25 mg (0.5 mL)
Children 5 to 10 kg: 50 mg (1 mL)
IRON DEXTRAN COMPLEX
Children >10 kg to adults: 100 mg (2 mL)
Dexferrum, INFeD
Anemia of prematurity:
Iron salt
Injection: 50 mg elemental iron/mL (2 mL)
Neonates: 0.2 to 1 mg/kg/day or 20 mg/kg/
week with epoietin alfa therapy
Pregnancy category C
Anemia of chronic renal failure:
Indications:
There is insufficient data to support IV iron
if the ferritin level is >500 ng/mL.
Treatment of microcytic hypochromic
anemia resulting from iron deficiency in
Children: predialysis or peritoneal dialysis, as
patients in whom oral administration is not
a single dose repeated as necessary (outside the
feasible or is ineffective. Approved in chil-
United States):
dren
4 months.
<10 kg: 125 mg
Dosage:
10 to 20 kg: 250 mg
>20 kg: 500 mg
Begin with a test dose
1 hour prior to
starting iron dextran therapy:
Children: hemodialysis, given during each
dialysis for 10 doses:
Infants <10 kg: 10 mg (0.2 mL)
<10 kg: 25 mg
Children 10 to 20 kg: 15 mg (0.3 mL)
10 to 20 kg: 50 mg
Children >20 kg to adult: 25 mg (0.5 mL)
>20 kg: 100 mg
Total replacement dose of iron dextran for
iron deficiency anemia:
Parenteral administration:
(mL) ¼ 0.0442
LBW (kg)
(Hgbn Hgbo)
IM: use Z-track technique (deep into upper
þ [0.26
LBW (kg)],
outer quadrant of buttock); test dose at
same site using the same method.
where
IV: infuse test dose over at least 5 minutes
LBW ¼ lean body weight
(Dexferrum) or
30
seconds
(INFeD);
Males: 50 kg þ 2.3 kg for every inch over 5 ft
dilute replacement dose in normal saline
in height
(50 to 100 mL) to maximum concentra-
Females: 45.5 kg þ 2.3 kg for every inch over
tion of 50 mg/mL and infuse over 1 to 6
5 ft in height
hours at a maximum rate of 50 mg/min-
Hgbn ¼ desired hemoglobin (g/dL) ¼ 12 if
ute. Avoid dilution in dextrose due to an
<15 kg or 14.8 if >15 kg
increased incidence of local pain and
phlebitis.
Hgbo ¼ measured hemoglobin (g/dL)
Monitor vital signs and for symptoms of
Total replacement dose of iron dextran for
anaphylaxis during the IV infusion.
acute blood loss:
(Assumes 1 mL of normocytic, normochro-
Notes:
mic red cells ¼ 1 mg elemental iron)
Do not give to patients with known hyper-
Replacement iron (mg) ¼ blood loss (mL)
sensitivity to iron dextran (anaphylaxis may
hematocrit
occur), in anemia not associated with iron
338
Formulary
deficiency, with hemochromatosis, or with
1 mg (elemental iron)/kg/dialysis treatment
hemolytic anemia. Use with caution in
(repletion)
patients with histories of significant allergies,
0.3 mg
(elemental iron)/kg/dialysis treat-
asthma, serious hepatic impairment, preex-
ment (maintenance)
isting cardiac diseases, and rheumatoid arthri-
Children: iron deficiency anemia
(off-label
tis (may cause an exacerbation of arthritis).
use):
Discontinue oral iron prior to initiating par-
enteral iron. Sweating, urticaria, arthralgia,
Calculation of iron deficit: body weight [kg]
(target Hgb-actual Hgb) [g/dL]
2.4
fever, chills, dizziness, headache, and nausea
may be delayed 24 to 48 hours after large doses
Dose: 5 mg/kg/day IV until iron deficit is
of IV administered drug or 3 to 4 days fol-
corrected
lowing IM administration. The IV route is
Adults: Hemodialysis dependent:
preferred for patients with chronic renal dis-
100 mg IV 1 to 3 times/week during dialysis,
ease and cancer related anemia (adults).
total of 10 doses (1000 mg); may continue to
Agents for treatment of acute anaphylaxis
administer at lowest dose possible to main-
should be readily available. Adverse events
tain target hemoglobin and iron storage
are much more associated with the high
parameters
molecular weight formulation (Dexferrum)
than with the low molecular weight formu-
Adults: nondialysis-dependent chronic renal
lation (INFeD). We recommend usage of only
failure: 200 mg on 5 different days over a 2-
the low molecular weight formulation.
week period (total dose 1000 mg)
Total-dose infusions have been used
safely and are the preferred method of
Notes:
administration, though not currently
Do not give to patients with known hyper-
approved in the United States. Must wait
sensitivity to iron formulations, anemia not
14 days after a dose for reequilibration if
associated with iron deficiency, hemochro-
retesting iron stores.
matosis, hemolytic anemia, or iron overload.
Use with caution in patients with history
IRON SUCROSE
of significant allergies, asthma, hepatic
Venofer
impairment, or rheumatoid arthritis.
Iron salt
Injection: 20 mg elemental iron/mL (5, 10 mL)
ITRACONAZOLE
Pregnancy category B
Sporanox, generic
Antifungal
Indications:
Capsule: 100 mg
Solution: 100 mg/10 mL
Treatment of microcytic, hypochromic ane-
mia resulting from iron deficiency in
Pregnancy category C
chronic kidney disease patients, either dial-
ysis-dependent or nondialysis-dependent,
Indications:
who may or may not be receiving
Treatment of susceptible systemic fungal
erythropoietin
infections including blastomycosis, coccidio-
mycosis,histoplasmosis,paracocciodiomyco-
Dosage:
sis, and aspergillosis in patients who do not
Children: end stage renal disease:
respond to or cannot tolerate amphotericin B;
Formulary
339
treatment of oropharyngeal or esophageal
Maintenance dose:
0.5 mg/kg/dose IM/IV
candidiasis (oral solution only).
q6h; maximum dose 30 mg q6h or 120 mg
q24h
Dosage:
Children >50 kg and adults: 10 mg PO q6h
Children: limited data: 3 to 5 mg/kg/day PO
PRN; maximum dose 40 mg/24 h
once daily
Doses as high as 5 to 10 mg/kg/day divided
Notes:
q12-24h have been used in children with
Do not give to patients with known hyper-
chronic granulomatous disease and for pro-
sensitivity to ketorolac or other NSAIDs,
phylaxis against Aspergillus infection; 6 to
patients with active peptic ulcer disease, gas-
8 mg/kg/day has been utilized in the treat-
trointestinal bleeding or perforation, renal
ment of disseminated histoplasmosis.
dysfunction, bleeding diathesis, thrombocy-
topenia, or cerebrovascular bleeding. Use
Notes:
with caution in patients with congestive heart
Do not give to patients with known hyper-
failure, hypertension, or decreased renal or
sensitivity to itraconzaole or other azole
hepatic function. May cause impaired plate-
drugs. Do not use to treat onychomycosis
let function, nausea, dyspepsia, drowsiness,
in patients with evidence of left ventricular
and interstitial nephritis.
dysfunction, congestive heart failure (CHF)
or a history of CHF, or women who are
LEUCOVORIN CALCIUM
pregnant or intending to become pregnant.
Generic
Use with caution in patients with renal or
Antidote, methotrexate; folic acid derivative
hepatic impairment. Many drug interac-
Tablets: 5, 10, 15, 25 mg
tions exist; consult the Physician’s Desk
Injection: 50, 100, 200, 350 mg vials
Reference. Avoid drinking grapefruit juice
or soda while taking oral itraconazole due to
Pregnancy category C
altered absorption.
Indications:
KETOROLAC TROMETHAMINE
Reduction of toxic effects (leucovorin res-
Generic, previously available as Toradol
cue) of high-dose methotrexate, to coun-
NSAID
teract effects of impaired methotrexate
Tablet: 10 mg
elimination, or as an antidote for
Injection: 15, 30 mg/mL
folic acid antagonist overdose. Indicated for
the treatment of folate-deficient megalo-
Pregnancy category C/D
blastic anemia of infancy, sprue, or preg-
nancy; treatment of nutritional deficiencies
Indications:
when oral folate therapy is not possible.
Short-term management of pain (up to 5
days for parenteral therapy, 5 to 14 days for
Dosage:
oral therapy).
Rescue dose
(following administration of
high-dose methotrexate), see protocols:
Dosage:
15 mg/m2 IV to start, then 15 mg/m2 q6h; if
Loading dose: 1 mg/kg IM; maximum dose
serum creatinine 48 hours after the start of
60 mg (loading dose not necessary in IV
the methotrexate is elevated more than 50%
administration)
or the serum methotrexate concentration is
340
Formulary
>5
106 M, increase dose to 150 mg/m2
Usual dose is 75 to 130 mg/m2 as a single
dose q3h until serum methotrexate level is
dose every
6
weeks; subsequent doses
<1
107 M or per protocol (refer to pub-
adjusted per platelet and leukocyte counts.
lished graphs for methotrexate clearance per
protocol). If methotrexate clearance is
Notes:
delayed, continue leucovorin until level
Do not give to patients with known hyper-
<1
107 M.
sensitivity to lomustine. May need to
Folate-deficient megaloblastic anemia:
adjust dose due to prolonged myelosup-
pression. Delayed pulmonary fibrosis may
1 mg/day IM/IV
occur with high cumulative doses (e.g.,
Megaloblastic anemia secondary to congenital
1g/m2).
deficiency of dihydrofolate reductase:
3 to 6 mg/day IM
MECLORETHAMINE
Folic acid antagonist (e.g., pyrimethamine,
Mustargen
trimethoprim) overdose:
Antineoplastic, alkylating agent
5 to 15 mg/day PO for 3 days or until the
Injection: 10 mg
blood counts are normal or
5 mg every
Pregnancy category D
3 days; doses of 6 mg/day are needed for
patients with platelet counts <100
109/L.
Indications:
Following intrathecal methotrexate (investi-
Treatment of Hodgkin and non-Hodgkin
gational):
lymphoma and brain tumors; sclerosing
12 mg/m2 PO/IV as a single dose
agent in intracavitary therapy of pleural,
pericardial, and other malignant effusions.
Notes:
Do not give to patients with known hyper-
Dosage:
sensitivity to leucovorin, pernicious anemia,
Refer to individual protocol.
or other megaloblastic anemia, such as sec-
ondary to vitamin B12 deficiency. Do not
Children: MOPP regimen
(Mustargen
administer intrathecal or intraventricular
[mechlorethamine], Oncovin
[vincristine],
(may be harmful or fatal).
procarbazine, prednisone):
6 mg/m2 IV on
days 1 and 6 of a 28 day cycle
LOMUSTINE
Brain tumors: MOPP regimen: 3 mg/m2 IV
CCNU
on days 1 and 8 of a 28 day cycle
CeeNU
Intracavitary (adults): 10 to 30 mg or 0.2 to
Antineoplastic, alkylating agent
0.4 mg/kg
Capsules: 10, 40, 100 mg
Pregnancy category D
Notes:
Do not give to patients with known hyper-
Indications:
sensitivity to mechlorethamine, preexisting
Treatment of primary or metastatic brain
profound myelosuppression, or pregnancy.
tumors and Hodgkin lymphoma.
Extravasation may result in severe tissue
damage; treat promptly with cold com-
Dosage:
presses
(6
to
12
hours) and sodium
Refer to individual protocol.
thiosulfate.
Formulary
341
MELPHALAN
MEPERIDINE HYDROCHLORIDE
Alkeran
Demerol, generic
Antineoplastic, alkylating agent
Narcotic, analgesic
Tablet: 2 mg
Tablets: 50, 100 mg
Injection: 50 mg
Elixir: 50 mg/5 mL
Injection: 10, 25, 50, 75, 100 mg/mL
Pregnancy category D
Pregnancy category C/D
Indications:
Indications:
Treatment of neuroblastoma, rhabdomyo-
Management of moderate to severe pain;
sarcoma, brain tumors, acute myelogenous
used as an adjunct to anesthesia and pre-
leukemia, Ewing sarcoma, medulloblas-
operative sedation; treatment of rigors fol-
toma, Hodgkin lymphoma, and for condi-
lowing administration of amphotericin B.
tioning prior to hematopoietic stem cell
transplantation (HSCT).
Dosage:
Dosage:
Children: 1 to 1.5 mg/kg/dose PO, IM, IV, or
SC q3-4h PRN; maximum dose 100 mg
Refer to individual protocol.
Adults: 50 to 150 mg/dose q3-4h PRN
Children: rhabdomyosarcoma: 10 to 35 mg/
m2/dose IV every 21 to 28 days
Notes:
High-dose therapy with HSCT conditioning:
70 to 100 mg/m2 IV on days -7 and -6 prior
Do not give to patients with known
to HSCT; or 140 to 220 mg/m2 IV single
hypersensitivity to meperidine. Avoid in
dose prior to HSCT; or 50 mg/m2/day
4
patients receiving acyclovir, cimetidine, tri-
days; or 70 mg/m2/day
3 days
cyclic antidepressants, and monoamine
oxidase inhibitors within the past 14 days.
Children: oral dosing:
4 to 20 mg/m2/day
Use with caution in renal or hepatic dys-
days 1 to 21
function, sickle cell disease, and seizure
disorders. Accumulation of normeperidine
Notes:
metabolites may precipitate seizures. May
Do not give to patients with known hyper-
cause central nervous system and respira-
sensitivity or to those whose disease was
tory depression, nausea, vomiting, consti-
resistant to prior therapy. Long-term oral
pation, bradycardia, hypotension, periph-
therapy and high cumulative doses (i.e.,
eral vasodilation, miosis, sedation, drows-
>600 mg) can increase the incidence of
iness, biliary or urinary tract spasm,
secondary leukemia. May lead to amenor-
increased intracranial pressure, and physi-
rhea. Causes bone marrow suppression; use
cal and psychological dependence. Other
with caution in those with prior myelosup-
narcotics are generally preferred for pain
pressive chemotherapy or radiation therapy.
management.
Use with caution and consider dose reduc-
tion in patients with renal impairment. May
MERCAPTOPURINE
increase the effects/levels of cyclosporine
6-MP, 6-mercaptopurine
and carmustine.
Purinethol
342
Formulary
Antineoplastic, antimetabolite
With ifosfamide and cyclophosphamide:
Tablet: 50 mg
Mesna dose is 20% of alkylator dose IV
15 minutes before or combined with alky-
Pregnancy category D
lator, followed by repeat doses 4
and 8
hours later; for high-dose alkylator therapy,
Indications:
give dose 15 minutes before alkylator then
Used in conjunction with methotrexate for
q3h for 3 to 6 doses. Total daily Mesna dose
maintenance therapy in acute lymphoblastic
ranges from 60% to 160% of the daily
leukemia; combination regimen in acute
alkylator dose.
myelogenous and chronic myelogenous leu-
IV continuous infusion of Mesna is given at
kemias; non-Hodgkin lymphoma.
doses equivalent to 60% to 100% of the
ifosfamide or cyclophosphamide dose.
Dosage:
Refer to individual protocol.
Mesna is given by IV infusion over 15 to 30
minutes or by continuous IV infusion, or
50 to 100 mg/m2 once daily
per protocol.
Notes:
Notes:
Do not give to patients with known hyper-
sensitivity to mercaptopurine, severe liver
Do not give to patients with known hyper-
disease, or severe bone marrow suppression.
sensitivity to Mesna or thiol compounds.
Use with caution and adjust dose in patients
May cause false positive urinary ketone
with renal or hepatic dysfunction. Patients
measurements.
who receive allopurinol concurrently
should have their mercaptopurine dose
METHADONE HYDROCHLORIDE
reduced by 33%.
Dolophine
Antidote, analgesic
MESNA
Tablet: 5, 10 mg
Mesnex
Solution: 5 mg/5 mL, 10 mg/5 mL
Prophylaxis, cyclophosphamide or ifosfa-
Injection: 10 mg/mL (20 mL)
mide-induced hemorrhagic cystitis.
Pregnancy category B/D
Tablet: 400 mg
Injection: 100 mg/mL
Indications:
Pregnancy category B
Management of severe pain; used in nar-
cotic detoxification maintenance programs
Indications:
and for the treatment of iatrogenic narcotic
Detoxifying agent used to inhibit hemor-
dependency.
rhagic cystitis induced by ifosfamide and
cyclophosphamide.
Dosage:
Dosage:
Children: analgesia:
Refer to individual protocol.
0.7 mg/kg/24 hour divided q4-6h PRN PO,
SC, IM, or IV
Dose is dependent on which antineoplastic
agent it is used with.
Adults: analgesia:
Formulary
343
2.5 to 10 mg PO, IM, IV, or SC q3-4h PRN,
dosing or by continuous infusion IV over 6
up to 5 to 20 mg q6-8h
to 42 hours (dependent on phase of therapy);
50 to 400 mg/m2 IV bolus every 10 days
Detoxification: see Physician’s Desk Reference:
Doses 100 to 500 mg/m2 may require leu-
15 to 40 mg/day PO
covorin and doses >500 mg/m2 require leu-
covorin rescue.
Notes:
Osteosarcoma/solid tumors:
Do not give to patients with known hyper-
sensitivity to methadone. Use with caution
Children <12 years: 12 g/m2 IV over 4 hours
in patients with respiratory disease as respi-
(dose range 12 to 18 g) þ leucovorin rescue
ratory depression lasts longer than analgesic
Children
12 years: 8 g/m2 IV over 4 hours
effects. May cause cardiac arrhythmias,
(maximum dose 18 g) þ leucovorin rescue
sedation, increased intracranial pressure,
Non-Hodgkin lymphoma:
hypotension, and bradycardia, in addition
to respiratory depression. Prolonged half-
200 to 500 mg/m2 IV; repeat every 4 weeks,
life; average
19 hours in children and 35
as per protocol.
hours in adults. Repeated use can result in
Meningeal leukemia (and prophylaxis):
cumulative effects necessitating adjustment
10 to 15 mg/m2 intrathecal (IT) (maximum
to the dose and frequency of administration.
15 mg) per protocol; dosed by age:
Conversion from other narcotics to meth-
Children <1 year: 6 mg
adone can be quite challenging; discussion
with a provider with experience in this area
Children 1 to <2 years: 8 mg
is warranted (see Chapter 28).
Children 2 to <3 years: 10 mg
Children
3 years: 12 to 15 mg
METHOTREXATE
Dilute in 4 to 6 mL 0.9% NaCl or Elliotts B
Rheumatrex, Trexall, generic
solution.
Antineoplastic, antimetabolite, antirheumatic
Intrathecal methotrexate may be combined
Tablet (Trexall): 2.5, 5, 7.5, 10, 15 mg
with other agents for IT administration (i.e.,
Injection: 1 g vial
cytarabine, hydrocortisone).
Pregnancy category X
Trophoblastic neoplasms (adults):
15 to 30 mg/day PO/IM for 5 days; repeat
Indications:
weekly for three to five courses.
Treatment of acute lymphoblastic leukemia
(ALL; including meningeal leukemia), tro-
Notes:
phoblastic neoplasms, osteosarcoma, non-
Do not give to patients with known hyper-
Hodgkin lymphoma, rheumatoid arthritis,
sensitivity to methotrexate, severe renal or
dermatomyositis, and psoriasis.
hepatic impairment, or preexisting profound
bone marrow suppression. High-dose meth-
Dosage:
otrexate (>1 g/m2) should not be adminis-
Refer to individual protocol.
tered to patients with a creatinine clearance of
Acute lymphoblastic leukemia (refer to pro-
less than 50% to 75% of normal. Patients
tocol, phase):
should receive alkaline fluids to maintain
7.5 to 30 mg/m2 once per week or every 2
urine pH of 7 or higher while receiving
weeks PO/IM; 10 to 18,000 mg/m2 bolus
high-dose methotrexate. Follow serum levels
344
Formulary
per protocol and administer leucovorin res-
Methemoglobinemia: 1 to 2 mg/kg (25 to
cue per protocol; follow methotrexate degra-
50 mg/m2) IV as a single dose; may be
dation curve per protocol. Methotrexate has
repeated after 1 hour as necessary
been associated with acute and severe chronic
NADPH-methemoglobin reductase defi-
hepatotoxicity, severe bone marrow suppres-
ciency: 1 to 1.5 mg/kg/day PO (maximum
sion, and renal failure with delayed clearance,
dose 300 mg/day) given with ascorbic acid 5
high-dose administration, concurrent neph-
to 8 mg/kg/day
rotoxic drugs, or inadequate hydration. Intra-
thecal and parenteral administration of meth-
Notes:
otrexate have been associated with acute neu-
Use with caution in patients with G6PD
rotoxicity. Severe dermatologic reactions and
deficiency or renal insufficiency. May cause
radiation dermatitis have been reported. May
nausea, vomiting, dizziness, headache,
accumulate in fluid collections (pleural effu-
abdominal pain, diaphoresis, phototoxicity,
sions, ascites) increasing local toxicity.
and skin staining. May cause transient blue-
Ensure no TMP-SMX, penicillin, NSAIDs,
green coloration of urine and stool. At high
or PPIs are given until the methotrexate level
doses, may cause methemoglobinemia.
is <1
107 M due to competitive excretion
and risk of toxicity secondary to delayed
methotrexate clearance.
METHYLPREDNISOLONE
Intrathecal drugs should be prepared
Medrol, Medrol dose pack, Depo-Medrol,
and administered separately from other che-
Solu-Medrol
motherapeutic drugs to avoid inappropriate
Corticosteroid
administration.
Tablets: 2, 4, 8, 16, 24, 32 mg
Recommend giving patients one pill size
Tablets (dose pack): 4 mg
only to avoid accidental overdose with dose
Injection, sodium succinate
(Solu-Medrol):
modifications during maintenance therapy
40, 125, 500, 1000, 2000 mg (IV/IM)
for ALL.
Injection
(acetate)
(Depo-Medrol): 20, 40,
80 mg/mL (IM)
METHYLENE BLUE
Pregnancy category C
Urolene blue, generic
Antidote, drug-induced methemoglobinemia
Indications:
Tablet (Urolene Blue): 65 mg
Anti-inflammatory or immunosuppressive
Injection: 10 mg/mL
agent used to treat a variety of diseases of
Pregnancy category C/D
hematologic, allergic, inflammatory, neo-
plastic, and autoimmune origin.
Indications:
Dosage:
Antidote for cyanide poisoning and drug-
induced methemoglobinemia; treatment of
Anti-inflammatory/immunosuppressive:
NADPH-methemoglobin reductase defi-
0.5 to 1.7 mg/kg/24 hours PO, IM, or IV
ciency; treatment and prevention of ifosfa-
divided q6-12h
mide-induced encephalopathy (adults).
Chemotherapy:
Refer to individual protocols for dosing (in
Dosage:
lieu of prednisone; convert dose for steroid
Children:
potency, 80% of prednisone dose).
Formulary
345
Notes:
seizure disorder. Sedation, headache, anxi-
Do not give to patients with hypersensitivity
ety, depression, leukopenia, and diarrhea
may occur.
to methylprednisolone. Do not administer
with live-virus vaccines or during active infec-
tion with varicella or herpes zoster. Avoid use
MICAFUNGIN
in patients with systemic fungal infections.
Mycamine
May cause hypertension, glucose intolerance,
Antifungal, echinocandin
gastrointestinal bleeding, osteoporosis, pseu-
Injection: 50, 100 mg
dotumor cerebri, Cushing’s syndrome, adre-
nal axis suppression, and acne. May increase
Pregnancy category C
levels of cyclosporine and tacrolimus.
Indications:
METOCLOPRAMIDE
Treatment of patients with candidemia,
esophageal candidiasis,
disseminated
Reglan, Maxolon, generic
candidiasis; prophylaxis of Candida infec-
Antiemetic
tions in patients undergoing hematopoie-
Tablets: 5, 10 mg
tic stem cell transplantation; treatment
Syrup: 5 mg/5 mL
of invasive Aspergillosis. Not effective
Injection: 5 mg/mL
against cryptococcus, fusariosis, and
Pregnancy category B
zygomycosis.
Indications:
Dosage:
Treatment of gastroesophageal reflux (GER)
Not FDA approved for use in children.
and prevention of nausea and vomiting
Guidelines are per pharmacokinetic
associated with chemotherapy.
studies, short duration trials, and case
reports.
Dosage:
Prophylaxis of Candida infections in HSCT
GER or gastrointestinal (GI) dysmotility:
recipients:
Infants and children: 0.1 to 0.2 mg/kg/dose
Infants, children, and adolescents:
1.5
to
up to QID, PO, IV, or IM; maximum dose
2 mg/kg/day IV daily
0.8 mg/kg/24 hours
Adults: 50 mg IV daily
Adults: 10 to 15 mg/dose on awakening and
Disseminated candidiasis:
at night, IM, PO, or IV
Neonates <1000 g: 10 mg/kg/day IV daily
Antiemetic:
Neonates
1000 g: 7 mg/kg/day IV daily
1 to 2 mg/kg/dose q2-6h PO, IV, or IM.
Give first dose 30 minutes prior to emeto-
Infants, children, and adolescents: 2 to 4 mg/
genic drug. Premedicate with diphenhydra-
kg/day IV daily (maximum dose 200 mg)
mine to reduce incidence of extrapyramidal
Adults: 100 mg IV daily
symptoms.
Aspergillosis, esophageal candidiasis:
Neonates: 8 to 12 mg/kg/IV daily
Notes:
Infants, children, and adolescents:
4
to
Do not give to patients with known hyper-
8.6 mg/kg IV daily (maximum dose 325 mg)
sensitivity to metoclopramide, GI obstruc-
tion, pheochromocytoma, or history of
Adults: 150 mg IV once daily
346
Formulary
Notes:
occur as the cumulative dosage approaches
Do not give to patients with known hyper-
550 mg/m2 (or 400 mg/m2 with chest irra-
diation or concomitant cyclophosphamide
sensitivity to micafungin or other echino-
candins. Use with caution in patients with
administration; based on anthracycline dose
equivalents, see Chapter 30). This may be an
renal or hepatic impairment and in patients
acute or late effect and monitoring with
receiving concomitant hepatotoxic drugs;
ECHO or MUGA and ECG is required per
monitor for evidence of worsening. May
protocol. Extravasation may lead to severe
cause electrolyte disturbances, cytopenias,
tissue damage. Dosage may need to be
skin rash, central nervous system, and car-
reduced in patients with impaired hepato-
diovascular effects.
biliary function, preexisting bone marrow
suppression, or previous treatment with
MITOXANTRONE
cardiotoxic drugs or chest radiation.
Novantrone, generic
Antineoplastic, anthracenedione
MORPHINE SULFATE
Injection, solution: 2 g/mL
Roxanol, Oramorph SR, MS Contin
Pregnancy category D
Narcotic, analgesic
Tablets: 15, 30 mg
Indications:
Controlled-release tablets: 15, 30, 60 mg (100
Treatment of acute myelogenous leukemia;
and 200 mg for opioid tolerant patients)
active in pediatric sarcoma, Hodgkin and
Extended-release capsules:
30 mg (60,
90,
non-Hodgkin lymphoma, acute lympho-
120 mg for opioid tolerant patients)
blastic leukemia, and myelodysplastic
Sustained-release pellets in capsules: 10, 20,
syndrome.
30, 50, 60 mg (100, 200 mg for opioid tol-
erant patients)
Dosage:
Solution: 10, 20, 100 mg/5 mL
Suppository: 5, 10, 20, 30 mg
Leukemia: children
2 years: 0.4 mg/kg IV
Injection:
0.5,
1,
2,
3,
4,
5,
8,
10, 15, 25,
daily for 3 to 5 days
50 mg/mL
Children >2 years and adults: 12 mg/m2
IV daily for 2 to 3 days; acute leukemia
Pregnancy category C/D
in relapse:
8 to
12 mg/m2 IV daily for
5 days; AML: 10 mg/m2 IV daily for 3 to
Indications:
5 days
Relief of moderate to severe pain, acute and
Solid tumors: children: 18 to 20 mg/m2 IV
chronic, after nonnarcotic analgesics have
q3-4 weeks or 5 to 8 mg/m2 IV weekly
failed; preanesthetic medication; relief of
dyspnea from acute left ventricular failure
Adults:
12
to
14 mg/m2 IV q3-4 weeks
and pulmonary edema.
(maximum total 80 to 120 mg/m2)
Dosage:
Notes:
Dose should be titrated to effect.
Do not give to patients with known hyper-
sensitivity to mitoxantrone. May cause
Neonates:
severe myelosuppression; use with caution
0.1 to 0.2 mg/kg IV q3-4h; q6h dosing may
in patients with preexisting myelosuppres-
be appropriate for extremely premature
sion. Irreversible myocardial toxicity may
infants or infants with hepatic dysfunction
Formulary
347
Infants and children:
MYCOPHENOLATE MOFETIL (MMF)
Tablet and solution (immediate release): 0.2
CellCept, Myfortic, generic (capsule, tablet)
to 0.5 mg/kg/dose PO q4-6h PRN
Immunosuppressive agent
Capsule, as mofetil or CellCept: 250 mg
Tablet (controlled release): 0.3 to 0.6 mg/kg/
Tablet, as mofetil or CellCept: 500 mg
dose PO q12h
Tablet, delayed release, as mycophenolic acid,
Injection: 0.05 to 0.2 mg/kg/dose IM, IV, or
Myfortic: 180, 360 mg (not recommended
SC q2-4h PRN; maximum 15 mg/dose
for children whose total body surface area
Adults:
[TBSA] is <1.19 m2)
Tablets and solution
(immediate release):
Injection, as mofetil or CellCept: 500 mg
10 to 30 mg PO q4h PRN
Powder for oral suspension, CellCept:
200 mg/mL
Tablet (controlled release): 15 to 30 mg PO
q8-12h PRN
Pregnancy category D
Injection: 2 to 15 mg/dose IV, IM, or SC
Indications:
Patient-controlled analgesia
(PCA) dosing
guidelines:
Used as an immunosuppressant drug fre-
quently in combination with other immuno-
PCA (on demand) dose: 0.01 to 0.04 mg/kg
suppressants (e.g., cyclosporine, corticoster-
Suggested lock out (between PCA doses): 10
oids) for the prophylaxis of organ rejection
to 20 minutes
(renal, hepatic, cardiac, and bone marrow
Continuous dose:
transplants), chronic graft-versus-host dis-
Neonate 0.01 to 0.02 mg/kg/h
ease, myasthenia gravis, proliferative lupus
Infant/child: 0.01 to 0.07 mg/kg/h
nephritis, and relapsing nephrotic syndrome.
Adult: 0.8 to 10 mg/h (higher doses with
tolerance)
Dosage:
Bolus (loading): 0.03 to 0.1 mg/kg
Limited data are available for pediatric
The one hour maximum is 0.1 to 0.15 mg/kg
dosing. Pharmacokinetic studies have indi-
(6 to 8 mg/h in tolerant adults), and typi-
cated that doses based on body surface area
cally equals the continuous dose plus 2 to 3
result in a more appropriate area under the
PCA bolus doses.
curve than doses based on body weight.
Notes:
Children: 600 mg/m2/dose BID or
Do not give to patients with severe renal or
by TBSA:
liver insufficiency. May cause severe pruritus,
1.25 to 1.5 mg/m2: 750 mg BID
urinary retention, respiratory depression,
>1.5 m2: 1 g BID
central nervous system depression, constipa-
tion, and ileus. Prolonged use can result in
physical and psychological dependence.
Notes:
Consider nighttime low continuous dose
Do not give to patients with known hyper-
infusion of 0.01 to 0.02 mg/kg/h without a
sensitivity to mycophenolate mofetil.
change in lock out so as to allow sleep. Avoid
Risk appears to be associated with the
narcotization with sedation and respiratory
intensity and duration of immunosup-
depression. Monitor vitals and encourage use
pression; may result in an increased inci-
of incentive spirometry and ambulation with
dence of lymphoma or other malignancies
repetitive dosing or PCA use.
especially in combination with other
348
Formulary
immunosuppressants. Patients should
Notes:
avoid excessive exposure to sunlight and
Do not give to patients with known hyper-
should use sun protection factor. Use with
sensitivity to naloxone. The dose for pediatric
caution and adjust dose in patients with
postoperative narcotic reversal is one-tenth
renal impairment.
of the dose for opiate intoxication. Endotra-
cheal administration can be done safely by
NALOXONE
diluting in 1 to 2 mL of normal saline. Will
Narcan, generic
produce narcotic withdrawal in patients with
Narcotic antagonist
dependence. Use with caution in patients
Injection: 0.4 mg/mL
with chronic heart disease. Abrupt reversal
of narcotic dependency may result in nausea,
Pregnancy category C
vomiting, diaphoresis, tachycardia, hyper-
tension, and tremulousness.
Indications:
Used to reverse central nervous system and
NELARABINE
respiratory depression in suspected narcotic
Arranon, ara-G
overdose; treatment of coma of unknown
Antineoplastic, antimetabolite
etiology.
Injection: 5 mg/mL
Pregnancy category D
Dosage:
Treatment of opiate intoxication:
Indications:
Neonates, children
<5 years or
<20 kg:
Treatment of T-cell acute lymphoblastic
0.1 mg/kg/dose; repeat q2-3 minutes PRN
leukemia and T-cell lymphoblastic
IM, IV, SC, or via ETT
lymphoma.
Children
>5 years or
20 kg:
2 mg/dose;
if no response, repeat q2-3 minutes PRN
Dosage:
IM, IV, SC, or via ETT
Refer to individual protocol.
Adults:
0.4
to
2.0 mg/dose q2-3 minutes
Children:
650 mg/m2 daily, days
1 to
5;
PRN IM, IV, SC, or via ETT. Use in 0.1
repeat cycle q21 days
to 0.2 mg increments in opioid-dependent
Adults: 1500 mg/m2/dose on days 1, 3, and
patients. If no response and cumulative dose
5; repeat cycle q21 days
>10 mg, reevaluate diagnosis.
IV continuous infusion: 0.005 mg/kg loading
Notes:
dose, then 0.0025 to 0.16 mg/kg/h. Taper
Do not give to patients with known hyper-
gradually to avoid relapse.
sensitivity to nelarabine. May cause severe
Patient-controlled analgesia side effect rever-
neurotoxicity, including severe somnolence,
sal
(pruritus and/or urinary retention with
seizure, and peripheral neuropathy. Observe
morphine):
closely for neurotoxicity. Adverse effects
IV continuous infusion: begin with 1 mcg/kg/
associated with demyelination or similar to
h, then titrate up or down to 0.25 to 2 mcg/
Guillain-Barre syndrome (ascending periph-
kg/h to abate opioid-related side effects;
eral neuropathies) have been reported.
taper infusion gradually over 2 to 4 hours
Neurotoxicity is dose limited and may not
when discontinuing.
reverse completely. Risk of neurotoxicity
Formulary
349
may increase in patients with concurrent
a history of cardiac arrhythmias. Long term
intrathecal chemotherapy or history of cra-
use may increase the incidence of gallstones
nial irradiation. May cause bone marrow
or sludge formation. May cause neuromus-
suppression. Use with caution in patients
cular, central nervous system, or cardiovas-
with renal or hepatic impairment. Monitor
cular toxicities. May decrease level/effects
blood counts, electrolytes, renal function,
of cyclosporine. Do not give with meals;
and transaminases frequently.
may decrease absorption of vitamin B12
and dietary fats.
OCTREOTIDE ACETATE
Sandostatin, generic
ONDANSETRON
Antidiarrheal, antidote, antihemorrhagic,
Zofran
antisecretory, somatostain analog
Antiemetic, serotonin 5-HT3 antagonist
Injection: 0.2, 1 mg/mL
Tablets: 4, 8, 16, 24 mg
Injection, preservative free:
0.05,
0.1,
Tablets (ODT, orally disintegrating): 4, 8 mg
0.5 mg/mL
Solution: 4 mg/5 mL
Pregnancy category B
Injection: 2 mg/mL
Injection, premixed in D5W: 32 mg/50 mL
Indications:
Pregnancy category B
Treatment of secretory diarrhea in patients
with metastatic carcinoid or vasoactive
Indications:
intestinal peptide-secreting tumors, chemo-
Prevention of nausea and vomiting associ-
therapy-induced diarrhea, graft-versus-host
ated with initial and repeat courses of eme-
disease associated diarrhea, and esophageal
togenic cancer chemotherapy or radiation,
varices/gastrointestinal (GI) bleeding.
prevention of postoperative nausea and
vomiting, treatment of emesis induced by
Dosage:
acute gastroenteritis.
Infants and children: (data limited to small
studies): diarrhea:
1
to
10 mcg/kg IV/SC
Dosage:
q12h; begin at low dose and titrate to effect
Prevention of chemotherapy-induced nausea
or IV continuous infusion 1 mcg/kg bolus
and vomiting:
followed by continuous infusion 1 mcg/kg/h.
IV: 0.15 mg/kg/dose 30 minutes prior to the
Esophageal varices/GI bleeding: 1 to 2 mcg/kg
start of emetogenic chemotherapy, with sub-
initial IV bolus followed by 1 to 2 mcg/kg/h
sequent doses administered 4 and 8 hours
continuous infusion; titrate to response,
after the first dose or every 8 hours until the
taper doses by 50% q12h when no active
chemotherapy is complete. For highly eme-
bleeding occurs for 24 hours, may discon-
togenic drugs, give 0.45 mg/kg as a single
tinue dose when at 25% of initial dose.
dose 30 minutes prior to the start of chemo-
therapy, followed by 0.15 mg/kg/dose q4h
Notes:
PRN. Maximum single dose 32 mg.
Do not give to patients with known hyper-
Oral: Dose based on total body surface area
sensitivity to octreotide. May need to adjust
(TBSA), weight, or age:
dose for patients with diabetes; may alter
TBSA:
insulin requirements. Use with caution in
patients with renal or hepatic impairment or
<0.3 m2: 1 mg TID PRN
350
Formulary
0.3 to 0.6 m2: 2 mg TID PRN
Indications:
0.6 to 1 m2: 3 mg TID PRN
Treatment of relapsed or refractory solid
tumors, brain tumors, and non-Hodgkin
>1 m2: 4 to 8 mg TID PRN
lymphoma.
Weight:
Same as IV dosing; round to closest conve-
Dosage:
nient dose
Refer to individual protocol.
Age:
Children:
1 year: 4.3 mg/kg IV over 2 hours
<11 years: dose based on TBSA
q3 weeks
Children
11 years and adults: 8 mg TID
Children:
>1 year: 130 mg/m2 IV over 2
PRN; first dose 30 minutes prior to start of
hours q3 weeks
chemotherapy
Notes:
Notes:
Do not give to patients with known hypersen-
Do not give to patients with known hyper-
sitivity to oxaliplatin, in pregnancy, or in those
sensitivity to ondansetron. Ondansetron is a
with grade 3 or 4 neuropathy (usually due to
substrate for the cytochrome P450 enzyme
prior exposure). Anaphylaxis may occur
system, so inducers or inhibitors of this
within minutes of administration; appropri-
system may affect the elimination of ondan-
ate supportive and resuscitative medications
setron. Data are limited for use in children
and equipment should be available. Two dif-
under 3 years of age. May need to adjust
ferent types of neuropathy may occur: (1) an
dose and interval for severe hepatic
acute
(within
2 days) reversible
(resolves
impairment. Side effects are usually mild,
within 14 days) sensory neuropathy with
with headache, sedation, constipation, and
peripheral symptoms that are often exacer-
dry mouth being the most common. May
bated by cold (may include pharyngolaryngeal
also cause bronchospasm, tachycardia,
dysesthesia), and (2) persistent (over 14 days)
hypokalemia, seizures, lightheadedness,
sensory neuropathy that presents with par-
diarrhea, transient increases in bilirubin
esthesias, dysesthesias, hypoesthesias, and
or transaminases, and transient blindness
impaired proprioception that interferes with
(minutes to 48 hours). ECG changes (QT
activities of daily living. Symptoms may
prolongation) have been reported. Data are
improve with discontinuing treatment. May
not available for use in children with radi-
cause pulmonary fibrosis or hepatotoxicity.
ation-induced nausea and vomiting. How-
When administered as sequential infusions,
ever, based on efficacy in adults and safety in
taxane derivatives should be administered
children, it is commonly used. Give 1 to 2
before platinum derivatives to limit myelo-
hours prior to radiation; may need round
suppression and enhance efficacy.
the clock dosing for abdominal radiation.
PENTAMIDINE ISETHIONATE
OXALIPLATIN
Pentam 300, NebuPent
Antibiotic, antiprotozoal
Eloxatin, generic
Antineoplastic, alkylating agent
Injection: 300 mg vial (Pentam 300)
Injection: 5 mg/mL
Inhalation: 300 mg vial (NebuPent)
Pregnancy category D
Pregnancy category C
Formulary
351
Indications:
Suspension: 40 mg/mL
Treatment of Pneumocystis jiroveci pneumo-
Pregnancy category C
nia (PCP) in patients who cannot tolerate or
who fail to respond to trimethoprim-sulfa-
Indications:
methoxazole, prevention of PCP infection in
Prophylaxis of invasive Aspergillus and
immunocompromised hosts, treatment of
Candida infections in high risk, severely
African trypanosomiasis, and treatment of
immunocompromised patients (e.g., he-
visceral and cutaneous leishmaniasis caused
matopoietic stem cell transplant recipient,
by Leishmania donovani.
graft-versus-host disease, hematologic
Dosage:
malignancy with prolonged chemother-
Prophylaxis for PCP:
apy-induced neutropenia); treatment of
oropharyngeal candidiasis
(including
4 mg/kg/24 hours IM/IV (over 1 to 2 hours)
those refractory to itraconzaole and/or
every 2 to 4 weeks
fluconazole); treatment of serious invasive
Inhalation (
5 years): 300 mg in 6 mL water
fungal infections including zygomycosis
via inhalation qmonth (use with Respirgard
and coccidioidomycosis in patients intol-
II nebulizer)
erant or refractory to other antifungal
Maximum single dose 300 mg
therapy.
Treatment of PCP:
4 mg/kg/dose IM/IV daily for 14 to 21 days
Dosage:
(preferably IV)
Children
13 years and adults:
Prophylaxis of invasive Aspergillus and Can-
Notes:
dida infections: 200 mg TID
Do not give to patients with known hypersen-
Treatment of refractory, invasive fungal infec-
sitivity to pentamidine isethionate. Adjust
tions:
800 mg/day in divided doses (given
dose in renal impairment. Use with caution
BID to QID)
in patients with diabetes mellitus, renal or
Treatment of orophargyngeal candidiasis:
hepatic dysfunction, hypertension, or hypo-
tension. Additive toxicity may occur with
Initial: 100 mg BID on day 1, then 100 mg
aminoglycosides, amphotericin B, cisplatin,
daily for 13 days
and vancomycin. Can see Jarisch-Herxhei-
Refractory: 400 mg BID
mer-like reaction
(fever, chills, headache,
myalgia). Inhalation therapy may cause irrita-
Notes:
tion of the airway, bronchospasm, cough,
Do not give to patients with known hyper-
oxygen desaturation, dyspnea, and loss of
sensitivity to posaconazole and avoid con-
appetite. May cause hypoglycemia, hypergly-
current administration with ergot alka-
cemia, hypotension (with infusion <2 hours),
loids. Cross sensitivity reactions with other
nausea, vomiting, fever, mild hepatotoxicity,
azoles have not yet been studied but may
pancreatitis, hypocalcemia, megaloblastic ane-
exist. May cause hepatotoxicity, and fre-
mia, granulocytopenia, and nephrotoxicity.
quent monitoring is recommended. May
alter drug levels of cyclosporine
(also
POSACONAZOLE
tacrolimus and sirolimus) resulting in
Noxafil
nephrotoxicity, leukoencephalopathy, and
Antifungal
death. Use with caution in neonates (oral
352
Formulary
solution contains sodium benzoate, a
thromboembolic disorders. May cause adre-
metabolite of benzyl alcohol, which has
nal axis suppression, hypertension, hyper-
been associated with a potentially fatal
glycemia, irritability, gastritis, skin atrophy,
toxicity).
osteoporosis, cataracts, fluid retention,
mood lability, nausea, diarrhea, bone pain,
PREDNISONE
acne, and weight gain. Consider use of
antacid in long-term therapy. Avascular
Deltasone, Liquid Pred, Orasone, generic
necrosis and growth retardation are seen
Corticosteroid
in patients with long-term or repeated
Tablets: 1, 2.5, 5, 10, 20, 50 mg
high-dose therapy. Ensure patients are
Solution: 1, 5 mg/mL
aware of risk of varicella infection in non-
Pregnancy category C/D
immune states.
Indications:
PROCARBAZINE HYDROCHLORIDE
Management of adrenocortical insuffi-
Matulane
ciency; used for anti-inflammatory or
Antineoplastic, alkylating agent
immunosuppressant effects in autoimmune
Capsule: 50 mg
diseases such as immune thrombocytopenia
Pregnancy category D
purpura; chemotherapy for acute lympho-
blastic leukemia and Hodgkin and non-
Indications:
Hodgkin lymphoma.
Treatment of Hodgkin lymphoma and
brain tumors.
Dosage:
Dose is dependent on condition being trea-
Dosage:
ted and response of patient. Consider alter-
Refer to individual protocol.
nate day dosing for long-term therapy. Dis-
continuation of long-term therapy requires
50 to 100 mg/m2 daily for 10 to 14 days
gradual tapering.
Notes:
Anti-inflammatory or immunosuppressant
(includes immune thrombocytopenic pur-
Avoid in patients with known hypersensi-
pura, aplastic anemia):
tivity to procarbazine or with preexisting
bone marrow aplasia. May cause nausea,
0.5 to 2 mg/kg/day PO divided 1-3 times daily
vomiting, dry mouth, constipation, head-
Chemotherapy:
ache, dizziness, and hair loss. Use with
40 to 180 mg/m2/day, as per protocol
caution and reduce dose in patients with
renal or hepatic impairment or marrow
Notes:
suppression. May potentiate central ner-
Avoid use in patients with life-threatening
vous system depression when used with
infections (except septic shock or tubercu-
phenothiazine derivatives, barbiturates,
lous meningitis), systemic fungal infections,
narcotics, alcohol, tricyclic antidepres-
varicella, and in those with hypersensitivity
sants, and methyldopa. Drug (e.g., mono-
to prednisone. Use with caution in patients
amine oxidase inhibitors) and food inter-
with hypothyroidism, cirrhosis, hyperten-
actions are common. Avoid food with high
sion, congestive heart failure, ulcerative
tyramine content
(i.e., aged cheese or
colitis,
gastrointestinal bleeding, and
meats, tea, dark beer, coffee, cola drinks,
Formulary
353
wine, soybean products, peanuts, avoca-
bath and analyzed within 4 hours. Collec-
does, bananas) because hypertensive crisis,
tion at room temperature may lead to
tremor, excitation, cardiac palpitations,
spuriously low uric acid levels due to con-
and angina may occur.
tinued degradation by rasburicase in the
sample.
RASBURICASE
RHD IMMUNE GLOBULIN
Elitek
Recombinant urate oxidase, uric acid low-
WinRho SDF, Rhophylac
ering agent, antigout agent
Immune globulin
Injection: 1.5, 7.5 mg vials
Injection: 600, 1500, 2500, 5000, 15,000 IU
(1 mcg ¼ 5 IU)
Pregnancy category C
Pregnancy category C
Indications:
Initial management or prevention of hyper-
Indications:
uricemia in high risk patients with leuke-
Treatment of immune thrombocytopenic
mia, lymphoma, or a select group of patients
purpura (ITP) in nonsplenectomized RhD-
with solid tumors at risk for tumor lysis
positive patients; suppression of RhD isoim-
syndrome with initiation of chemotherapy.
munization in an RhD-negative individual
Indicated only for short-term use; maxi-
exposed to RhD-positive blood or during
mum of 5 days.
delivery (or pregnancy) of an RhD-positive
infant (if father known to be RhD-positive
Dosage:
or status unknown).
0.15 to 0.2 mg/kg/dose IV over 30 minutes
(round down to nearest 1.5 mg). May repeat
Dosage:
q24h PRN up to 4 additional doses. Admin-
ITP:
ister until the uric acid is normal and the
Hemoglobin
10 g/dL: 50 mcg/kg IV (250 IU/
patient is stable. Most patients respond to
kg), administer dose over 3 to 5 minutes
1 dose.
Hemoglobin <10 g/dL: 25 to 40 mcg/kg IV
Give prior to chemotherapy if expected
(125 to 200 IU/kg)
massive tumor lysis and/or renal involve-
New onset ITP: 75 mcg/kg (375 IU/kg) as a
ment/dysfunction (e.g., Burkitt lymphoma
single dose has been used safely and effica-
with bulky disease).
ciously in pediatric patients with platelets
20
109/L
Notes:
Dose frequency is dependent on duration of
Do not give to patients with known hyper-
response, clinical symptoms, and adverse
sensitivity to rasburicase. Contraindicated
effects; may be given q2-4 weeks.
in patients with G6PD deficiency
(may
cause hemolysis) or severe asthma. Methe-
Notes:
moglobinemia has been reported with
use. May cause vomiting, nausea, fever,
Avoid in patients with known hypersensi-
headache, abdominal pain, constipation,
tivity to immunoglobulins or thimerosal
diarrhea, and rash. When measuring serum
and in patients with IgA deficiency. Use
uric acid levels, ensure tubes are prechilled
with extreme caution in patients with a
and samples are placed in an ice water
hemoglobin less than 8 g/dL. Adverse events
354
Formulary
associated with administration in ITP in-
Notes:
clude headache, chills, fever, and reduction
Rare cases of progressive multifocal leu-
in hemoglobin (resulting from the destruc-
koencephalopathy due to Creutzfeldt-Ja-
tion of RhD-positive red cells). May inter-
kob virus have been reported. Infusional
fere with immune response to live-virus
toxicity is common and primarily avoided
vaccines. Causes acute hemolysis with
with a slow infusion and premedication.
an average hemoglobin decrease of 1.5 to
Most common, infusional toxicity is seen
2 g/dL at 1 week. Do not use in patients with
with the first infusion and reactions may
active bleeding. Current FDA guidelines
include development of hives, broncho-
strongly recommend that patients be mon-
spasm, hypoxia, hypotension, pulmonary
itored in a health care center for 8 hours
infiltrates, and respiratory distress syn-
after a dose of RhD immune globulin, with
drome. Some reactions have been fatal.
urinalyses at baseline and 2, 4, and 8 hours
Close monitoring is required for all infu-
after administration due to risk of severe
sions and appropriate resuscitative medi-
hemolysis.
cations and equipment must be close by. In
the case of mild reactions, temporary ces-
sation of the infusion is indicated with
RITUXIMAB
acetaminophen, diphenhydramine, and
Rituxan
fluids as indicated. Hydrocortisone may
Antineoplastic, monoclonal antibody
also be indicated. If symptoms resolve,
Injection: 10 mg/mL
reinitiate the infusion at a 50% reduced
Pregnancy category C
rate and monitor closely. Subsequent infu-
sions should run slowly and the patient
Indications:
should be premedicated. Rituximab should
be discontinued altogether following a life-
Treatment of relapsed or refractory low
threatening reaction. Use with caution in
grade or follicular CD20
positive, B-cell
patients with preexisting pulmonary or
non-Hodgkin lymphoma; first-line therapy
cardiac conditions and in those at risk for
for diffuse large B-cell non-Hodgkin lym-
development of tumor lysis syndrome.
phoma in combination with other che-
Causes immunosuppression. Quantitative
motherapeutics; systemic autoimmune dis-
immunoglobulins and specific antibody
orders including autoimmune hemolytic
response should be checked prior to and
anemia and immune thrombocytopenia
following treatment with rituximab to
purpura; posttransplant lymphoprolifera-
determine if immunoglobulin infusions
tive disorder; rheumatoid arthritis; refrac-
might be needed post treatment until
tory graft-versus-host disease; refractory
recovery of immune function. Reactivation
systemic lupus erythematosis.
of hepatitis B has been noted (mainly in
adult patients); thus, immune status to
Dosage:
hepatitis B virus should be documented.
Refer to individual protocol.
Although specific to B-cells, Pneumocystis
Children and adults: 375 mg/m2 IV qweek
jiroveci pneumonia (PCP) infection after
for 4 weeks (range 3 to 6 weeks)
rituximab has been reported; therefore,
Courses may be repeated for treatment of
patients should receive PCP prophylaxis
malignancies or recurrent symptoms with
for 6 months or until recovery of immune
autoimmune disorders.
function is documented.
Formulary
355
SARGRAMOSTIM
purpura, known hypersensitivity to gran-
GM-CSF
ulocyte-macrophage colony-stimulating
factors, or yeast-derived products. Use
Leukine
Colony-stimulating factor
with caution in patients with autoimmune
or chronic inflammatory conditions,
Injection: 250, 500 mcg
hypertension, cardiovascular disease, pul-
Pregnancy category C
monary disease, or renal or hepatic
impairment.
Indications:
Accelerates myeloid recovery and engraft-
SIROLIMUS
ment after autologous or allogeneic
Rapamune
hematopoietic stem cell transplantation
Immunosuppressant agent
(HSCT); mobilizes hematopoietic progen-
Solution: 1 mg/mL
itor cells into peripheral blood for collec-
Tablet: 1, 2 mg
tion by leukapheresis; increases neutrophil
counts in patients receiving myelosup-
Pregnancy category C
pressive chemotherapy; used in neonatal
neutropenia.
Indications:
Graft-versus-host disease prophylaxis in
Dosage:
hematopoietic stem cell transplant recipi-
Neonates with neutropenia and sepsis:
ents; prophylaxis and treatment of rejection
in renal transplant patients; primary immu-
10 mcg/kg/day IV/SC for 5 days
nosuppression in other organ transplant.
Children and adults (no FDA approved dos-
ing in children) after HSCT:
Dosage:
250 mcg/kg/day IV/SC for 21 days, begin-
Refer to individual protocol.
ning 2 to 4 hours after marrow infusion on
Children
13 years of age and <40 kg: Load-
day 0 of HSCT or not sooner than 24 hours
ing dose 3 mg/m2 on day 1 followed by
after chemotherapy. Administer as a 30-
maintenance
1 mg/m2/day divided q12h
minute, 2-hour, or 6-hour infusion. Reduce
or once daily; adjust dose to achieve target
dose to 125 mcg/kg if adverse effect (after
sirolimus trough blood concentration.
resolution).
Administer daily until the absolute neutro-
Notes:
phil count is
5.0
109/L for 1 day or until
Do not give to patients with known hyper-
day þ21 post-HSCT, whichever comes first
sensitivity to sirolimus. Immunosuppres-
(or per protocol).
sion may result in increased susceptibility
Do not administer within 24 hours prior to
to infection. May increase risk for develop-
or after chemotherapy or 12 hours prior to
ment of lymphoma or other malignancy.
radiation therapy.
Avoid excessive sun exposure and use sun
protection factor. Severe hypersensitivity
Notes:
and skin reactions have been reported.
May lead to excessive immature myeloid
Monitor renal and hepatic function; dose
cells in the peripheral blood or bone mar-
reduction may be necessary. Use with cau-
row (>10%). Avoid in patients with a
tion in the perioperative period due to an
history of immune thrombocytopenic
increased risk of surgical complications
356
Formulary
from wound dehiscence and impaired
Dosage:
wound and tissue healing. Prophylaxis
Lead chelation, children:
against Pneumocystis jiroveci and cytomeg-
10 mg/kg/dose (350 mg/m2/dose) PO q8h
alovirus is recommended.
for 5 days, followed by 10 mg/kg/dose PO
q12h for 14 days. Repeat courses separated
SODIUM THIOSULFATE
by a minimum of 2 weeks may be necessary
Versiclear
to treat rebound lead concentrations
Antidote, extravasation of mechloretha-
(resulting from mobilization of lead from
mine, cisplatin, or cyanide; antifungal agent
bone stores).
(topical)
Injection: 100, 250 mg/mL
Notes:
Do not give to patients with known hyper-
Indications:
sensitivity to succimer. Use with caution
Treatment of extravasations of selected che-
in impaired hepatic or renal function. May
motherapeutic agents.
see gastrointestinal symptoms, increased
transaminases, rash, headache, and dizzi-
Dosage:
ness. Do not administer with other chela-
Chemotherapy infiltration: children and
tors. Treat iron deficiency and eliminate
environmental exposure to lead. Monitor
adults:
lead levels. May sprinkle contents of the
Mechlorethamine: use
2 mL for each mg
capsules on food if unable to swallow.
infiltrated
TACROLIMUS
Notes:
Prograf, generic
Do not give to patients with known hyper-
Immunosuppressant agent
sensitivity to sodium thiosulfate. Inject
Capsule: 0.5, 1, 5 mg
slowly, over at least
10
minutes; rapid
Injection: 5 mg/mL
administration may cause hypotension.
SUCCIMER
Indications:
Chemet
(2,3-dimercaptosuccinic acid;
Prevention of organ rejection in solid organ
DMSA)
transplant patients; prevention or treat-
Antidote (lead toxicity), chelating agent
ment of graft-versus-host disease (GVHD)
Capsule: 100 mg
in allogeneic hematopoietic stem cell trans-
plantation.
Pregnancy category C
Dosage:
Indications:
Children: younger children usually require
Treatment of lead poisoning in asymptom-
higher dosing on a mg/kg basis than older
atic children with blood levels between 45
and
69.9 mcg/dL. Can be considered at
children.
lower lead levels
(20
to
44.9 mcg/dL) if
Solid organ transplant: initial dose 0.15 to
aggressive environmental interventions
0.3 mg/kg/day PO divided q12h; may give
have not impacted level although not noted
IV continuous infusion 0.01 to 0.06 mg/kg/
to improve neurocognitive outcome.
day (refer to specific organ protocol)
Formulary
357
Prevention of GVHD: initial 0.03 mg/kg/day
Metronomic dosing: 75 mg/m2/day 5 days/
(based on lean body weight) as IV contin-
week, 21 to 42 day cycles
uous infusion. Begin 24 hours prior to stem
May also be administered concurrently with
cell infusion and continue until oral med-
radiation therapy.
ication can be tolerated. May occasionally
be given as a q12h IV infusion.
Notes:
Conversion from IV to PO dose (1:4 ratio):
Do not give to patients with known
Multiply total daily IV dose by 4 and admin-
hypersensitivity to temozolomide, dacar-
ister in two divided oral doses per day.
bazine, or any component. Thrombocyto-
penia and neutropenia are dose-limiting
Notes:
toxicities and may occur late in the treat-
Do not give to patients with known hypersen-
ment cycle and take 14 days to resolve.
sitivity to tacrolimus, castor oil, or any com-
Monitor blood counts and ensure platelet
ponent. Immunosuppression may increase
count
100
109/L and ANC
1.5
109/L
risk of infection or development of lymphoma
prior to initiation of each cycle of therapy.
or other malignancies. Risk is related to inten-
Rare cases of aplastic anemia, myelodys-
sity and duration of use. May cause nephro-
plasia, and secondary malignancies have
toxicity and neurotoxicity. Do not administer
been reported. Prophylaxis against Pneu-
concurrently with cyclosporine. Monitor elec-
mocystis jiroveci is required, especially
trolytes. Monitor tacrolimus drug levels and
with concurrent administration of irradia-
adjust dosages per protocol.
tion. Avoid in pregnancy; may cause fetal
harm.
TEMOZOLOMIDE
Temodar
THIOGUANINE
Antineoplastic, alkylating agent
6-TG, 6-Thioguanine
Capsule: 5, 20, 100, 140, 180, 250 mg
Antineoplastic, antimetabolite
Injection: 100 mg
Tablet (scored): 40 mg
Pregnancy category D
Pregnancy category D
Indications:
Indications:
Treatment of refractory anaplastic astrocy-
Remission induction and consolidation
toma; treatment of newly diagnosed glio-
phases in acute myelogenous leukemia;
blastoma multiforme; active against recur-
delayed intensification phase in acute lym-
rent glioblastoma multiforme, metastatic
phoblastic leukemia.
melanoma, brain stem glioma, ependy-
moma, medulloblastoma, primitive neu-
roectodermal tumor
(PNET), neuroblas-
Dosage:
toma, and anaplastic oligodendroglioma.
Refer to individual protocol.
Infants and children <3 years: 3.3 mg/kg/day
Dosage:
PO divided q12h for 4 days
Refer to individual protocol.
Children
3 years and adults: 50 to 200 mg/
Children:
100 to 200 mg/m2 IV/PO once
m2/day PO divided q12-24h for 4 to 14 days
daily for 5 days every 28 days
or per protocol
358
Formulary
Notes:
THROMBIN, TOPICAL
Do not give to patients with known hyper-
Evithrom, Recothrom, Thrombi-Gel,
sensitivity to thioguanine. Use with caution
Thrombi-Pad, Thrombin-JMI (epistaxis kit,
and reduce dosage in patients with renal or
spray kit)
hepatic impairment. May cause nausea,
Hemostatic agent
vomiting, anorexia, stomatitis, diarrhea,
Powder (Recothrom): 5000, 20,000 unit vials
myelosuppression, and veno-occlusive dis-
Powder, topical
(bovine, Thrombin-JMI):
ease
(sinusoidal obstructive syndrome).
available in epistaxis kit
5000 IU, spray
Increases busulfan toxicity.
20,000 IU
Solution, topical (Evithrom): 800 to 1200 IU/
THIOTEPA
mL
Sponge, topical
(Thrombi-Gel):
1000,
Thioplex, generic
2000 IU
Antineoplastic, alkylating agent
Pad, topical (bovine, Thrombi-Pad):
200 IU
Powder for injection: 15, 30 mg
Pregnancy category C
Pregnancy category D
Indications:
Indications:
Hemostasis for minor bleeding from acces-
Treatment of superficial tumors of the blad-
sible capillaries and small venules.
der, brain tumors, and other meningeal
neoplasms (including intrathecal adminis-
Dosage:
tration); control of pleural, pericardial, or
Apply powder directly to the site of bleeding
peritoneal effusions caused by metastatic
or on oozing surface, or use
1000
to
tumors; also used in high-dose regimens
2000 IU/mL of solution where bleeding is
with autologous hematopoietic stem cell
profuse. May be diluted with NS to other
transplantation (HSCT).
concentrations as needed. Use 100 IU/mL
for bleeding from skin or mucosal surfaces.
Dosage:
May utilize epistaxis/spray kit for mucosal/
Refer to individual protocol.
nose bleeds.
Children, HSCT:
300 mg/m2/dose over
3
hours IV; repeat q24h for 3 doses. Maximum
Notes:
tolerated dose over
3
days is
900
to
Avoid in patients with known hypersensitiv-
1125 mg/m2.
ity to thrombin. Some forms are of human or
Intrathecal: 5 to 11.5 mg/m2 per dose weekly
bovine origin. May cause fever or allergic
for 2 to 7 doses
reactions. Topical use only. Not for admin-
istration systemically or directly into sites of
Notes:
brisk arterial bleeding. Avoid in patients with
factor V deficiency due to cross-reactivity.
Do not give to patients with known hypersen-
sitivity to thiotepa or severe myelosuppression.
TOPOTECAN
Reduce dose in patients with renal, hepatic, or
bone marrow dysfunction. May cause central
Hycamtin
nervous system changes, skin hyperpigmenta-
Antineoplastic, camptothecin, topoisomer-
tion, nausea, vomiting, hematuria, and eleva-
ase inhibitor
tion of liver transaminases and bilirubin.
Capsule: 0.25, 1 mg
Formulary
359
Injection: 4 mg
Indications:
Short-term use (2 to 8 days) in hemophilia
Pregnancy category D
or von Willebrand disease patients for
mucosal bleeding (including tooth extrac-
Indications:
tion) to reduce or prevent hemorrhage and
Treatment of pediatric solid tumors, includ-
reduce need for factor replacement. May
ing sarcoma and neuroblastoma.
be used to treat primary menorrhagia or
recurrent epistaxis, to prevent gastrointes-
Dosage:
tinal or ocular hemorrhage following
Refer to individual protocol.
trauma, and to decrease perioperative
Children:
blood loss and need for transfusion in
patients undergoing congenital heart dis-
Pediatric solid tumors: 1 mg/m2/day (range
ease or scoliosis surgery.
0.75
to
1.9 mg/m2/day) for
3 days as a
continuous IV infusion; repeat q3 weeks
Dosage:
Single agent therapy for refractory solid tumors
Children and adults:
or hematologic malignancy:
2.4 mg/m2/day
once daily for 5 days of a 21 day course
Tooth extraction in patients with hemophilia:
Combination therapy for solid tumors:
10 mg/kg immediately before oral surgery
0.75 mg/m2/dose once daily for
5
days
IV, then 10 mg/kg/dose IV/PO q6-8h; may
every
21
days in combination with
be used for 2 to 8 days
cyclophosphamide
Menorrhagia:
1300 mg PO TID for up to 5 days during
Notes:
monthly menstruation
Do not give to patients with known hyper-
sensitivity to topotecan. Bone marrow tox-
Notes:
icity is dose limiting, primarily neutropenia.
Do not use in patients with known sub-
Has been associated with neutropenic coli-
arachnoid hemorrhage, active intravascu-
tis; should be a strong consideration in
lar clotting process, or acquired defective
patients with neutropenia, fever, and
color vision. Use with caution in patients
abdominal pain. Severe diarrhea has been
with cardiovascular or cerebrovascular dis-
reported, may require dose adjustment.
ease. Dose modification is required in
May cause fetal harm; must ensure contra-
patients with renal impairment. May cause
ception to avoid pregnancy. Use with cau-
hypotension, thromboembolic complica-
tion in patients with renal or hepatic
tions, headache, and visual abnormalities
impairment.
(seen in animals). Do not administer con-
comitantly with factor IX and prothrom-
bin complex concentrates or hormonal
TRANEXAMIC ACID
contraception due to increased risk of
Cyklokapron, Lysteda
thrombosis. Use with caution in patients
Antifibrinolytic, antihemophilic, hemo-
with upper urinary tract bleeding due to
static agent
potential for clot formation and ureteral
Tablet (Lysteda): 650 mg
obstruction. Use with extreme caution in
Injection (Cyklokapron): 100 mg/mL
patients with disseminated intravascular
Pregnancy category B
coagulation.
360
Formulary
TRIMETHOPRIM/
with G6PD deficiency and impaired renal or
SULFAMETHOXAZOLE
hepatic impairment. Serious adverse reac-
tions include Stevens-Johnson syndrome,
Bactrim, Septra, Co-trimoxazole, Sulfatrim,
TMP-SMX, generic
toxic epidermal necrolysis, hepatic necrosis,
agranulocytosis, aplastic anemia, and other
Antibiotic, sulfonamide derivative
blood dyscrasias. Discontinue with rash.
Tablet, single strength: 80 mg trimethoprim
May need to be held temporarily in oncology
(TMP) and 400 mg sulfamethoxazole (SMX)
patients who are on TMP-SMX for PCP
Tablet, double strength:
160 mg TMP and
prophylaxis and subsequently develop neu-
800 mg SMX
tropenia. Numerous drug interactions are
Suspension: 40 mg TMP/200 mg SMX per
reported. TMP-SMX decreases the clearance
5 mL
of warfarin and methotrexate, decreases
Injection: 16 mg TMP/80 mg SMX per mL
serum cyclosporine concentrations, and
Pregnancy category C/D
increases the effect of sulfonylureas, pheny-
toin, and thiopental. Do not give to patients
Indications:
within 24 hours of receiving high-dose meth-
Oral treatment of urinary tract infection
otrexate and until level is <1
107 M due
(UTI) and otitis media; prophylaxis for
to competitive excretion and increased risk of
Pneumocystis jiroveci pneumonia (PCP); IV
methotrexate toxicity due to delayed clearance.
treatment of documented or suspected PCP
infection in immunocompromised patients.
VALACYCLOVIR
Valtrex, generic
Dosage:
Antiviral
Dosage recommendations are based on the
Caplet: 500 mg, 1 g
TMP component. May be given PO or IV.
Pregnancy category B
Children >2 months of age:
Minor/moderate infection:
Indications:
8 to 12 mg TMP/kg/day PO divided q12h
Treatment of herpes zoster and herpes sim-
plex virus (HSV) in immunocompromised
Serious infection/PCP:
patients; treatment of varicella zoster virus
20 mg TMP/kg/day divided q6-8h PO/IV
(VZV) in immunocompetent children (ages
UTI or otitis media prophylaxis:
2 to 18 years); treatment of HSV labialis in
2 to 4 mg TMP/kg once daily
adolescents and adults.
Prophylaxis for PCP:
Dosage:
5 mg TMP/kg/day divided q12h 2 to 3 con-
Children:
secutive days a week
(maximum dose
Varicella, immunocompetent: 2 years to <18
320 mg TMP/day)
years:
20 mg/kg/dose PO TID for 5 days
(maximum 1 g TID); initiate within 24
Notes:
hours of onset of rash
Do not give to patients with known hyper-
Herpes labialis (cold sores): >12 years: 2 g
sensitivity to sulfa drugs or any component,
q12h for 1 day; initiated at earliest symptoms
porphyria, or megaloblastic anemia due to
folate deficiency. Do not use in infants under
Immunocompromised children at risk for
2 months of age. Use with caution in patients
HSV or VZV infection with normal renal
Formulary
361
function: (limited data) 15 to 30 mg/kg/dose
myalgia, paresthesia, constipation, abdom-
PO TID (maximum 2 g/dose)
inal pain, ileus, and mild alopecia.
Intrathecal administration is fatal.
Notes:
Do not give to patients with known hypersen-
VINCRISTINE SULFATE
sitivity to valacyclovir or acyclovir. Use with
Vincasar, generic
caution in patients with renal impairment or
Antineoplastic, mitotic inhibitor
those receiving concomitant nephrotoxic
Injection: 1, 2 mg vials (1 mg/mL)
drugs. Adjust dose in patients with renal
Pregnancy category D
impairment. Monitor renal function.
VINBLASTINE SULFATE
Indications:
Velban, generic
Treatment of acute lymphoblastic leukemia
Antineoplastic, mitotic inhibitor
(ALL), Hodgkin and non-Hodgkin lym-
Injection: 10 mg vial
phoma, neuroblastoma, Wilms tumor, and
rhabdomyosarcoma.
Pregnancy category D
Indications:
Dosage:
Treatment of Hodgkin and non-Hodgkin
Refer to individual protocol.
lymphoma, pediatric brain tumors (glio-
Children
10 kg or total body surface area
mas), Langerhans cell histiocytosis, chorio-
(TBSA) <1 m2:
carcinoma, and advanced testicular germ cell
0.05 mg/kg/dose IV once weekly; maximum
tumors.
single dose 2 mg
Children >10 kg or TBSA
1m2:
Dosage:
1 to 2 mg/m2 IV; may repeat weekly for 3 to
Refer to individual protocol.
6
weeks. Maximum single dose is
2 mg
Hodgkin lymphoma:
(some protocols allow for higher dosing in
2.5 to 6 mg/m2/dose IV once every 1 to 2
Hodgkin lymphoma and high risk ALL).
weeks for 3 to 6 cycles; maximum weekly
Neuroblastoma:
dose 12.5 mg/m2
IV continuous infusion with doxorubicin:
Langerhans cell histiocytosis:
1 mg/m2/day for 72 hours
6 mg/m2/dose IV once every 1 to 3 weeks
Brain tumors:
Notes:
6 mg/m2/dose IV weekly
Do not give to patients with known hyper-
sensitivity to vincristine. Avoid in patients
Notes:
with the demyelinating form of Charcot-
Do not give to patients with known hyper-
Marie-Tooth disease. Asparaginase may
sensitivity to vinblastine or with severe leu-
decrease clearance of vincristine. Dose mod-
kopenia. Dose modification may be needed
ification may be required in patients with
in patients with hepatic impairment or neu-
impaired hepatic function, preexisting neu-
rotoxicity. Extravasation may cause local
romuscular disease, or severe side effects of
severe tissue damage. May cause peripheral
treatment with vincristine. Extravasation
neuropathy, myelosuppression, jaw pain,
may cause local severe tissue damage.
362
Formulary
May cause peripheral neuropathy, paresthe-
Adults: 2.5 to 5 mg/24 hours PO or 10 mg/
sias, ileus, jaw pain, cranial nerve paralysis
dose IM/SC/IV
(ptosis), vocal cord paralysis, hyponatremia,
syndrome of inappropriate antidiuretic hor-
Notes:
mone (SIADH) secretion, alopecia, and
Do not give to patients with known hyper-
constipation.
sensitivity to phytonadione. Antagonizes
Intrathecal administration is fatal. Vin-
action of warfarin. Protect product from
cristine should never be taken into the proce-
light. Parenteral dosing may cause flushing,
dure room for a patient undergoing a lumbar
dizziness, cardiac or respiratory arrest,
puncture for instillation of intrathecal
hypotension, or anaphylaxis. High doses
chemotherapy.
(10 to 20 mg) in neonates may cause hyper-
bilirubinemia and severe hemolytic anemia.
VITAMIN K1/PHYTONADIONE
Monitor prothrombin time (PT), activated
AquaMEPHYTON, Mephyton
partial thromboplastin time (APTT); blood
Vitamin, water soluble
coagulation factors may increase within 6 to
Tablet: 5 mg
12 hours after oral dosing and within 1 to 2
Suspension: 1 mg/mL
hours after parenteral dosing.
Injection: 2, 10 mg/mL
VORICONAZOLE
Pregnancy category C
VFEND
Indications:
Antifungal agent
Prevention and treatment of hypopro-
Injection: 200 mg
thrombinemia caused by anticoagulants or
Powder for suspension: 200 mg/5 mL
drug-induced vitamin K deficiency; hemor-
Tablet: 50, 200 mg
rhagic disease of the newborn.
Pregnancy category D
Dosage:
Indications:
Hemorrhagic disease of the newborn:
Treatment of invasive aspergillosis, espe-
Prophylaxis: 0.5 to 1 mg IM within 1 hour of
cially in immunocompromised patients;
birth
treatment of candidemia in nonneutropenic
Treatment: 1 to 2 mg/24 hours IM, SC, or IV
patients; deep tissue Candida infections
Oral anticoagulant overdose:
including esophageal; treatment of serious
fungal infections caused by Scedosporium
Infants and children: 0.5 to 5 mg/dose PO,
apiospermum or Fusarium species in
IM, IV, or SC (give 5 mg for major bleeding,
patients intolerant or refractory to conven-
0.5 to 2.5 mg for minor bleeding)
tional antifungal therapy.
Adults: 2.5 to 10 mg/dose PO, IM, IV, or SC
Dose may be repeated 12 to 48 hours after
Dosage:
PO dose or 6 to 8 hours after parenteral dose
Limited information on pediatric dosing;
Vitamin K deficiency (due to drugs, malab-
children
12 years of age appear to require
sorption, or decreased synthesis of vitamin K
higher dosing than adults.
by the liver):
Children 2 to 11 years: loading dose 6 mg/kg/
Infants and children: 2.5 to 5 mg/24 hours PO
dose IV q12h for 2 doses on day 1 followed
or 1 to 2 mg/dose IM/IV/SC as a single dose
by maintenance dose 4 mg/kg/dose IV q12h
Formulary
363
(approximates exposure of adult dose of
Indications:
3 mg/kg/dose)
Prophylaxis and treatment of venous
Infectious Diseases Society of America (IDSA)
thromboembolic disorders; prevention of
guidelines: invasive aspergillosis: mainte-
arterial thromboembolism in patients with
nance dose 5 to 7 mg/kg/dose IV q12h
prosthetic heart valves or atrial fibrillation;
prevention of death, venous thromboembo-
Children
12 years and adults: loading dose
lism, and recurrent myocardial infarction
6 mg/kg/dose IV q12h for 2 doses on day 1;
(MI) after acute MI.
followed by maintenance dose:
Invasive aspergillosis and other deep tissue
fungal infections: 4 mg/kg/dose IV q12h
Dosage:
Candidemia: 3 to 4 mg/kg/dose IV q12h
Infants and children (to maintain an inter-
national normalized ratio
[INR] between
2 and 3):
Notes:
Initial dose: 0.2 mg/kg PO for 1 to 2 days;
Do not give to patients with known hyper-
maximum dose 10 mg
sensitivity to voriconazole. Use with cau-
Maintenance dose: 0.1 mg/kg/24 hours PO
tion in patients with known hypersensi-
daily; range
0.05
to
0.34 mg/kg/24 hours
tivity to azoles; cross-reactivity studies
(consult published charts based on INR)
have not been done. Tablets may contain
lactose and should be used with caution
Adults: 5 to 10 mg PO daily
2 to 5 days,
in patients with lactose intolerance. Avoid
adjust for desired INR; maintenance dose
concurrent administration with rifampin,
range 2 to 10 mg/day PO
CYP3A4 substrates, and ergot alkaloids.
Onset of action is within 36 to 72 hours and
Voriconazole is metabolized by cyto-
peak effects occur within 5 to 7 days. Mon-
chrome P450
enzymes and many drug
itor INR after 5 to 7 days of new dosage;
interactions exist. May cause severe
high risk patients may require more fre-
hepatic toxicity, visual changes, cardiac
quent monitoring.
arrhythmias, auditory hallucinations, and
Usual duration of therapy for first venous
photosensitivity. May cause fetal harm;
thrombotic event is 3 months (depending
must ensure contraception to avoid preg-
on elimination of procoagulant factor and
nancy. Use with caution in neonates; oral
based on resolution of clot).
suspension contains sodium benzoate, a
metabolite of benzyl alcohol, which may
cause a potentially fatal toxicity. Use with
Notes:
caution in patients with renal or hepatic
Do not give to patients with known hyper-
toxicity;
dose adjustment may be
sensitivity to warfarin, severe liver or kidney
necessary.
disease, uncontrolled bleeding, gastrointes-
tinal (GI) ulcers, status-post neurosurgical
procedures, and malignant hypertension.
WARFARIN SODIUM
Concomitant use with vitamin K may
Coumadin, Jantoven, generic
decrease anticoagulant effect. Concomitant
Anticoagulant
use with aspirin, nonsteroidal anti-inflam-
Tablets: 1, 2, 2.5, 3, 4, 5, 6, 7.5, 10 mg
matory drugs, or indomethacin may
Injection: 5 mg
increase warfarin’s anticoagulant effect and
Pregnancy category X
cause severe GI irritation. May cause fever,
364
Formulary
skin lesions, necrosis (especially in protein C
is vitamin K and prothrombin complex con-
deficiency),
hemorrhage, hemoptysis,
centrates (PCC; fresh frozen plasma if PCC
anorexia, nausea, vomiting, and diarrhea.
unavailable).
Many drug interactions exist; review all
medications prior to initiation of therapy.
The INR is the recommended test to mon-
References
itor anticoagulant effect. The absolute INR
desired is dependent on the indication and
Taketomo CK, Hodding JH, Kraus DM. Pediatric
has been extrapolated from adults. An INR
Dosage Handbook. 17th ed. Hudson, OH:
of 2 to 3 has been recommended for pro-
Lexi-Comp, 2010.
phylaxis and treatment of deep vein throm-
Physicians’ Desk Reference, 65th ed. Montvale,
bosis, pulmonary emboli, and bioprosthetic
NJ: Medical Economics Company, Inc., 2011.
heart valves. Younger children may require
Custer JW, Rau RE. The Johns Hopkins Hospital:
higher dosing. Certain foods and medica-
The Harriet Lane Handbook, A Manual for
tions may alter levels and should be
Pediatric House Officers, 18th ed. Philadel-
reviewed in detail with the patient. Antidote
phia, PA: Elsevier-Mosby, 2009.
Index
abdominal mass, 130, 178
clinical manifestations of, 152
evaluation of child with, 131
morphologic classification for, 153
ABO-incompatible transplantation
relapse, 155-6
transfusion in, 223
risk group classification, 153-6
absolute neutrophil count (ANC), 92-3, 215,
treatment, 154-5
226, 244
adenopathy
acquired von Willebrand syndrome (AVWS), 77
evaluation of child with, 126
acute chest syndrome (ACS), 19, 24-6
adrenocortical carcinoma (ACC), 204-5
etiology of, 26
afibrinogenemia, 79
laboratory evaluation of, 26
allergic transfusion reactions, 54
acute hemolytic transfusion reactions
allogeneic transplant
(ATHRs), 52-3
malignant conditions, 213
acute lymphoblastic leukemia (ALL), 144-51, 238
nonmalignant conditions, 214
clinical and laboratory features, 145
all-trans retinoic acid (ATRA), 154-5
clinical presentation, 145-6
alpha 2-antiplasmin deficiency, 80
diagnostic evaluation, 146-7
alpha-fetoprotein (AFP), 194, 203
differential diagnosis of, 147
alpha-thalassemia, 18, 36-8
initiation of therapy,
147-8
classification of, 41
risk group classification, 148-50
alveolar rhabdomyosarcoma (ARMS), 183, 185
new agents, 150
aminocaproic acid, 110
relapse, 150
amphotericin-B, 253
treatment, 148-50
analgesics
CNS directed therapy, 149
morphine, 23
complications of therapy, 150
patient-controlled analgesia, 23-4
consolidation, 149
anaplastic large cell lymphoma (ALCL), 170
delayed intensification, 149
anaplastic lymphoma kinase (ALK), 172
induction, 149
anemia, 1-17, 29-30, 47-8, 139
maintenance, 149
acute, 29-30
acute myelogenous leukemia (AML),
128,
anemic child
151-6, 244
approach to, 1-9
clinical presentation, 151-2
medical history of, 4
complications of therapy, 155
physical examination of, 5
diagnostic evaluation, 152-3
case study for, 8-9
etiology of, 151
child with, diagnostic approach to, 2
hyperleukocytosis
definition of, 1
Handbook of Pediatric Hematology and Oncology: Children’s Hospital & Research Center Oakland,
Second Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
2012 John Wiley & Sons, Ltd. Published 2012 by John Wiley & Sons, Ltd.
366
Index
anemia (Continued )
evaluation of, 62-3
erythropoietin, 6-8
history, 63
evaluation of, 1-2
initial laboratory evaluation, 63-5
full-term newborn, approach to,
7
management, 64-70
interventions, 2-8
physical examination, 63
investigation steps, 1-2
bleeding disorder, 62, 129
oral iron challenge, 2-3
menorrhagia in young women with, 67
parenteral iron therapy, 3-6
bleomycin
treatment, 139
mechanism of action, 282
Ann Arbor staging system, 168
side effects, monitoring, and treatment, 282
anorexia/weight loss, 269
utilized in, 282
anthracyclines, 285
blood
induced cardiac toxicity, 169
loss, acute, 47
antibiotics
products, irradiation of, 46
as anaerobic drugs, 248
blood transfusion
dual therapy, 248
approach to fever, 53
monotherapy, 248
chronic transfusion therapy, 27, 33-4, 57
treatment modification, 249-51
guidelines, 45
vancomycin, 248
Bloom syndrome, 144
anticoagulants, 90
body surface area (BSA), 108, 281
anticoagulation, 88-9
bone marrow aspiration, 108, 160, 296-8
antithrombin III (ATIII), 53, 282
indications, 296
arsenic trioxide (ATO), 155
materials, 296
ascorbic acid (vitamin C) , 58
post-procedure monitoring and
asparaginase, 281-2
complications, 298
mechanism of action, 281
pretreatment evaluation, 296
side effects, monitoring, and treatment,
282
procedure, 297-8
utilized in, 282
bone marrow biopsy, 296-8
ataxia telangiectasia, 144
indications, 296
autoimmune hemolytic anemia
materials, 296
(AIHA), 13, 47
post-procedure monitoring and
autoimmune neutropenia, 94
complications, 298
autologous transplant
pretreatment evaluation to, 296
malignant conditions, 213
procedure, 297-8
avascular necrosis, 30-1
bone marrow transplantation, 40, 98, 101,
214,
223, 265
bacterial infections, 55, 93
bone pain,
127
Bart’s hemoglobin, 41
bone sarcomas, 183, 187-92
Beckwith-Wiedemann syndrome, 122, 202
case for review, 191-2
benign familial neutropenia, 98
clinical presentation, 187
beta subunit of human chorionic gonadotropin
diagnostic evaluation, 188
(b-HCG), 194, 195, 203
genetics, 183-4
beta-thalassemia, 38-40
late effects, 191
intermedia, 39
pathologic diagnosis, 188-9
clinical management, 40
prognosis, 190-1
major, 40
treatment, 189-90
Bethesda titers, 84
brain tumors, 123, 136, 142, 157-65, 197-8, 213,
Bethesda unit (BU), 84
221, 270. See also central nervous system
bleeding child, approach, 62-70
tumours
case study for, 70
Burkitt lymphoma (BL), 141, 170
Index
367
cancer
gliomas, 163-5
bleeding, 129
imaging studies, 159
fever management in child with, 244-55
lumbar puncture, 160
duration of therapy, 251-2
magnetic resonance imaging, 159
febrile neutropenia, initial evaluation of,
neurosurgery, 160-1
245-7
positron emission tomography, 159
fever in nonneutropenic oncology patient,
primitive neuroectodermal tumor
255
(PNETs), 162-3
initial antibiotic treatment modification,
radiation therapy, 161
249-51
single-photon emission CT, 160
initial management, 247-9
specific tumor types, 162-5
new site infections, 254-5
treatment, 160-2
persistent fever and neutropenia (FN),
central venous catheters (CVCs), 235-43,
empiric antifungal therapy for, 252-3
247, 298
Pneumocystis jiroveci (P. carinii)
catheter-related thrombosis
pneumonia, 254
assessment and management of, 240-2
viral infection, 253-4
complications
initial symptoms of, 122
infectious, 238-40
supportive care of child with, 226-34
mechanical, 238
adverse effects, 233
types of, 237
chemotherapy induced nausea and
maintenance, 237-8
vomiting, prevention of, 229-30
types of, 235
febrile neutropenia, treatment of, 233
use of, 238
granulocyte colony-stimulating factor
chelation therapy, 57-61
(filgrastim), 230-3
institution of, 57
hematopoietic growth factors
lead toxicity, 59-61
in children, 230-3
transfusional iron overload, 57-8
immunization during chemotherapy, 228-9
chemotherapy, 136, 162, 281-90
infection prophylaxis, 226-7
asparaginase, 281-2
monitoring, 233
mechanism of action, 281
primary prophylaxis, 233
side effects, monitoring, and treatment, 282
recombinant human erythropoietin, 233-4
utilized in, 282
viral prophylaxis and treatment, 227-8
bleomycin
carboplatin, 282-3
mechanism of action, 282
mechanism of action, 282
side effects, monitoring, and treatment, 282
side effects, monitoring, and treatment, 282
utilized in, 282
utilized in, 282
cisplatin, 282-3
catheter-related infections, 238
mechanism of action, 282
catheter-related thrombosis
side effects, monitoring, and treatment, 283
assessment and management of, 240-2
utilized in, 282
central nervous system (CNS) tumors, 157-65
cyclophosphamide, 283-4
bone marrow aspiration, 160
mechanism of action, 283
bone scan, 160
side effects, monitoring, and treatment,
cause of, 157
283-4
chemotherapy, 162
utilized in, 283
clinical presentation, 157-9
cytarabine (Ara-C)
computerized tomography (CT), 159
mechanism of action, 284
diagnosis of, 157
side effects, monitoring, and treatment, 284
diagnostic evaluation, 159-60
utilized in, 284
germ cell tumors, 165
dactinomycin (actinomycin-D), 284-5
368
Index
chemotherapy (Continued )
vincristine, 289-90
mechanism of action, 285
mechanism of action, 290
side effects, monitoring, and treatment, 285
side effects, monitoring, and treatment, 290
utilized in, 285
utilized in, 290
daunorubicin, 285-6
chemotherapy induced nausea and
mechanism of action, 285
vomiting (CINV), 229
side effects, monitoring, and treatment,
treatment of, 230
285-6
chemotherapy injection, 295-6
utilized in, 285
indications, 295
etoposide
materials, 295
mechanism of action, 286
post-procedure monitoring and
side effects, monitoring, and treatment, 286
complications, 296
utilized in, 286
pretreatment evaluation, 295
extravasations treatment, 300-3
procedure, 295-6
imatinib (Gleevec
)
cisplatin, 282-3
mechanism of action, 286
side effects, monitoring, and treatment, 283
side effects, monitoring, and treatment, 286
utilized in, 282
utilized in, 286
coagulopathy, 139-40
induced nausea and vomiting, prevention of,
evaluation, 140
229-30
management, 140
intrathecal administration, 294
Cockcroft-Gault equation, 283
irinotecan, 286-7
cognitive-behavioral therapies (CBT), 263
mechanism of action, 286
combination estrogen-progestin oral
side effects, monitoring, and treatment,
contraceptive pills (COCP), 68
286-7
preparations of, 69
utilized in, 286
congenital amegakaryocytic thrombocytopenia
mercaptopurine (6-MP)
(CAMT), 118
mechanism of action, 287
constipation, 268-69
side effects, monitoring, and treatment, 287
causes of, 268
utilized in, 287
history, 268
methotrexate, 287-8
medications, 269
mechanism of action, 287
physical examination, 268-69
side effects, monitoring, and treatment,
consumptive thrombocytopenia
287-8
immune thrombocytopenic purpura (ITP), 1,
utilized in, 287
48-9, 64, 103, 111, 128
steroids
Coombs test. See direct antiglobulin test
mechanism of action, 288
craniospinal irradiation (CSI), 162
side effects, monitoring, and treatment, 288
cryoprecipitate, 50
utilized in, 288
dosing of, 51
temozolomide, 289
indications for, 50
mechanism of action, 289
Cushing’s syndrome, 204
side effects, monitoring, and treatment, 289
cyclic neutropenia, 95
utilized in, 289
cyclophosphamide, 168, 283-4
thioguanine (6-TG)
mechanism of action, 283
mechanism of action, 289
side effects, monitoring, and treatment,
side effects, monitoring, and treatment, 289
283-4
utilized in, 289
utilized in, 283
topotecan
cytarabine (Ara-C)
mechanism of action, 289
mechanism of action, 284
side effects, monitoring, and treatment, 289
side effects, monitoring, and treatment, 284
utilized in, 289
utilized in, 284
Index
369
cytomegalovirus (CMV), 228
epoetin alfa. See erythropoietin (EPO)
negative blood, 46
Erdheim-Chester disease, 207
cytosine arabinoside. See cytarabine (Ara-C)
erythropoietin (EPO), 8, 230
indications for, 8
dactinomycin (actinomycin-D), 284-5
side effects, 8
mechanism of action, 285
etoposide
side effects, monitoring, and treatment, 285
mechanism of action, 286
utilized in, 285
side effects, monitoring, and treatment, 286
dactylitis, 21
utilized in, 286
daunorubicin, 285-6
Evans syndrome, 13
mechanism of action, 285
Ewing sarcoma, 128, 183
side effects, monitoring, and treatment, 285-6
Ewing sarcoma family of tumors
utilized in, 285
(ESFTs), 184
decompression surgery, 31
active chemotherapy agents in, 190
delayed hemolytic transfusion reactions
immunophenotypic features, 189
(DHTRs), 52
excessive bruising, 63
delayed intensification (DI), 149
extravasation
Denys-Drash syndrome, 174
causes of, 300-1
desferoxamine (Desferal), 58
definition of, 300
desmopressin (DDAVP), 75, 77
of irritant drug, 300
intranasal form of, 75
recommendations to prevent, 301
diabetes insipidus (DI), 198
signs and symptoms, 303
Diamond-Blackfan anemia, 98
treatment, 302-3
diencephalic syndrome, 158
of vesicant drug, 300
diffuse large B-cell lymphoma (DLBCL), 170
direct antiglobulin testing (DAT), 13-14, 50,
faces, legs, activity, cry, consolability (FLACC)
55, 108
scale, 258
disseminated intravascular coagulation (DIC),
factor IX deficiency, 79
48, 116, 129
factor replacement therapy, 76
DNA index, 178
factor V deficiency, 79
donor selection criteria, 44
factor VII deficiency, 80
Down syndrome (DS), 122, 144, 194
factor VIII coagulant, 73
doxorubicin, 285-6
factor V Leiden mutation, 90
mechanism of action, 285
factor X deficiency, 80
side effects, monitoring, and treatment, 285-6
factor XI deficiency, 80
utilized in, 285
factor XIII deficiency, 80
dyskeratosis congenita (DKC), 98
familial adenomatous polyposis, 202
dyspnea, 271-2
Fanconi anemia, 98, 144
distress associated with, 275
fatigue, 269-71
treatment of, 271-2
model in life-threatening illness, 270
treatments for, 271
echocardiogram, 85
FDG-PET imaging. See fluorine-18-fluorodeoxy-
ektacytometry, 15
glucose positron emission tomography
electrophoresis, 41
febrile neutropenia
Embden-Meyerhof pathway (EMP), 11
initial evaluation of, 246-7
embryonal rhabdomyosarcoma (ERMS),
treatment of, 233
184-5
febrile nonhemolytic transfusion reactions
engraftment syndrome, 216, 224
(FNHTRs), 46, 52-4
epistaxis, 81
fetal alcohol syndrome, 202
management of, 65
fetal hemoglobin, 34
370
Index
fever
acute and chronic, signs and symptoms of, 219
definiton of, 245
grading of, 220
in nonneutropenic oncology patient, 255
prophylaxis and treatment, agents used for,
fever and neutropenia (FN), 245-6
220
empiric antifungal therapy for, 252-3
graft-versus-leukemia effect, 212
empiric therapy in, 252
granulocyte colony-stimulating factor (G-CSF),
evaluation and initial management of, 246
98, 212, 230-3, 255
laboratory evaluation, 247
granulocyte-macrophage colony-stimulating
on-going management of, 250
factor (GM-CSF), 230
pediatric oncology patients, risk stratification
granulocyte transfusions, 51-2
models for, 247
filgrastim. See granulocyte colony-stimulating
headaches, 28, 123, 293
factor (G-CSF)
Heinz bodies, 12
fingerstick hemoglobin, 8
hemarthroses, 81
Fitzgerald’s factor, 80
hematopoietic growth factors in children, 230-3
Fletcher’s factor, 80
hematopoietic stem cell transplantation
fluorescent in situ hybridization (FISH), 145
(HSCT), 34, 150, 212-25
fluorine-18-fluorodeoxyglucose positron
benefits of, 212
emission tomography (FDG-PET) imag-
case study for, 221-4
ing, 185, 188, 205
complications of, 216
folate
diet, 217-18
deficiencies of, 95
differential diagnosis, 224
free erythrocyte protoporphyrin (FEP), 61
graft-versus-host disease, 217
French-American-British (FAB) classification,
immune reconstitution, 221
153
infections, 216-17
fresh frozen plasma (FFP), 50, 140
late effects, 221
dosing of, 50
late sequelae, 217
indications for, 50
risks, 212
routine infection prophylaxis, 222
Gann Act, 44
sinusoidal obstructive syndrome (SOS),
Gardener syndrome, 202
216-18
germ cell tumors (GCTs), 160, 165, 193-9
surveillance in, 222
of central nervous system, 197-9
transfusion guidelines, 218-21
clinical presentation, 195-6
transplantable conditions, 212
diagnostic evaluation and risk stratification,
transplant patients, supportive care in, 217
196
types of transplantation, 212-13
epidemiology, 193-4
veno-occlusive disease (VOD), 216-17
histologic variants, 194
hematuria, 81
pathology and serum tumor markers, 194-5
hemoglobin E, 38
treatment and prognosis, 196-7
hemoglobin H, 37
gliomas, 163-5
hemoglobin H-constant spring, 37
glucose-6-phosphate dehydrogenase (G6PD)
hemoglobinopathies
deficiency, 12, 142
neonatal screening for, 40-3
diagnosis of, 13
hemoglobin S (Hgb S), 18
Mediterranean/Asian forms of, 12
hemolytic anemia, 10-17
variants of, 12
autoimmune hemolytic anemia, 13
glycogen storage disease 1b, 98
case study for, 16-17
glycoprotein Ib (GPIb), 71
child with, diagnostic approach to, 15
graft-versus-host disease (GVHD), 44, 139,
evaluation of, 14-15
213, 217
hemolytic disease of newborn, 14
Index
371
intrinsic causes, 13-14
hydroxyurea (HU)
microangiopathic hemolytic anemia, 14
therapy, 34
red cell enzyme defects, 11-13
hyperbilirubinemia, 31-2
red cell membrane disorders, 10-11
hypercalcemia, 142-3
treatment, 16
evaluation, 142-3
hemolytic transfusion reactions, 52-4
treatment, 143
hemolytic uremic syndrome (HUS), 116
hyperleukocytosis, 137-8
hemophagocytic lymphohistiocytosis (HLH),
definition of, 137
209-10
hypersplenism, 29, 117
diagnostic criteria for, 210
hyphema, 31
genetic mutations associated with, 209
hypoprothrombinemia, 79
hemophilia, 79-84
clinical presentation, 80-2
idarubicin, 285-6
definition of, 79
mechanism of action, 285
diagnosis, 82
side effects, monitoring, and treatment, 285-6
hemophilia A and B
utilized in, 285
guidelines for factor replacement in,
83
immune thrombocytopenic purpura (ITP), 103
inhibitors, 84
acute, 103-13
treatment, 82-3
natural history of, 109
hemostasis, 62
chronic, 111-13
heparin-induced thrombocytopenia
emergency therapy, 111-12
(HIT), 88, 115
evaluation, 105-9
hepatocellular carcinoma (HCC), 202-3
bone marrow examination, 129
hereditary spherocytosis (HS), 10
prognosis of, 120
family history of, 11
treatment for, 109-11
herpes simplex (HSV) infections, 228
ifosfamide, 283-4
high grade gliomas (HGG)
mechanism of action, 283
definition of, 163
side effects, monitoring, and treatment, 283-4
histiocytic disorders, 207-11
utilized in, 283
case study for, 210-11
iliopsoas hemorrhage, 81
classification of, 208
imatinib (Gleevec
)
hemophagocytic lymphohistiocytosis
mechanism of action, 286
(HLH), 209-10
side effects, monitoring, and treatment, 286
Langerhans cell histiocytosis (LCH),
utilized in, 286
207-9
immunization during chemotherapy, 228-9
Hodgkin and Reed-Sternberg (HRS) cells, 166
increased intracranial pressure
Hodgkin lymphoma (HL), 133, 166-70
(ICP), 136-7, 157
clinical presentation, 167
evaluation, 136
complications of treatment, 169-70
management, 136-7
evaluation, 167-8
indirect antiglobulin test (IAT), 13
intermediate and high-risk disease, 169
individualized care planning and coordination
low-risk disease,
169
model, 265
nodular lymphocyte predominant HL, 169
induced nausea and vomiting, prevention of,
staging, 168
229-30
treatment, 168
International Neuroblastoma Staging System,
homovanillic acid (HVA), 180
180-81
hook effect, 203
international normalized ratio (INR), 64, 89
Horner’s syndrome, 179
intracranial hemorrhage (ICH), 80-81, 105
human leukocyte antigen (HLA) system, 213
intracranial mass, 123
human platelet antigen 1a (HPA-1a), 113
intracranial pressure (ICP), 136
372
Index
intra-Ommaya reservoir tap, 295-6
low-molecular-weight heparin (LMWH), 88, 242
indications, 295
lumbar puncture (LP), 160, 185, 247, 291, 292-5
materials, 295
indications, 292
post-procedure monitoring and
materials, 293
complications, 296
post-procedure monitoring and
pretreatment evaluation, 295
complications, 294-5
procedure, 295-6
pretreatment evaluation, 292-3
intrathecal chemotherapy, 292-5
procedure, 293-4
indications, 292
lupus anticoagulant, 64
materials, 293
lymphadenopathy, 124
post-procedure monitoring and
differential diagnosis of, 124
complications, 294-5
physical examination, 125
pretreatment evaluation, 292-4
procedure, 293-4
macrophage activation syndrome (MAS), 210
intravenous immunoglobulin (IVIG), 110,
114
major histocompatibility complex (MHC), 213
involved field radiation therapy (IFRT),
malignancy
role of, 169
evaluation of child with, 122-32
irinotecan, 286-7
malignancy-associated hypercalcemia
mechanism of action, 286
(MAH), 142
side effects, monitoring, and
malignant tumors, 133
treatment, 286-7
massive hepatomegaly
utilized in, 286
evaluation, 137
iron chelators, 58
treatment, 137
iron deficiency
mean corpuscular volume (MCV), 1, 10
prevalence of, 9
mediastinal masses, 130
iron dextran, 3
membrane protein defects, 10
allergic reactions, 6
menorrhagia
management of, 68-70
Kasabach-Merritt syndrome, 117
mercaptopurine (6-MP)
ketorolac, 23
mechanism of action, 287
Knudson’s two-hit hypothesis, 200
side effects, monitoring, and treatment, 287
Kostmann syndrome, 95, 99-100
utilized in, 287
mesna, 284
lactate dehydrogenase (LDH), 16, 125
metaiodobenzylguanidine (MIBG)
Langerhans cell histiocytosis (LCH), 207-9
scan, 180
pathogenesis of, 207
methotrexate, 287-8
lead toxicity, 59-61
mechanism of action, 287
management of, 61
side effects, monitoring, and treatment, 287-8
screening for, 59-61
utilized in, 287
treatment, recommendations for, 60
microangiopathic hemolytic anemia
leukemias, acute, 144-56
(MAHA), 14, 115
evaluation of child with, 129
microcytic anemia
acute lymphoblastic leukemia (ALL), 144-8
child with, evaluation of, 6
acute myelogenous leukemia (AML), 151-6
minimal residual disease detection (MRD), 148
radiographic changes of bones, 146
mitosis-karyorrexis index, 178
leukopenia, 139
mixed lineage leukemia (MLL), 145
leukoreduction, 46
monoclonal antibodies
loss of heterozygosity (LOH), 175
epratuzumab, 150
low-estrogen oral contraceptives, 76
inotuzumab, 150
low grade gliomas (LGGs), 163
rituximab, 112
Index
373
Mosteller formula, 281
newborn
99m-technetium bone scan, 188
hemolytic disease of, 14
multimeric analysis, 73
screening, interpretation of, 41
myeloid cell surface markers, 147
nodular lymphocyte predominant Hodgkin
lymphoma (NLPHL), 166
narcotics
nongerminomatous germ cell tumors
side effects of, 24
(NGGCT), 197-9
National Marrow Donor Program, 214
diagnosis of, 161
nausea and vomiting, 267-8
non-Hodgkin lymphoma (NHL), 128, 133, 170-3
management of, 267
clinical presentation, 170-1
medications for treatment, 268
diagnostic evaluation, 171
neonatal alloimmune neutropenia, 95
histologic classification of, 170
neonatal alloimmune thrombocytopenia
staging, 171
(NAIT), 113-14
St. Jude staging system, 172
neonatal autoimmune thrombocytopenia, 114
subtypes of, 170
Neonatal Infant Pain Scale (NIPS), 257-8
treatment, 172-3
neuroblastoma, 178-82
nonimmune hemolytic anemia, 47
chemotherapy, 181
nonimmune thrombocytopenia, 115-17
clinical presentation, 178-9
noninvasive positive pressure ventilation
diagnostic evaluation, 179-80
(NIPPV), 272, 275
diagnostic studies, 180-1
decision making approach, 273-4
high-dose chemotherapy with autologous
nonrhabodomyomatous soft tissue sarcomas
transplant, 181-2
(NRSTS), 183, 187
history, 179
Nplate
,
113
laboratory studies, 179-80
nuclear medicine thyroid scintigraphy, 206
observation, 181
nutritional supplementation, 269
physical examination, 179
signs and symptoms, 179
oncologic emergencies, 133-43
staging, 180-2
anemia, 139
treatment, 181
brain herniation, 136-7
neurofibromatosis, 122
caused by abnormalities of blood and blood
neurokinin-1, 230
vessels, 137-9
neuron-specific enolase (NSE), 180
caused by space-occupying lesions, 133-4
neutropenia, 92, 102
coagulopathy, 139-40
autoimmune (See autoimmune neutropenia)
evaluation, 134
case study for, 101-2
hypercalcemia, 142-3
causes of, 94, 96-7
increased intracranial pressure, 136-7
chronic idiopathic (See chronic idiopathic
leukopenia, 139
neutropenia)
management, 134-5
cyclic (See cyclic neutropenia)
massive hepatomegaly, 137
etiology of, 93-9
metabolic emergencies, 140-2
fever management, 99-101
evaluation, 141
inherited conditions, 95
therapy, 141-2
initial evaluation, 99
tumor lysis syndrome, 140-1
management of, 99
spinal cord compression, 135-6
medications, 93
superior mediastinal syndrome, 133-4
neonatal alloimmune (See neonatal
superior vena cava syndrome, 133-4
alloimmune neutropenia)
opioids, 267
risk assessment, 92-3
opsoclonus-myoclonus-ataxia syndrome
neutrophils, 92
(OMAS), 179
374
Index
oral contraceptive pills (OCPs), 90
constipation, 268-9
oral pain therapy
dyspnea, 271-2
medications, 263
fatigue, 269-71
osmotic fragility test, 11, 15
nausea and vomiting, 267-8
osteonecrosis, 19
nutritional supplementation, 269
osteosarcomas, 184, 188
pain, 266
end of life care, 266
packed red blood cells (PRBCs), 46
individualized care planning and
transfusion, 44-6, 218
coordination, 264-6
acute blood loss, 47
questions, 276-8
autoimmune hemolytic anemia, 47
role for, 264
chronically transfused patients, 47-8
treatment, 272-5
dosing of, 48
sensation of dyspnea, 275
exchange transfusion, 48
support of gas exchange, 275
indications for, 46-8
support of respiration, 272-5
neonates, 46-7
World Health Organization definition of, 264
nonimmune hemolytic anemia, 47
parenteral iron therapy, 3-8
oncology patients, 47
Parinaud’s syndrome, 158
severe chronic anemia, 47
partial thromboplastin time (PTT), 63, 88
washing of, 46
validity of, 65
pain, 266
patient-controlled analgesia (PCA) pumps, 261
abdominal, 22
pediatric antiemetic agents, 232
assessment, 257-9
pediatric cancers
neonates and infants, 257
differential diagnosis, 123
preverbal/nonverbal children, 257-8
signs and symptoms of, 123
school aged and older children, 258-9
pediatric chemotherapeutic agents
nociceptive/neuropathic, 259
emetogenic potential, 231
nonpharmacological approaches, 261-4
penicillin
pediatric pain management
prophylaxis, 30
principles of, 266
perioperative transfusion, 33
pharmacology, 259-61
peripheral blood stem cell transplant
nonopioid analgesics, 259
(PBSCT), 213
strong opioids, 260-1
peripheral chemotherapy administration, 298-9
weak opioids, 259-60
indications, 298
therapy
materials, 299
medications, 260-61
post-procedure monitoring and
pain management, 22-4, 27, 32, 266
complications, 299
acute, 256-63
pretreatment evaluation, 298-9
in inpatient setting, 256-64
procedure, 299
pain assessment, 257-9
PET-CT, 167, 171
pain nonpharmacological approaches,
PFA-100 platelet function analyzer, 72
261-4
pit count, 29
pain pharmacology, 259-61
platelet alloantigen
world health organization analgesic
evaluation, 114
ladder, 256-7
platelet surface glycoprotein (Gp), 62
chronic, 256
platelet transfusions, 48-50
palliative care, 264-79
dosing of, 49-50
case study for, 276
indications for, 48-9
common symptoms at end of life, 266-75
Pneumocystis jiroveci (P. carinii) pneumonia, 100,
anorexia/weight loss, 269
112, 227, 246, 254
Index
375
pneumonia, 24-6
prothrombin time (PT), 63
positron emission tomography, 159
proton magnetic resonance imaging, 57
postthrombotic syndrome (PTS), 88
pulmonary sickling, 19
pretransplant preparative regimens
pyruvate kinase (PK) deficiency, 12
common agents in, 215
priapism, 27
radiation therapy (RT), 161, 189
definition of, 27
use of, 168
primary thrombophilia traits, 87
radical retroperitoneal lymph node
primitive neuroectodermal tumor (PNETs), 160,
dissection (RPLND), 205
162-3. See also Ewing sarcoma family of
radioiodine ablation, 206
tumors (ESFTs)
radioisotope bone scan, 185
primordial germ cells (PGCs), 193
rare coagulation factor deficiencies, 80
procedures
rare tumors of childhood, 200-206
bone marrow aspiration and biopsy,
296-8
adrenocortical carcinoma (ACC), 204-5
indications, 296
clinical presentation, 203
materials, 296
diagnosis and staging, 203-4
post-procedure monitoring and
liver tumors, 202-3
complications, 298
retinoblastoma, 200-202
pretreatment evaluation, 296
thyroid tumors, 205-6
procedure, 297-8
Raynaud’s phenomenon, 282
guide to, 291-9
rearranged during transfection (RET)
initiation of, 292
proto-oncogene, 206
intra-Ommaya reservoir tap and
recombinant human erythropoietin, 233-4
chemotherapy injection, 295-6
pediatric guidelines for, 233
indications, 295
red blood cells (RBCs), 1, 10
materials, 295
destruction of, 10
post-procedure monitoring and
enzyme defects, 11-13
complications, 296
membrane disorders, 10-11
pretreatment evaluation, 295
osmotic fragility of, 12
procedure, 295-6
values, 3
lumbar puncture, 292-5
respiratory failure
indications, 292
classification and features, 272
materials, 293
reticulocyte index (RI), 16
post-procedure monitoring and
reticulocytopenia, 30
complications, 294-5
retinal vascular occlusion, 31
pretreatment evaluation, 292-3
retinoblastoma, 200-202
procedure, 293-4
treatment for, 201
peripheral chemotherapy administration,
retinopathy, 19, 31
298-9
rhabdomyosarcoma (RMS), 118, 127-8, 130, 133,
indications, 298
142, 147, 180, 183, 186, 189, 202
materials, 299
Rho[D] Immune Globulin (RhoGAM ), 14
post-procedure monitoring and
ristocetin cofactor, 71
complications, 299
rituximab, 112
pretreatment evaluation, 298-9
Rosai-Dorfman disease, 207
procedure, 299
principles for, 291
sacrococcygeal tumors, 197
prolonged gingival oozing, 81
sargramostim. See granulocyte-macrophage
Promacta
,
113
colony-stimulating factor (GM-CSF)
prophylactic antibiotics, 100
screening coagulation tests
prothrombin complex concentrates (PCCs),
84
interpretation of, 66
376
Index
seizures, 158
clinical presentation, 184
selective IgA deficiency, 98
diagnostic evaluation, 184-5
septic shock, symptoms of, 245
genetics, 183-4
serotonin receptor antagonist antiemetics,
pathologic diagnosis and treatment, 185-6
ondansetron, 169
prognosis, 186-7
severe chronic anemia, 47
staging and classification, 185
severe congenital neutropenia (SCN), 95. See also
spinal cord compression, 135-6, 158
Kostmann syndrome
evaluation, 135
Shwachman-Diamond syndrome, 98
management, 135-6
sickle cell disease, 18-35
splenectomy, 11-12, 16, 29, 112
acute anemia, 29-30
splenomegaly, 95, 125
acute chest syndrome (ACS), 24-6
hemolytic anemia, 125
with acute pulmonary infiltrate, 25
immunological disease, 125
aplastic crisis, 30
infection, 125
avascular necrosis, 30-1
laboratory and imaging, 127
benign cholestasis, 32
malignancy, 126
case study for, 34-5
physical examination, 126
chronic transfusion therapy, 33-4
storage disease, 125
definition of, 18
steroids
fever and infection in, 19-20
mechanism of action, 288
fever in child, 20
side effects, monitoring, and treatment, 288
gallstones, 31-2
utilized in, 288
hematopoietic stem cell transplant in, 34
stroke (cerebrovascular accident), 28-9
hydroxyurea (HU) therapy, 34
Stroke Prevention Trial in Sickle Cell Anemia
hyperbilirubinemia, 31-2
(STOP) trial, 28
hyphema, 31
strong opioid
management, 26-7
characteristics, 262-3
pain management, 22-4
subacute bacterial endocarditis
analgesics, 23-4
prophylaxis for, 227
inpatient, 23
subarachnoid hemorrhage, 29
mild pain, 22
substance P receptors, 230
outpatient, 22
superior mediastinal syndrome, 133-4
severe pain, 22-3
superior vena cava syndrome (SVCS), 133-4
perioperative management of patients, 32-3
syndrome of inappropriate diuretic hormone
perioperative transfusion, 33
(SIADH), 284
pneumonia, 24-6
systemic lupus erythematosis (SLE), 112
priapism, 27
systemic multiagent chemotherapy, 189
retinopathy, 31
treatment of, 31
T-cell acute lymphoblastic leukemia (T-ALL), 133
splenic sequestration, 29-30
temozolomide, 289
stroke (cerebrovascular accident), 28-9
mechanism of action, 289
transfusion therapy, 33
side effects, monitoring, and treatment, 289
vaso-occlusive episodes, 21-2
utilized in, 289
single-photon emission CT, 160
thalassemia, 36-43
single-trait thrombophilia, 86
alpha-thalassemia, 36-8
sinusoidal obstructive syndrome
beta-thalassemia, 38-40
(SOS), 216-18
case study for, 42-3
treatments for, 219
hemoglobinopathies, neonatal screening
soft tissue hemorrhages, 81
for, 40-3
soft tissue sarcoma (STS), 184-7
phenotypic characteristics, 36
Index
377
thioguanine (6-TG)
bacterial infections, 55
mechanism of action, 289
case study for, 55-6
side effects, monitoring, and treatment, 289
granulocyte transfusion, 51-2
utilized in, 289
packed red blood cell transfusion,
thrombin time (TT), 63, 79
44-6
thrombocytopenia, 103-20
platelet transfusions, 48-50
acute immune thrombocytopenic
PRBC transfusion, 46-8
purpura, 103-13
hemolytic transfusion reactions, 52-4
approach to child with, 106-8
reactions, 52
case study for, 119-21
transfusion-associated acute lung injury
decreased platelet production, 117-19
(TRALI), 54
degree of, 115
transfusion-associated circulatory overload
differential diagnosis of, 104-5
(TACO), 54
drug-induced thrombocytopenia, 115
transfusion-associated graft-versus-host
neonatal alloimmune thrombocytopenia
disease (TA-GVHD), 54
(NAIT), 113-14
transfusion-associated acute lung injury
neonatal autoimmune thrombocytopenia,
(TRALI), 50, 54
114-15
transfusion-associated circulatory overload
nonimmune thrombocytopenia, 115-17
(TACO), 54
thrombocytopenia with absent radii (TAR), 118
transfusion-associated graft-versus-host disease
thromboembolism (TE)
(TA-GVHD), 54
evaluation, 241
transfusion factor, 55
thrombolysis, 88-9
transient ischemic attacks, 29
anticoagulation, 88-9
transplantation
duration of therapy, 89
conditions, 212
new agents, 89
types of, 212-13
thrombopoietin (TPO) mimetics, 112
traumatic lumbar punctures
thrombosis
definition of, 147
case study for, 89-91
trimethoprim/sulfamethoxazole
child with, 85-91
(TMP/SMX), 227, 254
evaluation of, 85-8
tumor lysis syndrome (TLS), 135, 140-1
management of, 88
tunneled central venous catheters
thrombolysis, 88-9
and related care, 236
thrombophilia, testing for, 86-7
thrombotic thrombocytopenic purpura
umbilical cord blood (UCB)
(TTP), 116
transplantation, 212
thyroid tumors, 205-6
unfractionated heparin (UH), 88-9
tissue plasminogen activator (TPA), 88
concomitant use, 89
vancomycin, 250
topotecan
indications for, 248
mechanism of action, 289
vanillylmandelic acid (VMA), 180
side effects, monitoring, and treatment, 289
Varicella Zoster Immune Globulin
utilized in, 289
(VariZIG), 228
total iron-binding capacity (TIBC), 3
vasoactive intestinal peptide (VIP), 179
total parenteral nutrition (TPN), 218
vaso-occlusive episodes (VOEs), 18, 21-2
transcranial Doppler (TCD), 28
veno-occlusive disease (VOD), 216-18
transfusion
treatments for, 219
allergic transfusion reactions, 54
venous thromboembolism (VTE), 85-6, 88,
guidelines,
44-56, 218-21
233, 240
antithrombin III (ATIII) concentrate, 51
evaluation of child with, 86
378
Index
vinblastine, 289-90
WAS protein (WASp), 99
mechanism of action, 290
white blood cells (WBCs), 1
side effects, monitoring, and treatment, 290
Wilms tumor, 174-7
utilized in, 290
clinical presentation, 174-5
vinca alkaloids, 113
evaluation, 175
vincristine, 289-90
genetics, 174-5
mechanism of action, 289-90
pathologic diagnosis, 175
side effects, monitoring, and treatment, 290
staging, 176
utilized in, 290
treatment, 176-7
vitamin B12
Wilms tumor 1 (WT1) gene, 174
deficiencies of, 95
Wiskott-Aldrich syndrome (WAS), 64, 98
von Willebrand disease (VWD), 71-8
Wong-Baker FACES Pain Rating Scale,
acquired von Willebrand syndrome
258-9
(AVWS), 77
World Health Organization (WHO), 153, 170,
classification of, 71, 73
256, 264
clinical features of, 72
analgesic ladder, 256-7
diagnosis, 72-3
adaptation of, 257
algorithm for, 74
recommendations, 256
tests, 72-3
classification system for brain
treatment, 73-7
tumors, 157
von Willebrand factor (VWF), 62, 71, 175
von Willebrand factor activity (VWF:RCo), 73
X-linked neutropenia, 98
von Willebrand factor antigen (VWF:Ag), 73
X-linked red cell enzyme deficiency, 12
~StormRG~